Improving immunological tumor microenvironment using electro-hyperthermia followed by dendritic cell immunotherapy

Yuk-Wah Tsang, Cheng-Chung Huang, Kai-Lin Yang, Mau-Shin Chi, Hsin-Chien Chiang, Yu-Shan Wang, Gabor Andocs, Andras Szasz, Wen-Tyng Li, Kwan-Hwa Chi, Yuk-Wah Tsang, Cheng-Chung Huang, Kai-Lin Yang, Mau-Shin Chi, Hsin-Chien Chiang, Yu-Shan Wang, Gabor Andocs, Andras Szasz, Wen-Tyng Li, Kwan-Hwa Chi

Abstract

Background: The treatment of intratumoral dentritic cells (DCs) commonly fails because it cannot evoke immunity in a poor tumor microenvironment (TME). Modulated electro-hyperthermia (mEHT, trade-name: oncothermia) represents a significant technological advancement in the hyperthermia field, allowing the autofocusing of electromagnetic power on a cell membrane to generate massive apoptosis. This approach turns local immunogenic cancer cell death (apoptosis) into a systemic anti-tumor immune response and may be implemented by treatment with intratumoral DCs.

Methods: The CT26 murine colorectal cancer model was used in this investigation. The inhibition of growth of the tumor and the systemic anti-tumor immune response were measured. The tumor was heated to a core temperature of 42 °C for 30 min. The matured synergetic DCs were intratumorally injected 24 h following mEHT was applied.

Results: mEHT induced significant apoptosis and enhanced the release of heat shock protein70 (Hsp70) in CT26 tumors. Treatment with mEHT-DCs significantly inhibited CT26 tumor growth, relative to DCs alone or mEHT alone. The secondary tumor protection effect upon rechallenging was observed in mice that were treated with mEHT-DCs. Immunohistochemical staining of CD45 and F4/80 revealed that mEHT-DC treatment increased the number of leukocytes and macrophages. Most interestingly, mEHT also induced infiltrations of eosinophil, which has recently been reported to be an orchestrator of a specific T cell response. Cytotoxic T cell assay and ELISpot assay revealed a tumor-specific T cell activity.

Conclusions: This study demonstrated that mEHT induces tumor cell apoptosis and enhances the release of Hsp70 from heated tumor cells, unlike conventional hyperthermia. mEHT can create a favorable tumor microenvironment for an immunological chain reaction that improves the success rate of intratumoral DC immunotherapy.

Figures

Fig. 1
Fig. 1
Apoptosis in mEHT-treated CT26 cells. One and a half million CT26 cells were heated to 42 °C for 30 min using LabEHY or a water bath (control). The apoptosis of CT26 cells after 24 h of hyperthermia treatment was analyzed using Annexin-V assay. (*, p < 0.05; n = 3)
Fig. 2
Fig. 2
Expression and release of Hsp70 following hyperthermia treatment. One and a half million CT26 cells were heated to 42 °C for 30 min using LabEHY or a water bath (control). a After 24 h of incubation, cells were harvested for western blot analysis of Hsp70 or HMGB1 protein in lysates of CT26 (control) (37 °C), a water bath (control) (42 °C) or mEHT. β-actin was used as an internal control. b After 24 h of incubation, supernatants were harvested and concentration of Hsp70 was measured using ELISA (n = 3). Data are presented as mean +/− SD from three independent experiments. * indicates p < 0.05
Fig. 3
Fig. 3
Inhibition of tumor growth and rechallenge inoculation. a Mice in different groups were injected with 5 × 105 CT26 tumor cells (subcutaneously) in right femoral area on day zero and treated with mEHT on day 14, before receiving DC injection on day 15. Data obtained from each mouse after tumor-cell inoculation (n = 7) were plotted. b A secondary rechallenge with CT26 tumor cells was administered to mice 30 days after first injection with DC alone or following mEHT or mEHT-DC therapy. Contra-lateral flanks of mice in treated groups and untreated control BALB/c mice were inoculated subcutaneously (1 × 105 parental CT26 cells). Percentage of mice that developed tumors at contra-lateral site was obtained using Kaplan–Meier method. (n = 7 mice per group.)
Fig. 4
Fig. 4
Areas of tumor infiltrated by immune cells after mEHT and DC treatment. Representative images of immunohistochemical straining revealed that quantities of CD45 (a) and F4/80 (b) were increased in tumors that were treated with DC and mEHT, either alone or in combination. Proportion of positive cells in one field that was randomly selected from ten fields, was calculated. c Amount of eosinophil increased significantly upon treatment of tumor; error bars represent standard errors. (***) P < 0.001 (t-test) relative to control. EH: electro-hyperethermia
Fig. 5
Fig. 5
Tumor-specific CTLs. Mice in various groups were injected with 5 × 105 CT26 tumor cells subcutaneously on day zero and with mEHT on day 14, before being given DC injection on day 15. On day 30 following tumor injection, splenocytes were harvested for CTL assay. Cytotoxic activity of splenocytes was determined by LDH-release assay at various effector/target cells (E/T) ratios
Fig. 6
Fig. 6
ELISpot assays. a Representative results of ELISpot assay of mice splenocytes that were pulsed with AH1. Top two rows, 2 × 105 splenocytes/well; bottom two rows, 105 splenocytes/well. PC: positive control, splenocytes treated with ConA (5 mg/ml) for 24 h. NC: negative control, splenocytes treated with BSA for 24 h. b Numbers of IFN-γ-secreting T-cells in DC-treated and mEHT + DC-treated mice significantly exceeded those in mice treated with mEHT alone and untreated control mice. Error bars represent standard errors. (***) P < 0.001 (t-test) relative to control. EH: electro-hyperethermia

