361
Views
4
CrossRef citations to date
0
Altmetric
REVIEW

Monitoring and Management of the Patient with Immune Checkpoint Inhibitor-Induced Inflammatory Arthritis: Current Perspectives

& ORCID Icon
Pages 3105-3118 | Published online: 25 May 2022

References

  • Brahmer JR, Tykodi SS, Chow LQM, et al. Safety and activity of anti–PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366(26):2455–2465. doi:10.1056/NEJMoa1200694
  • Wolchok JD, Chiarion-Sileni V, Gonzalez R, et al. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med. 2017;377(14):1345–1356. doi:10.1056/NEJMoa1709684
  • Motzer RJ, Tannir NM, McDermott DF, et al. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med. 2018;378(14):1277–1290. doi:10.1056/NEJMoa1712126
  • Hellmann MD, Paz-Ares L, Bernabe Caro R, et al. Nivolumab plus ipilimumab in advanced non–small-cell lung cancer. N Engl J Med. 2019;381:2020–2031. doi:10.1056/nejmoa1910231
  • Eggermont AMM, Blank CU, Mandala M, et al. Adjuvant pembrolizumab versus placebo in resected stage III melanoma. N Engl J Med. 2018;378(19):1789–1801. doi:10.1056/NEJMOA1802357
  • Cortes J, Cescon DW, Rugo HS, et al. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): a randomised, placebo-controlled, double-blind, phase 3 clinical trial. Lancet. 2020;396(10265):1817–1828. doi:10.1016/S0140-6736(20)32531-9
  • Marcus L, Lemery SJ, Keegan P, Pazdur R. FDA approval summary: pembrolizumab for the treatment of microsatellite instability-high solid tumors. Clin Cancer Res. 2019;25(13):3753–3758. doi:10.1158/1078-0432.CCR-18-4070
  • André T, Shiu -K-K, Kim TW, et al. Pembrolizumab in microsatellite-instability-high advanced colorectal cancer. N Engl J Med. 2020;383(23):2207–2218. doi:10.1056/NEJMOA2017699
  • Bagchi S, Yuan R, Engleman EG. Immune checkpoint inhibitors for the treatment of cancer: clinical impact and mechanisms of response and resistance. Annu Rev Pathol. 2021;16:223–249. doi:10.1146/ANNUREV-PATHOL-042020-042741
  • Postow MA, Sidlow R, Hellmann MD. Immune-related adverse events associated with immune checkpoint blockade. N Engl J Med. 2018;378(2):158–168. doi:10.1056/NEJMra1703481
  • Wang R, Singaraju A, Marks KE, et al. Clonally expanded CD38. 2021.
  • Chan KK, Tirpack A, Vitone G, et al. Higher checkpoint inhibitor arthritis disease activity may be associated with cancer progression: results from an observational registry. ACR Open Rheumatol. 2020;2(10):595–604. doi:10.1002/ACR2.11181
  • Braaten TJ, Brahmer JR, Forde PM, et al. Immune checkpoint inhibitor-induced inflammatory arthritis persists after immunotherapy cessation. Ann Rheum Dis. 2019;79:332–338. doi:10.1136/annrheumdis-2019-216109
  • Cappelli LC, Gutierrez AK, Bingham CO, Shah AA. Rheumatic and musculoskeletal immune-related adverse events due to immune checkpoint inhibitors: a systematic review of the literature. Arthritis Care Res. 2017;69(11):1751–1763. doi:10.1002/ACR.23177
  • Arnaud-Coffin P, Maillet D, Gan HK, et al. A systematic review of adverse events in randomized trials assessing immune checkpoint inhibitors. Int J Cancer. 2019;145(3):639–648. doi:10.1002/IJC.32132
