1
|
Pluetrattanabha N, Direksunthorn T, Ahmad I, Jyothi SR, Shit D, Singh AK, Chauhan AS. Inflammasome activation in melanoma progression: the latest update concerning pathological role and therapeutic value. Arch Dermatol Res 2025; 317:258. [PMID: 39820618 DOI: 10.1007/s00403-025-03802-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 12/11/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025]
Abstract
The progression of melanoma is a complex process influenced by both internal and external cues which encourage the transition of tumour cells, uncontrolled growth, migration, and metastasis. Additionally, inflammation allows tumours to evade the immune system, contributing to cancer development. The inflammasome, a complex of many proteins, is crucial in enhancing immune responses to external and internal triggers. As a critical inflammatory mechanism, it contributes to the development of melanoma. These mechanisms may be triggered via various internal and external stimuli, causing the induction of specific enzymes such as caspase-1, caspase-11, or caspase-8. This, in turn, leads to the release of interleukin (IL)-1β and IL-18 and cell death by apoptosis and pyroptosis. Proper inflammasome stimulation is crucial for the host to deal with invading pathogens or tissue injury. However, inappropriate inflammasome stimulation can result in unregulated tissue reactions, thus easing many diseases, including melanoma. Hence, keeping a delicate equilibrium between the stimulation and prohibition of inflammasomes is crucial, necessitating meticulous control of the assembly and functional aspects of inflammasomes. This review examines the latest advancements in inflammasome studies, specifically focusing on the molecular processes that control inflammasome formation, signalling, and modulation in melanoma.
Collapse
Affiliation(s)
| | | | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
- Health and Medical Research Center, King Khalid University, P.O. Box 960, AlQura'a, Abha, Saudi Arabia
| | - S Renuka Jyothi
- Department of Biotechnology and Genetics, JAIN (Deemed to be University) School of Sciences, Bangalore, Karnataka, India
| | - Debasish Shit
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
| | | | - Ashish Singh Chauhan
- Uttaranchal Institute of Pharmaceutical Sciences, Division of research and innovation, Uttaranchal University, Dehradun, Uttarakhand, India
| |
Collapse
|
2
|
Xu Y, Ding L, Wu M, Wang X, Wang L, Xu Z, Xia Y, Cao Z, Zhang Y, Song R, Deng B, Chen G. Lipid metabolic remodeling delays senescence of T cells to potentiate their immunity against solid tumors. J Immunother Cancer 2025; 13:e010403. [PMID: 39762081 PMCID: PMC11749770 DOI: 10.1136/jitc-2024-010403] [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: 08/28/2024] [Accepted: 12/14/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Tumor cells can drive the senescence of effector T cells by unbalancing their lipid metabolism, thereby limiting adoptive T cell therapy and contributing to tumor immune evasion. Our objective is to provide a feasible strategy for enhancing T cell treatment efficacy against solid tumors. METHODS In this study, liposomal arachidonyl trifluoromethyl ketone (ATK) was anchored onto the adoptive T cell surface via bioorthogonal reactions, aiming to specifically inhibit the group IVA cytosolic phospholipase A2α (cPLA2α), a key enzyme facilitating phospholipid metabolism and senescent state of T cells. RESULTS The surface engineering exerted rare side effects on the activation and migration of T cells, but local and sustained extravasation of ATK downregulated cPLA2α expression, reprogrammed lipid metabolism, and inhibited lipid droplet accumulation. This endows T cells with delayed senescence and declined apoptosis to maintain their tumor-killing potency. Systemic administration of surface-engineered T cells resulted in superior infiltration in solid tumors and improved antitumor efficacy by enhancing the secretion of cytotoxic molecules, thereby prolonging the survival of mice bearing colorectal carcinoma and melanoma xenografts. CONCLUSIONS Lipid-metabolically remodeled T cells with delayed senescence increase efficacy in tumor microenvironment, highlighting a novel strategy for solid tumor immunotherapy.
Collapse
Affiliation(s)
- Yemin Xu
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, People's Republic of China
- Department of Gastroenterology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, People's Republic of China
- Qingdao Key Laboratory of Materials for Tissue Repair and Rehabilitation, School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, Shandong, People's Republic of China
- Shandong Provincial Key Medical and Health Laboratory of Neuro-oncology of Innovative Integrated Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Yangzhou, Jiangsu, People's Republic of China
| | - Li Ding
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, Jiangsu, People's Republic of China
| | - Mengyue Wu
- Department of Anesthesiology, The Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, People's Republic of China
| | - Xiya Wang
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, People's Republic of China
| | - Lu Wang
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, People's Republic of China
- Department of Gastroenterology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, People's Republic of China
| | - Zhou Xu
- Qingdao Key Laboratory of Materials for Tissue Repair and Rehabilitation, School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, Shandong, People's Republic of China
- Shandong Provincial Key Medical and Health Laboratory of Neuro-oncology of Innovative Integrated Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Yangzhou, Jiangsu, People's Republic of China
| | - Yinhe Xia
- Qingdao Key Laboratory of Materials for Tissue Repair and Rehabilitation, School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, Shandong, People's Republic of China
- Shandong Provincial Key Medical and Health Laboratory of Neuro-oncology of Innovative Integrated Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Yangzhou, Jiangsu, People's Republic of China
| | - Zhennan Cao
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, People's Republic of China
| | - Yanqing Zhang
- Medical College, Yangzhou University, Yangzhou, Jiangsu, People's Republic of China
| | - Ruilong Song
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, People's Republic of China
| | - Bin Deng
- Department of Gastroenterology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, People's Republic of China
| | - Gang Chen
- Qingdao Key Laboratory of Materials for Tissue Repair and Rehabilitation, School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, Shandong, People's Republic of China
- Shandong Provincial Key Medical and Health Laboratory of Neuro-oncology of Innovative Integrated Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Yangzhou, Jiangsu, People's Republic of China
| |
Collapse
|
3
|
Liu H, Gou X, Tan Y, Fan Q, Chen J. Immunotherapy and delivery systems for melanoma. Hum Vaccin Immunother 2024; 20:2394252. [PMID: 39286868 PMCID: PMC11409522 DOI: 10.1080/21645515.2024.2394252] [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: 05/10/2024] [Revised: 08/01/2024] [Accepted: 08/15/2024] [Indexed: 09/19/2024] Open
Abstract
Melanoma is a highly malignant tumor of melanocyte origin that is prone to early metastasis and has a very poor prognosis. Early melanoma treatment modalities are mainly surgical, and treatment strategies for advanced or metastatic melanoma contain chemotherapy, radiotherapy, targeted therapy and immunotherapy. The efficacy of chemotherapy and radiotherapy has been unsatisfactory due to low sensitivity and strong toxic side effects. And targeted therapy is prone to drug resistance, so its clinical application is limited. Melanoma has always been the leader of immunotherapy for solid tumors, and how to maximize the role of immunotherapy and how to implement immunotherapy more accurately are still urgent to be explored. This review summarizes the common immunotherapies and applications for melanoma, illustrates the current research status of melanoma immunotherapy delivery systems, and discusses the advantages and disadvantages of each delivery system and its prospects for clinical application.
Collapse
Affiliation(s)
- Hui Liu
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xi Gou
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yuanfang Tan
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Qiuying Fan
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Juanjuan Chen
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
4
|
Liu C, Liu X, Hu L, Li X, Xin H, Zhu S. Global, regional, and national burden of cutaneous malignant melanoma from 1990 to 2021 and prediction to 2045. Front Oncol 2024; 14:1512942. [PMID: 39777336 PMCID: PMC11703817 DOI: 10.3389/fonc.2024.1512942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Background Cutaneous Malignant Melanoma (CMM) is a significant global health challenge. Understanding regional differences in CMM prevalence and trends is crucial for developing targeted strategies. To address this, we analyzed epidemiological patterns and investigated risk factors for CMM-related mortality. Methods This study analyzed CMM using data from the 2021 Global Burden of Diseases survey, covering 204 countries and territories. We evaluated the number and age-standardized rates of prevalence (ASPR), mortality (ASMR), disability-adjusted life years (ASDR), and annual percentage changes (EAPCs). Trends were stratified by region, country, age, sex, and Sociodemographic Index (SDI). A Bayesian Age-Period-Cohort model projected future prevalence, mortality, and DALYs, while decomposition analysis identified key drivers of CMM burden. Frontier analysis further associated CMM outcomes with socio-demographic development. Results In 2021, the global prevalence of CMM reached 833,215 cases, a 161.3% increase since 1990. During this period, the ASPR rose from 19.13 to 25.37 per 100,000, while the ASMR declined from 0.84 to 0.73 per 100,000. DALYs increased by 60.5%, from 1,045,777 to 1,678,836. The high SDI region had the highest ASPR, ASMR, and ASDR. Decomposition analysis identified population growth, demographic aging, and epidemiological changes as equal drivers of CMM DALYs globally. Countries like New Zealand and Australia demonstrated the most significant effective differences, indicating potential for improvement in CMM management. By 2045, the global ASPR is projected to rise to 36.61, with ASMR and ASDR expected to decrease to 0.79 and 10.21 per 100,000. Conclusion CMM poses an increasing global health concern, with ASPR steadily rising. While this analysis shows a decline in global ASMR and ASDR overall, these rates are actually increasing in low SDI regions, and projections indicate that this trend will likely continue until 2045.
Collapse
Affiliation(s)
- Chengling Liu
- Center of Burns and Plastic Surgery and Dermatology, The 924th Hospital of Joint Logistics Support Force of the People’s Liberation Army (PLA), Guilin, China
| | - Xingchen Liu
- Department of Pathology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Li Hu
- Department of the Hematology and Oncology, The 924th Hospital of Joint Logistics Support Force of the Chinese People’s Liberation Army (PLA), Guilin, China
| | - Xin Li
- Center of Burns and Plastic Surgery and Dermatology, The 924th Hospital of Joint Logistics Support Force of the People’s Liberation Army (PLA), Guilin, China
| | - Haiming Xin
- Center of Burns and Plastic Surgery and Dermatology, The 924th Hospital of Joint Logistics Support Force of the People’s Liberation Army (PLA), Guilin, China
| | - Sailin Zhu
- Center of Burns and Plastic Surgery and Dermatology, The 924th Hospital of Joint Logistics Support Force of the People’s Liberation Army (PLA), Guilin, China
| |
Collapse
|
5
|
Chen Y, Zhang L, Shi H, Shen Z, Huang D, Tang S, He Y, Wang G, Pan H, Wang Z. Thermosensitive hydrogel delivery of BCG lysates and tumor antigens: A novel strategy for melanoma immunoprevention and therapeutics. Biochem Biophys Res Commun 2024; 745:151215. [PMID: 39732120 DOI: 10.1016/j.bbrc.2024.151215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 12/18/2024] [Indexed: 12/30/2024]
Abstract
Melanoma, recognized as one of the most aggressive forms of skin cancer, continues to show a steady rise in global incidence. While Bacillus Calmette-Guérin (BCG) has been identified as a potential intralesional therapy for melanoma, its therapeutic efficacy remains suboptimal. This study introduces a novel thermosensitive hydrogel formulated with BCG lysates and either OVA peptide or tumor cell lysates (PPP-BCG-OVA/TL). The hydrogel demonstrated the ability to elicit a robust systemic tumor antigen-specific T cell response and significantly enhanced the infiltration of CD45+ leukocytes, macrophages, CD8+ T cells, and natural killer (NK) cells into the tumor microenvironment. As a result, vaccination with PPP-BCG-OVA/TL hydrogel effectively inhibited tumor growth and extended survival in mouse models of subcutaneous melanoma in both preventive and therapeutic contexts. These findings highlight the potential to markedly improve the therapeutic efficacy of BCG lysates through the addition of tumor antigens and the use of a hydrogel-based delivery platform.
Collapse
Affiliation(s)
- Yanwei Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Huashan Shi
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhixue Shen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Dan Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shuo Tang
- Chengdu Institute of Biological Products Company Limited, Chengdu, 610023, China
| | - Yueyue He
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Guilan Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hailong Pan
- China National Biotec Group Company Limited, Beijing, 100000, China
| | - Zhenling Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
6
|
Shi Y, Zhao W, Ding Y, Ge X, Ju M. Research on the influence of radiotherapy-related genes on immune infiltration, immunotherapy response and prognosis in melanoma based on multi-omics. Front Immunol 2024; 15:1467098. [PMID: 39687627 PMCID: PMC11647020 DOI: 10.3389/fimmu.2024.1467098] [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: 07/19/2024] [Accepted: 11/14/2024] [Indexed: 12/18/2024] Open
Abstract
Background Skin cutaneous melanoma (SKCM) is a significant oncological challenge due to its aggressive nature and poor treatment outcomes. This study explores the comprehensive effects of radiotherapy (RT) in SKCM, focusing on cell signaling pathways, immune infiltration, immune gene correlations, immunotherapy response, and prognosis. Methods Using the Cancer Genome Atlas (TCGA) database, differentially expressed genes (DEGs) in SKCM patients undergoing RT were identified. A risk score model based on these DEGs was developed to assess the effects of RT-related genes on drug sensitivity, immune cell infiltration, immunotherapy response, and prognosis through multi-omics analysis. Human melanoma cells UACC62 and UACC257 were irradiated with 8 Gy gamma ray to establish an in vitro model, verifying the impact of radiotherapy on gene expression. Results The risk score demonstrated significant prognostic value and emerged as an independent prognostic factor. miRNA-mRNA and transcription factor regulatory networks underscored its clinical significance. Four key genes were identified: DUSP1, CXCL13, SLAMF7, and EVI2B. Analysis of single-cell and immunotherapy datasets indicated that these genes enhance immune response and immunotherapy efficacy in melanoma patients. PCR results confirmed that gamma rays increased the expression of these genes in human melanoma cells UACC62 and UACC257. Conclusion Using a multi-omics approach, we analyzed and validated the impact of RT on the immune landscape of melanoma patients. Our findings highlight the critical role of RT-related genes in predicting SKCM prognosis and guiding personalized therapy strategies, particularly in the context of immunotherapy. These contribute to understanding the role of radiotherapy combined with immunotherapy in melanoma.
Collapse
Affiliation(s)
- Yujing Shi
- Department of Oncology, Affiliated Jurong Hospital of Jiangsu University, Zhenjiang, ;China
| | - Wantong Zhao
- Department of Radiation Oncology, Affiliated Zhongshan Hospital of Dalian University, Dalian, ;China
| | - Yuanjian Ding
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Osaka, ;Japan
| | - Xiaolin Ge
- Department of Radiation Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, ;China
| | - Mengyang Ju
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Osaka, ;Japan
| |
Collapse
|
7
|
Liu J, Luo Q, Zhao H, Yang M, Yang J, Wang Y, Zhao M, Mao J, Chen J, Guo B, Zhang L. Comprehensive gene set enrichment and variation analyses identify SUV39H1 as a potential prognostic biomarker for glioblastoma immunorelevance. Comput Struct Biotechnol J 2024; 23:4161-4176. [PMID: 39640533 PMCID: PMC11617780 DOI: 10.1016/j.csbj.2024.11.016] [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: 07/10/2024] [Revised: 10/25/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024] Open
Abstract
Glioblastoma (GBM) is the most common intracranial malignancy. SUV39H1 encodes a histone H3 lysine 9 methyltransferase that acts as an oncogene in several cancers; however, its role in GBM remains unknown. We obtained GBM transcriptome and clinical data from The Cancer Genome Atlas (TCGA) database on the UCSC Xena platform to perform differential and enrichment analyses of genes in the SUV39H1 high- and low-expression groups to construct a prognostic risk model. Analysis of SUV39H1 related biological processes in GBM was performed by gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA). High- and low-risk subgroup mutation signatures were analyzed using maftools. Immune infiltration was evaluated using IOBR and CIBERSORT algorithms. We analyzed the cell types and intercellular communication networks in glioma stem cells (GSCs) using scRNA-seq. The effects on GBM cells and GSCs after inhibition of SUV39H1 were investigated in vitro. SUV39H1 was significantly overexpressed in GBM and associated with poor prognosis. SUV39H1-related differentially expressed genes were enriched in immune and inflammation related pathways, and GSEA revealed that these genes were significantly enriched in signaling pathways such as IL-18, oxidative phosphorylation, and regulation of TP53 activity. Mutational analysis revealed frequent alterations in TP53 and PTEN expression. In addition, the infiltration abundances of the five immune cell types were significantly different between the high- and low-expression groups. Analysis of cellular communication networks by scRNA-seq revealed a strong interaction between CRYAB-GSC and PTPRZ1-GSC in GSCs. In vitro experiments verified that knockdown of SUV39H1 inhibited the viability and proliferation of U87 and U251 glioblastoma cells and downregulated the expression of stemness markers Nestin and SOX2 in CSC1589 and TS576 GSC lines. Increased SUV39H1 expression is associated with immune cell infiltration and poor prognosis in patients with GBM. Inhibition of SUV39H1 restrains GBM growth and reduces the stem cell properties of GSC. Thus, SUV39H1 might be a prognostic predictor and immunotherapeutic target in patients with GBM.
