1
|
Seldomridge AN, Weiser R, Holder AM. Systemic Therapy for Melanoma: What Surgeons Need to Know. Surg Oncol Clin N Am 2025; 34:359-374. [PMID: 40413004 DOI: 10.1016/j.soc.2025.01.003] [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: 05/27/2025]
Abstract
Immune checkpoint inhibitors and targeted therapies (BRAF/MEK inhibitors) have transformed the care of patients with stage IV melanoma, now with 5-year overall survival rates around 50%. Surgeons should be acquainted with these drugs, the multidisciplinary considerations of their use, and the unique immune-related adverse events (irAEs) they can cause. In this review, we discuss systemic therapies for cutaneous melanoma, including the biology of immune checkpoint inhibition, treatment indications, and toxicities. We also explain how these irAEs and other toxicities can impact surgical planning and perioperative management.
Collapse
Affiliation(s)
- Ashlee N Seldomridge
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Roi Weiser
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Ashley M Holder
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| |
Collapse
|
2
|
Michaels L, Noor M, Aslam T. Clinical and imaging strategies for the assessment of the ocular side effects of systemic targeted anti-cancer therapies. Eur J Cancer 2025; 222:115452. [PMID: 40306116 DOI: 10.1016/j.ejca.2025.115452] [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: 03/06/2025] [Revised: 04/14/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025]
Abstract
Systemic targeted anti-cancer therapies selectively target cancerous cells whilst limiting systemic side effects. The eye however, is a particularly sensitive organ and the expanding use of the newer targeted chemotherapy agents has been associated with multiple ocular side effects. In this review we provide an update of the ocular side effects of the newer targeted chemotherapy agents along with suggested minimum, pragmatic, evidence-based strategies for effective screening or monitoring for potential ocular side effects. This framework is designed to guide oncologists, trial managers, protocol developers and regulatory authorities so that appropriate ophthalmic clinical examinations and non-invasive modern imaging can be requested and commissioned according to a patient's specific treatment.
Collapse
Affiliation(s)
- Luke Michaels
- St Thomas' Hospital, Westminster Bridge Road, London SE1 7EH, United Kingdom
| | - Maha Noor
- Manchester Royal Eye Hospital, Oxford Road, Manchester M13 9WL, United Kingdom
| | - Tariq Aslam
- Manchester Royal Eye Hospital, Oxford Road, Manchester M13 9WL, United Kingdom; School of Health Sciences, University of Manchester, Oxford Road, Manchester M139PL, United Kingdom.
| |
Collapse
|
3
|
Dedeilia A, Braun T, Boland GM. Melanoma in Special Populations: Pediatrics, Elders, Pregnant Women. Surg Clin North Am 2025; 105:513-541. [PMID: 40412884 DOI: 10.1016/j.suc.2024.12.003] [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: 05/27/2025]
Abstract
Melanomas in special populations constitute a unique challenge. Pediatric melanomas present with atypical histopathological features, and the most common subtypes include congenital nevi-associated, Spitzoid, and conventional melanomas. Although they present in more advanced stages than in adults, pediatric melanomas show better prognosis. In the elderly, melanomas are common and often more aggressive than in younger adults, contributing to poorer outcomes and lower survival rates. Pregnancy-associated melanomas (PAMs) do not present with a worse prognosis than all other melanomas. PAM management involves balancing maternal and fetal safety and tailoring surgical treatment according to pregnancy timeline and tumor stage.
Collapse
Affiliation(s)
- Aikaterini Dedeilia
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA. https://twitter.com/dedeilia
| | - Tatum Braun
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA; Tufts University School of Medicine, Boston, MA, USA
| | - Genevieve M Boland
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Robert C, Kicinski M, Dutriaux C, Routier É, Govaerts AS, Bührer E, Neidhardt EM, Durando X, Baroudjian B, Saiag P, Gaudy-Marqueste C, Ascierto PA, Arance A, Russillo M, Perrot JL, Mortier L, Aubin F, Dalle S, Grange F, Muñoz-Couselo E, Mary-Prey S, Amini-Adle M, Mansard S, Lebbe C, Funck-Brentano E, Monestier S, Eggermont AMM, Oppong F, Wijnen L, Schilling B, MandalÁ M, Lorigan P, van Akkooi ACJ. Combination of encorafenib and binimetinib followed by ipilimumab and nivolumab versus ipilimumab and nivolumab in patients with advanced melanoma with BRAF V600E or BRAF V600K mutations (EBIN): an international, open-label, randomised, controlled, phase 2 study. Lancet Oncol 2025; 26:781-794. [PMID: 40449497 DOI: 10.1016/s1470-2045(25)00133-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 06/03/2025]
Abstract
BACKGROUND Current first-line treatment for patients with metastatic melanoma with BRAFV600E or BRAFV600K mutations includes immunotherapy with immune checkpoint inhibitors and targeted therapy; however, the optimal sequencing of these treatments is unclear. We aimed to investigate the use of a targeted-therapy induction regimen before treatment with immune checkpoint inhibitors. METHODS This open-label, randomised, controlled, phase 2 trial (EBIN) was conducted at 37 centres in eight European countries. Eligible patients were 18 years or older and had previously untreated, unresectable, stage III or IV melanoma with BRAFV600E or BRAFV600K mutations and an Eastern Cooperative Oncology Group performance status of 0 or 1. Patients were randomly assigned (1:1) to one of two groups. Those in the induction group received targeted therapy (oral encorafenib 450 mg once a day plus oral binimetinib 45 mg twice a day for 12 weeks) followed by immune checkpoint inhibitors (intravenous nivolumab 3 mg/kg plus intravenous ipilimumab 1 mg/kg once every 3 weeks for four doses, followed by intravenous nivolumab 480 mg once every 4 weeks until unacceptable toxicity, disease progression, or 2 years of treatment). Patients in the control group received immune checkpoint inhibitors as above without any induction targeted therapy. Randomisation was conducted using a minimisation technique and was stratified by centre and a variable defined using stage and lactate dehydrogenase activity. The primary outcome was progression-free survival in the intention-to-treat population. Safety was assessed in all patients who initiated the protocol treatment. In this Article we report the primary analysis. The study is registered with ClinicalTrials.gov, NCT03235245, and is ongoing. FINDINGS Between Nov 12, 2018, and July 11, 2022, 271 patients were randomly assigned: 136 to the induction group and 135 to the control group. 103 (38%) patients were female, 168 (62%) were male, and the median age was 55 years (IQR 43-66). The median follow-up time was 21 months (IQR 13-33). There was no evidence of a longer progression-free survival in the induction group than in the control group (hazard ratio 0·87, 90% CI 0·67-1·12; p=0·36). The median progression-free survival was 9 months (95% CI 7-13) in the induction group and 9 months (5-14) in the control group. Grade 3-5 treatment-related adverse events occurred in 57 (42%) of 136 patients who started treatment in the induction group and in 42 (32%) of 131 patients who started treatment in the control group. The most common grade 3-4 treatment-related adverse event was hepatitis (17 [13%] of 136 patients in the induction group and nine [7%] of 131 patients in the control group). Serious treatment-related adverse events occurred in 45 (33%) of 136 patients in the induction group and 33 (25%) of 131 patients in the control group. There were three treatment-related deaths: two from cardiac events (heart failure and arrhythmia) in the induction group and one from meningitis in the control group. INTERPRETATION The targeted-therapy induction regimen did not improve progression-free survival compared with first-line treatment with immune checkpoint inhibitors in unselected patients with advanced melanoma with BRAFV600E or BRAFV600K mutations. FUNDING Bristol Myers Squibb and Pierre Fabre.
Collapse
Affiliation(s)
- Caroline Robert
- Department of Cancer Medicine, Gustave Roussy Cancer Campus, University of Paris-Saclay, Villejuif, France.
| | | | - Caroline Dutriaux
- Department of Dermatology, Hôpital Saint-André, CHU de Bordeaux, Bordeaux, France
| | - Émilie Routier
- Department of Cancer Medicine, Gustave Roussy Cancer Campus, University of Paris-Saclay, Villejuif, France
| | | | | | | | - Xavier Durando
- INSERM U1240 IMoST, Université Clermont Auvergne, Clermont-Ferrand, France; Département de Recherche Clinique, Délégation Recherche Clinique et Innovation, Centre Jean Perrin, Clermont-Ferrand, France; Département d'Oncologie Médicale, Centre Jean Perrin, Clermont-Ferrand, France; Centre d'Investigation Clinique UMR501, Clermont-Ferrand, France
| | - Barouyr Baroudjian
- Université Paris Cité, AP-HP Dermato-oncology, Cancer Institute AP-HP, Nord Paris Cité, INSERM U976, Saint Louis Hospital, Paris, France
| | - Philippe Saiag
- Department of General and Oncologic Dermatology, Ambroise Paré Hospital, APHP & EA 4340 "Biomarkers in cancerology and hemato-oncology", UVSQ, Université Paris-Saclay, Boulogne-Billancourt, France
| | - Caroline Gaudy-Marqueste
- Dermatology and Skin Cancer Department, Aix Marseille Univ, APHM, La Timone Hospital, Marseille, France
| | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS 'Fondazione G Pascale', Naples, Italy
| | - Ana Arance
- Department of Medical Oncology and IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - Michelangelo Russillo
- Sarcoma and Rare Tumours Departmental Unit, IRCCS Regina Elena National Cancer Institute Rome, Rome, Italy
| | - Jean-Luc Perrot
- Groupe d'Imagerie Cutanée Non Invasive (GICNI), Société Française de Dermatologie (SFD), Paris, France; Department of Dermatology, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Laurent Mortier
- Department of Dermatology, INSERM U1189, CHU Lille, CARADERM, Lille University, Lille, France
| | - Francois Aubin
- Department of Dermatology, UHC and INSERM 1098, Besançon, France
| | - Stéphane Dalle
- Dermatology Department, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France
| | - Florent Grange
- Dermatology/Oncology, CHU Reims-Hôpital Robert Debre, Reims, France; Dermatology Department, Valence Hospital, Valence, France
| | - Eva Muñoz-Couselo
- Department of Oncology, University Hospital Vall d'Hebron, Barcelona, Spain
| | - Sorilla Mary-Prey
- Service de Dermatologie, CHU de Bordeaux, Bordeaux, France; BRIC (Bordeaux Institute of Oncology), INSERM UMR1312, Université de Bordeaux, Bordeaux, France
| | | | - Sandrine Mansard
- Service de Dermatologie, Centre Hospitalo-Universitaire de Clermont Auvergne, Clermont-Ferrand, France
| | - Céleste Lebbe
- Université Paris Cité, AP-HP Dermato-oncology, Cancer Institute AP-HP, Nord Paris Cité, INSERM U976, Saint Louis Hospital, Paris, France
| | - Elisa Funck-Brentano
- Department of General and Oncologic Dermatology, Ambroise Paré Hospital, APHP & EA 4340 "Biomarkers in cancerology and hemato-oncology", UVSQ, Université Paris-Saclay, Boulogne-Billancourt, France
| | - Sandrine Monestier
- Dermatology and Skin Cancer Department Aix Marseille Univ, APHM, La Timone Hospital, Marseille, France
| | - Alexander M M Eggermont
- Board of Comprehensive Cancer Center Munich of the Technical University Munich and the Ludwig Maximilians University, Munich, Germany; Princess Máxima Center, Utrecht, Netherlands
| | | | | | - Bastian Schilling
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany; Department of Dermatology, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
| | - Mario MandalÁ
- Unit of Oncology, Santa Maria Misericordia Hospital, University of Perugia, Perugia, Italy
| | - Paul Lorigan
- Division of Cancer Sciences, University of Manchester and Christie NHS Foundation Trust, Manchester, UK
| | - Alexander C J van Akkooi
- Melanoma Institute Australia, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| |
Collapse
|
5
|
González-Barrallo I, Castellón Rubio VE, Medina J, España Fernández S, Mujika K, Majem M, Aguado C, Cabrera Suárez MÁ, Palacio I, Osterloh L, Martínez-Fernández A, García-Castaño A. Relation between dabrafenib plus trametinib-induced pyrexia and age in BRAF V600-mutated metastatic melanoma patients: A post hoc analysis of the real-world ELDERLYMEL study. Melanoma Res 2025; 35:170-175. [PMID: 39918334 PMCID: PMC12043263 DOI: 10.1097/cmr.0000000000001020] [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: 07/11/2024] [Accepted: 11/22/2024] [Indexed: 05/02/2025]
Abstract
Pyrexia is the most common adverse event in patients treated with dabrafenib plus trametinib. However, the pathogenesis of pyrexia and factors to identify patients at higher risk of developing pyrexia remain unknown. The ELDERLYMEL study was a multicenter, noninterventional, retrospective, real-world study comparing the effectiveness and safety of dabrafenib plus trametinib between elderly (≥75 years, n = 29) and younger (<75 years, n = 130) advanced melanoma BRAF V600-mutated patients in Spain. Surprisingly, pyrexia was significantly less frequent in elderly patients (13.8%) than in younger (42.3%). The post hoc analysis presented here aimed to investigate the relationship between age and pyrexia, applying logistic regression models. Patients <75 years had 4.59 more possibilities to develop pyrexia than elderly patients. The possibility of developing pyrexia increased by 1.03 as age decreased by 1 year. Receiver operating characteristics curves identified 61.5 years as the optimal cutoff value to predict the onset of pyrexia. The age-adjusted regression model revealed that patients <61.5 years had 2.53 more possibilities to develop pyrexia than those ≥61.5. This study demonstrates, for the first time, that age significantly influences the development of pyrexia in patients with BRAF V600-mutated advanced melanoma receiving dabrafenib plus trametinib. Age should be considered in the management and follow-up of these patients but should not limit treatment decisions. These findings provide important insights for clinical practice and contribute to a better understanding of pyrexia in elderly patients. The constructed nomogram based on age could serve as a useful tool for estimating the risk of pyrexia in patients receiving this treatment.
