1
|
Amagai R, Fujimura T, Kambayashi Y, Takahashi-Watanabe M, Yamazaki E, Tamabuchi E, Oka K, Muto Y, Hashimoto A, Asano Y. Two cases of advanced melanoma with BRAF L597 mutation: Options for systemic treatment. J Dermatol 2025; 52:1090-1093. [PMID: 40130804 DOI: 10.1111/1346-8138.17718] [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: 02/26/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/26/2025]
Abstract
The combination of BRAF and MEK kinase inhibitors is a well-established treatment for BRAF V600-mutated advanced melanoma. However, the efficacy of these therapies against less common BRAF mutations, such as BRAF L597, remains unclear. We report two cases of advanced melanoma harboring the BRAF L597 mutation. In the first case, a 77-year-old man with metastatic melanoma achieved complete remission following combination therapy with nivolumab and ipilimumab. In the second case, a 50-year-old woman with metastatic melanoma exhibited resistance to multiple systemic therapies, including nivolumab, ipilimumab, and targeted therapy with encorafenib and binimetinib. These cases highlight the variable therapeutic responses in melanoma with the BRAF L597 mutation, suggesting that immune checkpoint inhibitors may be a viable first-line treatment, particularly for patients with a high tumor mutational burden. Further studies are needed to establish optimal treatment strategies for this rare mutation.
Collapse
Affiliation(s)
- Ryo Amagai
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Taku Fujimura
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yumi Kambayashi
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | - Emi Yamazaki
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Erika Tamabuchi
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kenta Oka
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yusuke Muto
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akira Hashimoto
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoshihide Asano
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
2
|
Gianmarco M, Carolina P, Gregorio M, Michela V, Monica P, Claire GG, Michele M, Giulia M, Roberta M, Cinzia A, Lorena B, Marcello T, Fabiana P, Roberta M. Circulating tumor DNA monitoring in advanced mutated melanoma (LIQUID-MEL). THE JOURNAL OF LIQUID BIOPSY 2025; 8:100295. [PMID: 40276578 PMCID: PMC12019447 DOI: 10.1016/j.jlb.2025.100295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025]
Abstract
Introduction Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of metastatic melanoma, but a percentage of patients did not show benefit. Circulating tumor DNA (ctDNA) has emerged as a potential non-invasive tool for monitoring disease evolution and treatment response. The present study aimed to evaluate the clinical utility of ctDNA dynamics in patients with metastatic melanoma receiving ICIs, while exploring its role in the oncological course. Materials and methods The LIQUID-MEL study is a prospective, single-centre pilot study including patients with BRAF/NRAS-mutant metastatic melanoma. ctDNA was quantified using digital droplet PCR (ddPCR) at four different time points. Uni- and multivariable Cox regression models were used to assess the correlation between shedding and progression-free survival (PFS), and overall survival (OS). Results Overall, 23 patients were included. At baseline, ctDNA was detectable in 5/23 (21.7 %) cases. Baseline ctDNA shedding was associated with shorter PFS (3.88 months vs. 0.69 months, p=0.012). A strong numerical trend was observed also in OS (12.66 months vs. 2.53 months, p=0.287). Shedding at baseline did not demonstrate independent prognostic or predictive value in the uni- and multivariable analysis. The longitudinal analysis revealed intriguing patterns of ctDNA shedding in individual patients. Conclusion ctDNA detectability and its dynamic changes during treatment may have potential clinical utility in patients with metastatic melanoma, offering a valuable non-invasive tool for monitoring disease and treatment response. The small sample size limited the statistical power of the analysis. Further studies with larger cohorts are needed to validate its role in routine clinical practice.
Collapse
Affiliation(s)
| | - Palazzi Carolina
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Monica Gregorio
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Verzè Michela
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Pluchino Monica
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Maffezzoli Michele
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Portsmouth Hospital University NHS Trust, Portsmouth, United Kingdom
| | - Mazzaschi Giulia
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Manuguerra Roberta
- Pathology Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Azzoni Cinzia
- Pathology Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Bottarelli Lorena
- Pathology Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Tiseo Marcello
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Perrone Fabiana
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Minari Roberta
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| |
Collapse
|
3
|
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
|
4
|
Thakur V, Thakur VS, Wang D, de Freitas JT, Bianchi A, Nivelo LA, Umland O, Welford SM, Bedogni B. Co-inhibition of Notch1 and ChK1 triggers genomic instability and melanoma cell death increasing the lifespan of mice bearing melanoma brain metastasis. J Exp Clin Cancer Res 2025; 44:163. [PMID: 40437523 PMCID: PMC12117938 DOI: 10.1186/s13046-025-03411-w] [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: 02/18/2025] [Accepted: 05/06/2025] [Indexed: 06/01/2025] Open
Abstract
BACKGROUND Melanoma brain metastases (MBM) are a leading cause of death in patients with advanced disease. MBM treatment relay on targeted and immunotherapy and on stereotactic radiosurgery as gold standard. Life expectancy has improved significantly with these therapies however, targeted therapy is short lived and only about half of the patients respond to immunotherapy, while radiation is limited by melanoma cells intrinsic resistance to DNA damage. New therapeutic approaches are therefore needed to treat MBM. Here we investigate a new role of Notch1 in genomic instability and demonstrate that blockade of both Notch1 and the DNA repair factor ChK1 causes extensive DNA damage and tumor cell death increasing survival in MBM bearing mice. METHODS Anti-Notch1 (anti-N1) was previously described. Prexaserib, a ChK1 inhibitor, is currently in clinical trials. K457 and A375 melanoma cells were used. RNA sequencing was performed in K457 cells treated with anti-N1 and Gene Set Enrichment Analysis performed. DNA damage was evaluated by a DNA fiber assay to assess replication fork speed; and γH2AX foci count and neutral comet assay to quantify double strand breaks. Cell survival was evaluated by trypan blue and a colony formation assay. Luciferase expressing A375 cells were orthotopically inoculated in the right cerebral cortex of athymic nude mice, for in vivo evaluation of a therapy with anti-N1 and prexasertib. Survival was assessed by Kaplan-Meyer survival curves and significance assessed by a Log-rank test. RESULTS Notch1 blockade caused genomic instability by reducing histone availability, leading to DNA replication stress and DNA damage. This in turn, resulted in the activation of the DNA Damage Response pathway ATR/ChK1 to counter the damage. Co-inhibition of Notch1, via anti-N1, and ChK1, via prexasertib (prex), exacerbated DNA damage increasing melanoma cell death. Importantly, combination anti-N1/prex significantly improved survival of mice bearing MBMs. CONCLUSIONS A therapy with anti-N1/prexasertib could represent a novel treatment strategy, alone or in combination with current treatment regimens, for melanoma brain metastases.
Collapse
Affiliation(s)
- Varsha Thakur
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL, 33136, USA
| | - Vijay S Thakur
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL, 33136, USA
| | - Dazhi Wang
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL, 33136, USA
| | - Juliano Tiburcio de Freitas
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL, 33136, USA
| | - Anna Bianchi
- Division of Surgical Oncology, Dewitt Daughtry Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Luis Alberto Nivelo
- Division of Surgical Oncology, Dewitt Daughtry Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Oliver Umland
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Scott M Welford
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL, 33136, USA
| | - Barbara Bedogni
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL, 33136, USA.
| |
Collapse
|
5
|
Kitadai R, Okuma Y, Shibata T, Kohno T, Koyama T. Tissue-agnostic target profiles and treatment efficacy in cancer patients: Insights from the C-CAT clinicogenomic repository. Eur J Cancer 2025; 220:115380. [PMID: 40163907 DOI: 10.1016/j.ejca.2025.115380] [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: 01/12/2025] [Revised: 03/02/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
PURPOSE Utilizing real-world data from Japan's C-CAT clinicogenomic repository, our study demonstrates a significant shift in cancer management through cancer genomic profiling (CGP) tests. The aim of this study is assessing the prevalence of cross-organ targeted genetic alterations and exploring the differences in treatment responses among cancer patients who underwent CGP tests. METHODS Analyzing data from 60,256 patients in the C-CAT repository, we documented the prevalence of FDA-approved biomarkers, cross-organ genetic alterations, and treatment outcomes for tissue-agnostic therapies from June 2019 to December 2023. RESULTS Biomarkers including RET rearrangement, BRAFV600E mutation, and NTRK rearrangement showed varied therapeutic responses, underscoring the need for universal CGP testing to optimize patient outcomes. Notably, our findings highlight variations in response across different age groups, suggesting the potential for age-specific treatment strategies. Comparisons with the AACR Project GENIE database revealed broader implications for the global genomic landscape. CONCLUSION This study emphasizes the crucial role of clinicogenomic repositories in advancing precision oncology across diverse populations, underscoring the utility of integrating clinical and genomic data in national repositories.
Collapse
Affiliation(s)
- Rui Kitadai
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan; Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yusuke Okuma
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Taro Shibata
- Biostatistics Division, Center for Research Administration and Support, National Cancer Center, Tokyo, Japan
| | - Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Takafumi Koyama
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
6
|
Beckhorn CB, Rhodin KE, Leraas HJ, Farrow NE, Lee JS, Beasley GM, Tracy ET. National Trends in Management of the Nodal Basin for Pediatric Patients With Occult Stage III Melanoma in the United States. Pediatr Blood Cancer 2025; 72:e31627. [PMID: 40038909 DOI: 10.1002/pbc.31627] [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: 09/20/2024] [Revised: 01/12/2025] [Accepted: 02/04/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND Following the publication of recent trials (MSLT-I, MSLT-II, DeCOG), routine completion lymph node dissection (CLND) after positive sentinel lymph node biopsy (SLNB) is no longer recommended for adults with melanoma, while adjuvant immunotherapy (IO) was approved for selected patients with positive SLNB. Given the exclusion of pediatric patients from these studies, we aimed to characterize trends in nodal management for pediatric patients with Stage III melanoma. PROCEDURE The National Cancer Database (NCDB) was queried for pediatric patients (age ≤20 years) with melanoma (clinical Stage I/II; pathologic Stage III) who underwent resection from 2012 to 2019. The primary objective was to examine trends in the extent of nodal surgery, the number of lymph nodes examined, and adjuvant IO utilization. Secondary objectives included comparing overall survival (OS) by nodal management and receipt of adjuvant IO using Kaplan-Meier methods. RESULTS Overall, 98 patients met inclusion criteria. From 2012 to 2019, the percentage of patients receiving SLNB alone increased (from 13% to 89%); conversely, therapeutic lymph node dissection (TLND) decreased (from 60% to 0%), as did CLND (from 27% to 11%). Median lymph nodes examined decreased from 2012 to 2019 (from 22 to 2), while receipt of adjuvant IO increased (from 33% to 44%). OS did not differ by nodal management nor receipt of adjuvant IO. CONCLUSIONS The findings of this study support clinical observation after SLNB in pediatric patients with melanoma, as we noted de-escalation in the extent of nodal surgery without compromising OS. We also noted increasing utilization of adjuvant IO among patients with positive SLNB. Multidisciplinary discussion remains vital for managing melanoma in pediatric patients.
Collapse
Affiliation(s)
| | - Kristen E Rhodin
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Harold J Leraas
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Norma E Farrow
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jay S Lee
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
- Division of Surgical Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Georgia M Beasley
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
- Division of Surgical Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Elisabeth T Tracy
- Division of Pediatric Surgery, UNC Children's Hospital, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
7
|
Reitmajer M, Schäffeler N, Bach A, Nanz L, Amaral T, Leiter U, Flatz L, Forschner A. Psychosocial distress and persistent adverse events in long-term survivors of stage IV melanoma - a cross-sectional questionnaire study. J Dtsch Dermatol Ges 2025. [PMID: 40277327 DOI: 10.1111/ddg.15712] [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: 10/30/2024] [Accepted: 02/02/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors and targeted therapies have improved survival in patients with stage IV melanoma. However, the challenges faced by long-term survivors remain unclear. The long-term toxicity and psychosocial impact of these treatments in real-world patients have yet to be reported. MATERIAL AND METHODS We conducted a cross-sectional questionnaire study using established screening tools, including the Hornheide Screening Instrument (HSI), the Distress Thermometer (DT) with the National Comprehensive Cancer Network (NCCN) problem list, and melanoma-specific questions addressing persistent adverse events, social impairments, emotional needs, and financial concerns. RESULTS A total of 159 patients with stage IV melanoma (≥5 years after the initial diagnosis) were enrolled, of whom 93 completed the questionnaire. Approximately one-third of DT/HSI values exceeded the threshold, indicating a need for psycho-oncological support. More than 40% of patients reported persistent treatment-related complaints. Financial and work-related impacts were rare, affecting approximately 8% and 1% of patients, respectively. CONCLUSIONS High rates of psychosocial distress and persistent adverse events were observed, highlighting the need for cancer survivorship programs in the follow-up care of melanoma patients.