Collapse
Affiliation(s)
- Jixuan Liu
- Key Laboratory of Pathobiology, Ministry of Education, Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Qian Luo
- Key Laboratory of Pathobiology, Ministry of Education, Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Haoran Zhao
- Key Laboratory of Pathobiology, Ministry of Education, Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Mei Yang
- Key Laboratory of Pathobiology, Ministry of Education, Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Jiaying Yang
- Key Laboratory of Pathobiology, Ministry of Education, Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yingtong Wang
- The Undergraduate Center of Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Mengxin Zhao
- Key Laboratory of Pathobiology, Ministry of Education, Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Juanjuan Mao
- Key Laboratory of Pathobiology, Ministry of Education, Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Jiasi Chen
- Key Laboratory of Pathobiology, Ministry of Education, Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Baofeng Guo
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun 130033, China
| | - Ling Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
8
|
Wang X, Li Z, Xu J, Wang J, Li Y, Li Q, Niu J, Yang R. HSPA4 Expression is Correlated with Melanoma Cell Proliferation, Prognosis, and Immune Regulation. Clin Cosmet Investig Dermatol 2024; 17:2733-2746. [PMID: 39629045 PMCID: PMC11614586 DOI: 10.2147/ccid.s477870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 11/15/2024] [Indexed: 12/06/2024]
Abstract
Purpose Heat shock protein A4 (HSPA4) is associated with a variety of human diseases. However, its function in cutaneous malignant melanoma (CMM) remains uncertain. Patients and Methods The gene and protein expression level of HSPA4 in CMM was investigated with public databases. Cell Counting Kit-8 (CCK8) assay was performed to assess the effect of HSPA4 on the proliferation of melanoma cells. Then, the diagnostic and prognostic value of HSPA4 in CMM were analyzed. Gene variations and methylation levels, and the correlation between HSPA4 expression and immune cell infiltration were evaluated, followed by the construction of HSPA4 related protein-protein interaction networks and functional enrichment analysis. Results The mRNA and protein expression level of HSPA4 was significantly higher in CMM. Knocking down HSPA4 in A-375 cell line could inhibit tumor cell growth. The receiver operating characteristic (ROC) curve analysis confirmed the diagnostic value of HSPA4. Survival analysis showed that high expression of HSPA4 was associated with poor prognosis. HSPA4 gene alterations were observed in 3% of CMM patients. Five CpG sites are associated with the prognosis of CMM. HSPA4 is negatively correlated with most immune cells in CMM. The protein interaction network shows that HSPA4 is closely related to proteins such as DnaJ heat shock protein family (Hsp40) member B1 (DNAJB1) and DnaJ heat shock protein family (Hsp40) member B6 (DNAJB6), and the expression of DNAJB1 is positively correlated with HSPA4. Functional enrichment analysis indicated that HSPA4 may be associated with immune suppression and immune escape within the tumor microenvironment of CMM. Conclusion HSPA4 may participate in the regulation of tumor development and microenvironment, which may be a potential diagnostic and prognostic marker of CMM.
Collapse
Affiliation(s)
- Xudong Wang
- Outpatient Department of Yangfangdian, Southern Medical District of Chinese PLA General Hospital, Beijing, 100843, People’s Republic of China
- Department of Dermatology, Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100010, People’s Republic of China
- Medical School of Chinese PLA, Beijing, 100853, People’s Republic of China
| | - Zhiyong Li
- Outpatient Department of Yangfangdian, Southern Medical District of Chinese PLA General Hospital, Beijing, 100843, People’s Republic of China
| | - Jianhong Xu
- Outpatient Department of Yangfangdian, Southern Medical District of Chinese PLA General Hospital, Beijing, 100843, People’s Republic of China
| | - Jun Wang
- Outpatient Department of Yangfangdian, Southern Medical District of Chinese PLA General Hospital, Beijing, 100843, People’s Republic of China
| | - Ying Li
- Outpatient Department of Yangfangdian, Southern Medical District of Chinese PLA General Hospital, Beijing, 100843, People’s Republic of China
| | - Qiang Li
- Medical Health Care Dept, Air Force Medical Center PLA, Beijing, 100142, People’s Republic of China
| | - Jianrong Niu
- Department of Dermatology, Air Force Medical Center PLA, Beijing, 100142, People’s Republic of China
| | - Rongya Yang
- Department of Dermatology, Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100010, People’s Republic of China
| |
Collapse
|
9
|
Zhao C. Correlation between thyroid dysfunction and efficacy of immune checkpoint inhibitors in patients with advanced solid tumors. Discov Oncol 2024; 15:663. [PMID: 39549217 PMCID: PMC11569323 DOI: 10.1007/s12672-024-01546-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 11/06/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are currently the first line of tumor immunotherapy for the treatment of a wide range of tumors. However, the main side effect of immune checkpoint inhibitors is that they cause immune-related adverse events (irAEs) in patients. The relationship between the occurrence of immune side effects in patients and efficacy is controversial. The objective of this study was to confirm the relationship between patients who develop thyroid dysfunction after immune checkpoint inhibitors and efficacy. METHODS This study was a retrospective real-world clinical study, and a total of 50 patients with advanced tumors treated with immune checkpoint inhibitors at Anqing People's Hospital from 2020.8 to 2022.5 were retrospectively collected. Among them, 30 patients with hypothyroidism and 20 patients with normal or hyperthyroidism occurred, and the treatment effects and prognostic differences between the two groups were compared. FINDING After PD1 treatment in all patients, there were 10 cases of partial remission(PR), 18 cases of stable disease(SD) and 2 cases of progressive disease(PD) in patients who developed hypothyroidism, with a disease control rate(DCR) of 93.3% and objective remission rate(ORR) of 33.3%. There were 0 complete remission (CR) patient, 3 PR patients, 11 SD patients, 6 PD patients, 70.0% DCR and 15.0% ORR in patients who did not develop hypothyroidism. The DCR and ORR of patients who developed hypothyroidism were better than those of non-hypothyroid patients, and the difference was statistically significant (P < 0.05). Kaplan-Meier survival analysis showed that progression-free survival (PFS) reached 9.2 months (95% CI 7.726-10.779) in patients who developed hypothyroidism and did not hypothyroidism patients have a PFS of 7.3 months (95% CI 5.604-8.505), with a statistically significant difference (P = 0.0341 < 0.05). Further subgroup analysis revealed that among patients who developed hypothyroidism, PFS was 3.3 months (95% CI 0.630-5.440) in patients with cholangiocarcinoma, 6.89 months (95% CI 5.604-8.505) in patients with non-small cell lung cancer, > 12 months in patients with hepatocellular carcinoma, and 11.05 months (95% CI 9.308-12.792) in patients with esophageal cancer months and PFS for patients with gastric cancer was 9.74 months (95% CI 6.979-12.502). INTERPRETATION When patients with advanced tumors were treated with immune checkpoint inhibitors, DCR and ORR were higher in patients who developed hypothyroidism, and patients had better PFS. The benefits were greater in patients with gastric, esophageal, and hepatocellular carcinomas.
Collapse
Affiliation(s)
- Cheng Zhao
- Anqing Petrochemical Hospital of Nanjing Drum Tower Hospital Group, Anhui, China.
| |
Collapse
|
10
|
Venturi F, Veronesi G, Scotti B, Dika E. Cutaneous Toxicities of Advanced Treatment for Cutaneous Melanoma: A Prospective Study from a Single-Center Institution. Cancers (Basel) 2024; 16:3679. [PMID: 39518117 PMCID: PMC11545238 DOI: 10.3390/cancers16213679] [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: 10/01/2024] [Revised: 10/19/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES The landscape of advanced melanoma treatments has shifted dramatically in recent years. Target therapy and immunotherapy have changed the management of patients with both metastatic (stage IV according to AJCC 8th ed.) and nodal (stage IIB/C and III) disease. As the use of novel agents has increased, so have the cutaneous toxicities associated with these medications. While most skin reactions are low-grade and can be managed conservatively with topical therapies, high-grade or life-threatening drug reactions can arise during therapy, requiring prompt dermatologic recognition and treatment. Given the survival benefit attributed to these new agents, treating skin toxicity and maintaining a patient's quality of life is of paramount importance. METHODS We undertook a prospective, monocentric, and descriptive study in Bologna, Italy, including patients referred to the Oncologic Dermatology Unit of IRCCS AOU of Bologna who developed biopsy-proven cutaneous adverse events (AE) under treatment with immunotherapy for cutaneous melanoma with nodal (stage IIB/C, III) and metastatic (stage IV) disease from January 2016 to April 2024. RESULTS In 202 identified patients, 75 (37.5%) developed skin AEs. Ipilimumab was causal for 48.1% of skin AEs, followed by nivolumab (37%) and pembrolizumab (31.4%). Recorded types of skin AEs included erythematous rash, vitiligo, alopecia, lichenoid, maculopapular, acneiform, urticarial, psoriasiform, granulomatous, eczematous, and severe cutaneous AEs, such as Erythema multiforme/Stevens-Johnson syndrome and bullous autoimmune dermatoses. Most AEs were low-grade [CTCAE 1-2] (97%) and typically occurred after 10 weeks of treatment. CONCLUSIONS This study comprehensively describes skin AEs occurring during systemic treatment with ICIs for cutaneous melanoma at a single center.
Collapse
Affiliation(s)
- Federico Venturi
- Oncologic Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy (E.D.)
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum of Bologna, 40126 Bologna, Italy
| | - Giulia Veronesi
- Oncologic Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy (E.D.)
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum of Bologna, 40126 Bologna, Italy
| | - Biagio Scotti
- Oncologic Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy (E.D.)
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum of Bologna, 40126 Bologna, Italy
| | - Emi Dika
- Oncologic Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy (E.D.)
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum of Bologna, 40126 Bologna, Italy
| |
Collapse
|
11
|
Liu H, Lu Y, Zong J, Zhang B, Li X, Qi H, Yu T, Li Y. Engineering dendritic cell biomimetic membrane as a delivery system for tumor targeted therapy. J Nanobiotechnology 2024; 22:663. [PMID: 39465376 PMCID: PMC11520105 DOI: 10.1186/s12951-024-02913-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 10/07/2024] [Indexed: 10/29/2024] Open
Abstract
Targeted immunotherapies make substantial strides in clinical cancer care due to their ability to counteract the tumor's capacity to suppress immune responses. Advances in biomimetic technology with minimally immunogenic and highly targeted, are addressing issues of targeted drug delivery and disrupting the tumor's immunosuppressive environment to trigger immune activation. Specifically, the use of dendritic cell (DC) membranes to coat nanoparticles ensures targeted delivery due to DC's unique ability to activate naive T cells, spotlighting their role in immunotherapy aimed at disrupting the tumor microenvironment. The potential of DC's biomimetic membrane to mediate immune activation and target tumors is gaining momentum, enhancing the effectiveness of cancer treatments in conjunction with other immune responses. This review delves into the methodologies behind crafting DC membranes and the fusion of dendritic and tumor cell membranes for encapsulating therapeutic nanoparticles. It explores their applications and recent advancements in combating cancer, offering an all-encompassing perspective on DC biomimetic nanosystems, immunotherapy driven by antigen presentation, and the collaborative efforts of drug delivery in chemotherapy and photodynamic therapies. Current evidence shows promise in augmenting combined therapeutic approaches for cancer treatment and holds translational potential for various cancer treatments in a clinical setting.
Collapse
Affiliation(s)
- Huiyang Liu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, People's Republic of China
| | - Yiming Lu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, People's Republic of China
| | - Jinbao Zong
- Clinical Laboratory, Central Laboratory, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, People's Republic of China
| | - Bei Zhang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Xiaolu Li
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China
| | - Hongzhao Qi
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao, 266021, People's Republic of China
| | - Tao Yu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China.
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao, 266021, People's Republic of China.
| | - Yu Li
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, People's Republic of China.
| |
Collapse
|
12
|
Khamaru S, Mukherjee T, Tung KS, Kumar PS, Bandyopadhyay S, Mahish C, Chattopadhyay S, Chattopadhyay S. Chikungunya virus infection inhibits B16 melanoma-induced immunosuppression of T cells and macrophages mediated by interleukin 10. Microb Pathog 2024; 197:107022. [PMID: 39419458 DOI: 10.1016/j.micpath.2024.107022] [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: 06/03/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 10/19/2024]
Abstract
Immunosuppression in cancer poses challenges for immunotherapy and highlights the vulnerability of immunocompromised patients to viral infections. This study explored how Chikungunya virus (CHIKV) infection potentially inhibits B16-F10 melanoma-induced immunosuppressive effects on T cells and RAW 264.7 macrophages. We found high expression of CHIKV entry genes in melanoma and other cancers, with B16-F10 cells demonstrating greater susceptibility to CHIKV infection than non-tumorigenic cells. Interestingly, the CHIKV-infected B16-F10 cell culture supernatant (B16-F10-CS) reversed the immunosuppressive effects of uninfected B16-F10-CS on T cells. This reversal was characterised by decreased STAT3 activation and increased MAPK activation in T cells, an effect amplified by interleukin 10 (IL-10) receptor blockade. In RAW 264.7 cells, B16-F10-CS enhanced CHIKV infectivity without triggering activation. However, blocking the IL-10 receptor (IL-10R) in RAW 264.7 reduced CHIKV infection. CHIKV infection and IL-10R blockade synergistically inhibited B16-F10-CS-mediated polarisation of RAW 264.7 cells towards immunosuppressive macrophage. Our findings suggest that CHIKV modulates cancer-induced immunosuppression through IL-10-dependent pathways, providing new insights into viral-cancer interactions. This research may contribute to developing novel antiviral immunotherapies and virotherapies beneficial for cancer patients and immunocompromised individuals.
