Abstract
Background:
Cancer patients with autoimmune disease have been excluded from randomized trials of immune checkpoint blockers (ICBs). We conducted a systematic review of observational studies and uncontrolled trials including cancer patients with pre-existing autoimmune disease who received ICIs.
Methods:
We searched 5 electronic databases through November 2023. Study selection, data collection, and quality assessment were performed independently by 2 investigators. We performed a meta-analysis to pool incidence of immune-related adverse events (irAEs), including de novo events and flares of existing autoimmune disease, hospitalizations due to irAEs, as well as deaths.
Results:
A total of 95 studies were included (23,897 patients with cancer and preexisting autoimmune disease). The most common cancer evaluated was lung cancer (30.7%) followed by skin cancer (15.7%). Patients with autoimmune disease were more likely to report irAEs compared to patients without autoimmune disease (relative risk 1.3, 95% CI 1.0 to 1.6). The pooled occurrence rate of any irAEs (flares or de novo) was 61% (95% CI 54% to 68%); that of flares was 36% (95% CI 30% to 43%), and that of de novo irAEs was 23% (95% CI 16% to 30%). Flares were mild (grade <3) in half of cases and more commonly reported in patients with psoriasis/psoriatic arthritis (39%), inflammatory bowel disease (37%), and rheumatoid arthritis (36%). 32% of the patients with irAEs required hospitalization and treatment of irAEs included corticosteroids in 72% of the cases. The irAEs mortality rate was 0.07%. There were no statistically significant differences in cancer response to ICIs between patients with and without autoimmune disease.
Conclusions:
Although more patients with pre-existing autoimmune disease had irAEs, these were mild and managed with corticosteroids in most cases, with no impact on cancer response. These results suggest that ICIs can be used in these patients, but careful monitoring is required, as over a third of the patients will experience a flare of their autoimmune disease and/or require hospitalization. These findings provide a crucial foundation for oncologists to refine their monitoring and management strategies, ensuring that the benefits of ICI therapy are maximized while minimizing its risks.
Keywords: immune checkpoint blockers, autoimmune diseases, anti–cytotoxic T-lymphocyte-associated protein 4, anti–programmed cell death protein 1, anti–programmed death ligand 1
Immune checkpoint blockers (ICBs) have revolutionized the treatment and prognosis of many cancers, including those in advanced stages. Three major classes of ICIs are currently approved by the U.S. Food and Drug Administration (FDA): anti–cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) antibodies, anti–programmed cell death protein and anti–programmed death ligand 1 (PD-1/PD-L1) antibodies, and lymphocyte activation gene 3 (LAG-3) (Supplementary Table 1).
When compared to alternative treatment options, ICI therapy, whether as monotherapy or in combination, resulted in considerable improvements in survival rates for various malignancies.1–6 Despite their benefits, ICIs frequently generate immune-related adverse events (irAEs), which can impact practically any system or organ and can be severe.7,8
The precise pathophysiology of irAEs is unknown and may vary depending on the toxicity phenotype, however irAEs are commonly related to the widespread activation of immune pathways, culminating in inflammatory and autoimmune symptoms. As a result, cancer patients with autoimmune conditions have been excluded from participating in ICI trials due to concerns about increasing their risk for immune toxicities and/or flares of their concomitant autoimmune disease. Limited data exist regarding the safety of ICIs in patients with cancer and autoimmune disease.7,9,10 About 15% of the U.S. population suffers from an autoimmune disorder (approximately 50 million people).11–13 Furthermore, autoimmune and chronic inflammatory diseases have been significantly associated with an increased risk for cancer.14 It is thus critical to quantify the potential risk for immune toxicity and for increased disease activity in patients with cancer who could benefit from ICI therapy but have a concomitant autoimmune disorder.
To the best of our knowledge, no controlled clinical trials have been reported to date evaluating the use of ICIs in patients with autoimmune disease. We systematically reviewed observational studies and uncontrolled trials reporting on outcomes of cancer patients with pre-existing autoimmune disease who received ICIs to determine the frequency of flares of autoimmune disease, de novo irAEs, hospitalizations, and deaths. Previous reviews have summarized irAE rates in patients with an underlying autoimmune disease (Supplementary Table 2), but the evolving nature of the topic, new ICI approvals, and the heterogeneity of autoimmune diseases (e.g., a psoriasis flare may not have the same outcome as a lupus flare) points to the need for a more comprehensive and systematic synthesis that estimates the incidence of irAEs, overall and according to specific condition among patients with cancer and pre-existing autoimmune disease.
Methods
For this systematic review, we followed the Cochrane Collaboration methodology and the 2020 PRISMA statement for reporting our results.15,16
Eligibility criteria
We included prospective or retrospective studies reporting the effects of ICIs in cancer patients with pre-existing autoimmune disorders. We excluded animal studies, clinical trials not reporting on autoimmune disease, and review articles. We also excluded case reports or series, studies where it was unclear whether all patients had an autoimmune disease (e.g., nonspecific interstitial lung disease, thyroid dysfunction, autoantibodies, paraneoplastic syndromes), studies exploring association between comorbidities and irAEs, and studies that did not provide separate data or outcome data (e.g., proportion of irAEs, flares, or deaths) for the patients with autoimmune disease.
Information sources
The databases searched were Medline (through Ovid), EMBASE (through Ovid), The Cochrane Library, Web of Science, and PubMed (non-Medline), from the start of the database through November 2023. In addition, the reference lists of included articles and prior comprehensive reviews were hand-searched. EndNote software (Clarivate Analytics) was used to store all citations and for duplicate checking.
Search strategy
The first search strategy was developed and conducted by a librarian (G.P.) with expertise in systematic reviews in the medical field to encompass all ICIs approved by the FDA. As opposed to previous reviews, ours was an exceptionally comprehensive search aimed to find all published studies on the topic. We included broad terms such as “cancer”, “tumor”, and “neoplasms” and drug-related terms such as “ipilimumab” and “nivolumab”, among others. The second search was run by another expert librarian (Y.G.) and incorporated terms for ICIs approved from 2019 onward. The final search was done by the first author of this study using the same strategy as the second but incorporating the FDA-approved ICIs in 2023 with the help of the librarian (Y.G.) with an end date of November 2023 and restricting results to new citations and human studies. The Medline search strategies are shown in Supplementary Table 3.
Selection process
The eligibility of the identified citations was assessed independently by pairs of reviewers each time the search was updated (N.A.; M.A.L.-O.; J.D.A.; M.K.; X.P.; J.J.K.; and P.C.) in a 2-step approach. Titles and abstracts of all retrieved citations were screened first, and the full text of those deemed potentially relevant was retrieved for further screening. Disagreements at either stage were resolved through discussion and, if needed, third-party adjudication (M.A.L.-O. or M.S.A.). We used DistillerSR (Evidence Partners Incorporated; Ottawa, Canada) to manage selection. For the last update (years 2022 and 2023), we used a modified screening approach using DistillerSR’s machine learning tool to estimate the likelihood of relevance based on a previously validated approach.17
Data collection process
One reviewer extracted data from individual studies, and another reviewer cross-checked the information (N.A.; M.A.L.-O.; J.D.A.; M.K.; X.P.; J.J.K.; and P.C.). Disagreements were resolved through discussion. We used Microsoft Excel to extract and tabulate the data.
Data items
We collected the following information: i) general information including title, authors, contact address, publication source, publication year, country, study sponsor; ii) study characteristics including type of study design, study period, sample size, exclusion criteria, and number of patients with and without pre-existing autoimmune disease; iii) participants’ baseline characteristics (age, gender), type of autoimmune disease, time since autoimmune disease diagnosis, autoimmune disease treatments, number of patients discontinuing their autoimmune disease treatment before the initiation of ICI, number of patients with autoimmune disease activity (i.e., symptoms from the autoimmune disease at the time of ICI treatment initiation); and iv) cancer characteristics including type of cancer, cancer stage, number of patients with previous use of ICI, current ICI treatment, and ICI doses. Outcome data included number of patients experiencing an irAE, categorized as a flare of their concomitant autoimmune disease or an unrelated de novo irAE. We also recorded irAE grade (based on the Common Terminology Criteria for Adverse Events), median time to flares after ICI initiation, treatment used for management of irAEs (i.e., corticosteroids, immunosuppressants), hospitalizations due to irAEs, permanent discontinuation of ICIs after irAEs, and deaths due to irAEs. The cancer outcomes collected included complete responder, partial responders and progressive disease.
Study risk of bias assessment
One reviewer assessed the risk of bias of the included studies, and another reviewer cross-checked the information (N.H.A.-W.; M.A.L.-O.; J.J.K.; and P.C.). Studies were appraised using The Newcastle Ottawa Scale (NOS).18 This instrument assesses the potential for bias due to confounding, in selection of participants, in measurement of interventions, due to departures from intended interventions, due to missing data, and in measurement of outcomes. Studies can be awarded a maximum of 1 star for each domain (2 if studies controlled for additional confounders). The maximum score allocated is 4 in the selection domain, 2 in the comparability domain, and 3 in the outcome (or exposure, for case-control studies) domain, with a total maximum score of 9. A higher score indicates a lower risk of bias.19
Effect measures
Our primary outcomes measures were flare and any irAEs occurrence. Secondary outcomes included de novo irAEs, median time to flare after ICI initiation, use of corticosteroids and/or immunosuppressants to manage irAEs, deaths related to irAEs, hospitalizations, permanent ICI discontinuation after an irAE, and cancer outcomes (i.e., complete responders, partial responders, and progressive disease). For the primary outcome, our numerator was the number of cancer patients with autoimmune disease and flares in each study. For the denominator, we considered all patients with cancer and autoimmune disease. We excluded from the denominator non-autoimmune diseases included in the individual analysis as pre-existing autoimmune conditions, such as gout, asthma, atopic dermatitis, Bell’s palsy, eczema, fibromyalgia, idiopathic pulmonary fibrosis, lipoid nephrosis, mucositis, reactive arthritis, and sicca syndrome. We pooled the proportions and calculated relative risks and their 95% CIs. For comparisons of dichotomous variables, we estimated the relative risk and 95% CIs.
