Skip to main content

Advertisement

Log in

Beta-glucan-induced inflammatory monocytes mediate antitumor efficacy in the murine lung

  • Original Article
  • Published:
Cancer Immunology, Immunotherapy Aims and scope Submit manuscript

Abstract

β-Glucan is a naturally occurring glucose polysaccharide with immunostimulatory activity in both infection and malignancy. β-Glucan’s antitumor effects have been attributed to the enhancement of complement receptor 3-dependent cellular cytotoxicity, as well as modulation of suppressive and stimulatory myeloid subsets, which in turn enhances antitumor T cell immunity. In the present study, we demonstrate antitumor efficacy of yeast-derived β-glucan particles (YGP) in a model of metastatic-like melanoma in the lung, through a mechanism that is independent of previously reported β-glucan-mediated antitumor pathways. Notably, efficacy is independent of adaptive immunity, but requires inflammatory monocytes. YGP-activated monocytes mediated direct cytotoxicity against tumor cells in vitro, and systemic YGP treatment upregulated inflammatory mediators, including TNFα, M-CSF, and CCL2, in the lungs. Collectively, these studies identify a novel role for inflammatory monocytes in β-glucan-mediated antitumor efficacy, and expand the understanding of how this immunomodulator can be used to generate beneficial immune responses against metastatic disease.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

Abbreviations

β-Glucan:

β-1,3 Backbone of d-glucose with β-1,6 branches of variable length and complexity

CR3:

Complement receptor 3

DT:

Diphtheria toxin

iC3b:

Inactivated complement component 3b

Mφ:

Macrophages

Mo:

Monocytes

mHIF1α−/− :

Myeloid-specific knockout of HIF1α

TII:

Trained innate immunity

YGP:

Yeast-derived β-glucan particles

References

  1. Aleem E (2013) β-Glucans and their applications in cancer therapy: focus on human studies. Anticancer Agents Med Chem 13:709–719

    Article  CAS  PubMed  Google Scholar 

  2. Vannucci L, Krizan J, Sima P et al (2013) Immunostimulatory properties and antitumor activities of glucans. Int J Oncol 43:357–364. https://doi.org/10.3892/ijo.2013.1974 (review)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Rahar S, Swami G, Nagpal N et al (2011) Preparation, characterization, and biological properties of β-glucans. J Adv Pharm Technol Res 2:94–103. https://doi.org/10.4103/2231-4040.82953

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Chan G, Chan W, Sze D (2009) The effects of β-glucan on human immune and cancer cells. J Hematol Oncol 2:25. https://doi.org/10.1186/1756-8722-2-25

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Hong F, Yan J, Baran JT et al (2004) Mechanism by which orally administered beta-1,3-glucans enhance the tumoricidal activity of antitumor monoclonal antibodies in murine tumor models. J Immunol 173:797–806. https://doi.org/10.4049/jimmunol.173.2.797

    Article  PubMed  CAS  Google Scholar 

  6. Vetvicka V, Thornton BP, Wieman TJ, Ross GD (1997) Targeting of natural killer cells to mammary carcinoma via naturally occurring tumor cell-bound iC3b and beta-glucan-primed CR3 (CD11b/CD18). J Immunol 159:599–605

    PubMed  CAS  Google Scholar 

  7. Cheung N-KV, Modak S, Vickers A, Knuckles B (2002) Orally administered beta-glucans enhance anti-tumor effects of monoclonal antibodies. Cancer Immunol Immunother 51:557–564. https://doi.org/10.1007/s00262-002-0321-3

    Article  PubMed  CAS  Google Scholar 

  8. Yan J, Vetvicka V, Xia Y et al (1999) Beta-glucan, a “specific” biologic response modifier that uses antibodies to target tumors for cytotoxic recognition by leukocyte complement receptor type 3 (CD11b/CD18). J Immunol 163:3045–3052

    PubMed  CAS  Google Scholar 

  9. Li B, Cai Y, Qi C et al (2010) Orally administered particulate beta-glucan modulates tumor-capturing dendritic cells and improves antitumor T-cell responses in cancer. Clin Cancer Res 16:5153–5164. https://doi.org/10.1158/1078-0432.CCR-10-0820