Collapse
Affiliation(s)
- Somlata Khamaru
- National Institute of Science Education and Research, School of Biological Sciences, Bhubaneswar, Odisha, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India
| | - Tathagata Mukherjee
- National Institute of Science Education and Research, School of Biological Sciences, Bhubaneswar, Odisha, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India; Institute of Life Sciences, Bhubaneswar, India
| | - Kshyama Subhadarsini Tung
- National Institute of Science Education and Research, School of Biological Sciences, Bhubaneswar, Odisha, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India
| | - P Sanjai Kumar
- Institute of Life Sciences, Bhubaneswar, India; Division of Neonatology and Newborn Nursery, University of Wisconsin, Madison, USA
| | - Saumya Bandyopadhyay
- National Institute of Science Education and Research, School of Biological Sciences, Bhubaneswar, Odisha, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India; Johns Hopkins University School of Medicine, Department of Biological Chemistry, 725 North Wolfe Street, Baltimore, Maryland, USA
| | - Chandan Mahish
- National Institute of Science Education and Research, School of Biological Sciences, Bhubaneswar, Odisha, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India
| | | | - Subhasis Chattopadhyay
- National Institute of Science Education and Research, School of Biological Sciences, Bhubaneswar, Odisha, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India.
| |
Collapse
|
13
|
Choi JH, Kim JY, Lee KR, Lee GY, Hong M, Hwang HW, Lee MY, Kim MK, Hong SA. Prognostic Significance of CD11b-, CD8-, and CD163-Positive Tumor-Infiltrating Immune Cells in Distal Bile Duct Cancer. J Pers Med 2024; 14:1033. [PMID: 39452540 PMCID: PMC11508419 DOI: 10.3390/jpm14101033] [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: 08/24/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
Background: Distal bile duct cancer is an aggressive malignancy. Tumor-infiltrating immune cells (TIICs) in the tumor microenvironment are crucial for predicting prognosis in various cancers. In this study, we analyzed TIICs based on CD11b, CD163, and CD8 expression, and evaluated their association with clinicopathologic factors and prognosis in distal bile duct cancer. Methods: A total of 90 patients who underwent curative resection for distal bile duct cancer were enrolled. We analyzed CD11b+ tumor-infiltrating myeloid cells (TIMs), CD163+ tumor-infiltrating macrophages (TAMs), and CD8+ tumor-infiltrating lymphocytes (TILs) using immunohistochemistry and tissue microarrays. The correlation between TIICs and clinicopathologic characteristics was assessed. Results: Low levels of CD11b+ TIMs (p < 0.001) and high levels of CD8+ TILs (p = 0.003) were significantly associated with improved overall survival (OS). A combined low level of CD11b+ TIMs and high level of CD8+ TILs was identified as an independent favorable prognostic factor (hazard ratio, 0.159; confidence interval, 0.061-0.410; p < 0.001). Conclusions: CD11b+ TIMs play a crucial role in the tumor microenvironment and the prognosis of distal bile duct cancer. The combined analysis of CD11b+ TIMs and CD8+ TILs can predict survival in patients with distal bile duct cancer.
Collapse
Affiliation(s)
- Jae Hyung Choi
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea;
| | - Joo Young Kim
- Department of Pathology, College of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea; (J.Y.K.); (K.R.L.); (G.Y.L.); (M.H.); (H.W.H.); (M.K.K.)
| | - Ki Rim Lee
- Department of Pathology, College of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea; (J.Y.K.); (K.R.L.); (G.Y.L.); (M.H.); (H.W.H.); (M.K.K.)
| | - Gyeong Yun Lee
- Department of Pathology, College of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea; (J.Y.K.); (K.R.L.); (G.Y.L.); (M.H.); (H.W.H.); (M.K.K.)
| | - Mineui Hong
- Department of Pathology, College of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea; (J.Y.K.); (K.R.L.); (G.Y.L.); (M.H.); (H.W.H.); (M.K.K.)
| | - Hye Won Hwang
- Department of Pathology, College of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea; (J.Y.K.); (K.R.L.); (G.Y.L.); (M.H.); (H.W.H.); (M.K.K.)
| | - Moo Yeol Lee
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea;
| | - Mi Kyung Kim
- Department of Pathology, College of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea; (J.Y.K.); (K.R.L.); (G.Y.L.); (M.H.); (H.W.H.); (M.K.K.)
| | - Soon Auck Hong
- Department of Pathology, College of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea; (J.Y.K.); (K.R.L.); (G.Y.L.); (M.H.); (H.W.H.); (M.K.K.)
| |
Collapse
|
14
|
Basir S, Bosiers J, Westgeest HM, Yick DCY, van Werven JR, van der Leest CH. Bronchiolitis after Combination Immunotherapy With Ipilimumab and Nivolumab in a Melanoma Patient. J Immunother 2024; 47:263-265. [PMID: 38409751 DOI: 10.1097/cji.0000000000000509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 01/27/2024] [Indexed: 02/28/2024]
Abstract
Therapy with immune checkpoint inhibitors (ICIs) has significantly improved the prognosis of metastatic melanoma but is also associated with various immune-related adverse events (AE), including pulmonary toxicity. Herein, we describe the case of a 60-year-old female with metastasized melanoma with BRAF mutation under combination immunotherapy with ipilimumab and nivolumab, who presented with a persistent, nonproductive cough for the last two months. Her CT-scan showed de novo bronchial inflammation and wall thickening in all lung fields. Initial treatment with antimicrobial treatment and inhalation corticosteroids did not resolve her symptoms, nor the radiologic abnormalities. Additional testing with transbronchial cryobiopsy showed a histologic picture of diffuse ill-formed granulomas and the presence of moderate chronic active inflammation of the respiratory epithelium, consistent with medication-related bronchiolitis. Bronchiolitis, as present in this case, has rarely been reported as an immune-related AE. A thorough diagnostic workup is mandatory as it remains a diagnosis of exclusion. Management consists of discontinuing ICIs and administering systemic corticosteroids. The addition of immunosuppressive agents (e, infliximab, cyclophosphamide, or mycophenolate mofetil) can be considered in refractory cases. In our case, clinical and radiologic resolution was achieved after discontinuing the ICI and treatment with high-dose prednisone. This case shows that although bronchiolitis is a rare immune-related side effect of ICIs, oncologists, and pulmonologists should always be aware of this relatively easily treatable AE.
Collapse
Affiliation(s)
- Shahir Basir
- Department of Respiratory Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Jana Bosiers
- Department of Respiratory Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Hans M Westgeest
- Department of Internal Medicine, Amphia Hospital, Breda, the Netherlands
| | - David C Y Yick
- Department of Pathology, Amphia Hospital, Breda, The Netherlands
| | | | | |
Collapse
|
15
|
Benedusi M, Lee H, Lim Y, Valacchi G. Oxidative State in Cutaneous Melanoma Progression: A Question of Balance. Antioxidants (Basel) 2024; 13:1058. [PMID: 39334716 PMCID: PMC11428248 DOI: 10.3390/antiox13091058] [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: 06/12/2024] [Revised: 08/02/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
Reactive oxygen species (ROS) are highly bioactive molecules involved not only in tissue physiology but also in the development of different human conditions, including premature aging, cardiovascular pathologies, neurological and neurodegenerative disorders, inflammatory diseases, and cancer. Among the different human tumors, cutaneous melanoma, the most aggressive and lethal form of skin cancer, is undoubtedly one of the most well-known "ROS-driven tumor", of which one of the main causes is represented by ultraviolet (UV) rays' exposure. Although the role of excessive ROS production in melanoma development in pro-tumorigenic cell fate is now well established, little is known about its contribution to the progression of the melanoma metastatic process. Increasing evidence suggests a dual role of ROS in melanoma progression: excessive ROS production may enhance cellular growth and promote therapeutic resistance, but at the same time, it can also have cytotoxic effects on cancer cells, inducing their apoptosis. In this context, the aim of the present work was to focus on the relationship between cell redox state and the signaling pathways directly involved in the metastatic processes. In addition, oxidative or antioxidant therapeutic strategies for metastatic melanoma were also reviewed and discussed.
Collapse
Affiliation(s)
- Mascia Benedusi
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy
| | - Heaji Lee
- Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yunsook Lim
- Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Giuseppe Valacchi
- Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Republic of Korea
- Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, NC 28081, USA
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
16
|
Neill B, Romero AR, Fenton OS. Advances in Nonviral mRNA Delivery Materials and Their Application as Vaccines for Melanoma Therapy. ACS APPLIED BIO MATERIALS 2024; 7:4894-4913. [PMID: 37930174 PMCID: PMC11220486 DOI: 10.1021/acsabm.3c00721] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Messenger RNA (mRNA) vaccines are promising platforms for cancer immunotherapy because of their potential to encode for a variety of tumor antigens, high tolerability, and capacity to induce strong antitumor immune responses. However, the clinical translation of mRNA cancer vaccines can be hindered by the inefficient delivery of mRNA in vivo. In this review, we provide an overview of mRNA cancer vaccines by discussing their utility in treating melanoma. Specifically, we begin our review by describing the barriers that can impede mRNA delivery to target cells. We then review native mRNA structure and discuss various modification methods shown to enhance mRNA stability and transfection. Next, we outline the advantages and challenges of three nonviral carrier platforms (lipid nanoparticles, polymeric nanoparticles, and lipopolyplexes) frequently used for mRNA delivery. Last, we summarize preclinical and clinical studies that have investigated nonviral mRNA vaccines for the treatment of melanoma. In writing this review, we aim to highlight innovative nonviral strategies designed to address mRNA delivery challenges while emphasizing the exciting potential of mRNA vaccines as next-generation therapies for the treatment of cancers.
Collapse
Affiliation(s)
- Bevin Neill
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Adriana Retamales Romero
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Owen S. Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
17
|
Luo S, Wang D, Chen J, Hong S, Fang Y, Cao L, Yong L, Liu S. The combination of single-cell and RNA sequencing analysis decodes the melanoma tumor microenvironment and identifies novel T cell-associated signature genes. J Cancer 2024; 15:5085-5100. [PMID: 39132169 PMCID: PMC11310880 DOI: 10.7150/jca.96484] [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: 03/21/2024] [Accepted: 07/15/2024] [Indexed: 08/13/2024] Open
Abstract
Skin cutaneous melanoma (SKCM), a malignant melanocyte-derived skin cancer, potentially leads to fatal outcomes without effective treatment. The variability in immunotherapy responses among melanoma patients is significantly influenced by the intricate immune microenvironment, particularly due to the status of tumor T cells, encompassing their activity, exhaustion levels, and antigen recognition capabilities. This study utilized single-cell RNA sequencing (scRNA-seq) to analyze 34 melanoma samples from two public datasets (GSE215120 and GSE115978). Herein, we extracted 706 marker genes associated with immune checkpoint (ICP) therapy from these T cells, 509 markers of T cells from 11 melanoma tissues, and eventually identified 33 candidate genes. These genes underwent LASSO and COX regression analyses to identify the signature genes. Of the initial 33 candidate genes, we successfully isolated six distinct T cell-associated immunotherapy-related genes (IRTGs). Additionally, the computation of each patient risk score proved beneficial in evaluating the immune cell infiltration level and functions as an independent prognostic factor for melanoma patient survival. The risk score results revealed promising predictive outcomes in determining the response of melanoma patients to immunotherapy. Notably, our study is the first to reveal the potential correlation between signature gene PEB4B and the immune microenvironment in melaoma, which was explored with multiple immunofluorescence (IF) and Immune Infiltration Assessment. In a conclusion, our findings demonstrate the potential utility of a risk score dependent on signature genes as a predictive tool for assessing the prognosis and response to immunotherapeutic interventions in melanoma patients.
Collapse
Affiliation(s)
- Sihan Luo
- Department of Dermatology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, 230022, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, 230022, China
| | - Daiyue Wang
- Department of Dermatology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, 230022, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, 230022, China
| | - Jiajie Chen
- Department of Dermatology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, 230022, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, 230022, China
| | - Shaocheng Hong
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Yuanyuan Fang
- Department of Obstetrics and Gynecology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Lu Cao
- Department of Dermatology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Liang Yong
- Laboratory of Stem Cell, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, P R China
| | - Shengxiu Liu
- Department of Dermatology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, 230022, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, 230022, China
| |
Collapse
|
18
|
Peisen F, Gerken A, Hering A, Dahm I, Nikolaou K, Gatidis S, Eigentler TK, Amaral T, Moltz JH, Othman AE. Can Delta Radiomics Improve the Prediction of Best Overall Response, Progression-Free Survival, and Overall Survival of Melanoma Patients Treated with Immune Checkpoint Inhibitors? Cancers (Basel) 2024; 16:2669. [PMID: 39123397 PMCID: PMC11312160 DOI: 10.3390/cancers16152669] [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: 06/17/2024] [Revised: 07/16/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND The prevalence of metastatic melanoma is increasing, necessitating the identification of patients who do not benefit from immunotherapy. This study aimed to develop a radiomic biomarker based on the segmentation of all metastases at baseline and the first follow-up CT for the endpoints best overall response (BOR), progression-free survival (PFS), and overall survival (OS), encompassing various immunotherapies. Additionally, this study investigated whether reducing the number of segmented metastases per patient affects predictive capacity. METHODS The total tumour load, excluding cerebral metastases, from 146 baseline and 146 first follow-up CTs of melanoma patients treated with first-line immunotherapy was volumetrically segmented. Twenty-one random forest models were trained and compared for the endpoints BOR; PFS at 6, 9, and 12 months; and OS at 6, 9, and 12 months, using as input either only clinical parameters, whole-tumour-load delta radiomics plus clinical parameters, or delta radiomics from the largest ten metastases plus clinical parameters. RESULTS The whole-tumour-load delta radiomics model performed best for BOR (AUC 0.81); PFS at 6, 9, and 12 months (AUC 0.82, 0.80, and 0.77); and OS at 6 months (AUC 0.74). The model using delta radiomics from the largest ten metastases performed best for OS at 9 and 12 months (AUC 0.71 and 0.75). Although the radiomic models were numerically superior to the clinical model, statistical significance was not reached. CONCLUSIONS The findings indicate that delta radiomics may offer additional value for predicting BOR, PFS, and OS in metastatic melanoma patients undergoing first-line immunotherapy. Despite its complexity, volumetric whole-tumour-load segmentation could be advantageous.
Collapse
Affiliation(s)
- Felix Peisen
- Department of Diagnostic and Interventional Radiology, Eberhard Karls University, Tuebingen University Hospital, Hoppe-Seyler-Straße 3, 72076 Tuebingen, Germany; (I.D.); (K.N.); (S.G.)
| | - Annika Gerken
- Fraunhofer Institute for Digital Medicine MEVIS, Max-von-Laue-Straße 2, 28359 Bremen, Germany; (A.G.); (A.H.); (J.H.M.)
| | - Alessa Hering
- Fraunhofer Institute for Digital Medicine MEVIS, Max-von-Laue-Straße 2, 28359 Bremen, Germany; (A.G.); (A.H.); (J.H.M.)
- Diagnostic Image Analysis Group, Radboudumc, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Isabel Dahm
- Department of Diagnostic and Interventional Radiology, Eberhard Karls University, Tuebingen University Hospital, Hoppe-Seyler-Straße 3, 72076 Tuebingen, Germany; (I.D.); (K.N.); (S.G.)
| | - Konstantin Nikolaou
- Department of Diagnostic and Interventional Radiology, Eberhard Karls University, Tuebingen University Hospital, Hoppe-Seyler-Straße 3, 72076 Tuebingen, Germany; (I.D.); (K.N.); (S.G.)
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, Faculty of Medicine, Eberhard Karls University, 72076 Tuebingen, Germany
| | - Sergios Gatidis
- Department of Diagnostic and Interventional Radiology, Eberhard Karls University, Tuebingen University Hospital, Hoppe-Seyler-Straße 3, 72076 Tuebingen, Germany; (I.D.); (K.N.); (S.G.)
- Max Planck Institute for Intelligent Systems, Max-Planck-Ring 4, 72076 Tuebingen, Germany
| | - Thomas K. Eigentler
- Center of Dermato-Oncology, Department of Dermatology, Eberhard Karls University, Tuebingen University Hospital, Liebermeisterstraße 25, 72076 Tuebingen, Germany; (T.K.E.); (T.A.)
