327
Views
1
CrossRef citations to date
0
Altmetric
Review

Circulating biomarkers of response to immunotherapy and immune-related adverse events

, , &
Pages 855-865 | Received 11 Jul 2022, Accepted 27 Sep 2022, Published online: 07 Oct 2022

References

  • Jiang Y, Zhao X, Fu J, et al. Progress and challenges in precise treatment of tumors with PD-1/PD-L1 blockade. Front Immunol. 2020;11:1664–3224.
  • 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.
  • Lee EY, Kulkarni RP. Circulating biomarkers predictive of tumor response to cancer immunotherapy. Expert Rev Mol Diagn. 2019;19(10):895–904.
  • Bettegowda C, Sausen M, Leary R. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med. 2014;6(224). DOI:10.1126/scitranslmed.3007094
  • Yeh CH, Spurgin J, Ford A, et al. Next-generation sequencing analysis of high-quality and high-quantity cell-free circulating DNA prepared from droplet volumes of patient plasma. J Clin Oncol. 2015;33(15_suppl):abstr e22008.
  • Diaz JL, Bardelli A. Liquid biopsies: genotyping circulating tumor DNA. J clin oncol. 2014;32(6):579–586.
  • Zhang Q, Luo J, Wu S, et al. Prognostic and predictive impact of circulating tumor DNA in patients with advanced cancers treated with immune checkpoint blockade. Cancer Discov. 2020;10(12):1842–1853.
  • Lee JH, Long GV, Boyd S, et al. Circulating tumour DNA predicts response to anti-PD1 antibodies in metastatic melanoma. Ann Oncol. 2017;28(5):1130–1136.
  • Powles T, Carroll D, Chowdhury S, et al. An adaptive, biomarker-directed platform study of durvalumab in combination with targeted therapies in advanced urothelial cancer. Nat Med. 2021;27(5):793–801.
  • Sobczuk P, Kozak K, Kopeć S, et al. The use of ctDNA for BRAF mutation testing in routine clinical practice in patients with advanced melanoma. Cancers (Basel). 2022;14(3):777.
  • Pu M, Chen J, Tao Z, et al. Regulatory network of miRNA on its target: coordination between transcriptional and post-transcriptional regulation of gene expression. Cell Mol Life Sci. 2019;76(3):441–451.
  • Gerin I, Bommer GT, McCoin CS, et al. Roles for mirna-378/378* in adipocyte gene expression and lipogenesis. Am J Physiol Endocrinol Metab. 2010;299(2):E198–206.
  • Gurtan AM, Sharp PA. The role of mirnas in regulating gene expression networks. J Mol Biol. 2013;425(19):3582–3600.
  • Calin GA, Dumitru CD, Shimizu M, et al. Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci USA. 2002;99(24):15524–15529.
  • Kluiver S, Poppema D, de Jong T, et al. BIC and miR-155 are highly expressed in Hodgkin, primary mediastinal and diffuse large B cell lymphomas. J Pathol. 2005;207(2):243–249.
  • Ota A, Tagawa H, Karnan S, et al. Identification and characterization of a novel gene, C13orf25, as a target for 13q31-q32 amplification in malignant lymphoma. Cancer Res. 2004;64(9):3087–3095.
  • Fornari F, Gramantieri L, Ferracin M, et al. MiR-221 controls CDKN1C/p57 and CDKN1B/p27 expression in human hepatocellular carcinoma. Oncogene. 2008;27(43):5651–5661.
  • Vahed S, Barzegari A, Saadat Y, et al. A microRNA isolation method from clinical samples. BioImpacts. 2016;6(1):25–31.
  • Schneegans S, Lück L, Besler K, et al. Pre-analytical factors affecting the establishment of a single tube assay for multiparameter liquid biopsy detection in melanoma patients. Mol Oncol. 2020;14(5):1001–1015.
  • Halvorsen A, Sandhu V, Sprauten M, et al. Circulating microRNAs associated with prolonged overall survival in lung cancer patients treated with nivolumab. Acta Oncologica. 2018;57(9):1225–1231.
  • Huber V, Vallacchi V, Fleming V, et al. Tumor-Derived MicroRNAs induce myeloid suppressor cells and predict immunotherapy resistance in melanoma. J Clin Investi. 2018;128(12):5505–5516.
  • Wang S, Song R, Wang Z, et al. S100A8/A9 in Inflammation. Front Immunol. 2018;9:1298.
