Pretreatment antigen-specific immunity and regulation - association with subsequent immune response to anti-tumor DNA vaccination

Laura E Johnson, Brian M Olson, Douglas G McNeel, Laura E Johnson, Brian M Olson, Douglas G McNeel

Abstract

Background: Immunotherapies have demonstrated clinical benefit for many types of cancers, however many patients do not respond, and treatment-related adverse effects can be severe. Hence many efforts are underway to identify treatment predictive biomarkers. We have reported the results of two phase I trials using a DNA vaccine encoding prostatic acid phosphatase (PAP) in patients with biochemically recurrent prostate cancer. In both trials, persistent PAP-specific Th1 immunity developed in some patients, and this was associated with favorable changes in serum PSA kinetics. In the current study, we sought to determine if measures of antigen-specific or antigen non-specific immunity were present prior to treatment, and associated with subsequent immune response, to identify possible predictive immune biomarkers.

Methods: Patients who developed persistent PAP-specific, IFNγ-secreting immune responses were defined as immune "responders." The frequency of peripheral T cell and B cell lymphocytes, natural killer cells, monocytes, dendritic cells, myeloid derived suppressor cells, and regulatory T cells were assessed by flow cytometry and clinical laboratory values. PAP-specific immune responses were evaluated by cytokine secretion in vitro, and by antigen-specific suppression of delayed-type hypersensitivity to a recall antigen in an in vivo SCID mouse model.

Results: The frequency of peripheral blood cell types did not differ between the immune responder and non-responder groups. Non-responder patients tended to have higher PAP-specific IL-10 production pre-vaccination (p = 0.09). Responder patients had greater preexisting PAP-specific bystander regulatory responses that suppressed DTH to a recall antigen (p = 0.016).

Conclusions: While our study population was small (n = 38), these results suggest that different measures of antigen-specific tolerance or regulation might help predict immunological outcome from DNA vaccination. These will be prospectively evaluated in an ongoing randomized, phase II trial.

Keywords: Biomarker; DNA vaccine; Interleukin 10; Prostate cancer; Prostatic acid phosphatase.

Conflict of interest statement

Ethics approval and consent to participate

Samples were collected under University of Wisconsin IRB-approved protocols from two clinical trials (NCT00582140 and NCT00849121), and all patients gave written, informed consent for remaining samples to be used for research.

Consent for publication

Not applicable.

