1
|
An Y, Zhao F, Jia H, Meng S, Zhang Z, Li S, Zhao J. Inhibition of programmed cell death by melanoma cell subpopulations reveals mechanisms of melanoma metastasis and potential therapeutic targets. Discov Oncol 2025; 16:62. [PMID: 39832036 PMCID: PMC11747064 DOI: 10.1007/s12672-025-01789-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
Melanoma is an aggressive type of skin cancer that arises from melanocytes, the cells responsible for producing skin pigment. In contrast to non-melanoma skin cancers like basal cell carcinoma and squamous cell carcinoma, melanoma is more invasive. Melanoma was distinguished by its rapid progression, high metastatic potential, and significant resistance to conventional therapies. Although it accounted for a small proportion of skin cancer cases, melanoma accounts for the majority of deaths caused by skin cancer due to its ability to invade deep tissues, adapt to diverse microenvironments, and evade immune responses. These unique features highlighted the challenges of treating melanoma and underscored the importance of advanced tools, such as single-cell sequencing, to unravel its biology and develop personalized therapeutic strategies. Thus, we conducted a single-cell analysis of the cellular composition within melanoma tumor tissues and further subdivided melanoma cells into subpopulations. Through analyzing metabolic pathways, stemness genes, and transcription factors (TFs) among cells in different phases (G1, G2/M, and S) as well as between primary and metastatic foci cells, we investigated the specific mechanisms underlying melanoma metastasis. We also revisited the cellular stemness and temporal trajectories of melanoma cell subpopulations, identifying the core subpopulation as C0 SOD3 + Melanoma cells. Our findings revealed a close relationship between the pivotal C0 SOD3 + Melanoma cells subpopulation and oxidative pathways in metastatic tumor tissues. Additionally, we analyzed prognostically relevant differentially expressed genes (DEGs) within the C0 SOD3 + Melanoma cells subpopulation and built a predictive model associated with melanoma outcomes. We selected the gene IGF1 with the highest coefficient (coef) value for further analysis, and experimentally validated its essential function in the proliferation and invasive metastasis of melanoma. In immune infiltration analysis, we discovered the critical roles played by M1/M2 macrophages in melanoma progression and immune evasion. Furthermore, the development and progression of malignant melanoma were closely associated with various forms of programmed cell death (PCD), including apoptosis, autophagic cell death, ferroptosis, and pyroptosis. Melanoma cells often resisted cell death mechanisms, maintaining their growth by inhibiting apoptosis and evading autophagic cell death. Meanwhile, the induction of ferroptosis and pyroptosis was thought to trigger immune responses that helped suppress melanoma dissemination. A deeper understanding of the relationship between melanoma and PCD pathways provided a critical foundation for developing novel targeted therapies, with the potential to enhance melanoma treatment efficacy. These findings contributed to the development of novel prognostic models for melanoma and shed light on research directions concerning melanoma metastasis mechanisms and therapeutic targets.
Collapse
Affiliation(s)
- Yuepeng An
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Fu Zhao
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Hongling Jia
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Siyu Meng
- Northeast International Hospital, Shenyang, 110180, China
| | - Ziwei Zhang
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Shuxiao Li
- Department of Burns and Plastic Reconstructive Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi Province, China.
- Life Science and Clinical Medicine Research Center, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi Province, China.
| | - Jiusi Zhao
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| |
Collapse
|
2
|
Minowa T, Murata K, Mizue Y, Murai A, Nakatsugawa M, Sasaki K, Tokita S, Kubo T, Kanaseki T, Tsukahara T, Handa T, Sato S, Horimoto K, Kato J, Hida T, Hirohashi Y, Uhara H, Torigoe T. Single-cell profiling of acral melanoma infiltrating lymphocytes reveals a suppressive tumor microenvironment. Sci Transl Med 2024; 16:eadk8832. [PMID: 39630887 DOI: 10.1126/scitranslmed.adk8832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 06/12/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024]
Abstract
Acral lentiginous melanoma (ALM) is the most common melanoma subtype in non-Caucasians. Despite advances in cancer immunotherapy, current immune checkpoint inhibitors remain unsatisfactory for ALM. Hence, we conducted comprehensive immune profiling using single-cell phenotyping with reactivity screening of the T cell receptors of tumor-infiltrating T lymphocytes (TILs) in ALM. Compared with cutaneous melanoma, ALM showed a lower frequency of tumor-reactive CD8 clusters and an enrichment of regulatory T cells with direct tumor recognition ability, suggesting a suppressive immune microenvironment in ALM. Tumor-reactive CD8 TILs showed heterogeneous expression of coinhibitory molecules, including KLRC1 (NKG2A), in subpopulations with therapeutic implications. Overall, our study provides a foundation for enhancing the efficacy of immunotherapy in ALM.
