Update on immunotherapy-mediated colitis: Clinical features, mechanisms, and management

Dandan Wang , Yiwei Zhao , Yiyun Zeng , Lanlin Hu , Chuan Xu

Malignancy Spectrum ›› 2024, Vol. 1 ›› Issue (4) : 225 -242.

PDF (737KB)
Malignancy Spectrum ›› 2024, Vol. 1 ›› Issue (4) : 225 -242. DOI: 10.1002/msp2.50
REVIEW

Update on immunotherapy-mediated colitis: Clinical features, mechanisms, and management

Author information +
History +
PDF (737KB)

Abstract

Immunotherapy, particularly immune checkpoint inhibitors (ICIs), has radically transformed the field of oncological therapy. However, immune‐related adverse events (irAEs) associated with the treatment might affect the life quality and even threaten the life of cancer patients. Immunotherapy-mediated colitis (IMC) is one of the most prevalent irAEs, especially in anti-cytotoxic T lymphocyte antigen 4 antibody (CTLA4) therapy. Current management of IMC includes administering immunosuppressants and discontinuing the treatment, which might affect the therapeutic efficacy. In this review, we briefly summarize the development of ICIs in cancer immunotherapy and the incidence, diagnosis, and management of IMC. Recent insights into the cellular and molecular pathways that underpin IMC, perspective research, and promising therapeutic strategies are highlighted. Further understanding of IMC and its implications will assist clinicians in optimizing treatment strategies to mitigate this side effect while maximizing the benefits of ICIs.

Keywords

immune checkpoint inhibitor / immune-related adverse event / immunotherapy-mediated colitis

Cite this article

Download citation ▾
Dandan Wang, Yiwei Zhao, Yiyun Zeng, Lanlin Hu, Chuan Xu. Update on immunotherapy-mediated colitis: Clinical features, mechanisms, and management. Malignancy Spectrum, 2024, 1(4): 225-242 DOI:10.1002/msp2.50

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Yousif LI, Screever EM, Versluis D, et al. Risk factors for immune checkpoint inhibitor–mediated cardiovascular toxicities. Curr Oncol Rep. 2023;25(7):753-763.

[2]

Muntyanu A, Netchiporouk E, Gerstein W, Gniadecki R, Litvinov IV. Cutaneous immune-related adverse events (irAEs) to immune checkpoint inhibitors: a dermatology perspective on management. J Cutan Med Surg. 2021;25(1):59-76.

[3]

Dougan M. Checkpoint blockade toxicity and immune homeostasis in the gastrointestinal tract. Front Immunol. 2017;8:1547.

[4]

Wang Y, Abu-Sbeih H, Mao E, et al. Endoscopic and histologic features of immune checkpoint inhibitor-related colitis. Inflamm Bowel Dis. 2018;24(8):1695-1705.

[5]

Marthey L, Mateus C, Mussini C, et al. Cancer immunotherapy with anti-CTLA4 monoclonal antibodies induces an inflammatory bowel disease. J Crohns Colitis. 2016;10(4):395-401.

[6]

Dougan M, Blidner AG, Choi J, et al. Multinational association of supportive care in cancer (MASCC) 2020 clinical practice recommendations for the management of severe gastro-intestinal and hepatic toxicities from checkpoint inhibitors. Support Care Cancer. 2020;28(12):6129-6143.

[7]

Ling SP, Ming LC, Dhaliwal JS, et al. Role of immunotherapy in the treatment of cancer: systematic review. Cancers. 2022;14(21):5205.

[8]

Waldman AD, Fritz JM, Lenardo MJ. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat Rev Immunol. 2020;20(11):651-668.

[9]

Dougan M, Dranoff G, Dougan SK. Cancer immunotherapy: beyond checkpoint blockade. Annu Rev Cancer Biol. 2019;3:55-75.

[10]

Saxena M, van der Burg SH, Melief CJM, Bhardwaj N. Therapeutic cancer vaccines. Nat Rev Cancer. 2021;21(6):360-378.

[11]

Zhang Y, Zhang Z. The history and advances in cancer immunotherapy: understanding the characteristics of tumorinfiltrating immune cells and their therapeutic implications. Cell Mol Immunol. 2020;17(8):807-821.

[12]

Forde PM, Spicer J, Lu S, et al. Neoadjuvant nivolumab plus chemotherapy in resectable lung cancer. N Engl J Med. 2022;386(21):1973-1985.

[13]

Gao S, Li N, Gao S, et al. Neoadjuvant PD-1 inhibitor (sintilimab) in NSCLC. J Thorac Oncol. 2020;15(5):816-826.

[14]

Antonia SJ, Villegas A, Daniel D, et al. Durvalumab after chemoradiotherapy in stage III non-small-Cell lung cancer. N Engl J Med. 2017;377(20):1919-1929.

[15]

Felip E, Altorki N, Zhou C, et al. Adjuvant atezolizumab after adjuvant chemotherapy in resected stage IB-IIIA non-small-cell lung cancer (IMpower010): a randomised, multicentre, open-label, phase 3 trial. The Lancet. 2021;398(10308):1344-1357.

[16]

Yang Y, Wu M, Cao D, et al. ZBP1-MLKL necroptotic signaling potentiates radiation-induced anti-tumor immunity via intratumoral STING pathway activation. Sci Adv. 2021;7(41): eabf6290.

[17]

Benci JL, Johnson LR, Choa R, et al. Opposing functions of interferon coordinate adaptive and innate immune responses to cancer immune checkpoint blockade. Cell. 2019;178(4):933-948.e14.

