1
|
Petit PF, Daoudlarian D, Latifyan S, Bouchaab H, Mederos N, Doms J, Abdelhamid K, Ferahta N, Mencarelli L, Joo V, Bartolini R, Stravodimou A, Shabafrouz K, Pantaleo G, Peters S, Obeid M. Tocilizumab provides dual benefits in treating immune checkpoint inhibitor-associated arthritis and preventing relapse during ICI rechallenge: the TAPIR study. Ann Oncol 2025; 36:43-53. [PMID: 39241964 DOI: 10.1016/j.annonc.2024.08.2340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/04/2024] [Accepted: 08/19/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND The aim of this retrospective study was to evaluate the dual efficacy of tocilizumab (TCZ) in the treatment of immune checkpoint inhibitor (ICI)-associated arthritis (ICI-AR) and the prevention of relapses after rechallenge. PATIENTS AND METHODS We identified 26 patients with ICI-AR. The primary objectives were to evaluate TCZ efficacy in ICI-AR treatment and as secondary prophylaxis during ICI rechallenge in 11 of them. Patients received prednisone (CS) at 0.3 mg/kg tapered at 0.05 mg/kg weekly for six weeks. TCZ was administered at a dose of 8 mg/kg every 2 weeks. In the subgroup receiving secondary prophylaxis (rechallenge n = 11), TCZ was reintroduced with the same regimen concurrently with ICI rechallenge, and without the addition of CS. A control group of patients (rechallenge n = 5) was rechallenged without TCZ. Secondary endpoints included post-rechallenge evaluation of ICI duration, reintroduction of CS >0.1 mg/kg/day, ICI-AR flares, and disease control rate. RESULTS The median age of the patients was 70 years. The median follow-up from ICI initiation was 864 days. Among the 20 patients treated with TCZ for ICI-AR, all (100%) achieved an ACR70 response rate, defined as greater than 70% improvement, at 10 weeks. Some 81% of these patients achieved steroid-free remission after 24 weeks on TCZ. The median follow-up period was 552 days in rechallenged patients. The results demonstrated a reduction in ICI-AR relapses upon ICI rechallenge in patients receiving TCZ prophylaxis compared with patients who did not receive prophylaxis (17% versus 40%). The requirement for CS was completely abolished with prophylaxis (0% versus 20%), and the mean duration of ICI treatment was notably extended from 113 to 206 days. The 12-month post-rechallenge outcomes showed a disease control rate of 77%. During TCZ prophylaxis, CXCL9 remained elevated, showing no decline from their concentrations at the onset of ICI-AR. CONCLUSIONS In addition to treating ICI-AR, TCZ demonstrated efficacy as a secondary prophylactic agent, preventing the recurrence of symptoms and lengthening ICI treatment duration after ICI rechallenge.
Collapse
Affiliation(s)
- P-F Petit
- Medical Oncology Service, CHU Helora, La Louvière, Belgium
| | - D Daoudlarian
- Department of Medicine, Immunology and Allergy Service
| | - S Latifyan
- Department of Oncology, Medical Oncology Service, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - H Bouchaab
- Department of Oncology, Medical Oncology Service, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - N Mederos
- Department of Oncology, Medical Oncology Service, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - J Doms
- Department of Medicine, Immunology and Allergy Service
| | - K Abdelhamid
- Department of Oncology, Medical Oncology Service, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - N Ferahta
- Department of Oncology, Medical Oncology Service, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - L Mencarelli
- Department of Medicine, Immunology and Allergy Service
| | - V Joo
- Department of Medicine, Immunology and Allergy Service
| | - R Bartolini
- Department of Medicine, Immunology and Allergy Service
| | - A Stravodimou
- Department of Oncology, Medical Oncology Service, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - K Shabafrouz
- Department of Oncology, Medical Oncology Service, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - G Pantaleo
- Department of Medicine, Immunology and Allergy Service
| | - S Peters
- Department of Oncology, Medical Oncology Service, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - M Obeid
- Department of Medicine, Immunology and Allergy Service.
| |
Collapse
|
2
|
Hu W, Bian Y, Ji H. TIL Therapy in Lung Cancer: Current Progress and Perspectives. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2409356. [PMID: 39422665 PMCID: PMC11633538 DOI: 10.1002/advs.202409356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/03/2024] [Indexed: 10/19/2024]
Abstract
Lung cancer remains the most prevalent malignant tumor worldwide and is the leading cause of cancer-related mortality. Although immune checkpoint blockade has revolutionized the treatment of advanced lung cancer, many patients still do not respond well, often due to the lack of functional T cell infiltration. Adoptive cell therapy (ACT) using expanded immune cells has emerged as an important therapeutic modality. Tumor-infiltrating lymphocytes (TIL) therapy is one form of ACT involving the administration of expanded and activated autologous T cells derived from surgically resected cancer tissues and reinfusion into patients and holds great therapeutic potential for lung cancer. In this review, TIL therapy is introduced and its suitability for lung cancer is discussed. Then its historical and clinical developments are summarized, and the methods developed up-to-date to identify tumor-recognizing TILs and optimize TIL composition. Some perspectives toward future TIL therapy for lung cancer are also provided.
Collapse
Affiliation(s)
- Weilei Hu
- Key Laboratory of Systems Health Science of Zhejiang ProvinceSchool of Life ScienceHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhou310024China
- Key Laboratory of Multi‐Cell SystemsShanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yifei Bian
- Key Laboratory of Multi‐Cell SystemsShanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
| | - Hongbin Ji
- Key Laboratory of Systems Health Science of Zhejiang ProvinceSchool of Life ScienceHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhou310024China
- Key Laboratory of Multi‐Cell SystemsShanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
- School of Life Science and TechnologyShanghai Tech UniversityShanghai200120China
| |
Collapse
|
3
|
Kao CJ, Charmsaz S, Alden SL, Brancati M, Li HL, Balaji A, Munjal K, Howe K, Mitchell S, Leatherman J, Griffin E, Nakazawa M, Tsai HL, Danilova L, Thoburn C, Gizzi J, Gross NE, Hernandez A, Coyne EM, Shin SM, Babu JS, Apostol GW, Durham J, Christmas BJ, Konig MF, Lipson EJ, Naidoo J, Cappelli LC, Pabani A, Ged Y, Baretti M, Brahmer J, Hoffman-Censits J, Seiwert TY, Garonce-Hediger R, Guha A, Bansal S, Tang L, Jaffee EM, Chandler GS, Mohindra R, Ho WJ, Yarchoan M. Immune-related events in individuals with solid tumors on immunotherapy associate with Th17 and Th2 signatures. J Clin Invest 2024; 134:e176567. [PMID: 39403935 PMCID: PMC11473156 DOI: 10.1172/jci176567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 08/20/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUNDImmune-related adverse events (irAEs) and their associated morbidity/mortality are a key concern for patients receiving immune checkpoint inhibitors (ICIs). Prospective evaluation of the drivers of irAEs in a diverse pan-tumor cohort is needed to identify patients at greatest risk and to develop rational treatment and interception strategies.METHODSIn an observational study, we prospectively collected blood samples and performed regular clinical evaluations for irAEs in patients receiving ICI therapy as standard of care for solid tumors. We performed in-parallel analysis of cytokines by Luminex immunoassay and circulating immune cells by cytometry by time-of-flight (CyTOF) at baseline and on treatment to investigate mechanisms of irAEs.RESULTSWe enrolled 111 patients, of whom 40.5% developed a symptomatic irAE (grade ≥ 2). Development of a grade ≥ 2 irAE was positively associated with the use of combination ICI and a history of an autoimmune disorder. Early changes in T helper 17 (Th17) (IL-6, IL-17f), type 2 (IL-5, IL-13, IL-25), and type 1 (TNF-α) cytokine signatures and congruent on-treatment expansions of Th17 and Th2 effector memory (Th2EM) T cell populations in peripheral blood were positively associated with the development of grade ≥2 irAEs. IL-6 levels were also associated with inferior cancer-specific survival and overall survival.CONCLUSIONSIn a diverse, prospective pan-tumor cohort, Th17 and Th2 skewing during early ICI treatment was associated with the development of clinically relevant irAEs but not antitumor responses, providing possible targets for monitoring and therapeutic interventions.FUNDINGJohns Hopkins Bloomberg-Kimmel Institute for Cancer Immunotherapy, the NCI SPORE in Gastrointestinal Cancers (P50 CA062924), NCI grant (R50CA243627 to LD), the NIH Center Core Grant (P30 CA006973), Swim Across America (to MY), NIAMS (K23AR075872 to LC), and imCORE-Genentech grant 137515 (to Johns Hopkins Medicine on behalf of MY).
