1
|
Gray-Gaillard EF, Shah AA, Bingham Iii CO, Elisseeff JH, Murray J, Brahmer J, Forde P, Anagnostou V, Mammen J, Cappelli LC. Higher levels of VEGF-A and TNFα in patients with immune checkpoint inhibitor-induced inflammatory arthritis. Arthritis Res Ther 2025; 27:74. [PMID: 40170117 PMCID: PMC11959780 DOI: 10.1186/s13075-025-03546-3] [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: 11/25/2024] [Accepted: 03/23/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI), a type of cancer immunotherapy, can cause side effects including inflammatory arthritis (ICI-IA). Previous studies of ICI-IA do not include a thorough characterization of associated immune responses to provide potential targets for treatment. We aimed to identify cytokines uniquely increased in ICI-IA and determine correlations with IA severity and persistence. METHODS We evaluated patients diagnosed with ICI-IA by a rheumatologist (n = 80); control serum was obtained from ICI-treated cancer patients without any diagnosed irAEs (n = 17) or diagnosed with an unrelated irAE (n = 19). Serum was assayed to quantify 9 cytokine levels (IFN-γ, IL-4, IL-6, IL-10, IL-12p70, IL-1α, TNF-α, IL-17a, VEGF-A) using MSD U-PLEX assay. Mann-Whitney U tests were performed to evaluate differences in cytokine levels between control and ICI-IA groups. The Kruskal-Wallis test and multivariable ordinal logistic regression were used to determine difference in cytokine levels between patients of differing disease activity. RESULTS VEGF-A and TNFα were significantly elevated in patients with ICI-IA compared to ICI-controls; results persisted when restricting analyses to patients not treated with immunosuppressants at the time of sampling. ICI-IA patients were stratified by IA severity using CDAI score; there was significantly higher VEGF-A in those with higher disease activity. Ordinal logistic regression showed higher levels of IL-6 and VEGF-A were associated with higher disease activity. CONCLUSION Elevated levels of VEGF-A and TNFα are associated with ICI-IA. There was also higher IL-6 and VEGF-A among those with higher disease activity when controlling for confounding. These cytokines could be used as biomarkers of ICI-IA severity and present therapeutic targets.
Collapse
Affiliation(s)
- Elise F Gray-Gaillard
- Johns Hopkins School of Medicine, Bloomberg Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, USA
| | - Ami A Shah
- Division of Rheumatology, Johns Hopkins School of Medicine, 5501 Hopkins Bayview Circle Suite 1B1, Baltimore, MD, 21224, USA
| | - Clifton O Bingham Iii
- Division of Rheumatology, Johns Hopkins School of Medicine, 5501 Hopkins Bayview Circle Suite 1B1, Baltimore, MD, 21224, USA
| | - Jennifer H Elisseeff
- Johns Hopkins School of Medicine, Bloomberg Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, USA
| | - Joseph Murray
- Johns Hopkins School of Medicine, Bloomberg Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Julie Brahmer
- Johns Hopkins School of Medicine, Bloomberg Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Patrick Forde
- Johns Hopkins School of Medicine, Bloomberg Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Valsamo Anagnostou
- Johns Hopkins School of Medicine, Bloomberg Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jennifer Mammen
- Division of Endocrinology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Laura C Cappelli
- Division of Rheumatology, Johns Hopkins School of Medicine, 5501 Hopkins Bayview Circle Suite 1B1, Baltimore, MD, 21224, USA.
| |
Collapse
|
2
|
Matheson BE, Jaremko JL, Dowhanik A, Gill J, Gallant C, Walker J, Armani N, Leslie WD, Kolinsky M, Boyd SK, Ye C. Assessing the effects of immune checkpoint inhibitors on bone utilizing machine learning-assisted opportunistic quantitative computed tomography. J Bone Miner Res 2025; 40:396-403. [PMID: 39849845 PMCID: PMC11909731 DOI: 10.1093/jbmr/zjaf009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/10/2024] [Accepted: 01/14/2025] [Indexed: 01/25/2025]
Abstract
Immune checkpoint inhibitors (ICIs) are widely used in cancer treatment, yet their impact on bone health remains unclear. This study aimed to perform a retrospective cohort study utilizing routine CT scans from patients with melanoma to perform opportunistic QCT analysis to investigate the effects of ICI treatment on skeletal health, including volumetric BMD (vBMD) measurements and osteoarthritis (OA) parameters. A previously established machine learning-assisted opportunistic QCT pipeline was used to estimate lumbar spine vBMD from baseline and 12-mo follow-up CT scans in patients with melanoma treated with ICI therapy and those not treated with ICI therapy. Facet joint OA, osteophyte formation, and endplate sclerosis were also graded. Independent and paired t tests were used to determine any differences in vBMD and OA parameters between ICI users and non-ICI users. Multivariable linear regression models were used to control for confounding variables. Non-ICI users had a significant decrease in vBMD of -6.96 mg/cm3 from baseline to follow-up, whereas the ICI users had no significant change. There was a significant difference in change in vBMD from baseline to follow-up between the 2 groups, with the non-ICI users experiencing a 11.22 mg/cm3 larger decrease in vBMD. After adjusting for baseline age, sex, baseline vBMD, and change in OA parameters, this difference remained significant at -13.04 mg/cm3. Among the ICI users, those who had a decline in vBMD had a lower baseline vBMD compared with those who had increased vBMD. Neither group showed a significant change in OA parameters over the follow-up period, nor a difference in change between ICI and non-ICI users, even after adjusting for sex, age, and baseline OA parameters. While the effects of ICI treatment on vBMD may vary based on baseline bone health, ICIs do not significantly impact OA parameters in the short term.
Collapse
Affiliation(s)
- Bryn E Matheson
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Jacob L Jaremko
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
- Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | - Alexandra Dowhanik
- Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | - Jasmine Gill
- Division of Dermatology, Department of Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - Cassandra Gallant
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - John Walker
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - Nathan Armani
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - William D Leslie
- Departments of Internal Medicine and Radiology, University of Manitoba, Winnipeg, MB R3A 1R9, Canada
| | - Michael Kolinsky
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - Steven K Boyd
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
- Department of Radiology, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Carrie Ye
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada
| |
Collapse
|
3
|
Bruera S, Riveiro-Barciela M, Meara A, Suarez-Almazor ME. Expert Clinical Management of Inflammatory Immune-Related Arthritis in Patients with Cancer Receiving Immune Checkpoint Inhibitors. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2025; 8:64-70. [PMID: 39811420 PMCID: PMC11728383 DOI: 10.36401/jipo-24-15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/27/2024] [Accepted: 10/14/2024] [Indexed: 01/16/2025]
Abstract
Introduction Treatment guidelines for immune-related inflammatory arthritis (irAE-IA) in patients with cancer receiving immune checkpoint inhibitors (ICIs) are vague with respect to the use of specific agents. Patients are usually referred to rheumatologists for treatment. We conducted a survey of expert rheumatologists to determine current practices. We also assessed experts' views on the potential deleterious effects of various agents on tumor progression. Methods We conducted a survey of international experts in the treatment of irAE-IA, identified as members of collaborative scientific workgroups in this area. Experts were presented with a case of a patient with moderate irAE-IA and were asked about their preferred management including glucocorticoids, timing and initial choice of disease-modifying antirheumatic drugs (DMARDs), and perception of the deleterious effects of different agents on tumor progression. Results We approached 25 experts, of whom 19 (76%) responded. Most experts (63%) agreed on 20 mg or less of prednisone as initial dose. Experts selected methotrexate (41%) or tumor necrosis factor inhibitor (TNFi) (23%) as the initial DMARD if there was no improvement with corticosteroids; most experts (42%) would initiate DMARDs after 4 weeks. For patients whose initial DMARD therapy failed, the second choice was either a tumor necrosis factor inhibitor (TNFi) (38%) or interleukin-6 receptor antagonist (IL6ri) (33%). Experts were most concerned about the potential deleterious effects on tumor progression of abatacept and prednisone at doses of 20 mg or higher. Conclusion There was substantial heterogeneity in the initial management of irAE-IA. Further understanding of the pathophysiology of this immunotoxicity can assist in the classification of different presentations, selection of relevant outcomes, and planning of clinical trials to establish optimal therapeutic efficacy while minimizing potential deleterious effects of treatment on immune tumor responses.
Collapse
Affiliation(s)
- Sebastian Bruera
- Department of Immunology, Allergy & Rheumatology, Baylor College of Medicine, Houston, TX, USA
| | - Mar Riveiro-Barciela
- Department of Internal Medicine, Hospital Universitari Vall d’Hebron, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Biomedical Research Network on Hepatic and Digestive Diseases, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Alexa Meara
- Internal Medicine Department, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Maria E. Suarez-Almazor
- Department of Health Services and Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
4
|
Cappelli LC. Immune checkpoint inhibitors and rheumatoid arthritis: All roads lead to PD-1? Semin Arthritis Rheum 2025; 70S:152582. [PMID: 39578183 PMCID: PMC11761356 DOI: 10.1016/j.semarthrit.2024.152582] [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: 09/23/2024] [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
|
5
|
Ladouceur A, Jamal S, Saltman A, Himmel M, Hudson M, Pope J, Hoa S, Roberts J, Colmegna I, Arreola LG, Nevskaya T, Karmali A, Moon D, Schmidt E, Toban N, Cho L, Barnetche T, Ye C. Chronicity of Immune Checkpoint Inhibitor-Associated Inflammatory Arthritis After Immunotherapy Discontinuation: Results From the Canadian Research Group of Rheumatology in Immuno-Oncology Database. ACR Open Rheumatol 2025; 7:e70002. [PMID: 39964344 PMCID: PMC11834585 DOI: 10.1002/acr2.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/05/2025] [Accepted: 01/05/2025] [Indexed: 02/21/2025] Open
Abstract
OBJECTIVE Immune checkpoint inhibitors (ICIs) improve overall survival (OS) and progression-free survival (PFS) in many types of malignancies but can result in off-target immune-related adverse events including inflammatory arthritis (ICI-associated inflammatory arthritis [ICI-IA]), which can persist even after ICI cessation. We aimed to examine the proportion of patients with ICI-IA who develop chronic ICI-IA and describe characteristics and outcomes associated with chronic ICI-IA. METHODS We identified patients from the Canadian Research Group of Rheumatology in Immuno-Oncology retrospective cohort who developed de novo ICI-IA with at least three months of follow-up after ICI cessation. Chronic ICI-IA was defined as symptoms or ongoing immunosuppression lasting beyond three months after ICI discontinuation. Acute ICI-IA was defined as resolution of ICI-IA symptoms and discontinuation of immunosuppression within three months of ICI discontinuation. OS and PFS were assessed with Kaplan-Meier curves. Landmark multivariable Cox proportional hazard models for OS and PFS were conducted. RESULTS The study cohort included 119 patients. A total of 15 patients (13%) had acute ICI-IA, whereas 104 (87%) had chronic ICI-IA. Patients with chronic ICI-IA were more likely to be White and to have polyarthritis at presentation. After adjusting for age, sex, tumor type, stage of cancer, ICI-IA treatment, and time from ICI initiation to ICI-IA onset, patients with chronic ICI-IA had greater PFS from ICI initiation (adjusted hazard ratio 0.27, 95% confidence interval 0.08-0.98; P = 0.046). Adjusted hazard ratio for OS was similar between those with acute versus chronic ICI-IA. CONCLUSION ICI-IA frequently persists after ICI discontinuation. Chronic ICI-IA is associated with improved PFS, but not OS, as compared to acute ICI-IA.
Collapse
Affiliation(s)
| | - Shahin Jamal
- University of British ColumbiaVancouverBritish ColumbiaCanada
| | | | | | | | - Janet Pope
- University of Western OntarioLondonOntarioCanada
| | | | | | | | | | | | - Ammana Karmali
- Arthritis Research CanadaVancouverBritish ColumbiaCanada
| | - David Moon
- University of AlbertaEdmontonAlbertaCanada
| | - Emma Schmidt
- University of British ColumbiaVancouverBritish ColumbiaCanada
| | | | - Lindsay Cho
- Dalhousie UniversityHalifaxNova ScotiaCanada
| | | | - Carrie Ye
- University of AlbertaEdmontonAlbertaCanada
| |
Collapse
|
6
|
Pan S, Wang Z. Antiviral therapy can effectively suppress irAEs in HBV positive hepatocellular carcinoma treated with ICIs: validation based on multi machine learning. Front Immunol 2025; 15:1516524. [PMID: 39931579 PMCID: PMC11807960 DOI: 10.3389/fimmu.2024.1516524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 12/30/2024] [Indexed: 02/13/2025] Open
Abstract
Background Immune checkpoint inhibitors have proven efficacy against hepatitis B-virus positive hepatocellular. However, Immunotherapy-related adverse reactions are still a major challenge faced by tumor immunotherapy, so it is urgent to establish new methods to effectively predict immunotherapy-related adverse reactions. Objective Multi-machine learning model were constructed to screen the risk factors for irAEs in ICIs for the treatment of HBV-related hepatocellular and build a prediction model for the occurrence of clinical IRAEs. Methods Data from 274 hepatitis B virus positive tumor patients who received PD-1 or/and CTLA4 inhibitor treatment and had immune cell detection results were collected from Henan Cancer Hospital for retrospective analysis. Models were established using Lasso, RSF (RandomForest), and xgBoost, with ten-fold cross-validation and resampling methods used to ensure model reliability. The impact of influencing factors on irAEs (immune-related adverse events) was validated using Decision Curve Analysis (DCA). Both uni/multivariable analysis were accomplished by Chi-square/Fisher's exact tests. The accuracy of the model is verified in the DCA curve. Results A total of 274 HBV-related liver cancer patients were enrolled in the study. Predictive models were constructed using three machine learning algorithms to analyze and statistically evaluate clinical characteristics, including immune cell data. The accuracy of the Lasso regression model was 0.864, XGBoost achieved 0.903, and RandomForest reached 0.961. Resampling internal validation revealed that RandomForest had the highest recall rate (AUC = 0.892). Based on machine learning-selected indicators, antiviral therapy and The HBV DNA copy number showed a significant correlation with both the occurrence and severity of irAEs. Antiviral therapy notably reduced the incidence of IRAEs and may modulate these events through regulation of B cells. The DCA model also demonstrated strong predictive performance. Effective control of viral load through antiviral therapy significantly mitigates the occurrence of irAEs. Conclusion ICIs show therapeutic potential in the treatment of HBV-HCC. Following antiviral therapy, the incidence of severe irAEs decreases. Even in cases where viral load control is incomplete, continuous antiviral treatment can still mitigate the occurrence of irAEs.
Collapse
Affiliation(s)
| | - Zibing Wang
- Department of Immunotherapy, The Affiliated Cancer Hospital of Zhengzhou University
& Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
7
|
Deng H, Zhou J, Liu Z, Huang L, Gu Y, Chen P, Xiao H. Concomitant medication effects on patients with lung cancer taking immune checkpoint inhibitors a review. Med Oncol 2025; 42:40. [PMID: 39762456 DOI: 10.1007/s12032-024-02591-3] [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: 10/25/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025]
Abstract
In the past decade, a variety of immune checkpoint inhibitors (ICIs) are currently approved for lung cancer in the world. As a new therapeutic approach, ICIs have shown significant clinical benefits in the first-line or second-line treatment for advanced lung cancer, improving the survival and quality of life of patients. Patients need to take multiple drugs in the meantime due to their own disease or side effects during treatment. In view of drug interactions, concomitant medications have a positive or negative impact on the prognosis of lung cancer patients. In this review, we reviewed the effects of multiple drugs on the prognosis of patients with lung cancer taking ICIs. Several studies indicate that antibiotics, proton pump inhibitors (PPIs), corticosteroids, and opioid analgesics can decrease the efficacy of ICIs. Aspirin and bone-targeting drugs can enhance the efficacy of ICIs and improve the survival rate. The effects of metformin (MET), renin-angiotensin-aldosterone system inhibitors (RASI), nonsteroidal anti-inflammatory drug (NSAIDS) (except aspirin), and statins on ICIs are controversial. Future research should further explore the effects of these concomitant medications on ICIs and develop personalized prescriptions based on the specific needs of patients.