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Liu M, Luo F, Ding C et al (2015) Dectin-1 activation by a natural product β-glucan converts immunosuppressive macrophages into an M1-like phenotype. J Immunol 195:5055–5065. https://doi.org/10.4049/jimmunol.1501158

    Article  PubMed  CAS  Google Scholar 

  11. Tian J, Ma J, Ma K et al (2013) β-Glucan enhances antitumor immune responses by regulating differentiation and function of monocytic myeloid-derived suppressor cells. Eur J Immunol 43:1220–1230. https://doi.org/10.1002/eji.201242841

    Article  PubMed  CAS  Google Scholar 

  12. Albeituni SH, Ding C, Liu M et al (2016) Yeast-derived particulate β-glucan treatment subverts the suppression of myeloid-derived suppressor cells (MDSC) by Inducing polymorphonuclear MDSC apoptosis and monocytic MDSC differentiation to APC in cancer. J Immunol 196:2167–2180. https://doi.org/10.4049/jimmunol.1501853

    Article  PubMed  CAS  Google Scholar 

  13. Osorio F, LeibundGut-Landmann S, Lochner M et al (2008) DC activated via dectin-1 convert Treg into IL-17 producers. Eur J Immunol 38:3274–3281. https://doi.org/10.1002/eji.200838950

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. LeibundGut-Landmann S, Osorio F, Brown GD, Reis e Sousa C (2008) Stimulation of dendritic cells via the dectin-1/Syk pathway allows priming of cytotoxic T-cell responses. Blood 112:4971–4980. https://doi.org/10.1182/blood-2008-05-158469

    Article  PubMed  CAS  Google Scholar 

  15. Netea MGG, Quintin J, van der Meer JWM, van der Meer JWM (2011) Trained immunity: a memory for innate host defense. Cell Host Microbe 9:355–361. https://doi.org/10.1016/j.chom.2011.04.006

    Article  PubMed  CAS  Google Scholar 

  16. Hohl TM, Rivera A, Lipuma L et al (2009) Inflammatory monocytes facilitate adaptive CD4 T cell responses during respiratory fungal infection. Cell Host Microbe 6:470–481. https://doi.org/10.1016/j.chom.2009.10.007

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Cheng S-C, Quintin J, Cramer RA et al (2014) mTOR- and HIF-1α-mediated aerobic glycolysis as metabolic basis for trained immunity. Science 345:1250684. https://doi.org/10.1126/science.1250684

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Clancy-Thompson E, Perekslis TJ, Croteau W et al (2015) Melanoma induces, and adenosine suppresses, CXCR3-cognate chemokine production and T-cell infiltration of lungs bearing metastatic-like disease. Cancer Immunol Res. https://doi.org/10.1158/2326-6066.CIR-15-0015

    Article  PubMed  PubMed Central  Google Scholar 

  19. Cramer T, Yamanishi Y, Clausen BE et al (2003) HIF-1alpha is essential for myeloid cell-mediated inflammation. Cell 112:645–657

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Clancy-Thompson E, Perekslis TJ, Croteau W et al (2015) Melanoma induces, and adenosine suppresses, CXCR3-cognate chemokine production and T-cell infiltration of lungs bearing metastatic-like disease. Cancer Immunol Res 3:956–967. https://doi.org/10.1158/2326-6066.CIR-15-0015

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Huang H, Ostroff GR, Lee CK et al (2012) Relative contributions of dectin-1 and complement to immune responses to particulate β-glucans. J Immunol 189:312–317. https://doi.org/10.4049/jimmunol.1200603

    Article  PubMed  CAS  Google Scholar 

  22. Wang X, Wang W, Xu J, Le Q (2013) Effect of rapamycin and interleukin-2 on regulatory CD4+CD25+Foxp3+ T cells in mice after allogenic corneal transplantation. Transplant Proc 45:528–537. https://doi.org/10.1016/j.transproceed.2012.06.064

    Article  PubMed  CAS  Google Scholar 

  23. Bekkering S, Blok BA, Joosten LAB et al (2016) In vitro experimental model of trained innate immunity in human primary monocytes. Clin Vaccine Immunol 23:926–933. https://doi.org/10.1128/CVI.00349-16