Collapse
Affiliation(s)
- Chester J. Kao
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Soren Charmsaz
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | | | - Madelena Brancati
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Howard L. Li
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aanika Balaji
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kabeer Munjal
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Kathryn Howe
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Sarah Mitchell
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - James Leatherman
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Ervin Griffin
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Mari Nakazawa
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Hua-Ling Tsai
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ludmila Danilova
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Convergence Institute and
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland, USA
| | - Chris Thoburn
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jennifer Gizzi
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nicole E. Gross
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Alexei Hernandez
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Erin M. Coyne
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Sarah M. Shin
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Jayalaxmi Suresh Babu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - George W. Apostol
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Jennifer Durham
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Brian J. Christmas
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Maximilian F. Konig
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Evan J. Lipson
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jarushka Naidoo
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Beaumont Hospital, Dublin, Ireland
- RCSI University of Health Sciences, Dublin, Ireland
| | - Laura C. Cappelli
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aliyah Pabani
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yasser Ged
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Marina Baretti
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Julie Brahmer
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jean Hoffman-Censits
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tanguy Y. Seiwert
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Aditi Guha
- Genentech Inc., a member of the imCORE network, South San Francisco, California, USA
| | - Sanjay Bansal
- Genentech Inc., a member of the imCORE network, South San Francisco, California, USA
| | - Laura Tang
- Genentech Inc., a member of the imCORE network, South San Francisco, California, USA
| | - Elizabeth M. Jaffee
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Convergence Institute and
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland, USA
| | - G. Scott Chandler
- F. Hoffmann-La Roche Ltd., a member of the imCORE network, Basel, Switzerland
| | - Rajat Mohindra
- F. Hoffmann-La Roche Ltd., a member of the imCORE network, Basel, Switzerland
| | - Won Jin Ho
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Convergence Institute and
| | - Mark Yarchoan
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Convergence Institute and
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Turley JL, Ward RW, Huete-Carrasco J, Muñoz-Wolf N, Roche K, Jin L, Bowie A, Andersson M, Lavelle EC. Intratumoral delivery of the chitin-derived C100 adjuvant promotes robust STING, IFNAR, and CD8 + T cell-dependent anti-tumor immunity. Cell Rep Med 2024; 5:101560. [PMID: 38729159 PMCID: PMC11148802 DOI: 10.1016/j.xcrm.2024.101560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 02/07/2024] [Accepted: 04/17/2024] [Indexed: 05/12/2024]
Abstract
Stimulator of IFN genes (STING) is a promising target for adjuvants utilized in in situ cancer vaccination approaches. However, key barriers remain for clinical translation, including low cellular uptake and accessibility, STING variability necessitating personalized STING agonists, and interferon (IFN)-independent signals that can promote tumor growth. Here, we identify C100, a highly deacetylated chitin-derived polymer (HDCP), as an attractive alternative to conventional STING agonists. C100 promotes potent anti-tumor immune responses, outperforming less deacetylated HDCPs, with therapeutic efficacy dependent on STING and IFN alpha/beta receptor (IFNAR) signaling and CD8+ T cell mediators. Additionally, C100 injection synergizes with systemic checkpoint blockade targeting PD-1. Mechanistically, C100 triggers mitochondrial stress and DNA damage to exclusively activate the IFN arm of the cGAS-STING signaling pathway and elicit sustained IFNAR signaling. Altogether, these results reveal an effective STING- and IFNAR-dependent adjuvant for in situ cancer vaccines with a defined mechanism and distinct properties that overcome common limitations of existing STING therapeutics.
Collapse
Affiliation(s)
- Joanna L Turley
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin 2, Ireland
| | - Ross W Ward
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin 2, Ireland
| | - Jorge Huete-Carrasco
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin 2, Ireland
| | - Natalia Muñoz-Wolf
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin 2, Ireland
| | - Kate Roche
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin 2, Ireland
| | - Lei Jin
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Andrew Bowie
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, D02 R590 Dublin, Ireland
| | - Mats Andersson
- Division Bioscience and Materials, RISE (Research Institutes of Sweden), Forskargatan 18, 151 36 Södertälje, Sweden
| | - Ed C Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin 2, Ireland; Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN) & Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, D02 PN40 Dublin 2, Ireland.
| |
Collapse
|
5
|
Alserawan L, Mulet M, Anguera G, Riudavets M, Zamora C, Osuna-Gómez R, Serra-López J, Barba Joaquín A, Sullivan I, Majem M, Vidal S. Kinetics of IFNγ-Induced Cytokines and Development of Immune-Related Adverse Events in Patients Receiving PD-(L)1 Inhibitors. Cancers (Basel) 2024; 16:1759. [PMID: 38730712 PMCID: PMC11083441 DOI: 10.3390/cancers16091759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/25/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024] Open
Abstract
Immune checkpoint inhibitors (ICI) have the potential to trigger unpredictable immune-related adverse events (irAEs), which can be severe. The underlying mechanisms of these events are not fully understood. As PD-L1 is upregulated by IFN, the heightened immune activation resulting from PD-1/PD-L1 inhibition may enhance the IFN response, triggering the expression of IFN-inducible genes and contributing to irAE development and its severity. In this study, we investigated the interplay between irAEs and the expression of IFN-inducible chemokines and cytokines in 134 consecutive patients with solid tumours treated with PD-(L)1 inhibitors as monotherapy or in combination with chemotherapy or other immunotherapy agents. We compared the plasma levels of IFN-associated cytokines (CXCL9/10/11, IL-18, IL-10, IL-6 and TGFβ) at various time points (at baseline, at the onset of irAE and previous to irAE onset) in three patient groups categorized by irAE development and severity: patients with serious irAEs, mild irAEs and without irAEs after PD-(L)1 inhibitors. No differences were observed between groups at baseline. However, patients with serious irAEs exhibited significant increases in CXCL9/10/11, IL-18 and IL-10 levels at the onset of the irAE compared to baseline. A network analysis and correlation patterns highlighted a robust relationship among these chemokines and cytokines at serious-irAE onset. Combining all of the analysed proteins in a cluster analysis, we identified a subgroup of patients with a higher incidence of serious irAEs affecting different organs or systems. Finally, an ROC analysis and a decision tree model proposed IL-18 levels ≥ 807 pg/mL and TGFβ levels ≤ 114 pg/mL as predictors for serious irAEs in 90% of cases. In conclusion, our study elucidates the dynamic changes in cytokine profiles associated with serious irAE development during treatment with PD-(L)1 inhibitors. The study's findings offer valuable insights into the intricate IFN-induced immune responses associated with irAEs and propose potential predictive markers for their severity.