  • Gebhardt C, Németh J, Angel P, et al. S100A8 and S100A9 in inflammation and cancer. Biochem Pharmacol. 2006;72(11):1622–1631.
  • Wagner NB, Weide B, Gries M, et al. Tumor microenvironment-derived S100A8/A9 is a novel prognostic biomarker for advanced melanoma patients and during immunotherapy with anti-PD-1 antibodies. J Immunother Cancer. 2019;7(1):343.
  • Wu Z, Jiang D, Huang X, et al. S100A8 as a promising biomarker and oncogenic immune protein in the tumor microenvironment: an integrative pancancer analysis. J Oncol. 2022;1-15:2022.
  • Ş Ş, Ö M, Mersin S, et al. Serum calprotectin (S100A8/A9) levels as a new potential biomarker of treatment response in hodgkin lymphoma. Int J Lab Hematol. 2021;43(4):638–644.
  • Simetić L, Blažičević K, Međugorac K, et al. Relative change in s100 as a biomarker of survival in patients with metastatic melanoma treated with pembrolizumab. Anticancer Res. 2020;40(4):2157–2163.
  • Wagner NB, Forschner A, Leiter U, et al. S100B and LDH as early prognostic markers for response and overall survival in melanoma patients treated with anti-PD-1 or combined anti-PD-1 plus anti-CTLA-4 antibodies. Br J Cancer. 2018;119(3):339–346.
  • Rozeman EA, Versluis JM, Moritz R, et al. Diagnostic performance of early increase in S100B or LDH as outcome predictor for non-responsiveness to anti-PD-1 monotherapy in advanced melanoma. Clin Chim Acta. 2022;533:71–78.
  • Sproston NR, Ashworth JJ. Role of C-reactive protein at sites of inflammation and infection. Front Immunol. 2018;9(754). DOI:10.3389/fimmu.2018.00754
  • Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.
  • Aref H, Refaat S. CRP evaluation in non-small cell lung cancer. Egyptian J Chest Dis Tuberculosis. 2014;63(3):717–722.
  • Villaseñor A, Flatt SW, Marinac C, et al. Postdiagnosis C - reactive protein and breast cancer survivorship: findings from the WHEL study. Cancer Epidemiol Biomarkers Prev. 2013;23(1):189–199.
  • Nimptsch K, Aleksandrova K, Boeing H, et al. Association of CRP genetic variants with blood concentrations of C-reactive protein and colorectal cancer risk. Int J Cancer. 2015;136(5):1181–1192.
  • Shibutani M, Maeda K, Nagahara H, et al. Elevated preoperative serum C-reactive protein levels are associated with poor survival in patients with colorectal cancer. Hepatogastroenterology. 2014;61(136):2236–2240.
  • Badakhshi H, Kaul D, Zhao KL. Association between the inflammatory biomarker, C-reactive protein, and the response to radiochemotherapy in patients with esophageal cancer. Mol Clin Oncol. 2016;4(4):643–647.
  • Huang Y, Feng JF, Liu JS, et al. Prognostic role of serum C-reactive protein in esophageal cancer: a systematic review and meta-analysis. Ther Clin Risk Manag. 2015;11:89–94.
  • Baba H, Kuwabara K, Ishiguro T, et al. C-reactive protein as a significant prognostic factor for stage iv gastric cancer patients. Anticancer Res. 2013;33(12):5591–5596.
  • Oliveira KG, von Zeidler SV, Lamas AZ, et al. Relationship of inflammatory markers and pain in patients with head and neck cancer prior to anticancer therapy. Braz J Med Biol Res. 2014;47(7):600–604.
  • Klümper N, Saal J, Berner F, et al. Creactive protein flare predicts response to checkpoint inhibitor treatment in non-small cell lung cancer. J Immunother Cancer. 2022;10(3):e004024.
  • Chen J, Jing X, Deng X, et al. Prognostic value of serum C-reactive protein in pancreatic cancer: a meta-analysis. Int J Clin Exp Med. 2018;11(11):11789–11796.
  • Inoue D, Ozaka M, Matsuyama M, et al. Prognostic value of neutrophil–lymphocyte ratio and level of C-reactive protein in a large cohort of pancreatic cancer patients: a retrospective study in a single institute in Japan. Japanese J Clin Oncol. 2015;45(1):61–66.
