Prospective development and validation of a liquid immune profile-based signature (LIPS) to predict response of patients with recurrent/metastatic cancer to immune checkpoint inhibitors

Jian-Guo Zhou, Anna-Jasmina Donaubauer, Benjamin Frey, Ina Becker, Sandra Rutzner, Markus Eckstein, Roger Sun, Hu Ma, Philipp Schubert, Claudia Schweizer, Rainer Fietkau, Eric Deutsch, Udo Gaipl, Markus Hecht, Jian-Guo Zhou, Anna-Jasmina Donaubauer, Benjamin Frey, Ina Becker, Sandra Rutzner, Markus Eckstein, Roger Sun, Hu Ma, Philipp Schubert, Claudia Schweizer, Rainer Fietkau, Eric Deutsch, Udo Gaipl, Markus Hecht

Abstract

Background: The predictive power of novel biological markers for treatment response to immune checkpoint inhibitors (ICI) is still not satisfactory for the majority of patients with cancer. One should identify valid predictive markers in the peripheral blood, as this is easily available before and during treatment. The current interim analysis of patients of the ST-ICI cohort therefore focuses on the development and validation of a liquid immune profile-based signature (LIPS) to predict response of patients with metastatic cancer to ICI targeting the programmed cell death protein 1 (PD-1)/programmed cell death-ligand 1 (PD-L1) axis.

Methods: A total of 104 patients were prospectively enrolled. 54 immune cell subsets were prospectively analyzed in patients' peripheral blood by multicolor flow cytometry before treatment with ICI (pre-ICI; n=89), and after the first application of ICI (n=65). Pre-ICI, patients were randomly allocated to a training (n=56) and a validation cohort (n=33). Univariate Cox proportional hazards regression analysis and least absolute shrinkage and selection operator Cox model were used to create a predictive immune signature, which was also checked after the first ICI, to consider the dynamics of changes in the immune status.

Results: Whole blood samples were provided by 89 patients pre-ICI and by 65 patients after the first ICI. We identified a LIPS which is based on five immune cell subtypes: CD14high monocytes, CD8+/PD-1+ T cells, plasmacytoid dendritic cells, neutrophils, and CD3+/CD56+/CD16+ natural killer (NK)T cells. The signature achieved a high accuracy (C-index 0.74 vs 0.71) for predicting overall survival (OS) benefit in both the training and the validation cohort. In both cohorts, the low-risk group had significantly longer OS than the high-risk group (HR 0.26, 95% CI 0.12 to 0.56, p=0.00025; HR 0.30, 95% CI 0.10 to 0.91, p=0.024, respectively). Regarding the whole cohort, LIPS also predicted progression-free survival (PFS). The identified LIPS was not affected by clinicopathological features with the exception of brain metastases. NKT cells and neutrophils of the LIPS can be used as dynamic predictive biomarkers for OS and PFS after first administration of the ICI.

Conclusion: Our study identified a predictive LIPS for survival of patients with cancer treated with PD-1/PD-L1 ICI, which is based on immune cell subsets in the peripheral whole blood.

Trial registration number: NCT03453892.

Keywords: biomarkers; immunotherapy; programmed cell death 1 receptor; tumor; tumor biomarkers; tumor microenvironment.