Collapse
Affiliation(s)
- Markus Reitmajer
- Department of Dermatology, University Hospital Tuebingen, Tübingen, Germany
| | - Norbert Schäffeler
- Department of Psychosomatic Medicine and Psychotherapy, University Hospital Tuebingen, Tübingen, Germany
| | - Anne Bach
- Department of Psychosomatic Medicine and Psychotherapy, University Hospital Tuebingen, Tübingen, Germany
| | - Lena Nanz
- Department of Dermatology, University Hospital Tuebingen, Tübingen, Germany
| | - Teresa Amaral
- Department of Dermatology, University Hospital Tuebingen, Tübingen, Germany
| | - Ulrike Leiter
- Department of Dermatology, University Hospital Tuebingen, Tübingen, Germany
| | - Lukas Flatz
- Department of Dermatology, University Hospital Tuebingen, Tübingen, Germany
| | - Andrea Forschner
- Department of Dermatology, University Hospital Tuebingen, Tübingen, Germany
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Fager A, Samuelsson M, Olofsson Bagge R, Pivodic A, Bjursten S, Levin M, Jespersen H, Ny L. Immune checkpoint inhibitor therapy is associated with a decreased risk of developing melanoma brain metastases. BJC REPORTS 2025; 3:22. [PMID: 40217072 PMCID: PMC11992042 DOI: 10.1038/s44276-025-00137-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 02/27/2025] [Accepted: 03/23/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Despite recent advancements in metastatic melanoma treatment, the emergence of melanoma brain metastases (MBM) continues to pose a challenge. This study aimed to explore factors associated with MBM development. METHODS This retrospective study included patients diagnosed with advanced melanoma (unresectable stages III and IV [M1a-c]) between 2013 and 2019 at Sahlgrenska University Hospital, Gothenburg, Sweden. Differences in baseline and primary tumor characteristics, mutational status, biomarker levels, and first-line treatment between patients who developed MBM (BM+) and patients who did not develop MBM (BM-) were analyzed using univariable and multivariable Cox proportional hazard regression. RESULT Of 395 patients, 91 subsequently developed MBM. Patients who received immune checkpoint inhibitors (ICI) as first-line treatment had a reduced risk of MBM development (p ≤ 0.001). None of the eleven patients who received CTLA-4 inhibitors as monotherapy or in combination with PD-1 inhibitors as first-line treatment developed brain metastases. Elevated plasma levels of S100B (p = 0.021) and higher metastatic stage (p = 0.047) were also associated with an increased risk of MBM development. CONCLUSION ICI treatment is associated with a decreased risk of MBM development, suggesting a protective role. Elevated S100B levels and stage IV disease at advanced melanoma diagnosis might indicate an increased risk of MBM development.
Collapse
Affiliation(s)
- Anna Fager
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden.
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Matilda Samuelsson
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Roger Olofsson Bagge
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | - Sara Bjursten
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Max Levin
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Jespersen
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Lars Ny
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
11
|
Topouzis S, Papapetropoulos A, Alexander SPH, Cortese-Krott M, Kendall DA, Martemyanov K, Mauro C, Nagercoil N, Panettieri RA, Patel HH, Schulz R, Stefanska B, Stephens GJ, Teixeira MM, Vergnolle N, Wang X, Ferdinandy P. Novel drugs approved by the EMA, the FDA and the MHRA in 2024: A year in review. Br J Pharmacol 2025; 182:1416-1445. [PMID: 39971274 DOI: 10.1111/bph.17458] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 01/04/2025] [Indexed: 02/21/2025] Open
Abstract
In the past year, the European Medicines Agency (EMA), the Food and Drug Administration (FDA) and the Medicines and Healthcare Products Regulatory Agency (MHRA) authorised 53 novel drugs. While the 2024 harvest is not as rich as in 2023, when 70 new chemical entities were approved, the number of 'orphan' drug authorisations in 2024 (21) is similar to that of 2023 (24), illustrating the dynamic development of therapeutics in areas of unmet need. The 2024 approvals of novel protein therapeutics (15) and advanced therapy medicinal products (ATMPs, 6) indicate a sustained trend also noticeable in the 2023 new drugs reviewed in this journal last year (16 and 11, respectively). Clearly, the most striking characteristic of the 2024 drug yield is the creative pharmacological design, which allows these medicines to employ a novel approach to target a disease. Some notable examples are the first drug successfully using a 'dock-and-block' mechanism of inhibition (zenocutuzumab), the first approved drug for schizophrenia designed as an agonist of M1/M4 muscarinic receptors (xanomeline), the first biparatopic antibody (zanidatamab), binding two distinct epitopes of the same molecule, the first haemophilia therapy that instead of relying on external supplementation of clotting factors, restores Factor Xa activity by inhibiting TFPI (marstacimab), or the first ever authorised direct telomerase inhibitor (imetelstat) that reprogrammes the oncogenic drive of tumour cells. In addition, an impressive percentage of novel drugs were first in class (28 out of 53 or 53% of the total) and a substantial number can be considered disease agnostic, indicating the possibility of future approved extensions of their use for additional indications. The 2024 harvest demonstrates the therapeutic potential of innovative pharmacological design, which allows the effective targeting of intractable disorders and addresses crucial, unmet therapeutic needs.
Collapse
Affiliation(s)
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
- Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Steve P H Alexander
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Miriam Cortese-Krott
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pneumology, Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
| | - Dave A Kendall
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, UK
| | | | - Claudio Mauro
- College of Medicine and Health, University of Birmingham, Birmingham, UK
| | | | | | - Hemal H Patel
- VA San Diego Healthcare System and University of California/San Diego, San Diego, California, USA
| | | | | | | | | | - Nathalie Vergnolle
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France
| | - Xin Wang
- University of Manchester, Manchester, UK
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research and Development, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| |
Collapse
|
12
|
Hazariwala V, Rossen JL, Lasky-Zeid J, Waanders A, Bohnsack BL. Baseline ophthalmic findings and complications in pediatric patients treated with MEK inhibitors. J AAPOS 2025; 29:104157. [PMID: 40032239 DOI: 10.1016/j.jaapos.2025.104157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 10/21/2024] [Accepted: 12/15/2024] [Indexed: 03/05/2025]
Abstract
BACKGROUND MEK inhibitors are an increasingly used class of medications associated with ocular complications, including serous retinal detachments, vein occlusions, uveitis, and dry eye syndrome in adults. However, baseline ophthalmic findings and MEK inhibitor-related complications in children are underreported. METHODS The medical records of patients <21 years of age who underwent MEK inhibitor treatment at our institution between 2016 and 2023 were reviewed retrospectively. Patients with baseline examination and at least one follow-up eye examination were included. Systemic and ophthalmic diagnoses, MEK inhibitor treatment course, and ocular findings were extracted from the record. RESULTS A total of 45 patients (23 males) were treated with a MEK inhibitor for tumor with neurofibromatosis type 1 (NF1, n = 20), tumor without NF1 (n = 22), or non-tumor (n = 3) at a mean age of 9.4 ± 4.8 years. Nineteen patients were treated with trametinib, 20 with selumetinib, and 6 with combined trametinib and dabrafenib. Thirty-three patients (73%), all with neurological tumors, had abnormal baseline eye examinations, including optic nerve abnormalities (n = 20), visual acuity impairment (n = 17), visual field defect (n = 8), strabismus (n = 10), or anterior segment abnormalities (n = 14). Average time on MEK inhibitors was 445.5 ± 377.8 days. Three patients with neurologic tumors without NF1 developed ophthalmic side effects of dry eye syndrome (n = 1) or symptomatic anterior uveitis (n = 2) at 6-7 months after initiation of MEK inhibitor. All 3 patients responded to topical medications. CONCLUSIONS The majority of children started on MEK inhibitors had baseline eye abnormalities due to ocular or visual pathway involvement of neurological tumors. Ophthalmic side effects of MEK inhibitors occurred in 7% of children. Additional studies are required to formulate screening guidelines for the pediatric population.
Collapse
Affiliation(s)
- Vikisha Hazariwala
- Division of Ophthalmology, Ann & Robert H. Lurie Children's Hospital of Chicago, Illinois; Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Jennifer L Rossen
- Division of Ophthalmology, Ann & Robert H. Lurie Children's Hospital of Chicago, Illinois; Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Janice Lasky-Zeid
- Division of Ophthalmology, Ann & Robert H. Lurie Children's Hospital of Chicago, Illinois; Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Angela Waanders
- Division of Hematology, Oncology, NeuroOncology, and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Brenda L Bohnsack
- Division of Ophthalmology, Ann & Robert H. Lurie Children's Hospital of Chicago, Illinois; Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
13
|
Galus Ł, Tusień-Małecka D, Mackiewicz J. Elevated lactate dehydrogenase in adjuvant BRAF and MEK inhibitor therapy does not have diagnostic significance in detecting melanoma recurrence. Contemp Oncol (Pozn) 2025; 29:107-112. [PMID: 40330447 PMCID: PMC12051877 DOI: 10.5114/wo.2025.149038] [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: 12/19/2024] [Accepted: 02/16/2025] [Indexed: 05/08/2025] Open
Abstract
Introduction Lactate dehydrogenase (LDH) is an intracellular enzyme the concentration of which in the serum of melanoma patients is a commonly used biomarker for detecting recurrence, monitoring the effectiveness of ongoing systemic treatment, and for determination of prognosis. Material and methods In this report we evaluated the clinical value of elevated LDH during adjuvant BRAF (dabrafenib) and MEK (trametinib) inhibitors in 23 patients after resection of stage III cutaneous, BRAF-mutated melanoma. Results The treatment was administered for one year or until disease progression or unactable toxicity. In all patients, an increase in LDH was observed during treatment, of whom 18 patients had an increase to values above the upper limit of normal, while 4 patients had an increase within normal limits. After discontinuation of dabrafenib with trametinib, a decrease in LDH levels was observed in all patients except one, in whom treatment was discontinued due to disease progression. The increase in LDH was not associated with disease progression. Hypotheses explaining the increase in LDH include, among others, the immunomodulatory effect of BRAF and MEK inhibitors and the effect of drugs in question on the MAPK pathway in wild-type BRAF cells. Conclusions Information on the common increase in LDH in patients undergoing adjuvant therapy with dabrafenib with trametinib will avoid additional imaging studies in many situations and may prevent unnecessary emotional stress for patients.
Collapse
Affiliation(s)
- Łukasz Galus
- Department of Medical and Experimental Oncology, Institute of Oncology, Poznań University of Medical Sciences, Poznań, Poland
| | - Daria Tusień-Małecka
- Department of Medical and Experimental Oncology, Institute of Oncology, Poznań University of Medical Sciences, Poznań, Poland
| | - Jacek Mackiewicz
- Department of Medical and Experimental Oncology, Institute of Oncology, Poznań University of Medical Sciences, Poznań, Poland
| |
Collapse
|
14
|
Peng J, Liu W, Tian J, Shu Y, Zhao R, Wang Y. Non-coding RNAs as key regulators of epithelial-mesenchymal transition in breast cancer. Front Cell Dev Biol 2025; 13:1544310. [PMID: 40201201 PMCID: PMC11975958 DOI: 10.3389/fcell.2025.1544310] [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: 12/13/2024] [Accepted: 03/06/2025] [Indexed: 04/10/2025] Open
Abstract
This study examines the critical role of non-coding RNAs (ncRNAs) in regulating epithelial-mesenchymal transition (EMT) in breast cancer, a prevalent malignancy with significant metastatic potential. EMT, wherein cancer cells acquire mesenchymal traits, is fundamental to metastasis. ncRNAs-such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs)-modulate EMT by influencing gene expression and signaling pathways, affecting cancer cell migration and invasion. This review consolidates recent findings on ncRNA-mediated EMT regulation and explores their diagnostic and therapeutic potential. Specifically, miRNAs inhibit EMT-related transcription factors, while lncRNAs and circRNAs regulate gene expression through interactions with miRNAs, impacting EMT progression. Given the influence of ncRNAs on metastasis and therapeutic resistance, advancing ncRNA-based biomarkers and treatments holds promise for improving breast cancer outcomes.
Collapse
Affiliation(s)
- Jing Peng
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Wenhui Liu
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Jiaju Tian
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yuncong Shu
- School of life science, Lanzhou University, Lanzhou, China
| | - Rui Zhao
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yuping Wang
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
15
|
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
|
16
|
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
|
17
|
Shi Q, Yang Z, Yang H, Xu L, Xia J, Gu J, Chen M, Wang Y, Zhao X, Liao Z, Mou Y, Gu X, Xie T, Sui X. Targeting ion channels: innovative approaches to combat cancer drug resistance. Theranostics 2025; 15:521-545. [PMID: 39744692 PMCID: PMC11671388 DOI: 10.7150/thno.103384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 10/21/2024] [Indexed: 01/11/2025] Open
Abstract
Ion channels, as functional molecules that regulate the flow of ions across cell membranes, have emerged as a promising target in cancer therapy due to their pivotal roles in cell proliferation, metastasis, apoptosis, drug resistance, and so on. Recently, increasing evidence suggests that dysregulation of ion channels is a common characteristic of cancer cells, contributing to their survival and the resistance to conventional therapies. For example, the aberrant expression of sodium (Na+) and potassium ion (K+) channels is significantly correlated with the sensitivity of chemotherapy drugs. The endogenous calcium (Ca2+) channels contribute to the acquired resistance of osimertinib in epidermal growth factor receptor (EGFR) mutant non-small cell lung cancer cell lines. Ferrous ions (Fe2+) enhance the sensitivity of breast cancer cells to doxorubicin treatment. Preclinical models have also demonstrated the effect of specific ion channel blockers or modulators on anticancer drug resistance. This review describes the current understanding about the interaction between ion channels and the therapeutic efficacy of anticancer drugs. Then, the therapeutic potential of ion channel blockers or modulators in enhancing the sensitivity or overcoming the resistance of cancer cells to anticancer therapies is discussed. Targeting ion channels will hopefully offer a novel and promising strategy for overcoming cancer drug resistance.