Collapse
Affiliation(s)
- Inés González-Barrallo
- Departament Medical Oncology, Hospital Clínico Universitario, Valencia, Biomedical Research Institute, INCLIVA, Valencia
| | | | - Javier Medina
- Division of Medical Oncology, Hospital Universitario de Toledo, Toledo
| | - Sofia España Fernández
- Medical Oncology Service, Catala Institute of Oncology – Badalona, Germans Trias i Pujol University Hospital, Barcelona
| | - Karmele Mujika
- Medical Oncology Department, Hospital Universitario de Donostia – Onkologikoa, Gipuzkoa
| | - Margarita Majem
- Medical Oncology Department, Hospital de la Santa Creu i Sant Pau, Barcelona
| | - Carlos Aguado
- Medical Oncology Department, Hospital Universitario Clínico San Carlos, Madrid
| | | | - Isabel Palacio
- Medical Oncology, Hospital Universitario Central de Asturias, Oviedo
| | - Lisa Osterloh
- Medical Department, Novartis Farmacéutica, S.A., Barcelona
| | | | - Almudena García-Castaño
- Department of Medical Oncology, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| |
Collapse
|
6
|
van der Hiel B, de Wit-van der Veen BJ, van den Eertwegh AJM, Vogel WV, Stokkel MPM, Lopez-Yurda M, Boellaard R, Kapiteijn EW, Hospers GAP, Aarts MJB, de Vos FYFL, Boers-Sonderen MJ, van der Veldt AAM, de Groot JWB, Haanen JBAG. Metabolic parameters on baseline and early [ 18F]FDG PET/CT as a predictive biomarker for resistance to BRAF/MEK inhibition in advanced cutaneous BRAFV600-mutated melanoma. EJNMMI Res 2025; 15:60. [PMID: 40434500 PMCID: PMC12119442 DOI: 10.1186/s13550-025-01259-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND [18F]FDG PET/CT plays a crucial role in evaluating cancer patients and assessing treatment response, including in BRAF-mutated melanoma. Metabolic tumor volume (MTV) and total lesion glycolysis (TLG) have emerged as promising alternatives to standardized uptake value (SUV)-based measures for tumor assessment. This study evaluates the predictive value of SUVpeak, MTV, and TLG in predicting progression-free survival (PFS) in advanced BRAF-mutated melanoma treated with BRAF/MEK inhibitors. RESULTS Seventy-five patients with metastatic melanoma were enrolled in a multi-center trial and treated with vemurafenib/cobimetinib. [18F]FDG-PET/CT scans were performed at baseline, week-2, and week-7. Imaging analysis included SUVpeak, MTV, and TLG of summed metastases, as well as percentage changes over time (∆). Baseline median PET-parameters were SUVpeak 12.59 (range 3.13-50.59), MTV 159mL (range 0-1897 mL), and TLG 1013 (range 1-13162). Baseline MTV was the strongest predictor (AUCT=6 months=0.714), while early changes in MTV, TLG, and especially week-7 ΔSUVpeak% showed similar or improved performance (P = 0.017 vs. baseline SUVpeak). Patients with TLG below the median had significantly prolonged PFS (15.4 vs. 8.5 months, P = 0.024). MTV above optimal cutoff (45.3 mL) was associated with an increased risk of progression/death, even after adjusting for LDH, ECOG status, and metastatic sites (HR = 2.97, 95% CI 1.17-7.52, P = 0.022). At week-2, ∆SUVpeak% was not predictive in a multivariable analysis, but became predictive at week-7 (median ∆SUVpeak%: 64), with a more than three-fold hazard of progression for patients with ∆SUVpeak% below 64% (P = 0.0014); PFS was 5.0 months (95% CI: 4.3-NA) for patients below the median versus 14.7 months (95% CI: 9.2-20.2) for those above or with non-quantifiable scans (P = 0.0002). Median ∆MTV was 95.5% at week-2 and 97.6% at week-7, with significant PFS differences at both time points (week-2: P = 0.020, week-7: P < 0.001). As expected, TLG mirrored MTV. Patients with MTV increases at week-7 after an initial response at week-2 had a median PFS of 5.3 vs. 12.6 months for those with stable or declining MTV (P = 0.0023). Intra-patient metabolic heterogeneity was also associated with outcome, with early reductions in SUVpeak variation between lesions correlating with better PFS. CONCLUSION This study supports the use of MTV and TLG as robust predictive markers for PFS in advanced melanoma treated with BRAF/MEK-inhibitors. Monitoring early PET parameters changes can provide valuable insights into therapeutic response and disease progression. TRIAL REGISTRATION Clinicaltrials.gov identifier: NCT02414750. Registered 10 April 2015, retrospectively registered.
Collapse
Affiliation(s)
- Bernies van der Hiel
- Department of Nuclear Medicine, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Plesmanlaan 121- Room C0.137, Amsterdam, 1066 CX, The Netherlands.
| | - Berlinda J de Wit-van der Veen
- Department of Nuclear Medicine, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Plesmanlaan 121- Room C0.137, Amsterdam, 1066 CX, The Netherlands
| | - Alfons J M van den Eertwegh
- Department of Medical Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Wouter V Vogel
- Department of Nuclear Medicine, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Plesmanlaan 121- Room C0.137, Amsterdam, 1066 CX, The Netherlands
| | - Marcel P M Stokkel
- Department of Nuclear Medicine, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Plesmanlaan 121- Room C0.137, Amsterdam, 1066 CX, The Netherlands
| | - Marta Lopez-Yurda
- Department of Biometrics, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Department of Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ellen W Kapiteijn
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Geke A P Hospers
- Department of Medical Oncology, University Medical Center Groningen, Groningen, The Netherlands
| | - Maureen J B Aarts
- Department of Medical Oncology, GROW-School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Filip Y F L de Vos
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marye J Boers-Sonderen
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | | | - John B A G Haanen
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Medical Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Carvalho DG, Kenski JCN, Moreira DA, Rajão MA, Krijgsman O, Furtado C, Boroni M, Viola JPB, Peeper DS, Possik PA. Resistance to BRAF inhibitors drives melanoma sensitivity to Chk1 inhibition. Pharmacol Res 2025; 217:107797. [PMID: 40414585 DOI: 10.1016/j.phrs.2025.107797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 05/20/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
BRAF inhibitor-resistant melanomas (BRAFiR) acquire (epi)genetic and functional alterations that enable them to evade alternative treatments. Identifying these alterations is critical to advancing treatment strategies. Here, we explored the effect of Chk1 inhibition (Chk1i) on BRAFiR cells, revealing higher sensitivity compared to treatment-naïve cells both in vitro and in vivo. Using FUCCI-labeling and time-lapse microscopy, we show that S phase progression is required for Chk1i-induced cytotoxicity in BRAFiR cells, but not in treatment-naïve cells. Replication stress markers, including reduced BrdU incorporation and increased phospho-RPA and γH2AX, were observed mostly in BRAFiR cells with increased sensitivity to Chk1i. Untreated BRAFiR cells exhibited upregulated DNA replication genes, reduced progressing forks and increased origin firing, suggesting intrinsic replication changes. MAPK pathway reactivation in treatment-naïve cells mimicked BRAFiR traits, increasing sensitivity to Chk1i. These findings indicate that Chk1i exploits elevated replication stress specifically in BRAFiR cells, highlighting its therapeutic potential in overcoming MAPK inhibitor resistance in BRAF600-mutant melanoma.
Collapse
Affiliation(s)
- Danielle G Carvalho
- Program of Immunology and Tumor Biology, Division of Basic and Experimental Research, Brazilian National Cancer Institute, Rio de Janeiro, Brazil; Division of Molecular Oncology and Immunology, Oncode institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Juliana C N Kenski
- Division of Molecular Oncology and Immunology, Oncode institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Daniel A Moreira
- Laboratory of Bioinformatics and Computational Biology, Division of Basic and Experimental Research, Brazilian National Cancer Institute, Rio de Janeiro, Brazil; Bioinformatics Core Facility, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Matheus A Rajão
- Program of Immunology and Tumor Biology, Division of Basic and Experimental Research, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - Oscar Krijgsman
- Division of Molecular Oncology and Immunology, Oncode institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Carolina Furtado
- Genomics core facility, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - Mariana Boroni
- Laboratory of Bioinformatics and Computational Biology, Division of Basic and Experimental Research, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - João P B Viola
- Program of Immunology and Tumor Biology, Division of Basic and Experimental Research, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - Daniel S Peeper
- Division of Molecular Oncology and Immunology, Oncode institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - Patricia A Possik
- Program of Immunology and Tumor Biology, Division of Basic and Experimental Research, Brazilian National Cancer Institute, Rio de Janeiro, Brazil; Wellcome Sanger Institute, Hinxton, Cambridgeshire, United Kingdom.
| |
Collapse
|
8
|
Hanna KS, Palaia J, Patel D, Moshyk A, Zhou ZY, Yang F, Xin Y, Garcia-Horton V. Cost per outcome of nivolumab + relatlimab vs BRAF + MEK inhibitor combinations for first-line treatment of BRAF-mutant advanced melanoma. J Manag Care Spec Pharm 2025:1-9. [PMID: 40391872 DOI: 10.18553/jmcp.2025.25015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
BACKGROUND The National Comprehensive Cancer Network guidelines list combination immunotherapy as the preferred first-line (1L) treatment for unresectable or metastatic melanoma over BRAF and MEK inhibitor (BRAFi/MEKi) therapy, regardless of BRAF mutation status. However, the economic impact of 1L treatment with nivolumab plus relatlimab (NIVO + RELA) vs BRAFi/MEKi therapies for BRAF-mutated advanced melanoma has not been assessed. OBJECTIVE To compare the health care costs, cost per progression-free life-year (PFLY), and cost per life-year (LY) of NIVO + RELA vs dabrafenib plus trametinib (DAB + TRAM), encorafenib plus binimetinib (ENCO + BINI), and vemurafenib plus cobimetinib (VEM + COBI) as 1L treatment for BRAF-mutated, unresectable or metastatic melanoma. METHODS A cost-per-outcome model compared the economic value of NIVO + RELA vs each BRAFi/MEKi therapy. Clinical inputs were derived from previous matching-adjusted indirect comparisons using individual patient data from the BRAF-mutant subgroup of RELATIVITY-047 and published data pooled from COMBI-d, COMBI-v, COLUMBUS, and coBRIM. LYs, PFLYs per investigator, and treatment duration were estimated using the restricted mean survival time. Health care costs (2024 US dollars), including drug acquisition and administration costs, disease management costs over the preprogression and postprogression periods, and adverse event management costs, were calculated over 5 years. Several scenario analyses were performed, including adding subsequent treatment costs. RESULTS Over 5 years, NIVO + RELA was associated with improved PFLYs and LYs compared with DAB + TRAM (mean PFLY: 1.94 vs 1.82 years, mean LY: 3.41 vs 2.77 years), ENCO + BINI (1.87 vs 1.78 years and 3.40 vs 2.91 years, respectively), and VEM + COBI (2.12 vs 1.80 years and 3.39 vs 2.63 years). The estimated total costs over 5 years were lower for NIVO + RELA vs DAB + TRAM ($300,479 vs $519,770), ENCO + BINI ($343,996 vs $572,556), and VEM + COBI ($296,361 vs $317,851). Main cost drivers were drug acquisition and administration costs. NIVO + RELA had lower costs per PFLY and per LY than DAB + TRAM ($155,107 vs $285,617 and $88,203 vs $187,699, respectively); ENCO + BINI ($183,628 vs $322,113 and $101,151 vs $196,924); and VEM + COBI ($139,688 vs $176,645 and $87,315 vs $121,086). The sensitivity analyses' results supported the base-case results. CONCLUSIONS NIVO + RELA showed improved LYs and PFLYs at lower cost than all 3 BRAFi/MEKi comparators over 5 years. These results support the economic value of NIVO + RELA for patients with previously untreated, BRAF-mutated, unresectable or metastatic melanoma.
Collapse
Affiliation(s)
| | | | | | | | | | - Fan Yang
- Analysis Group, Inc., London, United Kingdom
| | - Yiqiao Xin
- Analysis Group, Inc., London, United Kingdom
| | | |
Collapse
|
9
|
Zhang P, Zhang Y, Sun Y, Chen Y, Cao X, He Y, Tan Y, Zhang Z, Deng S, Zhou W, Zhong C, Zeng A, Liu H, Xu JY, Zhou Y, Shen S, Li P, Li Y. Discovery of 2-Amino-7-Amide Quinazoline Derivatives as Potent and Orally Bioavailable Inhibitors Targeting Extracellular Signal-Regulated Kinase 1/2. J Med Chem 2025. [PMID: 40370105 DOI: 10.1021/acs.jmedchem.5c01055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Aberrant activation of the ERK/MAPK pathway is closely associated with various cancers. Directly targeting ERK1/2, the most distal node of this cascade, is not only a rational therapeutic approach for cancers harboring pathway-activating alterations, but also provides a potential solution for overcoming resistance from upstream signaling. Herein, we described the discovery of potent and orally bioavailable ERK1/2 inhibitors featuring 2-amino-7-amide quinazoline skeletons through structure-based drug design. Among them, the optimal compound 23 inhibited ERK1/2 at single-digital nanomolar concentrations with good specificity, and exhibited great potencies in preventing cell growth, migration and invasion, disrupting cell cycle, and inducing cell apoptosis. Further mechanism studies demonstrated that 23 dose-dependently suppressed the phosphorylation of the downstream substrate RSK. Remarkably, 23 exerted favorable ADMET and PK profiles, as well as significant in vivo antitumor efficacy with excellent tolerance. Collectively, this work offers a novel and highly promising candidate targeting ERK1/2 for further drug development.
Collapse
Affiliation(s)
- Peili Zhang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Yu Zhang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Yuelan Sun
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Yanyan Chen
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Xiang Cao
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Yu He
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Ye Tan
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Zian Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shengyi Deng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenjuan Zhou
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Chuhai Zhong
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Ai Zeng
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Huanhuan Liu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Jun-Yu Xu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yubo Zhou
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Shiyang Shen
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ping Li
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yi Li
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
10
|
Jia DD, Xu Y, Ren ZW, Li LQ, Zhang L, Li Y, Lou LS, Yao WT, Liu Z, Li XA, Yang JL, Chen Y, Li T. Is One Year Enough? Extended Adjuvant Dabrafenib Plus Trametinib for Chinese Patients With Resected Stage III Melanoma. J Dermatol 2025. [PMID: 40366077 DOI: 10.1111/1346-8138.17779] [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: 04/21/2025] [Revised: 04/21/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Pathogenic BRAF mutations drive constitutive MAPK pathway activation in melanoma, and targeted therapies with dabrafenib plus trametinib have improved outcomes in the adjuvant setting. However, the optimal duration of adjuvant therapy remains unclear. This retrospective study examined whether extending dabrafenib plus trametinib beyond 1 year offers additional clinical benefit in Chinese patients with resected Stage III melanoma. METHODS Medical records from six centers were reviewed for adults with BRAF V600E/K-positive, completely resected Stage III melanoma who received at least 12 months of adjuvant dabrafenib plus trametinib. Patients were divided into a 1-year therapy group and a more-than-1-year therapy group. Relapse-free survival (RFS) was the primary end point; adverse events were also assessed. RESULTS Of the 122 patients included, 77 received more than 1 year of adjuvant therapy. The more-than-1-year group experienced significantly better RFS (log-rank p = 0.04), and longer therapy independently reduced recurrence risk in multivariate analysis (HR, 2.42; p = 0.035). Adverse event profiles did not differ between groups, and toxicity-related treatment modifications occurred primarily within the first year. CONCLUSIONS Extending dabrafenib plus trametinib beyond 1 year may provide improved RFS without increasing toxicity. Further prospective trials are warranted to confirm the impact on overall survival and identify optimal patient subsets for prolonged therapy.
Collapse
Affiliation(s)
- Dong-Dong Jia
- Department of Bone and Soft-Tissue Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, Hangzhou, China
| | - Yu Xu
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhi-Wu Ren
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Lin-Qing Li
- Department of Bone and Soft-Tissue Surgery, Hunan Cancer Hospital, Changsha, Hunan, China
| | - Lei Zhang
- Bone and Soft Tissue Oncology Department, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
| | - Yang Li
- Department of Bone and Soft Tissue, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Li-Shu Lou
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, Hangzhou, China
| | - Wei-Tao Yao
- Department of Bone and Soft Tissue, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Zhe Liu
- Bone and Soft Tissue Oncology Department, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
| | - Xian-An Li
- Department of Bone and Soft-Tissue Surgery, Hunan Cancer Hospital, Changsha, Hunan, China
| | - Ji-Long Yang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yong Chen
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tao Li
- Department of Bone and Soft-Tissue Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, Hangzhou, China
| |
Collapse
|
11
|
Sugitani I, Kiyota N, Ito Y, Onoda N, Hiromasa T, Horiuchi K, Kinuya S, Kondo T, Moritani S, Sugino K, Hara H. The 2024 revised clinical guidelines on the management of thyroid tumors by the Japan Association of Endocrine Surgery. Endocr J 2025; 72:545-635. [PMID: 40058844 PMCID: PMC12086281 DOI: 10.1507/endocrj.ej24-0644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 12/31/2024] [Indexed: 05/09/2025] Open
Abstract
The Japan Association of Endocrine Surgery published the first edition of the "Clinical guidelines on the management of thyroid tumors" in 2010 and the revised edition in 2018. The guideline presented herein is the English translation of the revised third edition, issued in 2024. The aim is to enhance health outcomes for patients suffering from thyroid tumors by facilitating evidence-based shared decision-making between healthcare providers and patients, as well as standardizing the management of thyroid tumors. The focus is on adult patients with thyroid tumors, addressing clinically significant issues categorized into areas such as an overview of the diagnosis and treatment of thyroid nodules, treatment strategies by histological type, radioactive iodine therapy, treatment of advanced differentiated carcinoma, pharmacotherapy, and complications and safety management associated with thyroid surgery. Thirty-two clinical questions were established in these areas. Following a comprehensive search of the literature and systematic review to evaluate the overall evidence, we aimed to present optimal recommendations by considering the balance of benefits and harms from the patient's perspective. We integrated evidence and clinical experience to determine the "Certainty of evidence" and "Strength of recommendations". Based on these, we illustrated overall flows of care as "Clinical algorithms". Necessary background knowledge of diseases and established clinical procedures for understanding the recommendations are presented in "Notes", while information that may be clinically useful but for which evidence remains insufficient is included in "Columns", based on the current state of evidence. Finally, future challenges for the next revision are presented as "Future research questions".