References

    1. Formenti SC, Demaria S. Systemic effects of local radiotherapy. Lancet Oncol. 2009;10(7):718–726. doi: 10.1016/S1470-2045(09)70082-8.
    1. Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392(6673):245–252. doi: 10.1038/32588.
    1. Chi KH, Liu SJ, Li CP, Kuo HP, Wang YS, Chao Y, Hsieh SL. Combination of conformal radiotherapy and intratumoral injection of adoptive dendritic cell immunotherapy in refractory hepatoma. J Immunother. 2005;28(2):129–135. doi: 10.1097/01.cji.0000154248.74383.5e.
    1. Rabinovich GA, Gabrilovich D, Sotomayor EM. IMMUNOSUPPRESSIVE STRATEGIES THAT ARE MEDIATED BY TUMOR CELLS. Annu Rev Immunol. 2007;25:267–296. doi: 10.1146/annurev.immunol.25.022106.141609.
    1. Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, Liu YJ, Pulendran B, Palucka K. Immunobiology of dendritic cells. Annu Rev Immunol. 2000;18:767–811. doi: 10.1146/annurev.immunol.18.1.767.
    1. Chi KH, Wang YS, Kao SJ. Improving radioresponse through modification of the tumor immunological microenvironment. Cancer Biother Radiopharm. 2012;27(1):6–11. doi: 10.1089/cbr.2011.1048.
    1. Wang YS, Liu SJ, Huang SC, Chang CC, Huang YC, Fong WL, Chi MS, Chi KH. Recombinant heat shock protein 70 in combination with radiotherapy as a source of tumor antigens to improve dendritic cell immunotherapy. Front Oncol. 2012;2:149.
    1. Chi CH, Wang YS, Lai YS, Chi KH. Anti-tumor effect of in vivo IL-2 and GM-CSF electrogene therapy in murine hepatoma model. Anticancer Res. 2003;23(1A):315–321.
    1. Wang YS, Tsang YW, Chi CH, Chang CC, Chu RM, Chi KH. Synergistic anti-tumor effect of combination radio- and immunotherapy by electro-gene therapy plus intra-tumor injection of dendritic cells. Cancer Lett. 2008;266(2):275–285. doi: 10.1016/j.canlet.2008.02.063.
    1. Petrulio CA, Kim-Schulze S, Kaufman HL. The tumour microenvironment and implications for cancer immunotherapy. Expert Opin Biol Ther. 2006;6(7):671–684. doi: 10.1517/14712598.6.7.671.
    1. Kaufman HL. Manipulation of the Local Tumor Microenvironment for Cancer Immunotherapy. ASCO Education Book. 2004;23:165–171.
    1. Andocs G, Renner H, Balogh L, Fonyad L, Jakab C, Szasz A. Strong synergy of heat and modulated electromagnetic field in tumor cell killing. Strahlenther Onkol. 2009;185(2):120–126. doi: 10.1007/s00066-009-1903-1.
    1. Szasz A, Vincze G, Szasz O, Szasz N. An Energy Analysis of Extracellular Hyperthermia. Electromagn Biol Med. 2003;22(2–3):103–115. doi: 10.1081/JBC-120024620.
    1. Andocs G, Szasz O, Szasz A. Oncothermia treatment of cancer: from the laboratory to clinic. Electromagn Biol Med. 2009;28(2):148–165. doi: 10.1080/15368370902724633.
    1. Szasz A, Szasz N, Szasz O. Oncothermia: Principles and Practices. Netherlands: Springer; 2010. Oncothermia: A New Kind of Oncologic Hyperthermia; pp. 173–392.
    1. Szasz A, Vincze G. Dose concept of oncological hyperthermia: heat-equation considering the cell destruction. J Cancer Res Ther. 2006;2(4):171–181. doi: 10.4103/0973-1482.29827.
    1. Hegyi G, Szigeti GP, Szasz A. Hyperthermia versus Oncothermia: Cellular Effects in Complementary Cancer Therapy. Evid Based Complement Alternat Med. 2013;2013:672873. doi: 10.1155/2013/672873.
