1
|
Wang C, Niu X, Xia T, Wang P, Wang Y, Zhang Z, Zhang J, Ju S, Xiao Z. Predicting c-KIT Inhibitor Efficacy in Patient-Derived Models of Sinonasal Mucosal Melanomas through Integrated Histogram Analysis of Whole-Tumor DKI, IVIM, and DCE-MRI. Clin Cancer Res 2025; 31:1686-1699. [PMID: 39937224 DOI: 10.1158/1078-0432.ccr-24-3765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/21/2025] [Accepted: 02/10/2025] [Indexed: 02/13/2025]
Abstract
PURPOSE To evaluate whole-tumor histogram analysis of diffusion kurtosis imaging (DKI), intravoxel incoherent motion (IVIM), and dynamic contrast-enhanced MRI (DCE-MRI) in predicting the efficacy of imatinib, a c-KIT inhibitor, for treating patient-derived models derived from sinonasal mucosal melanomas (MM). EXPERIMENTAL DESIGN This study included 38 patients with histologically confirmed sinonasal MM, who underwent DKI, IVIM, and DCE-MRI. Patient-derived tumor xenograft models and precision-cut tumor slices were established to evaluate tumor response to imatinib. Whole-tumor histogram analysis was conducted on imaging parameters, and logistic regression models were applied to determine the predictive value of these metrics in differentiating responders from nonresponders. RESULTS Among the 38 patients with sinonasal MM, 12 were classified as responders and 26 as nonresponders based on patient-derived tumor xenograft and precision-cut tumor slice model responses to imatinib. The DKI model revealed significant differences in mean, median, 10th percentile, and 90th percentile values of Dk and K between responders and nonresponders (P < 0.05). The IVIM model indicated significant differences in 10th percentile and mean values of D, with kurtosis f being a strong predictor. The DCE-MRI model, using the 90th percentile Ktrans metric, demonstrated robust predictive performance, achieving an AUC of 0.89, with 80.77% specificity and 91.67% sensitivity. The combined logistic model integrating DKI, IVIM, and DCE-MRI metrics produced the highest predictive accuracy, with an AUC of 0.90. CONCLUSIONS Whole-tumor histogram analysis of DKI, IVIM, and DCE-MRI offers a noninvasive method for predicting the efficacy of c-KIT inhibitors in sinonasal MMs, presenting valuable implications for guiding targeted treatment in this rare cancer type.
Collapse
Affiliation(s)
- Cong Wang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
- Department of Nuclear Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Xuewei Niu
- Department of Nuclear Medicine, Hebei Medical University, Shijiazhuang, China
| | - Tianyi Xia
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Peng Wang
- Department of Radiology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yuzhe Wang
- Department of Radiology, Zhongshan Hospital of Fudan University, Fudan University, Shanghai, China
| | | | - Jianyuan Zhang
- Department of Nuclear Medicine, Baoding No. 1 Central Hospital, China
| | - Shenghong Ju
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Zebin Xiao
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
2
|
Blacklock KLB, Donnelly K, Lu Y, del Pozo J, Glendinning L, Polton G, Selmic L, Tanis J, Killick D, Parys M, Morris JS, Breathnach I, Zago S, Gould SM, Shaw DJ, Tivers MS, Malucelli D, Marques A, Purzycka K, Cantatore M, Mathers ME, Stares M, Meynert A, Patton EE. Oronasal mucosal melanoma is defined by two transcriptional subtypes in humans and dogs with implications for diagnosis and therapy. J Pathol 2025; 265:245-259. [PMID: 39828982 PMCID: PMC11794980 DOI: 10.1002/path.6377] [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: 07/30/2024] [Accepted: 10/31/2024] [Indexed: 01/22/2025]
Abstract
Mucosal melanoma is a rare melanoma subtype associated with a poor prognosis and limited existing therapeutic interventions, in part due to a lack of actionable targets and translational animal models for preclinical trials. Comprehensive data on this tumour type are scarce, and existing data often overlooks the importance of the anatomical site of origin. We evaluated human and canine oronasal mucosal melanoma (OMM) to determine whether the common canine disease could inform the rare human equivalent. Using a human and canine primary OMM cohort of treatment-naive archival tissue, alongside clinicopathological data, we obtained transcriptomic, immunohistochemical, and microbiome data from both species. We defined the transcriptomic landscape in both species and linked our findings to immunohistochemical, microbiome, and clinical data. Human and dog OMM stratified into two distinctive transcriptional groups, which we defined using a species-independent 41-gene signature. These two subgroups are termed CTLA4-high and MET-high and indicate actionable targets for OMM patients. To guide clinical decision-making, we developed immunohistochemical diagnostic tools that distinguish between transcriptomic subgroups. We found that OMM had conserved transcriptomic subtypes and biological similarity between human and canine OMM, with significant implications for patient classification, treatment, and clinical trial design. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Kelly L Bowlt Blacklock
