Elsevier

Pharmacology & Therapeutics

Volume 189, September 2018, Pages 31-44
Pharmacology & Therapeutics

Natural killer cells and their therapeutic role in pancreatic cancer: A systematic review

https://doi.org/10.1016/j.pharmthera.2018.04.003Get rights and content

Abstract

Pancreatic cancer is among the three deadliest cancers worldwide with the lowest 5-year survival of all cancers. Despite all efforts, therapeutic improvements have barely been made over the last decade. Even recent highly promising targeted and immunotherapeutic approaches did not live up to their expectations. Therefore, other horizons have to be explored. Natural Killer (NK) cells are gaining more and more interest as a highly attractive target for cancer immunotherapies, both as pharmaceutical target and for cell therapies. In this systematic review we summarise the pathophysiological adaptions of NK cells in pancreatic cancer and highlight possible (future) therapeutic NK cell-related targets. Furthermore, an extensive overview of recent therapeutic approaches with an effect on NK cells is given, including cytokine-based, viro- and bacteriotherapy and cell therapy. We also discuss ongoing clinical trials that might influence NK cells. In conclusion, although several issues regarding NK cells in pancreatic cancer remain unsolved and need further investigation, extensive evidence is already provided that support NK cell oriented approaches in pancreatic cancer.

Introduction

Pancreatic ductal adenocarcinoma (PDAC) is among the three most deadliest cancers in Western countries, recently overtaking breast cancer (Dreyer, Chang, Bailey, and Biankin, 2017). The 5-year survival of 7% has barely changed in 50 years and is stated as the worst of any cancer type (Dreyer et al., 2017; Evan et al., 2017). PDAC has proven to be an extremely difficult-to-treat cancer because of its rapidly progressive nature and high grade of resistance to all conventional, targeted and immunotherapies (Heinemann et al., 2014; Seicean, Petrusel, and Seicean, 2015). This is painfully evidenced by an unforgiving reality test when preclinical insights are tested in the clinic (Bates, 2017; Borazanci et al., 2017). Indeed, 47 clinical trials failed to show improvement over gemcitabine treatment, which is in sharp contrast with recent, encouraging discoveries in cancer immunotherapy in other tumour types (Bates, 2017). Even the highly promising immune checkpoint inhibitors programmed cell death-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) have had no significant effect in PDAC (Jimenez-Luna et al., 2016; Mei, Du, and Ma, 2016).

The tumour microenvironment (TME) is believed to be a major confounding factor involved in the failure of all these new approaches (Haqq et al., 2014; Lunardi, Muschel, and Brunner, 2014; Tjomsland et al., 2011; Watt and Kocher, 2013; Wilson, Pirola, and Apte, 2014). A hallmark of this TME in PDAC is the strong desmoplastic reaction which results in a dense fibrotic/desmoplastic stroma that surrounds the pancreatic cancer cells (Heinemann et al., 2014; Watt and Kocher, 2013; Wilson et al., 2014). By acting as a mechanical and functional shield around the tumour, it is responsible for diminished delivery of anticancer agents because it causes a high intratumoural pressure and low microvascular density. The main orchestrator of this fierce stromal barrier is the pancreatic stellate cell (PSC). Once activated, these cells enhance the development, progression and invasion of PDAC through their extensive crosstalk with the tumour, resulting in reciprocal stimulation and therapy resistance (Feig et al., 2012; Heinemann et al., 2014; Watt and Kocher, 2013; Wilson et al., 2014).

Immune cells also comprise part of the TME. In this review we focus on natural killer (NK) cells. Although one of their primary functions is to kill cancer cells, less attention has been paid to these cytotoxic immune cells compared to T cells. NK cells are a subset of innate lymphoid cells (ILCs) and comprise about 5–15% of the circulating cell population (Chiossone, Vienne, Kerdiles, and Vivier, 2017; Fang, Xiao, and Tian, 2017). They were originally identified as an immune cell population with profound tumour cell killing abilities in vitro (Kiessling, Klein, Pross, and Wigzell, 1975; Kiessling, Klein, and Wigzell, 1975). However, numerous studies have since demonstrated their anticancer effect in different animal models as well as their benefit in human studies (Fang, Xiao, and Tian, 2017; Lopez-Soto, Gonzalez, Smyth, and Galluzzi, 2017; Pahl and Cerwenka, 2017). By using a well-defined set of activating and inhibitory receptors, NK cells are able to recognize and kill tumour cells while sparing healthy cells, more specifically because they sense a certain lack of major histocompatibility complex (MHC)-I molecules via their killer-cell immunoglobulin-like receptors (KIRs) (Chiossone et al., 2017). Moreover, once activated, NK cells can secrete a vast number of cytokines and chemokines such as interferon (IFN)γ, tumour necrosis factor (TNF)α, granulocyte-macrophage colony-stimulating factor (GM-CSF), chemokine (C-C motif) ligand (CCL) 1–5 and chemokine (C-X-C motif) ligand (CXCL) 8, which trigger activation and recruitment of other innate and adaptive immune cells that broaden and strengthen the anti-tumour immune response (Paul and Lal, 2017). These unique features make NK cells ideal targets for cancer immunotherapy as evidenced by an increasing number of both preclinical and clinical studies that show promising results in different tumour types (Chiossone et al., 2017; Fang, Xiao, and Tian, 2017).