  • Riedl JM, Barth DA, Brueckl WM, et al. C-Reactive Protein (CRP) levels in immune checkpoint inhibitor response and progression in advanced non-small cell lung cancer: a Bi-center study. Cancers (Basel). 2020;12(8):2319.
  • Simeone E, Gentilcore G, Giannarelli D, et al. Immunological and biological changes during ipilimumab treatment and their potential correlation with clinical response and survival in patients with advanced melanoma. Cancer Immunol Immunother. 2014;63(7):675–683.
  • Carter J, Preall J, Grigaityte K, et al. Single T cell sequencing demonstrates the functional role of αβ TCR pairing in cell lineage and antigen specificity. Front Immunol. 2019;10:1664–3224.
  • Laino AS, Woods D, Vassallo M, et al. Serum interleukin-6 and C-reactive protein are associated with survival in melanoma patients receiving immune checkpoint inhibition. J Immunother Cancer. 2020;8(1):e000842.
  • Park JE, Botting R, Conde CD, et al. A cell atlas of human thymic development defines T cell repertoire formation. Science. 2020;367(6480). DOI:10.1126/science.aay3224
  • Postow MA, Manuel M, Wong P, et al. Peripheral T cell receptor diversity is associated with clinical outcomes following ipilimumab treatment in metastatic melanoma. J Immunother Cancer. 2015;3(1):23.
  • Arakawa A, Vollmer S, Tietze J, et al. Clonality of CD4(+) blood T cells predicts longer survival with CTLA4 or PD-1 checkpoint inhibition in advanced melanoma. Front Immunol. 2019;10:1336.
  • Zhang J, Ji Z, Caushi JX, et al. Compartmental analysis of T-cell clonal dynamics as a function of pathologic response to neoadjuvant PD-1 blockade in resectable non–small cell lung cancer. Clin Cancer Res. 2020;26(6):1327–1337.
  • Yao L, Jia G, Lu L, et al. Factors affecting tumor responders and predictive biomarkers of toxicities in cancer patients treated with immune checkpoint inhibitors. Int Immunopharmacol. 2020;85:106628.
  • 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.
  • Eggermont AM, Chiarion-Sileni V, Grob JJ, et al. Prolonged survival in stage III Melanoma with Ipilimumab adjuvant therapy. N Engl J Med. 2016;375(19):1845–1855.
  • Schadendorf D, Wolchok JD, Hodi FS, et al. Efficacy and safety outcomes in patients with advanced melanoma who discontinued treatment with nivolumab and ipilimumab because of adverse events: a pooled analysis of randomized phase II and III trials. J Clin Oncol. 2017;35(34):3807–3814.
  • Kumar V, Chaudhary N, Garg M, et al. Current diagnosis and management of immune related adverse events (irAEs) induced by immune checkpoint inhibitor therapy. Front Pharmacol. 2017;8:49–62.
  • Bertrand A, Kostine M, Barnetche T, et al. Immune related adverse events associated with anti-CTLA-4 antibodies: systematic review and meta-analysis. BMC Med. 2015;13(1):211.
  • Wang PF, Chen Y, Song SY, et al. Immune-related adverse events associated with anti-PD-1/PD-L1 treatment for malignancies: a meta-analysis. Front Pharmacol. 2017;8:730–741.
  • Khoja L, Day D, Wei-Wu Chen T, et al. Tumour- and class-specific patterns of immune-related adverse events of immune checkpoint inhibitors: a systematic review. Ann Oncol. 2017;28(10):2377–2385.
  • Hu Y-B, Zhang Q, Li H-J, et al. Evaluation of rare but severe immune related adverse effects in PD-1 and PD-L1 inhibitors in non-small cell lung cancer: a meta-analysis. Transl Lung Cancer Res. 2017;6(S1):S8–S20.
  • Nobashi T, Baratto L, Reddy SA, et al. Predicting response to immunotherapy by evaluating tumors, lymphoid cell-rich organs, and immune-related adverse events using FDG-PET/CT. Clin Nucl Med. 2019;44(4):e272–e279.
  • Albandar HJ, Fuqua J, Albandar JM, et al. Immune-related adverse events (irAE) in cancer Immune Checkpoint Inhibitors (ICI) and survival outcomes correlation: to rechallenge or not? Cancers (Basel). 2021;13(5):989.
