Endogenous Heat-Shock Protein Induction with or Without Radiofrequency Ablation or Cryoablation in Patients with Stage IV Melanoma

Evidio Domingo-Musibay, James M Heun, Wendy K Nevala, Matthew Callstrom, Thomas Atwell, Evanthia Galanis, Lori A Erickson, Svetomir N Markovic, Evidio Domingo-Musibay, James M Heun, Wendy K Nevala, Matthew Callstrom, Thomas Atwell, Evanthia Galanis, Lori A Erickson, Svetomir N Markovic

Abstract

Lessons learned: Percutaneous thermal ablation combined with in situ granulocyte-macrophage colony-stimulating factor cytokine therapy was technically feasible and well tolerated.No significant clinical or immunologic responses were seen.

Background: Melanoma tumor-derived heat-shock proteins (HSPs) and HSP-peptide complexes can elicit protective antitumor responses. The granulocyte-macrophage colony-stimulating factor (GM-CSF) chemokine can also promote uptake and processing by professional antigen presenting cells (APCs). On this basis, we designed a pilot study of percutaneous thermal ablation as a means to induce heat-shock protein vaccination plus GM-CSF to determine safety and preliminary antitumor activity of this combination.

Materials and methods: This study was designed to assess overall safety of percutaneous ablation combined with GM-CSF for unresectable, metastatic melanoma including uveal and mucosal types. All patients received heat-shock therapy (42°C for 30 minutes), then received one of three treatments: (a) intralesional GM-CSF (500 mcg standard dose); (b) radiofrequency ablation (RFA) + GM-CSF; or (c) cryoablation plus GM-CSF. The primary endpoint of the study was the induction of endogenous HSP70 and melanoma-specific cytotoxic T lymphocytes (CTL).

Results: Nine patients (three per study arm) were enrolled. No dose-limiting toxicity was observed as specified per protocol. All patients developed progressive disease and went on to receive alternative therapy. Median overall survival (OS) was 8.2 months (95% confidence interval [CI] 2-17.2). The study was not powered to detect a difference in clinical outcome among treatment groups.

Conclusion: Percutaneous thermal ablation plus GM-CSF was well tolerated, technically feasible, and demonstrated an acceptable adverse event profile comparable to conventional RFA and cryoablation. While HSP70 was induced following therapy, the degree of HSP70 elevation was not associated with clinical outcome or induced CTL responses. While percutaneous thermal ablation plus GM-CSF combinations including checkpoint inhibitors could be considered in future studies, the use of GM-CSF remains experimental and for use in the context of clinical trials.

Trial registration: ClinicalTrials.gov NCT00568763.

© AlphaMedPress; the data published online to support this summary is the property of the authors.

Figures

Figure 1.
Figure 1.
Plasma HSP70 levels. Baseline circulating HSP70 was detectable in all patients. (A): Individual patient levels of plasma HSP70 over 24 hours shown for heat shock therapy (HST), RFA, and cryoablation cohorts. (B): HSP70 fold‐change above baseline level measurements for patients receiving cryoablation (top, arm C), RFA (middle, arm B), and HST (lower, arm A), with sustained elevations of HSP70 seen in patients receiving cryoablation therapy. Abbreviations: HSP70, 70 kilodalton heat‐shock protein; RFA, radiofrequency ablation.
Figure 2.
Figure 2.
Major histocompatibility complex‐tetramer testing identified cluster of differentiation 8 positive (CD8+) cytotoxic T lymphocytes (CTLs) for each tumor antigen. Pre‐specified criteria for positive tetramer results was >0.02% tetramer positive CD8+ CTLs if baseline negative, or >2‐fold increase in the baseline detectable number. We detected low baseline levels of peripheral CD8+ CTLs against known melanoma antigens MART‐1, GP100, and Tyrosinase (7 of 8 evaluable patients). There was no significant induction of CD8+ CTL responses to melanoma antigens following treatment. Abbreviations: GP100, glycoprotein 100; MART‐1, melanoma antigen recognized by T cells 1.

