1
|
Moon S, Zhao F, Uddin MN, Tucker CJ, Karmaus PW, Fessler MB. Flotillin-2 dampens T cell antigen sensitivity and functionality. JCI Insight 2024; 9:e182328. [PMID: 39499901 DOI: 10.1172/jci.insight.182328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 10/30/2024] [Indexed: 11/13/2024] Open
Abstract
T cell receptor (TCR) engagement triggers T cell responses, yet how TCR-mediated activation is regulated at the plasma membrane remains unclear. Here, we report that deleting the membrane scaffolding protein Flotillin-2 (Flot2) increases T cell antigen sensitivity, resulting in enhanced TCR signaling and effector function in response to weak TCR stimulation. T cell-specific Flot2-deficient mice exhibited reduced tumor growth and enhanced immunity to infection. Flot2-null CD4+ T cells exhibited increased Th1 polarization, proliferation, Nur77 induction, and phosphorylation of ZAP70 and ERK1/2 upon weak TCR stimulation, indicating a sensitized TCR-triggering threshold. Single-cell RNA-Seq suggested that Flot2-null CD4+ T cells follow a similar route of activation as WT CD4+ T cells but exhibit higher occupancy of a discrete activation state under weak TCR stimulation. Given prior reports that TCR clustering influences sensitivity of T cells to stimuli, we evaluated TCR distribution with super-resolution microscopy. Flot2 ablation increased the number of surface TCR nanoclusters on naive CD4+ T cells. Collectively, we posit that Flot2 modulates T cell functionality to weak TCR stimulation, at least in part, by regulating surface TCR clustering. Our findings have implications for improving T cell reactivity in diseases with poor antigenicity, such as cancer and chronic infections.
Collapse
MESH Headings
- Animals
- Membrane Proteins/metabolism
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Mice
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Lymphocyte Activation/immunology
- Mice, Knockout
- CD4-Positive T-Lymphocytes/immunology
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 1/immunology
- Signal Transduction/immunology
- Mice, Inbred C57BL
- Phosphorylation
Collapse
Affiliation(s)
- Sookjin Moon
- Immunity, Inflammation and Disease Laboratory and
| | - Fei Zhao
- Immunity, Inflammation and Disease Laboratory and
| | | | - Charles J Tucker
- Fluorescence Microscopy and Imaging Center, National Institute of Environmental Health Sciences (NIEHS), NIH, Research Triangle Park, North Carolina, USA
| | | | | |
Collapse
|
2
|
Triantafyllou E, Gudd CLC, Possamai LA. Immune-mediated liver injury from checkpoint inhibitors: mechanisms, clinical characteristics and management. Nat Rev Gastroenterol Hepatol 2024:10.1038/s41575-024-01019-7. [PMID: 39663461 DOI: 10.1038/s41575-024-01019-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/05/2024] [Indexed: 12/13/2024]
Abstract
Immunotherapy has changed the treatment landscape for patients with cancer in the past decade. Immune checkpoint inhibitor (ICI)-based therapies have proven effective in a range of malignancies, including liver and gastrointestinal cancers, but they can cause diverse off-target organ toxicities. With the increasingly wider application of these drugs, immune-mediated liver injury from ICIs has become a commonly encountered challenge in clinical hepatology and gastroenterology. In this Review, we discuss the evidence from human and animal studies on the immunological mechanisms of immune-mediated liver injury from ICIs and summarize its clinical features and practical considerations for its management.
Collapse
Affiliation(s)
- Evangelos Triantafyllou
- Section of Hepatology and Gastroenterology, Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom.
| | - Cathrin L C Gudd
- Section of Hepatology and Gastroenterology, Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Lucia A Possamai
- Section of Hepatology and Gastroenterology, Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom.
- Liver and Antiviral Unit, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom.
| |
Collapse
|
3
|
Roberts J, Barmettler S, Murray J, Melvin JE, Ye C. Musculoskeletal immune-related adverse events of PD-(L)1 inhibitors in melanoma: a systematic review and meta-analysis. Immunotherapy 2024:1-8. [PMID: 39648646 DOI: 10.1080/1750743x.2024.2436838] [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: 08/20/2024] [Accepted: 11/27/2024] [Indexed: 12/10/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are first-line treatment for melanoma. The incidence of musculoskeletal immune-related adverse events (MSK irAEs) remains unclear. OBJECTIVE To estimate the relative risk of MSK irAEs in melanoma patients treated with ICIs targeting programmed cell death-1 or its ligand PD-(L)1 as compared to placebo. METHODS We performed a systematic literature review including phase III randomized controlled trials of adult melanoma patients comparing a PD-(L)1 inhibitor to a placebo arm. Outcomes of interest included arthralgias, arthritis, back pain and myalgias. Meta-analysis was performed to estimate the pooled relative risk of MSK irAEs over the treatment course. RESULTS Four RCTs met the inclusion criteria (n = 3,041 subjects). Use of PD-(L)1 inhibitors was associated with an increased risk of developing arthralgias (RR 1.30 [95% CI: 1.13-1.49]) and myalgias (RR 1.48 [95% CI: 1.17-1.87]) as compared to placebo. Back pain and arthritis were not reported. CONCLUSIONS Use of PD-(L)1 inhibitors is associated with a significantly increased risk of arthralgias and myalgias in melanoma patients. The risk of back pain and arthritis is unknown. IMPLICATIONS FOR PRACTICE MSK irAEs can impact quality of life and should be considered, particularly in the adjuvant setting when risks and benefits are carefully weighed.
Collapse
Affiliation(s)
- Janet Roberts
- Department of Medicine, Division of Rheumatology, Dalhousie University, Halifax, NS, Canada
- Arthritis Research Canada, Vancouver, BC, Canada
| | - Sara Barmettler
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jenna Murray
- Pediatric cardiology, Stanford Children's Health, Palo Alto, CA, USA
| | - Jennifer E Melvin
- Department of Medicine, Division of Medical Oncology, Dalhousie University, Halifax, NS, Canada
| | - Carrie Ye
- Department of Medicine, University of Alberta, Edmonton, Canada
| |
Collapse
|
4
|
Nesbitt C, Van Der Walt A, Butzkueven H, Devitt B, Jokubaitis VG. Multiple sclerosis and cancer: Navigating a dual diagnosis. Mult Scler 2024; 30:1714-1736. [PMID: 39347791 DOI: 10.1177/13524585241274523] [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] [Indexed: 10/01/2024]
Abstract
Healthcare breakthroughs are extending the lives of multiple sclerosis (MS) patients and cancer survivors, creating a growing cohort of individuals navigating a dual diagnosis. Determining the relationship between MS and cancer risk remains challenging, with inconclusive findings confounded by age, risk exposures, comorbidities, genetics and the ongoing introduction of new MS disease-modifying therapies (DMTs) across study periods.This research places significant emphasis on cancer survival, with less attention given to the impact on MS outcomes. Our review explores the existing literature on MS, cancer risk and the intersection of DMTs and cancer treatments. We aim to navigate the complexities of managing MS in cancer survivors to optimise outcomes for both conditions. Continuous research and the formulation of treatment guidelines are essential for guiding future care. Collaboration between neuro-immunology and oncology is crucial, with a need to establish databases for retrospective and ultimately prospective analysis of outcomes in these rapidly evolving fields.
Collapse
Affiliation(s)
- Cassie Nesbitt
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Department of Neurology, MSNI Multiple Sclerosis and Neuro-Immunology, Alfred Health, Melbourne, VIC, Australia
- Department of Neuroscience, Monash University, The Alfred Centre, Melbourne, VIC, Australia
| | - Anneke Van Der Walt
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Department of Neurology, MSNI Multiple Sclerosis and Neuro-Immunology, Alfred Health, Melbourne, VIC, Australia
| | - Helmut Butzkueven
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Department of Neurology, MSNI Multiple Sclerosis and Neuro-Immunology, Alfred Health, Melbourne, VIC, Australia
| | - Bianca Devitt
- Department of Oncology, Eastern Health Clinical School, Monash University, Melbourne, VIC, Australia
- Oncology Clinical Trials Unit, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Vilija G Jokubaitis
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Department of Neurology, MSNI Multiple Sclerosis and Neuro-Immunology, Alfred Health, Melbourne, VIC, Australia
- Department of Neuroscience, Monash University, The Alfred Centre, Melbourne, VIC, Australia
| |
Collapse
|
5
|
Cao L, Wang X, Ma X, Xu M, Li J. Potential of natural products and gut microbiome in tumor immunotherapy. Chin Med 2024; 19:161. [PMID: 39567970 PMCID: PMC11580227 DOI: 10.1186/s13020-024-01032-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 11/01/2024] [Indexed: 11/22/2024] Open
Abstract
Immunotherapy is a novel treatment approach for malignant tumors, which has opened a new journey of anti-tumor therapy. Although some patients will show a positive response to immunotherapy, unfortunately, most patients and cancer types do not achieve an ideal response to immunotherapy. Therefore, it is urgent to search for the pathogenesis of sensitized immunotherapy. This review indicates that Fusobacterium nucleatum, Coprobacillus cateniformis, Akkermansia muciniphila, Bifidobacterium, among others, as well as intestinal microbial metabolites are closely associated with resistance to anti-tumor immunotherapy. While natural products of pectin, inulin, jujube, anthocyanins, ginseng polysaccharides, diosgenin, camu-camu, and Inonotus hispidus (Bull).Fr. P. Karst, Icariside I, Safflower yellow, Ganoderma lucidum, and Ginsenoside Rk3, and other Chinese native medicinal compound prescriptions to boost their efficacy of anti-tumor immunotherapy through the regulation of microbiota and microbiota metabolites. However, current research mainly focuses on intestinal, liver, and lung cancer. In the future, natural products could be a viable option for treating malignant tumors, such as pancreatic, esophageal, and gastric malignancies, via sensitizing immunotherapy. Besides, the application characteristics of different types, sources and efficacy of natural products in different immune resistance scenarios also need to be further clarified through the development of future immunotherapy-related studies.
Collapse
Affiliation(s)
- Luchang Cao
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixian'ge Street, Xicheng District, Beijing, China
| | - Xinmiao Wang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixian'ge Street, Xicheng District, Beijing, China
| | - Xinyi Ma
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixian'ge Street, Xicheng District, Beijing, China
| | - Manman Xu
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixian'ge Street, Xicheng District, Beijing, China
| | - Jie Li
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixian'ge Street, Xicheng District, Beijing, China.
| |
Collapse
|
6
|
Cappelli LC. Immune checkpoint inhibitors and rheumatoid arthritis: All roads lead to PD-1? Semin Arthritis Rheum 2024:152582. [PMID: 39578183 DOI: 10.1016/j.semarthrit.2024.152582] [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: 09/23/2024] [Accepted: 10/28/2024] [Indexed: 11/24/2024]
Abstract
Immune checkpoint molecules like PD-1 and its ligand PD-L1 and CTLA-4 are important regulators of the immune system. Medications blocking these pathways, immune checkpoint inhibitors, have been used to treat a variety of malignancies, while drugs agonizing these pathways, like abatacept, have been used in treating autoimmune diseases. Modulation of the PD-1/PD-L1 axis has become important for rheumatologists to understand in several different clinical scenarios. Currently, PD-1 agonists are being developed for treatment of rheumatoid arthritis (RA). In addition to patients with RA being potentially treated with PD-1 agonists, patients with rheumatoid arthritis may be treated with anti-PD-1/PD-L1 immune checkpoint inhibitors if they develop cancer. Finally, patients treated with immune checkpoint inhibitors may develop de novo inflammatory arthritis and be referred to rheumatology for management. In all three scenarios, there remain many unanswered clinical and translational questions. The parallel development of therapeutics antagonizing and agonizing the PD-1/PD-L1 pathway presents a unique chance for discovery in inflammatory arthritis.
Collapse
Affiliation(s)
- Laura C Cappelli
- Johns Hopkins School of Medicine, Division of Rheumatology, 5501 Hopkins Bayview Circle, Suite 1B1, Baltimore, MD 21212, USA..
| |
Collapse
|
7
|
Jia Y, Wu Q, Yang Z, Sun R, Zhang K, Guo X, Xu R, Guo Y. Mechanisms of myocardial toxicity of antitumor drugs and potential therapeutic strategies: A review of the literature. Curr Probl Cardiol 2024; 49:102782. [PMID: 39134104 DOI: 10.1016/j.cpcardiol.2024.102782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 08/28/2024]
Abstract
With the successive development of chemotherapy drugs, good results have been achieved in clinical application. However, myocardial toxicity is the biggest challenge. Anthracyclines, immune checkpoint inhibitors, and platinum drugs are widely used. Targeted drug delivery, nanomaterials and dynamic imaging evaluation are all emerging research directions. This article reviews the recent literature on the use of targeted nanodrug delivery and imaging techniques to evaluate the myocardial toxicity of antineoplastic drugs, and discusses the potential mechanisms.
Collapse
Affiliation(s)
- Yang Jia
- Department of Radiology, West China Second University Hospital, Sichuan University, 20# South Renmin Road, Chengdu, Sichuan 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education; 20# South Renmin Road, Chengdu, Sichuan 610041, China
| | - Qihong Wu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education; 20# South Renmin Road, Chengdu, Sichuan 610041, China
| | - Zhigang Yang
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Street, Chengdu 610041, China
| | - Ran Sun
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education; 20# South Renmin Road, Chengdu, Sichuan 610041, China
| | - Kun Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education; 20# South Renmin Road, Chengdu, Sichuan 610041, China
| | - Xia Guo
- Department of Hematology, West China Second University Hospital, Sichuan University; 20# South Renmin Road, Chengdu, Sichuan 610041, China
| | - Rong Xu
- Department of Radiology, West China Second University Hospital, Sichuan University, 20# South Renmin Road, Chengdu, Sichuan 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education; 20# South Renmin Road, Chengdu, Sichuan 610041, China.
| | - Yingkun Guo
- Department of Radiology, West China Second University Hospital, Sichuan University, 20# South Renmin Road, Chengdu, Sichuan 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education; 20# South Renmin Road, Chengdu, Sichuan 610041, China.
| |
Collapse
|
8
|
Gohil S, Chen H, Lewis WE, Trivedi M, Aparasu RR, Johnson ML. Factors associated with the use of immune checkpoint inhibitors in older adults with metastatic non-small cell lung cancer and pre-existing autoimmune disease: A SEER-Medicare study. J Geriatr Oncol 2024; 15:102071. [PMID: 39312847 DOI: 10.1016/j.jgo.2024.102071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 08/27/2024] [Accepted: 09/16/2024] [Indexed: 09/25/2024]
Abstract
INTRODUCTION The presence of pre-existing autoimmune disease (PAD) with metastatic non-small cell lung cancer (mNSCLC) poses challenges in the use of immune checkpoint inhibitors (ICI). This study investigated factors influencing ICI utilization in older adults with mNSCLC and PAD. MATERIALS AND METHODS A retrospective cohort study with a 12-month baseline prior to treatment initiation was conducted using the SEER-Medicare data. Patients aged 66 years and above diagnosed with mNSCLC between January 2015 and December 2017, who initiated immunotherapy only/chemoimmunotherapy (IT/CIT) or chemotherapy only (CIT) and had at least one PAD diagnosed any time before treatment initiation, were included. Multiple factors, guided by the Model of Health Services Utilization, were analyzed using multivariable logistic regression. Adjusted odds ratios (aORs) and 95.0% CIs were reported. RESULTS Among 1,319 patients initiating first-line (1L) systemic therapy, 22.3% received IT/CIT and 77.7% received CT. Patients initiating IT/CIT were more likely to be 76-80 years old (aOR = 1.70, 95.0% CI = 1.02-2.81) and > 80 years old (aOR = 2.49, 95.0% CI = 1.46-4.25), reside in South (aOR = 2.32, 95.0% CI = 1.36-3.96) and West (aOR = 2.27, 95.0% CI = 1.44-3.60) SEER regions, diagnosed in 2016 (aOR = 6.36, 95.0% CI = 3.06-13.22) and 2017 (aOR = 40.45, 95.0% CI = 19.70-83.07), having a longer time to treatment initiation (aOR = 1.14, 95.0% CI = 1.08-1.19), having non-squamous tumor histology (aOR = 1.511, 95.0% CI = 1.048-2.179), and having a prior hospitalization (aOR = 1.63, 95.0% CI = 1.14-2.33). These patients were less likely to have recently used an immunosuppressant (IS) (aOR = 0.06, 95.0% CI = 0.04-0.10). DISCUSSION Several factors, such as age, region, cancer diagnosis year, time to treatment initiation, and recent IS use, intricately shape treatment decisions. Further in-depth research on each of these factors is imperative to optimize strategies for this distinctive patient population.