[18]

Nandi D, Pathak S, Verma T, et al. T cell costimulation, check-point inhibitors and anti-tumor therapy. J Biosci. 2020;45:50.

[19]

Jhunjhunwala S, Hammer C, Delamarre L. Antigen presentation in cancer: insights into tumour immunogenicity and immune evasion. Nat Rev Cancer. 2021;21(5):298-312.

[20]

Spurrell EL, Lockley M. Adaptive immunity in cancer immunology and therapeutics. Ecancermedicalscience. 2014;8:441.

[21]

Wei SC, Duffy CR, Allison JP. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 2018;8(9):1069-1086.

[22]

Brunet JF, Denizot F, Luciani MF, et al. A new member of the immunoglobulin superfamily—CTLA4. Nature. 1987;328(6127):267-270.

[23]

Martin-Liberal J, Ochoa de Olza M, Hierro C, Gros A, Rodon J, Tabernero J. The expanding role of immunotherapy. Cancer Treat Rev. 2017;54:74-86.

[24]

Sharma P, Allison JP. The future of immune checkpoint therapy. Science. 2015;348(6230):56-61.

[25]

Krummel MF, Allison JP. CD28 and CTLA4 have opposing effects on the response of T cells to stimulation. J Exp Med. 1995;182(2):459-465.

[26]

Wing K, Onishi Y, Prieto-Martin P, et al. CTLA4 control over Foxp3+ regulatory T cell function. Science. 2008;322:271-275.

[27]

Peggs KS, Quezada SA, Chambers CA, Korman AJ, Allison JP. Blockade of CTLA4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA4 antibodies. J Exp Med. 2009;206(8):1717-1725.

[28]

Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711-723.

[29]

Franzen D, Schad K, Kowalski B, et al. Ipilimumab and early signs of pulmonary toxicity in patients with metastastic melanoma: a prospective observational study. Cancer Immunol Immunother. 2018;67(1):127-134.

[30]

Robert C, Schachter J, Long GV, et al. Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med. 2015;372(26):2521-2532.

[31]

FDA. 2011 Notifications. Via the FDA website. 2018. Accessed August 26, 2023.

[32]

Pitt JM, Vétizou M, Daillère R, et al. Resistance mechanisms to immune-checkpoint blockade in cancer: tumor-Intrinsic and -extrinsic factors. Immunity. 2016;44(6):1255-1269.

[33]

FDA. Oncology (cancer)/hematologic malignancies approval notifications. Via the FDA website. 2023. Accessed October 8, 2023.

[34]

Sangro B, Chan SL, Kelley RK, et al. Four-year overall survival update from the phase III HIMALAYA study of tremelimumab plus durvalumab in unresectable hepatocellular carcinoma. Ann Oncol. 2024;35(5):448-457.

[35]

Johnson ML, Cho BC, Luft A, et al. Durvalumab with or without tremelimumab in combination with chemotherapy as first-line therapy for metastatic non-small-cell llung cancer: the phase III POSEIDON study. J Clin Oncol. 2023;41(6):1213-1227.

[36]

Zhang JY, Yan YY, Li JJ, Adhikari R, Fu LW. PD-1/PD-L1 based combinational cancer therapy: icing on the cake. Front Pharmacol. 2020;11:722.

[37]

Wang X, Yang X, Zhang C, et al. Tumor cell-intrinsic PD-1 receptor is a tumor suppressor and mediates resistance to PD-1 blockade therapy. Proc Natl Acad Sci. 2020;117(12):6640-6650.

[38]

Ribas A, Puzanov I, Dummer R, et al. Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): a randomised, controlled, phase 2 trial. Lancet Oncol. 2015;16(8):908-918.

[39]

Robert C, Ribas A, Schachter J, et al. Pembrolizumab versus ipilimumab in advanced melanoma (KEYNOTE-006): post-hoc 5-year results from an open-label, multicentre, randomised, controlled, phase 3 study. Lancet Oncol. 2019;20(9):1239-1251.

[40]

Robert C, Carlino MS, McNeil C, et al. Seven-year follow-up of the phase III KEYNOTE-006 study: pembrolizumab versus ipilimumab in advanced melanoma. J Clin Oncol. 2023;41(24):3998-4003.

[41]

Weber JS, D’Angelo SP, Minor D, et al. Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2015;16(4):375-384.

[42]

Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366(26):2443-2454.

[43]

Ning YM, Suzman D, Maher VE, et al. FDA approval summary: atezolizumab for the treatment of patients with progressive advanced urothelial carcinoma after platinum-containing chemotherapy. Oncologist. 2017;22(6):743-749.

[44]

Research C for D. E. and Drug Approvals and Databases FDA. Via the FDA website; 2022.

[45]

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: Mech Dis. 2021;16:223-249.

[46]

Guigay J, Lee KW, Patel MR, et al. Avelumab for platinumineligible/refractory recurrent and/or metastatic squamous cell carcinoma of the head and neck: phase Ib results from the JAVELIN solid tumor trial. J Immunother Cancer. 2021;9(10): e002998.

[47]

Grosso JF, Kelleher CC, Harris TJ, et al. LAG-3 regulates CD8+ T cell accumulation and effector function in murine self-and tumor-tolerance systems. J Clin Invest. 2007;117(11):3383-3392.

[48]

Nguyen LT, Ohashi PS. Clinical blockade of PD1 and LAG3—potential mechanisms of action. Nat Rev Immunol. 2015;15(1):45-56.

[49]

Puhr HC, Ilhan-Mutlu A. New emerging targets in cancer immunotherapy: the role of LAG3. ESMO Open. 2019;4(2): e000482.