Collapse
Affiliation(s)
- Han Deng
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, Section 4, South Renmin Road, Chengdu, Sichuan, China
| | - Junxiang Zhou
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, Section 4, South Renmin Road, Chengdu, Sichuan, China
| | - Zhixi Liu
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, Section 4, South Renmin Road, Chengdu, Sichuan, China
| | - Lu Huang
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, Section 4, South Renmin Road, Chengdu, Sichuan, China
| | - Yanru Gu
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, Section 4, South Renmin Road, Chengdu, Sichuan, China
| | - Peng Chen
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, Section 4, South Renmin Road, Chengdu, Sichuan, China
| | - Hongtao Xiao
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, Section 4, South Renmin Road, Chengdu, Sichuan, China.
| |
Collapse
|
8
|
Petit PF, Daoudlarian D, Latifyan S, Bouchaab H, Mederos N, Doms J, Abdelhamid K, Ferahta N, Mencarelli L, Joo V, Bartolini R, Stravodimou A, Shabafrouz K, Pantaleo G, Peters S, Obeid M. Tocilizumab provides dual benefits in treating immune checkpoint inhibitor-associated arthritis and preventing relapse during ICI rechallenge: the TAPIR study. Ann Oncol 2025; 36:43-53. [PMID: 39241964 DOI: 10.1016/j.annonc.2024.08.2340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/04/2024] [Accepted: 08/19/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND The aim of this retrospective study was to evaluate the dual efficacy of tocilizumab (TCZ) in the treatment of immune checkpoint inhibitor (ICI)-associated arthritis (ICI-AR) and the prevention of relapses after rechallenge. PATIENTS AND METHODS We identified 26 patients with ICI-AR. The primary objectives were to evaluate TCZ efficacy in ICI-AR treatment and as secondary prophylaxis during ICI rechallenge in 11 of them. Patients received prednisone (CS) at 0.3 mg/kg tapered at 0.05 mg/kg weekly for six weeks. TCZ was administered at a dose of 8 mg/kg every 2 weeks. In the subgroup receiving secondary prophylaxis (rechallenge n = 11), TCZ was reintroduced with the same regimen concurrently with ICI rechallenge, and without the addition of CS. A control group of patients (rechallenge n = 5) was rechallenged without TCZ. Secondary endpoints included post-rechallenge evaluation of ICI duration, reintroduction of CS >0.1 mg/kg/day, ICI-AR flares, and disease control rate. RESULTS The median age of the patients was 70 years. The median follow-up from ICI initiation was 864 days. Among the 20 patients treated with TCZ for ICI-AR, all (100%) achieved an ACR70 response rate, defined as greater than 70% improvement, at 10 weeks. Some 81% of these patients achieved steroid-free remission after 24 weeks on TCZ. The median follow-up period was 552 days in rechallenged patients. The results demonstrated a reduction in ICI-AR relapses upon ICI rechallenge in patients receiving TCZ prophylaxis compared with patients who did not receive prophylaxis (17% versus 40%). The requirement for CS was completely abolished with prophylaxis (0% versus 20%), and the mean duration of ICI treatment was notably extended from 113 to 206 days. The 12-month post-rechallenge outcomes showed a disease control rate of 77%. During TCZ prophylaxis, CXCL9 remained elevated, showing no decline from their concentrations at the onset of ICI-AR. CONCLUSIONS In addition to treating ICI-AR, TCZ demonstrated efficacy as a secondary prophylactic agent, preventing the recurrence of symptoms and lengthening ICI treatment duration after ICI rechallenge.
Collapse
Affiliation(s)
- P-F Petit
- Medical Oncology Service, CHU Helora, La Louvière, Belgium
| | - D Daoudlarian
- Department of Medicine, Immunology and Allergy Service
| | - S Latifyan
- Department of Oncology, Medical Oncology Service, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - H Bouchaab
- Department of Oncology, Medical Oncology Service, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - N Mederos
- Department of Oncology, Medical Oncology Service, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - J Doms
- Department of Medicine, Immunology and Allergy Service
| | - K Abdelhamid
- Department of Oncology, Medical Oncology Service, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - N Ferahta
- Department of Oncology, Medical Oncology Service, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - L Mencarelli
- Department of Medicine, Immunology and Allergy Service
| | - V Joo
- Department of Medicine, Immunology and Allergy Service
| | - R Bartolini
- Department of Medicine, Immunology and Allergy Service
| | - A Stravodimou
- Department of Oncology, Medical Oncology Service, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - K Shabafrouz
- Department of Oncology, Medical Oncology Service, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - G Pantaleo
- Department of Medicine, Immunology and Allergy Service
| | - S Peters
- Department of Oncology, Medical Oncology Service, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - M Obeid
- Department of Medicine, Immunology and Allergy Service.
| |
Collapse
|
9
|
Ladouceur A, Barnetche T, Prey S, Dutriaux C, Gerard É, Pham-Ledard A, Beylot-Barry M, Zysman M, Veillon R, Domblides C, Daste A, Gross-Goupil M, Sionneau B, Lefort F, Mathieu L, Richez C, Truchetet ME, Schaeverbeke T, Kostine M. Resolution of immune checkpoint inhibitors-induced inflammatory arthritis while maintaining active treatment with checkpoint inhibitors and after its discontinuation: An observational study. Joint Bone Spine 2025; 92:105795. [PMID: 39447690 DOI: 10.1016/j.jbspin.2024.105795] [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: 03/20/2024] [Revised: 07/15/2024] [Accepted: 10/02/2024] [Indexed: 10/26/2024]
Abstract
INTRODUCTION Immune checkpoint inhibitors-induced inflammatory arthritis (ICI-IA) affects about 5% of ICI recipients. We aimed (1) to characterize the resolution of ICI-IA during ICI treatment and after ICI discontinuation and (2) to assess how ICI-IA influences ICI management across time. METHODS All ICI-treated patients referred to rheumatology at Bordeaux University Hospital were identified and patients with ICI-IA with a follow-up of≥6months after ICI-IA onset were included. Resolution of ICI-IA was defined by discontinuation of ICI-IA medications without recurrence of ICI-IA symptoms. RESULTS Resolution of ICI-IA occurred in 13 of 80 patients (16%) while maintaining active ICI treatment, mainly in patients with polymyalgia rheumatica (PMR)-like clinical presentation (P=0.03). Synovitis was more frequent in those whose ICI-IA persisted throughout ICI treatment. In patients with persistent ICI-IA throughout ICI treatment, 34 (50%) and 47 (70%) resolved at 6- and 12-months post-ICI discontinuation, respectively. Reason for terminating ICI was more frequently cancer stable or in remission in those who still had active ICI-IA at 6- and 12-months post-ICI discontinuation. Both progression-free survival and overall survival were longer in the groups with active ICI-IA at 6- and 12-months after ICI discontinuation. DISCUSSION In this cohort, ICI was safely continued in most patients experiencing ICI-IA. About one sixth of ICI-IA resolved despite maintaining active ICI treatment and allowing ICI-IA treatment discontinuation without recurrence of symptoms, mainly in those with PMR-like presentation. Larger studies are needed to determine predicting factors of resolving ICI-IA to minimize exposure to immunosuppressive treatment.
Collapse
Affiliation(s)
- Alexandra Ladouceur
- Department of Rheumatology, Hôpital Pellegrin, place Amélie-Raba-Léon, 33000 Bordeaux, France; Department of Medicine, McGill University, 845, rue Sherbrooke Ouest, Montréal, QC H3A 0G4, Canada.
| | - Thomas Barnetche
- Department of Rheumatology, Hôpital Pellegrin, place Amélie-Raba-Léon, 33000 Bordeaux, France
| | - Sorilla Prey
- Department of Dermatology, Hôpital Saint-André, 1, rue Jean-Burguet, 33000 Bordeaux, France
| | - Caroline Dutriaux
- Department of Dermatology, Hôpital Saint-André, 1, rue Jean-Burguet, 33000 Bordeaux, France
| | - Émilie Gerard
- Department of Dermatology, Hôpital Saint-André, 1, rue Jean-Burguet, 33000 Bordeaux, France
| | - Anne Pham-Ledard
- Department of Dermatology, Hôpital Saint-André, 1, rue Jean-Burguet, 33000 Bordeaux, France; Bordeaux Institute of Oncology, BRIC U1312 Team 5, Inserm, 2, rue du Dr-Hoffmann-Martinot, 33000 Bordeaux, France
| | - Marie Beylot-Barry
- Department of Dermatology, Hôpital Saint-André, 1, rue Jean-Burguet, 33000 Bordeaux, France
| | - Maeva Zysman
- Department of Pulmonology, Hôpital Haut-Lévêque, avenue Magellan, 33600 Pessac, France
| | - Rémi Veillon
- Department of Pulmonology, Hôpital Haut-Lévêque, avenue Magellan, 33600 Pessac, France
| | - Charlotte Domblides
- Department of Medical Oncology, Hôpital Saint-André, 1, rue Jean-Burguet, 33000 Bordeaux, France
| | - Amaury Daste
- Department of Medical Oncology, Hôpital Saint-André, 1, rue Jean-Burguet, 33000 Bordeaux, France
| | - Marine Gross-Goupil
- Department of Medical Oncology, Hôpital Saint-André, 1, rue Jean-Burguet, 33000 Bordeaux, France
| | - Baptiste Sionneau
- Department of Medical Oncology, Hôpital Saint-André, 1, rue Jean-Burguet, 33000 Bordeaux, France
| | - Felix Lefort
- Department of Medical Oncology, Hôpital Saint-André, 1, rue Jean-Burguet, 33000 Bordeaux, France
| | - Larroquette Mathieu
- Department of Medical Oncology, Hôpital Saint-André, 1, rue Jean-Burguet, 33000 Bordeaux, France
| | - Christophe Richez
- Department of Rheumatology, Hôpital Pellegrin, place Amélie-Raba-Léon, 33000 Bordeaux, France
| | - Marie-Elise Truchetet
- Department of Rheumatology, Hôpital Pellegrin, place Amélie-Raba-Léon, 33000 Bordeaux, France
| | - Thierry Schaeverbeke
- Department of Rheumatology, Hôpital Pellegrin, place Amélie-Raba-Léon, 33000 Bordeaux, France
| | - Marie Kostine
- Department of Rheumatology, Hôpital Pellegrin, place Amélie-Raba-Léon, 33000 Bordeaux, France
| |
Collapse
|
10
|
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; 16:1247-1254. [PMID: 39648646 PMCID: PMC11852740 DOI: 10.1080/1750743x.2024.2436838] [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: 08/20/2024] [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
|
11
|
Bernabela L, Bermas B. Immune Checkpoint Inhibitor Associated Rheumatoid Arthritis. Curr Rheumatol Rep 2024; 27:3. [PMID: 39589663 DOI: 10.1007/s11926-024-01173-6] [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: 09/27/2024] [Indexed: 11/27/2024]
Abstract
PURPOSE OF THIS REVIEW Immune checkpoint inhibitors (ICI) have revolutionized cancer therapy over the past decade. Unfortunately, immune related adverse events (irAEs) are common, including rheumatologic adverse events. These rheumatologic irAEs include de novo rheumatoid arthritis-like presentations or flares of pre-existing rheumatoid arthritis, collectively called ICI-associated rheumatoid arthritis. In this article we review the different mechanisms of disease activity and management approaches including use of conventional (cs) DMARDs and biologic (b) DMARDs in this patient population. Other forms of ICI-induced inflammatory arthritis e.g., PMR-like or Spondylarthritis-type IA, are beyond the scope of this review. RECENT FINDINGS The heterogeneous presentations of inflammatory arthritis in patients receiving immune checkpoint inhibitors has made this a challenging area to study. Nonetheless, recent studies are providing better understanding on the mechanisms of de novo disease and flares in patients with rheumatoid arthritis. About half of patients with pre-existing rheumatoid arthritis flare after receiving checkpoint inhibitors. Persistent arthritis is often encountered in patients receiving combination immune checkpoint inhibitors. Outcomes on overall survival do not differ in rheumatoid arthritis patients receiving checkpoint inhibitors compared to their non-arthritis counterparts. Rheumatologist play a critical role in the management of active rheumatoid arthritis induced by checkpoint inhibitors. Collaboration with oncology colleagues will continue to be a crucial component in providing quality care to these patients. While the use of glucocorticoids is often the first line therapy for active inflammatory arthritic disease, we recommend earlier consideration of DMARDs just as we inverted the treatment pyramid several decades ago, for rheumatoid arthritis.
Collapse
Affiliation(s)
- Luigino Bernabela
- Division of Rheumatic Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bonnie Bermas
- Division of Rheumatic Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
12
|
Watts T, Jennings S, Anstey S, Roche D. Supporting People Treated with Immune Checkpoint Inhibitors: A Qualitative Study Exploring Oncology Healthcare Professionals' Experiences. Semin Oncol Nurs 2024:151745. [PMID: 39482207 DOI: 10.1016/j.soncn.2024.151745] [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: 08/30/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 11/03/2024]
Abstract
OBJECTIVES Immune checkpoint inhibitors have recently developed successfully in treatment for several advanced cancers, including advanced renal cancer, where options have previously been limited. However, while some are able to tolerate these treatments, others may experience unpredictable and sometimes severe immune-related adverse events. Oncology health care professionals have vital roles in optimizing safety and supporting positive outcomes for people receiving these treatments. This study aimed to better understand these professionals' experiences of supporting people receiving immune checkpoint inhibitors. METHODS A qualitative exploratory methodology was adopted using semi-structured interviews with 18 purposively sampled senior oncology health professionals, including 12 nurses, who had experience caring for people being treated with checkpoint inhibitors. Data were collected between June and September 2020, transcribed verbatim, and analyzed using reflexive thematic analysis. RESULTS The analysis identified three main themes: First, participants were positive about the potential benefits that checkpoint inhibitors afforded patients, balanced against challenges associated with ambiguities of the treatments and potential impact on existing workloads. Secondly, participants identified the importance of proactive patient monitoring for early detection and reporting of adverse events. Participants highlighted potential challenges if these events went undetected, particularly in the context of the expectation for patient recognition and prompt reporting. Finally, participants identified the need for continual enhancement of health professionals' knowledge and understanding of immunotherapy, supported by the prioritizing of formal immunotherapy education. CONCLUSIONS Whilst immune checkpoint inhibitors offer the possibility for improved disease outcomes, this is balanced against uncertainties regarding potentially unpredictable, often complex, adverse treatment events. This study shows that nurses have vital roles in supporting people receiving these treatments. IMPLICATIONS FOR NURSING PRACTICE Effective care and treatment management for people receiving checkpoint inhibitors require nurses' support through their expert knowledge of immunotherapy and their skills for appropriate coordination and organization of cross-boundary care.
Collapse
Affiliation(s)
- Tessa Watts
- Reader, School of Healthcare Sciences, Cardiff University, Cardiff, Wales.
| | - Stephen Jennings
- Lecturer, Bristol Medical School, Bristol University, Cardiff, Wales
| | - Sally Anstey
- Reader Emirata, School of Healthcare Sciences, Cardiff University, Cardiff, Wales
| | - Dominic Roche
- Senior Lecturer, School of Healthcare Sciences, Cardiff University, Cardiff, Wales
| |
Collapse
|
13
|
Pasoto SG, Franco AS, Silva CA, Bonfa E. Sicca syndrome/Sjögren's disease associated with cancer immunotherapy: a narrative review on clinical presentation, biomarkers, and management. Expert Rev Clin Immunol 2024; 20:1149-1167. [PMID: 38903050 DOI: 10.1080/1744666x.2024.2370327] [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: 03/17/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
INTRODUCTION Almost one-quarter of immune checkpoint inhibitor (ICI) recipients experience sicca syndrome, while Sjögren's disease (SjD) is estimated at 0.3-2.5%, possibly underreported. AREAS COVERED This narrative review (Medline/Embase until January/31/2024) addresses the pathophysiology, incidence, demographic/clinical features, biomarkers, labial salivary gland biopsy (LSGB), fulfillment of the idiopathic SjD (iSjD) classificatory criteria, differential diagnosis, and management of sicca syndrome/SjD associated with ICIs. EXPERT OPINION SjD associated with ICIs is underdiagnosed, since studies that performed the mandatory SjD investigation identified that 40-60% of patients with sicca syndrome associated with ICIs meet the iSjD classificatory criteria. LSGB played a fundamental role in recognizing these cases, as most of them had negative anti-Ro/SS-A antibody. Despite the finding of focal lymphocytic sialoadenitis in LSGB samples mimicking iSjD, immunohistochemical analysis provided novel evidence of a distinct pattern for sicca syndrome/SjD associated with ICIs compared to iSjD. The former has scarcity of B lymphocytes, which are a hallmark of iSjD. Additionally, patients with sicca syndrome/SjD associated with ICIs have demographical/clinical/serological and treatment response dissimilarities compared to iSjD. Dryness symptoms are more acute in the former than in iSjD, with predominance of xerostomia over xerophthalmia, and partial/complete response to glucocorticoids. Dryness symptoms in ICI-treated patients warrant prompt SjD investigation.