Collapse
Affiliation(s)
- Shrey Gohil
- University of Houston College of Pharmacy, Houston, TX, USA.
| | - Hua Chen
- University of Houston College of Pharmacy, Houston, TX, USA
| | | | | | | | | |
Collapse
|
9
|
Mori S, Nakamura K, Shimamura M, Ohe K. Use of Upadacitinib to Treat a Severe Flare-Up of Rheumatoid Arthritis During Anti-PD-1 Immune Checkpoint Inhibitor Therapy for Stage IV Squamous Cell Carcinoma of the Lung. J Clin Med 2024; 13:6257. [PMID: 39458207 PMCID: PMC11508340 DOI: 10.3390/jcm13206257] [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: 09/23/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Immune checkpoint inhibitor (ICI) therapy is becoming the standard of care for the treatment of advanced non-small-cell lung cancer. However, T-cell activation by ICIs frequently induces a flare-up of preexisting autoimmune diseases such as rheumatoid arthritis (RA). Janus kinase (JAK) inhibitors are increasingly used in the treatment of RA, but they could interfere with the efficacy of ICIs by inhibiting interferon signaling. Case Report: Here, we describe a case in which upadacitinib, a JAK1-selective inhibitor, was used to manage a severe RA flare-up occurring during ICI therapy with pembrolizumab, an anti-programmed cell death protein-1 antibody. A 54-year-old man with RA was diagnosed with grade IV lung squamous cell carcinoma. The patient had maintained RA remission for 4 years at the time of lung cancer diagnosis. After seven cycles of pembrolizumab therapy, the size of the primary tumor was markedly reduced, but a severe RA flare-up and organizing pneumonia (OP)-like pulmonary lesions occurred. Considering the severity of the flare-up, pembrolizumab was discontinued. Upadacitinib induced swift recovery from the RA flare-up and OP. Eleven months after the last pembrolizumab use, almost all metastatic lesions in the body had disappeared. We did not observe recurrence of lung cancer for more than 1 year during upadacitinib therapy. Conclusions: Upadacitinib could be a safe and effective option to treat severe RA flare-ups occurring during anti-PD-1 ICI therapy.
Collapse
Affiliation(s)
- Shunsuke Mori
- Department of Rheumatology, Clinical Research Center for Rheumatic Diseases, National Hospital Organization (NHO) Kumamoto Saishun Medical Center, Kohshi 861-1196, Kumamoto, Japan
| | - Kazuyoshi Nakamura
- Department of Respiratory Medicine, National Hospital Organization (NHO) Kumamoto Saishun Medical Center, Kohshi 861-1196, Kumamoto, Japan
| | - Minori Shimamura
- Department of Respiratory Medicine, National Hospital Organization (NHO) Kumamoto Saishun Medical Center, Kohshi 861-1196, Kumamoto, Japan
| | - Kouhei Ohe
- Department of Respiratory Medicine, National Hospital Organization (NHO) Kumamoto Saishun Medical Center, Kohshi 861-1196, Kumamoto, Japan
| |
Collapse
|
10
|
Shin YE, Kim H, Yoo JJ, Kim SG, Kim YS. A Potential Pneumothorax Induced by Immune Checkpoint Inhibitors: A Case Report and Literature Review. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1634. [PMID: 39459421 PMCID: PMC11509409 DOI: 10.3390/medicina60101634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024]
Abstract
Background and Objectives: Immune checkpoint inhibitors (ICIs), which target immune checkpoints in cancer cells, are increasingly used as a mainstay in anticancer treatment. The combination of atezolizumab and bevacizumab is also a first-line treatment for hepatocellular carcinoma (HCC). However, ICIs can cause immune-related adverse events (IrAEs) which range from mild to severe, potentially leading to the need for discontinuing immunotherapy. We report a case of a pneumothorax, a rare side effect caused by IrAEs. Materials and Methods: This paper reports a case of a 78-year-old male HCC patient who developed a recurrent pneumothorax, suspected to be an adverse effect of ICIs. Results: The patient was a current smoker with a 30 pack-year smoking history. Prior to initiating ICIs, a chest CT scan showed mild emphysema and fibrosis attributable to smoking. Following ICI treatment, the patient developed a recurrent pneumothorax. Further tests revealed no underlying cause for the pneumothorax other than the ICIs and smoking, and there were no signs of intrapulmonary metastasis or pneumonitis. Conclusions: When a pneumothorax occurs in a patient undergoing immunotherapy, it is important to consider it as a potential adverse effect of the treatment. Special attention should be given to the possibility that immunotherapy may exacerbate underlying lung conditions. Patients should be advised on the importance of smoking cessation. As there are currently no guidelines for resuming immunotherapy after a pneumothorax, it is crucial to weigh the risks and benefits and consider dose reduction or discontinuation of the medication.
Collapse
Affiliation(s)
| | | | - Jeong-Ju Yoo
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon 14584, Republic of Korea; (Y.-E.S.); (H.K.); (S.G.K.); (Y.S.K.)
| | | | | |
Collapse
|
11
|
Wang S, Luo C, Guo J, Hu R, Shen B, Lin F, Zhang C, Liao C, He J, Wang Y, Qu J, Liu L. Enhancing Therapeutic Response and Overcoming Resistance to Checkpoint Inhibitors in Ovarian Cancer through Cell Cycle Regulation. Int J Mol Sci 2024; 25:10018. [PMID: 39337506 PMCID: PMC11431879 DOI: 10.3390/ijms251810018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/11/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Tumor cells invade normal surrounding tissues through continuous division. In this study, we hypothesized that cell cycle regulation changes the immune efficacy of ovarian cancer. To investigate this hypothesis, a Förster resonance energy transfer (FRET) sensor was constructed to characterize the cell activity in real time. Cell shrinkage caused by apoptosis induces the aggregation of proteins on the cell membrane, leading to variations in the fluorescence lifetime of FRET sensors. Moreover, we tracked cell activity across various cycles following co-culture with an immune checkpoint inhibitor. Consequently, we assessed how cell cycle regulation influences immunotherapy in a tumor mouse model. This approach, which involves inhibiting typical cell cycle processes, markedly enhances the effectiveness of immunotherapy. Our findings suggest that modulating the cycle progression of cancer cells may represent a promising approach to enhance the immune response of ovarian cancer cells and the efficacy of immunotherapy based on immune checkpoint inhibitors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Liwei Liu
- State Key Laboratory of Radio Frequency Heterogeneous Integration, Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China; (S.W.); (C.L.); (J.G.); (R.H.); (B.S.); (F.L.); (C.Z.); (C.L.); (J.H.); (Y.W.); (J.Q.)
| |
Collapse
|
12
|
Meyers DE, Rittberg R, Dawe DE, Banerji S. Immunotherapy in Patients with Advanced Non-Small-Cell Lung Cancer Under-Represented by Clinical Trials. Curr Oncol 2024; 31:5498-5515. [PMID: 39330035 PMCID: PMC11431477 DOI: 10.3390/curroncol31090407] [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: 08/05/2024] [Revised: 09/08/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024] Open
Abstract
Since the initial US FDA approval of an immune checkpoint inhibitor (ICI) for the treatment of non-oncogene-driven non-small-cell lung cancer (NSCLC) nine years ago, this therapeutic strategy has been cemented as a crucial component of treatment for most of these patients. However, there is a clear efficacy-effectiveness gap whereby patients in the 'real world' seem to have more modest clinical outcomes compared to those enrolled in landmark clinical trials. This gap may be driven by the under-representation of important patient populations, including populations defined by clinical or molecular characteristics. In this review, we summarize the data outlining the evidence of ICIs in patients with poor Eastern Cooperative Oncology Group performance status (ECOG PS), underlying autoimmune disease (AID), older age, active brain metastases (BMs), and molecular aberrations such as EGFR mutations, ALK fusions, BRAF mutations and ROS1 fusions.
Collapse
Affiliation(s)
| | | | - David E Dawe
- CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
| | | |
Collapse
|
13
|
Tong W, Wang T, Bai Y, Yang X, Han P, Zhu L, Zhang Y, Shen Z. Spatial transcriptomics reveals tumor-derived SPP1 induces fibroblast chemotaxis and activation in the hepatocellular carcinoma microenvironment. J Transl Med 2024; 22:840. [PMID: 39267037 PMCID: PMC11391636 DOI: 10.1186/s12967-024-05613-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/13/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND The tumor microenvironment (TME) exerts profound effects on tumor progression and therapeutic efficacy. In hepatocellular carcinoma (HCC), the TME is enriched with cancer-associated fibroblasts (CAFs), which secrete a plethora of cytokines, chemokines, and growth factors that facilitate tumor cell proliferation and invasion. However, the intricate architecture of the TME in HCC, as well as the mechanisms driving interactions between tumor cells and CAFs, remains largely enigmatic. METHODS We analyzed 10 spatial transcriptomics and 12 single-cell transcriptomics samples sourced from public databases, complemented by 20 tumor tissue samples from liver cancer patients obtained in a clinical setting. RESULTS Our findings reveal that tumor cells exhibiting high levels of SPP1 are preferentially localized adjacent to hepatic stellate cells (HSCs). The SPP1 secreted by these tumor cells interacts with the CD44 receptor on HSCs, thereby activating the PI3K/AKT signaling pathway, which promotes the differentiation of HSCs into CAFs. Notably, blockade of the CD44 receptor effectively abrogates this interaction. Furthermore, in vivo studies demonstrate that silencing SPP1 expression in tumor cells significantly impairs HSC differentiation into CAFs, leading to a reduction in tumor volume and collagen deposition within the tumor stroma. CONCLUSIONS This study delineates the SPP1-CD44 signaling axis as a pivotal mechanism underpinning the interaction between tumor cells and CAFs. Targeting this pathway holds potential to mitigate liver fibrosis and offers novel therapeutic perspectives for liver cancer management.
Collapse
Affiliation(s)
- Wen Tong
- The First Central Clinical School, Tianjin Medical University, Tianjin, 300070, China
| | - Tianze Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yi Bai
- Department of Hepatobiliary Surgery, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Xingpeng Yang
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, Beijing, 100853, China
| | - Pinsheng Han
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Liuyang Zhu
- The First Central Clinical School, Tianjin Medical University, Tianjin, 300070, China
| | - Yamin Zhang
- Department of Hepatobiliary Surgery, Tianjin First Central Hospital, Tianjin, 300192, China.
| | - Zhongyang Shen
- Organ Transplantation Centre, Tianjin First Central Hospital, Tianjin, 300192, China.
| |
Collapse
|
14
|
You J, Guo Y, Dong Z. Polypeptides-Based Nanocarriers in Tumor Therapy. Pharmaceutics 2024; 16:1192. [PMID: 39339228 PMCID: PMC11435007 DOI: 10.3390/pharmaceutics16091192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/07/2024] [Accepted: 09/08/2024] [Indexed: 09/30/2024] Open
Abstract
Cancer remains a worldwide problem, and new treatment strategies are being actively developed. Peptides have the characteristics of good biocompatibility, strong targeting, functional diversity, modifiability, membrane permeable ability, and low immunogenicity, and they have been widely used to construct targeted drug delivery systems (DDSs). In addition, peptides, as endogenous substances, have a high affinity, which can not only regulate immune cells but also work synergistically with drugs to kill tumor cells, demonstrating significant potential for application. In this review, the latest progress of polypeptides-based nanocarriers in tumor therapy has been outlined, focusing on their applications in killing tumor cells and regulating immune cells. Additionally, peptides as carriers were found to primarily provide a transport function, which was also a subject of interest to us. At the end of the paper, the shortcomings in the construction of peptide nano-delivery system have been summarized, and possible solutions are proposed therein. The application of peptides provides a promising outlook for cancer treatment, and we hope this article can provide in-depth insights into possible future avenues of exploration.
Collapse
Affiliation(s)
- Juhua You
- School of Pharmacy, Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Xiangfang District, Harbin 150040, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Yifei Guo
- School of Pharmacy, Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Xiangfang District, Harbin 150040, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Zhengqi Dong
- School of Pharmacy, Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Xiangfang District, Harbin 150040, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| |
Collapse
|
15
|
Liu D, Li M, Zhao Z, Zhou L, Zhi F, Guo Z, Cui J. Targeting the TRIM14/USP14 Axis Enhances Immunotherapy Efficacy by Inducing Autophagic Degradation of PD-L1. Cancer Res 2024; 84:2806-2819. [PMID: 38924473 DOI: 10.1158/0008-5472.can-23-3971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/30/2024] [Accepted: 06/20/2024] [Indexed: 06/28/2024]
Abstract
Immunotherapy has greatly improved cancer treatment in recent years by harnessing the immune system to target cancer cells. The first immunotherapeutic agent approved by the FDA was IFNα. Treatment with IFNα can lead to effective immune activation and attenuate tumor immune evasion, but persistent treatment has been shown to elicit immunosuppressive effects. Here, we identified an autophagy-dependent mechanism by which IFNα triggers tumor immune evasion by upregulating PD-L1 to suppress the antitumor activity of CD8+ T cells. Mechanistically, IFNα increased the transcription of TRIM14, which recruited the deubiquitinase USP14 to inhibit the autophagic degradation of PD-L1. USP14 removed K63-linked ubiquitin chains from PD-L1, impairing its recognition by the cargo receptor p62 (also known as SQSTM1) for subsequent autophagic degradation. Combining the USP14 inhibitor IU1 with IFNα and anti-CTLA4 treatment effectively suppressed tumor growth without significant toxicity. This work suggests a strategy for targeting selective autophagy to abolish PD-L1-mediated cancer immune evasion. Significance: IFNα-induced TRIM14 transcription suppresses antitumor immunity by recruiting USP14 to inhibit autophagic degradation of PD-L1, indicating that targeting this axis could be an effective immunotherapeutic approach for treating cancer.