- Department of Dermatology, Venereology and Allergology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Luisenstraße 2, 10117 Berlin, Germany
| | - Teresa Amaral
- Center of Dermato-Oncology, Department of Dermatology, Eberhard Karls University, Tuebingen University Hospital, Liebermeisterstraße 25, 72076 Tuebingen, Germany; (T.K.E.); (T.A.)
| | - Jan H. Moltz
- Fraunhofer Institute for Digital Medicine MEVIS, Max-von-Laue-Straße 2, 28359 Bremen, Germany; (A.G.); (A.H.); (J.H.M.)
| | - Ahmed E. Othman
- Institute of Neuroradiology, Johannes Gutenberg University Hospital Mainz, Langenbeckstraße 1, 55131 Mainz, Germany;
| |
Collapse
|
19
|
Almeida TC, Giannotti KC, Ribeiro Silva LM, Marques-Porto R, DeOcesano-Pereira C, Camargo L, Chudzinski-Tavassi AM, Reid P, Picolo G. Crotoxin induces cytotoxic effects in human malignant melanoma cells in both native and detoxified forms. Front Pharmacol 2024; 15:1425446. [PMID: 39114354 PMCID: PMC11303296 DOI: 10.3389/fphar.2024.1425446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction: Melanoma, a highly aggressive skin cancer originating in melanocytes, poses a significant threat due to its metastatic potential. While progress has been made in treating melanoma with targeted therapies and immunotherapies, challenges persist. Crotoxin (CTX), the principal toxin in Crotalus durissus terrificus snake venom, exhibits various biological activities, including anti-tumoral effects across multiple cancers. However, its clinical use is limited by toxicity. Thus, exploring alternatives to mitigate adverse effects is crucial. Methods and Results: This study investigates the antitumoral potential of CTX in its native and in a detoxified form, in melanoma cells. Firstly, we demonstrated that detoxified CTX presented reduced phospholipase activity. Both forms proved to be more cytotoxic to SK-MEL-28 and MeWo melanoma cells than non-tumoral cells. In SK-MEL-28 cells, where cytotoxic effects were more pronounced, native and detoxified CTX induced increased necrosis and apoptosis rates. We also confirmed the apoptosis death demonstrated by the activation of caspase-3 and 7, and the formation of apoptotic bodies. Furthermore, both CTX caused cell cycle arrest at the G2/M phase, interfering with melanoma cell proliferation. Cell migration and invasion were also suppressed by both CTX. These results confirm the antitumoral potential of CTX. Discussion: The maintenance of the antiproliferative effects in the detoxified version, with reduced enzymatic activity often liked to harm effects, supports further studies to identify active parts of the molecule responsible for the interesting effects without causing substantial toxic events, contributing to the future use of CTX-derived drugs with safety and efficacy.
Collapse
Affiliation(s)
| | | | | | | | | | - Lauren Camargo
- Centre of Excellence in New Target Discovering (CENTD), Butantan Institute, Sao Paulo, Brazil
| | | | - Paul Reid
- Celtic Biotech Ltd., Dublin, Ireland
| | - Gisele Picolo
- Laboratory of Pain and Signaling, Butantan Institute, Sao Paulo, Brazil
| |
Collapse
|
20
|
Didier AJ, Nandwani SV, Watkins D, Fahoury AM, Campbell A, Craig DJ, Vijendra D, Parquet N. Patterns and trends in melanoma mortality in the United States, 1999-2020. BMC Cancer 2024; 24:790. [PMID: 38956559 PMCID: PMC11221171 DOI: 10.1186/s12885-024-12426-z] [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: 03/09/2024] [Accepted: 05/24/2024] [Indexed: 07/04/2024] Open
Abstract
INTRODUCTION Melanoma, a deadly form of skin cancer, has witnessed a notable increase in incidence over the past decades. Despite advancements in treatment, it remains a significant cause of cancer mortality. Understanding demographic trends and variations in melanoma mortality is crucial for addressing disparities and implementing effective interventions. METHODS Using the Centers for Disease Control Wide Ranging Online Data for Epidemiologic Research (CDC WONDER) database, we analyzed melanoma mortality data in the United States from 1999 to 2020. Data were stratified by demographic and regional variables, and age-adjusted mortality rates were calculated. Descriptive analysis was performed and Joinpoint regression analysis was employed to identify temporal trends. RESULTS Between 1999 and 2020, there were 184,416 melanoma-related deaths in the United States Overall, the age-adjusted mortality rate declined from 2.7 to 2.0 per 100,000 people at a rate of -1.3% annually, with significant variations across demographic groups and regions. Men, non-Hispanic White individuals, and those aged > 65 experienced higher mortality rates. Non-Hispanic White individuals noted the steepest decrease in AAMR after 2013 at a rate of -6.1% annually. Disparities were seen by geographic density, with rural populations exhibiting higher mortality compared to their urban and suburban counterparts. CONCLUSION The study highlights a significant reduction in melanoma mortality in the U.S. since 2013, potentially attributed to advancements in diagnostic techniques such as dermoscopy and the introduction of immune checkpoint inhibitors. Disparities persist, particularly among rural populations. Targeted interventions focusing on increased screening and education are warranted to further mitigate melanoma mortality and address demographic disparities.
Collapse
Affiliation(s)
- Alexander J Didier
- The University of Toledo College of Medicine and Life Sciences, 3000 Arlington Ave, Toledo, OH, USA.
| | - Swamroop V Nandwani
- The University of Toledo College of Medicine and Life Sciences, 3000 Arlington Ave, Toledo, OH, USA
| | - Dean Watkins
- The University of Toledo College of Medicine and Life Sciences, 3000 Arlington Ave, Toledo, OH, USA
| | - Alan M Fahoury
- The University of Toledo College of Medicine and Life Sciences, 3000 Arlington Ave, Toledo, OH, USA
| | - Andrew Campbell
- The University of Toledo College of Medicine and Life Sciences, 3000 Arlington Ave, Toledo, OH, USA
| | - Daniel J Craig
- The University of Toledo College of Medicine and Life Sciences, 3000 Arlington Ave, Toledo, OH, USA
| | - Divya Vijendra
- Division of Hematology and Oncology, Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Nancy Parquet
- Division of Dermatology, Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| |
Collapse
|
21
|
Kiraly P, Fischer MD. RETRACTED ARTICLE: Cystoid Macular Oedema in a Patient Treated with STING Agonist and Ezabenlimab for Disseminated Melanoma. Ophthalmol Ther 2024; 13:2061. [PMID: 38467992 PMCID: PMC11178736 DOI: 10.1007/s40123-024-00911-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 02/15/2024] [Indexed: 03/13/2024] Open
Affiliation(s)
- Peter Kiraly
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom.
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford, United Kingdom.
| | - M Dominik Fischer
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford, United Kingdom
- Centre for Ophthalmology, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
22
|
Rix A, Heinrichs H, Porte C, Leenaars C, Bleich A, Kiessling F. Ultrasound-induced immune responses in tumors: A systematic review and meta-analysis. J Control Release 2024; 371:146-157. [PMID: 38777126 DOI: 10.1016/j.jconrel.2024.05.030] [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: 02/16/2024] [Revised: 04/29/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024]
Abstract
Ultrasound is widely used in the diagnosis and therapy of cancer. Tumors can be treated by thermal or mechanical tissue ablation. Furthermore, tumors can be manipulated by hyperthermia, sonodynamic therapy and sonoporation, e.g., by increasing tumor perfusion or the permeability of biological barriers to enhance drug delivery. These treatments induce various immune responses in tumors. However, conflicting data and high heterogeneity between experimental settings make it difficult to generalize the effects of ultrasound on tumor immunity. Therefore, we performed a systematic review to answer the question: "Does ultrasound alter the immune reaction of peripheral solid tumors in humans and animals compared to control conditions without ultrasound?" A systematic literature search was performed in PubMed, EMBASE, and Web of Science and 24,401 potentially relevant publications were identified. Of these, 96 publications were eligible for inclusion in the systematic review. Experiments were performed in humans, rats, and mice and focused on different tumor types, primarily breast and melanoma. We collected data on thermal and non-thermal ultrasound settings, the use of sono-sensitizers or sono-enhancers, and anti-tumor therapies. Six meta-analyses were performed to quantify the effect of ultrasound on tumor infiltration by T cells (cytotoxic, helper, and regulatory T cells) and on blood cytokines (interleukin-6, interferon-γ, tumor necrosis factor-α). We provide robust scientific evidence that ultrasound alters T cell infiltration into tumors and increases blood cytokine concentrations. Furthermore, we identified significant differences in immune cell infiltration based on tumor type, ultrasound settings, and mouse age. Stronger effects were observed using hyperthermia in combination with sono-sensitizers and in young mice. The latter may impair the translational impact of study results as most cancer patients are older. Thus, our results may help refining ultrasound parameters to enhance anti-tumor immune responses for therapeutic use and to minimize immune effects in diagnostic applications.
Collapse
Affiliation(s)
- Anne Rix
- Institute for Experimental Molecular Imaging, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Helen Heinrichs
- Institute for Experimental Molecular Imaging, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Céline Porte
- Institute for Experimental Molecular Imaging, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Cathalijn Leenaars
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - André Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, Medical Faculty, RWTH Aachen University, Aachen, Germany; Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany.
| |
Collapse
|
23
|
Shalata W, Attal ZG, Solomon A, Shalata S, Abu Saleh O, Tourkey L, Abu Salamah F, Alatawneh I, Yakobson A. Melanoma Management: Exploring Staging, Prognosis, and Treatment Innovations. Int J Mol Sci 2024; 25:5794. [PMID: 38891988 PMCID: PMC11171767 DOI: 10.3390/ijms25115794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Melanoma, a malignant neoplasm originating from melanocytes, stands as one of the most prevalent cancers globally, ranking fifth in terms of estimated new cases in recent years. Its aggressive nature and propensity for metastasis pose significant challenges in oncology. Recent advancements have led to a notable shift towards targeted therapies, driven by a deeper understanding of cutaneous tumor pathogenesis. Immunotherapy and tyrosine kinase inhibitors have emerged as promising strategies, demonstrating the potential to improve clinical outcomes across all disease stages, including neoadjuvant, adjuvant, and metastatic settings. Notably, there has been a groundbreaking development in the treatment of brain metastasis, historically associated with poor prognosis in oncology but showcasing impressive results in melanoma patients. This review article provides a comprehensive synthesis of the most recent knowledge on staging and prognostic factors while highlighting emerging therapeutic modalities, with a particular focus on neoadjuvant and adjuvant strategies, notably immunotherapy and targeted therapies, including the ongoing trials.
Collapse
Affiliation(s)
- Walid Shalata
- The Legacy Heritage Cancer Center and Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Zoe Gabrielle Attal
- Medical School for International Health, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Adam Solomon
- Medical School for International Health, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Sondos Shalata
- Nutrition Unit, Galilee Medical Center, Nahariya 22000, Israel
| | - Omar Abu Saleh
- Department of Dermatology and Venereology, The Emek Medical Centre, Afula 18341, Israel
| | - Lena Tourkey
- The Legacy Heritage Cancer Center and Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Fahed Abu Salamah
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
- Department of Dermatology, Soroka Medical Center, Beer Sheva 84105, Israel
| | - Ibrahim Alatawneh
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
- Department of Dermatology, Soroka Medical Center, Beer Sheva 84105, Israel
| | - Alexander Yakobson
- The Legacy Heritage Cancer Center and Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| |
Collapse
|
24
|
Wu LY, Park SH, Jakobsson H, Shackleton M, Möller A. Immune Regulation and Immune Therapy in Melanoma: Review with Emphasis on CD155 Signalling. Cancers (Basel) 2024; 16:1950. [PMID: 38893071 PMCID: PMC11171058 DOI: 10.3390/cancers16111950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Melanoma is commonly diagnosed in a younger population than most other solid malignancies and, in Australia and most of the world, is the leading cause of skin-cancer-related death. Melanoma is a cancer type with high immunogenicity; thus, immunotherapies are used as first-line treatment for advanced melanoma patients. Although immunotherapies are working well, not all the patients are benefitting from them. A lack of a comprehensive understanding of immune regulation in the melanoma tumour microenvironment is a major challenge of patient stratification. Overexpression of CD155 has been reported as a key factor in melanoma immune regulation for the development of therapy resistance. A more thorough understanding of the actions of current immunotherapy strategies, their effects on immune cell subsets, and the roles that CD155 plays are essential for a rational design of novel targets of anti-cancer immunotherapies. In this review, we comprehensively discuss current anti-melanoma immunotherapy strategies and the immune response contribution of different cell lineages, including tumour endothelial cells, myeloid-derived suppressor cells, cytotoxic T cells, cancer-associated fibroblast, and nature killer cells. Finally, we explore the impact of CD155 and its receptors DNAM-1, TIGIT, and CD96 on immune cells, especially in the context of the melanoma tumour microenvironment and anti-cancer immunotherapies.
Collapse
Affiliation(s)
- Li-Ying Wu
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4059, Australia;
- JC STEM Lab, Department of Otorhinolaryngology, Chinese University of Hong Kong, Shatin, Hong Kong SAR, China;
- Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Su-Ho Park
- JC STEM Lab, Department of Otorhinolaryngology, Chinese University of Hong Kong, Shatin, Hong Kong SAR, China;
- Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Haakan Jakobsson
- Department of Medical Oncology, Paula Fox Melanoma and Cancer Centre, Alfred Health, Melbourne, VIC 3004, Australia;
| | - Mark Shackleton
- Department of Medical Oncology, Paula Fox Melanoma and Cancer Centre, Alfred Health, Melbourne, VIC 3004, Australia;
- School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Andreas Möller
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4059, Australia;
- JC STEM Lab, Department of Otorhinolaryngology, Chinese University of Hong Kong, Shatin, Hong Kong SAR, China;
- Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
25
|
Wang F, Chen P, Ouyang S, Xiong K, Liu Z, Wang Y. Identification of prognostic m6A modification patterns and score system in melanoma patients. Medicine (Baltimore) 2024; 103:e37950. [PMID: 38669381 PMCID: PMC11049698 DOI: 10.1097/md.0000000000037950] [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: 11/30/2023] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
N6-methyladenosine (m6A) is the most common modification on RNAs and LncRNAs. It plays an important role in cancer stem cell differentiation, T cell differentiation, and immune homeostasis. In this study, we explored the potential roles of m6A modification of RNA in melanoma and investigated the immune cell infiltration in tumor microenvironment in diverse m6Aclusters and different m6Ascore groups. A consensus clustering algorithm determined m6A modification patterns based on 14 m6A regulators, and further explored the biological functions and the connection with TME. An m6A-related gene signature (m6Ascore) was constructed based on m6A-related genes using principal component analysis. Three m6A modification patterns were identified based on 14 m6A regulators, named as m6Aclusters A-C. The prognosis of m6Acluster A was more favorable than m6Aclusters B and C, and it was more closely associated with immune regulation. To quantify the m6A modification patterns of individual tumor, an m6Ascore was constructed, and patients were classified into high and low m6Ascore groups. The low m6Ascore group, which had a favorable prognosis, was more relevant to immunology. The expression of PD-L1 was higher and the immunophenoscore (IPS) revealed stronger response to immunotherapy in the low m6Ascore group. This study identified 3 m6A modification patterns with different immune characteristics and constructed an m6Ascore system to predict prognosis and immunogenicity of patients, which is conducive to clinical prognosis judgment and individual treatment.
