1
|
Lei W, Zhou K, Lei Y, Li Q, Zhu H. Cancer vaccines: platforms and current progress. MOLECULAR BIOMEDICINE 2025; 6:3. [PMID: 39789208 PMCID: PMC11717780 DOI: 10.1186/s43556-024-00241-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 01/12/2025] Open
Abstract
Cancer vaccines, crucial in the immunotherapeutic landscape, are bifurcated into preventive and therapeutic types, both integral to combating oncogenesis. Preventive cancer vaccines, like those against HPV and HBV, reduce the incidence of virus-associated cancers, while therapeutic cancer vaccines aim to activate dendritic cells and cytotoxic T lymphocytes for durable anti-tumor immunity. Recent advancements in vaccine platforms, such as synthetic peptides, mRNA, DNA, cellular, and nano-vaccines, have enhanced antigen presentation and immune activation. Despite the US Food and Drug Administration approval for several vaccines, the full therapeutic potential remains unrealized due to challenges such as antigen selection, tumor-mediated immunosuppression, and optimization of delivery systems. This review provides a comprehensive analysis of the aims and implications of preventive and therapeutic cancer vaccine, the innovative discovery of neoantigens enhancing vaccine specificity, and the latest strides in vaccine delivery platforms. It also critically evaluates the role of adjuvants in enhancing immunogenicity and mitigating the immunosuppressive tumor microenvironment. The review further examines the synergistic potential of combining cancer vaccines with other therapies, such as chemotherapy, radiotherapy, and immune checkpoint inhibitors, to improve therapeutic outcomes. Overcoming barriers such as effective antigen identification, immunosuppressive microenvironments, and adverse effects is critical for advancing vaccine development. By addressing these challenges, cancer vaccines can offer significant improvements in patient outcomes and broaden the scope of personalized cancer immunotherapy.
Collapse
Affiliation(s)
- Wanting Lei
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Kexun Zhou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ye Lei
- College of Liberal Arts, Neijiang Normal University, Neijiang, 641100, Sichuan, China
| | - Qiu Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
2
|
Vitale E, Rizzo A, Maistrello L, Guven DC, Massafra R, Mollica V, Monteiro FSM, Santoni M, Massari F. Sex differences in adverse events among cancer patients receiving immune checkpoint inhibitors: the MOUSEION-07 systematic review and meta-analysis. Sci Rep 2024; 14:28309. [PMID: 39550353 PMCID: PMC11569249 DOI: 10.1038/s41598-024-71746-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/30/2024] [Indexed: 11/18/2024] Open
Abstract
Immune checkpoint inhibitors have revolutionized cancer treatment, but they are associated with a range of immune-related adverse events (irAEs), and emerging evidence suggests significant sex differences in the incidence, type, and severity of these toxicities, suggesting an influential factor and understanding sex-related differences in irAEs as crucial for optimizing patient care and improving clinical outcomes. In MOUSEION-07 study, we aimed to assess the association between sex and treatment-related adverse events in cancer patients treated with immunotherapy through a large up-to-date meta-analysis of available clinical trials. The present systematic review and meta-analysis was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-analysis. The protocol was registered with PROSPERO no. CRD42024549518. Sixteen studies encompassing a total of 4658 patients were included, and 2133 adverse effects were highlighted. The analysis observed a not statistically significant difference in terms of immune-related adverse events (irAEs) between males and females (Odds Ratio 1.19; CI 0.88-1.63) and revealed the presence of publication bias (β = -2.53; 95% CI = [-4.03; -1.04]; P = 0.006). Sex differences in immunotherapy-related adverse events are a significant factor in cancer treatment, necessitating a personalized approach to patient care. Further research is needed to fully understand the mechanisms driving these differences and to develop optimized strategies for monitoring and managing irAEs in both females and males.
Collapse
Affiliation(s)
- Elsa Vitale
- Scientific Directorate, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Alessandro Rizzo
- S.S.D. C.O.R.O. Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco 65, 70124, Bari, Italy.
| | | | - Deniz Can Guven
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Raffaella Massafra
- Scientific Directorate, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Fernando Sabino Marques Monteiro
- Latin American Cooperative Oncology Group - LACOG, Porto Alegre, RS, Brazil
- Oncology and Hematology Department, Hospital Sírio-Libanês, Brasilia, DF, Brazil
| | | | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| |
Collapse
|
3
|
Boucher R, Haigh O, Barreau E, Champiat S, Lambotte O, Adam C, Labetoulle M, Rousseau A. Ocular surface toxicities associated with modern anticancer therapies. Surv Ophthalmol 2024; 69:198-210. [PMID: 37806566 DOI: 10.1016/j.survophthal.2023.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/25/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023]
Abstract
Cancer treatments have recently shifted from broad-spectrum cytotoxic therapies to more focused treatments, maximizing anticancerous activity while reducing toxicity to healthy cells. These modern anticancer therapies (MATs) encompass a wide range of innovative molecules that include immune checkpoint inhibitors and other targeted anticancer therapies, comprising antibody drug conjugates and inhibitors of signal transduction. Some MATs are associated with ocular surface adverse events that can cause severe discomfort and even lead to loss of vision. While these complications remain rare, they are probably underreported. It is likely that both oncologists and ophthalmologists will come across MATs-associated ocular surface-adverse events in their practices, owing to the increasing number of patients being treated with MATs. Rapid identification of ocular surface-adverse events is crucial, as early intervention can manage these conditions to avoid vision loss and reduce negative impacts on quality of life. We discuss characteristics of ocular surface pathologies attributed to MATs, describe the suspected underlying pathophysiological mechanisms, and outline the main lines of treatment.
Collapse
Affiliation(s)
- Rafael Boucher
- Service d'Ophtalmologie, Assistance Publique Hôpitaux de Paris (AP-HP), Université Paris-Saclay. Centre de Référence pour les maladies rares en ophtalmologie (OPHTARA), Le Kremlin-Bicêtre, France; Department of Immunology of Viral and Auto-immune Disease (IMVA DSV/iMETI/IDMIT), UMR1184, CEA, Fontenay-aux-Roses, France
| | - Oscar Haigh
- Department of Immunology of Viral and Auto-immune Disease (IMVA DSV/iMETI/IDMIT), UMR1184, CEA, Fontenay-aux-Roses, France
| | - Emmanuel Barreau
- Service d'Ophtalmologie, Assistance Publique Hôpitaux de Paris (AP-HP), Université Paris-Saclay. Centre de Référence pour les maladies rares en ophtalmologie (OPHTARA), Le Kremlin-Bicêtre, France
| | - Stéphane Champiat
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Villejuif, France
| | - Olivier Lambotte
- Department of Immunology of Viral and Auto-immune Disease (IMVA DSV/iMETI/IDMIT), UMR1184, CEA, Fontenay-aux-Roses, France; Department of Internal Medicine and Immunology, Assistance Publique Hôpitaux de Paris (AP-HP), Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Clovis Adam
- Department of Pathology, Assistance Publique Hôpitaux de Paris (AP-HP), Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Marc Labetoulle
- Service d'Ophtalmologie, Assistance Publique Hôpitaux de Paris (AP-HP), Université Paris-Saclay. Centre de Référence pour les maladies rares en ophtalmologie (OPHTARA), Le Kremlin-Bicêtre, France; Department of Immunology of Viral and Auto-immune Disease (IMVA DSV/iMETI/IDMIT), UMR1184, CEA, Fontenay-aux-Roses, France
| | - Antoine Rousseau
- Service d'Ophtalmologie, Assistance Publique Hôpitaux de Paris (AP-HP), Université Paris-Saclay. Centre de Référence pour les maladies rares en ophtalmologie (OPHTARA), Le Kremlin-Bicêtre, France; Department of Immunology of Viral and Auto-immune Disease (IMVA DSV/iMETI/IDMIT), UMR1184, CEA, Fontenay-aux-Roses, France.
| |
Collapse
|
4
|
Liu Y, Lu L, Yang H, Wu X, Luo X, Shen J, Xiao Z, Zhao Y, Du F, Chen Y, Deng S, Cho CH, Li Q, Li X, Li W, Wang F, Sun Y, Gu L, Chen M, Li M. Dysregulation of immunity by cigarette smoking promotes inflammation and cancer: A review. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 339:122730. [PMID: 37838314 DOI: 10.1016/j.envpol.2023.122730] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/26/2023] [Accepted: 10/11/2023] [Indexed: 10/16/2023]
Abstract
Smoking is a serious global health issue. Cigarette smoking contains over 7000 different chemicals. The main harmful components include nicotine, acrolein, aromatic hydrocarbons and heavy metals, which play the key role for cigarette-induced inflammation and carcinogenesis. Growing evidences show that cigarette smoking and its components exert a remarkable impact on regulation of immunity and dysregulated immunity promotes inflammation and cancer. Therefore, this comprehensive and up-to-date review covers four interrelated topics, including cigarette smoking, inflammation, cancer and immune system. The known harmful chemicals from cigarette smoking were summarized. Importantly, we discussed in depth the impact of cigarette smoking on the formation of inflammatory or tumor microenvironment, primarily by affecting immune effector cells, such as macrophages, neutrophils, and T lymphocytes. Furthermore, the main molecular mechanisms by which cigarette smoking induces inflammation and cancer, including changes in epigenetics, DNA damage and others were further summarized. This article will contribute to a better understanding of the impact of cigarette smoking on inducing inflammation and cancer.
Collapse
Affiliation(s)
- Yubin Liu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
| | - Lan Lu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Huan Yang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Xinyue Luo
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Shuai Deng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Chi Hin Cho
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
| | - Qianxiu Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
| | - Xiaobing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Wanping Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Fang Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yuhong Sun
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Li Gu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Meijuan Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China.
| |
Collapse
|
5
|
Kumar PPP, Lim DK. Photothermal Effect of Gold Nanoparticles as a Nanomedicine for Diagnosis and Therapeutics. Pharmaceutics 2023; 15:2349. [PMID: 37765317 PMCID: PMC10534847 DOI: 10.3390/pharmaceutics15092349] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Gold nanoparticles (AuNPs) have received great attention for various medical applications due to their unique physicochemical properties. AuNPs with tunable optical properties in the visible and near-infrared regions have been utilized in a variety of applications such as in vitro diagnostics, in vivo imaging, and therapeutics. Among the applications, this review will pay more attention to recent developments in diagnostic and therapeutic applications based on the photothermal (PT) effect of AuNPs. In particular, the PT effect of AuNPs has played an important role in medical applications utilizing light, such as photoacoustic imaging, photon polymerase chain reaction (PCR), and hyperthermia therapy. First, we discuss the fundamentals of the optical properties in detail to understand the background of the PT effect of AuNPs. For diagnostic applications, the ability of AuNPs to efficiently convert absorbed light energy into heat to generate enhanced acoustic waves can lead to significant enhancements in photoacoustic signal intensity. Integration of the PT effect of AuNPs with PCR may open new opportunities for technological innovation called photonic PCR, where light is used to enable fast and accurate temperature cycling for DNA amplification. Additionally, beyond the existing thermotherapy of AuNPs, the PT effect of AuNPs can be further applied to cancer immunotherapy. Controlled PT damage to cancer cells triggers an immune response, which is useful for obtaining better outcomes in combination with immune checkpoint inhibitors or vaccines. Therefore, this review examines applications to nanomedicine based on the PT effect among the unique optical properties of AuNPs, understands the basic principles, the advantages and disadvantages of each technology, and understands the importance of a multidisciplinary approach. Based on this, it is expected that it will help understand the current status and development direction of new nanoparticle-based disease diagnosis methods and treatment methods, and we hope that it will inspire the development of new innovative technologies.