[50]

Long L, Zhang X, Chen F, et al. The promising immune checkpoint LAG-3: from tumor microenvironment to cancer immunotherapy. Genes Cancer. 2018;9(5–6):176-189.

[51]

Kraman M, Faroudi M, Allen NL, et al. FS118, a bispecific antibody targeting LAG-3 and PD-L1, enhances T-Cell acti-vation resulting in potent antitumor activity. Clin Cancer Res. 2020;26(13):3333-3344.

[52]

Huang RY, Eppolito C, Lele S, Shrikant P, Matsuzaki J, Odunsi K. LAG3 and PD1 co-inhibitory molecules collaborate to limit CD8+ T cell signaling and dampen antitumor immunity in a murine ovarian cancer model. Oncotarget. 2015;6(29):27359-27377.

[53]

Tawbi HA, Schadendorf D, Lipson EJ, et al. Relatlimab and nivolumab versus nivolumab in untreated advanced melanoma. N Engl J Med. 2022;386(1):24-34.

[54]

Rotte A, Jin JY, Lemaire V. Mechanistic overview of immune checkpoints to support the rational design of their combinations in cancer immunotherapy. Ann Oncol. 2018;29(1):71-83.

[55]

Andrews LP, Yano H, Vignali DAA. Inhibitory receptors and ligands beyond PD-1, PD-L1 and CTLA4: breakthroughs or backups. Nature Immunol. 2019;20(11):1425-1434.

[56]

Anderson AC. Tim-3, a negative regulator of anti-tumor immunity. Curr Opin Immunol. 2012;24(2):213-216.

[57]

Gefen T, Castro I, Muharemagic D, Puplampu-Dove Y, Patel S, Gilboa E. A TIM-3 oligonucleotide aptamer enhances T cell functions and potentiates tumor immunity in mice. Mol Ther. 2017;25(10):2280-2288.

[58]

Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12(4):252-264.

[59]

Sakuishi K, Apetoh L, Sullivan JM, Blazar BR, Kuchroo VK, Anderson AC. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J Exp Med. 2010;207(10):2187-2194.

[60]

Fourcade J, Sun Z, Benallaoua M, et al. Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients. J Exp Med. 2010;207(10):2175-2186.

[61]

ElHalabi L, Adam J, Gravelle P, et al. Expression of the immune checkpoint regulators LAG-3 and TIM-3 in classical Hodgkin lymphoma. Clin Lymphoma Myeloma Leuk. 2021;21(4):257-266.e3.

[62]

Curigliano G, Gelderblom H, Mach N, et al. Phase I/Ib clinical trial of sabatolimab, an anti-TIM-3 antibody, alone and in combination with spartalizumab, an Anti-PD-1 antibody, in advanced solid tumors. Clin Cancer Res. 2021;27(13):3620-3629.

[63]

Harding JJ, Moreno V, Bang YJ, et al. Blocking TIM-3 in treatment-refractory advanced solid tumors: a phase Ia/b study of LY3321367 with or without an anti-PD-L1 antibody. Clin Cancer Res. 2021;27:2168-2178.

[64]

Hollebecque A, Chung HC, de Miguel MJ, et al. Safety and antitumor activity of α-PD-L1 antibody as monotherapy or in combination with α-TIM-3 antibody in patients with micro-satellite instability-high/mismatch repair-deficient tumors. Clin Cancer Res. 2021;27(23):6393-6404.

[65]

Hellmann MD, Bivi N, Calderon B, et al. Safety and immunogenicity of LY3415244, a bispecific antibody against TIM-3 and PD-L1, in patients with advanced solid tumors. Clin Cancer Res. 2021;27(10):2773-2781.

[66]

Manieri NA, Chiang EY, Grogan JL. TIGIT: a key inhibitor of the cancer immunity cycle. Trends Immunol. 2017;38(1):20-28.

[67]

Joller N, Lozano E, Burkett PR, et al. Treg cells expressing the co-inhibitory molecule TIGIT selectively inhibit proinflammatory TH1 and TH17 cell responses. Immunity. 2014;40(4):569-581.

[68]

Dougall WC, Kurtulus S, Smyth MJ, Anderson AC. TIGIT and CD96: new checkpoint receptor targets for cancer immunotherapy. Immunol Rev. 2017;276(1):112-120.

[69]

Chen X, Xue L, Ding X, et al. An Fc-competent anti-human TIGIT blocking antibody ociperlimab (BGB-A1217) elicits strong immune responses and potent anti-tumor efficacy in pre-clinical models. Front Immunol. 2022;13:828319.

[70]

Ge Z, Peppelenbosch MP, Sprengers D, Kwekkeboom J. TIGIT, the next step towards successful combination immune check-point therapy in cancer. Front Immunol. 2021;12:699895.

[71]

Yuan L, Tatineni J, Mahoney KM, Freeman GJ. VISTA: a mediator of quiescence and a promising target in cancer immunotherapy. Trends Immunol. 2021;42(3):209-227.

[72]

Getu AA, Tigabu A, Zhou M, Lu J, Fodstad Ø, Tan M. New frontiers in immune checkpoint B7-H3 (CD276) research and drug development. Mol Cancer. 2023;22(1):43.

[73]

Pulanco MC, Madsen AT, Tanwar A, Corrigan DT, Zang X. Recent advancements in the B7/CD28 immune checkpoint families: new biology and clinical therapeutic strategies. Cell Mol Immunol. 2023;20(7):694-713.

[74]

Shenderov E, De Marzo AM, Lotan TL, et al. Neoadjuvant enoblituzumab in localized prostate cancer: a single-arm, phase 2 trial. Nature Med. 2023;29(4):888-897.