Collapse
Affiliation(s)
- Qian Shi
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Zijing Yang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Huan Yang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Lihui Xu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Jing Xia
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Jie Gu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Mengting Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yan Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xiaohong Zhao
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Zehua Liao
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yiping Mou
- General Surgery, Cancer Center, Department of Gastrointestinal-Pancreatic Surgery, Zhejiang Provincial People's Hospital, Hangzhou Medical University, Hangzhou, Zhejiang, China
| | - Xidong Gu
- Department of Breast Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xinbing Sui
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
18
|
Swalduz A, Beau-Faller M, Planchard D, Mazieres J, Bayle-Bleuez S, Debieuvre D, Fallet V, Geier M, Cortot A, Couraud S, Daniel C, Domblides C, Pichon E, Fabre E, Larivé S, Lerolle U, Tomasini P, Wislez M, Missy P, Morin F, Westeel V, Auliac JB. Real-world efficacy of the dabrafenib-trametinib (D-T) combination in BRAF V600E-mutated metastatic non-small cell lung cancer (NSCLC): Results from the IFCT-2004 BLaDE cohort. Lung Cancer 2025; 199:108038. [PMID: 39616778 DOI: 10.1016/j.lungcan.2024.108038] [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: 08/06/2024] [Revised: 11/08/2024] [Accepted: 11/23/2024] [Indexed: 02/02/2025]
Abstract
BACKGROUND BRAF V600E mutations occur in 2-5 % of advanced non-small cell lung cancer (NSCLC) patients. The dabrafenib-trametinib (D-T) combination was associated with improved and durable OS in patients in phase II. This study (IFCT-2004 BLaDE study) reported the efficacy of D-T combination in a large retrospective French real-world multicenter cohort of patients with advanced BRAF V600E-mutated NSCLC. METHOD Patients with advanced BRAF V600E-mutated NSCLC diagnosed between 01.01.2016 and 31.12.2019 and treated with D-T in combination, regardless of the treatment line, were included. The primary endpoint was the 12-month OS rate (%) in patients receiving D-T as a second-line therapy or beyond. RESULTS A total of 163 patients were included: 50.3 % were female, 30.2 % were never smokers, 95.1 % had adenocarcinoma, and 78.2 % had a PDL1 ≥ 1 %. The median age was 68.3 years. At D-T initiation, 80.8 % of patients had a PS of 0/1, 78.6 % had stage IV disease, and 20.9 % had brain metastasis. At the cutoff, the median follow-up was 27.4 months. The 12-month OS rate in patients receiving D + T as a second-line therapy or beyond (n = 119) was 67.4 %, with a median progression-free survival (mPFS) of 10.4 months. Among the 44 patients who received D + T as a first-line therapy, the 12-month OS rate was 67.4 %, with an mPFS of 18.2 months. D-T discontinuation for toxicity was reported in 10.3 % of patients. CONCLUSIONS To our knowledge, this is the largest retrospective cohort of BRAF-mutated patients reported. The findings confirmed the significant efficacy of D-T in combination with BRAF V600E-mutated metastatic NSCLC in pretreated and untreated patients. These results under real-world conditions are consistent with those of other registered studies.
Collapse
Affiliation(s)
- Aurélie Swalduz
- Centre Léon Bérard, Department of Medical Oncology, Lyon, France.
| | - Michèle Beau-Faller
- Centre Hospitalier Universitaire Strasbourg, Laboratoire d'Onco-Biologie & Oncologie Thoracique Hôpital de Hautepierre & Nouvel Hôpital Civil, INSERM UMR 1260 - Nanomédecine Régénérative, Université de Strasbourg - CRBS, Strasbourg, France
| | - David Planchard
- Gustave Roussy, Cancer Medicine Department, Villejuif, France
| | | | | | - Didier Debieuvre
- Groupe Hospitalier de la Région Mulhouse Sud-Alsace, Hôpital Emile Muller, GHRMSA - Mulhouse, Mulhouse, France
| | - Vincent Fallet
- Tenon Hospital, Assistance Publique Hôpitaux de Paris, Department of Pneumology and Thoracic Oncology and GRC4, Theranoscan, Sorbonne Université, Paris, France
| | - Margaux Geier
- University Hospital of Brest, Department of Medical Oncology France
| | - Alexis Cortot
- Department of Thoracic Oncology, CHU de Lille, CNRS, Inserm, Institut Pasteur de Lille, UMR9020-U1277-CANTHER, Lille, France
| | - Sébastien Couraud
- Acute Respiratory Medicine and Thoracic Oncology Department, & CIRCAN Program Coordinator, Cancer Institute of Hospices Civils de Lyon, Lyon Sud Hospital, Pierre Bénite, France
| | | | - Charlotte Domblides
- Department of Medical Oncology, University Hospital of Bordeaux, 33000 Bordeaux, France
| | - Eric Pichon
- Centre Hospitalier Universitaire, Tours, France
| | - Elizabeth Fabre
- Department of Thoracic Oncology, Hôpital européen Georges Pompidou, APHP-Centre, Carpem Cancer Institute, Paris, France
| | | | - Ulrike Lerolle
- Clinique Saint-Joseph, Service de Pneumologie, Trélazé, France
| | - Pascale Tomasini
- APHM, Hôpital Nord, Service d'Oncologie Multidisciplinaire & Innovations Thérapeutiques, Marseille, France
| | - Marie Wislez
- APHP, Hôpital Cochin, Service de Pneumologie, Unité d'Oncologie Thoracique, Paris, France
| | - Pascale Missy
- Intergroupe Francophone de Cancérologie Thoracique, Paris, France
| | - Franck Morin
- Intergroupe Francophone de Cancérologie Thoracique, Paris, France
| | | | | |
Collapse
|
19
|
Qian J, Wan J, Yao Q, Chen Y, Ling T, Zhang Y, Tang Z. Cutaneous adverse events associated with BRAF and MEK inhibitors: a systematic review and meta-analysis. Front Pharmacol 2024; 15:1457226. [PMID: 39776585 PMCID: PMC11703664 DOI: 10.3389/fphar.2024.1457226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Aim Cutaneous adverse events (CAEs) after treatment with BRAF and MEK inhibitors in patients with melanoma remain incompletely characterized. To determine the association of BRAF and MEK inhibitor treatment with CAEs in patients with melanoma compared with BRAF inhibitor alone. Method PubMed, Cochrane, Embase and Web of Science were systematically searched for BRAF and MEK inhibitors from database inception through 10 May 2024. Randomized clinical trials reporting on CAEs in patients with melanoma being treated with BRAF and MEK inhibitors compared with patients with melanoma being treated with BRAF inhibitor monotherapy were selected. Pooled Risk ratios (RRs) and 95% CIs were determined using random-effects analyses. The selected end points were alopecia, cutaneous squamous-cell carcinoma, hyperkeratosis, keratoacanthoma, palmoplantar erythrodysaesthesia syndrome, palmoplantar keratoderma, rash, photosensitivity reaction, and skin papilloma. All-grade and high-grade (≥3) CAEs were recorded. Results Comparing with BRAF and MEK inhibitors, treatment with BRAF inhibitors alone was associated with an increased risk of rash (RR, 0.73; 95% CI, 0.54-0.99; p = 0.039; I2 = 88%), alopecia (RR, 0.28; 95% CI, 0.20-0.41; P < 0.001; I2 = 76%), hyperkeratosis (RR, 0.30; 95% CI, 0.22-0.41; P < 0.001; I2 = 56%), palmoplantar erythrodysaesthesia syndrome (RR, 0.21; 95% CI, 0.10-0.47; P < 0.001; I2 = 81%), palmoplantar keratoderma (RR, 0.39; 95% CI, 0.26-0.57; P < 0.001; I2 = 29%), Skin papilloma (RR, 0.25; 95% CI, 0.12-0.52; P < 0.001; I2 = 77%), cutaneous squamous-cell carcinoma (RR, 0.21; 95% CI, 0.11-0.42; P < 0.001; I2 = 50%), and keratoacanthoma (RR, 0.22; 95% CI, 0.12-0.40; P < 0.001; I2 = 0%). Conclusion Therapy with BRAF and MEK inhibitors was associated with a lower risk of CAEs, especially rash, alopecia, hyperkeratosis, palmoplantar erythrodysaesthesia syndrome, palmoplantar keratoderma, skin papilloma, cutaneous squamous-cell carcinoma, and keratoacanthoma, compared with BRAF inhibitor alone. The risks of photosensitivity reaction was similar between the assessed groups. The findings may help to balance between beneficial melanoma treatment and cutaneous morbidity and mortality.
Collapse
Affiliation(s)
- Junhui Qian
- Department of Pharmacy, Shaoxing People’s Hospital, Shaoxing, China
| | - Jinlong Wan
- Department of Gastroenterology, Gaozhou People’s Hospital, Maoming, China
| | - Qin Yao
- Department of Pharmacy, Shaoxing People’s Hospital, Shaoxing, China
| | - Yin Chen
- Department of Pharmacy, Suqian First Hospital, Suqian, China
| | - Tao Ling
- Department of Pharmacy, Suqian First Hospital, Suqian, China
| | - Yuejuan Zhang
- Department of Pharmacy, Shaoxing People’s Hospital, Shaoxing, China
| | - Zhihua Tang
- Department of Pharmacy, Shaoxing People’s Hospital, Shaoxing, China
| |
Collapse
|
20
|
Padovano F, Villa C. The development of drug resistance in metastatic tumours under chemotherapy: An evolutionary perspective. J Theor Biol 2024; 595:111957. [PMID: 39369787 DOI: 10.1016/j.jtbi.2024.111957] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/08/2024]
Abstract
We present a mathematical model of the evolutionary dynamics of a metastatic tumour under chemotherapy, comprising non-local partial differential equations for the phenotype-structured cell populations in the primary tumour and its metastasis. These equations are coupled with a physiologically-based pharmacokinetic model of drug administration and distribution, implementing a realistic delivery schedule. The model is carefully calibrated from the literature, focusing on BRAF-mutated melanoma treated with Dabrafenib as a case study. By means of long-time asymptotic and global sensitivity analyses, as well as numerical simulations, we explore the impact of cell migration from the primary to the metastatic site, physiological aspects of the tumour tissues and drug dose on the development of chemoresistance and treatment efficacy. Our findings provide a possible explanation for empirical evidence indicating that chemotherapy may foster metastatic spread and that metastases may be less impacted by the chemotherapeutic agent.
Collapse
Affiliation(s)
- Federica Padovano
- Sorbonne Université, CNRS, Université de Paris, Laboratoire Jacques-Louis Lions UMR 7598, 4 place Jussieu, 75005 Paris, France.
| | - Chiara Villa
- Sorbonne Université, CNRS, Université de Paris, Inria, Laboratoire Jacques-Louis Lions UMR 7598, 4 place Jussieu, 75005 Paris, France.
| |
Collapse
|
21
|
Mun J, Lim B. The hallmarks of tissue-agnostic therapies and strategies for early anticancer drug discovery. Drug Discov Today 2024; 29:104203. [PMID: 39389456 DOI: 10.1016/j.drudis.2024.104203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 09/24/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024]
Abstract
Recently, precision medicine has enabled biomarker-driven treatment to be administered in a pan-tumor setting, so-called tissue-agnostic therapy. This represents a paradigm shift in cancer care from conventional cancer type-specific to biomarker-centric care, where patients are selected for this type of therapy based on molecular aberrations that occur across cancer types, regardless of tumor site or histology. This approach is particularly important because it offers new treatment options for patients with rare cancers or for whom there are no adequate treatments. Therefore drug development with the goal of tissue agnosticism can accelerate drug approval and address unmet medical needs. Herein, we discuss our opinions on the hallmarks of tissue-agnostic treatments and propose strategies for early drug discovery based on these hallmarks.
Collapse
Affiliation(s)
- Jihyeob Mun
- Center for Supercomputing Applications, Division of National Supercomputing R&D, Korea Institute of Science and Technology Information (KISTI), Daejeon, Republic of Korea
| | - Byungho Lim
- Data Convergence Drug Research Center, Division of Drug Discovery Research, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea; Department of Medicinal Chemistry and Pharmacology, University of Science and Technology (UST), Daejeon, Republic of Korea.
| |
Collapse
|
22
|
Lengyel AS, Meznerics FA, Galajda NÁ, Gede N, Kói T, Mohammed AA, Péter PN, Lakatos AI, Krebs M, Csupor D, Bánvölgyi A, Hegyi P, Holló P, Kemény LV. Safety and Efficacy Analysis of Targeted and Immune Combination Therapy in Advanced Melanoma-A Systematic Review and Network Meta-Analysis. Int J Mol Sci 2024; 25:12821. [PMID: 39684531 DOI: 10.3390/ijms252312821] [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: 10/15/2024] [Revised: 11/23/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
The combinations of BRAF inhibitor-based targeted therapies with immune checkpoint inhibitors currently represent less common therapeutic approaches in advanced melanoma. The aim of this study was to assess the safety and efficacy of currently available melanoma treatments by conducting a systematic review and network meta-analysis. Four databases were systematically searched for randomized clinical studies that included patients with advanced/metastatic melanoma receiving chemotherapy, immune checkpoint inhibitors, BRAF/MEK inhibitor therapy, or combinations thereof. The primary endpoints were treatment-related adverse events (TRAE), serious adverse events (SAE) of grade ≥ 3 adverse events, therapy discontinuation, progression-free survival (PFS), as well as objective response rate (ORR) and complete response rate (CRR). A total of 63 articles were eligible for our systematic review; 59 of them were included in the statistical analysis. A separate subgroup analysis was conducted to evaluate the efficacy outcomes, specifically in BRAF-positive patients. Triple combination therapy or triple therapy (inhibiting BRAF, MEK and PD1/PDL1 axis) showed significantly longer progression-free survival compared to BRAF + MEK combination therapies (HR = 0.76; 95% CI 0.64-0.9), but similar objective and complete response rates in BRAF-mutated melanoma. This safety analysis suggests that triple therapy is not inferior to combined immune checkpoint inhibitors (ICI) and BRAF/MEK therapies in terms of serious adverse events and therapy discontinuation rates. However, monotherapies and BRAF/MEK combinations showed notable advantage over triple therapy in terms of treatment-related adverse events. Combination strategies including BRAF/MEK-targeted therapies with ICI therapies are effective first-line options for advanced, BRAF-mutant melanoma; however, they are associated with more frequent side effects. Therefore, future RCTs are required to evaluate and identify high-risk subpopulations where triple therapy therapies should be considered.
