784
Views
7
CrossRef citations to date
0
Altmetric
Letter to the Editor

Hyperthermia classic commentary: Activation of natural killer (NK) cells by heat shock protein 70, Gabriele Multhoff, International Journal of Hyperthermia, 2002;18:576–585

Pages 176-179 | Published online: 09 Jul 2009

Hsp70 activated NK cells in cancer therapy

Dear Sir

Identification of molecular markers that can be utilized as targets for cancer immunotherapy is a critical goal for the development of therapeutics and also for improved diagnostic applications. Ideally, a biomarker should be selectively expressed on tumor but not on normal cells, or at least, should be expressed at a much lower level on normal cells. A large scale screening program on more than 900 tumor biopsies derived from patients with different tumor entities including gastrointestinal, urogenital and bronchial tumors, as well as hematological diseases, and their corresponding normal tissues revealed that a membrane-bound form of Hsp70 could fulfill this criterion. However, an Hsp70 membrane-positive phenotype has also been associated with a higher metastatic potential and an unfavorable prognosis in malignant melanoma Citation1, lower rectal cancer, squamous cell carcinoma of the lung Citation2 and in acute myeloid leukemia Citation3, Citation4. These latter findings suggest that elevated Hsp70 expression at the plasma membrane may predict a more severe cancer pathology. In line with these findings, the membrane density of Hsp70 has been found to be enhanced in cells within metastases when compared to those in primary tumors using a xenograft tumor mouse model Citation5.

How might plasma membrane-associated Hsp70 be useful as a therapeutic target? In 1995, we were the first to demonstrate that membrane-bound Hsp70 provides a tumor-specific target structure for lytic natural killer (NK) cells of the innate immune system Citation6, Citation7. Moreover, the expression of Hsp70 on the cell surface of tumors plays an immunomodulatory role by increasing tumor cell lysis by NK cells when in the presence of pro-inflammatory cytokines Citation6, Citation8, Citation9. Furthermore, a correlation of the cytolytic activity of NK cells with the Hsp70 density on tumor cells also has been determined Citation10, Citation11. Unstimulated NK cells derived from patients did not show any killing activity towards autologous tumors. This might be due to the fact that these NK cells might be in a resting stage or that the tumor has evolved tumor escape mechanisms.

In this commentary we provide an update on the role of Hsp70 for the innate immune system. We will discuss research regarding the anchorage Citation12 and the export of Hsp70 from tumor cells Citation13, as well as the mechanism of kill of Hsp70 membrane-positive tumors Citation14. Furthermore, we will summarize the results of a phase I clinical trial which has been performed in patients with metastasized colorectal and non-small lung cell carcinoma (NSCLC), applying ex vivo Hsp70 peptide-activated, autologous NK cells. Finally, we will report on a targeted NK cell-based adjuvant immunotherapy for the treatment of patients with NSCLC after standard radiochemotherapy which will be initiated this year as a ‘proof of principle’ clinical phase II randomized trial.

Anchorage of Hsp70 in the plasma membrane of tumor cells

As mentioned before, we found Hsp70 to be selectively expressed on the cell surface of human tumors, but not on normal cells. However, it remains unclear how this tumor-specificity is maintained. Hsp70, the major stress-inducible member of the HSP70 family, is a cytosolic protein which lacks a classical transmembrane domain. Therefore, it was assumed that Hsp70 is presented on the plasma membrane via linkage to a cell surface receptor. However, neither high salt conditions nor changes in the pH altered the Hsp70 membrane expression pattern (unpublished observations). Therefore it appears highly unlikely that Hsp70 is bound to a receptor on the plasma membrane. With respect to previously published data by Hightower and Guidon Citation15 and the group of Antonio DeMaio Citation16–18 showing a physical interaction of Hsp70 with fatty acid components, in collaboration with Gerd Schmitz, Claudia Steinem and Cliff Lingwood, we performed lipidomic analyses of isolated plasma membranes and cholesterol rich microdomains (CRMs) from tumor cell lines with differential Hsp70 membrane expression. It became apparent that the lipid composition of CRMs of Hsp70 membrane-positive and–negative tumor cells differed in the content of globoyltriaosylceramide (Gb3) suggesting the hypothesis that there is an interaction between Hsp70 and the glycosphingolipid Gb3. This hypothesis could be further supported by vesicle aggregation assays using artificial unilamellar raft-like liposomes which contain Gb3. Since Gb3 is frequently over-expressed in the plasma membrane of human colorectal tumors and metastases derived thereof, this may provide a mechanism by which Hsp70 is expressed on the tumor cell surface Citation19. The fact that apart from germline B cells, Gb3 is not found in the plasma membrane of normal cells and is over-expressed in CRMs of tumors might explain the tumor-specificity of the Hsp70 cell surface expression. Ongoing research is aiming to further characterize the molecular nature of the Hsp70-lipid interaction.