Collapse
Affiliation(s)
- Di Liu
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, Joint Lab of First Affiliated Hospital and School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Mengqiu Li
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, Joint Lab of First Affiliated Hospital and School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhiyao Zhao
- Greater Bay Area Institute of Precision Medicine, Guangzhou, China
| | - Liang Zhou
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, Joint Lab of First Affiliated Hospital and School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Feng Zhi
- Department of Neurosurgery, Third Affiliated Hospital of Soochow University, Changzhou, China
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| | - Zhiyong Guo
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, Joint Lab of First Affiliated Hospital and School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Jun Cui
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, Joint Lab of First Affiliated Hospital and School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
16
|
Cai W, Xiao C, Fan T, Deng Z, Wang D, Liu Y, Li C, He J. Targeting LSD1 in cancer: Molecular elucidation and recent advances. Cancer Lett 2024; 598:217093. [PMID: 38969160 DOI: 10.1016/j.canlet.2024.217093] [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: 05/09/2024] [Revised: 06/18/2024] [Accepted: 06/27/2024] [Indexed: 07/07/2024]
Abstract
Histones are the main components of chromatin, functioning as an instructive scaffold to maintain chromosome structure and regulate gene expression. The dysregulation of histone modification is associated with various pathological processes, especially cancer initiation and development, and histone methylation plays a critical role. However, the specific mechanisms and potential therapeutic targets of histone methylation in cancer are not elucidated. Lys-specific demethylase 1A (LSD1) was the first identified demethylase that specifically removes methyl groups from histone 3 at lysine 4 or lysine 9, acting as a repressor or activator of gene expression. Recent studies have shown that LSD1 promotes cancer progression in multiple epigenetic regulation or non-epigenetic manners. Notably, LSD1 dysfunction is correlated with repressive cancer immunity. Many LSD1 inhibitors have been developed and clinical trials are exploring their efficacy in monotherapy, or combined with other therapies. In this review, we summarize the oncogenic mechanisms of LSD1 and the current applications of LSD1 inhibitors. We highlight that LSD1 is a promising target for cancer treatment. This review will provide the latest theoretical references for further understanding the research progress of oncology and epigenetics, deepening the updated appreciation of epigenetics in cancer.
Collapse
Affiliation(s)
- Wenpeng Cai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Di Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yixiao Liu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
17
|
Wei Z, Guo X, Li D, Wang J, Lin C, Tan C, Wang Y, Zhu X, Tan S. Prognostic value of CMTM6 protein in hepatocellular carcinoma involving the regulation of the immune microenvironment. Int J Biol Macromol 2024; 275:133618. [PMID: 38971275 DOI: 10.1016/j.ijbiomac.2024.133618] [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: 05/20/2024] [Revised: 06/25/2024] [Accepted: 06/30/2024] [Indexed: 07/08/2024]
Abstract
There have been notable irregularities in CMTM6 expression observed in hepatocellular carcinoma (HCC), with an evident correlation between CMTM6 dysregulation and patient prognosis. The cell cycle progression came to a halt at the G2/M phase. In-depth RNA-sequencing analysis of CMTM6 knockdown Hep3B cells revealed that the most prominent effect of CMTM6 perturbation was on the expression of CXCL8, a chemokine involved in immune responses, particularly through the interleukin-17F (IL-17F) signaling pathway. By carefully examining the RNA-sequencing data obtained from CMTM6 knockdown Hep3B cells and cross-referencing it with the TCGA-LIHC database, we were able to discern that CMTM6 and programmed death-ligand 1 (PD-L1) collaboratively partake in immune regulation within T cells. Furthermore, CMTM6 exerted an influential role in modulating the infiltration of CD4+ and CD8+ T cells in the HCC microenvironment, thereby impacting the overall immune response. Our investigation found that HCC cases characterized by an elevated co-expression of CMTM6 and PD-L1, along with augmented CD4+ T cell infiltration, demonstrated comparatively longer overall and progression-free survival rates when contrasted with those displaying lower CD4+ T cell infiltration.
Collapse
Affiliation(s)
- Zhongheng Wei
- Youjiang Medical University for Nationalities Affiliated Hospital, Baise, Guangxi 533000, China
| | - Xuefeng Guo
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Di Li
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Jianchu Wang
- Youjiang Medical University for Nationalities Affiliated Hospital, Baise, Guangxi 533000, China
| | - Cheng Lin
- Youjiang Medical University for Nationalities Affiliated Hospital, Baise, Guangxi 533000, China
| | - Chao Tan
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Yue Wang
- Youjiang Medical University for Nationalities Affiliated Hospital, Baise, Guangxi 533000, China; Department of Pharmacology and Toxicology, Wright State University, Dayton, OH 45435, USA
| | - Xiaonian Zhu
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, Guangxi 541199, China.
| | - Shengkui Tan
- Youjiang Medical University for Nationalities Affiliated Hospital, Baise, Guangxi 533000, China; Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, Guangxi 541199, China.
| |
Collapse
|
18
|
Yu L, Huang K, Liao Y, Wang L, Sethi G, Ma Z. Targeting novel regulated cell death: Ferroptosis, pyroptosis and necroptosis in anti-PD-1/PD-L1 cancer immunotherapy. Cell Prolif 2024; 57:e13644. [PMID: 38594879 PMCID: PMC11294428 DOI: 10.1111/cpr.13644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/02/2024] [Accepted: 03/30/2024] [Indexed: 04/11/2024] Open
Abstract
Chemotherapy, radiotherapy, and immunotherapy represent key tumour treatment strategies. Notably, immune checkpoint inhibitors (ICIs), particularly anti-programmed cell death 1 (PD1) and anti-programmed cell death ligand 1 (PD-L1), have shown clinical efficacy in clinical tumour immunotherapy. However, the limited effectiveness of ICIs is evident due to many cancers exhibiting poor responses to this treatment. An emerging avenue involves triggering non-apoptotic regulated cell death (RCD), a significant mechanism driving cancer cell death in diverse cancer treatments. Recent research demonstrates that combining RCD inducers with ICIs significantly enhances their antitumor efficacy across various cancer types. The use of anti-PD-1/PD-L1 immunotherapy activates CD8+ T cells, prompting the initiation of novel RCD forms, such as ferroptosis, pyroptosis, and necroptosis. However, the functions and mechanisms of non-apoptotic RCD in anti-PD1/PD-L1 therapy remain insufficiently explored. This review summarises the emerging roles of ferroptosis, pyroptosis, and necroptosis in anti-PD1/PD-L1 immunotherapy. It emphasises the synergy between nanomaterials and PD-1/PD-L1 inhibitors to induce non-apoptotic RCD in different cancer types. Furthermore, targeting cell death signalling pathways in combination with anti-PD1/PD-L1 therapies holds promise as a prospective immunotherapy strategy for tumour treatment.
Collapse
Affiliation(s)
- Li Yu
- Health Science CenterYangtze UniversityJingzhouHubeiChina
- Department of UrologyJingzhou Central Hospital, Jingzhou Hospital Affiliated to Yangtze UniversityJingzhouHubeiChina
| | - Ke Huang
- Health Science CenterYangtze UniversityJingzhouHubeiChina
| | - Yixiang Liao
- Department of UrologyJingzhou Central Hospital, Jingzhou Hospital Affiliated to Yangtze UniversityJingzhouHubeiChina
| | - Lingzhi Wang
- Department of PharmacologyYong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- Cancer Science Institute of Singapore, National University of SingaporeSingaporeSingapore
- NUS Centre for Cancer Research (N2CR), National University of SingaporeSingaporeSingapore
| | - Gautam Sethi
- Department of PharmacologyYong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- NUS Centre for Cancer Research (N2CR), National University of SingaporeSingaporeSingapore
| | - Zhaowu Ma
- Health Science CenterYangtze UniversityJingzhouHubeiChina
| |
Collapse
|
19
|
Wallwork RS, Kotzin JJ, Cappelli LC, Mecoli C, Bingham CO, Wigley FM, Wilson PC, DiRenzo DD, Shah AA. Immune checkpoint inhibitor therapy in patients with cancer and pre-existing systemic sclerosis. Semin Arthritis Rheum 2024; 67:152460. [PMID: 38733668 PMCID: PMC11211049 DOI: 10.1016/j.semarthrit.2024.152460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/02/2024] [Accepted: 04/11/2024] [Indexed: 05/13/2024]
Abstract
OBJECTIVE Immune checkpoint inhibitor (ICI) therapies have dramatically improved outcomes in multiple cancers. ICI's mechanism of action involves immune system activation to augment anti-tumor immunity. Patients with pre-existing autoimmune diseases, such as systemic sclerosis (SSc), were excluded from initial ICI clinical trials due to concern that such immune system activation could precipitate an autoimmune disease flare or new, severe immune related adverse events (irAE). In the present study, we report our experience with ICIs in patients with pre-existing SSc. METHODS Patients with SSc who received ICI therapy for cancer were identified from the Johns Hopkins Scleroderma Center Research Registry. Through chart review and prespecified definitions, we identified whether patients experienced worsening SSc activity or new irAEs. SSc disease activity worsening was pre-defined as an increase in modified Rodnan skin score (mRSS), new scleroderma renal crisis, progression of interstitial lung disease (ILD) on CT scan, increased Raynaud's phenomenon frequency or severity, new pulmonary hypertension, or myositis flare. IrAEs also included active inflammatory arthritis and dermatitis. RESULTS Eight patients with SSc who received ICI therapy for cancer were included. Overall, SSc symptoms remained stable during and after ICI therapy. None of the patients with long-standing sine or limited cutaneous SSc (lcSSc) had progressive skin thickening after ICI therapy. One patient, who was early in his diffuse cutaneous SSc (dcSSc) disease course, experienced worsening skin thickening and renal crisis. Three patients (38 %) experienced a total of five irAEs (grade 2: diarrhea, mucositis and dermatitis; grade 3: pneumonitis, and grade 4: nephritis). The patient with grade 4 nephritis developed scleroderma renal crisis and immune checkpoint related nephritis simultaneously. There were no deaths due to irAEs. CONCLUSION In this study, ICI therapy was well tolerated in patients with longstanding, sine or lcSSc. IrAE were common but generally manageable. Patients with early, active SSc may be at greater risk from ICI therapy, but more research is needed.
Collapse
Affiliation(s)
- Rachel S Wallwork
- Department of Medicine, Division of Rheumatology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Jonathan J Kotzin
- Department of Medicine, Division of Rheumatology, University of Pennsylvania, Philadelphia, PA, United States
| | - Laura C Cappelli
- Department of Medicine, Division of Rheumatology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Christopher Mecoli
- Department of Medicine, Division of Rheumatology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Clifton O Bingham
- Department of Medicine, Division of Rheumatology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Fredrick M Wigley
- Department of Medicine, Division of Rheumatology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Parker C Wilson
- Department of Medicine, Division of Rheumatology, University of Pennsylvania, Philadelphia, PA, United States
| | - Dana D DiRenzo
- Department of Medicine, Division of Rheumatology, University of Pennsylvania, Philadelphia, PA, United States
| | - Ami A Shah
- Department of Medicine, Division of Rheumatology, Johns Hopkins School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
20
|
Urias Rivera AC, Pizuorno Machado A, Shatila M, Triadafilopoulos G, McQuade JL, Altan M, Zhao D, Wang Y, Shafi MA. The Effect of Immune Checkpoint Inhibitor Therapy on Pre-Existing Gastroparesis and New Onset of Symptoms of Delayed Gastric Emptying. Cancers (Basel) 2024; 16:2658. [PMID: 39123385 PMCID: PMC11311627 DOI: 10.3390/cancers16152658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/17/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) can cause myenteric plexopathy, which could result in delayed gastric emptying (GE) and possibly gastroparesis. We assessed the clinical outcomes of patients who had pre-existing gastroparesis or who developed symptoms of delayed GE following ICI therapy. We retrospectively identified adults with ICD-9 and ICD-10 codes for gastroparesis who received ICI therapy between 1 January 2020 and 31 December 2022 at a tertiary cancer center. Of 76 eligible patients, 37 had pre-existing gastroparesis; 39 (0.2% of the more than 18,000 screened) developed symptoms of delayed GE after ICI therapy, of which 27 (69%) patients had an alternative etiology for delayed GE. Four patients (11%) with pre-existing gastroparesis had a flare-up after ICI, and the median time to flare-up was 10.2 months (IQR, 0.7-28.6 months); for patients with new onset of suspected delayed GE after ICI, the median time to symptom onset was 12.8 months (IQR, 4.4-35.5 months). The clinical symptom duration of patients without an alternative etiology (74.5 days (IQR, 21.5-690 days)) and those with an alternative etiology (290 days (IQR, 147-387 days)) did not differ significantly (p = 1.00). Delayed GE after ICI therapy is a rare presentation but has a late onset and a prolonged symptom duration.
Collapse
Affiliation(s)
| | - Antonio Pizuorno Machado
- Department of Internal Medicine, The University of Texas Health Science Center, Houston, TX 77030, USA
| | - Malek Shatila
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (Y.W.)
| | - George Triadafilopoulos
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (Y.W.)
| | - Jennifer L. McQuade
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mehmet Altan
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dan Zhao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yinghong Wang
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (Y.W.)
| | - Mehnaz A. Shafi
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (Y.W.)
| |
Collapse
|
21
|
Valdez CN, Sánchez-Zuno GA, Osmani L, Ibrahim W, Galan A, Bacchiocchi A, Halaban R, Kulkarni RP, Kang I, Bucala R, Tran T. Prognostic and therapeutic insights into MIF, DDT, and CD74 in melanoma. Oncotarget 2024; 15:507-520. [PMID: 39028303 PMCID: PMC11259151 DOI: 10.18632/oncotarget.28615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/08/2024] [Indexed: 07/20/2024] Open
Abstract
Macrophage Migration Inhibitory Factor (MIF) and its homolog D-dopachrome Tautomerase (DDT) have been implicated as drivers of tumor progression across a variety of cancers. Recent evidence suggests MIF as a therapeutic target in immune checkpoint inhibition (ICI) resistant melanomas, however clinical evidence of MIF and particularly of DDT remain limited. This retrospective study analyzed 97 patients treated at Yale for melanoma between 2002-2020. Bulk-RNA sequencing of patient tumor samples from the Skin Cancer SPORE Biorepository was used to evaluate for differential gene expression of MIF, DDT, CD74, and selected inflammatory markers, and gene expression was correlated with patient survival outcomes. Our findings revealed a strong correlation between MIF and DDT levels, with no statistically significant difference across common melanoma mutations and subtypes. Improved survival was associated with lower MIF and DDT levels and higher CD74:MIF and CD74:DDT levels. High CD74:DDT and CD74:MIF levels were also associated with enrichment of infiltrating inflammatory cell markers. These data suggest DDT as a novel target in immune therapy. Dual MIF and DDT blockade may provide synergistic responses in patients with melanoma, irrespective of common mutations, and may overcome ICI resistance. These markers may also provide prognostic value for further biomarker development.