Collapse
Affiliation(s)
- Iwao Sugitani
- Department of Endocrine Surgery, Nippon Medical School, Tokyo 113-8603, Japan
- the Task Force of the Japan Association of Endocrine Surgery on the Guidelines for Thyroid Tumors
| | - Naomi Kiyota
- Department of Medical Oncology and Hematology, Cancer Center, Kobe University Hospital, Kobe 650-0017, Japan
- the Task Force of the Japan Association of Endocrine Surgery on the Guidelines for Thyroid Tumors
| | - Yasuhiro Ito
- Department of Surgery, Kuma Hospital, Kobe 650-0011, Japan
- the Task Force of the Japan Association of Endocrine Surgery on the Guidelines for Thyroid Tumors
| | - Naoyoshi Onoda
- Department of Surgery, Kuma Hospital, Kobe 650-0011, Japan
- the Task Force of the Japan Association of Endocrine Surgery on the Guidelines for Thyroid Tumors
| | - Tomo Hiromasa
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa 920-8641, Japan
- the Task Force of the Japan Association of Endocrine Surgery on the Guidelines for Thyroid Tumors
| | - Kiyomi Horiuchi
- Department of Endocrine Surgery, Tokyo Women’s Medical University, Tokyo 162-8666, Japan
- the Task Force of the Japan Association of Endocrine Surgery on the Guidelines for Thyroid Tumors
| | - Seigo Kinuya
- Department of Nuclear Medicine, Kanazawa University, Kanazawa 920-8641, Japan
- the Task Force of the Japan Association of Endocrine Surgery on the Guidelines for Thyroid Tumors
| | - Tetsuo Kondo
- Department of Pathology, University of Yamanashi, Yamanashi 409-3898, Japan
- the Task Force of the Japan Association of Endocrine Surgery on the Guidelines for Thyroid Tumors
| | - Sueyoshi Moritani
- Center for Head and Neck Thyroid Surgery, Oumi Medical Center, Shiga 525-8585, Japan
- the Task Force of the Japan Association of Endocrine Surgery on the Guidelines for Thyroid Tumors
| | - Kiminori Sugino
- Surgical Branch, Ito Hospital, Tokyo 150-8308, Japan
- the Task Force of the Japan Association of Endocrine Surgery on the Guidelines for Thyroid Tumors
| | - Hisato Hara
- Department of Breast and Endocrine Surgery, University of Tsukuba, Tsukuba 305-8576, Japan
- the Task Force of the Japan Association of Endocrine Surgery on the Guidelines for Thyroid Tumors
| |
Collapse
|
12
|
Borbényi-Galambos K, Erdélyi K, Ditrói T, Jurányi EP, Szántó N, Szatmári R, Czikora Á, Schmidt EE, Garai D, Cserepes M, Liszkay G, Tóth E, Tóvári J, Nagy P. Realigned transsulfuration drives BRAF-V600E-targeted therapy resistance in melanoma. Cell Metab 2025; 37:1171-1188.e9. [PMID: 40037361 DOI: 10.1016/j.cmet.2025.01.021] [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: 08/26/2023] [Revised: 12/13/2024] [Accepted: 01/22/2025] [Indexed: 03/06/2025]
Abstract
BRAF V600E-inhibition effectively treats melanoma, but acquired resistance rapidly develops. Protein expression profiles, mitochondrial energetics, metabolomics and fluxomics data in cell line, xenograft, and patient-derived xenograft systems revealed that concerted reprogramming of metabolic pathways (including glutaminolysis, glycolysis, TCA cycle, electron transport chain [ETC], and transsulfuration), along with an immediate cytoprotective response to drug-induced oxidative stress, underpins drug-tolerant persister cancer cell survival. Realignment of cysteine (Cys) metabolism, in particular an immediate upregulation of cystathionine-γ-lyase (CSE), was vital in persister cells. The oxidative cellular environment, drug-induced elevated cystine uptake and oxidative Cys catabolism, increased intracellular cystine/Cys ratios, thereby favoring cystine as a CSE substrate. This produces persulfides and hydrogen sulfide to protect protein thiols and support elevated energy demand in persister cells. Combining BRAF V600E inhibitors with CSE inhibitors effectively diminished proliferative relapse in culture models and increased progression-free survival of xenografted mice. This, together with induced CSE expression in patient samples under BRAF-V600E-inhibition, reveals an approach to increase BRAF-V600E-targeted therapeutic efficacy.
Collapse
Affiliation(s)
- Klaudia Borbényi-Galambos
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary; Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hajdú-Bihar County, 4032, Hungary
| | - Katalin Erdélyi
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary
| | - Tamás Ditrói
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary
| | - Eszter Petra Jurányi
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary; Semmelweis University Doctoral School, Semmelweis University, Budapest, 1094, Hungary
| | - Noémi Szántó
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary
| | - Réka Szatmári
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary; Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hajdú-Bihar County, 4032, Hungary; Chemistry Coordinating Institute, University of Debrecen, Debrecen, Hajdú-Bihar County, 4012, Hungary
| | - Ágnes Czikora
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary
| | - Edward E Schmidt
- Department of Anatomy and Histology, HUN-REN-UVMB Laboratory of Redox Biology, University of Veterinary Medicine, Budapest, 1078, Hungary; Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, 59717, United States of America
| | - Dorottya Garai
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary; Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hajdú-Bihar County, 4032, Hungary
| | - Mihály Cserepes
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary
| | - Gabriella Liszkay
- Department of Dermatology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary
| | - Erika Tóth
- Department of Surgical and Molecular Pathology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary
| | - József Tóvári
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary
| | - Péter Nagy
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary; Chemistry Coordinating Institute, University of Debrecen, Debrecen, Hajdú-Bihar County, 4012, Hungary; Department of Anatomy and Histology, HUN-REN-UVMB Laboratory of Redox Biology, University of Veterinary Medicine, Budapest, 1078, Hungary.
| |
Collapse
|
13
|
Roerden M, Spranger S. Cancer immune evasion, immunoediting and intratumour heterogeneity. Nat Rev Immunol 2025; 25:353-369. [PMID: 39748116 DOI: 10.1038/s41577-024-01111-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2024] [Indexed: 01/04/2025]
Abstract
Cancers can avoid immune-mediated elimination by acquiring traits that disrupt antitumour immunity. These mechanisms of immune evasion are selected and reinforced during tumour evolution under immune pressure. Some immunogenic subclones are effectively eliminated by antitumour T cell responses (a process known as immunoediting), which results in a clonally selected tumour. Other cancer cells arise to resist immunoediting, which leads to a tumour that includes several distinct cancer cell populations (referred to as intratumour heterogeneity (ITH)). Tumours with high ITH are associated with poor patient outcomes and a lack of responsiveness to immune checkpoint blockade therapy. In this Review, we discuss the different ways that cancer cells evade the immune system and how these mechanisms impact immunoediting and tumour evolution. We also describe how subclonal antigen presentation in tumours with high ITH can result in immune evasion.
Collapse
Affiliation(s)
- Malte Roerden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute for Technology, Cambridge, MA, USA
| | - Stefani Spranger
- Koch Institute for Integrative Cancer Research, Massachusetts Institute for Technology, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute for Technology, Cambridge, MA, USA.
- Ragon Institute of Mass General Hospital, Massachusetts Institute for Technology and Harvard, Cambridge, MA, USA.
| |
Collapse
|
14
|
Steinberg FT, Simon M, Wawer-Matos Reimer PA, Rokohl AC, Heindl LM. [New systemic treatment approaches for conjunctival melanoma]. DIE OPHTHALMOLOGIE 2025; 122:349-356. [PMID: 40067450 DOI: 10.1007/s00347-025-02207-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/30/2024] [Accepted: 02/05/2025] [Indexed: 05/08/2025]
Abstract
Conjunctival melanoma is a rare disease that nevertheless has a high tumor-associated mortality rate. A resection in sano with adjuvant local treatment currently represents the therapeutic gold standard and systemic treatment is used for metastasized conjunctival melanoma and/or very advanced nonresectable local findings. New knowledge on molecular changes in conjunctival melanoma shows a clear similarity to those of cutaneous melanoma. Therefore, many findings on new systemic forms of treatment for cutaneous melanoma can be transferred to conjunctival melanoma. In the clinical application BRAF/MEK inhibitors and immune checkpoint inhibitors are already in use and good response rates have been shown in small retrospective studies and case reports. Due to the rarity of conjunctival melanoma, there are no larger prospective studies comparing different systemic therapeutic agents. In a nonrandomized retrospective comparison, a better overall survival was shown for a combination of BRAF/MEK inhibitors (progression-free 1‑year survival probability of 54.7%; overall survival of 29.1 months) compared to a combination of PD1/CTLA4 antibodies (progression-free 1‑year survival probability of 42%; overall survival of 18 months). The current recommendation is to perform genomic profiling for every conjunctival melanoma, particularly to investigate a BRAF mutation. If a BRAF mutation is present, BRAF/MEK inhibitor treatment should preferably be initiated. Treatment with immune checkpoint inhibitors can be used in the case of BRAF-negative mutation status or treatment failure with BRAF/MEK inhibitors. Monotherapy with the CTLA4 antibody ipilimumab is not recommended due to its inferiority to PD1 antibodies. New knowledge in the genomic profiling of conjunctival melanoma could enable further targeted treatment options in the future.
Collapse
Affiliation(s)
| | - Michael Simon
- Zentrum für Augenheilkunde, Universitätsklinikum Köln, Köln, Deutschland
| | | | - Alexander C Rokohl
- Zentrum für Augenheilkunde, Universitätsklinikum Köln, Köln, Deutschland
| | - Ludwig M Heindl
- Zentrum für Augenheilkunde, Universitätsklinikum Köln, Köln, Deutschland
| |
Collapse
|
15
|
Ueki Y, Naylor RM, Ghozy SA, Thirupathi K, Rinaldo L, Kallmes DF, Kadirvel R. Advances in sporadic brain arteriovenous malformations: Novel genetic insights, innovative animal models and emerging therapeutic approaches. J Cereb Blood Flow Metab 2025; 45:793-799. [PMID: 39948029 PMCID: PMC11826813 DOI: 10.1177/0271678x251319913] [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: 07/10/2024] [Revised: 01/27/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025]
Abstract
Brain arteriovenous malformations (bAVMs) are a notable cause of intracranial hemorrhage, strongly associated with severe morbidity and mortality. Contemporary treatment options include surgery, stereotactic radiosurgery, and endovascular embolization, each of which has limitations. Hence, development of pharmacological interventions is urgently needed. The recent discovery of the presence of activating Kirsten rat sarcoma (KRAS) viral oncogene homologue mutations in most sporadic bAVMs has opened the door for a more comprehensive understanding of the pathogenesis of bAVMs and has pointed to entirely novel possible therapeutic targets. Herein, we review the status quo of genetics, animal models, and therapeutic approaches in bAVMs.
Collapse
Affiliation(s)
- Yasuhito Ueki
- Department of Neurologic Surgery, Mayo Clinic Rochester, MN, USA
- Department of Neurosurgery, Faculty of Medicine, The University of Juntendo, Tokyo, Japan
| | - Ryan M Naylor
- Department of Neurologic Surgery, Mayo Clinic Rochester, MN, USA
| | - Sherief A Ghozy
- Department of Neurologic Surgery, Mayo Clinic Rochester, MN, USA
| | | | - Lorenzo Rinaldo
- Department of Neurologic Surgery, Mayo Clinic Rochester, MN, USA
| | | | - Ramanathan Kadirvel
- Department of Neurologic Surgery, Mayo Clinic Rochester, MN, USA
- Department of Radiology, Mayo Clinic Rochester, MN, USA
| |
Collapse
|
16
|
Nakano F, Kan T, Ushio Y, Ito H, Kamegashira A, Sone T, Hiyama T, Harada Y, Tanaka A. A case of malignant melanoma in which adalimumab was effective for steroid-resistant uveitis caused by dabrafenib/trametinib combination therapy. J Dermatol 2025; 52:e334-e336. [PMID: 39698756 DOI: 10.1111/1346-8138.17594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/29/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024]
Affiliation(s)
- Fumi Nakano
- Department of Dermatology, The Medical Bulletin of Onomichi General Hospital, Onomichi, Japan
| | - Takanobu Kan
- Department of Dermatology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yukiko Ushio
- Department of Dermatology, The Medical Bulletin of Onomichi General Hospital, Onomichi, Japan
| | - Haruna Ito
- Department of Dermatology, The Medical Bulletin of Onomichi General Hospital, Onomichi, Japan
| | - Akiko Kamegashira
- Department of Dermatology, The Medical Bulletin of Onomichi General Hospital, Onomichi, Japan
| | - Takashi Sone
- Department of Ophthalmology, The Medical Bulletin of Onomichi General Hospital, Onomichi, Japan
| | - Tomona Hiyama
- Department of Ophthalmology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yosuke Harada
- Department of Ophthalmology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Akio Tanaka
- Department of Dermatology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
17
|
Alamoudi MK, Alsaleh AA, Thyagarajan A, Alkholifi FK, Raza ML, Sahu RP. Elevated HDAC4 Expression Is Associated with Reduced T-Cell Inflamed Tumor Microenvironment Gene Signatures and Immune Checkpoint Inhibitor Effectiveness in Melanoma. Cancers (Basel) 2025; 17:1518. [PMID: 40361444 PMCID: PMC12070970 DOI: 10.3390/cancers17091518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/17/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
Background/Objectives: Melanoma remains a difficult malignancy to treat because it employs tolerance mechanisms like negative immune checkpoint (IC) molecules to avoid antitumor immune responses. Thus, immune checkpoint inhibitors (ICIs) are increasingly used to treat melanoma. However, many patients do not respond, indicating resistance mechanisms like intrinsic tumor characteristics and an immunosuppressive tumor microenvironment (TME). An inflamed TME was associated with improved ICI efficacy by upregulating the T-cell inflamed TME gene signatures, an array of genes associated with dendritic cells (DCs) and cytotoxic CD8+ T-cell-mediated anti-tumor responses. As histone deacetylases (HDACs) have been shown to play crucial roles in regulating gene expression and aberrant HDAC expression has been reported in melanoma and also implicated in the regulation of IC, programmed cell death protein 1 (PD-1), and its ligand (PD-L1) and various immune evasion genes, we investigated the relationship between T-cell inflamed TME gene signatures and the HDAC family, particularly HDAC4. Methods: We used the skin cutaneous melanoma (SKCM) database, ICI-pretreated melanoma dataset, and other platforms including cBioPortal, TIMER 2.0, TISIDB, and UALCAN for the analysis. Results: We identified that high HDAC4 expression negatively modulated the TME by decreasing the abundance of DCs and cytotoxic CD8+ T-cells. The group of melanoma patients with elevated HDAC4 expression exhibited not only poor prognosis but also diminished transcription of T-cell inflamed TME gene signatures and increased DNA methylation of T-cell inflamed TME gene signatures. Importantly, elevated HDAC4 expression was associated with decreased CD8+ T-cells and a decreased ESTIMATE immune score in ICI-pretreated melanoma patients. Conclusions: Our findings suggest that HDAC4 may transform the TME into a non-inflamed phenotype, thereby reducing ICI efficacy in melanoma. Overall, this research shows that a combination of HDAC4 inhibitors and ICIs could result in better melanoma prognosis.