  • Richter MD, Crowson C, Kottschade LA, 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. Ann Rheum Dis. 2019;17(3):1–10. doi:10.1007/s00262-017-2069-9
  • Cunningham-Bussel A, Wang J, Prisco LC, et al. Predictors of rheumatic immune-related adverse events and de novo inflammatory arthritis after immune checkpoint inhibitor treatment for cancer. Arthritis Rheumatol. 2022;74(3):527–540. doi:10.1002/ART.41949/ABSTRACT
  • Cappelli LC, Dorak MT, Bettinotti MP, Bingham CO, Shah AA. Association of HLA-DRB1 shared epitope alleles and immune checkpoint inhibitor-induced inflammatory arthritis. Rheumatology. 2019;58(3):476–478. doi:10.1093/RHEUMATOLOGY/KEY358
  • Ghosh N, Tiongson MD, Stewart C, et al. Checkpoint inhibitor-associated arthritis: a systematic review of case reports and case series. J Clin Rheumatol. 2021;27(8):E317–E322. doi:10.1097/RHU.0000000000001370
  • Calabrese C, Cappelli LC, Kostine M, Kirchner E, Braaten T, Calabrese L. Polymyalgia rheumatica-like syndrome from checkpoint inhibitor therapy: case series and systematic review of the literature. RMD Open. 2019;5(1):e000906. doi:10.1136/rmdopen-2019-000906
  • Martin De Fremont G, Belkhir R, Henry J, et al. Features of polymyalgia rheumatica-like syndrome after immune checkpoint inhibitor therapy. Ann Rheum Dis. 2022;81(3):E52. doi:10.1136/annrheumdis-2020-217225
  • Reid P, Liew DFL, Akruwala R, Bass AR, Chan KK. Activated osteoarthritis following immune checkpoint inhibitor treatment: an observational study. J Immunother Cancer. 2021;9(9). doi:10.1136/JITC-2021-003260
  • National Cancer Institute. Common terminology criteria for adverse events (CTCAE) common terminology criteria for adverse events (CTCAE) v5.0; 2017. Available from: https://www.meddra.org/. Accessed January 15, 2022.
  • Anderson J, Caplan L, Yazdany J, et al. Rheumatoid arthritis disease activity measures: American College of Rheumatology recommendations for use in clinical practice. Arthritis Care Res. 2012;64(5):640–647. doi:10.1002/ACR.21649
  • Bruce B, Fries JF. The Stanford health assessment questionnaire: dimensions and practical applications. Health Qual Life Outcomes. 2003;1:20. doi:10.1186/1477-7525-1-20
  • Ghosh N, Chan KK, Jivanelli B, Bass AR, Ayaz NA. Autoantibodies in patients with immune-related adverse events from checkpoint inhibitors. J Clin Rheumatol. 2021;27:1–8. doi:10.1097/rhu.0000000000001777
  • De Moel EC, Rozeman EA, Kapiteijn EH, et al. Autoantibody development under treatment with immune-checkpoint inhibitors. Cancer Immunol Res. 2019;7(1):6–11. doi:10.1158/2326-6066.CIR-18-0245
  • Cappelli LC, Darrah E, Shah AA, Bingham CO. Patients with checkpoint inhibitor‐induced inflammatory arthritis do not become seropositive for anti‐cyclic citrullinated peptide when followed over time. ACR Open Rheumatol. 2022;4(1):83–84. doi:10.1002/acr2.11363
  • Subedi A, Williams SG, Yao L, et al. Use of magnetic resonance imaging to identify immune checkpoint inhibitor-induced inflammatory arthritis. JAMA Netw open. 2020;3(2):e200032. doi:10.1001/JAMANETWORKOPEN.2020.0032
  • Cappelli LC, Gutierrez AK, Baer AN, et al. Inflammatory arthritis and sicca syndrome induced by nivolumab and ipilimumab. Ann Rheum Dis. 2017;76(1):43–50. doi:10.1136/annrheumdis-2016-209595