Collapse
Affiliation(s)
- Feixiang Wang
- Medical Oncology Department, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangdong, Guangzhou, China
| | - Peijie Chen
- Medical Oncology Department, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangdong, Guangzhou, China
| | - Si Ouyang
- Medical Oncology Department, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangdong, Guangzhou, China
| | - Kaixin Xiong
- Medical Oncology Department, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangdong, Guangzhou, China
| | - Zichuan Liu
- Medical Oncology Department, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangdong, Guangzhou, China
| | - Yao Wang
- Medical Oncology Department, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangdong, Guangzhou, China
| |
Collapse
|
26
|
Fu YB, Liu CF, Wang JJ, Ji XL, Tang RH, Liao KY, Chen LY, Hong YZ, Fan BB, Wang SC, Liu WH. Immunomodulatory Function of Pien Tze Huang in T Cell-Mediated Anti-tumor Activity against B16-F10, MC38 and Hep1-6 Tumor Models. Chin J Integr Med 2024; 30:348-358. [PMID: 38212499 DOI: 10.1007/s11655-023-3749-2] [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: 05/10/2023] [Indexed: 01/13/2024]
Abstract
OBJECTIVE To investigate the anti-tumor effects of Pien Tze Huang (PZH) in mouse models of B16-F10 melanoma, MC38 colorectal cancer, Hep1-6 hepatocellular carcinoma and chemically induced hepatocellular carcinoma model. METHODS Various tumor models, including B16-F10, MC38 and Hep1-6 tumor hypodermic inoculation models, B16-F10 and Hep1-6 pulmonary metastasis models, Hep1-6 orthotopic implantation model, and chemically induced hepatocellular carcinoma model, were utilized to evaluate the anti-tumor function of PZH. Tumor growth was assessed by measuring tumor size and weight of solid tumors isolated from C57BL/6 mice. For cell proliferation and death of tumor cells in vitro, as well as T cell activation markers, cytokine production and immune checkpoints analysis, single-cell suspensions were prepared from mouse spleen, lymph nodes, and tumors after PZH treatment. RESULTS PZH demonstrated significant therapeutic efficacy in inhibiting tumor growth (P<0.01). Treatment with PZH resulted in a reduction in tumor size in subcutaneous MC38 colon adenocarcinoma and B16-F10 melanoma models, and decreased pulmonary metastasis of B16-F10 melanoma and Hep1-6 hepatoma (P<0.01). However, in vitro experiments showed that PZH only had slight impact on the cell proliferation and survival of tumor cells (P>0.05). Nevertheless, PZH exhibited a remarkable ability to enhance T cell activation and the production of interferon gamma, tumor necrosis factor alpha, and interleukin 2 in CD4+ T cells in vitro (P<0.01 or P<0.05). Importantly, PZH substantially inhibited T cell exhaustion and boosted cytokine production by tumor-infiltrating CD8+ T cells (P<0.01 or P<0.05). CONCLUSION This study has confirmed a novel immunomodulatory function of PZH in T cell-mediated anti-tumor immunity, indicating that PZH holds promise as a potential therapeutic agent for cancer treatment.
Collapse
Affiliation(s)
- Yu-Bing Fu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Science, Xiamen University, Xiamen, Fujian Province, 361102, China
| | - Chen-Feng Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Science, Xiamen University, Xiamen, Fujian Province, 361102, China
| | - Jin-Jia Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Science, Xiamen University, Xiamen, Fujian Province, 361102, China
| | - Xiao-Lin Ji
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Science, Xiamen University, Xiamen, Fujian Province, 361102, China
| | - Rong-Han Tang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Science, Xiamen University, Xiamen, Fujian Province, 361102, China
| | - Kun-Yu Liao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Science, Xiamen University, Xiamen, Fujian Province, 361102, China
| | - Ling-Yue Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Science, Xiamen University, Xiamen, Fujian Province, 361102, China
| | - Ya-Zhen Hong
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Science, Xiamen University, Xiamen, Fujian Province, 361102, China
| | - Bin-Bin Fan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Science, Xiamen University, Xiamen, Fujian Province, 361102, China
| | - Shi-Cong Wang
- Fujian Pien Tze Huang Enterprise Key Laboratory of Natural Medicine Research and Development, Zhangzhou, Fujian Province, 363000, China
| | - Wen-Hsien Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Science, Xiamen University, Xiamen, Fujian Province, 361102, China.
| |
Collapse
|
27
|
Yel I, Koch V, Gruenewald LD, Mahmoudi S, Alizadeh LS, Goekduman A, Eichler K, Vogl TJ, Dimitrova M, Booz C. Advancing Differentiation of Hepatic Metastases in Malignant Melanoma through Dual-Energy Computed Tomography Rho/Z Maps. Diagnostics (Basel) 2024; 14:742. [PMID: 38611654 PMCID: PMC11012221 DOI: 10.3390/diagnostics14070742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/21/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
OBJECTIVES The aim of this study is to evaluate the diagnostic accuracy of dual-energy computed tomography (DECT)-based Rho/Z maps in differentiating between metastases and benign liver lesions in patients diagnosed with malignant melanoma compared to conventional CT value measurements. METHODS This retrospective study included 73 patients (mean age, 70 ± 13 years; 43 m/30 w) suffering from malignant melanoma who had undergone third-generation DECT as part of tumor staging between December 2017 and December 2021. For this study, we measured Rho (electron density) and Z (effective atomic number) values as well as Hounsfield units (HUs) in hypodense liver lesions. Values were compared, and diagnostic accuracy for differentiation was computed using receiver operating characteristic (ROC) curve analyses. Additional performed MRI or biopsies served as a standard of reference. RESULTS A total of 136 lesions (51 metastases, 71 cysts, and 14 hemangiomas) in contrast-enhanced DECT images were evaluated. The most notable discrepancy (p < 0.001) between measured values and the highest diagnostic accuracy for distinguishing melanoma metastases from benign cysts was observed for the Z (0.992; 95% CI, 0.956-1) parameters, followed by Rho (0.908; 95% CI, 0.842-0.953) and finally HU120kV (0.829; 95% CI, 0.751-0.891). Conversely, when discriminating between liver metastases and hemangiomas, the HU120kV parameters showed the most significant difference (p < 0.001) and yielded the highest values for diagnostic accuracy (0.859; 95% CI, 0.740-0.937), followed by the Z parameters (0.790; 95% CI, 0.681-0.876) and finally the Rho values (0.621; 95% CI, 0.501-0.730). CONCLUSIONS Rho and Z measurements derived from DECT allow for improved differentiation of liver metastases and benign liver cysts in patients with malignant melanoma compared to conventional CT value measurements. In contrast, in differentiation between liver hemangiomas and metastases, Rho/Z maps show inferior diagnostic accuracy. Therefore, differentiation between these two lesions remains a challenge for CT imaging.
Collapse
|
28
|
Ji Y, Li C, Wan S, Zhang K, Liu Y, Shi S. Comprehensive pan-cancer analysis reveals SIRT5 is a predictive biomarker for prognosis and immunotherapy response. Funct Integr Genomics 2024; 24:60. [PMID: 38499806 DOI: 10.1007/s10142-024-01338-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/10/2024] [Accepted: 03/12/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND Sirtuin 5 (SIRT5) is a promising therapeutic target involved in regulating multiple metabolic pathways in cells and organisms. The role of SIRT5 in cancer is currently unclear, and a comprehensive systematic pan-cancer analysis is required to explore its value in diagnosis, prognosis, and immune function. METHODS We investigated the role of SIRT5 in tumorigenesis, diagnosis, prognosis, metabolic pathways, the immune microenvironment, and pan-cancer therapeutic response. Moreover, we explored chemicals affecting the expression of SIRT5 and computed the relationship between SIRT5 and drug sensitivity. Finally, the role of SIRT5 in melanoma was analyzed using a series of experiments in vitro and in vivo. RESULTS We found that SIRT5 is differentially expressed and shows early diagnostic value in various tumors and that somatic cell copy number alterations and DNA methylation contribute to its aberrant expression. SIRT5 expression correlates with clinical features. Besides, it is negatively (positively) correlated with several metabolic pathways and positively (negatively) correlated with several important metastasis-related and immune-related pathways. High SIRT5 expression predicts poor (or good) prognosis in various tumors and can affect drug sensitivity. We also demonstrated that SIRT5 expression significantly correlates with immunomodulator-associated molecules, lymphocyte subpopulation infiltration, and immunotherapeutic response biomarkers. In addition, we showed that SIRT5 is differentially expressed in immunotherapy cohorts. In addition, we explored various chemicals that may affect SIRT5 expression. In conclusion, we demonstrated that SIRT5 is a key pathogenic gene that promotes melanoma progression. CONCLUSION Our study provides a systematic analysis of SIRT5 and its regulatory genes. SIRT5 has excellent diagnostic and prognostic capabilities for many cancers. This may remodel the tumor microenvironment. The potential of SIRT5-based cancer therapies is emphasized and helps predict the response to immunotherapy.
Collapse
Affiliation(s)
- Yacong Ji
- Department of Dermatology, The Third Hospital of Hebei Medical University, No.139 Ziqiang Road, Qiaoxi District, Shijiazhuang, Hebei Province, 050051, China
| | - Chongyang Li
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Sicheng Wan
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, 400715, China
| | - Kui Zhang
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, USA
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Yaling Liu
- Department of Dermatology, The Third Hospital of Hebei Medical University, No.139 Ziqiang Road, Qiaoxi District, Shijiazhuang, Hebei Province, 050051, China.
| | - Shaomin Shi
- Department of Dermatology, The Third Hospital of Hebei Medical University, No.139 Ziqiang Road, Qiaoxi District, Shijiazhuang, Hebei Province, 050051, China.
| |
Collapse
|
29
|
Pears G, Mahajan A, Olsson-Brown A, Sacco J. A case of immunotherapy-induced thyroiditis. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:225-231. [PMID: 38464389 PMCID: PMC10918230 DOI: 10.37349/etat.2024.00214] [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: 03/20/2023] [Accepted: 12/21/2023] [Indexed: 03/12/2024] Open
Abstract
Immunotherapy treatments for cancer are known to cause adverse thyroid events which present a diagnostic challenge to clinicians and radiologists. This case report highlights the importance of a high clinical index of suspicion and careful assessment of the thyroid on serial imaging studies to make the diagnosis. The case involves a 65-year-old male with malignant melanoma who was started on immunotherapy as part of a clinical trial. He developed thyroid dysfunction followed by an attack of acute neck pain. Ultrasound of his thyroid was performed which showed significant atrophy. A review of previous imaging was undertaken which confirmed the patient had suffered from thyroiditis and subsequent atrophy. Following this, the diagnosis of immunotherapy-induced thyroid dysfunction was made. Thyroxine supplementation and steroid dose were then adjusted causing his thyroid function and symptoms to improve. Immunotherapy agents for cancers are becoming more and more common. As the case report shows, physicians and radiologists will need to be vigilant to diagnose and treat any adverse events.
Collapse
Affiliation(s)
- George Pears
- Department of Imaging, The Clatterbridge Cancer Centre NHS Foundation Trust, L7 8YA Liverpool, UK
| | - Abhishek Mahajan
- Department of Imaging, The Clatterbridge Cancer Centre NHS Foundation Trust, L7 8YA Liverpool, UK
- Faculty of Health and Life Sciences, University of Liverpool, L7 8TX Liverpool, UK
| | - Anna Olsson-Brown
- Oncology, The Clatterbridge Cancer Centre NHS Foundation Trust, L7 8YA Liverpool, UK
| | - Joseph Sacco
- Oncology, The Clatterbridge Cancer Centre NHS Foundation Trust, L7 8YA Liverpool, UK
| |
Collapse
|
30
|
Bundgaard Kjellingbro K, Naranjo Freixa C, Hjorth Mikkelsen L, Heegaard S. Challenges in diagnosing canine spindle cell tumours using immunohistochemistry, illustrated by three nonpigmented malignant cases from the nictitating membrane. Acta Vet Scand 2024; 66:7. [PMID: 38396026 PMCID: PMC10893616 DOI: 10.1186/s13028-024-00727-z] [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: 09/12/2023] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Nonpigmented malignant spindle cell tumours of the membrana nictitans are rare in dogs. In twenty-three years only three cases have been diagnosed in Scandinavia. This study describes the three cases of malignant tumours of the membrana nictitans recorded by the Eye Pathology Section, University of Copenhagen, Denmark, with reference to the clinical appearance and work-up, the treatment and prognosis, and the histopathological description including immunohistochemistry. The three cases are compared to previous publications on canine tumours of the nictitating membrane. We emphasize the importance of using protocols that are adapted to the specific species such as dogs. Opposite the human tissue responses, we even need more than one marker when diagnosing melanomas in dogs. RESULTS The dogs presented were an 8-year-old Dachshund, a 12-year-old Akita and a 14-year-old Shetland Sheepdog. All three dogs were entire females. All three nictitating membrane tumours developed on the right nictitating membrane as firm or multilobulated hyperaemic masses. Two of the tumours were macroscopically nonpigmented, the third being partly pigmented on the surface and ulcerated. According to the histopathology and for two of the cases immunohistochemistry with dog-adapted protocols the diagnoses included one hemangiosarcoma and two amelanotic melanomas. Tumour regrowth developed in all three cases and repeated resections were completed 1, 2 and 3 times, respectively, with recurrence experienced within 1.5 months - 3 years. CONCLUSIONS Nonpigmented malignant spindle cell tumours of the canine membrana nictitans are rare. Treatment of choice should be complete excision with a minimal histologic tumour-free distance and in case of a recurrence a full resection of the nictitating membrane. We strongly recommend a dog-adapted protocol for immunohistochemistry.
Collapse
Affiliation(s)
| | | | - Lauge Hjorth Mikkelsen
- Eye Pathology Section, Department of Pathology, Rigshospitalet, University of Copenhagen, Frederik V's Vej 11, 1st Floor, 2100, Copenhagen Ø, Denmark
- Department of Ophthalmology, Rigshospitalet, University of Copenhagen, Frederik V's Vej 11, 1st Floor, 2100, Copenhagen Ø, Denmark
| | - Steffen Heegaard
- Eye Pathology Section, Department of Pathology, Rigshospitalet, University of Copenhagen, Frederik V's Vej 11, 1st Floor, 2100, Copenhagen Ø, Denmark
- Department of Ophthalmology, Rigshospitalet, University of Copenhagen, Frederik V's Vej 11, 1st Floor, 2100, Copenhagen Ø, Denmark
| |
Collapse
|
31
|
Bonnet B, Tournier L, Deschamps F, Yevich S, Marabelle A, Robert C, Albiges L, Besse B, Bonnet V, De Baère T, Tselikas L. Thermal Ablation Combined with Immune Checkpoint Blockers: A 10-Year Monocentric Experience. Cancers (Basel) 2024; 16:855. [PMID: 38473217 DOI: 10.3390/cancers16050855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 03/14/2024] Open
Abstract
PURPOSE We report a 10-year experience in cancer therapy with concomitant treatment of percutaneous thermal ablation (PTA) and immune checkpoint blockers (ICBs). MATERIAL AND METHODS This retrospective cohort study included all patients at a single tertiary cancer center who had received ICBs at most 90 days before, or 30 days after, PTA. Feasibility and safety were assessed as the primary outcomes. The procedure-related complications and immune-related adverse events (irAEs) were categorized according to the Common Terminology Criteria for Adverse Events v5.0 (CTCAE). Efficacy was evaluated based on overall survival (OS), progression-free survival (PFS), and local progression-free survival (LPFS) according to the indication, ablation modality, neoplasm histology, and ICB type. RESULTS Between 2010 and 2021, 78 patients (57% male; median age: 61 years) were included. The PTA modality was predominantly cryoablation (CA) (61%), followed by radiofrequency ablation (RFA) (31%). PTA indications were the treatment of oligo-persistence (29%), oligo-progression (14%), and palliation of symptomatic lesions or prevention of skeletal-related events (SREs) (56%). Most patients received anti-PD1 ICB monotherapy with pembrolizumab (n = 35) or nivolumab (n = 24). The feasibility was excellent, with all combined treatment performed and completed as planned. Ten patients (13%) experienced procedure-related complications (90% grade 1-2), and 34 patients (44%) experienced an irAE (86% grade 1-2). The only factor statistically associated with better OS and PFS was the ablation indication, favoring oligo-persistence (p = 0.02). Tumor response was suggestive of an abscopal effect in four patients (5%). CONCLUSIONS The concomitant treatment of PTA and ICBs within 2-4 weeks is feasible and safe for both palliative and local control indications. Overall, PTA outcomes were found to be similar to standards for patients not on ICB therapy. While a consistently reproducible abscopal effect remains elusive, the safety profile of concomitant therapy provides the framework for continued assessment as ICB therapies evolve.