Export of Hsp70 from tumor cells

It is known that HSPs are found not only in the cytosol, on the plasma membrane but are also found in the extracellular space Citation20, Citation21. Members of the Hsp70 family in cooperation with other chaperones are major players in transport mechanisms across lipid membranes Citation22. Therefore, it is conceivable that Hsp70 might fulfill shuttle functions from the cytosol into the extracellular milieu. Although the mechanism of export remains elusive, release of Hsp70 from viable cells with intact plasma membrane has been documented Citation23, Citation24. Blocking experiments indicated that Hsp70 secretion is mediated through a non-classical ER-Golgi pathway since neither brefeldin A nor monensin affect Hsp70 export. Since there is very little spontaneous release of free Hsp70 protein from viable cells, it was speculated that Hsp70 might be released in vesicles. Indeed, following ultracentrifugation of supernatants of tumor cells, detergent-soluble liposomes could be isolated which present Hsp70 on their surface Citation25. An enrichment of the small GTPase Rab-4 within these vesicles supports the idea of an intracellular transport route from the early endosome to the plasma membrane and finally into the extracellular space. A recent study has documented the presence of Gb3-containing lipid rafts in the vesicular membranes Citation19. This finding might suggest a mechanism by which Hsp70 can be anchored within the vesicular membranes.

Mechanism of tumor cell kill

Cell surface-bound Hsp70 renders tumor cells more sensitive to the cytolytic attack mediated by NK cells which have been stimulated with a 14-mer amino acid Hsp70 sequence TKD (aa 450–463) plus low dose IL-2. Flow cytometry analysis of these stimulated NK cells compared to unstimulated NK cells revealed an up-regulation of markers including CD56, CD94, NKG2D, natural cytotoxicity receptors (NCRs) and activation markers CD69 and CD25. In contrast, killer cell immunoglobulin-like receptors (KIRs) appeared to be unaffected and the low affinity receptor CD16 was found to be down-regulated by TKD-stimulation Citation26. Due to their cytosolic motifs KIRs can either inhibit (long ITIM) or activate (short ITAM) the cytolytic activity of NK cells. Overall, the phenotype of the NK cells after TKD/IL-2 stimulation resembled the CD16dim/CD56 bright phenotype, which are considered as cytokine producers such as IFN-gamma upon stimulation with interleukins Citation27. This NK cell type has been found to possess low amounts of perforin and thus is expected to have a low cytotoxic activity. However, we have shown that our TKD/IL-2 activated NK cells are highly efficient in killing Hsp70 membrane-positive tumor cells. With regard to these findings we studied the mechanism of kill of Hsp70 membrane-positive tumors in more detail. By affinity chromatography, it was shown that the apoptosis-inducing enzyme granzyme B interacts with full length Hsp70 as well as with the Hsp70 peptide TKD. An incubation of tumor cells with differential Hsp70 membrane expression pattern with purified granzyme B in the absence of perforin indicated that only Hsp70 membrane-positive tumors were able to internalize granzyme B into the cytosol. Following uptake and activation of the caspase pathway these tumor cells underwent apoptotic cell death Citation14. From these results, we propose that Hsp70 on the plasma membrane of tumor cells might serve as an entry port for granzyme B, even in the absence of perforin, and thus might enable apoptotic cell death caused by CD16dim/CD56bright NK cells.

