Trough levels of ipilimumab in serum as a potential biomarker of clinical outcomes for patients with advanced melanoma after treatment with ipilimumab

Yoshinobu Koguchi, Noriko Iwamoto, Takashi Shimada, Shu-Ching Chang, John Cha, Brendan D Curti, Walter J Urba, Brian D Piening, William L Redmond, Yoshinobu Koguchi, Noriko Iwamoto, Takashi Shimada, Shu-Ching Chang, John Cha, Brendan D Curti, Walter J Urba, Brian D Piening, William L Redmond

Abstract

Background: Immune checkpoint blockade (ICB) using anti-CTLA-4 and anti-PD-1/PD-L1 has revolutionized the treatment of advanced cancer. However, ICB is effective for only a small fraction of patients, and biomarkers such as expression of PD-L1 in tumor or serum levels of CXCL11 have suboptimal sensitivity and specificity. Exposure-response (E-R) relationships have been observed with other therapeutic monoclonal antibodies. There are many factors influencing E-R relationships, yet several studies have shown that trough levels of anti-PD-1/PD-L1 correlated with clinical outcomes. However, the potential utility of anti-CTLA-4 levels as a biomarker remains unknown.

Methods: Serum was obtained at trough levels at weeks 7 and 12 (after doses 2 and 4) from patients with advanced melanoma who received ipilimumab alone (3 mg/kg every 3 weeks for four treatments) via an expanded access program (NCT00495066). We have successfully established a proteomics assay to measure the concentration of ipilimumab in serum using an liquid chromatography with tandem mass spectrometry-based nanosurface and molecular-orientation limited proteolysis (nSMOL) approach. Serum samples from 38 patients were assessed for trough levels of ipilimumab by the nSMOL assay.

Results: We found that trough levels of ipilimumab were higher in patients who developed immune-related adverse events but did not differ based on the presence or absence of disease progression. We found that patients with higher trough levels of ipilimumab had better overall survival when grouped based on ipilimumab trough levels. Trough levels of ipilimumab were inversely associated with pretreatment serum levels of CXCL11, a predictive biomarker we previously identified, and soluble CD25 (sCD25), a prognostic biomarker for advanced melanoma, as well as C reactive protein (CRP) and interleukin (IL)-6 levels at week 7.

Conclusions: Our results suggest that trough levels of ipilimumab may be a useful biomarker for the long-term survival of patients with advanced melanoma treated with ipilimumab. The association of ipilimumab trough levels with pretreatment serum levels of CXCL11 and sCD25 is suggestive of a baseline-driven E-R relationship, and the association of ipilimumab trough levels with on-treatment levels of CRP and IL-6 is suggestive of response-driven E-R relationship. Our findings highlight the potential utility of trough levels of ipilimumab as a biomarker.

Trial registration number: NCT00495066.

Keywords: CTLA-4 antigen; biomarkers; immunotherapy; melanoma; translational medical research; tumor.

Conflict of interest statement

Competing interests: YK: research support from Bristol Myers Squibb (BMS), GlaxoSmithKline, and Shimadzu. NI and TS: employees of Shimadzu Scientific Instruments. BDC: research support from AstraZeneca, Clinigen, and Galectin Therapeutics; patents/royalties: none; advisory boards: Clinigen, Nektar, and Merck. WJU: research support from BMS; royalties: data safety monitoring board for AstraZeneca. BDP: research support from Heat Biologics and Shimadzu; advisory boards: Bayer and UnitedHealthcare. WLR: research support from Galectin Therapeutics, BMS, GlaxoSmithKline, MiNA Therapeutics, Inhibrx, Veana Therapeutics, Aeglea Biotherapeutics, Shimadzu, OncoSec, and Calibr; patents/licensing fees: Galectin Therapeutics; advisory boards: Nektar Therapeutics and Vesselon.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Establishment of nSMOL assay for ipilimumab detection. (A) ClustalW sequence alignment of ipilimumab, nivolumab, avelumab, and ramucirumab. The black area indicates the common sequence for all antibodies. The red line describes the selected signature peptide for ipilimumab quantification. The peptides marked with blue and green lines did not meet the validation criteria. (B) Calibration curve of ipilimumab-spiked serum analyzed by nSMOL method. (C) MRM chromatograms of the signature ipilimumab peptide TGWLGPFDYWGQGTLVTVSSASTK. The peptide peak was observed at a retention time at 6.03 min (red arrow). Representative chromatograms of 853.5→780.4 transition from non-spiked (left) and ipilimumab-spiked (right) serum are shown. MRM, multiple reaction monitoring; nSMOL, nanosurface and molecular-orientation limited proteolysis.
Figure 2
Figure 2
Trough levels of ipilimumab were associated with irAE status but not with clinical benefit. (A) The trough levels at weeks 7 and 12 of ipilimumab in individual patients were compared. (B) The trough levels of ipilimumab at week 7 were compared between patients with or without PD (n=27 and n=10, respectively). (C) The trough levels of ipilimumab at week 12 were compared between patients with or without PD (n=22 and n=10, respectively). (D) The trough levels of ipilimumab at week 7 were compared between patients who did not develop irAE (no irAE) and those who developed irAE G1–4 (n=10 and n=27, respectively). (E) The trough levels of ipilimumab at week 12 were compared between patients who did not develop irAE (no irAE) and those who developed irAE G1–4 (n=8 and n=24, respectively). CR, complete response; ipi, ipilimumab; irAE, immune-related adverse event; irAE G1–4, grades 1–4 irAE; PD, disease progression; PR, partial response; SD, stable disease.
Figure 3
Figure 3
Patients with lower serum trough levels of ipilimumab had worse OS. Curves for OS obtained by applying a median value of ipilimumab trough levels as cut points for week 7 (A) and week 12 (B). The survival curves are significantly different by the log-rank test (A, p=0.0021; B, p=0.0177). ipi, ipilimumab; OS, overall survival.
Figure 4
Figure 4
Relationship between trough levels of ipilimumab and biomarkers of clinical outcomes and inflammation. The relationship between pre-treatment levels of CXCL11 (A), sCD25 (B), or LDH (C) and the trough levels of ipilimumab at week seven was shown. The relationship between levels of CRP (D) or IL-6 (E) at week seven and the trough levels of ipilimumab at week seven are shown. CRP, C reactive protein; ipi, ipilimumab; IL, interleukin; pre-tx, pre-treatment; sCD25, soluble CD25.