Competing interests

DGM has ownership interest, receives research support, and serves as consultant to Madison Vaccines, Inc. which has licensed material described in this manuscript. The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Schema of vaccination schedule of two phase I trials in which prostate cancer patients were treated with a DNA vaccine encoding PAP. Panel a: in Trial 1 (NCT00582140), patients received six biweekly immunization. Panel b: In Trial 2 (NCT00849121), patients received six biweekly immunizations followed by booster immunization every 3 months or based on immune monitoring. Grey arrows represent immunization time points. PAP-specific immune responses were monitored every 3 months after the initial six biweekly immunizations for each trial up to one year, as demonstrated by asterisks. Samples obtained before immunization were used for the biomarker analysis. Panel c: Shown are the number of immune responders and non-responders as defined for each trial
Fig. 2
Fig. 2
Differences in absolute lymphocyte or monocyte counts in immune responding and non-responding patients were not detected. Panel a: Absolute lymphocyte and monocyte counts per μl of blood of responder and non-responder patients. Panel b: The lymphocyte-to-monocyte and neutrophil-to-lymphocyte ratios were calculated. Each dot represents the absolute lymphocyte or monocyte counts and lymphocyte ratios from individual subjects (closed squares, immune responders (n = 12); open squares, non-responders (n = 25)). Lines show median values
Fig. 3
Fig. 3
Differences in peripheral blood cellular subsets were not detected in immune responding and non-responding patients. The percentage of T cells (CD3 + CD4+ T cells or CD3 + CD8+ T cells), B cells (CD3-CD19+), monocytes (CD14+), (Panel a); NK cells (CD56 + CD3-), NK T cells (CD56 + CD3+), (Panel b); dendritic cells (HLADR + CD11c + CD14-), (Panel c); and MDSCs (CD3-HLA-DRlow/negCD33 + CD11b + CD14+) and T regulatory cells (CD3 + CD4 + CD127-CD25 + FoxP3+) (Panel d) was evaluated in PBMCs from immune responding and non-responding patients. Each dot represents the percentage of each cell type among total live PBMCs from an individual subject (closed squares, immune-responders (n = 7); open squares, non-responders (n = 22)). Lines represent median values for each data set. Samples were analyzed in duplicate and averaged. Cryopreserved samples were not available for this analysis for 9 subjects
Fig. 4
Fig. 4
Prostate cancer patients had preexisting immune responses to the PAP protein. PBMCs from responder or non-responder patients were stimulated 1 or 3 day(s) with PAP, PSA, AR LBD (AR), and Con A. Supernatants from cultures were collected and analyzed for cytokine concentrations (a) Granzyme B, (b) IFNγ, (c) IL-2, (d) IL-4, (e) IL-10, (f) IL-6, and (g) IL-17a using a cytokine bead array. Each dot represents the cytokine expression level for an individual prostate cancer patient (closed squares-responders (n = 7) and open square-non-responders (n = 23)). Samples were analyzed in triplicates and averaged. Lines represent median values for each data set. Statistical comparisons were made using a Mann Whitney t test. As in Fig. 3, cryopreserved samples were not available for 8 subjects
Fig. 5
Fig. 5
Patients that were long term immune responders had preexisting PAP-specific regulatory responses. Pretreatment PBMCs from immune responder (n = 9) and non-responder patients (n = 23) were injected into the footpads of SCID mice with TT/D (recall antigen) alone or in combination with PAP or PSA. Each dot represents the net swelling (10−4 in.) after 24 h. This was defined as the antigen-specific swelling measurement minus the swelling due to the PBMCs and PBS alone. Shown is the net DTH immune response (10−4 in.) to TT/D alone (closed circles, responders; closed squares, non-responders) or in combination with PAP (a) or PSA (b) (circles, immune responders; squares, immune non-responders) for each patient. Statistical comparisons were made using a paired, non-parametric t test analysis. The log-transformed fold change is shown comparing c) TT/D PAP to TT/D and d) TT/D PSA to TT/D for immune responders and non-responders. Comparison of fold change was made using a Mann Whitney t test

