1
|
Ito T, Tanaka Y, Tanegashima K, Nishio K, Hashimoto H, Ichiki T, Ohno F, Kaku-Ito Y, Nakahara T. KS-NailMel-1: a novel cell line of nail apparatus melanoma. Hum Cell 2025; 38:112. [PMID: 40437181 PMCID: PMC12119781 DOI: 10.1007/s13577-025-01242-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 05/17/2025] [Indexed: 06/01/2025]
Abstract
Nail apparatus melanoma (NAM) is a specific type of cutaneous melanoma that develops in the nail apparatus of the hands and feet. The prognosis for metastatic NAM is poor due to a lack of fully effective systemic therapies. However, the difficulty in obtaining a NAM model has hindered basic research aimed at discovering effective treatment strategies. In this study, we established a NAM cell line, named KS-NailMel-1, from a primary tumor located on the nail apparatus of the left ring finger of a 68-year-old Japanese female. The cells were successfully maintained for over 9 months, exhibiting a doubling time of 38.6 ± 1.94 h. KS-NailMel-1 displayed consistent growth, spheroid formation, and invasiveness, and was confirmed to be identical to the original tumor through short tandem repeat analyses, whole-exome sequencing, and immunohistochemistry. Western blotting of the cells demonstrated the protein expression of NECTIN4, which has recently attracted attention as a potential therapeutic target for melanoma. The KS-NailMel-1 cell line represents a valuable resource for basic and preclinical research on NAM, deepening our understanding of the tumor characteristics and facilitating the development of treatment strategies for this rare form of cancer.
Collapse
Affiliation(s)
- Takamichi Ito
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Yuka Tanaka
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Keiko Tanegashima
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kiichiro Nishio
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hiroki Hashimoto
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Toshio Ichiki
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Fumitaka Ohno
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yumiko Kaku-Ito
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takeshi Nakahara
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
2
|
Prasai A, Baral N, Elajami MK, Vallabhaneni E, Mitchell JD, Iqbal U, Bahmad HF, Welty FK, Elajami TK. Trends of all-cause, melanoma-specific, and cardiovascular mortality in melanoma patients from 2005 to 2020. Cancer Epidemiol 2025; 97:102848. [PMID: 40381435 DOI: 10.1016/j.canep.2025.102848] [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: 02/15/2025] [Revised: 05/11/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND Recent advances in melanoma treatment, including immunotherapy and targeted therapy, have significantly improved survival among melanoma patients after 2010. However, these changes may have influenced mortality trends, including those related to cardiovascular (CV) events. OBJECTIVE In this study, we assess mortality trends including CV mortality in melanoma patients. METHODS Using data from the Surveillance, Epidemiology, and End Results (SEER) 17 registry, we examined melanoma incidence and CV, melanoma-specific, and all-cause mortalities from 2005 to 2020. We utilized Joinpoint software to estimate the annual percentage change (APC). RESULTS Our study included 299,993 melanoma patients (173,889 males; 126,104 females). The incidence of melanoma increased by 0.6 % annually (95 % CI: 0.3-1.5, p < 0.05) from 2005 to 2019, followed by a 6.3 % decrease (95 % CI: -9.9 to -0.7, p < 0.05) in 2020. All-cause mortality increased annually by 8.3 % (95 % CI: 6.7-11.3, p < 0.05) from 2005 to 2010, then by 3.3 % (95 % CI: 2.8-3.7, p < 0.05) after 2010. Melanoma-specific mortality increased by 2.0 % annually (95 % CI: 0.8-3.5, p < 0.05) from 2005 until 2013, after which it declined by 5.1 % (95 % CI: -7.3 to -3.8, p < 0.05). CV mortality increased by 7.1 % annually (95 % CI: 4.6-9.4, p < 0.05) from 2005 to 2020. Trends were similar across sexes, with a non-significant higher APC in CV mortality noted among females from 2017 to 2020. CONCLUSIONS Our study shows despite the significant decrease in all-cause and melanoma-specific mortalities after 2010, likely reflecting the benefits of modern therapies, CV mortality continued to rise. These findings underscore the need for long-term surveillance and CV risk management in melanoma patients.
Collapse
Affiliation(s)
- Astha Prasai
- Department of Internal Medicine, Rochester Regional Health, Rochester, NY, USA
| | - Nischit Baral
- Columbia University Division of Cardiology, Mount Sinai Heart Institute, Miami Beach, FL, USA
| | - Mohamad K Elajami
- Department of Internal Medicine, Hartford Hospital, Hartford, CT, USA
| | - Esha Vallabhaneni
- Columbia University Division of Cardiology, Mount Sinai Heart Institute, Miami Beach, FL, USA
| | - Joshua D Mitchell
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Uzma Iqbal
- Department of Internal Medicine, Rochester Regional Health, Rochester, NY, USA; Department of Cardiology, Sands, Constellation Heart Institute, Rochester Regional Health, Rochester, NY, USA
| | - Hisham F Bahmad
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Francine K Welty
- Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Tarec K Elajami
- Columbia University Division of Cardiology, Mount Sinai Heart Institute, Miami Beach, FL, USA.
| |
Collapse
|
3
|
Corica DA, Bell SD, Miller PJ, Kasperbauer DT, Lawler NJ, Wakefield MR, Fang Y. Into the Future: Fighting Melanoma with Immunity. Cancers (Basel) 2024; 16:4002. [PMID: 39682188 DOI: 10.3390/cancers16234002] [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: 10/31/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Immunotherapy offers a novel and promising option in the treatment of late-stage melanoma. By utilizing the immune system to assist in tumor destruction, patients have additional options after tumor progression. Immune checkpoint inhibitors reduce the ability for tumors to evade the immune system by inhibiting key surface proteins used to inactivate T-cells. Without these surface proteins, T-cells can induce cytotoxic responses against tumors. Tumor infiltrating lymphocyte therapy is a form of adoptive cell therapy that takes advantage of a small subset of T-cells that recognize and infiltrate tumors. Isolation and rapid expansion of these colonies assist the immune system in mounting a charged response that can induce remission. Tumor vaccines deliver a high dose of unique antigens expressed by tumor cells to the entire body. The introduction of large quantities of tumor antigens upregulates antigen presenting cells and leads to effective activation of the immune system against tumors. Cytokine therapy introduces high amounts of chemical messengers that are endogenous to the immune system and support T-cell expansion. While other methods of immunotherapy exist, immune checkpoint inhibitors, tumor infiltrating lymphocytes, tumor vaccines, and cytokine therapy are commonly used to treat melanoma. Like many other cancer treatments, immunotherapy is not without adverse effects, as toxicities represent a major obstacle. However, immunotherapy has been efficacious in the treatment of melanoma.
Collapse
Affiliation(s)
- Derek A Corica
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA
| | - Scott D Bell
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA
| | - Peyton J Miller
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA
| | - Daniel T Kasperbauer
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA
| | - Nicholas J Lawler
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA
| | - Mark R Wakefield
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Yujiang Fang
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
| |
Collapse
|
4
|
Wang J, Wang Y, Jiang X. Targeting anticancer immunity in melanoma tumour microenvironment: unleashing the potential of adjuvants, drugs, and phytochemicals. J Drug Target 2024; 32:1052-1072. [PMID: 39041142 DOI: 10.1080/1061186x.2024.2384071] [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: 05/08/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
Melanoma poses a challenge in oncology because of its aggressive nature and limited treatment modalities. The tumour microenvironment (TME) in melanoma contains unique properties such as an immunosuppressive and high-density environment, unusual vasculature, and a high number of stromal and immunosuppressive cells. In recent years, numerous experiments have focused on boosting the immune system to effectively remove malignant cells. Adjuvants, consisting of phytochemicals, toll-like receptor (TLR) agonists, and cytokines, have shown encouraging results in triggering antitumor immunity and augmenting the therapeutic effectiveness of anticancer therapy. These adjuvants can stimulate the maturation of dendritic cells (DCs) and infiltration of cytotoxic CD8+ T lymphocytes (CTLs). Furthermore, nanocarriers can help to deliver immunomodulators and antigens directly to the tumour stroma, thereby improving their efficacy against malignant cells. The remodelling of melanoma TME utilising phytochemicals, agonists, and other adjuvants can be combined with current modalities for improving therapy outcomes. This review article explores the potential of adjuvants, drugs, and their nanoformulations in enhancing the anticancer potency of macrophages, CTLs, and natural killer (NK) cells. Additionally, the capacity of these agents to repress the function of immunosuppressive components of melanoma TME, such as immunosuppressive subsets of macrophages, stromal and myeloid cells will be discussed.
Collapse
Affiliation(s)
- Jingping Wang
- Emergency Department, Zhejiang Provincial General Hospital of the Chinese People's Armed Police Force, Zhejiang, China
| | - Yaping Wang
- Respiratory and Oncology Department, Zhejiang Provincial General Hospital of the Chinese People's Armed Police Force, Zhejiang, China
| | - Xiaofang Jiang
- Respiratory and Oncology Department, Zhejiang Provincial General Hospital of the Chinese People's Armed Police Force, Zhejiang, China
| |
Collapse
|
5
|
Jia DD, Li T. Comprehensive insights on genetic alterations and immunotherapy prognosis in Chinese melanoma patients. Sci Rep 2024; 14:16607. [PMID: 39025927 PMCID: PMC11258252 DOI: 10.1038/s41598-024-65065-6] [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: 12/16/2023] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
Immune checkpoint inhibitors (ICI) have emerged as a promising therapeutic option for melanoma, which demonstrating improved clinical outcomes in melanoma patients regardless of specific genetic mutations. However, the identification of reliable biomarkers for predicting immunotherapy response and prognosis remains a challenge. In this study, we performed genetic profiling of the melanoma patients with different subtypes and evaluated the efficacy of ICI treatment. A total of 221 melanoma patients were included in our cohort, consisting primarily of acral lentiginous melanoma (ALM), cutaneous malignant melanoma (CMM), and mucosal malignant melanoma (MMM). Genetic analysis revealed BRAF mutations was predominant in CMM and NRAS mutations was prevalent in ALM. Copy number variants (CNVs) and structural variants (SV) were also detected, with CCND1 and CDK4 being the most affected genes in CNV and BRAF, ALK and RAF1 being the druggable targets in SV. Furthermore, NRAS mutations were associated with a poor prognosis in ALM, while TERT mutations were linked to unfavorable outcomes in CMM after receiving PD-1 therapy. Additionally, ALK expression exhibited improved outcomes in both ALM and CMM subtypes. Our study provides a comprehensive genomic and pathological profiling of Chinese melanoma patients, shedding light on the molecular landscape of the disease. Furthermore, numbers of gene mutations and ALK expression were identified as prognostic indicators. These findings contribute to the understanding of melanoma genetics in the Chinese population and have implications for personalized treatment approaches.
Collapse
Affiliation(s)
- Dong-Dong Jia
- Department of Bone and Soft-Tissue Surgery, Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou Institute of Medicine (HIM), No.1 Banshan East Road, Hangzhou, 310022, Zhejiang, China
| | - Tao Li
- Department of Bone and Soft-Tissue Surgery, Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou Institute of Medicine (HIM), No.1 Banshan East Road, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
6
|
Takiwaki M, Umemura H, Kikutani Y, Fukuzawa S, Abe K, Fujino K, Sugihara S, Tachibana K, Morizane S, Satoh M, Nakayama T, Yamasaki O. A method for measuring serum levels of melanin-associated indole metabolites using LC-MS/MS and its application to malignant melanoma. Clin Chim Acta 2024; 557:117873. [PMID: 38493943 DOI: 10.1016/j.cca.2024.117873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 02/29/2024] [Accepted: 03/09/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND AND AIMS With the development of novel therapies for advanced malignant melanoma (MM), biomarkers that can accurately reflect the progression of MM are needed. Serum levels of melanin-related indole metabolites such as 5-hydroxy-6-methoxyindole-2-carboxylic acid (5H6MI2C) and 6-hydroxy-5-methoxyindole-2-carboxylic acid (6H5MI2C) are potential biomarkers for MM. Here, we describe the development of a mass spectrometry (MS)-based assay to determine serum levels of 5H6MI2C and 6H5MI2C. MATERIALS AND METHODS We developed a stable isotope dilution-selective reaction monitoring-MS protocol using liquid chromatography tandem mass spectrometry (LC-MS/MS) to measure human serum 5H6MI2C and 6H5MI2C levels. Analytical evaluations of the method were performed and the method was applied to serum samples from MM patients (n = 81). RESULTS The method established in this study showed high reproducibility and linearity. This novel method also found that serum 6H5MI2C levels were significantly elevated in patients with metastatic MM compared to those with non-metastatic MM. Unfortunately, 5H6MI2C did not show a comparable significant difference. CONCLUSION We successfully established measurement methods for serum 5H6MI2C and 6H5MI2C levels using LC-MS/MS. Serum 6H5MI2C levels offer a potential marker for MM.