References

    1. Wei SC, Duffy CR, Allison JP. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov 2018;8:1069–86. 10.1158/-18-0367
    1. Emens LA, Ascierto PA, Darcy PK, et al. . Cancer immunotherapy: opportunities and challenges in the rapidly evolving clinical landscape. Eur J Cancer 2017;81:116–29. 10.1016/j.ejca.2017.01.035
    1. Ku GY, Yuan J, Page DB, et al. . Single-institution experience with ipilimumab in advanced melanoma patients in the compassionate use setting: lymphocyte count after 2 doses correlates with survival. Cancer 2010;116:1767–75. 10.1002/cncr.24951
    1. Ng Tang D, Shen Y, Sun J, et al. . Increased frequency of ICOS+ CD4 T cells as a pharmacodynamic biomarker for anti-CTLA-4 therapy. Cancer Immunol Res 2013;1:229–34. 10.1158/2326-6066.CIR-13-0020
    1. Snyder A, Makarov V, Merghoub T, et al. . Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med 2014;371:2189–99. 10.1056/NEJMoa1406498
    1. Gao J, Shi LZ, Zhao H, et al. . Loss of IFN-γ pathway genes in tumor cells as a mechanism of resistance to anti-CTLA-4 therapy. Cell 2016;167:397–404. 10.1016/j.cell.2016.08.069
    1. Koguchi Y, Hoen HM, Bambina SA, et al. . Serum immunoregulatory proteins as predictors of overall survival of metastatic melanoma patients treated with ipilimumab. Cancer Res 2015;75:5084–92. 10.1158/0008-5472.CAN-15-2303
    1. Tumeh PC, Harview CL, Yearley JH, et al. . PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014;515:568–71. 10.1038/nature13954
    1. 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;366:2443–54. 10.1056/NEJMoa1200690
    1. Le DT, Uram JN, Wang H, et al. . PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med 2015;372:2509–20. 10.1056/NEJMoa1500596
    1. Ayers M, Lunceford J, Nebozhyn M, et al. . IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest 2017;127:2930–40. 10.1172/JCI91190
    1. Huang AC, Postow MA, Orlowski RJ, et al. . T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature 2017;545:60–5. 10.1038/nature22079
    1. Dai HI, Vugmeyster Y, Mangal N. Characterizing exposure-response relationship for therapeutic monoclonal antibodies in Immuno-Oncology and beyond: challenges, perspectives, and prospects. Clin Pharmacol Ther 2020;108:1156–70. 10.1002/cpt.1953
    1. Basak EA, Koolen SLW, Hurkmans DP, et al. . Correlation between nivolumab exposure and treatment outcomes in non-small-cell lung cancer. Eur J Cancer 2019;109:12–20. 10.1016/j.ejca.2018.12.008
    1. Hurkmans DP, Basak EA, van Dijk T, et al. . A prospective cohort study on the pharmacokinetics of nivolumab in metastatic non-small cell lung cancer, melanoma, and renal cell cancer patients. J Immunother Cancer 2019;7:192. 10.1186/s40425-019-0669-y
    1. Agrawal S, Feng Y, Roy A, et al. . Nivolumab dose selection: challenges, opportunities, and lessons learned for cancer immunotherapy. J Immunother Cancer 2016;4:72. 10.1186/s40425-016-0177-2
    1. Turner DC, Kondic AG, Anderson KM, et al. . Pembrolizumab exposure-response assessments challenged by association of cancer cachexia and catabolic clearance. Clin Cancer Res 2018;24:5841–9. 10.1158/1078-0432.CCR-18-0415
    1. Li H, Yu J, Liu C, et al. . Time dependent pharmacokinetics of pembrolizumab in patients with solid tumor and its correlation with best overall response. J Pharmacokinet Pharmacodyn 2017;44:403–14. 10.1007/s10928-017-9528-y
    1. Baverel PG, Dubois VFS, Jin CY, et al. . Population pharmacokinetics of Durvalumab in cancer patients and association with longitudinal biomarkers of disease status. Clin Pharmacol Ther 2018;103:631–42. 10.1002/cpt.982