References

    1. Topalian SL, et al. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol. 2014;32(10):1020–1030. doi: 10.1200/JCO.2013.53.0105.
    1. Weber JS, et al. Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2015;16(4):375–384. doi: 10.1016/S1470-2045(15)70076-8.
    1. Garon EB, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med. 2015;372(21):2018–2028. doi: 10.1056/NEJMoa1501824.
    1. Motzer RJ, et al. Nivolumab for metastatic renal cell carcinoma: results of a randomized phase II trial. J Clin Oncol. 2015;33(13):1430–1437. doi: 10.1200/JCO.2014.59.0703.
    1. Postow MA, et al. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N Engl J Med. 2015;372(21):2006–2017. doi: 10.1056/NEJMoa1414428.
    1. Larkin J, Hodi FS, Wolchok JD. Combined Nivolumab and Ipilimumab or Monotherapy in untreated melanoma. N Engl J Med. 2015;373(13):1270–1271. doi: 10.1056/NEJMc1509660.
    1. Wolchok JD, et al. Ipilimumab monotherapy in patients with pretreated advanced melanoma: a randomised, double-blind, multicentre, phase 2, dose-ranging study. Lancet Oncol. 2010;11(2):155–164. doi: 10.1016/S1470-2045(09)70334-1.
    1. McDermott DF, et al. Atezolizumab, an anti-Programmed Death-Ligand 1 antibody, in metastatic renal cell carcinoma: long-term safety, clinical activity, and immune correlates from a phase Ia study. J Clin Oncol. 2016;34(8):833–842. doi: 10.1200/JCO.2015.63.7421.
    1. Rosenberg JE, et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet. 2016;387(10031):1909–1920. doi: 10.1016/S0140-6736(16)00561-4.
    1. Kantoff PW, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363(5):411–422. doi: 10.1056/NEJMoa1001294.
    1. Galon J, et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science. 2006;313(5795):1960–1964. doi: 10.1126/science.1129139.
    1. Galon J, et al. Cancer classification using the Immunoscore: a worldwide task force. J Transl Med. 2012;10:205. doi: 10.1186/1479-5876-10-205.
    1. Topalian SL, et al. 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.
    1. Gajewski TF, Schreiber H, Fu YX. Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol. 2013;14(10):1014–1022. doi: 10.1038/ni.2703.
    1. Weide B, et al. Baseline biomarkers for outcome of melanoma patients treated with Pembrolizumab. Clin Cancer Res, 2016;22:5487–96.
    1. Martens A, et al. Baseline peripheral blood biomarkers associated with clinical outcome of advanced melanoma patients treated with Ipilimumab. Clin Cancer Res. 2016;22(12):2908–2918. doi: 10.1158/1078-0432.CCR-15-2412.
    1. Martens A, 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(19):4848–4858. doi: 10.1158/1078-0432.CCR-16-0249.
    1. Slovin SF, et al. Ipilimumab alone or in combination with radiotherapy in metastatic castration-resistant prostate cancer: results from an open-label, multicenter phase I/II study. Ann Oncol. 2013;24(7):1813–1821. doi: 10.1093/annonc/mdt107.
    1. Kwon ED, et al. Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): a multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol. 2014;15(7):700–712. doi: 10.1016/S1470-2045(14)70189-5.
    1. McNeel DG, et al. Phase I trial of a monoclonal antibody specific for {alpha}v{beta}3 Integrin (MEDI-522) in patients with advanced malignancies, including an assessment of effect on tumor perfusion. Clin Cancer Res. 2005;11(21):7851–7860. doi: 10.1158/1078-0432.CCR-05-0262.
    1. Brahmer JR, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366(26):2455–2465. doi: 10.1056/NEJMoa1200694.
    1. Kantoff P, et al. Randomized, double-blind, vector-controlled study of targeted immunotherapy in patients with hormone-refractory prostate cancer [abstract] J Clin Oncol. 2005;24(18S):2501.
    1. Kantoff PW, et al. Randomized, double-blind, vector-controlled study of targeted immunotherapy in patients (pts) with hormone-refractory prostate cancer (HRPC) Proc Am Soc Clin Oncol. 2006;24:100s.
    1. Kantoff PW, et al. Overall survival analysis of a phase II randomized controlled trial of a Poxviral-based PSA-targeted immunotherapy in metastatic castration-resistant prostate cancer. J Clin Oncol. 2010;28(7):1099–1105. doi: 10.1200/JCO.2009.25.0597.
    1. Small EJ, et al. Placebo-controlled phase III trial of immunologic therapy with sipuleucel-T (APC8015) in patients with metastatic, asymptomatic hormone refractory prostate cancer. J Clin Oncol. 2006;24(19):3089–3094. doi: 10.1200/JCO.2005.04.5252.
    1. Gulley JL, et al. Immunologic and prognostic factors associated with overall survival employing a poxviral-based PSA vaccine in metastatic castrate-resistant prostate cancer. Cancer Immunol Immunother. 2010;59(5):663–674. doi: 10.1007/s00262-009-0782-8.