Collapse
Affiliation(s)
- Mariam K. Alamoudi
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia;
| | - Abdulmonem A. Alsaleh
- Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard-Health Affairs (MNG-HA), Riyadh 11481, Saudi Arabia;
| | - Anita Thyagarajan
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA;
| | - Faisal K. Alkholifi
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia;
| | - Muhammad Liaquat Raza
- Department of Infection Prevention Control, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNG-HA), Riyadh 11481, Saudi Arabia;
| | - Ravi P. Sahu
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA;
| |
Collapse
|
18
|
Mechahougui H, Gutmans J, Gouasmi R, Smekens L, Friedlaender A. BRAF Targeting Across Solid Tumors: Molecular Aspects and Clinical Applications. Int J Mol Sci 2025; 26:3757. [PMID: 40332392 PMCID: PMC12027668 DOI: 10.3390/ijms26083757] [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: 03/08/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
BRAF mutations are critical drivers in cancers such as melanoma, colorectal cancer, and non-small-cell lung cancer. The most common mutation, BRAF V600E, is a key therapeutic target. Targeted treatments with BRAF and MEK inhibitors have significantly improved progression-free and overall survival in melanoma patients. However, in cancers like metastatic colorectal cancer, BRAF mutations are associated with poor outcomes due to aggressive disease behavior and resistance to conventional chemotherapy. Despite progress, resistance to BRAF/MEK inhibitors remains a major challenge, often driven by secondary mutations in the mitogen-activated protein kinase (MAPK) pathway, activation of alternative pathways such as phosphoinositide 3-kinases (PI3Ks)/protein kinase B (AKT), or changes in the tumor microenvironment. These challenges have motivated ongoing research into combining BRAF inhibitors with immunotherapies to enhance and prolong treatment effectiveness. Future research must also account for the role of the cancer's tissue of origin, as the biological context significantly influences response to targeted therapies, highlighting the need for a deeper understanding of tumor biology, micro-environment, and genetics.
Collapse
Affiliation(s)
- Hiba Mechahougui
- Oncology Department, Geneva University Hospital (HUG), 1205 Geneva, Switzerland; (J.G.); (L.S.)
| | - James Gutmans
- Oncology Department, Geneva University Hospital (HUG), 1205 Geneva, Switzerland; (J.G.); (L.S.)
| | - Roumaïssa Gouasmi
- Cancer Research Center of Lyon, CNRS UMR5286, Inserm U1052, University of Lyon, 69100 Lyon, France;
| | - Laure Smekens
- Oncology Department, Geneva University Hospital (HUG), 1205 Geneva, Switzerland; (J.G.); (L.S.)
| | | |
Collapse
|
19
|
Hernandez-Barry H, dela Cruz-Chuh J, Kajihara KK, Asundi J, Vandlen R, Zhang D, Hazenbos WL, Pillow T, Liu Y, Wu C, Kozak KR, Loyet KM. Mechanistic Characterization of the Potency of THIOMAB Antibody-Drug Conjugates Targeting Staphylococcus aureus and ETbR-Expressing Tumor Cells Using Quantitative LC-MS/MS Analysis of Intracellular Drug Accumulation. Bioconjug Chem 2025; 36:652-661. [PMID: 40179311 PMCID: PMC12007502 DOI: 10.1021/acs.bioconjchem.4c00533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
THIOMAB drug conjugate (TDC) technology provides site-specific conjugation of linker drugs to antibodies, allowing for targeted delivery of the payload. While a direct measurement of TDC cytotoxic potency allows efficient screening and confirmation that new drugs conjugated to antibodies result in proper processing in cells, additional mechanistic characterization is often needed to provide information-rich data to guide further optimization of TDC design. For example, a quantitative understanding of how TDCs are processed intracellularly can help determine which processing step is impacting payload delivery and thereby inform the basis of the TDC efficacy. Here, we measure the cellular accumulation of two different TDC drug payloads: MAPK (mitogen-activated protein kinase) pathway inhibitor targeting ETbR-expressing tumor cells and an antibiotic active against Staphylococcus aureus with an in vitro cell-based drug release LC-MS/MS assay in a 96-well format. This assay allowed us to correlate the cellular potency of each unconjugated molecule with the amount of payload that accumulated inside the cell. In the case of the pathway inhibitor drug, the biochemical characterization of TDC processing by cathepsin B and purified human liver enzyme extract demonstrated a correlation between the efficiency of the linker drug cleavage and intracellular payload accumulation. For the antibody-antibiotic conjugate, kinetic analysis of intracellular free drug retention provided valuable insight into the chemistry modifications needed for an efficient TDC. Taken together, we demonstrated the utility of quantitative LC-MS/MS assays as one tool in guiding the design of more effective TDCs via the mechanistic release characterization of two distinct payloads.
Collapse
Affiliation(s)
| | | | | | - Jyoti Asundi
- Genentech, Inc., South San Francisco, California 94080, United States
| | - Richard Vandlen
- Genentech, Inc., South San Francisco, California 94080, United States
| | - Donglu Zhang
- Genentech, Inc., South San Francisco, California 94080, United States
| | | | - Thomas Pillow
- Genentech, Inc., South San Francisco, California 94080, United States
| | - Yichin Liu
- Genentech, Inc., South San Francisco, California 94080, United States
| | - Cong Wu
- Genentech, Inc., South San Francisco, California 94080, United States
| | | | - Kelly M. Loyet
- Genentech, Inc., South San Francisco, California 94080, United States
| |
Collapse
|
20
|
Pélouard F, Chedru-Legros V, Nganoa C, Dompmartin A, L'Orphelin JM. Optimizing targeted therapy for metastatic melanoma: a combination of encorafenib and trametinib beyond standard protocols. Dermatol Reports 2025. [PMID: 40237146 DOI: 10.4081/dr.2025.10036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 02/04/2025] [Indexed: 04/18/2025] Open
Abstract
Only three combinations of BRAF inhibitor (BRAFi) and MEK inhibitor (MEKi) targeted therapies are marketed for the treatment of BRAF-mutated metastatic melanoma. The use of these combinations can be limited by the occurrence of severe adverse events (AEs) that may lead to discontinuation of treatment or contraindication. We present the case of a 45-year-old male diagnosed with stage III melanoma of the left thigh, as classified by the 8th edition of the American Joint Committee on Cancer (AJCC), exhibiting rapid recurrence of inguinal lymph node metastasis following complete surgical resection. Molecular biology revealed a mutated BRAFV600E status, indicating treatment associated with BRAFi/MEKi. First-line treatments were introduced successively with dabrafenib-trametinib and then encorafenib-binimetinib, both stopped for fever and severe digestive AEs. After the failure of a third line with an immune checkpoint inhibitor, a new rechallenge of targeted therapy (TT) was introduced with encorafenib-trametinib to increase tolerance. This unusual and innovative combination allowed a spectacular tolerance and complete oncological response for 39 months after the failure of the usual combinations. This is the first case in the literature to show the potential efficacy of a non-standard combination of encorafenib and trametinib, which are commercialized in two different market combinations. A pharmacological evidence-based analysis was performed to understand these good clinical results.
Collapse
Affiliation(s)
| | | | | | | | - Jean-Matthieu L'Orphelin
- Interdisciplinary Research Unit for the Prevention and Treatment of Cancer (ANTICIPE), Dermatology Department, Caen University Hospital.
| |
Collapse
|
21
|
Oddershede JK, Meklenborg IK, Bastholt L, Guldbrandt LM, Schmidt H, Friis RB. Cardiotoxicity in patients with metastatic melanoma treated with BRAF/MEK inhibitors: a real-world analysis of incidence, risk factors, and reversibility. Acta Oncol 2025; 64:507-515. [PMID: 40223207 PMCID: PMC12012651 DOI: 10.2340/1651-226x.2025.42567] [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: 11/24/2024] [Accepted: 03/01/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND BRAF/MEK inhibitors (BRAFi/MEKi) improve outcome in patients with BRAF-mutated metastatic melanoma but are associated with cardiotoxicity, leading to a decline in left ventricular ejection fraction (LVEF). This study aimed to evaluate the incidence, timeline, risk factors, and reversibility of BRAFi/MEKi-induced cardiotoxicity in a real-world setting. PATIENTS/MATERIALS AND METHODS Patients with metastatic melanoma (n = 170) treated with Encorafenib/Binimetinib, Vemurafenib/Cobimetinib, or Dabrafenib/Trametinib at Aarhus and Odense University Hospital, Denmark, from 2015 to 2023 were included. Cardiac function was assessed at baseline and every 3 months during treatment with either echocardiograms or multigated acquisition scans. Cardiotoxicity was defined as a reduction of LVEF by ≥10 percentage points (pp) to an LVEF < 50% (Major cardiotoxicity) or a reduction of LVEF by ≥15 pp but remaining > 50% (Minor cardiotoxicity). RESULTS Cardiotoxicity occurred in 21% of patients, with 14% experiencing major cardiotoxicity. The mean time to LVEF decline was 187 days, with 92% of major cardiotoxicity cases occurring within the first year. Cardiotoxicity was reversible in 79% of patients following dose reduction, treatment pauses, heart failure therapy, or continued treatment with monitoring. Baseline atrial fibrillation (odds ratio 13.67, p = 0.008) was identified as a risk factor for major cardiotoxicity. INTERPRETATION BRAFi/MEKi-induced cardiotoxicity is a significant but manageable complication, often reversible with timely interventions. Routine LVEF monitoring is recommended. The majority (92%) of major cardiac events were diagnosed within the first year of treatment, which might warrant a discontinuation of routine LVEF monitoring after 1 year of BRAFi/MEKi treatment.
Collapse
Affiliation(s)
| | - Ida K Meklenborg
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Lars Bastholt
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | | | - Henrik Schmidt
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Rasmus B Friis
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|
22
|
Tóth K, Gaál Z. Impact of Tyrosine Kinase Inhibitors on the Expression Pattern of Epigenetic Regulators. Cancers (Basel) 2025; 17:1282. [PMID: 40282457 PMCID: PMC12025482 DOI: 10.3390/cancers17081282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 04/03/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Advances in molecular genetic diagnostics and emerging opportunities for targeted treatment have opened new horizons in precision oncology. Tyrosine kinase inhibitors (TKI) are the subgroup of these agents with which the most clinical experience has been gathered so far. However, little data is available on the effect of TKI agents on the expression levels of molecules responsible for epigenetic regulation. Methods: In this study, we investigated the effect of in vitro and in vivo treatment with tyrosine kinase inhibitor agents on the expression of epigenetic regulators in hematological malignancies and solid tumors, based on data included in the functional genomics repository Gene Expression Omnibus. Results: Statistical analysis of datasets and series of gene expression patterns revealed numerous significant changes in the levels of epigenetic writers, erasers, microRNAs and members of chromatin-remodeling complexes following TKI treatment. Previously published data about the role of these epigenetic modifiers in malignant diseases has also been summarized. Conclusions: Our results may contribute to the establishment of novel treatment strategies aiming at the combinatorial administration of TKI and epidrugs in cancer, leading to less toxic therapy with further improved results.
Collapse
Affiliation(s)
| | - Zsuzsanna Gaál
- Institute of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
23
|
Hosoya K, Ozasa H, Yoshida H, Ajimizu H, Tsuji T, Yamazoe M, Ogimoto T, Hashimoto K, Funazo Yamamoto T, Suminaga K, Shima Y, Yoshida H, Nomizo T, Ito H, Terada K, Nishikawa S, Menju T, Yoshizawa A, Date H, Hirai T. Novel TEAD1 Inhibitor VT103 Enhances Dabrafenib Efficacy in BRAF V600E Mutated Lung Adenocarcinoma via Survivin Downregulation. Cancer Sci 2025. [PMID: 40202586 DOI: 10.1111/cas.70075] [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: 11/14/2024] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/10/2025] Open
Abstract
The BRAF V600E mutation is observed in 2% of the patients with lung adenocarcinoma (LUAD), and combination therapy targeting BRAF and mitogen-activated protein kinase (MEK) is the standard treatment for this population. However, acquired resistance inevitably develops, which highlights the need for novel therapeutic strategies. In this study, we established a patient-derived BRAF V600E-mutated LUAD cell line, KTOR81, and investigated the potential of targeting the Yes-associated protein 1 (YAP1)/transcriptional enhanced associate domain 1 (TEAD1) pathway in combination with BRAF inhibition. We observed that the novel TEAD1 inhibitor VT103 enhanced the efficacy of the BRAF inhibitor dabrafenib in KTOR81 cells and xenograft models. The combination of dabrafenib and VT103 downregulated the expression of the antiapoptotic protein survivin, which is transcriptionally regulated by the YAP1/TEAD1 complex, leading to increased apoptosis. Moreover, we used a LUAD tissue microarray to compare the staining patterns of YAP1, TEAD1, and survivin, and examined their association with prognosis. These analyses revealed a strong correlation between YAP1, TEAD1, and survivin expression in LUAD, suggesting the relevance of the YAP1/TEAD1-survivin axis beyond BRAF V600E-mutated cases. While no statistically significant association was observed between survivin expression and prognosis, when limited to driver oncogene-positive patients, high survivin expression was suggested to be associated with poor prognosis. These findings provide preclinical evidence for the efficacy of combining TEAD1 inhibition with BRAF-targeted therapy in BRAF V600E-mutated LUAD and highlight the YAP1/TEAD1-survivin axis as a potential therapeutic target especially in the driver oncogene-positive LUAD patients.
Collapse
Affiliation(s)
- Kazutaka Hosoya
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroaki Ozasa
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hironori Yoshida
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hitomi Ajimizu
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takahiro Tsuji
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Anatomy and Molecular Cell Biology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Masatoshi Yamazoe
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tatsuya Ogimoto
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kentaro Hashimoto
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomoko Funazo Yamamoto
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keiichiro Suminaga
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yusuke Shima
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Yoshida
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Nomizo
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroaki Ito
- Department of Diagnostic Pathology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuhiro Terada
- Department of Diagnostic Pathology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shigeto Nishikawa
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toshi Menju
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akihiko Yoshizawa
- Department of Diagnostic Pathology, Graduate School of Medicine, Nara Medical University, Nara, Japan
| | - Hiroshi Date
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
24
|
Al-Naesan I, Krepper D, Sparano F, Sztankay M, Efficace F, Giesinger JM. Patient-reported outcomes in randomized controlled trials evaluating BRAF inhibitors in patients with cutaneous melanoma: a systematic scoping review of quality of reporting and trial results. Melanoma Res 2025; 35:77-86. [PMID: 39668671 DOI: 10.1097/cmr.0000000000001014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
The objective of this study was to provide an overview of the current practice of patient-reported outcome (PRO) assessments in trials investigating treatment with BRAF inhibitors in patients with advanced melanomas. In addition, we extracted information on symptomatic adverse events (AEs) reported by clinicians to inform future PRO measurement strategies. For our systematic scoping review, we investigated randomized controlled trials (RCTs) evaluating treatment with BRAF inhibitors that had a primary, secondary or exploratory PRO endpoint and were indexed on PubMed. Two independent reviewers extracted information on general RCT characteristics, clinical results (e.g. survival, treatment response and symptomatic AEs) and the PRO measurement and results. Quality of PRO reporting using the CONSORT-PRO checklist was also assessed. We identified nine RCTs meeting the inclusion criteria, in which PROs were secondary or exploratory endpoints. In all trials but one, PROs were measured with the generic EORTC QLQ-C30 questionnaire. The quality of PRO reporting showed substantial variation across the different types of information, with information on handling of missing data and on PRO hypotheses lacking most frequently. Our analysis identified 29 relevant symptomatic AEs that could be reported directly by patients. Our findings may inform the planning of the PRO component of future RCTs, in particular regarding what symptoms and AEs should be covered by PRO measures to provide a comprehensive assessment of treatment tolerability. Our results also indicate a need for improving the quality of PRO reporting, to maximize the impact of PRO findings in real-word practice.