Collapse
Affiliation(s)
- Tomoyuki Minowa
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
- Department of Dermatology, Sapporo Medical University School of Medicine, 060-8543 Sapporo, Hokkaido, Japan
| | - Kenji Murata
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
- Joint Research Center for Immunoproteogenomics, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
| | - Yuka Mizue
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
| | - Aiko Murai
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
| | - Munehide Nakatsugawa
- Department of Pathology, Tokyo Medical University Hachioji Medical Center, 193-0998 Hachioji, Tokyo, Japan
| | - Kenta Sasaki
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
| | - Serina Tokita
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
- Joint Research Center for Immunoproteogenomics, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
| | - Terufumi Kubo
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
| | - Takayuki Kanaseki
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
- Joint Research Center for Immunoproteogenomics, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
| | - Tomohide Tsukahara
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
| | - Toshiya Handa
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
- Department of Dermatology, Sapporo Medical University School of Medicine, 060-8543 Sapporo, Hokkaido, Japan
| | - Sayuri Sato
- Department of Dermatology, Sapporo Medical University School of Medicine, 060-8543 Sapporo, Hokkaido, Japan
| | - Kohei Horimoto
- Department of Dermatology, Sapporo Medical University School of Medicine, 060-8543 Sapporo, Hokkaido, Japan
| | - Junji Kato
- Department of Dermatology, Sapporo Medical University School of Medicine, 060-8543 Sapporo, Hokkaido, Japan
| | - Tokimasa Hida
- Department of Dermatology, Sapporo Medical University School of Medicine, 060-8543 Sapporo, Hokkaido, Japan
| | - Yoshihiko Hirohashi
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
| | - Hisashi Uhara
- Department of Dermatology, Sapporo Medical University School of Medicine, 060-8543 Sapporo, Hokkaido, Japan
| | - Toshihiko Torigoe
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
| |
Collapse
|
3
|
Jiang Y, Bei W, Li W, Huang Y, He S, Zhu X, Zheng L, Xia W, Dong S, Liu Q, Zhang C, Lv S, Xie C, Xiang Y, Liu G. Single-cell transcriptome analysis reveals evolving tumour microenvironment induced by immunochemotherapy in nasopharyngeal carcinoma. Clin Transl Med 2024; 14:e70061. [PMID: 39415331 PMCID: PMC11483602 DOI: 10.1002/ctm2.70061] [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: 05/09/2024] [Revised: 09/28/2024] [Accepted: 10/04/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Combinatory therapeutic strategy containing immunochemotherapy as part of induction therapy components is one of the current trends in the treatment of high-risk metastatic locally advanced nasopharyngeal carcinoma (NPC). However, the mechanism underlying the heterogeneity of response at the single-cell level has not been underexplored. METHODS 18 bulks and 11 single-cell RNA sequencing from paired before-treatment and on-treatment samples in patients with treatment-naive high-risk metastatic locally advanced NPCs were obtained. Following quality control, a total of 87 191 cells were included in the subsequence bioinformatics analysis. RESULTS Immunochemotherapy was associated with on-treatment tumour microenvironment (TME) remodelling, including upregulation of anti-TMEs signatures, downregulation of pro-TMEs signatures, reversing CD8+ T exhaustion, and repolarizing proinflammatory TAMs. For the patients achieving a complete response, the cytotoxic activity of CD8+ T cells was stimulated and more interferon-gamma was provided, which would be the key for TAMs proinflammatory repolarization and eventually promote the CD8+ T cells maturation in turn. Among patients who did not reach complete response, differentiation and hypoxia signatures for endothelial cells were elevated after therapy. These patients exhibited higher levels of immune checkpoint genes in malignant cells at the baseline (before treatment), and decreased tumour antigen presentation activity, which may underlie the resistance mechanism to therapy. CONCLUSIONS This study pictures a map of TME modulation following immunochemotherapy-based combination induction therapy and provides potential future approaches. HIGHLIGHTS Immunochemotherapy remodeled T cell phenotypes. For the patients achieving complete response, more interferon gamma was provided by CD8+ T cells after therapy, which would be the key for TAMs pro-inflammatory repolarization and eventually promote the CD8+ T cells maturation in turns. Among patients who did not reach complete response, malignant cells exhibited higher level of immune checkpoint genes before therapy, and decreased tumor antigen presentation activity, which may underlie the resistance mechanism to therapy.