Collapse
Affiliation(s)
- Anna Sára Lengyel
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
- HCEMM-SU Translational Dermatology Research Group, Semmelweis University, 1094 Budapest, Hungary
- Department of Physiology, Semmelweis University, Tűzoltó Str. 37-47, 1094 Budapest, Hungary
| | - Fanni Adél Meznerics
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
| | - Noémi Ágnes Galajda
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
| | - Noémi Gede
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, 7623 Pécs, Hungary
| | - Tamás Kói
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
- Department of Stochastics, Institute of Mathematics, Budapest University of Technology and Economics, 1111 Budapest, Hungary
| | - Alzahra Ahmed Mohammed
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
- HCEMM-SU Translational Dermatology Research Group, Semmelweis University, 1094 Budapest, Hungary
| | - Petra Nikolett Péter
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
- HCEMM-SU Translational Dermatology Research Group, Semmelweis University, 1094 Budapest, Hungary
- Department of Physiology, Semmelweis University, Tűzoltó Str. 37-47, 1094 Budapest, Hungary
| | - Alexandra It Lakatos
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
- HCEMM-SU Translational Dermatology Research Group, Semmelweis University, 1094 Budapest, Hungary
- Department of Physiology, Semmelweis University, Tűzoltó Str. 37-47, 1094 Budapest, Hungary
| | - Máté Krebs
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
- HCEMM-SU Translational Dermatology Research Group, Semmelweis University, 1094 Budapest, Hungary
- Department of Physiology, Semmelweis University, Tűzoltó Str. 37-47, 1094 Budapest, Hungary
| | - Dezső Csupor
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, 7623 Pécs, Hungary
- Institute of Clinical Pharmacy, Faculty of Pharmacy, University of Szeged, 6725 Szeged, Hungary
| | - András Bánvölgyi
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
| | - Péter Hegyi
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, 7623 Pécs, Hungary
- Institute of Pancreatic Diseases, Semmelweis University, 1083 Budapest, Hungary
| | - Péter Holló
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary
| | - Lajos V Kemény
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
- HCEMM-SU Translational Dermatology Research Group, Semmelweis University, 1094 Budapest, Hungary
- Department of Physiology, Semmelweis University, Tűzoltó Str. 37-47, 1094 Budapest, Hungary
- MTA-SE Lendület "Momentum" Dermatooncology Research Group, 1094 Budapest, Hungary
| |
Collapse
|
23
|
Senechal I, Andres MS, Tong J, Ramalingam S, Nazir MS, Rosen SD, Young K, Idaikkadar P, Larkin J, Lyon AR. Risk Stratification, Screening and Treatment of BRAF/MEK Inhibitors-Associated Cardiotoxicity. Curr Oncol Rep 2024; 26:1431-1441. [PMID: 39316222 DOI: 10.1007/s11912-024-01599-2] [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] [Accepted: 08/13/2024] [Indexed: 09/25/2024]
Abstract
PURPOSE OF REVIEW In this review article we describe the cardiovascular adverse events associated with BRAF and MEK inhibitors as well as their pathophysiologic mechanisms and provide up to date guidance for risk stratified surveillance of patients on treatment and the optimal management of emergent cardiotoxicities. RECENT FINDINGS Combination BRAF/MEK inhibition has become an established standard treatment option for patients with a wide variety of BRAF mutant haematological and solid organ cancers, its use is most commonly associated with stage three and metastatic melanoma. The introduction of these targeted drugs has significantly improved the prognosis of previously treatment resistant cancers. It is increasingly recognised that these drugs have a number of cardiovascular toxicities including left ventricular systolic dysfunction, hypertension and QTc interval prolongation. Whilst cardiotoxicity is largely reversible and manageable with medical therapy, it does limit the effective use of these highly active agents.
Collapse
Affiliation(s)
- Isabelle Senechal
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK.
- Centre Hospitalier Universitaire de Québec, Québec City, Québec, Canada.
| | - Maria Sol Andres
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Jieli Tong
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Sivatharshini Ramalingam
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Muhummad Sohaib Nazir
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
- School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Stuart D Rosen
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Kate Young
- Royal Marsden Hospital Foundation Trust, London, UK
| | | | - James Larkin
- Royal Marsden Hospital Foundation Trust, London, UK
| | - Alexander R Lyon
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
24
|
Misiąg P, Molik K, Kisielewska M, Typek P, Skowron I, Karwowska A, Kuźnicki J, Wojno A, Ekiert M, Choromańska A. Amelanotic Melanoma-Biochemical and Molecular Induction Pathways. Int J Mol Sci 2024; 25:11502. [PMID: 39519055 PMCID: PMC11546312 DOI: 10.3390/ijms252111502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/19/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Amelanotic melanoma (AM) is a subtype of hypomelanotic or completely amelanotic melanoma. AM is a rare subtype of melanoma that exhibits a higher recurrence rate and aggressiveness as well as worse surveillance than typical melanoma. AM shows a dysregulation of melanin production, cell cycle control, and apoptosis pathways. Knowing these pathways has an application in medicine due to targeted therapies based on the inhibiting elements of the abovementioned pathways. Therefore, we summarized and discussed AM biochemical and molecular induction pathways and personalized medicine approaches, clinical management, and future directions due to the fact that AM is relatively rare. AM is commonly misdiagnosed. Hence, the role of biomarkers is becoming significant. Nonetheless, there is a shortage of biomarkers specific to AM. BRAF, NRAS, and c-KIT genes are the main targets of therapy. However, the role of BRAF and KIT in AM varied among studies. BRAF inhibitors combined with MAK inhibitors demonstrate better results. Immune checkpoint inhibitors targeting CTLA-4 combined with a programmed death receptor 1 (PD-1) show better outcomes than separately. Fecal microbiota transplantation may overcome resistance to immune checkpoint therapy of AM. Immune-modulatory vaccines against indoleamine 2,3-dioxygenase (IDO) and PD ligand (PD-L1) combined with nivolumab may be efficient in melanoma treatment.
Collapse
Affiliation(s)
- Piotr Misiąg
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (P.M.); (K.M.); (M.K.); (P.T.); (I.S.); (A.K.); (J.K.); (A.W.)
- Students Scientific Group No. 148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Klaudia Molik
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (P.M.); (K.M.); (M.K.); (P.T.); (I.S.); (A.K.); (J.K.); (A.W.)
- Students Scientific Group No. 148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Monika Kisielewska
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (P.M.); (K.M.); (M.K.); (P.T.); (I.S.); (A.K.); (J.K.); (A.W.)
- Students Scientific Group No. 148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Paulina Typek
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (P.M.); (K.M.); (M.K.); (P.T.); (I.S.); (A.K.); (J.K.); (A.W.)
- Students Scientific Group No. 148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Izabela Skowron
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (P.M.); (K.M.); (M.K.); (P.T.); (I.S.); (A.K.); (J.K.); (A.W.)
- Students Scientific Group No. 148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Anna Karwowska
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (P.M.); (K.M.); (M.K.); (P.T.); (I.S.); (A.K.); (J.K.); (A.W.)
- Students Scientific Group No. 148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Jacek Kuźnicki
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (P.M.); (K.M.); (M.K.); (P.T.); (I.S.); (A.K.); (J.K.); (A.W.)
- Students Scientific Group No. 148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Aleksandra Wojno
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (P.M.); (K.M.); (M.K.); (P.T.); (I.S.); (A.K.); (J.K.); (A.W.)
- Students Scientific Group No. 148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Marcin Ekiert
- Department of Oncology, Wroclaw Medical University, pl. L. Hirszfelda 12, 53-413 Wroclaw, Poland;
| | - Anna Choromańska
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| |
Collapse
|
25
|
Brbić M, Yasunaga M, Agarwal P, Leskovec J. Predicting drug outcome of population via clinical knowledge graph. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.06.24303800. [PMID: 38496488 PMCID: PMC10942490 DOI: 10.1101/2024.03.06.24303800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Optimal treatments depend on numerous factors such as drug chemical properties, disease biology, and patient characteristics to which the treatment is applied. To realize the promise of AI in healthcare, there is a need for designing systems that can capture patient heterogeneity and relevant biomedical knowledge. Here we present PlaNet, a geometric deep learning framework that reasons over population variability, disease biology, and drug chemistry by representing knowledge in the form of a massive clinical knowledge graph that can be enhanced by language models. Our framework is applicable to any sub-population, any drug as well drug combinations, any disease, and a wide range of pharmacological tasks. We apply the PlaNet framework to reason about outcomes of clinical trials: PlaNet predicts drug efficacy and adverse events, even for experimental drugs and their combinations that have never been seen by the model. Furthermore, PlaNet can estimate the effect of changing population on trial outcomes with direct implications for patient stratification in clinical trials. PlaNet takes fundamental steps towards AI-guided clinical trials design, offering valuable guidance for realizing the vision of precision medicine using AI.
Collapse
Affiliation(s)
- Maria Brbić
- School of Computer and Communication Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Michihiro Yasunaga
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA
| | - Prabhat Agarwal
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA
| | - Jure Leskovec
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
26
|
Amarillo D, Flaherty KT, Sullivan RJ. Targeted Therapy Innovations for Melanoma. Hematol Oncol Clin North Am 2024; 38:973-995. [PMID: 38971651 DOI: 10.1016/j.hoc.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2024]
Abstract
Melanoma, a malignant tumor of melanocytes, poses a significant clinical challenge due to its aggressive nature and high potential for metastasis. The advent of targeted therapy has revolutionized the treatment landscape of melanoma, particularly for tumors harboring specific genetic alterations such as BRAF V600E mutations. Despite the initial success of targeted agents, resistance inevitably arises, underscoring the need for novel therapeutic strategies. This review explores the latest advances in targeted therapy for melanoma, focusing on new molecular targets, combination therapies, and strategies to overcome resistance.
Collapse
Affiliation(s)
- Dahiana Amarillo
- Oncóloga Médica, Departamento Básico de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Keith T Flaherty
- Mass General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Ryan J Sullivan
- Mass General Cancer Center, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
| |
Collapse
|
27
|
Lee Boniao E, Allen RC, Sundar G. Targeted therapy and immunotherapy for orbital and periorbital tumors: a major review. Orbit 2024; 43:656-673. [PMID: 37728602 DOI: 10.1080/01676830.2023.2256848] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 09/01/2023] [Indexed: 09/21/2023]
Abstract
Traditionally, for patients who are poor candidates for surgery and/or radiotherapy, palliative chemotherapy is often offered but with significant toxic side effects. However, recent advancements in our understanding of tumor biology and molecular genetics have brought new understanding to the molecular pathways of certain tumors and cancers. This has ushered in a new era of precision medicine specific to a tumor or cancer treatment pathway (targeted therapy) or directed to host-tumor responses (immunotherapy). This article will focus on recent updates in the application of available targeted and immunotherapy for managing orbital and periorbital tumors and tumor-like conditions, which include cutaneous basal cell carcinoma, cutaneous squamous cell carcinoma, cutaneous melanoma, Merkel cell carcinoma, sebaceous gland carcinoma, solitary fibrous tumor, dermatofibrosarcoma protuberans, orbital meningioma, neurofibromatosis, Langerhans cell histiocytosis, ocular adnexal lymphoma, orbital lymphatic malformation, and adenoid cystic carcinoma.
Collapse
Affiliation(s)
- Emmanuel Lee Boniao
- Orbit & Oculofacial Surgery, Ophthalmic Oncology, Department of Ophthalmology, National University Hospital, National University of Singapore, Singapore, Singapore
- Department of Ophthalmology, Amai Pakpak Medical Center, Marawi City, Philippines
| | - Richard C Allen
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA
| | - Gangadhara Sundar
- Orbit & Oculofacial Surgery, Ophthalmic Oncology, Department of Ophthalmology, National University Hospital, National University of Singapore, Singapore, Singapore
| |
Collapse
|
28
|
Dini F, Susini P, Zuccaro B, Nisi G, Cuomo R, Grimaldi L, Perillo G, Tinunin L, Antonini P, Innocenti A, Cecchi G, Gambale E, Doni L, Mazzini C, Santoro N, De Giorgi V. Head and neck melanoma: the eyelid region has a better prognosis and easier management. A retrospective survey and systematic review. Melanoma Res 2024; 34:429-438. [PMID: 38833343 DOI: 10.1097/cmr.0000000000000984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Eyelid melanoma (EM) is a malignant neoplasm accounting for around 1% of eyelid malignancies. Because of its rarity, most of our knowledge of EM is currently based on studies of cutaneous melanomas located elsewhere. Accordingly, this study aimed to specifically evaluate EM characteristics, management strategies, and prognosis. A retrospective study was carried out on patients diagnosed with EM at Careggi University Hospital, Florence between May 2012 and May 2022. In addition, a systematic review of relevant literature was conducted, encompassing studies published from 2013 to 2023. Clinical, histopathological, therapeutical, and prognostic data were analyzed to assess the metastasis rate and the 5-year survival rate of patients with EM. Separate data were extracted for in situ and invasive disease. Our original study included 19 patients diagnosed with EM with a 5-year survival rate of 100% for in situ and 83.3% for invasive EM. The literature review identified five poorly detailed large database reviews and 14 original studies on EM with an overall 5-year survival rate of 79.7%. The present research indicates that EM is a challenging malignancy, but has a relatively better prognosis and easier management than other melanomas of the head and neck region. These are probably related to the anatomical location which leads to early diagnosis. Therefore, EM should be considered as a specific disease requiring dedicated treatment. Based on the personal authors' experience and comprehensive overview of the current knowledge, a dedicated protocol is proposed.
