356
Views
0
CrossRef citations to date
0
Altmetric
ORIGINAL RESEARCH

Predictive Nomogram for Hyperprogressive Disease During Anti-PD-1/PD-L1 Treatment in Patients with Advanced Non-Small Cell Lung Cancer

ORCID Icon, ORCID Icon, , , , , , , , & show all
Pages 1-16 | Received 18 May 2022, Accepted 29 Jul 2022, Published online: 04 Jan 2023

References

  • Topalian SL, Taube JM, Anders RA, Pardoll DM. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer. 2016;16(5):275–287. doi:10.1038/nrc.2016.36
  • 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):1853–1862. doi:10.1016/S0140-6736(17)31601-X
  • Betof WA, Palmer JS, Shoushtari AN, et al. Long-Term outcomes and responses to retreatment in patients with melanoma treated with PD-1 blockade. J Clin Oncol. 2020;38(15):1655–1663. doi:10.1200/JCO.19.01464
  • Socinski MA, Jotte RM, Cappuzzo F, et al. Atezolizumab for First-Line treatment of metastatic nonsquamous NSCLC. N Engl J Med. 2018;378(24):2288–2301. doi:10.1056/NEJMoa1716948
  • Rittmeyer A, Barlesi F, Waterkamp D, et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet. 2017;389(10066):255–265. doi:10.1016/S0140-6736(16)32517-X
  • Bellmunt J, de Wit R, Vaughn DJ, et al. Pembrolizumab as Second-Line therapy for advanced urothelial carcinoma. N Engl J Med. 2017;376(11):1015–1026. doi:10.1056/NEJMoa1613683
  • Powles T, Duran I, van der Heijden MS, et al. Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): a multicentre, open-label, phase 3 randomised controlled trial. Lancet. 2018;391(10122):748–757. doi:10.1016/S0140-6736(17)33297-X
  • Harrington KJ, Ferris RL, Blumenschein GJ, et al. Nivolumab versus standard, single-agent therapy of investigator’s choice in recurrent or metastatic squamous cell carcinoma of the head and neck (CheckMate 141): health-related quality-of-life results from a randomised, phase 3 trial. Lancet Oncol. 2017;18(8):1104–1115. doi:10.1016/S1470-2045(17)30421-7
  • Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020;70(1):7–30. doi:10.3322/caac.21590
  • Dawe DE, Harlos CH, Juergens RA. Immuno-oncology—the new paradigm of lung cancer treatment. Curr Oncol. 2020;27(Suppl 12):S78–S86. doi:10.3747/co.27.5183
  • Larkin J, Minor D, D’Angelo S, et al. Overall survival in patients with advanced melanoma who received nivolumab versus investigator’s choice chemotherapy in CheckMate 037: a randomized, controlled, Open-Label Phase III trial. J Clin Oncol. 2018;36(4):383–390. doi:10.1200/JCO.2016.71.8023
  • Gray JE, Villegas A, Daniel D, et al. Three-year overall survival with durvalumab after chemoradiotherapy in stage III NSCLC—update from PACIFIC. J Thorac Oncol. 2020;15(2):288–293. doi:10.1016/j.jtho.2019.10.002
  • Chung HC, Piha-Paul SA, Lopez-Martin J, et al. Pembrolizumab after two or more lines of previous therapy in patients with recurrent or metastatic SCLC: results from the KEYNOTE-028 and KEYNOTE-158 studies. J Thorac Oncol. 2020;15(4):618–627. doi:10.1016/j.jtho.2019.12.109
  • Travert C, Barlesi F, Greillier L, Tomasini P. Immune oncology biomarkers in lung cancer: an overview. Curr Oncol Rep. 2020;22(11):107. doi:10.1007/s11912-020-00970-3