Collapse
Affiliation(s)
- Leticia Alserawan
- Immunology-Inflammatory Diseases, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025 Barcelona, Spain; (L.A.); (M.M.); (C.Z.); (R.O.-G.)
- Department of Immunology, Hospital Clínic Barcelona, 08036 Barcelona, Spain
| | - Maria Mulet
- Immunology-Inflammatory Diseases, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025 Barcelona, Spain; (L.A.); (M.M.); (C.Z.); (R.O.-G.)
| | - Geòrgia Anguera
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain; (G.A.); (M.R.); (J.S.-L.); (A.B.J.); (I.S.); (M.M.)
| | - Mariona Riudavets
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain; (G.A.); (M.R.); (J.S.-L.); (A.B.J.); (I.S.); (M.M.)
- Department of Pneumologie, Hôpital Cochin—APHP Centre, 75014 Paris, France
| | - Carlos Zamora
- Immunology-Inflammatory Diseases, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025 Barcelona, Spain; (L.A.); (M.M.); (C.Z.); (R.O.-G.)
| | - Rubén Osuna-Gómez
- Immunology-Inflammatory Diseases, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025 Barcelona, Spain; (L.A.); (M.M.); (C.Z.); (R.O.-G.)
| | - Jorgina Serra-López
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain; (G.A.); (M.R.); (J.S.-L.); (A.B.J.); (I.S.); (M.M.)
| | - Andrés Barba Joaquín
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain; (G.A.); (M.R.); (J.S.-L.); (A.B.J.); (I.S.); (M.M.)
| | - Ivana Sullivan
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain; (G.A.); (M.R.); (J.S.-L.); (A.B.J.); (I.S.); (M.M.)
| | - Margarita Majem
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain; (G.A.); (M.R.); (J.S.-L.); (A.B.J.); (I.S.); (M.M.)
| | - Silvia Vidal
- Immunology-Inflammatory Diseases, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025 Barcelona, Spain; (L.A.); (M.M.); (C.Z.); (R.O.-G.)
| |
Collapse
|
6
|
Zhang H, Houadj L, Wu KY, Tran SD. Diagnosing and Managing Uveitis Associated with Immune Checkpoint Inhibitors: A Review. Diagnostics (Basel) 2024; 14:336. [PMID: 38337852 PMCID: PMC10855398 DOI: 10.3390/diagnostics14030336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
This review aims to provide an understanding of the diagnostic and therapeutic challenges of uveitis associated with immune checkpoint inhibitors (ICI). In the wake of these molecules being increasingly employed as a treatment against different cancers, cases of uveitis post-ICI therapy have also been increasingly reported in the literature, warranting an extensive exploration of the clinical presentations, risk factors, and pathophysiological mechanisms of ICI-induced uveitis. This review further provides an understanding of the association between ICIs and uveitis, and assesses the efficacy of current diagnostic tools, underscoring the need for advanced techniques to enable early detection and accurate assessment. Further, it investigates the therapeutic strategies for ICI-related uveitis, weighing the benefits and limitations of existing treatment regimens, and discussing current challenges and emerging therapies in the context of their potential efficacy and side effects. Through an overview of the short-term and long-term outcomes, this article suggests recommendations and emphasizes the importance of multidisciplinary collaboration between ophthalmologists and oncologists. Finally, the review highlights promising avenues for future research and development in the field, potentially informing transformative approaches in the ocular assessment of patients under immunotherapy and the management of uveitis following ICI therapy.
Collapse
Affiliation(s)
- Huixin Zhang
- Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada;
| | - Lysa Houadj
- Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada;
| | - Kevin Y. Wu
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada
| | - Simon D. Tran
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
| |
Collapse
|
7
|
Soussan S, Pupier G, Cremer I, Joubert PE, Sautès-Fridman C, Fridman W, Sibéril S. Unraveling the complex interplay between anti-tumor immune response and autoimmunity mediated by B cells and autoantibodies in the era of anti-checkpoint monoclonal antibody therapies. Front Immunol 2024; 15:1343020. [PMID: 38318190 PMCID: PMC10838986 DOI: 10.3389/fimmu.2024.1343020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/05/2024] [Indexed: 02/07/2024] Open
Abstract
The intricate relationship between anti-tumor immunity and autoimmunity is a complex yet crucial aspect of cancer biology. Tumor microenvironment often exhibits autoimmune features, a phenomenon that involves natural autoimmunity and the induction of humoral responses against self-antigens during tumorigenesis. This induction is facilitated by the orchestration of anti-tumor immunity, particularly within organized structures like tertiary lymphoid structures (TLS). Paradoxically, a significant number of cancer patients do not manifest autoimmune features during the course of their illness, with rare instances of paraneoplastic syndromes. This discrepancy can be attributed to various immune-mediated locks, including regulatory or suppressive immune cells, anergic autoreactive lymphocytes, or induction of effector cells exhaustion due to chronic stimulation. Overcoming these locks holds the risk to induce autoimmune mechanisms during cancer progression, a phenomenon notably observed with anti-immune checkpoint therapies, in contrast to more conventional treatments like chemotherapy or radiotherapy. Therefore, the challenge arises in managing immune-related adverse events (irAEs) induced by immune checkpoint inhibitors treatment, as decoupling them from the anti-tumor activity poses a significant clinical dilemma. This review summarizes recent advances in understanding the link between B-cell driven anti-tumor responses and autoimmune reactions in cancer patients, and discusses the clinical implications of this relationship.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sophie Sibéril
- Centre de recherche des Cordeliers, INSERM U1138, Sorbonne Université, Université Paris Cité, Paris, France
| |
Collapse
|
8
|
Kozubek P, Wołoszczak J, Gomułka K. Immune Reactions in Major Types of Oncological Treatment. Int J Mol Sci 2023; 24:11257. [PMID: 37511017 PMCID: PMC10378943 DOI: 10.3390/ijms241411257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/24/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
In recent years, there has been a noticeable development in oncological treatment, including chemotherapy and biological treatment. Despite their significant effectiveness, they are not free from side effects, such as allergic and dermatological reactions. These reactions can vary in severity and outcome, including potential death. Examples, among others, are type I-IV hypersensitivity reactions of various origins and skin reactions including rashes, itching and redness, but also severe cutaneous syndromes. Due to the therapy used, these may include Stevens-Johnson syndrome, toxic epidermal necrolysis, drug rash with eosinophilia and systemic symptoms, drug-induced hypersensitivity syndrome and acute generalized exanthematous pustulosis. In some cases, it is necessary to interrupt therapy, which may result in a poorer outcome and shorten the patient's survival. This paper reviews various types of research documents published since 2016. It aims to systematize the latest knowledge and highlight the need for further research into ways to avoid adverse reactions.