Collapse
Affiliation(s)
| | | | - Lais Osmani
- Department of Medicine, Section of Rheumatology, Allergy and Immunology, Yale University, New Haven, CT 06520, USA
| | - Wael Ibrahim
- Department of Dermatology, Yale University, New Haven, CT 06520, USA
| | - Anjela Galan
- Department of Dermatology, Yale University, New Haven, CT 06520, USA
| | | | - Ruth Halaban
- Department of Dermatology, Yale University, New Haven, CT 06520, USA
| | - Rajan P. Kulkarni
- Department of Dermatology, Oregon Health and Science University, Portland, OR 97239, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
- Department of Veterans Affairs Portland Health Care System, Operative Care Division, U.S. Portland, OR 97239, USA
| | - Insoo Kang
- School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Medicine, Section of Rheumatology, Allergy and Immunology, Yale University, New Haven, CT 06520, USA
| | - Richard Bucala
- School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Medicine, Section of Rheumatology, Allergy and Immunology, Yale University, New Haven, CT 06520, USA
- Yale Cancer Center, Yale University, New Haven, CT 06520, USA
| | - Thuy Tran
- School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Medicine, Section of Medical Oncology, Yale University, New Haven, CT 06520, USA
- Yale Cancer Center, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
22
|
Sumimoto H, Noda S, Koide H, Douke Y, Sakai K, Nishikawa A, Tomioka A, Hori M, Nakato H, Kimura Y, Tokuda A, Takano A, Teramoto K, Murata S, Daigo Y. Pre-existing autoimmune disease as a risk factor for immune-related adverse events in cancer patients receiving immune checkpoint inhibitors. PLoS One 2024; 19:e0306995. [PMID: 39012903 PMCID: PMC11251620 DOI: 10.1371/journal.pone.0306995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 06/26/2024] [Indexed: 07/18/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have been widely used as standard therapies for various cancers. However, in 20-30% of cases, ICIs can lead to immune-related adverse events (irAEs), which sometimes require discontinuation of treatment. Due to the increased risk of irAEs, patients with pre-existing autoimmune diseases (AI) are often advised against receiving ICIs. However, there has not been sufficient objective risk assessment for AI. In our study, we conducted logistic regression analysis to assess the risk of irAEs by analyzing 478 cases that received anti-PD-(L)1 Ab and/or anti-CTLA4 Ab at our hospital between April 3, 2017, and May 24, 2022. Among these cases, 28 (5.9%) had pre-existing AI. We selected several independent factors for analysis: gender, age, performance status (PS), cancer type, type of ICI, type of combined anti-cancer agents, best overall response, and pre-existing AI. The adjusted odds ratio (OR) of AI for irAE occurrence was 2.52 [95% CI: 1.08-5.86] (p = 0.033), and the adjusted OR of AI for ICI discontinuation due to irAE was 3.32 [1.41-7.78] (p = 0.006). Patients with pre-existing AI experienced a significantly shorter irAE-free survival time compared to those without AI (median irAE-free survival: 5.7 months [95% CI: 3.5-7.8] vs 10.4 months [95% CI: 7.9-12.9], respectively, p = 0.035). Frequently observed irAEs in full ICI cohort, such as dermatologic issues (7.5%), pneumonitis (7.1%), hepatitis (4.6%), and hypothyroidism (4.2%), were often accompanied by pre-existing AI. Furthermore, pre-existing AI flared up in 6 cases (37.5% in AI-positive irAE-positive cases). The activity of AI was not related to the occurrence of irAEs. Grade 3 or higher irAEs were observed in 6 out of 20 (30.0%) cases in AI-accompanied patients complicated with irAEs. Although having a complicated AI increases the risk of irAEs, it may not necessarily be a contraindication for ICI treatment if closely monitored. (292<300 characters).
Collapse
Affiliation(s)
- Hidetoshi Sumimoto
- Department of Medical Oncology, Shiga University of Medical Science, Otsu, Shiga, Japan
- Cancer Center, Shiga University of Medical Science, Otsu, Shiga, Japan
- Center for Advanced Medicine Against Cancer, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Satoshi Noda
- Department of Pharmacy, Shiga University of Medical Science Hospital, Otsu, Shiga, Japan
| | - Hiroyoshi Koide
- Department of Pharmacy, Shiga University of Medical Science Hospital, Otsu, Shiga, Japan
| | - Yutaro Douke
- Department of Pharmacy, Shiga University of Medical Science Hospital, Otsu, Shiga, Japan
| | - Kosuke Sakai
- Department of Pharmacy, Shiga University of Medical Science Hospital, Otsu, Shiga, Japan
| | - Akihito Nishikawa
- Cancer Center, Shiga University of Medical Science, Otsu, Shiga, Japan
- Nursing Department, Shiga University of Medical Science Hospital, Otsu, Shiga, Japan
| | - Azumi Tomioka
- Cancer Center, Shiga University of Medical Science, Otsu, Shiga, Japan
- Nursing Department, Shiga University of Medical Science Hospital, Otsu, Shiga, Japan
| | - Maki Hori
- Cancer Center, Shiga University of Medical Science, Otsu, Shiga, Japan
- Nursing Department, Shiga University of Medical Science Hospital, Otsu, Shiga, Japan
| | - Hiromi Nakato
- Cancer Center, Shiga University of Medical Science, Otsu, Shiga, Japan
- Nursing Department, Shiga University of Medical Science Hospital, Otsu, Shiga, Japan
| | - Yuri Kimura
- Cancer Center, Shiga University of Medical Science, Otsu, Shiga, Japan
- Nursing Department, Shiga University of Medical Science Hospital, Otsu, Shiga, Japan
| | - Aya Tokuda
- Cancer Center, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Atsushi Takano
- Department of Medical Oncology, Shiga University of Medical Science, Otsu, Shiga, Japan
- Cancer Center, Shiga University of Medical Science, Otsu, Shiga, Japan
- Center for Advanced Medicine Against Cancer, Shiga University of Medical Science, Otsu, Shiga, Japan
- Center for Antibody and Vaccine Therapy, Research Hospital, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Koji Teramoto
- Department of Medical Oncology, Shiga University of Medical Science, Otsu, Shiga, Japan
- Cancer Center, Shiga University of Medical Science, Otsu, Shiga, Japan
- Center for Advanced Medicine Against Cancer, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Satoshi Murata
- Cancer Center, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Yataro Daigo
- Department of Medical Oncology, Shiga University of Medical Science, Otsu, Shiga, Japan
- Cancer Center, Shiga University of Medical Science, Otsu, Shiga, Japan
- Center for Advanced Medicine Against Cancer, Shiga University of Medical Science, Otsu, Shiga, Japan
- Center for Antibody and Vaccine Therapy, Research Hospital, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
23
|
Emile J, Cauquil C, Carpentier D, Routier E, Robert C. Fatal myasthenia gravis (MG) associated with myositis and myocarditis in a patient with pre-existing MG treated by adjuvant nivolumab for a stage III melanoma. Eur J Cancer 2024; 205:114098. [PMID: 38761774 DOI: 10.1016/j.ejca.2024.114098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 04/23/2024] [Indexed: 05/20/2024]
Affiliation(s)
- Jeanne Emile
- Service of Dermatology, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Cécile Cauquil
- Department of Neurology, CHU Bicetre, APHP, Le Kremlin Bicêtre, France
| | | | - Emilie Routier
- Service of Dermatology, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Caroline Robert
- Service of Dermatology, Gustave Roussy, Université Paris Saclay, Villejuif, France.
| |
Collapse
|
24
|
Chen Q, Ao L, Zhao Q, Tang L, Xiong Y, Yuan Y, Wu X, Xing W, Li Z, Guo W, Liang H, Zheng SG, Lian Q, Lu D, Wan W, Xu X. WTAP/YTHDF1-mediated m 6A modification amplifies IFN-γ-induced immunosuppressive properties of human MSCs. J Adv Res 2024:S2090-1232(24)00256-X. [PMID: 38944238 DOI: 10.1016/j.jare.2024.06.019] [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: 05/14/2024] [Accepted: 06/21/2024] [Indexed: 07/01/2024] Open
Abstract
INTRODUCTION The immunosuppressive capacity of mesenchymal stem cells (MSCs) is dependent on the "license" of several pro-inflammatory factors to express immunosuppressive molecular profiles, which determines the therapeutic efficacy of MSCs in immune-mediated inflammatory diseases. Of those, interferon-γ (IFN-γ) is a key inducer for the expression of immunosuppressive molecular profiles; however, the mechanism underlying this effect is unknown. OBJECTIVES To elucidate the regulation mechanism and biological functions of N6-methyladenosine (m6A) modification in the immunosuppressive functions by the IFN-γ-licensing MSCs. METHODS Epitranscriptomic microarray analysis and MeRIP-qPCR assay were performed to identify the regulatory effect of WTAP in the IFN-γ-licensing MSCs. RIP-qPCR, western blot, qRT-PCR and RNA stability assays were used to determine the regulation of WTAP/m6A/YTHDF1 signaling axis in the expression of immunosuppressive molecules. Further, functional capacity of T cells was tested using flow cytometry, and both DSS-induced colitis mice and CIA mice were constructed to clarify the effect of WTAP and YTHDF1 in MSC-mediated immunosuppression. RESULTS We identified that IFN-γ increased the m6A methylation levels of immunosuppressive molecules, while WTAP deficiency abolished the IFN-γ-induced promotion of m6A modification. IFN-γ activated ERK signaling, which induced WTAP phosphorylation. Additionally, the stabilization of WTAP post-transcriptionally increased the mRNA expression of immunosuppressive molecules (IDO1, PD-L1, ICAM1, and VCAM1) in an m6A-YTHDF1-dependent manner; this effect further impacted the immunosuppressive capacity of IFN-γ licensing MSCs on activated T cells. Notably, WTAP/YTHDF1 overexpression enhanced the therapeutic efficacy of IFN-γ licensing MSCs and restructures the ecology of inflammation in both colitis and arthritis models. CONCLUSION Our results showed that m6A modification of IDO1, PD-L1, ICAM1, and VCAM1 mRNA mediated by WTAP-YTHDF1 is involved in the regulation of IFN-γ licensing MSCs immunosuppressive abilities, and shed a light to enhance the clinical therapeutic potential of IFN-γ-licensing MSCs.
Collapse
Affiliation(s)
- Quan Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing 400042, China; Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming 650500, China
| | - Luoquan Ao
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Qing Zhao
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Lu Tang
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Yanli Xiong
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing 400042, China; Cancer Center, Daping Hospital, Army Medical University, Chongqing, China, No.10 Changjiang Zhi Rd, Yuzhong District, Chongqing 400042, China
| | - Yuchuan Yuan
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Xiaofeng Wu
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Wei Xing
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Zhan Li
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Wei Guo
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Huaping Liang
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Song Guo Zheng
- Department of Immunology, School of Cell and Gene Therapy, Songjiang Research Institute, Shanghai Songjiang District Central Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 201600, China
| | - Qizhou Lian
- Faculty of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518000, China; Cord Blood Bank, Guangzhou Institute of Eugenics and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510000, China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Di Lu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming 650500, China
| | - Weijun Wan
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing 400042, China.
| | - Xiang Xu
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing 400042, China; Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming 650500, China; Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China.
| |
Collapse
|
25
|
Angerilli V, Vanoli A, Celin G, Ceccon C, Gasparello J, Sabbadin M, De Lisi G, Paudice M, Lenti MV, Rovedatti L, Di Sabatino A, Bazzocchi F, Lonardi S, Savarino E, Luchini C, Parente P, Grillo F, Mastracci L, Fassan M. Gastric Carcinoma in Autoimmune Gastritis: A Histopathologic and Molecular Study. Mod Pathol 2024; 37:100491. [PMID: 38588886 DOI: 10.1016/j.modpat.2024.100491] [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: 11/25/2023] [Revised: 03/04/2024] [Accepted: 04/02/2024] [Indexed: 04/10/2024]
Abstract
Patients with autoimmune gastritis (AIG) have a 13-fold risk of developing type-1 neuroendocrine tumors, whereas the risk for gastric adenocarcinoma is still uncertain. Here we describe the clinicopathologic and molecular features of a series of gastric carcinomas (GC) arising in the context of AIG. A total of 26 AIG-associated GC specimens were collected from 4 Italian Institutions. Immunohistochemistry for MUC1, MUC2, MUC5AC, MUC6, CDX2, HER2, PD-L1, CLDN18, mismatch repair (MMR) proteins, and p53 and EBV-encoded RNA (EBER) in situ hybridization were performed. Histologic and immunohistochemical features were jointly reviewed by 5 expert gastrointestinal pathologists. Next-generation sequencing analysis (TrueSight Oncology 500, Illumina) of 523 cancer-related genes was performed on 19 cases. Most tumors were diagnosed as pT1 (52%) and they were located in the corpus/fundus (58%) and associated with operative link for gastritis assessment stage II gastritis (80.8%), absence of parietal cells, complete intestinal metaplasia, and enterochromaffin-like-cell micronodular hyperplasia. Only 4 (15.4%) GCs were diagnosed during follow-up for AIG. The following histotypes were identified: 20 (77%) adenocarcinomas; 3 (11%) mixed neuroendocrine-non-neuroendocrine neoplasms, and 2 (8%) high-grade solid adenocarcinomas with focal neuroendocrine component, 1 (4%) adenocarcinoma with an amphicrine component. Overall, 7 cases (27%) showed MMR deficiency, 3 (12%) were positive (score 3+) for HER2, 6 (23%) were CLDN18 positive, and 11 (42%) had PD-L1 combined positive score ≥ 10. EBER was negative in all cases. Molecular analysis revealed 5/19 (26%) microsatellite instability (MSI) cases and 7 (37%) tumor mutational burden (TMB) high. The most frequently altered genes were TP53 (8/19, 42%), RNF43 (7/19, 37%), ERBB2 (7/19, 37% [2 amplified and 5 mutated cases]), ARID1A (6/19, 32%), and PIK3CA (4/19, 21%). In summary, AIG-associated GCs are often diagnosed at low stage in patients with longstanding misrecognized severe AIG; they often display a neuroendocrine component or differentiation, have relatively higher rates of MMR deficiency, and TMB high.