  • Van Der Geest KSM, Sandovici M, Rutgers A, et al. Imaging in immune checkpoint inhibitor-induced polymyalgia rheumatica. Ann Rheum Dis. 2020;1–2. doi:10.1136/annrheumdis-2020-217381
  • Medina H, Eickhoff J, Edison J. Thinking inside the box. J Clin Rheumatol. 2019. doi:10.1097/RHU.0000000000001088
  • Murray-Brown W, Wilsdon TD, Weedon H, et al. Nivolumab-induced synovitis is characterized by florid T cell infiltration and rapid resolution with synovial biopsy-guided therapy. J Immunother Cancer. 2020;8(1):1–6. doi:10.1136/jitc-2019-000281
  • Wang R, Singaraju A, Marks KE, et al. Clonally expanded CD38hi cytotoxic CD8 T cells define the T cell infiltrate in checkpoint inhibitor-associated arthritis. bioRxiv. 2021. doi:10.1101/2021.10.19.464961
  • Zaretsky JM, Garcia-Diaz A, Shin DS, et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med. 2016;375(9):819–829. doi:10.1056/NEJMOA1604958/SUPPL_FILE/NEJMOA1604958_DISCLOSURES.PDF
  • Murray K, Floudas A, Murray C, et al. First use of tofacitinib to treat an immune checkpoint inhibitor-induced arthritis. BMJ Case Rep CP. 2021;14(2):e238851. doi:10.1136/BCR-2020-238851
  • Belkhir R, Le Burel S, Dunogeant L, et al. Rheumatoid arthritis and polymyalgia rheumatica occurring after immune checkpoint inhibitor treatment. Ann Rheum Dis. 2017;76(10):1747–1750. doi:10.1136/annrheumdis-2017-211216
  • Calabrese C, Kirchner E, Kontzias K, 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:10.1136/rmdopen-2016-000412
  • Leipe J, Christ LA, Arnoldi AP, et al. Characteristics and treatment of new-onset arthritis after checkpoint inhibitor therapy. RMD Open. 2018;4(2):1–7. doi:10.1136/rmdopen-2018-000714
  • Roberts J, Smylie M, Walker J, et al. Hydroxychloroquine is a safe and effective steroid-sparing agent for immune checkpoint inhibitor–induced inflammatory arthritis. Clin Rheumatol. 2019;38(5):1513–1519. doi:10.1007/s10067-019-04451-2
  • Smith MH, Bass AR. Arthritis after cancer immunotherapy: symptom duration and treatment response. Arthritis Care Res. 2017. doi:10.1002/acr.23467
  • Lee J, Graham A, Sion A. Evaluation of arthralgias in adult oncology patients receiving immune checkpoint inhibitors. J Oncol Pharm Pract. 2019;25(8):1867–1872. doi:10.1177/1078155218822707
  • Lidar M, Giat E, Garelick D, et al. Rheumatic manifestations among cancer patients treated with immune checkpoint inhibitors. Autoimmun Rev. 2018;17(3):284–289. doi:10.1016/j.autrev.2018.01.003
  • 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):1127–1132. doi:10.1016/j.semarthrit.2018.10.012
  • Verspohl SH, Holderried T, Behning C, Brossart P, Schäfer 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;13:1–14. doi:10.1177/1759720X211006963
  • 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(2018):88–102. doi:10.1016/j.ejca.2018.09.027
  • Van Der Geest KSM, Sandovici M, Rutgers A, et al. Management of immune checkpoint inhibitor-induced polymyalgia rheumatica. Ann Rheum Dis. 2020;1–2. doi:10.1136/annrheumdis-2020-218276
  • Ceccarelli F, Botticelli A, Gelibter AJ, et al. Correspondence on “Immune checkpoint inhibitor-induced inflammatory arthritis persists after immunotherapy cessation” by Braaten et al. Ann Rheum Dis. 2022;81(1):1–2. doi:10.1136/annrheumdis-2019-216867