Collapse
Affiliation(s)
| | - Dong-Kwon Lim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea;
- Department of Integrative Energy Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
- Brain Science Institute, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| |
Collapse
|
6
|
Chen L, Xu N, Wang P, Zhu H, Zhang Z, Yang Z, Zhang W, Guo H, Lin J. Nanoalbumin-prodrug conjugates prepared via a thiolation-and-conjugation method improve cancer chemotherapy and immune checkpoint blockade therapy by promoting CD8 + T-cell infiltration. Bioeng Transl Med 2023; 8:e10377. [PMID: 36684090 PMCID: PMC9842047 DOI: 10.1002/btm2.10377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/07/2022] [Accepted: 07/16/2022] [Indexed: 01/25/2023] Open
Abstract
Protein-drug conjugates are emerging tools to combat cancers. Here, we adopted an indirect thiolation-and-conjugation method as a general strategy to prepare protein-drug conjugates. We found for the first time that this method led to the formation of nanometric conjugates, probably due to the formation of intermolecular disulfide bonds, which facilitated enhanced uptake by cancer cells. As a proof-of-concept application in cancer therapy, a nanometric albumin-doxorubicin prodrug conjugate (NanoAlb-proDOX) was prepared. The nanometric size promoted its uptake by cancer cells, and the prodrug characteristic defined its selective cytotoxicity toward cancer cells in vitro and reduced side effects in vivo. In multiple tumor xenograft models, nanometric NanoAlb-proDOX showed superior antitumor activity and synergy with immune checkpoint blockade, probably due to the synergistically enhanced tumor CD8+ T-cell infiltration and activation. Hence, the thiolation-and-conjugation strategy may serve as a generally applicable method for preparing drug conjugates, and the proof-of-concept nanometric albumin-doxorubicin conjugate may be a good choice for antitumor therapy with the ability to co-stimulate the efficacy of immune checkpoint blockade.
Collapse
Affiliation(s)
- Long Chen
- Department of PharmacyPeking University Third Hospital, College of Chemistry and Molecular Engineering, Peking UniversityBeijingChina
| | - Nuo Xu
- Department of PharmacyPeking University Third Hospital, College of Chemistry and Molecular Engineering, Peking UniversityBeijingChina
| | - Pan Wang
- Department of Obstetrics and GynecologyPeking University Third HospitalBeijingChina
| | - Haichuan Zhu
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and TechnologyWuhanChina
| | - Zijian Zhang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and TechnologyWuhanChina
| | - Zhanqun Yang
- Department of PharmacyPeking University Third Hospital, College of Chemistry and Molecular Engineering, Peking UniversityBeijingChina
| | - Wenyuan Zhang
- Department of PharmacyPeking University Third Hospital, College of Chemistry and Molecular Engineering, Peking UniversityBeijingChina
| | - Hongyan Guo
- Department of Obstetrics and GynecologyPeking University Third HospitalBeijingChina
| | - Jian Lin
- Department of PharmacyPeking University Third Hospital, College of Chemistry and Molecular Engineering, Peking UniversityBeijingChina
| |
Collapse
|
7
|
Gao W, Wu L, Jin S, Li J, Liu X, Xu J, Zhang W, Gong Q, Sun C, Wang W, Wang Z, Shao YW, Yin JC, Shen L, Chen L, Wang Q, Guo R. Rechallenge of immune checkpoint inhibitors in a case with adverse events inducing myasthenia gravis. J Immunother Cancer 2022; 10:jitc-2022-005970. [PMID: 36450378 PMCID: PMC9716945 DOI: 10.1136/jitc-2022-005970] [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] [Accepted: 10/21/2022] [Indexed: 12/02/2022] Open
Abstract
The mechanism(s) of immune checkpoint inhibitor (ICI)-induced myasthenia gravis (MG), an immune-related adverse event (irAE) that is fatal and limits subsequent ICI use, remain unexplored. Here, through comparative genomic analysis, we identified a pathogenic p.S467C germline variant in SLC22A5 in a thymoma case with ICI-induced MG, which was found to be associated with fatty acid oxidation through its regulation on L-carnitine levels. Remarkably, ICI rechallenge with L-carnitine pretreatment led to durable response without MG-related symptoms. Thus, we provide the first clinical evidence of genetic test-directed irAE management, which integrates individualized ICI treatment into the evolving paradigm of cancer management.
Collapse
Affiliation(s)
- Wen Gao
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lingxiang Wu
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China,Department of Bioinformatics, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shidai Jin
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jun Li
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xinyin Liu
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiali Xu
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wei Zhang
- Department of Radiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qixing Gong
- Department of Pathology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chunxiao Sun
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wei Wang
- Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zidun Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yang W Shao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, Jiangsu, China,School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiani C Yin
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, Jiangsu, China
| | - Lu Shen
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, Jiangsu, China
| | - Liang Chen
- Department of Thoracic Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qianghu Wang
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China,Department of Bioinformatics, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Renhua Guo
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
8
|
Ocular Inflammation Induced by Immune Checkpoint Inhibitors. J Clin Med 2022; 11:jcm11174993. [PMID: 36078923 PMCID: PMC9456546 DOI: 10.3390/jcm11174993] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Ocular immunotherapy-related adverse events (IRAEs), although rare, can be sight-threatening. Our objective was to analyze ocular IRAEs diagnosed in France from the marketing of immune checkpoint inhibitors (ICPIs) until June 2021 and to review the literature. We collected the cases of 28 patients (36 ocular IRAEs), occurring after an average of 17 weeks (±19). Forty-six percent of patients were treated for metastatic melanoma. Anti-PD1 agents were responsible for 57% of the IRAEs. Anterior uveitis was the most common (44%), followed by panuveitis (28%). Of 25 uveitis cases, 80% were bilateral and 60% were granulomatous. We found one case with complete Vogt-Koyanagi–Harada syndrome and one case of birdshot retinochoroidopathy. The other IRAEs were eight ocular surface disorders, one optic neuropathy, and one inflammatory orbitopathy. Seventy percent of the IRAEs were grade 3 according to the common terminology of AEs. ICPIs were discontinued in 60% of patients and 50% received local corticosteroids alone. The literature review included 230 uveitis cases, of which 7% were granulomatous. The distributions of ICPIs, cancer, and type of uveitis were similar to our cohort. Ocular IRAEs appeared to be easily controlled by local or systemic corticosteroids and did not require routine discontinuation of ICPIs. Further work is still warranted to define the optimal management of ocular IRAEs.
Collapse
|
9
|
V. Lima B, Oliveira MJ, Barbosa MA, Gonçalves RM, Castro F. Harnessing chitosan and poly-(γ-glutamic acid)-based biomaterials towards cancer immunotherapy. MATERIALS TODAY ADVANCES 2022; 15:100252. [DOI: 10.1016/j.mtadv.2022.100252] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
10
|
Li H, Sahu KK, Maughan BL. Mechanism and Management of Checkpoint Inhibitor-Related Toxicities in Genitourinary Cancers. Cancers (Basel) 2022; 14:2460. [PMID: 35626064 PMCID: PMC9139183 DOI: 10.3390/cancers14102460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 12/10/2022] Open
Abstract
The use of immune checkpoint inhibitors (ICIs) is rapidly increasing as more combinations and clinical indications are approved in the field of genitourinary malignancies. Most immunotherapeutic agents being approved are for the treatment of renal cell carcinoma and bladder cancer, which mainly involve PD-1/PD-L1 and CTLA-4 pathways. There is an ongoing need for recognizing and treating immunotherapy-related autoimmune adverse effects (irAEs). This review aims to critically appraise the recent literature on the mechanism, common patterns, and treatment recommendations of irAEs in genitourinary malignancies. We review the epidemiology of these adverse effects as well as general treatment strategies. The underlying mechanisms will also be discussed. Diagnostic considerations including differential diagnosis are also included in this review.
Collapse
Affiliation(s)
| | | | - Benjamin L. Maughan
- Division of Medical Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84108, USA; (H.L.); (K.K.S.)
| |
Collapse
|
11
|
Hui G, Drolen C, Hannigan CA, Drakaki A. Treatment Equity in the Immunotherapy Era: Options for Patients with Both Autoimmune Disease and GU Cancers. Life (Basel) 2022; 12:360. [PMID: 35330111 PMCID: PMC8949122 DOI: 10.3390/life12030360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/31/2022] [Accepted: 02/22/2022] [Indexed: 11/16/2022] Open
Abstract
Numerous immunotherapeutic agents, such as immune checkpoint inhibitors (ICIs), have been approved for the treatment of genitourinary (GU) malignancies. While ICIs have improved treatment outcomes and expanded treatment options, they can cause immune-related adverse events (irAEs). The scope of irAEs is broad, and this paper aims to review the rheumatologic side effects associated with immunotherapy drugs approved for bladder cancer and renal cell carcinoma. IrAEs are graded by the common terminology criteria for adverse events (CTCAE), which ranges from 1 to 5. The management of irAEs includes corticosteroids or other immunosuppressive therapies, and it may require discontinuation of immunotherapy. Several real world experience studies suggest that most patients with pre-existing autoimmune diseases treated with ICI did not have to discontinue treatment due to immune-mediated side effects. While data suggest autoimmune side effects are manageable, patients with pre-existing autoimmune diseases are often excluded from immunotherapy clinical trials. Better understanding of these irAEs will improve its safety and expand its use in those with underlying autoimmune disease.
Collapse
Affiliation(s)
- Gavin Hui
- Department of Medicine, Stanford University, Stanford, CA 94305, USA;
| | - Claire Drolen
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA;
| | - Christopher A. Hannigan
- Department of Medicine, Division of Hematology/Oncology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA;
| | - Alexandra Drakaki
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA;
- Department of Medicine, Division of Hematology/Oncology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA;
| |
Collapse
|
12
|
Villa-Crespo L, Podlipnik S, Anglada N, Izquierdo C, Giavedoni P, Iglesias P, Dominguez M, Aya F, Arance A, Malvehy J, Puig S, Carrera C. Timeline of Adverse Events during Immune Checkpoint Inhibitors for Advanced Melanoma and Their Impacts on Survival. Cancers (Basel) 2022; 14:cancers14051237. [PMID: 35267545 PMCID: PMC8909485 DOI: 10.3390/cancers14051237] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/13/2022] [Accepted: 02/19/2022] [Indexed: 02/05/2023] Open
Abstract
Immune-related adverse events (irAEs) are frequent and could be associated with improved response to immune checkpoint inhibitors (ICIs). A prospective cohort of advanced melanoma patients receiving ICI as first-line therapy was retrospectively reviewed (January 2011−February 2019). A total of 116 of 153 patients presented with at least one irAE (75.8%). The most frequent irAEs were dermatological (derm irAEs, 50%), asthenia (38%), and gastrointestinal (29%). Most irAEs appeared within the first 90 days, while 11.2% appeared after discontinuation of the therapy. Mild grade 1−2 derm irAEs tended to appear within the first 2 months of therapy with a median time of 65.5 days (IQR 26-139.25), while grade 3−4 derm irAEs appeared later (median 114 days; IQR 69-218) and could be detected at any time during therapy. Only derm irAE occurrence was related to improved survival (HR 6.46). Patients presenting derm irAEs showed better 5-year overall survival compared to those with no derm irAEs (53.1% versus 24.9%; p < 0.001). However, the difference was not significant when adjusting for the duration of therapy. In conclusion: the timeline of immune-related-AEs differs according to the organ involved. The (apparent) improved survival of patients who present derm AEs during immunotherapy could be partially explained by longer times under treatment.