Collapse
Affiliation(s)
- Sandra Gofinet Pasoto
- Rheumatology Division, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brasil
| | - André Silva Franco
- Rheumatology Division, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brasil
| | - Clovis Artur Silva
- Pediatric Rheumatology Unit, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brasil
| | - Eloisa Bonfa
- Rheumatology Division, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brasil
| |
Collapse
|
14
|
Abdel-Wahab N, Suarez-Almazor ME. Rheumatic adverse events of immune checkpoint inhibitors in cancer immunotherapy. Expert Rev Clin Immunol 2024; 20:873-893. [PMID: 38400840 PMCID: PMC11449381 DOI: 10.1080/1744666x.2024.2323966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/19/2024] [Indexed: 02/26/2024]
Abstract
INTRODUCTION The advent of immune checkpoint inhibitors (ICIs) in cancer treatment has marked a transformative era, albeit tempered by immune-related adverse events (irAEs), including those impacting the musculoskeletal system. The lack of precise epidemiologic data on rheumatic irAEs is attributed to factors such as potential underrecognition, underreporting in clinical trials, and the tendency to overlook manifestations without immediate life-threatening implications, further complicating the determination of accurate incidence rates, while the complete understanding of the mechanisms driving rheumatic irAEs remains elusive. AREAS COVERED This literature review comprehensively examines rheumatic irAEs in cancer patients undergoing ICI therapy, encompassing epidemiology, risk factors, mechanisms, clinical manifestations, and current management guidance for prevalent conditions such as inflammatory arthritis, polymyalgia rheumatica, and myositis. Less frequent rheumatic and musculoskeletal irAEs are also explored, alongside insights into ongoing clinical trials testing therapeutic and preventive strategies for irAEs. A thorough literature search on Medline and the National Cancer Institute Clinical Trials Database was conducted up to October 2023 to compile relevant information. EXPERT OPINION In light of the evolving landscape of cancer immunotherapy, there is a compelling need for prospective longitudinal studies to enhance understanding and inform clinical management strategies for rheumatic irAEs.
Collapse
Affiliation(s)
- Noha Abdel-Wahab
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine; and Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Rheumatology and Rehabilitation, Assiut University Faculty of Medicine, Assiut University Hospitals, Assiut, Egypt
| | - Maria E Suarez-Almazor
- Department of Health Services Research; and Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
15
|
Li Y, Li J, Dang Y, Chen Y, Tao C. Adverse Events of COVID-19 Vaccines in the United States: Temporal and Spatial Analysis. JMIR Public Health Surveill 2024; 10:e51007. [PMID: 39008362 PMCID: PMC11287098 DOI: 10.2196/51007] [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: 07/19/2023] [Revised: 04/07/2024] [Accepted: 05/23/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND The COVID-19 pandemic, caused by SARS-CoV-2, has had a profound impact worldwide, leading to widespread morbidity and mortality. Vaccination against COVID-19 is a critical tool in controlling the spread of the virus and reducing the severity of the disease. However, the rapid development and deployment of COVID-19 vaccines have raised concerns about potential adverse events following immunization (AEFIs). Understanding the temporal and spatial patterns of these AEFIs is crucial for an effective public health response and vaccine safety monitoring. OBJECTIVE This study aimed to analyze the temporal and spatial characteristics of AEFIs associated with COVID-19 vaccines in the United States reported to the Vaccine Adverse Event Reporting System (VAERS), thereby providing insights into the patterns and distributions of the AEFIs, the safety profile of COVID-19 vaccines, and potential risk factors associated with the AEFIs. METHODS We conducted a retrospective analysis of administration data from the Centers for Disease Control and Prevention (n=663,822,575) and reports from the surveillance system VAERS (n=900,522) between 2020 and 2022. To gain a broader understanding of postvaccination AEFIs reported, we categorized them into system organ classes (SOCs) according to the Medical Dictionary for Regulatory Activities. Additionally, we performed temporal analysis to examine the trends of AEFIs in all VAERS reports, those related to Pfizer-BioNTech and Moderna, and the top 10 AEFI trends in serious reports. We also compared the similarity of symptoms across various regions within the United States. RESULTS Our findings revealed that the most frequently reported symptoms following COVID-19 vaccination were headache (n=141,186, 15.68%), pyrexia (n=122,120, 13.56%), and fatigue (n=121,910, 13.54%). The most common symptom combination was chills and pyrexia (n=56,954, 6.32%). Initially, general disorders and administration site conditions (SOC 22) were the most prevalent class reported. Moderna exhibited a higher reporting rate of AEFIs compared to Pfizer-BioNTech. Over time, we observed a decreasing reporting rate of AEFIs associated with COVID-19 vaccines. In addition, the overall rates of AEFIs between the Pfizer-BioNTech and Moderna vaccines were comparable. In terms of spatial analysis, the middle and north regions of the United States displayed a higher reporting rate of AEFIs associated with COVID-19 vaccines, while the southeast and south-central regions showed notable similarity in symptoms reported. CONCLUSIONS This study provides valuable insights into the temporal and spatial patterns of AEFIs associated with COVID-19 vaccines in the United States. The findings underscore the critical need for increasing vaccination coverage, as well as ongoing surveillance and monitoring of AEFIs. Implementing targeted monitoring programs can facilitate the effective and efficient management of AEFIs, enhancing public confidence in future COVID-19 vaccine campaigns.
Collapse
Affiliation(s)
- Yiming Li
- McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jianfu Li
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Jacksonville, FL, United States
| | - Yifang Dang
- McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Yong Chen
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Cui Tao
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
16
|
Fletcher K, Johnson DB. Chronic immune-related adverse events arising from immune checkpoint inhibitors: an update. J Immunother Cancer 2024; 12:e008591. [PMID: 38964785 PMCID: PMC11227828 DOI: 10.1136/jitc-2023-008591] [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: 06/18/2024] [Indexed: 07/06/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have transformed cancer treatment, improving outcomes for many patients. However, toxicities termed immune-related adverse events (irAEs) are limitations of these revolutionary treatments. These irAEs may resolve with treatment or ICI cessation (acute) or persist many months beyond therapy cessation (chronic). Acute irAEs were the first to be recognized and are thus more well studied. However, chronic irAEs have been highlighted in recent years and are becoming a topic of more intensive investigation. These chronic irAEs have been noted to affect many different organ systems, including endocrine, rheumatologic, gastrointestinal, dermatologic, neurologic, and cardiovascular systems. In this review, we discuss current knowledge surrounding the frequency, time course, and risk factors associated with chronic irAEs affecting various organ systems, treatment approaches, and future directions.
Collapse
Affiliation(s)
- Kylie Fletcher
- Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Douglas B Johnson
- Department of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
17
|
Afshari AR, Sanati M, Ahmadi SS, Kesharwani P, Sahebkar A. Harnessing the capacity of phytochemicals to enhance immune checkpoint inhibitor therapy of cancers: A focus on brain malignancies. Cancer Lett 2024; 593:216955. [PMID: 38750720 DOI: 10.1016/j.canlet.2024.216955] [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: 04/05/2024] [Revised: 05/02/2024] [Accepted: 05/08/2024] [Indexed: 05/23/2024]
Abstract
Brain cancers, particularly glioblastoma multiforme (GBM), are challenging health issues with frequent unmet aspects. Today, discovering safe and effective therapeutic modalities for brain tumors is among the top research interests. Immunotherapy is an emerging area of investigation in cancer treatment. Since immune checkpoints play fundamental roles in repressing anti-cancer immunity, diverse immune checkpoint inhibitors (ICIs) have been developed, and some monoclonal antibodies have been approved clinically for particular cancers; nevertheless, there are significant concerns regarding their efficacy and safety in brain tumors. Among the various tools to modify the immune checkpoints, phytochemicals show good effectiveness and excellent safety, making them suitable candidates for developing better ICIs. Phytochemicals regulate multiple immunological checkpoint-related signaling pathways in cancer biology; however, their efficacy for clinical cancer immunotherapy remains to be established. Here, we discussed the involvement of immune checkpoints in cancer pathology and summarized recent advancements in applying phytochemicals in modulating immune checkpoints in brain tumors to highlight the state-of-the-art and give constructive prospects for future research.
Collapse
Affiliation(s)
- Amir R Afshari
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran; Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran; Experimental and Animal Study Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Seyed Sajad Ahmadi
- Department of Ophthalmology, Khatam-Ol-Anbia Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
18
|
Silverstein J, Goyal N, Tsai KK. For the Long Haul: Management of Long-Term Survivors after Melanoma Systemic Therapy. Curr Oncol Rep 2024; 26:804-817. [PMID: 38780676 DOI: 10.1007/s11912-024-01541-6] [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: 04/29/2024] [Indexed: 05/25/2024]
Abstract
PURPOSE OF REVIEW This review summarizes the latest advancements in survivorship care for patients with advanced melanoma who received systemic therapy and emphasizes the areas where more research is needed. RECENT FINDINGS Over the last decade there have been remarkable advances in the treatment of advanced and metastatic melanoma. Due to these novel treatments, including several immune checkpoint inhibitors and tyrosine kinase inhibitors, there are and will continue to be increasing numbers of long-term melanoma survivors who have been treated with systemic therapy. These patients will navigate new challenges are they are essentially among the first long term survivors after these novel therapies. Survivorship care focuses on improving the health-related quality of life of patients including the physical, emotional, social and functional effects of cancer that begin at diagnosis and continue through the end of life. Survivorship also includes screening for cancer recurrence and second cancers. As the number of melanoma survivors who received systemic therapy continues to grow, the survivorship care plan will become increasingly important for optimal care of patients even after their cancer treatments. Understanding the many domains of survivorship care for this group of patients is imperative for their care now and to identify unmet needs for future research.
Collapse
Affiliation(s)
- Jordyn Silverstein
- Department of Medicine, Division of Hematology/Oncology, University of California, Los Angeles (UCLA), 757 Westwood Plaza, Los Angeles, CA, 90095, USA.
| | - Neha Goyal
- Department of Psycho-Oncology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Katy K Tsai
- Department of Medicine, Division of Hematology/Oncology, University of California, San Francisco (UCSF), San Francisco, CA, USA
| |
Collapse
|
19
|
Takada H, Demoruelle MK, Deane KD, Nakamura S, Katsumata Y, Ikari K, Buckner JH, Robinson WH, Seifert JA, Feser ML, Moss L, Norris JM, Harigai M, Hsieh EW, Holers VM, Okamoto Y. Expansion of HLA-DR Positive Peripheral Helper T and Naive B Cells in Anticitrullinated Protein Antibody-Positive Individuals At Risk for Rheumatoid Arthritis. Arthritis Rheumatol 2024; 76:1023-1035. [PMID: 38412870 PMCID: PMC11213678 DOI: 10.1002/art.42839] [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: 07/31/2023] [Revised: 12/23/2023] [Accepted: 02/26/2024] [Indexed: 02/29/2024]
Abstract
OBJECTIVE To investigate immune dysregulation in the peripheral blood that contributes to the pre-rheumatoid arthritis (RA) stage of RA development in anticitrullinated protein antibody (ACPA)+ individuals. METHODS Using 37 markers by mass cytometry, we investigated peripheral blood mononuclear cells (PBMCs) from ACPA+ at-risk individuals, ACPA+ early untreated patients with RA, and ACPA- controls in the Tokyo Women's Medical University cohort (n = 17 in each group). Computational algorithms, FlowSOM and Optimized t-Distributed Stochastic Neighbor Embedding, were employed to explore specific immunologic differences between study groups. These findings were further evaluated, and longitudinal changes were explored, using flow cytometry and PBMCs from the US-based Targeting Immune Responses for Prevention of RA cohort that included 11 ACPA+ individuals who later developed RA (pre-RA), of which 9 had post-RA diagnosis PBMCs (post-RA), and 11 ACPA- controls. RESULTS HLA-DR+ peripheral helper T (Tph) cells, activated regulatory T cells, PD-1hi CD8+ T cells, and CXCR5-CD11c-CD38+ naive B cells were significantly expanded in PBMCs from at-risk individuals and patients with early RA from the Tokyo Women's Medical University cohort. Expansion of HLA-DR+ Tph cells and CXCR5-CD11c-CD38+ naive B cells was likewise found in both pre-RA and post-RA time points in the Targeting Immune Responses for Prevention of RA cohort. CONCLUSION The expansion of HLA-DR+ Tph cells and CXCR5-CD11c-CD38+ naive B cells in ACPA+ individuals, including those who developed inflammatory arthritis and classified RA, supports a key role of these cells in transition from pre-RA to classified RA. These findings may identify a new mechanistic target for treatment and prevention in RA.
Collapse
Affiliation(s)
- Hideto Takada
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - M. Kristen Demoruelle
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kevin D. Deane
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Shohei Nakamura
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
| | - Yasuhiro Katsumata
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
| | - Katsunori Ikari
- Department of Orthopedic Surgery, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
- Division of Multidisciplinary Management of Rheumatic Diseases, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
| | | | - William H. Robinson
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, USA
| | - Jennifer A. Seifert
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Marie L. Feser
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - LauraKay Moss
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jill M. Norris
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Masayoshi Harigai
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
| | - Elena W.Y. Hsieh
- University of Colorado School of Medicine, Department of Immunology and Microbiology Aurora, CO, USA
- University of Colorado School of Medicine, Children’s Hospital Colorado, Department of Pediatrics, Section of Allergy & Immunology, Aurora, CO, USA
| | - V. Michael Holers
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Yuko Okamoto
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
| |
Collapse
|
20
|
Gault A, Hogarth L, Williams KC, Greystoke A, Rajan N, Speight A, Lamb CA, Bridgewood A, Brown-Schofield LJ, Rayner F, Isaacs JD, Nsengimana J, Stewart CJ, Anderson AE, Plummer R, Pratt AG. Monitoring immunE DysregulAtion foLLowing Immune checkpOint-inhibitioN (MEDALLION): protocol for an observational cancer immunotherapy cohort study. BMC Cancer 2024; 24:733. [PMID: 38877461 PMCID: PMC11179333 DOI: 10.1186/s12885-024-12468-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 06/03/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND Checkpoint inhibitors (CPIs) are widely used in cancer treatment, with transformative impacts on survival. They nonetheless carry a significant risk of toxicity in the form of immune-related adverse events (IrAEs), which may be sustained and life-altering. IrAEs may require high-dose and/or prolonged steroid use and represent a significant healthcare burden. They mimic immune-mediated inflammatory diseases (IMIDs) but understanding of their pathogenesis is limited. The MEDALLION project aims to determine targetable mechanisms of immune dysregulation in IrAE development, employing an immune monitoring approach to determine changes in circulating and tissue resident cells of CPI recipients who do/do not develop them and assessing the contribution of the microbiome in parallel. METHODS MEDALLION is a non-randomised longitudinal cohort study aiming to recruit 66 cancer patient recipients of anti-PD1/PD-L1, anti-CTLA-4 or combination therapy. Eligible participants include those with malignant melanoma in the adjuvant or metastatic setting, mesothelioma and non-small cell lung carcinoma (NSCLC) treated in the metastatic setting. Comprehensive clinical evaluation is carried out alongside blood, skin swab and stool sampling at the time of CPI initiation (baseline) and during subsequent routine hospital visits on 6 occasions over a 10-month follow-up period. It is conservatively anticipated that one third of enrolled patients will experience a "significant IrAE" (SirAE), defined according to pre-determined criteria specific to the affected tissue/organ system. Those developing such toxicity may optionally undergo a biopsy of affected tissue where appropriate, otherwise being managed according to standard of care. Peripheral blood mononuclear cells will be analysed using multi-parameter flow cytometry to investigate immune subsets, their activation status and cytokine profiles. Stool samples and skin swabs will undergo DNA extraction for 16 S ribosomal RNA (rRNA) sequencing and internal transcribed spacer (ITS) gene sequencing to determine bacterial and fungal microbiome diversity, respectively, including species associated with toxicity. Stored tissue biopsies will be available for in situ and single-cell transcriptomic evaluation. Analysis will focus on the identification of biological predictors and precursors of SirAEs. DISCUSSION The pathogenesis of IrAEs will be assessed through the MEDALLION cohort, with the potential to develop tools for their prediction and/or strategies for targeted prevention or treatment. TRIAL REGISTRATION The study was registered on 18/09/2023 in the ISRCTN registry (43,419,676).