  • Okada N, Kawazoe H, Takechi K, et al. Association between immune-related adverse events and clinical efficacy in patients with melanoma treated with nivolumab: a multicenter retrospective study. Clin Ther. 2019;41(1):59–67.
  • Petrelli F, Grizzi G, Ghidini M, et al. Immune-related adverse events and survival in solid tumors treated with immune checkpoint inhibitors: a systematic review and meta-analysis. J Immunother. 2019;43(1):1–7.
  • Wang DY, Salem JE, Cohen JV, et al. Fatal toxic effects associated with immune checkpoint inhibitors: a systematic review and meta-analysis. JAMA Oncol. 2018;4(12):1721–1728.
  • 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.
  • Genova C, Rossi G, Rijavec E, et al. Releasing the brake: safety profile of immune check-point inhibitors in non-small cell lung cancer. Expert Opin Drug Saf. 2017;16(5):573–585.
  • Uciechowski P, Dempke WCM. Interleukin-6: a Masterplayer in the cytokine network. Oncology. 2020;98(3):131–137.
  • Jordan SC, Choi J, Kim I, et al. Interleukin-6, a cytokine critical to mediation of inflammation, autoimmunity and allograft rejection. Transplantation. 2017;101(1):32–44.
  • Hong DS, Angelo LS, Kurzrock R. Interleukin-6 and its receptor in cancer. Cancer. 2007;110(9):1911–1928.
  • Liu C, Yang L, Xu H, et al. Systematic analysis of IL-6 as a predictive biomarker and desensitizer of immunotherapy responses in patients with non-small cell lung cancer. BMC Med. 2022;20(187). DOI:10.1186/s12916-022-02356-7
  • Valpione S, Pasquali S, Campana LG, et al. Sex and interleukin-6 are prognostic factors for autoimmune toxicity following treatment with anti-CTLA4 blockade. J Transl Med. 2018;16(94). DOI:10.1186/s12967-018-1467-x
  • 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.
  • Tanaka R, Okiyama N, Okune M, et al. Serum level of interleukin-6 is increased in nivolumab-associated psoriasiform dermatitis and tumor necrosis factor-alpha is a biomarker of nivolumab recativity. J Dermatol Sci. 2017;86(1):71–73.
  • McGeachy MJ, Cua DJ, Gaffen SL. The Il-17 family of cytokines in health and disease. Immunity. 2019;50(4):892–906.
  • Schindler K, Harmankaya K, Kuk D, et al. Correlation of absolute and relative eosinophil counts with immune-related adverse events in melanoma patients treated with ipilimumab. J clin oncol. 2014;32(15_suppl):9096.
  • 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.
  • Kurimoto C, Inaba H, Ariyasu H, et al. Predictive and sensitive biomarkers for thyroid dysfunctions during treatment with immune‐checkpoint inhibitors. Cancer Sci. 2020;111(5):1468–1477.
  • Tarhini AA, Zahoor H, Lin Y, et al. Baseline circulating IL-17 predicts toxicity while TGF-beta1 and IL-10 are prognostic of relapse in ipilimumab neoadjuvant therapy of melanoma. J Immunother Cancer. 2015;3(39). DOI:10.1186/s40425-015-0081-1
  • Abraham C, Cho J. Interleukin-23/Th17 pathways and inflammatory bowel disease. Inflamm Bowel Dis. 2009;15(7):1090–1100.
  • Keeley EC, Mehrad B, Strieter RM. CXC chemokines in cancer angiogenesis and metastases. Adv Cancer Res. 2010;106:91–111.
  • Rumble JM, Huber AK, Krishnamoorthy G, et al. Neutrophil-related factors as biomarkers in EAE and MS. J Exp Med. 2015;212(1):23–35.
  • Fujimura T, Kakizaki A, Furudate S, et al. A possible interaction between periostin and CD163 + skin-resident macrophages in pemphigus vulgaris and bullous pemphigoid. Exp Dermatol. 2017;26(12):1193–1198.
  • Buckland J. Rheumatoid arthritis: citrullination alters the inflammatory properties of chemokines in inflammatory arthritis. Nat Rev Rheumatol. 2014;10(8):446.
  • Fujimura T, Sato Y, Tanita K, et al. Serum levels of soluble CD163 and CXCL5 may be predictive markers for immune-related adverse events in patients with advanced melanoma treated with nivolumab: a pilot study. Oncotarget. 2018;9(21):15542–15551.