Collapse
Affiliation(s)
- Federica Dini
- Department of Neurosciences, Psychology, Drug Research and Child Health Eye Clinic, University of Florence, Florence
| | - Pietro Susini
- Plastic Surgery Unit, Department of Medicine, Surgery, and Neuroscience, University of Siena, Siena
| | | | - Giuseppe Nisi
- Plastic Surgery Unit, Department of Medicine, Surgery, and Neuroscience, University of Siena, Siena
| | - Roberto Cuomo
- Plastic Surgery Unit, Department of Medicine, Surgery, and Neuroscience, University of Siena, Siena
| | - Luca Grimaldi
- Plastic Surgery Unit, Department of Medicine, Surgery, and Neuroscience, University of Siena, Siena
| | | | - Luca Tinunin
- Section of Anatomic Pathology, Department of Health Sciences, University of Florence, Florence
| | - Pietro Antonini
- Section of Anatomic Pathology, Department of Health Sciences, University of Florence, Florence
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona, Verona
| | | | | | | | - Laura Doni
- Clinical Oncology Unit, Careggi University Hospital
| | - Cinzia Mazzini
- Unit of Ocular Oncology, Department of Neuromuscular and Sense Organs
| | - Nicola Santoro
- Unit of Ocular Oncology, Department of Surgery and Translational Medicine, Careggi University Hospital, Florence, Italy
| | | |
Collapse
|
29
|
Okuda-Hiwatashi S, Amagai R, Fujimura T, Kambayashi Y, Watanabe-Takahashi M, Yamazaki E, Tamabuchi E, Itabashi C, Hashimoto A, Asano Y. The Evaluation of Immune Checkpoint Inhibitors and BRAF/MEK Inhibitors in Different Therapy Lines for Metastatic Melanoma: A Retrospective Study. J Clin Med 2024; 13:5560. [PMID: 39337055 PMCID: PMC11432506 DOI: 10.3390/jcm13185560] [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: 08/16/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Background: Nivolumab plus ipilimumab (nivo/ipi) combination therapy is highly effective in treating advanced melanoma, but serious immune-related adverse events (irAEs) are prevalent. The overall response rate (ORR) of the BRAF inhibitor plus MEK inhibitor (BRAFi/MEKi) combination therapy for BRAFV600-mutant advanced melanoma surpasses that of immune checkpoint inhibitors (ICIs). However, the OS and PFS of BRAFi/MEKi combination therapy are inferior to those of ICIs. Methods: We retrospectively evaluated 22 melanoma patients treated with nivo/ipi therapy and 13 patients treated with encorafenib plus binimetinib (enco/bini) between November 2018 and July 2023. Results: The ORR of nivo/ipi for metastatic melanoma patients was significantly higher in the first-line cohort [60.0% (95% CI: 31.2-83.3%)] than in the second-line or beyond cohort [8.3% (95% CI: 0-37.5%)], whereas the ORR of enco/bini was comparable between the first-line cohort [75.0% (95% CI: 28.9-96.6%)] and the second-line or beyond cohort [77.8% (95% CI: 44.3-94.7%)]. The median PFS of nivo/ipi significantly improved in the first-line cohort [7.7 months (95% CI: 2.0-11.9)] compared to the second-line or beyond cohort [2.3 months (95% CI: 0.5-6.0)] (p = 0.0109). In addition to efficacy, the incidence of grade 3 or greater AEs was comparable in the first-line and second-line or beyond cohorts. Conclusions: Although our present data are based on a small number of cases, they suggest that nivo/ipi should be administered as the first-line therapy for the treatment of BRAFV600-mutant metastatic melanoma, rather than enco/bini, aligning with findings from previous clinical trials.
Collapse
Affiliation(s)
| | | | - Taku Fujimura
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan; (S.O.-H.); (R.A.); (Y.K.); (M.W.-T.); (E.Y.); (E.T.); (C.I.); (A.H.); (Y.A.)
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Subbiah V, Gouda MA, Ryll B, Burris HA, Kurzrock R. The evolving landscape of tissue-agnostic therapies in precision oncology. CA Cancer J Clin 2024; 74:433-452. [PMID: 38814103 DOI: 10.3322/caac.21844] [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: 01/20/2024] [Revised: 03/25/2024] [Accepted: 04/09/2024] [Indexed: 05/31/2024] Open
Abstract
Tumor-agnostic therapies represent a paradigm shift in oncology by altering the traditional means of characterizing tumors based on their origin or location. Instead, they zero in on specific genetic anomalies responsible for fueling malignant growth. The watershed moment for tumor-agnostic therapies arrived in 2017, with the US Food and Drug Administration's historic approval of pembrolizumab, an immune checkpoint inhibitor. This milestone marked the marriage of genomics and immunology fields, as an immunotherapeutic agent gained approval based on genomic biomarkers, specifically, microsatellite instability-high or mismatch repair deficiency (dMMR). Subsequently, the approval of NTRK inhibitors, designed to combat NTRK gene fusions prevalent in various tumor types, including pediatric cancers and adult solid tumors, further underscored the potential of tumor-agnostic therapies. The US Food and Drug Administration approvals of targeted therapies (BRAF V600E, RET fusion), immunotherapies (tumor mutational burden ≥10 mutations per megabase, dMMR) and an antibody-drug conjugate (Her2-positive-immunohistochemistry 3+ expression) with pan-cancer efficacy have continued, offering newfound hope to patients grappling with advanced solid tumors that harbor particular biomarkers. In this comprehensive review, the authors delve into the expansive landscape of tissue-agnostic targets and drugs, shedding light on the rationale underpinning this approach, the hurdles it faces, presently approved therapies, voices from the patient advocacy perspective, and the tantalizing prospects on the horizon. This is a welcome advance in oncology that transcends the boundaries of histology and location to provide personalized options.
Collapse
Affiliation(s)
- Vivek Subbiah
- Sarah Cannon Research Institute, Nashville, Tennessee, USA
| | - Mohamed A Gouda
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Bettina Ryll
- Melanoma Patient Network Europe, Uppsala, Sweden
- The Stockholm School of Economics Institute for Research (SIR), Stockholm, Sweden
| | | | | |
Collapse
|
31
|
Martínez-Vila C, González-Navarro EA, Teixido C, Martin R, Aya F, Juan M, Arance A. Lymphocyte T Subsets and Outcome of Immune Checkpoint Inhibitors in Melanoma Patients: An Oncologist's Perspective on Current Knowledge. Int J Mol Sci 2024; 25:9506. [PMID: 39273452 PMCID: PMC11394732 DOI: 10.3390/ijms25179506] [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: 07/17/2024] [Revised: 08/09/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Melanoma is the most aggressive and deadly form of skin cancer, and its incidence has been steadily increasing over the past few decades, particularly in the Caucasian population. Immune checkpoint inhibitors (ICI), anti-PD-1 monotherapy or in combination with anti-CTLA-4, and more recently, anti-PD-1 plus anti-LAG-3 have changed the clinical evolution of this disease. However, a significant percentage of patients do not benefit from these therapies. Therefore, to improve patient selection, it is imperative to look for novel biomarkers. Immune subsets, particularly the quantification of lymphocyte T populations, could contribute to the identification of ICI responders. The main purpose of this review is to thoroughly examine significant published data on the potential role of lymphocyte T subset distribution in peripheral blood (PB) or intratumorally as prognostic and predictive of response biomarkers in advanced melanoma patients treated with ICI regardless of BRAFV600 mutational status.
Collapse
Affiliation(s)
- Clara Martínez-Vila
- Department of Medical Oncology, Althaia Xarxa Assistencial Universitària de Manresa, Dr. Joan Soler, 1-3, 08243 Manresa, Spain
- Programa de Doctorat en Medicina i Recerca Translacional, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
- Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central (IRIS-CC), Roda 70, 08500 Vic, Spain
| | - Europa Azucena González-Navarro
- Department of Immunology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain
| | - Cristina Teixido
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain
- Department of Pathology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain
| | - Roberto Martin
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain
- Department of Medical Oncology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain
- Grupo Español de Terapias Inmunobiológicas en Cáncer (GETICA), Velázquez 7, 28001 Madrid, Spain
| | - Francisco Aya
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain
- Department of Medical Oncology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain
- Grupo Español de Terapias Inmunobiológicas en Cáncer (GETICA), Velázquez 7, 28001 Madrid, Spain
| | - Manel Juan
- Department of Immunology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain
- Grupo Español de Terapias Inmunobiológicas en Cáncer (GETICA), Velázquez 7, 28001 Madrid, Spain
| | - Ana Arance
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain
- Department of Medical Oncology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain
- Grupo Español de Terapias Inmunobiológicas en Cáncer (GETICA), Velázquez 7, 28001 Madrid, Spain
| |
Collapse
|
32
|
Pedersen S, Nielsen MØ, Donia M, Svane IM, Zerahn B, Ellebaek E. Real-World Cardiotoxicity in Metastatic Melanoma Patients Treated with Encorafenib and Binimetinib. Cancers (Basel) 2024; 16:2945. [PMID: 39272803 PMCID: PMC11394091 DOI: 10.3390/cancers16172945] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/16/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Modern therapies targeting the BRAF gene mutation in advanced melanoma have significantly improved patient outcomes but pose cardiovascular risks. This retrospective study in Eastern Denmark (2019-2022) assessed 108 melanoma patients treated with encorafenib and binimetinib. Patients were monitored for heart function using multigated acquisition (MUGA) scans. The study defined major cardiotoxicity as a decline in left ventricular ejection fraction (LVEF) by more than 10 percentage points to below 50%, and minor cardiotoxicity as a decrease in LVEF by more than 15 points but remaining above 50%. Results showed that 19 patients (18%) developed minor cardiotoxicity and were asymptomatic, while 7 (6%) experienced major cardiotoxicity, with two requiring intervention. Notably, no significant declines in LVEF were observed after six months of treatment. The study concluded that significant cardiotoxicity occurred in 6% of cases, mostly asymptomatic and reversible, and suggests that monitoring LVEF could potentially be reduced after 6-9 months if no early signs of cardiotoxicity are detected. This provides valuable insights into the cardiac safety of these treatments in real-world settings.
Collapse
Affiliation(s)
- Sidsel Pedersen
- Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev and Gentofte, 2730 Herlev, Denmark
| | - Marc Østergaard Nielsen
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital, Herlev and Gentofte, 2730 Herlev, Denmark
| | - Marco Donia
- Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev and Gentofte, 2730 Herlev, Denmark
| | - Inge Marie Svane
- Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev and Gentofte, 2730 Herlev, Denmark
| | - Bo Zerahn
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital, Herlev and Gentofte, 2730 Herlev, Denmark
| | - Eva Ellebaek
- Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev and Gentofte, 2730 Herlev, Denmark
| |
Collapse
|
33
|
Dastgheib ZS, Abolmaali SS, Farahavar G, Salmanpour M, Tamaddon AM. Gold nanostructures in melanoma: Advances in treatment, diagnosis, and theranostic applications. Heliyon 2024; 10:e35655. [PMID: 39170173 PMCID: PMC11336847 DOI: 10.1016/j.heliyon.2024.e35655] [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: 02/15/2024] [Revised: 06/16/2024] [Accepted: 08/01/2024] [Indexed: 08/23/2024] Open
Abstract
Melanoma, a lethal form of skin cancer, poses a significant challenge in oncology due to its aggressive nature and high mortality rates. Gold nanostructures, including gold nanoparticles (GNPs), offer myriad opportunities in melanoma therapy and imaging due to their facile synthesis and functionalization, robust stability, tunable physicochemical and optical properties, and biocompatibility. This review explores the emerging role of gold nanostructures and their composites in revolutionizing melanoma treatment paradigms, bridging the gap between nanotechnology and clinical oncology, and offering insights for researchers, clinicians, and stakeholders. It begins by elucidating the potential of nanotechnology-driven approaches in cancer therapy, highlighting the unique physicochemical properties and versatility of GNPs in biomedical applications. Various therapeutic modalities, including photothermal therapy, photodynamic therapy, targeted drug delivery, gene delivery, and nanovaccines, are discussed in detail, along with insights from ongoing clinical trials. In addition, the utility of GNPs in melanoma imaging and theranostics is explored, showcasing their potential in diagnosis, treatment monitoring, and personalized medicine. Furthermore, safety considerations and potential toxicities associated with GNPs are addressed, underscoring the importance of comprehensive risk assessment in clinical translation. Finally, the review concludes by discussing current challenges and future directions, emphasizing the need for innovative strategies to maximize the clinical impact of GNPs in melanoma therapy.