Collapse
Affiliation(s)
- Abigail Gault
- Translational and Clinical Research Institute, The Medical School, Newcastle University, William Leech Building, Framlington Place, Newcastle upon Tyne, NE4 2HH, UK
- Northern Centre for Cancer Care, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Linda Hogarth
- Northern Centre for Cancer Care, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Kristian C Williams
- Translational and Clinical Research Institute, The Medical School, Newcastle University, William Leech Building, Framlington Place, Newcastle upon Tyne, NE4 2HH, UK
| | - Alastair Greystoke
- Northern Centre for Cancer Care, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Neil Rajan
- Translational and Clinical Research Institute, The Medical School, Newcastle University, William Leech Building, Framlington Place, Newcastle upon Tyne, NE4 2HH, UK
- Department of Dermatology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Ally Speight
- Department of Gastroenterology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Christopher A Lamb
- Translational and Clinical Research Institute, The Medical School, Newcastle University, William Leech Building, Framlington Place, Newcastle upon Tyne, NE4 2HH, UK
- Department of Gastroenterology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Alison Bridgewood
- Northern Centre for Cancer Care, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Lisa-Jayne Brown-Schofield
- Northern Centre for Cancer Care, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Fiona Rayner
- Translational and Clinical Research Institute, The Medical School, Newcastle University, William Leech Building, Framlington Place, Newcastle upon Tyne, NE4 2HH, UK
- Department of Rheumatology, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - John D Isaacs
- Translational and Clinical Research Institute, The Medical School, Newcastle University, William Leech Building, Framlington Place, Newcastle upon Tyne, NE4 2HH, UK
- Department of Rheumatology, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Jérémie Nsengimana
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Christopher J Stewart
- Translational and Clinical Research Institute, The Medical School, Newcastle University, William Leech Building, Framlington Place, Newcastle upon Tyne, NE4 2HH, UK
| | - Amy E Anderson
- Translational and Clinical Research Institute, The Medical School, Newcastle University, William Leech Building, Framlington Place, Newcastle upon Tyne, NE4 2HH, UK
| | - Ruth Plummer
- Translational and Clinical Research Institute, The Medical School, Newcastle University, William Leech Building, Framlington Place, Newcastle upon Tyne, NE4 2HH, UK
- Northern Centre for Cancer Care, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Arthur G Pratt
- Translational and Clinical Research Institute, The Medical School, Newcastle University, William Leech Building, Framlington Place, Newcastle upon Tyne, NE4 2HH, UK.
- Department of Rheumatology, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.
| |
Collapse
|
21
|
Vetsika EK, Fragoulis GE, Kyriakidi M, Verrou KM, Tektonidou MG, Alissafi T, Sfikakis PP. Insufficient PD-1 expression during active autoimmune responses: a deep single-cell proteomics analysis in inflammatory arthritis. Front Immunol 2024; 15:1403680. [PMID: 38911848 PMCID: PMC11190177 DOI: 10.3389/fimmu.2024.1403680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/27/2024] [Indexed: 06/25/2024] Open
Abstract
Objectives Programmed cell death protein-1 (PD-1) maintains peripheral immune tolerance by preventing T cell continuous activation. Aiming to understand the extent of PD-1 expression in inflammatory arthritis beyond its involvement with T cells, we assess its presence on various circulating single cells. Methods Mass cytometry analysis of patients with active seropositive/seronegative rheumatoid (RA; n=9/8) and psoriatic (PsA; n=9) arthritis versus healthy controls (HC; n=13), re-evaluating patients after 3 months of anti-rheumatic treatment. Results PD-1 was expressed in all leukocyte subpopulations, with the highest PD-1+ cell frequencies in eosinophils (59-73%) and T cells (50-60%), and the lowest in natural-killer cells (1-3%). PD-1+ cell frequencies and PD-1 median expression were comparable between patient subgroups and HC, in the majority of cell subsets. Exceptions included increases in certain T cell/B cell subsets of seropositive RA and specific monocyte subsets and dendritic cells of PsA; an expanded PD-1+CD4+CD45RA+CD27+CD28+ T subset, denoting exhausted T cells, was common across patient subgroups. Strikingly, significant inverse correlations between individual biomarkers of systemic inflammation (ESR and/or serum CRP) and PD-1+ cell frequencies and/or median expression were evident in several innate and adaptive immunity cell subsets of RA and PsA patients. Furthermore, all inverse correlations noted in individuals with active arthritis were no longer discernible in those who attained remission/low disease activity post-treatment. Conclusion PD-1 expression may be insufficient, relative to the magnitude of the concomitant systemic inflammatory response on distinct leukocyte subsets, varying between RA and PsA. Our results point to the potential therapeutic benefits of pharmacological PD-1 activation, to rebalance the autoimmune response and reduce inflammation.
Collapse
Affiliation(s)
- Eleni-Kyriaki Vetsika
- Centre of New Biotechnologies and Precision Medicine (CNBPM), School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - George E. Fragoulis
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Kyriakidi
- Centre of New Biotechnologies and Precision Medicine (CNBPM), School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Kleio-Maria Verrou
- Centre of New Biotechnologies and Precision Medicine (CNBPM), School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria G. Tektonidou
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Themis Alissafi
- Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Laboratory of Immune Regulation, Center of Basic Sciences, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Petros P. Sfikakis
- Centre of New Biotechnologies and Precision Medicine (CNBPM), School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
22
|
Ferguson L, Hooper S. Lidocaine Infusion: An Analgesic Option for Checkpoint Inhibitor Arthritis: A Case Report. J Pain Palliat Care Pharmacother 2024; 38:153-156. [PMID: 38718037 DOI: 10.1080/15360288.2024.2345334] [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: 02/21/2024] [Accepted: 04/13/2024] [Indexed: 06/06/2024]
Abstract
This report describes the use of subcutaneous lidocaine infusion to manage complex pain associated with checkpoint inhibitor inflammatory arthritis. In addition, the safe administration of lidocaine in the home setting is described. A 49-year-old man with metastatic melanoma to lung, right axilla and posterior chest wall on regular pembrolizumab developed checkpoint inhibitor inflammatory arthritis. Pain associated with this was unresponsive to simple analgesia, escalating opioids and adjuvant analgesics. Lidocaine infusion was used on separate occasions (inpatient unit and home setting) to gain rapid and sustained control of inflammatory pain. Inflammatory pain responded well to 2 mg/kg/h lidocaine infusion over 4 days with sustained response between infusions of up to 6 wk. Resulting in improved mobility, functional status, and overall quality of life. Lidocaine infusion should be considered as an option for analgesic management of checkpoint inhibitor inflammatory arthritis in patients for whom usual treatment is ineffective, and as an opioid-sparing intervention.
Collapse
Affiliation(s)
- Lana Ferguson
- Dr. Lana Ferguson, BHB, MBChB, DipPallMed (RA CP), FRA CP, FA ChPM, Hospice Waikato, Hamilton, New Zealand
| | - Stacey Hooper
- Dr. Stacey Hooper, MBChB, DipPallMed (RACP), Waikato District Health Board, Hamilton, New Zealand
| |
Collapse
|
23
|
Kachi S, Sumitomo S, Oka H, Hata A, Ohmura K. Case report: Inflammatory sternoclavicular joint arthritis induced by an immune checkpoint inhibitor with remarkable responsiveness to infliximab. Front Immunol 2024; 15:1400097. [PMID: 38799449 PMCID: PMC11116605 DOI: 10.3389/fimmu.2024.1400097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024] Open
Abstract
This report describes the case of a 48-year-old woman who presented with sternoclavicular joint arthritis after administration of an immune checkpoint inhibitor (ICI), durvalumab, for small cell lung carcinoma. The onset of arthritis transpired 18 months after the commencement of the ICI therapeutic regimen and demonstrated resilience to glucocorticoid treatment. After excluding infectious aetiologies and metastatic involvement, the patient was diagnosed with ICI-induced arthritis (ICI-IA). Considering the articular implications akin to the SAPHO syndrome, the patient was treated with infliximab, resulting in complete resolution. This finding implies that biological DMARDs can serve as effective interventions for ICI-induced sternoclavicular joint arthritis. Given the heterogeneous nature of its pathogenesis, the selection of therapeutic agents may require customization based on the distinct clinical presentation of each individual case.
Collapse
Affiliation(s)
- Shion Kachi
- Department of Rheumatology, Kobe City Medical Center General Hospital, Kobe, Hyogo, Japan
| | - Shuji Sumitomo
- Department of Rheumatology, Kobe City Medical Center General Hospital, Kobe, Hyogo, Japan
| | - Hideki Oka
- Department of Rheumatology, Kobe City Medical Center General Hospital, Kobe, Hyogo, Japan
| | - Akito Hata
- Department of Thoracic Oncology, Kobe Minimally Invasive Cancer Center, Kobe, Hyogo, Japan
| | - Koichiro Ohmura
- Department of Rheumatology, Kobe City Medical Center General Hospital, Kobe, Hyogo, Japan
| |
Collapse
|
24
|
Cappelli LC, Kamal O, Jones M, Bingham CO, Shah AA. Baseline predictors of disease severity in immune checkpoint inhibitor-induced inflammatory arthritis. Rheumatology (Oxford) 2024; 63:1518-1522. [PMID: 37647635 PMCID: PMC11147537 DOI: 10.1093/rheumatology/kead438] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/24/2023] [Accepted: 08/08/2023] [Indexed: 09/01/2023] Open
Abstract
OBJECTIVES The objective of this study was to determine baseline risk factors for requiring immunosuppression and having persistent arthritis in patients with immune checkpoint inhibitor-induced inflammatory arthritis (ICI-inflammatory arthritis). METHODS Participants were adults with rheumatologist diagnosed ICI-inflammatory arthritis. The primary outcome was requirement of conventional synthetic (cs) or biologic (b) DMARDs; other outcomes were persistence of inflammatory arthritis >6 months after ICI cessation and requirement of CSs. Logistic regression models evaluated associations between clinical features and primary and secondary outcomes, with adjustment for potential confounders, as appropriate. RESULTS One hundred and twenty-six patients with ICI-inflammatory arthritis were included; 53 patients (42%) required a csDMARD/bDMARD. In the univariate logistic regression analysis, higher clinical disease activity index (CDAI), tenosynovitis, longer symptom duration before first rheumatology visit and longer ICI duration were significantly associated with a higher likelihood of requiring DMARDs; in addition, there was a trend towards those treated with prior chemotherapy being less likely to need DMARDs. After adjustment, tenosynovitis, longer symptom duration and higher CDAI remained associated with requiring DMARDs, while those with prior chemotherapy were significantly less likely to require DMARDs. Combination anti-CTLA-4 (cytotoxic T-lymphocyte-associated protein-4)/PD-1 (Programmed cell death protein-1) therapy and CS use at baseline were associated with a higher risk of persistent inflammatory arthritis. CONCLUSION Higher levels of disease activity, tenosynovitis and longer symptom duration prior to rheumatology referral were associated with requiring DMARDs for ICI-inflammatory arthritis, while those treated previously with chemotherapy were less likely to require additional immunosuppression. The presence of risk factors for severe disease at baseline may indicate a role for higher initial CS dose, earlier rheumatology referral, and adoption of immunosuppression beyond CSs to improve outcomes.
Collapse
Affiliation(s)
- Laura C Cappelli
- Division of Rheumatology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Omer Kamal
- Department of Medicine, St. Agnes Hospital, Baltimore, MD, USA
| | - Michelle Jones
- Division of Rheumatology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Clifton O Bingham
- Division of Rheumatology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ami A Shah
- Division of Rheumatology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
25
|
Ruf T, Kramer R, Forschner A, Leiter U, Meier F, Reinhardt L, Dücker P, Ertl C, Tomsitz D, Tietze JK, Gutzmer R, Dabrowski E, Zimmer L, Gesierich A, Zierold S, French LE, Eigentler T, Amaral T, Heinzerling L. Second-line therapies for steroid-refractory immune-related adverse events in patients treated with immune checkpoint inhibitors. Eur J Cancer 2024; 203:114028. [PMID: 38652976 DOI: 10.1016/j.ejca.2024.114028] [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: 01/10/2024] [Revised: 03/03/2024] [Accepted: 03/12/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) induce adverse events (irAEs) that do not respond to steroids, i.e. steroid-refractory (sr) irAEs, and irAEs in which steroids cannot be tapered, i.e. steroid-dependent (sd) irAEs, in about 10% of cases. An evidence-based analysis of the effectiveness of second-line immunosuppressive agents with regard to irAE and tumor control is lacking. METHODS The international web-based Side Effect Registry Immuno-Oncology (SERIO; http://serio-registry.org) is a collaborative initiative with the Paul-Ehrlich-Institute to document rare, severe, complex or therapy-refractory immunotherapy-induced side effects. The registry was queried on August 1, 2023 for cases of irAEs which were treated with second-line therapies. RESULTS From a total of 1330 cases, 217 patients (16.3%) received 249 second-line therapies. A total of 19 different second-line therapies were employed, including TNF-alpha antagonists (46.5%), intravenous immunoglobulins (IVIG; 19.1%), mycophenolate mofetil (15.9%), and methotrexate (3.6%). Therapy choices were determined by the type of irAE. The time to onset of sr-/sd-irAEs after ICI initiation did not consistently differ from steroid-responsive irAEs. While 74.3% of sr-/sd-irAEs resolved and 13.1% had improved, 4.3% persisted, 3.9% resulted in permanent sequelae, and 4.3% in death with ongoing symptoms. Infliximab exhibited potential for earlier symptom improvement compared to mycophenolate mofetil or IVIG. Tumor response in patients with second-line treated sd-/sr-irAE was similar to patients with irAEs treated with steroids only. CONCLUSION Several second-line therapies are effective against sr-/sd-irAEs, the second-line therapies show no clear negative impact on tumor response, and infliximab shows potential for faster improvement of symptoms. However, prospective comparative data are needed.
Collapse
Affiliation(s)
- Theresa Ruf
- Department of Dermatology and Allergology, University Hospital, LMU Munich, Munich, Germany; SERIO registry
| | - Rafaela Kramer
- Department of Dermatology, University Clinic Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany; SERIO registry
| | - Andrea Forschner
- Department of Dermatology, Eberhard-Karls-University of Tübingen, Tübingen, Germany
| | - Ulrike Leiter
- Department of Dermatology, Eberhard-Karls-University of Tübingen, Tübingen, Germany
| | - Friedegund Meier
- Department of Dermatology, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Lydia Reinhardt
- Department of Dermatology, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Pia Dücker
- Department of Dermatology, Hospital Dortmund, Dortmund, Germany
| | - Carolin Ertl
- Department of Dermatology and Allergology, University Hospital, LMU Munich, Munich, Germany; SERIO registry
| | - Dirk Tomsitz
- Department of Dermatology and Allergology, University Hospital, LMU Munich, Munich, Germany
| | - Julia K Tietze
- Clinic for Dermatology and Venerology, University Medical Center Rostock, Rostock, Germany
| | - Ralf Gutzmer
- Department of Dermatology, Johannes Wesling Medical Center, Ruhr-University Bochum, Minden, Germany
| | | | - Lisa Zimmer
- Department of Dermatology, Essen University Hospital, West German Cancer Center, University of Duisburg-Essen and the German Cancer Consortium (DKTK), Partner site Essen/Düsseldorf, Germany
| | - Anja Gesierich
- Department of Dermatology, University Hospital Würzburg, Germany
| | - Sarah Zierold
- Department of Dermatology and Allergology, University Hospital, LMU Munich, Munich, Germany; SERIO registry
| | - Lars E French
- Department of Dermatology and Allergology, University Hospital, LMU Munich, Munich, Germany; Dr. Philip Frost Department of Dermatology & Cutaneous Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Thomas Eigentler
- Department of Dermatology, Charité University Medicine Berlin, Berlin, Germany
| | - Teresa Amaral
- Department of Dermatology, Eberhard-Karls-University of Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC 2180), Tübingen, Germany
| | - Lucie Heinzerling
- Department of Dermatology and Allergology, University Hospital, LMU Munich, Munich, Germany; Department of Dermatology, University Clinic Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany; SERIO registry.
| |
Collapse
|
26
|
Singh N, Shahane A, Sparks JA, Bitoun S, Cappelli LC. Immune Checkpoint Inhibitor-induced Inflammatory Arthritis: Current Approaches to Management. Rheum Dis Clin North Am 2024; 50:269-279. [PMID: 38670725 PMCID: PMC11139458 DOI: 10.1016/j.rdc.2024.02.002] [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] [Indexed: 04/28/2024]
Abstract
The introduction of immune checkpoint inhibitors (ICIs) has changed the landscape of the treatment of cancer. Several immune-related adverse events (irAEs) have now been described such as ICI-inflammatory arthritis (IA), sicca syndrome, polymyalgia rheumatica, myositis, and vasculitis as a consequence of immune activation. The onset of the ICI-IA can vary from after the first infusion of ICIs to a delayed presentation a year or more after ICI initiation. Ultimately, baseline patient and tumor characteristics, the types of immunotherapies used, pre-existing autoimmune diseases, and/or other irAEs, as well as patient preferences will all shape the discussions around ICI-IA management.
