Skip to main content

Advertisement

Log in

Combined treatment with anti-HER2/neu and anti-4-1BB monoclonal antibodies induces a synergistic antitumor effect but requires dose optimization to maintain immune memory for protection from lethal rechallenge

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

Abstract

Human epidermal growth factor receptor type 2 (HER2)-positive breast cancer that is treated with anti-HER2/neu monoclonal antibody (mAb) is not free from late recurrences. Addition of anti-4-1BB mAb to anti-HER2/neu mAb has been demonstrated to strengthen the cytotoxic antitumor response. Our study expands on this by revealing the influence of anti-4-1BB mAb addition on the immune memory of anti-HER2/neu mAb. We designed murine breast cancer models by implanting TUBO and TUBO-P2J cell lines in mice, which were then treated with anti-HER2/neu and/or anti-4-1BB mAb. After complete surgical and/or chemical regression of the tumor, the mice were rechallenged with a second injection of cancer cells. Notably, anti-HER2/neu and anti-4-1BB mAb combination therapy had a synergistic antitumor effect at the initial treatment. However, the combination therapy did not evoke immune memory, allowing the tumors to thrive at rechallenge with reduced CD44+ expression in CD8+ T cells. Immune memory was also impaired when anti-4-1BB mAb was administered to naive CD8+ T cells but was sustained when this was administered to activated CD8+ T cells. In an attempt to resist the loss of immune memory, we controlled the dose of anti-4-1BB mAb to optimize the stimulation of activated CD8+ T cells. Immune memory was achieved with the dose regulation of anti-4-1BB mAb to 1 mg/kg in our model. Our study demonstrates the importance in understanding the adaptive immune mechanism of anti-HER2/neu and anti-4-1BB mAb combination therapy and suggests a dose optimization strategy is necessary to ensure development of successful immune memory.

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

Similar content being viewed by others

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

References

  1. Hudis CA (2007) Trastuzumab–mechanism of action and use in clinical practice. N Engl J Med 357(1):39–51

    Article  CAS  Google Scholar 

  2. Clynes RA et al (2000) Inhibitory Fc receptors modulate in vivo cytotoxicity against tumor targets. Nat Med 6(4):443–446

    Article  CAS  Google Scholar 

  3. Park S et al (2010) The therapeutic effect of anti-HER2/neu antibody depends on both innate and adaptive immunity. Cancer Cell 18(2):160–170

    Article  CAS  Google Scholar 

  4. Cameron D et al (2017) 11 years’ follow-up of trastuzumab after adjuvant chemotherapy in HER2-positive early breast cancer: final analysis of the HERceptin Adjuvant (HERA) trial. Lancet 389(10075):1195–1205

    Article  CAS  Google Scholar 

  5. Watts TH (2005) TNF/TNFR family members in costimulation of T cell responses. Annu Rev Immunol 23:23–68

    Article  CAS  Google Scholar 

  6. Melero I et al (1997) Monoclonal antibodies against the 4–1BB T-cell activation molecule eradicate established tumors. Nat Med 3(6):682–685

    Article  CAS  Google Scholar 

  7. Takahashi C et al (1999) Cutting edge: 4–1BB is a bona fide CD8 T cell survival signal. J Immunol 162(9):5037–5040

    CAS  PubMed  Google Scholar 

  8. Stagg J et al (2011) Anti-ErbB-2 mAb therapy requires type I and II interferons and synergizes with anti-PD-1 or anti-CD137 mAb therapy. Proc Natl Acad Sci USA 108(17):7142–7147

    Article  CAS  Google Scholar 

  9. Song H et al (2014) Intratumoral heterogeneity impacts the response to anti-neu antibody therapy. BMC Cancer 14:647

    Article  Google Scholar 

  10. Kang SW et al (2017) Anti-CD137 Suppresses Tumor Growth by Blocking Reverse Signaling by CD137 Ligand. Cancer Res 77(21):5989–6000

    Article  CAS  Google Scholar 

  11. Masu T et al (2018) Anti-CD137 monoclonal antibody enhances trastuzumab-induced, natural killer cell-mediated cytotoxicity against pancreatic cancer cell lines with low human epidermal growth factor-like receptor 2 expression. PLoS ONE 13(12):e0200664

    Article  CAS  Google Scholar 

  12. Sallusto F et al (1999) Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 401(6754):708–712

    Article  CAS  Google Scholar 

  13. Moasser MM (2007) Targeting the function of the HER2 oncogene in human cancer therapeutics. Oncogene 26(46):6577–6592

    Article  CAS  Google Scholar 

  14. Kiessling R et al (2002) Cellular immunity to the Her-2/neu protooncogene. Adv Cancer Res 85:101–144

    Article  CAS  Google Scholar 

  15. Le XF et al (2005) Genes affecting the cell cycle, growth, maintenance, and drug sensitivity are preferentially regulated by anti-HER2 antibody through phosphatidylinositol 3-kinase-AKT signaling. J Biol Chem 280(3):2092–2104

    Article  CAS  Google Scholar 

  16. Mittendorf EA et al (2010) A novel interaction between HER2/neu and cyclin E in breast cancer. Oncogene 29(27):3896–3907