Collapse
Affiliation(s)
- Valentina Angerilli
- Department of Medicine, Surgical Pathology Unit, University of Padua, Padua, Italy
| | - Alessandro Vanoli
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia, Pavia, Italy; Anatomic Pathology Unit, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Giulia Celin
- Department of Medicine, Surgical Pathology Unit, University of Padua, Padua, Italy
| | - Carlotta Ceccon
- Department of Medicine, Surgical Pathology Unit, University of Padua, Padua, Italy
| | - Jessica Gasparello
- Department of Medicine, Surgical Pathology Unit, University of Padua, Padua, Italy
| | | | - Giuseppe De Lisi
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Michele Paudice
- Anatomic Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DICS), University of Genova, Italy; Ospedale Policlinico San Martino, IRCCS for Oncology and Neuroscience, Genova, Italy
| | - Marco Vincenzo Lenti
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; First Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Laura Rovedatti
- Gastroenterology and Digestive Endoscopy Unit, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Antonio Di Sabatino
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; First Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Francesca Bazzocchi
- Surgical Abdominal Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Sara Lonardi
- Department of Oncology, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Edoardo Savarino
- Division of Gastroenterology, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Paola Parente
- Pathology Unit, Fondazione IRCCS Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Federica Grillo
- Anatomic Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DICS), University of Genova, Italy; Ospedale Policlinico San Martino, IRCCS for Oncology and Neuroscience, Genova, Italy
| | - Luca Mastracci
- Anatomic Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DICS), University of Genova, Italy; Ospedale Policlinico San Martino, IRCCS for Oncology and Neuroscience, Genova, Italy
| | - Matteo Fassan
- Department of Medicine, Surgical Pathology Unit, University of Padua, Padua, Italy; Veneto Institute of Oncology, IOV-IRCCS, Padua, Italy.
| |
Collapse
|
26
|
Nagasaka H, Kishida T, Kouro T, Igarashi Y, Takebe S, Yamamoto S, Kondo T, Koizumi M, Terao H, Suzuki T, Nakaigawa N, Himuro H, Wei F, Sasada T. MMP1, IL-1β, sTNFR-1, and IL-6 are prognostic factors for patients with unresectable or metastatic renal cell carcinoma treated with immune checkpoint inhibitors. Int J Clin Oncol 2024; 29:832-839. [PMID: 38580797 PMCID: PMC11130069 DOI: 10.1007/s10147-024-02477-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 01/11/2024] [Indexed: 04/07/2024]
Abstract
BACKGROUND Few studies have reported reliable prognostic factors for immune checkpoint inhibitors (ICIs) in renal cell carcinoma (RCC). Therefore, we investigated prognostic factors in patients treated with ICIs for unresectable or metastatic RCC. METHODS We included 43 patients who received ICI treatment for RCC between January 2018 and October 2021. Blood samples were drawn before treatment, and 73 soluble factors in the plasma were analyzed using a bead-based multiplex assay. We examined factors associated with progression-free survival (PFS), overall survival (OS), and immune-related adverse events (irAE) using the Chi-squared test, Kaplan-Meier method, and the COX proportional hazards model. RESULTS Patients exhibited a median PFS and OS of 212 and 783 days, respectively. Significant differences in both PFS and OS were observed for MMP1 (PFS, p < 0.001; OS, p = 0.003), IL-1β (PFS, p = 0.021; OS, p = 0.008), sTNFR-1 (PFS, p = 0.017; OS, p = 0.005), and IL-6 (PFS, p = 0.004; OS, p < 0.001). Multivariate analysis revealed significant differences in PFS for MMP1 (hazard ratio [HR] 5.305, 95% confidence interval [CI], 1.648-17.082; p = 0.005) and OS for IL-6 (HR 23.876, 95% CI, 3.426-166.386; p = 0.001). Moreover, 26 patients experienced irAE, leading to ICI discontinuation or withdrawal. MMP1 was significantly associated with irAE (p = 0.039). CONCLUSION MMP1 may be associated with severe irAE, and MMP1, IL-1β, sTNFR-1, and IL-6 could serve as prognostic factors in unresectable or metastatic RCC treated with ICIs. MMP1 and IL-6 were independent predictors of PFS and OS, respectively. Thus, inhibiting these soluble factors may be promising for enhancing antitumor responses in patients with RCC treated with ICIs.
Collapse
Affiliation(s)
- Hirotaka Nagasaka
- Department of Urology, Kanagawa Cancer Center, Yokohama, Kanagawa, 241-8515, Japan
| | - Takeshi Kishida
- Department of Urology, Kanagawa Cancer Center, Yokohama, Kanagawa, 241-8515, Japan.
| | - Taku Kouro
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa, 241-8515, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Kanagawa, Japan
| | - Yuka Igarashi
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa, 241-8515, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Kanagawa, Japan
| | - Shinichi Takebe
- Department of Urology, Kanagawa Cancer Center, Yokohama, Kanagawa, 241-8515, Japan
| | - Shotaro Yamamoto
- Department of Urology, Kanagawa Cancer Center, Yokohama, Kanagawa, 241-8515, Japan
| | - Takuya Kondo
- Department of Urology, Kanagawa Cancer Center, Yokohama, Kanagawa, 241-8515, Japan
| | - Mitsuyuki Koizumi
- Department of Urology, Kanagawa Cancer Center, Yokohama, Kanagawa, 241-8515, Japan
| | - Hideyuki Terao
- Department of Urology, Kanagawa Cancer Center, Yokohama, Kanagawa, 241-8515, Japan
| | - Takahisa Suzuki
- Department of Urology, Kanagawa Cancer Center, Yokohama, Kanagawa, 241-8515, Japan
| | - Noboru Nakaigawa
- Department of Urology, Kanagawa Cancer Center, Yokohama, Kanagawa, 241-8515, Japan
| | - Hidetomo Himuro
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa, 241-8515, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Kanagawa, Japan
| | - Feifei Wei
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa, 241-8515, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Kanagawa, Japan
| | - Tetsuro Sasada
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa, 241-8515, Japan.
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Kanagawa, Japan.
| |
Collapse
|
27
|
Singh AK, Duddempudi PK, Kenchappa DB, Srivastava N, Amdare NP. Immunological landscape of solid cancer: Interplay between tumor and autoimmunity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 389:163-235. [PMID: 39396847 DOI: 10.1016/bs.ircmb.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
The immune system, a central player in maintaining homeostasis, emerges as a pivotal factor in the pathogenesis and progression of two seemingly disparate yet interconnected categories of diseases: autoimmunity and cancer. This chapter delves into the intricate and multifaceted role of the immune system, particularly T cells, in orchestrating responses that govern the delicate balance between immune surveillance and self-tolerance. T cells, pivotal immune system components, play a central role in both diseases. In autoimmunity, aberrant T cell activation drives damaging immune responses against normal tissues, while in cancer, T cells exhibit suppressed responses, allowing the growth of malignant tumors. Immune checkpoint receptors, example, initially explored in autoimmunity, now revolutionize cancer treatment via immune checkpoint blockade (ICB). Though effective in various tumors, ICB poses risks of immune-related adverse events (irAEs) akin to autoimmunity. This chapter underscores the importance of understanding tumor-associated antigens and their role in autoimmunity, immune checkpoint regulation, and their implications for both diseases. It also explores autoimmunity resulting from cancer immunotherapy and shared molecular pathways in solid tumors and autoimmune diseases, highlighting their interconnectedness at the molecular level. Additionally, it sheds light on common pathways and epigenetic features shared by autoimmunity and cancer, and the potential of repurposing drugs for therapeutic interventions. Delving deeper into these insights could unlock therapeutic strategies for both autoimmunity and cancer.
Collapse
Affiliation(s)
- Ajay K Singh
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY, United States; Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | | | | | - Nityanand Srivastava
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Nitin P Amdare
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States.
| |
Collapse
|
28
|
Chan KK, Bass AR. Impact of Non-steroidal Anti-inflammatory Drugs, Glucocorticoids, and Disease-Modifying Anti-Rheumatic Drugs on Cancer Response to Immune Checkpoint Inhibitor Therapy. Rheum Dis Clin North Am 2024; 50:337-357. [PMID: 38670731 DOI: 10.1016/j.rdc.2024.02.007] [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] [Indexed: 04/28/2024]
Abstract
Immune checkpoint inhibitor (ICI) therapy for advanced malignancies often leads to off-target adverse events. Rheumatic immune-related adverse events can often linger beyond the duration of ICI therapy and sometimes requires the use of immunomodulator therapy. A key question, therefore, is if the commonly used therapies affect cancer outcomes. In this review, the authors summarize the state of the data as it currently stands, taking into consideration the limitations of the various source studies. The most information is known about glucocorticoids, which appear to be harmful especially when used early and at high doses.
Collapse
Affiliation(s)
- Karmela K Chan
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021, USA; Department of Medicine, Division of Rheumatology, Weill Cornell Medicine.
| | - Anne R Bass
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021, USA; Department of Medicine, Division of Rheumatology, Weill Cornell Medicine
| |
Collapse
|
29
|
Sparks JA. Pre-existing Autoimmune Diseases and Immune Checkpoint Inhibitors for Cancer Treatment: Considerations About Initiation, Flares, Immune-Related Adverse Events, and Cancer Progression. Rheum Dis Clin North Am 2024; 50:147-159. [PMID: 38670718 DOI: 10.1016/j.rdc.2024.01.001] [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] [Indexed: 04/28/2024]
Abstract
Immune checkpoint inhibitors (ICIs) are increasingly used to treat a variety of cancer types. Patients with preexisting autoimmune diseases may be vulnerable to underlying disease flare as well as immune-related adverse events from ICIs. There has also been concern that immunosuppression needed to control the autoimmune disease may blunt ICI efficacy. Much of the literature is focused on diverse preexisting autoimmune diseases, which may limit conclusions to specific diseases. There is a growing literature of specific diseases, such as preexisting rheumatoid arthritis, investigating outcomes after ICI.
Collapse
Affiliation(s)
- Jeffrey A Sparks
- Department of Medicine, Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, 60 Fenwood Road, Suite 6016U, Boston, MA 02115, USA.
| |
Collapse
|
30
|
Pedrero Prieto M, Gorriz Romero D, Gómez Roch E, Pérez Miralles FC, Casanova Estruch B. Neuromyelitis optica associated with the use of Atezolizumab in a patient with advanced lung adenocarcinoma. Neurol Sci 2024; 45:2199-2202. [PMID: 38091210 DOI: 10.1007/s10072-023-07252-3] [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: 08/07/2023] [Accepted: 12/04/2023] [Indexed: 04/17/2024]
Abstract
Immune checkpoint inhibitors (ICIs) are a pharmacological group increasingly used in Oncology and Hematology. These treatments can lead to autoimmune complications, with neurological conditions, especially central nervous system (CNS) involvement, being rare. We describe a case of seropositive neuromyelitis optica in a patient with locally advanced lung adenocarcinoma treated with Atezolizumab.
Collapse
Affiliation(s)
- Manuel Pedrero Prieto
- Neurology Department, University and Polytechnic Hospital La Fe, Fernando Abril Martorell Avenue 106, 46026, Valencia, Spain.
| | - David Gorriz Romero
- Neurology Department, University and Polytechnic Hospital La Fe, Fernando Abril Martorell Avenue 106, 46026, Valencia, Spain.
| | - Elisa Gómez Roch
- Oncology Department, University and Polytechnic Hospital La Fe, Fernando Abril Martorell Avenue 106, 46026, Valencia, Spain
| | - Francisco-Carlos Pérez Miralles
- Neurology Department, University and Polytechnic Hospital La Fe, Fernando Abril Martorell Avenue 106, 46026, Valencia, Spain
| | - Bonaventura Casanova Estruch
- Neurology Department, University and Polytechnic Hospital La Fe, Fernando Abril Martorell Avenue 106, 46026, Valencia, Spain
| |
Collapse
|
31
|
Moon S, Zhao F, Uddin MN, Tucker CJ, Karmaus PWF, Fessler MB. Flotillin-2 dampens T cell antigen-sensitivity and functionality. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.26.591344. [PMID: 38746431 PMCID: PMC11092481 DOI: 10.1101/2024.04.26.591344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
T cell receptor (TCR) engagement triggers T cell responses, yet how TCR-mediated activation is regulated at the plasma membrane remains unclear. Here, we report that deleting the membrane scaffolding protein Flotillin-2 (Flot2) increases T cell antigen sensitivity, resulting in enhanced TCR signaling and effector function to weak TCR stimulation. T cell-specific Flot2-deficient mice exhibited reduced tumor growth and enhanced immunity to infection. Flot2-null CD4 + T cells exhibited increased T helper 1 polarization, proliferation, Nur77 induction, and phosphorylation of ZAP70 and LCK upon weak TCR stimulation, indicating a sensitized TCR-triggering threshold. Single cell-RNA sequencing suggested that Flot2 - null CD4 + T cells follow a similar route of activation as wild-type CD4 + T cells but exhibit higher occupancy of a discrete activation state under weak TCR stimulation. Given prior reports that TCR clustering influences sensitivity of T cells to stimuli, we evaluated TCR distribution with super-resolution microscopy. Flot2 ablation increased the number of surface TCR nanoclusters on naïve CD4 + T cells. Collectively, we posit that Flot2 modulates T cell functionality to weak TCR stimulation, at least in part, by regulating surface TCR clustering. Our findings have implications for improving T cell reactivity in diseases with poor antigenicity, such as cancer and chronic infections.
Collapse
|
32
|
Ceccarelli F, Natalucci F, Picciariello L, Cirillo A, Olivieri G, Veroli M, Pisegna S, Ciancarella C, Gelibter A, Picone V, Santini D, Botticelli A, Conti F. Antinuclear antibodies may predict the development of immune-related adverse events in asymptomatic patients treated with immune checkpoint inhibitors: results from a single-center cohort. Clin Exp Med 2024; 24:72. [PMID: 38598028 PMCID: PMC11006777 DOI: 10.1007/s10238-024-01317-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 02/29/2024] [Indexed: 04/11/2024]
Abstract
We aim at investigating the association between subclinical autoimmunity and immune-related adverse events (irAEs) in a cohort of patients treated by immune checkpoint inhibitors for solid metastatic cancer. In the context of an oncology/rheumatology outpatient clinic, we evaluated patients treated with anti-PD-1 or anti-PD-L1. Before treatment, each patient underwent a physical evaluation and a blood sample to identify the presence of a set of autoantibodies. Indeed, all the patients were followed during treatment to identify irAEs and to assess the association with autoantibodies. Fifty-one patients (M/F 16/35; median age 70 years, IQR 16.5) were evaluated; 34.8% of patients showed ANA positivity, 6.5% ENA positivity (anti-SSA), 4.3% Ratest positivity, and 2.1% (one patient) ACPA positivity. During a median period of 21 months (IQR 38.75), 39.2% of patients developed irAEs. Musculo-skeletal manifestations, in particular arthritis, were the most frequent. We found a significant association between the positivity for ANA and the development of irAES (p = 0.03, RR 2.01, 95% CI 1.03-3.92). Furthermore, the progression-free survival was significantly longer in patients developing irAEs compared to those who are not experiencing these events (p = 0.007). This study underlines the potential role of ANA positivity as a predictive biomarker for the development of irAEs.
