Skip to main content
Top
Published in: Cancer Immunology, Immunotherapy 1/2021

Open Access 01-01-2021 | Colorectal Cancer | Original Article

Mechanism of action of a T cell-dependent bispecific antibody as a breakthrough immunotherapy against refractory colorectal cancer with an oncogenic mutation

Authors: Daisuke Kamakura, Ryutaro Asano, Hiroki Kawai, Masahiro Yasunaga

Published in: Cancer Immunology, Immunotherapy | Issue 1/2021

Login to get access

Abstract

T cell-dependent bispecific antibody (TDB)-induced T cell activation, which can eliminate tumor cells independent of MHC engagement, is expected to be a novel breakthrough immunotherapy against refractory cancer. However, the mechanism of action of TDBs has not been fully elucidated thus far. We focused on TDB-induced T cell–tumor cell contact as an important initial step in direct T cell-mediated tumor cell killing via transport of cytotoxic cell proteases (e.g., granzymes) with or without immunological synapse formation. Using an anti-EGFR/CD3 TDB, hEx3, we visualized and quantified T cell–tumor cell contact and demonstrated a correlation between the degree of cell contact and TDB efficacy. We also found that cytokines, including interferon-gamma (IFNγ) and tumor necrosis factor-alpha (TNFα) secreted by activated T cells, damaged tumor cells in a cell contact-independent manner. Moreover, therapeutic experiences clearly indicated that hEx3, unlike conventional anti-EGFR antibodies, was effective against colorectal cancer (CRC) cells with mutant KRAS, BRAF, or PIK3CA. In a pharmacokinetic analysis, T cells spread gradually in accordance with the hEx3 distribution within tumor tissue. Accordingly, we propose that TDBs should have four action steps: 1st, passive targeting via size-dependent tumor accumulation; 2nd, active targeting via specific binding to tumor cells; 3rd, T cell redirection toward tumor cells; and 4th, TDB-induced cell contact-dependent (direct) or -independent (indirect) tumor cell killing. Finally, our TDB hEx3 may be a promising reagent against refractory CRC with an oncogenic mutation associated with a poor prognosis.
Appendix
Available only for authorised users
Literature
2.
go back to reference Sankaranarayanan R, Swaminathan R, Brenner H, Chen K, Chia KS, Chen JG, Law SCK, Ahn Y-O, Xiang YB, Yeole BB, Shin HR, Shanta V, Woo ZH, Martin N, Sumitsawan Y, Sriplung H, Barboza AO, Eser S, Nene BM, Suwanrungruang K, Jayalekshmi P, Dikshit R, Wabinga H, Esteban DB, Laudico A, Bhurgri Y, Bah E, Al-Hamdan N (2010) Cancer survival in Africa, Asia, and Central America: a population-based study. Lancet Oncol 11(2):165–173. https://doi.org/10.1016/s1470-2045(09)70335-3CrossRefPubMed Sankaranarayanan R, Swaminathan R, Brenner H, Chen K, Chia KS, Chen JG, Law SCK, Ahn Y-O, Xiang YB, Yeole BB, Shin HR, Shanta V, Woo ZH, Martin N, Sumitsawan Y, Sriplung H, Barboza AO, Eser S, Nene BM, Suwanrungruang K, Jayalekshmi P, Dikshit R, Wabinga H, Esteban DB, Laudico A, Bhurgri Y, Bah E, Al-Hamdan N (2010) Cancer survival in Africa, Asia, and Central America: a population-based study. Lancet Oncol 11(2):165–173. https://​doi.​org/​10.​1016/​s1470-2045(09)70335-3CrossRefPubMed
5.