[75]

Aggarwal C, Prawira A, Antonia S, et al. Dual checkpoint targeting of B7-H3 and PD-1 with enoblituzumab and pembrolizumab in advanced solid tumors: interim results from a multicenter phase I/II trial. J Immunother Cancer. 2022;10(4):e004424.

[76]

Das S, Johnson DB. Immune-related adverse events and anti-tumor efficacy of immune checkpoint inhibitors. J Immunother Cancer. 2019;7(1):306.

[77]

Pasquali S, Hadjinicolaou AV, Chiarion Sileni V, Rossi CR, Mocellin S. Systemic treatments for metastatic cutaneous melanoma. Cochrane Database Syst Rev. 2018;2(2):011123.

[78]

John AO, Ramnath N. Neoadjuvant versus adjuvant systemic therapy for early-stage non-small cell lung cancer: the changing landscape due to immunotherapy. Oncologist. 2023;28(9):752-764.

[79]

de Castro G, Kudaba I, Wu Y, et al. 363 KEYNOTE-042 5-year survival update: pembrolizumab versus chemotherapy in patients with previously untreated, PD-L1–positive, locally advanced or metastatic non–small-cell lung cancer. J Immunother Cancer. 2021;9(Suppl 3):A390.

[80]

Hussaini S, Chehade R, Boldt RG, et al. Association between immune-related side effects and efficacy and benefit of immune checkpoint inhibitors–a systematic review and meta-analysis. Cancer Treat Rev. 2021;92:102134.

[81]

Ricciuti B, Genova C, De Giglio A, et al. Impact of immune-related adverse events on survival in patients with advanced non-small cell lung cancer treated with nivolumab: long-term outcomes from a multi-institutional analysis. J Cancer Res Clin Oncol. 2019;145(2):479-485.

[82]

Sato K, Akamatsu H, Murakami E, et al. Correlation between immune-related adverse events and efficacy in non-small cell lung cancer treated with nivolumab. Lung Cancer. 2018;115:71-74.

[83]

Indini A, Di Guardo L, Cimminiello C, et al. Immune-related adverse events correlate with improved survival in patients undergoing anti-PD1 immunotherapy for metastatic melanoma. J Cancer Res Clin Oncol. 2019;145(2):511-521.

[84]

Weber JS, Hodi FS, Wolchok JD, et al. Safety profile of nivolumab monotherapy: a pooled analysis of patients with advanced melanoma. J Clin Oncol. 2017;35(7):785-792.

[85]

Brahmer JR, Lacchetti C, Schneider BJ, et al. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American society of clinical oncology clinical practice guideline. J Clin Oncol. 2018;36(17):1714-1768.

[86]

Tandon P, Bourassa-Blanchette S, Bishay K, Parlow S, Laurie SA, McCurdy JD. The risk of diarrhea and colitis in patients with advanced melanoma undergoing immune checkpoint inhibitor therapy: a systematic review and meta-analysis. J Immunother. 2018;41(3):101-108.

[87]

Khoja L, Day D, Wei-Wu Chen T, Siu LL, Hansen AR. Tumour-and class-specific patterns of immune-related adverse events of immune checkpoint inhibitors: a systematic review. Ann Oncol. 2017;28(10):2377-2385.

[88]

Wang DY, Ye F, Zhao S, Johnson DB. Incidence of immune checkpoint inhibitor-related colitis in solid tumor patients: a systematic review and meta-analysis. Oncoimmunology. 2017;6(10):e1344805.

[89]

Borghaei H, Paz-Ares L, Horn L, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 2015;373(17):1627-1639.

[90]

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):123-135.

[91]

Yasuda Y, Urata Y, Tohnai R, et al. Immune-related colitis induced by the long-term use of nivolumab in a patient with non-small cell lung cancer. Intern Med. 2018;57(9):1269-1272.

[92]

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):576-583.

[93]

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;91:21-29.

[94]

Dougan M, Pietropaolo M. Time to dissect the autoimmune etiology of cancer antibody immunotherapy. J Clin Invest. 2020;130(1):51-61.

[95]

Knights D, Lassen KG, Xavier RJ. Advances in inflammatory bowel disease pathogenesis: linking host genetics and the microbiome. Gut. 2013;62(10):1505-1510.

[96]

Abraham C, Cho JH. Inflammatory bowel disease. N Engl J Med. 2009;361(21):2066-2078.

[97]

Zhang Y-Z. Inflammatory bowel disease: pathogenesis. World J Gastroenterol. 2014;20(1):91-99.

[98]

Beck KE, Blansfield JA, Tran KQ, et al. Enterocolitis in patients with cancer after antibody blockade of cytotoxic T-lymphocyte-associated antigen 4. J Clin Oncol. 2006;24(15):2283-2289.

[99]

Zhang ML, Neyaz A, Patil D, Chen J, Dougan M, Deshpande V. Immune-related adverse events in the gastro-intestinal tract: diagnostic utility of upper gastrointestinal biopsies. Histopathology. 2020;76(2):233-243.

[100]

Burdine L, Lai K, Laryea JA. Ipilimumab-induced colonic perforation. J Surg Case Rep. 2014;2014(3):rju010.

[101]

Shah R, Witt D, Asif T, Mir FF. Ipilimumab as a cause of severe pan-colitis and colonic perforation. Cureus. 2017;9(4):e1182.

[102]

Dilling P, Walczak J, Pikiel P, Kruszewski WJ. Multiple colon perforation as a fatal complication during treatment of meta-static melanoma with ipilimumab–case report. Polish J Surg. 2014;86(2):94-96.