More recent data have shown that Hsp70 membrane-negative tumors which do express NKG2D ligands such as MICA/B on their cell surface can also be killed by TKD/IL-2 activated NK cells Citation28. The best lysis was observed in tumors that express both Hsp70 and NKG2D ligands on their cell surface Citation28. It can be speculated that the synergistic effects of two immunological danger signals might result in an enhanced cytotoxic activity of NK cells against tumors. The role of standard radiochemotherapy Citation29 and hyperthermia as potential tools to enhance the expression of danger signals on the cell surface of tumor cells is presently under investigation Citation30. Indeed, recent studies show the potential for hyperthermia to increase the expression of NKG2D ligands on the surface of tumor cells Citation31.

Clinical application of Hsp70 peptide activated NK cells

An Hsp70 membrane-positive phenotype was associated with poor clinical outcome in a large number of tumor entities Citation2. We have shown in tumor mouse models that TKD/IL-2 activated NK cells have the capacity to kill Hsp70 membrane-positive tumors and to enhance overall survival of tumor-bearing animals. As mentioned earlier, unstimulated NK cells of patients are unable to kill Hsp70 membrane-positive tumor cells. These promising pre-clinical results encouraged us to initiate a clinical phase I trial in order to study feasibility, safety, and tolerability of ex vivo Hsp70-peptide TKD plus low dose IL-2 activated NK cells Citation32. Patients with multiple metastasized colorectal and non-small lung cell carcinoma (NSCLC) that were refractory to standard radiochemotherapy were enrolled into the trial. Up to six repeated reinfusions of complete autologous leukapheresis products that had been stimulated with TKD/IL-2 was found to be feasible, safe and well tolerated by all patients. Although the leukapheresis and reinfusions were performed every two weeks, the total number of lymphocytes and the composition of lymphocyte subpopulations in the leukapheresis product remained stable. A key finding of the trial was that the cytolytic of NK cells derived from pre-treated patients following radiochemotherapy could be stimulated by in vitro incubation with TKD/IL-2. Patients who received 4 reinfusions of TKD/IL-2 activated effector cells showed an enhanced in vivo cytolytic activity corresponding to an enhanced cell surface density of activating NK cell surface markers. Moreover, 2 of 5 patients receiving more than 4 treatment cycles showed clinical responses: a stable disease and a mixed response Citation32.

Based on these promising findings a 'proof of principle’ randomized phase II clinical study is underway in NSCLC patients. Lung cancer is the second most frequent tumor type in western societies and the leading cause of cancer death. Previously, we have shown that radiochemotherapy efficiently enhances Hsp70 membrane expression on tumor cells (unpublished observation). Therefore, we now want to adoptively transfer TKD/IL-2-activated, autologous NK cells into NSCLC patients stage IIIA and IIIB following successful radiochemotherapy. An Hsp70 membrane positive tumor phenotype is a key inclusion criteria for enrolling patients into the study. Recent data have shown that the DNA damage response induced by ionizing radiation enhances the expression of NKG2D ligands Citation33, Citation34 and thus this could be beneficial for clinical outcome. Another approach to increase radiosensitization is the use of Hsp90 inhibitors which are known to enhance the Hsp70 synthesis Citation34. A major goal of the study will be is to prolong survival of patients with a better quality of life. An additional goal in this study will be to measure the phenotype and the cytolytic capacity of immune effector cells in the patients before, during and after immunotherapy. The follow up of the patients will be 18 months from start of the NK cell infusions.

In summary, our paper republished here in the International Journal of Hyperthermia was the start of a new direction in our research which has taken us from basic laboratory observations to the implementation of new clinical trials. Overall, we expect that membrane-bound Hsp70 might provide a tumor-specific target structure for future cell-based and/or antibody-based therapeutic approaches.

Acknowledgements

The author would like to thank Armin Bareiss for technical assistance.

Grant support: Deutsche Forschungsgemeinschaft (DFG, MU 1238/7-2), European Union EU-STEMDIAGNOSTICS (LSHB CT 2007 037703), EU-CARDIORISK (FP7 2007–2001, #211403), BMBF–MOBITUM (Watching Tumor Biology by Molecular Imaging–MoBiMed), BMBF–Innovative Therapies (funding in progress) and multimmune GmbH.

Declaration of interest: The author reports no conflicts of interest. The author alone is responsible for the content and writing of the paper.

