671
Views
0
CrossRef citations to date
0
Altmetric
Editorial

Use of the TLR2 ligand polysaccharide krestin to maximize conventional cancer therapy

ORCID Icon, &
Pages 133-135 | Received 18 Nov 2016, Accepted 25 Apr 2017, Published online: 10 May 2017

The HER2+ breast cancer subtype accounts for 20–30% of cases and represents a particularly aggressive form of breast cancer. As this type of breast cancer is dependent on the HER2 receptor for proliferation, the introduction of the anti-HER2 monoclonal antibody (mAb) trastuzumab revolutionized the management of HER2+ disease and remains the foundation of anti-HER2 treatment [Citation1]. Despite this improvement in treatment, patients with advanced HER2+ disease will progress on trastuzumab. The primary antitumor activity of trastuzumab is mediated by the immune process of antibody (Ab)-dependent cellular cytotoxicity (ADCC) [Citation2]. Functionally, ADCC occurs when the Fc portion of an Ab binds to the Fcg receptor of a natural killer (NK) cell, inducing NK cell cytokine release (interferon gamma (IFN-γ) and cytolysis of the Ab-bound cell. In HER2+ breast cancer, studies have shown that the functional activity of NK cells impacts the antitumor effects of trastuzumab [Citation3]. In addition, treatment with trastuzumab affects the localization of NK cells as was shown in patients undergoing neoadjuvant chemo and trastuzumab therapy, where trastuzumab treatment was associated with a significant increase in the numbers of tumor-associated NK cells [Citation4]. Despite these observations, NK cell function is inhibited in cancer patients by multiple mechanisms, including the secretion of the immunosuppressive transforming growth factor-beta cytokine release by tumors [Citation5] and the downregulation of the activating NK cell CD16 receptor [Citation6] (a critical receptor for ADCC).

The presence of HER2-specific Type I or Th1 immunity is critical to antitumor efficacy in HER2+ disease as it represents an adaptive immune response that mediates a direct cytotoxic effect on tumor cells [Citation7]. A recent study has documented that there is a progressive loss of a HER2-specific Th1 immune response through growth of HER2+ breast cancer [Citation8]. It has been demonstrated that trastuzumab induces HER2-specific Th1 immunity in a minority of patients (30%), and that levels of the induced HER2-specific T cell immunity are variable [Citation9]. We have shown that HER2 vaccination can induce additional immune response above what is generated with trastuzumab such that 70% of patients develop HER2-specific immunity following trastuzumab and vaccination [Citation9]. Clinically, this is an important observation as measurable HER2-specific immunity has been linked to improved survival in HER2+ breast cancer [Citation10]. This finding has been shown in several studies, including a recent investigation that revealed in patients who received chemo + trastuzumab, Th1-nonresponsive patients had a worse disease-free survival (median, 47 vs. 113 months; P < .001) compared with Th1-responsive patients [Citation10].

Our investigation into interventions to augment trastuzumab-mediated ADCC and HER2-specific Th1 immunity led us to polysaccharide krestin (PSK), an extract from the mycelium of the mushroom Trametes versicolor. First approved in the 1970s, PSK has been used for decades in Japan as an anticancer therapy. Its clinical cancer use was supported by human trials that suggested improved survival when PSK was administered in gastric [Citation11], colorectal [Citation12], and lung cancers [Citation13]. Despite this survival benefit, the exact mechanism of PSK’s antitumor activity has been unclear. Prior publications have suggested that PSK’s activity may be immune mediated, with studies reporting that PSK induces the gene expression of IL-8 in peripheral blood mononuclear cells (PBMCs) after oral administration [Citation14], stimulates T-cell proliferation [Citation15], and improves the function of CD4+ T cells in gut-associated lymphoid tissue [Citation16].