[103]

Geukes Foppen MH, Rozeman EA, van Wilpe S, et al. Immune checkpoint inhibition-related colitis: symptoms, endoscopic features, histology and response to management. ESMO Open. 2018;3(1):e000278.

[104]

Common Terminology Criteria for Adverse Events (CTCAE). (2017).

[105]

Gupta A, De Felice KM, Loftus EV, Khanna S. Systematic review: colitis associated with anti-CTLA4 therapy. Aliment Pharmacol Ther. 2015;42(4):406-417.

[106]

Bellaguarda E, Hanauer S. Checkpoint inhibitor-induced colitis. Am J Gastroenterol. 2020;115(2):202-210.

[107]

Shah R, Witt D, Asif T, Mir FF. Ipilimumab as a cause of severe Pan-Colitis and colonic perforation. Cureus. 2017;9(4): e1182.

[108]

Cho HJ, Kim WR, Kim JH, Kim DH, Kim DJ, Kang H. Colonic perforation after treatment with nivolumab in esophageal cancer: a case report. Ann Coloproctol. 2021;37(Suppl 1): S39-S43.

[109]

Nakai M, Kai Y, Suzuki K, et al. A case of perforated immune-related colitis complicated by cytomegalovirus infection during treatment of immune-related adverse effect in lung cancer immunotherapy. Respir Med Case Rep. 2023;41:101794.

[110]

De Silva S, Trieu H, Rajan A, Liang Y, Lin JL, Kidambi TD. Flexible sigmoidoscopy may be sufficient for initial evaluation of suspected immunotherapy-mediated colitis: a cross-sectional study. J Gastroenterol Hepatol. 2022;37(2):284-290.

[111]

Abu-Sbeih H, Ali FS, Luo W, Qiao W, Raju GS, Wang Y. Importance of endoscopic and histological evaluation in the management of immune checkpoint inhibitor-induced colitis. J Immunother Cancer. 2018;6(1):95.

[112]

Lerrer S, Mor A. Immune checkpoint inhibitors and the shared epitope theory: from hypothesis to practice. Transl Cancer Res. 2019;8(Suppl 6):S625-S627.

[113]

Berner F, Bomze D, Diem S, et al. Association of checkpoint inhibitor-induced toxic effects with shared cancer and tissue antigens in non-small cell lung cancer. JAMA Oncol. 2019;5(7):1043-1047.

[114]

Passat T, Touchefeu Y, Gervois N, Jarry A, Bossard C, Bennouna J. Mécanismes physiopathologiques des effets secondaires des immunothérapies par anticorps anti-CTLA-4, anti-PD-1 et anti-PD-L1 dans le traitement du cancer [Physiopathological mechanisms of immune-related adverse events induced by anti-CTLA-4, anti-PD-1 and anti-PD-L1 antibodies in cancer treatment]. Bull Cancer. 2018;105(11):1033-1041.

[115]

Vétizou M, Pitt JM, Daillère R, et al. Anticancer immunotherapy by CTLA4 blockade relies on the gut microbiota. Science. 2015;350(6264):1079-1084.

[116]

Davar D, Kirkwood JM. PD-1 immune checkpoint inhibitors and immune-related adverse events: understanding the upside of the downside of checkpoint blockade. JAMA Oncology. 2019;5(7):942-943.

[117]

Luoma AM, Suo S, Williams HL, et al. Molecular pathways of colon inflammation induced by cancer immunotherapy. Cell. 2020;182(3):655-671.e22.

[118]

Lu CH, Li KJ, Wu CH, et al. The FcγRIII engagement augments PMA-stimulated neutrophil extracellular traps (NETs) formation by granulocytes partially via cross-talk between Syk-ERK-NF-κB and PKC-ROS signaling pathways. Biomedicines. 2021;9(9):1127.

[119]

Eitler J, Wotschel N, Miller N, et al. Inability of granule polarization by NK cells defines tumor resistance and can be overcome by CAR or ADCC mediated targeting. J Immunother Cancer. 2021;9(1):e001334.

[120]

Vijayaraghavan S, Lipfert L, Chevalier K, et al. Amivantamab (JNJ-61186372), an Fc enhanced EGFR/cMet bispecific anti-body, induces receptor downmodulation and antitumor activity by monocyte/macrophage trogocytosis. Mol Cancer Ther. 2020;19(10):2044-2056.

[121]

Selby MJ, Engelhardt JJ, Quigley M, et al. Anti-CTLA4 anti-bodies of IgG2a isotype enhance antitumor activity through reduction of intratumoral regulatory T cells. Cancer Immunol Res. 2013;1(1):32-42.

[122]

Simpson TR, Li F, Montalvo-Ortiz W, et al. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA4 therapy against melanoma. J Exp Med. 2013;210(9):1695-1710.

[123]

Bauché D, Mauze S, Kochel C, et al. Antitumor efficacy of combined CTLA4/PD-1 blockade without intestinal inflammation is achieved by elimination of FcγR interactions. J Immunother Cancer. 2020;8(2):e001584.

[124]

Das R, Bar N, Ferreira M, et al. Early B cell changes predict autoimmunity following combination immune checkpoint blockade. J Clin Invest. 2018;128(2):715-720.

[125]

Isnardi I, Ng YS, Menard L, et al. Complement receptor 2/CD21 human naive B cells contain mostly autoreactive unresponsive clones. Blood. 2010;115(24):5026-5036.

[126]

Tipton CM, Fucile CF, Darce J, et al. Diversity, cellular origin and autoreactivity of antibody-secreting cell population expansions in acute systemic lupus erythematosus. Nature Immunol. 2015;16(7):755-765.