Collapse
Affiliation(s)
- Masaki Takiwaki
- Medical Equipment Business Operations, JEOL Ltd., Tokyo, Japan
| | - Hiroshi Umemura
- Division of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan.
| | | | | | - Kentaro Abe
- Medical Equipment Business Operations, JEOL Ltd., Tokyo, Japan
| | - Kiyotaka Fujino
- Medical Equipment Business Operations, JEOL Ltd., Tokyo, Japan
| | - Satoru Sugihara
- Melanoma Center, Okayama University Hospital, Okayama, Japan
| | - Kota Tachibana
- Melanoma Center, Okayama University Hospital, Okayama, Japan
| | - Shin Morizane
- Melanoma Center, Okayama University Hospital, Okayama, Japan
| | - Mamoru Satoh
- Division of Clinical Mass Spectrometry, Chiba University, Chiba, Japan
| | - Tomohiro Nakayama
- Division of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan; Division of Clinical Proteomics, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Osamu Yamasaki
- Melanoma Center, Okayama University Hospital, Okayama, Japan
| |
Collapse
|
7
|
Diaz MJ, Quach J, Song J, Milanovic S, Tran JT, Ladehoff LC, Batchu S, Whitman P, Kaffenberger BH, Montanez-Wiscovich ME. Hypoxic transcriptomes predict survival and tumor-infiltrating immune cell composition in cutaneous melanoma. Melanoma Res 2024; 34:118-124. [PMID: 38329217 DOI: 10.1097/cmr.0000000000000938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Hypoxia has established associations with aggressive tumor phenotypes in many cancers. However, it is not currently understood whether tumor hypoxia levels map to distinct immune infiltrates in cutaneous melanoma, potentially unveiling novel therapeutic targets. To this end, we leveraged a previously identified seven-gene hypoxia signature to grade hypoxia levels of 460 cutaneous melanomas obtained from the Broad Institute GDAC Firehose portal. CIBERSORTx ( https://cibersortx.stanford.edu/ ) was employed to calculate the relative abundance of 22 mature human hematopoietic populations. Clinical outcomes and immune cell associations were assessed by computational means. Results indicated that patients with high-hypoxia tumors reported significantly worse overall survival and correlated with greater Breslow depth, validating the in-silico methodology. High-hypoxia tumors demonstrated increased infiltration of activated and resting dendritic cells, resting mast cells, neutrophils, and resting NK cells, but lower infiltration of gamma-delta T cells. These data suggest that high tumor hypoxia correlates with lower survival probability and distinct population differences of several tumor-infiltrating leukocytes in cutaneous melanomas.
Collapse
Affiliation(s)
| | - Jessica Quach
- Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Joanna Song
- Morsani College of Medicine, University of South Florida, Tampa, Florida
| | | | - Jasmine T Tran
- School of Medicine, Indiana University, Indianapolis, Indiana
| | - Lauren C Ladehoff
- Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Sai Batchu
- Cooper Medical School, Rowan University, Camden, New Jersey
| | | | | | | |
Collapse
|
8
|
Dehghankhold M, Sadat Abolmaali S, Nezafat N, Mohammad Tamaddon A. Peptide nanovaccine in melanoma immunotherapy. Int Immunopharmacol 2024; 129:111543. [PMID: 38301413 DOI: 10.1016/j.intimp.2024.111543] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 01/03/2024] [Accepted: 01/11/2024] [Indexed: 02/03/2024]
Abstract
Melanoma is an especially fatal neoplasm resistant to traditional treatment. The advancement of novel therapeutical approaches has gained attention in recent years by shedding light on the molecular mechanisms of melanoma tumorigenesis and their powerful interplay with the immune system. The presence of many mutations in melanoma cells results in the production of a varied array of antigens. These antigens can be recognized by the immune system, thereby enabling it to distinguish between tumors and healthy cells. In the context of peptide cancer vaccines, generally, they are designed based on tumor antigens that stimulate immunity through antigen-presenting cells (APCs). As naked peptides often have low potential in eliciting a desirable immune reaction, immunization with such compounds usually necessitates adjuvants and nanocarriers. Actually, nanoparticles (NPs) can provide a robust immune response to peptide-based melanoma vaccines. They improve the directing of peptide vaccines to APCs and induce the secretion of cytokines to get maximum immune response. This review provides an overview of the current knowledge of the utilization of nanotechnology in peptide vaccines emphasizing melanoma, as well as highlights the significance of physicochemical properties in determining the fate of these nanovaccines in vivo, including their drainage to lymph nodes, cellular uptake, and influence on immune responses.
Collapse
Affiliation(s)
- Mahvash Dehghankhold
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Samira Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Navid Nezafat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Computational vaccine and Drug Design Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ali Mohammad Tamaddon
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
9
|
Liao C, Yang J, Chen L, Ye Z. Identification of hypoxic-related lncRNAs prognostic model for revealing clinical prognostic and immune infiltration characteristic of cutaneous melanoma. Aging (Albany NY) 2024; 16:3734-3749. [PMID: 38364250 PMCID: PMC10929800 DOI: 10.18632/aging.205556] [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: 09/08/2023] [Accepted: 12/26/2023] [Indexed: 02/18/2024]
Abstract
BACKGROUND Cutaneous melanoma (CM) remains a significant threat to human health. There are clues to the potential role of hypoxia in CM progression. However, the role of hypoxia-related lncRNAs (HRLs) in CM has not been clarified. METHODS We obtained hypoxia related genes from MSigDB database and subsequently identified HRLs by applying TCGA database. LASSO-univariate and multivariate Cox analysis were used to comprehensively analyze the survival characteristics and HRLs expressions, and a novel HRLs-related prognostic risk model was subsequently established for comprehensive analysis. RESULTS The established risk model could evaluate the clinical outcome of CM accurately. The ability of the model-related risk score was also validated as an independent prognostic indicator of CM. Immune infiltration, TMB analysis, drug sensitivity analysis and immunotherapy evaluation were conducted to comprehensively assess the possible causes of the difference in prognosis. The reliability of bioinformatics results was partially verified by RT-qPCR. CONCLUSION We established a new HRLs related risk model and discussed the potential role of hypoxia in the development of CM, which provided a novel basis for CM risk stratification.
Collapse
Affiliation(s)
- Congjuan Liao
- Dermatology and STD Department of The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen and Longgang District People’s Hospital of Shenzhen, Shenzhen 518172, China
| | - Jiabao Yang
- Dermatology and STD Department of The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen and Longgang District People’s Hospital of Shenzhen, Shenzhen 518172, China
| | - Liuting Chen
- Beijing University of Chinese Medicine Shenzhen Hospital (Long Gang), Shenzhen 518116, China
| | - Zhiguang Ye
- Dermatology and STD Department of The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen and Longgang District People’s Hospital of Shenzhen, Shenzhen 518172, China
| |
Collapse
|
10
|
Ito T, Kaku-Ito Y, Ohno F, Nakahara T. A real-world study on the safety profile of extended-interval dosing of immune checkpoint inhibitors for melanoma: a single-center analysis in Japan. Front Med (Lausanne) 2023; 10:1293397. [PMID: 38143437 PMCID: PMC10740208 DOI: 10.3389/fmed.2023.1293397] [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: 09/13/2023] [Accepted: 11/27/2023] [Indexed: 12/26/2023] Open
Abstract
Background Anti-programmed death-1 (PD-1) antibodies are the mainstay for the treatment of unresectable or high-risk melanoma. However, real-world data on the safety profile of their extended-interval doses (EDs) are limited, particularly in Asian patients with melanoma. Materials and methods In this single-center retrospective study, we analyzed the risks of immune-related adverse events (irAEs) among 71 Japanese patients (36 males; mean age, 65.0 years) who received anti-PD-1 monotherapy for melanoma at our institute. Patients who were administered ipilimumab prior to anti-PD-1 monotherapy were excluded. Patients were divided into three groups: canonical-interval dose (CD) group (n = 50, body weight-based dosing or 240 mg Q2W for nivolumab and body weight-based dosing or 200 mg Q3W for pembrolizumab), ED group (n = 14, 480 mg Q4W for nivolumab and 400 mg Q6W for pembrolizumab), and dose-switch (DS) group (n = 7, upfront CD followed by ED). Results The CD group received nivolumab more frequently in the metastatic setting. There were no significant differences in baseline characteristics among the three groups, including in sex, age, primary tumor site, tumor subtype, and follow-up period. irAEs occurred in 36.6% (26 patients) of all patients (32.0% of the CD group, 35.7% of the ED group, and 71.4% of the DS group), while severe (grade ≥ 3) irAEs occurred in only two patients, both of whom were in the CD group. Most of the irAEs occurred during the first 6 months of anti-PD-1 therapy and, interestingly, all of the irAEs in the DS group occurred before the switch (during the CD). There was no significant difference among the three groups in the probability of irAE estimated by the Kaplan-Meier method. Conclusion These findings may highlight the safety of ED of anti-PD-1 monotherapy in the treatment of Asian patients with melanoma.
Collapse
Affiliation(s)
- Takamichi Ito
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | |
Collapse
|
11
|
Jackson-Carroll N, Whisenant M, Crane S, Johnson C. Impact of Immune Checkpoint Inhibitor Therapy on Quality of Life in Patients With Advanced Melanoma: A Systematic Review. Cancer Nurs 2023:00002820-990000000-00189. [PMID: 37976054 DOI: 10.1097/ncc.0000000000001299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) were approved to treat advanced melanoma (AM) because of meaningful clinical benefit. These early data reported that ICI therapy is generally well tolerated, and despite symptoms, patients reported a high global health-related quality of life (HRQOL). OBJECTIVE Immune checkpoint inhibitors are widely used in the oncology community; the aim of this systematic review was to evaluate current data on ICI therapy and its impact on HRQOL of patients with AM. METHODS The Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines were followed during this systematic review to identify and select studies from the PubMed, OVID, EMBASE, and Cochrane databases. Selected studies were downloaded into Covidence and analyzed for trends in how ICI therapy impacts HRQOL in patients with AM. Multiple tools were used to assess the quality of the studies. RESULTS The 16 studies included 12 quantitative, 2 qualitative, and 2 mixed-methods studies. The quality of the studies was moderate (n = 7) or strong (n = 9). Symptoms that impacted HRQOL were fatigue, endocrine dysfunction, rash, diarrhea, cognitive impairment, emotional impact (anxiety and depression), and financial toxicity. Suicidal ideation and 1 attempt were reported in 2 studies, which had not been previously published. CONCLUSION Patient-reported symptoms due to ICI negatively impacted HRQOL. Anxiety and depression are prevalent. Current QOL instruments do not capture the entire patient experience. IMPLICATIONS FOR PRACTICE Patients need to be asked if their symptoms are impacting their HRQOL. Further prospective research is needed to develop or adjust current patient-reported outcome instruments to adequately capture the impact of ICIs on HRQOL.
Collapse
Affiliation(s)
- Natalie Jackson-Carroll
- Author Affiliations: Cizik School of Nursing, The University of Texas Health Science Center at Houston (Drs Jackson-Carroll, Whisenant, Crane, and Johnson); and Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston (Dr Jackson-Carroll)
| | | | | | | |
Collapse
|
12
|
Ito T, Hashimoto H, Kaku-Ito Y, Tanaka Y, Nakahara T. Nail Apparatus Melanoma: Current Management and Future Perspectives. J Clin Med 2023; 12:jcm12062203. [PMID: 36983205 PMCID: PMC10057171 DOI: 10.3390/jcm12062203] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/10/2023] [Accepted: 03/10/2023] [Indexed: 03/16/2023] Open
Abstract
Nail apparatus melanoma (NAM) is a rare type of cutaneous melanoma that belongs to the acral melanoma subtype. NAM is managed principally in accordance with the general treatment for cutaneous melanoma, but there is scarce evidence in support of this in the literature. Acral melanoma is genetically different from non-acral cutaneous melanoma, while recently accumulated data suggest that NAM also has a different genetic background from acral melanoma. In this review, we focus on recent advances in the management of NAM. Localized NAM should be surgically removed; amputation of the digit and digit-preserving surgery have been reported. Sentinel lymph node biopsy can be considered for invasive NAM for the purpose of accurate staging. However, it is yet to be clarified whether patients with metastatic sentinel lymph nodes can be safely spared completion lymph node dissection. Similar to cutaneous melanoma, immune checkpoint inhibitors and BRAF/MEK inhibitors are used as the first-line treatment for metastatic NAM, but data on the efficacy of these therapies remain scarce. The therapeutic effects of immune checkpoint inhibitors could be lower for NAM than for cutaneous melanoma. This review highlights the urgent need to accumulate data to better define the optimal management of this rare melanoma.
Collapse
Affiliation(s)
- Takamichi Ito
- Correspondence: ; Tel.: +81-92-642-5585; Fax: +81-92-642-5600
| | | | | | | | | |
Collapse
|
13
|
Bahreyni A, Mohamud Y, Luo H. Recent advancements in immunotherapy of melanoma using nanotechnology-based strategies. Biomed Pharmacother 2023; 159:114243. [PMID: 36641926 DOI: 10.1016/j.biopha.2023.114243] [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: 12/07/2022] [Revised: 01/07/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
Melanoma is a malignant tumor that accounts for the deadliest form of skin cancers. Despite the significant efforts made recently for development of immunotherapeutic strategies including using immune checkpoint inhibitors and cancer vaccines, the clinical outcomes are unsatisfying. Different factors affect efficient cancer immunotherapy such as side-effects, immunosuppressive tumor microenvironment, and tumor heterogeneity. In the past decades, various nanotechnology-based approaches have been developed to enhance the efficacy of cancer immunotherapy, in addition to diminishing the toxicity associated with it. Several studies have shown that proper application of nanomaterials can revolutionize the outcome of immunotherapy in diverse melanoma models. This review summarizes the recent advancement in the integration of nanotechnology and cancer immunotherapy in melanoma treatment. The importance of nanomaterials and their therapeutic advantages for patients with melanoma are also discussed.