Synthesis methods
Analyses were conducted using Stata statistical software version 18 (StataCorp). Descriptive statistics were used to report frequencies of autoimmune disease and cancer types. Pooled frequencies were calculated using the Freeman-Tukey arcsine transformation to stabilize variances and conducted a meta-analysis using inverse variance weights. Then, estimates and their 95% CI boundaries were back-transformed into proportions. We used random effects models to calculate more conservative pooled estimates (incidences/proportions and RR). Study heterogeneity was assessed by using the I2 statistic and subsequent χ2 test. We performed exploratory subgroup analyses to evaluate: 1) the impact of the treatment strategy (combination therapy (2 ICIs or an ICI combined with another treatment) vs monotherapy); and 2) study design (prospective vs retrospective studies). A meta-regression was performed to investigate the extent to which statistical heterogeneity was explained by study sample size, year in which the study was conducted, type of ICI agent received, and grade of irAE reported.
Reporting bias assessment
The risk of publication bias was assessed through funnel plots. When the regression slope significantly deviated from the vertical slope, this was considered to indicate significant potential for bias.
Certainty assessment
The strength of the evidence was evaluated using the GRADE system.20 It divides findings into four categories: i) high quality, which indicates that there is little chance that future research will alter our conclusions; ii) moderate quality, which indicates that future research will probably have an impact on our conclusions; iii) low quality, which indicates that future research will likely have a significant impact on our conclusions; and iv) very low quality, which indicates that there is uncertainty in our conclusions. Findings based on observational studies are rated as low or very low quality of evidence with the GRADE system; however, they can lead to strong recommendations in clinical settings where randomized trials are not available.
Results
Study selection
Flow of studies.
Our searches found 67,006 citations; after exclusion of duplicates, we screened titles and abstracts of 37,163 citations. Of these, we excluded 29,649 citations. The reasons for exclusion at each screening step are shown in Figure 1. We screened the full text of the remaining 7,514 citations, and we found 120 publications representing 95 studies meeting our eligibility criteria.9,21–139
Figure 1.
PRISMA flow diagram: Study selection.
Excluded studies.
There were 7,301 citations excluded because they were case reports or series (with or without pre-existing autoimmune disease) or observational studies without patients with pre-existing autoimmune disease. The remaining 93 excluded were studies with an unclear number of patients with pre-existing autoimmune disease, studies reporting on patients with pre-existing autoantibodies (e.g., rheumatoid factor, anti-nuclear antibodies), studies exploring association between comorbidities and irAEs, or studies with insufficient data provided for analysis (i.e., outcome data not reported or preliminary data from studies included in the analysis). These references are listed in Supplementary Table 4.
Study characteristics
Fifty-two studies (54.1%) also reported data in cancer patients without a pre-existing autoimmune disease; 11 (11.5%) were results from national registries, regulatory agencies, or claims data; 4 (4.2%) were data from uncontrolled trials (single arm or phase IV). In total, the included studies evaluated 23,897 patients with cancer and autoimmune diseases and 51,676 cancer patients without autoimmune disease. Thirty-three studies (34.4%) were multicenter. The studies were conducted in Australia, Canada, China, Europe, Israel, Japan, Turkey, and the United States. The most commonly reported autoimmune diseases were inflammatory bowel diseases (34.7%), type 1 diabetes (13.1%), rheumatoid arthritis (11.6%), psoriasis (7.5%), and Addison disease (3.0%) (Table 1).
Table 1.
Characteristics of the studies included in our analysis (n=95 studies).
Characteristic | Value |
---|---|
Design | |
Uncontrolled cohort | 28 (29.2%) |
Controlled cohort | 51 (54.1%) |
Registries | 11 (11.5%) |
Uncontrolled trials | 4 (4.2%) |
Individual patient data from multiple controlled trials | 1 (1.0%) |
Centers | |
Single-center | 63 (65.6%) |
Multi-center | 32 (34.4%) |
Countries | |
Australia | 4 (4.2%) |
Canada | 3 (3.1%) |
China | 2 (2.1%) |
Europe | 30 (31.3%) |
Israel | 1 (1.0%) |
Japan | 6 (6.3%) |
United States | 40 (42.7%) |
Global collaboration | 9 (9.4%) |
Time period (range) | 1999 to 2022 |
Number of patients without pre-existing autoimmune disease | 51,676 |
Number of patients with pre-existing autoimmune disease | 23,897 |
Number of autoimmune diseases reported (n= 30,493a) | |
Inflammatory bowel disease | 10,585 (34.7%) |
Type 1 diabetes | 4,007 (13.1%) |
Rheumatoid arthritis | 3,542 (11.6%) |
Unspecified | 2,436 (8.0%) |
Psoriasis | 2,296 (7.5%) |
Addison’s disease | 922 (3.0%) |
Hashimoto’s thyroiditis | 758 (2.5%) |
Systemic lupus erythematosus | 595 (2.0%) |
Dermatologic (unspecified) | 525 (1.7%) |
Vasculitis | 515 (1.7%) |
Autoimmune thyroiditis | 467 (1.5%) |
Graves’ disease | 458 (1.5%) |
Multiple sclerosis | 339 (1.1%) |
Rheumatic (unspecified) | 325 (1.1%) |
Lichen | 300 (1.0%) |
Otherb | 2,423 (7.9%) |
Some patients had overlapping disease, therefore the number of autoimmune diseases is larger than the number of patients with pre-existing autoimmune diseases reported.
Other autoimmune disease with less than 1% individually presence were: celiac disease, vitiligo, CREST syndrome (calcinosis, Raynaud phenomenon, esophageal dysmotility, sclerodactyly, and telangiectasia), spondyloarthropathies, endocrine (unspecified), systemic sclerosis, myasthenia gravis, ulcerative colitis, autoimmune hepatitis, Crohn’s disease, alopecia areata, polymyalgia rheumatica, dermatomyositis, sarcoidosis, bullous pemphigus, psoriasis with psoriatic arthritis, psoriatic arthritis, neurologic (unspecified), pernicious anemia, immune thrombocytopenic purpura, gastrointestinal (unspecified), inflammatory arthritis, Sjogren, Guillain-Barré syndrome, Raynaud’s phenomenon, uveitis, discoid lupus, IgA nephropathy, mixed or undifferentiated connective tissue disorder, cutaneous lupus, myositis, hemolytic anemia, polymyositis, granulomatosis with polyangiitis, autoimmune hypophysitis, Behçet’s disease, primary biliary cholangitis, chronic inflammatory demyelinating neuropathy, giant cell arteritis, hidradenitis suppurativa, rheumatic fever, autoimmune urticaria, anti-phospholipid syndrome, Churg-Strauss syndrome, erythema nodosum, IgG4-sclerosing, juvenile idiopathic arthritis, glomerulonephritis, myelitis, autoimmune pancreatitis, polyarteritis nodosa, autoimmune retinopathy.
Participants’ characteristics
Table 2 shows the baseline demographic characteristics of participants (n=23,897) in the included studies. The mean age ranged from 54 to 76 years (pooled mean 66.1, 95% CI 65.3 to 67.0). The proportion of male participants ranged between 19% and 100%. Lung cancer (30.7%) was the most commonly reported cancer, followed by skin cancer (15.7%). Studies included patients with resectable cancer (i.e., melanoma) and advanced cancer stages (i.e., II to IV). Four studies reported use of previous ICIs in the included patients (CTLA-4 in 3 and PD-1/PD-L1 in 2).9,40,62,89,140 Nivolumab, pembrolizumab, and ipilimumab were the ICIs most commonly evaluated. Duration of current ICI treatment ranged from 120 to 196 days, and the number of cycles ranged between 1 and 20. The percentage of patients on treatment for autoimmune disease in each study ranged from 0% to 87.5%; this treatment varied and included nonsteroidal anti-inflammatory drugs, corticosteroids (topical, oral, or systemic), and conventional and targeted disease-modifying anti-rheumatic drugs (e.g., hydroxychloroquine, methotrexate, sulfasalazine, infliximab, apremilast). At the time of initiating ICI, the percentage of patients with active autoimmune disease ranged from 0% to 67% across studies.
Table 2.
Baseline demographic characteristics of participants with autoimmune disease in the included studies.
Characteristic | Value |
---|---|
Age, pooled mean (95% CI) | 66.1 (65.3 to 67.0) |
Age, range of means | 54 to 76 years |
Male sex, range | 19% to 100% |
Cancer type, no. (%) (n=23,897 participants with autoimmune disease) | |
Lung (non–small cell carcinoma, small cell carcinoma, mesothelioma, unspecified) | 7,336 (30.7%) |
Skin (melanoma, squamous cell carcinoma, Merkel cell carcinoma) | 3,752 (15.7%) |
Genitourinary (renal cell, urothelial, bladder, prostate, penile, unspecified) | 2,127 (8.9%) |
Gastrointestinal (colorectal, hepatocellular, esophageal, pancreatic, unspecified) | 1,936 (8.1%) |
Head and neck (sinonasal, unspecified) | 263 (1.1%) |
Gynecological (breast, ovarian, endometrial, epithelial trophoblastic, unspecified) | 134 (0.56%) |
Hematological (Hodgkin’s lymphoma, B-cell lymphoma, unspecified) | 127 (0.53%) |
Unspecified | 8,149 (34.1%) |
Othersa | 74 (0.31%) |
Distribution of ICIs, no. (%) (n=23,897 participants with autoimmune disease) | |
Anti–PD-1 | 5,807 (24.3%) |
Nivolumab | 4,134 (17.3%) |
Pembrolizumab | 2,557 (10.7%) |
Ipilimumab | 2,246 (9.4%) |
ICI monotherapy | 1,768 (7.4%) |
Anti–PD-1/PD-L1 | 1,147 (4.8%) |
ICI combination | 550 (2.3%) |
Combination | 502 (2.1%) |
Durvalumab | 406 (1.7%) |
Anti-CTLA–4 | 263 (1.1%) |
Atezolizumab | 48 (0.2%) |
Avelumab | 24 (0.2%) |
ICI + non-ICI | 27 (0.1%) |
Unspecified ICI | 4397 (18.4%) |
Duration of current ICI treatment, range | 120 to 196 days |
Number of cycles, range | 1 to 20 |
Patients on treatment for autoimmune disease, range | 0% to 87.5% |
Active autoimmune disease, range | 0% to 67% |
Others included neuroendocrine, brain unspecified, sarcoma, thymoma, thyroid.