[127]

Lau D, Lan LYL, Andrews SF, et al. Low CD21 expression defines a population of recent germinal center graduates primed for plasma cell differentiation. Sci Immunol. 2017;2(7):eaai8153.

[128]

Rakhmanov M, Gutenberger S, Keller B, Schlesier M, Peter HH, Warnatz K. CD21 low B cells in common variable immunodeficiency do not show defects in receptor editing, but resemble tissue-like memory B cells. Blood. 2010;116(18):3682-3683.

[129]

Martin JC, Chang C, Boschetti G, et al. Single-cell analysis of Crohn’s disease lesions identifies a pathogenic cellular module associated with resistance to anti-TNF therapy. Cell. 2019;178(6):1493-1508.e20.

[130]

Smillie CS, Biton M, Ordovas-Montanes J, et al. Intra- and inter-cellular rewiring of the human colon during ulcerative colitis. Cell. 2019;178(3):714-730.e22.

[131]

Ramos-Casals M, Brahmer JR, Callahan MK, et al. Immune-related adverse events of checkpoint inhibitors. Nat Rev Dis Primers. 2020;6(1):38.

[132]

Alissafi T, Hatzioannou A, Legaki AI, Varveri A, Verginis P. Balancing cancer immunotherapy and immune-related adverse events: the emerging role of regulatory T cells. J Autoimmun. 2019;104:102310.

[133]

Liu Y, Zheng P. Preserving the CTLA4 checkpoint for safer and more effective cancer immunotherapy. Trends Pharmacol Sci. 2020;41(1):4-12.

[134]

Simpson TR, Li F, Montalvo-Ortiz W, et al. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA4 therapy against melanoma. J Exp Med. 2013;210(9):1695-1710.

[135]

Ha D, Tanaka A, Kibayashi T, et al. Differential control of human Treg and effector T cells in tumor immunity by Fc-engineered anti-CTLA4 antibody. Proc Natl Acad Sci. 2019;116(2):609-618.

[136]

Lo BC, Kryczek I, Yu J, et al. Microbiota-dependent activation of CD4+ T cells induces CTLA4 blockade-associated colitis via Fcγ receptors. Science. 2024;383(6678):62-70.

[137]

von Euw E, Chodon T, Attar N, et al. CTLA4 blockade increases TH17 cells in patients with metastatic melanoma. J Transl Med. 2009;7:35.

[138]

Harbour SN, Maynard CL, Zindl CL, Schoeb TR, Weaver CT. TH17 cells give rise to TH1 cells that are required for the pathogenesis of colitis. Proc Natl Acad Sci. 2015;112(22):7061-7066.

[139]

Maddur MS, Miossec P, Kaveri SV, Bayry J. TH17 cells. Am J Pathol. 2012;181(1):8-18.

[140]

Tarhini AA, Zahoor H, Lin Y, et al. Baseline circulating IL-17 predicts toxicity while TGF-β1 and IL-10 are prognostic of relapse in ipilimumab neoadjuvant therapy of melanoma. J Immunother Cancer. 2015;3:39.

[141]

Bamias G, Delladetsima I, Perdiki M, et al. Immunological characteristics of colitis associated with anti-CTLA4 antibody therapy. Cancer Invest. 2017;35(7):443-455.

[142]

Perez-Ruiz E, Minute L, Otano I, et al. Prophylactic TNF blockade uncouples efficacy and toxicity in dual CTLA4 and PD-1 immunotherapy. Nature. 2019;569(7756):428-432.

[143]

Lo JW, Cozzetto D, Alexander JL, et al. Immune checkpoint inhibitor-induced colitis is mediated by polyfunctional lymphocytes and is dependent on an IL23/IFNγ axis. Nat Commun. 2023;14(1):6719.

[144]

Hailemichael Y, Johnson DH, Abdel-Wahab N, et al. Interleukin-6 blockade abrogates immunotherapy toxicity and promotes tumor immunity. Cancer Cell. 2022;40(5):509-523.e6.

[145]

Jones SA, Jenkins BJ. Recent insights into targeting the IL-6 cytokine family in inflammatory diseases and cancer. Nat Rev Immunol. 2018;18(12):773-789.

[146]

Holmstroem RB, Nielsen OH, Jacobsen S, et al. COLAR: open-label clinical study of IL-6 blockade with tocilizumab for the treatment of immune checkpoint inhibitor-induced colitis and arthritis. J Immunother Cancer. 2022;10(9):e005111.

[147]

Neurath MF. Targeting immune cell circuits and trafficking in inflammatory bowel disease. Nature Immunol. 2019;20(8):970-979.

[148]

Abu-Sbeih H, Ali FS, Alsaadi D, et al. Outcomes of vedolizumab therapy in patients with immune checkpoint inhibitor-induced colitis: a multi-center study. J Immunother Cancer. 2018;6(1):142.

[149]

Diana P, Mankongpaisarnrung C, Atkins MB, Zeck JC, Charabaty A. Emerging role of vedolizumab in managing refractory immune checkpointinhibitor-induced enteritis. ACG Case Rep J. 2018;5:e17.

[150]

Horvat TZ, Adel NG, Dang TO, et al. Immune-related adverse events, need for systemic immunosuppression, and effects on survival and time to treatment failure in patients with melanoma treated with ipilimumab at memorial sloan kettering cancer center. J Clin Oncol. 2015;33(28):3193-3198.