Collapse
Affiliation(s)
- Yaofei Jiang
- Department of Nasopharyngeal CarcinomaState Key Laboratory of Oncology in South ChinaGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapyGuangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
- Department of Oncologythe First Affiliated Hospital of NanChang UniversityNanChangChina
| | - Weixin Bei
- Department of Nasopharyngeal CarcinomaState Key Laboratory of Oncology in South ChinaGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapyGuangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Wangzhong Li
- Department of Thoracic Surgery and OncologyThe First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory HealthGuangzhouChina
| | - Ying Huang
- Department of RadiotherapySun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Shuiqing He
- Department of Nasopharyngeal CarcinomaState Key Laboratory of Oncology in South ChinaGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapyGuangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Xiaobin Zhu
- Thoracic and GI Malignancies BranchNational Cancer Institute, National Institutes of HealthBethesdaUSA
| | - Lisheng Zheng
- Department of PathologyGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Weixiong Xia
- Department of Nasopharyngeal CarcinomaState Key Laboratory of Oncology in South ChinaGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapyGuangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Shuhui Dong
- Department of Nasopharyngeal CarcinomaState Key Laboratory of Oncology in South ChinaGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapyGuangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Qin Liu
- Department of Nasopharyngeal CarcinomaState Key Laboratory of Oncology in South ChinaGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapyGuangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Chuanrun Zhang
- Department of Nasopharyngeal CarcinomaState Key Laboratory of Oncology in South ChinaGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapyGuangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Shuhui Lv
- Department of Nasopharyngeal CarcinomaState Key Laboratory of Oncology in South ChinaGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapyGuangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Changqing Xie
- Department of PathologyGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Yanqun Xiang
- Department of Nasopharyngeal CarcinomaState Key Laboratory of Oncology in South ChinaGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapyGuangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Guoying Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationDepartment of Radiation OncologyMedical Research CenterSun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| |
Collapse
|
4
|
Alfei S, Zuccari G, Athanassopoulos CM, Domenicotti C, Marengo B. Strongly ROS-Correlated, Time-Dependent, and Selective Antiproliferative Effects of Synthesized Nano Vesicles on BRAF Mutant Melanoma Cells and Their Hyaluronic Acid-Based Hydrogel Formulation. Int J Mol Sci 2024; 25:10071. [PMID: 39337557 PMCID: PMC11432396 DOI: 10.3390/ijms251810071] [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: 09/04/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Cutaneous metastatic melanoma (CMM) is the most aggressive form of skin cancer with a poor prognosis. Drug-induced secondary tumorigenesis and the emergency of drug resistance worsen an already worrying scenario, thus rendering urgent the development of new treatments not dealing with mutable cellular processes. Triphenyl phosphonium salts (TPPSs), in addiction to acting as cytoplasmic membrane disruptors, are reported to be mitochondria-targeting compounds, exerting anticancer effects mainly by damaging their membranes and causing depolarization, impairing mitochondria functions and their DNA, triggering oxidative stress (OS), and priming primarily apoptotic cell death. TPP-based bola amphiphiles are capable of self-forming nanoparticles (NPs) with enhanced biological properties, as commonly observed for nanomaterials. Already employed in several other biomedical applications, the per se selective potent antibacterial effects of a TPP bola amphiphile have only recently been demonstrated on 50 multidrug resistant (MDR) clinical superbugs, as well as its exceptional and selective anticancer properties on sensitive and MDR neuroblastoma cells. Here, aiming at finding new molecules possibly developable as new treatments for counteracting CMM, the effects of this TPP-based bola amphiphile (BPPB) have been investigated against two BRAF mutants CMM cell lines (MeOV and MeTRAV) with excellent results (even IC50 = 49 nM on MeOV after 72 h treatment). With these findings and considering the low cytotoxicity of BPPB against different mammalian non-tumoral cell lines and red blood cells (RBCs, selectivity indexes up to 299 on MeOV after 72 h treatment), the possible future development of BPPB as topical treatment for CMM lesions was presumed. With this aim, a biodegradable hyaluronic acid (HA)-based hydrogel formulation (HA-BPPB-HG) was prepared without using any potentially toxic crosslinking agents simply by dispersing suitable amounts of the two ingredients in water and sonicating under gentle heating. HA-BPPB-HA was completely characterized, with promising outcomes such as high swelling capability, high porosity, and viscous elastic rheological behavior.
Collapse
Affiliation(s)
- Silvana Alfei
- Department of Pharmacy, University of Genoa, Viale Cembrano, 16148 Genoa, Italy;
| | - Guendalina Zuccari
- Department of Pharmacy, University of Genoa, Viale Cembrano, 16148 Genoa, Italy;
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147 Genoa, Italy
| | | | - Cinzia Domenicotti
- Department of Experimental Medicine (DIMES), University of Genova, Via Alberti L.B., 16132 Genoa, Italy;
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Barbara Marengo
- Department of Experimental Medicine (DIMES), University of Genova, Via Alberti L.B., 16132 Genoa, Italy;
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| |
Collapse
|
5
|
Valdez CN, Sánchez-Zuno GA, Osmani L, Ibrahim W, Galan A, Bacchiocchi A, Halaban R, Kulkarni RP, Kang I, Bucala R, Tran T. Prognostic and therapeutic insights into MIF, DDT, and CD74 in melanoma. Oncotarget 2024; 15:507-520. [PMID: 39028303 PMCID: PMC11259151 DOI: 10.18632/oncotarget.28615] [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: 05/16/2024] [Accepted: 07/08/2024] [Indexed: 07/20/2024] Open
Abstract
Macrophage Migration Inhibitory Factor (MIF) and its homolog D-dopachrome Tautomerase (DDT) have been implicated as drivers of tumor progression across a variety of cancers. Recent evidence suggests MIF as a therapeutic target in immune checkpoint inhibition (ICI) resistant melanomas, however clinical evidence of MIF and particularly of DDT remain limited. This retrospective study analyzed 97 patients treated at Yale for melanoma between 2002-2020. Bulk-RNA sequencing of patient tumor samples from the Skin Cancer SPORE Biorepository was used to evaluate for differential gene expression of MIF, DDT, CD74, and selected inflammatory markers, and gene expression was correlated with patient survival outcomes. Our findings revealed a strong correlation between MIF and DDT levels, with no statistically significant difference across common melanoma mutations and subtypes. Improved survival was associated with lower MIF and DDT levels and higher CD74:MIF and CD74:DDT levels. High CD74:DDT and CD74:MIF levels were also associated with enrichment of infiltrating inflammatory cell markers. These data suggest DDT as a novel target in immune therapy. Dual MIF and DDT blockade may provide synergistic responses in patients with melanoma, irrespective of common mutations, and may overcome ICI resistance. These markers may also provide prognostic value for further biomarker development.