Collapse
Affiliation(s)
- Patrycja Kozubek
- Student Scientific Group of Adult Allergology, 50-369 Wrocław, Poland
| | - Julia Wołoszczak
- Student Scientific Group of Adult Allergology, 50-369 Wrocław, Poland
| | - Krzysztof Gomułka
- Clinical Department of Internal Medicine, Pneumology and Allergology, Wroclaw Medical University, 50-369 Wrocław, Poland
| |
Collapse
|
9
|
Deligiorgi MV, Trafalis DT. A Concerted Vision to Advance the Knowledge of Diabetes Mellitus Related to Immune Checkpoint Inhibitors. Int J Mol Sci 2023; 24:ijms24087630. [PMID: 37108792 PMCID: PMC10146255 DOI: 10.3390/ijms24087630] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/03/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
The rubric of immune-related (ir) diabetes mellitus (DM) (irDM) encompasses various hyperglycemic disorders related to immune checkpoint inhibitors (ICPis). Beyond sharing similarities with conventional DM, irDM is a distinct, yet important, entity. The present narrative review provides a comprehensive overview of the literature regarding irDM published in major databases from January 2018 until January 2023. Initially considered rare, irDM is increasingly being reported. To advance the knowledge of irDM, the present review suggests a concerted vision comprising two intertwined aspects: a scientific-centered and a patient-centered view. The scientific-centered aspect addresses the pathophysiology of irDM, integrating: (i) ICPi-induced pancreatic islet autoimmunity in genetically predisposed patients; (ii) altered gut microbiome; (iii) involvement of exocrine pancreas; (iv) immune-related acquired generalized lipodystrophy. The patient-centered aspect is both nurtured by and nurturing the four pillars of the scientific-centered aspect: awareness, diagnosis, treatment, and monitoring of irDM. The path forward is a multidisciplinary initiative towards: (i) improved characterization of the epidemiological, clinical, and immunological profile of irDM; (ii) standardization of reporting, management, and surveillance protocols for irDM leveraging global registries; (iii) patient stratification according to personalized risk for irDM; (iv) new treatments for irDM; and (v) uncoupling ICPi efficacy from immunotoxicity.
Collapse
Affiliation(s)
- Maria V Deligiorgi
- Department of Pharmacology-Clinical Pharmacology Unit, Faculty of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Dimitrios T Trafalis
- Department of Pharmacology-Clinical Pharmacology Unit, Faculty of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
10
|
Reyna Villasmil E. Anticuerpos inmunomoduladores en el tratamiento del cáncer. REPERTORIO DE MEDICINA Y CIRUGÍA 2023. [DOI: 10.31260/repertmedcir.01217372.1361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023] Open
Abstract
Los anticuerpos inmunomoduladores (Aim) tienen la capacidad de modificar el funcionamiento del sistema inmune. Sus efectos sobre los receptores CTLA-4 y PD-1 producen disminución de la activación celular, afectando las acciones de los linfocitos T. La función de ambos receptores es cesar las funciones de las células inmunes autorreactivas que no son destruidas en las estructuras inmunes correspondientes y proteger los tejidos inflamados. Los tumores que expresan estos receptores evitan el reconocimiento por parte de las células inmunes. Los Aim bloquean los receptores y permiten a los linfocitos reconocer y responder ante antígenos neoplásicos. Las investigaciones sobre los fármacos con Aim muestran eficacia moderada en el tratamiento de algunos casos de cáncer en estadios avanzados. El uso combinado de fármacos tiene potenciales efectos sinérgicos con resultados positivos. Aún deben establecerse los posibles indicadores de éxito terapéutico y la posibilidad de reducir los efectos adversos en el uso clínico. El objetivo de esta revisión fue analizar las funciones y utilidad terapéutica de los anticuerpos inmunomoduladores en el tratamiento del cáncer.
Collapse
|
11
|
Wlosik J, Fattori S, Rochigneux P, Goncalves A, Olive D, Chretien AS. Immune biology of NSCLC revealed by single-cell technologies: implications for the development of biomarkers in patients treated with immunotherapy. Semin Immunopathol 2023; 45:29-41. [PMID: 36414693 PMCID: PMC9974692 DOI: 10.1007/s00281-022-00973-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 10/31/2022] [Indexed: 11/23/2022]
Abstract
First-line immunotherapy in non-small-cell lung cancer largely improved patients' survival. PD-L1 testing is required before immune checkpoint inhibitor initiation. However, this biomarker fails to accurately predict patients' response. On the other hand, immunotherapy exposes patients to immune-related toxicity, the mechanisms of which are still unclear. Hence, there is an unmet need to develop clinically approved predictive biomarkers to better select patients who will benefit the most from immune checkpoint inhibitors and improve risk management. Single-cell technologies provide unprecedented insight into the tumor and its microenvironment, leading to the discovery of immune cells involved in immune checkpoint inhibitor response or toxicity. In this review, we will underscore the potential of the single-cell approach to identify candidate biomarkers improving non-small-cell lung cancer patients' care.
Collapse
Affiliation(s)
- J Wlosik
- Team Immunity and Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille University UM105, Inserm U1068, 13009, Marseille, France. .,Immunomonitoring Department, Institut Paoli-Calmettes, 13009, Marseille, France.
| | - S Fattori
- Team Immunity and Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille University UM105, Inserm U1068, 13009, Marseille, France.,Immunomonitoring Department, Institut Paoli-Calmettes, 13009, Marseille, France
| | - P Rochigneux
- Team Immunity and Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille University UM105, Inserm U1068, 13009, Marseille, France.,Immunomonitoring Department, Institut Paoli-Calmettes, 13009, Marseille, France.,Department of Medical Oncology, Inserm U1068, Aix-Marseille University UM105, CNRS UMR7258, Institute Paoli-Calmettes, 13009, Marseille, France
| | - A Goncalves
- Department of Medical Oncology, Inserm U1068, Aix-Marseille University UM105, CNRS UMR7258, Institute Paoli-Calmettes, 13009, Marseille, France.,Team Cell Polarity, Cell Signaling and Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille University, Inserm U1068UM 105, 13009, Marseille, France
| | - D Olive
- Team Immunity and Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille University UM105, Inserm U1068, 13009, Marseille, France.,Immunomonitoring Department, Institut Paoli-Calmettes, 13009, Marseille, France
| | - A S Chretien
- Team Immunity and Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille University UM105, Inserm U1068, 13009, Marseille, France. .,Immunomonitoring Department, Institut Paoli-Calmettes, 13009, Marseille, France.
| |
Collapse
|
12
|
Fridman WH, Sibéril S, Pupier G, Soussan S, Sautès-Fridman C. Activation of B cells in Tertiary Lymphoid Structures in cancer: Anti-tumor or anti-self? Semin Immunol 2023; 65:101703. [PMID: 36481358 DOI: 10.1016/j.smim.2022.101703] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
Whereas T cells in the tumor microenvironment have been the main focus as cancer controlling cells and targets of immunotherapies, B cells have recently gained strong attention. Being associated to Tertiary Lymphoid Structures (TLS) located at the vicinity of tumor nests, the fate of B cell depends on TLS maturity. In immature TLS they may evolve as regulatory B cells producing immunosuppressive cytokines and promote tumor growth. In mature TLS with a germinal center, B cells are selected, amplified, undergo affinity maturation and isotypic switching, resulting in plasma cell generation and production of anti-tumor antibodies. In that case, they are associated with longer patient's survival and therapeutic response to immunotherapy. Identification of tumor specific, or tumor overexpressed, antigens recognized by "in situ" produced antibodies and their discrimination from self-antigens induced by ICI treatments is a major challenge to develop novel antibody-based immunotherapies.