Collapse
Affiliation(s)
- Amirhossein Bahreyni
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Yasir Mohamud
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Honglin Luo
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada.
| |
Collapse
|
14
|
Kaehler M, Cascorbi I. Molecular Mechanisms of Tyrosine Kinase Inhibitor Resistance in Chronic Myeloid Leukemia. Handb Exp Pharmacol 2023; 280:65-83. [PMID: 36882601 DOI: 10.1007/164_2023_639] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
The hematopoietic neoplasm chronic myeloid leukemia (CML) is a rare disease caused by chromosomal reciprocal translocation t(9;22)(q34:q11) with subsequent formation of the BCR-ABL1 fusion gene. This fusion gene encodes a constitutively active tyrosine kinase, which results in malignant transformation of the cells. Since 2001, CML can be effectively treated using tyrosine kinase inhibitors (TKIs) such as imatinib, which prevent phosphorylation of downstream targets by blockade of the BCR-ABL kinase. Due to its tremendous success, this treatment became the role model of targeted therapy in precision oncology. Here, we review the mechanisms of TKI resistance focusing on BCR-ABL1-dependent and -independent mechanisms. These include the genomics of the BCR-ABL1, TKI metabolism and transport and alternative signaling pathways.
Collapse
Affiliation(s)
- Meike Kaehler
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Ingolf Cascorbi
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Kiel, Germany.
| |
Collapse
|
15
|
Li M, Long X, Bu W, Zhang G, Deng G, Liu Y, Su J, Huang K. Immune-related risk score: An immune-cell-pair-based prognostic model for cutaneous melanoma. Front Immunol 2023; 14:1112181. [PMID: 36875110 PMCID: PMC9975150 DOI: 10.3389/fimmu.2023.1112181] [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: 11/30/2022] [Accepted: 01/26/2023] [Indexed: 02/17/2023] Open
Abstract
Background Melanoma is among the most malignant immunologic tumor types and is associated with high mortality. However, a considerable number of melanoma patients cannot benefit from immunotherapy owing to individual differences. This study attempts to build a novel prediction model of melanoma that fully considers individual differences in the tumor microenvironment. Methods An immune-related risk score (IRRS) was constructed based on cutaneous melanoma data from The Cancer Genome Atlas (TCGA). Single-sample gene set enrichment analysis (ssGSEA) was used to calculate immune enrichment scores of 28 immune cell signatures. We performed pairwise comparisons to obtain scores for cell pairs based on the difference in the abundance of immune cells within each sample. The resulting cell pair scores, in the form of a matrix of relative values of immune cells, formed the core of the IRRS. Results The area under the curve (AUC) for the IRRS was over 0.700, and when the IRRS was combined with clinical information, the AUC reached 0.785, 0.817, and 0.801 for the 1-, 3-, and 5-year survival, respectively. Differentially expressed genes between the two groups were enriched in staphylococcal infection and estrogen metabolism pathway. The low IRRS group showed a better immunotherapeutic response and exhibited more neoantigens, richer T-cell receptor and B-cell receptor diversity, and higher tumor mutation burden. Conclusion The IRRS enables a good prediction of prognosis and immunotherapy effect, based on the difference in the relative abundance of different types of infiltrating immune cells, and could provide support for further research in melanoma.
Collapse
Affiliation(s)
- Mingjia Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China.,Hunan Engineering Research Center of Skin Health and Disease, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Department of Dermatology, Peking University First Hospital, Peking University, Beijing, China
| | - Xinrui Long
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China.,Hunan Engineering Research Center of Skin Health and Disease, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wenbo Bu
- Department of Dermatological Surgery, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences, Peking Union Medical College, Nanjing, China
| | - Guanxiong Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China.,Hunan Engineering Research Center of Skin Health and Disease, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China.,Hunan Engineering Research Center of Skin Health and Disease, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yuancheng Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China.,Hunan Engineering Research Center of Skin Health and Disease, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Juan Su
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China.,Hunan Engineering Research Center of Skin Health and Disease, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Kai Huang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China.,Hunan Engineering Research Center of Skin Health and Disease, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
16
|
De Sousa-Coelho AL, Aureliano M, Fraqueza G, Serrão G, Gonçalves J, Sánchez-Lombardo I, Link W, Ferreira BI. Decavanadate and metformin-decavanadate effects in human melanoma cells. J Inorg Biochem 2022; 235:111915. [PMID: 35834898 DOI: 10.1016/j.jinorgbio.2022.111915] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/21/2022] [Accepted: 07/03/2022] [Indexed: 10/17/2022]
Abstract
Decavanadate is a polyoxometalate (POMs) that has shown extensive biological activities, including antidiabetic and anticancer activity. Importantly, vanadium-based compounds as well as antidiabetic biguanide drugs, such as metformin, have shown to exert therapeutic effects in melanoma. A combination of these agents, the metformin-decavanadate complex, was also recognized for its antidiabetic effects and recently described as a better treatment than the monotherapy with metformin enabling lower dosage in rodent models of diabetes. Herein, we compare the effects of decavanadate and metformin-decavanadate on Ca2+-ATPase activity in sarcoplasmic reticulum vesicles from rabbit skeletal muscles and on cell signaling events and viability in human melanoma cells. We show that unlike the decavanadate-mediated non-competitive mechanism, metformin-decavanadate inhibits Ca2+-ATPase by a mixed-type competitive-non-competitive inhibition with an IC50 value about 6 times higher (87 μM) than the previously described for decavanadate (15 μM). We also found that both decavanadate and metformin-decavanadate exert antiproliferative effects on melanoma cells at 10 times lower concentrations than monomeric vanadate. Western blot analysis revealed that both, decavanadate and metformin-decavanadate increased phosphorylation of extracellular signal-regulated kinase (ERK) and serine/threonine protein kinase AKT signaling proteins upon 24 h drug exposure, suggesting that the anti-proliferative activities of these compounds act independent of growth-factor signaling pathways.
Collapse
Affiliation(s)
- Ana Luísa De Sousa-Coelho
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve, Faro, Portugal; Algarve Biomedical Center (ABC), Faro, Portugal; Escola Superior de Saúde (ESS), Universidade do Algarve, Faro, Portugal.
| | - Manuel Aureliano
- Faculdade de Ciências e Tecnologia (FCT), Universidade do Algarve, Faro, Portugal; Centro de Ciências do Mar (CCMar), Universidade do Algarve, Faro, Portugal.
| | - Gil Fraqueza
- Centro de Ciências do Mar (CCMar), Universidade do Algarve, Faro, Portugal; Instituto Superior de Engenharia (ISE), Universidade do Algarve, Faro, Portugal
| | - Gisela Serrão
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve, Faro, Portugal
| | - João Gonçalves
- Faculdade de Medicina e Ciências Biomédicas, Universidade do Algarve, Faro, Portugal
| | - Irma Sánchez-Lombardo
- División Académica de Ciencias Básicas, Universidad Juárez Autónoma de Tabasco, Cunduacán, Mexico
| | - Wolfgang Link
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM). Madrid, Spain
| | - Bibiana I Ferreira
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve, Faro, Portugal; Algarve Biomedical Center (ABC), Faro, Portugal; Faculdade de Medicina e Ciências Biomédicas, Universidade do Algarve, Faro, Portugal
| |
Collapse
|
17
|
Marconcini R, Fava P, Nuzzo A, Manacorda S, Ferrari M, De Rosa F, De Tursi M, Tanda ET, Consoli F, Minisini A, Pimpinelli N, Morgese F, Bersanelli M, Tucci M, Saponara M, Parisi A, Ocelli M, Bazzurri S, Massaro G, Morganti R, Ciardetti I, Stanganelli I. Comparison Between First Line Target Therapy and Immunotherapy in Different Prognostic Categories of BRAF Mutant Metastatic Melanoma Patients: An Italian Melanoma Intergroup Study. Front Oncol 2022; 12:917999. [PMID: 36046043 PMCID: PMC9421680 DOI: 10.3389/fonc.2022.917999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundBRAF and MEK inhibitors target therapies (TT) and AntiPD1 immunotherapies (IT) are available first-line treatments for BRAF v600 mutant metastatic melanoma patients. ECOG PS (E), baseline LDH (L), and baseline number of metastatic sites (N) are well-known clinical prognostic markers that identify different prognostic categories of patients. Direct comparison between first-line TT and IT in different prognostic categories could help in first line treatment decision.MethodsThis is a retrospective analysis conducted in 14 Italian centers on about 454 metastatic melanoma patients, divided in 3 groups: group A—patients with E = 0, L within normal range, and N less than 3; group B—patients not included in group A or C; group C—patients with E > 0, L over the normal range, and N more than 3. For each prognostic group, we compared TT and IT in terms of progression free survival (PFS), overall survival (OS), and disease control rate (DCR).ResultsIn group A, results in 140 TT and 36 IT-treated patients were, respectively, median PFS 35.5 vs 11.6 months (HR (95% CI) 1.949 (1.180–3.217) p value 0.009); median OS not reached vs 55 months (HR (95% CI) 1.195 (0.602–2.373) p value 0.610); DCR 99% vs 75% p value <0.001). In group B, results in 196 TT and 38 IT-treated patients were, respectively, median PFS 11.5 vs 5 months (HR 1.535 (1.036–2.275) p value 0.033); median OS 19 vs 20 months (HR 0.886 (0.546–1.437) p value 0.623); DCR 85% vs 47% p value <0.001). In group C, results in 41 TT and 3 IT-treated patients were, respectively, median PFS 6.4 vs 1.8 months (HR 4.860 (1.399–16) p value 0.013); median OS 9 vs 5 months (HR 3.443 (0.991–11.9) p value 0.052); DCR 66% vs 33% p value 0.612).ConclusionsIn good prognosis, group A—TT showed statistically significant better PFS than IT, also in a long-term period, suggesting that TT can be a good first line option for this patient category. It is only in group B that we observed a crossing of the survival curves after the 3rd year of observation in favor of IT. Few patients were enrolled in group C, so few conclusions can be made on it.
Collapse
Affiliation(s)
- Riccardo Marconcini
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
- *Correspondence: Riccardo Marconcini,
| | - Paolo Fava
- Struttura Complessa (S.C.) Dermatologia Azienda Ospedaliero Universitaria (AOU) Città della Salute e della Scienza di Torino, Torino, Italy
| | - Amedeo Nuzzo
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Simona Manacorda
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Marco Ferrari
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Francesco De Rosa
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Romagnolo per lo studio dei Tumori “Dino Amadori”, Meldola, Italy
| | - Michele De Tursi
- Dipartimento di Tecnologie Innovative in Medicina & Odontoiatria Sezione di Oncologia Università G. D’Annunzio Chieti-Pescara, Chieti-Pescara, Italy
| | - Enrica Teresa Tanda
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, Genoa, Italy
- Genetics of Rare Cancers, Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Genoa, Italy
| | - Francesca Consoli
- Unitá Operativa (U.O.) Oncologia Medica, Azienda Socio Sanitaria Territoriale (ASST) Spedali Civili, Brescia, Italy
| | - Alessandro Minisini
- Dipartimento di Oncologia Azienda Sanitaria Universitaria del Friuli Centrale P.le Santa Maria (SM) della Misericordia, Udine, Italy
| | - Nicola Pimpinelli
- Dipartimento Di Scienze Della Salute (DSS), Sezione Dermatologia, Università di Firenze, Melanoma & Skin Cancer Unit Area Vasta Centro, Firenze, Italy
| | - Francesca Morgese
- Clinica Oncologica, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I, G.M. Lancisi, G. Salesi di Ancona, Ancona, Italy
| | - Melissa Bersanelli
- Unità Operativa di Oncologia Medica, Azienda Ospedaliero-Universitaria di Parma e Dipartimento di Medicina e Chirurgia, Università degli Studi di Parma, Parma, Italy
| | - Marco Tucci
- Medical Oncology Unit, Department of interdisciplinary Medicine (DIM), University of Bari ‘Aldo Moro’, Bari, Italy
| | | | | | | | - Serena Bazzurri
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Giulia Massaro
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | | | - Isabella Ciardetti
- Dipartimento Di Scienze Della Salute (DSS), Sezione Dermatologia, Università di Firenze, Melanoma & Skin Cancer Unit Area Vasta Centro, Firenze, Italy
| | - Ignazio Stanganelli
- Skin Cancer Unit, Scientific Institute of Romagna for the Study of Cancer, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto per La Ricerca Scientifica e Tecnologica (IRST), Meldola, Italy
- Department of Dermatology, University of Parma, Parma, Italy
| |
Collapse
|
18
|
Schauer F, Rafei-Shamsabadi D, Mai S, Mai Y, Izumi K, Meiss F, Kiritsi D. Hemidesmosomal Reactivity and Treatment Recommendations in Immune Checkpoint Inhibitor-Induced Bullous Pemphigoid-A Retrospective, Monocentric Study. Front Immunol 2022; 13:953546. [PMID: 35936009 PMCID: PMC9355658 DOI: 10.3389/fimmu.2022.953546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Immune checkpoint inhibitors (ICI) induce T-cell-mediated antitumour responses. While ICI were initially successfully applied in metastasized melanoma, they are now approved for several tumour entities. Numerous autoimmune disorders have been reported to occur as adverse events of the treatment, among them bullous pemphigoid (BP), with less than 1% of the patients experiencing ICI-induced BP. This number is higher than the estimated prevalence of autoimmune bullous diseases in the general population of Germany, which lies around 0.05%. We here describe our cohort of eight patients, who developed a bullous pemphigoid under or shortly after ICI treatment. Half of them had a severe subtype (as shown by BPDAI >57) and showed a median onset of ICI-BP after 10 months of ICI initiation. Six patients had a palmar and/or plantar involvement, while oral involvement occurred in one case. All patients had linear epidermal IgG depositions in split skin in the indirect immunofluorescence. In four out of five biopsies available for direct immunofluorescence, linear IgG and C3 depositions were detected at the basement membrane, while one patient showed linear IgM staining. Moderate to high levels of FLBP180 autoantibodies were found in seven of eight cases. The disease can still be active after ICI discontinuation, while rituximab might be required for remission. Finally, four tumour samples were stained histochemically for collagen XVII (BP180), but no enhanced expression was found.