References

  • Farkas B, Hantschel M, Magyarlaki M, Becker B, Scherer K, Landthaler M, Pfister K, Gehrmann M, Gross C, Mackensen A, Multhoff G. Heat shock protein 70 membrane expression and melanoma-associated marker phenotype in primary and metastatic melanoma. Melanoma Res 2003; 13: 147–152
  • Pfister K, Radons J, Busch R, Tidball JG, Pfeifer M, Freitag L, Feldmann HJ, Milani V, Issels R, Multhoff G. Patient survival by Hsp70 membrane phenotype: Association with different routes of metastasis. Cancer 2007; 110: 926–935
  • Steiner K, Graf M, Hecht K, Reif S, Rossbacher L, Pfister K, Kolb HJ, Schmetzer HM, Multhoff G. High HSP70-membrane expression on leukemic cells from patients with acute myeloid leukemia is associated with a worse prognosis. Leukemia 2006; 20: 2076–2079
  • Gehrmann M, Schmetzer H, Eissner G, Haferlach T, Hiddemann W, Multhoff G. Membrane-bound heat shock protein 70 (Hsp70) in acute myeloid leukemia: A tumor specific recognition structure for the cytolytic activity of autologous NK cells. Haematologica 2003; 88: 474–476
  • Stangl S, Wortmann A, Guertler U, Multhoff G. Control of metastasized pancreatic carcinomas in SCID/beige mice with human IL-2/TKD-activated NK cells. J Immunol 2006; 176: 6270–6276
  • Multhoff G, Botzler C, Wiesnet M, Eissner G, Issels R. CD3- large granular lymphocytes recognize a heat-inducible immunogenic determinant associated with the 72-kD heat shock protein on human sarcoma cells. Blood 15-8-1995; 86: 1374–1382
  • Multhoff G, Botzler C, Jennen L, Schmidt J, Ellwart J, Issels R. Heat shock protein 72 on tumor cells: A recognition structure for natural killer cells. J Immunol 1997; 158: 4341–4350
  • Radons J, Multhoff G. Immunostimulatory functions of membrane-bound and exported heat shock protein 70. Exerc Immunol Rev 2005; 11: 17–33
  • Multhoff G, Mizzen L, Winchester CC, Milner CM, Wenk S, Eissner G, Kampinga HH, Laumbacher B, Johnson J. Heat shock protein 70 (Hsp70) stimulates proliferation and cytolytic activity of natural killer cells. Exp Hematol 1999; 27: 1627–1636
  • Botzler C, Kolb HJ, Issels RD, Multhoff G. Noncytotoxic alkyl-lysophospholipid treatment increases sensitivity of leukemic K562 cells to lysis by natural killer (NK) cells. Int J Cancer 1996; 65: 633–638
  • Multhoff G, Hightower LE. Cell surface expression of heat shock proteins and the immune response. Cell Stress Chaperones 1996; 1: 167–176
  • Gehrmann M, Liebisch G, Schmitz G, Anderson R, Steinem C, De MA, Pockley G, Multhoff G. Tumor-specific Hsp70 plasma membrane localization is enabled by the glycosphingolipid Gb3. PLoS ONE 2008; 3: e1925
  • Gastpar R, Gross C, Rossbacher L, Ellwart J, Riegger J, Multhoff G. The cell surface-localized heat shock protein 70 epitope TKD induces migration and cytolytic activity selectively in human NK cells. J Immunol 2004; 172: 972–980
  • Gross C, Koelch W, DeMaio A, Arispe N, Multhoff G. Cell surface-bound heat shock protein 70 (Hsp70) mediates perforin-independent apoptosis by specific binding and uptake of granzyme B. J Biol Chem 2003; 278: 41173–41181
  • Hightower LE, Guidon PT, Jr. Selective release from cultured mammalian cells of heat-shock (stress) proteins that resemble glia-axon transfer proteins. J Cell Physiol 1989; 138: 257–266
  • Arispe N, Doh M, Simakova O, Kurganov B, De Maio A. Hsc70 and Hsp70 interact with phosphatidylserine on the surface of PC12 cells resulting in a decrease of viability. FASEB J 2004; 18: 1636–1645