Collapse
Affiliation(s)
- Wolf H Fridman
- Centre de recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, F-75006 Paris, France; Equipe labellisée Ligue Contre le Cancer, 75006 Paris, France.
| | - Sophie Sibéril
- Centre de recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, F-75006 Paris, France
| | - Guilhem Pupier
- Centre de recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, F-75006 Paris, France; Equipe labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Sarah Soussan
- Centre de recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, F-75006 Paris, France
| | - Catherine Sautès-Fridman
- Centre de recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, F-75006 Paris, France; Equipe labellisée Ligue Contre le Cancer, 75006 Paris, France
| |
Collapse
|
13
|
Lacombe RV, Sibéril S, Dimitrov JD. Immature and mature antibodies as defenders against cancer. Cell Mol Immunol 2023; 20:3-5. [PMID: 36369369 PMCID: PMC9794785 DOI: 10.1038/s41423-022-00951-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 11/01/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Robin V Lacombe
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, Paris, France
| | - Sophie Sibéril
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, Paris, France
| | - Jordan D Dimitrov
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, Paris, France.
| |
Collapse
|
14
|
Immune-checkpoint inhibitor use in patients with cancer and pre-existing autoimmune diseases. Nat Rev Rheumatol 2022; 18:641-656. [PMID: 36198831 DOI: 10.1038/s41584-022-00841-0] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2022] [Indexed: 11/08/2022]
Abstract
Immune-checkpoint inhibitors (ICIs) have dramatically changed the management of advanced cancers. Designed to enhance the antitumour immune response, they can also cause off-target immune-related adverse events (irAEs), which are sometimes severe. Although the efficacy of ICIs suggests that they could have wide-ranging benefits, clinical trials of the drugs have so far excluded patients with pre-existing autoimmune disease. However, evidence is accumulating with regard to the use of ICIs in this 'at-risk' population, with retrospective data suggesting that they have an acceptable safety profile, but that there is a risk of disease flare or other irAE occurrence. The management of immunosuppressive drugs at ICI initiation in patients with autoimmune disease (or later in instances of disease flare or irAE) remains a question of particular interest in clinical practice, in which there is always a search for the balance between protecting against autoimmunity and ensuring a good tumour response. Although temporary use of immunosuppressants seems safe, prolonged use or use at ICI initiation might hamper the antitumour immune response, prompting clinicians to use the minimal efficient immunosuppressive regimen. However, a new paradigm is emerging, in which inhibitors of TNF or IL-6 could have synergistic effects with ICIs on tumour response, while also preventing severe irAEs. If confirmed, this 'decoupling' effect on toxicity and efficacy could change therapeutic practice in this field. Knowledge of the current use of ICIs in patients with pre-existing autoimmune disease, particularly with regard to the use of immunosuppressive drugs and/or biologic DMARDs, can help to guide clinical practice.
Collapse
|
15
|
Aging-associated and CD4 T-cell–dependent ectopic CXCL13 activation predisposes to anti–PD-1 therapy-induced adverse events. Proc Natl Acad Sci U S A 2022; 119:e2205378119. [PMID: 35858347 PMCID: PMC9303859 DOI: 10.1073/pnas.2205378119] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Immune-related adverse events (irAEs) induced by immune-checkpoint blockade including antiprogrammed death receptor (PD)-1 therapy are a major problematic issue in cancer immunotherapy. Preclinical models for more physiologically occurring irAEs are potentially useful for the clarification of fundamental causes and natural developmental course of irAEs. Here, we found that in tumor-bearing aged, but not young, mice, anti–PD-(L)1 therapy alone induces irAE-like multiorgan toxicities through CD4 T-cell–derived interleukin (IL)-21 and subsequent age-specific CXCL13 expression in tertiary lymphoid structure. Consistent with this animal model, a systemic increase in CXCL13 correlates with irAE incidence in cancer patients. These findings provide insight into the development of management strategies for irAE that balance both irAE-related immune response and antitumor immune surveillance. Clinical success of immune-checkpoint blockade (ICB) cancer immunotherapy is compromised by increased risk of immune-related adverse events (irAEs). However, mechanistic action(s) of immune responses underlying development of irAE remain not fully explored. Here, we found that in tumor-bearing aged, but not young, mice, antiprogrammed death receptor (PD)-1 therapy elicited irAE-like multiorgan dysfunctions with ectopic accumulation of T and B cells in damaged organs. In this preclinical model, the organ toxicities were mediated by immunoglobulin G (IgG) deposition because administration of IG from ICB-treated aged mice induced the pathogenicity specifically in naïve aged hosts. Mechanistically, CD4 T-cell–derived interleukin (IL)-21 upregulated B-cell–homing chemokine, CXCL13, preferentially in irAE organs from aged mice treated with anti–PD-1 therapy. The ICB-induced pathogenicity was alleviated by B-cell depletion or by blockade of IL-21 or CXCL13 activity. These results suggest that age-associated immune regulatory milieu contributes to the formation of tertiary lymphoid structure-like lymphocytic aggregates in irAE organs and irAE-related toxicity employing IL-21-CXCL13-auto-antibody axis. Supporting this, a systemic increase in CXCL13 and Il21 expression in CD4 T cells correlated with irAE incidence in ICB-treated patients. These findings provide rationale for therapeutic usefulness of CXCL13 in irAE management.
Collapse
|
16
|
Deligiorgi MV, Trafalis DT. The continuum of care of anticancer treatment-induced hypothyroidism in patients with solid non thyroid tumors: time for an intimate collaboration between oncologists and endocrinologists. Expert Rev Clin Pharmacol 2022; 15:531-549. [PMID: 35757870 DOI: 10.1080/17512433.2022.2093714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Hypothyroidism is a common adverse event of various anticancer treatment modalities, constituting a notable paradigm of the integration of the endocrine perspective into precision oncology. AREAS COVERED The present narrative review provides a comprehensive and updated overview of anticancer treatment-induced hypothyroidism in patients with solid non-thyroid tumors. A study search was conducted on the following electronic databases: PubMed, Google Scholar, Scopus.com, ClinicalTrials.gov, and European Union Clinical Trials Register from 2011 until August 2021. EXPERT OPINION In patients with solid non-thyroid tumors, hypothyroidism is a common adverse event of radiotherapy, high dose interleukin 2 (HD IL-2), interferon alpha (IFN-α), bexarotene, immune checkpoint inhibitors (ICPi), and tyrosine kinase inhibitors (TKIs), while chemotherapy may induce hypothyroidism more often than initially considered. The path forward for the management of anticancer treatment-induced hypothyroidism in patients with solid non-thyroid tumors is an integrated approach grounded on 5 pillars: prevention, vigilance, diagnosis, treatment and monitoring. Current challenges concerning anticancer treatment-induced hypothyroidism await counteraction, namely awareness of the growing list of related anticancer treatments, identification of predictive factors, counteraction of diagnostic pitfalls, tuning of thyroid hormone replacement, and elucidation of its prognostic significance. Close collaboration of oncologists with endocrinologists will provide optimal patient care.