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Liu C, Yu S, Kappes J et al (2007) Expansion of spleen myeloid suppressor cells represses NK cell cytotoxicity in tumor-bearing host. Blood 109:4336–4342. https://doi.org/10.1182/blood-2006-09-046201

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Butler KL, Clancy-Thompson E, Mullins DW (2017) CXCR3+ monocytes/macrophages are required for establishment of pulmonary metastases. Sci Rep 7:45593. https://doi.org/10.1038/srep45593

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Sorensen MR, Pedersen SR, Lindkvist A et al (2014) Quantification of B16 melanoma cells in lungs using triplex Q-PCR—a new approach to evaluate melanoma cell metastasis and tumor control. PLoS One 9:e87831. https://doi.org/10.1371/journal.pone.0087831

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Misharin AV, Morales-Nebreda L, Mutlu GM et al (2013) Flow cytometric analysis of macrophages and dendritic cell subsets in the mouse lung. Am J Respir Cell Mol Biol 49:503–510. https://doi.org/10.1165/rcmb.2013-0086MA

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Hong F, Hansen RD, Yan J et al (2003) Beta-glucan functions as an adjuvant for monoclonal antibody immunotherapy by recruiting tumoricidal granulocytes as killer cells. Cancer Res 63:9023–9031

    PubMed  CAS  Google Scholar 

  29. Yoon TJ, Kim TJ, Lee H et al (2008) Anti-tumor metastatic activity of β-glucan purified from mutated Saccharomyces cerevisiae. Int Immunopharmacol 8:36–42. https://doi.org/10.1016/j.intimp.2007.10.005

    Article  PubMed  CAS  Google Scholar 

  30. Ballas ZK, Buchta CM, Rosean TR et al (2013) Role of NK cell subsets in organ-specific murine melanoma metastasis. PLoS One 8:e65599. https://doi.org/10.1371/JOURNAL.PONE.0065599

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Noy R, Pollard JW (2014) Tumor-associated macrophages: from mechanisms to therapy. Immunity 41:49–61. https://doi.org/10.1016/j.immuni.2014.06.010

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Pommier A, Audemard A, Durand A et al (2013) Inflammatory monocytes are potent antitumor effectors controlled by regulatory CD4+ T cells. Proc Natl Acad Sci 110:13085–13090. https://doi.org/10.1073/pnas.1300314110

    Article  PubMed  PubMed Central  Google Scholar 

  33. Vetvicka V, Vetvickova J (2015) Glucan supplementation has strong anti-melanoma effects: role of NK cells. Anticancer Res 35:5287–5292

    PubMed  CAS  Google Scholar 

  34. Boring L, Gosling J, Chensue SW et al (1997) Impaired monocyte migration and reduced type 1 (Th1) cytokine responses in C–C chemokine receptor 2 knockout mice. J Clin Invest 100:2552–2561. https://doi.org/10.1172/JCI119798

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Fu X, Tao L, Rivera A et al (2010) A simple and sensitive method for measuring tumor-specific T cell cytotoxicity. PLoS One 5:e11867. https://doi.org/10.1371/journal.pone.0011867

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Yamamoto K, Kimura T, Sugitachi A, Matsuura N (2009) Anti-angiogenic and anti-metastatic effects of β-1,3-D-glucan purified from Hanabiratake, Sparassis crispa. Biol Pharm Bull 32:259–263. https://doi.org/10.1248/bpb.32.259

    Article  PubMed  CAS  Google Scholar 

  37. Cooper PD, Sim RB (1984) Substances that can trigger activation of the alternative pathway of complement have anti-melanoma activity in mice. Int J Cancer 33:683–687. https://doi.org/10.1002/ijc.2910330520

    Article  PubMed  CAS  Google Scholar 

  38. Qi C, Cai Y, Gunn L et al (2011) Differential pathways regulating innate and adaptive antitumor immune responses by particulate and soluble yeast-derived β-glucans. Blood 117:6825–6836. https://doi.org/10.1182/blood-2011-02-339812

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Baran J, Allendorf DJ, Hong F, Ross GD (2007) Oral beta-glucan adjuvant therapy converts nonprotective Th2 response to protective Th1 cell-mediated immune response in mammary tumor-bearing mice. Folia Histochem Cytobiol 45:107–114

