Molecular signatures associated with tumor-specific immune response in melanoma patients treated with dendritic cell-based immunotherapy

Tamara García-Salum, Andrea Villablanca, Franziska Matthäus, Andrés Tittarelli, Mauricio Baeza, Cristián Pereda, M Alejandra Gleisner, Fermín E González, Mercedes N López, Jörg D Hoheisel, Johannes Norgauer, Peter J Gebicke-Haerter, Flavio Salazar-Onfray, Tamara García-Salum, Andrea Villablanca, Franziska Matthäus, Andrés Tittarelli, Mauricio Baeza, Cristián Pereda, M Alejandra Gleisner, Fermín E González, Mercedes N López, Jörg D Hoheisel, Johannes Norgauer, Peter J Gebicke-Haerter, Flavio Salazar-Onfray

Abstract

Purpose: We previously showed that autologous dendritic cells (DCs) loaded with an allogeneic heat shock (HS)-conditioned melanoma cell-derived lysate, called TRIMEL, induce T-cell-mediated immune responses in stage IV melanoma patients. Importantly, a positive delayed-type hypersensitivity (DTH) reaction against TRIMEL after vaccination, correlated with patients prolonged survival. Furthermore, we observed that DTH reaction was associated with a differential response pattern reflected in the presence of distinct cell subpopulations in peripheral blood. Detected variations in patient responses encouraged molecular studies aimed to identify gene expression profiles induced after vaccination in treated patients, allowing the identification of new molecular predictive markers.

Methods: Gene expression patterns were analyzed by microarrays during vaccination, and some of them confirmed by quantitative real-time reverse transcriptase PCR (qRT-PCR) in the total leukocyte population of a representative group of responder and non-responder patients. New candidates for biomarkers with predictive value were identified using bioinformatics, molecular analysis, and flow cytometry.

Results: Seventeen genes overexpressed in responder patients after vaccination respect to non-responders were identified after a mathematical analysis, from which ten were linked to immune responses and five related to cell cycle control and signal transduction. In immunological responder patients, increased protein levels of the chemokine receptor CXCR4 and the Fc-receptor CD32 were observed on cell membranes of CD8+ T and B cells and the monocyte population, respectively, confirming gene expression results.

Conclusions: Our study contributes to finding new molecular markers associated with clinical outcome and better understanding of clinically relevant immunological responses induced by anti-tumor DC-vaccines.

Keywords: CD32; CXCR4; immunotherapy; melanoma; molecular signatures.

Conflict of interest statement

CONFLICTS OF INTEREST The authors declare that they have no conflicts of interest.

Figures

Figure 1. TRIMEL-loaded DCs-treated immunological responder patients…
Figure 1. TRIMEL-loaded DCs-treated immunological responder patients have a higher median survival than non-immunological responder patients
Post-TRIMEL-loaded DCs immunotherapy overall survival curves for immunological responder (DTH+: full line) and immunological non-responder (DTH-/NT: dash line) patients. *p < 0.05 **p < 0.01. NT: Not tested.
Figure 2. Differences in temporal gene expression…
Figure 2. Differences in temporal gene expression of potential molecular markers between clinical responder and non-responder patients
Vaccinated patients were reclassified according DTH reaction and overall survival as described in Material and methods. (A) Relative gene expression of genes CLEC2D, CXCR4, FCGR2A, GIT2, MS4A7, PRDM1, PRDX3, SDCBP, SPG21 and VNN2 were determined by microarrays. Black dots: Clinical responders (DTH+/long survivor patients); samples from 1st-4th vaccination (n = 5), samples from DTH evaluation (n = 6); red triangles: Non-responder patients (DTH− patients), samples from 1st vaccination (n = 3), 2nd vaccination (n = 4), 3rd vaccination (n = 1), 4th vaccination (n = 2) and DTH evaluation (n = 1). (B) Relative expression of genes CREB5, CSNK1A1, EIF4G2, LOC648210, STRN3, TROVE2 and unknown (probe ID 2120450) were determined by microarrays. Black dots: Clinical responder patients, samples from 1st to 4th vaccination (n = 5), DTH evaluation (n = 6); red triangles: Non-responder patients, samples from 1st vaccination (n = 3), 2nd vaccination (n = 4), 3rd vaccination (n = 1). 4th vaccination (n = 2) and DTH evaluation (n = 1). (C) Relative gene expression determined by qRT-PCR of genes CLEC2D, CXCR4, FCGR2A, MS4A7 and CSNK1A1. Black dots: Clinical responder patients, samples from 1st vaccination (n = 4–6), 2nd vaccination (n = 3–4), 3rd vaccination (n = 6), 4th vaccination (n = 5–7), and DTH evaluation (n = 7); red triangles: non-responder patients, samples from 1st vaccination (n = 2–3), 2nd vaccination (n = 4); 3rd vaccination (n = 1), 4th vaccination (n = 2) and DTH evaluation (n = 1). Fold changes relative to the respective reference sample are given. Each data dot represents one patient's sample, each line represents the mean of the data dots. Vaccination (vac.); evaluation (eval.); ID, identification number.
Figure 3. Theoretical network of genetic interactions…
Figure 3. Theoretical network of genetic interactions associated to DC vaccine-induced immune response
Fifteen genes significantly regulated during the vaccination period (large, blue circles) were subjected to analyses of direct genetic interactions using STRING and BioGrid platforms. Most interactions were unique for each gene, but a few appeared in two or more lists of the 15 genes attributing to them a connector function (yellow circles; the more of the 15 genes are connected by a connector gene, the larger its diameter) and hence giving rise to a molecular network. For instance, ubiquitin C (UBC) (the connector gene with the most connections) connects 9 out of the 15 genes. The combined STRING and BioGrid genes were entered into “genes.R” available in “R”, and further processed in “igraph”.
Figure 4. Increased CXCR4 surface expression on…
Figure 4. Increased CXCR4 surface expression on CD8+ T-cells and B-cells, and CD32 surface expression on monocyte populations in immunological responder compared to non-responder patients
(A, C) Cryopreserved PBMCs, obtained from healthy donors (HD; n = 4–5), immunological responder (n = 4–9) and non-responder patients (n = 7–10), at the beginning (pre-tx) and at the end (post-tx) of TRIMEL-loaded DCs immunization protocol, were analyzed for the CXCR4 (A) and CD32 (C) surface expression by flow cytometry. Each data point represents one patient sample. MFI: mean of fluorescence intensity. Mann-Whitney test; *p < 0.05 **p < 0.01. (B, D) Representative histograms compare the analysis of one healthy donor (HD), one responder and one non-responder patient to TRIMEL-loaded DCs immunotherapy. CD4+ T-cells: CD45+/CD3+/CD4+; CD8+ T-cells: CD3+/CD8+; B-cells: CD3–/CD19+; NK-cells: CD45+/CD16+/CD56+ and monocyte population: CD45+/CD14+. Black lines: immunological responder patients; red lines: non-responder patients; continuous lines: pre-tx; dashed lines: post-tx; fill curve: HD; tx, treatment.