Collapse
Affiliation(s)
- Franziska Schauer
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - David Rafei-Shamsabadi
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Shoko Mai
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yosuke Mai
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kentaro Izumi
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Frank Meiss
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Dimitra Kiritsi
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany
| |
Collapse
|
19
|
Zheng W, Sun G, Li Z, Wu F, Sun G, Cao H, Zhou J, Ma Y. The Effect of Anlotinib Combined with anti-PD-1 in the Treatment of Gastric Cancer. Front Surg 2022; 9:895982. [PMID: 35495754 PMCID: PMC9039330 DOI: 10.3389/fsurg.2022.895982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 03/29/2022] [Indexed: 12/02/2022] Open
Abstract
Background Protein tyrosine kinase (PTK) signaling pathway has been confirmed to be involved in the proliferation, differentiation and migration of tumor cells. Anlotinib, as a multi-target tyrosine kinase inhibitor, which can inhibit the expression of vascular endothelial growth factor receptor (VEGFR), has been confirmed to have significant therapeutic effects on non-small cell lung cancer, medullary thyroid carcinoma, and soft tissue sarcoma, but the therapeutic effect on gastric cancer (GC) is still unclear. Methods Anlotinib was screened out of 880 drugs through Cell Counting Kit 8 (CCK-8) technology. TCGA was used to detect the expression of VEGFR in GC, and Kaplan-Meier Plotter was used to analyze the correlation between the expression of VEGFR and the survival rate of GC patients. The impacts exerted by anlotinib to GC cell proliferating, migrating and invading processes were assessed through wound healing assay, transwell assay, and proliferation assay in vitro. In vivo experiments of GC were performed in C57/B6 mouse model to evaluate the function of anlotinib and PD-1 antibody. Results It was found from more than compunds that anlotinib has a significant inhibitory effect on GC cells. In vitro experiments show that anlotinib can significantly inhibit the proliferation, invasion and proliferation of GC cells. The expression level of VEGFR is related to the prognosis and survival of GC. GC patients with low expression of VEGFR have better survival. Anlotinib can inhibit the expression of PD-L1, and achieve better therapeutic effects after combined with PD-1 antibody. Conclusion The present study reveals that anlotinib down regulates PD-L1. The combination of anlotinib and PD-1 monoclonal antibody is beneficial to GC therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Hongyong Cao
- Correspondence: Hongyong Cao ; Jin Zhou ; Yong Ma
| | - Jin Zhou
- Correspondence: Hongyong Cao ; Jin Zhou ; Yong Ma
| | - Yong Ma
- Correspondence: Hongyong Cao ; Jin Zhou ; Yong Ma
| |
Collapse
|
20
|
Del Sordo R, Volta U, Lougaris V, Parente P, Sidoni A, Facchetti M, Bassotti G, Carosi I, Clemente C, Villanacci V. Histological Features of Celiac-Disease-like Conditions Related to Immune Checkpoint Inhibitors Therapy: A Signal to Keep in Mind for Pathologists. Diagnostics (Basel) 2022; 12:395. [PMID: 35204486 PMCID: PMC8871268 DOI: 10.3390/diagnostics12020395] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/19/2022] [Accepted: 02/02/2022] [Indexed: 11/17/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) targeting cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), programmed cell death protein (PD-1), and its ligand PDL-1, are finding increasing application in the treatment of malignant neoplasms. The widespread clinical use of these drugs, however, resulted in the discovery of side effects. The occurrence of celiac disease (CD) after ICIs therapy has been reported in the literature, but its incidence remains unknown and the role of ICIs in its onset is not yet clear. In this review, we examine the published data on this topic in order to better understand and define this entity from a histological point of view. We performed an electronic literature search to identify original reports in which CD or pathological CD-like conditions were documented histologically in patients treated with ICIs. We identified ten papers. A total of twenty-five patients were included in these publications, eleven of them receiving a serologic and histological diagnosis of CD, and four a histological diagnosis of CD-like conditions, in which pathogenesis appears to be multifactorial. ICIs can cause a CD-like enteropathy and biopsies with clinical integration are crucial to diagnose this condition. CD rarely has been observed during treatment with ICIs and its morphological aspects are similar to ICIs-CD enteropathy. Moreover, the onset of ICIs-CD may have a distinct immune mechanism compared to classical CD. Thus, the pathologists must make a histological diagnosis of CD with caution and only in adequate clinical and serological context.
Collapse
Affiliation(s)
- Rachele Del Sordo
- Department of Medicine and Surgery, Section of Anatomic Pathology and Histology, Medical School, University of Perugia, 06132 Perugia, Italy; (R.D.S.); (A.S.)
| | - Umberto Volta
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy;
| | - Vassilios Lougaris
- Pediatrics Clinic, Department of Clinical and Experimental Sciences, University of Brescia and Children’s Hospital, ASST-Spedali Civili, 25123 Brescia, Italy;
| | - Paola Parente
- Pathology Unit, Fondazione IRCCS Ospedale Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, FG, Italy; (I.C.); (C.C.)
| | - Angelo Sidoni
- Department of Medicine and Surgery, Section of Anatomic Pathology and Histology, Medical School, University of Perugia, 06132 Perugia, Italy; (R.D.S.); (A.S.)
| | - Mattia Facchetti
- Institute of Pathology, ASST-Spedali Civili Brescia, 25123 Brescia, Italy;
| | - Gabrio Bassotti
- Gastroenterology & Hepatology Section, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy;
| | - Illuminato Carosi
- Pathology Unit, Fondazione IRCCS Ospedale Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, FG, Italy; (I.C.); (C.C.)
| | - Celeste Clemente
- Pathology Unit, Fondazione IRCCS Ospedale Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, FG, Italy; (I.C.); (C.C.)
| | | |
Collapse
|
21
|
Oba U, Koga Y, Hata N, Oda Y, Ohga S. Nivolumab therapy for a pediatric-onset primary intracranial melanoma. Pediatr Int 2022; 64:e14956. [PMID: 35484924 DOI: 10.1111/ped.14956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 07/29/2021] [Accepted: 08/11/2021] [Indexed: 01/05/2023]
Affiliation(s)
- Utako Oba
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuhki Koga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Nobuhiro Hata
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate of School of Medical Science, Kyushu University, Fukuoka, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
22
|
Boussadia Z, Gambardella AR, Mattei F, Parolini I. Acidic and Hypoxic Microenvironment in Melanoma: Impact of Tumour Exosomes on Disease Progression. Cells 2021; 10:3311. [PMID: 34943819 PMCID: PMC8699343 DOI: 10.3390/cells10123311] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/12/2021] [Accepted: 11/19/2021] [Indexed: 12/12/2022] Open
Abstract
The mechanisms of melanoma progression have been extensively studied in the last decade, and despite the diagnostic and therapeutic advancements pursued, malignant melanoma still accounts for 60% of skin cancer deaths. Therefore, research efforts are required to better define the intercellular molecular steps underlying the melanoma development. In an attempt to represent the complexity of the tumour microenvironment (TME), here we analysed the studies on melanoma in acidic and hypoxic microenvironments and the interactions with stromal and immune cells. Within TME, acidity and hypoxia force melanoma cells to adapt and to evolve into a malignant phenotype, through the cooperation of the tumour-surrounding stromal cells and the escape from the immune surveillance. The role of tumour exosomes in the intercellular crosstalk has been generally addressed, but less studied in acidic and hypoxic conditions. Thus, this review aims to summarize the role of acidic and hypoxic microenvironment in melanoma biology, as well as the role played by melanoma-derived exosomes (Mexo) under these conditions. We also present a perspective on the characteristics of acidic and hypoxic exosomes to disclose molecules, to be further considered as promising biomarkers for an early detection of the disease. An update on the use of exosomes in melanoma diagnosis, prognosis and response to treatment will be also provided and discussed.
Collapse
Affiliation(s)
- Zaira Boussadia
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| | - Adriana Rosa Gambardella
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
| | - Fabrizio Mattei
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| | - Isabella Parolini
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| |
Collapse
|
23
|
A Pan-Cancer Analysis of SLC12A5 Reveals Its Correlations with Tumor Immunity. DISEASE MARKERS 2021; 2021:3062606. [PMID: 34630736 PMCID: PMC8495467 DOI: 10.1155/2021/3062606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/05/2021] [Accepted: 09/08/2021] [Indexed: 11/17/2022]
Abstract
Background Solute carrier family 12 member 5 (SLC12A5) has been reported to play an oncogenic role in certain malignancies. Its prognostic roles and immune mechanisms of action in human cancers, however, remain largely unknown. Methods Data derived from TCGA, GEPIA, and TIMER databases were utilized to delve into the expressing patterns, prognostic values, clinical significances, and tumor immunity of SLC12A5 in tumors. Additionally, the association of SLC12A5 expressions with tumor mutation burden (TMB), methyltransferases, and mismatch repairs (MMRs) was also analyzed. Results Herein, we observed that SLC12A5 was significantly overexpressed in various malignancies, and SLC12A5 levels correlated with overall survival, disease-specific survival, and tumor stage of certain cancers. Furthermore, we noticed that SLC12A5 was distinctly associated with methyltransferases, mismatch repair proteins, TMB, and MSI in human cancers. Conclusions SLC12A5 may act as a potential prognostic and immunological biomarker and therapeutic target for human cancers.
Collapse
|
24
|
Qin H, Vlaminck B, Owoyemi I, Herrmann SM, Leung N, Markovic SN. Successful Treatment of Pembrolizumab-Induced Severe Capillary Leak Syndrome and Lymphatic Capillary Dysfunction. Mayo Clin Proc Innov Qual Outcomes 2021; 5:670-674. [PMID: 34195558 PMCID: PMC8240169 DOI: 10.1016/j.mayocpiqo.2021.01.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Although capillary leak syndrome has a high mortality rate, its trigger, diagnosis, and treatment remain a challenge to clinicians because of the poor understanding of its mechanism and lack of treatment guidelines. With the extended use of immune checkpoint inhibitors in modern oncology, immune checkpoint inhibitor–associated immune-related adverse events have also expanded. We present a case of pembrolizumab-induced capillary leak syndrome and lymphatic capillary dysfunction in which the patient had an excellent clinical response to a tailored treatment strategy.
Collapse
Affiliation(s)
- Haixia Qin
- Department of Medicine, Mayo Clinic, Rochester, MN
| | | | - Itunu Owoyemi
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | | | - Nelson Leung
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
25
|
Kaehler M, Cascorbi I. Pharmacogenomics of Impaired Tyrosine Kinase Inhibitor Response: Lessons Learned From Chronic Myelogenous Leukemia. Front Pharmacol 2021; 12:696960. [PMID: 34262462 PMCID: PMC8273252 DOI: 10.3389/fphar.2021.696960] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 06/17/2021] [Indexed: 12/25/2022] Open
Abstract
The use of small molecules became one key cornerstone of targeted anti-cancer therapy. Among them, tyrosine kinase inhibitors (TKIs) are especially important, as they were the first molecules to proof the concept of targeted anti-cancer treatment. Since 2001, TKIs can be successfully used to treat chronic myelogenous leukemia (CML). CML is a hematologic neoplasm, predominantly caused by reciprocal translocation t(9;22)(q34;q11) leading to formation of the so-called BCR-ABL1 fusion gene. By binding to the BCR-ABL1 kinase and inhibition of downstream target phosphorylation, TKIs, such as imatinib or nilotinib, can be used as single agents to treat CML patients resulting in 80 % 10-year survival rates. However, treatment failure can be observed in 20-25 % of CML patients occurring either dependent or independent from the BCR-ABL1 kinase. Here, we review approved TKIs that are indicated for the treatment of CML, their side effects and limitations. We point out mechanisms of TKI resistance focusing either on BCR-ABL1-dependent mechanisms by summarizing the clinically observed BCR-ABL1-mutations and their implications on TKI binding, as well as on BCR-ABL1-independent mechanisms of resistances. For the latter, we discuss potential mechanisms, among them cytochrome P450 implications, drug efflux transporter variants and expression, microRNA deregulation, as well as the role of alternative signaling pathways. Further, we give insights on how TKI resistance could be analyzed and what could be learned from studying TKI resistance in CML in vitro.