Using knockout mouse models, our investigations of PSK have established that PSK selectively binds to the toll-like receptor 2 (TLR2) [Citation17]. Toll-like receptors are elements of the innate immune system tasked with the detection of foreign microbes and viruses and the activation of an immune response against these foreign bodies. When bound by ligands and activated, TLR2 induces the activation of multiple immune cell subtypes. Specifically, TLR2 is primarily found on dendritic cells (DCs), and to a lesser extent on T cells and NK cells. In our preclinical models, PSK induces the maturation and activation of DCs, resulting in an increased percentage of CD86+ MHCHhigh DCs. These PSK-activated DCs in turn release the Th1-linked cytokine, IL-12. Secretion of IL-12 induces the antitumor activities of multiple other immune cells including CD8+ T cells and NK cells. PSK administration also induced the secretion of other Th1-related cytokines including IFN-γ, TNF-alpha, and IL-2. We have demonstrated that PSK administration inhibits the growth of implanted or spontaneous breast tumors in neu-transgenic mice. To investigate which immune cells were responsible for the antitumor activity of PSK, we performed selective immune cell depletion in our mouse models. These depletion studies revealed that the antitumor activity of PSK is primarily due to CD8+ T cells and NK cells.

As we had established that PSK could induce NK cell maturation, we next investigated if PSK could also augment the functional activity of NK cells and enhance the ADCC of tumor antigen-specific mAbs. In these evaluations, we revealed that the addition of PSK to PBMCs augments the activation of NK cells, as determined by increased CD25 and CD69 expression, and induces the NK cell secretion of IFN-γ [Citation18]. We confirmed that these effects on NK cells are mediated through both direct TLR2 binding of PSK and indirectly through the induction of DC secretion of IL-12, as noted earlier. We also showed that when added to PBMCs, PSK augments the cytolytic function of NK cells against cancer cell lines. In order to evaluate if PSK could augment trastuzumab-mediated ADCC, we exposed HER2+ breast cancer cell lines (SKBR3 and MDA-MB-231) to PSK-treated PBMCs, and verified that indeed PSK significantly enhanced trastuzumab-mediated ADCC against these cells. Lastly, in neu-transgenic mice bearing neu+ tumors, the combination of PSK and an anti-HER2 mAb exhibited enhanced antitumor effects compared to either therapy alone.

Based on these preclinical investigations, we have initiated a Phase II randomized clinical trial to investigate the immune and therapeutic effects of the addition of PSK to anti-HER2 therapy in advanced HER2+ breast cancer. Patients on this randomized study receive trastuzumab and a HER2-directed vaccine with or without PSK. To date, we have enrolled 24 patients of a planned 30 patients. In the treated patients, there have been no grade 3 or higher adverse events on the study. This trial will complete enrollment and treatment in 2017. The immune analysis from this study will investigate changes in NK cell functional activity and HER2-specific immunity from baseline and between patients who received PSK vs. those who did not. The ability of PSK to augment NK cell functional activity will be measured by NK cell secretion of IFN-γ and a CD107a NK cell degranulation assay (to measure NK cell cytotoxicity). We will also investigate changes in HER2-specific Th1 immunity with HER2 IFN-γ ELISPOT assays. The clinical and correlative science results from this trial will guide future development and application of PSK and similar therapies. Our long-term clinical plan is to integrate PSK into neoadjuvant anti-HER2 therapy to significantly improve upon the current pathologic complete response rate of 40% [Citation19].

Declaration of interest

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

Additional information

Funding

Funding received from the National Institutes of Health, National Cancer Institute 1U19AT006028-04.

