open access

Vol 72, No 3 (2022)
Review paper
Published online: 2022-04-15
Get Citation

Immunological aspects of heat shock protein functions and their significance in the development of cancer vaccines

Iryna Boliukh1, Agnieszka Rombel-Bryzek1, Barbara Radecka23
DOI: 10.5603/NJO.a2022.0024
·
Nowotwory. Journal of Oncology 2022;72(3):174-183.
Affiliations
  1. Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medical Sciences, University of Opole, Opole, Poland
  2. Department of Oncology, Institute of Medical Sciences, University of Opole, Opole, Poland
  3. Department of Clinical Oncology, Tadeusz Koszarowski Cancer Centre in Opole, Opole, Poland

open access

Vol 72, No 3 (2022)
Review article
Published online: 2022-04-15

Abstract

The primary function of intracellular heat shock proteins (HSPs) is to protect the cell by suppressing the effects of va­rious stress factors by either refolding misfolded proteins or blocking apoptosis. After neoplastic transformation, cells overexpress HSPs, which act as factors promoting the neoplastic process by stabilizing proteins responsible for carci­nogenesis, however, HSPs can be released into the extracellular environment where they act as important modulators of the immune response. In a tumor microenvironment, extracellular HSPs are able to induce a pro- or anti-neoplastic response, using various mechanisms of affecting immune cells, The study of the role of extracellular HSPs in immuno­modulation processes is a very important direction in the search for new methods of cancer treatment. This review summarizes reports on the use of HSPs in immunotherapeutic cancer strategies, in particular in cancer vaccine design.

Abstract

The primary function of intracellular heat shock proteins (HSPs) is to protect the cell by suppressing the effects of va­rious stress factors by either refolding misfolded proteins or blocking apoptosis. After neoplastic transformation, cells overexpress HSPs, which act as factors promoting the neoplastic process by stabilizing proteins responsible for carci­nogenesis, however, HSPs can be released into the extracellular environment where they act as important modulators of the immune response. In a tumor microenvironment, extracellular HSPs are able to induce a pro- or anti-neoplastic response, using various mechanisms of affecting immune cells, The study of the role of extracellular HSPs in immuno­modulation processes is a very important direction in the search for new methods of cancer treatment. This review summarizes reports on the use of HSPs in immunotherapeutic cancer strategies, in particular in cancer vaccine design.

Get Citation

Keywords

heat shock proteins; cancer immunotherapy; vaccine

About this article
Title

Immunological aspects of heat shock protein functions and their significance in the development of cancer vaccines

Journal

Nowotwory. Journal of Oncology

Issue

Vol 72, No 3 (2022)

Article type

Review paper

Pages

174-183

Published online

2022-04-15

Page views

4637

Article views/downloads

453

DOI

10.5603/NJO.a2022.0024

Bibliographic record

Nowotwory. Journal of Oncology 2022;72(3):174-183.