Collapse
Affiliation(s)
- Fulvia Ceccarelli
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Arthritis Center, Rheumatology, Sapienza University of Rome, Rome, Italy
| | - Francesco Natalucci
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Arthritis Center, Rheumatology, Sapienza University of Rome, Rome, Italy.
| | - Licia Picciariello
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Arthritis Center, Rheumatology, Sapienza University of Rome, Rome, Italy
| | - Alessio Cirillo
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, Italy
| | - Giulio Olivieri
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- PhD Program in Immunology, Molecular Medicine and Applied Biotechnology, University of Rome Tor Vergata, Rome, Italy
| | - Margherita Veroli
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, Italy
| | - Simona Pisegna
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, Italy
| | - Claudia Ciancarella
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Arthritis Center, Rheumatology, Sapienza University of Rome, Rome, Italy
| | - Alain Gelibter
- Division of Medical Oncology B, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Vincenzo Picone
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, Italy
| | - Daniele Santini
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, Italy
| | - Andrea Botticelli
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, Italy
| | - Fabrizio Conti
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Arthritis Center, Rheumatology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
33
|
Casagrande S, Sopetto GB, Bertalot G, Bortolotti R, Racanelli V, Caffo O, Giometto B, Berti A, Veccia A. Immune-Related Adverse Events Due to Cancer Immunotherapy: Immune Mechanisms and Clinical Manifestations. Cancers (Basel) 2024; 16:1440. [PMID: 38611115 PMCID: PMC11011060 DOI: 10.3390/cancers16071440] [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: 03/03/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
The landscape of cancer treatment has undergone a significant transformation with the introduction of Immune Checkpoint Inhibitors (ICIs). Patients undergoing these treatments often report prolonged clinical and radiological responses, albeit with a potential risk of developing immune-related adverse events (irAEs). Here, we reviewed and discussed the mechanisms of action of ICIs and their pivotal role in regulating the immune system to enhance the anti-tumor immune response. We scrutinized the intricate pathogenic mechanisms responsible for irAEs, arising from the evasion of self-tolerance checkpoints due to drug-induced immune modulation. We also summarized the main clinical manifestations due to irAEs categorized by organ types, detailing their incidence and associated risk factors. The occurrence of irAEs is more frequent when ICIs are combined; with neurological, cardiovascular, hematological, and rheumatic irAEs more commonly linked to PD1/PD-L1 inhibitors and cutaneous and gastrointestinal irAEs more prevalent with CTLA4 inhibitors. Due to the often-nonspecific signs and symptoms, the diagnosis of irAEs (especially for those rare ones) can be challenging. The differential with primary autoimmune disorders becomes sometimes intricate, given the clinical and pathophysiological similarities. In conclusion, considering the escalating use of ICIs, this area of research necessitates additional clinical studies and practical insights, especially the development of biomarkers for predicting immune toxicities. In addition, there is a need for heightened education for both clinicians and patients to enhance understanding and awareness.
Collapse
Affiliation(s)
- Silvia Casagrande
- Unit of Neurology, Rovereto Hospital, Azienda Provinciale per i Servizi Sanitari-APSS, 38122 Trento, Italy; (S.C.); (B.G.)
| | - Giulia Boscato Sopetto
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38122 Trento, Italy; (G.B.S.); (G.B.); (V.R.)
| | - Giovanni Bertalot
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38122 Trento, Italy; (G.B.S.); (G.B.); (V.R.)
- Center for Medical Sciences (CISMed), University of Trento, 38122 Trento, Italy
- Multizonal Unit of Pathology, APSS, 38122 Trento, Italy
| | - Roberto Bortolotti
- Unit of Rheumatology, Santa Chiara Regional Hospital, APSS, 38122 Trento, Italy;
| | - Vito Racanelli
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38122 Trento, Italy; (G.B.S.); (G.B.); (V.R.)
- Center for Medical Sciences (CISMed), University of Trento, 38122 Trento, Italy
- Unit of Internal Medicine, Santa Chiara Regional Hospital, APSS, 38122 Trento, Italy
| | - Orazio Caffo
- Unit of Oncology, Santa Chiara Regional Hospital, APSS, 38122 Trento, Italy; (O.C.); (A.V.)
| | - Bruno Giometto
- Unit of Neurology, Rovereto Hospital, Azienda Provinciale per i Servizi Sanitari-APSS, 38122 Trento, Italy; (S.C.); (B.G.)
- Center for Medical Sciences (CISMed), University of Trento, 38122 Trento, Italy
- Department of Psychology and Cognitive Sciences (DIPSCO), University of Trento, 38122 Trento, Italy
| | - Alvise Berti
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38122 Trento, Italy; (G.B.S.); (G.B.); (V.R.)
- Center for Medical Sciences (CISMed), University of Trento, 38122 Trento, Italy
- Unit of Rheumatology, Santa Chiara Regional Hospital, APSS, 38122 Trento, Italy;
| | - Antonello Veccia
- Unit of Oncology, Santa Chiara Regional Hospital, APSS, 38122 Trento, Italy; (O.C.); (A.V.)
| |
Collapse
|
34
|
Zhang Y, Cao M, Wu Y, Malih S, Xu D, Yang E, Younis MH, Lin W, Zhao H, Wang C, Liu Q, Engle JW, Rasaee MJ, Guan Y, Huang G, Liu J, Cai W, Xie F, Wei W. Preclinical development of novel PD-L1 tracers and first-in-human study of [ 68Ga]Ga-NOTA-RW102 in patients with lung cancers. J Immunother Cancer 2024; 12:e008794. [PMID: 38580333 PMCID: PMC11002357 DOI: 10.1136/jitc-2024-008794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND The programmed cell death protein-1 (PD-1)/programmed death receptor ligand 1 (PD-L1) axis critically facilitates cancer cells' immune evasion. Antibody therapeutics targeting the PD-1/PD-L1 axis have shown remarkable efficacy in various tumors. Immuno-positron emission tomography (ImmunoPET) imaging of PD-L1 expression may help reshape solid tumors' immunotherapy landscape. METHODS By immunizing an alpaca with recombinant human PD-L1, three clones of the variable domain of the heavy chain of heavy-chain only antibody (VHH) were screened, and RW102 with high binding affinity was selected for further studies. ABDRW102, a VHH derivative, was further engineered by fusing RW102 with the albumin binder ABD035. Based on the two targeting vectors, four PD-L1-specific tracers ([68Ga]Ga-NOTA-RW102, [68Ga]Ga-NOTA-ABDRW102, [64Cu]Cu-NOTA-ABDRW102, and [89Zr]Zr-DFO-ABDRW102) with different circulation times were developed. The diagnostic efficacies were thoroughly evaluated in preclinical solid tumor models, followed by a first-in-human translational investigation of [68Ga]Ga-NOTA-RW102 in patients with non-small cell lung cancer (NSCLC). RESULTS While RW102 has a high binding affinity to PD-L1 with an excellent KD value of 15.29 pM, ABDRW102 simultaneously binds to human PD-L1 and human serum albumin with an excellent KD value of 3.71 pM and 3.38 pM, respectively. Radiotracers derived from RW102 and ABDRW102 have different in vivo circulation times. In preclinical studies, [68Ga]Ga-NOTA-RW102 immunoPET imaging allowed same-day annotation of differential PD-L1 expression with specificity, while [64Cu]Cu-NOTA-ABDRW102 and [89Zr]Zr-DFO-ABDRW102 enabled longitudinal visualization of PD-L1. More importantly, a pilot clinical trial shows the safety and diagnostic value of [68Ga]Ga-NOTA-RW102 immunoPET imaging in patients with NSCLCs and its potential to predict immune-related adverse effects following PD-L1-targeted immunotherapies. CONCLUSIONS We developed and validated a series of PD-L1-targeted tracers. Initial preclinical and clinical evidence indicates that immunoPET imaging with [68Ga]Ga-NOTA-RW102 holds promise in visualizing differential PD-L1 expression, selecting patients for PD-L1-targeted immunotherapies, and monitoring immune-related adverse effects in patients receiving PD-L1-targeted treatments. TRIAL REGISTRATION NUMBER NCT06165874.
Collapse
Affiliation(s)
- You Zhang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Min Cao
- Department of Thoracic Surgery,Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanfei Wu
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Sara Malih
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Dong Xu
- Department of Thoracic Surgery, Huashan Hospital Fudan University, Shanghai, China
| | - Erpeng Yang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Muhsin H Younis
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Wilson Lin
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Haitao Zhao
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Cheng Wang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiufang Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jonathan W Engle
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Mohammad J Rasaee
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Yihui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
35
|
Hansen I, Gebhardt C, Booken N, Schneider SW. Erfolgreiche Behandlung eines Checkpoint‐Inhibitor‐assoziierten bullösen Pemphigoids mit Dupilumab bei Angiosarkom. J Dtsch Dermatol Ges 2024; 22:587-590. [PMID: 38574016 DOI: 10.1111/ddg.15340_g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 11/16/2023] [Indexed: 04/06/2024]
Affiliation(s)
- Inga Hansen
- Klinik und Poliklinik für Dermatologie und Venerologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg
| | - Christoffer Gebhardt
- Klinik und Poliklinik für Dermatologie und Venerologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg
| | - Nina Booken
- Klinik und Poliklinik für Dermatologie und Venerologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg
| | - Stefan W Schneider
- Klinik und Poliklinik für Dermatologie und Venerologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg
| |
Collapse
|
36
|
Hansen I, Gebhardt C, Booken N, Schneider SW. Successful treatment of checkpoint inhibitor-associated bullous pemphigoid with dupilumab in a patient with angiosarcoma. J Dtsch Dermatol Ges 2024; 22:587-589. [PMID: 38379256 DOI: 10.1111/ddg.15340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 11/16/2023] [Indexed: 02/22/2024]
Affiliation(s)
- Inga Hansen
- Klinik und Poliklinik für Dermatologie und Venerologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg
| | - Christoffer Gebhardt
- Klinik und Poliklinik für Dermatologie und Venerologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg
| | - Nina Booken
- Klinik und Poliklinik für Dermatologie und Venerologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg
| | - Stefan W Schneider
- Klinik und Poliklinik für Dermatologie und Venerologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg
| |
Collapse
|
37
|
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
|
38
|
Ma F, Lin Y, Ni Z, Wang S, Zhang M, Wang X, Zhang Z, Luo X, Miao X. Microwave ablation enhances the systemic immune response in patients with lung cancer. Oncol Lett 2024; 27:106. [PMID: 38298427 PMCID: PMC10829076 DOI: 10.3892/ol.2024.14239] [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: 05/02/2023] [Accepted: 11/06/2023] [Indexed: 02/02/2024] Open
Abstract
Microwave ablation (MWA) is a key alternative therapy to conventional surgery for the treatment of lung cancer. In addition to eliminating local tumors, MWA may promote antitumor immunological responses, such as abscopal effects in distant lesions. However, the intensity of MWA is limited and the underlying mechanisms are not well-defined. The present study assessed the impact of MWA on immune cell subsets and cytokines in patients with lung cancer. A total of 45 patients with lung cancer who underwent percutaneous lung tumor MWA were enrolled. Peripheral blood samples were collected before and 24 h after MWA and changes in immune cell subsets [lymphocytes, CD3+, CD4+ and CD8+ T cells, B cells and natural killer (NK) cells] and serum cytokine levels (IL-1β, IL-2, IL-4-6, IL-8, IL-10, IL-12p70, IL-17A and F, IL-22, TNF-α, TNF-β and IFN-γ) were assessed by flow cytometry and ELISA. The number of total lymphocytes, CD4+ T and NK cells in the peripheral blood significantly decreased 24 h after MWA, while number of CD8+ T cells remained stable, leading to a higher proportion of CD8+ T cells. In addition, the serum levels of IL-2, IL-1β, IL-6, IL-12p70, IL-22, TNF-α and IFN-γ were significantly increased 24 h after MWA, indicating a T helper 1 type immune response. The immune response in patients with advanced stage disease was comparable with patients in the early stage group; however, the number of total lymphocytes and CD3+ T cells significantly decreased and the ratio of CD4/CD8 and IL-2 levels significantly increased. The early immune response after MWA may contribute to systemic antitumor immunity in patients with both early and advanced disease. Thus, MWA may exhibit potential as a local therapy and trigger abscopal effects in distant lesions in patients with lung cancer.
Collapse
Affiliation(s)
- Fuqi Ma
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Yuhua Lin
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Zhenhua Ni
- Central Lab, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Shiqiang Wang
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Mengjie Zhang
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Xiaoe Wang
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Zhuhua Zhang
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Xuming Luo
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Xiayi Miao
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| |
Collapse
|
39
|
Dimitrova M, Weber J. Melanoma-Modern Treatment for Metastatic Melanoma. Cancer J 2024; 30:79-83. [PMID: 38527260 DOI: 10.1097/ppo.0000000000000707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
ABSTRACT Traditional chemotherapy has been ineffective in the treatment of metastatic melanoma. Until the use of checkpoint inhibitors, patients had very limited survival. Since the original US Food and Drug Administration approval of ipilimumab over a decade ago, the armamentarium of immunotherapeutic agents has expanded to include programmed cell death protein 1 and lymphocyte activation gene 3 antibodies, requiring a nuanced approach to the selection of frontline treatments, managing patients through recurrence and progression, and determining length of therapy. Herein, we review the existing evidence supporting current standard immunotherapy regimens and discuss the clinical decision-making involved in treating patients with metastatic melanoma with checkpoint inhibitors.
Collapse
Affiliation(s)
- Maya Dimitrova
- From the Laura and Isaac Perlmutter Comprehensive Cancer Center, NYU Grossman School of Medicine, New York, NY
| | | |
Collapse
|
40
|
Seidel MG, Hauck F. Multilayer concept of autoimmune mechanisms and manifestations in inborn errors of immunity: Relevance for precision therapy. J Allergy Clin Immunol 2024; 153:615-628.e4. [PMID: 38185417 DOI: 10.1016/j.jaci.2023.12.022] [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: 08/25/2023] [Revised: 12/12/2023] [Accepted: 12/21/2023] [Indexed: 01/09/2024]
Abstract
Autoimmunity in inborn errors of immunity (IEIs) has a multifactorial pathogenesis and develops subsequent to a genetic predisposition in conjunction with gene regulation, environmental modifiers, and infectious triggers. On the basis of incremental data availability owing to upfront application of omics technologies, a more granular and dynamic view of mechanisms and manifestations is warranted. Here, we present a comprehensive novel concept of autoimmunity in IEIs that considers multiple layers of interdependent elements and connects 101 causative genes or deletions according to the quality of the allelic variants with 47 molecular pathways and 22 immune effector mechanisms. Furthermore, we list 50 resulting manifestations together with the corresponding Human Phenotype Ontology terms and review the types and frequencies of the most relevant clinical presentations. When all of its elements are taken together, this concept (1) extends the historical anatomic view of central versus peripheral tolerance toward multiple interdependent mechanisms of immune tolerance, (2) delineates the mechanisms underlying the protean clinical manifestations, and thereby, (3) points toward the most suitable precision therapy for autoimmunity in IEIs. The multilayer concept of autoimmune mechanisms and manifestations in IEIs will facilitate research design and provide clinical guidance on the use of precision medicine irrespective of the data depth available in each health care scenario.