Risk of bias within studies
Supplementary Figure 1 shows the risk of bias assessment of the included studies in an aggregate manner. NOS score ranged from 4 to 7 stars. For 28 studies (29.2%), there was no control group (i.e., patients without autoimmune disease receiving ICIs). Few studies adjusted their analysis for potential confounders. In 1 study, the follow-up time was not reported.125 Four studies reported no source of funding, 1 cited the U.S. National Institutes of Health,62 and 1 reported 7 funding sources.80
Safety of ICIs in patients with pre-existing autoimmune diseases
Table 3 shows the reported number of patients not experiencing irAEs and those experiencing de novo irAEs, flares, and deaths. Patients with autoimmune disease were 30% more likely to report an irAE compared to patients without autoimmune disease (relative risk 1.3, 95% CI 1.0 to 1.6). The pooled frequency of any irAEs (flares or de novo) was 61% (95% CI 54%-68%); the pooled frequency of flares was 36% (95% CI 29% to 43%), and that of de novo irAEs was 23% (95% CI 16% to 30%). Out of 1,827 de novo irAEs reported, the most common were colitis (17%), thyroid dysfunction (11%), skin rash (6%), and arthritis/arthralgia (5%). When categorized by type of ICI, 25% (95% CI 15% to 36%) of the patients receiving anti–PD-1/PD-L1 agents had irAEs, as did 38% (95% CI 22% to 54%) of those who received anti–CTLA-4 agents. Although most irAEs were mild, 30% of the irAEs were grade 3–4, and 32% of all irAEs in patients with pre-existing autoimmune disease required hospitalization. Pooled mortality related to ICIs was less than 0.1%; however, the pooled frequency of permanent discontinuation of the ICI due to irAEs was 35% (95% CI 24% to 46%). Most irAEs were managed with local or systemic corticosteroids (72%), and less than a third of patients in the pooled analysis required immunosuppression (27%; 95% CI 15% to 40%). The irAEs rates were similar when patients with combination therapy were removed from the analysis (Supplementary Table 5).
Table 3.
Effects associated with ICIs in patients with pre-existing autoimmune diseases in the included studies.
Number of studies with these data | Number of patientsa | Pooled incidence or relative risk (95% CI) | |
---|---|---|---|
Patients not experiencing irAEs | 68 | 23430 | 35.7% (28.5% to 43.3%) |
Risk of irAEs (autoimmune vs no autoimmune disease), relative risk | 21 | 2399 vs 20348 | 1.3 (1.0 to 1.6) |
Risk of irAEs in patients receiving treatment for autoimmune disease vs no treatment), relative risk | 4 | 183 | 1.1 (CI 0.72–1.6) |
irAEs (any) | 68 | 4350 | 61.0% (54.3% to 67.5%) |
Grade of irAEs | |||
1–2 | 29 | 966 | 50.1% (39.7% to 60.5%) |
3–4 | 25 | 713 | 29.6% (20.6% to 39.4%) |
Management of irAEs | |||
Corticosteroids | 35 | 478 | 71.6% (59.2% to 82.9%) |
Immunosuppressants/biologics | 26 | 570 | 26.5% (14.7% to 39.7%) |
irAEs per type of ICI agent | |||
Anti–PD-1/PD-L1 | 3 | 74 | 25.2% (15.4% to 36.3%) |
Anti–CTLA-4 | 4 | 98 | 37.7% (22.2% to 54.4%) |
De novo irAEs (not flares) | 59 | 1827 | 22.9% (16.3% to 30.0%) |
Patients with irAEs requiring hospitalization | 6 | 103 | 31.7% (13.8% to 52.0%) |
Permanent ICI discontinuation after irAEs | 48 | 968 | 34.9% (24.4% to 46.0%) |
Deaths related to irAEs | 43 | 943 | 0.07% (<0.01% to 3.9%) |
Flares (any autoimmune disease) | 74 | 2753 | 36.3% (29.5% to 43.3%) |
Median time to flare after ICI initiation, range | 25 | 687 | 7 to 470 days |
Flares per type of autoimmune disease (most commonly reported) | |||
Inflammatory bowel disease | 10 | 480 | 36.9% (28.5% to 45.7%) |
Rheumatoid arthritis | 13 | 789 | 35.9% (22.2% to 50.4%) |
Psoriasis/psoriatic arthritis | 13 | 560 | 38.9% (22.9% to 55.8%) |
Autoimmune thyroiditis (any) | 10 | 458 | 15.4% (8.2% to 23.8%) |
Multiple sclerosis | 8 | 291 | 18.0% (<1% to 49.3%) |
Flares per use of DMARDs/corticosteroids at ICI start | 1 | 47 | 29.4% (9.7% to 53.6%) |
Complete responders (autoimmune vs no autoimmune disease), relative risk | 3 | 94 vs 780 | 1.2 (0.47 to 2.9) |
Partial responders (autoimmune vs no autoimmune disease), relative risk | 5 | 172 vs 1179 | 1.1 (0.83 to 1.3) |
Progressive disease (autoimmune vs no autoimmune disease), relative risk | 6 | 129 vs 629 | 1.4 (0.79 to 2.3) |
DMARD, disease-modifying antirheumatic drugs
Number of patients in studies reporting the data.
Flares
The median time to flare after ICI initiation ranged from 7 to 470 days. Flares were most commonly reported in patients with psoriasis/psoriatic arthritis (39%), inflammatory bowel disease (37%), rheumatoid arthritis (36%), multiple sclerosis (18%), and any type of autoimmune thyroiditis (15%). The pooled incidence of flares for people using immunosuppressive agents or corticosteroids at the start of ICI therapy was 29% (95% CI 10% to 54%).
Cancer response to ICI in patients with cancer and pre-existing autoimmune diseases
Rates of complete response after treatment with any ICI did not significantly differ between patients with autoimmune disease and patients without autoimmune disease (relative risk 1.2, 95% CI 0.47 to 2.9). Similarly, no significant differences between those groups were observed in the number of partial responders or patients with progressive disease (Table 3).
Risk of bias across studies
Supplementary Figure 2 shows the funnel plot of the occurrence of flares in the reported studies. The plot looks asymmetrical, with greater numbers of studies with less variability reporting lower occurrence rates and studies with higher variability reporting higher occurrence rates. The Egger test for small-study effects was significant (p=0.006), indicating smaller studies reporting larger effect sizes than the larger studies. However, when studies with sample sizes less than 5 patients (33%, 95% CI 27% to 40%; 58 studies; Egger test p=0.11) and less than 10 patients (29%, 95% CI 24% to 36%; 41 studies, Egger test p=0.66) were removed, the pooled occurrence rates remained similar.
DISCUSSION
We synthetized current published observational studies reporting on the use of ICI in patients with cancer and pre-existing autoimmune disease. We found that irAEs of any type were commonly reported in more than two thirds of the patients, with autoimmune disease flares reported in over a third of the patients and de novo irAEs in over 20% of the patients. The frequency of flares appeared to differ according to the type of autoimmune disease and the ICI received. Most studies reported flares or de novo irAEs that were mild to moderate with low rates of discontinuations and a rate of less than 0.1% for deaths related to irAEs. However, our findings also suggest that these patients required careful monitoring given that a third required hospitalization due to irAEs. We also found that responses to the ICI treatment were similar between cancer patients with and without pre-existing autoimmune disease.
Our systematic review differs from other comprehensive reviews published in the literature (Supplementary Table 2).7,141–162 We found 23 reviews with information on this topic, with the number of included studies ranging from 5 to 53. Eleven were narrative, 12 were systematic reviews and of these, 5 included meta-analyses (2 out of the 5 included non-autoimmune comorbid conditions). None of the search strategies reported were built as a broad search to cover any type of publication reporting on adverse events with ICIs like ours. Although our method increased the number of citations to screen, which we leveraged using machine learning screening methods, our search proved more sensitive and retrieve 95 relevant citations. This facilitated the exclusion of conditions that are not autoimmune (e.g., gout). We excluded studies in which it was unclear if all patients had an autoimmune condition such as interstitial lung disease and thyroid dysfunction; inclusion of this type of disorders could have overestimated the rates of flares or de novo irAEs, given the frequency of these disorders (such as subclinical hypothyroidism), which are also found in the general population, and in fact only few in this category can be labeled as autoimmune disease.143 Nonetheless, our pooled estimate for incidence of flares was similar to those reported in prior meta-analyses (range 23% to 37%).143,161,162 Previously, we reported a review of 123 patients in 49 publications with 92 (75%) reporting an exacerbation of autoimmune disease, irAEs, or both.7 In our previous report, more than 50% of the total number of cases had disease exacerbation, more than a third had de novo irAEs, and 9% irAEs were fatal. In our current review, the frequency of irAEs in patients with autoimmune disease was lower. The differences could be due to increased reporting of poor outcomes in case reports, versus more unbiased reporting in series that systematically identified patients with autoimmune disease receiving ICIs, which is the study design for publications included in the current review.
