Skip to main content

Advertisement

Log in

Multiple antibodies targeting tumor-specific mutations redirect immune cells to inhibit tumor growth and increase survival in experimental animal models

  • Research Article
  • Published:
Clinical and Translational Oncology Aims and scope Submit manuscript

Abstract

Background

T cell therapy for cancer involves genetic introduction of a target-binding feature into autologous T cells, ex vivo expansion and single large bolus administration back to the patient. These reprogrammed T cells can be highly effective in killing cells, but tumor heterogeneity results in regrowth of cells that do not sufficiently express the single antigen being targeted. We describe a cell-based therapy that simultaneously targets multiple tumor-specific antigens.

Methods

High-affinity polyclonal rabbit antibodies were generated against nine different surface-related tumor-specific mutations on B16F10 cells. Unsorted splenic effector cells from syngeneic mice were incubated with a cocktail of the nine anti-B16F10 antibodies. These ‘armed’ effector cells were used to treat mice previously inoculated with B16F10 melanoma cells.

Results

The cocktail of nine antibodies resulted in dense homogeneous binding to histological sections of B16F10 cells. Five treatments with the armed effector cells and PD1 inhibition inhibited tumor growth and improved survival. Shortening the interval of the five treatments from every three days to every day increased survival. Arming effector cells with the four antibodies showing best binding to B16F10 cells even further increased survival.

Conclusions

This study demonstrates that ex vivo arming a mixed population of immune effector cells with antibodies targeting multiple tumor-specific mutated proteins in conjunction with PD1 inhibition delayed tumor growth and prolonged survival in mice inoculated with an aggressive melanoma. A remarkably low total antibody dose of less than 5 µg was sufficient to accomplish tumor inhibition. Scaling up to clinical level may be feasible.

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
Fig. 6

Similar content being viewed by others

References

  1. Topalian SL, Muul LM, Solomon D, Rosenberg SA. Expansion of human tumor infiltrating lymphocytes for use in immunotherapy trials. J Immunol Methods. 1987;102:127–41.

    Article  CAS  Google Scholar 

  2. Rosenberg SA, Packard BS, Aebersold PM, et al. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report. N Engl J Med 1988;319:1676–80

    Article  CAS  Google Scholar 

  3. Rosenberg SA, Dudley ME. Cancer regression in patients with metastatic melanoma after the transfer of autologous antitumor lymphocytes. Proc Natl Acad Sci USA. 2004;101(Suppl 2):14639–45.

    Article  CAS  Google Scholar 

  4. Yannelli JR, Hyatt C, McConnell S, et al. Growth of tumor-infiltrating lymphocytes from human solid cancers: summary of a 5-year experience. Int J Cancer. 1996;65:413–21.

    Article  CAS  Google Scholar 

  5. Minutolo NG, Hollander EE, Powell DJ Jr. The Emergence of universal immune receptor T cell therapy for cancer. Front Oncol. 2019;9:176.

    Article  Google Scholar 

  6. Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365:725–33.

    Article  CAS  Google Scholar 

  7. Castle JC, Kreiter S, Diekmann J, et al. Exploiting the mutanome for tumor vaccination. Cancer Res. 2012;72:1081–91.

    Article  CAS  Google Scholar 

  8. Shukla GS, Sun YJ, Pero SC, Sholler GS, Krag DN. Immunization with tumor neoantigens displayed on T7 phage nanoparticles elicits plasma antibody and vaccine-draining lymph node B cell responses. J Immunol Methods. 2018;460:51–62.

    Article  CAS  Google Scholar 

  9. Shukla GS, Olson WC, Pero SC, et al. Vaccine-draining lymph nodes of cancer patients for generating anti-cancer antibodies. J Transl Med. 2017;15:180.

    Article  Google Scholar 

  10. McGray AJ, Hallett R, Bernard D, et al. Immunotherapy-induced CD8+ T cells instigate immune suppression in the tumor. Mol Ther. 2014;22:206–18.

    Article  CAS  Google Scholar 

  11. Dominguez-Soto A, de las Casas-Engel M, Bragado R, et al. Intravenous immunoglobulin promotes antitumor responses by modulating macrophage polarization. J Immunol 2014;193:5181–9.

    Article  Google Scholar 

  12. Hassannia H, Amiri MM, Jadidi-Niaragh F, et al. Inhibition of tumor growth by mouse ROR1 specific antibody in a syngeneic mouse tumor model. Immunol Lett. 2018;193:35–41.

    Article  CAS  Google Scholar 

  13. Cho HI, Barrios K, Lee YR, Linowski AK, Celis E. BiVax: a peptide/poly-IC subunit vaccine that mimics an acute infection elicits vast and effective anti-tumor CD8 T-cell responses. Cancer Immunol Immunother. 2013;62:787–99.

    Article  CAS  Google Scholar 

  14. Buhtoiarov IN, Sondel PM, Eickhoff JC, Rakhmilevich AL. Macrophages are essential for antitumour effects against weakly immunogenic murine tumours induced by class B CpG-oligodeoxynucleotides. Immunology. 2007;120:412–23.