Keywords

heat shock proteins
cancer immunotherapy
vaccine

Authors

Iryna Boliukh
Agnieszka Rombel-Bryzek
Barbara Radecka

References (87)
  1. Smyth MJ, Crowe NY, Godfrey DI. NK cells and NKT cells collaborate in host protection from methylcholanthrene-induced fibrosarcoma. Int Immunol. 2001; 13(4): 459–463.
  2. Simson L, Ellyard JI, Dent LA, et al. Regulation of carcinogenesis by IL-5 and CCL11: a potential role for eosinophils in tumor immune surveillance. J Immunol. 2007; 178(7): 4222–4229.
  3. Derhovanessian E, Solana R, Larbi A, et al. Immunity, ageing and cancer. Immun Ageing. 2008; 5: 11.
  4. Frisch M, Biggar RJ, Engels EA, et al. AIDS-Cancer Match Registry Study Group. Association of cancer with AIDS-related immunosuppression in adults. JAMA. 2001; 285(13): 1736–1745.
  5. Das JK, Xiong X, Ren X, et al. Heat Shock Proteins in Cancer Immunotherapy. J Oncol. 2019; 2019: 3267207.
  6. Banerjee S, Lin CFL, Skinner KA, et al. Heat shock protein 27 differentiates tolerogenic macrophages that may support human breast cancer progression. Cancer Res. 2011; 71(2): 318–327.
  7. Calderwood SK, Gong J. Heat Shock Proteins Promote Cancer: It's a Protection Racket. Trends Biochem Sci. 2016; 41(4): 311–323.
  8. Thériault JR, Adachi H, Calderwood SK. Role of scavenger receptors in the binding and internalization of heat shock protein 70. J Immunol. 2006; 177(12): 8604–8611.
  9. Delneste Y, Magistrelli G, Gauchat JF, et al. Involvement of LOX-1 in Dendritic Cell-Mediated Antigen Cross-Presentation. Immunity. 2002; 17(3): 353–362.
  10. Stocki P, Morris NJ, Preisinger C, et al. Identification of potential HLA class I and class II epitope precursors associated with heat shock protein 70 (HSPA). Cell Stress Chaperones. 2010; 15(5): 729–741.
  11. Mambula SS, Calderwood SK. Heat shock protein 70 is secreted from tumor cells by a nonclassical pathway involving lysosomal endosomes. J Immunol. 2006; 177(11): 7849–7857.
  12. Pockley AG, Muthana M, Calderwood SK. The dual immunoregulatory roles of stress proteins. Trends Biochem Sci. 2008; 33(2): 71–79.
  13. Chalmin F, Ladoire S, Mignot G, et al. Membrane-associated Hsp72 from tumor-derived exosomes mediates STAT3-dependent immunosuppressive function of mouse and human myeloid-derived suppressor cells. J Clin Invest. 2010; 120(2): 457–471.
  14. Borges TJ, Wieten L, van Herwijnen MJC, et al. The anti-inflammatory mechanisms of Hsp70. Front Immunol. 2012; 3: 95.
  15. de Kleer I, Vercoulen Y, Klein M, et al. CD30 discriminates heat shock protein 60-induced FOXP3+ CD4+ T cells with a regulatory phenotype. J Immunol. 2010; 185(4): 2071–2079.
  16. Aalberse JA, Kapitein B, de Roock S, et al. Cord blood CD4+ T cells respond to self heat shock protein 60 (HSP60). PLoS One. 2011; 6(9): e24119.
  17. Cohen-Sfady M, Nussbaum G, Pevsner-Fischer M, et al. Heat shock protein 60 activates B cells via the TLR4-MyD88 pathway. J Immunol. 2005; 175(6): 3594–3602.
  18. Tsai YP, Yang MH, Huang CH, et al. Interaction between HSP60 and beta-catenin promotes metastasis. Carcinogenesis. 2009; 30(6): 1049–1057.
  19. Stocki P, Wang XN, Dickinson AM. Inducible heat shock protein 70 reduces T cell responses and stimulatory capacity of monocyte-derived dendritic cells. J Biol Chem. 2012; 287(15): 12387–12394.
  20. Gastpar R, Gehrmann M, Bausero MA, et al. Heat shock protein 70 surface-positive tumor exosomes stimulate migratory and cytolytic activity of natural killer cells. Cancer Res. 2005; 65(12): 5238–5247.
  21. Gehrmann M, Marienhagen J, Eichholtz-Wirth H, et al. Dual function of membrane-bound heat shock protein 70 (Hsp70), Bag-4, and Hsp40: protection against radiation-induced effects and target structure for natural killer cells. Cell Death Differ. 2005; 12(1): 38–51.
  22. Maki RG, Old LJ, Srivastava PK. Human homologue of murine tumor rejection antigen gp96: 5'-regulatory and coding regions and relationship to stress-induced proteins. Proc Natl Acad Sci U S A. 1990; 87(15): 5658–5662.