Collapse
Affiliation(s)
- Maria V Deligiorgi
- Department of Pharmacology - Clinical Pharmacology Unit, National and Kapodistrian University of Athens, Faculty of Medicine, Athens, Greece
| | - Dimitrios T Trafalis
- Department of Pharmacology - Clinical Pharmacology Unit, National and Kapodistrian University of Athens, Faculty of Medicine, Athens, Greece
| |
Collapse
|
17
|
Aran A, Garrigós L, Curigliano G, Cortés J, Martí M. Evaluation of the TCR Repertoire as a Predictive and Prognostic Biomarker in Cancer: Diversity or Clonality? Cancers (Basel) 2022; 14:cancers14071771. [PMID: 35406543 PMCID: PMC8996954 DOI: 10.3390/cancers14071771] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/22/2022] [Accepted: 03/29/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary The TCR is the T cell antigen receptor, and it is responsible of the T cell activation, through the HLA-antigen complex recognition. Studying the TCR repertoire in patients with cancer can help to better understand the anti-tumoural responses and it has been suggested to have predictive and or/prognostic values, both for the disease and in response to treatments. The aim of this review is to summarize TCR repertoire studies performed in patients with cancer found in the literature, thoroughly analyse the different factors that can be involved in shaping the TCR repertoire, and draw the current conclusions in this field, especially focusing on whether the TCR diversity—or its opposite, the clonality—can be used as predictors or prognostic biomarkers of the disease. Abstract T cells play a vital role in the anti-tumoural response, and the presence of tumour-infiltrating lymphocytes has shown to be directly correlated with a good prognosis in several cancer types. Nevertheless, some patients presenting tumour-infiltrating lymphocytes do not have favourable outcomes. The TCR determines the specificities of T cells, so the analysis of the TCR repertoire has been recently considered to be a potential biomarker for patients’ progression and response to therapies with immune checkpoint inhibitors. The TCR repertoire is one of the multiple elements comprising the immune system and is conditioned by several factors, including tissue type, tumour mutational burden, and patients’ immunogenetics. Its study is crucial to understanding the anti-tumoural response, how to beneficially modulate the immune response with current or new treatments, and how to better predict the prognosis. Here, we present a critical review including essential studies on TCR repertoire conducted in patients with cancer with the aim to draw the current conclusions and try to elucidate whether it is better to encounter higher clonality with few TCRs at higher frequencies, or higher diversity with many different TCRs at lower frequencies.
Collapse
Affiliation(s)
- Andrea Aran
- Immunology Unit, Department of Cell Biology, Physiology and Immunology, Institut de Biotecnologia I Biomedicina (IBB), Universitat Autònoma de Barcelona (UAB), 08193 Bellaterra, Spain;
| | - Laia Garrigós
- International Breast Cancer Center (IBCC), 08017 Barcelona, Spain; (L.G.); (J.C.)
| | - Giuseppe Curigliano
- Division of Early Drug Development, European Institute of Oncology, IRCCS, 20141 Milano, Italy;
- Department of Oncology and Hemato-Oncology, University of Milano, 20122 Milano, Italy
| | - Javier Cortés
- International Breast Cancer Center (IBCC), 08017 Barcelona, Spain; (L.G.); (J.C.)
- Medica Scientia Innovation Research (MedSIR), 08018 Barcelona, Spain
- Medica Scientia Innovation Research (MedSIR), Ridgewood, NJ 07450, USA
- Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain
| | - Mercè Martí
- Immunology Unit, Department of Cell Biology, Physiology and Immunology, Institut de Biotecnologia I Biomedicina (IBB), Universitat Autònoma de Barcelona (UAB), 08193 Bellaterra, Spain;
- Correspondence: ; Tel.: +34-935812409
| |
Collapse
|
18
|
Motofei IG. Nobel Prize for immune checkpoint inhibitors, understanding the immunological switching between immunosuppression and autoimmunity. Expert Opin Drug Saf 2021; 21:599-612. [PMID: 34937484 DOI: 10.1080/14740338.2022.2020243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) are a revolutionary form of immunotherapy in cancer. However, the percentage of patients responding to therapy is relatively low, while adverse effects occur in a large number of patients. In addition, the therapeutic mechanisms of ICIs are not yet completely described. AREAS COVERED The initial view (articles published in PubMed, Scopus, Web of Science, etc.) was that ICIs increase tumor-specific immunity. Recent data (collected from the same databases) suggest that the ICIs pharmacotherapy actually extends beyond the topic of immune reactivity, including additional immune pathways, such as disrupting immunosuppression and increasing tumor-specific autoimmunity. Unfortunately, there is no clear delimitation between these specific autoimmune reactions that are therapeutically beneficial, and nonspecific autoimmune reactions/toxicity that can be extremely severe side effects. EXPERT OPINION Immune checkpoint mechanisms perform a non-selective immune regulation, maintaining a dynamic balance between immunosuppression and autoimmunity. By blocking these mechanisms, ICIs actually perform an immunological reset, decreasing immunosuppression and increasing tumor-specific immunity and predisposition to autoimmunity. The predisposition to autoimmunity induces both side effects and beneficial autoimmunity. Consequently, further studies are necessary to maximize the beneficial tumor-specific autoimmunity, while reducing the counterproductive effect of associated autoimmune toxicity.
Collapse
Affiliation(s)
- Ion G Motofei
- Department of Surgery/ Oncology, Carol Davila University, Bucharest, Romania.,Department of Surgery/ Oncology, St. Pantelimon Hospital, Bucharest, Romania
| |
Collapse
|
19
|
Arai K, Matsuda M, Nakayasu H, Meguro S, Kurokami T, Kubota A, Iwasaki T, Suzuki M, Kawaguchi S, Iwashita T. Nivolumab-induced liver injury with a steroid-refractory increase in biliary enzymes, in a patient with malignant mesothelioma: An autopsy case report. Clin Case Rep 2021; 9:e05174. [PMID: 34987810 PMCID: PMC8697699 DOI: 10.1002/ccr3.5174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 09/29/2021] [Accepted: 11/07/2021] [Indexed: 12/19/2022] Open
Abstract
This is the first autopsy report of hepatotoxicity from nivolumab immunotherapy for malignant mesothelioma. The increase in levels of biliary enzymes and randomly distributed endothelial damage were steroid-refractory, but second-line option was abandoned because of cachexia. Further discussions are needed regarding the customized management of immune-related toxicities.