  • Arispe N, Doh M, De Maio A. Lipid interaction differentiates the constitutive and stress-induced heat shock proteins Hsc70 and Hsp70. Cell Stress Chaperones 2002; 7: 330–338
  • Vega VL, Rodriguez-Silva M, Frey T, Gehrmann M, Diaz JC, Steinem C, Multhoff G, Arispe N, De Maio A. Hsp70 translocates into the plasma membrane after stress and is released into the extracellular environment in a membrane-associated form that activates macrophages. J Immunol 2008; 180: 4299–4307
  • Falguieres T, Maak M, von WC, Sarr M, Sastre X, Poupon MF, Robine S, Johannes L, Janssen KP. Human colorectal tumors and metastases express Gb3 and can be targeted by an intestinal pathogen-based delivery tool. Mol Cancer Ther 2008; 7: 2498–2508
  • Yeh C-H, Tseng R, Zhang Z, Cortes J, O`Brien S, Giles F, Hannah G, Estrov Z, Keating M, Kantarjian H, Albitar M. Circulating Hsp70 and progression in patients with chronic myeloid leukemia. Leukemia Res 2009; 33: 212–217
  • Njemini R, Demanet C, Mets T. Comparison of two ELISAs for the determination of Hsp70 in serum. J Immunol Methods 2005; 306: 176–182
  • Hartl FU, Hayer-Hartl M. Molecular chaperones in the cytosol: From nascent chain to folded protein. Science 2002; 295: 1852–1858
  • Pockley AG, Shepherd J, Corton JM. Detection of heat shock protein 70 (Hsp70) and anti-Hsp70 antibodies in the serum of normal individuals. Immunol Invest 1998; 27: 367–377
  • Pockley AG, Georgiades A, Thulin T, deFaire U, Frostegard J. Serum heat shock protein 70 levels predict the development of atherosclerosis in subjects with established hypertension. Hypertension 2003; 42: 235–238
  • De Gassart A, Geminard C, Fevrier B, Raposo G, Vidal M. Lipid raft-associated protein sorting in exosomes. Blood 2003; 102: 4336–4344
  • Gross C, Hansch D, Gastpar R, Multhoff G. Interaction of heat shock protein 70 peptide with NK cells involves the NK receptor CD94. Biol Chem 2003; 384: 267–279
  • Ferlazzo G, Pack M, Thomas D, Paludan C, Schmid D, Strowig T, Bougras G, Muller WA, Moretta L, Munz C. Distinct roles of IL-12 and IL-15 in human natural killer cell activation by dendritic cells from secondary lymphoid organs. Proc Natl Acad Sci USA 2004; 101: 16606–16611
  • Elsner L, Muppala V, Gehrmann M, Lozano J, Malzahn D, Bickeboller H, Brunner E, Zientkowska M, Herrmann T, Walter L, Alves F, Multhoff G, Dressel R. The heat shock protein (Hsp70) promotes mouse NK cell activity against tumors that express inducible NKG2D ligands. J Immunol 2007; 179: 5523–5533
  • Gehrmann M, Radons J, Molls M, Multhoff G. The therapeutic implications of clinically applied modifiers of the Hsp70 expression by tumors. Cell Stress Chaperones 2008; 13: 1–10
  • Gasser S, Orsulic S, Brown EJ, Raulet DH. The DNA damage pathway regulates innate immune system ligands of the NKG2D receptor. Nature 2005; 436: 1186–1190
  • Dayanc BE, Beachy SH, Ostberg JR, Repasky EA. Dissecting the role of hyperthermia in natural killer cell mediated anti-tumor responses. Int J Hyperthermia 2008; 24: 41–56
  • Krause SW, Gastpar R, Andreesen R, Gross C, Ullrich H, Thonigs G, Pfister K, Multhoff G. 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: 3699–3707
  • Zitvogel L, Casares N, Pequignot MO, Chaput N, Albert ML, Kroemer G. Immune response against dying tumor cells. Adv Immunol 2004; 84: 131–179
  • Camphausen K, Tofilon PJ. Inhibition of Hsp90: A multi-target approach to radiosensitization. Clin Cancer Res 2007; 13: 4326–4330

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.