[151]

Abu-Sbeih H, Ali FS, Wang X, et al. Early introduction of selective immunosuppressive therapy associated with favorable clinical outcomes in patients with immune checkpoint inhibitor-induced colitis. J Immunother Cancer. 2019;7(1):93.

[152]

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):103.

[153]

Chaput N, Lepage P, Coutzac C, et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann Oncol. 2017;28(6):1368-1379.

[154]

Dubin K, Callahan MK, Ren B, et al. Intestinal microbiome analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis. Nat Commun. 2016;7:10391.

[155]

Usyk M, Pandey A, Hayes RB, et al. Bacteroides vulgatus and bacteroides dorei predict immune-related adverse events in immune checkpoint blockade treatment of metastatic melanoma. Genome Med. 2021;13(1):160.

[156]

Zhang J, Hoedt EC, Liu Q, et al. Elucidation of Proteus mirabilis as a key bacterium in Crohn’s disease inflammation. Gastroenterology. 2021;160(1):317-330.e11.

[157]

Herp S, Brugiroux S, Garzetti D, et al. Mucispirillum schaedleri antagonizes salmonella virulence to protect mice against colitis. Cell Host Microbe. 2019;25(5):681-694.e8.

[158]

Liu W, Ma F, Sun B, et al. Intestinal microbiome associated with immune-related adverse events for patients treated with Anti-PD-1 inhibitors, a real-world study. Front Immunol. 2021;12:756872.

[159]

Zhou G, Zhang N, Meng K, Pan F. Interaction between gut microbiota and immune checkpoint inhibitor-related colitis. Front Immunol. 2022;13:1001623.

[160]

Palleja A, Mikkelsen KH, Forslund SK, et al. Recovery of gut microbiota of healthy adults following antibiotic exposure. Nat Microbiol. 2018;3(11):1255-1265.

[161]

Mohiuddin JJ, Chu B, Facciabene A, et al. Association of antibiotic exposure with survival and toxicity in patients with melanoma receiving immunotherapy. J Natl Cancer Inst. 2020;113(2):162-170.

[162]

Mao J, Wang D, Long J, et al. Gut microbiome is associated with the clinical response to anti-PD-1 based immunotherapy in hepatobiliary cancers. J Immunother Cancer. 2021;9(12):e003334.

[163]

Zhao L, Li Y, Jiang N, et al. Association of blood biochemical indexes and antibiotic exposure with severe immune-related adverse events in patients with advanced cancers receiving PD-1 inhibitors. J Immunother. 2022;45(4):210-216.

[164]

Hoefsmit EP, Rozeman EA, Haanen JBAG, Blank CU. Susceptible loci associated with autoimmune disease as potential biomarkers for checkpoint inhibitor-induced immune-related adverse events. ESMO Open. 2019;4(4):e000472.

[165]

Jostins L, Ripke S, Weersma RK, et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature. 2012;491(7422):119-124.

[166]

Huang H, Fang M, Jostins L, et al. Fine-mapping inflammatory bowel disease loci to single-variant resolution. Nature. 2017;547(7662):173-178.

[167]

Shek D, Read SA, Akhuba L, et al. Non-coding RNA and immune-checkpoint inhibitors: friends or foes? Immunotherapy. 2020;12(7):513-529.

[168]

Schneider C, Setty M, Holmes AB, et al. MicroRNA 28 controls cell proliferation and is down-regulated in b-cell lymphomas. Proc Natl Acad Sciences. 2014;111(22):8185-8190.

[169]

Fujita Y, Yagishita S, Hagiwara K, et al. The clinical relevance of the miR-197/CKS1B/STAT3-mediated PD-L1 network in chemoresistant non-small-cell lung cancer. Mol Ther. 2015;23(4):717-727.

[170]

Marschner D, Falk M, Javorniczky NR, et al. MicroRNA-146a regulates immune-related adverse events caused by immune checkpoint inhibitors. JCI Insight. 2020;5(6):e132334.

[171]

Park N, Pandey K, Chang SK, et al. Preclinical platform for long-term evaluation of immuno-oncology drugs using hCD34+ humanized mouse model. J Immunother Cancer. 2020;8(2):e001513.

[172]

Adam K, Iuga A, Tocheva AS, Mor A. A novel mouse model for checkpoint inhibitor-induced adverse events. PLoS One. 2021;16(2):e0246168.

[173]

Xu Y, Fu Y, Zhu B, Wang J, Zhang B. Predictive biomarkers of immune checkpoint inhibitors-related toxicities. Front Immunol. 2020;11:2023.

[174]

Hommes JW, Verheijden RJ, Suijkerbuijk KPM, Hamann D. Biomarkers of checkpoint inhibitor induced immune-related adverse events – a comprehensive review. Front Oncol. 2021;10:585311.

[175]

Kang JH, Bluestone JA, Young A. Predicting and preventing immune checkpoint inhibitor toxicity: targeting cytokines. Trends Immunol. 2021;42:293-311.

[176]

Fujisawa Y, Yoshino K, Otsuka A, et al. Fluctuations in routine blood count might signal severe immune-related adverse events in melanoma patients treated with nivolumab. J Dermatol Sci. 2017;88(2):225-231.

[177]

Liu C, Shatila M, Mathew A, et al. Role of C-reactive protein in predicting the severity and response of immune-mediated diarrhea and colitis in patients with cancer. J Cancer. 2023;14(10):1913-1919.

[178]

Zou F, Wang X, Glitza Oliva IC, et al. Fecal calprotectin concentration to assess endoscopic and histologic remission in patients with cancer with immune-mediated diarrhea and colitis. J Immunother Cancer. 2021;9(1):e002058.