  • Buder-Bakhaya K, Benesova K, Schulz C, et al. Characterization of arthralgia induced by PD-1 antibody treatment in patients with metastasized cutaneous malignancies. Cancer Immunol Immunother. 2018;67(2):175–182. doi:10.1007/s00262-017-2069-9
  • Cappelli LC, Brahmer JR, Forde PM, et al. Clinical presentation of immune checkpoint inhibitor-induced inflammatory arthritis differs by immunotherapy regimen. Semin Arthritis Rheum. 2018;48:1–5. doi:10.1016/j.semarthrit.2018.02.011
  • Roberts J, Ennis D, Hudson M, et al. Rheumatic immune-related adverse events associated with cancer immunotherapy: a nationwide multi-center cohort. Autoimmun Rev. 2020;19(8):102595. doi:10.1016/j.autrev.2020.102595
  • 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):393–398. doi:10.1136/annrheumdis-2017-212257
  • Liu Y, Jaquith JM, Mccarthy-Fruin K, et al. Immune checkpoint inhibitor-induced inflammatory arthritis: a novel clinical entity with striking similarities to seronegative rheumatoid arthritis. Clin Rheumatol. 2020;39(12):3631–3637. doi:10.1007/s10067-020-05162-9
  • Ornstein MC, Calabrese C, Wood LS, et al. Myalgia and arthralgia immune-related adverse events (irAEs) in patients with genitourinary malignancies treated with immune checkpoint inhibitors. Clin Genitourin Cancer. 2019;17(3):177–182. doi:10.1016/j.clgc.2019.01.021
  • 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):468–475. doi:10.1002/art.40745
  • Haanen JB, Carbonnel F, Robert C, et al. Management of toxicities from immunotherapy: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2017;28(Supplement 4):iv119–iv142. doi:10.1093/annonc/mdx225
  • Schneider BJ, Naidoo J, Santomasso BD, et al. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: ASCO guideline update. J Clin Oncol. 2021;39:4073–4126. doi:10.1200/JCO.21.01440
  • Thompson JA, Schneider BJ, Brahmer J, et al. NCCN guidelines version 4.2021 management of immunotherapy-related toxicities NCCN guidelines panel disclosures continue NCCN; 2021. Available from: https://www.nccn.org/home/member-. Accessed December 18, 2021.
  • Brahmer JR, Abu-Sbeih H, Ascierto PA, et al. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immune checkpoint inhibitor-related adverse events. J Immunother Cancer. 2021;9(6):e002435. doi:10.1136/JITC-2021-002435
  • Kostine M, Finckh A, Bingham CO, et al. EULAR points to consider for the diagnosis and management of rheumatic immune-related adverse events due to cancer immunotherapy with checkpoint inhibitors. Ann Rheum Dis. 2021;80(1):36–48. doi:10.1136/ANNRHEUMDIS-2020-217139
  • Schrezenmeier E, Dörner T. Mechanisms of action of hydroxychloroquine and chloroquine: implications for rheumatology. Nat Rev Rheumatol. 2020;16(3):155–166. doi:10.1038/S41584-020-0372-X
  • Choi J, Fenando A. Sulfasalazine. Intraocular Inflamm. 2021;373–377. doi:10.1007/978-3-540-75387-2_30
  • Gerriets V, Bansal P, Goyal A, Khaddour K. Tumor necrosis factor inhibitors. StatPearls; 2021. Available from: https://www.ncbi.nlm.nih.gov/books/NBK482425/. Accessed January 15, 2022.