Collapse
Affiliation(s)
- Namrata Singh
- Division of Rheumatology, Department of Medicine, University of Washington, 4245 Roosevelt Way Northeast, Seattle, WA 98105, USA
| | - Anupama Shahane
- Division of Rheumatology, Department of Medicine, University of Pennsylvania, 220 South 40th Street, Philadelphia, PA 19104, USA
| | - Jeffrey A Sparks
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, 12 Braddock Park, Unit 2, Boston, MA 02116, USA
| | - Samuel Bitoun
- Department of Rheumatology, Université Paris-Saclay, Hôpital Bicêtre, Assistance Publique - Hôpitaux de Paris, FHU CARE, INSERM UMR1184, 78, Avenue du General Leclerc, Le Kremlin Bicêtre 94270, France
| | - Laura C Cappelli
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Suite 1B1, Baltimore, MD 21224, USA.
| |
Collapse
|
27
|
Zhang J, Ni R, Oke I, Calabrese C, Strouse J, Weinmann S, Ladouceur A. Imaging in Rheumatic Immune-related Adverse Events. Rheum Dis Clin North Am 2024; 50:313-323. [PMID: 38670729 DOI: 10.1016/j.rdc.2024.02.005] [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
Since their introduction, immune checkpoint inhibitors have revolutionized cancer treatment by harnessing the body's own immune system as a defense against tumor growth. The downside of activating the immune system is the development of immune-related adverse events (irAEs), which mimic autoimmune disease of various organ systems. The musculoskeletal system is an uncommon, but substantial one for patients and can lead to long-term pain and disability that affects their quality of life. This review summarizes recent literature on imaging forms utilized for diagnosis and assessing treatment response in rheumatic irAEs.
Collapse
Affiliation(s)
- Jeremy Zhang
- Department of Internal Medicine, University of Iowa Carver College of Medicine, 200 Hawkins Drive, C310 GH, Iowa City, IA 52242-1009, USA
| | - Ruoning Ni
- Division of Immunology, University of Iowa Carver College of Medicine, 200 Hawkins Drive, C42 GH, Iowa City, IA 52242-1009, USA
| | - Ibiyemi Oke
- Division of Immunology, University of Iowa Carver College of Medicine, 200 Hawkins Drive, C42 GH, Iowa City, IA 52242-1009, USA
| | - Cassandra Calabrese
- Department of Rheumatologic and Immunologic Disease, Cleveland Clinic, 9500 Euclid Avenue, Mail Code A50, Cleveland, OH 44195, USA
| | - Jennifer Strouse
- Division of Immunology, University of Iowa Carver College of Medicine, 200 Hawkins Drive, C42 GH, Iowa City, IA 52242-1009, USA
| | - Sophia Weinmann
- Division of Rheumatology & Immunology, Duke University School of Medicine, 40 Duke Medicine Circle, Clinic 1J, Durham, NC 27710, USA
| | - Alexandra Ladouceur
- Division of Rheumatology, Department of Medicine, Jewish General Hospital and McGill University, 3755 Côte Ste-Catherine Road, Room A725, Montreal, Quebec H3T 1E2, Canada.
| |
Collapse
|
28
|
Shah NJ, Bottini N. Inhibiting the Inhibitor in Synovial Macrophages and Cancer Immunotherapy-Associated Inflammatory Arthritis. Arthritis Rheumatol 2024; 76:505-506. [PMID: 37909274 PMCID: PMC10965387 DOI: 10.1002/art.42743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 10/25/2023] [Indexed: 11/03/2023]
Affiliation(s)
- Nisarg J. Shah
- University of California, San Diego, La Jolla, CA 92093, USA
| | | |
Collapse
|
29
|
Tran SD, Lin J, Galvez C, Rasmussen LV, Pacheco J, Perottino GM, Rahbari KJ, Miller CD, John JD, Theros J, Vogel K, Dinh PV, Malik S, Ramzan U, Tegtmeyer K, Mohindra N, Johnson JL, Luo Y, Kho A, Sosman J, Walunas TL. Rapid identification of inflammatory arthritis and associated adverse events following immune checkpoint therapy: a machine learning approach. Front Immunol 2024; 15:1331959. [PMID: 38558818 PMCID: PMC10978703 DOI: 10.3389/fimmu.2024.1331959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction Immune checkpoint inhibitor-induced inflammatory arthritis (ICI-IA) poses a major clinical challenge to ICI therapy for cancer, with 13% of cases halting ICI therapy and ICI-IA being difficult to identify for timely referral to a rheumatologist. The objective of this study was to rapidly identify ICI-IA patients in clinical data and assess associated immune-related adverse events (irAEs) and risk factors. Methods We conducted a retrospective study of the electronic health records (EHRs) of 89 patients who developed ICI-IA out of 2451 cancer patients who received ICI therapy at Northwestern University between March 2011 to January 2021. Logistic regression and random forest machine learning models were trained on all EHR diagnoses, labs, medications, and procedures to identify ICI-IA patients and EHR codes indicating ICI-IA. Multivariate logistic regression was then used to test associations between ICI-IA and cancer type, ICI regimen, and comorbid irAEs. Results Logistic regression and random forest models identified ICI-IA patients with accuracies of 0.79 and 0.80, respectively. Key EHR features from the random forest model included ICI-IA relevant features (joint pain, steroid prescription, rheumatoid factor tests) and features suggesting comorbid irAEs (thyroid function tests, pruritus, triamcinolone prescription). Compared to 871 adjudicated ICI patients who did not develop arthritis, ICI-IA patients had higher odds of developing cutaneous (odds ratio [OR]=2.66; 95% Confidence Interval [CI] 1.63-4.35), endocrine (OR=2.09; 95% CI 1.15-3.80), or gastrointestinal (OR=2.88; 95% CI 1.76-4.72) irAEs adjusting for demographics, cancer type, and ICI regimen. Melanoma (OR=1.99; 95% CI 1.08-3.65) and renal cell carcinoma (OR=2.03; 95% CI 1.06-3.84) patients were more likely to develop ICI-IA compared to lung cancer patients. Patients on nivolumab+ipilimumab were more likely to develop ICI-IA compared to patients on pembrolizumab (OR=1.86; 95% CI 1.01-3.43). Discussion Our machine learning models rapidly identified patients with ICI-IA in EHR data and elucidated clinical features indicative of comorbid irAEs. Patients with ICI-IA were significantly more likely to also develop cutaneous, endocrine, and gastrointestinal irAEs during their clinical course compared to ICI therapy patients without ICI-IA.
Collapse
Affiliation(s)
- Steven D. Tran
- Center for Health Information Partnerships, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jean Lin
- Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Carlos Galvez
- Hematology and Oncology, University of Illinois Health, Chicago, IL, United States
| | - Luke V. Rasmussen
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jennifer Pacheco
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | | | - Kian J. Rahbari
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Charles D. Miller
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jordan D. John
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jonathan Theros
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Kelly Vogel
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Patrick V. Dinh
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Sara Malik
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Umar Ramzan
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Kyle Tegtmeyer
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Nisha Mohindra
- Department of Medicine, Division of Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, United States
| | - Jodi L. Johnson
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, United States
- Departments of Pathology and Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Yuan Luo
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Abel Kho
- Center for Health Information Partnerships, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Medicine, Division of General Internal Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jeffrey Sosman
- Department of Medicine, Division of Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, United States
| | - Theresa L. Walunas
- Center for Health Information Partnerships, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Medicine, Division of General Internal Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
30
|
Zhou Z, Zhou X, Jiang X, Yang B, Lu X, Fei Y, Zhao L, Chen H, Zhang L, Si X, Liang N, Wang Y, Yang D, Peng Y, Yang Y, Yao Z, He Y, Wu X, Zhang W, Wang M, Yang H, Zhang X. Single-cell profiling identifies IL1B hi macrophages associated with inflammation in PD-1 inhibitor-induced inflammatory arthritis. Nat Commun 2024; 15:2107. [PMID: 38453911 PMCID: PMC10920757 DOI: 10.1038/s41467-024-46195-x] [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: 07/08/2023] [Accepted: 02/14/2024] [Indexed: 03/09/2024] Open
Abstract
Inflammatory arthritis (IA) is a common rheumatic adverse event following immune checkpoint inhibitors treatment. The clinical disparities between IA and rheumatoid arthritis (RA) imply disease heterogeneity and distinct mechanisms, which remain elusive. Here, we profile CD45+ cells from the peripheral blood or synovial fluid (SF) of patients with PD-1-induced IA (PD-1-IA) or RA using single-cell RNA sequencing. We report the predominant expansion of IL1Bhi myeloid cells with enhanced NLRP3 inflammasome activity, in both the SF and peripheral blood of PD-1-IA, but not RA. IL1Bhi macrophages in the SF of PD-1-IA shared similar inflammatory signatures and might originate from peripheral IL1Bhi monocytes. Exhausted CD8+ T cells (Texs) significantly accumulated in the SF of patients with PD-1-IA. IL1Bhi myeloid cells communicated with CD8+ Texs possibly via the CCR1-CCL5/CCL3 and CXCL10-CXCR3 axes. Collectively, these results demonstrate different cellular and molecular pathways in PD-1-IA and RA and highlight IL1Bhi macrophages as a possible therapeutic target in PD-1-IA.
Collapse
Affiliation(s)
- Ziyue Zhou
- Department of Rheumatology and Clinical Immunology, National Clinical Research Center for Dermatologic and Immunologic Diseases, the Ministry of Education Key Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Xiaoxiang Zhou
- 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, 100021, Beijing, China
| | - Xu Jiang
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- National Infrastructure for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Bo Yang
- Department of Orthopedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Xin Lu
- Department of Orthopedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Yunyun Fei
- Department of Rheumatology and Clinical Immunology, National Clinical Research Center for Dermatologic and Immunologic Diseases, the Ministry of Education Key Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Lidan Zhao
- Department of Rheumatology and Clinical Immunology, National Clinical Research Center for Dermatologic and Immunologic Diseases, the Ministry of Education Key Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Hua Chen
- Department of Rheumatology and Clinical Immunology, National Clinical Research Center for Dermatologic and Immunologic Diseases, the Ministry of Education Key Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Li Zhang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Xiaoyan Si
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Naixin Liang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Yadong Wang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Dan Yang
- Department of Rheumatology and Clinical Immunology, National Clinical Research Center for Dermatologic and Immunologic Diseases, the Ministry of Education Key Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Yezi Peng
- Department of Rheumatology and Clinical Immunology, National Clinical Research Center for Dermatologic and Immunologic Diseases, the Ministry of Education Key Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Yiying Yang
- Department of Rheumatology and Clinical Immunology, National Clinical Research Center for Dermatologic and Immunologic Diseases, the Ministry of Education Key Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Zhuoran Yao
- Department of Thoracic Oncology, Cancer Center, and Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Yangzhige He
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- National Infrastructure for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Xunyao Wu
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Wen Zhang
- Department of Rheumatology and Clinical Immunology, National Clinical Research Center for Dermatologic and Immunologic Diseases, the Ministry of Education Key Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Min Wang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Huaxia Yang
- Department of Rheumatology and Clinical Immunology, National Clinical Research Center for Dermatologic and Immunologic Diseases, the Ministry of Education Key Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China.
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China.
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences, 100730, Beijing, China.
| |
Collapse
|
31
|
Springer R, Lange K, Homey B, Meller S, Lindhof HH. Steroid-dependent polyarthritis induced by immune checkpoint inhibitor therapy successfully treated with bimekizumab. Immunotherapy 2024; 16:287-293. [PMID: 38264842 DOI: 10.2217/imt-2023-0252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 12/14/2023] [Indexed: 01/25/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) are an integral part of modern-day cancer therapy. Along with a greatly improved antitumor response come a number of immune-related adverse events (irAEs), musculoskeletal irAEs rank among the less frequent manifestations. The mechanisms behind these events are poorly understood, and so far clear guidelines for therapeutic management beyond treatment with glucocorticosteroids are lacking. We present the case of a 72-year-old patient who developed a severe ICI-induced polyarthritis that could not be controlled by glucocorticosteroids. We initiated an immunomodulating therapy with the IL-17A/F/AF-inhibitor bimekizumab, which lead to a full clinical and sonographic remission.
Collapse
Affiliation(s)
- Robin Springer
- Department of Dermatology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Kristin Lange
- Department of Dermatology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Bernhard Homey
- Department of Dermatology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Stephan Meller
- Department of Dermatology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Harm-Henning Lindhof
- Department of Dermatology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Moorenstr. 5, 40225, Duesseldorf, Germany
| |
Collapse
|
32
|
Daetwyler E, Wallrabenstein T, König D, Cappelli LC, Naidoo J, Zippelius A, Läubli H. Corticosteroid-resistant immune-related adverse events: a systematic review. J Immunother Cancer 2024; 12:e007409. [PMID: 38233099 PMCID: PMC10806650 DOI: 10.1136/jitc-2023-007409] [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: 10/28/2023] [Indexed: 01/19/2024] Open
Abstract
Immune checkpoint inhibitor (ICI) treatment has become an important therapeutic option for various cancer types. Although the treatment is effective, ICI can overstimulate the patient's immune system, leading to potentially severe immune-related adverse events (irAEs), including hepatitis, colitis, pneumonitis and myocarditis. The initial mainstay of treatments includes the administration of corticosteroids. There is little evidence how to treat steroid-resistant (sr) irAEs. It is mainly based on small case series or single case reports. This systematic review summarizes available evidence about sr-irAEs. We conducted a systematic literature search in PubMed. Additionally, we included European Society for Medical Oncology, Society for Immunotherapy of Cancer, National Comprehensive Cancer Network and American Society of Clinical Oncology Guidelines for irAEs in our assessment. The study population of all selected publications had to include patients with cancer who developed hepatitis, colitis, pneumonitis or myocarditis during or after an immunotherapy treatment and for whom corticosteroid therapy was not sufficient. Our literature search was not restricted to any specific cancer diagnosis. Case reports were also included. There is limited data regarding life-threatening sr-irAEs of colon/liver/lung/heart and the majority of publications are single case reports. Most publications investigated sr colitis (n=26), followed by hepatitis (n=21), pneumonitis (n=17) and myocarditis (n=15). There is most data for mycophenolate mofetil (MMF) to treat sr hepatitis and for infliximab, followed by vedolizumab, to treat sr colitis. Regarding sr pneumonitis there is most data for MMF and intravenous immunoglobulins (IVIG) while data regarding infliximab are conflicting. In sr myocarditis, most evidence is available for the use of abatacept or anti-thymocyte globulin (ATG) (both with or without MMF) or ruxolitinib with abatacept. This review highlights the need for prompt recognition and treatment of sr hepatitis, colitis, pneumonitis and myocarditis. Guideline recommendations for sr situations are not defined precisely. Based on our search, we recommend-as first line treatment-(1) MMF for sr hepatitis, (2) infliximab for sr colitis, followed by vedolizumab, (3) MMF and IVIG for sr pneumonitis and (4) abatacept or ATG (both with or without MMF) or ruxolitinib with abatacept for sr myocarditis. These additional immunosuppressive agents should be initiated promptly if there is no sufficient response to corticosteroids within 3 days.
Collapse
Affiliation(s)
- Eveline Daetwyler
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
| | - Till Wallrabenstein
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Division of Hematology and Medical Oncology, University Medical Center Freiburg, Freiburg, Germany
| | - David König
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
| | - Laura C Cappelli
- Divison of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Alfred Zippelius
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Heinz Läubli
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
33
|
Rose N, Furer V, Polachek A, Elkayam O, Gertel S. Immune Checkpoint Inhibitor-Induced Inflammatory Arthritis: Overview of Therapies and a Personalized Approach to Optimized Combined Therapy. Eur J Rheumatol 2024; 11. [PMID: 38477323 PMCID: PMC11365019 DOI: 10.5152/eurjrheum.2024.23091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 10/04/2023] [Indexed: 03/14/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs), including anti-cytotoxic T lymphocyte antigen 4, anti-programmed cell death 1, and anti-programmed cell death ligand 1 (PD-L1) antibodies, are currently widely used in oncology clinical practice, achieving considerable success in improving disease outcomes. New checkpoint targets are being discovered and investigated through basic science research and clinical trials. ICI remove negative regulatory immune signals on T cells, leading to immune activation and induction of antitumor immunity. Patients who receive ICI, however, are at risk for developing immune-related adverse events (irAEs), which are attributed to increased T cell activity against antigens in both tumors and in healthy tissues, to increased inflammatory cytokine levels, to increased levels of preexisting autoantibodies, and to enhanced complement-mediated inflammation. Arthritis is one of the most common irAEs. ICI-induced rheumatic irAEs are categorized by levels of severity which guide the choice of treatment options. Management of ICI-induced rheumatic irAEs includes the use of glucocorticoids, disease-modifying antirheumatic drugs (mainly methotrexate), and biological agents (e.g., tumor necrosis factor, interleukin-6 receptor, and CD20 inhibitors). This review aims to summarize the current ICI subtypes, their role in rheumatic irAEs development, and therapies currently used in clinical practice to manage irAEs. In addition, we propose to use an ex vivo personalized diagnostic assay for the selection of the most effective ICI with antirheumatic drugs combinations that will inhibit the advancement of ICI-induced adverse events.