    Article  CAS  Google Scholar 

  17. Pegram M et al (1999) Inhibitory effects of combinations of HER-2/neu antibody and chemotherapeutic agents used for treatment of human breast cancers. Oncogene 18(13):2241–2251

    Article  CAS  Google Scholar 

  18. Clynes RA et al (2000) Inhibitory Fc receptors modulate in vivo cytoxicity against tumor targets. Nat Med 6(4):443–446

    Article  CAS  Google Scholar 

  19. Musolino A et al (2008) Immunoglobulin G fragment C receptor polymorphisms and clinical efficacy of trastuzumab-based therapy in patients with HER-2/neu-positive metastatic breast cancer. J Clin Oncol 26(11):1789–1796

    Article  CAS  Google Scholar 

  20. Arnould L et al (2006) Trastuzumab-based treatment of HER2-positive breast cancer: an antibody-dependent cellular cytotoxicity mechanism? Br J Cancer 94(2):259–267

    Article  CAS  Google Scholar 

  21. Varchetta S et al (2007) Elements related to heterogeneity of antibody-dependent cell cytotoxicity in patients under trastuzumab therapy for primary operable breast cancer overexpressing Her2. Cancer Res 67(24):11991–11999

    Article  CAS  Google Scholar 

  22. Gennari R et al (2004) Pilot study of the mechanism of action of preoperative trastuzumab in patients with primary operable breast tumors overexpressing HER2. Clin Cancer Res 10(17):5650–5655

    Article  CAS  Google Scholar 

  23. Ladjemi MZ et al (2010) Anti-HER2 vaccines: new prospects for breast cancer therapy. Cancer Immunol Immunother 59(9):1295–1312

    Article  CAS  Google Scholar 

  24. Wang W et al (2015) NK cell-mediated antibody-dependent cellular cytotoxicity in cancer immunotherapy. Front Immunol 6:368

    PubMed  PubMed Central  Google Scholar 

  25. Chester C et al (2016) 4–1BB agonism: adding the accelerator to cancer immunotherapy. Cancer Immunol Immunother 65(10):1243–1248

    Article  CAS  Google Scholar 

  26. Dranoff G (2004) Cytokines in cancer pathogenesis and cancer therapy. Nat Rev Cancer 4(1):11–22

    Article  CAS  Google Scholar 

  27. Topalian SL et al (2016) Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer 16(5):275–287

    Article  CAS  Google Scholar 

  28. Farhood B et al (2019) CD8(+) cytotoxic T lymphocytes in cancer immunotherapy: A review. J Cell Physiol 234(6):8509–8521

    Article  CAS  Google Scholar 

  29. Etxeberria I et al (2020) New emerging targets in cancer immunotherapy: CD137/4–1BB costimulatory axis. ESMO Open 4(Suppl 3):e000733

    PubMed  PubMed Central  Google Scholar 

  30. Ahmadzadeh M et al (2009) (2009) Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 114(8):1537–1544

    Article  CAS  Google Scholar 

  31. Miller BC et al (2019) Subsets of exhausted CD8(þ) T cells differentially mediate tumor control and respond to checkpoint blockade. Nat Immunol 20(3):326–336

    Article  CAS  Google Scholar 

  32. Paley MA et al (2012) Progenitor and terminal subsets of CD8þ T cells cooperate to contain chronic viral infection. Science 338(6111):1220–1225

    Article  CAS  Google Scholar 

  33. Krasniqi E et al (2019) Immunotherapy in HER2-positive breast cancer: state of the art and future perspectives. J Hematol Oncol 12(1):111

    Article  CAS  Google Scholar 

Download references

Funding

This work was supported by the Bio & Medical Technology Development Program of the National Research Foundation (NRF) of Korea funded by the Korean Government (MSIT) Under Grant (No.2019M3A9H1103607); Under Grant (No.2017R1C1B5076247); Under Grant (No.2016R1D1A1B03935426); and the SAMJINPHARM.CO., LTD Under Grant (SJ-IIT-17-07).

Author information

Authors and Affiliations

Authors

Contributions

HYK, J-HC and SGP conceptualized the study and reviewed the writing of the manuscript. HYK, J-HC and MMH wrote the original draft of the manuscript and analyzed the data. JHP, I-HK and BKC investigated the immune cell population and activity. AL and SGP were involved in supervision, funding acquisition and project administration.

Corresponding authors

Correspondence to Anbok Lee or SaeGwang Park.

Ethics declarations

Conflict of interest

The researcher claims no conflicts of interest.

Ethics approval and consent to participate

The experimental procedures were reviewed and approved by the Institutional Animal Care and Use Committee of Inje University (2019-002).

Consent for publication

I would like to declare that the manuscript is approved by all authors for publication.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kim, H.Y., Choi, JH., Haque, M.M. et al. Combined treatment with anti-HER2/neu and anti-4-1BB monoclonal antibodies induces a synergistic antitumor effect but requires dose optimization to maintain immune memory for protection from lethal rechallenge. Cancer Immunol Immunother 71, 967–978 (2022). https://doi.org/10.1007/s00262-021-03120-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00262-021-03120-1

Keywords

Navigation