    1. Gadaleta-Caldarola G, Infusino S, Galise I, Ranieri G, Vinciarelli G, Fazio V, Divella R, Daniele A, Filippelli G, Gadaleta CD. Sorafenib and locoregional deep electro-hyperthermia in advanced hepatocellular carcinoma: A phase II study. Oncol lett. 2014;8(4):1783–1787.
    1. Fiorentini G, Giovanis P, Rossi S, Dentico P, Paola R, Turrisi G, Bernardeschi P. A phase II clinical study on relapsed malignant gliomas treated with electro-hyperthermia. In Vivo. 2006;20(6A):721–724.
    1. Szasz A. Current status of oncothermia therapy for lung cancer. The Korean journal of thoracic and cardiovascular surgery. 2014;47(2):77–93. doi: 10.5090/kjtcs.2014.47.2.77.
    1. Qin W, Akutsu Y, Andocs G, Suganami A, Hu X, Yusup G, Komatsu-Akimoto A, Hoshino I, Hanari N, Mori M, et al. Modulated electro-hyperthermia enhances dendritic cell therapy through an abscopal effect in mice. Oncol Rep. 2014;32(6):2373–2379.
    1. Dudek AM, Martin S, Garg AD, Agostinis P. Immature, Semi-Mature, and Fully Mature Dendritic Cells: Toward a DC-Cancer Cells Interface That Augments Anticancer Immunity. Front Immunol. 2013;4:438. doi: 10.3389/fimmu.2013.00438.
    1. Mole RH. Whole body irradiation; radiobiology or medicine? Br J Radiol. 1953;26(305):234–241. doi: 10.1259/0007-1285-26-305-234.
    1. Mukhopadhaya A, Mendecki J, Dong X, Liu L, Kalnicki S, Garg M, Alfieri A, Guha C. Localized Hyperthermia Combined with Intratumoral Dendritic Cells Induces Systemic Antitumor Immunity. Cancer Res. 2007;67(16):7798–7806. doi: 10.1158/0008-5472.CAN-07-0203.
    1. Friedman EJ. Immune modulation by ionizing radiation and its implications for cancer immunotherapy. Curr Pharm Des. 2002;8(19):1765–1780. doi: 10.2174/1381612023394089.
    1. Lugade AA, Sorensen EW, Gerber SA, Moran JP, Frelinger JG, Lord EM. Radiation-induced IFN-gamma production within the tumor microenvironment influences antitumor immunity. J Immunol. 2008;180(5):3132–3139. doi: 10.4049/jimmunol.180.5.3132.
    1. Teitz-Tennenbaum S, Li Q, Okuyama R, Davis MA, Sun R, Whitfield J, Knibbs RN, Stoolman LM, Chang AE. Mechanisms involved in radiation enhancement of intratumoral dendritic cell therapy. J Immunother. 2008;31(4):345–358. doi: 10.1097/CJI.0b013e318163628c.
    1. Andocs G, Meggyeshazi N, Balogh L, Spisak S, Maros ME, Balla P, Kiszner G, Teleki I, Kovago C, Krenacs T. Upregulation of heat shock proteins and the promotion of damage-associated molecular pattern signals in a colorectal cancer model by modulated electrohyperthermia. Cell Stress Chaperones. 2015;20(1):37–46. doi: 10.1007/s12192-014-0523-6.
    1. Szasz A. Challenges and Solutions in Oncological Hyperthermia. Thermal Med. 2013;29(1):1–23. doi: 10.3191/thermalmed.29.1.
    1. Wang YS, Chi KH, Liao KW, Liu CC, Cheng CL, Lin YC, Cheng CH, Chu RM. Characterization of canine monocyte-derived dendritic cells with phenotypic and functional differentiation. Can J Vet Res. 2007;71(3):165–174.
    1. Hirasawa M, Ito Y, Shibata MA, Otsuki Y. Mechanism of inflammation in murine eosinophilic myocarditis produced by adoptive transfer with ovalbumin challenge. Int Arch Allergy Immunol. 2007;142(1):28–39. doi: 10.1159/000095996.
    1. Huang AY, Gulden PH, Woods AS, Thomas MC, Tong CD, Wang W, Engelhard VH, Pasternack G, Cotter R, Hunt D, et al. The immunodominant major histocompatibility complex class I-restricted antigen of a murine colon tumor derives from an endogenous retroviral gene product. Proc Natl Acad Sci U S A. 1996;93(18):9730–9735. doi: 10.1073/pnas.93.18.9730.