Collapse
Affiliation(s)
- Imad Al-Naesan
- Department of Psychiatry, Psychotherapy, Psychosomatics, and Medical Psychology, Innsbruck Medical University, University Hospital of Psychiatry II, Innsbruck, Austria
| | - Daniela Krepper
- Department of Psychiatry, Psychotherapy, Psychosomatics, and Medical Psychology, Innsbruck Medical University, University Hospital of Psychiatry II, Innsbruck, Austria
| | - Francesco Sparano
- Data Center and Health Outcomes Research Unit, Italian Group for Adult Haematologic Diseases (GIMEMA), Rome, Italy
| | - Monika Sztankay
- Department of Psychiatry, Psychotherapy, Psychosomatics, and Medical Psychology, Innsbruck Medical University, University Hospital of Psychiatry II, Innsbruck, Austria
| | - Fabio Efficace
- Data Center and Health Outcomes Research Unit, Italian Group for Adult Haematologic Diseases (GIMEMA), Rome, Italy
| | - Johannes M Giesinger
- Department of Psychiatry, Psychotherapy, Psychosomatics, and Medical Psychology, Innsbruck Medical University, University Hospital of Psychiatry II, Innsbruck, Austria
| |
Collapse
|
25
|
Yamazaki N, Sakata H, Iida O, Katayama T, Uhara H. Post-marketing surveillance of encorafenib in combination with binimetinib in Japanese patients with BRAF-mutant melanoma. Int J Clin Oncol 2025; 30:814-823. [PMID: 39918770 PMCID: PMC11946937 DOI: 10.1007/s10147-025-02693-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/04/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND A BRAF inhibitor, encorafenib, combined with a MEK inhibitor, binimetinib, was approved in Japan in early 2019 for the treatment of BRAF V600-mutant, unresectable malignant melanoma based on results of the global phase III trial, COLUMBUS, conducted in various countries including Japan. This post-marketing surveillance (PMS) assessed the combination in real-world clinical practice in Japan. METHODS We performed a prospective, multicentre, 12-month PMS of the safety and effectiveness of encorafenib plus binimetinib for radically unresectable, BRAF-mutant malignant melanoma in Japan. RESULTS Among 174 survey forms collected from 85 centres between February 2019 and August 2020, 172 were included for safety and effectiveness analysis. Patients (male [52.3%], median age 62.0 years) had Eastern Cooperative Oncology Group Performance Status 0 or 1 (91.8%) and comorbidities (55.2%). Respective encorafenib and binimetinib median dosages were 450 mg/day and 90 mg/day; median treatment duration, 24.1 and 24.2 weeks, and discontinuation, 71.5% for each, primarily for disease progression (56.9%) and adverse drug reactions (ADRs, 38.2%). Safety assessment ADRs occurred in 99 patients (57.6%), including eye disorders (40.7%), hepatic dysfunction (20.3%), rhabdomyolysis (4.7%), haemorrhage (2.3%), palmar-plantar erythrodysaesthesia syndrome (1.7%), and hypertension (1.7%); 19.8% were grade ≥ 3, none were grade 5, most resolved with/without treatment modification. At 12 months, the objective response rate was 48.8% (95% CI 41.2, 56.6; complete [19.2%], partial [29.7%]), overall survival was 40.1%. CONCLUSION The safety and effectiveness of encorafenib plus binimetinib in Japanese patients with BRAF-mutant malignant melanoma were similar to data reported in COLUMBUS; no new safety concerns were identified.
Collapse
Affiliation(s)
- Naoya Yamazaki
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hidenori Sakata
- Department of Pharmacovigilance, Ono Pharmaceutical Co., Ltd, Osaka, Japan
| | - Osamu Iida
- Department of Pharmacovigilance, Ono Pharmaceutical Co., Ltd, Osaka, Japan
| | - Teruaki Katayama
- Department of Oncology Medical Affairs, Ono Pharmaceutical Co., Ltd, Osaka, Japan
| | - Hisashi Uhara
- Department of Dermatology, Sapporo Medical University, South 1, West 16, Chuo-Ku, Sapporo, Hokkaido, 060-8556, Japan.
| |
Collapse
|
26
|
Ferrari M, Facchini BA, Ascierto PA, Sparano F. Melanoma neoadjuvant treatment: review and update of recent trials. Expert Rev Anticancer Ther 2025; 25:383-392. [PMID: 40043281 DOI: 10.1080/14737140.2025.2474182] [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: 10/16/2024] [Accepted: 02/26/2025] [Indexed: 03/16/2025]
Abstract
INTRODUCTION Neoadjuvant immunotherapy is emerging as an effective approach for resectable stage III/IV melanoma, showing improvements in disease response and survival outcomes. AREAS COVERED This review summarizes findings from neoadjuvant treatment trials in melanoma patients. Using the PubMed search engine and including the keywords 'neoadjuvant,' 'immunotherapy,' and 'melanoma,' we selected 18 trials that showed efficacy in patients with melanoma, mainly testing checkpoint inhibitors alone or in combination. Across all trials examined, treatments showed objective disease responses, which frequently translated into improved disease-free survival. EXPERT OPINION Additional phase III studies comparing neoadjuvant and adjuvant therapies are needed to establish the optimal standard of care. The variety of regimens and dosing schedules investigated highlights the need for further research to determine the most appropriate treatments in this clinical setting. Advances in the study of biomarkers that can identify specific subgroups of patients will guide future research in this field.
Collapse
Affiliation(s)
- Marco Ferrari
- Azienda USL Toscana centro, U.O. Oncologia Medica, Ponte a Niccheri, Bagno a Ripoli, FI, Italy
| | - Bianca Arianna Facchini
- Istituto Nazionale Tumori IRCCS Fondazione Giovanni Pascale - Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Napoli, Napoli, Italy
| | - Paolo Antonio Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione Giovanni Pascale - Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Napoli, Napoli, Italy
| | - Francesca Sparano
- Istituto Nazionale Tumori IRCCS Fondazione Giovanni Pascale - Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Napoli, Napoli, Italy
| |
Collapse
|
27
|
Huang K, Kim MO. Therapeutic Strategies for Drug-resistant Melanoma and Their Clinical Implications. J Cancer Prev 2025; 30:7-11. [PMID: 40201029 PMCID: PMC11973462 DOI: 10.15430/jcp.24.028] [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: 12/02/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 04/10/2025] Open
Abstract
Melanoma is a malignant tumor originating from melanocytes, characterized by its high invasiveness and metastasis, leading to poor prognosis and high mortality. Early-stage melanoma is primarily treated with surgery; however, due to its metastatic nature, surgery becomes challenging in advanced stages. Treatment strategies for advanced or metastatic melanoma include chemotherapy, radiation therapy, and targeted therapy. However, melanoma's propensity for rapid drug resistance remains a significant clinical challenge. This review summarizes the developments in the treatment of drug-resistant melanoma over the past decade and discusses the advantages and disadvantages of various therapeutic approaches and their clinical significance implications.
Collapse
Affiliation(s)
- Ke Huang
- Department of Animal Science and Biotechnology, Research Institute for Innovative Animal Sciences, Kyungpook National University, Sangju, Korea
| | - Myoung Ok Kim
- Department of Animal Science and Biotechnology, Research Institute for Innovative Animal Sciences, Kyungpook National University, Sangju, Korea
| |
Collapse
|
28
|
Chang MC, Malut VR, Mahar R, Rushin A, McLeod MA, Pierre GL, Malut IR, Staklinski SJ, Glanz ME, Ragavan M, Sharma G, Madheswaran M, Badar A, Rao AD, Law BK, Kilberg MS, Collins JHP, Kodibagkar VD, Bankson JA, DeBerardinis RJ, Merritt ME. Assessing cancer therapeutic efficacy in vivo using [ 2H 7]glucose deuterium metabolic imaging. SCIENCE ADVANCES 2025; 11:eadr0568. [PMID: 40138413 PMCID: PMC11939044 DOI: 10.1126/sciadv.adr0568] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 02/24/2025] [Indexed: 03/29/2025]
Abstract
Metabolic imaging produces powerful visual assessments of organ function in vivo. Current techniques can be improved by safely increasing metabolic contrast. The gold standard, 2-[18F]fluorodeoxyglucose-positron emission tomography (FDG-PET) imaging, is limited by radioactive exposure and sparse assessment of metabolism beyond glucose uptake and retention. Deuterium magnetic resonance imaging (DMRI) with [6,6-2H2]glucose is nonradioactive, achieves tumor metabolic contrast, but can be improved by enriched contrast from deuterated water (HDO) based imaging. Here, we developed a DMRI protocol employing [2H7]glucose. Imaging 2H-signal and measuring HDO production in tumor-bearing mice detected differential glucose utilization across baseline tumors, tumors treated with vehicle control or anti-glycolytic BRAFi and MEKi therapy, and contralateral healthy tissue. Control tumors generated the most 2H-signal and HDO. To our knowledge this is the first application of DMRI with [2H7]glucose for tumoral treatment monitoring. This approach demonstrates HDO as a marker of tumor glucose utilization and suggests translational capability in humans due to its safety, noninvasiveness, and suitability for serial monitoring.
Collapse
Affiliation(s)
- Mario C. Chang
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Vinay R. Malut
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Rohit Mahar
- Department of Chemistry, Hemvati Nandan Bahuguna Garhwal University (A Central University), Srinagar Garhwal, Uttarakhand 246174, India
| | - Anna Rushin
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Marc A. McLeod
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Geraldine L. Pierre
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Indu R. Malut
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Stephen J. Staklinski
- School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Max E. Glanz
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Mukundan Ragavan
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Gaurav Sharma
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Manoj Madheswaran
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Arshee Badar
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Aparna D. Rao
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Brian K. Law
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Michael S. Kilberg
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - James H. P. Collins
- National High Magnetic Field Laboratory, University of Florida, Gainesville, FL 32610, USA
| | - Vikram D. Kodibagkar
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85281, USA
| | - James A. Bankson
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ralph J. DeBerardinis
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Matthew E. Merritt
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| |
Collapse
|
29
|
Gao T, He X, Wang J, Liu J, Hu X, Bai C, Yin S, Shi Y, Wang Y, Tan Z, Cao F, Li S, Shi YJ, Xue R, Li J, He Y, Li J, Lu H, Zhang H, Zhang L, Fang Z, Wang X, Liu M, Fu W, Tang L, Ye B, Fan Z, Xi JJ. Self-assembled patient-derived tumor-like cell clusters for personalized drug testing in diverse sarcomas. Cell Rep Med 2025; 6:101990. [PMID: 40054460 PMCID: PMC11970405 DOI: 10.1016/j.xcrm.2025.101990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 10/30/2024] [Accepted: 02/04/2025] [Indexed: 03/21/2025]
Abstract
Several patient-derived tumor models have emerged recently. However, soft tissue sarcomas (STSs) present a challenge in developing preclinical drug-testing models due to their non-epithelial and complex nature. Here, we report a model termed patient-derived tumor-like cell clusters (PTCs) derived from STS patients. PTCs result from the self-assembly and proliferation of mesenchymal stem cells (MSCs), epithelial cells, and immune cells, faithfully recapitulating the morphology and function of the original tumors. Through standardized culture and drug-response assessment protocols, PTCs facilitate personalized drug testing, evaluating hundreds of therapies within two weeks. Notably, PTCs exhibit 100% accuracy in distinguishing between complete or partial response and disease progression. We demonstrate the utility of PTCs in guiding chemotherapy selection for a patient with relapse and metastases following conventional therapy, who exhibited a positive response after non-conventional therapy identified through PTC. These findings underscore the potential of PTCs for prospective use in clinical decision-making regarding therapy selection.
Collapse
Affiliation(s)
- Tian Gao
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xinyu He
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Junyi Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Jiayong Liu
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xiongbing Hu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Chujie Bai
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Shenyi Yin
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China; GeneX Health Co., Ltd., Beijing 100195, China
| | - Yunfei Shi
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yanmin Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Zhichao Tan
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Fang Cao
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Shu Li
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yan-Jie Shi
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ruifeng Xue
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Juan Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Yang He
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Jiaxin Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China; Peking University Yangtze Center of Future Health Technology, Wuxi 214111, China
| | - Huinan Lu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China; Peking University Yangtze Center of Future Health Technology, Wuxi 214111, China
| | - Hanshuo Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China; GeneX Health Co., Ltd., Beijing 100195, China
| | - Lu Zhang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhiwei Fang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xinyu Wang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Mengmeng Liu
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Wenjun Fu
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Lei Tang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Buqing Ye
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Zhengfu Fan
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China.
| | - Jianzhong Jeff Xi
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China; Peking University Yangtze Center of Future Health Technology, Wuxi 214111, China.
| |
Collapse
|
30
|
Mahipal A, Bucheit L, Zhang N, Barnett RM, Storandt MH, Chakrabarti S. Frequency and outcomes of BRAF alterations identified by liquid biopsy in metastatic, non-colorectal gastrointestinal cancers. Oncologist 2025; 30:oyaf044. [PMID: 40163685 PMCID: PMC11957259 DOI: 10.1093/oncolo/oyaf044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 02/04/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Impact of BRAF V600E mutations (BRAFV600E), a poor prognostic factor in metastatic colorectal cancer, is lacking in non-CRC gastrointestinal (GI) cancers including pancreatic (PDAC), gastric/gastroesophageal (GEA), hepatocellular carcinoma (HCC), and cholangiocarcinoma (CCA). Due to tumor-agnostic approvals for patients with BRAFV600E, understanding the frequency and impact of BRAF alterations across non-CRC GI cancers is essential for clinical decision-making. METHODS Patients with PDAC, GEA, HCC, or CCA who had cell-free DNA detected on Guardant360 (Guardant Health) from 2020 to 2023 were queried. Prevalence of characterized BRAF genomic alterations (GA) was calculated; GAs were grouped by class (Class I/II/III). The Chi-squared test assessed differences between cancer types. A subset of patients had outcomes analysis using GuardantINFORM, a real-world clinicogenomic database, to derive real-world overall survival (rwOS). RESULTS Of 32 480 included patients, BRAF GAs were identified in 4.4%; 19% were BRAFV600E (0.81% prevalence overall). CCA had the highest rate of BRAF GAs and BRAFV600E (P < .01); HCC and GEA had the highest rates of BRAF class II/III alterations. There were no significant differences in rwOS by alteration class or cancer type; numeric differences were observed by alteration class. Few patients were treated with BRAF inhibitors (2.2%). Prevalence of co-occurring alterations was unique by cancer type. CONCLUSIONS Frequency of BRAF GAs, including BRAFV600E, in non-CRC GI cancers detected by liquid biopsy is similar to tissue-based rates and can be reliably used to assess BRAF status. BRAF GAs have mixed prognostic implications on survival for patients with non-CRC GI malignancies that warrant further exploration.
Collapse
Affiliation(s)
- Amit Mahipal
- University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, United States
- Mayo Clinic, Rochester, MN, United States
| | | | - Nicole Zhang
- Guardant Health Inc, Palo Alto, CA, United States
| | | | | | - Sakti Chakrabarti
- University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
31
|
Lubrano S, Cervantes-Villagrana RD, Faraji F, Ramirez S, Sato K, Adame-Garcia SR, Officer A, Arang N, Rigiracciolo DC, Anguiano Quiroz PY, Martini C, Wang Y, Ferguson FM, Bacchiocchi A, Halaban R, Coma S, Holmen SL, Pachter JA, Aplin AE, Gutkind JS. FAK inhibition combined with the RAF-MEK clamp avutometinib overcomes resistance to targeted and immune therapies in BRAF V600E melanoma. Cancer Cell 2025; 43:428-445.e6. [PMID: 40020669 PMCID: PMC11903146 DOI: 10.1016/j.ccell.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 10/17/2024] [Accepted: 02/04/2025] [Indexed: 03/03/2025]
Abstract
Widespread BRAF mutations result in persistent RAS-RAF-MEK-ERK (MAPK) signaling in melanoma. BRAF (BRAFi) and MEK (MEKi) inhibitors are approved for BRAF V600E melanomas, including those progressing on immunotherapy; however, rapid resistance to these agents highlights the need for novel strategies. Here, transcriptome analysis of BRAF V600E melanomas from patients resistant to BRAFi and MEKi shows activation of focal adhesion signaling. Consistently, BRAFi, MEKi, and the RAF-MEK clamp avutometinib activate focal adhesion kinase (FAK) in melanoma cells. Mechanistically, inhibition of an MAPK-RhoE (RND3) feedback loop results in the adaptive activation of RhoA-FAK-AKT. In turn, FAK inhibitors (FAKi) exert potent pro-apoptotic activity when combined with MAPK pathway inhibition. FAKi plus avutometinib overcomes resistance in multiple models derived from BRAFi plus MEKi-resistant melanoma patients and immunotherapy-resistant syngeneic mouse models. These findings provide a rationale for the development of avutometinib in combination with FAKi for patients with BRAF V600E melanoma progressing on BRAFi plus MEKi or immunotherapy.