Collapse
Affiliation(s)
- Noa Rose
- Department of Rheumatology, Tel Aviv Sourasky Medical Center affiliated to the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Victoria Furer
- Department of Rheumatology, Tel Aviv Sourasky Medical Center affiliated to the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ari Polachek
- Department of Rheumatology, Tel Aviv Sourasky Medical Center affiliated to the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ori Elkayam
- Department of Rheumatology, Tel Aviv Sourasky Medical Center affiliated to the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Smadar Gertel
- Department of Rheumatology, Tel Aviv Sourasky Medical Center affiliated to the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
34
|
Ladouceur A, Barnetche T, Mouterde G, Tison A, Bitoun S, Prey S, Dutriaux C, Gerard E, Pham-Ledard A, Beylot-Barry M, Zysman M, Veillon R, Domblides C, Daste A, Gross-Goupil M, Sionneau B, Lefort F, Larroquette M, Richez C, Truchetet ME, Schaeverbeke T, Kostine M. Immune checkpoint inhibitor rechallenge in patients who previously experienced immune-related inflammatory arthritis: a multicentre observational study. RMD Open 2023; 9:e003795. [PMID: 38030233 PMCID: PMC10689372 DOI: 10.1136/rmdopen-2023-003795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 11/02/2023] [Indexed: 12/01/2023] Open
Abstract
OBJECTIVE Another course of immune checkpoint inhibitors (ICIs) is often considered in patients with cancer progression and previous immune-related adverse events, including inflammatory arthritis (ICI-IA), but there are limited data regarding safety of ICI rechallenge in this setting. We aimed to assess the rate and clinical features associated with ICI-IA flare/recurrence on ICI rechallenge. METHODS We conducted a multicentre observational study including cancer patients with ICI-IA who started a second course of ICI more than 3 months after ICI discontinuation in four French university hospitals. Primary outcome was the frequency of ICI flare/recurrence after ICI rechallenge. RESULTS Twenty-three patients were included. At the time of ICI rechallenge, 18 patients reported no symptoms of ICI-IA (78%) and 5 had grade 1 (22%), 11 patients (48%) were not receiving any ICI-IA treatment, 11 (48%) were still on prednisone, 2 (9%) were on conventional synthetic disease-modifying antirheumatic drugs and 1 (4%) on anti-IL-6. ICI-IA flare/recurrence occurred in 12 patients (52%) with a median time of 1 month after ICI rechallenge. ICI-IA phenotype, disease activity and ICI-IA treatment at the time of ICI rechallenge did not differ according to ICI-IA flare/recurrence status. CONCLUSION In this first observational study of ICI-IA patients rechallenged with ICI, about half of the patients experienced ICI-IA flare/recurrence with a similar phenotype but occurring earlier than the initial ICI-IA, warranting close monitoring during the first month of retreatment. Risk of flare did not differ according to baseline immunosuppressive treatment at the time of rechallenge.
Collapse
Affiliation(s)
- Alexandra Ladouceur
- Rheumatology, Centre Hospitalier Universitaire de Bordeaux Groupe hospitalier Pellegrin, Bordeaux, France
- Rheumatology, McGill University Faculty of Medicine, Montreal, Quebec, Canada
| | - Thomas Barnetche
- Rheumatology, Centre Hospitalier Universitaire de Bordeaux Groupe hospitalier Pellegrin, Bordeaux, France
| | - Gael Mouterde
- Rheumatology, Hôpital Lapeyronie, Montpellier, France
| | | | - Samuel Bitoun
- Rheumatology, Université Paris Saclay, Hôpital Bicetre, Assistance Publique-Hôpitaux de Paris, FHU CARE, Le Kremlin-Bicetre, France
| | - Sorilla Prey
- Dermatology, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
- Pediatric Dermatology Unit, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Caroline Dutriaux
- Dermatology, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Emilie Gerard
- Dermatology, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Anne Pham-Ledard
- Dermatology, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Marie Beylot-Barry
- Dermatology, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Maeva Zysman
- Pulmonology, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Rémi Veillon
- Pulmonology, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | | | - Amaury Daste
- Oncology, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | | | - Baptiste Sionneau
- Oncology, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Felix Lefort
- Oncology, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | | | - Christophe Richez
- Rheumatology, Centre Hospitalier Universitaire de Bordeaux Groupe hospitalier Pellegrin, Bordeaux, France
- UMR CNRS 5164, Université de Bordeaux Collège Sciences de la Santé, Bordeaux, France
| | - Marie-Elise Truchetet
- Rheumatology, Centre Hospitalier Universitaire de Bordeaux Groupe hospitalier Pellegrin, Bordeaux, France
| | - Thierry Schaeverbeke
- Rheumatology, Centre Hospitalier Universitaire de Bordeaux Groupe hospitalier Pellegrin, Bordeaux, France
| | - Marie Kostine
- Rheumatology, Centre Hospitalier Universitaire de Bordeaux Groupe hospitalier Pellegrin, Bordeaux, France
| |
Collapse
|
35
|
Small A, Lowe K, Wechalekar MD. Immune checkpoints in rheumatoid arthritis: progress and promise. Front Immunol 2023; 14:1285554. [PMID: 38077329 PMCID: PMC10704353 DOI: 10.3389/fimmu.2023.1285554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/07/2023] [Indexed: 12/18/2023] Open
Abstract
Rheumatoid arthritis (RA) is one of the most prevalent autoimmune inflammatory conditions, and while the mechanisms driving pathogenesis are yet to be completely elucidated, self-reactive T cells and immune checkpoint pathways have a clear role. In this review, we provide an overview of the importance of checkpoint pathways in the T cell response and describe the involvement of these in RA development and progression. We discuss the relationship between immune checkpoint therapy in cancer and autoimmune adverse events, draw parallels with the involvement of immune checkpoints in RA pathobiology, summarise emerging research into some of the lesser-known pathways, and the potential of targeting checkpoint-related pathways in future treatment approaches to RA management.
Collapse
Affiliation(s)
- Annabelle Small
- Department of Rheumatology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Katie Lowe
- Department of Rheumatology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Mihir D Wechalekar
- Department of Rheumatology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Department of Rheumatology, Flinders Medical Centre, Adelaide, SA, Australia
| |
Collapse
|
36
|
Paroli M, Sirinian MI. When Autoantibodies Are Missing: The Challenge of Seronegative Rheumatoid Arthritis. Antibodies (Basel) 2023; 12:69. [PMID: 37987247 PMCID: PMC10660552 DOI: 10.3390/antib12040069] [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: 09/01/2023] [Revised: 09/25/2023] [Accepted: 10/23/2023] [Indexed: 11/22/2023] Open
Abstract
Seronegative rheumatoid arthritis (SNRA) is characterized by the absence of both rheumatoid factor (RF) and antibodies against the cyclic citrullinated protein (ACPA) in serum. However, the differences between the two forms of RA are more complex and have not yet been definitively characterized. Several lines of evidences support the idea that there are specific elements of the two forms, including genetic background, epidemiology, pathogenesis, severity of progression over time, and response to therapy. Clinical features that may differentiate SNRA from SPRA are also suggested by data obtained from classical radiology and newer imaging techniques. Although new evidence seems to provide additional help in differentiating the two forms of RA, their distinguishing features remain largely elusive. It should also be emphasized that the distinctive features of RA forms, if not properly recognized, can lead to the underdiagnosis of SNRA, potentially missing the period called the "window of opportunity" that is critical for early diagnosis, timely treatment, and better prognosis. This review aims to summarize the data provided in the scientific literature with the goal of helping clinicians diagnose SNRA as accurately as possible, with emphasis on the most recent findings available.
Collapse
Affiliation(s)
- Marino Paroli
- Center for Allergy and Immunology, Department of Clinical, Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome c/o Polo Pontino, 04100 Latina, Italy
| | | |
Collapse
|
37
|
Parvathareddy V, Selamet U, Sen AA, Mamlouk O, Song J, Page VD, Abdelrahim M, Diab A, Abdel-Wahab N, Abudayyeh A. Infliximab for Treatment of Immune Adverse Events and Its Impact on Tumor Response. Cancers (Basel) 2023; 15:5181. [PMID: 37958355 PMCID: PMC10649345 DOI: 10.3390/cancers15215181] [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: 09/17/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Background: Immune-related adverse events (irAEs) challenge the use of immune checkpoint inhibitors (ICIs). We performed a retrospective study to evaluate response to infliximab for immune-related adverse event management, and infliximab's effect on progression-free survival (PFS) and overall survival (OS) with a focus on melanoma and genitourinary cancers. Methods: We retrospectively reviewed records of all cancer patients exposed to infliximab after immune checkpoint inhibitor (ICI) treatment from 2004 to 2021 at the MD Anderson Cancer Center. Survival was assessed utilizing the Kaplan-Meier method. Univariate and multivariate logistic regression was utilized to evaluate predictors of infliximab response, OS, and PFS. Results: We identified 185 cancer patients (93 melanoma and 37 genitourinary cancers) treated with ICI and who received infliximab to treat irAEs. Within 3 months of treatment initiation, 71% of the patients responded to infliximab, 27% had no response, and 2% had unknown response. Among different irAEs, colitis was associated with increased response to infliximab at 3 months, irrespective of the type of malignancy. We evaluated best tumor response before and after infliximab in the entire cohort and again in the melanoma and genitourinary (GU); the findings were similar in the melanoma cohort and the entire cohort, where best tumor response before and after infliximab was not significantly different. In the melanoma cohort, acute kidney injury (AKI) was associated with increased risk of death, p = 0.0109, and having response to infliximab was associated with decreased risk of death, p = 0.0383. Interestingly in GU cancer patients, myositis was associated with increased risk of death, p = 0.0041, and having a response to infliximab was marginally associated with decreased risk of death, p = 0.0992. As regards PFS, in a multivariate Cox regression model, having a history of cardiovascular disease remained significantly associated with shorter PFS in the melanoma cohort. For patients with GU cancers, response to infliximab was associated with longer PFS. Conclusions: Our study is among the largest retrospective analyses of infliximab use for irAE management. Patients with colitis were the best responders to infliximab. AKI before initiation of infliximab in the melanoma subcohort and myositis in GU subcohort are associated with higher risk of death. Our results indicate no association between infliximab and cancer progression with the exception of genitourinary cancers.
Collapse
Affiliation(s)
| | - Umut Selamet
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA;
| | - Aditi A. Sen
- Department of Nephrology, Baylor College of Medicine, Houston, TX 77030, USA; (V.P.); (A.A.S.)
| | - Omar Mamlouk
- Section of Nephrology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Unit 1468, 1400 Pressler Street, Houston, TX 77030, USA; (O.M.); (V.D.P.)
| | - Juhee Song
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Valda D. Page
- Section of Nephrology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Unit 1468, 1400 Pressler Street, Houston, TX 77030, USA; (O.M.); (V.D.P.)
| | - Maen Abdelrahim
- Institute of Academic Medicine and Weill Cornell Medical College, Houston Methodist Cancer Center, Houston, TX 77479, USA;
| | - Adi Diab
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.D.); (N.A.-W.)
| | - Noha Abdel-Wahab
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.D.); (N.A.-W.)
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Rheumatology and Rehabilitation Department, Assiut University Hospitals, Faculty of Medicine, Assiut 71515, Egypt
| | - Ala Abudayyeh
- Section of Nephrology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Unit 1468, 1400 Pressler Street, Houston, TX 77030, USA; (O.M.); (V.D.P.)
| |
Collapse
|
38
|
Gente K, Diekmann L, Daniello L, Will J, Feisst M, Olsavszky V, Günther J, Lorenz HM, Souto-Carneiro MM, Hassel JC, Christopoulos P, Leipe J. Sex and anti-inflammatory treatment affect outcome of melanoma and non-small cell lung cancer patients with rheumatic immune-related adverse events. J Immunother Cancer 2023; 11:e007557. [PMID: 37730272 PMCID: PMC10510926 DOI: 10.1136/jitc-2023-007557] [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: 08/21/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND Rheumatic immune-related adverse events (R-irAEs) occur in 5-15% of patients receiving immune checkpoint inhibitors (ICI) and, unlike other irAEs, tend to be chronic. Herein, we investigate the factors influencing cancer and R-irAEs outcomes with particular focus on adverse effects of anti-inflammatory treatment. METHODS In this prospective, multicenter, long-term, observational study, R-irAEs were comprehensively analyzed in patients with malignant melanoma (MM, n=50) and non-small cell lung cancer (NSCLC, n=41) receiving ICI therapy who were enrolled in the study between August 1, 2018, and December 11, 2022. RESULTS After a median follow-up of 33 months, progressive disease or death occurred in 66.0% and 30.0% of MM and 63.4% and 39.0% of patients with NSCLC. Male sex (progression-free survival (PFS): p=0.013, and overall survival (OS): p=0.009), flare of a pre-existing condition (vs de novo R-irAE, PFS: p=0.010) and in trend maximum glucocorticoid (GC) doses >10 mg and particularly ≥1 mg/kg prednisolone equivalent (sex-adjusted PFS: p=0.056, OS: p=0.051) were associated with worse cancer outcomes. Patients receiving disease-modifying antirheumatic drugs (DMARDs) showed significantly longer PFS (n=14, p=0.011) and OS (n=20, p=0.018). Effects of these variables on PFS and/or OS persisted in adjusted Cox regression models. Additionally, GC treatment negatively correlated with the time from diagnosis of malignancy and the latency from ICI start until R-irAE onset (all p<0.05). R-irAE features and outcomes were independent of other baseline patient characteristics in both studied cancer entities. CONCLUSION Male sex, flare of pre-existing rheumatologic conditions and extensive GC treatment appeared to be linked with unfavorable cancer outcomes, while DMARD use had a favorable impact. These findings challenge the current dogma of restrictive DMARD use for R-irAE and thus may pave the way to better strategies and randomized controlled trials for the growing number of patients with R-irAE.
Collapse
Affiliation(s)
- Karolina Gente
- Department of Internal Medicine V - Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Leonore Diekmann
- Department of Internal Medicine V - Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Lea Daniello
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor Diseases (NCT) at University Hospital Heidelberg, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC-H), Member of German Center for Lung Research, Heidelberg, Germany
| | - Julia Will
- Department of Internal Medicine V - Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Manuel Feisst
- Institute of Medical Biometry (IMBI), University Hospital Heidelberg, Heidelberg, Germany
| | - Victor Olsavszky
- Department of Dermatology, University Medical Centre Mannheim, Mannheim, Germany
| | - Janine Günther
- Department of Internal Medicine V - Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Hanns-Martin Lorenz
- Department of Internal Medicine V - Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - M Margarida Souto-Carneiro
- Department of Internal Medicine V - Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Jessica C Hassel
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor Diseases (NCT) at University Hospital Heidelberg, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC-H), Member of German Center for Lung Research, Heidelberg, Germany
| | - Jan Leipe
- Department of Medicine V - Division of Rheumatology, University Medical Centre Mannheim, Mannheim, Germany
| |
Collapse
|
39
|
Reynolds G. Rheumatic complications of checkpoint inhibitors: Lessons from autoimmunity. Immunol Rev 2023; 318:51-60. [PMID: 37435963 PMCID: PMC10952967 DOI: 10.1111/imr.13242] [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: 05/13/2023] [Accepted: 06/26/2023] [Indexed: 07/13/2023]
Abstract
Immune checkpoint inhibitors are now an established treatment in the management of a range of cancers. Their success means that their use is likely to increase in future in terms of the numbers of patients treated, the indications and the range of immune checkpoints targeted. They function by counteracting immune evasion by the tumor but, as a consequence, can breach self-tolerance at other sites leading to a range of immune-related adverse events. Included among these complications are a range of rheumatologic complications, including inflammatory arthritis and keratoconjunctivitis sicca. These superficially resemble immune-mediated rheumatic diseases (IMRDs) such as rheumatoid arthritis and Sjogren's disease but preliminary studies suggest they are clinically and immunologically distinct entities. However, there appear to be common processes that predispose to the development of both that may inform preventative interventions and predictive tools. Both groups of conditions highlight the centrality of immune checkpoints in controlling tolerance and how it can be restored. Here we will discuss some of these commonalities and differences between rheumatic irAEs and IMRDs.