However, the role of NK cells in PDAC has not been well defined, given only a few articles have focused specifically on NK cells. Nonetheless, data on their function and importance in PDAC are available in some studies. This systematic review summarises the current evidence on NK cells in PDAC and highlights several possible approaches that could be pursued in future PDAC research. To our knowledge, this is the first systematic review which focusses on the NK cells in pancreatic cancer.

Section snippets

Methodology

We employed the Preferred Reporting Items for Systematic Reviews and Meta – Analyses (PRISMA) methodology to conduct this systematic review (Moher et al., 2009). We performed a search in the highly relevant MEDLINE database (1973 – present) using a list of four terms:

  • -

    “NK cells”, “pancreatic cancer” and the Boolean operator “AND”

  • -

    “NK cell”, “pancreatic cancer” and the Boolean operator “AND”

  • -

    “natural killer cells”, “pancreatic cancer” and the Boolean operator “AND”

  • -

    “natural killer cell”, “pancreatic

The biology and physiological changes of NK cells in PDAC

Tumours have an extensive crosstalk with their surrounding microenvironment, including the presence of NK cells and other immune cells (Varn, Wang, Mullins, Fiering, and Cheng, 2017; Vitale, Cantoni, Pietra, Mingari, and Moretta, 2014). Therefore, we first summarise the main facts of NK cells in PDAC and their adaptations caused by the tumour and its TME (Fig. 2).

The effect of the current standard of care in PDAC on NK cells

At present, the standard of care for pancreatic cancer remains surgical removal of the tumour with consecutive chemotherapy (10–20% of the patients) or chemotherapy alone when the patient has locally advanced or metastatic disease (Hidalgo et al., 2015). Current chemotherapy approaches comprise 2 options: the first one is FOLFIRINOX, a combination regimen of 4 different chemotherapeutic agents (5-FU, oxaliplatin, irinotecan and leucoverin), used in adjuvant setting for patients with metastatic

Immunomodulating therapies in PDAC with an effect on NK cells

Several agents have been tested in the past for their effect in PDAC resulting in enhancing NK cell function.. This includes the use of cytokines and chemokines.

Cell therapies in PDAC

In this last part, we discuss cell therapies involving NK cells in pancreatic cancer. Several studies have explored adoptive NK cell therapy in PDAC (Table 11). In one study, NK cells were ex vivo expanded with antibodies directed against CD3 and CD52 that resulted in higher in vitro cytotoxicity and tumour suppression and resulted in better survival in patients up to 13 months following their adoptive transfer. In these patients metastatic lesions also shrunk as well as tumour markers (

Conclusion

This systematic review provides substantial evidence for the important role NK cells play in PDAC and their potential therapeutic impact. PDAC substantially impairs NK cell functions by downregulation of effector molecules, reduced cytokine secretion capacity and decreased expression of virtually every activation receptor, including the NCRs, nectin- and nectin-like binding molecules and above all NKG2D. Also the TME plays a major role in the reduced cytotoxicity of NK cells. It is therefore of

Conflict of interest

The authors declare to have no conflict of interest. Jonas RM Van Audenaerde is a research fellow of the Research Foundation Flanders (fellowship number 1S32316N). He received a travel grant from the Research Foundation Flanders (V4.090.17N) to visit the PeterMacCallum Cancer Centre. We also want to express our special gratitude to Mr. Willy Floren and the Vereycken family for their kind gifts which enabled us to perform this work.

References (126)

  • S. Lunardi et al.

    The stromal compartments in pancreatic cancer: Are there any therapeutic targets?

    Cancer Letters

    (2014)
  • J. Masuyama et al.

    Ex vivo expansion of natural killer cells from human peripheral blood mononuclear cells co-stimulated with anti-CD3 and anti-CD52 monoclonal antibodies

    Cytotherapy

    (2016)
  • A.M. Noonan et al.