Our study also reports findings according to different autoimmune diseases, which is more clinically relevant, given the different presentations of each autoimmune condition. A broad spectrum of autoimmune diseases was reported, including psoriasis and/or psoriatic arthritis, rheumatoid arthritis, autoimmune thyroid disease, ulcerative colitis, Crohn disease, multiple sclerosis, myasthenia gravis, and sarcoidosis. Previous comprehensive reviews included meta-analysis according to organ system rather than disease.143,160 Another meta-analysis found that patients undergoing immunosuppression had a similar risk of flare compared to those without immunosuppression.161 When the authors compared the risk of developing flares between rheumatoid arthritis and other autoimmune diseases, the only significant difference observed was between rheumatoid arthritis and autoimmune thyroiditis after removal of the study with a higher proportion of flares reported for autoimmune thyroiditis.161 A prior systematic review including 1 observational study and 3 case reports also found that the frequency of irAEs differed by type of ICI. In our study, we found that flares were more commonly reported with anti–CTLA-4.145
Previous studies have also reported no differences in cancer outcomes between patients with and without autoimmune disease,144 except when the data were analyzed by type of cancer in patients with interstitial lung disease.143 Patients with non–small cell lung cancer and pre-existing interstitial lung disease were more likely to achieve complete response than those without interstitial lung disease.143 Another study pooled the objective response rate (combining complete, partial, and stable response and progressive disease) and found that patients with flares had better objective response rates than those without flares.160
The results of our systematic review and meta-analysis were limited by the available data. Although our search was run recently, we acknowledge the potential of missing newly indexed articles. This is because of the nature of the research question, with multiple studies being published rapidly and continuously. There is also the possibility of newer studies being more methodologically rigorous, which could impact the confidence in our findings. The current state of the evidence is derived from observational/pharmacovigilance studies. However, we are confident that our estimates are robust, and new evidence from observational studies could only change the magnitude of the occurrence.
While some of our results corroborate existing knowledge, the methodological rigor, information on hospitalization rates due to irAEs, inclusion of the most recent ICI approved agents, and the detailed breakdown by type of autoimmune disease add a layer of depth to the current understanding. These enhancements help refine clinical approaches and inform future research directions. We believe these aspects underscore the value of our study and hope that this clarification helps emphasize how our findings contribute to the broader scientific and clinical discourse on the treatment of cancer patients with preexisting autoimmune diseases using ICIs.
Given the high occurrence rate of irAEs, oncologists should implement enhanced monitoring protocols for patients with pre-existing autoimmune diseases undergoing ICI therapy. 1) A thorough baseline assessment of the patient’s autoimmune condition before ICI initiation, could help anticipate the likelihood and severity of flares and potentially guide the choice and timing of ICI therapy, balancing cancer treatment efficacy with potential autoimmune exacerbations. 2) An integrated care approach during ICI treatment would help promptly address irAEs, optimize immunosuppressive therapy, and ensure continuity of cancer care. 3) The risk-benefit ratios of immunotherapy in patients with pre-existing autoimmune diseases needs to be carefully discussed with those patients. Patients should be thoroughly informed about the risks of irAEs, potential symptoms, and the importance of prompt reporting of any changes in their health, and the potential for modifications to their immunosuppressive or cancer treatments. Educated patients are better prepared to manage their conditions in partnership with their healthcare providers. 4) Oncology facilities should be prepared for the potential need for a rapid response to severe irAEs, including immediate access to autoimmune disease treatment interventions and hospital care.
Our results can assist in providing evidence-based data to patients with cancer and pre-existing autoimmune diseases. We combined data from observational studies and uncontrolled trials and found that the rate of flares and de novo irAEs was substantial, but these events were generally mild in most patients. Tumor response rates in patients with autoimmune disease were similar to those in patients without autoimmune disease. In conclusion, immune checkpoint blockade in patients with known autoimmune diseases is possible but requires careful monitoring due to the potential need of hospitalization due to irAEs. Our findings provide a crucial foundation for oncologists to refine their monitoring and management strategies, ensuring with more frequent clinical assessments, closer communication with other autoimmune disease specialists, and patient education about signs and symptoms of irAEs that the benefits of ICI therapy are maximized while minimizing its risks.
Supplementary Material
Highlights.
Having an autoimmune disease (AD) increases the risk of irAEs.
Over 1/3 of patients with AD on ICIs will have AD flares, but most are mild.
Those with AD and irAEs need close monitoring as over 1/3 will need hospitalization.
Studies are needed to quantify irAEs by cancer type and AD treatment at ICI start.
ACKNOWLEDGMENTS
We would like to thank Houssein Safa, MD; Mohsin Shah, MD; Ali Al-Tarbsheh, MD; and Nikitha Vobugari, MD; for their contributions during the study selection. The manuscript was edited by Sarah Bronson, ELS, of the Research Medical Library at The University of Texas MD Anderson Cancer Center.
Source(s) of funding:
This study was supported by the National Cancer Institute (Project number: CA237619), National Institute of Arthritis and Musculoskeletal and Skin Diseases (project number: K08AR079587), and the Rheumatology Research Foundation.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Presentation at a meeting: Lopez-Olivo M, Abdel-Wahab N, Suarez-Almazor M. A Systematic Review and Meta-analysis of Observational Studies Reporting on the Use of Checkpoint Inhibitors in Patients with Cancer and Pre-existing Autoimmune Disease [abstract]. Arthritis Rheumatol. 2019; 71 (suppl 10). https://acrabstracts.org/abstract/a-systematic-review-and-meta-analysis-of-observational-studies-reporting-on-the-use-of-checkpoint-inhibitors-in-patients-with-cancer-and-pre-existing-autoimmune-disease/.
Declaration of Competing Interest
The authors declare the following financial interests/personal relationships which may be considered as potential competing interests. Dr. Suarez-Almazor has received consultant fees from participation on advisory boards for Syneos Health and Celgene. All activities are unrelated to this work. Dr. Xerxes Pundole is an employee and shareholder of Amgen, Inc. The remaining authors have no conflict of interest to declare
REFERENCES
- 1.Marrone KA, Ying W, Naidoo J. Immune-Related Adverse Events From Immune Checkpoint Inhibitors. Clin Pharmacol Ther 2016; 100(3): 242–51. [DOI] [PubMed] [Google Scholar]
- 2.Hodi FS, Chiarion-Sileni V, Gonzalez R, et al. Nivolumab plus ipilimumab or nivolumab alone versus ipilimumab alone in advanced melanoma (CheckMate 067): 4-year outcomes of a multicentre, randomised, phase 3 trial. Lancet Oncol 2018; 19(11): 1480–92. [DOI] [PubMed] [Google Scholar]
- 3.Schachter J, Ribas A, Long GV, et al. Pembrolizumab versus ipilimumab for advanced melanoma: final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006). Lancet 2017; 390(10105): 1853–62. [DOI] [PubMed] [Google Scholar]
- 4.Brahmer J, Reckamp KL, Baas P, et al. Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer. N Engl J Med 2015; 373(2): 123–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Herbst RS, Baas P, Kim DW, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet 2016; 387(10027): 1540–50. [DOI] [PubMed] [Google Scholar]
- 6.Motzer RJ, Escudier B, McDermott DF, et al. Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. N Engl J Med 2015; 373(19): 1803–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Abdel-Wahab N, Shah M, Lopez-Olivo MA, Suarez-Almazor ME. Use of Immune Checkpoint Inhibitors in the Treatment of Patients With Cancer and Preexisting Autoimmune Disease: A Systematic Review. Ann Intern Med 2018; 168(2): 121–30. [DOI] [PubMed] [Google Scholar]
- 8.Postow MA, Sidlow R, Hellmann MD. Immune-Related Adverse Events Associated with Immune Checkpoint Blockade. N Engl J Med 2018; 378(2): 158–68. [DOI] [PubMed] [Google Scholar]
- 9.Menzies AM, Johnson DB, Ramanujam S, et al. Anti-PD-1 therapy in patients with advanced melanoma and preexisting autoimmune disorders or major toxicity with ipilimumab. Ann Oncol 2017; 28(2): 368–76. [DOI] [PubMed] [Google Scholar]
- 10.Johnson DB, Sullivan RJ, Ott PA, et al. Ipilimumab Therapy in Patients With Advanced Melanoma and Preexisting Autoimmune Disorders. JAMA Oncol 2016; 2(2): 234–40. [DOI] [PubMed] [Google Scholar]