Collapse
Affiliation(s)
- Simone Lubrano
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Rodolfo Daniel Cervantes-Villagrana
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Farhoud Faraji
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA; Department of Otolaryngology - Head and Neck Surgery, UC San Diego Health, La Jolla, CA 92037, USA
| | - Sydney Ramirez
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kuniaki Sato
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sendi R Adame-Garcia
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Adam Officer
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nadia Arang
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA 94158, USA
| | | | - Paola Y Anguiano Quiroz
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA; John Muir College, University of California, San Diego, La Jolla, CA 92093, USA
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | - YiYu Wang
- Department of Chemistry and Biochemistry and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Fleur M Ferguson
- Department of Chemistry and Biochemistry and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | | | - Ruth Halaban
- Department of Dermatology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | - Sheri L Holmen
- Department of Surgery, University of Utah Health Sciences Center, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | | | - Andrew E Aplin
- Department of Pharmacology, Physiology and Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, US
| | - J Silvio Gutkind
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
32
|
Tan SL, Ho BLS, Yew TT, Yunus D. An 8-year-old girl with secondary histiocytic sarcoma with BRAF V600 mutation following T-cell acute lymphoblastic leukemia demonstrating stable disease for 3 years on dabrafenib and trametinib - a case report and literature review. BMC Pediatr 2025; 25:178. [PMID: 40057687 PMCID: PMC11889787 DOI: 10.1186/s12887-025-05539-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 02/25/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Histiocytic sarcoma as a secondary malignancy following childhood leukemia is extremely uncommon with fewer than 20 cases reported worldwide. They often pose a diagnostic challenge and prognosis is dismal. There is a lack of well-established clinical treatment protocols owing to rarity of disease. Majority were managed with chemotherapy with variable outcomes. CASE PRESENTATION Herein we report a rare case of an 8-year-old girl with secondary BRAFV600-mutant histiocytic sarcoma following T-cell acute lymphoblastic leukemia. After poor disease control with salvage chemotherapy, she was treated with MAPK-targeted therapy with dabrafenib and trametinib. She demonstrated excellent response and remained in partial remission with no signs of disease progression 3 years later. CONCLUSIONS There is yet to be consensus on the optimal management for this neoplasm. Description of our successful clinical experience highlights that investigation for BRAF mutations in histiocytic sarcoma is potentially advantageous. It also adds to the growing evidence that precision medicine may be a promising avenue to target this aggressive tumor and lays the foundation for future research.
Collapse
Affiliation(s)
- Sue Lyn Tan
- Department of Paediatrics, Faculty of Medicine and Health Sciences, Universiti Malaysia Sarawak, Jalan Datuk Muhammad Musa, Kota Samarahan, Sarawak, 94300, Malaysia.
| | - Betty Lee Sue Ho
- Department of Paediatrics, Sarawak General Hospital, Jalan Hospital, Kuching, Sarawak, 93586, Malaysia
| | - Ting Ting Yew
- Department of Radiology, Faculty of Medicine and Health Sciences, Universiti Malaysia Sarawak, Jalan Datuk Muhammad Musa, Kota Samarahan, Sarawak, 94300, Malaysia
| | - Dahziela Yunus
- Department of Pathology, Queen Elizabeth Hospital, Kota Kinabalu Sabah, 88586, Malaysia
| |
Collapse
|
33
|
Rubinstein JC, Domanskyi S, Sheridan TB, Sanderson B, Park S, Kaster J, Li H, Anczukow O, Herlyn M, Chuang JH. Spatiotemporal Profiling Defines Persistence and Resistance Dynamics during Targeted Treatment of Melanoma. Cancer Res 2025; 85:987-1002. [PMID: 39700408 PMCID: PMC11875961 DOI: 10.1158/0008-5472.can-24-0690] [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: 07/16/2024] [Revised: 10/03/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024]
Abstract
Resistance of BRAF-mutant melanomas to targeted therapy arises from the ability of cells to enter a persister state, evade treatment with relative dormancy, and repopulate the tumor when reactivated. A better understanding of the temporal dynamics and specific pathways leading into and out of the persister state is needed to identify strategies to prevent treatment failure. Using spatial transcriptomics in patient-derived xenograft models, we captured clonal lineage evolution during treatment. The persister state showed increased oxidative phosphorylation, decreased proliferation, and increased invasive capacity, with central-to-peripheral gradients. Phylogenetic tracing identified intrinsic and acquired resistance mechanisms (e.g., dual-specific phosphatases, reticulon-4, and cyclin-dependent kinase 2) and suggested specific temporal windows of potential therapeutic susceptibility. Deep learning-enabled analysis of histopathologic slides revealed morphologic features correlating with specific cell states, demonstrating that juxtaposition of transcriptomics and histologic data enabled identification of phenotypically distinct populations from using imaging data alone. In summary, this study defined state change and lineage selection during melanoma treatment with spatiotemporal resolution, elucidating how choice and timing of therapeutic agents will impact the ability to eradicate resistant clones. Significance: Tracking clonal progression during treatment uncovers conserved, global transcriptional changes and local clone-clone and spatial patterns underlying the emergence of resistance, providing insights into therapy-induced tumor evolution.
Collapse
Affiliation(s)
- Jill C. Rubinstein
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Department of Surgery, Hartford Healthcare, Hartford, CT, USA
- Department of Surgery, UCONN School of Medicine, Farmington, CT, USA
- These authors provided equal contribution to this work
- Further information and requests for resources should be directed to and will be fulfilled by the lead contact, Jill Rubinstein
| | - Sergii Domanskyi
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- These authors provided equal contribution to this work
| | - Todd B. Sheridan
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Department of Surgery, Hartford Healthcare, Hartford, CT, USA
| | - Brian Sanderson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - SungHee Park
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Jessica Kaster
- The Wistar Institute, Philadelphia, PA, USA
- Saint Joseph’s University, Philadelphia, PA, USA
| | - Haiyin Li
- The Wistar Institute, Philadelphia, PA, USA
| | - Olga Anczukow
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | | |
Collapse
|
34
|
Sad K, Fawwal D, Jones C, Hill E, Skinner K, Adams M, Lustenberger S, Lee R, Lohano S, Elayavalli S, Farhi J, Mehta C, Lemon L, Fasken MB, Hong AL, Sloan SA, Corbett A, Spangle JM. Histone H3E50K remodels chromatin to confer oncogenic activity and support an EMT phenotype. NAR Cancer 2025; 7:zcaf002. [PMID: 39901931 PMCID: PMC11788928 DOI: 10.1093/narcan/zcaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/08/2025] [Accepted: 01/17/2025] [Indexed: 02/05/2025] Open
Abstract
Sequencing of human patient tumors has identified recurrent missense mutations in genes encoding core histones. We report that mutations that convert histone H3 amino acid 50 from a glutamate to a lysine (H3E50K) support an oncogenic phenotype. Expression of H3E50K is sufficient to transform human cells as evidenced by an increase in cell migration and invasion, and an increase in proliferation and clonogenicity. H3E50K also increases the invasive phenotype in the context of co-occurring BRAF mutations, which are present in patient tumors characterized by H3E50K. H3E50 lies on the globular domain surface in a region that contacts H4 within the nucleosome. We find that H3E50K selectively increases chromatin accessibility and perturbs proximal H3 post-translational modifications including H3K27me3; together these changes to chromatin dynamics dysregulate gene expression to support the epithelial-to-mesenchymal transition. Functional studies using Saccharomyces cerevisiae reveal that, while yeast cells that express H3E50K as the sole copy of histone H3 show sensitivity to cellular stressors, including caffeine, H3E50K cells display some genetic interactions that are distinct from the characterized H3K36M oncohistone yeast model. Taken together, these data suggest that additional H3 mutations have the potential to support oncogenic activity and function through distinct mechanisms that dysregulate gene expression.
Collapse
Affiliation(s)
- Kirti Sad
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, United States
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, United States
| | - Dorelle V Fawwal
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, United States
- Graduate Program in Biochemistry, Cell, and Developmental Biology, Emory University, Atlanta, GA 30322, United States
| | - Celina Y Jones
- Department of Biology, Emory College of Arts and Sciences, Atlanta, GA 30322, United States
| | - Emily J Hill
- Graduate Program in Genetics and Molecular Biology, Emory University, Atlanta, GA 30322, United States
| | - Katie T Skinner
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, United States
- Graduate Program in Cancer Biology, Emory University, Atlanta, GA 30322, United States
| | - Miranda L Adams
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, United States
- Graduate Program in Cancer Biology, Emory University, Atlanta, GA 30322, United States
| | - Severin Lustenberger
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02115, United States
| | - Richard S Lee
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, United States
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, United States
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, United States
| | - Sandhya V Lohano
- Department of Biology, Emory College of Arts and Sciences, Atlanta, GA 30322, United States
| | - Satvik R Elayavalli
- Department of Biology, Emory College of Arts and Sciences, Atlanta, GA 30322, United States
| | - Jonathan Farhi
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, United States
- Graduate Program in Cancer Biology, Emory University, Atlanta, GA 30322, United States
| | - Christina C Mehta
- Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, United States
| | - Laramie D Lemon
- Department of Biology, Emory College of Arts and Sciences, Atlanta, GA 30322, United States
| | - Milo B Fasken
- Department of Biology, Emory College of Arts and Sciences, Atlanta, GA 30322, United States
| | - Andrew L Hong
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, United States
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, United States
| | - Steven A Sloan
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, United States
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, United States
| | - Anita H Corbett
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, United States
- Department of Biology, Emory College of Arts and Sciences, Atlanta, GA 30322, United States
| | - Jennifer M Spangle
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, United States
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, United States
| |
Collapse
|
35
|
Wang H, Qiao S, Huang L, Zhang Z, Wang J, Tian W. PTPN9 promotes melanoma progression by regulating the ferroptosis pathway. FASEB J 2025; 39:e70394. [PMID: 39937573 DOI: 10.1096/fj.202402285r] [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: 09/24/2024] [Revised: 01/21/2025] [Accepted: 02/05/2025] [Indexed: 02/13/2025]
Abstract
In recent years, there has been a gradual increase in the incidence and mortality rates of melanoma, posing a significant threat to human health and life. Protein tyrosine phosphatases (PTPNs) have been implicated in the progression of various human cancers, including breast, lung, and cervical cancer. To investigate PTPN9 expression in melanoma, impacting the disease's survival and prognosis. Our study, which involved an analysis of The Cancer Genome Atlas database and immunohistochemical staining of pathological sections, identified an upregulation of PTPN9 expression in melanoma, impacting the disease's survival and prognosis. At the cellular level, we investigated the effects of PTPN9 on the proliferation, invasion, and metastasis of A375 and SK-MEL-28 cells. Through various experimental techniques such as Western blot protein detection, electron microscopy, and oil red O staining, we observed that PTPN9 potentially contributes to the development of skin cutaneous melanoma (SKCM) by regulating ferroptosis-related proteins ACSL4, FTH1, and P53, thereby influencing lipid metabolism. The results of this study highlight the unique role of PTPN9 in SKCM and suggest its potential as a biomarker for the disease.
Collapse
Affiliation(s)
- Hongmei Wang
- Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
- Center of Translational Medicine, Zibo Central Hospital, Zibo, China
| | - Sen Qiao
- Assisted Reproduction Center, Northwest Women's and Children's Hospital, Xi'an, China
| | - Lingyan Huang
- Pathological Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Zhengping Zhang
- Department of Radiology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Jiao Wang
- Clinical Laboratory, Zibo Central Hospital, Zibo, China
| | - Wenxiu Tian
- Center of Translational Medicine, Zibo Central Hospital, Zibo, China
| |
Collapse
|
36
|
Soria A, Sanchez Mauriño P, Serrano Domingo JJ, García Galindo R, Sequero S, Gutiérrez Sanz L, Crespo G, Díaz-Beveridge R, Puértolas T, López P, Fra Rodríguez J, López Castro R, Aguayo C, Valdivia J, Cunquero-Tomás AJ, Benítez G, Ayala de Miguel P, Espinosa E, Muñoz-Couselo E, Campos B, García Sánchez L, Cerezuela-Fuentes P. Real-life evidence of encorafenib plus binimetinib in patients with unresectable advanced or metastatic BRAF V600-mutant melanoma in Spain: the BECARE (GEM-2002) trial. Front Oncol 2025; 15:1466185. [PMID: 40078188 PMCID: PMC11897558 DOI: 10.3389/fonc.2025.1466185] [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: 07/17/2024] [Accepted: 01/17/2025] [Indexed: 03/14/2025] Open
Abstract
Purpose Combined BRAF/MEK inhibition with encorafenib (E) plus binimetinib (B) has demonstrated efficacy and tolerability in phase III clinical trials, and is the standard of care for advanced/metastatic BRAFV600-mutant melanoma. However, real-life evidence is limited, particularly in patients pre-treated with immune checkpoint inhibitors (ICI). Patients and methods BECARE GEM 2002 was a retrospective, non-interventional study aimed at investigating the real-world effectiveness and tolerability of EB in patients with unresectable or metastatic BRAFV600-mutant melanoma conducted at 21 sites in Spain. The primary objective of this study was to characterise the population of patients receiving EB and assess the efficacy and tolerability of EB in real life. The study included patients treated according to standard clinical practice with EB as the 1st line or 2nd line after progression to ICI for an unresectable or metastatic stage. Patients who previously received treatment with BRAF and/or MEK inhibitor, other than as adjuvants, that ended ≥ 6 m before EB were not eligible. Results From September 2021 to March 2023, 117 patients were included; 89 (76.1%) and 28 (23.9%) patients received EB as 1st line and 2nd line, respectively. The median follow-up was 13.8 months (95% CI: 12.0-17.4). In patients with EB as 1st line treatment, ORR and median PFS were 75% and 12 months (95% CI: 9.4-18.6), respectively. In patients with EB as 2nd line treatment after ICI, ORR and median PFS were 77.8% and 12.5 months (95% CI: 6.6-NA), respectively. In patients with brain metastasis ORR and median PFS were 70.8% and 6.3 months (95% CI: 6.1-10.3). Treatment-related adverse events of grade ≥3 were reported in 17 (14.5%) patients; transaminitis (9.4%) and diarrhoea (2.6%) were the most frequent adverse events. Conclusion In this real-world study, EB treatment demonstrated effectiveness and a consistent safety profile in patients with BRAFV600-mutant melanoma treated according to standard clinical practice, including in those with prior ICI treatment and of brain metastasis; therefore, EB is a feasible treatment option for unresectable and metastatic melanoma.Clinical trial identification: REec: 0004-2021-OBS. Clinical trial identification REec: 0004-2021-OBS.
Collapse
Affiliation(s)
- Ainara Soria
- Medical Oncology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | | | | | - Regina García Galindo
- Medical Oncology, Hospital Universitario de Jerez de la Frontera, Jerez de la Frontera, Spain
| | - Silvia Sequero
- Medical Oncology, Hospital Universitario San Cecilio, Granada, Spain
| | | | - Guillermo Crespo
- Medical Oncology, Hospital Universitario de Burgos, Burgos, Spain
| | - Roberto Díaz-Beveridge
- Medical Oncology, Hospital Universitario y Politécnico la Fe de Valencia, Valencia, Spain
| | - Teresa Puértolas
- Medical Oncology, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Pedro López
- Medical Oncology, Complejo Hospitalario de Jaén, Jaén, Spain
| | | | - Rafael López Castro
- Medical Oncology, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Cristina Aguayo
- Medical Oncology, Hospital Universitario Infanta Sofía, San Sebastián de los Reyes, Spain
| | - Javier Valdivia
- Medical Oncology, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | | | - Gretel Benítez
- Medical Oncology, Complejo Hospitalario Universitario Insular-Materno infantil de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Pablo Ayala de Miguel
- Medical Oncology, Hospital Universitario San Pedro de Alcántara, Cáceres, Cáceres, Spain
| | - Enrique Espinosa
- Universidad Autónoma de Madrid, School of Medicine - Hospital Universitario La Paz - CIBERONC - Madrid, Madrid, Spain
| | - Eva Muñoz-Couselo
- Medical Oncology, Hospital Universitario Vall d’Hebron & Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Begoña Campos
- Medical Oncology, Hospital Universitario Lucus Augusti de Lugo, Lugo, Spain
| | | | - Pablo Cerezuela-Fuentes
- Medical Oncology, Hospital Clínico Universitario Virgen de la Arrixaca, Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Ciudad de Murcia, Spain
| |
Collapse
|
37
|
Nakajima I, Yoshino K, Tsuji H. Incidence and Management of Retinopathy and Uveitis in Patients Receiving BRAF/MEK Inhibitor Therapy. Semin Ophthalmol 2025:1-7. [PMID: 39991926 DOI: 10.1080/08820538.2025.2468381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/05/2025] [Accepted: 02/12/2025] [Indexed: 02/25/2025]
Abstract
PURPOSE To assess the clinical features of ocular adverse events in patients receiving BRAF/MEK inhibitor therapy. METHODS In this retrospective study, 65 patients were treated with BRAF/MEK inhibitors (dabrafenib/trametinib or encorafenib/binimetinib). RESULTS Of the 65 patients, 28 had malignant melanoma and 37 had non-melanoma malignancies. Bilateral MEK-associated retinopathy was observed in nine cases; none experienced vision loss due to MEK-associated retinopathy. Uveitis was diagnosed in four patients (6.1%), three of whom presented with Vogt-Koyanagi-Harada (VKH)-like uveitis. All VKH-like uveitis cases were observed in patients with melanoma and their incidences were significantly higher in these patients than in those without melanoma (p = .04). Treatment with corticosteroids resulted in either resolution or control of symptoms in all cases of VKH-like uveitis, enabling continuation of BRAF/MEK inhibitor therapy. CONCLUSION VKH-like uveitis was found to be significantly more frequent in patients with melanoma than in those with other malignancies.