  23. Udono H, Srivastava PK. Heat shock protein 70-associated peptides elicit specific cancer immunity. J Exp Med. 1993; 178(4): 1391–1396.
  24. Multhoff G. Activation of natural killer cells by heat shock protein 70. 2002. Int J Hyperthermia. 2009; 25(3): 169–175.
  25. Gross C, Hansch D, Gastpar R, et al. Interaction of heat shock protein 70 peptide with NK cells involves the NK receptor CD94. Biol Chem. 2003; 384(2): 267–279.
  26. Multhoff G. Heat shock protein 70 (Hsp70) stimulates proliferation and cytolytic activity of natural killer cells. Exp Hematol. 1999; 27(11): 1627–1636.
  27. Tsan MF, Gao B. Cytokine function of heat shock proteins. Am J Physiol Cell Physiol. 2004; 286(4): C739–C744.
  28. McNulty S, Colaco CA, Blandford LE, et al. Heat-shock proteins as dendritic cell-targeting vaccines--getting warmer. Immunology. 2013; 139(4): 407–415.
  29. Asea A, Kraeft SK, Kurt-Jones EA, et al. HSP70 stimulates cytokine production through a CD14-dependant pathway, demonstrating its dual role as a chaperone and cytokine. Nat Med. 2000; 6(4): 435–442.
  30. Singh-Jasuja H, Scherer H, Hilf N, et al. The heat shock protein gp96 induces maturation of dendritic cells and down-regulation of its receptor. Eur J Immunol. 2000; 30(8): 2211–2215, doi: 10.1002/1521-4141(2000)30:8<2211::aid-immu2211>3.0.co;2-0.
  31. Panjwani NN, Popova L, Srivastava PK. Heat shock proteins gp96 and hsp70 activate the release of nitric oxide by APCs. J Immunol. 2002; 168(6): 2997–3003.
  32. Chen T, Guo J, Han C, et al. Heat shock protein 70, released from heat-stressed tumor cells, initiates antitumor immunity by inducing tumor cell chemokine production and activating dendritic cells via TLR4 pathway. J Immunol. 2009; 182(3): 1449–1459.
  33. Murshid A, Gong J, Stevenson MA, et al. Heat shock proteins and cancer vaccines: developments in the past decade and chaperoning in the decade to come. Expert Rev Vaccines. 2011; 10(11): 1553–1568.
  34. Murshid A, Theriault J, Gong J, et al. Investigating receptors for extracellular heat shock proteins. Methods Mol Biol. 2011; 787: 289–302.
  35. Linder M, Pogge von Strandmann E. The Role of Extracellular HSP70 in the Function of Tumor-Associated Immune Cells. Cancers (Basel). 2021; 13(18).
  36. Castelli C, Ciupitu AM, Rini F, et al. Human heat shock protein 70 peptide complexes specifically activate antimelanoma T cells. Cancer Res. 2001; 61(1): 222–227.
  37. Antony GK, Dudek AZ. Interleukin 2 in cancer therapy. Curr Med Chem. 2010; 17(29): 3297–3302.
  38. Parker BS, Rautela J, Hertzog PJ. Antitumour actions of interferons: implications for cancer therapy. Nat Rev Cancer. 2016; 16(3): 131–144.
  39. Metcalf D. The colony-stimulating factors and cancer. Nat Rev Cancer. 2010; 10(6): 425–434.
  40. Goldenberg M. Trastuzumab, a recombinant DNA-derived humanized monoclonal antibody, a novel agent for the treatment of metastatic breast cancer. Clin Ther. 1999; 21(2): 309–318.
  41. June CH. Adoptive T cell therapy for cancer in the clinic. J Clin Invest. 2007; 117(6): 1466–1476.
  42. Farkona S, Diamandis EP, Blasutig IM. Cancer immunotherapy: the beginning of the end of cancer? BMC Med. 2016; 14: 73.
  43. Palucka K, Banchereau J. Dendritic-cell-based therapeutic cancer vaccines. Immunity. 2013; 39(1): 38–48.
  44. Plosker GL. Sipuleucel-T in metastatic castration-resistant prostate cancer: profile report. BioDrugs. 2011; 25(4): 255–256.
  45. Yu M, Finn OJ. DNA vaccines for cancer too. Cancer Immunol Immunother. 2006; 55(2): 119–130.
  46. Zahm CD, Colluru VT, McNeel DG. DNA vaccines for prostate cancer. Pharmacol Ther. 2017; 174: 27–42.
  47. Snook AE, Baybutt TR, Hyslop T, et al. Preclinical Evaluation of a Replication-Deficient Recombinant Adenovirus Serotype 5 Vaccine Expressing Guanylate Cyclase C and the PADRE T-helper Epitope. Hum Gene Ther Methods. 2016; 27(6): 238–250.