Collapse
Affiliation(s)
- Lorena Villa-Crespo
- Melanoma Group, Institut d’Investigacions Biomediques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain; (L.V.-C.); (S.P.); (P.G.); (P.I.); (M.D.); (J.M.); (S.P.)
| | - Sebastian Podlipnik
- Melanoma Group, Institut d’Investigacions Biomediques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain; (L.V.-C.); (S.P.); (P.G.); (P.I.); (M.D.); (J.M.); (S.P.)
| | - Natalia Anglada
- Medicine Department, Medicine Faculty, Campus Clínic, University of Barcelona, 08036 Barcelona, Spain; (N.A.); (C.I.)
| | - Clara Izquierdo
- Medicine Department, Medicine Faculty, Campus Clínic, University of Barcelona, 08036 Barcelona, Spain; (N.A.); (C.I.)
| | - Priscila Giavedoni
- Melanoma Group, Institut d’Investigacions Biomediques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain; (L.V.-C.); (S.P.); (P.G.); (P.I.); (M.D.); (J.M.); (S.P.)
| | - Pablo Iglesias
- Melanoma Group, Institut d’Investigacions Biomediques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain; (L.V.-C.); (S.P.); (P.G.); (P.I.); (M.D.); (J.M.); (S.P.)
| | - Mireia Dominguez
- Melanoma Group, Institut d’Investigacions Biomediques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain; (L.V.-C.); (S.P.); (P.G.); (P.I.); (M.D.); (J.M.); (S.P.)
| | - Francisco Aya
- Medical Oncology Department, Hospital Clinic of Barcelona, University of Barcelona, 08036 Barcelona, Spain; (F.A.); (A.A.)
| | - Ana Arance
- Medical Oncology Department, Hospital Clinic of Barcelona, University of Barcelona, 08036 Barcelona, Spain; (F.A.); (A.A.)
| | - Josep Malvehy
- Melanoma Group, Institut d’Investigacions Biomediques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain; (L.V.-C.); (S.P.); (P.G.); (P.I.); (M.D.); (J.M.); (S.P.)
- Medicine Department, Medicine Faculty, Campus Clínic, University of Barcelona, 08036 Barcelona, Spain; (N.A.); (C.I.)
- Biomedical Research Networking Center on Rare Diseases (CIBERER), ISCIII, 08036 Barcelona, Spain
| | - Susana Puig
- Melanoma Group, Institut d’Investigacions Biomediques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain; (L.V.-C.); (S.P.); (P.G.); (P.I.); (M.D.); (J.M.); (S.P.)
- Medicine Department, Medicine Faculty, Campus Clínic, University of Barcelona, 08036 Barcelona, Spain; (N.A.); (C.I.)
- Biomedical Research Networking Center on Rare Diseases (CIBERER), ISCIII, 08036 Barcelona, Spain
| | - Cristina Carrera
- Melanoma Group, Institut d’Investigacions Biomediques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain; (L.V.-C.); (S.P.); (P.G.); (P.I.); (M.D.); (J.M.); (S.P.)
- Medicine Department, Medicine Faculty, Campus Clínic, University of Barcelona, 08036 Barcelona, Spain; (N.A.); (C.I.)
- Biomedical Research Networking Center on Rare Diseases (CIBERER), ISCIII, 08036 Barcelona, Spain
- Correspondence: ; Tel.: +34-93-2275400
| |
Collapse
|
13
|
Homan D, Ruissen-Luijt P, Boekhout A, Maaskant J. A historic cohort study of nutritional status related side effects and weight loss of cancer patients treated with immunotherapy. Clin Nutr ESPEN 2022; 47:163-169. [DOI: 10.1016/j.clnesp.2021.12.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/24/2022]
|
14
|
Sun MM, Seleme N, Chen JJ, Zekeridou A, Sechi E, Walsh RD, Beebe JD, Sabbagh O, Mejico LJ, Gratton S, Skidd PM, Bellows DA, Falardeau J, Fraser CL, Cappelen-Smith C, Haines SR, Hassanzadeh B, Seay MD, Subramanian PS, Williams Z, Gordon LK. Neuro-Ophthalmic Complications in Patients Treated With CTLA-4 and PD-1/PD-L1 Checkpoint Blockade. J Neuroophthalmol 2021; 41:519-530. [PMID: 33136674 DOI: 10.1097/wno.0000000000001148] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND In recent years, CTLA-4 and PD-1/PD-L1 checkpoint inhibitors have proven to be effective and have become increasingly popular treatment options for metastatic melanoma and other cancers. These agents work by enhancing autologous antitumor immune responses. Immune-related ophthalmologic complications have been reported in association with checkpoint inhibitor use but remain incompletely characterized. This study seeks to investigate and further characterize the neuro-ophthalmic and ocular complications of immune checkpoint blockade treatment. METHODS A survey was distributed through the secure electronic data collection tool REDCap to neuro-ophthalmology specialists in the North American Neuro-Ophthalmology Society listserv. The study received human subjects approval through the University of California at Los Angeles Institutional Review Board. The survey identified patients sent for neuro-ophthalmic consultation while receiving one or more of a PD-1 inhibitor (pembrolizumab, nivolumab, or cemiplimab); PD-L1 inhibitor (atezolizumab, avelumab, or durvalumab); or the CTLA-4 inhibitor ipilimumab. Thirty-one patients from 14 institutions were identified. Patient demographics, neuro-ophthalmic diagnosis, diagnostic testing, severity, treatment, clinical response, checkpoint inhibitor drug used, and cancer diagnosis was obtained. RESULTS The checkpoint inhibitors used in these patients included pembrolizumab (12/31), nivolumab (6/31), combined ipilimumab with nivolumab (7/31, one of whom also received pembrolizumab during their course of treatment), durvalumab (3/31), ipilimumab (2/31), and cemiplimab (1/31). Malignant melanoma (16/31) or nonsmall cell lung carcinoma (6/31) were the most common malignancies. The median time between first drug administration and the time of ophthalmological symptom onset was 14.5 weeks. Eleven patients had involvement of the optic nerve, 7 patients had inflammatory orbital or extraocular muscle involvement, 6 patients had ocular involvement from neuromuscular junction dysfunction, 4 patients had cranial nerve palsy, and 4 patients had non neuro-ophthalmic complications. Use of systemic corticosteroids with or without stopping the checkpoint inhibitor resulted in improvement of most patients with optic neuropathy, and variable improvement for the other ophthalmic conditions. CONCLUSION This study describes the variable neuro-ophthalmic adverse events associated with use of immune checkpoint inhibitors and contributes a more thorough understanding of their clinical presentations and treatment outcomes. We expect this will increase awareness of these drug complications and guide specialists in the care of these patients.
Collapse
Affiliation(s)
- Michel M Sun
- Department of Ophthalmology (MMS, NS, LKG), Jules Stein Eye Institute, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California; Department of Ophthalmology (JJC), Mayo Clinic, Rochester, Minnesota; Department of Neurology (JJC, AZ, ES), Mayo Clinic, Rochester, Minnesota; Department of Ophthalmology & Visual Sciences (RDW), Medical College of Wisconsin, Milwaukee, Wisconsin; Department of Ophthalmology (JDB), Park Nicollet Health Services, Minneapolis, Minnesota; Department of Ophthalmology (OS), University of Kentucky/Retina Associates of Kentucky, Lexington, Kentucky; Department of Neurology (LJM), SUNY Upstate Medical University, Syracuse, New York; Department of Ophthalmology (LJM), SUNY Upstate Medical University, Syracuse, New York; Department of Neurology (SG), University of Missouri-Kansas City, Kansas City, Missouri; Department of Ophthalmology (PMS), University of Vermont Medical Center, Burlington, Vermont; The Medical Eye Center (DAB), Manchester, New Hampshire; Department of Ophthalmology (JF), Oregon Health & Science University, Portland, Oregon; Department of Ophthalmology (CLF), University of Sydney, Sydney, Australia ; Department of Neurology & Neurophysiology (CC-S), Liverpool Hospital, NSW, Australia ; Department of Neurology (SRH), Virginia Commonwealth University School of Medicine, Richmond, Virginia; Department of Neurology (BH), INI Eye Center, OSF Healthcare, University of Illinois College of Medicine, Peoria, Illinois; Department of Ophthalmology (MDS), University of Utah, Salt Lake City, Utah; Department of Ophthalmology (PSS), University of Colorado, Aurora, Colorado; and Department of Ophthalmology (ZW), University of Rochester Medical Center, Rochester, New York
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Mineiro dos Santos Garrett NF, Carvalho da Costa AC, Barros Ferreira E, Damiani G, Diniz dos Reis PE, Inocêncio Vasques C. Prevalence of dermatological toxicities in patients with melanoma undergoing immunotherapy: Systematic review and meta-analysis. PLoS One 2021; 16:e0255716. [PMID: 34358260 PMCID: PMC8345892 DOI: 10.1371/journal.pone.0255716] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 07/22/2021] [Indexed: 12/18/2022] Open
Abstract
Background Checkpoint inhibitors have revolutionized advanced melanoma care; however, their cutaneous side effects have not been definitively elucidated. Objective To identify the prevalence of cutaneous toxicity in patients with melanoma treated with immune checkpoint inhibitors as monotherapy and/or in combination with chemotherapy and/or radiotherapy. Materials and methods We performed a systematic review and meta-analysis, which encompassed both clinical trials and observational studies describing the dermatological toxicities in patients treated with immune checkpoint inhibitors. The protocol was registered in the International Prospective Register of Systematic Review under the number CRD42018091915. The searches were performed using the CINAHL, Cochrane CENTRAL, LILACS, LIVIVO, PubMed, Scopus, and Web of Science databases. The methodological quality of the studies was evaluated with the JBI Critical Appraisal Checklist for Studies Reporting Prevalence Data Results A total of 9,802 articles were identified in the databases. The final sample comprised 39 studies. The evaluated drugs were ipilimumab, tremelimumab, pembrolizumab, and nivolumab. The results suggest that the most prevalent side effect was grade 1 and 2 pruritus (24%), followed by grade 1 and 2 rash (21%) and grade 1 and 2 vitiligo (10%). Conclusion The most prevalent side effects in patients treated with checkpoint inhibitors are pruritus, rash, and vitiligo, and they are rated mostly as grades 1 and 2 adverse events. Remarkably, vitiligo is most commonly found in patients treated with PD-1 inhibitors.
Collapse
Affiliation(s)
| | | | | | - Giovanni Damiani
- Clinical Dermatology, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
- Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States of America
- Department of Drug Sciences, University of Padua, Padua, Italy
| | | | | |
Collapse
|
16
|
Kaźmierczak-Siedlecka K, Roviello G, Catalano M, Polom K. Gut Microbiota Modulation in the Context of Immune-Related Aspects of Lactobacillus spp. and Bifidobacterium spp. in Gastrointestinal Cancers. Nutrients 2021; 13:2674. [PMID: 34444834 PMCID: PMC8401094 DOI: 10.3390/nu13082674] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 07/16/2021] [Accepted: 07/30/2021] [Indexed: 12/17/2022] Open
Abstract
Accumulating evidence has revealed the critical roles of commensal microbes in cancer progression and recently several investigators have evaluated the therapeutic effectiveness of targeting the microbiota. This gut microbiota-related approach is especially attractive in the treatment of gastrointestinal cancers. Probiotics supplementation is a microbiota-targeted strategy that appears to improve treatment efficacy; Lactobacillus spp. and Bifidobacterium spp. are among the most commonly used probiotic agents. These bacteria seem to exert immunomodulatory effects, impacting on the immune system both locally and systemically. The gut microbiota are able to affect the efficiency of immunotherapy, mainly acting as inhibitors at immune checkpoints. The effects of immunotherapy may be modulated using traditional probiotic strains and/or next generation probiotics, such as Akkermansia municiphila. It is possible that probiotics might enhance the efficiency of immunotherapy based on PD-1/PD-L1 and CTLA-4 but more data are needed to confirm this speculation. Indeed, although there is experimental evidence for the efficacy of several strains, the health-promoting effects of numerous probiotics have not been demonstrated in human patients and furthermore the potential risks of these products, particularly in oncologic patients, are rarely mentioned.