    Randomized phase 2 trial of the oncolytic virus pelareorep (Reolysin) in upfront treatment of metastatic pancreatic adenocarcinoma

    Molecular Therapy: The Journal of the American Society of Gene Therapy

    (2016)
  • J. Pahl et al.

    Tricking the balance: NK cells in anti-cancer immunity

    Immunobiology

    (2017)
  • M.T. Saung et al.

    Current standards of chemotherapy for pancreatic cancer

    Clinical Therapeutics

    (2017)
  • M. Abdel-Wahab et al.

    Evaluation of cell mediated immunity in advanced pancreatic carcinoma before and after treatment with interleukin-2 (IL-2)

    Hepato-Gastroenterology

    (1999)
  • E. Ames et al.

    NK cells preferentially target tumor cells with a cancer stem cell phenotype

    Journal of Immunology (Baltimore, Md.: 1950)

    (2015)
  • M.J. Baine et al.

    Transcriptional profiling of peripheral blood mononuclear cells in pancreatic cancer patients identifies novel genes with potential diagnostic utility

    PLoS One

    (2011)
  • C.P. Bailey et al.

    New interleukin-15 superagonist (IL-15SA) significantly enhances graft-versus-tumor activity

    Oncotarget

    (2017)
  • S. Bang et al.

    Differences in immune cells engaged in cell-mediated immunity after chemotherapy for far advanced pancreatic cancer

    Pancreas

    (2006)
  • S.E. Bates

    Pancreatic cancer: Challenge and inspiration

    Clinical Cancer Research: An Official Journal of the American Association for Cancer Research

    (2017)
  • A.V. Bazhin et al.

    The novel mitochondria-targeted antioxidant SkQ1 modulates angiogenesis and inflammatory micromilieu in a murine orthotopic model of pancreatic cancer

    International Journal of Cancer

    (2016)
  • R. Bhat et al.

    Enhancement of NK cell antitumor responses using an oncolytic parvovirus

    International Journal of Cancer

    (2011)
  • S.J. Blake et al.

    Suppression of metastases using a new lymphocyte checkpoint target for cancer immunotherapy

    Cancer Discovery

    (2016)
  • E. Borazanci et al.

    Pancreatic cancer: "A riddle wrapped in a mystery inside an enigma"

    Clinical Cancer Research: An Official Journal of the American Association for Cancer Research

    (2017)
  • V.T. Bui et al.

    Augmented IFN-gamma and TNF-alpha induced by probiotic bacteria in NK cells mediate differentiation of stem-like tumors leading to inhibition of tumor growth and reduction in inflammatory cytokine release; regulation by IL-10

    Frontiers in Immunology

    (2015)
  • N. Carrere et al.

    Characterization of the bystander effect of somatostatin receptor sst2 after in vivo gene transfer into human pancreatic cancer cells

    Human Gene Therapy

    (2005)
  • J.P. Cata et al.

    Lidocaine stimulates the function of natural killer cells in different experimental settings

    Anticancer Research

    (2017)
  • C.J. Chan et al.

    The receptors CD96 and CD226 oppose each other in the regulation of natural killer cell functions

    Nature Immunology

    (2014)
  • L.S. Chard et al.

    A vaccinia virus armed with interleukin-10 is a promising therapeutic agent for treatment of murine pancreatic cancer

    Clinical Cancer Research: An Official Journal of the American Association for Cancer Research

    (2015)
  • G. Chitadze et al.

    Shedding of endogenous MHC class I-related chain molecules A and B from different human tumor entities: heterogeneous involvement of the “a disintegrin and metalloproteases” 10 and 17

    International Journal of Cancer

    (2013)
  • H.W. Chung et al.

    Clinical implications and diagnostic usefulness of correlation between soluble major histocompatibility complex class I chain-related molecule a and protumorigenic cytokines in pancreatic ductal adenocarcinoma

    Cancer

    (2013)
  • R. Cubas et al.

    Chimeric Trop2 virus-like particles: A potential immunotherapeutic approach against pancreatic cancer

    Journal of Immunotherapy (Hagerstown, Md: 1997)

    (2011)
  • L.M. Curd et al.

    Pro-tumour activity of interleukin-22 in HPAFII human pancreatic cancer cells

    Clinical and Experimental Immunology

    (2012)
  • T. Daikeler et al.

    The influence of gemcitabine on the CD4/CD8 ratio in patients with solid tumours

    Oncology Reports

    (1997)
  • M. D'Alincourt Salazar et al.

    Evaluation of innate and adaptive immunity contributing to the antitumor effects of PD1 blockade in an orthotopic murine model of pancreatic cancer

    Oncoimmunology

    (2016)
  • Z. Dambrauskas et al.