- 11.Lerner A, Jeremias P, Matthias T. The World Incidence and Prevalence of Autoimmune Diseases is Increasing. International Journal of Celiac Disease 2015; 3(4): 151–5. [Google Scholar]
- 12.Dooley MA, Hogan SL. Environmental epidemiology and risk factors for autoimmune disease. Curr Opin Rheumatol 2003; 15(2): 99–103. [DOI] [PubMed] [Google Scholar]
- 13.Cooper GS, Bynum ML, Somers EC. Recent insights in the epidemiology of autoimmune diseases: improved prevalence estimates and understanding of clustering of diseases. J Autoimmun 2009; 33(3–4): 197–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Franks AL, Slansky JE. Multiple Associations Between a Broad Spectrum of Autoimmune Diseases, Chronic Inflammatory Diseases and Cancer. Anticancer Res 2012; 32(4): 1119–36. [PMC free article] [PubMed] [Google Scholar]
- 15.Higgins J, Thomas J, Chandler J, et al. Cochrane Handbook for Systematic Reviews of Interventions version 6.4 (updated August 2023). The Cochrane Collaboration; 2023: Available from www.training.cochrane.org/handbook. [Google Scholar]
- 16.Page MJ, McKenzie JE, Bossuyt PM, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ 2021; 372: n71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Hamel C, Kelly SE, Thavorn K, Rice DB, Wells GA, Hutton B. An evaluation of DistillerSR’s machine learning-based prioritization tool for title/abstract screening - impact on reviewer-relevant outcomes. Bmc Med Res Methodol 2020; 20: 256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Wells G, Shea B, O’Connell D, et al. The Newcastle-Ottawa Scale (NOS) for assessing the quality of nonrandomised studies in meta-analyses. Ottawa Hospital Research Institute 2008: Available at: https://www.ohri.ca/programs/clinical_epidemiology/oxford.asp; Accessed on: December 12, 2023. [Google Scholar]
- 19.Stang A. Critical evaluation of the Newcastle-Ottawa scale for the assessment of the quality of nonrandomized studies in meta-analyses. Eur J Epidemiol 2010; 25(9): 603–5. [DOI] [PubMed] [Google Scholar]
- 20.Schünemann H, Brożek J, Guyatt G, Oxman A, and editors. GRADE handbook for grading quality of evidence and strength of recommendations. The GRADE Working Group 2013. [Google Scholar]
- 21.Abu-Sbeih H, Faleck DM, Ricciuti B, et al. Immune Checkpoint Inhibitor Therapy in Patients With Preexisting Inflammatory Bowel Disease. J Clin Oncol 2020; 38(6): 576–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Ak N, Aydiner A. Nivolumab treatment for metastatic thymic epithelial tumors. J Oncol Pharm Pract 2021; 27(7): 1710–5. [DOI] [PubMed] [Google Scholar]
- 23.Alexander S, Swami U, Kaur A, et al. Safety of immune checkpoint inhibitors in patients with cancer and pre-existing autoimmune disease. Ann Transl Med 2021; 9(12): 1033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Ansel S, Rulach R, Trotter N, Steele N. Pembrolizumab for advanced non-small cell lung cancer (NSCLC): Impact of autoimmune comorbidity and outcomes following treatment completion. J Oncol Pharm Pract 2023; 29(4): 785–93. [DOI] [PubMed] [Google Scholar]
- 25.Ardizzoni A, Azevedo S, Rubio-Viqueira B, et al. Primary results from TAIL: a global single-arm safety study of atezolizumab monotherapy in a diverse population of patients with previously treated advanced non-small cell lung cancer. J Immunother Cancer 2021; 9(3): e001865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Arreola L, Tan J, Ye C, et al. Safety and Efficacy of Immune Checkpoint Inhibitors in Patients with Rheumatic Autoimmune Disease: Experience from the Canadian Research Group of Rheumatology in Immuno-Oncology (CANRIO). Ann Rheum Dis 2023; 82: 2118–9. [Google Scholar]
- 27.Bhatlapenumarthi V, Patwari A, Harb AJ. Immune-related adverse events and immune checkpoint inhibitor tolerance on rechallenge in patients with irAEs: a single-center experience. J Cancer Res Clin Oncol 2021; 147(9): 2789–800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Braga Neto MB, Ramos GP, Loftus EV, Jr., Faubion WA, Raffals LE. Use of Immune Checkpoint Inhibitors in Patients With Pre-established Inflammatory Bowel Diseases: Retrospective Case Series. Clin Gastroenterol Hepatol 2021; 19(6): 1285–7 e1. [DOI] [PubMed] [Google Scholar]
- 29.Brown LJ, Weppler A, Bhave P, et al. Combination anti-PD1 and ipilimumab therapy in patients with advanced melanoma and pre-existing autoimmune disorders. J Immunother Cancer 2021; 9(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Calabrese C, Kirchner E, Kontzias A, Velcheti V, Calabrese LH. Rheumatic immune-related adverse events of checkpoint therapy for cancer: case series of a new nosological entity. RMD Open 2017; 3(1): e000412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Calvo V, Fernandez MA, Collazo-Lorduy A, Franco F, Nunez B, Provencio M. Use of immune checkpoint inhibitors in patients with solid tumors and pre-existing autoimmune or inflammatory disease: real-world data. Lung Cancer Manag 2021; 10(4): LMT51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Chan TSY, Hwang YY, Khong PL, et al. Low-dose pembrolizumab and nivolumab were efficacious and safe in relapsed and refractory classical Hodgkin lymphoma: Experience in a resource-constrained setting. Hematol Oncol 2020; 38(5): 726–36. [DOI] [PubMed] [Google Scholar]
- 33.Chanza NM, Xie WL, Issa M, et al. Safety and efficacy of immune checkpoint inhibitors in advanced urological cancers with pre-existing autoimmune disorders: a retrospective international multicenter study. Journal for Immunotherapy of Cancer 2020; 8(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Chanza NM, Xie WL, Issa M, et al. Retrospective analysis of the safety and efficacy of immune checkpoint inhibitors (CPI) among patients (pts) with pre-existing autoimmune disorders (AD) and renal cell carcinoma (RCC) or urothelial carcinoma (UC). J Clin Oncol 2019; 37(15). [Google Scholar]
- 35.Chavaz L, Guyon D, Boespflug A, Cauquil C, Michot JM. Tolerability of immune checkpoint inhibitors in patients with cancer and pre-existing multiple sclerosis. Eur J Cancer 2023; 189: 112928. [DOI] [PubMed] [Google Scholar]
- 36.Cortellini A, Buti S, Santini D, et al. Clinical Outcomes of Patients with Advanced Cancer and Pre-Existing Autoimmune Diseases Treated with Anti-Programmed Death-1 Immunotherapy: A Real-World Transverse Study. Oncologist 2019; 24(6): e327–e37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Cutroneo P, Ingrasciotta Y, Isgro V, et al. Psoriasis and psoriasiform reactions secondary to immune checkpoint inhibitors. Dermatol Ther 2021; 34(2): e14830. [DOI] [PubMed] [Google Scholar]
- 38.Cytryn S, Efuni E, Sandigursky S. Toxicities of single agent and combination immune checkpoint inhibitors in patients with autoimmune diseases. J Clin Oncol 2019; 37(15). [Google Scholar]
- 39.Cytryn S, Efuni E, Velcheti V, Sandigursky S. Use of Immune Checkpoint Inhibitors in Patients with Advanced Lung Cancer and Pre-Existing Autoimmune Diseases. J Thorac Oncol 2019; 14(10): S726–S7. [Google Scholar]
- 40.Danlos FX, Voisin AL, Dyevre V, et al. Tolerance and Efficiency of Anti-Programmed Death 1 Antibodies in Patients with Cancer and Preexisting Autoimmune or Inflammatory Diseases. Arthritis Rheumatol 2017; 69. [Google Scholar]
- 41.Danlos FX, Voisin AL, Dyevre V, et al. Safety and efficacy of anti-programmed death 1 antibodies in patients with cancer and pre-existing autoimmune or inflammatory disease. Eur J Cancer 2018; 91: 21–9. [DOI] [PubMed] [Google Scholar]
- 42.De La Fuente F, Belkhir R, Henry J, et al. Use of a bDMARD or tsDMARD for the management of inflammatory arthritis under checkpoint inhibitors: an observational study. RMD Open 2022; 8(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Debieuvre D, Juergens RA, Asselain B, et al. Two-year survival with nivolumab in previously treated advanced non-small-cell lung cancer: A real-world pooled analysis of patients from France, Germany, and Canada. Lung Cancer 2021; 157: 40–7. [DOI] [PubMed] [Google Scholar]
- 44.Duong SL, Barbiero FJ, Nowak RJ, Baehring JM. Neurotoxicities associated with immune checkpoint inhibitor therapy. J Neurooncol 2021; 152(2): 265–77. [DOI] [PubMed] [Google Scholar]
- 45.Efuni E, Cytryn S, Boland P, et al. Risk of Toxicity After Initiating Immune Checkpoint Inhibitor Treatment in Patients With Rheumatoid Arthritis. J Clin Rheumatol 2021; 27(7): 267–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Efuni E, Cytryn S, Boland P, Sandigursky S. Risk of Immunotherapy Related Toxicity in Patients with Rheumatoid Arthritis. Arthritis Rheumatol 2019; 71. [Google Scholar]
- 47.Faehling M, Schumann C, Christopoulos P, et al. Durvalumab after definitive chemoradiotherapy in locally advanced unresectable non-small cell lung cancer (NSCLC): Real-world data on survival and safety from the German expanded-access program (EAP). Lung Cancer 2020; 150: 114–22. [DOI] [PubMed] [Google Scholar]
- 48.Fountzilas E, Lampaki S, Koliou GA, et al. Real-world safety and efficacy data of immunotherapy in patients with cancer and autoimmune disease: the experience of the Hellenic Cooperative Oncology Group. Cancer Immunol Immunother 2022; 71(2): 327–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Fredrick TW, Braga Neto MB, Ramos GP, Kane S, Pardi DS, Raffals LE. Letter: safety of immune checkpoint inhibitors in patients with pre-established microscopic colitis-a single-centre experience. Aliment Pharmacol Ther 2021; 54(2): 217–8. [DOI] [PubMed] [Google Scholar]