  48. Madan RA, Bilusic M, Heery C, et al. Clinical evaluation of TRICOM vector therapeutic cancer vaccines. Semin Oncol. 2012; 39(3): 296–304.
  49. Larocca C, Schlom J. Viral vector-based therapeutic cancer vaccines. Cancer J. 2011; 17(5): 359–371.
  50. Tangney M, Gahan CGM. Listeria monocytogenes as a vector for anti-cancer therapies. Curr Gene Ther. 2010; 10(1): 46–55.
  51. Bolhassani A, Rafati S. Heat-shock proteins as powerful weapons in vaccine development. Expert Rev Vaccines. 2008; 7(8): 1185–1199.
  52. Pakravan N, Soudi S, Hassan ZM. N-terminally fusion of Her2/neu to HSP70 decreases efficiency of Her2/neu DNA vaccine. Cell Stress Chaperones. 2010; 15(5): 631–638.
  53. Garrod TJ, Grubor-Bauk B, Gargett T, et al. DNA vaccines encoding membrane-bound or secreted forms of heat shock protein 70 exhibit improved potency. Eur J Immunol. 2014; 44(7): 1992–2002.
  54. Wang L, Rollins L, Gu Q, et al. A Mage3/Heat Shock Protein70 DNA vaccine induces both innate and adaptive immune responses for the antitumor activity. Vaccine. 2009; 28(2): 561–570.
  55. Delpino A, Falcioni R, Ferrini U. Modulation of Heat Shock Protein Synthesis in Two Human Melanoma Cell Lines. Tumori. 1984; 70(5): 393–398.
  56. Gross L. Intradermal immunization of C3H mice against a sarcoma that originated in an animal of the same line. Cancer Res. 1943: 323–326.
  57. Srivastava PK, Das MR. The serologically unique cell surface antigen of Zajdela ascitic hepatoma is also its tumor-associated transplantation antigen. Int J Cancer. 1984; 33(3): 417–422.
  58. Ullrich SJ, Robinson EA, Law LW, et al. A mouse tumor-specific transplantation antigen is a heat shock-related protein. Proc Natl Acad Sci U S A. 1986; 83(10): 3121–3125.
  59. Baldin AV, Zamyatnin AA, Bazhin AV, et al. Advances in the Development of Anticancer HSP-based Vaccines. Curr Med Chem. 2019; 26(3): 427–445.
  60. Chandawarkar RY, Wagh MS, Kovalchin JT, et al. Immune modulation with high-dose heat-shock protein gp96: therapy of murine autoimmune diabetes and encephalomyelitis. Int Immunol. 2004; 16(4): 615–624.
  61. Binder RJ, Zhou YuJ, Messmer MN, et al. CD91-Dependent Modulation of Immune Responses by Heat Shock Proteins: A Role in Autoimmunity. Autoimmune Dis. 2012; 2012: 863041.
  62. Bu N, Li QL, Feng Qi, et al. Immune protection effect of exosomes against attack of L1210 tumor cells. Leuk Lymphoma. 2006; 47(5): 913–918.
  63. Cho Ja, Lee YS, Kim SH, et al. MHC independent anti-tumor immune responses induced by Hsp70-enriched exosomes generate tumor regression in murine models. Cancer Lett. 2009; 275(2): 256–265.
  64. Xie Y, Bai Ou, Zhang H, et al. Membrane-bound HSP70-engineered myeloma cell-derived exosomes stimulate more efficient CD8(+) CTL- and NK-mediated antitumour immunity than exosomes released from heat-shocked tumour cells expressing cytoplasmic HSP70. J Cell Mol Med. 2010; 14(11): 2655–2666.
  65. Shevtsov M, Pitkin E, Ischenko A, et al. Hsp70-Activated NK Cells in Combination With PD-1 Inhibition Significantly Increase Overall Survival in Preclinical Models of Glioblastoma and Lung Cancer. Front Immunol. 2019; 10: 454.
  66. Komarova EY, Suezov RV, Nikotina AD, et al. Hsp70-containing extracellular vesicles are capable of activating of adaptive immunity in models of mouse melanoma and colon carcinoma. Sci Rep. 2021; 11(1): 21314.
  67. Behzadi E, Hosseini HM, Halabian R, et al. Macrophage cell-derived exosomes/staphylococcal enterotoxin B against fibrosarcoma tumor. Microb Pathog. 2017; 111: 132–138.
  68. Eton O, Ross MI, East MJo, et al. Autologous tumor-derived heat-shock protein peptide complex-96 (HSPPC-96) in patients with metastatic melanoma. J Transl Med. 2010; 8: 9.
  69. Bloch O, Crane CA, Fuks Y, et al. Heat-shock protein peptide complex-96 vaccination for recurrent glioblastoma: a phase II, single-arm trial. Neuro Oncol. 2014; 16(2): 274–279.