Collapse
Affiliation(s)
| | | | - Lais Osmani
- Department of Medicine, Section of Rheumatology, Allergy and Immunology, Yale University, New Haven, CT 06520, USA
| | - Wael Ibrahim
- Department of Dermatology, Yale University, New Haven, CT 06520, USA
| | - Anjela Galan
- Department of Dermatology, Yale University, New Haven, CT 06520, USA
| | | | - Ruth Halaban
- Department of Dermatology, Yale University, New Haven, CT 06520, USA
| | - Rajan P. Kulkarni
- Department of Dermatology, Oregon Health and Science University, Portland, OR 97239, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
- Department of Veterans Affairs Portland Health Care System, Operative Care Division, U.S. Portland, OR 97239, USA
| | - Insoo Kang
- School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Medicine, Section of Rheumatology, Allergy and Immunology, Yale University, New Haven, CT 06520, USA
| | - Richard Bucala
- School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Medicine, Section of Rheumatology, Allergy and Immunology, Yale University, New Haven, CT 06520, USA
- Yale Cancer Center, Yale University, New Haven, CT 06520, USA
| | - Thuy Tran
- School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Medicine, Section of Medical Oncology, Yale University, New Haven, CT 06520, USA
- Yale Cancer Center, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
6
|
Valdez CN, Sánchez-Zuno GA, Bucala R, Tran TT. Macrophage Migration Inhibitory Factor (MIF) and D-Dopachrome Tautomerase (DDT): Pathways to Tumorigenesis and Therapeutic Opportunities. Int J Mol Sci 2024; 25:4849. [PMID: 38732068 PMCID: PMC11084905 DOI: 10.3390/ijms25094849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Discovered as inflammatory cytokines, MIF and DDT exhibit widespread expression and have emerged as critical mediators in the response to infection, inflammation, and more recently, in cancer. In this comprehensive review, we provide details on their structures, binding partners, regulatory mechanisms, and roles in cancer. We also elaborate on their significant impact in driving tumorigenesis across various cancer types, supported by extensive in vitro, in vivo, bioinformatic, and clinical studies. To date, only a limited number of clinical trials have explored MIF as a therapeutic target in cancer patients, and DDT has not been evaluated. The ongoing pursuit of optimal strategies for targeting MIF and DDT highlights their potential as promising antitumor candidates. Dual inhibition of MIF and DDT may allow for the most effective suppression of canonical and non-canonical signaling pathways, warranting further investigations and clinical exploration.
Collapse
Affiliation(s)
- Caroline Naomi Valdez
- School of Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA; (C.N.V.); (R.B.)
| | - Gabriela Athziri Sánchez-Zuno
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA;
| | - Richard Bucala
- School of Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA; (C.N.V.); (R.B.)
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA;
- Yale Cancer Center, Yale University, 333 Cedar St., New Haven, CT 06510, USA
| | - Thuy T. Tran
- School of Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA; (C.N.V.); (R.B.)
- Yale Cancer Center, Yale University, 333 Cedar St., New Haven, CT 06510, USA
- Section of Medical Oncology, Department of Internal Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA
| |
Collapse
|
7
|
Wang L, Shen K, Gao Z, Ren M, Wei C, Yang Y, Li Y, Zhu Y, Zhang S, Ding Y, Zhang T, Li J, Zhu M, Zheng S, Yang Y, Du S, Wei C, Gu J. Melanoma Derived Exosomes Amplify Radiotherapy Induced Abscopal Effect via IRF7/I-IFN Axis in Macrophages. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304991. [PMID: 38286661 PMCID: PMC10987102 DOI: 10.1002/advs.202304991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 12/11/2023] [Indexed: 01/31/2024]
Abstract
Radiotherapy (RT) can induce tumor regression outside the irradiation field, known as the abscopal effect. However, the detailed underlying mechanisms remain largely unknown. A tumor-bearing mouse model is successfully constructed by inducing both subcutaneous tumors and lung metastases. Single-cell RNA sequencing, immunofluorescence, and flow cytometry are performed to explore the regulation of tumor microenvironment (TME) by RT. A series of in vitro assays, including luciferase reporter, RNA Pulldown, and fluorescent in situ hybridization (FISH) assays, are performed to evaluate the detailed mechanism of the abscopal effect. In addition, in vivo assays are performed to investigate combination therapy strategies for enhancing the abscopal effect. The results showed that RT significantly inhibited localized tumor and lung metastasis progression and improved the TME. Mechanistically, RT promoted the release of tumor-derived exosomes carrying circPIK3R3, which is taken up by macrophages. circPIK3R3 promoted Type I interferon (I-IFN) secretion and M1 polarization via the miR-872-3p/IRF7 axis. Secreted I-IFN activated the JAK/STAT signaling pathway in CD8+ T cells, and promoted IFN-γ and GZMB secretion. Together, the study shows that tumor-derived exosomes promote I-IFN secretion via the circPIK3R3/miR-872-3p/IRF7 axis in macrophages and enhance the anti-tumor immune response of CD8+ T cells.