    1. Wilkins JJ, Brockhaus B, Dai H, et al. . Time-Varying clearance and impact of disease state on the pharmacokinetics of Avelumab in Merkel cell carcinoma and urothelial carcinoma. CPT Pharmacometrics Syst Pharmacol 2019;8:415–27. 10.1002/psp4.12406
    1. Bajaj G, Wang X, Agrawal S, et al. . Model-Based population pharmacokinetic analysis of nivolumab in patients with solid tumors. CPT Pharmacometrics Syst Pharmacol 2017;6:58–66. 10.1002/psp4.12143
    1. Feng Y, Wang X, Bajaj G, et al. . Nivolumab exposure-response analyses of efficacy and safety in previously treated squamous or Nonsquamous non-small cell lung cancer. Clin Cancer Res 2017;23:5394–405. 10.1158/1078-0432.CCR-16-2842
    1. Feng Y, Roy A, Masson E, et al. . Exposure-response relationships of the efficacy and safety of ipilimumab in patients with advanced melanoma. Clin Cancer Res 2013;19:3977–86. 10.1158/1078-0432.CCR-12-3243
    1. Iwamoto N, Shimada T, Umino Y, et al. . Selective detection of complementarity-determining regions of monoclonal antibody by limiting protease access to the substrate: nano-surface and molecular-orientation limited proteolysis. Analyst 2014;139:576–80. 10.1039/C3AN02104A
    1. Iwamoto N, Shimada T, Terakado H, et al. . Validated LC-MS/MS analysis of immune checkpoint inhibitor nivolumab in human plasma using a Fab peptide-selective quantitation method: nano-surface and molecular-orientation limited (nSMOL) proteolysis. J Chromatogr B Analyt Technol Biomed Life Sci 2016;1023-1024:9–16. 10.1016/j.jchromb.2016.04.038
    1. Iwamoto N, Yonezawa A, Matsubara K, et al. . Acceleration of nano-surface and molecular-orientation limited (nSMOL) proteolysis with acidified reduction pretreatment for quantification of tocilizumab. J Pharm Biomed Anal 2019;164:467–74. 10.1016/j.jpba.2018.11.019
    1. Feng Y, Masson E, Dai D, et al. . Model-based clinical pharmacology profiling of ipilimumab in patients with advanced melanoma. Br J Clin Pharmacol 2014;78:106–17. 10.1111/bcp.12323
    1. Ballman KV. Biomarker: predictive or prognostic? J Clin Oncol 2015;33:3968–71. 10.1200/JCO.2015.63.3651
    1. Mouawad R, Spano J-P, Khayat D. Old and new serological biomarkers in melanoma: where we are in 2009. Melanoma Res 2010;20:67–76. 10.1097/CMR.0b013e328335a8c1
    1. Laws LH, Parker CE, Cherala G, et al. . Inflammation causes resistance to Anti-CD20-Mediated B cell depletion. Am J Transplant 2016;16:3139–49. 10.1111/ajt.13902
    1. Baverel P, Roskos L, Tatipalli M, et al. . Exposure-Response analysis of overall survival for tremelimumab in unresectable malignant mesothelioma: the confounding effect of disease status. Clin Transl Sci 2019;12:450–8. 10.1111/cts.12633
    1. Curti BD, Koguchi Y, Leidner RS, et al. . Enhancing clinical and immunological effects of anti-PD-1 with belapectin, a galectin-3 inhibitor. J Immunother Cancer 2021;9:e002371. 10.1136/jitc-2021-002371
    1. Dreesen E, Bossuyt P, Mulleman D, et al. . Practical recommendations for the use of therapeutic drug monitoring of biopharmaceuticals in inflammatory diseases. Clin Pharmacol 2017;9:101–11. 10.2147/CPAA.S138414
    1. Vermeire S, Dreesen E, Papamichael K, et al. . How, when, and for whom should we perform therapeutic drug monitoring? Clin Gastroenterol Hepatol 2020;18:1291–9. 10.1016/j.cgh.2019.09.041
    1. Centanni M, Moes DJAR, Trocóniz IF, et al. . Clinical pharmacokinetics and pharmacodynamics of immune checkpoint inhibitors. Clin Pharmacokinet 2019;58:835–57. 10.1007/s40262-019-00748-2