    PubMed  CAS  Google Scholar 

  40. Netea MG, Quintin J, van der Meer JWM (2011) Trained Immunity: a memory for innate host defense. Cell Host Microbe 9:355–361. https://doi.org/10.1016/j.chom.2011.04.006

    Article  PubMed  CAS  Google Scholar 

  41. Goodridge HS, Wolf AJ, Underhill DM (2009) β-Glucan recognition by the innate immune system. Immunol Rev 230:38–50. https://doi.org/10.1111/j.1600-065X.2009.00793.x

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  42. Quintin J, Saeed S, Martens JHA et al (2012) Candida albicans infection affords protection against reinfection via functional reprogramming of monocytes. Cell Host Microbe 12:223–232. https://doi.org/10.1016/j.chom.2012.06.006

    Article  PubMed  CAS  Google Scholar 

  43. Demir G, Klein HO, Mandel-Molinas N, Tuzuner N (2007) Beta glucan induces proliferation and activation of monocytes in peripheral blood of patients with advanced breast cancer. Int Immunopharmacol 7:113–116. https://doi.org/10.1016/j.intimp.2006.08.011

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

We thank Brent L. Berwin, Constance E. Brinkerhoff, Marc S. Ernstoff, John Frelinger, and Irene M. Mullins for helpful suggestions and feedback. We thank Jennifer L. Vella and Yina H. Huang for assistance with B16.F10-luciferase cells.

Funding

Matthew P. Alexander was supported, in part, through the Joanna M. Nicolay Melanoma Foundation. David W. Mullins was supported, in part, through USPHS R03 CA188418. Flow cytometry and multiplex assays were carried out in DartLab, the Immune Monitoring and Flow Cytometry Shared Resource, supported by a National Cancer Institute Cancer Center Support Grant to the Norris Cotton Cancer Center (P30CA023108-37) and an Immunology COBRE Grant (P30GM103415-15) from the National Institute of General Medical Sciences.

Author information

Authors and Affiliations

Authors

Contributions

MPA performed all in vitro and mouse model studies, participated in data and statistical analyses, and contributed to the drafting of the manuscript. SNF contributed to study design and data analysis, and participated in drafting of the manuscript. GRO generated YGP β-glucan, contributed to study design and data analysis, and participated in drafting of the manuscript. RAC maintained and provided Myeloid-HIF1α−/− mice, contributed to study design and data analysis, and participated in drafting of the manuscript. DWM participated in in vitro and mouse model studies, oversaw study design and data/statistical analyses, and contributed to the drafting of the manuscript.

Corresponding author

Correspondence to David W. Mullins.

Ethics declarations

Conflict of interest

David W. Mullins is a senior scientific advisor and has received research grants, unrelated to the current work, from Qu Biologics (Vancouver, BC). The remaining authors declare no conflict of interest.

Animal studies

C57BL/6J (stock no. 000664) and CCR2−/− mice (stock no. 004999) mice were obtained from Jackson Laboratories (Bar Harbor, ME). CCR2.DTR mice were a gift from Dr. Tobias Hohl (Memorial Sloan Kettering Cancer Center). Myeloid-HIF1α−/− mice (LysM-Cre floxed-HIF1α exon 2) were generated and bred in house at Dartmouth by Dr. Robert A. Cramer.

Ethical approval

All animal care procedures and experimental protocols were performed in accordance with all applicable international, national, and/or institutional guidelines for the care and use of animals. All animal care procedures and experimental protocols were approved by the Institutional Animal Care and Use Committee (IACUC) of Dartmouth College (protocol 1141).

Cell line authentication

B16-F10 were newly acquired from ATCC, then expanded and cryopreserved to create individual aliquots that were used for each study. B16.F10-luciferase cells were generated from new aliqouts of B16-F10. Cells were confirmed to be free of Mycoplasma infection using the Hek Blue Mycoplasma Detection Kit from Invivogen.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (PDF 1850 KB)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Alexander, M.P., Fiering, S.N., Ostroff, G.R. et al. Beta-glucan-induced inflammatory monocytes mediate antitumor efficacy in the murine lung. Cancer Immunol Immunother 67, 1731–1742 (2018). https://doi.org/10.1007/s00262-018-2234-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00262-018-2234-9

Keywords

Navigation