References

  • Dawood S, Broglio K, Buzdar AU, et al. Prognosis of women with metastatic breast cancer by HER2 status and trastuzumab treatment: an institutional-based review. J Clin Oncol. 2010;28:92–98.
  • Spiridon CI, Guinn S, Vitetta ES. A comparison of the in vitro and in vivo activities of IgG and F(ab’)2 fragments of a mixture of three monoclonal anti-Her-2 antibodies. Clin Cancer Res. 2004;10:3542–3551.
  • Ferris RL, Jaffee EM, Ferrone S. Tumor antigen-targeted, monoclonal antibody-based immunotherapy: clinical response, cellular immunity, and immunoescape. J Clin Oncol. 2010;28:4390–4399.
  • Arnould L, Gelly M, Penault-Llorca F, et al. Trastuzumab-based treatment of HER2-positive breast cancer: an antibody-dependent cellular cytotoxicity mechanism? Br J Cancer. 2006;94:259–267.
  • Lee JC, Lee KM, Kim DW, et al. Elevated TGF-beta1 secretion and down-modulation of NKG2D underlies impaired NK cytotoxicity in cancer patients. J Immunol. 2004;172:7335–7340.
  • Kono K, Takahashi A, Ichihara F, et al. Impaired antibody-dependent cellular cytotoxicity mediated by herceptin in patients with gastric cancer. Cancer Res. 2002;62:5813–5817.
  • Sica A, Larghi P, Mancino A, et al. Macrophage polarization in tumour progression. Semin Cancer Biol. 2008;18:349–355.
  • Datta J, Rosemblit C, Berk E. Progressive loss of anti-HER2 CD4+ T-helper type 1 response in breast tumorigenesis and the potential for immune restoration. Oncoimmunology. 2015;4:e1022301.
  • Disis ML, Wallace DR, Gooley TA, et al. Concurrent trastuzumab and HER2/neu-specific vaccination in patients with metastatic breast cancer. J Clin Oncol. 2009;27:4685–4692.
  • Datta J, Fracol M, McMillan MT, et al. Association of depressed anti-HER2 T-helper type 1 response with recurrence in patients with completely treated HER2-positive breast cancer: role for immune monitoring. JAMA Oncol. 2016;2:242–246.
  • Nakazato H, Koike A, Saji S, et al. Efficacy of immunochemotherapy as adjuvant treatment after curative resection of gastric cancer. Study Group of Immunochemotherapy with PSK for Gastric Cancer. Lancet. 1994;343:1122–1126.
  • Torisu M, Hayashi Y, Ishimitsu T, et al. Significant prolongation of disease-free period gained by oral polysaccharide K (PSK) administration after curative surgical operation of colorectal cancer. Cancer Immunol Immunother. 1990;31:261–268.
  • Hayakawa K, Mitsuhashi N, Saito Y, et al. Effect of krestin (PSK) as adjuvant treatment on the prognosis after radical radiotherapy in patients with non-small cell lung cancer. Anticancer Res. 1993;13:1815–1820.
  • Kato M, Hirose K, Hakozaki M, et al. Induction of gene expression for immunomodulating cytokines in peripheral blood mononuclear cells in response to orally administered PSK, an immunomodulating protein-bound polysaccharide. Cancer Immunol Immunother. 1995;40:152–156.
  • Hirai R, Oguchi Y, Sugita N, et al. Enhancement of T-cell proliferation by PSK. Int J Immunopharmacol. 1993;15:745–750.
  • Harada M, Matsunaga K, Oguchi Y, et al. Oral administration of PSK can improve the impaired anti-tumor CD4+ T-cell response in gut-associated lymphoid tissue (GALT) of specific-pathogen-free mice. Int J Cancer. 1997;70:362–372.
  • Lu H, Yang Y, Gad E, et al. Polysaccharide krestin is a novel TLR2 agonist that mediates inhibition of tumor growth via stimulation of CD8 T cells and NK cells. Clin Cancer Res. 2011;17:67–76.
  • Lu H, Yang Y, Gad E, et al. TLR2 agonist PSK activates human NK cells and enhances the antitumor effect of HER2-targeted monoclonal antibody therapy. Clin Cancer Res. 2011;17:6742–6753.
  • Gianni L, Pienkowski T, Im Y-H, et al. 5-year analysis of neoadjuvant pertuzumab and trastuzumab in patients with locally advanced, inflammatory, or early-stage HER2-positive breast cancer (NeoSphere): a multicentre, open-label, phase 2 randomised trial. Lancet Oncol. 2016;17:791–800.

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.