    1. Coss CC, Clinton SK, Phelps MA. Cachectic cancer patients: immune to checkpoint inhibitor therapy? Clin Cancer Res 2018;24:5787–9. 10.1158/1078-0432.CCR-18-1847
    1. Dromain C, Beigelman C, Pozzessere C, et al. . Imaging of tumour response to immunotherapy. Eur Radiol Exp 2020;4:2. 10.1186/s41747-019-0134-1
    1. Das S, Johnson DB. Immune-related adverse events and anti-tumor efficacy of immune checkpoint inhibitors. J Immunother Cancer 2019;7:306. 10.1186/s40425-019-0805-8
    1. 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;77:1322–30. 10.1158/0008-5472.CAN-16-2324
    1. 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:e000842. 10.1136/jitc-2020-000842
    1. Wochner RD. Hypercatabolism of normal IgG; an unexplained immunoglobulin abnormality in the connective tissue diseases. J Clin Invest 1970;49:454–64. 10.1172/JCI106254
    1. Ovacik M, Lin K. Tutorial on monoclonal antibody pharmacokinetics and its considerations in early development. Clin Transl Sci 2018;11:540–52. 10.1111/cts.12567
    1. Pyzik M, Sand KMK, Hubbard JJ, et al. . The neonatal Fc receptor (FcRn): a misnomer? Front Immunol 2019;10:1540. 10.3389/fimmu.2019.01540
    1. Martins JP, Kennedy PJ, Santos HA, et al. . A comprehensive review of the neonatal Fc receptor and its application in drug delivery. Pharmacol Ther 2016;161:22–39. 10.1016/j.pharmthera.2016.03.007
    1. Pyzik M, Rath T, Lencer WI, et al. . Fcrn: the architect behind the immune and nonimmune functions of IgG and albumin. J Immunol 2015;194:4595–603. 10.4049/jimmunol.1403014
    1. Oldham RJ, Mockridge CI, James S, et al. . FcγRII (CD32) modulates antibody clearance in NOD SCID mice leading to impaired antibody-mediated tumor cell deletion. J Immunother Cancer 2020;8:e000619. 10.1136/jitc-2020-000619
    1. Li F, Ulrich ML, Shih VF-S, et al. . Mouse strains influence clearance and efficacy of antibody and antibody-drug conjugate via Fc-FcγR interaction. Mol Cancer Ther 2019;18:780–7. 10.1158/1535-7163.MCT-18-0977
    1. Anania JC, Chenoweth AM, Wines BD, et al. . The human FcγRII (CD32) family of leukocyte FcR in health and disease. Front Immunol 2019;10:464. 10.3389/fimmu.2019.00464
    1. Engblom C, Pfirschke C, Pittet MJ. The role of myeloid cells in cancer therapies. Nat Rev Cancer 2016;16:447–62. 10.1038/nrc.2016.54
    1. Dahan R, Sega E, Engelhardt J, et al. . FcγRs modulate the anti-tumor activity of antibodies targeting the PD-1/PD-L1 axis. Cancer Cell 2015;28:285–95. 10.1016/j.ccell.2015.08.004
    1. Lo Russo G, 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;25:989–99. 10.1158/1078-0432.CCR-18-1390
    1. Arlauckas SP, Garris CS, Kohler RH, et al. . In vivo imaging reveals a tumor-associated macrophage-mediated resistance pathway in anti-PD-1 therapy. Sci Transl Med 2017;910.1126/scitranslmed.aal3604
    1. Arce Vargas F, Furness AJS, Litchfield K, et al. . Fc effector function contributes to the activity of human anti-CTLA-4 antibodies. Cancer Cell 2018;33:649–63. 10.1016/j.ccell.2018.02.010
    1. Vogelpoel LTC, Baeten DLP, de Jong EC, et al. . Control of cytokine production by human Fc gamma receptors: implications for pathogen defense and autoimmunity. Front Immunol 2015;6:79. 10.3389/fimmu.2015.00079
    1. Neubert NJ, Schmittnaegel M, Bordry N, et al. . T cell–induced CSF1 promotes melanoma resistance to PD1 blockade. Sci Transl Med 2018;1010.1126/scitranslmed.aan3311

Source: PubMed

3
Prenumerera