Collapse
Affiliation(s)
- Lu Wang
- Department of Plastic SurgeryZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Kangjie Shen
- Department of Plastic SurgeryZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Zixu Gao
- Department of Plastic SurgeryZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Ming Ren
- Department of Plastic SurgeryZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Chenlu Wei
- Department of Plastic SurgeryZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Yang Yang
- Department of Plastic SurgeryZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Yinlam Li
- Department of Plastic SurgeryZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Yu Zhu
- Department of Plastic SurgeryZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Simin Zhang
- Department of Plastic SurgeryShanghai Geriatric Medical CenterShanghai201104P. R. China
| | - Yiteng Ding
- Department of Plastic SurgeryZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Tianyi Zhang
- Department of Plastic SurgeryZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Jianrui Li
- Department of Plastic SurgeryZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Ming Zhu
- Department of Plastic SurgeryZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Shaoluan Zheng
- Department of Plastic SurgeryZhongshan Hospital Xiamen BranchFudan UniversityXiamen361015P. R. China
| | - Yanwen Yang
- Department of Plastic SurgeryZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Shisuo Du
- Department of RadiotherapyZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Chuanyuan Wei
- Department of Plastic SurgeryZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Jianying Gu
- Department of Plastic SurgeryZhongshan HospitalFudan UniversityShanghai200032P. R. China
- Department of Plastic SurgeryZhongshan Hospital Xiamen BranchFudan UniversityXiamen361015P. R. China
| |
Collapse
|
8
|
Zhou H, He X, Huang J, Zhong Y, Zhang L, Ao X, Zhao H, Hu S, Li H, Huang J, Huang H, Liang H. Single-cell sequencing reveals the immune landscape of breast cancer patients with brain metastasis. Thorac Cancer 2024; 15:702-714. [PMID: 38316626 PMCID: PMC10961220 DOI: 10.1111/1759-7714.15243] [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: 01/20/2024] [Accepted: 01/22/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Breast cancer has the highest incidence rate of cancer worldwide, and brain metastases (BrM) are among the most malignant cases. While some patients have benefited from immune checkpoint inhibitors (ICIs), the complex anatomical structure of the brain and the heterogeneity of metastatic tumors have made it difficult to characterize the tumor immune microenvironment (TME) of metastatic tumors. METHODS To address this, we used single-cell RNA sequencing (scRNA-seq) to analyze immune cells in the cerebrospinal fluid (CSF) of BrM patients with breast cancer, thereby providing a comprehensive view of the immune microenvironment landscape of BrM. RESULTS Based on canonical marker genes, we identified nine cell types, and further identified their subtypes through differential expression gene (DEG) analysis. We compared the changes in cells and functions in the immune microenvironment of patients with different prognoses. Our analysis revealed a series of genes that promote tumor immune function (CCR5, LYZ, IGKC, MS4A1, etc.) and inhibit tumor immune function (SCGB2A2, CD24, etc.). CONCLUSIONS The scRNA-seq in CSF provides a noninvasive method to describe the TME of breast cancer patients and guide immunotherapy.
Collapse
Grants
- 202102080096, HL Liang, 201904010331, JQ Huang Guangzhou S&T Project
- 2023A03J0430, HL Liang Guangzhou S&T City and University United Project
- 2022A1515012376, JQ Huang Project Natural Science Foundation of Guangdong Province
- 2021KTSCX091, HL Liang, 2020KTSCX105, JQ Huang Guangdong Provincial Bureau of Education Project
- 20191A011097, HL Liang Guangzhou Health S&T Project
- 202005, HS Li Clinical Key Specialty Project of Guangzhou Medical University
Collapse
Affiliation(s)
- Huaping Zhou
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityGuangzhouChina
| | - Xiang He
- Key Laboratory of Molecular Radiation Oncology Hunan ProvinceXiangya Hospital, Central South UniversityChangshaChina
| | - Jia Huang
- School of Health ManagementGuangzhou Medical UniversityGuangzhouChina
| | - Yumin Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of TechnologyGuangzhouChina
| | - Leyao Zhang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityGuangzhouChina
| | - Xiang Ao
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityGuangzhouChina
| | - Hailin Zhao
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityGuangzhouChina
| | - Su Hu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityGuangzhouChina
| | - Hongsheng Li
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityGuangzhouChina
| | - Jianqing Huang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityGuangzhouChina
- School of Health ManagementGuangzhou Medical UniversityGuangzhouChina
| | - Hongxin Huang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityGuangzhouChina
| | - Hongling Liang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityGuangzhouChina
| |
Collapse
|
9
|
Lim SY, Rizos H. Single-cell RNA sequencing in melanoma: what have we learned so far? EBioMedicine 2024; 100:104969. [PMID: 38241976 PMCID: PMC10831183 DOI: 10.1016/j.ebiom.2024.104969] [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: 10/29/2023] [Revised: 12/18/2023] [Accepted: 01/03/2024] [Indexed: 01/21/2024] Open
Abstract
Over the past decade, there have been remarkable improvements in the treatment and survival rates of melanoma patients. Treatment resistance remains a persistent challenge, however, and is partly attributable to intratumoural heterogeneity. Melanoma cells can transition through a series of phenotypic and transcriptional cell states that vary in invasiveness and treatment responsiveness. The diverse stromal and immune contexture of the tumour microenvironment also contributes to intratumoural heterogeneity and disparities in treatment response in melanoma patients. Recent advances in single-cell sequencing technologies have enabled a more detailed understanding of melanoma heterogeneity and the underlying transcriptional programs that regulate melanoma cell diversity and behaviour. In this review, we examine the concept of intratumoural heterogeneity and the challenges it poses to achieving long-lasting treatment responses. We focus on the significance of next generation single-cell sequencing in advancing our understanding of melanoma diversity and the unique insights gained from single-cell studies.