Collapse
Affiliation(s)
- Markus G Seidel
- Research Unit for Pediatric Hematology and Immunology, Division of Pediatric Hemato-Oncology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria.
| | - Fabian Hauck
- Division of Pediatric Immunology and Rheumatology, Department of Pediatrics, Dr von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, München, Germany.
| |
Collapse
|
41
|
Isaacs DJ, Kathuria-Prakash N, Hilder R, Lechner MG, Drakaki A. Risk of severe immune-related adverse events in cancer patients with pre-existing autoimmunity receiving immune checkpoint inhibitor therapy. CURRENT CANCER REPORTS 2024; 5:168-180. [PMID: 39397890 PMCID: PMC11469208 DOI: 10.25082/ccr.2023.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Purpose To evaluate the frequency and severity of irAEs in patients with pre-existing autoimmunity, including irAE-related morbidity and mortality, irAE-related management and resolution, and outcome of ICI rechallenge, to better understand the treatment options for this vulnerable patient population. Methods We designed a retrospective, single-center, case-control study at a large, academic medical center to evaluate the incidence and severity of irAEs in patients with pre-existing autoimmunity compared to controls. Controls were matched 2:1 for age, sex, cancer histology, and ICI class. Patients were identified with ICD 9 and 10 codes followed by manual chart extraction. Cases were defined as patients with pre-existing, systemic autoimmunity. The primary outcome was severe irAE (Grade 3 or higher by Common Terminology Criteria for Adverse Events) within 6 months of ICI therapy. Secondary outcomes included response to ICIs, resolution of the irAE, ICI rechallenge success, and survival. Statistical analyses were performed by Chi-square, Fishers exact, Mann-Whitney, and Log-rank tests. Results Of 3,130 patients treated with ICIs from 2015-2021, 28 cases with pre-existing autoimmune disease were identified and were matched with 56 controls. Pre-existing autoimmune conditions included antiphospholipid syndrome, inflammatory polyarthritis, juvenile rheumatoid arthritis, multiple sclerosis, psoriatic arthritis, rheumatoid arthritis, and type I diabetes. Multiple cancer histologies, including genitourinary, gynecologic, head & neck, hepatobiliary, lung, melanoma, and pancreatic, were represented. Six of 28 cases (21.4%) experienced severe irAEs compared to 9/56 (16.1%) controls; the odds of developing a severe irAE were not significantly different (OR 0.43, 95% CI 0.083-2.33, p = 0.627, ns). Moreover, there were no significant differences in overall survival or tumor response between the two groups. The majority of irAEs resolved without long-term sequelae (66.7% of cases, 55.6% of controls). The majority of patients who were rechallenged with ICIs were successful in continuing therapy (66.7% of cases, 100% of controls). Conclusion Our study suggests that patients with pre-existing autoimmune disease can be treated with ICI cancer therapies and experience rates of severe irAEs and overall survival that are similar to those of the general population. These data can aid oncologists in discussing risks and benefits of ICIs when treating patients with pre-existing autoimmunity and solid tumors.
Collapse
Affiliation(s)
- Dayna Jill Isaacs
- Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Nikhita Kathuria-Prakash
- Division of Hematology and Oncology, Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Robin Hilder
- Division of Endocrinology, Diabetes, and Metabolism, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Melissa G. Lechner
- Division of Endocrinology, Diabetes, and Metabolism, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Alexandra Drakaki
- Division of Hematology and Oncology, Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| |
Collapse
|
42
|
McCarter KR, Arabelovic S, Wang X, Wolfgang T, Yoshida K, Qian G, Kowalski EN, Vanni KMM, LeBoeuf NR, Buchbinder EI, Gedmintas L, MacFarlane LA, Rao DA, Shadick NA, Gravallese EM, Sparks JA. Immunomodulator use, risk factors and management of flares, and mortality for patients with pre-existing rheumatoid arthritis after immune checkpoint inhibitors for cancer. Semin Arthritis Rheum 2024; 64:152335. [PMID: 38100899 PMCID: PMC10842881 DOI: 10.1016/j.semarthrit.2023.152335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 11/17/2023] [Accepted: 11/28/2023] [Indexed: 12/17/2023]
Abstract
OBJECTIVE To investigate immunomodulator use, risk factors and management for rheumatoid arthritis (RA) flares, and mortality for patients with pre-existing RA initiating immune checkpoint inhibitors (ICI) for cancer. METHODS We performed a retrospective cohort study of all patients with RA meeting 2010 ACR/EULAR criteria that initiated ICI for cancer at Mass General Brigham or Dana-Farber Cancer Institute in Boston, MA (2011-2022). We described immunomodulator use and changes at baseline of ICI initiation. We identified RA flares after baseline, categorized the severity, and described the management. Baseline factors were examined for RA flare risk using Fine and Gray competing risk models. We performed a landmark analysis to limit the potential for immortal time bias, where the analysis started 3 months after ICI initiation. Among those who survived at least 3 months, we examined whether RA flare within 3 months after ICI initiation was associated with mortality using Cox regression. RESULTS Among 11,901 patients who initiated ICI for cancer treatment, we analyzed 100 pre-existing RA patients (mean age 70.3 years, 63 % female, 89 % on PD-1 monotherapy, 50 % lung cancer). At ICI initiation, 71 % were seropositive, 82 % had remission/low RA disease activity, 24 % were on glucocorticoids, 35 % were on conventional synthetic disease-modifying antirheumatic drugs (DMARDs), and 10 % were on biologic or targeted synthetic DMARDs. None discontinued glucocorticoids and 3/35 (9 %) discontinued DMARDs in anticipation of starting ICI. RA flares occurred in 46 % (incidence rate 1.84 per 1000 person-months, 95 % CI 1.30, 2.37); 31/100 flared within 3 months of baseline. RA flares were grade 1 in 16/46 (35 %), grade 2 in 25/46 (54 %), and grade 3 in 5/46 (11 %); 2/46 (4 %) required hospitalization for RA flare. Concomitant immune-related adverse events occurred in 15/46 (33 %) that flared. A total of 72/100 died during follow-up; 21 died within 3 months of baseline. Seropositivity had an age-adjusted sdHR of 1.95 (95 % CI 1.02, 3.71) for RA flare compared to seronegativity, accounting for competing risk of death. Otherwise, no baseline factors were associated with RA flare, including cancer type, disease activity, RA duration, and deformities. 9/46 (20 %) patients had their ICI discontinued/paused due to RA flares. In the landmark analysis among 79 patients who survived at least 3 months, RA flare in the first 3 months was not associated with lower mortality (adjusted HR 1.24, 95 % CI 0.71, 2.16) compared to no RA flare. CONCLUSION Among patients with pre-existing RA, few changed immunomodulator medications in anticipation of starting ICI, but RA flares occurred in nearly half. RA flares were mostly mild and treated with typical therapies. Seropositivity was associated with RA flare risk. A minority had severe RA flares requiring disruption of ICI, and RA flares were not associated with mortality.
Collapse
Affiliation(s)
- Kaitlin R McCarter
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States of America; Harvard Medical School, Boston, Massachusetts, United States of America
| | - Senada Arabelovic
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States of America; Harvard Medical School, Boston, Massachusetts, United States of America; Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Xiaosong Wang
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States of America; Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Taylor Wolfgang
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States of America; Harvard Medical School, Boston, Massachusetts, United States of America; Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Kazuki Yoshida
- Harvard Medical School, Boston, Massachusetts, United States of America; Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Grace Qian
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States of America; Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Emily N Kowalski
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States of America; Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Kathleen M M Vanni
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States of America; Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Nicole R LeBoeuf
- Harvard Medical School, Boston, Massachusetts, United States of America; Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts, United States of America; Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Elizabeth I Buchbinder
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States of America; Harvard Medical School, Boston, Massachusetts, United States of America; Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Lydia Gedmintas
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States of America; Harvard Medical School, Boston, Massachusetts, United States of America; Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Lindsey A MacFarlane
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States of America; Harvard Medical School, Boston, Massachusetts, United States of America; Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Deepak A Rao
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States of America; Harvard Medical School, Boston, Massachusetts, United States of America; Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Nancy A Shadick
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States of America; Harvard Medical School, Boston, Massachusetts, United States of America; Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Ellen M Gravallese
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States of America; Harvard Medical School, Boston, Massachusetts, United States of America; Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Jeffrey A Sparks
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States of America; Harvard Medical School, Boston, Massachusetts, United States of America; Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, United States of America.
| |
Collapse
|
43
|
Farina A, Villagrán-García M, Vogrig A, Zekeridou A, Muñiz-Castrillo S, Velasco R, Guidon AC, Joubert B, Honnorat J. Neurological adverse events of immune checkpoint inhibitors and the development of paraneoplastic neurological syndromes. Lancet Neurol 2024; 23:81-94. [PMID: 38101905 DOI: 10.1016/s1474-4422(23)00369-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 09/14/2023] [Accepted: 09/20/2023] [Indexed: 12/17/2023]
Abstract
Immune checkpoint inhibitors, a class of oncological treatments that enhance antitumour immunity, can trigger neurological adverse events closely resembling paraneoplastic neurological syndromes. Unlike other neurological adverse events caused by these drugs, post-immune checkpoint inhibitor paraneoplastic neurological syndromes predominantly affect the CNS and are associated with neural antibodies and cancer types commonly found also in spontaneous paraneoplastic neurological syndromes. Furthermore, post-immune checkpoint inhibitor paraneoplastic neurological syndromes have poorer neurological outcomes than other neurological adverse events of immune checkpoint inhibitors. Early diagnosis and initiation of immunosuppressive therapy are likely to be crucial in preventing the accumulation of neurological disability. Importantly, the neural antibodies found in patients with post-immune checkpoint inhibitor paraneoplastic neurological syndromes are sometimes detected before treatment, indicating that these antibodies might help to predict the development of neurological adverse events. Experimental and clinical evidence suggests that post-immune checkpoint inhibitor paraneoplastic neurological syndromes probably share immunological features with spontaneous paraneoplastic syndromes. Hence, the study of post-immune checkpoint inhibitor paraneoplastic neurological syndromes can help in deciphering the immunopathogenesis of paraneoplastic neurological syndromes and in identifying novel therapeutic targets.
Collapse
Affiliation(s)
- Antonio Farina
- Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, Neurological Hospital, Bron, France; MeLiS, UCBL-CNRS UMR 5284, INSERM U1314, Université Claude Bernard Lyon 1, Lyon, France; Department of Neuroscience, Psychology, Pharmacology and Child Health, University of Florence, Florence, Italy
| | - Macarena Villagrán-García
- Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, Neurological Hospital, Bron, France; MeLiS, UCBL-CNRS UMR 5284, INSERM U1314, Université Claude Bernard Lyon 1, Lyon, France
| | - Alberto Vogrig
- Clinical Neurology, Santa Maria della Misericordia University Hospital, Azienda Sanitaria Universitaria Friuli Centrale (ASU FC), Udine, Italy; Department of Medicine (DAME), University of Udine Medical School, Udine, Italy
| | - Anastasia Zekeridou
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Sergio Muñiz-Castrillo
- Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, Neurological Hospital, Bron, France; MeLiS, UCBL-CNRS UMR 5284, INSERM U1314, Université Claude Bernard Lyon 1, Lyon, France; Stanford Center for Sleep Sciences and Medicine, Palo Alto, CA, USA
| | - Roser Velasco
- Neuro-Oncology Unit, Hospital Universitari de Bellvitge-Institut Català d Oncologia L'Hospitalet, Institut d'Investigació Biomèdica de Bellvitge, l'Hospitalet de Llobregat, Barcelona, Spain; Institute of Neurosciences and Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Amanda C Guidon
- Harvard Medical School, Boston, MA, USA; Division of Neuromuscular Medicine, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Bastien Joubert
- Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, Neurological Hospital, Bron, France; MeLiS, UCBL-CNRS UMR 5284, INSERM U1314, Université Claude Bernard Lyon 1, Lyon, France; Department of Neurology, Hôpital Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Jérôme Honnorat
- Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, Neurological Hospital, Bron, France; MeLiS, UCBL-CNRS UMR 5284, INSERM U1314, Université Claude Bernard Lyon 1, Lyon, France.
| |
Collapse
|
44
|
Wuyts SCM, Cappelle CAH, Verhaert M, Bravenboer B, Aspeslagh S. Immunosuppressive therapy management in cancer patients with autoimmune diseases treated with immune checkpoint inhibitors: A case series and systematic literature review. J Oncol Pharm Pract 2024; 30:55-66. [PMID: 37051622 DOI: 10.1177/10781552231167824] [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] [Indexed: 04/14/2023]
Abstract
INTRODUCTION Prescribing immune checkpoint inhibitors (ICIs) to cancer patients with an autoimmune disease (AID) is presumed safe when cautious adverse event management is applied. However, guidelines on immunosuppressant (IS) adaptations are limited and real-world evidence is scarce. METHODS Current practice of IS adaptations is described in a case series of AID patients treated with ICIs in a tertiary university hospital in Belgium (1/1/2016-31/12/2021). Patient, drug and disease-related data were documented using retrospective chart review. A systematic search of the PubMed database was performed to identify similar cases (1/1/2010-30/11/2022). RESULTS Sixteen patients were described in the case series (62% with active AID). Systemic IS were changed before ICI initiation in 5/9 patients. Four patients continued therapy, of which one achieved partial remission. Patients who had IS (partially) stopped before ICI start (n = 4) had AID flares in two cases; immune-related adverse events in three cases. In the systematic review, 37 cases were identified in 9 articles. Corticosteroids (n = 12) and non-selective IS (n = 27) were continued in, respectively, 66% and 68% of patients. Methotrexate was frequently discontinued (13/21). Biologicals, excluding tocilizumab and vedolizumab, were withheld during ICI treatment. Out of all patients with flares (n = 15), 47% had stopped IS therapy before ICI start and 53% had continued their AID drugs. CONCLUSIONS A detailed overview of IS management in patients with AID receiving ICI therapy is presented. Expanding the knowledge base germane to IS management with ICI therapy in the diverse population is essential to evaluate their mutual impact, thus advancing responsible patient care.