  70. Testori A, Richards J, Whitman E, et al. C-100-21 Study Group. Phase III comparison of vitespen, an autologous tumor-derived heat shock protein gp96 peptide complex vaccine, with physician's choice of treatment for stage IV melanoma: the C-100-21 Study Group. J Clin Oncol. 2008; 26(6): 955–962.
  71. Wood C, Srivastava P, Bukowski R, et al. An adjuvant autologous therapeutic vaccine (HSPPC-96; vitespen) versus observation alone for patients at high risk of recurrence after nephrectomy for renal cell carcinoma: a multicentre, open-label, randomised phase III trial. Lancet. 2008; 372(9633): 145–154.
  72. Li Z, Qiao Yi, Liu B, et al. Combination of imatinib mesylate with autologous leukocyte-derived heat shock protein and chronic myelogenous leukemia. Clin Cancer Res. 2005; 11(12): 4460–4468.
  73. Roman LD, Wilczynski S, Muderspach LI, et al. A phase II study of Hsp-7 (SGN-00101) in women with high-grade cervical intraepithelial neoplasia. Gynecol Oncol. 2007; 106(3): 558–566.
  74. Victora GD, Socorro-Silva A, Volsi EC, et al. Immune response to vaccination with DNA-Hsp65 in a phase I clinical trial with head and neck cancer patients. Cancer Gene Ther. 2009; 16(7): 598–608.
  75. Trimble CL, Peng S, Kos F, et al. A phase I trial of a human papillomavirus DNA vaccine for HPV16+ cervical intraepithelial neoplasia 2/3. Clin Cancer Res. 2009; 15(1): 361–367.
  76. Zappasodi R, Pupa SM, Ghedini GC, et al. Improved clinical outcome in indolent B-cell lymphoma patients vaccinated with autologous tumor cells experiencing immunogenic death. Cancer Res. 2010; 70(22): 9062–9072.
  77. Krause SW, Gastpar R, Andreesen R, et al. Treatment of colon and lung cancer patients with ex vivo heat shock protein 70-peptide-activated, autologous natural killer cells: a clinical phase i trial. Clin Cancer Res. 2004; 10(11): 3699–3707.
  78. Matsui H, Hazama S, Tamada K, et al. Identification of a Promiscuous Epitope Peptide Derived From HSP70. J Immunother. 2019; 42(7): 244–250.
  79. Shimizu Y, Yoshikawa T, Kojima T, et al. Heat shock protein 105 peptide vaccine could induce antitumor immune reactions in a phase I clinical trial. Cancer Sci. 2019; 110(10): 3049–3060.
  80. Shevtsov MA, Pozdnyakov AV, Mikhrina AL, et al. Effective immunotherapy of rat glioblastoma with prolonged intratumoral delivery of exogenous heat shock protein Hsp70. Int J Cancer. 2014; 135(9): 2118–2128.
  81. Chang CL, Tsai YC, He L, et al. Cancer immunotherapy using irradiated tumor cells secreting heat shock protein 70. Cancer Res. 2007; 67(20): 10047–10057.
  82. Abkin SV, Pankratova KM, Komarova EYu, et al. Hsp70 chaperone-based gel composition as a novel immunotherapeutic anti-tumor tool. Cell Stress Chaperones. 2013; 18(3): 391–396.
  83. Shevtsov MA, Kim AV, Samochernych KA, et al. Pilot study of intratumoral injection of recombinant heat shock protein 70 in the treatment of malignant brain tumors in children. Onco Targets Ther. 2014; 7: 1071–1081.
  84. Tischer S, Basila M, Maecker-Kolhoff B, et al. Heat shock protein 70/peptide complexes: potent mediators for the generation of antiviral T cells particularly with regard to low precursor frequencies. J Transl Med. 2011; 9: 175.
  85. Jiang J, Xie D, Zhang W, et al. Fusion of Hsp70 to Mage-a1 enhances the potency of vaccine-specific immune responses. J Transl Med. 2013; 11: 300.
  86. Schildkopf P, Frey B, Ott OJ, et al. Radiation combined with hyperthermia induces HSP70-dependent maturation of dendritic cells and release of pro-inflammatory cytokines by dendritic cells and macrophages. Radiother Oncol. 2011; 101(1): 109–115.
  87. Ito A, Matsuoka F, Honda H, et al. Antitumor effects of combined therapy of recombinant heat shock protein 70 and hyperthermia using magnetic nanoparticles in an experimental subcutaneous murine melanoma. Cancer Immunol Immunother. 2004; 53(1): 26–32.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

Wydawcą serwisu jest VM Media Group sp. z o.o., ul. Świętokrzyska 73, 80–180 Gdańsk

tel.:+48 58 320 94 94, faks:+48 58 320 94 60, e-mail: viamedica@viamedica.pl