Collapse
Affiliation(s)
- Su Yin Lim
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Australia; Melanoma Institute Australia, Sydney, Australia.
| | - Helen Rizos
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Australia; Melanoma Institute Australia, Sydney, Australia
| |
Collapse
|
10
|
Carvalho LAD, Aguiar FC, Smalley KSM, Possik PA. Acral melanoma: new insights into the immune and genomic landscape. Neoplasia 2023; 46:100947. [PMID: 37913653 PMCID: PMC10637990 DOI: 10.1016/j.neo.2023.100947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 10/25/2023] [Indexed: 11/03/2023]
Abstract
Acral melanoma is a rare subtype of melanoma that arises on the non-hair bearing skin of the nail bed, palms of the hand and soles of the feet. It is unique among melanomas in not being linked to ultraviolet radiation (UVR) exposure from the sun, and, as such, its incidence is similar across populations who are of Asian, Hispanic, African and European origin. Although research into acral melanoma has lagged behind that of sun-exposed cutaneous melanoma, recent studies have begun to address the unique genetics and immune features of acral melanoma. In this review we will discuss the latest progress in understanding the biology of acral melanoma across different ethnic populations and will outline how these new discoveries can help to guide the therapeutic management of this rare tumor.
Collapse
Affiliation(s)
| | - Flavia C Aguiar
- Division of Basic and Experimental Research, Brazilian National Cancer Institute, Rua Andre Cavalcanti 37, Rio de Janeiro, RJ, 20231-050, Brazil
| | - Keiran S M Smalley
- Department of Tumor Biology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612 USA.
| | - Patricia A Possik
- Division of Basic and Experimental Research, Brazilian National Cancer Institute, Rua Andre Cavalcanti 37, Rio de Janeiro, RJ, 20231-050, Brazil
| |
Collapse
|
11
|
Shen K, Song W, Wang H, Wang L, Yang Y, Hu Q, Ren M, Gao Z, Wang Q, Zheng S, Zhu M, Yang Y, Zhang Y, Wei C, Gu J. Decoding the metastatic potential and optimal postoperative adjuvant therapy of melanoma based on metastasis score. Cell Death Discov 2023; 9:397. [PMID: 37880239 PMCID: PMC10600209 DOI: 10.1038/s41420-023-01678-6] [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: 07/05/2023] [Revised: 09/26/2023] [Accepted: 10/11/2023] [Indexed: 10/27/2023] Open
Abstract
Metastasis is a formidable challenge in the prognosis of melanoma. Accurately predicting the metastatic potential of non-metastatic melanoma (NMM) and determining effective postoperative adjuvant treatments for inhibiting metastasis remain uncertain. In this study, we conducted comprehensive analyses of melanoma metastases using bulk and single-cell RNA sequencing data, enabling the construction of a metastasis score (MET score) through diverse machine-learning algorithms. The reliability and robustness of the MET score were validated using various in vitro assays and in vivo models. Our findings revealed a distinct molecular landscape in metastatic melanoma characterized by the enrichment of metastasis-related pathways, intricate cell-cell communication, and heightened infiltration of pro-angiogenic tumor-associated macrophages compared to NMM. Importantly, patients in the high MET score group exhibited poorer prognoses and an immunosuppressive microenvironment, featuring increased infiltration of regulatory T cells and decreased infiltration of CD8+ T cells, compared to the low MET score patient group. Expression of PD-1 was markedly higher in patients with low MET scores. Anti-PD-1 (aPD-1) therapy profoundly affected antitumor immunity activation and metastasis inhibition in these patients. In summary, our study demonstrates the effectiveness of the MET score in predicting melanoma metastatic potential. For patients with low MET scores, aPD-1 therapy may be a potential treatment strategy to inhibit metastasis. Patients with high MET scores may benefit from combination therapies.
Collapse
Affiliation(s)
- Kangjie Shen
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenyu Song
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hongye Wang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Lu Wang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yang Yang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qianrong Hu
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Min Ren
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zixu Gao
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiangcheng Wang
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Shaoluan Zheng
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, China
| | - Ming Zhu
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanwen Yang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yong Zhang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chuanyuan Wei
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Jianying Gu
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China.