References

    1. Winnepenninckx V, Lazar V, Michiels S, Dessen P, Stas M, Alonso SR, Avril MF, Ortiz Romero PL, Robert T, Balacescu O, Eggermont AM, Lenoir G, Sarasin A, et al. Melanoma Group of the European Organization for Research and Treatment of Cancer Gene expression profiling of primary cutaneous melanoma and clinical outcome. J Natl Cancer Inst. 2006;98:472–482.
    1. Bogunovic D, O'Neill DW, Belitskaya-Levy I, Vacic V, Yu YL, Adams S, Darvishian F, Berman R, Shapiro R, Pavlick AC, Lonardi S, Zavadil J, Osman I, Bhardwaj N. Immune profile and mitotic index of metastatic melanoma lesions enhance clinical staging in predicting patient survival. Proc Natl Acad Sci U S A. 2009;106:20429–20434.
    1. Jönsson G, Busch C, Knappskog S, Geisler J, Miletic H, Ringnér M, Lillehaug JR, Borg A, Lønning PE. Gene expression profiling-based identification of molecular subtypes in stage IV melanomas with different clinical outcome. Clin Cancer Res. 2010;16:3356–3367.
    1. Couzin-Frankel J, Breakthrough of the year 2013 Cancer immunotherapy. Science. 2013;342:1432–1433.
    1. Yang Y. Cancer immunotherapy: harnessing the immune system to battle cancer. J Clin Invest. 2015;125:3335–3337.
    1. Farolfi A, Ridolfi L, Guidoboni M, Nicoletti SV, Piciucchi S, Valmorri L, Costantini M, Scarpi E, Amadori D, Ridolfi R. Ipilimumab in advanced melanoma: reports of long-lasting responses. Melanoma Res. 2012;22:263–270.
    1. Mackensen A, Meidenbauer N, Vogl S, Laumer M, Berger J, Andreesen R. Phase I study of adoptive T-cell therapy using antigen-specific CD8+ T cells for the treatment of patients with metastatic melanoma. J Clin Oncol. 2006;24:5060–5069.
    1. Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley M. Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Nat Rev Cancer. 2008;8:299–308.
    1. López MN, Pereda C, Segal G, Muñoz L, Aguilera R, González FE, Escobar A, Ginesta A, Reyes D, González R, Mendoza-Naranjo A, Larrondo M, Compán A, et al. Prolonged survival of dendritic cell-vaccinated melanoma patients correlates with tumor-specific delayed type IV hypersensitivity response and reduction of tumor growth factor beta-expressing T cells. J Clin Oncol. 2009;27:945–952.
    1. González FE, Gleisner A, Falcón-Beas F, Osorio F, López MN, Salazar-Onfray F. Tumor cell lysates as immunogenic sources for cancer vaccine design. Hum Vaccin Immunother. 2014;10:3261–3269.
    1. Kyte JA, Kvalheim G, Lislerud K, thor Straten P, Dueland S, Aamdal S, Gaudernack G. T cell responses in melanoma patients after vaccination with tumor-mRNA transfected dendritic cells. Cancer Immunol Immunother. 2007;56:659–675.
    1. Sabado RL, Bhardwaj N. Dendritic cell immunotherapy. Ann N Y Acad Sci. 2013;1284:31–45.
    1. O'Rourke MG, Johnson M, Lanagan C, See J, Yang J, Bell JR, Slater GJ, Kerr BM, Crowe B, Pur die DM, Elliott SL, Ellem KA, Schmidt CW. Durable complete clinical responses in a phase I/II trial using an autologous melanoma cell/dendritic cell vaccine. Cancer Immunol Immunother. 2003;52:387–395.
    1. Aguilera R, Saffie C, Tittarelli A, González FE, Ramírez M, Reyes D, Pereda C, Hevia D, García T, Salazar L, Ferreira A, Hermoso M, Mendoza-Naranjo A, et al. Heat-shock induction of tumor-derived danger signals mediates rapid monocyte differentiation into clinically effective dendritic cells. Clin Cancer Res. 2011;17:2474–2483.