    1. Sheikh NA, et al. Sipuleucel-T immune parameters correlate with survival: an analysis of the randomized phase 3 clinical trials in men with castration-resistant prostate cancer. Cancer Immunol Immunother. 2013;62(1):137–147. doi: 10.1007/s00262-012-1317-2.
    1. McNeel DG, et al. Naturally occurring prostate cancer antigen-specific T cell responses of a Th1 phenotype can be detected in patients with prostate cancer. Prostate. 2001;47(3):222–229. doi: 10.1002/pros.1066.
    1. Olson BM, McNeel DG. Antibody and T-cell responses specific for the androgen receptor in patients with prostate cancer. Prostate. 2007;67(16):1729–1739. doi: 10.1002/pros.20652.
    1. Olson BM, et al. Human prostate tumor antigen-specific CD8+ regulatory T cells are inhibited by CTLA-4 or IL-35 blockade. J Immunol. 2012;189(12):5590–5601. doi: 10.4049/jimmunol.1201744.
    1. Santegoets SJ, et al. T cell profiling reveals high CD4+CTLA-4 + T cell frequency as dominant predictor for survival after prostate GVAX/ipilimumab treatment. Cancer Immunol Immunother. 2013;62(2):245–256. doi: 10.1007/s00262-012-1330-5.
    1. Farsaci B, et al. Analyses of Pretherapy peripheral Immunoscore and response to vaccine therapy. Cancer Immunol Res. 2016;4(9):755–765. doi: 10.1158/2326-6066.CIR-16-0037.
    1. McNeel DG, et al. Safety and immunological efficacy of a DNA vaccine encoding prostatic acid phosphatase in patients with stage D0 prostate cancer. J Clin Oncol. 2009;27(25):4047–4054. doi: 10.1200/JCO.2008.19.9968.
    1. McNeel DG, et al. Real-time immune monitoring to guide plasmid DNA vaccination schedule targeting prostatic acid phosphatase in patients with castration-resistant prostate cancer. Clin Cancer Res. 2014;20(14):3692–3704. doi: 10.1158/1078-0432.CCR-14-0169.
    1. Becker JT, et al. DNA vaccine encoding prostatic acid phosphatase (PAP) elicits long-term T-cell responses in patients with recurrent prostate cancer. J Immunother. 2010;33(6):639–647. doi: 10.1097/CJI.0b013e3181dda23e.
    1. Maecker HT, McCoy JP, Nussenblatt R. Standardizing immunophenotyping for the human immunology project. Nat Rev Immunol. 2012;12(3):191–200.
    1. Idorn M, et al. Correlation between frequencies of blood monocytic myeloid-derived suppressor cells, regulatory T cells and negative prognostic markers in patients with castration-resistant metastatic prostate cancer. Cancer Immunol Immunother. 2014;63(11):1177–1187. doi: 10.1007/s00262-014-1591-2.
    1. Spencer WF, et al. Immunotherapy with interleukin-2 and alpha-interferon in patients with metastatic renal cell cancer with in situ primary cancers: a pilot study. J Urol. 1992;147(1):24–30. doi: 10.1016/S0022-5347(17)37124-0.
    1. Slovin SF. Biomarkers for immunotherapy in genitourinary malignancies. Urol Oncol. 2016;34(4):205–213. doi: 10.1016/j.urolonc.2015.02.007.
    1. Patel SP, Kurzrock R. PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther. 2015;14(4):847–856. doi: 10.1158/1535-7163.MCT-14-0983.
    1. Topalian SL, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366(26):2443–2454. doi: 10.1056/NEJMoa1200690.
    1. Herbst RS, 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(10027):1540–1550. doi: 10.1016/S0140-6736(15)01281-7.
    1. Seiwert TY, et al. Safety and clinical activity of pembrolizumab for treatment of recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-012): an open-label, multicentre, phase 1b trial. Lancet Oncol. 2016;17(7):956–965. doi: 10.1016/S1470-2045(16)30066-3.
    1. Daud AI, et al. Programmed Death-Ligand 1 expression and response to the anti-Programmed Death 1 antibody Pembrolizumab in melanoma. J Clin Oncol. 2016;34(34):4102–4109. doi: 10.1200/JCO.2016.67.2477.
    1. Minardi D, et al. Neutrophil-to-lymphocyte ratio may be associated with the outcome in patients with prostate cancer. Spring. 2015;4:255. doi: 10.1186/s40064-015-1036-1.
    1. Langsenlehner T, et al. Validation of the neutrophil-to-lymphocyte ratio as a prognostic factor in a cohort of European prostate cancer patients. World J Urol. 2015;33(11):1661–1667. doi: 10.1007/s00345-015-1494-7.
    1. Yao A, et al. High neutrophil-to-lymphocyte ratio predicts poor clinical outcome in patients with castration-resistant prostate cancer treated with docetaxel chemotherapy. Int J Urol, 2015;22:827–33.
    1. Garcia-Hernandez ML, et al. Interleukin-10 promotes B16-melanoma growth by inhibition of macrophage functions and induction of tumour and vascular cell proliferation. Immunology. 2002;105(2):231–243. doi: 10.1046/j.1365-2567.2002.01363.x.
    1. Kalli F, et al. Comparative analysis of cancer vaccine settings for the selection of an effective protocol in mice. J Transl Med. 2013;11:120. doi: 10.1186/1479-5876-11-120.

Source: PubMed

3
Prenumerera