Vaccine-draining lymph nodes of cancer patients for generating anti-cancer antibodies

Girja S Shukla, Walter C Olson, Stephanie C Pero, Yu-Jing Sun, Chelsea L Carman, Craig L Slingluff Jr, David N Krag, Girja S Shukla, Walter C Olson, Stephanie C Pero, Yu-Jing Sun, Chelsea L Carman, Craig L Slingluff Jr, David N Krag

Abstract

Background: Our research is focused on using the vaccine draining lymph node to better understand the immune response to cancer vaccines and as a possible source of anti-cancer reagents. We evaluated vaccine draining lymph nodes archived from a clinical study in melanoma patients and determined the reaction of B cells to the vaccine peptides.

Methods: Mononuclear cells (MNCs) were recovered from cryopreserved lymph nodes that were directly receiving drainage from multi-peptide melanoma vaccine. The patients were enrolled on a vaccine study (NCT00089219, FDA, BB-IND No. 10825). B cell responses in the vaccine-draining lymph nodes were studied under both stimulated and un-stimulated conditions. Cryopreserved cells were stimulated with CD40L, stained with multiple human cell-surface markers (CD19, CD27, IgM) to identify different categories of B cell sub populations with flow cytometry. Hybridomas were generated from the lymph node cells after CD40L-stimulation. Cells were fused to murine plasmacytoma P3X63.Ag8.653 using Helix electrofusion chamber. ELISA was used to evaluate hybridoma derived antibody binding to vaccine peptides.

Results: Viable MNCs were satisfactorily recovered from lymph nodes cryopreserved from six vaccine study patients 8-14 years previously. B cell ELISPOT demonstrated responses for each patient to multiple vaccine peptides. CD40L stimulation of lymph node cells increased the proportion of CD19+ CD27+ cells from 12 to 65% of the sample and increased the proportion of class-switched cells. Screening of IgG secreting clones demonstrated binding to melanoma vaccine peptides.

Conclusions: B cells were successfully recovered and expanded from human cryopreserved vaccine-draining lymph nodes. Individual B cells were identified that secreted antibodies that bound to cancer vaccine peptides. The ability to reliably generate in vitro the same antibodies observed in the blood of vaccinated patients will facilitate research to understand mechanisms of human antibody activity and possibly lead to therapeutic antibodies.

Keywords: Antibodies; B cells; Cancer vaccine; Human; Melanoma.

Figures

Fig. 1
Fig. 1
B cell ELISPOT analyses showing the number of vaccine-draining lymph node B cells per patient that secreted IgG Abs against individual and pooled (6MEL) vaccine peptides. The number of spots were plotted after subtracting the values of irrelevant peptide negative control
Fig. 2
Fig. 2
Representative density plots and histograms from flow cytometric analyses of vaccine-draining lymph node cells, showing enrichment of a CD19+CD27+ cells and b class-switched IgM− CD19+ CD27+ cells following subsequent rounds of CD40L-stimulated amplification
Fig. 3
Fig. 3
Screening of IgG secreting clones and their binding to vaccine antigens. a ELISA screening of a limited set of hybridoma culture supernatant samples identified high numbers of IgG secreting clones. b Further analysis of these supernatant samples identified two clones, A11 and C3, for their binding to the mixture of six vaccine peptides
Fig. 4
Fig. 4
Specificity of the clones that bind to vaccine antigens. Hybridoma culture supernatant samples from clones A11 and C3 specifically bind to FLL peptide and not to RNG peptide