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, China.
| |
Collapse
|
12
|
Augustin RC, Newman S, Li A, Joy M, Lyons M, Pham MP, Lucas P, Smith K, Sander C, Isett B, Davar D, Najjar YG, Zarour HM, Kirkwood JM, Luke JJ, Bao R. Identification of tumor-intrinsic drivers of immune exclusion in acral melanoma. J Immunother Cancer 2023; 11:e007567. [PMID: 37857525 PMCID: PMC10603348 DOI: 10.1136/jitc-2023-007567] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2023] [Indexed: 10/21/2023] Open
Abstract
Acral melanoma (AM) has distinct characteristics as compared with cutaneous melanoma and exhibits poor response to immune checkpoint inhibitors (ICIs). Tumor-intrinsic mechanisms of immune exclusion have been identified in many cancers but less studied in AM. We characterized clinically annotated tumors from patients diagnosed with AM at our institution in correlation with ICI response using whole transcriptome RNAseq, whole exome sequencing, CD8 immunohistochemistry, and multispectral immunofluorescence imaging. A defined interferon-γ-associated T cell-inflamed gene signature was used to categorize tumors into non-T cell-inflamed and T cell-inflamed phenotypes. In combination with AM tumors from two published studies, we systematically assessed the immune landscape of AM and detected differential gene expression and pathway activation in a non-T cell-inflamed tumor microenvironment (TME). Two single-cell(sc) RNAseq AM cohorts and 11 bulk RNAseq cohorts of various tumor types were used for independent validation on pathways associated with lack of ICI response. In total, 892 specimens were included in this study. 72.5% of AM tumors showed low expression of the T cell-inflamed gene signature, with 23.9% of total tumors categorized as the non-T cell-inflamed phenotype. Patients of low CD3+CD8+PD1+ intratumoral T cell density showed poor prognosis. We identified 11 oncogenic pathways significantly upregulated in non-T cell-inflamed relative to T cell-inflamed TME shared across all three acral cohorts (MYC, HGF, MITF, VEGF, EGFR, SP1, ERBB2, TFEB, SREBF1, SOX2, and CCND1). scRNAseq analysis revealed that tumor cell-expressing pathway scores were significantly higher in low versus high T cell-infiltrated AM tumors. We further demonstrated that the 11 pathways were enriched in ICI non-responders compared with responders across cancers, including AM, cutaneous melanoma, triple-negative breast cancer, and non-small cell lung cancer. Pathway activation was associated with low expression of interferon stimulated genes, suggesting suppression of antigen presentation. Across the 11 pathways, fatty acid synthase and CXCL8 were unifying downstream target molecules suggesting potential nodes for therapeutic intervention. A unique set of pathways is associated with immune exclusion and ICI resistance in AM. These data may inform immunotherapy combinations for immediate clinical translation.
Collapse
Affiliation(s)
- Ryan C Augustin
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Medical Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sarah Newman
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Aofei Li
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Marion Joy
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Maureen Lyons
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Mary P Pham
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Peter Lucas
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Katelyn Smith
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Cindy Sander
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Brian Isett
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Diwakar Davar
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yana G Najjar
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Hassane M Zarour
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - John M Kirkwood
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jason John Luke
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Riyue Bao
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
13
|
Augustin RC, Newman S, Li A, Joy M, Lyons M, Pham M, Lucas PC, Smith K, Sander C, Isett B, Davar D, Najjar YG, Zarour HM, Kirkwood JM, Luke JJ, Bao R. Identification of tumor-intrinsic drivers of immune exclusion in acral melanoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.24.554717. [PMID: 37662409 PMCID: PMC10473736 DOI: 10.1101/2023.08.24.554717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Background Acral melanoma (AM) has distinct characteristics as compared to cutaneous melanoma and exhibits poor response to immune checkpoint inhibitors (ICI). Tumor-intrinsic mechanisms of immune exclusion have been identified in many cancers but less studied in AM. Methods We characterized clinically annotated tumors from patients diagnosed with AM at our institution in correlation with ICI response using whole transcriptome RNAseq, whole exome sequencing, CD8 immunohistochemistry, and multispectral immunofluorescence imaging. A defined interferon-γ-associated T cell-inflamed gene signature was used to categorize tumors into non-T cell-inflamed and T cell-inflamed phenotypes. In combination with AM tumors from two published studies, we systematically assessed the immune landscape of AM and detected differential gene expression and pathway activation in a non-T cell-inflamed tumor microenvironment (TME). Two single-cell(sc) RNAseq AM cohorts and 11 bulk RNAseq cohorts of various tumor types were used for independent validation on pathways associated with lack of ICI response. In total, 892 specimens were included in this study. Results 72.5% of AM tumors showed low expression of the T cell-inflamed gene signature, with 23.9% of total tumors categorized as the non-T cell-inflamed phenotype. Patients of low CD3 + CD8 + PD1 + intratumoral T cell density showed poor prognosis. We identified 11 oncogenic pathways significantly upregulated in non-T cell-inflamed relative to T cell-inflamed TME shared across all three acral cohorts (MYC, HGF, MITF, VEGF, EGFR, SP1, ERBB2, TFEB, SREBF1, SOX2, and CCND1). scRNAseq analysis revealed that tumor cell-expressing pathway scores were significantly higher in low vs high T cell-infiltrated AM tumors. We further demonstrated that the 11 pathways were enriched in ICI non-responders compared to responders across cancers, including acral melanoma, cutaneous melanoma, triple-negative breast cancer, and non-small cell lung cancer. Pathway activation was associated with low expression of interferon stimulated genes, suggesting suppression of antigen presentation. Across the 11 pathways, fatty acid synthase and CXCL8 were unifying downstream target molecules suggesting potential nodes for therapeutic intervention. Conclusions A unique set of pathways is associated with immune exclusion and ICI resistance in AM. These data may inform immunotherapy combinations for immediate clinical translation.