  • Tanaka T, Narazaki M, Kishimoto T. IL-6 in inflammation, immunity, and disease. Cold Spring Harb Perspect Biol. 2014;6(10):16295–16296. doi:10.1101/CSHPERSPECT.A016295
  • Faje AT, Lawrence D, Flaherty K, et al. High-dose glucocorticoids for the treatment of ipilimumab-induced hypophysitis is associated with reduced survival in patients with melanoma. Cancer. 2018;124(18):3706–3714. doi:10.1002/cncr.31629
  • Maslov DV, Tawagi K, Kc M, et al. Timing of steroid initiation and response rates to immune checkpoint inhibitors in metastatic cancer. J Immunother Cancer. 2021;9(7):e002261. doi:10.1136/JITC-2020-002261
  • Wang Y, Abu-Sbeih H, Mao E, et al. Immune-checkpoint inhibitor-induced diarrhea and colitis in patients with advanced malignancies: retrospective review at MD Anderson. J Immunother Cancer. 2018;6(1). doi:10.1186/s40425-018-0346-6
  • Krueger J, Santinon F, Kazanova A, et al. Hydroxychloroquine (HCQ) decreases the benefit of anti-PD-1 immune checkpoint blockade in tumor immunotherapy. PLoS One. 2021;16(6):e0251731. doi:10.1371/JOURNAL.PONE.0251731
  • Chen AY, Wolchok JD, Bass AR. TNF in the era of immune checkpoint inhibitors: friend or foe? Nat Rev Rheumatol. 2021;17(4):213–223. doi:10.1038/s41584-021-00584-4
  • Johnson DH, Zobniw CM, Trinh VA, et al. Infliximab associated with faster symptom resolution compared with corticosteroids alone for the management of immune-related enterocolitis. J Immunother Cancer. 2018;6(1). doi:10.1186/s40425-018-0412-0
  • Verheijden RJ, May AM, Blank CU, et al. Association of anti-TNF with decreased survival in steroid refractory ipilimumab and anti-PD1 treated patients in the Dutch Melanoma Treatment Registry. Clin Cancer Res. 2020;26:2268–2274. doi:10.1158/1078-0432.ccr-19-3322
  • Zou F, Faleck D, Thomas A, et al. Original research: efficacy and safety of vedolizumab and infliximab treatment for immune-mediated diarrhea and colitis in patients with cancer: a two-center observational study. J Immunother Cancer. 2021;9(11):e003277. doi:10.1136/JITC-2021-003277
  • Stroud CRG, Hegde A, Cherry C, et al. Tocilizumab for the management of immune mediated adverse events secondary to PD-1 blockade. J Oncol Pharm Pract. 2019;25(3):551–557. doi:10.1177/1078155217745144
  • Dimitriou F, Hogan S, Menzies AM, Dummer R, Long GV. Interleukin-6 blockade for prophylaxis and management of immune-related adverse events in cancer immunotherapy. Eur J Cancer. 2021;157:214–224. doi:10.1016/j.ejca.2021.08.031
  • Fa’ak F, Zobniw CM, Buni M, et al. Selective immune suppression using interleukin-6 blockade in immune related adverse events. J Immunother Cancer. 2021;9(Suppl 2):A853–A853. doi:10.1136/JITC-2021-SITC2021.816
  • Damuzzo V, Solito S, Pinton L, et al. Clinical implication of tumor-associated and immunological parameters in melanoma patients treated with ipilimumab. Oncoimmunology. 2016;5(12):e1249559. doi:10.1080/2162402X.2016.1249559
  • Tachibana H, Nemoto Y, Ishihara H, et al. Predictive impact of early changes in serum C-reactive protein levels in nivolumab plus ipilimumab therapy for metastatic renal cell carcinoma. Clin Genitourin Cancer. 2021;20:e81–e88. doi:10.1016/J.CLGC.2021.10.005
  • Shi Y, Liu X, Du J, et al. Circulating cytokines associated with clinical outcomes in advanced non-small cell lung cancer patients who received chemoimmunotherapy. Thorac Cancer. 2022;13(2):219–227. doi:10.1111/1759-7714.14248
  • Tsukamoto H, Fujieda K, Miyashita A, et al. Combined blockade of IL6 and PD-1/PD-L1 signaling abrogates mutual regulation of their immunosuppressive effects in the tumor microenvironment. Cancer Res. 2018;78(17):5011–5022. doi:10.1158/0008-5472.CAN-18-0118