Collapse
Affiliation(s)
- Gary Reynolds
- Institute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUK
- Center for Immunology and Inflammatory DiseasesMassachusetts General HospitalBostonMassachusettsUSA
| |
Collapse
|
40
|
Wang R, Singaraju A, Marks KE, Shakib L, Dunlap G, Adejoorin I, Greisen SR, Chen L, Tirpack AK, Aude C, Fein MR, Todd DJ, MacFarlane L, Goodman SM, DiCarlo EF, Massarotti EM, Sparks JA, Jonsson AH, Brenner MB, Postow MA, Chan KK, Bass AR, Donlin LT, Rao DA. Clonally expanded CD38 hi cytotoxic CD8 T cells define the T cell infiltrate in checkpoint inhibitor-associated arthritis. Sci Immunol 2023; 8:eadd1591. [PMID: 37506196 PMCID: PMC10557056 DOI: 10.1126/sciimmunol.add1591] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/05/2023] [Indexed: 07/30/2023]
Abstract
Immune checkpoint inhibitor (ICI) therapies used to treat cancer, such as anti-PD-1 antibodies, can induce autoimmune conditions in some individuals. The T cell mechanisms mediating such iatrogenic autoimmunity and their overlap with spontaneous autoimmune diseases remain unclear. Here, we compared T cells from the joints of 20 patients with an inflammatory arthritis induced by ICI therapy (ICI-arthritis) with two archetypal autoimmune arthritides, rheumatoid arthritis (RA) and psoriatic arthritis (PsA). Single-cell transcriptomic and antigen receptor repertoire analyses highlighted clonal expansion of an activated effector CD8 T cell population in the joints and blood of patients with ICI-arthritis. These cells were identified as CD38hiCD127- CD8 T cells and were uniquely enriched in ICI-arthritis joints compared with RA and PsA and also displayed an elevated interferon signature. In vitro, type I interferon induced CD8 T cells to acquire the ICI-associated CD38hi phenotype and enhanced cytotoxic function. In a cohort of patients with advanced melanoma, ICI therapy markedly expanded circulating CD38hiCD127- T cells, which were frequently bound by the therapeutic anti-PD-1 drug. In patients with ICI-arthritis, drug-bound CD8 T cells in circulation showed marked clonal overlap with drug-bound CD8 T cells from synovial fluid. These results suggest that ICI therapy directly targets CD8 T cells in patients who develop ICI-arthritis and induces an autoimmune pathology that is distinct from prototypical spontaneous autoimmune arthritides.
Collapse
Affiliation(s)
- Runci Wang
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Shanghai Institute of Rheumatology / Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Anvita Singaraju
- HSS Research Institute, Hospital for Special Surgery, New York, NY 10021, USA
- Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| | - Kathryne E. Marks
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Lorien Shakib
- HSS Research Institute, Hospital for Special Surgery, New York, NY 10021, USA
- Graduate Program in Physiology, Biophysics and Systems Biology, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| | - Garrett Dunlap
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA
| | - Ifeoluwakiisi Adejoorin
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Stinne R. Greisen
- Department of Biomedicine, Aarhus University / Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
| | - Lin Chen
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Aidan K. Tirpack
- Medical School at Campbell University School of Osteopathic Medicine, Lillington, NC 27546, USA
| | - Carlos Aude
- Division of Rheumatology, Hospital for Special Surgery / Weill Cornell Medicine, New York, NY 10021, USA
| | - Miriam R. Fein
- HSS Research Institute, Hospital for Special Surgery, New York, NY 10021, USA
| | - Derrick J. Todd
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Lindsey MacFarlane
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Susan M. Goodman
- Division of Rheumatology, Hospital for Special Surgery / Weill Cornell Medicine, New York, NY 10021, USA
| | - Edward F. DiCarlo
- Division of Pathology, Hospital for Special Surgery / Weill Cornell Medicine, New York, NY 10021, USA
| | - Elena M. Massarotti
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jeffrey A. Sparks
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - A. Helena Jonsson
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Michael B. Brenner
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Michael A. Postow
- Melanoma Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Karmela K. Chan
- Division of Rheumatology, Hospital for Special Surgery / Weill Cornell Medicine, New York, NY 10021, USA
| | - Anne R. Bass
- Division of Rheumatology, Hospital for Special Surgery / Weill Cornell Medicine, New York, NY 10021, USA
| | - Laura T. Donlin
- HSS Research Institute, Hospital for Special Surgery, New York, NY 10021, USA
- Graduate Program in Physiology, Biophysics and Systems Biology, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| | - Deepak A. Rao
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
41
|
Curkovic NB, Johnson DB. Updates in toxicities associated with immune checkpoint inhibitors. Expert Rev Clin Immunol 2023; 19:1117-1129. [PMID: 37276071 PMCID: PMC10527235 DOI: 10.1080/1744666x.2023.2221434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/31/2023] [Indexed: 06/07/2023]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) have become a pillar of treatment for numerous cancers with increasing use in combination with other ICIs and in earlier stages of disease treatment. Although effective, ICI use is accompanied by a milieu of potentially bothersome or even life-threatening toxicities known as immune-related adverse events (irAEs), necessitating careful monitoring and early intervention. AREAS COVERED In this review, we provide an overview of recent advances surrounding toxicity pathophysiology and treatment in the context of relevant organ systems. An emphasis on current treatments by toxicity, as well as updates on steroid-refractory toxicities, chronic toxicities, and biomarkers will be a focus of this update on the current understanding of irAEs. EXPERT OPINION ICI toxicities are a major limitation on the deployment of multi-agent ICI regimens and are thus a major priority to understand, treat, and prevent. Recent developments have led to greater understanding of the pathophysiology of these events, which may lead to improved prevention or mitigation strategies. Further, early studies have also suggested steroid-sparing approaches that may be useful. Ultimately, preventing and managing irAEs will be a key goal toward successful ICI treatment across a broader range of patients with cancer.
Collapse
Affiliation(s)
| | - Douglas B. Johnson
- Department of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
42
|
Bass AR, Abdel-Wahab N, Reid PD, Sparks JA, Calabrese C, Jannat-Khah DP, Ghosh N, Rajesh D, Aude CA, Gedmintas L, MacFarlane L, Arabelovic S, Falohun A, Mushtaq K, Haj FA, Diab A, Shah AA, Bingham CO, Chan KK, Cappelli LC. Comparative safety and effectiveness of TNF inhibitors, IL6 inhibitors and methotrexate for the treatment of immune checkpoint inhibitor-associated arthritis. Ann Rheum Dis 2023; 82:920-926. [PMID: 37019614 PMCID: PMC10330686 DOI: 10.1136/ard-2023-223885] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/24/2023] [Indexed: 04/07/2023]
Abstract
OBJECTIVES To compare the safety and effectiveness of biologic and conventional disease-modifying antirheumatic drugs (DMARDs) for immune checkpoint inhibitor-associated inflammatory arthritis (ICI-IA). METHODS The retrospective multicentre observational study included patients with a diagnosis of ICI-IA treated with a tumour necrosis factor inhibitor (TNFi), interleukin-6 receptor inhibitor (IL6Ri) and/or methotrexate (MTX); patients with pre-existing autoimmune disease were excluded. The primary outcome was time to cancer progression from ICI initiation; the secondary outcome was time to arthritis control from DMARD initiation. Cox proportional hazard models were used to compare medication groups, adjusting for confounders. RESULTS 147 patients were included (mean age 60.3 (SD 11.9) years, 66 (45%) women). ICI-IA treatment was TNFi in 33 (22%), IL6Ri 42 (29%) and MTX 72 (49%). After adjustment for time from ICI initiation to DMARD initiation, time to cancer progression was significantly shorter for TNFi compared with MTX (HR 3.27 (95% CI 1.21 to 8.84, p=0.019)) while the result for IL6Ri was HR 2.37 (95% CI 0.94 to 5.98, p=0.055). Time to arthritis control was faster for TNFi compared with MTX (HR 1.91 (95% CI 1.06 to 3.45, p=0.032)) while the result for IL6Ri was HR 1.66 (95% CI 0.93 to 2.97, p=0.089). A subset analysis in patients with melanoma gave similar results for both cancer progression and arthritis control. CONCLUSION The treatment of ICI-IA with a biologic DMARD is associated with more rapid arthritis control than with MTX, but may be associated with a shorter time to cancer progression.
Collapse
Affiliation(s)
- Anne R Bass
- Rheumatology, Hospital for Special Surgery, New York, New York, USA
- Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Noha Abdel-Wahab
- Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Pankti D Reid
- Rheumatology, University of Chicago Medical Center, Chicago, Illinois, USA
| | - Jeffrey A Sparks
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | | | - Deanna P Jannat-Khah
- Jannat Khah: Medicine; Aude: Rheumatology, Hospital for Special Surgery, New York, New York, USA
- Epidemiology in Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Nilasha Ghosh
- Rheumatology, Hospital for Special Surgery, New York, New York, USA
- Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Divya Rajesh
- Harvard Medical School, Boston, Massachusetts, USA
| | - Carlos Andres Aude
- Jannat Khah: Medicine; Aude: Rheumatology, Hospital for Special Surgery, New York, New York, USA
| | - Lydia Gedmintas
- Rheumatology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | | | - Senada Arabelovic
- Rheumatology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Adewunmi Falohun
- Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Farah Al Haj
- Hematology and Medical Oncology, Tulane University Health Sciences Center, New Orleans, Louisiana, USA
| | - Adi Diab
- Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ami A Shah
- Medicine/Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Clifton O Bingham
- Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Karmela Kim Chan
- Rheumatology, Hospital for Special Surgery, New York, New York, USA
- Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Laura C Cappelli
- Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
43
|
Ghosh N, Reid P, Aude CA, Kirschman J, Goodman S, Bykerk VP, Lakhanpal A, Rajesh D, Chan KK, Robinson WH, Bass AR. Anticitrullinated peptide antibody epitope expansion and the HLA DRB1 'shared epitope' are less common in seropositive checkpoint inhibitor-induced inflammatory arthritis than in longstanding rheumatoid arthritis. RMD Open 2023; 9:e003012. [PMID: 37355249 PMCID: PMC10314674 DOI: 10.1136/rmdopen-2023-003012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 05/22/2023] [Indexed: 06/26/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) can potentially cause ICI-inflammatory arthritis (ICI-IA), which often resembles rheumatoid arthritis (RA). In this study, we examined the degree of anticitrullinated peptide antibodies (ACPA) epitope expansion in CCP+ICI-IA and patients with RA. METHODS We used clinical data and serum from ICI-IA and patients with RA with early disease as well as longstanding disease. A custom, bead-based antigen array was used to identify IgG ACPA reactivities to 18 putative RA-associated citrullinated proteins. Hierarchical clustering software was used to create a heatmap to identify ACPA levels. Additionally, HLA DRB1 typing was performed on ICI-IA patients as well as controls of patients treated with ICI that did not develop ICI-IA (ICI controls). RESULTS Compared to patients with CCP+RA, patients with CCP+ICI-IA were older (p<0.001), less likely to have positive rheumatoid factor (p<0.001) and had a shorter duration of symptoms (p<0.001). There were less ACPA levels and a lower number of distinct ACPA epitopes in the serum of patients with ICI-IA compared with longstanding patients with RA (p<0.001). Among those tested for HLA DRB1, there were no differences in the frequency of the shared epitope between those with ICI-IA and ICI controls. CONCLUSION Patients with ICI-IA had lower ACPA titres and targeted fewer ACPA epitopes than longstanding patients with RA, and there were no significant differences in the presence of the shared epitope between those that developed ICI-IA and ICI controls. It remains to be determined if ICI-IA represents an accelerated model of RA pathogenesis with ICI triggering a transition from preclinical to clinical disease.
Collapse
Affiliation(s)
- Nilasha Ghosh
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery, New York, New York, USA
| | - Pankti Reid
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, Illinois, USA
| | - Carlos Andres Aude
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery, New York, New York, USA
| | - Jessica Kirschman
- Department of Medicine, Stanford University, Stanford, California, USA
| | - Susan Goodman
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery, New York, New York, USA
| | - Vivian P Bykerk
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery, New York, New York, USA
| | - Amit Lakhanpal
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery, New York, New York, USA
| | - Diviya Rajesh
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery, New York, New York, USA
| | - Karmela K Chan
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery, New York, New York, USA
| | | | - Anne R Bass
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery, New York, New York, USA
| |
Collapse
|
44
|
Fa'ak F, Buni M, Falohun A, Lu H, Song J, Johnson DH, Zobniw CM, Trinh VA, Awiwi MO, Tahon NH, Elsayes KM, Ludford K, Montazari EJ, Chernis J, Dimitrova M, Sandigursky S, Sparks JA, Abu-Shawer O, Rahma O, Thanarajasingam U, Zeman AM, Talukder R, Singh N, Chung SH, Grivas P, Daher M, Abudayyeh A, Osman I, Weber J, Tayar JH, Suarez-Almazor ME, Abdel-Wahab N, Diab A. Selective immune suppression using interleukin-6 receptor inhibitors for management of immune-related adverse events. J Immunother Cancer 2023; 11:e006814. [PMID: 37328287 PMCID: PMC10277540 DOI: 10.1136/jitc-2023-006814] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2023] [Indexed: 06/18/2023] Open
Abstract
BACKGROUND Management of immune-related adverse events (irAEs) is important as they cause treatment interruption or discontinuation, more often seen with combination immune checkpoint inhibitor (ICI) therapy. Here, we retrospectively evaluated the safety and effectiveness of anti-interleukin-6 receptor (anti-IL-6R) as therapy for irAEs. METHODS We performed a retrospective multicenter study evaluating patients diagnosed with de novo irAEs or flare of pre-existing autoimmune disease following ICI and were treated with anti-IL-6R. Our objectives were to assess the improvement of irAEs as well as the overall tumor response rate (ORR) before and after anti-IL-6R treatment. RESULTS We identified a total of 92 patients who received therapeutic anti-IL-6R antibodies (tocilizumab or sarilumab). Median age was 61 years, 63% were men, 69% received anti-programmed cell death protein-1 (PD-1) antibodies alone, and 26% patients were treated with the combination of anti-cytotoxic T lymphocyte antigen-4 and anti-PD-1 antibodies. Cancer types were primarily melanoma (46%), genitourinary cancer (35%), and lung cancer (8%). Indications for using anti-IL-6R antibodies included inflammatory arthritis (73%), hepatitis/cholangitis (7%), myositis/myocarditis/myasthenia gravis (5%), polymyalgia rheumatica (4%), and one patient each with autoimmune scleroderma, nephritis, colitis, pneumonitis and central nervous system vasculitis. Notably, 88% of patients had received corticosteroids, and 36% received other disease-modifying antirheumatic drugs (DMARDs) as first-line therapies, but without adequate improvement. After initiation of anti-IL-6R (as first-line or post-corticosteroids and DMARDs), 73% of patients showed resolution or change to ≤grade 1 of irAEs after a median of 2.0 months from initiation of anti-IL-6R therapy. Six patients (7%) stopped anti-IL-6R due to adverse events. Of 70 evaluable patients by RECIST (Response Evaluation Criteria in Solid Tumors) V.1.1 criteria; the ORR was 66% prior versus 66% after anti-IL-6R (95% CI, 54% to 77%), with 8% higher complete response rate. Of 34 evaluable patients with melanoma, the ORR was 56% prior and increased to 68% after anti-IL-6R (p=0.04). CONCLUSION Targeting IL-6R could be an effective approach to treat several irAE types without hindering antitumor immunity. This study supports ongoing clinical trials evaluating the safety and efficacy of tocilizumab (anti-IL-6R antibody) in combination with ICIs (NCT04940299, NCT03999749).