Conflict of interest statement

Competing interests: MH reports conflict of interest with Merck Serono (advisory boards, honoraria for lectures, travel grants, research funding); MSD (advisory boards, travel grants, research funding); AstraZeneca (research funding); Novartis (research funding); BMS (advisory boards, honoraria for lectures); Teva (travel grants). USG received support for presentation activities for Dr Sennewald Medizintechnik GmbH, has received support for investigator initiated clinical studies (IITs) from MSD and AstraZeneca and contributed at Advisory Boards Meetings of AstraZeneca and Bristol-Myers Squibb. JGZ received support from AstraZeneca (travel grants). HM reports conflict of interest with Merck Serono (advisory boards, honoraria for lectures, travel grants); MSD (advisory boards, honoraria for lectures, travel grants); AstraZeneca (advisory boards, honoraria for lectures, travel grants); BMS (advisory boards, honoraria for lectures, travel grants). RS received travel and accommodation expenses from AstraZeneca. ED reports grants and honoraria from ROCHE GENENTECH, grants from SERVIER, grants and honoraria from ASTRAZENECA, MERCK SERONO, BMS and MSD, outside the submitted work. SR conflict of interest with AstraZeneca (research funding); MSD (research funding). ME conflict of interest with Diaceutics (employment, honoraria, advisory role, speakers’ bureau, travel expenses); AstraZeneca (honoraria, advisory role, speakers’ bureau, travel expenses); Roche (honoraria, travel expenses); MSD (honoraria, speakers’ bureau); GenomicHealth (honoraria, advisory role, speakers bureau, travel expenses); Astellas (honoraria, speakers’ bureau); Janssen-Cilag (honoraria, advisory role, research funding, travel expenses); Stratifyer (research funding, patents). RF conflict of interest with MSD (honoraria, advisory role, research funding, travel expenses); Fresenius (honoraria); BrainLab (honoraria); AstraZeneca (honoraria, advisory role, research funding, travel expenses); Merck Serono (advisory role, research funding, travel expenses); Novocure (advisory role, speakers’ bureau, research funding); Sennewald (speakers’ bureau, travel expenses).

© 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
Research design (A) and flowchart (B) for the identification of a predictive signature by immunophenotyping (IPT) for patients with metastatic cancer treated with immune checkpoint inhibitors (ICI). ROC, receiver operating characteristic.
Figure 2
Figure 2
Characteristics of the liquid immune profile-based signature (LIPS) in the training and validation cohorts. (A) The risk score of each patient with metastatic cancer (patient ID) treated with immune checkpoint inhibitors (ICI) in the training cohort. (B) Overall survival and survival status of patients with metastatic cancer in the training cohort. (C) Heat map of immune cell counts of patients with metastatic cancer in the training cohort. (D) The risk score of each patient with metastatic cancer treated with ICI in the validation cohort. (E) Overall survival and survival status of patients with metastatic cancer in the validation cohort. (F) Heat map of immune cell counts of patients with metastatic cancer in the validation cohort. NKT cells, natural killer T cells; pDCs, plasmacytoid dendritic cells; PD-1, programmed cell death protein 1.
Figure 3
Figure 3
The ability of the liquid immune profile-based signature (LIPS) to predict the overall survival (OS) in the training and validation cohorts. (A) The OS in training cohort stratified by the LIPS into high-risk and low-risk with the p value. (B) Time-dependent receiver operating characteristic (ROC) curves of LIPS in the training cohort. (C) The OS in validation cohort stratified by the LIPS into high-risk and low-risk with the p value. (D) Time-dependent ROC curves of LIPS in the validation cohort. AUC, area under the ROC curve.
Figure 4
Figure 4
Liquid immune profile-based signature (LIPS) predicts survival benefit from immune checkpoint inhibitors (ICI) treatment in all patients with metastatic cancer of the ST-ICI cohort. (A) The overall survival (OS) in all patients stratified by the LIPS into high-risk and low-risk with the p value. (B) Time-dependent receiver operating characteristic (ROC) curves of LIPS for OS in all patients. (C) The progression-free survival (PFS) in all patients stratified by the LIPS into high-risk and low-risk with the p value. (D) Time-dependent ROC curves of LIPS for PFS in all patients. AUC, area under the ROC curve.
Figure 5
Figure 5
Subgroup analysis of the liquid immune profile-based signature (LIPS) predict survival benefit of patients treated with ICI by baseline characteristics. Subgroup analysis of the LIPS predict overall survival (OS) benefit (A) and progression-free survival (PFS) benefit (B) of patients treated with ICI. HR and 95% CIs. HNSCC, head and neck squamous cell carcinoma; ICI, immune checkpoint inhibitors; NSCLC, non-small cell lung cancer; TPS, tumor proportion score.