Collapse
Affiliation(s)
| | - Ingolf Cascorbi
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
26
|
Bioinformatic Analysis Reveals Central Role for Tumor-Infiltrating Immune Cells in Uveal Melanoma Progression. J Immunol Res 2021; 2021:9920234. [PMID: 34195299 PMCID: PMC8214507 DOI: 10.1155/2021/9920234] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/05/2021] [Accepted: 05/23/2021] [Indexed: 12/13/2022] Open
Abstract
Tumor-infiltrating immune cells are capable of effective cancer surveillance, and their abundance is linked to better prognosis in numerous tumor types. However, in uveal melanoma (UM), extensive immune infiltrate is associated with poor survival. This study aims to decipher the role of different tumor-infiltrating cell subsets in UM in order to identify potential targets for future immunotherapeutic treatment. We have chosen the TCGA-UVM cohort as a training dataset and GSE22138 as a testing dataset by mining publicly available databases. The abundance of 22 immune cell types was estimated using CIBERSORTx. Then, to determine the significance of tumor-infiltrating cell subsets in UM, we built a multicell type prognostic signature, which was validated in the testing cohort. The created signature was built upon the negative prognostic role of CD8+ T cells and M0 macrophages and the positive role of neutrophils. Based on the created signature score, we divided the patients into low- and high-risk groups. Kaplan-Meier, Cox, and ROC analyses demonstrated superior performance of our risk score compared to either clinical or pathologic characteristics of both cohorts. Further, we found the molecular pathways associated with cancer immunoevasion and metastasis to be enriched in the high-risk group, explaining both the lack of adequate immune surveillance despite increased infiltration of CD8+ T cells as well as the higher metastatic potential. Genes associated with tryptophan metabolism (IDO1 and KYNU) and metalloproteinases were among the most differentially expressed between the high- and low-risk groups. Our correlation analyses interpreted in context of published in vitro data strongly suggest the central role of CD8+ T cells in shifting the UM tumor microenvironment towards suppressive and metastasis-promoting. Therefore, we propose further investigations of IDO1 and metalloproteinases as novel targets for immunotherapy in lymphocyte-rich metastatic UM patients.
Collapse
|
27
|
Dalle S, Mortier L, Corrie P, Lotem M, Board R, Arance AM, Meiss F, Terheyden P, Gutzmer R, Buysse B, Oh K, Brokaw J, Le TK, Mathias SD, Scotto J, Lord-Bessen J, Moshyk A, Kotapati S, Middleton MR. Long-term real-world experience with ipilimumab and non-ipilimumab therapies in advanced melanoma: the IMAGE study. BMC Cancer 2021; 21:642. [PMID: 34051732 PMCID: PMC8164785 DOI: 10.1186/s12885-021-08032-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 03/15/2021] [Indexed: 12/14/2022] Open
Abstract
Background Ipilimumab has shown long-term overall survival (OS) in patients with advanced melanoma in clinical trials, but robust real-world evidence is lacking. We present long-term outcomes from the IMAGE study (NCT01511913) in patients receiving ipilimumab and/or non-ipilimumab (any approved treatment other than ipilimumab) systemic therapies. Methods IMAGE was a multinational, prospective, observational study assessing adult patients with advanced melanoma treated with ipilimumab or non-ipilimumab systemic therapies between June 2012 and March 2015 with ≥3 years of follow-up. Adjusted OS curves based on multivariate Cox regression models included covariate effects. Safety and patient-reported outcomes were assessed. Results Among 1356 patients, 1094 (81%) received ipilimumab and 262 (19%) received non-ipilimumab index therapy (systemic therapy [chemotherapy, anti–programmed death 1 antibodies, or BRAF ± MEK inhibitors], radiotherapy, and radiosurgery). In the overall population, median age was 64 years, 60% were male, 78% were from Europe, and 78% had received previous treatment for advanced melanoma. In the ipilimumab-treated cohort, 780 (71%) patients did not receive subsequent therapy (IPI-noOther) and 314 (29%) received subsequent non-ipilimumab therapy (IPI-Other) on study. In the non-ipilimumab–treated cohort, 205 (78%) patients remained on or received other subsequent non-ipilimumab therapy (Other-Other) and 57 (22%) received subsequent ipilimumab therapy (Other-IPI) on study. Among 1151 patients who received ipilimumab at any time during the study (IPI-noOther, IPI-Other, and Other-IPI), 296 (26%) reported CTCAE grade ≥ 3 treatment-related adverse events, most occurring in year 1. Ipilimumab-treated and non-ipilimumab–treated patients who switched therapy (IPI-Other and Other-IPI) had longer OS than those who did not switch (IPI-noOther and Other-Other). Patients with prior therapy who did not switch therapy (IPI-noOther and Other-Other) showed similar OS. In treatment-naive patients, those in the IPI-noOther group tended to have longer OS than those in the Other-Other group. Patient-reported outcomes were similar between treatment cohorts. Conclusions With long-term follow-up (≥ 3 years), safety and OS in this real-world population of patients treated with ipilimumab 3 mg/kg were consistent with those reported in clinical trials. Patient-reported quality of life was maintained over the study period. OS analysis across both pretreated and treatment-naive patients suggested a beneficial role of ipilimumab early in treatment. Trial registration ClinicalTrials.gov, NCT01511913. Registered January 19, 2012 – Retrospectively registered, https://clinicaltrials.gov/ct2/show/NCT01511913 Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08032-y.
Collapse
Affiliation(s)
- Stéphane Dalle
- Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, 69495, Pierre-Bénite, France.
| | - Laurent Mortier
- Université de Lille, INSERM U1189, CHRU Lille, 59037, Lille, France
| | - Pippa Corrie
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB0 2QQ, UK
| | - Michal Lotem
- Hadassah Hebrew University Hospital, 91120, Jerusalem, Israel
| | - Ruth Board
- Royal Preston Hospital, Preston, PR2 9HT, UK
| | | | - Frank Meiss
- Department of Dermatology, Faculty of Medicine, Medical Center - University of Freiburg, 79104, Freiburg, Germany
| | | | - Ralf Gutzmer
- Medizinische Hochschule Hannover, 30625, Hanover, Germany
| | | | - Kelly Oh
- Syneos Health, Morrisville, NC, 27560, USA
| | - Jane Brokaw
- Bristol Myers Squibb, Princeton, NJ, 08543, USA
| | - T Kim Le
- Bristol Myers Squibb, Princeton, NJ, 08543, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Gangliosides as Signaling Regulators in Cancer. Int J Mol Sci 2021; 22:ijms22105076. [PMID: 34064863 PMCID: PMC8150402 DOI: 10.3390/ijms22105076] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/07/2021] [Accepted: 05/07/2021] [Indexed: 02/06/2023] Open
Abstract
At the plasma membrane, gangliosides, a group of glycosphingolipids, are expressed along with glycosphingolipids, phospholipids, and cholesterol in so-called lipid rafts that interact with signaling receptors and related molecules. Most cancers present abnormalities in the intracellular signal transduction system involved in tumor growth, invasion, and metastasis. To date, the roles of gangliosides as regulators of signal transduction have been reported in several cancer types. Gangliosides can be expressed by the exogenous ganglioside addition, with their endogenous expression regulated at the enzymatic level by targeting specific glycosyltransferases. Accordingly, the relationship between changes in the composition of cell surface gangliosides and signal transduction has been investigated by controlling ganglioside expression. In cancer cells, several types of signaling molecules are positively or negatively regulated by ganglioside expression levels, promoting malignant properties. Moreover, antibodies against gangliosides have been shown to possess cytotoxic effects on ganglioside-expressing cancer cells. In the present review, we highlight the involvement of gangliosides in the regulation of cancer cell signaling, and we explore possible therapies targeting ganglioside-expressing cancer.
Collapse
|
29
|
Ryan AL, Burns C, Gupta AK, Samarasekera R, Ziegler DS, Kirby ML, Alvaro F, Downie P, Laughton SJ, Cross S, Hassall T, McCowage GB, Hansford JR, Kotecha RS, Gottardo NG. Malignant Melanoma in Children and Adolescents Treated in Pediatric Oncology Centers: An Australian and New Zealand Children's Oncology Group (ANZCHOG) Study. Front Oncol 2021; 11:660172. [PMID: 33996584 PMCID: PMC8117414 DOI: 10.3389/fonc.2021.660172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/23/2021] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVES Unlike adults, malignant melanoma in children and adolescents is rare. In adult melanoma, significant progress in understanding tumor biology and new treatments, including targeted therapies and immunotherapy have markedly improved overall survival. In sharp contrast, there is a paucity of data on the biology and clinical behavior of pediatric melanoma. We report a national case series of all pediatric and adolescent malignant melanoma presenting to ANZCHOG Childhood Cancer Centers in Australia and New Zealand. METHODS A retrospective, descriptive, multi-center study was undertaken to identify patients less than 18 years of age treated for cutaneous malignant melanoma over a twenty-year period (1994 to 2014). Data on clinical characteristics, histopathology, and extent of disease, treatment and follow-up are described. RESULTS A total of 37 cases of malignant melanoma were identified from all of the Australasian tertiary Childhood Cancer Centers. The median age was 10 years (range 1 month - 17 years). Clinically, the most common type of lesion was pigmented, occurring in sixteen (57%) patients, whilst amelanotic was seen in 7 patients (25%). In 11 (27.9%) the Breslow thickness was greater than 4mm. A total of 11 (29.7%) patients relapsed and 90% of these died of disease. Five-year event free survival (EFS) and overall survival were 63.2 (95% CI: 40.6 - 79.1) and 67.7% (95% CI: 45.1 - 82.6) respectively. CONCLUSION Our data confirms that melanoma is a rare presentation of cancer to tertiary Australasian Childhood Cancer Centers with only 37 cases identified over two decades. Notably, melanoma managed in Childhood Cancer Centers is frequently at an advanced stage, with a high percentage of patients relapsing and the majority of these patients who relapsed died of disease. This study confirms previous clinical and prognostic information to support the early multidisciplinary management in Childhood Cancer Centers, in conjunction with expert adult melanoma centers, of this rare and challenging patient group.
Collapse
Affiliation(s)
- Anne L. Ryan
- Department of Haematology, Oncology and Bone Marrow Transplant, Perth Children’s Hospital, Perth, WA, Australia
| | - Charlotte Burns
- Children’s Cancer Centre, The Royal Children’s Hospital, Melbourne, VIC, Australia
| | - Aditya K. Gupta
- Cancer Centre for Children, The Children’s Hospital at Westmead, Westmead, NSW, Australia
| | | | - David S. Ziegler
- Kids Cancer Centre, Sydney Children’s Hospital, Randwick, NSW, Australia
| | - Maria L. Kirby
- Department of Haematology/Oncology, Women’s and Children’s Hospital, Adelaide, SA, Australia
| | - Frank Alvaro
- Department of Haematology/Oncology, John Hunter Children’s Hospital, Newcastle, NSW, Australia
| | - Peter Downie
- Children’s Cancer Centre, The Royal Children’s Hospital, Melbourne, VIC, Australia
- Department of Haematology/Oncology, Monash Children’s Hospital, Melbourne, VIC, Australia
| | - Stephen J. Laughton
- Starship Blood and Cancer Centre, Starship Children’s Hospital, Auckland, New Zealand
| | - Siobhan Cross
- Children’s Haematology/Oncology Centre, Christchurch Hospital, Christchurch, New Zealand
| | - Timothy Hassall
- Department of Haematology/Oncology, Queensland Children’s Hospital, Brisbane, QLD, Australia
| | - Geoff B. McCowage
- Cancer Centre for Children, The Children’s Hospital at Westmead, Westmead, NSW, Australia
| | - Jordan R. Hansford
- Children’s Cancer Centre, The Royal Children’s Hospital, Melbourne, VIC, Australia
- Murdoch Children’s Research Institute; Department of Pediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Rishi S. Kotecha
- Department of Haematology, Oncology and Bone Marrow Transplant, Perth Children’s Hospital, Perth, WA, Australia
- Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
- Curtin Medical School, Curtin University, Perth, WA, Australia
| | - Nicholas G. Gottardo
- Department of Haematology, Oncology and Bone Marrow Transplant, Perth Children’s Hospital, Perth, WA, Australia
- Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
| |
Collapse
|
30
|
Caplan IF, Prasad A, Carey RM, Brody RM, Cannady SB, Rajasekaran K, Bur AM, Lukens JN, Briceño CA, Newman JG, Brant JA. Primary Orbital Melanoma: An Investigation of a Rare Malignancy Using the National Cancer Database. Laryngoscope 2021; 131:1790-1797. [PMID: 33570180 DOI: 10.1002/lary.29428] [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: 11/19/2020] [Revised: 12/29/2020] [Accepted: 01/16/2021] [Indexed: 11/11/2022]
Abstract
OBJECTIVES Primary orbital melanoma (POM) is a rare disease with limited data on survival and best treatment practices. Here we utilize the National Cancer Database (NCDB) to determine the overall survival (OS) and covariates that influence mortality. STUDY DESIGN Retrospective cohort study. METHODS All patients diagnosed with POM from 2004 to 2016 were identified in the NCDB. Patient and oncologic data were analyzed using the Kaplan-Meier method and multivariate models for the primary outcome of OS. RESULTS A total of 129 patients were identified. Median OS was 36.9 months (95% confidence interval [CI] 24.1-78.7 months) with mean 5-year survival of 42.0% (CI 33.2%-53.2%). Treatments received included surgery alone (43.4%), radiation alone (23.3%), and surgery followed by radiation (20.2%). The multivariate model demonstrated an increased risk of death associated with age over 80 years (hazard ratio [HR] 3.41, CI 1.31-8.86, P = .012), a Charlson-Deyo comorbidity score of 2 or greater (HR 5.30, CI 1.87-15.03, P = .002), and no treatment (HR 2.28, CI 1.03-5.06, P = .042). For every 1 cm increase in tumor size, there was an increased risk of death (HR 1.06, CI 1.00-1.13, P = .039). When compared to surgery alone, no other treatment modality had an effect on OS. CONCLUSIONS This study leveraged multiyear data from the NCDB to provide prognostic and demographic information on the largest known cohort of POM cases. Increased age, increased comorbidities, not receiving treatment, and larger tumor size were associated with increased mortality. There was no clear survival advantage for specific treatments. LEVEL OF EVIDENCE 4 Laryngoscope, 131:1790-1797, 2021.