  • Strangfeld A, Richter A, Siegmund B, et al. Risk for lower intestinal perforations in patients with rheumatoid arthritis treated with tocilizumab in comparison to treatment with other biologic or conventional synthetic DMARDs. Ann Rheum Dis. 2017;76(3):504–510. doi:10.1136/ANNRHEUMDIS-2016-209773
  • Danese S, Vermeire S, Hellstern P, et al. Randomised trial and open-label extension study of an anti-interleukin-6 antibody in Crohn’s disease (ANDANTE I and II). Gut. 2019;68(1):40–48. doi:10.1136/gutjnl-2017-314562
  • Harrington R, Al Nokhatha SA, Conway R. JAK inhibitors in rheumatoid arthritis: an evidence-based review on the emerging clinical data. J Inflamm Res. 2020;13:519. doi:10.2147/JIR.S219586
  • Salem J-E, Allenbach Y, Vozy A, et al. Abatacept for severe immune checkpoint inhibitor-associated myocarditis. N Engl J Med. 2019;380(24):2377–2379. doi:10.1056/NEJMC1901677
  • Tyan K, Baginska J, Brainard M, et al. Cytokine changes during immune-related adverse events and corticosteroid treatment in melanoma patients receiving immune checkpoint inhibitors. Cancer Immunol Immunother. 2021;70(8):2209–2221. doi:10.1007/s00262-021-02855-1
  • Husain B, Kirchberger MC, Erdmann M, et al. Inflammatory markers in autoimmunity induced by checkpoint inhibitors. J Cancer Res Clin Oncol. 2021;147(6):1623–1630. doi:10.1007/S00432-021-03550-5
  • Esfahani K, Miller WH. Reversal of autoimmune toxicity and loss of tumor response by interleukin-17 blockade. N Engl J Med. 2017;376(20):1989–1991. doi:10.1056/NEJMC1703047/SUPPL_FILE/NEJMC1703047_DISCLOSURES.PDF
  • Thomas AS, Ma W, Wang Y. Ustekinumab for refractory colitis associated with immune checkpoint inhibitors. N Engl J Med. 2021;384(6):581–583. doi:10.1056/NEJMC2031717
  • Deftereos SN, Georgonikou D. Effectiveness of rituximab in treating immune-checkpoint-inhibitor-induced immune-related adverse events: results of a systematic review. Ann Oncol. 2021;32(2):282–283. doi:10.1016/J.ANNONC.2020.12.001
  • Simonaggio A, Michot JM, Voisin AL, et al. Evaluation of readministration of immune checkpoint inhibitors after immune-related adverse events in patients with cancer. JAMA Oncol. 2019;5(9):1310–1317. doi:10.1001/JAMAONCOL.2019.1022
  • Dolladille C, Ederhy S, Sassier M, et al. Immune checkpoint inhibitor rechallenge after immune-related adverse events in patients with cancer. JAMA Oncol. 2020;6(6):865–871. doi:10.1001/JAMAONCOL.2020.0726
  • van der Kooij MK, Suijkerbuijk KP, Dekkers OM, Kapiteijn E. Safety and efficacy of checkpoint inhibition in patients with melanoma and preexisting autoimmune disease. Ann Intern Med. 2021;174(9):1345. doi:10.7326/L21-0441
  • 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):121–130. doi:10.7326/M17-2073
  • Arbour KC, Mezquita L, Long N, et al. Impact of baseline steroids on efficacy of programmed cell death-1 and programmed death-ligand 1 blockade in patients with non–small-cell lung cancer. J Clin Oncol. 2018;36(28):2872–2878. doi:10.1200/JCO.2018.79.0006
  • 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):368–376. doi:10.1093/annonc/mdw443
  • Scott DL, Wolfe F, Huizinga TWJ. Rheumatoid arthritis. Lancet. 2010;376(9746):1094–1108. doi:10.1016/S0140-6736(10)60826-4
  • Ghosh N, Postow M, Zhu C, et al. Lower baseline autoantibody levels are associated with immune-related adverse events from immune checkpoint inhibition. J Immunother Cancer. 2022;10(1):e004008. doi:10.1136/jitc-2021-004008