References

    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. Burtness B, Harrington KJ, Greil R, et al. . KEYNOTE-048: phase III study of first-line pembrolizumab (P) for recurrent/metastatic head and neck squamous cell carcinoma (r/m HNSCC). Annals of Oncology 2018;29:viii729 10.1093/annonc/mdy424.045
    1. Mok TSK, Wu Y-L, Kudaba I, et al. . Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): a randomised, open-label, controlled, phase 3 trial. Lancet 2019;393:1819–30. 10.1016/S0140-6736(18)32409-7
    1. Derer A, Spiljar M, Bäumler M. Chemoradiation increases PD-L1 expression in certain melanoma and glioblastoma cells. Front Immunol 2016:7.
    1. Hecht M, Büttner-Herold M, Erlenbach-Wünsch K, et al. . Pd-L1 is upregulated by radiochemotherapy in rectal adenocarcinoma patients and associated with a favourable prognosis. Eur J Cancer 2016;65:52–60. 10.1016/j.ejca.2016.06.015
    1. Hopkins AM, Rowland A, Kichenadasse G, et al. . Predicting response and toxicity to immune checkpoint inhibitors using routinely available blood and clinical markers. Br J Cancer 2017;117:913–20. 10.1038/bjc.2017.274
    1. 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;375:819–29. 10.1056/NEJMoa1604958
    1. Vokes NI, Liu D, Ricciuti B. Harmonization of tumor mutational burden quantification and association with response to immune checkpoint blockade in non-small-cell lung cancer. JCO Precis Oncol 2019;3. 10.1200/PO.19.00171. [Epub ahead of print: 12 Nov 2019].
    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. Chai LF, Prince E, Pillarisetty VG, et al. . Challenges in assessing solid tumor responses to immunotherapy. Cancer Gene Ther 2020;27:528–38. 10.1038/s41417-019-0155-1
    1. Seymour L, Bogaerts J, Perrone A, et al. . iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol 2017;18:e143–52. 10.1016/S1470-2045(17)30074-8
    1. Zhang H, Houghton AM. Good cops turn bad: the contribution of neutrophils to immune-checkpoint inhibitor treatment failures in cancer. Pharmacol Ther 2021;217:107662. 10.1016/j.pharmthera.2020.107662
    1. Chen JA, Ma W, Yuan J, et al. . Translational biomarkers and rationale strategies to overcome resistance to immune checkpoint inhibitors in solid tumors. Cancer Treat Res 2020;180:251–79. 10.1007/978-3-030-38862-1_9
    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. Kim KH, Cho J, Ku BM, et al. . The First-week Proliferative Response of Peripheral Blood PD-1+CD8+ T Cells Predicts the Response to Anti-PD-1 Therapy in Solid Tumors. Clin Cancer Res 2019;25:2144–54. 10.1158/1078-0432.CCR-18-1449
    1. Krieg C, Nowicka M, Guglietta S, et al. . High-Dimensional single-cell analysis predicts response to anti-PD-1 immunotherapy. Nat Med 2018;24:144–53. 10.1038/nm.4466
    1. Rühle PF, Fietkau R, Gaipl US, et al. . Development of a modular assay for detailed immunophenotyping of peripheral human whole blood samples by multicolor flow cytometry. Int J Mol Sci 2016;17:1316. 10.3390/ijms17081316
    1. Donaubauer A-J, Rühle PF, Becker I. One-Tube Multicolor Flow Cytometry Assay (OTMA) for Comprehensive Immunophenotyping of Peripheral Blood : Steinitz M, Human monoclonal antibodies: methods and protocols. New York, NY: Springer New York, 2019: 189–212.
    1. Simon N, Friedman J, Hastie T, et al. . Regularization paths for COX's proportional hazards model via coordinate descent. J Stat Softw 2011;39:1–13. 10.18637/jss.v039.i05
    1. Zhou J-G, Zhao H-T, Jin S-H, et al. . Identification of a RNA-seq-based signature to improve prognostics for uterine sarcoma. Gynecol Oncol 2019;155:499–507. 10.1016/j.ygyno.2019.08.033