Collapse
Affiliation(s)
- Ian F Caplan
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, U.S.A
| | - Aman Prasad
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, U.S.A
| | - Ryan M Carey
- Department of Otorhinolaryngology - Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, U.S.A
| | - Robert M Brody
- Department of Otorhinolaryngology - Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, U.S.A.,Corporal Michael J. Crescenz VAMC, Philadelphia, Pennsylvania, U.S.A
| | - Steven B Cannady
- Department of Otorhinolaryngology - Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, U.S.A
| | - Karthik Rajasekaran
- Department of Otorhinolaryngology - Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, U.S.A
| | - Andrés M Bur
- Department of Otolaryngology - Head and Neck Surgery, University of Kansas School of Medicine, University of Kansas, Kansas City, Kansas, U.S.A
| | - John N Lukens
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, U.S.A
| | - César A Briceño
- Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, U.S.A
| | - Jason G Newman
- Department of Otorhinolaryngology - Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, U.S.A
| | - Jason A Brant
- Department of Otorhinolaryngology - Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, U.S.A.,Corporal Michael J. Crescenz VAMC, Philadelphia, Pennsylvania, U.S.A
| |
Collapse
|
31
|
Almutairi AR, Slack M, Erstad BL, McBride A, Abraham I. Association of immune-checkpoint inhibitors and the risk of immune-related colitis among elderly patients with advanced melanoma: real-world evidence from the SEER-Medicare database. Ther Adv Drug Saf 2021; 12:2042098621991279. [PMID: 33796257 PMCID: PMC7970695 DOI: 10.1177/2042098621991279] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
Background: The use of anti-cytotoxic T-lymphocyte antigen 4 (anti-CTLA4) therapy (ipilimumab) and anti-programmed cell-death 1 (anti-PD1) agents (nivolumab and pembrolizumab) in advanced melanoma have been associated with immune-related adverse events (irAEs) including colitis. We aimed to estimate the incidence and the risk of colitis in elderly patients with advanced melanoma treated with anti-CTLA4 and anti-PD1 in the real-world setting. Methods: Elderly patients (age ⩾ 65 years) diagnosed with advanced melanoma between 2011 and 2015 and treated with anti-CTLA4 or anti-PD1 agents were identified from the Surveillance, Epidemiology, and End Results (SEER)–Medicare data. We estimated the risk of colitis from start of treatment up to 90 days from the last dose of therapy. We used the log-rank test and logistic regression with adjustment for potential confounders using the inverse probability of treatment weighting method. We conducted several sensitivity analyses. Results: A total of 274 elderly patients with advanced melanoma were included in our cohort. The risk of colitis was similar between anti-PD1 users and anti-CTLA4 users based on log-rank test (p = 0.17) and logistic regression [odds ratio (OR) = 0.35, 95% confidence interval (95%CI) 0.04–2.79]. Sensitivity analyses for patients with all-stage melanoma showed a significantly lower risk of colitis in anti-PD1 compared with anti-CTLA4 treated patients based on log-rank test (p = 0.017) and logistic regression (OR = 0.21, 95%CI 0.09–0.53). Conclusion: Elderly with advanced melanoma treated with anti-CTLA4 or anti-PD1 had a similar risk of developing colitis. However, there was a statistically significant difference in the risk of colitis between anti-CTLA4 or anti-PD1 users among all-stage-melanoma patients. Plain Language Summary Risk of colitis (inflammation of the large intestine) in elderly patients with melanoma treated with immune-checkpoint inhibitors (a group of medications that uses the patient’s immune system to fight cancer) While the anti-cancer agents known as immune-checkpoint inhibitors have had a great impact on the treatment of melanoma, they may also have side effects. This study estimated the risk of colitis, a chronic inflammation of the colon, in elderly patients with melanoma treated with anti-cytotoxic T-lymphocyte antigen 4 (anti-CTLA4) or anti-programmed cell-death 1 (anti-PD1) agents, using data from the Surveillance, Epidemiology, and End Results (SEER)–Medicare linked database. Overall, we found that the risk of colitis was not different between anti-PD1 users and anti-CTLA4 users with advanced-stage melanoma. However, after including patients across all stages of melanoma, we found a significantly lower risk of colitis with anti-PD1 compared with anti-CTLA4.
Collapse
Affiliation(s)
- Abdulaali R. Almutairi
- Center for Health Outcomes and PharmacoEconomic Research, College of Pharmacy, University of Arizona, Tucson, AZ, USA
- Department of Pharmacy Practice and Science, University of Arizona, Tucson, AZ, USA; Drug Sector, Saudi Food and Drug Authority, Riyadh, Saudi Arabia
| | - Marion Slack
- Department of Pharmacy Practice and Science, University of Arizona, Tucson, AZ, USA
| | - Brian L. Erstad
- Department of Pharmacy Practice and Science, University of Arizona, Tucson, AZ, USA
- University of Arizona Cancer Center, Tucson, AZ, USA
| | - Ali McBride
- Department of Pharmacy Practice and Science, University of Arizona, Tucson, AZ, USA
- University of Arizona Cancer Center, Tucson, AZ, USA
| | | |
Collapse
|
32
|
Qu H, Zhao H, Zhang X, Liu Y, Li F, Sun L, Song Z. Integrated Analysis of the ETS Family in Melanoma Reveals a Regulatory Role of ETV7 in the Immune Microenvironment. Front Immunol 2020; 11:612784. [PMID: 33424867 PMCID: PMC7786291 DOI: 10.3389/fimmu.2020.612784] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/19/2020] [Indexed: 12/22/2022] Open
Abstract
The ETS family modulates immune response and drug efficiency to targeted therapies, but their role in melanoma is largely unclear. In this study, the ETS family was systematically analyzed in multiple public data sets. Bioinformatics tools were used to characterize the function of ETV7 in melanoma. A prognostic model was constructed using the LASSO Cox regression method. We found that ETV7 was the only differentially expressed gene with significant prognostic relevance in melanoma. Enrichment analysis of seven independent data sets indicated ETV7 participation in various immune-related pathways. ETV7 particularly showed a strong positive correlation with CD8+ T cell infiltration. The prognostic model based on ETV7 and its hub genes showed a relatively good predictive value in training and testing data sets. Thus, ETV7 can potentially regulate the immune microenvironment in melanoma.
Collapse
Affiliation(s)
- Hui Qu
- Department of Plastic Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Hui Zhao
- Department of Urology, The Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Xi Zhang
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yang Liu
- Department of Pathology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Feng Li
- Department of Plastic Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Liyan Sun
- Department of Plastic Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Zewen Song
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
33
|
Pavlick AC, Zhao R, Lee CH, Ritchings C, Rao S. First-line immunotherapy versus targeted therapy in patients with BRAF-mutant advanced melanoma: a real-world analysis. Future Oncol 2020; 17:689-699. [PMID: 33084375 DOI: 10.2217/fon-2020-0643] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aim: To compare effectiveness of nivolumab + ipilimumab (NIVO + IPI) versus BRAF + MEK inhibitors (BRAFi + MEKi) in patients with BRAF-mutant advanced melanoma in the real-world setting. Materials & methods: This study used the Flatiron Health electronic medical record database. Results: After adjusting for differences in baseline characteristics, NIVO + IPI was associated with a 32% reduction in risk of death versus BRAFi + MEKi. At a mean follow-up of 15-16 months, 64% of NIVO + IPI patients and 43% of BRAFi + MEKi patients were alive; subsequent therapy was administered to 33 and 41% of patients, respectively. After first-line NIVO + IPI, 20% of patients died before subsequent therapy, whereas 32% died after first-line BRAFi + MEKi. Conclusion: In this real-world study, patients treated with first-line NIVO + IPI showed significant survival benefit versus those receiving first-line BRAFi + MEKi.
Collapse
Affiliation(s)
| | - Ruizhi Zhao
- Bristol Myers Squibb, Princeton, NJ 08540, USA
| | - Cho-Han Lee
- Bristol Myers Squibb, Princeton, NJ 08540, USA
| | | | - Sumati Rao
- Bristol Myers Squibb, Princeton, NJ 08540, USA
| |
Collapse
|
34
|
Seyed Jafari SM, Feldmeyer L, Hunger RE. Development of Extragenital Lichen Sclerosus in Malignant Melanoma Patients Treated With Ipilimumab in Combination With Nivolumab. Front Oncol 2020; 10:573527. [PMID: 33117707 PMCID: PMC7577175 DOI: 10.3389/fonc.2020.573527] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/25/2020] [Indexed: 12/26/2022] Open
Abstract
The immune checkpoint inhibitors opened a new era for the treatment of melanoma. Nowadays, combined immune checkpoint inhibitors are administered to provide additive or synergistic effects on anti-melanoma immunity. The use of these drugs comes with serious adverse events related to excessive immune activation. Here, we present development of extragenital lichen sclerosus in a patient with metastatic malignant melanoma, during the combined therapy with checkpoint inhibitors.
Collapse
Affiliation(s)
- S. Morteza Seyed Jafari
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | | |
Collapse
|
35
|
Saraiva DP, Matias AT, Braga S, Jacinto A, Cabral MG. Establishment of a 3D Co-culture With MDA-MB-231 Breast Cancer Cell Line and Patient-Derived Immune Cells for Application in the Development of Immunotherapies. Front Oncol 2020; 10:1543. [PMID: 32974189 PMCID: PMC7482668 DOI: 10.3389/fonc.2020.01543] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022] Open
Abstract
3D cell culture including different cell types, such as immune cells, is a representative platform that mimics the tumor microenvironment. Here we disclose an easy-to-handle 3D co-culture protocol using a scaffold-free technique with the breast cancer cell line MDA-MB-231 and breast cancer patient-derived immune cells from peripheral blood. The method presented is simple, less time-consuming and less expensive when compared to other 3D techniques. Additionally, this is an optimized protocol for the establishment of a 3D system for this cell line, which is normally seen as challenging to spontaneously form spheroids. The addition of patient-derived immune cells to the cancer cells' spheroid allows the study of the crosstalk between both cell types, as well as the assessment of individual therapeutic approaches to intensify the antitumor immune response. In fact, with this model, we observed that patients' immune cells exhibit a wide range of antitumor responses and we further demonstrated that it is possible to manipulate the less effective ones with a canonical stimulus, as a proof-of-concept, in order to improve their ability to lower the viability of tumor cells. Therefore, this platform could be applied for a personalized immune-based drug screening, with results after a maximum of 10 days of culture, in order to develop more tailored breast cancer treatments and ameliorate patients' survival rate.
Collapse
Affiliation(s)
- Diana P Saraiva
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas da Universidade Nova de Lisboa, Lisbon, Portugal
| | - Ana T Matias
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas da Universidade Nova de Lisboa, Lisbon, Portugal
| | - Sofia Braga
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas da Universidade Nova de Lisboa, Lisbon, Portugal.,Instituto CUF de Oncologia, Lisbon, Portugal
| | - António Jacinto
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas da Universidade Nova de Lisboa, Lisbon, Portugal
| | - M Guadalupe Cabral
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas da Universidade Nova de Lisboa, Lisbon, Portugal
| |
Collapse
|
36
|
MicroRNA-21-Enriched Exosomes as Epigenetic Regulators in Melanomagenesis and Melanoma Progression: The Impact of Western Lifestyle Factors. Cancers (Basel) 2020; 12:cancers12082111. [PMID: 32751207 PMCID: PMC7464294 DOI: 10.3390/cancers12082111] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/16/2020] [Accepted: 07/24/2020] [Indexed: 02/06/2023] Open
Abstract
DNA mutation-induced activation of RAS-BRAF-MEK-ERK signaling associated with intermittent or chronic ultraviolet (UV) irradiation cannot exclusively explain the excessive increase of malignant melanoma (MM) incidence since the 1950s. Malignant conversion of a melanocyte to an MM cell and metastatic MM is associated with a steady increase in microRNA-21 (miR-21). At the epigenetic level, miR-21 inhibits key tumor suppressors of the RAS-BRAF signaling pathway enhancing proliferation and MM progression. Increased MM cell levels of miR-21 either result from endogenous upregulation of melanocytic miR-21 expression or by uptake of miR-21-enriched exogenous exosomes. Based on epidemiological data and translational evidence, this review provides deeper insights into environmentally and metabolically induced exosomal miR-21 trafficking beyond UV-irradiation in melanomagenesis and MM progression. Sources of miR-21-enriched exosomes include UV-irradiated keratinocytes, adipocyte-derived exosomes in obesity, airway epithelium-derived exosomes generated by smoking and pollution, diet-related exosomes and inflammation-induced exosomes, which may synergistically increase the exosomal miR-21 burden of the melanocyte, the transformed MM cell and its tumor environment. Several therapeutic agents that suppress MM cell growth and proliferation attenuate miR-21 expression. These include miR-21 antagonists, metformin, kinase inhibitors, beta-blockers, vitamin D, and plant-derived bioactive compounds, which may represent new options for the prevention and treatment of MM.