    1. Simson L, Ellyard JI, Dent LA, Matthaei KI, Rothenberg ME, Foster PS, Smyth MJ, Parish CR. Regulation of carcinogenesis by IL-5 and CCL11: a potential role for eosinophils in tumor immune surveillance. J Immunol. 2007;178(7):4222–4229. doi: 10.4049/jimmunol.178.7.4222.
    1. Cormier SA, Taranova AG, Bedient C, Nguyen T, Protheroe C, Pero R, Dimina D, Ochkur SI, O'Neill K, Colbert D, et al. Pivotal Advance: eosinophil infiltration of solid tumors is an early and persistent inflammatory host response. J Leukoc Biol. 2006;79(6):1131–1139. doi: 10.1189/jlb.0106027.
    1. Carretero R, Sektioglu IM, Garbi N, Salgado OC, Beckhove P, Hammerling GJ. Eosinophils orchestrate cancer rejection by normalizing tumor vessels and enhancing infiltration of CD8(+) T cells. Nat Immunol. 2015;16(6):609–617. doi: 10.1038/ni.3159.
    1. Meggyeshazi N, Andocs G, Balogh L, Balla P, Kiszner G, Teleki I, et al. DNA fragmentation and caspase-independent programmed cell death by modulated electrohyperthermia. Strahlenther Onkol. 2014;1–8.
    1. Feng H, Zeng Y, Graner MW, Katsanis E. Stressed apoptotic tumor cells stimulate dendritic cells and induce specific cytotoxic T cells. Blood. 2002;100(12):4108–4115. doi: 10.1182/blood-2002-05-1389.
    1. Jaffe ML, Arai H, Nabel GJ. Mechanisms of tumor-induced immunosuppression: evidence for contact-dependent T cell suppression by monocytes. Mol Med. 1996;2(6):692–701.
    1. Brusa D, Migliore E, Garetto S, Simone M, Matera L. Immunogenicity of 56 degree C and UVC-treated prostate cancer is associated with release of HSP70 and HMGB1 from necrotic cells. Prostate. 2009;69(12):1343–1352. doi: 10.1002/pros.20981.
    1. Gallucci S, Lolkema M, Matzinger P. Natural adjuvants: Endogenous activators of dendritic cells. Nat Med. 1999;5(11):1249–1255. doi: 10.1038/15200.
    1. Lutz MB, Kukutsch N, Ogilvie AL, Rossner S, Koch F, Romani N, Schuler G. An advanced culture method for generating large quantities of highly pure dendritic cells from mouse bone marrow. J Immunol Methods. 1999;223(1):77–92. doi: 10.1016/S0022-1759(98)00204-X.
    1. Candido KA, Shimizu K, McLaughlin JC, Kunkel R, Fuller JA, Redman BG, Thomas EK, Nickoloff BJ, Mule JJ. Local administration of dendritic cells inhibits established breast tumor growth: implications for apoptosis-inducing agents. Cancer Res. 2001;61(1):228–236.
    1. Nosho K, Baba Y, Tanaka N, Shima K, Hayashi M, Meyerhardt JA, Giovannucci E, Dranoff G, Fuchs CS, Ogino S. Tumour-infiltrating T-cell subsets, molecular changes in colorectal cancer, and prognosis: cohort study and literature review. J Pathol. 2010;222(4):350–366. doi: 10.1002/path.2774.
    1. Benencia F, Sprague L, McGinty J, Pate M, Muccioli M. Dendritic cells the tumor microenvironment and the challenges for an effective antitumor vaccination. J Biomed Biotechnol. 2012;2012:425476. doi: 10.1155/2012/425476.
    1. Harvey RD. Immunologic and Clinical Effects of Targeting PD-1 in Lung Cancer. Clin Pharmacol Ther. 2014;96(2):214–223. doi: 10.1038/clpt.2014.74.
    1. Sharma P, Allison JP. The future of immune checkpoint therapy. Science. 2015;348(6230):56–61. doi: 10.1126/science.aaa8172.

Source: PubMed

3
Tilaa