Collapse
Affiliation(s)
- Faisal Fa'ak
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - Maryam Buni
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Adewunmi Falohun
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Huifang Lu
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Juhee Song
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | - Van A Trinh
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | - Khaled M Elsayes
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kaysia Ludford
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Emma J Montazari
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Julia Chernis
- University of Texas Health Science Center at Houston John P and Katherine G McGovern Medical School, Houston, Texas, USA
| | - Maya Dimitrova
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - Sabina Sandigursky
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - Jeffrey A Sparks
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Osama Abu-Shawer
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Osama Rahma
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | | | - Rafee Talukder
- Fred Hutchinson Cancer Center, University of Washington School of Medicine, Seattle, Washington, USA
| | - Namrata Singh
- Fred Hutchinson Cancer Center, University of Washington School of Medicine, Seattle, Washington, USA
| | - Sarah H Chung
- Fred Hutchinson Cancer Center, University of Washington School of Medicine, Seattle, Washington, USA
| | - Petros Grivas
- Fred Hutchinson Cancer Center, University of Washington School of Medicine, Seattle, Washington, USA
| | - May Daher
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ala Abudayyeh
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Iman Osman
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - Jeffrey Weber
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - Jean H Tayar
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Noha Abdel-Wahab
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Assiut University Faculty of Medicine, Assiut University Hospitals, Assiut, Egypt
| | - Adi Diab
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Immune check point inhibitors (ICIs) are a unique class of cancer treatments that harness the body's innate antitumor response. Although these medications have transformed oncology care, they also lead to generalized immune activation that can result in toxicities across a spectrum of organ systems called immune-related adverse events. This article reviews the most common rheumatologic immune-related adverse events and their management. RECENT FINDINGS Inflammatory arthritis, polymyalgia rheumatic, sicca symptoms, systemic sclerosis, myositis, and vasculitis have all been reported as ICI adverse events. Treatment includes nonsteroidal anti-inflammatory drugs, glucocorticoids, traditional DMARDs, and biologics. SUMMARY Rheumatologists have an important role in the management of patients with rheumatologic immune-related adverse events. Working with our oncology colleagues, we can help manage rheumatologic immune-related adverse events while optimally preserving ICI's antitumor effects.
Collapse
Affiliation(s)
- Melissa Defoe
- Division of Rheumatic Diseases, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | |
Collapse
|
46
|
Yang X, Zhang W, Zhu W. Profiling of immune responses by lactate modulation in cervical cancer reveals key features driving clinical outcome. Heliyon 2023; 9:e14896. [PMID: 37151676 PMCID: PMC10161385 DOI: 10.1016/j.heliyon.2023.e14896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 04/07/2023] Open
Abstract
Cervical cancer is still an important problem perplexing health management in developing countries. Previous studies have shown that cervical cancer cells show markers of aerobic glycolysis, suggesting that these tumors may secrete lactic acid. Through the biological characterization of lactate gene in tumor and its relationship with immune cells in tumor microenvironment, a lactate scoring system capable of evaluating cancer prognosis was constructed to explore the molecular mechanism of lactate metabolism disorder affecting prognosis. 29 hub genes in this study were differentially expressed in cervical cancer, including 24 genes related to lactate metabolism, LDHA in Lactate dehydrogenase (LDH) group, SLC16A3 in Monocarboxylate transporters (MCT) group and three Histone lactation modification related genes (EP300, ACAT1, ACACA). More importantly, we found that from an epigenetic point of view, histone lactation plays an important role in the pathogenesis and prognosis of cervical cancer. Mainly affect the prognosis of the disease through changes in the infiltration of plasmacytoid Dendritic Cell (pDC) and Central Memory T cell (Tcm) in the tumor immune microenvironment. Lactate inhibition may be a useful tool for anticancer therapy.
Collapse
|
47
|
Pacholczak-Madej R, Kosałka-Węgiel J, Kuszmiersz P, Mituś JW, Püsküllüoğlu M, Grela-Wojewoda A, Korkosz M, Bazan-Socha S. Immune Checkpoint Inhibitor Related Rheumatological Complications: Cooperation between Rheumatologists and Oncologists. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:4926. [PMID: 36981837 PMCID: PMC10049070 DOI: 10.3390/ijerph20064926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 06/18/2023]
Abstract
In cancer, immune checkpoint inhibitors (ICIs) improve patient survival but may lead to severe immune-related adverse events (irAEs). Rheumatic irAEs are a distinct entity that are much more common in a real-life than in clinical trial reports due to their unspecific symptoms and them being a rare cause of hospitalization. This review focuses on an interdisciplinary approach to the management of rheumatic irAEs, including cooperation between oncologists, rheumatologists, and immunologists. We discuss the immunological background of rheumatic irAEs, as well as their unique clinical characteristics, differentiation from other irAEs, and treatment strategies. Importantly, steroids are not the basis of therapy, and nonsteroidal anti-inflammatory drugs should be administered in the front line with other antirheumatic agents. We also address whether patients with pre-existing rheumatic autoimmune diseases can receive ICIs and how antirheumatic agents can interfere with ICIs. Interestingly, there is a preclinical rationale for combining ICIs with immunosuppressants, particularly tumor necrosis factor α and interleukin 6 inhibitors. Regardless of the data, the mainstay in managing irAEs is interdisciplinary cooperation between oncologists and other medical specialties.
Collapse
Affiliation(s)
- Renata Pacholczak-Madej
- Department of Clinical Oncology, The Maria Skłodowska-Curie National Research Institute of Oncology, Kraków Branch, 31-115 Kraków, Poland
- Department of Anatomy, Jagiellonian University Medical College, 33-332 Kraków, Poland
| | - Joanna Kosałka-Węgiel
- Department of Rheumatology and Immunology, Jagiellonian University Medical Kraków, 30-688 Krakow, Poland
- Division of Rheumatology and Immunology Clinical, University Hospital, 30-688 Kraków, Poland
| | - Piotr Kuszmiersz
- Department of Rheumatology and Immunology, Jagiellonian University Medical Kraków, 30-688 Krakow, Poland
- Division of Rheumatology and Immunology Clinical, University Hospital, 30-688 Kraków, Poland
| | - Jerzy W. Mituś
- Department of Anatomy, Jagiellonian University Medical College, 33-332 Kraków, Poland
- Department of Surgical Oncology, National Research Institute of Oncology, Kraków Branch, 31-115 Kraków, Poland
| | - Mirosława Püsküllüoğlu
- Department of Clinical Oncology, The Maria Skłodowska-Curie National Research Institute of Oncology, Kraków Branch, 31-115 Kraków, Poland
| | - Aleksandra Grela-Wojewoda
- Department of Clinical Oncology, The Maria Skłodowska-Curie National Research Institute of Oncology, Kraków Branch, 31-115 Kraków, Poland
| | - Mariusz Korkosz
- Department of Rheumatology and Immunology, Jagiellonian University Medical Kraków, 30-688 Krakow, Poland
- Division of Rheumatology and Immunology Clinical, University Hospital, 30-688 Kraków, Poland
| | - Stanisława Bazan-Socha
- Department of Internal Medicine, Jagiellonian University Medical College, 30-688 Kraków, Poland
| |
Collapse
|
48
|
Kubo M, Miyagaki T, Ohashi H, Kishi A, Miyano K, Okano T, Muto M, Takeuchi S, Imamura M, Kadono T. A case of ruptured Baker's cyst induced by nivolumab and ipilimumab. JOURNAL OF CUTANEOUS IMMUNOLOGY AND ALLERGY 2023. [DOI: 10.1002/cia2.12290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023] Open
Affiliation(s)
- Marie Kubo
- Department of Dermatology St. Marianna University School of Medicine Kawasaki Japan
| | - Tomomitsu Miyagaki
- Department of Dermatology St. Marianna University School of Medicine Kawasaki Japan
| | - Hiroyuki Ohashi
- Department of Dermatology St. Marianna University School of Medicine Kawasaki Japan
| | - Arisa Kishi
- Department of Dermatology St. Marianna University School of Medicine Kawasaki Japan
| | - Kaoru Miyano
- Department of Dermatology St. Marianna University School of Medicine Kawasaki Japan
| | - Tatsuro Okano
- Department of Dermatology St. Marianna University School of Medicine Kawasaki Japan
| | - Mayuko Muto
- Department of Dermatology St. Marianna University School of Medicine Kawasaki Japan
| | - Sora Takeuchi
- Department of Dermatology St. Marianna University School of Medicine Kawasaki Japan
| | - Mitsuru Imamura
- Division of Rheumatology and Allergology, Department of Internal Medicine St. Marianna University School of Medicine Kawasaki Japan
| | - Takafumi Kadono
- Department of Dermatology St. Marianna University School of Medicine Kawasaki Japan
| |
Collapse
|
49
|
Xin Z, You L, Na F, Li J, Chen M, Song J, Bai L, Chen J, Zhou J, Ying B. Immunogenetic variations predict immune-related adverse events for PD-1/PD-L1 inhibitors. Eur J Cancer 2023; 184:124-136. [PMID: 36917924 DOI: 10.1016/j.ejca.2023.01.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 01/30/2023] [Indexed: 02/24/2023]
Abstract
BACKGROUND PD-1/PD-L1 inhibitors have brought remarkable benefits but can cause profound immune-related adverse events (irAEs). The host immunogenetic background is likely to play a role in irAE susceptibility. In this study, we aimed to identify potential immunogenetic biomarkers to predict irAEs. METHODS Patients with solid tumours receiving PD-1/PD-L1 blockade were recruited and followed up. Genes considered pivotal contributors to tumour-immunity and autoimmune diseases were screened out via protein-protein interaction network and Cytoscape. Consequently, thirty-nine variants in eighteen genes were genotyped using the multiplex genotyping assay. Association analysis between genetic variants and irAEs as well as irAEs-free survival was performed. RESULTS Four immunogenetic variants as predictive biomarkers of irAEs were identified. The C allele of Mitogen-Activated Protein Kinase 1 (MAPK1) rs3810610 (odds ratio [OR] = 1.495, 95% confidence interval [CI] = 1.093-2.044, P = 0.012) was a risk predictor while the A allele of PTPRC rs6428474 (OR = 0.717, 95% CI = 0.521-0.987, P = 0.041) was a protective factor for all-grade irAEs. The A allele of ADAD1 rs17388568 (OR = 2.599, 95% CI = 1.355-4.983, P = 0.003) increased the risk while the G allele of IL6 rs1800796 (OR = 0.425, 95% CI = 0.205-0.881, P = 0.018) protected patients from high-grade irAEs. Significant immunogenetic variants reached a similar tendency in PD-1 blockade or lung cancer subgroups. In multivariate Cox regression analysis, the MAPK1 rs3810610 was an independent factor regarding all-grade irAEs-free survival (CC versus CT or TT: hazard ratio [HR] = 0.71, 95% CI = 0.52-0.99, P = 0.042). ADAD1 rs17388568 (AA versus AG or GG: HR = 0.11, 95% CI = 0.025-0.49, P = 0.004) and IL6 rs1800796 (GG or GC versus CC: HR = 3.10, 95% CI = 1.315-7.29, P = 0.01) were independent variables for high-grade irAEs-free survival. CONCLUSION We first identified several immunogenetic polymorphisms associated with irAEs and irAEs-free survival in PD-1/PD-L1 blockade-treated tumour patients, and they may serve as potential predictive biomarkers.
Collapse
Affiliation(s)
- Zhaodan Xin
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Liting You
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, PR China; Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Feifei Na
- Department of Thoracic Cancer, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Jin Li
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Min Chen
- Department of Laboratory Medicine, The First Affiliated Hospital of Hainan Medical College, Haikou, Hainan Province 570100, PR China
| | - Jiajia Song
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Ling Bai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Jie Chen
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, PR China.
| | - Juan Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, PR China.
| | - Binwu Ying
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, PR China.
| |
Collapse
|
50
|
Ghosh N, Couette N, van Binsbergen WH, Weinmann SC, Jivanelli B, Shea B, Bass AR, Benesova K, Bingham CO, Calabrese C, Cappelli LC, Chan KK, Choy E, Daoussis D, Goodman S, Hudson M, Jamal S, Leipe J, Lopez-Olivo MA, Suarez-Almazor M, van der Laken CJ, Meara AS, Liew D, Kostine M. Identification of outcome domains in immune checkpoint inhibitor-induced inflammatory arthritis and polymyalgia rheumatica: A scoping review by the OMERACT irAE working group. Semin Arthritis Rheum 2023; 58:152110. [PMID: 36372016 PMCID: PMC10026626 DOI: 10.1016/j.semarthrit.2022.152110] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 07/22/2022] [Accepted: 08/05/2022] [Indexed: 11/11/2022]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICI), increasingly used cancer therapeutics, can cause off-target inflammatory effects called immune-related adverse events (irAEs), including ICI-induced inflammatory arthritis (ICI-induced IA) and polymyalgia rheumatica (ICI-induced PMR). There are no validated classification criteria or outcome measures for these conditions, and adaptation of treatment recommendations from corresponding rheumatic diseases may not be appropriate. We summarized clinical descriptors of ICI-induced IA and ICI-induced PMR and aggregated domains used for these conditions in order to inform the development of a core set of outcome domains. METHODS As the initial step of the core domain set generation process, we systemically searched Medline (Pubmed), EMBASE, Cochrane, and CINHL through March 2021 to identify all studies that provide both clinical descriptions and domains relevant to ICI-induced IA and ICI-induced PMR. Domains were mapped to core areas, such as pathophysiological manifestations, life impact, resource use, and longevity/survival, as suggested by the OMERACT 2.1 Filter. RESULTS We identified 69 publications, over a third of which utilized non-specific diagnoses of "arthritis," "arthralgia," and/or "PMR". Other publications provided the number, the distribution and/or names of specific joints affected, while others labeled the irAE as the corresponding rheumatic disease, such as rheumatoid arthritis or spondyloarthritis. Most distinct domains mapped to the pathophysiology/manifestations core area (24 domains), such as signs/symptoms (13 domains), labs (6 domains), and imaging (5 domains), with harm domains of adverse effects from irAE treatment and fear of irAE treatment decreasing ICI efficacy. Forty-three publications also referenced irAE treatment and 35 subsequent response, as well as 32 tumor response. CONCLUSION There is considerable heterogeneity in the domains used to clinically characterize ICI-induced IA and ICI-induced PMR. There were several domains mapped to the pathophysiologic manifestations core area, although several publications highlighted domains evenly distributed among the other core areas of life impact, longevity/survival and resource use.
Collapse
Affiliation(s)
- Nilasha Ghosh
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery/Weill Cornell Medical College, 535 E 70th St, New York, NY, USA.
| | - Nina Couette
- Department of Internal Medicine. Division of Rheumatology & Immunology, The Ohio State University, Columbus, OH, USA
| | - Wouter H van Binsbergen
- Amsterdam Rheumatology and Immunology Center, Department of Rheumatology & Clinical Immunology, Amsterdam University Medical Center location DBL Amsterdam, the Netherlands
| | - Sophia C Weinmann
- Department of Rheumatology & Immunology, Duke University, Durham, NC, USA
| | - Bridget Jivanelli
- Hospital for Special Surgery, Kim Barrett Memorial Library, HSS Education Institute, New York, NY, USA
| | - Beverley Shea
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada
| | - Anne R Bass
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery/Weill Cornell Medical College, 535 E 70th St, New York, NY, USA
| | - Karolina Benesova
- Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, Heidelberg, Germany
| | - Clifton O Bingham
- Division of Rheumatology, Johns Hopkins University, Baltimore MD, USA
| | - Cassandra Calabrese
- Department of Rheumatologic and Immunologic Diseases, Cleveland Clinic, Cleveland, OH, USA
| | - Laura C Cappelli
- Division of Rheumatology, Johns Hopkins University, Baltimore MD, USA
| | - Karmela Kim Chan
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery/Weill Cornell Medical College, 535 E 70th St, New York, NY, USA
| | - Ernest Choy
- CREATE Centre, Section of Rheumatology, Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Dimitrios Daoussis
- Department of Rheumatology, University of Patras Medical School, Patras University Hospital, Patras, Greece
| | - Susan Goodman
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery/Weill Cornell Medical College, 535 E 70th St, New York, NY, USA
| | - Marie Hudson
- Jewish General Hospital, Lady Davis Institute for medical research and McGill University, Montreal, Quebec, Canada
| | - Shahin Jamal
- Vancouver Coastal Health, University of British Columbia, Vancouver, BC, Canada
| | - Jan Leipe
- Division of Rheumatology, Department of Medicine V, University Hospital Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Germany
| | | | - Maria Suarez-Almazor
- Department of Health Services Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Conny J van der Laken
- Amsterdam Rheumatology and Immunology Center, Department of Rheumatology & Clinical Immunology, Amsterdam University Medical Center location DBL Amsterdam, the Netherlands
| | - Alexa Simon Meara
- Department of Internal Medicine. Division of Rheumatology & Immunology, The Ohio State University, Columbus, OH, USA
| | - David Liew
- Department of Rheumatology, Austin Health, Department of Clinical Pharmacology and Therapeutics, Austin Health, Australia, Department of Medicine, University of Melbourne, Parkville VIC, Australia
| | - Marie Kostine
- Bordeaux University Hospital, Department of Rheumatology, France
| |
Collapse
|