- Royal (Dick) School of Veterinary Studies and the Roslin InstituteEdinburghUK
- MRC Human Genetics Unit, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
- Edinburgh Cancer Research, CRUK Scotland Centre, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
| | - Kevin Donnelly
- MRC Human Genetics Unit, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
| | - Yuting Lu
- MRC Human Genetics Unit, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
- Edinburgh Cancer Research, CRUK Scotland Centre, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
| | - Jorge del Pozo
- Royal (Dick) School of Veterinary Studies and the Roslin InstituteEdinburghUK
| | - Laura Glendinning
- Royal (Dick) School of Veterinary Studies and the Roslin InstituteEdinburghUK
| | | | - Laura Selmic
- Department of Veterinary Clinical SciencesThe Ohio State UniversityColumbusOHUSA
| | - Jean‐Benoit Tanis
- Department of Small Animal Clinical Sciences, Institute of Infection, Veterinary and Ecological ScienceUniversity of LiverpoolNestonUK
| | - David Killick
- Department of Small Animal Clinical Sciences, Institute of Infection, Veterinary and Ecological ScienceUniversity of LiverpoolNestonUK
| | - Maciej Parys
- Royal (Dick) School of Veterinary Studies and the Roslin InstituteEdinburghUK
| | | | | | | | | | - Darren J Shaw
- Royal (Dick) School of Veterinary Studies and the Roslin InstituteEdinburghUK
| | - Michael S Tivers
- Paragon Veterinary Referrals, Paragon Point, Red Hall CrescentWakefieldUK
| | - Davide Malucelli
- Paragon Veterinary Referrals, Paragon Point, Red Hall CrescentWakefieldUK
| | | | - Katarzyna Purzycka
- Anderson Moores Veterinary Specialists, The Granary, Bunstead BarnsHampshireUK
| | - Matteo Cantatore
- Anderson Moores Veterinary Specialists, The Granary, Bunstead BarnsHampshireUK
| | | | - Mark Stares
- Edinburgh Cancer Research, CRUK Scotland Centre, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
- Edinburgh Cancer Centre, Western General Hospital, Crewe RoadEdinburghUK
| | - Alison Meynert
- MRC Human Genetics Unit, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
| | - E Elizabeth Patton
- MRC Human Genetics Unit, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
- Edinburgh Cancer Research, CRUK Scotland Centre, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
| |
Collapse
|
3
|
Xu J, Jin M, Mu Z, Li Z, Qi R, Han X, Jiang H. Inhibiting melanoma tumor growth: the role of oxidative stress-associated LINC02132 and COPDA1 long non-coding RNAs. Front Immunol 2025; 16:1558292. [PMID: 40092985 PMCID: PMC11906686 DOI: 10.3389/fimmu.2025.1558292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 02/11/2025] [Indexed: 03/19/2025] Open
Abstract
Background Cutaneous melanoma is a type of malignant tumor that is challenging to predict and is readily stimulated by various factors. Oxidative stress can induce damage and alterations in melanocytes, subsequently triggering immune responses. Given that oxidative stress is a prevalent tumor stimulus, we aimed to enhance melanoma prediction by identifying lncRNA signatures associated with oxidative stress. Methods We screened for oxidative stress-related lncRNAs that could improve melanoma patient prognosis using the TCGA and GTEx databases. Utilizing differentially expressed oxidative stress-related lncRNAs (DE-OSlncRNAs), we constructed a Lasso regression model. The accuracy of the model was validated using univariate and multivariate regression, Kaplan-Meier (K-M) curves, and ROC curves. Subsequently, we conducted immune infiltration analysis, immune checkpoint differential analysis, IC50 pharmaceutical analysis, and gene set enrichment analysis. Investigating the effects of the target gene on melanoma using fluorescence in situ hybridization (FISH), quantitative real-time PCR (qRT-PCR), Edu assay, wound healing assay, transwell assay, flow cytometry, and reactive oxygen species (ROS) detection. Results Thirteen lncRNAs were identified as significant prognostic factors. Four oxidative stress-related lncRNAs (COPDA1, LINC02132, LINC02812, and MIR205HG) were further validated by fluorescence in situ hybridization (FISH), with results consistent with our data analysis. LINC02132 and COPDA1 can influence the proliferation, invasion, migration, and apoptosis of melanoma. Conclusion Our findings suggest that upregulation of the LINC02132 or COPDA1 genes elevates intracellular reactive oxygen species (ROS) levels in melanoma cells, suppresses tumor cell proliferation, migration, and invasion, and promotes apoptosis. These results suggest a novel therapeutic strategy for melanoma treatment.