  • Van Gorp H, Delputte PL, Nauwynck HJ. Scavenger receptor CD163, a Jack-of-all-trades and potential target for cell-directed therapy. Mol Immunol. 2010;47(7–8):1650–1660.
  • Greisen SR, Møller HJ, Stengaard-Pedersen K, et al. Macrophage activity assessed by soluble CD163 in early rheumatoid arthritis: association with disease activity but different response patterns to synthetic and biologic DMARDs. Clin Exp Rheumatol. 2015;33(4):498–502.
  • Fujimura T. Stromal factors as a target for immunotherapy in melanoma and non-melanoma skin cancers. Int J Mol Sci. 2022;23(7):4044.
  • Furudate S, Fujimura T, Kakizaki A, et al. The possible interaction between periostin expressed by cancer stroma and tumor-associated macrophages in developing mycosis fungoides. Exp Dermatol. 2016;25(2):107–112.
  • Gudd CLC, Au L, Triantafyllou E, et al. Activation and transcriptional profile of monocytes and CD8+ T cells are altered in checkpoint inhibitor-related hepatitis. J Hepatol. 2021;75(1):177–189.
  • Diehl A, Yarchoan M, Hopkins A, et al. Relationships between lymphocyte counts and treatment-related toxicities and clinical responses in patients with solid tumors treated with PD-1 checkpoint inhibitors. Oncotarget. 2017;8(69):114268–114280.
  • Nakamura Y, Tanaka R, Maruyama H, et al. Correlation between blood cell count and outcome of melanoma patients treated with anti-PD-1 antibodies. Jpn J Clin Oncol. 2019;49(5):431–437.
  • 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.
  • Da Gama Duarte J, Parakh S, Andrews M, et al. Autoantibodies may predict immune-related toxicity: results from a Phase I study of intralesional Bacillus Calmette-Guérin followed by ipilimumab in patients with advanced metastatic melanoma. Front Immunol. 2018;9:411.
  • Iwama S, De Remigis A, Callahan MK, et al. Pituitary expression of CTLA-4 mediates hypophysitis secondary to administration of CTLA-4 blocking antibody. Sci Transl Med. 2014;6(230):230ra45.
  • Stamatouli AM, Quandt Z, Perdigoto AL, et al. Collateral damage: insulin-dependent diabetes induced with checkpoint inhibitors. Diabetes. 2018;67(8):1471–1480.
  • Umeguchi H, Takenoshita H, Inoue H, et al. Autoimmune-related primary hypoparathyroidism possibly induced by the administration of pembrolizumab: a case report. J Oncol Pract. 2018;14(7):449–451.
  • Lupi I, Brancatella A, Cetani F, et al. Activating antibodies to the calcium-sensing receptor in immunotherapy-induced hypoparathyroidism. J Clin Endocrinol Metab. 2020;105(5):dgaa092.
  • Piranavan P, Li Y, Brown E, et al. Immune checkpoint inhibitor-induced hypoparathyroidism associated with calcium-sensing receptor-activating autoantibodies. J Clin Endocrinol Metab. 2019;104(2):550–556.
  • 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):3583–3590.
  • Oh DY, Cham J, Zhang L, et al. Immune toxicities elicted by CTLA-4 blockade in cancer patients are associated with early diversification of the T-cell repertoire. Cancer Res. 2017;77(6):1322–1330.
  • Subudhi SK, Aparicio A, Gao J, et al. Clonal expansion of CD8 T cells in the systemic circulation precedes development of ipilimumab-induced toxicities. Proc Nat Acad Sci. 2016;113(42):11919–11924.
  • Lozano AX, Chaudhuri AA, Nene A, et al. T cell characteristics associated with toxicity to immune checkpoint blockade in patients with melanoma. Nat Med. 2022;28(2):353–362.
  • 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(1):10391.
  • Botticelli A, Vernocchi P, Marini F, et al. Gut metabolomics profiling of non-small cell lung cancer (NSCLC) patients under immunotherapy treatment. J Transl Med. 2020;18(1):49.
  • Schett A, Rothschild SI, Curioni-Fontecedro A, et al. Predictive impact of antibiotics in patients with advanced non small-cell lung cancer receiving immune checkpoint inhibitors: antibiotics immune checkpoint inhibitors in advanced NSCLC. Cancer Chemother Pharmacol. 2020;85(1):121–131.
  • Routy B, Le Chatelier E, Derosa L, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–97.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.