- 50.Garcia CR, Jayswal R, Adams V, Anthony LB, Villano JL. Multiple sclerosis outcomes after cancer immunotherapy. Clin Transl Oncol 2019; 21(10): 1336–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Gonzalez-Mazon I, Sanchez-Bilbao L, Martín-Varillas JL, et al. Immune-related adverse events in patients with solid-organ tumours treated with immunotherapy: a 3-year study of 102 cases from a single centre. Clin Exp Rheumatol 2021; 39(3): 612–20. [PubMed] [Google Scholar]
- 52.Grover S, Ruan A, Srivoleti P, et al. Safety of Immune Checkpoint Inhibitors in Patients with Preexisting Inflammatory Bowel Disease and Microscopic Colitis. Gastroenterology 2020; 158(6): S537–S. [DOI] [PubMed] [Google Scholar]
- 53.Grover S, Ruan AB, Srivoleti P, et al. Safety of Immune Checkpoint Inhibitors in Patients With Pre-Existing Inflammatory Bowel Disease and Microscopic Colitis. JCO Oncol Pract 2020; 16(9): e933–e42. [DOI] [PubMed] [Google Scholar]
- 54.Guaraldi F, La Selva R, Sama MT, et al. Characterization and implications of thyroid dysfunction induced by immune checkpoint inhibitors in real-life clinical practice: a long-term prospective study from a referral institution. J Endocrinol Invest 2018; 41(5): 549–56. [DOI] [PubMed] [Google Scholar]
- 55.Gulati N, Celen A, Johannet P, et al. Preexisting immune-mediated inflammatory disease is associated with improved survival and increased toxicity in melanoma patients who receive immune checkpoint inhibitors. Cancer Med 2021; 10(21): 7457–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Gutzmer R, Koop A, Meier F, et al. Programmed cell death protein-1 (PD-1) inhibitor therapy in patients with advanced melanoma and preexisting autoimmunity or ipilimumab-triggered autoimmunity. Eur J Cancer 2017; 75: 24–32. [DOI] [PubMed] [Google Scholar]
- 57.Han CY, Fitzgerald C, Lee M, et al. Association Between Toxic Effects and Survival in Patients With Cancer and Autoimmune Disease Treated With Checkpoint Inhibitor Immunotherapy. Jama Oncol 2022; 8(9): 1352–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Hayashi Y, Hosoe N, Takabayashi K, et al. Clinical, Endoscopic, and Pathological Characteristics of Immune Checkpoint Inhibitor-Induced Gastroenterocolitis. Dig Dis Sci 2021; 66(6): 2129–34. [DOI] [PubMed] [Google Scholar]
- 59.Hoa S, Laaouad L, Roberts J, et al. Preexisting autoimmune disease and immune-related adverse events associated with anti-PD-1 cancer immunotherapy: a national case series from the Canadian Research Group of Rheumatology in Immuno-Oncology. Cancer Immunol Immun 2021; 70(8): 2197–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Jamal S, Ye C, Fifi-Mah A, et al. Rheumatic Immune Related Adverse Events Associated with Cancer Immunotherapy: Experience from the Canadian Research Group of Rheumatology in Immuno-Oncology (Can-RIO). J Rheumatol 2019; 46(7): 822–3. [Google Scholar]
- 61.Johnson DB, Khushalani NI, Puzanov I, et al. Ipilimumab in metastatic melanoma patients with pre-existing autoimmune disorders. J Clin Oncol 2015; 33(15). [Google Scholar]
- 62.Johnson DB, Sullivan RJ, Ott PA, et al. Ipilimumab Therapy in Patients With Advanced Melanoma and Preexisting Autoimmune Disorders. Jama Oncol 2016; 2(2): 234–40. [DOI] [PubMed] [Google Scholar]
- 63.Kahler KC, Eigentler TK, Gesierich A, et al. Ipilimumab in metastatic melanoma patients with pre-existing autoimmune disorders. Cancer Immunol Immunother 2018; 67(5): 825–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Kartolo A, Sattar J, Sahai V, Baetz T, Lakoff JM. Predictors of immunotherapy-induced immune-related adverse events. Curr Oncol 2018; 25(5): e403–e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Kaur A, Doberstein T, Amberker RR, Garje R, Field EH, Singh N. Immune-related adverse events in cancer patients treated with immune checkpoint inhibitors: A single-center experience. Medicine (Baltimore) 2019; 98(41): e17348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Khan A, Martin H, Khattak M. Rheumatologic immune related adverse events (irAEs) secondary to immune checkpoint inhibitor (ICI) therapy: A Western Australia experience. Annals of Oncology 2020; 31: S1376–S. [Google Scholar]
- 67.Khozin S, Walker MS, Jun M, et al. Real-world outcomes of patients with advanced non-small cell lung cancer (aNSCLC) and autoimmune disease (AD) receiving immune checkpoint inhibitors (ICIs). J Clin Oncol 2019; 37(15). [Google Scholar]
- 68.Kim ST, Pundole X, Dadu R, Lambotte O, Ramos-Casals M, Suarez-Almazor ME. Use of immune checkpoint inhibitors in cancer patients with pre-existing sarcoidosis. Immunotherapy 2021; 13(6): 465–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Kimbara S, Fujiwara Y, Iwama S, et al. Association of antithyroglobulin antibodies with the development of thyroid dysfunction induced by nivolumab. Cancer Sci 2018; 109(11): 3583–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Kobayashi T, Iwama S, Yasuda Y, et al. Patients With Antithyroid Antibodies Are Prone To Develop Destructive Thyroiditis by Nivolumab: A Prospective Study. J Endocr Soc 2018; 2(3): 241–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Kostine M, Rouxel L, Barnetche T, et al. Rheumatic disorders associated with immune checkpoint inhibitors in patients with cancer-clinical aspects and relationship with tumour response: a single-centre prospective cohort study. Ann Rheum Dis 2018; 77(3): 393–8. [DOI] [PubMed] [Google Scholar]
- 72.Kotwal A, Kottschade L, Ryder M. PD-L1 Inhibitor-Induced Thyroiditis Is Associated with Better Overall Survival in Cancer Patients. Thyroid 2020; 30(2): 177–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Kwok TSH, Sweezie R, Bell MJ. A Clinical Profile of Patients on Immune Checkpoint Inhibitors Referred to Rheumatology. J Clin Rheumatol 2020; 26(6): 265–9. [DOI] [PubMed] [Google Scholar]
- 74.Laaouad L, Roberts J, Ennis D, et al. Preexisting Autoimmune Disease and Rheumatic Immune-Related Adverse Events Associated with Cancer Immunotherapy: A Case Series from the Canadian Research Group of Rheumatology in Immuno-Oncology (CanRIO). Arthritis Rheumatol 2019; 71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Ladouceur A, Arreola L, Ye C, et al. The Effect of Sex on Patients With Pre-Existing Rheumatoid Arthritis and Immune Checkpoint Inhibitor-Induced Inflammatory Arthritis: Data From the Canadian Research Group of Rheumatology in Immuno-Oncology (CanRIO) Prospective Cohort. J Rheumatol 2023; 50(7): 89–90. [Google Scholar]
- 76.Lau KS, Liu R, Wong CC, Siu WKS, Yuen KK. Clinical outcome and toxicity for immunotherapy treatment in metastatic cancer patients. Ann Palliat Med 2020; 9(6): 4446–57. [DOI] [PubMed] [Google Scholar]
- 77.Le Burel S, Champiat S, Routier E, et al. Onset of connective tissue disease following anti-PD1/PD-L1 cancer immunotherapy. Ann Rheum Dis 2018; 77(3): 468–70. [DOI] [PubMed] [Google Scholar]
- 78.Lee B, Wong A, Kee D, et al. The use of ipilimumab in patients with rheumatoid arthritis and metastatic melanoma. Ann Oncol 2016; 27(6): 1174–7. [DOI] [PubMed] [Google Scholar]
- 79.Lee C, Drobni ZD, Zafar A, et al. Pre-Existing Autoimmune Disease Increases the Risk of Cardiovascular and Noncardiovascular Events After Immunotherapy. JACC CardioOncol 2022; 4(5): 660–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Leonardi GC, Gainor JF, Altan M, et al. Safety of Programmed Death-1 Pathway Inhibitors Among Patients With Non-Small-Cell Lung Cancer and Preexisting Autoimmune Disorders. J Clin Oncol 2018; 36(19): 1905–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Lidar M, Giat E, Garelick D, et al. Rheumatic manifestations among cancer patients treated with immune checkpoint inhibitors. Autoimmunity Reviews 2018; 17(3): 284–9. [DOI] [PubMed] [Google Scholar]
- 82.Llano EM, Rubin DT, Luke J. Safety of Immune Checkpoint Inhibitors for Cancer Therapy in IBD Patients. Am J Gastroenterol 2018; 113: S406–S7. [Google Scholar]
- 83.Loriot Y, Sternberg CN, Castellano D, et al. Safety and efficacy of atezolizumab in patients with autoimmune disease: Subgroup analysis of the SAUL study in locally advanced/metastatic urinary tract carcinoma. Eur J Cancer 2020; 138: 202–11. [DOI] [PubMed] [Google Scholar]
- 84.Loriot Y, Sternberg CN, Gauna DC, et al. Safety and efficacy of atezolizumab (atezo) in patients (pts) with autoimmune disease (AID): Subgroup analysis of the SAUL study in locally advanced/metastatic urinary tract carcinoma. Annals of Oncology 2019; 30: 368-+. [Google Scholar]
- 85.Lusa A, Alvarez C, Saxena Beem S, Schwartz TA, Ishizawar R. Immune-related adverse events in patients with pre-existing autoimmune rheumatologic disease on immune checkpoint inhibitor therapy. BMC Rheumatol 2022; 6(1): 64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Matsuoka H, Hayashi T, Takigami K, et al. Correlation between immune-related adverse events and prognosis in patients with various cancers treated with anti PD-1 antibody. Bmc Cancer 2020; 20: 656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Melia A, Fockens E, Sfumato P, et al. Musculoskeletal immune-related adverse events in 927 patients treated with immune checkpoint inhibitors for solid cancer. Joint Bone Spine 2023; 90(1): 105457. [DOI] [PubMed] [Google Scholar]