Collapse
Affiliation(s)
- Kazumori Arai
- Department of PathologyShizuoka General HospitalShizuokaJapan
| | - Masanori Matsuda
- Department of GastroenterologyShizuoka General HospitalShizuokaJapan
| | - Hiromasa Nakayasu
- Department of Respiratory MedicineShizuoka General HospitalShizuokaJapan
| | - Shiori Meguro
- Department of Regenerative and Infectious PathologyHamamatsu University School of MedicineHamamatsuJapan
| | - Takafumi Kurokami
- Department of GastroenterologyShizuoka General HospitalShizuokaJapan
| | - Aki Kubota
- Department of PathologyShizuoka General HospitalShizuokaJapan
| | | | - Makoto Suzuki
- Department of PathologyShizuoka General HospitalShizuokaJapan
| | - Shinya Kawaguchi
- Department of GastroenterologyShizuoka General HospitalShizuokaJapan
| | - Toshihide Iwashita
- Department of Regenerative and Infectious PathologyHamamatsu University School of MedicineHamamatsuJapan
| |
Collapse
|
20
|
Deligiorgi MV, Sagredou S, Vakkas L, Trafalis DT. The Continuum of Thyroid Disorders Related to Immune Checkpoint Inhibitors: Still Many Pending Queries. Cancers (Basel) 2021; 13:5277. [PMID: 34771441 PMCID: PMC8582503 DOI: 10.3390/cancers13215277] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/11/2021] [Accepted: 10/18/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Until more data are available to shed light on the thyroid disorders related to immune checkpoint inhibitors (ICPi) implemented for the treatment of hematological malignancies, the decision-making is guided by pertinent data derived mostly from solid tumors. METHODS The present review provides a comprehensive and updated overview of the thyroid disorders related to ICPi, namely to inhibitors of cytotoxic T-lymphocyte antigen 4 (CTLA-4), programmed cell death (PD) 1 (PD-1), and the ligand of the latter (PD-L1). RESULTS With the increasing recognition of ir thyroid disorders, many outstanding issues have emerged. Ir thyroid disorders are reminiscent of, but not identical to, thyroid autoimmunity. Interclass and intraclass ICPi differences regarding thyroid immunotoxicity await interpretation. The available data concerning the predictive value of thyroid autoantibodies for the development of ir thyroid disorders are inconclusive. Mounting data indicate an association of ir thyroid disorders with ICPi efficacy, but a causative link is still lacking. The path forward is a tailored approach, entailing: (i) the validation of tumor-specific, patient-specific, and ICPi-specific predictive factors; (ii) appropriate patient selection; (iii) the uncoupling of antitumor immunity from immunotoxicity; (iv) a multidisciplinary initiative; and (v) global registry strategies. CONCLUSIONS Untangling and harnessing the interrelationship of immuno-oncology with endocrinology underlying the ir thyroid disorders will yield the optimal patient care.
Collapse
Affiliation(s)
- Maria V. Deligiorgi
- Department of Pharmacology—Clinical Pharmacology Unit, Faculty of Medicine, National and Kapodistrian University of Athens, Building 16, 1st Floor, 75 Mikras Asias Str., Goudi, 11527 Athens, Greece; (S.S.); (L.V.); (D.T.T.)
| | | | | | | |
Collapse
|
21
|
Ma P, Jin X, Fan Z, Wang Z, Yue S, Wu C, Chen S, Wu Y, Chen M, Gu D, Zhang S, Mao R, Fan Y. Super-enhancer receives signals from the extracellular matrix to induce PD-L1-mediated immune evasion via integrin/BRAF/TAK1/ERK/ETV4 signaling. Cancer Biol Med 2021; 19:j.issn.2095-3941.2021.0137. [PMID: 34623791 PMCID: PMC9196059 DOI: 10.20892/j.issn.2095-3941.2021.0137] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
OBJECTIVE PD-L1 and PD-L2 expression levels determine immune evasion and the therapeutic efficacy of immune checkpoint blockade. The factors that drive inducible PD-L1 expression have been extensively studied, but mechanisms that result in constitutive PD-L1 expression in cancer cells are largely unknown. METHODS DNA elements were deleted in cells by CRISPR/Cas9-mediated knockout. Protein function was inhibited by chemical inhibitors. Protein levels were examined by Western blot, mRNA levels were examined by real-time RT-PCR, and surface protein expression was determined by cellular immunofluorescence and flow cytometry. Immune evasion was examined by in vitro T cell-mediated killing. RESULTS We determined the core regions (chr9: 5, 496, 378-5, 499, 663) of a previously identified PD-L1L2-super-enhancer (SE). Through systematic analysis, we found that the E26 transformation-specific (ETS) variant transcription factor (ETV4) bound to this core DNA region but not to DNA surrounding PD-L1L2SE. Genetic knockout of ETV4 dramatically reduced the expressions of both PD-L1 and PD-L2. ETV4 transcription was dependent on ERK activation, and BRAF/TAK1-induced ERK activation was dependent on extracellular signaling from αvβ3 integrin, which profoundly affected ETV4 transcription and PD-L1/L2 expression. Genetic silencing or pharmacological inhibition of components of the PD-L1L2-SE-associated pathway rendered cancer cells susceptible to T cell-mediated killing. CONCLUSIONS We identified a pathway originating from the extracellular matrix that signaled via integrin/BRAF/TAK1/ERK/ETV4 to PD-L1L2-SE to induce PD-L1-mediated immune evasion. These results provided new insights into PD-L1L2-SE activation and pathways associated with immune checkpoint regulation in cancer.
Collapse
Affiliation(s)
- Panpan Ma
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong 226001, China
- Department of Clinical Laboratory, Yancheng No. 1 People's Hospital, Yancheng 224005, China
| | - Xinxin Jin
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Zhiwei Fan
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Zhou Wang
- School of Life Sciences, Nantong University, Nantong 226001, China
| | - Suhui Yue
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Changyue Wu
- Department of Dermatology, Affiliated Hospital of Nantong University, Nantong University, Nantong 226001, China
| | - Shiyin Chen
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Yuanyuan Wu
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Miaomiao Chen
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Donghua Gu
- The Department of Urology, the Second Affiliated Hospital of Nantong University, Nantong University, Nantong 226001, China
| | - Siliang Zhang
- The Department of Radiotherapy Oncology, Harbin Medical University Cancer Hospital, Harbin 150086, China
| | - Renfang Mao
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong 226001, China
| | - Yihui Fan
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong 226001, China
| |
Collapse
|
22
|
Zhu L, Wang Z, Stebbing J, Wang Z, Peng L. Immunotherapy-Related Cystitis: Case Report and Review of the Literature. Onco Targets Ther 2021; 14:4321-4328. [PMID: 34366676 PMCID: PMC8336986 DOI: 10.2147/ott.s321965] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/19/2021] [Indexed: 12/20/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) including anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA4) and anti-programmed death cell protein 1 (anti-PD1) have extended patient survival benefit and revolutionized cancer treatment. As ICIs rely on immune regeneration to eliminate tumor cells, they can also lead to an imbalance of immune reactions often called immune-related adverse events (irAEs). Rare irAEs such as ocular or cardiac toxicity or vasculitis are seen in less than 1% of patients receiving ICIs. Immune-related cystitis remains a rare occurrence. Herein, we describe a patient with extensive-stage small cell lung cancer (SCLC) and a history of syphilis with a complete response to second-line treatment using nivolumab plus paclitaxel who complained of urinary irritation symptoms. At biopsy, we found infiltration of CD3+ and CD8+ lymphocytes in the urothelium. Although there are reports describing immune-related cystitis in cancer patients, our case has comprehensive pathological confirmation and a differentiation diagnosis. In this report, we review other cases to elucidate clinical characteristics and discuss suitable management of this rare irAE.