References

    1. Barth A, Wanek LA, Morton DL. Prognostic factors in 1,521 melanoma patients with distant metastases. J Am Coll Surg 1995;181:193–201.
    1. Zou W, Wolchok JD, Chen L. PD‐L1 (B7‐H1) and PD‐1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations. Sci Transl Med 2016;8:328rv4.
    1. Lau PK, Ascierto PA, McArthur G. Melanoma: The intersection of molecular targeted therapy and immune checkpoint inhibition. Curr Opin Immunol 2016;39:30–38.
    1. Morris ZS, Guy EI, Francis DM et al. In situ tumor vaccination by combining local radiation and tumor‐specific antibody or immunocytokine treatments. Cancer Res 2016;76:3929–3941.
    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin 2015;65:5–29.
    1. Ribas A, Hamid O, Daud A et al. Association of pembrolizumab with tumor response and survival among patients with advanced melanoma. JAMA 2016;315:1600–1609.
    1. Robert C, Long GV, Brady B et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med 2015;372:320–330.
    1. Algazi AP, Tsai KK, Shoushtari AN et al. Clinical outcomes in metastatic uveal melanoma treated with PD‐1 and PD‐L1 antibodies. Cancer 2016;122:3344–3353.
    1. Doussot A, Nardin C, Takaki H et al. Liver resection and ablation for metastatic melanoma: A single center experience. J Surg Oncol 2015;111:962–968.
    1. Bendz H, Ruhland SC, Pandya MJ et al. Human heat shock protein 70 enhances tumor antigen presentation through complex formation and intracellular antigen delivery without innate immune signaling. J Biol Chem 2007;282:31688–31702.
    1. Castelli C, Ciupitu AM, Rini F et al. Human heat shock protein 70 peptide complexes specifically activate antimelanoma T cells. Cancer Res 2001;61:222–227.
    1. Andtbacka RH, Ross M, Puzanov I et al. Patterns of clinical response with talimogene laherparepvec (T‐VEC) in patients with melanoma treated in the OPTiM phase III clinical trial. Ann Surg Oncol 2016;23:4169–4177.
    1. Harper JW, Bennett EJ. Proteome complexity and the forces that drive proteome imbalance. Nature 2016;537:328–338.
    1. Kampinga HH, Hageman J, Vos MJ et al. Guidelines for the nomenclature of the human heat shock proteins. Cell Stress Chaperones 2009;14:105–111.
    1. Murphy ME. The HSP70 family and cancer. Carcinogenesis 2013;34:1181–1188.
    1. Budina‐Kolomets A, Webster MR, Leu JI et al. HSP70 inhibition limits FAK‐dependent invasion and enhances the response to melanoma treatment with BRAF inhibitors. Cancer Res 2016;76:2720–2730.
    1. Delneste Y, Magistrelli G, Gauchat J et al. Involvement of LOX‐1 in dendritic cell‐mediated antigen cross‐presentation. Immunity 2002;17:353–362.
    1. Theriault JR, Mambula SS, Sawamura T et al. Extracellular HSP70 binding to surface receptors present on antigen presenting cells and endothelial/epithelial cells. FEBS Lett 2005;579:1951–1960.
    1. Basu S, Binder RJ, Ramalingam T et al. CD91 is a common receptor for heat shock proteins gp96, hsp90, hsp70, and calreticulin. Immunity 2001;14:303–313.
    1. Theriault JR, Adachi H, Calderwood SK. Role of scavenger receptors in the binding and internalization of heat shock protein 70. J Immunol 2006;177:8604–8611.
    1. Shevtsov M, Multhoff G. Heat shock protein‐peptide and HSP‐based immunotherapies for the treatment of cancer. Front Immunol 2016;7:171.
    1. Figueiredo C, Wittmann M, Wang D, Dressel R, Seltsam A, Blasczyk R, et al. Heat shock protein 70 (HSP70) induces cytotoxicity of T-helper cells. Blood. 2009;113:3008–3016.
    1. Sluijter BJ, van den Hout MF, Koster BD et al. Arming the melanoma sentinel lymph node through local administration of CpG‐B and GM‐CSF: Recruitment and activation of BDCA3/CD141(+) dendritic cells and enhanced cross‐presentation. Cancer Immunol Res 2015;3:495–505.
    1. Geng H, Zhang GM, Xiao H et al. HSP70 vaccine in combination with gene therapy with plasmid DNA encoding sPD‐1 overcomes immune resistance and suppresses the progression of pulmonary metastatic melanoma. Int J Cancer 2006;118:2657–2664.
    1. 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;10:1553–1568.

Source: PubMed

3
Abonnere