Collapse
Affiliation(s)
- Baptiste Bonnet
- Gustave Roussy, Département d'Anesthésie, Chirurgie et Interventionnel (DACI), F-94805 Villejuif, France
| | - Louis Tournier
- Gustave Roussy, Département d'Anesthésie, Chirurgie et Interventionnel (DACI), F-94805 Villejuif, France
- Department of Radiology, Saint-Louis Hospital, Université de Paris, F-75010 Paris, France
| | - Frédéric Deschamps
- Gustave Roussy, Département d'Anesthésie, Chirurgie et Interventionnel (DACI), F-94805 Villejuif, France
| | - Steven Yevich
- Department of Interventional Radiology, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Aurélien Marabelle
- Drug Development Department (DITEP), F-94805 Villejuif, France
- Laboratoire de Recherche Translationnelle en Immunothérapies (LRTI), Inserm U1015, F-94805 Villejuif, France
- Faculty of Medicine, Paris-Saclay University, F-94276 Le Kremlin Bicêtre, France
| | - Caroline Robert
- Faculty of Medicine, Paris-Saclay University, F-94276 Le Kremlin Bicêtre, France
- Gustave Roussy, Département de Médecine Oncologique, F-94805 Villejuif, France
| | - Laurence Albiges
- Faculty of Medicine, Paris-Saclay University, F-94276 Le Kremlin Bicêtre, France
- Gustave Roussy, Département de Médecine Oncologique, F-94805 Villejuif, France
| | - Benjamin Besse
- Faculty of Medicine, Paris-Saclay University, F-94276 Le Kremlin Bicêtre, France
- Gustave Roussy, Département de Médecine Oncologique, F-94805 Villejuif, France
| | - Victoire Bonnet
- Medicine Department, Campus Pierre et Marie Curie, Sorbonne University, 4 Place Jussieu, F-75005 Paris, France
| | - Thierry De Baère
- Gustave Roussy, Département d'Anesthésie, Chirurgie et Interventionnel (DACI), F-94805 Villejuif, France
- Faculty of Medicine, Paris-Saclay University, F-94276 Le Kremlin Bicêtre, France
| | - Lambros Tselikas
- Gustave Roussy, Département d'Anesthésie, Chirurgie et Interventionnel (DACI), F-94805 Villejuif, France
- Laboratoire de Recherche Translationnelle en Immunothérapies (LRTI), Inserm U1015, F-94805 Villejuif, France
- Faculty of Medicine, Paris-Saclay University, F-94276 Le Kremlin Bicêtre, France
| |
Collapse
|
32
|
Natarelli N, Aleman SJ, Mark IM, Tran JT, Kwak S, Botto E, Aflatooni S, Diaz MJ, Lipner SR. A Review of Current and Pipeline Drugs for Treatment of Melanoma. Pharmaceuticals (Basel) 2024; 17:214. [PMID: 38399429 PMCID: PMC10892880 DOI: 10.3390/ph17020214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Malignant melanoma is the most aggressive form of skin cancer. Standard treatment options include surgery, radiation therapy, systemic chemotherapy, targeted therapy, and immunotherapy. Combining these modalities often yields better responses. Surgery is suitable for localized cases, sometimes involving lymph node dissection and biopsy, to assess the spread of the disease. Radiation therapy may be sometimes used as a standalone treatment or following surgical excision. Systemic chemotherapy, while having low response rates, is utilized as part of combination treatments or when other methods fail. The development of resistance to systemic chemotherapies and associated side effects have prompted further research and clinical trials for novel approaches. In the case of advanced-stage melanoma, a comprehensive approach may be necessary, incorporating targeted therapies and immunotherapies that demonstrate significant antitumor activity. Targeted therapies, including inhibitors targeting BRAF, MEK, c-KIT, and NRAS, are designed to block the specific molecules responsible for tumor growth. These therapies show promise, particularly in patients with corresponding mutations. Combination therapy, including BRAF and MEK inhibitors, has been evidenced to improve progression-free survival; however, concerns about resistance and cutaneous toxicities highlight the need for close monitoring. Immunotherapies, leveraging tumor-infiltrating lymphocytes and CAR T cells, enhance immune responses. Lifileucel, an FDA-approved tumor-infiltrating lymphocyte therapy, has demonstrated improved response rates in advanced-stage melanoma. Ongoing trials continue to explore the efficacy of CAR T-cell therapy for advanced melanoma. Checkpoint inhibitors targeting CTLA-4 and PD-1 have enhanced outcomes. Emerging IL-2 therapies boost dendritic cells, enhancing anticancer immunity. Oncolytic virus therapy, approved for advanced melanoma, augments treatment efficacy in combination approaches. While immunotherapy has significantly advanced melanoma treatment, its success varies, prompting research into new drugs and factors influencing outcomes. This review provides insights into current melanoma treatments and recent therapeutic advances.
Collapse
Affiliation(s)
- Nicole Natarelli
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Sarah J. Aleman
- School of Medicine, Louisiana State University, New Orleans, LA 70112, USA
| | - Isabella M. Mark
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Jasmine T. Tran
- School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Sean Kwak
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Elizabeth Botto
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Shaliz Aflatooni
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Michael J. Diaz
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Shari R. Lipner
- Department of Dermatology, Weill Cornell Medicine, New York City, NY 10021, USA
| |
Collapse
|
33
|
He W, Zhang Y, Qu Y, Liu M, Li G, Pan L, Xu X, Shi G, Hao Q, Liu F, Gao Y. Research progress on hydrogel-based drug therapy in melanoma immunotherapy. BMB Rep 2024; 57:71-78. [PMID: 38053295 PMCID: PMC10910090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/04/2023] [Accepted: 11/15/2023] [Indexed: 12/07/2023] Open
Abstract
Melanoma is one of the most aggressive skin tumors, and conventional treatment modalities are not effective in treating advanced melanoma. Although immunotherapy is an effective treatment for melanoma, it has disadvantages, such as a poor response rate and serious systemic immune-related toxic side effects. The main solution to this problem is the use of biological materials such as hydrogels to reduce these side effects and amplify the immune killing effect against tumor cells. Hydrogels have great advantages as local slow-release drug carriers, including the ability to deliver antitumor drugs directly to the tumor site, enhance the local drug concentration in tumor tissue, reduce systemic drug distribution and exhibit good degradability. Despite these advantages, there has been limited research on the application of hydrogels in melanoma treatment. Therefore, this article provides a comprehensive review of the potential application of hydrogels in melanoma immunotherapy. Hydrogels can serve as carriers for sustained drug delivery, enabling the targeted and localized delivery of drugs with minimal systemic side effects. This approach has the potential to improve the efficacy of immunotherapy for melanoma. Thus, the use of hydrogels as drug delivery vehicles for melanoma immunotherapy has great potential and warrants further exploration. [BMB Reports 2024; 57(2): 71-78].
Collapse
Affiliation(s)
- Wei He
- College of Life Science, Northwest University, Xi’an 710069, China
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Yanqin Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Air Force Military Medical University, Xi’an 710032, China
| | - Yi Qu
- Department of Xi’an Shunmei Medical Cosmetology Outpatient, Xi’an 710075, China
| | - Mengmeng Liu
- College of Life Science, Northwest University, Xi’an 710069, China
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Guodong Li
- College of Life Science, Northwest University, Xi’an 710069, China
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Luxiang Pan
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Xinyao Xu
- College of Life Science, Northwest University, Xi’an 710069, China
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Gege Shi
- College of Life Science, Northwest University, Xi’an 710069, China
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Qiang Hao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Fen Liu
- Department of Periodontology, Shenzhen Stomatological Hospital (Pingshan), Southern Medical University, Shenzhen 510515, China
| | - Yuan Gao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| |
Collapse
|
34
|
Xuan L, Bai C, Ju Z, Luo J, Guan H, Zhou PK, Huang R. Radiation-targeted immunotherapy: A new perspective in cancer radiotherapy. Cytokine Growth Factor Rev 2024; 75:1-11. [PMID: 38061920 DOI: 10.1016/j.cytogfr.2023.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 02/16/2024]
Abstract
In contemporary oncology, radiation therapy and immunotherapy stand as critical treatments, each with distinct mechanisms and outcomes. Radiation therapy, a key player in cancer management, targets cancer cells by damaging their DNA with ionizing radiation. Its effectiveness is heightened when used alongside other treatments like surgery and chemotherapy. Employing varied radiation types like X-rays, gamma rays, and proton beams, this approach aims to minimize damage to healthy tissue. However, it is not without risks, including potential damage to surrounding normal cells and side effects ranging from skin inflammation to serious long-term complications. Conversely, immunotherapy marks a revolutionary step in cancer treatment, leveraging the body's immune system to target and destroy cancer cells. It manipulates the immune system's specificity and memory, offering a versatile approach either alone or in combination with other treatments. Immunotherapy is known for its targeted action, long-lasting responses, and fewer side effects compared to traditional therapies. The interaction between radiation therapy and immunotherapy is intricate, with potential for both synergistic and antagonistic effects. Their combined use can be more effective than either treatment alone, but careful consideration of timing and sequence is essential. This review explores the impact of various radiation therapy regimens on immunotherapy, focusing on changes in the immune microenvironment, immune protein expression, and epigenetic factors, emphasizing the need for personalized treatment strategies and ongoing research to enhance the efficacy of these combined therapies in cancer care.
Collapse
Affiliation(s)
- Lihui Xuan
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China; Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Chenjun Bai
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Zhao Ju
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China; Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Jinhua Luo
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China; Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Hua Guan
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China.
| | - Ping-Kun Zhou
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China.
| | - Ruixue Huang
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China.
| |
Collapse
|
35
|
Su J, Fu Y, Cui Z, Abidin Z, Yuan J, Zhang X, Li R, Zhao C. Relatlimab: a novel drug targeting immune checkpoint LAG-3 in melanoma therapy. Front Pharmacol 2024; 14:1349081. [PMID: 38269271 PMCID: PMC10806167 DOI: 10.3389/fphar.2023.1349081] [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: 12/04/2023] [Accepted: 12/27/2023] [Indexed: 01/26/2024] Open
Abstract
Relatlimab is a type of human immunoglobulin G4 monoclonal blocking antibody. It is the world's first Lymphocyte-Activation Gene-3 (LAG-3) inhibitor and the third immune checkpoint inhibitor with clinical application, following PD-1 and CTLA-4. Relatlimab can bind to the LAG-3 receptor which blocks the interaction between LAG-3 and its ligand to reduce LAG-3 pathway-mediated immunosuppression and promote T-cell proliferation, inducing tumor cell death. On 18 March 2022, the U.S. FDA approved the fixed-dose combination of relatlimab developed by Bristol Myers Squibb with nivolumab, under the brand name Opdualag for the treatment of unresectable or metastatic melanoma in adult and pediatric patients aged 12 and older. This study comprehensively describes the mechanism of action and clinical trials of relatlimab and a brief overview of immune checkpoint drugs currently used for the treatment of melanoma.
Collapse
Affiliation(s)
- Jingjing Su
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Yiting Fu
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Zitong Cui
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Zain Abidin
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Jingsong Yuan
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Xinmiao Zhang
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Runmin Li
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Chunzhen Zhao
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| |
Collapse
|
36
|
Fanfarillo F, Ferraguti G, Lucarelli M, Francati S, Barbato C, Minni A, Ceccanti M, Tarani L, Petrella C, Fiore M. The Impact of ROS and NGF in the Gliomagenesis and their Emerging Implications in the Glioma Treatment. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:449-462. [PMID: 37016521 DOI: 10.2174/1871527322666230403105438] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/19/2022] [Accepted: 02/01/2023] [Indexed: 04/06/2023]
Abstract
Reactive oxygen species (ROS) are highly reactive molecules derived from molecular oxygen (O2). ROS sources can be endogenous, such as cellular organelles and inflammatory cells, or exogenous, such as ionizing radiation, alcohol, food, tobacco, chemotherapeutical agents and infectious agents. Oxidative stress results in damage of several cellular structures (lipids, proteins, lipoproteins, and DNA) and is implicated in various disease states such as atherosclerosis, diabetes, cancer, neurodegeneration, and aging. A large body of studies showed that ROS plays an important role in carcinogenesis. Indeed, increased production of ROS causes accumulation in DNA damage leading to tumorigenesis. Various investigations demonstrated the involvement of ROS in gliomagenesis. The most common type of primary intracranial tumor in adults is represented by glioma. Furthermore, there is growing attention on the role of the Nerve Growth Factor (NGF) in brain tumor pathogenesis. NGF is a growth factor belonging to the family of neurotrophins. It is involved in neuronal differentiation, proliferation and survival. Studies were conducted to investigate NGF pathogenesis's role as a pro- or anti-tumoral factor in brain tumors. It has been observed that NGF can induce both differentiation and proliferation in cells. The involvement of NGF in the pathogenesis of brain tumors leads to the hypothesis of a possible implication of NGF in new therapeutic strategies. Recent studies have focused on the role of neurotrophin receptors as potential targets in glioma therapy. This review provides an updated overview of the role of ROS and NGF in gliomagenesis and their emerging role in glioma treatment.
Collapse
Affiliation(s)
| | - Giampiero Ferraguti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Marco Lucarelli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Silvia Francati
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Antonio Minni
- Department of Sensory Organs, Sapienza University of Rome, Rome, Italy
| | - Mauro Ceccanti
- SITAC, Società Italiana per il Trattamento dell'Alcolismo e le sue Complicanze, Rome, Italy
| | - Luigi Tarani
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Carla Petrella
- Institute of Biochemistry and Cell Biology, IBBC-CNR, Rome, Italy
| | - Marco Fiore
- Institute of Biochemistry and Cell Biology, IBBC-CNR, Rome, Italy
| |
Collapse
|
37
|
Georgescu MT, Trifanescu OG, Serbanescu GL, Mitrica RI, Georgescu DE, Mihaila RI, Neagu A, Gaube A, Botezatu C, Manolescu Mastalier BS. Navigating a Complex Intersection: Immunotherapy and Radiotherapy Synergy in Squamous Cell Carcinoma of the Skin—A Comprehensive Literature Review. COSMETICS 2023; 10:165. [DOI: 10.3390/cosmetics10060165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025] Open
Abstract
Skin squamous cell carcinoma (SCC) represents a major public health concern due to its high incidence and potential for local invasion and metastasis. Compared to local recurrence, metastatic SCC represents an even greater therapeutic challenge. Once distant metastasis occurs, the disease becomes incurable, and treatment focuses on palliation and prolonging survival. The immune microenvironment of SCC is characterized by an infiltration of immune cells, including tumor-infiltrating lymphocytes. In addition to its direct cytotoxic effects, radiotherapy also induces immunomodulatory effects within the tumor microenvironment. Radiation can promote the release of tumor-associated antigens and induce immunogenic cell death, thereby enhancing the recognition of tumor cells by the immune system. Immunotherapy and radiotherapy have emerged as promising therapeutic modalities for metastatic SCC. This literature review aims to evaluate the potential synergy between these treatments and shed light on their combined efficacy. Within the manuscript, we present a compelling case report of a patient with advanced SCC who exhibited resistance to the combined regimen of immunotherapy and radiotherapy, leading to disease progression. Despite the increasing evidence supporting the synergy between these modalities, this case underscores the complex nature of treatment response and the importance of considering individual patient characteristics.