Collapse
Affiliation(s)
| | - Giandomenico Roviello
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy; (G.R.); (M.C.)
| | - Martina Catalano
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy; (G.R.); (M.C.)
| | - Karol Polom
- Department of Surgical Oncology, Medical University of Gdansk, 80-210 Gdańsk, Poland;
| |
Collapse
|
17
|
Kato T, Wakiyama H, Furusawa A, Choyke PL, Kobayashi H. Near Infrared Photoimmunotherapy; A Review of Targets for Cancer Therapy. Cancers (Basel) 2021; 13:cancers13112535. [PMID: 34064074 PMCID: PMC8196790 DOI: 10.3390/cancers13112535] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/12/2021] [Accepted: 05/18/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Near-infrared photoimmunotherapy (NIR-PIT) is a newly developed cancer treatment that uses an antibody-photoabsorber (IRDye700DX) conjugate (APC) that is activated by NIR light irradiation. A major benefit of NIR-PIT is that only APC-bound cancer cells that are exposed to NIR light are killed by NIR-PIT; thus, minimal damage occurs in adjacent normal cells. NIR-PIT has now been applied to many cancers expressing various cell-surface target proteins using monoclonal antibodies designed to bind to them. Moreover, NIR-PIT is not limited to tumor antigens but can also be used to kill specific host cells that create immune-permissive environments in which tumors grow. Moreover, multiple targets can be treated simultaneously with NIR-PIT using a cocktail of APCs. NIR-PIT has great potential to treat a wide variety of cancers by targeting appropriate tumor cells, immune cells, or both, and can be augmented by other immunotherapies. Abstract Near-infrared photoimmunotherapy (NIR-PIT) is a newly developed cancer treatment that uses an antibody-photoabsorber (IRDye700DX) conjugate (APC) that is activated by NIR light irradiation. In September 2020, the first APC and laser system were conditionally approved for clinical use in Japan. A major benefit of NIR-PIT is that only APC-bound cancer cells that are exposed to NIR light are killed by NIR-PIT; thus, minimal damage occurs in adjacent normal cells. These early trials have demonstrated that in addition to direct cell killing, there is a significant therapeutic host immune response that greatly contributes to the success of the therapy. Although the first clinical use of NIR-PIT targeted epidermal growth factor receptor (EGFR), many other targets are suitable for NIR-PIT. NIR-PIT has now been applied to many cancers expressing various cell-surface target proteins using monoclonal antibodies designed to bind to them. Moreover, NIR-PIT is not limited to tumor antigens but can also be used to kill specific host cells that create immune-permissive environments in which tumors grow. Moreover, multiple targets can be treated simultaneously with NIR-PIT using a cocktail of APCs. NIR-PIT can be used in combination with other therapies, such as immune checkpoint inhibitors, to enhance the therapeutic effect. Thus, NIR-PIT has great potential to treat a wide variety of cancers by targeting appropriate tumor cells, immune cells, or both, and can be augmented by other immunotherapies.
Collapse
|
18
|
Braun D, Getahun D, Chiu VY, Coleman AL, Holland GN, Yu F, Gordon LK, Sun MM. Population-Based Frequency of Ophthalmic Adverse Events in Melanoma, Other Cancers, and After Immune Checkpoint Inhibitor Treatment. Am J Ophthalmol 2021; 224:282-291. [PMID: 33359682 DOI: 10.1016/j.ajo.2020.12.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE To examine the frequency of ophthalmic immune-related adverse events (OirAEs) in melanoma, other cancers, and after immune checkpoint inhibitor (ICI) treatment. DESIGN Retrospective clinical cohort study. METHODS This study identified patients diagnosed with OirAEs between January 1, 2011, and December 31, 2018, in the Kaiser Permanente Southern California electronic health records. The primary exposures of interest were prior initiation of ICIs and underlying cancer diagnosis. Risk-adjusted prevalence of OirAEs was evaluated in patients with melanoma, with nonmelanoma cancer, and without cancer. The 1-year incidence of OirAEs and recurrence of prior ophthalmic disease were identified in ICI-receiving patients with melanoma and nonmelanoma. RESULTS Among 4,695,669 unique patients identified, 9.9% had a cancer diagnosis, of whom 2.8% had a diagnosis of melanoma. Overall prevalence for uveitis and selected neuro-ophthalmic diagnoses was 341.8/100,000 patient-years in patients with melanoma and 369.6/100,000 patient-years in patients with nonmelanoma cancer regardless of ICI treatment, compared with 142.2/100,000 patient-years in patients without cancer. A total of 2,911 unique patients received ICI therapy. Compared with patients with nonmelanoma cancer, patients with melanoma on any ICI had elevated 1-year incidence rates of uveitis (1.2% vs 0.2%; risk-adjusted odds ratio, 6.45). High 1-year recurrence rates for uveitis in ICI patients with a prior uveitis history were also observed. CONCLUSIONS The prevalence of all OirAEs was substantially higher in patients with cancer, with ICI-related uveitis risk specifically increased in patients with melanoma compared with patients with nonmelanoma cancer. Evidence-based guidelines for ophthalmic monitoring of patients undergoing ICI treatment may require different risk stratifications based on underlying cancer diagnosis, specific ICI used, and prior history of uveitis.
Collapse
Affiliation(s)
- David Braun
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, California, USA; Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, California, USA; Department of Pediatrics, Kaiser Permanente Panorama City, Panorama City, California, USA
| | - Darios Getahun
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, California, USA; Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, California, USA
| | - Vicki Y Chiu
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, California, USA
| | - Anne L Coleman
- UCLA Stein Eye Institute and the Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA; Jonathan and Karin Fielding School of Public Health, UCLA, Los Angeles, California, USA
| | - Gary N Holland
- UCLA Stein Eye Institute and the Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Fei Yu
- UCLA Stein Eye Institute and the Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA; Jonathan and Karin Fielding School of Public Health, UCLA, Los Angeles, California, USA
| | - Lynn K Gordon
- UCLA Stein Eye Institute and the Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Michel M Sun
- UCLA Stein Eye Institute and the Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.
| |
Collapse
|
19
|
Noronha V, Abraham G, Patil V, Joshi A, Menon N, Mahajan A, Janu A, Jain S, Talreja VT, Kapoor A, Kumar Singh G, Khaddar S, Gupta K, Rathinasamy N, Srinivas S, Agrawal A, Ventrapati P, Prabhash K. A real-world data of Immune checkpoint inhibitors in solid tumors from India. Cancer Med 2021; 10:1525-1534. [PMID: 33591635 PMCID: PMC7940210 DOI: 10.1002/cam4.3617] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/13/2020] [Accepted: 10/30/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Checkpoint inhibitors (Nivolumab and Pembrolizumab) are approved for multiple indications in solid tumors. However access to these therapies is limited in low and middle income countries. Hence we performed an audit to identify accessibility, adverse event rates, compliance, progression free survival and overall survival in solid tumors. METHODS This was a single center retrospective analysis of prospective data base of patients with non-melanoma solid tumors who were treated with immunotherapy from August 2015 to November 2018. Adverse events during immunotherapy were documented and graded using CTCAE (Common terminology criteria for adverse events), v. 4.02. The response rates to immunotherapy, toxicities and the time to onset and resolution of toxicities were also evaluated as secondary endpoints. RESULTS Out of 9610 patients, only 155 patients (1.61%) could receive immunotherapy. The most common malignancies included metastatic non-small cell lung cancer, metastatic renal cell carcinoma, metastatic urothelial carcinoma and relapsed/recurrent head and neck squamous cell carcinoma. Median overall survival in patients who received immunotherapy in non-melanoma solid malignancies was 5.37 months (95% CI, 3.73-9.73). Poor performance status at baseline was the only adverse prognostic factor. The median progression free survival was 2.57 months (95% CI, 1.73-3.83). Immunotherapy was well tolerated with most common side effects being fatigue 14.8% and anorexia 5.8%. The cumulative incidence of immune related adverse events like hepatitis, pneumonitis, colitis and nephritis was less than 10%. CONCLUSION Real-world data in Indian setting confirms the benefit of immunotherapy in patients with advanced non-melanoma solid tumors.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Anorexia/chemically induced
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/mortality
- Carcinoma, Renal Cell/drug therapy
- Carcinoma, Renal Cell/mortality
- Fatigue/chemically induced
- Female
- Head and Neck Neoplasms/drug therapy
- Head and Neck Neoplasms/mortality
- Humans
- Immune Checkpoint Inhibitors/adverse effects
- Immune Checkpoint Inhibitors/supply & distribution
- Immune Checkpoint Inhibitors/therapeutic use
- Immunotherapy, Adoptive/statistics & numerical data
- India
- Kidney Neoplasms/drug therapy
- Kidney Neoplasms/mortality
- Lung Neoplasms/drug therapy
- Lung Neoplasms/mortality
- Male
- Middle Aged
- Neoplasms/drug therapy
- Neoplasms/mortality
- Neoplasms/pathology
- Nivolumab/adverse effects
- Nivolumab/therapeutic use
- Progression-Free Survival
- Retrospective Studies
- Squamous Cell Carcinoma of Head and Neck/drug therapy
- Squamous Cell Carcinoma of Head and Neck/mortality
- Young Adult
Collapse
Affiliation(s)
- Vanita Noronha
- Department of Medical OncologyTata Memorial HospitalMumbaiIndia
| | - George Abraham
- Department of Medical OncologyTata Memorial HospitalMumbaiIndia
| | - Vijay Patil
- Department of Medical OncologyTata Memorial HospitalMumbaiIndia
| | - Amit Joshi
- Department of Medical OncologyTata Memorial HospitalMumbaiIndia
| | - Nandini Menon
- Department of Medical OncologyTata Memorial HospitalMumbaiIndia
| | | | - Amit Janu
- Department of RadiodiagnosisTata Memorial HospitalMumbaiIndia
| | - Srushti Jain
- Department of Medical OncologyTata Memorial HospitalMumbaiIndia
| | - Vikas T Talreja
- Department of Medical OncologyTata Memorial HospitalMumbaiIndia
| | - Akhil Kapoor
- Department of Medical OncologyTata Memorial HospitalMumbaiIndia
| | | | - Satvik Khaddar
- Department of Medical OncologyTata Memorial HospitalMumbaiIndia
| | - Kushal Gupta
- Department of Medical OncologyTata Memorial HospitalMumbaiIndia
| | | | - Sujay Srinivas
- Department of Medical OncologyTata Memorial HospitalMumbaiIndia
| | - Amit Agrawal
- Department of Medical OncologyTata Memorial HospitalMumbaiIndia
| | | | - Kumar Prabhash
- Department of Medical OncologyTata Memorial HospitalMumbaiIndia
| |
Collapse
|
20
|
Fenoglio R, Roccatello D, De Simone E, Del Vecchio G, Ferro M, Quattrocchio G, Barreca A, Sciascia S. The Challenging Management of Cancer: An Immunonephrologist's Perspective. Kidney Blood Press Res 2020; 46:114-120. [PMID: 33326976 DOI: 10.1159/000511256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/31/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Onconephrology is an emerging medical subspecialization that focuses on the numberless interrelations between cancer and kidney diseases. Tumor cells evade immune surveillance through activation of immune checkpoint pathways that suppress antitumor immune responses. By blocking checkpoints, new anticancer agents disrupt immune homeostasis but potentially induce immune-mediated diseases. Nephrologists and nephroimmunologists should be able to treat the nephrotoxic sequelae of cancer therapy and ensure continuation of the life-saving treatment. METHODS Thirty-seven renal biopsies have been carried out over 42 months in oncologic patients, that is, 5.2% of the total native renal biopsies were carried out in the same period. The commonest diagnoses (>6 cases) were interstitial tubular nephritis, membranous glomerulopathy, IgA nephropathy, vasculitis, and focal and segmental glomerulosclerosis. CASE PRESENTATION Three example cases, including focusing on key questions which could involve the nephrologists are reported in detail. They include a cancer-related Goodpasture Syndrome, the peculiar toxic effects of pemetrexed on tubular cells, and the intriguing relationship between bevacizumab and cryoglobulinemic glomerulonephritis. CONCLUSION As shown by these 3 example cases, nephrologists need to be open-minded with regard to kidney biopsy in order to get a timely diagnosis. Nephrologists also need to improve their knowledge of cancer biology and therapy in order to prevent kidney problems, manage therapy-related immune-mediated disorders, and improve patient life expectancy.