Collapse
Affiliation(s)
- JingWen Xu
- Department of Dermatology, Shengjing Hospital of China Medical University, Shenyang, China
| | - MingZhu Jin
- Department of Dermatology, Shengjing Hospital of China Medical University, Shenyang, China
| | - ZhenZhen Mu
- Department of Dermatology, Shengjing Hospital of China Medical University, Shenyang, China
| | - ZhengXiu Li
- Department of Dermatology & Key Lab of Dermatology, Ministry of Education and Public Health, National Joint Engineering Research Center for Theranostics of Immunology Skin Diseases, The First Hospital of China Medical University, Shenyang, China
| | - RuiQun Qi
- Department of Dermatology & Key Lab of Dermatology, Ministry of Education and Public Health, National Joint Engineering Research Center for Theranostics of Immunology Skin Diseases, The First Hospital of China Medical University, Shenyang, China
| | - XiuPing Han
- Department of Dermatology, Shengjing Hospital of China Medical University, Shenyang, China
| | - HangHang Jiang
- Department of Dermatology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
4
|
Kim SH, Tsao H. Acral Melanoma: A Review of Its Pathogenesis, Progression, and Management. Biomolecules 2025; 15:120. [PMID: 39858514 PMCID: PMC11763010 DOI: 10.3390/biom15010120] [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: 12/04/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Acral melanoma is a distinct subtype of cutaneous malignant melanoma that uniquely occurs on ultraviolet (UV)-shielded, glabrous skin of the palms, soles, and nail beds. While acral melanoma only accounts for 2-3% of all melanomas, it represents the most common subtype among darker-skinned, non-Caucasian individuals. Unlike other cutaneous melanomas, acral melanoma does not arise from UV radiation exposure and is accordingly associated with a relatively low tumor mutational burden. Recent advances in genomic, transcriptomic, and epigenomic sequencing have revealed genetic alterations unique to acral melanoma, including novel driver genes, high copy number variations, and complex chromosomal rearrangements. This review synthesizes the current knowledge on the clinical features, epidemiology, and treatment approaches for acral melanoma, with a focus on the genetic pathogenesis that gives rise to its unique tumor landscape. These findings highlight a need to deepen our genetic and molecular understanding to better target this challenging subtype of melanoma.
Collapse
Affiliation(s)
| | - Hensin Tsao
- Department of Dermatology, Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
5
|
Stoff R, Markovic SN, McWilliams RR, Kottschade LA, Montane HN, Dimou A, Dudek AZ, Tan W, Dronca RS, Seetharam M, Chen R, Block MS. Real-world evidence on efficacy and toxicity of targeted therapy in older melanoma patients treated in a tertiary-hospital setting. Melanoma Res 2024; 34:510-518. [PMID: 39207855 PMCID: PMC11524625 DOI: 10.1097/cmr.0000000000000997] [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: 05/15/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024]
Abstract
Melanoma is the deadliest form of skin cancer. The median age at diagnosis is 66. While most patients are treated with immunotherapy, the use of targeted therapy is a valid alternative for patients whose tumors harbor a BRAF or c-KIT driver mutation. These agents, while effective, come with a variety of side effects which limit their use, especially in older patients. We sought to assess the efficacy and toxicity of these agents in older melanoma patients. Melanoma patients over 65 treated with BRAF/MEK or c-KIT inhibitors were retrospectively identified, and their data were analyzed for treatment efficacy and toxicity. All data were compared using the Chi-square test for categorical comparisons and the Kruskal-Wallis method for median comparisons. One hundred and sixteen patients were identified. One hundred and six patients were treated with BRAF/MEK inhibitors. The assessed response rate (RR) was 83% and was comparable across different subgroups, including advanced line patients and those with a more aggressive disease. The median progression free survival (PFS) was 7.9 months, and the median overall survival (OS) was 15.7 months. Twenty-seven percent experienced grade 3-4 toxicity leading to a 24% treatment discontinuation rate. Another 10 patients were treated with the c-KIT inhibitor imatinib, for whom the assessed RR was 55%. The median PFS was 4.3 months, and the median OS was 22.6 months. Forty percent needed dose reductions, yet none had to stop treatment due to adverse effects. The use of targeted therapy in older patients is effective yet challenging due to toxicity. Deploying mitigation strategies can help maximizing their usefulness.