  • Wang X, Wang F, Zhong M, Yarden Y, Fu L. The biomarkers of hyperprogressive disease in PD-1/PD-L1 blockage therapy. Mol Cancer. 2020;19(1):81. doi:10.1186/s12943-020-01200-x
  • Champiat S, Dercle L, Ammari S, et al. Hyperprogressive disease is a new pattern of progression in cancer patients treated by Anti-PD-1/PD-L1. Clin Cancer Res. 2017;23(8):1920–1928. doi:10.1158/1078-0432.CCR-16-1741
  • Ferrara R, Mezquita L, Texier M, et al. Hyperprogressive disease in patients with advanced Non-Small cell lung cancer treated with PD-1/PD-L1 inhibitors or with Single-Agent chemotherapy. JAMA Oncol. 2018;4(11):1543–1552. doi:10.1001/jamaoncol.2018.3676
  • Lin Z, Zhang Y, Zhang L. Construction of an integrated prognostic classifier model for predicting the efficacy of immune checkpoint inhibitor therapy in non-small cell lung cancer. Cancer Commun. 2020;40(8):370–373. doi:10.1002/cac2.12070
  • Schwartz LH, Litiere S, de Vries E, et al. RECIST 1.1-Update and clarification: from the RECIST committee. Eur J Cancer. 2016;62:132–137. doi:10.1016/j.ejca.2016.03.081
  • Fang W, Chen Y, Sheng J, et al. Association between PD-L1 expression on tumour-infiltrating lymphocytes and overall survival in patients with gastric cancer. J Cancer. 2017;8(9):1579–1585. doi:10.7150/jca.18729
  • Ferte C, Fernandez M, Hollebecque A, et al. Tumor growth rate is an early indicator of antitumor drug activity in Phase I clinical trials. Clin Cancer Res. 2014;20(1):246–252. doi:10.1158/1078-0432.CCR-13-2098
  • Saada-Bouzid E, Defaucheux C, Karabajakian A, et al. Hyperprogression during anti-PD-1/PD-L1 therapy in patients with recurrent and/or metastatic head and neck squamous cell carcinoma. Ann Oncol. 2017;28(7):1605–1611. doi:10.1093/annonc/mdx178
  • Kato S, Goodman A, Walavalkar V, Barkauskas DA, Sharabi A, Kurzrock R. Hyperprogressors after immunotherapy: analysis of genomic alterations associated with accelerated growth rate. Clin Cancer Res. 2017;23(15):4242–4250. doi:10.1158/1078-0432.CCR-16-3133
  • Wu Q, Xu Y, Zhang KJ, Jiang SM, Zhou Y, Zhao Y. A clinical model for the prediction of acute exacerbation risk in patients with idiopathic pulmonary fibrosis. Biomed Res Int. 2020;2020:8848919. doi:10.1155/2020/8848919
  • Kim CG, Kim KH, Pyo KH, et al. Hyperprogressive disease during PD-1/PD-L1 blockade in patients with non-small-cell lung cancer. Ann Oncol. 2019;30(7):1104–1113. doi:10.1093/annonc/mdz123
  • Lo RG, Moro M, Sommariva M, et al. Antibody-Fc/FcR interaction on macrophages as a mechanism for hyperprogressive disease in non-small cell lung cancer subsequent to PD-1/PD-L1 blockade. Clin Cancer Res. 2019;25(3):989–999. doi:10.1158/1078-0432.CCR-18-1390
  • Locker GJ, Kapiotis S, Veitl M, et al. Activation of endothelium by immunotherapy with interleukin-2 in patients with malignant disorders. Br J Haematol. 1999;105(4):912–919. doi:10.1046/j.1365-2141.1999.01453.x
  • Richard V, Bernier M, Themelin L, Bron D, Sculier JP. Blood coagulation abnormalities during adoptive immunotherapy with interleukin-2 (r-Met Hu IL-2 [ala 125]). Ann Oncol. 1991;2(1):67–68. doi:10.1093/oxfordjournals.annonc.a057827
  • Curti BD, Longo DL, Ochoa AC, et al. Treatment of cancer patients with ex vivo anti-CD3-activated killer cells and interleukin-2. J Clin Oncol. 1993;11(4):652–660. doi:10.1200/JCO.1993.11.4.652