Collapse
Affiliation(s)
- Roberta Fenoglio
- Nephrology and Dialysis Unit & CMID (Center of Research of Immunopathology and Rare Diseases), Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hub Hospital of Turin, and Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Dario Roccatello
- Nephrology and Dialysis Unit & CMID (Center of Research of Immunopathology and Rare Diseases), Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hub Hospital of Turin, and Department of Clinical and Biological Sciences, University of Turin, Turin, Italy,
| | - Emanuele De Simone
- Nephrology and Dialysis Unit & CMID (Center of Research of Immunopathology and Rare Diseases), Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hub Hospital of Turin, and Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Giulio Del Vecchio
- Nephrology and Dialysis Unit & CMID (Center of Research of Immunopathology and Rare Diseases), Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hub Hospital of Turin, and Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Michela Ferro
- Nephrology and Dialysis Unit & CMID (Center of Research of Immunopathology and Rare Diseases), Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hub Hospital of Turin, and Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Giacomo Quattrocchio
- Nephrology and Dialysis Unit & CMID (Center of Research of Immunopathology and Rare Diseases), Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hub Hospital of Turin, and Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Antonella Barreca
- Division of Pathology, Città della Salute e della Scienza Hospital, Turin, Italy
| | - Savino Sciascia
- Nephrology and Dialysis Unit & CMID (Center of Research of Immunopathology and Rare Diseases), Coordinating Center of the Network for Rare Diseases of Piedmont and Aosta Valley, San Giovanni Bosco Hub Hospital of Turin, and Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| |
Collapse
|
21
|
Hamilton JA, Henry CJ. Aging and immunotherapies: New horizons for the golden ages. AGING AND CANCER 2020; 1:30-44. [PMID: 35874875 PMCID: PMC9307207 DOI: 10.1002/aac2.12014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The life expectancy of the world’s elderly population (65 and older) continues to reach new milestones with older individuals currently comprising greater than 8.5% (617 million) of the world’s population. This percentage is predicted to approach 20% of the world’s population by 2050 (representing 1.6 billion people). Despite this amazing feat, many healthcare systems are not equipped to handle the multitude of diseases that commonly manifest with age, including most types of cancers. As the world’s aging population grows, cancer treatments continue to evolve. Immunotherapies are a new drug class that has revolutionized our ability to treat previously intractable cancers; however, their efficacy in patients with compromised immune systems remains unclear. In this review, we will discuss how aging-associated losses in immune homeostasis impact the efficacy and safety of immunotherapy treatment in preclinical models of aging. We will also discuss how these findings translate to elderly patients receiving immunotherapy treatment for refractory and relapsed cancers, as well as, strategies that could be explored to improve the efficacy of immunotherapies in aged patients.
Collapse
Affiliation(s)
- Jamie A.G. Hamilton
- Department of Pediatrics Emory University School of Medicine Atlanta Georgia USA
- Aflac Cancer and Blood Disorders Center Children's Healthcare of Atlanta Atlanta Georgia USA
| | - Curtis J. Henry
- Department of Pediatrics Emory University School of Medicine Atlanta Georgia USA
- Aflac Cancer and Blood Disorders Center Children's Healthcare of Atlanta Atlanta Georgia USA
| |
Collapse
|
22
|
Sun MM, Kelly SP, Mylavarapu Bs AL, Holland GN, Coleman AL, Yu F, Hsu Ms S, Lum F, Gordon LK. Ophthalmic Immune-Related Adverse Events after Anti-CTLA-4 or PD-1 Therapy Recorded in the American Academy of Ophthalmology Intelligent Research in Sight Registry. Ophthalmology 2020; 128:910-919. [PMID: 33166553 DOI: 10.1016/j.ophtha.2020.11.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/21/2020] [Accepted: 11/02/2020] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Detailed study of ophthalmic immune-related adverse events (AEs), including determination of incidence and recurrence rates, is of integral importance in cancer immunotherapy to inform management and treatment guidelines. DESIGN Retrospective registry study. PARTICIPANTS Patients newly diagnosed with ophthalmic immune-related AEs between January 1, 2013, and December 31, 2017, in the American Academy of Ophthalmology's Intelligent Research in Sight (IRIS®) Registry. METHODS Data were collected from electronic health records of IRIS® Registry participating ophthalmology practices. Patients with select ophthalmic immune-related AEs were identified by International Classification of Diseases diagnosis codes. The primary exposure of interest was prior initiation of immune checkpoint inhibitors (ICIs). MAIN OUTCOME MEASURES Incidence of ophthalmic immune-related AEs within 1 year after initiation of ICI therapy was determined. Incidence rate ratios (IRRs) were derived by comparing incidence of ophthalmic immune-related AEs after ICIs versus rates of the same ocular complications in patients not taking ICIs in the entire registry population. Rates of ophthalmic immune-related AEs in patients with a past history of ocular inflammation or other specific ophthalmic condition before initiation of ICIs were examined further. RESULTS A total of 3123 patients who received anti-CTLA-4 or anti-programmed cell death 1 (PD-1) therapy were identified, 112 of whom demonstrated an ophthalmic immune-related AE. Incidence rates for anterior uveitis, the most common ophthalmic immune-related AE, were 8209 per 100 000 for ipilimumab (anti-CTLA-4), 2542 per 100 000 for nivolumab (anti-PD-1), 2451 per 100 000 for pembrolizumab (anti-PD-1), 5556 per 100 000 for ipilimumab plus nivolumab, and 3740 per 100 000 among all ICIs. Rates of ophthalmic immune-related AEs among patients receiving ICI therapy were higher compared with baseline rates in the general registry population (anterior uveitis IRR, 13.9; other uveitis IRR, 43.0; papilledema IRR, 38.3). Patients with a history of uveitis or other ocular inflammatory condition demonstrated high recurrence rates of ophthalmic immune-related AEs after initiating ICIs (up to 51.1%). CONCLUSIONS For patients initiating ICI therapy, early coordination with ophthalmic subspecialist care is important because rates of ophthalmic immune-related AEs are elevated compared with ocular complication rates in the entire registry population and patients with a history of prior autoimmune ocular disease are at high risk of recurrence of ocular complications.
Collapse
Affiliation(s)
- Michel M Sun
- Department of Ophthalmology, Stein Eye Institute, David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles, California
| | - Scott P Kelly
- American Academy of Ophthalmology, San Francisco, California
| | - Apoorva L Mylavarapu Bs
- Department of Ophthalmology, Stein Eye Institute, David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles, California
| | - Gary N Holland
- Department of Ophthalmology, Stein Eye Institute, David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles, California
| | - Anne L Coleman
- Department of Ophthalmology, Stein Eye Institute, David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles, California; Department of Epidemiology, Fielding School of Public Health, University of California at Los Angeles, Los Angeles, California
| | - Fei Yu
- Department of Ophthalmology, Stein Eye Institute, David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles, California
| | - Stephen Hsu Ms
- American Academy of Ophthalmology, San Francisco, California
| | - Flora Lum
- American Academy of Ophthalmology, San Francisco, California
| | - Lynn K Gordon
- Department of Ophthalmology, Stein Eye Institute, David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles, California.
| |
Collapse
|
23
|
Jacques FH, Apedaile E. Immunopathogenesis of COVID-19: Summary and Possible Interventions. Front Immunol 2020; 11:564925. [PMID: 33072103 PMCID: PMC7533543 DOI: 10.3389/fimmu.2020.564925] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/02/2020] [Indexed: 12/22/2022] Open
Abstract
Since the onset of the COVID-19 pandemic in the fall of 2019 over 4 million people have been infected and over 280,000 have died (1). Information about the SARS-CoV2 virus is evolving rapidly. At this time there are no interventions proven to be effective for cases infected with SARS-CoV2. Current knowledge about the clinical and laboratory manifestations of COVID-19 infection is reviewed and combined with knowledge about the immunopathogenic mechanisms of Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV1) and Middle East Respiratory Syndrome (MERS) to formulate theories and suggest possible therapeutic interventions. SARS-CoV2 immunopathogenic mechanisms vary from immunosuppression that initially enables viral escape to a hyperinflammatory immune response. Ultimately therapeutic intervention will be phase dependent.
Collapse
|
24
|
Voli F, Valli E, Lerra L, Kimpton K, Saletta F, Giorgi FM, Mercatelli D, Rouaen JRC, Shen S, Murray JE, Ahmed-Cox A, Cirillo G, Mayoh C, Beavis PA, Haber M, Trapani JA, Kavallaris M, Vittorio O. Intratumoral Copper Modulates PD-L1 Expression and Influences Tumor Immune Evasion. Cancer Res 2020; 80:4129-4144. [PMID: 32816860 DOI: 10.1158/0008-5472.can-20-0471] [Citation(s) in RCA: 235] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 06/19/2020] [Accepted: 08/03/2020] [Indexed: 11/16/2022]
Abstract
Therapeutic checkpoint antibodies blocking programmed death receptor 1/programmed death ligand 1 (PD-L1) signaling have radically improved clinical outcomes in cancer. However, the regulation of PD-L1 expression on tumor cells is still poorly understood. Here we show that intratumoral copper levels influence PD-L1 expression in cancer cells. Deep analysis of the The Cancer Genome Atlas database and tissue microarrays showed strong correlation between the major copper influx transporter copper transporter 1 (CTR-1) and PD-L1 expression across many cancers but not in corresponding normal tissues. Copper supplementation enhanced PD-L1 expression at mRNA and protein levels in cancer cells and RNA sequencing revealed that copper regulates key signaling pathways mediating PD-L1-driven cancer immune evasion. Conversely, copper chelators inhibited phosphorylation of STAT3 and EGFR and promoted ubiquitin-mediated degradation of PD-L1. Copper-chelating drugs also significantly increased the number of tumor-infiltrating CD8+ T and natural killer cells, slowed tumor growth, and improved mouse survival. Overall, this study reveals an important role for copper in regulating PD-L1 and suggests that anticancer immunotherapy might be enhanced by pharmacologically reducing intratumor copper levels. SIGNIFICANCE: These findings characterize the role of copper in modulating PD-L1 expression and contributing to cancer immune evasion, highlighting the potential for repurposing copper chelators as enhancers of antitumor immunity. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/19/4129/F1.large.jpg.