    1. Durán-Aniotz C, Segal G, Salazar L, Pereda C, Falcón C, Tempio F, Aguilera R, González R, Pérez C, Tittarelli A, Catalán D, Nervi B, Larrondo M, et al. The immunological response and post-treatment survival of DC-vaccinated melanoma patients are associated with increased Th1/Th17 and reduced Th3 cytokine responses. Cancer Immunol Immunother. 2013;62:761–772.
    1. Tittarelli A, González FE, Pereda C, Mora G, Muñoz L, Saffie C, García T, Díaz D, Falcón C, Hermoso M, López MN, Salazar-Onfray F. Toll-like receptor 4 gene polymorphism influences dendritic cell in vitro function and clinical outcomes in vaccinated melanoma patients. Cancer Immunol Immunother. 2012;61:2067–2077.
    1. Mimeault M, Batra S. Novel biomarkers and therapeutic targets for optimizing the therapeutic management of melanomas. World J Clin Oncol. 2012;3:32–42.
    1. Gebicke-Haerter P. Systems psychopharmacology: A network approach to developing novel therapies. World J Psychiatry. 2016;6:66–83.
    1. Schadendorf D, Fisher DE, Garbe C, Gershenwald JE, Grob JJ, Halpern A, Herlyn M, Marchetti MA, McArthur G, Ribas A, Roesch A, Hauschild A. Melanoma. Nat Rev Dis Primers. 2015;1:15003.
    1. Mines MA, Goodwin JS, Limbird LE, Cui FF, Fan G. Deubiquitination of CXCR4 by USP14 is critical for both CXCL12-induced CXCR4 degradation and chemotaxis but not ERK ativation. J Biol Chem. 2009;284:5742–5752.
    1. Hummel S, Van Aken H, Zarbock A. Inhibitors of CXC chemokine receptor type 4: putative therapeutic approaches in inflammatory diseases. Curr Opin Hematol. 2014;21:29–36.
    1. Zhang T, Somasundaram R, Berking C, Caputo L, Van Belle P, Elder D, Czerniecki B, Hotz S, Schuchter L, Spitz FR, Berencsi K, Rani P, Marincola F, et al. Preferential involvement of CX chemokine receptor 4 and CX chemokine ligand 12 in T-cell migration toward melanoma cells. Cancer Biol Ther. 2006;5:1304–1312.
    1. Lisi S, Sisto M, Lofrumento DD, D'Amore S, D'Amore M. Advances in the understanding of the Fc gamma receptors-mediated autoantibodies uptake. Clin Exp Med. 2011;11:1–10.
    1. Gillis C, Gouel-Cheron A, Jonsson F, Bruhns P. Contribution of Human FcgammaRs to Disease with Evidence from Human Polymorphisms and Transgenic Animal Studies. Front Immunol. 2014;5:254.
    1. Li F, Smith P, Ravetch J. Inhibitory Fcgamma receptor is required for the maintenance of tolerance through distinct mechanisms. J Immunol. 2014;192:3021–3028.
    1. Boles KS, Barten R, Kumaresan PR, Trowsdale J, Mathew P. Cloning of a new lectin-like receptor expressed on human NK cells. Immunogenetics. 1999;50:1–7.
    1. Rosen DB, Cao W, Avery DT, Tangye SG, Liu YJ, Houchins JP, Lanier LL. Functional consequences of interactions between human NKR-P1A and its ligand LLT1 expressed on activated dendritic cells and B cells. J Immunol. 2008;180:6508–6517.
    1. Germain C, Meier A, Jensen T, Knapnougel P, Poupon G, Lazzari A, Neisig A, Håkansson K, Dong T, Wagtmann N, Galsgaard ED, Spee P, Braud VM. Induction of lectin-like transcript 1 (LLT1) protein cell surface expression by pathogens and interferon-gamma contributes to modulate immune responses. J Biol Chem. 2011;286:37964–37975.
    1. Vandesompele J, De Preter K, Pattyn F, Poppe B, Van Roy N, De Paepe A, Speleman F. Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol. 2002;3 RESEARCH0034.

Source: PubMed

3
Subskrybuj