Collapse
|
14
|
Wang M, Zeng G, Xiong B, Zhu X, Guo J, Chen D, Zhang S, Luo M, Guo L, Cai L. ALOX5 Promotes Autophagy-dependent Ferroptosis by Activating the AMPK/mTOR Pathway in Melanoma. Biochem Pharmacol 2023; 212:115554. [PMID: 37080437 DOI: 10.1016/j.bcp.2023.115554] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 04/09/2023] [Accepted: 04/11/2023] [Indexed: 04/22/2023]
Abstract
BACKGROUND Melanoma has become more common, and its therapeutic management has remained challenging in recent decades. The purpose of our study is to explore new prognostic therapeutic markers of melanoma and to find new therapeutic methods and therapeutic targets of novel drugs, which have great significance. METHOD First, the arachidonate 5-lipoxygenase (ALOX5) gene associated with both autophagy and ferroptosis was identified by R version 4.2.0. We used human melanoma and para-cancer tissues, human melanoma cell lines, and melanoma-bearing mouse tissues. We used qRT-PCR, Western blotting, immunohistochemistry, immunofluorescence staining, CCK-8, iron ion assay, GSH assay, and MDA assay. In vivo, the ferroptosis activation and antitumor effects of recombinant human ALOX5 protein were evaluated using a xenograft model. RESULT We report that the downregulation of ALOX5 in melanoma is positively correlated with the prognosis of patients and is an independent prognostic factor. Elevated ALOX5 contributes to autophagy and ferroptosis in vitro and in vivo. At the same time, inhibition of autophagy can reduce ferroptosis enhanced by ALOX5, and autophagy and ALOX5 have a synergistic effect. The results of the mechanistic study showed that the increase in ALOX5 could activate the AMPK/mTOR pathway and inhibit GPX4 expression, promoting the occurrence of autophagy-dependent ferroptosis, while the decrease in p-AMPK/AMPK inhibited the occurrence of ferroptosis. CONCLUSION ALOX5 deficiency was resistant to autophagy and ferroptosis by inhibiting the AMPK/mTOR pathway. Therefore, it can provide new targets and methods for melanoma drug development.
Collapse
Affiliation(s)
- Min Wang
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, China; Department of Plastic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Guang Zeng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Bingrui Xiong
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Xiaobin Zhu
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jia Guo
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Danyang Chen
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shanshan Zhang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Min Luo
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University,China.
| | - Liang Guo
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Lin Cai
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
15
|
Li T, Xu Y, Sun W, Yan W, Wang C, Hu T, Zhang X, Luo Z, Liu X, Chen Y. Adjuvant Anti-PD-1 Immunotherapy versus Conventional Therapy for Stage III Melanoma: A Real-World Retrospective Cohort Study. Pharmaceuticals (Basel) 2022; 16:ph16010041. [PMID: 36678538 PMCID: PMC9867270 DOI: 10.3390/ph16010041] [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: 11/29/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/30/2022] Open
Abstract
The use of adjuvant therapy has provided survival benefits in patients with advanced melanoma. This study aimed to explore the recurrence and prognosis of the PD-1 inhibitor, conventional interferon (IFN), or observation (OBS) on resected stage III acral and cutaneous melanoma patients through a retrospective analysis. Patients with resected stage III melanoma at Fudan University Shanghai Cancer Center from 2017 to 2021 were enrolled with all of their clinicopathologic characteristics collected. They were divided into three groups: PD-1 inhibitor, IFN, and OBS. Survival analyses were performed to indicate the significance of different adjuvant therapies. A total of 199 patients were enrolled (PD-1 n = 126; IFN n = 31; and OBS n = 42), with their median follow-up times being 21 months, 24 months, and 49 months, respectively. The PD-1 inhibitor significantly improved relapse-free survival (p = 0.027) and overall survival (p = 0.033) compared with conventional treatment (IFN+OBS). The superiority of the PD-1 inhibitor was witnessed in stage IIIC/D (p = 0.000) acral (p = 0.05) melanoma patients with ulceration (p = 0.011) or lymph node macrometastasis (p = 0.010). The PD-1 inhibitor significantly reduced local recurrence and systemic metastasis compared with conventional therapy (p = 0.002). In conclusion, adjuvant anti-PD-1 immunotherapy can achieve better survival outcomes in acral and cutaneous melanoma patients compared with conventional treatment, without considering adverse events. More clinical benefits were seen in later-stage acral melanoma patients with ulceration or lymph node macrometastasis.
Collapse
Affiliation(s)
- Tong Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Yu Xu
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Wei Sun
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Wangjun Yan
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Chunmeng Wang
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Tu Hu
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Xiaowei Zhang
- Department of Internal Medicine of Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Gastrointestinal Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhiguo Luo
- Department of Internal Medicine of Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Gastrointestinal Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xin Liu
- Department of Internal Medicine of Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Head and Neck and Neuroendocrine Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yong Chen
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Correspondence:
| |
Collapse
|