Collapse
|
37
|
Narrow-Margin Excision for Invasive Acral Melanoma: Is It Acceptable? J Clin Med 2020; 9:jcm9072266. [PMID: 32708762 PMCID: PMC7408916 DOI: 10.3390/jcm9072266] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/15/2020] [Accepted: 07/15/2020] [Indexed: 12/23/2022] Open
Abstract
In this retrospective review of 100 patients with primary invasive acral melanoma, we examined whether narrow-margin excision is warranted for acral melanoma. Patients treated with surgical margins recommended by the National Comprehensive Cancer Network (R-group) were compared to those treated with narrow margins (N-group). A total of 65 patients underwent narrow-margin excision. Positive margin status or local recurrence rarely occurred regardless of the excision margins, whereas fatal events frequently occurred, particularly among the patients with T4 melanoma. The mortality rates of N- and R-group with T1–3 melanomas were similar (1.36 and 1.28 per 100 person-years, respectively). However, patients with T4 melanoma treated with narrow-margin excision had a higher mortality rate (11.44 vs. 5.03 per 100 person-years). Kaplan–Meier analyses showed a worse prognosis in the N-group (p = 0.045) but this group had thicker Breslow thickness (4.21 mm vs. 2.03 mm, p = 0.0013). A multivariate analysis showed that Breslow thickness was an independent risk factor, but surgical margin was not a risk factor for melanoma-specific survival or disease-free survival. In conclusion, although we could not find a difference between the narrow-margin excision and recommended-margin excision in this study, we suggest following current recommendations of guidelines. Our study warrants the prospective collection of data on acral melanoma to better define the prognosis of this infrequent type of melanoma.
Collapse
|
38
|
Abstract
Immune checkpoint inhibitors deeply improved the prognosis of metastatic melanoma or other types of cancer, but their related adverse effects (AEs) can be very severe, especially when the neurological system is touched, as in myasthenia gravis (MG). It is a rare immune AE that can be life-threatening and can be revealed by several symptoms. We report a case of our experience and review the current literature of MG exacerbated or occurring during immunotherapy to describe characteristics of this AE, warn the oncologist about this toxicity, and summarize the treatments conducted. Thirty-four cases of MG were reported, mostly with anti-programmed cell death protein 1 checkpoint inhibitor, and with melanoma. Onset was quick after the first or second infusion. Treatment comprised corticosteroids, prostigmine, and more or less plasmapheresis or immunoglobulins. Prognosis is poor, as 13 patients died after MG. MG is a rare immune-related AE that must be rapidly evoked and treated in case of neurological symptoms emerging after immunotherapy.
Collapse
|
39
|
Fahmy D, El‐Amawy H, El‐Samongy M, Fouda A, Soliman S, El‐Kady A, Farnetani F, Conti A, Zoeir A, Eissa A, Eissa R, Puliatti S, Sighinolfi M, Rocco B, Pellacani G. COVID-19 and dermatology: a comprehensive guide for dermatologists. J Eur Acad Dermatol Venereol 2020; 34:1388-1394. [PMID: 32428303 PMCID: PMC7276795 DOI: 10.1111/jdv.16545] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Linked articles: COVID‐19 SPECIAL FORUM. J Eur Acad Dermatol Venereol 2020; 34: e291–e310 .
Collapse
Affiliation(s)
- D.H. Fahmy
- Dermatology DepartmentFaculty of MedicineTanta UniversityTantaEgypt
| | - H.S. El‐Amawy
- Dermatology DepartmentFaculty of MedicineTanta UniversityTantaEgypt
| | - M.A. El‐Samongy
- Dermatology DepartmentFaculty of MedicineTanta UniversityTantaEgypt
| | - A.A. Fouda
- Dermatology DepartmentFaculty of MedicineTanta UniversityTantaEgypt
| | - S.H. Soliman
- Dermatology DepartmentFaculty of MedicineTanta UniversityTantaEgypt
| | - A. El‐Kady
- Dermatology DepartmentEl‐Menshawy HospitalMinistry of Health & PopulationTantaEgypt
| | - F. Farnetani
- Dermatology DepartmentUniversity of Modena & Reggio EmiliaModenaItaly
| | - A. Conti
- Dermatology DepartmentAzienda Ospedaliero Universitaria of ModenaModenaItaly
| | - A. Zoeir
- Urology DepartmentUniversity of Modena & Reggio EmiliaModenaItaly
- Urology DepartmentFaculty of MedicineTanta UniversityTantaEgypt
| | - A. Eissa
- Urology DepartmentUniversity of Modena & Reggio EmiliaModenaItaly
- Urology DepartmentFaculty of MedicineTanta UniversityTantaEgypt
| | - R. Eissa
- Microbiology & Virology DepartmentFaculty of MedicineTanta UniversityTantaEgypt
| | - S. Puliatti
- Urology DepartmentUniversity of Modena & Reggio EmiliaModenaItaly
- ORSI AcademyMelleBelgium
| | - M.C. Sighinolfi
- Urology DepartmentUniversity of Modena & Reggio EmiliaModenaItaly
| | - B. Rocco
- Urology DepartmentUniversity of Modena & Reggio EmiliaModenaItaly
| | - G. Pellacani
- Dermatology DepartmentUniversity of Modena & Reggio EmiliaModenaItaly
| |
Collapse
|
40
|
Takenaka W, Takahashi Y, Tamari K, Minami K, Katsuki S, Seo Y, Isohashi F, Koizumi M, Ogawa K. Radiation Dose Escalation is Crucial in Anti-CTLA-4 Antibody Therapy to Enhance Local and Distant Antitumor Effect in Murine Osteosarcoma. Cancers (Basel) 2020; 12:E1546. [PMID: 32545427 PMCID: PMC7352693 DOI: 10.3390/cancers12061546] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/06/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022] Open
Abstract
We previously reported that a combination of 10 Gy of X-ray irradiation and dual immune checkpoint blockade with anti-CTLA-4 (C4) and anti-PD-L1 antibodies produced a significant shrinkage of irradiated and unirradiated tumors (abscopal effect) and prolonged overall survival. However, the optimal radiation delivery regimen combined with single immune checkpoint blockade of C4 for inducing a maximum systemic antitumor response still remains unclear, particularly for patients with osteosarcoma. We used syngeneic C3H mice that were subcutaneously injected with LM8 osteosarcoma cells into both legs. C4 was administered three times, and one side of the tumor was irradiated by X-ray beams. The optimal radiation dose required to induce the abscopal effect was explored with a focus on the induction of the type-I interferon pathway. Radiation delivered in a single fraction of 10 Gy, 4.5 Gy × 3 fractions (fx), and 2 Gy × 8 fx with C4 failed to produce significant inhibition of unirradiated tumor growth compared with monotherapy with C4. Dose escalation to 16 Gy in a single fraction, or the equivalent hypofractionated dose of 8 Gy × 3 fx, which significantly increased secretion of IFN-β in vitro, produced a dramatic regression of both irradiated and unirradiated tumors and prolonged overall survival in combination with C4. Furthermore, irradiation at 16 Gy in both a single fraction and 8 Gy × 3 fx diminished regulatory T cells in the unirradiated tumor microenvironment. These results suggest that total dose escalation of radiation is crucial in C4 therapy to enhance the antitumor response in both local and distant tumors and prolonged overall survival regardless of fractionation for osteosarcoma.
Collapse
Affiliation(s)
- Wataru Takenaka
- Department of Medical Physics and Engineering, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan; (W.T.); (S.K.); (M.K.)
| | - Yutaka Takahashi
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan; (K.T.); (K.M.); (Y.S.); (F.I.); (K.O.)
| | - Keisuke Tamari
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan; (K.T.); (K.M.); (Y.S.); (F.I.); (K.O.)
| | - Kazumasa Minami
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan; (K.T.); (K.M.); (Y.S.); (F.I.); (K.O.)
| | - Shohei Katsuki
- Department of Medical Physics and Engineering, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan; (W.T.); (S.K.); (M.K.)
| | - Yuji Seo
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan; (K.T.); (K.M.); (Y.S.); (F.I.); (K.O.)
| | - Fumiaki Isohashi
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan; (K.T.); (K.M.); (Y.S.); (F.I.); (K.O.)
| | - Masahiko Koizumi
- Department of Medical Physics and Engineering, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan; (W.T.); (S.K.); (M.K.)
| | - Kazuhiko Ogawa
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan; (K.T.); (K.M.); (Y.S.); (F.I.); (K.O.)
| |
Collapse
|
41
|
Kyriakou G, Melachrinou M. Cancer stem cells, epigenetics, tumor microenvironment and future therapeutics in cutaneous malignant melanoma: a review. Future Oncol 2020; 16:1549-1567. [PMID: 32484008 DOI: 10.2217/fon-2020-0151] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
This review provides an overview of the current understanding of the ontogeny and biology of melanoma stem cells in cutaneous malignant melanoma. This article also summarizes and evaluates the current knowledge of the underlying epigenetic mechanisms, the regulation of melanoma progress by the tumor microenvironment as well as the therapeutic implications and applications of these novel insights, in the setting of personalized medicine. Unraveling the complex ecosystem of cutaneous malignant melanoma and the interplay between its components, aims to provide novel insights into the establishment of efficient therapeutic strategies.
Collapse
Affiliation(s)
- Georgia Kyriakou
- Department of Dermatology, University General Hospital of Patras, Rion 265 04, Greece
| | - Maria Melachrinou
- Department of Pathology, University General Hospital of Patras, Rion 265 04, Greece
| |
Collapse
|
42
|
Clinical Update on Checkpoint Inhibitor Therapy for Conjunctival and Eyelid Melanoma. Int Ophthalmol Clin 2020; 60:77-89. [PMID: 32205655 DOI: 10.1097/iio.0000000000000308] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
43
|
Immunohistochemical BRAF V600E Expression and Intratumor BRAF V600E Heterogeneity in Acral Melanoma: Implication in Melanoma-Specific Survival. J Clin Med 2020; 9:jcm9030690. [PMID: 32143442 PMCID: PMC7141227 DOI: 10.3390/jcm9030690] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 02/23/2020] [Accepted: 03/02/2020] [Indexed: 01/09/2023] Open
Abstract
Acral melanoma, a distinct form of cutaneous melanoma originating in the glabrous skin of the palms, soles, and nail beds, has a different genetic background from other subtypes of cutaneous melanoma. The roles of oncogenic BRAF mutations of acral melanoma in pathogenesis and patient outcomes have not been fully elucidated. We retrieved a total of 112 patients with primary acral melanoma and checked their BRAF V600E status using immunohistochemical staining of VE1 antibody. Among these cases, 21 acral melanoma samples (18.8%) showed positive BRAF V600E staining, and of those, 11 samples (9.8%) showed a heterogeneous staining pattern, with a mixture of VE1-positive and VE1-negative cells. BRAF V600E positivity was significantly associated with thicker melanoma (p = 0.0015). There was no significant difference in clinicopathological factors between homogeneous and heterogeneous VE1-positive acral melanoma. Both patients with BRAF V600E-positive acral melanoma and those with heterogeneous BRAF V600E had significantly shorter melanoma-specific survival than those with BRAF V600E-negative melanoma in Kaplan–Meier analysis (p = 0.0283 and p = 0.0065, respectively). These findings provide novel insights into the pathobiology of acral melanoma.
Collapse
|
44
|
Ito T, Kaku-Ito Y, Murata M, Ichiki T, Kuma Y, Tanaka Y, Ide T, Ohno F, Wada-Ohno M, Yamada Y, Oda Y, Furue M. Intra- and Inter-Tumor BRAF Heterogeneity in Acral Melanoma: An Immunohistochemical Analysis. Int J Mol Sci 2019; 20:E6191. [PMID: 31817947 PMCID: PMC6941107 DOI: 10.3390/ijms20246191] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/28/2019] [Accepted: 12/05/2019] [Indexed: 12/26/2022] Open
Abstract
The current development of BRAF inhibitors has revolutionized the treatment of unresectable melanoma. As the potential heterogeneity of BRAF mutations in melanoma has been reported, accurate detection of BRAF mutations are important. However, the genetic heterogeneity of acral melanoma-a distinct type of melanoma with a unique genetic background-has not fully been investigated. We conducted a retrospective review of our acral melanoma patients. Of the 196 patients with acral melanoma, we retrieved 31 pairs of primary and matched metastatic melanomas. We immunostained the 31 pairs with VE1, a BRAFV600E-mutation-specific monoclonal antibody. Immunohistochemistry with VE1 showed a high degree of sensitivity and specificity for detecting BRAFV600E mutations compared with the real-time polymerase chain reaction method. A total of nine primary (29.0%) and eight metastatic (25.8%) acral melanomas were positive for VE1. In three patients (9.7%), we observed a discordance of VE1 staining between the primary and metastatic lesions. Of note, VE1 immunohistochemical staining revealed a remarkable degree of intra-tumor genetic heterogeneity in acral melanoma. Our study reveals that VE1 immunostaining is a useful ancillary method for detecting BRAFV600E mutations in acral melanoma and allows for a clear visualization of intra- and inter-tumor BRAF heterogeneity.
Collapse
Affiliation(s)
- Takamichi Ito
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.-I.); (M.M.); (T.I.); (Y.K.); (Y.T.); (T.I.); (F.O.); (M.W.-O.); (M.F.)
| | - Yumiko Kaku-Ito
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.-I.); (M.M.); (T.I.); (Y.K.); (Y.T.); (T.I.); (F.O.); (M.W.-O.); (M.F.)
| | - Maho Murata
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.-I.); (M.M.); (T.I.); (Y.K.); (Y.T.); (T.I.); (F.O.); (M.W.-O.); (M.F.)
| | - Toshio Ichiki
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.-I.); (M.M.); (T.I.); (Y.K.); (Y.T.); (T.I.); (F.O.); (M.W.-O.); (M.F.)