Collapse
Affiliation(s)
- Zahra Sadat Dastgheib
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, 71345, Iran
| | - Samira Sadat Abolmaali
- Pharmaceutical Nanotechnology Department and Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, 71345, Iran
| | - Ghazal Farahavar
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, 71345, Iran
| | - Mohsen Salmanpour
- Cellular and Molecular Biology Research Center, School of Nursing, Larestan University of Medical Sciences, Larestan, Iran
| | - Ali Mohammad Tamaddon
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, 71345, Iran
| |
Collapse
|
34
|
Bialves TS, Bastos LL, Parra JAA, Moysés MN, Marques E, de Castro Pimenta AM, Quintela FM, Mariano DCB, Carvalho FC, de Melo-Minardi RC, Boyle RT. Interaction of DisBa01 peptide from Bothrops alternatus venom with BRAF melanoma receptors: Modeling and molecular docking. Int J Biol Macromol 2024; 274:133283. [PMID: 38909731 DOI: 10.1016/j.ijbiomac.2024.133283] [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: 01/09/2024] [Revised: 06/12/2024] [Accepted: 06/18/2024] [Indexed: 06/25/2024]
Abstract
Metastatic melanoma is highly aggressive and challenging, often leading to a grim prognosis. Its progression is swift, especially when mutations like BRAFV600E continuously activate pathways vital for cell growth and survival. Although several treatments target this mutation, resistance typically emerges over time. In recent decades, research has underscored the potential of snake venoms and peptides as bioactive substances for innovative drugs, including anti-coagulants, anti-microbial, and anti-cancer agents. Leveraging this knowledge, we propose employing a bioinformatics simulation approach to: a) Predict how well a peptide (DisBa01) from Bothrops alternatus snake venom binds to the melanoma receptor BRAFV600E via Molecular Docking. b) Identify the specific peptide binding sites on receptors and analyze their proximity to active receptor sites. c) Evaluate the behavior of resulting complexes through molecular dynamics simulations. d) Assess whether this peptide qualifies as a candidate for anti-melanoma therapy. Our findings reveal that DisBa01 enhances stability in the BRAFV600E melanoma receptor structure by binding to its RGD motif, an interaction absent in the BRAF WT model. Consequently, both docking and molecular dynamics simulations suggest that DisBa01 shows promise as a BRAFV600E inhibitor.
Collapse
Affiliation(s)
- Tatiane Senna Bialves
- Graduate Program in Physiological Sciences (PPGCF), Federal University of Rio Grande - FURG, Av. Italy, s/n - km 8 - Carreiros, Rio Grande, Rio Grande do Sul, Brazil.
| | - Luana Luiza Bastos
- Laboratory of Bioinformatics and Systems, Institute of Exact Sciences, Department of Computer Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - John Alexanders Amaya Parra
- Graduate Program in Biochemistry and Immunology, Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Maurício Nogueira Moysés
- Graduate Program in Biochemistry and Immunology, Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Edleusa Marques
- Graduate Program in Biochemistry and Immunology, Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Adriano Monteiro de Castro Pimenta
- Graduate Program in Biochemistry and Immunology, Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernando Marques Quintela
- Instituto Nacional de Pesquisas do Pantanal- Museu Paraense Emílio Goeldi, Av. Magalhães Barata, 376, Belém, Pará, Brazil
| | - Diego César Batista Mariano
- Laboratory of Bioinformatics and Systems, Institute of Exact Sciences, Department of Computer Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Frederico Chaves Carvalho
- Laboratory of Bioinformatics and Systems, Institute of Exact Sciences, Department of Computer Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Raquel C de Melo-Minardi
- Laboratory of Bioinformatics and Systems, Institute of Exact Sciences, Department of Computer Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Robert Tew Boyle
- Graduate Program in Physiological Sciences (PPGCF), Federal University of Rio Grande - FURG, Av. Italy, s/n - km 8 - Carreiros, Rio Grande, Rio Grande do Sul, Brazil
| |
Collapse
|
35
|
Del Pozzo-Magaña BR, Liy-Wong C. Drugs and the skin: A concise review of cutaneous adverse drug reactions. Br J Clin Pharmacol 2024; 90:1838-1855. [PMID: 35974692 DOI: 10.1111/bcp.15490] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/04/2022] [Accepted: 08/06/2022] [Indexed: 01/19/2023] Open
Abstract
Drug-induced skin disease or cutaneous adverse drug reactions (CADRs) are terms that encompass the clinical manifestations of the skin, mucosae and adnexa induced by a drug or its metabolites. The skin is the organ most frequently affected by drug reactions, which may affect up to 10% of hospitalized patients and occur in 1-3% of multimedicated patients. Most CADRs are mild or self-resolving conditions; however, 2-6.7% of could develop into potentially life-threatening conditions. CADRs represent a heterogeneous field and can be diagnostically challenging as they may potentially mimic any dermatosis. Currently, there are between 29-35 different cutaneous drug-reaction patterns reported ranging from mild dermatitis to an extensively burnt patient. The most frequently reported are maculopapular rash, urticaria/angioedema, fixed drug eruption and erythema multiforme. Less common but more severe patterns include erythroderma, drug reaction with eosinophilia and systemic symptoms, and Stevens-Johnson syndrome/toxic epidermal necrolysis spectrum. Almost any drug can induce a CADR, but antibiotics, nonsteroidal anti-inflammatory drugs and antiepileptics are the most frequently involved. Different mechanisms are involved in the pathogenesis of CADRs, although in some cases, these remain still unknown. CADRs could be classified in different ways: (i) type A (augmented) or type B (bizarre); (ii) immediate or delayed; (iii) immune-mediated or nonimmune-mediated; (iv) nonsevere or life-threatening; and (v) by their phenotype, including exanthematous, urticarial, pustular and blistering morphology. Recognizing a specific CADR will mostly depend on the ability of the physician to perform a detailed clinical examination, the proper description of the morphology of the skin lesions and supporting laboratory and/or skin biopsy findings.
Collapse
Affiliation(s)
- Blanca R Del Pozzo-Magaña
- Department of Pediatrics, Division of Pediatric Clinical Pharmacology, Children's Hospital of Western Ontario, Western University, London, ON, Canada
| | - Carmen Liy-Wong
- Department of Pediatrics, Division of Dermatology and Rheumatology, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
36
|
Wang M, Sullivan RJ, Mooradian MJ. Toxicities from BRAF and MEK Inhibitors: Strategies to Maximize Therapeutic Success. Curr Oncol Rep 2024; 26:934-944. [PMID: 38850505 DOI: 10.1007/s11912-024-01544-3] [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] [Accepted: 05/03/2024] [Indexed: 06/10/2024]
Abstract
PURPOSE OF REVIEW This report highlights several of the recent therapeutic advancements in the treatment of BRAF-mutant tumors, discusses the most common adverse events observed with BRAF-targeted agents, and suggests strategies to manage and mitigate treatment-related toxicities. RECENT FINDINGS BRAF and MEK inhibitors represent a significant advancement in the treatment of BRAF-mutated malignancies with data across tumor types demonstrating the anti-tumor efficacy of dual MAPK inhibition. Although these agents have a reasonable toxicity profile, variable side effects across organ systems can develop. The discovery of activating BRAF mutations and subsequent development of BRAF and MEK inhibitors has transformed the treatment algorithms of BRAF-mutant malignancies. With increased application of these targeted regimens, identification and prompt management of their unique adverse events are crucial.
Collapse
Affiliation(s)
- Mike Wang
- Division of Medical Oncology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Ryan J Sullivan
- Division of Medical Oncology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Meghan J Mooradian
- Division of Medical Oncology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
37
|
Park BS, Jeon H, Kim Y, Kwon H, Choi GE, Chi SG, Park HM, Lee H, Kim T. Polyamine and EIF5A hypusination downstream of c-Myc confers targeted therapy resistance in BRAF mutant melanoma. Mol Cancer 2024; 23:136. [PMID: 38965534 PMCID: PMC11223307 DOI: 10.1186/s12943-024-02031-w] [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: 01/02/2024] [Accepted: 05/24/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND BRAF inhibitors are widely employed in the treatment of melanoma with the BRAF V600E mutation. However, the development of resistance compromises their therapeutic efficacy. Diverse genomic and transcriptomic alterations are found in BRAF inhibitor resistant melanoma, posing a pressing need for convergent, druggable target that reverse therapy resistant tumor with different resistance mechanisms. METHODS CRISPR-Cas9 screens were performed to identify novel target gene whose inhibition selectively targets A375VR, a BRAF V600E mutant cell line with acquired resistance to vemurafenib. Various in vitro and in vivo assays, including cell competition assay, water soluble tetrazolium (WST) assay, live-dead assay and xenograft assay were performed to confirm synergistic cell death. Liquid Chromatography-Mass Spectrometry analyses quantified polyamine biosynthesis and changes in proteome in vemurafenib resistant melanoma. EIF5A hypusination dependent protein translation and subsequent changes in mitochondrial biogenesis and activity were assayed by O-propargyl-puromycin labeling assay, mitotracker, mitoSOX labeling and seahorse assay. Bioinformatics analyses were used to identify the association of polyamine biosynthesis with BRAF inhibitor resistance and poor prognosis in melanoma patient cohorts. RESULTS We elucidate the role of polyamine biosynthesis and its regulatory mechanisms in promoting BRAF inhibitor resistance. Leveraging CRISPR-Cas9 screens, we identify AMD1 (S-adenosylmethionine decarboxylase 1), a critical enzyme for polyamine biosynthesis, as a druggable target whose inhibition reduces vemurafenib resistance. Metabolomic and proteomic analyses reveal that polyamine biosynthesis is upregulated in vemurafenib-resistant cancer, resulting in enhanced EIF5A hypusination, translation of mitochondrial proteins and oxidative phosphorylation. We also identify that sustained c-Myc levels in vemurafenib-resistant cancer are responsible for elevated polyamine biosynthesis. Inhibition of polyamine biosynthesis or c-Myc reversed vemurafenib resistance both in vitro cell line models and in vivo in a xenograft model. Polyamine biosynthesis signature is associated with poor prognosis and shorter progression free survival after BRAF/MAPK inhibitor treatment in melanoma cohorts, highlighting the clinical relevance of our findings. CONCLUSIONS Our findings delineate the molecular mechanisms involving polyamine-EIF5A hypusination-mitochondrial respiration pathway conferring BRAF inhibitor resistance in melanoma. These targets will serve as effective therapeutic targets that can maximize the therapeutic efficacy of existing BRAF inhibitors.
Collapse
Affiliation(s)
- Byung-Sun Park
- Medicinal Materials Research Center, Korea Institute of Science and Technology, 5 Hwarangro-14-Gil, SeongbukGu, Seoul, 02792, Republic of Korea
- Department of Life Sciences, Korea University, 145 AnamRo, SeongbukGu, Seoul, 02841, Republic of Korea
| | - Heeju Jeon
- Medicinal Materials Research Center, Korea Institute of Science and Technology, 5 Hwarangro-14-Gil, SeongbukGu, Seoul, 02792, Republic of Korea
- Department of Life Sciences, Korea University, 145 AnamRo, SeongbukGu, Seoul, 02841, Republic of Korea
| | - Yeonseo Kim
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology, 5 Hwarangro- 14-Gil, SeongbukGu, Seoul, 02792, Republic of Korea
| | - Haejin Kwon
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, 5 Hwarangro-14-Gil, SeongbukGu, Seoul, 02792, Republic of Korea
| | - Ga-Eun Choi
- Medicinal Materials Research Center, Korea Institute of Science and Technology, 5 Hwarangro-14-Gil, SeongbukGu, Seoul, 02792, Republic of Korea
| | - Sung-Gil Chi
- Department of Life Sciences, Korea University, 145 AnamRo, SeongbukGu, Seoul, 02841, Republic of Korea
| | - Hyun-Mee Park
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, 5 Hwarangro-14-Gil, SeongbukGu, Seoul, 02792, Republic of Korea
| | - Hyunbeom Lee
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology, 5 Hwarangro- 14-Gil, SeongbukGu, Seoul, 02792, Republic of Korea
| | - Tackhoon Kim
- Medicinal Materials Research Center, Korea Institute of Science and Technology, 5 Hwarangro-14-Gil, SeongbukGu, Seoul, 02792, Republic of Korea.
- Department of Life Sciences, Korea University, 145 AnamRo, SeongbukGu, Seoul, 02841, Republic of Korea.
- Division of Bio-Medical Science and Technology, Korea University of Science and Technology, 217 GajeongRo YuseongGu, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
38
|
Takano Y, Shimokata T, Urakawa H, Kikumori T, Ando Y. Long-term response to MEK inhibitor monotherapy in a patient with papillary thyroid carcinoma harboring BRAF V600E mutation. Int Cancer Conf J 2024; 13:184-188. [PMID: 38962055 PMCID: PMC11217198 DOI: 10.1007/s13691-024-00670-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 03/04/2024] [Indexed: 07/05/2024] Open
Abstract
Solid tumors harboring mutations in the Braf gene (BRAF) are currently treated by combination Braf/MEK inhibitor therapy, and there is an extensive literature on patient response rates. Alternatively, few studies have documented the clinical response of BRAF mutation-positive solid tumors to MEK inhibitor monotherapy. We report the case of a 57-year-old female diagnosed with papillary thyroid carcinoma and progressive lung metastases initially treated by total thyroidectomy and subsequent thyroid-stimulating hormone suppression therapy. Next-generation sequencing revealed that the tumor harbored a BRAF V600E mutation, and the patient was enrolled in a clinical study of the oral MEK1/2 inhibitor binimetinib. Shortly after starting treatment, the patient experienced pneumothorax due to rapid regression of lung metastases, and computed tomography after 6 months of binimetinib treatment revealed a partial sustained response. One year later, the dose was reduced because of an acneiform rash. After 5 years of binimetinib treatment, lung metastases had regrown, and treatment was switched to the oral multikinase inhibitor lenvatinib. This case demonstrates the potential of MEK inhibitor monotherapy as an alternative treatment for BRAF mutation-positive papillary thyroid carcinoma.