Collapse
Affiliation(s)
- Mihai Teodor Georgescu
- Prof. Dr. Al. Trestioreanu Oncology Discipline, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 022328 Bucharest, Romania
| | - Oana Gabriela Trifanescu
- Prof. Dr. Al. Trestioreanu Oncology Discipline, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 022328 Bucharest, Romania
| | - Georgia Luiza Serbanescu
- Prof. Dr. Al. Trestioreanu Oncology Discipline, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 022328 Bucharest, Romania
| | - Radu Iulian Mitrica
- Prof. Dr. Al. Trestioreanu Oncology Discipline, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 022328 Bucharest, Romania
| | - Dragos Eugen Georgescu
- “Dr. Ion Cantacuzino” Surgery Discipline, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Raluca Ioana Mihaila
- “Prof. Dr. AL. Trestioreanu” Oncology I Department, “Carol Davila” University of Medicine and Pharmacy, 022328 Bucharest, Romania
| | - Alexandra Neagu
- Radiotherapy 2 Department, “Prof. Dr. Al. Trestioreanu” Oncology Institute, 022328 Bucharest, Romania
| | - Alexandra Gaube
- “Prof. Dr. Matei Bals” National Institute of Infectious Diseases, 021105 Bucharest, Romania
| | - Cristian Botezatu
- Colentina Hospital Surgery Discipline, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | | |
Collapse
|
38
|
Scheper J, Hildebrand LS, Faulhaber EM, Deloch L, Gaipl US, Symank J, Fietkau R, Distel LV, Hecht M, Jost T. Tumor-specific radiosensitizing effect of the ATM inhibitor AZD0156 in melanoma cells with low toxicity to healthy fibroblasts. Strahlenther Onkol 2023; 199:1128-1139. [PMID: 36229655 PMCID: PMC10673781 DOI: 10.1007/s00066-022-02009-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/15/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE Despite new treatment options, melanoma continues to have an unfavorable prognosis. DNA damage response (DDR) inhibitors are a promising drug class, especially in combination with chemotherapy (CT) or radiotherapy (RT). Manipulating DNA damage repair during RT is an opportunity to exploit the genomic instability of cancer cells and may lead to radiosensitizing effects in tumors that could improve cancer therapy. METHODS A panel of melanoma-derived cell lines of different origin were used to investigate toxicity-related clonogenic survival, cell death, and cell cycle distribution after treatment with a kinase inhibitor (KI) against ATM (AZD0156) or ATR (VE-822, berzosertib), irradiation with 2 Gy, or a combination of KI plus ionizing radiation (IR). Two fibroblast cell lines generated from healthy skin tissue were used as controls. RESULTS Clonogenic survival indicated a clear radiosensitizing effect of the ATM inhibitor (ATMi) AZD0156 in all melanoma cells in a synergistic manner, but not in healthy tissue fibroblasts. In contrast, the ATR inhibitor (ATRi) VE-822 led to additive enhancement of IR-related toxicity in most of the melanoma cells. Both inhibitors mainly increased cell death induction in combination with IR. In healthy fibroblasts, VE-822 plus IR led to higher cell death rates compared to AZD0156. A significant G2/M block was particularly induced in cancer cells when combining AZD0156 with IR. CONCLUSION ATMi, in contrast to ATRi, resulted in synergistic radiosensitization regarding colony formation in melanoma cancer cells, while healthy tissue fibroblasts were merely affected with respect to cell death induction. In connection with an increased number of melanoma cells in the G2/M phase after ATMi plus IR treatment, ATMi seems to be superior to ATRi in melanoma cancer cell treatments when combined with RT.
Collapse
Affiliation(s)
- Julian Scheper
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054, Erlangen, Germany
| | - Laura S Hildebrand
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054, Erlangen, Germany
| | - Eva-Maria Faulhaber
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054, Erlangen, Germany
| | - Lisa Deloch
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054, Erlangen, Germany
- Translational Radiobiology, Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Udo S Gaipl
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054, Erlangen, Germany
- Translational Radiobiology, Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Julia Symank
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054, Erlangen, Germany
| | - Luitpold V Distel
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054, Erlangen, Germany
| | - Markus Hecht
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054, Erlangen, Germany
| | - Tina Jost
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany.
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054, Erlangen, Germany.
- Translational Radiobiology, Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany.
| |
Collapse
|
39
|
Zhang C, Qin M. Extracellular vesicles targeting tumor microenvironment in ovarian cancer. Int J Biol Macromol 2023; 252:126300. [PMID: 37573911 DOI: 10.1016/j.ijbiomac.2023.126300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/17/2023] [Accepted: 08/10/2023] [Indexed: 08/15/2023]
Abstract
Ovarian cancer (OC) is a prevalent neoplastic condition affecting women. Extracellular vesicles (EVs), nano-sized membrane vesicles, are secreted by various cells in both physiological and pathological states. The profound interplay between EVs and the tumor microenvironment (TME) in ovarian cancer is crucial. In this review, we explores the pivotal role of EVs in facilitating intercellular communication between cancer cells and the TME, emphasizing the potential of EVs as promising diagnostic markers and innovative therapeutic targets for ovarian cancer. The comprehensive analysis outlines the specific mechanisms by which EVs engage in communication with the constituents of the TME, including the modulation of tumor growth through EVs carrying matrix metalloproteinases (MMPs) and EV-mediated inhibition of angiogenesis, among other factors. Additionally, the we discuss the potential clinical applications of EVs that target the TME in ovarian cancer, encompassing the establishment of novel treatment strategies and the identification of novel biomarkers for early detection and prognosis. Finally, this review identifies novel strategies for therapeutic interventions, such as utilizing EVs as carriers for drug delivery and targeting specific EV-mediated signaling pathways. In summary, this manuscript offers valuable insights into the role of EVs in ovarian cancer and highlights the significance of comprehending intercellular communication in the realm of cancer biology.
Collapse
Affiliation(s)
- Chunmei Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, China
| | - Meiying Qin
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, China.
| |
Collapse
|
40
|
Borges MI, Abreu JM, Ramalhosa F, Nogueira S, Corte Real A. Late-Onset Masseteric Metastasis of Malignant Melanoma in a Patient With Neurofibromatosis and Lipomatosis: A Diagnostic Challenge and Case Report. Cureus 2023; 15:e50847. [PMID: 38249267 PMCID: PMC10798650 DOI: 10.7759/cureus.50847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2023] [Indexed: 01/23/2024] Open
Abstract
Melanoma and neurofibromatosis (NF) are distinctly separate conditions, each characterized by unique pathophysiological processes. Nevertheless, their clinical presentations can share overlapping similarities. This report highlights a unique case involving a 68-year-old male with NF1 and lipomatosis, whose unwavering belief that a developing mass in the masseter region was benign and linked to the pre-existing diagnoses contributed to a significant delay in seeking healthcare. Consequently, this postponement resulted in the late diagnosis of disseminated malignant melanoma (stage IV, T4N0M1c). Given the patient's prognosis and poor general health, a palliative treatment plan was devised, entailing the complete excision of the masseteric mass and vertebral radiotherapy. Following a rapid and extensive progression of the cancerous lesions, the patient passed away in a palliative care infirmary four months after surgery. The significance of this case, justified not only by its uncommon presentation and atypical differential diagnosis, highlights the critical necessity of regular follow-up protocols for melanoma patients, particularly those prone to metastasis, while ensuring patient attendance. Furthermore, it underscores the necessity of patient education, particularly in recognizing early signs and symptoms, and timely intervention in cases with complex comorbidities.
Collapse
Affiliation(s)
- Maria Inês Borges
- Department of Stomatology, Clinical and Academic Centre of Coimbra, Coimbra, PRT
| | - João M Abreu
- Faculty of Medicine, Clinical and Academic Centre of Coimbra, Coimbra, PRT
- Department of Stomatology, Clinical and Academic Centre of Coimbra, Coimbra, PRT
| | - Fátima Ramalhosa
- Department of Pathology, Clinical and Academic Centre of Coimbra, Coimbra, PRT
| | - Simão Nogueira
- Department of Stomatology, Clinical and Academic Centre of Coimbra, Coimbra, PRT
| | - Ana Corte Real
- Faculty of Medicine, Clinical and Academic Centre of Coimbra, Coimbra, PRT
| |
Collapse
|
41
|
Shalata W, Attal ZG, Shhadi R, Abu Salman A, Abu Jama A, Shalata S, Halumi K, Yakobson A. Tolerated Re-Challenge of Immunotherapy in a Patient with ICI Associated Myocarditis: A Case Report and Literature Review. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1946. [PMID: 38003995 PMCID: PMC10673034 DOI: 10.3390/medicina59111946] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/28/2023] [Accepted: 11/01/2023] [Indexed: 11/26/2023]
Abstract
Many different types of cancer can be treated with immunotherapy drugs called immune checkpoint inhibitors (ICIs). These drugs have altered the landscape of cancer treatment options since they function by triggering a stronger immune response to malignancy. As expected, ICIs' modification of immune regulatory controls leads to a wide range of organ/gland-specific immune-related side effects. These adverse effects are uncommonly deadly and typically improve by discontinuing treatment or administering corticosteroid drugs. As a result of a number of factors-including a lack of specificity in the clinical presentation, the possibility of overlap with other cardiovascular and general medical illnesses, difficulties in diagnosis, and a general lack of awareness-the true incidence of ICI-associated myocarditis is likely underestimated. Currently, protocols for the surveillance, diagnosis, or treatment of this condition are unclear. Several questions remain unanswered, such as how to best screen for this rare toxin, what tests should be run on patients who are suspected of having it, how to treat myocarditis once it has developed, and who is at most risk. In this article, we provide a case study of ICI-associated myocarditis and explain its key characteristics and treatment options.
Collapse
Affiliation(s)
- Walid Shalata
- The Legacy Heritage Cancer Center & Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Zoé Gabrielle Attal
- Medical School for International Health, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Rajeh Shhadi
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Amjad Abu Salman
- Cardiology Division, Soroka Medical Center, Beer Sheva 84105, Israel
| | - Ashraf Abu Jama
- The Legacy Heritage Cancer Center & Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Sondos Shalata
- Nutrition Unit, Galilee Medical Center, Nahariya 22000, Israel
| | - Kais Halumi
- The Legacy Heritage Cancer Center & Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel
| | - Alexander Yakobson
- The Legacy Heritage Cancer Center & Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| |
Collapse
|
42
|
Fert S, River P, Bondonny L, Cauzinille L. Metastatic extradural melanoma of the lumbar spine in a cat. Vet Med Sci 2023; 9:2393-2398. [PMID: 37656442 PMCID: PMC10650352 DOI: 10.1002/vms3.1248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 07/20/2023] [Accepted: 08/18/2023] [Indexed: 09/02/2023] Open
Abstract
A 7-year-old neutered male Domestic shorthair cat, with a 1.5-year history of left eye enucleation secondary to a diffuse iris malignant melanoma, was evaluated for progressive onset of pelvic limb paresis and ataxia with severe thoracolumbar hyperaesthesia and dysorexia. Neurological examination localised a lesion to the T3-L3 spinal cord segments. Magnetic resonance imaging of the thoracolumbar spine showed a well-defined extradural T1-weighted hyperintense non-contrast-enhancing mass, initially suggesting a potential haemorrhagic component. Exploratory surgery revealed a brownish extradural lumbar mass. Histologic examination concluded to a melanoma, most probably metastatic given the animal's previous medical history. This report highlights the importance of collecting a complete medical history, which can help in obtaining a preliminary differential diagnosis in cats with clinical signs of myelopathy. Although the location of this metastasis is particularly unusual both in human and veterinary medicine, making optimal treatment challenging for neurosurgeon, our increased understanding of immune and tumour cell biology during the past decade is likely to improve the future treatments of feline melanoma and its metastases.
Collapse
Affiliation(s)
- Sabrina Fert
- Department of NeurologyVeterinary Hospital Center FrégisGentillyFrance
| | - Pablo River
- Department of SurgeryVeterinary Hospital Center OnlyvetSaint PriestFrance
| | - Laura Bondonny
- Department of SurgeryVeterinary Hospital Center OnlyvetSaint PriestFrance
| | | |
Collapse
|
43
|
Bai J, Wang H, Li C, Liu L, Wang J, Sun C, Zhang Q. A novel mitochondria-targeting compound exerts therapeutic effects against melanoma by inducing mitochondria-mediated apoptosis and autophagy in vitro and in vivo. ENVIRONMENTAL TOXICOLOGY 2023; 38:2608-2620. [PMID: 37466182 DOI: 10.1002/tox.23896] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 06/17/2023] [Accepted: 07/01/2023] [Indexed: 07/20/2023]
Abstract
Melanoma is the most invasive skin cancer, with a high mortality rate. However, existing therapeutic drugs have side effects, low reactivity, and lead to drug resistance. As the power source in cells, mitochondria play an important role in the survival of cancer cells and are an important target for tumor therapy. This study aimed to develop a new anti-melanoma compound that targets mitochondria, evaluate its effect on the proliferation and metastasis of melanoma cells, and explore its mechanism of action. The novel mitochondria-targeting compound, SCZ0148, was synthesized by modifying the structure of cyanine. Then, A375 and B16 cells were incubated with different concentrations of SCZ0148, and different doses of SCZ0148 were administered to A375 and B16 xenograft zebrafish. The results showed that SCZ0148 targeted mitochondria, had dose- and time-dependent effects on the proliferation of melanoma cell lines, and had no obvious side effects on normal cells. In addition, SCZ0148 induced melanoma cell apoptosis through the reactive oxygen species-mediated mitochondrial pathway of apoptosis and promoted autophagy. SCZ0148 significantly inhibited the migration of melanoma cells via a matrix metalloprotein 9-mediated pathway. Similarly, SCZ0148 inhibited melanoma cell proliferation in a concentration-dependent manner in vivo. In summary, SCZ0148 may be a novel anti-melanoma compound that targets mitochondria.
Collapse
Affiliation(s)
- Jun Bai
- Environmental Health Effects and Risk Assessment Key Laboratory of Luzhou, School of Public Health, Southwest Medical University, Luzhou, China
| | - Hailan Wang
- Environmental Health Effects and Risk Assessment Key Laboratory of Luzhou, School of Public Health, Southwest Medical University, Luzhou, China
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing, China
| | - Chenwen Li
- Environmental Health Effects and Risk Assessment Key Laboratory of Luzhou, School of Public Health, Southwest Medical University, Luzhou, China
| | - Li Liu
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jianv Wang
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Changzhen Sun
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Qingbi Zhang
- Environmental Health Effects and Risk Assessment Key Laboratory of Luzhou, School of Public Health, Southwest Medical University, Luzhou, China
| |
Collapse
|
44
|
Frederiksen H, Ashwath ML. Disseminated Melanoma: A Disappearing Left Ventricular Mass. CASE (PHILADELPHIA, PA.) 2023; 7:456-460. [PMID: 38028384 PMCID: PMC10679527 DOI: 10.1016/j.case.2023.08.003] [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] [Indexed: 12/01/2023]
Abstract
•Melanoma may lead to asymptomatic cardiac metastasis. •Resolution of the intracardiac mass occurred with nonsurgical treatment. •Echocardiography is important for screening for metastatic involvement.