References

    1. Krag DN, Anderson SJ, Julian TB, et al. Sentinel-lymph-node resection compared with conventional axillary-lymph-node dissection in clinically node-negative patients with breast cancer: overall survival findings from the NSABP B-32 randomised phase 3 trial. Lancet Oncol. 2010;11:927–933. doi: 10.1016/S1470-2045(10)70207-2.
    1. Reed CM, Cresce ND, Mauldin IS, Slingluff CL, Jr, Olson WC. Vaccination with melanoma helper peptides induces antibody responses associated with improved overall survival. Clin Cancer Res. 2015;21:3879–3887. doi: 10.1158/1078-0432.CCR-15-0233.
    1. Slingluff CL, Jr, Petroni GR, Olson WC, et al. A randomized pilot trial testing the safety and immunologic effects of a MAGE-A3 protein plus AS15 immunostimulant administered into muscle or into dermal/subcutaneous sites. Cancer Immunol Immunother. 2016;65:25–36. doi: 10.1007/s00262-015-1770-9.
    1. Slingluff CL, Jr, Yamshchikov GV, Hogan KT, et al. Evaluation of the sentinel immunized node for immune monitoring of cancer vaccines. Ann Surg Oncol. 2008;15:3538–3549. doi: 10.1245/s10434-008-0046-4.
    1. Slingluff CL, Jr, Petroni GR, Olson W, et al. Helper T-cell responses and clinical activity of a melanoma vaccine with multiple peptides from MAGE and melanocytic differentiation antigens. J Clin Oncol. 2008;26:4973–4980. doi: 10.1200/JCO.2008.17.3161.
    1. Kaumaya PT, Foy KC, Garrett J, et al. Phase I active immunotherapy with combination of two chimeric, human epidermal growth factor receptor 2, B-cell epitopes fused to a promiscuous T-cell epitope in patients with metastatic and/or recurrent solid tumors. J Clin Oncol. 2009;27:5270–5277. doi: 10.1200/JCO.2009.22.3883.
    1. Sittler T, Zhou J, Park J, et al. Concerted potent humoral immune responses to autoantigens are associated with tumor destruction and favorable clinical outcomes without autoimmunity. Clin Cancer Res. 2008;14:3896–3905. doi: 10.1158/1078-0432.CCR-07-4782.
    1. Park JM, Terabe M, Steel JC, et al. Therapy of advanced established murine breast cancer with a recombinant adenoviral ErbB-2/neu vaccine. Cancer Res. 2008;68:1979–1987. doi: 10.1158/0008-5472.CAN-07-5688.
    1. Yamshchikov GV, Barnd DL, Eastham S, et al. Evaluation of peptide vaccine immunogenicity in draining lymph nodes and peripheral blood of melanoma patients. Int J Cancer. 2001;92:703–711. doi: 10.1002/1097-0215(20010601)92:5<703::AID-IJC1250>;2-5.
    1. Jahnmatz M, Kesa G, Netterlid E, Buisman AM, Thorstensson R, Ahlborg N. Optimization of a human IgG B-cell ELISpot assay for the analysis of vaccine-induced B-cell responses. J Immunol Methods. 2013;391:50–59. doi: 10.1016/j.jim.2013.02.009.
    1. Adekar SP, Jones RM, Elias MD, et al. Hybridoma populations enriched for affinity-matured human IgGs yield high-affinity antibodies specific for botulinum neurotoxins. J Immunol Methods. 2008;333:156–166. doi: 10.1016/j.jim.2008.01.015.
    1. Schultze JL, Cardoso AA, Freeman GJ, et al. Follicular lymphomas can be induced to present alloantigen efficiently: a conceptual model to improve their tumor immunogenicity. Proc Natl Acad Sci USA. 1995;92:8200–8204. doi: 10.1073/pnas.92.18.8200.
    1. Kearney JF, Radbruch A, Liesegang B, Rajewsky K. A new mouse myeloma cell line that has lost immunoglobulin expression but permits the construction of antibody-secreting hybrid cell lines. J Immunol. 1979;123:1548–1550.
    1. Zimmermann U, Gessner P, Schnettler R, Perkins S, Foung SK. Efficient hybridization of mouse-human cell lines by means of hypo-osmolar electrofusion. J Immunol Methods. 1990;134:43–50. doi: 10.1016/0022-1759(90)90110-H.
    1. Cham S, Chen L, Burke WM, et al. Utilization and outcomes of sentinel lymph node biopsy for vulvar cancer. Obstet Gynecol. 2016;128:754–760. doi: 10.1097/AOG.0000000000001648.
    1. Liberale G, Vankerckhove S, Galdon MG, et al. Sentinel lymph node detection by blue dye versus indocyanine green fluorescence imaging in colon cancer. Anticancer Res. 2016;36:4853–4858. doi: 10.21873/anticanres.11048.
    1. Wit EM, Acar C, Grivas N, et al. Sentinel node procedure in prostate cancer: a systematic review to assess diagnostic accuracy. Eur Urol. 2017;71:596–605. doi: 10.1016/j.eururo.2016.09.007.
    1. Krag DN. Minimal access surgery for staging regional lymph nodes: the sentinel-node concept. Curr Probl Surg. 1998;35:951–1016. doi: 10.1016/S0011-3840(98)80008-7.
    1. Li Q, Lao X, Pan Q, et al. Adoptive transfer of tumor reactive B cells confers host T-cell immunity and tumor regression. Clin Cancer Res. 2011;17:4987–4995. doi: 10.1158/1078-0432.CCR-11-0207.
    1. Rothe A, Klimka A, Tur MK, et al. Construction of phage display libraries from reactive lymph nodes of breast carcinoma patients and selection for specifically binding human single chain Fv on cell lines. Int J Mol Med. 2004;14:729–735.
    1. Xu MY, Xu XH, Chen GZ, et al. Production of a human single-chain variable fragment antibody against esophageal carcinoma. World J Gastroenterol. 2004;10:2619–2623. doi: 10.3748/wjg.v10.i18.2619.
    1. Hosking CG, Driguez P, McWilliam HE, et al. Using the local immune response from the natural buffalo host to generate an antibody fragment library that binds the early larval stages of Schistosoma japonicum. Int J Parasitol. 2015;45:729–740. doi: 10.1016/j.ijpara.2015.05.002.
    1. Ramos TC, Vinageras EN, Ferrer MC, et al. Treatment of NSCLC patients with an EGF-based cancer vaccine: report of a phase I trial. Cancer Biol Ther. 2006;5:145–149. doi: 10.4161/cbt.5.2.2334.
    1. Silveira EL, Kasturi SP, Kovalenkov Y, et al. Vaccine-induced plasmablast responses in rhesus macaques: phenotypic characterization and a source for generating antigen-specific monoclonal antibodies. J Immunol Methods. 2015;416:69–83. doi: 10.1016/j.jim.2014.11.003.

Source: PubMed

3
Subskrybuj