Collapse
Affiliation(s)
- Yuko Takano
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Japan
- Department of Breast and Endocrine Surgery, Nagoya University Hospital, Nagoya, Japan
| | - Tomoya Shimokata
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Japan
| | - Hiroshi Urakawa
- Department of Advanced Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Toyone Kikumori
- Department of Breast and Endocrine Surgery, Nagoya University Hospital, Nagoya, Japan
| | - Yuichi Ando
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Japan
- Department of Advanced Medicine, Nagoya University Hospital, Nagoya, Japan
| |
Collapse
|
39
|
Perrone C, Angioli R, Luvero D, Giannini A, Di Donato V, Cuccu I, Muzii L, Raspagliesi F, Bogani G. Targeting BRAF pathway in low-grade serous ovarian cancer. J Gynecol Oncol 2024; 35:e104. [PMID: 38768941 PMCID: PMC11262891 DOI: 10.3802/jgo.2024.35.e104] [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: 02/19/2024] [Revised: 04/14/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024] Open
Abstract
Mutations in genes encoding for proteins along the RAS-RAF-MEK-ERK pathway have been detected in a variety of tumor entities including ovarian carcinomas. In the recent years, several inhibitors of this pathway have been developed, whose antitumor potential is currently being assessed in different clinical trials. Low grade serous ovarian carcinoma, is a rare gynecological tumor which shows favorable overall survival, compared to the general ovarian cancer population, but worrying resistance to conventional chemotherapies. The clinical behavior of low grade serous ovarian carcinoma reflects the different gene profile compared to high-grade serous carcinoma: KRAS/BRAF mutations. BRAF inhibitors as single agents were approved for the treatment of BRAF mutated tumors. Nevertheless, many patients face progressive disease. The understanding of the mechanisms of resistance to BRAF inhibitors therapy and preclinical studies showing that BRAF and mitogen-activated protein kinase kinase (MEK) inhibitors combined therapy delays the onset of resistance compared to BRAF inhibitor single agent, led to the clinical investigation of combined therapy. The aim of this paper is to review the efficacy and safety of the combination of BRAF plus MEK inhibitors on ovarian carcinomas, in particularly focusing on low grade serous ovarian carcinoma.
Collapse
Affiliation(s)
- Chiara Perrone
- Department of Gynecological, Obstetrical and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Roberto Angioli
- Department of Gynecology, Campus Bio-Medico University Hospital Foundation, Rome, Italy
| | - Daniela Luvero
- Department of Gynecology, Campus Bio-Medico University Hospital Foundation, Rome, Italy
| | - Andrea Giannini
- Department of Gynecological, Obstetrical and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Violante Di Donato
- Department of Gynecological, Obstetrical and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Ilaria Cuccu
- Department of Gynecological, Obstetrical and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Ludovico Muzii
- Department of Gynecological, Obstetrical and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Francesco Raspagliesi
- Gynecologic Oncologic Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Giorgio Bogani
- Gynecologic Oncologic Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy.
| |
Collapse
|
40
|
Slominski RM, Kim TK, Janjetovic Z, Brożyna AA, Podgorska E, Dixon KM, Mason RS, Tuckey RC, Sharma R, Crossman DK, Elmets C, Raman C, Jetten AM, Indra AK, Slominski AT. Malignant Melanoma: An Overview, New Perspectives, and Vitamin D Signaling. Cancers (Basel) 2024; 16:2262. [PMID: 38927967 PMCID: PMC11201527 DOI: 10.3390/cancers16122262] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Melanoma, originating through malignant transformation of melanin-producing melanocytes, is a formidable malignancy, characterized by local invasiveness, recurrence, early metastasis, resistance to therapy, and a high mortality rate. This review discusses etiologic and risk factors for melanoma, diagnostic and prognostic tools, including recent advances in molecular biology, omics, and bioinformatics, and provides an overview of its therapy. Since the incidence of melanoma is rising and mortality remains unacceptably high, we discuss its inherent properties, including melanogenesis, that make this disease resilient to treatment and propose to use AI to solve the above complex and multidimensional problems. We provide an overview on vitamin D and its anticancerogenic properties, and report recent advances in this field that can provide solutions for the prevention and/or therapy of melanoma. Experimental papers and clinicopathological studies on the role of vitamin D status and signaling pathways initiated by its active metabolites in melanoma prognosis and therapy are reviewed. We conclude that vitamin D signaling, defined by specific nuclear receptors and selective activation by specific vitamin D hydroxyderivatives, can provide a benefit for new or existing therapeutic approaches. We propose to target vitamin D signaling with the use of computational biology and AI tools to provide a solution to the melanoma problem.
Collapse
Affiliation(s)
- Radomir M. Slominski
- Department of Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Tae-Kang Kim
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.-K.K.); (Z.J.); (E.P.); (C.E.); (C.R.)
| | - Zorica Janjetovic
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.-K.K.); (Z.J.); (E.P.); (C.E.); (C.R.)
| | - Anna A. Brożyna
- Department of Human Biology, Institute of Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Torun, Poland;
| | - Ewa Podgorska
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.-K.K.); (Z.J.); (E.P.); (C.E.); (C.R.)
| | - Katie M. Dixon
- School of Medical Sciences, The University of Sydney, Sydney, NSW 2050, Australia; (K.M.D.); (R.S.M.)
| | - Rebecca S. Mason
- School of Medical Sciences, The University of Sydney, Sydney, NSW 2050, Australia; (K.M.D.); (R.S.M.)
| | - Robert C. Tuckey
- School of Molecular Sciences, University of Western Australia, Perth, WA 6009, Australia;
| | - Rahul Sharma
- Department of Biomedical Informatics and Data Science, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - David K. Crossman
- Department of Genetics and Bioinformatics, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Craig Elmets
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.-K.K.); (Z.J.); (E.P.); (C.E.); (C.R.)
| | - Chander Raman
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.-K.K.); (Z.J.); (E.P.); (C.E.); (C.R.)
| | - Anton M. Jetten
- Cell Biology Section, NIEHS—National Institutes of Health, Research Triangle Park, NC 27709, USA;
| | - Arup K. Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Andrzej T. Slominski
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.-K.K.); (Z.J.); (E.P.); (C.E.); (C.R.)
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Pathology and Laboratory Medicine Service, Veteran Administration Medical Center, Birmingham, AL 35233, USA
| |
Collapse
|
41
|
Fabiani I, Chianca M, Aimo A, Emdin M, Dent S, Fedele A, Cipolla CM, Cardinale DM. Use of new and emerging cancer drugs: what the cardiologist needs to know. Eur Heart J 2024; 45:1971-1987. [PMID: 38591670 DOI: 10.1093/eurheartj/ehae161] [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: 11/18/2022] [Revised: 02/28/2024] [Accepted: 03/04/2024] [Indexed: 04/10/2024] Open
Abstract
The last decade has witnessed a paradigm shift in cancer therapy, from non-specific cytotoxic chemotherapies to agents targeting specific molecular mechanisms. Nonetheless, cardiovascular toxicity of cancer therapies remains an important concern. This is particularly relevant given the significant improvement in survival of solid and haematological cancers achieved in the last decades. Cardio-oncology is a subspecialty of medicine focusing on the identification and prevention of cancer therapy-related cardiovascular toxicity (CTR-CVT). This review will examine the new definition of CTR-CVT and guiding principles for baseline cardiovascular assessment and risk stratification before cancer therapy, providing take-home messages for non-specialized cardiologists.
Collapse
Affiliation(s)
- Iacopo Fabiani
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy
| | - Michela Chianca
- Interdisciplinary Center for Health Science, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Alberto Aimo
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy
- Interdisciplinary Center for Health Science, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Michele Emdin
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy
- Interdisciplinary Center for Health Science, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Susan Dent
- Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Antonella Fedele
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Carlo Maria Cipolla
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Daniela Maria Cardinale
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| |
Collapse
|
42
|
Amouzegar A, Haig S, Kahn AM, Tawbi HA, Jones JA, Goldberg SB. Navigating the Complexities of Brain Metastases Management. Am Soc Clin Oncol Educ Book 2024; 44:e433694. [PMID: 38781565 DOI: 10.1200/edbk_433694] [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: 05/25/2024]
Abstract
The management of brain metastases, a potentially devastating complication of advanced cancers, has become increasingly complex with advancements in local and systemic therapies. Improved outcomes and extended survival for patients with metastatic solid tumors have led to a surge in the prevalence and possibly incidence of brain metastases, affecting up to 40% of individuals with solid tumors. Enhanced imaging technologies contribute to more accurate and early detection, shaping the understanding of the intricate landscape of this condition. Traditionally, surgery and radiation stood as the mainstays of treatment because of the limited efficacy of systemic therapies within the brain. However, emerging clinical data, particularly in melanoma, lung, and breast cancers, reveal promising results with novel systemic treatments such as immunotherapy and targeted therapies. Despite the historical exclusion of patients with active brain metastases from clinical trials, a shift is occurring toward a more inclusive approach. This chapter delves into the multifaceted challenges associated with managing brain metastases, with a focus on the evolving landscape of systemic approaches as well as the intricacies of shared decision making, providing a comprehensive overview of the current state and future directions in navigating the complexities of brain metastases management.
Collapse
Affiliation(s)
- Afsaneh Amouzegar
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Shannon Haig
- Lake Erie College of Osteopathic Medicine, Greensburg, PA
| | - Adriana M Kahn
- Section of Medical Oncology, Department of Medicine, Yale School of Medicine, New Haven, CT
| | - Hussein A Tawbi
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Joshua A Jones
- Department of Oncology, Division of Radiation Oncology and Division of Palliative Medicine, Rochester Regional Health System, Rochester, NY
| | - Sarah B Goldberg
- Section of Medical Oncology, Department of Medicine, Yale School of Medicine, New Haven, CT
| |
Collapse
|
43
|
Amagai R, Fujimura T, Yamazaki E, Takahashi M, Tamabuchi E, Kambayashi Y, Hashimoto A, Hashimoto K, Asano Y. Vogt-Koyanagi-Harada disease-like uveitis after drug therapy including BRAF/MEK inhibitors in melanoma patients with HLA-DRB1*04. J Dermatol 2024; 51:854-857. [PMID: 38111371 DOI: 10.1111/1346-8138.17082] [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: 09/25/2023] [Revised: 11/26/2023] [Accepted: 12/05/2023] [Indexed: 12/20/2023]
Abstract
The combination of BRAF kinase inhibitors (BRAFis) and MEK kinase inhibitors (MEKis) is one of the most promising chemotherapy regimens in the treatment of BRAF-mutant melanoma. Although BRAFi plus MEKi combined therapy is widely used for the treatment of BRAFV600-mutated melanoma, the incidence of uveitis caused by BRAFi plus MEKi is limited. In this report, we described five cases (two men and three women) of Vogt-Koyanagi-Harada (VKH) disease-like uveitis in melanoma patients who received BRAFi plus MEKi combined therapy. Of note, all the patients had the HLA-DRB1*04 haplotype, which is frequently detected in VKH-like non-infectious uveitis. On the other hand, among BRAFi plus MEKi-treated patients who did not develop VKH disease-like uveitis, only one of five (20%) patients had the HLA-DRB1*04 haplotype. Collectively, BRAFi/MEKi might induce severe VKH disease-like uveitis in melanoma patients with the HLA-DRB1*04 haplotype.
Collapse
Affiliation(s)
- Ryo Amagai
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Taku Fujimura
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Emi Yamazaki
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Manami Takahashi
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Erika Tamabuchi
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yumi Kambayashi
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akira Hashimoto
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuki Hashimoto
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoshihide Asano
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
44
|
Algazi AP, Moon J, Lao CD, Chmielowski B, Kendra KL, Lewis KD, Gonzalez R, Kim K, Godwin JE, Curti BD, Latkovic-Taber M, Lomeli SH, Gufford BT, Scumpia PO, Lo RS, Othus M, Ribas A. A phase 1 study of triple-targeted therapy with BRAF, MEK, and AKT inhibitors for patients with BRAF-mutated cancers. Cancer 2024; 130:1784-1796. [PMID: 38261444 DOI: 10.1002/cncr.35200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/30/2023] [Accepted: 11/21/2023] [Indexed: 01/25/2024]
Abstract
BACKGROUND Aberrant PI3K/AKT signaling in BRAF-mutant cancers contributes to resistance to BRAF inhibitors. The authors examined dual MAPK and PI3K pathway inhibition in patients who had BRAF-mutated solid tumors (ClinicalTrials.gov identifier NCT01902173). METHODS Patients with BRAF V600E/V600K-mutant solid tumors received oral dabrafenib at 150 mg twice daily with dose escalation of oral uprosertib starting at 50 mg daily, or, in the triplet cohorts, with dose escalation of both oral trametinib starting at 1.5 mg daily and oral uprosertib starting at 25 mg daily. Dose-limiting toxicities (DLTs) were assessed within the first 56 days of treatment. Radiographic responses were assessed at 8-week intervals. RESULTS Twenty-seven patients (22 evaluable) were enrolled in parallel doublet and triplet cohorts. No DLTs were observed in the doublet cohorts (N = 7). One patient had a DLT at the maximum administered dose of triplet therapy (dabrafenib 150 mg twice daily and trametinib 2 mg daily plus uprosertib 75 mg daily). Three patients in the doublet cohorts had partial responses (including one who had BRAF inhibitor-resistant melanoma). Two patients in the triplet cohorts had a partial response, and one patient had an unconfirmed partial response. Pharmacokinetic data suggested reduced dabrafenib and dabrafenib metabolite exposure in patients who were also exposed to both trametinib and uprosertib, but not in whose who were exposed to uprosertib without trametinib. CONCLUSIONS Concomitant inhibition of both the MAPK and PI3K-AKT pathways for the treatment of BRAF-mutated cancers was well tolerated, leading to objective responses, but higher level drug-drug interactions affected exposure to dabrafenib and its metabolites.