Collapse
Affiliation(s)
- Liping Zhu
- Department of Medical Oncology, Shouguang Hospital of Traditional Chinese Medicine, Shouguang, Shandong Province, People's Republic of China
| | - Zhiqiang Wang
- Department of Urology, Shouguang Hospital of Traditional Chinese Medicine, Shouguang, Shandong Province, People's Republic of China
| | - Justin Stebbing
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Zibing Wang
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan Province, People's Republic of China
| | - Ling Peng
- Department of Respiratory Disease, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang Province, People's Republic of China
| |
Collapse
|
23
|
Ikeda G, Yamamoto S, Kato K. The safety of current treatment options for advanced esophageal cancer after first-line chemotherapy. Expert Opin Drug Saf 2021; 21:55-65. [PMID: 34263677 DOI: 10.1080/14740338.2021.1955100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The prognosis of advanced esophageal cancer (EC) remains poor, and few effective agents are available. For advanced EC patients, a combination of platinum and fluoropyrimidine is recognized as the standard first-line treatment. After first-line treatment, taxane or irinotecan has been used. Based on the KEYNOTE-181 and the ATTRACTION-3 trials, immune checkpoint inhibitors (ICIs) such as pembrolizumab and nivolumab appear to prolong survival, compared with cytotoxic agents, as second-line treatments for advanced EC patients. In addition, ICIs have different safety profiles than conventional cytotoxic agents. Herein, we discuss the differences in the safety profiles of cytotoxic agents and ICIs for the treatment of advanced EC patients after first-line chemotherapy.ICIs as a second-line treatment are tolerable in advanced EC patients. Although infrequent, ICIs can cause immune-related adverse events that are sometimes fatal. Therefore, regular monitoring of physical and laboratory examinations is needed during and after the administration of ICIs. As the major toxicities of taxane are neutropenia and neuropathy, while those of irinotecan are neutropenia and diarrhea, appropriate supportive care or dose modification may be needed for individual patients. ICI-containing treatments have been developed not only as second-line treatments, but also as first-line treatments or for use in perioperative settings; thus, particular attention with regard to immune-related toxicities is needed.
Collapse
Affiliation(s)
- Go Ikeda
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Shun Yamamoto
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Ken Kato
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
24
|
George AS, Fernandez CJ, Eapen D, Pappachan JM. Organ-specific Adverse Events of Immune Checkpoint Inhibitor Therapy, with Special Reference to Endocrinopathies. TOUCHREVIEWS IN ENDOCRINOLOGY 2021; 17:21-32. [PMID: 35118443 PMCID: PMC8320015 DOI: 10.17925/ee.2021.17.1.21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/09/2021] [Indexed: 02/05/2023]
Abstract
Immune checkpoint inhibitors are potent and promising immunotherapeutic agents that are increasingly used for the management of various types of advanced cancers. The widespread approval of this group of drugs simultaneously revealed immune-related adverse events as unique side-effects. Endocrinopathies are one of the most common immune-related adverse events. The precise pathogenic mechanisms for these endocrinopathies are still unclear. Though few of the endocrinopathies are reversible, calling for only symptom control, most are irreversible, requiring multiple long-term hormone replacement therapies. However, in contrast to other organ-specific immune-related adverse events, patients with endocrinopathies can continue their immune checkpoint therapy, provided the hormone replacement therapy is adequate and the symptoms are controlled. Though patients who have developed immune-related adverse events demonstrate superior antitumor activity and overall survival, due to the high morbidity associated with the immune-related adverse events, researchers are trying to uncouple the antitumour activity associated with immune checkpoint inhibitor therapy from the immune-related adverse events, to preserve antitumour activity without adverse events.
Collapse
Affiliation(s)
- Annu Susan George
- Department of Medical Oncology, Lakeshore Hospital, Cochin, Kerala, India
| | - Cornelius J Fernandez
- Department of Endocrinology, Pilgrim Hospital, United Lincolnshire Hospitals NHS Trust, Boston, UK
| | - Dilip Eapen
- Department of Endocrinology, Pilgrim Hospital, United Lincolnshire Hospitals NHS Trust, Boston, UK
| | - Joseph M Pappachan
- Department of Endocrinology & Metabolism, Lancashire Teaching Hospitals NHS Trust, Preston, UK
- Manchester Metropolitan University, Manchester, UK
- The University of Manchester, Manchester, UK
| |
Collapse
|
25
|
Deligiorgi MV, Liapi C, Trafalis DT. Hypophysitis related to immune checkpoint inhibitors: An intriguing adverse event with many faces. Expert Opin Biol Ther 2021; 21:1097-1120. [PMID: 33393372 DOI: 10.1080/14712598.2021.1869211] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The incorporation of immune checkpoint inhibitors in the oncologists' arsenal is a milestone in cancer therapeutics, though not being devoid of toxicities.Areas covered: The present review provides a comprehensive and up-to-date overview of the immune-related hypophysitis with focus on the elusive biological background, the wide spectrum of the epidemiological profile, the varying clinical aspects, and the diagnostic and therapeutic challenges.Expert opinion: Historically considered distinctive of anti-cytotoxic T-lymphocyte antigen 4 (CTLA-4) monoclonal antibodies (mAbs), the immune-related hypophysitis is increasingly correlated with the anti-programmed cell-death (PD) protein 1 (PD-1)/anti-PD ligand 1 (PD-L1) mAbs. The distinct phenotype of hypophysitis related to anti-PD1/anti-PD-L1 mAbs is highlighted with focus on the immune-related isolated adrenocorticotropic (ACTH) deficiency. The immune-related central diabetes insipidus is discussed as a rare aspect of anti-CTL-A4 mAbs-induced hypophysitis, recently related to anti-PD1/anti-PD-L1 mAbs as well. The present review builds on existing literature concerning immune-related hypophysitis underscoring the pending issues still to be addressed, including (i) pathogenesis; (ii) correlation with preexisting autoimmunity; (iii) predictive value; (iv) utility of high-dose glucocorticoids; and (v) establishment of evidence-based diagnostic and therapeutic protocols. Increased awareness and constant vigilance are advocated as cornerstone of a multidisciplinary approach to ensure optimal patients' care.
Collapse
Affiliation(s)
- Maria V Deligiorgi
- Department of Pharmacology-Clinical Pharmacology Unit, National and Kapodistrian University of Athens, Faculty of Medicine, Athens, Greece
| | - Charis Liapi
- Department of Pharmacology-Clinical Pharmacology Unit, National and Kapodistrian University of Athens, Faculty of Medicine, Athens, Greece
| | - Dimitrios T Trafalis
- Department of Pharmacology-Clinical Pharmacology Unit, National and Kapodistrian University of Athens, Faculty of Medicine, Athens, Greece
| |
Collapse
|
26
|
George AS, Fernandez CJ, Eapen D, Pappachan JM. Organ-specific Adverse Events of Immune Checkpoint Inhibitor Therapy, with Special Reference to Endocrinopathies. EUROPEAN ENDOCRINOLOGY 2021. [DOI: 10.17925/ee.2021.1.1.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|