Collapse
Affiliation(s)
- Florida Voli
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, New South Wales, Australia.,School of Women's and Children's Health, UNSW Sydney, Sydney, Australia
| | - Emanuele Valli
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, New South Wales, Australia.,School of Women's and Children's Health, UNSW Sydney, Sydney, Australia
| | - Luigi Lerra
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, New South Wales, Australia
| | - Kathleen Kimpton
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, New South Wales, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, UNSW Sydney, New South Wales, Sydney, Australia
| | - Federica Saletta
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, New South Wales, Australia.,Children's Cancer Research Unit, Kids Research Institute, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - Federico M Giorgi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Daniele Mercatelli
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Jourdin R C Rouaen
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, New South Wales, Australia
| | - Sylvie Shen
- Cord & Marrow Transplant Facility, Kids Cancer Centre, Sydney Children's Hospital, Sydney, Australia
| | - Jayne E Murray
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, New South Wales, Australia
| | - Aria Ahmed-Cox
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, New South Wales, Australia.,School of Women's and Children's Health, UNSW Sydney, Sydney, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, UNSW Sydney, New South Wales, Sydney, Australia
| | - Giuseppe Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Chelsea Mayoh
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, New South Wales, Australia.,School of Women's and Children's Health, UNSW Sydney, Sydney, Australia
| | - Paul A Beavis
- Rosie Lew Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia
| | - Michelle Haber
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, New South Wales, Australia
| | - Joseph A Trapani
- Rosie Lew Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia
| | - Maria Kavallaris
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, New South Wales, Australia. .,School of Women's and Children's Health, UNSW Sydney, Sydney, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, UNSW Sydney, New South Wales, Sydney, Australia
| | - Orazio Vittorio
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, New South Wales, Australia. .,School of Women's and Children's Health, UNSW Sydney, Sydney, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, UNSW Sydney, New South Wales, Sydney, Australia
| |
Collapse
|
25
|
Lamberti G, Gelsomino F, Brocchi S, Poerio A, Melotti B, Sperandi F, Gargiulo M, Borghi C, Fiorentino M, Ardizzoni A. New disappearance of complicated atheromatous plaques on rechallenge with PD-1/PD-L1 axis blockade in non-small cell lung cancer patient: follow up of an unexpected event. Ther Adv Med Oncol 2020; 12:1758835920913801. [PMID: 32782484 PMCID: PMC7383634 DOI: 10.1177/1758835920913801] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 02/05/2020] [Indexed: 11/18/2022] Open
Abstract
Atherosclerosis is considered an irreversible process, with crucial contribution of inflammation and immune cells. Impact of cancer immunotherapy on a partly immune-driven disease, such as atherosclerosis, is poorly understood, but preclinical models suggest its worsening on programmed death/ligand-1 (PD-1/PD-L1) inhibitors. In a previously reported cohort of 11 patients with non-small cell lung cancer (NSCLC) treated with nivolumab and pre-existing complicated atheromatous plaques, 3 patients had a dramatic radiologic reduction of aortic plaques while on nivolumab; of these 3, 2 died receiving no further treatment. The remaining patient was an 83-year-old woman with history of arterial hypertension and hypothyroidism who was diagnosed with locally advanced squamous NSCLC. At relapse, complicated aortic atheromatous plaques were demonstrated on scans. The patient was then treated with nivolumab obtaining stable disease at radiological assessment, which also demonstrated almost complete vanishing of aortic plaques. After relapse and interval treatment with chemotherapy, she experienced new development of aortic atheromatous plaques. At further relapse she received atezolizumab, which yielded disease response and new reduction in aortic plaques, until nearly complete resolution. The observation of a repeated improvement of atheromatous plaques on treatment with PD-1/PD-L1 inhibitors favors the protective role of T cells on atheromatous plaques that is impaired by PD-L1 expression by plaque-associated macrophages. Validation by independent and prospective observation is needed.
Collapse
Affiliation(s)
- Giuseppe Lamberti
- Medical Oncology, Policlinico S.Orsola-Malpighi, University of Bologna, Bologna, Italy
| | - Francesco Gelsomino
- Medical Oncology Unit, Policlinico S.Orsola-Malpighi, Via P. Albertoni, 15, Bologna, 40138, Italy
| | - Stefano Brocchi
- Radiology Unit, Policlinico S.Orsola-Malpighi, Bologna, Italy
| | - Antonio Poerio
- Radiology Unit, Policlinico S.Orsola-Malpighi, University of Bologna, Bologna, Italy
| | - Barbara Melotti
- Medical Oncology Unit, Policlinico S.Orsola-Malpighi, Bologna, Italy
| | | | - Mauro Gargiulo
- Vascular Surgery, Policlinico S.Orsola-Malpighi, University of Bologna, Bologna, Italy
| | - Claudio Borghi
- Internal Medicine, Policlinico S.Orsola-Malpighi, University of Bologna, Bologna, Italy
| | | | - Andrea Ardizzoni
- Medical Oncology, Policlinico S.Orsola-Malpighi, University of Bologna, Bologna, Italy
| |
Collapse
|
26
|
Quiroga D, Liebner DA, Philippon JS, Hoffman S, Tan Y, Chen JL, Lenobel S, Wakely PE, Pollock R, Tinoco G. Activity of PD1 inhibitor therapy in advanced sarcoma: a single-center retrospective analysis. BMC Cancer 2020; 20:527. [PMID: 32503455 PMCID: PMC7275332 DOI: 10.1186/s12885-020-07021-x] [Citation(s) in RCA: 16] [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: 01/17/2020] [Accepted: 05/28/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Sarcomas constitute a heterogeneous group of tumors with different clinical behaviors and variable responses to systemic therapies. Recent immunotherapy studies with PD1 inhibitors (PD1i) show promising results with use in certain soft-tissue sarcomas; however, the clinical and molecular features that best predict response to PD1i remain unclear. METHODS Demographic, imaging, histologic, and genetic sequencing data was collected for sarcoma patients who received nivolumab or pembrolizumab (PD1i) treatment at our institution between January 1st 2015 and April 23rd 2018. The primary objective was to determine progression-free survival (PFS) in patients with advanced sarcomas receiving PD1i. Secondary objectives included determining overall survival (OS) and assessment of characteristics associated with response to PD1i. Fifty-six patients who were treated with PD1i therapy met inclusion criteria for this study. RESULTS Partial response towards PD1i treatment was seen in 3 in 26 evaluable patients, but no complete responses were observed (overall response rate 11.5%). Within this group of patients, the 90 day PFS was found to be 48.8%. In patients in whom PD1 expression was known, there was a statistically significant positive correlation between expression of PD1 and longer PFS and OS rates. Patients that were treated with more than four cycles of PD1i therapy were also more likely to have a greater OS. CONCLUSIONS This study suggests activity of PD1i in a pretreated cohort of advanced sarcoma patients, particularly for the subset of patients with PD1 positive tumors. Our results highlight the importance of further research to better target the optimal patient population and markers of response.
Collapse
Affiliation(s)
- Dionisia Quiroga
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, 410 W 12th Avenue, Columbus, OH, 43210, USA
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Starling Loving Hall, 320 W 10th Ave, Columbus, OH, 43210, USA
| | - David A Liebner
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, 410 W 12th Avenue, Columbus, OH, 43210, USA
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Starling Loving Hall, 320 W 10th Ave, Columbus, OH, 43210, USA
- Department of Biomedical Informatics, The Ohio State University, 250 Lincoln Tower, 1800 Cannon Dr, Columbus, OH, 43210, USA
| | - Jennifer S Philippon
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, 410 W 12th Avenue, Columbus, OH, 43210, USA
| | - Sarah Hoffman
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, 410 W 12th Avenue, Columbus, OH, 43210, USA
| | - Yubo Tan
- Center for Biostatistics, The Ohio State University, 2012 Kenny Rd, Columbus, OH, 43221, USA
| | - James L Chen
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, 410 W 12th Avenue, Columbus, OH, 43210, USA
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Starling Loving Hall, 320 W 10th Ave, Columbus, OH, 43210, USA
- Center for Biostatistics, The Ohio State University, 2012 Kenny Rd, Columbus, OH, 43221, USA
| | - Scott Lenobel
- Department of Radiology, The Ohio State University, 410 W 12th Avenue, Columbus, OH, 43210, USA
| | - Paul E Wakely
- Department of Pathology, The Ohio State University, 410 W 10th Ave, Columbus, OH, 43210, USA
| | - Raphael Pollock
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, 410 W 12th Avenue, Columbus, OH, 43210, USA
- Department of Surgery, The Ohio State University, 410 W 10th Ave, Columbus, OH, 43210, USA
| | - Gabriel Tinoco
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, 410 W 12th Avenue, Columbus, OH, 43210, USA.
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Starling Loving Hall, 320 W 10th Ave, Columbus, OH, 43210, USA.
- The Ohio State University Comprehensive Cancer Center, 320 W 10th Ave, A444 Starling Loving Hall, Columbus, OH, 43210, USA.
| |
Collapse
|
27
|
Shah M, Jomaa MK, Ferrarotto R, Yeung SCJ, Hanna EY, Reyes-Gibby CC. Serious immune-related adverse events in patients with head and neck cancer after checkpoint blockade: Systematic review. Head Neck 2019; 41:4036-4050. [PMID: 31414560 DOI: 10.1002/hed.25911] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 06/19/2019] [Accepted: 07/30/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Immune checkpoint inhibitors confer significant clinical benefit by bolstering immune-system activity, however, they also produce a spectrum of immune-related adverse events (irAEs). Rapid recognition and timely treatment of these patients is essential for improved outcomes. METHODS We conducted a systematic review of English-language articles in MEDLINE, EMBASE, Web of Science, PubMed, and Cochrane CENTRAL databases on patients with head and neck cancer treated with immune checkpoint inhibitors who developed treatment-related adverse events. RESULTS Of 1715 unique citations, 11 studies met inclusion criteria. Eight patients with serious irAEs were reported from case reports and case series. Overall, 46 treatment-related AEs were identified from the pooled 791 patients with at least 12 having potential relevance to irAEs. The most frequent AEs observed in patients receiving PD-1 inhibitors involved the endocrine, cutaneous, and gastrointestinal systems. CONCLUSIONS Characterizing irAEs in longitudinal studies is needed for developing strategies for their prompt recognition and management.
Collapse
Affiliation(s)
- Mohsin Shah
- Department of Emergency Medicine, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mona K Jomaa
- Department of Emergency Medicine, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Renata Ferrarotto
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sai-Ching J Yeung
- Department of Emergency Medicine, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ehab Y Hanna
- Department of Head and Neck Surgery, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cielito C Reyes-Gibby
- Department of Emergency Medicine, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
28
|
Shah M, Jizzini MN, Majzoub IE, Qdaisat A, Reyes-Gibby CC, Yeung SCJ. Safety of Immune Checkpoint Blockade in Patients with Cancer and Preexisting Autoimmune Diseases and/or Chronic Inflammatory Disorders. ACTA ACUST UNITED AC 2019. [DOI: 10.4103/jipo.jipo_11_19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Abstract
Background: Checkpoint blockade therapy, in the form of immune checkpoint inhibitors (ICIs), is increasingly being used to prolong survival in cancer patients, but its use is limited by the occurrence of immune-related adverse events (irAEs). These can be serious and occasionally fatal. However, the safety of ICIs is currently unknown in cancer patients with preexisting autoimmune diseases (PADs) and/or chronic inflammatory disorders (CIDs) such as eczema. Aim: The aim of this study is to evaluate the safety of ICIs in cancer patients with PAD and/or eczema at our institution. Patients and Methods: A retrospective study of cancer patients who presented to the Emergency Department between March 1, 2011, and February 29, 2016, after ICI therapy was previously conducted. Among these patients, those with PAD and/or eczema were further evaluated for safety by determining the occurrences of de novo irAEs or preexisting disease exacerbation. Results: Twenty-two cancer patients with PAD and/or eczema who received ICIs were reviewed, in which 15 were male (68%). Their median age was 63 years (range: 40–78 years). Most patients received anti-PD-1drugs (68%). Melanoma was the most common malignancy (45%). Autoimmune thyroiditis/primary hypothyroidism was the most common PAD. Four patients were receiving treatment for PAD at baseline using systemic corticosteroids, anti-inflammatory agents, and other immunosuppressants. Nineteen patients experienced de novo irAEs and/or PAD exacerbation. In three patients, the irAE was severe (grade ≥3). In six patients, the irAE or exacerbation was managed with systemic corticosteroids. Twelve patients experienced resolution of the de novo irAE or PAD exacerbation without the need to withhold or discontinue ICI therapy. The median time to last follow-up or death from the first dose of ICI was 16.8 months (range: 2–80 months). Death due to cancer progression was reported in 17 patients. Conclusion: Although de novo irAEs and PAD exacerbation were common, most patients with PAD and/or CIDs tolerated ICI therapy well.