Collapse
Affiliation(s)
- Hunter Frederiksen
- Division of Internal Medicine, Department of Cardiology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Mahi L. Ashwath
- Division of Internal Medicine, Department of Cardiology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| |
Collapse
|
45
|
Li X, He L, Ou Y, Wang S, Hu Y, Niu H. Oxymatrine inhibits melanoma development by modulating the immune microenvironment and targeting the MYC/PD-L1 pathway. Int Immunopharmacol 2023; 124:111000. [PMID: 37788594 DOI: 10.1016/j.intimp.2023.111000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/13/2023] [Accepted: 09/24/2023] [Indexed: 10/05/2023]
Abstract
Oxymatrine, also known as ammothamnine or oxysophoridine, is a natural compound isolated from Sophora flavescens (in Chinese, Kushen), and many previous researchers have characterized its anti-inflammatory, anti-fibrotic and anti-tumor properties. However, the underlying anti-tumor immunological mechanism of oxymatrine remains elusive. In this study, we carried out experiments both in vitro and in vivo and investigated the anti-tumor effect of oxymatrine to inhibit the proliferation and migration of melanoma B16 cells, while promoting apoptosis. Oxymatrine upregulated CD4+ T, CD8+ T and NKT cells, downregulated Treg cells, promoted TNF-α secretion, and successfully modulated the immune microenvironment and ultimately suppressed melanoma development in subcutaneous tumor models established in mice. Evidence from network pharmacology and RNAseq suggested that possible targets of oxymatrine for melanoma treatment included PD-L1 and MYC. We observed oxymatrine inhibited PD-L1 and MYC expression in melanoma cells via qRT-PCR and western blotting analysis, and found MYC potentially regulated PD-L1 to mediate anti-tumor effects. These findings provide insight into the mechanism by which oxymatrine inhibits melanoma and enhances the anti-tumor immune effect. In summary, our study proposes a novel approach to suppress melanoma by targeting the MYC/PD-L1 pathway using oxymatrine, which may develop into a less toxic and more efficient anti-tumor agent for melanoma treatment.
Collapse
Affiliation(s)
- Xin Li
- School of Medicine, Jinan University, Guangzhou, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Ministry of Education, Guangzhou, China; Guangzhou Key Laboratory for Germ-Free Animals and Microbiome Application, Guangzhou, China
| | - Lun He
- School of Medicine, Jinan University, Guangzhou, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Ministry of Education, Guangzhou, China; Guangzhou Key Laboratory for Germ-Free Animals and Microbiome Application, Guangzhou, China
| | - Yanhua Ou
- School of Medicine, Jinan University, Guangzhou, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Ministry of Education, Guangzhou, China; Guangzhou Key Laboratory for Germ-Free Animals and Microbiome Application, Guangzhou, China
| | - Shanshan Wang
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Ministry of Education, Guangzhou, China; Guangzhou Key Laboratory for Germ-Free Animals and Microbiome Application, Guangzhou, China; School of Basic Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Yaqian Hu
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Ministry of Education, Guangzhou, China; Guangzhou Key Laboratory for Germ-Free Animals and Microbiome Application, Guangzhou, China; School of Basic Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Haitao Niu
- School of Medicine, Jinan University, Guangzhou, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Ministry of Education, Guangzhou, China; Guangzhou Key Laboratory for Germ-Free Animals and Microbiome Application, Guangzhou, China; School of Basic Medicine, Yunnan University of Chinese Medicine, Kunming, China.
| |
Collapse
|
46
|
Muccioli S, Brillo V, Varanita T, Rossin F, Zaltron E, Velle A, Alessio G, Angi B, Severin F, Tosi A, D'Eletto M, Occhigrossi L, Falasca L, Checchetto V, Ciaccio R, Fascì A, Chieregato L, Rebelo AP, Giacomello M, Rosato A, Szabò I, Romualdi C, Piacentini M, Leanza L. Transglutaminase Type 2-MITF axis regulates phenotype switching in skin cutaneous melanoma. Cell Death Dis 2023; 14:704. [PMID: 37898636 PMCID: PMC10613311 DOI: 10.1038/s41419-023-06223-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 10/30/2023]
Abstract
Skin cutaneous melanoma (SKCM) is the deadliest form of skin cancer due to its high heterogeneity that drives tumor aggressiveness. Melanoma plasticity consists of two distinct phenotypic states that co-exist in the tumor niche, the proliferative and the invasive, respectively associated with a high and low expression of MITF, the master regulator of melanocyte lineage. However, despite efforts, melanoma research is still far from exhaustively dissecting this phenomenon. Here, we discovered a key function of Transglutaminase Type-2 (TG2) in regulating melanogenesis by modulating MITF transcription factor expression and its transcriptional activity. Importantly, we demonstrated that TG2 expression affects melanoma invasiveness, highlighting its positive value in SKCM. These results suggest that TG2 may have implications in the regulation of the phenotype switching by promoting melanoma differentiation and impairing its metastatic potential. Our findings offer potential perspectives to unravel melanoma vulnerabilities via tuning intra-tumor heterogeneity.
Collapse
Affiliation(s)
- Silvia Muccioli
- Department of Biology, University of Padua, Padua, Italy
- Laboratory of Translational Research, Azienda USL - IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | | | | | - Federica Rossin
- Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
| | | | - Angelo Velle
- Department of Biology, University of Padua, Padua, Italy
| | | | - Beatrice Angi
- Department of Biology, University of Padua, Padua, Italy
| | | | - Anna Tosi
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Manuela D'Eletto
- Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
| | - Luca Occhigrossi
- Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
| | - Laura Falasca
- National Institute for Infectious Diseases IRCCS "Lazzaro Spallanzani", Rome, Italy
| | | | | | - Amelia Fascì
- Department of Biology, University of Padua, Padua, Italy
| | | | | | | | - Antonio Rosato
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padua, Padua, Italy
| | - Ildikò Szabò
- Department of Biology, University of Padua, Padua, Italy
| | | | - Mauro Piacentini
- Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
- National Institute for Infectious Diseases IRCCS "Lazzaro Spallanzani", Rome, Italy
| | - Luigi Leanza
- Department of Biology, University of Padua, Padua, Italy.
| |
Collapse
|
47
|
Huang Y, Li C, Li Z, Wang Q, Huang S, Liu Q, Liang Y. Development and Preclinical Evaluation of [ 68Ga]BMSH as a New Potent Positron Emission Tomography Tracer for Imaging Programmed Death-Ligand 1 Expression. Pharmaceuticals (Basel) 2023; 16:1487. [PMID: 37895958 PMCID: PMC10610256 DOI: 10.3390/ph16101487] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/07/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023] Open
Abstract
Immunotherapy targeting the programmed death-ligand 1 (PD-L1)/programmed cell death protein 1 (PD-1) pathway has shown remarkable efficacy against various cancers, but the overall response rate (ORR) is still low. PD-L1 expression in tumors may predict treatment response to immunotherapy. Indeed, ongoing clinical studies utilize a few PD-L1 radiotracers to assess PD-L1 expression as a predictive biomarker for immunotherapy. Here, we present a novel positron emission tomography (PET) radiotracer called [68Ga]BMSH, which is derived from a small molecule inhibitor specifically targeting the binding site of PD-L1. The inhibitor was modified to optimize its in vivo pharmacokinetic properties and enable chelation of 68Ga. In vitro evaluation revealed [68Ga]BMSH possessed a strong binding affinity, high specificity, and rapid internalization in PD-L1 overexpressing cells. Biodistribution studies showed that PD-L1 overexpressing tumors had an uptake of [68Ga]BMSH at 4.22 ± 0.65%ID/g in mice, while the number was 2.23 ± 0.41%ID/g in PD-L1 low-expressing tumors. Micro-PET/CT imaging of tumor-bearing mice further confirmed that, compared to [18F]FDG, [68Ga]BMSH can specifically identify tumors with varying levels of PD-L1 expression. Our findings suggest that the [68Ga]BMSH is a PD-L1 radioligand with ideal imaging properties, and its further application in the clinical screening of PD-L1 overexpressing tumors may improve ORR for immunotherapy.
Collapse
Affiliation(s)
- Yong Huang
- Department of Nuclear Medicine, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China; (Y.H.); (C.L.); (Z.L.); (Q.W.); (S.H.)
| | - Chengze Li
- Department of Nuclear Medicine, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China; (Y.H.); (C.L.); (Z.L.); (Q.W.); (S.H.)
| | - Zhongjing Li
- Department of Nuclear Medicine, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China; (Y.H.); (C.L.); (Z.L.); (Q.W.); (S.H.)
| | - Qiong Wang
- Department of Nuclear Medicine, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China; (Y.H.); (C.L.); (Z.L.); (Q.W.); (S.H.)
| | - Size Huang
- Department of Nuclear Medicine, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China; (Y.H.); (C.L.); (Z.L.); (Q.W.); (S.H.)
| | - Qi Liu
- International Cancer Center, Shenzhen University School of Medicine, Shenzhen University, Shenzhen 518057, China
- Institute of Biomedical Engineering, Shenzhen Graduate School, Peking University, Shenzhen 518055, China
| | - Ying Liang
- Department of Nuclear Medicine, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China; (Y.H.); (C.L.); (Z.L.); (Q.W.); (S.H.)
| |
Collapse
|
48
|
Xu M, Li S. Nano-drug delivery system targeting tumor microenvironment: A prospective strategy for melanoma treatment. Cancer Lett 2023; 574:216397. [PMID: 37730105 DOI: 10.1016/j.canlet.2023.216397] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 08/30/2023] [Accepted: 09/11/2023] [Indexed: 09/22/2023]
Abstract
Melanoma, the most aggressive form of cutaneous malignancy arising from melanocytes, is frequently characterized by metastasis. Despite considerable progress in melanoma therapies, patients with advanced-stage disease often have a poor prognosis due to the limited efficacy, off-target effects, and toxicity associated with conventional drugs. Nanotechnology has emerged as a promising approach to address these challenges with nanoparticles capable of delivering therapeutic agents specifically to the tumor microenvironment (TME). However, the clinical approval of nanomedicines for melanoma treatment remains limited, necessitating further research to develop nanoparticles with improved biocompatibility and precise targeting capabilities. This comprehensive review provides an overview of the current research on nano-drug delivery systems for melanoma treatment, focusing on liposomes, polymeric nanoparticles, and inorganic nanoparticles. It discusses the potential of these nanoparticles for targeted drug delivery, as well as their ability to enhance the efficacy of conventional drugs while minimizing toxicity. Furthermore, this review emphasizes the significance of interdisciplinary collaboration between researchers from various fields to advance the development of nanomedicines. Overall, this review serves as a valuable resource for researchers and clinicians interested in the potential of nano-drug delivery systems for melanoma treatment and offers insights into future directions for research in this field.
Collapse
Affiliation(s)
- Mengdan Xu
- Department of Hematology and Breast Cancer, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Shenglong Li
- Second Ward of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China; The Liaoning Provincial Key Laboratory of Interdisciplinary Research on Gastrointestinal Tumor Combining Medicine with Engineering, China.
| |
Collapse
|
49
|
Pradhan AK, Modi J, Maji S, Kumar A, Bhoopathi P, Mannangatti P, Guo C, Afosah DK, Mochel MC, Mukhopadhyay ND, Kirkwood JM, Wang XY, Desai UR, Sarkar D, Emdad L, Das SK, Fisher PB. Dual Targeting of the PDZ1 and PDZ2 Domains of MDA-9/Syntenin Inhibits Melanoma Metastasis. Mol Cancer Ther 2023; 22:1115-1127. [PMID: 37721536 DOI: 10.1158/1535-7163.mct-22-0653] [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: 10/07/2022] [Revised: 04/14/2023] [Accepted: 06/27/2023] [Indexed: 09/19/2023]
Abstract
Genome-wide gene expression analysis and animal modeling indicate that melanoma differentiation associated gene-9 (mda-9, Syntenin, Syndecan binding protein, referred to as MDA-9/Syntenin) positively regulates melanoma metastasis. The MDA-9/Syntenin protein contains two tandem PDZ domains serving as a nexus for interactions with multiple proteins that initiate transcription of metastasis-associated genes. Although targeting either PDZ domain abrogates signaling and prometastatic phenotypes, the integrity of both domains is critical for full biological function. Fragment-based drug discovery and NMR identified PDZ1i, an inhibitor of the PDZ1 domain that effectively blocks cancer invasion in vitro and in vivo in multiple experimental animal models. To maximize disruption of MDA-9/Syntenin signaling, an inhibitor has now been developed that simultaneously binds and blocks activity of both PDZ domains. PDZ1i was joined to the second PDZ binding peptide (TNYYFV) with a PEG linker, resulting in PDZ1i/2i (IVMT-Rx-3) that engages both PDZ domains of MDA-9/Syntenin. IVMT-Rx-3 blocks MDA-9/Syntenin interaction with Src, reduces NF-κB activation, and inhibits MMP-2/MMP-9 expression, culminating in repression of melanoma metastasis. The in vivo antimetastatic properties of IVMT-Rx-3 are enhanced when combined with an immune-checkpoint inhibitor. Collectively, our results support the feasibility of engineering MDA-9 dual-PDZ inhibitors with enhanced antimetastatic activities and applications of IVMT-Rx-3 for developing novel therapeutic strategies effectively targeting melanoma and in principle, a broad spectrum of human cancers that also overexpress MDA-9/Syntenin.
Collapse
Affiliation(s)
- Anjan K Pradhan
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Jinkal Modi
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Santanu Maji
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Amit Kumar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Praveen Bhoopathi
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Padmanabhan Mannangatti
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Chunqing Guo
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
- VCU Institute of Molecular Medicine, Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Daniel K Afosah
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Mark C Mochel
- Department of Pathology, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Nitai D Mukhopadhyay
- Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia
| | - John M Kirkwood
- Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Xiang-Yang Wang
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
- VCU Institute of Molecular Medicine, Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Umesh R Desai
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
- VCU Institute of Molecular Medicine, Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
- VCU Institute of Molecular Medicine, Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
- VCU Institute of Molecular Medicine, Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
- VCU Institute of Molecular Medicine, Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| |
Collapse
|
50
|
Zhang M, Zuo Y, Guo J, Yang L, Wang Y, Tan M, Guo X. A novel signature for predicting prognosis and immune landscape in cutaneous melanoma based on anoikis-related long non-coding RNAs. Sci Rep 2023; 13:16332. [PMID: 37770477 PMCID: PMC10539372 DOI: 10.1038/s41598-023-39837-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 07/31/2023] [Indexed: 09/30/2023] Open
Abstract
Anoikis is a unique form of apoptosis associated with vascularization and distant metastasis in cancer. Eliminating anoikis resistance in tumor cells could be a promising target for improving the prognosis of terminal cancer patients. However, current studies have not elaborated on the prognosis effect of anoikis-related long non-coding RNAs (lncRNAs) in cutaneous melanoma. Pre-processed data, including RNA sequences and clinical information, were retrieved from TCGA and GTEx databases. After a series of statistical analyses, anoikis-related lncRNAs with prognostic significance were identified, and a unique risk signature was constructed. Risk scores were further analyzed in relation to the tumor microenvironment, tumor immune dysfunction and exclusion, immune checkpoint genes, and RNA methylation genes. The indicators were also used to predict the potentially sensitive anti-cancer drugs. An anoikis-related lncRNAs risk signature consisting of LINC01711, POLH-AS1, MIR205HG, and LINC02416 was successfully established in cutaneous melanoma. Overall survival and progression-free survival of patients were strongly linked with the risk score, independently of other clinical factors. The low-risk group exhibited a more beneficial immunological profile, was less affected by RNA methylation, and was more sensitive to the majority of anti-cancer drugs, all of which indicated a better prognostic outcome. The 4 hub lncRNAs may be fundamental to studying the mechanism of anoikis in cutaneous melanoma and provide personalized therapy for salvaging drug resistance.
Collapse
Affiliation(s)
- Miao Zhang
- Department of Plastic and Burns Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yuzhi Zuo
- Department of Plastic and Burns Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jian Guo
- Vascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Lushan Yang
- Department of Plastic and Burns Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yizhi Wang
- Department of Plastic and Burns Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Meiyun Tan
- Department of Orthopaedics, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Xing Guo
- Department of Plastic and Burns Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
- Center of Ambulatory Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|