Collapse
Affiliation(s)
- Stephanie C M Wuyts
- Pharmacy Department, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
- Research Group Clinical Pharmacology and Clinical Pharmacy, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Marthe Verhaert
- Department of Medical Oncology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Bert Bravenboer
- Department of Endocrinology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Sandrine Aspeslagh
- Department of Medical Oncology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| |
Collapse
|
45
|
Du Y, Liu X, Zhang L. Combining immune-checkpoint inhibitors with tocilizumab to treat a lung adenocarcinoma patient with pre-existing polymyalgia rheumatica: A case report. Thorac Cancer 2024; 15:198-200. [PMID: 38044322 PMCID: PMC10788468 DOI: 10.1111/1759-7714.15176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/14/2023] [Accepted: 11/20/2023] [Indexed: 12/05/2023] Open
Abstract
Immune-checkpoint inhibitors (ICIs) have changed the management of advanced cancers. However, patients with pre-existing autoimmune diseases (ADs) have usually been excluded from clinical trials of ICIs due to concerns about exacerbation of AD. Here, we combined ICIs with selective immunosuppressant treatment in a metastatic lung adenocarcinoma patient with active pre-existing polymyalgia rheumatica (PMR). Remarkably, the strategy led to durable response and no exacerbation of PMR. Thus, we provide the first clinical evidence of treating metastatic cancer with ICIs and concomitant use of tocilizumab and hydroxychloroquine for active pre-existing PMR.
Collapse
Affiliation(s)
- Yang Du
- Department of Respiratory and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Xiao‐Yan Liu
- Department of Respiratory and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Li Zhang
- Department of Respiratory and Critical Care MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| |
Collapse
|
46
|
Li R, Qiu J, Zhang Z, Qu C, Tang Z, Yu W, Tian Y, Tian H. Prognostic significance of Lymphocyte-activation gene 3 (LAG3) in patients with solid tumors: a systematic review, meta-analysis and pan-cancer analysis. Cancer Cell Int 2023; 23:306. [PMID: 38041068 PMCID: PMC10693146 DOI: 10.1186/s12935-023-03157-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/21/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND Lymphocyte-activation gene 3 (LAG3) is a recently discovered immune checkpoint molecule that has been linked to immunosuppression and the advancement of cancer in different types of solid tumors. This study aimed to evaluate the prognostic importance of LAG3 and its role in the immune system within solid tumors. METHODS Extensive literature searches were conducted using the Pubmed, EMBASE, and Cochrane Library databases to identify relevant studies exploring the effect of LAG3 on survival outcomes. Pooled hazard ratios (HRs) with its 95% confidence intervals (CIs) were calculated to evaluate the prognostic values of LAG3. Afterwards, subgroup analysis and sensitivity analysis were conducted. Pan-cancer analysis investigated the possible relationships between LAG3 expression and genetic alterations, RNA methylation modification-related genes, genomic instability, immune checkpoint genes, and infiltration of immune cells. RESULTS A total of 43 studies with 7,118 patients were included in this analysis. Higher expression of LAG3 was associated with worse overall survival (HR = 1.10, 95% CI 1.01-1.19, P = 0.023), but not disease-free survival (HR = 1.41, 95% CI 0.96-2.07, P = 0.078), progression-free survival (HR = 1.12, 95% CI 0.90-1.39, P = 0.317) or recurrence-free survival (HR = 0.98, 95% CI 0.81-1.19, P = 0.871). Subgroup analysis showed that LAG3 might play different prognostic roles in different solid tumors. LAG3 expression was positively associated with immune cell infiltration and immune checkpoint genes in all of the cancers included. LAG3 expression was also found to be associated with microsatellite instability (MSI), copy number variation (CNV), simple nucleoside variation (SNV), tumor mutation burden (TMB), and neoantigen in various types of cancers. CONCLUSIONS Elevated expression of LAG3 is linked to poorer prognosis among patients diagnosed with solid cancers. LAG3 might play varying prognostic roles in different types of solid tumors. Given its substantial involvement in cancer immunity and tumorigenesis, LAG3 has garnered attention as a promising prognostic biomarker and a potential target for immunotherapy.
Collapse
Affiliation(s)
- Rongyang Li
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Jianhao Qiu
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Zhan Zhang
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Chenghao Qu
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Zhanpeng Tang
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Wenhao Yu
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Yu Tian
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
| | - Hui Tian
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
47
|
Luo Y, Zeng Z, Liu Y, Liu A. Reflecting on the cardiac toxicity in non-small cell lung cancer in the era of immune checkpoint inhibitors therapy combined with thoracic radiotherapy. Biochim Biophys Acta Rev Cancer 2023; 1878:189008. [PMID: 37913939 DOI: 10.1016/j.bbcan.2023.189008] [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: 06/08/2023] [Revised: 08/31/2023] [Accepted: 10/16/2023] [Indexed: 11/03/2023]
Abstract
In recent years, immune checkpoint inhibitors (ICIs) have become a widely used treatment for non-small cell lung cancer (NSCLC), and the combination with traditional radiotherapy (RT) has shown significant potential in prolonging patient survival. However, both thoracic RT and ICIs can lead to cardiac toxicity, including radiation-induced heart damage (RIHD) and immunotherapy-related heart damage (IRHD). It still remains uncertain whether the combination of thoracic RT and immunotherapy will exacerbate acute or late cardiovascular (CV) toxicity and incidence. In this review, we summarize safety data from relevant clinical studies regarding CV toxicity for the combination therapy in NSCLC patients, explore the underlying synergetic mechanisms and common risk factors, and proposed treatment and management strategies. We hope to increase emphasis on the long-term assessment of CV toxicity risks associated with the combination therapy, and reduce the incidence of CV deaths resulting from such regimens.
Collapse
Affiliation(s)
- Yuxi Luo
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province 330006, China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province 330006, China; Radiation Induced Heart Damage Institute of Nanchang University, Nanchang, Jiangxi Province 330006, China
| | - Zhimin Zeng
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province 330006, China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province 330006, China; Radiation Induced Heart Damage Institute of Nanchang University, Nanchang, Jiangxi Province 330006, China
| | - Yunwei Liu
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province 330006, China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province 330006, China; Radiation Induced Heart Damage Institute of Nanchang University, Nanchang, Jiangxi Province 330006, China
| | - Anwen Liu
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province 330006, China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province 330006, China; Radiation Induced Heart Damage Institute of Nanchang University, Nanchang, Jiangxi Province 330006, China.
| |
Collapse
|
48
|
Ponvilawan B, Roth MT. Sequencing Systemic Therapy in Hepatocellular Carcinoma. Curr Treat Options Oncol 2023; 24:1580-1597. [PMID: 37843628 DOI: 10.1007/s11864-023-01135-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2023] [Indexed: 10/17/2023]
Abstract
OPINION STATEMENT Multiple treatment options are now approved for unresectable hepatocellular carcinoma (HCC). An immune checkpoint inhibitor (ICI)-containing regimen should be highly considered as the first-line treatment when there is no contraindication, especially in those with hepatitis virus-related HCC, due to proven superior overall survival (OS) compared to sorafenib. Atezolizumab plus bevacizumab and durvalumab plus tremelimumab remain the treatment of choice among all ICI-containing regimens, unless contraindications to either of the medications exist. Although sorafenib is still the only medication currently approved for select patients with Child-Pugh B (CP) HCC in the first-line setting, atezolizumab plus bevacizumab is being studied in this patient population. Moreover, patients with post-liver transplantation recurrence may benefit from tyrosine kinase inhibitors (TKIs), while more studies are still needed to determine the safety of ICIs in this setting. Interestingly, multiple potential biomarkers, including tumor mutational burden (TMB), microsatellite instability (MSI) status, and PD-L1 expression level, have inconsistently predicted response to ICIs in patients with HCC. Limited evidence is available to guide treatment choice in later-line settings after progressing on ICIs, and decisions should be based on the safety profile of the treatment regimen and patient preference. Multiple trials are ongoing to elucidate the optimal treatment sequence. Of note, we believe that TKIs (e.g., cabozantinib, regorafenib, lenvatinib, and sorafenib) could be more beneficial in later-line settings to broaden inhibition of other pathways apart from vascular endothelial growth factor (VEGF). When conventional treatment options are exhausted, tissue biopsy may be helpful to reveal rare targetable mutations, such as RET gene fusions.
Collapse
Affiliation(s)
- Ben Ponvilawan
- Department of Internal Medicine, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Marc T Roth
- Department of Internal Medicine, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA.
- Department of Hematology/Oncology, St. Luke's Cancer Institute, 4401 Wornall Road, Kansas City, MO, 64111, USA.
| |
Collapse
|
49
|
Liu Q, Li R, Li L, Wang G, Ji S, Zheng X, Jia X, Tao H, Hu Y. Efficacy and safety of anti-PD-1 monotherapy versus anti-PD-1 antibodies plus lenvatinib in patients with advanced hepatocellular carcinoma: a real-world experience. Ther Adv Med Oncol 2023; 15:17588359231206274. [PMID: 37885459 PMCID: PMC10599113 DOI: 10.1177/17588359231206274] [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: 08/06/2023] [Accepted: 09/21/2023] [Indexed: 10/28/2023] Open
Abstract
Background The efficacy of anti-programmed cell death (PD)-1 monotherapy in advanced hepatocellular carcinoma (aHCC) is limited, and combination therapy with lenvatinib and pembrolizumab has shown promising results. However, comparative studies between immune monotherapies and combination therapies are lacking. Objectives To investigate the efficacy and safety of anti-PD-1 monotherapy (PD-1) and anti-PD-1 plus lenvatinib (PD-1 + L) in patients with aHCC to guide clinical treatment decisions. Design A retrospective study was conducted on a cohort of patients with aHCC who received either PD-1 monotherapy or PD-1 + L combination therapy between January 2018 and January 2020. Methods The study retrospectively reviewed the medical records of 94 eligible patients with aHCC, with 39 in the PD-1 group and 55 in the PD-1 + L group. The efficacy outcomes, including objective response rate (ORR), disease control rate (DCR), duration of response (DOR), progression-free survival (PFS), overall survival (OS), and safety, were assessed. Results With a median follow-up of 30.1 months, the PD-1 + L group demonstrated a significantly higher ORR (32.7% versus 10.3%, p = 0.013), better DCR (80.0% versus 53.8%, p = 0.012), longer median PFS (10.6 versus 4.4 months, p < 0.001) and longer median OS (18.4 versus 8.5 months, p = 0.013) than PD-1 group. For the responders, the efficacy of the two groups was durable (DOR was 11.6 versus 3.5 months, p = 0.009). Subgroup analyses based on prior tyrosine kinase inhibitor (TKI) treatment and the presence or absence of macrovascular tumor thrombosis or extrahepatic metastases favored the PD-1 + L group. The combination therapy was a good predictor of PFS and OS in multivariate analysis. Grade 3/4 treatment-related adverse events were more common in PD-1 + L group, with higher incidences of hypertension and hand-foot skin reactions. Conclusions PD-1 monotherapy and PD-1 plus lenvatinib combination therapy were well-tolerated in patients with aHCC. PD-1 + L showed significantly better survival benefits than PD-1 monotherapy.
Collapse
Affiliation(s)
- Qingyan Liu
- Department of Oncology, Fifth Medical Center of the Chinese People’s Liberation Army General Hospital, Beijing, ChinaMedical School of Chinese People’s Liberation Army, Beijing, China
| | - Rong Li
- Department of Health Medicine, Second Medical Center of the Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Lingling Li
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Gaokun Wang
- Changchun Second Retired Cadre Rest Center of Jilin Provincial Military Region, Changchun, Jilin
| | - Shiyu Ji
- Department of Oncology, Fifth Medical Center of the Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Xuan Zheng
- Department of Oncology, Fifth Medical Center of the Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Xiaodong Jia
- Department of Oncology, Fifth Medical Center of the Chinese People’s Liberation Army General Hospital, 100 West Fourth Ring Road, Fengtai District, Beijing 100039, China
| | - Haitao Tao
- Department of Oncology, Fifth Medical Center of the Chinese People’s Liberation Army General Hospital, 28 Fuxing Road, Haidian District, Beijing 100000, China
| | - Yi Hu
- Department of Oncology, Fifth Medical Center of the Chinese People’s Liberation Army General Hospital, 28 Fuxing Road, Haidian District, Beijing 100000, China
| |
Collapse
|
50
|
Genta S, Lajkosz K, Yee NR, Spiliopoulou P, Heirali A, Hansen AR, Siu LL, Saibil S, Stayner LA, Yanekina M, Sauder MB, Keshavarzi S, Salawu A, Vornicova O, Butler MO, Bedard PL, Razak ARA, Rottapel R, Chruscinski A, Coburn B, Spreafico A. Autoimmune PaneLs as PrEdictors of Toxicity in Patients TReated with Immune Checkpoint InhibiTors (ALERT). J Exp Clin Cancer Res 2023; 42:276. [PMID: 37865776 PMCID: PMC10589949 DOI: 10.1186/s13046-023-02851-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/05/2023] [Indexed: 10/23/2023] Open
Abstract
BACKGROUND Immune-checkpoint inhibitors (ICI) can lead to immune-related adverse events (irAEs) in a significant proportion of patients. The mechanisms underlying irAEs development are mostly unknown and might involve multiple immune effectors, such as T cells, B cells and autoantibodies (AutoAb). METHODS We used custom autoantigen (AutoAg) microarrays to profile AutoAb related to irAEs in patients receiving ICI. Plasma was collected before and after ICI from cancer patients participating in two clinical trials (NCT03686202, NCT02644369). A one-time collection was obtained from healthy controls for comparison. Custom arrays with 162 autoAg were used to detect IgG and IgM reactivities. Differences of median fluorescent intensity (MFI) were analyzed with Wilcoxon sign rank test and Kruskal-Wallis test. MFI 500 was used as threshold to define autoAb reactivity. RESULTS A total of 114 patients and 14 healthy controls were included in this study. irAEs of grade (G) ≥ 2 occurred in 37/114 patients (32%). We observed a greater number of IgG and IgM reactivities in pre-ICI collections from patients versus healthy controls (62 vs 32 p < 0.001). Patients experiencing irAEs G ≥ 2 demonstrated pre-ICI IgG reactivity to a greater number of AutoAg than patients who did not develop irAEs (39 vs 33 p = 0.040). We observed post-treatment increase of IgM reactivities in subjects experiencing irAEs G ≥ 2 (29 vs 35, p = 0.021) and a decrease of IgG levels after steroids (38 vs 28, p = 0.009). CONCLUSIONS Overall, these results support the potential role of autoAb in irAEs etiology and evolution. A prospective study is ongoing to validate our findings (NCT04107311).
Collapse
Affiliation(s)
- Sofia Genta
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Katherine Lajkosz
- Department of Biostatistics, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Noelle R Yee
- Toronto General Research Institute, University Health Network Toronto, Toronto, ON, Canada
| | - Pavlina Spiliopoulou
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Alya Heirali
- Toronto General Research Institute, University Health Network Toronto, Toronto, ON, Canada
| | - Aaron R Hansen
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Lillian L Siu
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Sam Saibil
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Lee-Anne Stayner
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Maryia Yanekina
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Maxwell B Sauder
- Division of Dematology, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Sareh Keshavarzi
- Department of Biostatistics, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Abdulazeez Salawu
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Olga Vornicova
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Marcus O Butler
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Philippe L Bedard
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Albiruni R Abdul Razak
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Robert Rottapel
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | | | - Bryan Coburn
- Toronto General Research Institute, University Health Network Toronto, Toronto, ON, Canada
| | - Anna Spreafico
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|