- 88.Mirza S, Haque F, Hodgson S, Lind M, Maraveyas A, Brown V. Real-world experience of immunotherapy in elderly cancer patients in a UK cancer centre. Annals of Oncology 2020; 31: S1439–S40. [Google Scholar]
- 89.Mitchell E, Lau P, Khoo C, et al. Rheumatological Immune Related Adverse Events in Malignancy Patients Treated with Anti-Programmed Cell Death (PD) 1 Antibodies. Arthritis Rheumatol 2017; 69. [Google Scholar]
- 90.Mitchell EL, Lau PKH, Khoo C, et al. Rheumatic immune-related adverse events secondary to anti-programmed death-1 antibodies and preliminary analysis on the impact of corticosteroids on anti-tumour response: A case series. Eur J Cancer 2018; 105: 88–102. [DOI] [PubMed] [Google Scholar]
- 91.Mooradian MJ, Nasrallah M, Gainor JF, et al. Musculoskeletal rheumatic complications of immune checkpoint inhibitor therapy: A single center experience. Semin Arthritis Rheum 2019; 48(6): 1127–32. [DOI] [PubMed] [Google Scholar]
- 92.Naidoo J, Cappelli L, Lipson EJ, et al. A multidisciplinary toxicity team for cancer immunotherapy-related adverse events. J Clin Oncol 2018; 36(15). [DOI] [PubMed] [Google Scholar]
- 93.Naidoo J, Zhang J, Lipson EJ, et al. A Multidisciplinary Toxicity Team for Cancer Immunotherapy-Related Adverse Events. J Natl Compr Canc Netw 2019; 17(6): 712–20. [DOI] [PubMed] [Google Scholar]
- 94.Nardin C, Jeand’heur A, Bouiller K, et al. Vitiligo under anti-programmed cell death-1 therapy is associated with increased survival in melanoma patients. Journal of the American Academy of Dermatology 2020; 82(3): 770–2. [DOI] [PubMed] [Google Scholar]
- 95.Ngo T, Hossain C, Guzman AK, Halmos B, Balagula Y, McLellan B. Spectrum of PD-1 and PD-L1 inhibitor cutaneous adverse events in skin of color: a retrospective, single-institutional study in an urban community. Acta Oncol 2021; 60(4): 559–63. [DOI] [PubMed] [Google Scholar]
- 96.Olsson-Brown AC, Baxter M, Dobeson C, et al. Real-world outcomes of immune-related adverse events in 2,125 patients managed with immunotherapy: A United Kingdom multicenter series. J Clin Oncol 2020; 38(15). [Google Scholar]
- 97.Pan EY, Merl MY, Lin K. The impact of corticosteroid use during anti-PD1 treatment. Journal of Oncology Pharmacy Practice 2020; 26(4): 814–22. [DOI] [PubMed] [Google Scholar]
- 98.Panhaleux M, Espitia O, Terrier B, et al. Anti-programmed death ligand 1 immunotherapies in cancer patients with pre-existing systemic sclerosis: A postmarketed phase IV safety assessment study. Eur J Cancer 2022; 160: 134–9. [DOI] [PubMed] [Google Scholar]
- 99.Patel G, Sawhney P, Ohana D, et al. Safety and efficacy of immune checkpoint inhibitors in cancer patients with pre-existing autoimmune disease: A UK tertiary cancer centre experience. Annals of Oncology 2021; 32: S12–S3. [Google Scholar]
- 100.Patwari A, Bhatlapenumarthi V, Harb AJ, Curtis A. mmune related adverse effects (IRAEs) in patients with cancer who received immune checkpoint inhibitors (ICIs): A single center experience from rural Maine. J Clin Oncol 2020; 38(15). [Google Scholar]
- 101.Pease M, Shyne S, Joshi A, Lee M, Patel. Effectiveness and tolerance of immune checkpoint blockade in a real-world lung cancer patient population. Journal for ImmunoTherapy of Cancer 2018; 6: Conference: 33rd Annual Meeting and Pre Conference Programs of the Society for Immunotherapy of Cancer. [Google Scholar]
- 102.Pizuorno Machado A, Shatila M, Liu C, et al. Immune-related adverse events after immune checkpoint inhibitor exposure in adult cancer patients with pre-existing autoimmune diseases. J Cancer Res Clin Oncol 2023; 149(9): 6341–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Placais L, Dalle S, Dereure O, et al. Risk of irAEs in patients with autoimmune diseases treated by immune checkpoint inhibitors for stage III or IV melanoma: results from a matched case-control study. Ann Rheum Dis 2022; 81(10): 1445–52. [DOI] [PubMed] [Google Scholar]
- 104.Pundole X, Lambotte O, Ramos-Casals M, Suarez-Almazor M. Immune Checkpoint Inhibitor Therapy in Patients with Preexisting Sarcoidosis. Ann Rheum Dis 2020; 79: 1221–2. [Google Scholar]
- 105.Richter MD, Crowson C, Kottschade LA, Finnes HD, Markovic SN, Thanarajasingam U. Rheumatic Syndromes Associated With Immune Checkpoint Inhibitors: A Single-Center Cohort of Sixty-One Patients. Arthritis Rheumatol 2019; 71(3): 468–75. [DOI] [PubMed] [Google Scholar]
- 106.Richter MD, Pinkston O, Kottschade LA, Finnes HD, Markovic SN, Thanarajasingam U. Brief Report: Cancer Immunotherapy in Patients With Preexisting Rheumatic Disease: The Mayo Clinic Experience. Arthritis Rheumatol 2018; 70(3): 356–60. [DOI] [PubMed] [Google Scholar]
- 107.Roberts J, Ennis D, Hudson M, et al. Rheumatic Immune Related Adverse Events Associated with Cancer Immunotherapy: A Nationwide Multi-centre Canadian Cohort from the Canadian Research Group of Rheumatology in Immuno-oncology (CanRIO). Arthritis Rheumatol 2019; 71. [Google Scholar]
- 108.Romao R, Mendes AS, Ranchor R, et al. Impact of Immune-Related Adverse Events on Immune Checkpoint Inhibitors Treated Cancer Patients’ Survival: Single Center Experience and Literature Review. Cancers (Basel) 2023; 15(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Rubin SJS, Balabanis T, Gubatan J, Habtezion A. Disease exacerbation is common in inflammatory bowel disease patients treated with immune checkpoint inhibitors for malignancy. World J Clin Cases 2022; 10(6): 1787–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Saberian C, Fa’ak F, Tayar J, et al. Interleukin-6 Receptor Blockade for Management of Immune Checkpoint Inhibitor Related Adverse Events in Patients with Melanoma. Journal for Immunotherapy of Cancer 2020; 8: A394–A. [Google Scholar]
- 111.Safa H, Johnson DH, Trinh VA, et al. Immune checkpoint inhibitor related myasthenia gravis: single center experience and systematic review of the literature. Journal for Immunotherapy of Cancer 2019; 7(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Sakakida T, Ishikawa T, Chihara Y, et al. Safety and efficacy of PD-1/PD-L1 blockade in patients with preexisting antinuclear antibodies. Clin Transl Oncol 2020; 22(6): 919–27. [DOI] [PubMed] [Google Scholar]
- 113.Sakakida T, Ishikawa T, Chihara Y, et al. Safety of PD-1/PD-L1 blockade in patients with preexisting positive antinuclear antibodies and autoimmune disease. Annals of Oncology 2019; 30. [Google Scholar]
- 114.Sandigursky S, Houssein S, Pundole X, et al. Toxicities of Single Agent and Combination Immune Checkpoint Inhibitors in Patients with Pre-Existing Autoimmune Diseases. Journal for Immunotherapy of Cancer 2020; 8: A395–A. [Google Scholar]
- 115.Schadendorf D, Ascierto PA, Haanen J, et al. Safety and efficacy of nivolumab in challenging subgroups with advanced melanoma who progressed on or after ipilimumab treatment: A single-arm, open-label, phase II study (CheckMate 172). Eur J Cancer 2019; 121: 144–53. [DOI] [PubMed] [Google Scholar]
- 116.Sechi E, Markovic SN, McKeon A, et al. Neurologic autoimmunity and immune checkpoint inhibitors: Autoantibody profiles and outcomes. Neurology 2020; 95(17): e2442–e52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Shah M, Jizzini MN, Majzoub IE, Qdaisat A, Reyes-Gibby CC, Yeung S-CJ. Safety of Immune Checkpoint Blockade in Patients with Cancer and Preexisting Autoimmune Diseases and/or Chronic Inflammatory Disorders. Journal of Immunotherapy and Precision Oncology 2019; 2(3): 59–64. [Google Scholar]
- 118.Shah MN, Jizzini I, El Majzoub A, Qdaisat CC, Reyes SCJ, Yeung SJ. Safety of cancer patients with preexisting autoimmune diseases presenting in the emergency center following immune checkpoint blockade. Journal for ImmunoTherapy of Cancer 2018; 6: Conference: 33rd Annual Meeting and Pre Conference Programs of the Society for Immunotherapy of Cancer. [Google Scholar]
- 119.Snavely A, Perez-Torres EJ, Weber JS, Sandigursky S, Thawani SP. Immune checkpoint inhibition in patients with inactive pre-existing neuromuscular autoimmune diseases. J Neurol Sci 2022; 438: 120275. [DOI] [PubMed] [Google Scholar]
- 120.So AC, Board RE. Real-world experience with pembrolizumab toxicities in advanced melanoma patients: a single-center experience in the UK. Melanoma Manag 2018; 5(1): MMT05. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Sternberg CN, Loriot Y, James N, et al. Primary Results from SAUL, a Multinational Single-arm Safety Study of Atezolizumab Therapy for Locally Advanced or Metastatic Urothelial or Nonurothelial Carcinoma of the Urinary Tract. Eur Urol 2019; 76(1): 73–81. [DOI] [PubMed] [Google Scholar]
- 122.Tang K, Tiu BC, Wan G, et al. Pre-Existing Autoimmune Disease and Mortality in Patients Treated with Anti-PD-1 and Anti-PD-L1 Therapy. J Natl Cancer Inst 2022; 114(8): 1200–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Tison A, Quere G, Misery L, et al. Safety and Efficacy of Immune Checkpoint Inhibitors in Patients with Cancer and Preexisting Autoimmune Diseases: A Nationwide Multicenter Retrospective Study. Ann Rheum Dis 2018; 77: 147. [Google Scholar]
- 124.Tison A, Quere G, Misery L, et al. Safety and Efficacy of Immune Checkpoint Inhibitors in Patients With Cancer and Preexisting Autoimmune Disease: A Nationwide, Multicenter Cohort Study. Arthritis Rheumatol 2019; 71(12): 2100–11. [DOI] [PubMed] [Google Scholar]