    Expression of major histocompatibility complex class I-related chain A/B (MICA/B) in pancreatic carcinoma

    International Journal of Oncology

    (2014)
  • G. Damia et al.

    Flavone acetic acid antitumour activity against a mouse pancreatic adenocarcinoma is mediated by natural killer cells

    Cancer Immunology, Immunotherapy: CII

    (1990)
  • M. Davis et al.

    Effect of pemetrexed on innate immune killer cells and adaptive immune T cells in subjects with adenocarcinoma of the pancreas

    Journal of Immunotherapy (Hagerstown, Md: 1997)

    (2012)
  • L. Degrate et al.

    Interleukin-2 immunotherapy action on innate immunity cells in peripheral blood and tumoral tissue of pancreatic adenocarcinoma patients

    Langenbeck's Archives of Surgery

    (2009)
  • S. Dempe et al.

    Antitumoral activity of parvovirus-mediated IL-2 and MCP-3/CCL7 delivery into human pancreatic cancer: Implication of leucocyte recruitment

    Cancer Immunology, Immunotherapy: CII

    (2012)
  • S.B. Dreyer et al.

    Pancreatic cancer genomes: Implications for clinical management and therapeutic development

    Clinical Cancer Research: An Official Journal of the American Association for Cancer Research

    (2017)
  • X. Duan et al.

    Clinical significance of the immunostimulatory MHC class I chain-related molecule A and NKG2D receptor on NK cells in pancreatic cancer

    Medical Oncology (Northwood, London, England)

    (2011)
  • G.I. Evan et al.

    Re-engineering the pancreas tumor microenvironment: A “regenerative program” hacked

    Clinical Cancer Research: An Official Journal of the American Association for Cancer Research

    (2017)
  • S. Eisold et al.

    Induction of an antitumoral immune response by wild-type adeno-associated virus type 2 in an in vivo model of pancreatic carcinoma

    Pancreas

    (2007)
  • J. Ellermeier et al.

    Therapeutic efficacy of bifunctional siRNA combining TGF-beta1 silencing with RIG-I activation in pancreatic cancer

    Cancer Research

    (2013)
  • E. Eriksson et al.

    Shaping the tumor stroma and sparking immune activation by CD40 and 4-1BB signaling induced by an armed oncolytic virus

    Clinical Cancer Research: An Official Journal of the American Association for Cancer Research

    (2017)
  • R. Fahrig et al.

    RP101 improves the efficacy of chemotherapy in pancreas carcinoma cell lines and pancreatic cancer patients

    Anti-Cancer Drugs

    (2006)
  • C. Feig et al.

    The pancreas cancer microenvironment

    Clinical Cancer Research: An Official Journal of the American Association for Cancer Research

    (2012)
  • T.L. Frankel et al.

    Identification and characterization of a tumor infiltrating CD56(+)/CD16 (-) NK cell subset with specificity for pancreatic and prostate cancer cell lines

    Cancer Immunology, Immunotherapy: CII

    (2010)
  • Cited by (34)

    • PKCι regulates the expression of PDL1 through multiple pathways to modulate immune suppression of pancreatic cancer cells

      2021, Cellular Signalling
      Citation Excerpt :

      Compromised NK cell cytotoxicity has been indicated to be correlated with increased cancer risk and poor prognosis in various malignancies [20]. The number or the cytotoxic capacity of circulating NK cells is reduced and correlated with worse overall survival or advanced stage in PDAC patients, suggesting an important role that NK cells play in the PDAC development [21]. Since the expression of programmed cell death protein 1 (PD1), the receptor for PDL1, is shown to be upregulated and responsible for impaired degranulation and cytotoxic activity of NK in the tumor microenvironment, checkpoint blockade immunotherapy targeting the PD1/PDL1 axis may re-invigorate the anti-tumor response mediated by NK cells [22].

    • Mechanistic target of rapamycin in the tumor microenvironment and its potential as a therapeutic target for pancreatic cancer

      2020, Cancer Letters
      Citation Excerpt :

      However, Wang et al. examined mice with conditional NK cell-specific Raptor or Rictor deletion and concluded that mTORC1 maintains mTORC2 activity via CD122-mediated IL-15 signaling and that mTORC2 represses mTORC1 and controls NK cell functions by restraining the STAT5-SLC7A5 axis [114]. NK cells are downregulated in both abundance and function in PC, resulting in reduced cytotoxic capacity and reduced cell numbers [115]. mTOR signaling may be a promising approach to inhibit PC growth by promoting NK cell activation and development.

    View all citing articles on Scopus
    View full text