    1. Zhou J-G, Liang B, Jin S-H, et al. . Development and validation of an RNA-Seq-Based prognostic signature in neuroblastoma. Front Oncol 2019;9:1361. 10.3389/fonc.2019.01361
    1. Brentnall AR, Cuzick J. Use of the concordance index for predictors of censored survival data. Stat Methods Med Res 2018;27:2359–73. 10.1177/0962280216680245
    1. Haibe-Kains B, Desmedt C, Sotiriou C, et al. . A comparative study of survival models for breast cancer prognostication based on microarray data: does a single gene beat them all? Bioinformatics 2008;24:2200–8. 10.1093/bioinformatics/btn374
    1. Herbst RS, Baas P, Kim D-W, et al. . Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet 2016;387:1540–50. 10.1016/S0140-6736(15)01281-7
    1. Wang Z, Duan J, Cai S, et al. . Assessment of blood tumor mutational burden as a potential biomarker for immunotherapy in patients with non-small cell lung cancer with use of a next-generation sequencing cancer gene panel. JAMA Oncol 2019;5:696–702. 10.1001/jamaoncol.2018.7098
    1. Samstein RM, Lee C-H, Shoushtari AN, et al. . Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet 2019;51:202–6. 10.1038/s41588-018-0312-8
    1. Hellmann MD, Nathanson T, Rizvi H, et al. . Genomic features of response to combination immunotherapy in patients with advanced non-small-cell lung cancer. Cancer Cell 2018;33:843–52. 10.1016/j.ccell.2018.03.018
    1. Takada K, Toyokawa G, Okamoto T, et al. . A Comprehensive Analysis of Programmed Cell Death Ligand-1 Expression With the Clone SP142 Antibody in Non-Small-Cell Lung Cancer Patients. Clin Lung Cancer 2017;18:572–82. 10.1016/j.cllc.2017.02.004
    1. Lee H-H, Wang Y-N, Xia W, et al. . Removal of N-linked glycosylation enhances PD-L1 detection and predicts anti-PD-1/PD-L1 therapeutic efficacy. Cancer Cell 2019;36:168–78. 10.1016/j.ccell.2019.06.008
    1. Okuma Y, Wakui H, Utsumi H, et al. . Soluble programmed cell death ligand 1 as a novel biomarker for nivolumab therapy for non-small-cell lung cancer. Clin Lung Cancer 2018;19:410–7. 10.1016/j.cllc.2018.04.014
    1. Duruisseaux M, Martínez-Cardús A, Calleja-Cervantes ME, et al. . Epigenetic prediction of response to anti-PD-1 treatment in non-small-cell lung cancer: a multicentre, retrospective analysis. Lancet Respir Med 2018;6:771–81. 10.1016/S2213-2600(18)30284-4
    1. Gnjatic S, Bronte V, Brunet LR, et al. . Identifying baseline immune-related biomarkers to predict clinical outcome of immunotherapy. J Immunother Cancer 2017;5:44. 10.1186/s40425-017-0243-4
    1. Wang S, He Z, Wang X, et al. . Antigen presentation and tumor immunogenicity in cancer immunotherapy response prediction. Elife 2019;8:8. 10.7554/eLife.49020
    1. Bochem J, Zelba H, Amaral T, et al. . Peripheral PD-1+CD56+ T-cell frequencies correlate with outcome in stage IV melanoma under PD-1 blockade. PLoS One 2019;14:e0221301. 10.1371/journal.pone.0221301
    1. Martens A, Wistuba-Hamprecht K, Yuan J, et al. . Increases in absolute lymphocytes and circulating CD4+ and CD8+ T cells are associated with positive clinical outcome of melanoma patients treated with ipilimumab. Clin Cancer Res 2016;22:4848–58. 10.1158/1078-0432.CCR-16-0249
    1. Liang H, Xu Y, Chen M, et al. . Patterns of response in metastatic NSCLC during PD-1 or PD-L1 inhibitor therapy: comparison of the RECIST 1.1 and iRECIST criteria. Thorac Cancer 2020;11:1068–75. 10.1111/1759-7714.13367
    1. Haddad R, Blumenschein G, Fayette J, et al. . Treatment beyond progression with nivolumab in patients with recurrent or metastatic (r/m) squamous cell carcinoma of the head and neck (SCCHN) in the phase 3 checkmate 141 study: a biomarker analysis and updated clinical outcomes. Annals of Oncology 2017;28:v372–3. 10.1093/annonc/mdx374.001