Collapse
Affiliation(s)
- Alain P Algazi
- University of California-San Francisco, San Francisco, California, USA
| | - James Moon
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Southwest Oncology Group Statistical Center, Seattle, Washington, USA
| | | | - Bartosz Chmielowski
- Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, California, USA
| | - Kari L Kendra
- The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Karl D Lewis
- University of Colorado Comprehensive Cancer Center, Denver, Colorado, USA
| | - Rene Gonzalez
- University of Colorado Comprehensive Cancer Center, Denver, Colorado, USA
| | - Kevin Kim
- California Pacific Medical Center Research Institute, San Francisco, California, USA
| | | | | | | | - Shirley H Lomeli
- Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, California, USA
| | | | - Philip O Scumpia
- Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, California, USA
| | - Roger S Lo
- Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, California, USA
| | - Megan Othus
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Antoni Ribas
- Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, California, USA
| |
Collapse
|
45
|
Ichikawa K, Ohno S, Kubo S, Nakajima H. Large-vessel vasculitis possibly induced by BRAF and MEK inhibitors for BRAF V600E positive lung adenocarcinoma. BMJ Case Rep 2024; 17:e255958. [PMID: 38719253 PMCID: PMC11085983 DOI: 10.1136/bcr-2023-255958] [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: 05/13/2024] Open
Abstract
The combination therapy of v-Raf murine sarcoma viral oncogene homolog B1 (BRAF) and mitogen-activated protein kinase kinase (MEK) inhibitors is approved for treating patients with BRAF V600E-positive tumours, including melanoma and lung cancer. Several case reports indicated autoimmune side effects associated with the use of BRAF and MEK inhibitors. Still, the effects of these drugs on the immune system were not fully elucidated. Here, we report a patient with large-vessel vasculitis diagnosed after initiation of treatment with dabrafenib and trametinib for BRAF V600E-positive metastatic lung adenocarcinoma. She was a never-smoker woman in her early 70s who presented with a chronic cough and was diagnosed with BRAF V600E-positive metastatic lung adenocarcinoma by transbronchial lung biopsy. She was successfully treated with prednisolone and methotrexate while BRAF and MEK inhibitors were continued. We should be careful about autoimmune diseases using BRAF and MEK inhibitors.
Collapse
Affiliation(s)
- Kento Ichikawa
- Center for Rheumatic Diseases, Yokohama City University Medical Center, Yokohama, Japan
| | - Shigeru Ohno
- Center for Rheumatic Diseases, Yokohama City University Medical Center, Yokohama, Japan
| | - Sousuke Kubo
- Respiratory Disease Center, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Hideaki Nakajima
- Department of Stem Cell and Immune Regulation, Yokohama City University School of Medicine Graduate School of Medicine, Yokohama, Kanagawa, Japan
| |
Collapse
|
46
|
de Sauvage MA, Torrini C, Nieblas-Bedolla E, Summers EJ, Sullivan E, Zhang BS, Batchelor E, Marion B, Yamazawa E, Markson SC, Wakimoto H, Nayyar N, Brastianos PK. The ERK inhibitor LY3214996 augments anti-PD-1 immunotherapy in preclinical mouse models of BRAFV600E melanoma brain metastasis. Neuro Oncol 2024; 26:889-901. [PMID: 38134951 PMCID: PMC11066918 DOI: 10.1093/neuonc/noad248] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) have revolutionized cancer treatment; however, only a subset of patients with brain metastasis (BM) respond to ICI. Activating mutations in the mitogen-activated protein kinase signaling pathway are frequent in BM. The objective of this study was to evaluate whether therapeutic inhibition of extracellular signal-regulated kinase (ERK) can improve the efficacy of ICI for BM. METHODS We used immunotypical mouse models of BM bearing dual extracranial/intracranial tumors to evaluate the efficacy of single-agent and dual-agent treatment with selective ERK inhibitor LY3214996 (LY321) and anti-programmed death receptor 1 (PD-1) antibody. We verified target inhibition and drug delivery, then investigated treatment effects on T-cell response and tumor-immune microenvironment using high-parameter flow cytometry, multiplex immunoassays, and T-cell receptor profiling. RESULTS We found that dual treatment with LY321 and anti-PD-1 significantly improved overall survival in 2 BRAFV600E-mutant murine melanoma models but not in KRAS-mutant murine lung adenocarcinoma. We demonstrate that although LY321 has limited blood-brain barrier (BBB) permeability, combined LY321 and anti-PD-1 therapy increases tumor-infiltrating CD8+ effector T cells, broadens the T-cell receptor repertoire in the extracranial tumor, enriches T-cell clones shared by the periphery and brain, and reduces immunosuppressive cytokines and cell populations in tumors. CONCLUSIONS Despite the limited BBB permeability of LY321, combined LY321 and anti-PD-1 treatment can improve intracranial disease control by amplifying extracranial immune responses, highlighting the role of extracranial tumors in driving intracranial response to treatment. Combined ERK and PD-1 inhibition is a promising therapeutic approach, worthy of further investigation for patients with melanoma BM.
Collapse
Affiliation(s)
- Magali A de Sauvage
- Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts, USA
- University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Consuelo Torrini
- Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Edwin Nieblas-Bedolla
- Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Elizabeth J Summers
- Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Emily Sullivan
- Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Britney S Zhang
- Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Emily Batchelor
- Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Braxton Marion
- Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Erika Yamazawa
- Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Samuel C Markson
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Hiroaki Wakimoto
- Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Naema Nayyar
- Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Priscilla K Brastianos
- Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Division of Neuro-Oncology, Department of Neurology, Massachusetts General Hospital. Boston, Massachusetts, USA
| |
Collapse
|
47
|
Kikuchi Y, Shimada H, Yamasaki F, Yamashita T, Araki K, Horimoto K, Yajima S, Yashiro M, Yokoi K, Cho H, Ehira T, Nakahara K, Yasuda H, Isobe K, Hayashida T, Hatakeyama S, Akakura K, Aoki D, Nomura H, Tada Y, Yoshimatsu Y, Miyachi H, Takebayashi C, Hanamura I, Takahashi H. Clinical practice guidelines for molecular tumor marker, 2nd edition review part 2. Int J Clin Oncol 2024; 29:512-534. [PMID: 38493447 DOI: 10.1007/s10147-024-02497-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 02/21/2024] [Indexed: 03/19/2024]
Abstract
In recent years, rapid advancement in gene/protein analysis technology has resulted in target molecule identification that may be useful in cancer treatment. Therefore, "Clinical Practice Guidelines for Molecular Tumor Marker, Second Edition" was published in Japan in September 2021. These guidelines were established to align the clinical usefulness of external diagnostic products with the evaluation criteria of the Pharmaceuticals and Medical Devices Agency. The guidelines were scoped for each tumor, and a clinical questionnaire was developed based on a serious clinical problem. This guideline was based on a careful review of the evidence obtained through a literature search, and recommendations were identified following the recommended grades of the Medical Information Network Distribution Services (Minds). Therefore, this guideline can be a tool for cancer treatment in clinical practice. We have already reported the review portion of "Clinical Practice Guidelines for Molecular Tumor Marker, Second Edition" as Part 1. Here, we present the English version of each part of the Clinical Practice Guidelines for Molecular Tumor Marker, Second Edition.
Collapse
Affiliation(s)
| | - Hideaki Shimada
- Department of Clinical Oncology, Toho University, Tokyo, Japan.
- Department of Surgery, Toho University, Tokyo, Japan.
| | - Fumiyuki Yamasaki
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Taku Yamashita
- Department of Otorhinolaryngology-Head and Neck Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| | - Koji Araki
- Department of Otorhinolaryngology-Head and Neck Surgery, National Defense Medical College, Saitama, Japan
| | - Kohei Horimoto
- Department of Dermatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | | | - Masakazu Yashiro
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Keigo Yokoi
- Department of Lower Gastrointestinal Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| | - Haruhiko Cho
- Department of Surgery, Tokyo Metropolitan Komagome Hospital, Tokyo, Japan
| | - Takuya Ehira
- Department of Gastroenterology, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Kazunari Nakahara
- Department of Gastroenterology, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Hiroshi Yasuda
- Department of Gastroenterology, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Kazutoshi Isobe
- Division of Respiratory Medicine, Department of Internal Medicine (Omori), Toho University, Tokyo, Japan
| | - Tetsu Hayashida
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Shingo Hatakeyama
- Department of Urology, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | | | - Daisuke Aoki
- International University of Health and Welfare Graduate School, Tokyo, Japan
| | - Hiroyuki Nomura
- Department of Obstetrics and Gynecology, School of Medicine, Fujita Health University, Aichi, Japan
| | - Yuji Tada
- Department of Pulmonology, School of Medicine, International University of Health and Welfare, Chiba, Japan
| | - Yuki Yoshimatsu
- Department of Patient-Derived Cancer Model, Tochigi Cancer Center Research Institute, Tochigi, Japan
| | - Hayato Miyachi
- Faculty of Clinical Laboratory Sciences, Nitobe Bunka College, Tokyo, Japan
| | - Chiaki Takebayashi
- Division of Hematology and Oncology, Department of Internal Medicine (Omori), Toho University, Tokyo, Japan
| | - Ichiro Hanamura
- Division of Hematology, Department of Internal Medicine, Aichi Medical University, Aichi, Japan
| | | |
Collapse
|
48
|
Gowda S, Sandow L, Heinrich MC. Treatment of BRAF V600E mutant gastrointestinal stromal tumor with dabrafenib: a case report. J Gastrointest Oncol 2024; 15:788-793. [PMID: 38756640 PMCID: PMC11094487 DOI: 10.21037/jgo-23-767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 03/22/2024] [Indexed: 05/18/2024] Open
Abstract
Background Gastrointestinal stromal tumor (GIST) is a rare mesenchymal tumor arising in the gut, most commonly stomach or small bowel. The most common driver mutations are KIT and PDGFRA which can be treated with imatinib or avapritinib (for PDGFRA D842V-mutant GIST), respectively. BRAF V600E mutant GISTs are rare and these do not respond to imatinib. Multiple clinical trials have shown antitumor effects with dabrafenib in BRAF-mutant melanoma and a few case reports have demonstrated treatment of BRAF V600E mutant GIST with a BRAF kinase inhibitor. Case Description We present a case of a 67-year-old woman diagnosed with high-risk GIST following initial resection. She was initially treated with adjuvant imatinib which was discontinued after 7 months because molecular analysis of her tumor showed the absence of KIT and PDGFRA mutations and a BRAF V600E mutation. When her disease progressed, she was started on sunitinib and subsequently regorafenib. Both agents were discontinued due to severe palmar-plantar erythrodysesthesia and clinical progression. She was subsequently started on dabrafenib based on the presence of a BRAF V600E mutation; this therapy led to a partial response. Her disease remained stable on this medication for 19 months before progression and addition of trametinib to her treatment. Her disease continued to progress and she was switched to everolimus with mixed response before re-challenging with dabrafenib and trametinib. Her imaging showed a mixed response to the re-challenge before progressing after 5 months and transitioning to hospice. Conclusions We describe an uncommon molecular subtype of GIST with a BRAF V600E mutation. As expected, her disease was resistant to standard GIST therapy, however there was notable tumor regression following treatment with dabrafenib. This case shows the importance of molecular testing in GIST and adds to the current body of literature on the treatment of BRAF-mutant GIST.
Collapse
Affiliation(s)
- Sonia Gowda
- Oregon Health and Science University Knight Cancer Institute, Portland, OR, USA
| | - Lyndsey Sandow
- Oregon Health and Science University Knight Cancer Institute, Portland, OR, USA
| | - Michael C. Heinrich
- Oregon Health and Science University Knight Cancer Institute, Portland, OR, USA
- Portland VA Health Care System and Oregon Health and Science University Knight Cancer Institute, Portland, OR, USA
| |
Collapse
|
49
|
Pompili SVB, Fanzini S, Schachner M, Chen S. In Vitro and In Vivo Studies of Melanoma Cell Migration by Antagonistic Mimetics of Adhesion Molecule L1CAM. Int J Mol Sci 2024; 25:4811. [PMID: 38732030 PMCID: PMC11084881 DOI: 10.3390/ijms25094811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/08/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Melanoma, the deadliest type of skin cancer, has a high propensity to metastasize to other organs, including the brain, lymph nodes, lungs, and bones. While progress has been made in managing melanoma with targeted and immune therapies, many patients do not benefit from these current treatment modalities. Tumor cell migration is the initial step for invasion and metastasis. A better understanding of the molecular mechanisms underlying metastasis is crucial for developing therapeutic strategies for metastatic diseases, including melanoma. The cell adhesion molecule L1CAM (CD171, in short L1) is upregulated in many human cancers, enhancing tumor cell migration. Earlier studies showed that the small-molecule antagonistic mimetics of L1 suppress glioblastoma cell migration in vitro. This study aims to evaluate if L1 mimetic antagonists can inhibit melanoma cell migration in vitro and in vivo. We showed that two antagonistic mimetics of L1, anagrelide and 2-hydroxy-5-fluoropyrimidine (2H5F), reduced melanoma cell migration in vitro. In in vivo allograft studies, only 2H5F-treated female mice showed a decrease in tumor volume.
Collapse
Affiliation(s)
- Stefano Vito Boccadamo Pompili
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University, 00185 Rome, Italy
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA;
| | - Sophia Fanzini
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA;
| | - Melitta Schachner
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA;
| | - Suzie Chen
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA;
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
- VA New Jersey Health System, East Orange, NJ 07018, USA
| |
Collapse
|
50
|
Bergamaschi D. Improving cancer stem cells killing in malignant melanoma. Br J Dermatol 2024; 190:615-616. [PMID: 38282338 DOI: 10.1093/bjd/ljae035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 01/30/2024]
Affiliation(s)
- Daniele Bergamaschi
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Faculty of Medicine & Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|