  • Becker JC, Winkler B, Klingert S, Brocker EB. Antiphospholipid syndrome associated with immunotherapy for patients with melanoma. Cancer Am Cancer Soc. 1994;73(6):1621–1624. doi:10.1002/1097-0142(19940315)73:6<1621::aid-cncr2820730613>3.0.co;2-e
  • Buechner J, Grupp SA, Hiramatsu H, et al. Practical guidelines for monitoring and management of coagulopathy following tisagenlecleucel CAR T-cell therapy. Blood Adv. 2021;5(2):593–601. doi:10.1182/bloodadvances.2020002757
  • Tawbi HA, Burgess M, Bolejack V, et al. Pembrolizumab in advanced soft-tissue sarcoma and bone sarcoma (SARC028): a multicentre, two-cohort, single-arm, open-label, Phase 2 trial. Lancet Oncol. 2017;18(11):1493–1501. doi:10.1016/S1470-2045(17)30624-1
  • Kida W, Nakaya M, Ito A, Kozai Y, Bingo M. A case of acquired factor v inhibitor following nivolumab administration. Cureus. 2022;14(1):e21670. doi:10.7759/cureus.21670
  • Abbas M, Kassim SA, Wang ZC, Shi M, Hu Y, Zhu HL. Clinical evaluation of plasma coagulation parameters in patients with advanced-stage non-small cell lung cancer treated with palliative chemotherapy in China. Int J Clin Pract. 2020;74(12):e13619. doi:10.1111/ijcp.13619
  • Shu YJ, Weng H, Bao RF, et al. Clinical and prognostic significance of preoperative plasma hyperfibrinogenemia in gallbladder cancer patients following surgical resection: a retrospective and in vitro study. Bmc Cancer. 2014;14:566. doi:10.1186/1471-2407-14-566
  • Geng C, Yang G, Wang H, Zhang Z, Zhou H, Chen W. The prognostic role of prothrombin time and activated partial thromboplastin time in patients with newly diagnosed multiple myeloma. Biomed Res Int. 2021;2021:6689457. doi:10.1155/2021/6689457
  • Suzuki T, Shimada H, Nanami T, et al. Hyperfibrinogenemia is associated with inflammatory mediators and poor prognosis in patients with gastric cancer. Surg Today. 2016;46(12):1394–1401. doi:10.1007/s00595-016-1339-z
  • Dirix LY, Oeyen S, Buys A, et al. Coagulation/fibrinolysis and circulating tumor cells in patients with advanced breast cancer. Breast Cancer Res Treat. 2022;192(3):583–591. doi:10.1007/s10549-021-06484-1
  • Huang W, Li C, Wang Z, et al. Decreased serum albumin level indicates poor prognosis of COVID-19 patients: hepatic injury analysis from 2,623 hospitalized cases. Sci China Life Sci. 2020;63(11):1678–1687. doi:10.1007/s11427-020-1733-4
  • Kas B, Talbot H, Ferrara R, et al. Clarification of definitions of hyperprogressive disease during immunotherapy for Non-Small cell lung cancer. JAMA Oncol. 2020;6(7):1039–1046. doi:10.1001/jamaoncol.2020.1634
  • Matos I, Martin-Liberal J, Garcia-Ruiz A, et al. Capturing Hyperprogressive disease with immune checkpoint inhibitors using RECIST 1.1 criteria. Clin Cancer Res. 2019. doi:10.1158/1078-0432.CCR-19-2226
  • Klempner SJ, Fabrizio D, Bane S, et al. Tumor mutational burden as a predictive biomarker for response to immune checkpoint inhibitors: a review of current evidence. Oncologist. 2019. doi:10.1634/theoncologist.2019-0244
  • Martins F, Sofiya L, Sykiotis GP, et al. Adverse effects of immune-checkpoint inhibitors: epidemiology, management and surveillance. Nat Rev Clin Oncol. 2019;16(9):563–580. doi:10.1038/s41571-019-0218-0