    1. Petitprez F, Meylan M, de Reyniès A, et al. . The tumor microenvironment in the response to immune checkpoint blockade therapies. Front Immunol 2020;11:784. 10.3389/fimmu.2020.00784
    1. Tüting T, de Visser KE, KEd V. Cancer. How neutrophils promote metastasis. Science 2016;352:145–6. 10.1126/science.aaf7300
    1. Ocana A, Nieto-Jiménez C, Pandiella A, et al. . Neutrophils in cancer: prognostic role and therapeutic strategies. Mol Cancer 2017;16:137. 10.1186/s12943-017-0707-7
    1. Koucký V, Bouček J, Fialová A. Immunology of plasmacytoid dendritic cells in solid tumors: a brief review. Cancers 2019;11:470. 10.3390/cancers11040470
    1. Kini Bailur J, Gueckel B, Pawelec G. Prognostic impact of high levels of circulating plasmacytoid dendritic cells in breast cancer. J Transl Med 2016;14:151. 10.1186/s12967-016-0905-x
    1. Tjomsland V, Sandström P, Spångeus A, et al. . Pancreatic adenocarcinoma exerts systemic effects on the peripheral blood myeloid and plasmacytoid dendritic cells: an indicator of disease severity? BMC Cancer 2010;10:87. 10.1186/1471-2407-10-87
    1. Shimizu K, Asakura M, Shinga J, et al. . Invariant NKT cells induce plasmacytoid dendritic cell (dC) cross-talk with conventional DCs for efficient memory CD8+ T cell induction. J Immunol 2013;190:5609–19. 10.4049/jimmunol.1300033
    1. Youn J-I, Park S-M, Park S, et al. . Peripheral natural killer cells and myeloid-derived suppressor cells correlate with anti-PD-1 responses in non-small cell lung cancer. Sci Rep 2020;10:9050. 10.1038/s41598-020-65666-x
    1. Bae E-A, Seo H, Kim I-K, et al. . Roles of NKT cells in cancer immunotherapy. Arch Pharm Res 2019;42:543–8. 10.1007/s12272-019-01139-8
    1. Juliá EP, Mandó P, Rizzo MM, et al. . Peripheral changes in immune cell populations and soluble mediators after anti-PD-1 therapy in non-small cell lung cancer and renal cell carcinoma patients. Cancer Immunol Immunother 2019;68:1585–96. 10.1007/s00262-019-02391-z
    1. Thommen DS, Koelzer VH, Herzig P, et al. . A transcriptionally and functionally distinct PD-1+ CD8+ T cell pool with predictive potential in non-small-cell lung cancer treated with PD-1 blockade. Nat Med 2018;24:994–1004. 10.1038/s41591-018-0057-z
    1. Lee MY, Park C-J, Cho Y-U, et al. . Differences in PD-1 expression on CD8+ T-cells in chronic myeloid leukemia patients according to disease phase and TKI medication. Cancer Immunol Immunother 2020;69:2223-2232. 10.1007/s00262-020-02617-5
    1. La J, Cheng D, Brophy MT, et al. . Real-World outcomes for patients treated with immune checkpoint inhibitors in the Veterans Affairs system. JCO Clin Cancer Inform 2020;4:918–28. 10.1200/CCI.20.00084
    1. Di Giacomo AM, Valente M, Cerase A, et al. . Immunotherapy of brain metastases: breaking a "dogma". J Exp Clin Cancer Res 2019;38:419. 10.1186/s13046-019-1426-2
    1. Schmidberger H, Rapp M, Ebersberger A, et al. . Long-Term survival of patients after ipilimumab and hypofractionated brain radiotherapy for brain metastases of malignant melanoma: sequence matters. Strahlenther Onkol 2018;194:1144–51. 10.1007/s00066-018-1356-5
    1. Kocher M, Ruge MI, Galldiks N, et al. . Applications of radiomics and machine learning for radiotherapy of malignant brain tumors. Strahlenther Onkol 2020;196:856–67. 10.1007/s00066-020-01626-8
    1. Nabet BY, Esfahani MS, Moding EJ, et al. . Noninvasive early identification of therapeutic benefit from immune checkpoint inhibition. Cell 2020;183:363–76. 10.1016/j.cell.2020.09.001

Source: PubMed

3
S'abonner