    Article  CAS  Google Scholar 

  15. Nagato T, Lee YR, Harabuchi Y, Celis E. Combinatorial immunotherapy of polyinosinic-polycytidylic acid and blockade of programmed death-ligand 1 induce effective CD8 T-cell responses against established tumors. Clin Cancer Res. 2014;20:1223–344.

    Article  CAS  Google Scholar 

  16. Snyder A, Makarov V, Merghoub T, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371:2189–99.

    Article  Google Scholar 

  17. Wang L, Lawrence MS, Wan Y, et al. SF3B1 and other novel cancer genes in chronic lymphocytic leukemia. N Engl J Med. 2011;365:2497–506.

    Article  CAS  Google Scholar 

  18. Koboldt DC, Larson DE, Wilson RK. Using VarScan 2 for Germline Variant Calling and Somatic Mutation Detection. Curr Protoc Bioinform 2013;44:15 4 1–7

  19. Cingolani P, Platts A, le Wang L, et al. A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff: SNPs in the genome of Drosophila melanogaster strain w1118; iso-2; iso-3. Fly (Austin). 2012;6:80–92.

    Article  CAS  Google Scholar 

  20. McLaren W, Gil L, Hunt SE, et al. The Ensembl Variant Effect Predictor. Genome Biol. 2016;17:122.

    Article  Google Scholar 

  21. The UC. UniProt: the universal protein knowledgebase. Nucleic Acids Res. 2017;45:D158–D69.

    Article  Google Scholar 

  22. Fleit HB, Wright SD, Unkeless JC. Human neutrophil Fc gamma receptor distribution and structure. Proc Natl Acad Sci USA. 1982;79:3275–9.

    Article  CAS  Google Scholar 

  23. Patel KR, Roberts JT, Barb AW. Multiple variables at the leukocyte cell surface impact Fc γ receptor-dependent mechanisms. Front Immunol. 2019;10:223.

    Article  CAS  Google Scholar 

  24. Penfold PL, Walker LC, Roitt IM. Complex arming in antibody-dependent cell-mediated cytotoxicity: ultrastructural studies of the interaction between human effector cells armed with aggregated anti-DNP antibody and DNP-coated erythrocytes. Clin Exp Immunol. 1978;31:197–204.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Saksela E, Imir T, Makela O. Specifically cytotoxic human and mouse lymphoid cells induced with antibody or antigen-antibody complexes. J Immunol. 1975;115:1488–92.

    CAS  PubMed  Google Scholar 

  26. Park JH, Riviere I, Gonen M, et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N Engl J Med. 2018;378:449–59.

    Article  CAS  Google Scholar 

  27. Parrillo JE, Fauci AS. Apparent direct cellular cytotoxicity mediated via cytophilic antibody. Multiple Fc receptor bearing effector cell populations mediating cytophilic antibody induced cytotoxicity. Immunology 1977;33:839–50.

  28. Perlmann P, Perlmann H. Contactual lysis of antibody-coated chicken erythrocytes by purified lymphocytes. Cell Immunol. 1970;1:300–15.

    Article  CAS  Google Scholar 

  29. Matlung HL, Babes L, Zhao XW, et al. Neutrophils kill antibody-opsonized cancer cells by trogoptosis. Cell Rep. 2018;23(3946–59):e6.

    Google Scholar 

Download references

Acknowledgements

This work was supported by the SD Ireland Cancer Research Fund, Cast Off Chemo! Foundation, the Department of Surgery at the University of Vermont Larner College of Medicine internal grant, and the Beat Neuroblastoma Foundation. This work was also supported by an Institutional Development Award (IDeA) from the National Institute of General Medical Sciences of the National Institutes of Health under Grant Number P20GM103449. Flow cytometry data was obtained at the Harry Hood Bassett Flow Cytometry and Cell Sorting Facility at the University of Vermont Larner College of Medicine.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to D. N. Krag.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Ethical approval and ethical standards

All animal procedures used in the present study were approved by the UVM IACUC (Protocol # 18-002).

Animal source

Mice were obtained from the Jackson Laboratory (Bar Harbor, ME) in the present study.

Cell line authentication

We utilized confirmed sequence data of B16F10 mouse melanoma tumor cell line7 and selected multiple cell surface-related mutated proteins with a single amino acid substitution.

Informed consent

Informed consent is not applicable to this manuscript. We only report on mice and did refer to having the protocol correctly approved. The human sequence information comes from public databases.

Additional information

Publisher's Note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary file1 (PDF 1176 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shukla, G.S., Pero, S.C., Sun, Y.J. et al. Multiple antibodies targeting tumor-specific mutations redirect immune cells to inhibit tumor growth and increase survival in experimental animal models. Clin Transl Oncol 22, 1094–1104 (2020). https://doi.org/10.1007/s12094-019-02235-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12094-019-02235-3

Keywords

Navigation