- 125.Tison A, Quere G, Misery L, et al. Safety of Immune Checkpoint Inhibitors for the Treatment of Melanoma, Bronchopulmonary and Urologic Neoplasms in Patients with Preexisting Autoimmune Disease. Arthritis Rheumatol 2017; 69. [Google Scholar]
- 126.Tully KH, Cone EB, Cole AP, et al. Risk of Immune-related Adverse Events in Melanoma Patients With Preexisting Autoimmune Disease Treated With Immune Checkpoint Inhibitors: A Population-based Study Using SEER-Medicare Data. Am J Clin Oncol 2021; 44(8): 413–8. [DOI] [PubMed] [Google Scholar]
- 127.van der Kooij MK, Suijkerbuijk KPM, Aarts MJB, et al. Safety and Efficacy of Checkpoint Inhibition in Patients With Melanoma and Preexisting Autoimmune Disease A Cohort Study. Annals of Internal Medicine 2021; 174(5): 641-+. [DOI] [PubMed] [Google Scholar]
- 128.Verspohl S, Holderried T, Behning C, Brossart P, Schaefer V. Prevalence, Therapy and Tumor Response in Patients with Rheumatic Immune-related Adverse Events Following Immune Checkpoint Inhibitor Therapy: A Single-Centre Analysis. Arthritis Rheumatol 2020; 72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Verspohl SH, Holderried T, Behning C, Brossart P, Schafer VS. Prevalence, therapy and tumour response in patients with rheumatic immune-related adverse events following immune checkpoint inhibitor therapy: a single-centre analysis. Ther Adv Musculoskelet Dis 2021; 13: 1759720X211006963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Wan G, Nguyen N, Leung BW, et al. Pre-existing inflammatory disease predicts cutaneous immunotherapy toxicity development: A multi-institutional cohort study. J Am Acad Dermatol 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Wan G, Nguyen N, Leung BW, et al. Pre-Existing Inflammatory Disease Predicts Cutaneous Immunotherapy Toxicity Development: A Multi-Institutional Cohort Study. medRxiv 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Warner AB, Rotemberg V. Safety and efficacy of immune checkpoint inhibitors (ICI) in patients with autoimmune conditions. Pigment Cell and Melanoma Research 2020; 155(33 (1)). [Google Scholar]
- 133.Warner BM, Baer AN, Lipson EJ, et al. Sicca Syndrome Associated with Immune Checkpoint Inhibitor Therapy. Oncologist 2019; 24(9): 1259–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Weinstock C, Singh H, Maher VE, Kim G, Pazdur R. FDA analysis of patients with baseline autoimmune diseases treated with PD-1/PD-L1 immunotherapy agents. J Clin Oncol 2017; 35. [Google Scholar]
- 135.Weppler AM, Pattison A, Bhave P, et al. Clinical, FDG-PET and molecular markers of immune checkpoint inhibitor response in patients with metastatic Merkel cell carcinoma. J Immunother Cancer 2020; 8(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Yano S, Ashida K, Nagata H, et al. Nivolumab-induced thyroid dysfunction lacking antithyroid antibody is frequently evoked in Japanese patients with malignant melanoma. Bmc Endocr Disord 2018; 18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Yeung C, Kartolo A, Holstead R, et al. Safety and Clinical Outcomes of Immune Checkpoint Inhibitors in Patients With Cancer and Preexisting Autoimmune Diseases. J Immunother 2021; 44(9): 362–70. [DOI] [PubMed] [Google Scholar]
- 138.McCarter KR, Wolfgang T, Arabelovic S, et al. Mortality and immune-related adverse events after immune checkpoint inhibitor initiation for cancer among patients with pre-existing rheumatoid arthritis: a retrospective, comparative, cohort study. Lancet Rheumatol 2023; 5(5): E274–E83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Reid P, Sandigursky S, Song JH, et al. Safety and effectiveness of combination versus monotherapy with immune checkpoint inhibitors in patients with preexisting autoimmune diseases. Oncoimmunology 2023; 12(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Danlos FX, Voisin AL, Dyevre V, et al. Safety and efficacy of anti-programmed death 1 antibodies in patients with cancer and pre-existing autoimmune or inflammatory disease. Eur J Cancer 2018; 91: 21–9. [DOI] [PubMed] [Google Scholar]
- 141.Antonuzzo A, Calabro F, Quaglino P, et al. Immunotherapy in Underrepresented Populations of Patients with Cancer: Do We Have Enough Evidence at Present? A Focus on Patients with Major Viral Infections and Autoimmune Disorders. Oncologist 2020; 25(6): e946–e54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Aoun R, Gratch D, Kaminetzky D, Kister I. Immune Checkpoint Inhibitors in Patients with Pre-existing Neurologic Autoimmune Disorders. Curr Neurol Neurosci Rep 2023. [DOI] [PubMed] [Google Scholar]
- 143.Aung WY, Lee CS, Morales J, Rahman H, Seetharamu N. Safety and Efficacy of Immune Checkpoint Inhibitors in Cancer Patients and Preexisting Autoimmune Disease: A Systematic Review and Meta-Analysis in Non-Small-Cell Lung Cancer. Clin Lung Cancer 2023; 24(7): 598–612. [DOI] [PubMed] [Google Scholar]
- 144.Cai Q, Huo GW, Zhu FY, Yue P, Yuan DQ, Chen P. Safety and efficacy of immune checkpoint inhibitors in advanced cancer patients with autoimmune disease: A meta-analysis. Hum Vaccin Immunother 2022; 18(7): 2145102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Cappelli LC, Gutierrez AK, Bingham CO 3rd, Shah AA. Rheumatic and Musculoskeletal Immune-Related Adverse Events Due to Immune Checkpoint Inhibitors: A Systematic Review of the Literature. Arthritis Care Res (Hoboken) 2017; 69(11): 1751–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Coureau M, Meert AP, Berghmans T, Grigoriu B. Efficacy and Toxicity of Immune - Checkpoint Inhibitors in Patients With Preexisting Autoimmune Disorders. Front Med (Lausanne) 2020; 7: 137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Dietz H, Weinmann SC, Salama AK. Checkpoint Inhibitors in Melanoma Patients with Underlying Autoimmune Disease. Cancer Manag Res 2021; 13: 8199–208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Donia M, Pedersen M, Svane IM. Cancer immunotherapy in patients with preexisting autoimmune disorders. Semin Immunopathol 2017; 39(3): 333–7. [DOI] [PubMed] [Google Scholar]
- 149.Haanen J, Ernstoff MS, Wang Y, et al. Autoimmune diseases and immune-checkpoint inhibitors for cancer therapy: review of the literature and personalized risk-based prevention strategy. Ann Oncol 2020; 31(6): 724–44. [DOI] [PubMed] [Google Scholar]
- 150.Johnson DB, Beckermann KE, Wang DY. Immune Checkpoint Inhibitor Therapy in Patients With Autoimmune Disease. Oncology-Ny 2018; 32(4): 190–4. [PubMed] [Google Scholar]
- 151.Kennedy LC, Bhatia S, Thompson JA, Grivas P. Preexisting Autoimmune Disease: Implications for Immune Checkpoint Inhibitor Therapy in Solid Tumors. J Natl Compr Canc Netw 2019; 17(6): 750–7. [DOI] [PubMed] [Google Scholar]
- 152.Klavdianou K, Melissaropoulos K, Filippopoulou A, Daoussis D. Should we be Afraid of Immune Check Point Inhibitors in Cancer Patients with Pre-Existing Rheumatic Diseases? Immunotherapy in Pre-Existing Rheumatic Diseases. Mediterr J Rheumatol 2021; 32(3): 218–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Liu X, Shi Y, Zhang D, et al. Risk factors for immune-related adverse events: what have we learned and what lies ahead? Biomark Res 2021; 9(1): 79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Meserve J, Facciorusso A, Holmer AK, Annese V, Sandborn WJ, Singh S. Systematic review with meta-analysis: safety and tolerability of immune checkpoint inhibitors in patients with pre-existing inflammatory bowel diseases. Aliment Pharmacol Ther 2021; 53(3): 374–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Passiglia F, Cetoretta V, De Filippis M, Napoli V, Novello S. Exploring the immune-checkpoint inhibitors’ efficacy/tolerability in special non-small cell lung cancer (NSCLC) populations: focus on steroids and autoimmune disease. Transl Lung Cancer Res 2021; 10(6): 2876–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Rakshit S, Molina JR. Immunotherapy in patients with autoimmune disease. J Thorac Dis 2020; 12(11): 7032–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Sebastiani GD, Scirocco C, Galeazzi M. Rheumatic immune related adverse events in patients treated with checkpoint inhibitors for immunotherapy of cancer. Autoimmun Rev 2019; 18(8): 805–13. [DOI] [PubMed] [Google Scholar]
- 158.Shan Q, Lu H. Immune Checkpoint Inhibitors in Special Populations. Technol Cancer Res Treat 2021; 20: 15330338211036526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Tang H, Zhou J, Bai C. The Efficacy and Safety of Immune Checkpoint Inhibitors in Patients With Cancer and Preexisting Autoimmune Disease. Front Oncol 2021; 11: 625872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Wu C, Zhong L, Wu Q, Lin S, Xie X. The safety and efficacy of immune-checkpoint inhibitors in patients with cancer and pre-existing autoimmune diseases. Immunotherapy 2021; 13(6): 527–39. [DOI] [PubMed] [Google Scholar]
- 161.Xie W, Huang H, Xiao S, Fan Y, Deng X, Zhang Z. Immune checkpoint inhibitors therapies in patients with cancer and preexisting autoimmune diseases: A meta-analysis of observational studies. Autoimmun Rev 2020; 19(12): 102687. [DOI] [PubMed] [Google Scholar]
- 162.Yamaguchi A, Saito Y, Okamoto K, et al. Preexisting autoimmune disease is a risk factor for immune-related adverse events: a meta-analysis. Support Care Cancer 2021; 29(12): 7747–53. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.