Collapse
Affiliation(s)
- Isana Nakajima
- Department of Ophthalmology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Koji Yoshino
- Department of Dermatologic Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hideki Tsuji
- Department of Ophthalmology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
38
|
Connor C, Carr QL, Sweazy A, McMasters K, Hao H. Clinical Approaches for the Management of Skin Cancer: A Review of Current Progress in Diagnosis, Treatment, and Prognosis for Patients with Melanoma. Cancers (Basel) 2025; 17:707. [PMID: 40002300 PMCID: PMC11853469 DOI: 10.3390/cancers17040707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Melanoma represents a significant public health challenge due to its increasing incidence and potential for metastasis. This review will explore the current clinical approaches to the management of melanoma, focusing on advancements in diagnosis, treatment, and prognosis. Methods for early detection and accurate staging have been enhanced by new diagnostic strategies. Treatment modalities have expanded beyond traditional surgical excision to include targeted therapy and immunotherapy. Prognostic assessment has benefited from the development of novel biomarkers and genetic profiling. This review will highlight the progress made in the multidisciplinary management of melanoma, underscoring the importance of continuous research to improve patient outcomes.
Collapse
Affiliation(s)
- Colton Connor
- School of Medicine, University of Louisville, Louisville, KY 40202, USA; (C.C.); (Q.L.C.)
| | - Quinton L. Carr
- School of Medicine, University of Louisville, Louisville, KY 40202, USA; (C.C.); (Q.L.C.)
| | - Alisa Sweazy
- The Hiram C. Polk, Jr., MD Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (A.S.); (K.M.)
| | - Kelly McMasters
- The Hiram C. Polk, Jr., MD Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (A.S.); (K.M.)
| | - Hongying Hao
- The Hiram C. Polk, Jr., MD Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (A.S.); (K.M.)
| |
Collapse
|
39
|
Placke JM, Rajcsanyi LS, Herbst R, Terheyden P, Utikal J, Pföhler C, Kreuter A, Mohr P, Gutzmer R, Weichenthal M, Meier F, Berking C, Leiter U, Seier J, Krefting F, Tasdogan A, Lodde GC, Livingstone E, Zimmer L, Roesch A, Griewank K, Schadendorf D, Ugurel S. Presence of brain metastasis differentially impacts long-term survival after first-line therapy in melanoma depending on BRAF mutation status. Front Immunol 2025; 16:1536642. [PMID: 40028330 PMCID: PMC11868123 DOI: 10.3389/fimmu.2025.1536642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 01/29/2025] [Indexed: 03/05/2025] Open
Abstract
Background Modern therapeutic strategies have significantly improved the prognosis of advanced melanoma patients. Predictive factors of therapy response include serum LDH; however, predictive markers for long-term survival are currently largely lacking. Patients and methods Patients diagnosed with stage IV melanoma (AJCCv8) of cutaneous origin or unknown primary were identified from the prospective multicenter German Dermatologic Cooperative Oncology Group (DeCOG) skin cancer registry ADOREG. Baseline characteristics were compared between patient groups with short-term versus long-term survival. Statistical analysis included ROC analysis and multinomial regression analysis. Results Of 3066 stage IV melanoma patients entered into the ADOREG between 05/2014 and 06/2021, 395 were identified for this study, of whom 301 (76.2%) survived ≤1 year, and 94 (23.8%) survived ≥5 years after stage IV diagnosis. The median follow-up time was 6 months (range 0-129 months). Regarding the baseline characteristics, only elevated serum LDH (P <0.001) was found to be independently predicting survival ≤1 year. Type of first-line therapy, immune checkpoint inhibition (ICI) versus BRAF/MEK targeted therapy (TT), was not predictive of long-term survival ≥5 years. For survival ≤1 year, the presence of brain metastases at treatment start was an independent predictor in BRAF-mutated patients regardless if they received TT (N=113; P=0<0.001) or ICI (N=69; P=0.015), but not in BRAF-wildtype patients who received ICI (N=161; P=0.47). Conclusions Low serum LDH independently predicts long-term survival of stage IV melanoma patients in every subgroup of treatment type and BRAF status. Brain metastasis has a negative impact on long-term survival in BRAF-mutated, but not in BRAF-wildtype patients. Investigation of molecular features of brain metastases in BRAF-mutated vs. BRAF-wildtype melanomas may lead to new insights in tumor biology and may yield new therapeutic approaches.
Collapse
Affiliation(s)
- Jan-Malte Placke
- Department of Dermatology, University Hospital Essen, Essen, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Essen, Germany
| | - Luisa Sophie Rajcsanyi
- Section of Molecular Genetics in Mental Disorders, University Hospital Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, Germany
- Institute for Sex- and Gender-Sensitive Medicine, University Hospital Essen, Essen, Germany
| | - Rudolf Herbst
- Department of Dermatology, Helios Klinikum Erfurt, Erfurt, Germany
| | | | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Claudia Pföhler
- Department of Dermatology, Saarland University Medical School, Homburg/Saar, Germany
| | - Alexander Kreuter
- Department of Dermatology, Venereology and Allergology, HELIOS St. Elisabeth Klinik Oberhausen, University Witten-Herdecke, Oberhausen, Germany
| | - Peter Mohr
- Department of Dermatology, Elbe Kliniken Buxtehude, Buxtehude, Germany
| | - Ralf Gutzmer
- Department of Dermatology, Skin Cancer Center Minden, Minden, Germany
| | - Michael Weichenthal
- Department of Dermatology, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Friedegund Meier
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Carola Berking
- Department of Dermatology, Uniklinikum Erlangen, CCC Erlangen – EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ulrike Leiter
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Johanna Seier
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Frederik Krefting
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Alpaslan Tasdogan
- Department of Dermatology, University Hospital Essen, Essen, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Essen, Germany
| | - Georg C. Lodde
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | | | - Lisa Zimmer
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Alexander Roesch
- Department of Dermatology, University Hospital Essen, Essen, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Essen, Germany
| | - Klaus Griewank
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen, Essen, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Essen, Germany
| |
Collapse
|
40
|
Höppener DJ, Grünhagen DJ, Eggermont AMM, van der Veldt AAM, Verhoef C. An Overview of Liver Directed Locoregional Therapies. Hematol Oncol Clin North Am 2025; 39:103-123. [PMID: 39510668 DOI: 10.1016/j.hoc.2024.08.010] [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: 11/15/2024]
Abstract
An overview of all liver-directed locoregional therapies, including surgical resection for melanoma liver metastases (MLMs), is provided. MLM patients are divided by their primary melanoma location; cutaneous, uvea (eye), and mucosal melanoma. If patients with isolated cutaneous MLMs are considered for surgical resection, treatment with systemic therapy should be part of the treatment course. For uveal MLMs, complete surgical or ablative treatment of all MLMs suggests superior results compared with other liver-directed or systemic therapies, based on current evidence, no recommendations for any liver-directed regional therapy in the treatment of mucosal MLMs can be made.
Collapse
Affiliation(s)
- Diederik J Höppener
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Dirk J Grünhagen
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Alexander M M Eggermont
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, the Netherlands
| | - Astrid A M van der Veldt
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Cornelis Verhoef
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands.
| |
Collapse
|
41
|
Wladis EJ, Rothschild MI, Bohnak CE, Adam AP. New therapies for unresectable or metastatic cutaneous eyelid and orbital melanoma. Orbit 2025; 44:137-143. [PMID: 38796755 DOI: 10.1080/01676830.2024.2351514] [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: 04/06/2024] [Accepted: 04/30/2024] [Indexed: 05/28/2024]
Abstract
PURPOSE Newer treatment options offer the promise of improved outcomes for metastatic and unresectable melanoma. This investigation was performed to review these modalities for cutaneous eyelid and orbital disease. METHODS A search for articles that were related to this subject was performed in the PubMed database, and the bibliographies of these manuscripts were reviewed to ensure capture of the appropriate literature. Data was abstracted and analyzed. RESULTS Historically, patients who suffer from melanoma of the ocular adnexa have fared poorly. Approaches that employ BRAF and mitogen-associated protein kinase inhibitors, immunotherapy, and novel cellular therapies improve outcomes and survival rates, although the side effect profiles of these agents are problematic. Most of the existing strategies have not explored ocular adnexal disease specifically, and treatment plans are generally adapted from the general cutaneous oncology literature. CONCLUSIONS Thanks to advances in our comprehension of the cellular biology of the disease, the management of unresectable and metastatic melanoma has evolved considerably over the past several years. Newer modalities will likely continue to improve survival and reduce adverse events.
Collapse
Affiliation(s)
- Edward J Wladis
- Department of Ophthalmology, Lions Eye Institute, Albany Medical College, Albany, New York, USA
- Department of Otolaryngology, Albany Medical College, Albany, New York, USA
| | - Michael I Rothschild
- Department of Ophthalmology, Lions Eye Institute, Albany Medical College, Albany, New York, USA
| | - Carisa E Bohnak
- Department of Ophthalmology, Lions Eye Institute, Albany Medical College, Albany, New York, USA
| | - Alejandro P Adam
- Department of Ophthalmology, Lions Eye Institute, Albany Medical College, Albany, New York, USA
- Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| |
Collapse
|
42
|
Wikerholmen T, Taule EM, Rigg E, Berle BF, Sættem M, Sarnow K, Saed HS, Sundstrøm T, Thorsen F. Repurposing neuroleptics: clozapine as a novel, adjuvant therapy for melanoma brain metastases. Clin Exp Metastasis 2025; 42:12. [PMID: 39856383 PMCID: PMC11761981 DOI: 10.1007/s10585-025-10328-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/01/2025] [Indexed: 01/27/2025]
Abstract
The blood-brain barrier and the distinct brain immunology provide challenges in translating commonly used chemotherapeutics to treat intracranial tumors. Previous reports suggest anti-tumoral effects of antipsychotics, encouraging investigations into potential treatment effects of neuroleptics on brain metastases. For the first time, the therapeutic potential of the antipsychotic drug clozapine in treating melanoma brain metastases (MBM) was investigated using three human MBM cell lines. Through in vitro cell culture and viability experiments, clozapine displayed potent anti-tumoral effects on MBM cells with an exploitable therapeutic window when compared to normal human astrocytes or rat brain organoids. Further, it was shown that clozapine inhibited migration, proliferation, and colony formation in a dose-dependent manner. Through flow cytometry and proteome screening, we found that clozapine induced apoptosis in MBM cells and potentially altered the tumor immunological environment by upregulating proteins such as macrophage inflammatory protein-1 alpha (MIP-1α) and interleukin-8 (IL-8). In conclusion, clozapine shows significant and selective anti-tumoral effects on MBM cell lines in vitro. Further in vivo experiments are warranted to translate these results into clinical use.
Collapse
Affiliation(s)
- Tobias Wikerholmen
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, Bergen, 5009, Norway
| | - Erlend Moen Taule
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, Bergen, 5009, Norway
| | - Emma Rigg
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, Bergen, 5009, Norway
| | - Birgitte Feginn Berle
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, Bergen, 5009, Norway
| | - Magnus Sættem
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, Bergen, 5009, Norway
| | - Katharina Sarnow
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, Bergen, 5009, Norway
- Department of Neurosurgery, Boston Children's Hospital, 300 longwood Ave, Boston, MA, 02115, USA
| | - Halala Sdik Saed
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, Bergen, 5009, Norway
| | - Terje Sundstrøm
- Department of Neurosurgery, Haukeland University Hospital, Haukelandsveien 22, Bergen, 5021, Norway
- Department of Clinical Medicine, University of Bergen, Jonas Lies Vei 87, Bergen, 5009, Norway
| | - Frits Thorsen
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, Bergen, 5009, Norway.
- Department of Neurosurgery, Haukeland University Hospital, Haukelandsveien 22, Bergen, 5021, Norway.
- Department of Clinical Medicine, University of Bergen, Jonas Lies Vei 87, Bergen, 5009, Norway.
- Molecular Imaging Center, Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, Bergen, 5009, Norway.
| |
Collapse
|
43
|
Villarreal OE, Xu Y, Tran H, Machado A, Prescod D, Anderson A, Minelli R, Peoples M, Martinez AH, Lee HM, Wong CW, Fowlkes N, Kanikarla P, Sorokin A, Alshenaifi J, Coker O, Lin K, Bristow C, Viale A, Shen JP, Parseghian C, Marszalek JR, Corcoran R, Kopetz S. Adaptive Plasticity Tumor Cells Modulate MAPK-Targeting Therapy Response in Colorectal Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.22.634215. [PMID: 39896605 PMCID: PMC11785218 DOI: 10.1101/2025.01.22.634215] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
MAPK pathway inhibitors (MAPKi) are increasingly used in the treatment of advanced colorectal cancer, but often produce short-lived responses in patients. Although acquired resistance by de novo mutations in tumors have been found to reduce response in some patients, additional mechanisms underlying the limited response durability of MAPK targeting therapy remain unknown. Here, we denote new contributory tumor biology and provide insight on the impact of tumor plasticity on therapy response. Analysis of MAPKi treated patients revealed activation of stemness programs and increased ASCL2 expression, which are associated with poor outcomes. Greater ASCL2 with MAPKi treatment was also seen in patient-derived CRC models, independent of driver mutations. We find ASCL2 denotes a distinct cell population, arising from phenotypic plasticity, with a proliferative, stem-like phenotype, and decreased sensitivity to MAPKi therapy, which were named adaptive plasticity tumor (APT) cells. MAPK pathway suppression induces the APT phenotype in cells, resulting in APT cell enrichment in tumors and limiting therapy response in preclinical and clinical data. APT cell depletion improved MAPKi treatment efficacy and extended MAPKi response durability in mice. These findings uncover a cellular program that mitigates the impact of MAPKi therapies and highlights the importance of addressing tumor plasticity to improve clinical outcomes.
Collapse
|
44
|
Miranda I, Jahan N, Shevde LA. The metastatic cascade through the lens of therapeutic inhibition. Cell Rep Med 2025; 6:101872. [PMID: 39706193 PMCID: PMC11866422 DOI: 10.1016/j.xcrm.2024.101872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/21/2024] [Accepted: 11/18/2024] [Indexed: 12/23/2024]
Abstract
Metastasis is a main cause of cancer-related death, and a deeper understanding of the metastatic process will inform more targeted and mechanistic approaches that can abrogate challenges in treatment efficacy and toxicity. Several steps throughout the metastatic cascade, from angiogenesis to secondary tumor formation, offer specific vulnerabilities to therapies that can lead to the decline or cessation of metastatic progression. A deeper understanding of the metastatic cascade also allows combination systemic therapies to be used synergistically. In this review, we describe current treatment modalities in the context of multiple steps of the metastatic cascade. We highlight their mechanisms and present their efficacy across multiple cancers. This work also presents targets within the metastatic cascade in need of more research that can advance the landscape of treatments and lead to the goal of metastatic cancer remission.