[179]

Glehr G, Riquelme P, Yang Zhou J, et al. External validation of biomarkers for immune-related adverse events after immune checkpoint inhibition. Front Immunol. 2022;13:1011040.

[180]

Jing Y, Yang J, Johnson DB, Moslehi JJ, Han L. Harnessing big data to characterize immune-related adverse events. Nat Rev Clin Oncol. 2022;19(4):269-280.

[181]

Li J, Wuethrich A, Sina AAI, et al. A digital single-molecule nanopillar SERS platform for predicting and monitoring immune toxicities in immunotherapy. Nat Commun. 2021;12:1087.

[182]

Thompson JA, Schneider BJ, Brahmer J, et al. NCCN guidelines insights: management of immunotherapy-related toxicities, version 1.2020. J Natl Compr Canc Netw. 2020;18(3):230-241.

[183]

Dougan M. Gastrointestinal and hepatic complications of immunotherapy: current management and future perspectives. Curr Gastroenterol Rep. 2020;22(4):15.

[184]

Puzanov I, Diab A, Abdallah K, et al. Managing toxicities associated with immune checkpoint inhibitors: consensus recommendations from the society for immunotherapy of cancer (SITC) toxicity management working group. J Immunother Cancer. 2017;5(1):95.

[185]

Haanen JBAG, Carbonnel F, Robert C, et al. Management of toxicities from immunotherapy: ESMO clinical practice guide-lines for diagnosis, treatment and follow-up. Ann Oncol. 2017;28(Suppl 4):iv119-iv142.

[186]

Collins M, Michot JM, Danlos FX, et al. Inflammatory gastrointestinal diseases associated with PD-1 blockade antibodies. Ann Oncol. 2017;28(11):2860-2865.

[187]

Abu-Sbeih H, Ali FS, Naqash AR, et al. Resumption of immune checkpoint inhibitor therapy after immune-mediated colitis. J Clin Oncol. 2019;37(30):2738-2745.

[188]

Wolfe RM, Ang DC. Biologic therapies for autoimmune and connective tissue diseases. Immunol Allergy Clin North Am. 2017;37(2):283-299.

[189]

Sfikakis PP. The first decade of biologic TNF antagonists in clinical practice: lessons learned, unresolved issues and future directions. Curr Dir Autoimmun. 2010;11:180-210.

[190]

Bergqvist V, Hertervig E, Gedeon P, et al. Vedolizumab treatment for immune checkpoint inhibitor-induced enterocolitis. Cancer Immunol Immunother. 2017;66(5):581-592.

[191]

Dotan I, Ron Y, Yanai H, et al. Patient factors that increase infliximab clearance and shorten half-life in inflammatory bowel disease: a population pharmacokinetic study. Inflamm Bowel Dis. 2014;20(12):2247-2259.

[192]

Brahmer JR, Lacchetti C, Thompson JA. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American society of clinical oncology clinical practice guideline summary. J Oncol Pract. 2018;14(4):247-249.

[193]

Wang Y, Wiesnoski DH, Helmink BA, et al. Fecal microbiota transplantation for refractory immune checkpoint inhibitor-associated colitis. Nature Med. 2018;24(12):1804-1808.

[194]

Monsour EP, Pothen J, Balaraman R. A novel approach to the treatment of pembrolizumab-induced psoriasis exacerbation: a case report. Cureus. 2019;11(10):e5824.

[195]

Johnson D, Patel AB, Uemura MI, et al. IL17A blockade successfully treated psoriasiform dermatologic toxicity from immunotherapy. Cancer Immunol Res. 2019;7(6):860-865.

[196]

Keegan A, Ricciuti B, Garden P, et al. Plasma IL-6 changes correlate to PD-1 inhibitor responses in NSCLC. J Immunother Cancer. 2020;8(2):e000678.

[197]

Richards CD. The enigmatic cytokine oncostatin M and roles in disease. ISRN Inflamm. 2013;2013:512103.

[198]

Hermanns HM. Oncostatin M and interleukin-31: cytokines, receptors, signal transduction and physiology. Cytokine Growth Factor Rev. 2015;26(5):545-558.

[199]

West NR, Hegazy AN, Owens BMJ, et al. Oncostatin M drives intestinal inflammation and predicts response to tumor necrosis factor-neutralizing therapy in patients with inflammatory bowel disease. Nature Med. 2017;23(5):579-589.

[200]

Snyder M, Huang X-Y, Zhang JJ. Signal transducers and activators of transcription 3 (STAT3) directly regulates cytokineinduced fascin expression and is required for breast cancer cell migration. J Biol Chem. 2011;286(45):38886-38893.

[201]

Winder DM, Chattopadhyay A, Muralidhar B, et al. Over-expression of the oncostatin M receptor in cervical squamous cell carcinoma cells is associated with a pro-angiogenic phenotype and increased cell motility and invasiveness. J Pathol. 2011;225:448-462.

[202]

Smigiel JM, Parameswaran N, Jackson MW. Potent EMT and CSC phenotypes are induced by oncostatin-M in pancreatic cancer. Mol Cancer Res. 2017;15(4):478-488.

[203]

Xue G, Li X, Kalim M, et al. Clinical drug screening reveals clofazimine potentiates the efficacy while reducing the toxicity of anti-PD-1 and CTLA4 immunotherapy. Cancer Cell. 2024;42(5):780-796.e6.

RIGHTS & PERMISSIONS

2024 The Author(s). Malignancy Spectrum published by John Wiley & Sons Australia, Ltd on behalf of Higher Education Press.

AI Summary AI Mindmap
PDF (737KB)

597

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/