Collapse
Affiliation(s)
- Mohsin Shah
- Department of Emergency Medicine, Unit 1468, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mazen N. Jizzini
- Department of Emergency Medicine, Unit 1468, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Imad E. Majzoub
- Department of Emergency Medicine, Unit 1468, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Emergency Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Aiham Qdaisat
- Department of Emergency Medicine, Unit 1468, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cielito C. Reyes-Gibby
- Department of Emergency Medicine, Unit 1468, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sai-Ching J. Yeung
- Department of Emergency Medicine, Unit 1468, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
29
|
Role of glypicans in regulation of the tumor microenvironment and cancer progression. Biochem Pharmacol 2019; 168:108-118. [PMID: 31251939 DOI: 10.1016/j.bcp.2019.06.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 06/20/2019] [Indexed: 12/28/2022]
Abstract
Glypicans are evolutionary conserved, cell surface heparan sulfate (HS) proteoglycans that are attached to the cell membrane via a glycosylphosphatidylinositol (GPI) anchor. Glypicans interact with a broad class of soluble and insoluble ligands, such as morphogens, growth factors, chemokines, receptors and components of the extracellular matrix (ECM). Such versatility comes from their ability to interact through both their HS chains and core protein. Glypicans are involved in cellular and tissue development, morphogenesis and cell motility. They exhibit differential expression in several cancers, acting as both tumor promoters and inhibitors in a cancer type-specific manner. They also influence tumor stroma by facilitating angiogenesis, ECM remodeling and alteration of immune cell functions. Glypicans have emerged as a new therapeutic moiety, whose functions can be exploited in the field of targeted therapies and precision medicine in cancer. This is demonstrated by the emergence of several anti-glypican antibody-based immunologics that have been recently developed and are being evaluated in clinical trials. This review will focus on glypican structure and function with an emphasis on their expression in various cancers. Discussion will also center on the potential of glypicans to be therapeutic targets for inhibition of cancer cell growth.
Collapse
|
30
|
Matts C, Beck A. Immunotherapy-Related Adverse Effects When Treating Cancer #375. J Palliat Med 2019; 22:724-725. [DOI: 10.1089/jpm.2019.0179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
31
|
Berger G, Marloye M, Lawler SE. Pharmacological Modulation of the STING Pathway for Cancer Immunotherapy. Trends Mol Med 2019; 25:412-427. [PMID: 30885429 DOI: 10.1016/j.molmed.2019.02.007] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 02/13/2019] [Accepted: 02/14/2019] [Indexed: 02/06/2023]
Abstract
The advent of immunotherapy in recent years has shown the potential to revolutionize the treatment of cancer. Unleashing antitumor T cell responses via immune checkpoint blockade has led to remarkable responses in previously untreatable tumors. The master regulator of interferon-mediated antiviral responses - stimulator of interferon genes (STING) - has now emerged as a critical mediator of innate immune sensing of cancer, and is a promising target for local immunostimulation, promoting intratumoral inflammation, and facilitating antitumor T cell responses. Pharmacological activation of the STING pathway can lead to T cell-mediated tumor regression in preclinical tumor models, and novel STING activating small molecules are now being tested in clinical trials. Here we will introduce the STING pathway and review the current state of drug development.
Collapse
Affiliation(s)
- Gilles Berger
- Microbiology, Bioorganic and Macromolecular Chemistry, Faculty of Pharmacy, Université libre de Bruxelles, Boulevard du Triomphe, 1050 Brussels, Belgium; Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mickaël Marloye
- Microbiology, Bioorganic and Macromolecular Chemistry, Faculty of Pharmacy, Université libre de Bruxelles, Boulevard du Triomphe, 1050 Brussels, Belgium
| | - Sean E Lawler
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
32
|
Weaver JL, Zadrozny LM, Gabrielson K, Semple KM, Shea KI, Howard KE. BLT-Immune Humanized Mice as a Model for Nivolumab-Induced Immune-Mediated Adverse Events: Comparison of the NOG and NOG-EXL Strains. Toxicol Sci 2019; 169:194-208. [DOI: 10.1093/toxsci/kfz045] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Abstract
Checkpoint inhibitors represent a new class of therapeutics in the treatment of cancer that has demonstrated remarkable clinical effectiveness. However, some patients have experienced serious immune-mediated adverse effects including pneumonitis, hepatitis, colitis, nephritis, dermatitis, encephalitis, and adrenal or pituitary insufficiency. These adverse events were not predicted by nonclinical studies. To determine if bone marrow-liver-thymus (BLT) immune humanized mice could demonstrate these adverse effects, we studied the effect of nivolumab on 2 strains of BLT-humanized mice, NOD.Cg-Prkdcscid Il2rgtm1Sug/JicTac (NOG) and NOD.Cg-Prkdcscid Il2rgtm1Sug Tg(SV40/HTLV-IL3, CSF2)10-7Jic/JicTac (NOG-EXL). Mice were treated with 2.5, 5.0, or 10.0 mg/kg nivolumab or saline twice weekly for 28 days. BLT-NOG mice had significantly reduced survival compared with BLT-NOG-EXL mice. In spite of the difference in survival, both BLT-humanized strains showed adverse reactions similar to those reported in humans, including pneumonitis and hepatitis, with nephritis, dermatitis and adrenalitis also noted in some individuals. Additional histopathologic findings included pancreatic atrophy, myositis, and osteomyelitis in some animals. T-cell activation increased with concomitant loss of PD-1 detection. These findings show that BLT immune humanized mice can demonstrate immune-mediated adverse effects of antiPD1 therapy, and may represent a model that can be used to better understand toxicity of this class of drugs.
Collapse
Affiliation(s)
- James L Weaver
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food & Drug Administration, Silver Spring, Maryland 20993
| | - Leah M Zadrozny
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food & Drug Administration, Silver Spring, Maryland 20993
| | - Kathleen Gabrielson
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food & Drug Administration, Silver Spring, Maryland 20993
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, Maryland 21205-2196
| | - Kenrick M Semple
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food & Drug Administration, Silver Spring, Maryland 20993
- Division of Gastroenterology and Inborn Errors Products, Office of Drug Evaluation III, Office of New Drugs, Center for Drug Evaluation and Research, U.S. Food & Drug Administration, Silver Spring, Maryland 20993
| | - Katherine I Shea
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food & Drug Administration, Silver Spring, Maryland 20993
| | - Kristina E Howard
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food & Drug Administration, Silver Spring, Maryland 20993
| |
Collapse
|
33
|
Sun MM, Levinson RD, Filipowicz A, Anesi S, Kaplan HJ, Wang W, Goldstein DA, Gangaputra S, Swan RT, Sen HN, Gordon LK. Uveitis in Patients Treated with CTLA-4 and PD-1 Checkpoint Blockade Inhibition. Ocul Immunol Inflamm 2019; 28:217-227. [PMID: 30821569 PMCID: PMC6832811 DOI: 10.1080/09273948.2019.1577978] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 01/24/2019] [Accepted: 01/30/2019] [Indexed: 12/11/2022]
Abstract
Purpose: To investigate the link between treatment with CTLA-4 and PD-1 checkpoint blockade inhibitors and the development of noninfectious uveitis.Methods: A survey was distributed to uveitis specialists to identify patients who developed uveitis while receiving either PD-1 inhibitors pembrolizumab and nivolumab; PD-L1 inhibitors atezolizumab, avelumab, and durvalumab; or the CTLA-4 inhibitor ipilimumab.Results: Fifteen patients from seven institutions were identified. The most common cancer diagnosis (13/15) was malignant melanoma. Fourteen patients had a new uveitis diagnosis following checkpoint blockade administration (six anterior uveitis, six panuveitis, one posterior uveitis, one anterior/intermediate combined); one patient developed optic neuritis. Uveitis was diagnosed within 6 months after drug initiation for 11/12 patients (median 63 days). Corticosteroid treatment was effective for most patients, although two patients had permanent loss of vision.Conclusions: Patients on checkpoint inhibitor therapy should be educated to seek care if they develop ocular symptoms, and prompt referral to specialists should be incorporated into oncology protocols.
Collapse
Affiliation(s)
- Michel M Sun
- Department of Ophthalmology, Jules Stein Eye Institute, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA
| | - Ralph D Levinson
- Department of Ophthalmology, Jules Stein Eye Institute, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA
| | - Artur Filipowicz
- Department of Ophthalmology, Massachusetts Eye Research and Surgical Institution, Waltham, MA, USA
| | - Stephen Anesi
- Department of Ophthalmology, Massachusetts Eye Research and Surgical Institution, Waltham, MA, USA
| | - Henry J Kaplan
- Department of Ophthalmology, University of Louisville, Louisville, KY, USA
| | - Wei Wang
- Department of Ophthalmology, University of Louisville, Louisville, KY, USA
| | - Debra A Goldstein
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Sapna Gangaputra
- Department of Ophthalmology, Vanderbilt Eye Institute, Nashville, TN, USA
| | - Robert T Swan
- Department of Ophthalmology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - H Nida Sen
- National Eye Institute, NIH, Bethesda, MD, USA
| | - Lynn K Gordon
- Department of Ophthalmology, Jules Stein Eye Institute, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
34
|
Shah M, Tayar JH, Abdel-Wahab N, Suarez-Almazor ME. Myositis as an adverse event of immune checkpoint blockade for cancer therapy. Semin Arthritis Rheum 2019; 48:736-740. [DOI: 10.1016/j.semarthrit.2018.05.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/11/2018] [Accepted: 05/16/2018] [Indexed: 02/06/2023]
|
35
|
Young JS, Dayani F, Morshed RA, Okada H, Aghi MK. Immunotherapy for High Grade Gliomas: A Clinical Update and Practical Considerations for Neurosurgeons. World Neurosurg 2019; 124:397-409. [PMID: 30677574 PMCID: PMC6642850 DOI: 10.1016/j.wneu.2018.12.222] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 12/26/2018] [Accepted: 12/28/2018] [Indexed: 10/27/2022]
Abstract
The current standard of care for patients with high grade gliomas includes surgical resection, chemotherapy, and radiation; but even still the majority of patients experience disease progression and succumb to their illness within a few years of diagnosis. Immunotherapy, which stimulates an anti-tumor immune response, has been revolutionary in the treatment of some hematological and solid malignancies, generating substantial excitement for its potential for patients with glioblastoma. The most commonly used immunotherapies include dendritic cell and peptide vaccines, checkpoint inhibitors, and adoptive T cell therapy. However, to date, the preclinical success of these approaches against high-grade glioma models has not been replicated in human clinical trials. Moreover, the complex response to these biologically active treatments can complicate management decisions, and the neurosurgical oncology community needs to be actively involved in and up to date on the use of these agents in high grade glioma patients. In this review, we discuss the challenges immunotherapy faces for high grade gliomas, the completed and ongoing clinical trials for the major immunotherapies, and the nuances in management for patients being actively treated with one of these agents.