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.Y.); (Y.O.)
| | - Yuki Kuma
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.-I.); (M.M.); (T.I.); (Y.K.); (Y.T.); (T.I.); (F.O.); (M.W.-O.); (M.F.)
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.Y.); (Y.O.)
| | - Yuka Tanaka
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.-I.); (M.M.); (T.I.); (Y.K.); (Y.T.); (T.I.); (F.O.); (M.W.-O.); (M.F.)
| | - Taketoshi Ide
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.-I.); (M.M.); (T.I.); (Y.K.); (Y.T.); (T.I.); (F.O.); (M.W.-O.); (M.F.)
| | - Fumitaka Ohno
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.-I.); (M.M.); (T.I.); (Y.K.); (Y.T.); (T.I.); (F.O.); (M.W.-O.); (M.F.)
| | - Maiko Wada-Ohno
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.-I.); (M.M.); (T.I.); (Y.K.); (Y.T.); (T.I.); (F.O.); (M.W.-O.); (M.F.)
| | - Yuichi Yamada
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.Y.); (Y.O.)
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.Y.); (Y.O.)
| | - Masutaka Furue
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.-I.); (M.M.); (T.I.); (Y.K.); (Y.T.); (T.I.); (F.O.); (M.W.-O.); (M.F.)
| |
Collapse
|
45
|
Rimassa L, Pressiani T, Merle P. Systemic Treatment Options in Hepatocellular Carcinoma. Liver Cancer 2019; 8:427-446. [PMID: 31799201 PMCID: PMC6883446 DOI: 10.1159/000499765] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/21/2019] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Patients with advanced hepatocellular carcinoma (HCC) typically have poor survival outcomes. Until recently, sorafenib was the only systemic therapy option available and no agents were approved after sorafenib failure. However, rapid changes are beginning to emerge in the treatment landscape of advanced HCC, with approvals of regorafenib, nivolu-mab, lenvatinib, pembrolizumab, and cabozantinib and positive phase II/III clinical trial results with other agents. SUMMARY Here, we provide a comprehensive overview of the clinical trial data of systemic agents that are currently approved for advanced HCC (sorafenib, regorafenib, and nivolumab), including agents recently approved in 2018 (lenvatinib, pembrolizumab, and cabozantinib) and those with recent positive phase II/III results (ramucirumab). Key features of the clinical trial design, including patient selection criteria, the use of biomarkers in HCC, and criteria for efficacy assessment, and their implications in real-world practice are discussed. Important ongoing and planned trials in advanced HCC are summarized to provide a glimpse into the future of advanced HCC treatment. From a physician's viewpoint, the treatment algorithms for advanced HCC are undergoing significant changes, as additional and imminent approvals impact the choices of first- and second-line treatment and decisions regarding the timing of therapy initiation. With these additional choices at hand, treatment sequencing remains a complex task and should take patient selection and tolerance profiles into account. KEY MESSAGES The treatment of advanced HCC remains challenging and complex. The rapid developments in systemic therapy for advanced HCC should be considered when determining the best choice and sequence of treatment for patients with advanced HCC.
Collapse
Affiliation(s)
- Lorenza Rimassa
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, Humanitas Clinical and Research Center – IRCCS, Milan, Italy
| | - Tiziana Pressiani
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, Humanitas Clinical and Research Center – IRCCS, Milan, Italy
| | - Philippe Merle
- Hepatology Unit, Croix-Rousse Hospital, Groupement Hospitalier Lyon Nord, Lyon, France
| |
Collapse
|
46
|
Umemura H, Kaji T, Tachibana K, Morizane S, Yamasaki O. Serum 5-S-cysteinyl-dopa levels as a predictive marker for the efficacy of nivolumab in advanced malignant melanoma. Int J Biol Markers 2019; 34:414-420. [DOI: 10.1177/1724600819883658] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Purpose: With the recent developments in novel molecular targeted therapy such as immune-checkpoint blockades, serine/threonine-protein kinase B-Raf, and mitogen-activated protein kinase kinase inhibitors, the prognosis of advanced malignant melanoma has been improving. 5-S-cysteinyl-dopa (5-S-CD), a precursor of pheomelanin, has been previously revealed to be a useful biomarker for advanced-stage malignant melanoma, especially in patients with distant metastases. Here, we aimed to assess and compare the utility of serum 5-S-CD and lactate dehydrogenase levels as markers for predicting the effects of nivolumab in advanced malignant melanoma. Methods: Baseline serum 5-S-CD and lactate dehydrogenase levels in patients with unresectable stage IIIC and IV malignant melanoma treated with nivolumab ( n = 21) were analyzed to determine their utility as predictive markers for survival. We also analyzed the prognostic value of these markers among patients with only stage IV malignant melanoma ( n = 17). Results: Our analysis showed that patients with baseline serum 5-S-CD levels >25.0 nmol/L had significantly poor prognosis. In contrast, serum lactate dehydrogenase levels at the upper limit of the normal range did not exhibit such changes. Conclusions: Serum 5-S-CD levels have the potential to be an excellent predictive marker for the efficacy of nivolumab therapy in patients with advanced malignant melanoma.
Collapse
Affiliation(s)
- Hiroshi Umemura
- Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Division of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Tatsuya Kaji
- Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kota Tachibana
- Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shin Morizane
- Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Osamu Yamasaki
- Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
47
|
Kochin V, Nishikawa H. <Editors' Choice> Meddling with meddlers: curbing regulatory T cells and augmenting antitumor immunity. NAGOYA JOURNAL OF MEDICAL SCIENCE 2019; 81:1-18. [PMID: 30962651 PMCID: PMC6433633 DOI: 10.18999/nagjms.81.1.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
CD4+ regulatory T cells (Tregs) expressing the transcription factor forkhead
box P3 (FoxP3) play an important role in self-tolerance and immune homeostasis. Tregs have
evolved to protect the host from aberrant immune responses against self-components and
collateral damages occurring in the process of defense against invading pathogens by
softening immune responses. However, they turned to be a scourge in malignant tumors by
not only allowing and promoting tumor growth but also suppressing effective antitumor
actions, both inherent (host’s immune surveillance) and extrinsic (anticancer therapy). An
increase in the number of Tregs infiltrating into tumor sites and a concomitant decrease
in the number of CD8+ cytotoxic T lymphocytes are associated with a poor
prognosis for various types of cancers, marking Tregs as notorious meddlers with an
effective antitumor response. Various cancer immunotherapy approaches are often dampened
by meddling Tregs, making them one of the major targets in the treatment of cancer. The
recent success of immune checkpoint inhibitors (ICIs) that target immune checkpoint
molecules expressed by Tregs or effector T cells implies, that “meddling with meddlers”
represents an effective strategy in cancer immunotherapy. However, clinical responses to
ICIs are effective and durable only in some patients with cancer, whereas more than half
of them do not show significant clinical improvement. This implies that a therapeutic
approach based on the use of a single ICI, or targeting Tregs alone, is insufficient,
highlighting the need for combinatorial approaches. With regard to antitumor immune
stimulation, several approaches, such as vaccination with peptides (or the corresponding
DNA) to stimulate antigen-presenting CD8+ T cells with tumor-specific
neoantigens, cancer/testis antigens, or cancer stem cell antigens, that eventually boost
effective cytotoxic antitumor responses are being tested. This review describes the
immunosuppressive physiology of Tregs and their meddling with the host’s antitumor
immunity; current and prospective approaches to curb Tregs; and approaches to augment
antitumor immunity.
Collapse
Affiliation(s)
- Vitaly Kochin
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroyoshi Nishikawa
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo / Chiba, Japan
| |
Collapse
|
48
|
Hamuro L, Statkevich P, Bello A, Roy A, Bajaj G. Nivolumab Clearance Is Stationary in Patients With Resected Melanoma on Adjuvant Therapy: Implications of Disease Status on Time-Varying Clearance. Clin Pharmacol Ther 2019; 106:1018-1027. [PMID: 31090921 DOI: 10.1002/cpt.1502] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 03/21/2019] [Indexed: 12/24/2022]
Abstract
Nivolumab clearance (CL) in patients with advanced melanoma (MEL) decreases over the treatment duration, with change in CL associated with improved disease status, measured by reduced tumor burden. Here, we characterize the pharmacokinetics of nivolumab administered as adjuvant therapy for patients with MEL (AdjMEL) whose tumors were removed by surgical resection. A population pharmacokinetic model was developed using data from 1,773 patients with AdjMEL, MEL, non-small cell lung cancer, and other solid tumors who received nivolumab over a dose range of 0.1-20 mg/kg every 2 weeks. In patients with AdjMEL, the geometric mean nivolumab CL of 6.0 mL/hour was 40% lower at baseline and did not vary with time and 20% lower at steady state compared with patients with MEL. Lower nivolumab CL in patients with AdjMEL and absence of time dependence support the hypothesis that changes in nivolumab CL in the metastatic setting are associated with disease status after treatment.
Collapse
Affiliation(s)
- Lora Hamuro
- Clinical Pharmacology and Pharmacometrics, Bristol-Myers Squibb, Princeton, New Jersey, USA
| | - Paul Statkevich
- Clinical Pharmacology and Pharmacometrics, Bristol-Myers Squibb, Princeton, New Jersey, USA
| | - Akintunde Bello
- Clinical Pharmacology and Pharmacometrics, Bristol-Myers Squibb, Princeton, New Jersey, USA
| | - Amit Roy
- Clinical Pharmacology and Pharmacometrics, Bristol-Myers Squibb, Princeton, New Jersey, USA
| | - Gaurav Bajaj
- Clinical Pharmacology and Pharmacometrics, Bristol-Myers Squibb, Princeton, New Jersey, USA
| |
Collapse
|
49
|
Raman SS, Hecht JR, Chan E. Talimogene laherparepvec: review of its mechanism of action and clinical efficacy and safety. Immunotherapy 2019; 11:705-723. [PMID: 31045464 DOI: 10.2217/imt-2019-0033] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Intratumoral immunotherapy inhibits tumor growth by killing injected tumor cells, thus initiating local and systemic immune responses. Oncolytic viruses are a novel class of intratumoral immunotherapies that show promise for treating solid tumors. Talimogene laherparepvec is a first-in-class, genetically modified, herpes simplex virus type 1-based oncolytic immunotherapy approved for the local treatment of unresectable cutaneous, subcutaneous and nodal lesions in patients with melanoma recurrent after initial surgery. This review highlights the dual mechanism of action for talimogene laherparepvec (direct tumor cell lysis and stimulation of local response in tumor microenvironment and systemic immune response in distant metastases), summarizes key preclinical and clinical trials evaluating efficacy and safety of talimogene laherparepvec in melanoma, and describes studies ongoing in other solid tumors.
Collapse
Affiliation(s)
- Steven S Raman
- David Geffen School of Medicine, University of California, Los Angeles, CA, 90095 USA
| | - Joel Randolph Hecht
- David Geffen School of Medicine, University of California, Los Angeles, CA, 90095 USA
| | - Emily Chan
- Amgen Inc., Thousand Oaks, CA, 91320 USA
| |
Collapse
|
50
|
Wang F, Luo Y, Tian X, Ma S, Sun Y, You C, Gong Y, Xie C. Impact of Radiotherapy Concurrent with Anti-PD-1 Therapy on the Lung Tissue of Tumor-Bearing Mice. Radiat Res 2019; 191:271-277. [PMID: 30694722 DOI: 10.1667/rr15182.1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Pneumonitis is a common adverse effect found in non-small cell lung cancer patients after radiotherapy or immune checkpoint inhibitor treatment. We investigated the effects of these two therapies, combined, in the lung tissue of an orthotopic tumor-bearing mouse model. The mice received an 8 Gy dose three times with or without 200 µg anti-programmed death-1 (anti-PD-1) antibody intraperitoneal injection every three days. Lung tissues were H&E stained to determine histological changes. The serum levels of cytokines, such as interferon-γ, tumor necrosis factor and interleukin-5, were detected by cytometric bead array. The neutrophil infiltration was evaluated by immunohistochemical staining for myeloperoxidase. The lung injury score was higher in the treated groups than the control group, especially in the combined treatment group, in which the proportion of neutrophils in lung tissues was significantly higher compared to any other groups. Similarly, the CD4/CD8 ratio of the lung tissues in the combined treatment group, as well as the serum levels of interferon-γ, tumor necrosis factor and interleukin-5, were significantly higher than the other groups. These findings indicate that radiation combined with anti-PD-1 treatment leads to more severe lung injury in the orthotopic tumor-bearing mouse model, accompanied by increased neutrophil infiltration and increased inflammatory response.
Collapse
Affiliation(s)
- Feng Wang
- a Department of Radiation and Medical Oncology, Hubei Cancer Clinical Study Center.,b Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center
| | - Yuan Luo
- a Department of Radiation and Medical Oncology, Hubei Cancer Clinical Study Center.,b Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center
| | - Xiaoli Tian
- a Department of Radiation and Medical Oncology, Hubei Cancer Clinical Study Center.,b Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center
| | - Shijing Ma
- a Department of Radiation and Medical Oncology, Hubei Cancer Clinical Study Center.,b Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center
| | - Yingming Sun
- a Department of Radiation and Medical Oncology, Hubei Cancer Clinical Study Center.,b Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center
| | - Chengcheng You
- a Department of Radiation and Medical Oncology, Hubei Cancer Clinical Study Center.,b Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center
| | - Yan Gong
- c Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Conghua Xie
- a Department of Radiation and Medical Oncology, Hubei Cancer Clinical Study Center.,b Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center
| |
Collapse
|