Collapse
Affiliation(s)
- Ronen Stoff
- Department of Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota
| | - Svetomir N. Markovic
- Department of Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota
| | - Robert R. McWilliams
- Department of Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota
| | - Lisa A. Kottschade
- Department of Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota
| | - Heather N. Montane
- Department of Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota
| | - Anastasios Dimou
- Department of Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota
| | - Arkadiusz Z. Dudek
- Department of Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota
| | - Winston Tan
- Department of Hematology and Oncology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida
| | - Roxana S. Dronca
- Department of Hematology and Oncology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida
| | - Mahesh Seetharam
- Department of Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Scottsdale, Arizona, USA
| | - Ruqin Chen
- Department of Hematology and Oncology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida
| | - Matthew S. Block
- Department of Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota
| |
Collapse
|
6
|
De Sanctis JB, Garmendia JV, Duchová H, Valentini V, Puskasu A, Kubíčková A, Hajdúch M. Lck Function and Modulation: Immune Cytotoxic Response and Tumor Treatment More Than a Simple Event. Cancers (Basel) 2024; 16:2630. [PMID: 39123358 PMCID: PMC11311849 DOI: 10.3390/cancers16152630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Lck, a member of the Src kinase family, is a non-receptor tyrosine kinase involved in immune cell activation, antigen recognition, tumor growth, and cytotoxic response. The enzyme has usually been linked to T lymphocyte activation upon antigen recognition. Lck activation is central to CD4, CD8, and NK activation. However, recently, it has become clearer that activating the enzyme in CD8 cells can be independent of antigen presentation and enhance the cytotoxic response. The role of Lck in NK cytotoxic function has been controversial in a similar fashion as the role of the enzyme in CAR T cells. Inhibiting tyrosine kinases has been a highly successful approach to treating hematologic malignancies. The inhibitors may be useful in treating other tumor types, and they may be useful to prevent cell exhaustion. New, more selective inhibitors have been documented, and they have shown interesting activities not only in tumor growth but in the treatment of autoimmune diseases, asthma, and graft vs. host disease. Drug repurposing and bioinformatics can aid in solving several unsolved issues about the role of Lck in cancer. In summary, the role of Lck in immune response and tumor growth is not a simple event and requires more research.
Collapse
Affiliation(s)
- Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (J.V.G.); (V.V.); (A.K.); (M.H.)
- Czech Advanced Technologies and Research Institute (CATRIN), 77900 Olomouc, Czech Republic
| | - Jenny Valentina Garmendia
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (J.V.G.); (V.V.); (A.K.); (M.H.)
| | - Hana Duchová
- Faculty of Science, Palacky University, 77900 Olomouc, Czech Republic; (H.D.); (A.P.)
| | - Viktor Valentini
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (J.V.G.); (V.V.); (A.K.); (M.H.)
| | - Alex Puskasu
- Faculty of Science, Palacky University, 77900 Olomouc, Czech Republic; (H.D.); (A.P.)
| | - Agáta Kubíčková
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (J.V.G.); (V.V.); (A.K.); (M.H.)
- Czech Advanced Technologies and Research Institute (CATRIN), 77900 Olomouc, Czech Republic
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (J.V.G.); (V.V.); (A.K.); (M.H.)
- Czech Advanced Technologies and Research Institute (CATRIN), 77900 Olomouc, Czech Republic
- Laboratory of Experimental Medicine, University Hospital Olomouc, 77900 Olomouc, Czech Republic
| |
Collapse
|
7
|
Chan PY, Corrie PG. Curing Stage IV Melanoma: Where Have We Been and Where Are We? Am Soc Clin Oncol Educ Book 2024; 44:e438654. [PMID: 38669609 DOI: 10.1200/edbk_438654] [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: 04/28/2024]
Abstract
Little more than 10 years ago, metastatic melanoma was considered to have one of the poorest cancer outcomes, associated with a median overall survival of 6-8 months. Cytotoxic chemotherapy offered modest response rates of 20%-30%, but no clear survival benefit. Patients were routinely enrolled in clinical trials as their first-line therapy in the search for effective novel therapeutics. Remarkable developments in molecular biology, cancer genomics, immunology, and drug discovery have dominated the early part of the 21st century, and nowhere have the benefits been better realized than in the transformation of outcomes for patients with metastatic melanoma: since 2011, 14 new agents have been approved that significantly increase survival, with long-term remissions and, possibly now, potential for cure. Even so, there is still much work to be done, given that most treated patients still die of their disease. Although most survival gains have so far been realized for cutaneous melanoma, improving treatment options for those 10% of patients with rarer, noncutaneous melanomas is a high priority. Key novel therapeutic approaches aimed at improving outcomes with potential for curing patients with melanoma are considered.
Collapse
Affiliation(s)
- Pui Ying Chan
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Pippa G Corrie
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| |
Collapse
|