Collapse
Affiliation(s)
- Jacob S Young
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Fara Dayani
- School of Medicine, University of California, San Francisco
| | - Ramin A Morshed
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Hideho Okada
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Manish K Aghi
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| |
Collapse
|
36
|
Saucedo AM, De La Cerda J, Suami H, Serda RE. Multimodal imaging of the tumor microenvironment and biological responses to immune therapy. Biomed Microdevices 2018; 20:105. [PMID: 30535532 DOI: 10.1007/s10544-018-0347-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Beyond heterogeneous cancer cells, the tumor microenvironment includes stromal and immune cells, blood vessels, extracellular matrix and biologically active molecules. Abnormal signaling, uncontrolled proliferation and high interstitial pressure all contribute to a chaotic, non-hierarchical vascular organization. Using an immune competent 4T1 breast adenocarcinoma murine model, this study fully characterizes the architecture and immunocyte milieu of the tumor microenvironment. Heterogeneous vessel distribution, chaotic connectivity, limited perfusion, cancer cell density, immune phenotype, and biological responses to immune therapy are presented. Cancer cell density mirrored the distribution of large, perfusable vessels, both predominately in the tumor periphery. Intratumoral administration of the proinflammatory cytokine IL-12 led to an increase in CD45+ leukocytes, with a specific increase in CD4+ and CD8+ T cells, and a decrease in the percentage of Gr-llo myeloid-derived suppressor cells. Concomitantly, serum G-CSF, IL-10 and VEGF decreased, while CXCR9 and interferon gamma increased. The distribution pattern of infiltrating monocytes/macrophages, visualized using a fluorescent perfluorocarbon emulsion, indicated that macrophages predominately localize in the vicinity of large blood vessels. Electron microscopy supports the presence of dense tumor cell masses throughout the tumor, with the largest vessels present in the surrounding mammary fat pad. Overall, large vessels in the 4T1 tumor periphery support high, localized vascular perfusion and myeloid accumulation. The pro-inflammatory cytokine IL-12 stimulated a transition towards T helper 1 cytokines in serum, supporting suppression of tumor growth and angiostatic conditions.
Collapse
Affiliation(s)
- Alexander M Saucedo
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jorge De La Cerda
- Small Animal Imaging Resource, UT MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Hiroo Suami
- Department of Plastic Surgery, UT MD Anderson Cancer Center, Houston, TX, 77030, USA.,Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Rita E Serda
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Nanomedicine, Houston Methodist, Houston, TX, 77030, USA. .,Molecular Medicine, Internal Medicine, University of New Mexico Health Science Center, 915 Camino de Salud, CRF 307, Albuquerque, NM, 87131, USA.
| |
Collapse
|
37
|
|
38
|
AlHarbi M, Ali Mobark N, AlMubarak L, Aljelaify R, AlSaeed M, Almutairi A, Alqubaishi F, Hussain ME, Balbaid AAO, Said Marie A, AlSubaie L, AlShieban S, alTassan N, Ramkissoon SH, Abedalthagafi M. Durable Response to Nivolumab in a Pediatric Patient with Refractory Glioblastoma and Constitutional Biallelic Mismatch Repair Deficiency. Oncologist 2018; 23:1401-1406. [PMID: 30104292 DOI: 10.1634/theoncologist.2018-0163] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 05/31/2018] [Indexed: 01/05/2023] Open
Abstract
Primary brain tumors are a leading cause of cancer-related morbidity and mortality in children. Glioblastoma (GBM) is a high-grade astrocytoma that occurs in both children and adults and is associated with a poor prognosis. Despite extensive study in recent years, the clinical management of these tumors has remained largely unchanged, consisting of surgical resection, conventional chemotherapy, and radiotherapy. Although the etiology and genomic drivers in GBM are diverse, constitutional mismatch repair-deficiency (CMMRD) syndrome is a rare, recessively inherited disease with a predisposition to gliomagenesis. CMMRD results from biallelic mutations in one of the mismatch repair genes including mutL homolog 1 (MLH1), mutS homolog 2 (MSH2), mutS homolog 6 (MSH6), and post-meiotic segregation increased 2 (PMS2). In this report, we present the case of a 5-year-old female with GBM and CMMRD due to an MSH6 homozygous c.1883G>A mutation, who continues to experience an exceptional and durable response (9 months) to the immune checkpoint inhibitor (ICPI) nivolumab. Our patient presented with acute neurologic decline and increased intracranial pressure. Neuroimaging studies revealed a large left frontoparietal mass requiring neurosurgical decompression and resection. Histopathologic analyses resulted in a diagnosis of de novo GBM that was BRAF wild type and negative for programmed death-ligand 1 protein expression. She received standard-of-care treatment with surgery, radiation therapy, and temozolomide; however, the tumor recurred 3 months after the initial diagnosis. Molecular analyses of tumor and blood tissues revealed an MSH6 homozygous c.1883G>A mutation consistent with CMMRD. Given her CMMRD status, she was treated with nivolumab (3 mg/kg doses every 2 weeks for 36 weeks) and showed a 60% reduction in tumor size, improved clinical symptoms, and an ongoing durable response lasting 10 months to date. Our study highlights a durable response to the ICPI nivolumab in a pediatric patient with recurrent/refractory CMMRD-associated GBM. We show that incorporating genomic and/or molecular testing for CMMRD into routine pediatric oncology clinical care can identify a subset of patients likely to benefit from ICPI. KEY POINTS: Constitutional mismatch repair-deficiency (CMMRD) syndrome, alternatively known as biallelic mismatch repair deficiency syndrome, occurs in subset of pediatric cancer patients, including those with primary brain tumors.Patients from Arab and other developing countries are predicted to have higher incidence of CMMRD due to high prevalence of consanguinity.Integration of molecular and/or genomic testing into routine clinical care for pediatric cancer patients is important to identify patients with CMMRD syndrome.Patient with CMMRD-associated cancers may show increased responsiveness to immune checkpoint inhibitors.To the authors' knowledge, this is the first report in the Arab world of a durable response to immune checkpoint inhibitors in a pediatric glioblastoma patient.
Collapse
Affiliation(s)
- Musa AlHarbi
- Department of Oncology, Comprehensive Cancer Centre, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Nahla Ali Mobark
- Department of Oncology, Comprehensive Cancer Centre, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Latifa AlMubarak
- Genomics Research Department, Saudi Human Genome Project, King Fahad Medical City and King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Rasha Aljelaify
- Genomics Research Department, Saudi Human Genome Project, King Fahad Medical City and King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Mariam AlSaeed
- Genomics Research Department, Saudi Human Genome Project, King Fahad Medical City and King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Amal Almutairi
- Genomics Research Department, Saudi Human Genome Project, King Fahad Medical City and King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Fatmah Alqubaishi
- Genomics Research Department, Saudi Human Genome Project, King Fahad Medical City and King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - M Emarat Hussain
- Radiology Department, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Ali Abdullah O Balbaid
- Genomics Research Department, Saudi Human Genome Project, King Fahad Medical City and King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Amal Said Marie
- Department of Radiation Oncology, Comprehensive Cancer Centre, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Lamia AlSubaie
- Division of Genetics, Department of Pediatric, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Saeed AlShieban
- Department of Pathology, King Abdulaziz Medical City, National Guard Health Affair, Riyadh, Saudi Arabia
| | - Nada alTassan
- Genetics Department, King Faisal Specialists Hospital and Research Center, Riyadh, Saudi Arabia
| | - Shakti H Ramkissoon
- Wake Forest Comprehensive Cancer Center and Department of Pathology, Winston-Salem, North Carolina, USA
- Foundation Medicine, Inc., Morrisville, North Carolina, USA
| | - Malak Abedalthagafi
- Genomics Research Department, Saudi Human Genome Project, King Fahad Medical City and King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
39
|
Ali AK, Watson DE. Pharmacovigilance Assessment of Immune-Mediated Reactions Reported for Checkpoint Inhibitor Cancer Immunotherapies. Pharmacotherapy 2017; 37:1383-1390. [PMID: 28950039 DOI: 10.1002/phar.2035] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
STUDY OBJECTIVE Ipilimumab, pembrolizumab, and nivolumab are checkpoint inhibitors (CPIs) that activate T-cell-mediated immune response. CPI can provide durable benefits to some cancer patients with melanoma, renal cell cancer, non-small cell lung cancer, or a growing list of other cancers. However, CPI treatment is also associated with adverse immune-mediated reactions (IMRs) that can be life-threatening. This pharmacovigilance analysis aims to characterize IMR signals in relation to CPI treatment. DESIGN Retrospective pharmacovigilance disproportionality analysis. DATA SOURCE U.S. Food and Drug Administration Adverse Event Reporting System (FAERS). MEASUREMENTS AND MAIN RESULTS Adverse event reports submitted to FAERS between 2011 and 2015 were analyzed. CPIs were identified by generic names, and IMRs were identified by MedDRA Preferred Terms. Empirical Bayes geometric means with corresponding 95% confidence intervals (EB05-EB95) were calculated as CPI-IMR association metrics. Signals were defined as metrics with EB05 ≥ 2.0. Overall, 1,018 IMR events were submitted for CPIs, corresponding to 76% for ipilimumab, 15% for nivolumab, and 9% for pembrolizumab. The period of data collection precluded data on the most recently approved CPI agents. IMRs consisted of 51% colitis, 16% endocrinopathies, 12% pneumonitis, 11% hepatitis, 4% infusion-related reactions, 3% nephritis, and 3% other IMRs. Colitis contributed to 63% and 41% of IMRs for ipilimumab and nivolumab, respectively. Pneumonitis and hepatitis contributed to a majority of IMRs for pembrolizumab, for which nephritis and infusion-related reactions were not reported. Signals of IMRs were detected for CPIs as a class (EB05 = 12.4) and individual agents: ipilimumab (EB05 = 13.2), nivolumab (EB05 = 15.0), and pembrolizumab (EB05 = 7.3). Colitis and pneumonitis had the strongest signals for CPIs (EB05 = 45.6 and EB05 = 17.6, respectively). Colitis was the strongest signal for ipilimumab (EB05 = 54.2), and pneumonitis was the strongest signal for nivolumab (EB05 = 48.0) and pembrolizumab (EB05 = 21.8). CONCLUSION Cancer immunotherapy with CPIs is associated with a multitude of IMRs, especially colitis and pneumonitis. Individual CPIs had variable IMR signals, and pharmacoepidemiologic studies are required to evaluate the identified signals.
Collapse
Affiliation(s)
- Ayad K Ali
- Global Patient Safety, Eli Lilly and Company, Indianapolis, Indiana
| | - David E Watson
- Toxicology, Eli Lilly and Company, Indianapolis, Indiana
| |
Collapse
|
40
|
Associations between inflammatory gene polymorphisms (TNF-α 308G/A, TNF-α 238G/A, TNF-β 252A/G, TGF-β1 29T/C, IL-6 174G/C and IL-10 1082A/G) and susceptibility to osteosarcoma: a meta-analysis and literature review. Oncotarget 2017; 8:97571-97583. [PMID: 29228633 PMCID: PMC5722585 DOI: 10.18632/oncotarget.18813] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 06/12/2017] [Indexed: 12/18/2022] Open
Abstract
Associations between inflammatory gene polymorphisms (TNF-α 308G/A, TNF-α 238G/A, TNF-β 252A/G, TGF-β1 29T/C, IL-6 174G/C and IL-10 1082A/G) and osteosarcoma (OS) risk remain unclear. We conducted a systematic search to retrieve studies that investigated associations between inflammatory gene polymorphisms and OS risk. Nine studies that met the inclusion criteria were finally recruited in this meta-analysis. Overall, there was a significant association between TNF-α 308G/A, IL-10 1082A/G and OS risk, while there was no significant association between TNF-α 238G/A, TNF-β 252A/G and IL-6 174G/C and OS risk. Our subgroup analysis showed a significant association between IL-6 174G/C and IL-10 1082A/G and OS risk in Asians, while no such significant correlation was observed with TNF-α 308G/A, TNF-α 238G/A, TNF-β 252A/G and TGF-β1 29T/C polymorphisms. In Caucasians, there was a significant association between TNF-α 238G/A and the decreased incidence of OS. In conclusion, inflammatory gene polymorphisms play a key role in the occurrence and progression of OS. IL-6 174G/C polymorphism was obviously associated with OS risk in Asians, while TNF-α 238G/A polymorphism seemed to be associated with the decreased susceptibility to OS in Caucasians as Altman and Bland test indicated. Although controversial results were observed between IL-10 1082A/G and OS risk in Asians and Caucasians, it is difficult to make a definite conclusion about the role of IL-10 1082A/G polymorphism in the etiology of OS because our Altman and Bland test showed no good evidence to support a different effect in Asians and Caucasians.
Collapse
|