Collapse
Affiliation(s)
- Ian Miranda
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nusrat Jahan
- Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lalita A Shevde
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
45
|
Hossain MA. A comprehensive review of targeting RAF kinase in cancer. Eur J Pharmacol 2025; 986:177142. [PMID: 39577552 DOI: 10.1016/j.ejphar.2024.177142] [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: 08/25/2024] [Revised: 11/11/2024] [Accepted: 11/17/2024] [Indexed: 11/24/2024]
Abstract
RAF kinases, particularly the BRAF isoform, play a crucial role in the MAPK/ERK signaling pathway, regulating key cellular processes such as proliferation, differentiation, and survival. Dysregulation of this pathway often caused by mutations in the BRAF gene or alterations in upstream regulators like Ras and receptor tyrosine kinases contributes significantly to cancer development. Mutations, such as BRAF-V600E, are present in a variety of malignancies, with the highest prevalence in melanoma. Targeted therapies against RAF kinases have achieved substantial success, especially in BRAF-V600E-mutant melanomas, where inhibitors like vemurafenib and dabrafenib have demonstrated remarkable efficacy, leading to improved patient outcomes. These inhibitors have also shown clinical benefits in cancers such as thyroid and colorectal carcinoma, although to a lesser extent. Despite these successes, therapeutic resistance remains a major hurdle. Resistance mechanisms, including RAF dimerization, feedback reactivation of the MAPK pathway, and paradoxical activation of ERK signaling, often lead to diminished efficacy over time, resulting in disease progression or even secondary malignancies. In response, current research is focusing on novel therapeutic strategies, including combination therapies that target multiple components of the pathway simultaneously, such as MEK inhibitors used in tandem with RAF inhibitors. Additionally, next-generation RAF inhibitors are being developed to address resistance and enhance therapeutic specificity. This review discusses the clinical advancements in RAF-targeted therapies, with a focus on ongoing efforts to overcome therapeutic resistance and enhance outcomes for cancer patients. It also underscores the persistent challenges in effectively targeting RAF kinase in oncology.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh.
| |
Collapse
|
46
|
Baptista da Mata D, Coelho S, Vilas Boas MI, Silva MJ, Marques D, Ferreira P. Inequalities in Drug Access for Advanced Melanoma: The Prognostic Impact Resulting From the Approval Delay of the Combined Ipilimumab/Nivolumab Treatment in Portugal. Cureus 2025; 17:e78185. [PMID: 40027067 PMCID: PMC11870778 DOI: 10.7759/cureus.78185] [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] [Accepted: 01/29/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction A combination of ipilimumab/nivolumab has demonstrated a median overall survival (mOS) of 71.9 months in advanced melanoma, establishing it as the standard first-line (1L) therapy. However, the approval of this combination by the Portuguese Regulatory Authority occurred 76 months after its approval by the European Authority, leaving tyrosine kinase inhibitors as the only 1L option available for the BRAF-mutated melanoma population. Our study aims to evaluate real-world data from patients with advanced melanoma and assess the potential prognostic impact of the delayed availability of ipilimumab/nivolumab combination therapy on this population. Methods This was an observational, retrospective cohort study conducted at a Portuguese Comprehensive Cancer Center. The study included adult patients with melanoma who received innovative therapies in the 1L between May 2016 and December 2021 and who would meet the criteria for treatment with ipilimumab/nivolumab. The primary outcome measure was mOS; secondary outcome measures included median progression-free survival (mPFS), objective response rate (ORR), and safety data. Results Our study included 172 patients, of which 50% were male, and 32.6% (n = 56) had BRAF-mutated melanoma. In 1L setting, 70.9% received anti-programmed cell death protein 1 (anti-PD-1) monotherapy, while the rest were treated with targeted therapies. The median follow-up time was 57 months. Patients treated with anti-PD-1 had ORR of 36.0%, mPFS of seven months (95% CI 2.9-11.1), and mOS of 19 months (95% CI 7.5-30.4). Among patients treated with targeted therapies, the ORR was 56.0%, mPFS seven months (95% CI 5.1-8.9), and mOS 14 months (95% CI 5.9-22.1). In our population, 10% presented grade 3 or higher adverse events, with no drug-related deaths reported. Conclusion These findings underscore significant disparities in access to innovative therapies in Portugal, which may have adversely impacted patients' outcomes. The delay raises ethical concerns regarding equity in healthcare access and highlights the need for policy measures to expedite the approval and availability of life-extending treatments.
Collapse
Affiliation(s)
| | - Sara Coelho
- Medical Oncology, Instituto Português de Oncologia do Porto, Porto, PRT
| | | | - Maria João Silva
- Medical Oncology, Instituto Português de Oncologia do Porto, Porto, PRT
| | - Dânia Marques
- Medical Oncology, Instituto Português de Oncologia do Porto, Porto, PRT
| | - Paula Ferreira
- Medical Oncology, Instituto Português de Oncologia do Porto, Porto, PRT
| |
Collapse
|
47
|
Forschner A, Nanz L, Maczey-Leber Y, Amaral T, Flatz L, Leiter U. Response and outcome of patients with melanoma skin metastases and immune checkpoint inhibition. Int J Cancer 2025; 156:145-153. [PMID: 39032035 DOI: 10.1002/ijc.35103] [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: 02/27/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 07/22/2024]
Abstract
It is known, that different metastatic organ systems respond differently to immune checkpoint inhibitors (ICIs). In this study, we aimed to investigate the extent to which skin/subcutaneous metastases respond to ICI or targeted therapies (TTs) and whether the response rate differs from that of distant metastases in the same patient. Patients with melanoma diagnosed between January 2021 and September 2023 with at least one skin/subcutaneous metastasis who had received therapy with ICI or TT in an advanced setting were included in the analysis. Best overall response (BOR) was classified according to the revised response evaluation criteria in solid tumors (RECIST). The BOR of skin metastases and visceral metastases to ICI and TT was compared using the chi-square test. Skin metastases treated with ICI a first-line setting showed an overall response rate (ORR) of 44.1%. In contrast, visceral metastases had a higher ORR of 51.1%. However, the difference was not statistically significant (p = .77). Regarding TT, the ORR for skin metastases was 57.1%, compared to 38.5% for visceral metastases (p = .59). Interestingly, the ORR for skin/subcutaneous metastases was notably lower with ICI compared to visceral metastases, in contrast to patients who underwent TT. Skin metastases showed a poorer response to ICI than visceral metastases. Therefore, careful monitoring is recommended to detect non-response early in patients with skin metastases as skin metastases may have a worse response than TT. A larger cohort is needed for a comprehensive analysis and confirmation of our results.
Collapse
Affiliation(s)
- Andrea Forschner
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Lena Nanz
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Yves Maczey-Leber
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Teresa Amaral
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Lukas Flatz
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Ulrike Leiter
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|
48
|
Noguchi T, Ariga S, Moku R, Kikuchi J, Amano T, Maeda T, Ishikawa K, Maeda T, Shiiya A, Goda T, Ohhara Y, Hagio K, Saito Y, Hatanaka KC, Hatanaka Y, Taguchi J, Takeuchi S, Shimizu Y, Kinoshita I. Actionable Gene Alterations Identified in Patients With Malignant Melanoma by Targeted Sequencing in Japan. JCO Precis Oncol 2025; 9:e2400437. [PMID: 39823560 PMCID: PMC11753464 DOI: 10.1200/po-24-00437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/28/2024] [Accepted: 12/04/2024] [Indexed: 01/30/2025] Open
Abstract
PURPOSE Precision medicine plays an important role in the treatment of patients with advanced melanoma. Despite its high incidence in White patients, advanced melanoma is rare in Asian countries, hampering prospective clinical trials targeting the Asian population. This retrospective study aimed to elucidate the real-world molecular diagnoses and outcomes of Japanese patients with melanoma using comprehensive genome profiling (CGP). MATERIALS AND METHODS Patients with melanoma who completed standard anticancer medical treatments (including those expected to complete the treatments) underwent CGP, which is covered by the National Health Insurance. We analyzed the results and clinical annotations of 569 patients registered before August 2023 in a national database. RESULTS Skin, mucosal, and uveal melanomas accounted for 64%, 28%, and 7% of cases, respectively. Patients with BRAF, NRAS, NF1, and KIT variants represented 25%, 20%, 17%, and 17%, respectively. Eighty-two percent of BRAF, 97% of NRAS, 69% of NF1, and 54% of KIT were actionable alterations (ie, BRAF classes I, II, and III, NRAS Q61, G12, G13, NF1 loss-of-function, KIT gain-of-function variants). BRAF V600E/K variants occurred in 22% of skin and 2% of mucosal melanomas, but not in uveal melanomas. The mean tumor mutation burden in cutaneous melanomas was 4.2 variants/Mb. Patients previously treated with BRAF-targeted therapy harbored amplifications of BRAF and cell cycle genes more frequently than therapy-naive patients. Thirty-six patients (6.3%) were treated following the molecular tumor board (MTB) recommendations. CONCLUSION Actionable gene alterations in BRAF, NRAS, NF1, and KIT are common in Japanese patients with melanoma. However, few patients were treated according to the MTB recommendations, suggesting that there is an unmet need to increase accessibility to gene-matched clinical trials in Japan.
Collapse
Affiliation(s)
- Takuro Noguchi
- Department of Medical Oncology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
- Department of Medical Oncology, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Shin Ariga
- Department of Medical Oncology, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Rika Moku
- Department of Medical Oncology, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Junko Kikuchi
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Toraji Amano
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Takuya Maeda
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kosuke Ishikawa
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Taku Maeda
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Akihiko Shiiya
- Department of Medical Oncology, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Tomohiro Goda
- Department of Medical Oncology, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Yoshihito Ohhara
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Kanako Hagio
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Yusuke Saito
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Kanako C. Hatanaka
- Center for Development of Advanced Diagnostics, Hokkaido University Hospital, Sapporo, Japan
| | - Yutaka Hatanaka
- Center for Development of Advanced Diagnostics, Hokkaido University Hospital, Sapporo, Japan
| | - Jun Taguchi
- Department of Medical Oncology, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Satoshi Takeuchi
- Department of Medical Oncology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
- Department of Medical Oncology, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Yasushi Shimizu
- Department of Medical Oncology, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Ichiro Kinoshita
- Department of Medical Oncology, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| |
Collapse
|
49
|
Amaral T, Ottaviano M, Arance A, Blank C, Chiarion-Sileni V, Donia M, Dummer R, Garbe C, Gershenwald JE, Gogas H, Guckenberger M, Haanen J, Hamid O, Hauschild A, Höller C, Lebbé C, Lee RJ, Long GV, Lorigan P, Muñoz Couselo E, Nathan P, Robert C, Romano E, Schadendorf D, Sondak V, Suijkerbuijk KPM, van Akkooi ACJ, Michielin O, Ascierto PA. Cutaneous melanoma: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol 2025; 36:10-30. [PMID: 39550033 DOI: 10.1016/j.annonc.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 11/18/2024] Open
Affiliation(s)
- T Amaral
- Skin Cancer Clinical Trials Center-University of Tuebingen, Tuebingen, Germany
| | - M Ottaviano
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Napoli, Italy
| | - A Arance
- Department of Medical Oncology and IDIBAPS, Hospital Clinic y Provincial de Barcelona, Barcelona, Spain
| | - C Blank
- Department of Medical Oncology and Division of Immunology, The Netherlands Cancer Institute Antoni van Leeuwenhoek Ziekenhuis (NKI), Amsterdam; Leiden University Medical Center (LUMC), Leiden, The Netherlands; University Clinic Regensburg, Regensburg, Germany
| | - V Chiarion-Sileni
- Department of Oncology, Melanoma Unit, Istituto Oncologico Veneto, IOV-IRCCS, Padova, Italy
| | - M Donia
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev and Gentofte, Herlev, Denmark
| | - R Dummer
- Department of Dermatology, Skin Cancer Center, USZ-University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - C Garbe
- Department of Dermatology, Center for DermatoOncology, University Hospital Tuebingen, Tuebingen, Germany
| | - J E Gershenwald
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center and The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, USA
| | - H Gogas
- First Department of Medicine, School of Medicine, National and Kapodistrian University of Athens-School of Medicine, Athens, Greece
| | - M Guckenberger
- Department of Radiation Oncology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - J Haanen
- Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands; Department of Medical Oncology, Leiden University Medical Center (LUMC), Leiden, The Netherlands; Oncology Service, Melanoma Clinic, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - O Hamid
- Medical Oncology, Cutaneous Malignancies, The Angeles Clinic and Research Institute, A Cedars Sinai Affiliate, Los Angeles, USA
| | - A Hauschild
- Department of Dermatology, UKSH-Universitätsklinikum Schleswig-Holstein-Campus Kiel, Kiel, Germany
| | - C Höller
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - C Lebbé
- Université Paris Cite, AP-HP Dermato-oncology and CIC, Cancer Institute APHP, Nord Paris Cité, INSERM U976, Saint Louis Hospital, Paris, France
| | - R J Lee
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK; Faculty of Biology Medicine and Health, The University of Manchester, Manchester, UK
| | - G V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Department Medical Oncology, Royal North Shore Hospital, Sydney, Australia; Mater Hospital, Sydney, Australia
| | - P Lorigan
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK; Faculty of Biology Medicine and Health, The University of Manchester, Manchester, UK
| | - E Muñoz Couselo
- Department of Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - P Nathan
- Mount Vernon Cancer Centre, Northwood, UK
| | - C Robert
- Department of Oncology, Institut Gustave Roussy and Paris-Saclay University, Villejuif, France
| | - E Romano
- Department of Oncology, Center for Cancer Immunotherapy, Institut Curie, Paris, France
| | - D Schadendorf
- Department of Dermatology, WTZ-Westdeutsches Tumorzentrum Essen, National Center for Tumor Diseases (NCT-West), Campus Essen, Essen, Germany; University Alliance Ruhr, Research Center One Health, University Duisburg-Essen, Essen, Germany
| | - V Sondak
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, USA
| | - K P M Suijkerbuijk
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - A C J van Akkooi
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, Australia
| | - O Michielin
- Department of Oncology, Geneva University Hospital, Geneva, Switzerland
| | - P A Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Instituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| |
Collapse
|
50
|
Farma JM, Olszanski AJ, Messina JL, Sondak VK. Annals of Surgical Oncology Practice Guidelines Series: Adjuvant and Neoadjuvant Therapy for Melanoma. Ann Surg Oncol 2025; 32:3-11. [PMID: 39495363 DOI: 10.1245/s10434-024-16418-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 10/11/2024] [Indexed: 11/05/2024]
Abstract
Surgery has always been the mainstay of melanoma treatment, but the risk of recurrence after curative-intent surgery remains high for some stages of the disease. In this Annals of Surgical Oncology Guidelines Review, we provide an overview of practice changing studies, review international guidelines, and highlight current recommendations and areas of controversy when treating melanoma patients in the adjuvant and neoadjuvant setting. Recent clinical trials have established important roles for adjuvant and neoadjuvant therapy in conjunction with surgery for selected patients with stage II, stage III, and even resectable stage IV melanoma. Patients with melanoma should be evaluated for multimodality therapy, including a combination of surgery, systemic therapy (i.e., BRAF-targeted therapy or checkpoint blockade immunotherapy), and at times radiotherapy. With the rapid pace of advances in the field, it is crucial for surgical oncologists to remain updated on the latest guidelines and recommendations for adjuvant and neoadjuvant therapy and to continue to be leaders in this paradigm shift. Given the complex and evolving nature of treatment, this report reviews the latest practice guidelines in the context of modern multidisciplinary management of melanoma.
Collapse
Affiliation(s)
- Jeffrey M Farma
- Department of Surgery, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Anthony J Olszanski
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Jane L Messina
- Departments of Pathology and Cutaneous Oncology, Moffitt Cancer Center, and Department of Oncologic Sciences, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Vernon K Sondak
- Department of Cutaneous Oncology, Moffitt Cancer Center, and Department of Oncologic Sciences, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
| |
Collapse
|