go back to reference Karapetis CS, Khambata-Ford S, Jonker DJ, O'Callaghan CJ, Tu D, Tebbutt NC, Simes RJ, Chalchal H, Shapiro JD, Robitaille S, Price TJ, Shepherd L, Au HJ, Langer C, Moore MJ, Zalcberg JR (2008) K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med 359(17):1757–1765. https://doi.org/10.1056/NEJMoa0804385CrossRefPubMed Karapetis CS, Khambata-Ford S, Jonker DJ, O'Callaghan CJ, Tu D, Tebbutt NC, Simes RJ, Chalchal H, Shapiro JD, Robitaille S, Price TJ, Shepherd L, Au HJ, Langer C, Moore MJ, Zalcberg JR (2008) K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med 359(17):1757–1765. https://​doi.​org/​10.​1056/​NEJMoa0804385CrossRefPubMed
6.
go back to reference Misale S, Yaeger R, Hobor S, Scala E, Janakiraman M, Liska D, Valtorta E, Schiavo R, Buscarino M, Siravegna G, Bencardino K, Cercek A, Chen CT, Veronese S, Zanon C, Sartore-Bianchi A, Gambacorta M, Gallicchio M, Vakiani E, Boscaro V, Medico E, Weiser M, Siena S, Di Nicolantonio F, Solit D, Bardelli A (2012) Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature 486(7404):532–536. https://doi.org/10.1038/nature11156CrossRefPubMedPubMedCentral Misale S, Yaeger R, Hobor S, Scala E, Janakiraman M, Liska D, Valtorta E, Schiavo R, Buscarino M, Siravegna G, Bencardino K, Cercek A, Chen CT, Veronese S, Zanon C, Sartore-Bianchi A, Gambacorta M, Gallicchio M, Vakiani E, Boscaro V, Medico E, Weiser M, Siena S, Di Nicolantonio F, Solit D, Bardelli A (2012) Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature 486(7404):532–536. https://​doi.​org/​10.​1038/​nature11156CrossRefPubMedPubMedCentral
7.
17.
go back to reference Kim M, Kim SJ, Xu Z, Ha SY, Byeon JH, Kang EJ, Shin SH, Yoo SK, Jee HG, Yoon SG, Yi JW, Bae JM, Yu HW, Chai YJ, Cho SW, Choi JY, Lee KE, Han W (2020) BRAF(V600E) transduction of an SV40-immortalized normal human thyroid cell line induces dedifferentiated thyroid carcinogenesis in a mouse xenograft model. Thyroid 30(4):487–500. https://doi.org/10.1089/thy.2019.0301CrossRefPubMed Kim M, Kim SJ, Xu Z, Ha SY, Byeon JH, Kang EJ, Shin SH, Yoo SK, Jee HG, Yoon SG, Yi JW, Bae JM, Yu HW, Chai YJ, Cho SW, Choi JY, Lee KE, Han W (2020) BRAF(V600E) transduction of an SV40-immortalized normal human thyroid cell line induces dedifferentiated thyroid carcinogenesis in a mouse xenograft model. Thyroid 30(4):487–500. https://​doi.​org/​10.​1089/​thy.​2019.​0301CrossRefPubMed
20.
go back to reference Zaretsky JM, Garcia-Diaz A, Shin DS, Escuin-Ordinas H, Hugo W, Hu-Lieskovan S, Torrejon DY, Abril-Rodriguez G, Sandoval S, Barthly L, Saco J, Homet Moreno B, Mezzadra R, Chmielowski B, Ruchalski K, Shintaku IP, Sanchez PJ, Puig-Saus C, Cherry G, Seja E, Kong X, Pang J, Berent-Maoz B, Comin-Anduix B, Graeber TG, Tumeh PC, Schumacher TN, Lo RS, Ribas A (2016) Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med 375(9):819–829. https://doi.org/10.1056/NEJMoa1604958CrossRefPubMedPubMedCentral Zaretsky JM, Garcia-Diaz A, Shin DS, Escuin-Ordinas H, Hugo W, Hu-Lieskovan S, Torrejon DY, Abril-Rodriguez G, Sandoval S, Barthly L, Saco J, Homet Moreno B, Mezzadra R, Chmielowski B, Ruchalski K, Shintaku IP, Sanchez PJ, Puig-Saus C, Cherry G, Seja E, Kong X, Pang J, Berent-Maoz B, Comin-Anduix B, Graeber TG, Tumeh PC, Schumacher TN, Lo RS, Ribas A (2016) Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med 375(9):819–829. https://​doi.​org/​10.​1056/​NEJMoa1604958CrossRefPubMedPubMedCentral
21.
26.
go back to reference Scott AC, Dundar F, Zumbo P, Chandran SS, Klebanoff CA, Shakiba M, Trivedi P, Menocal L, Appleby H, Camara S, Zamarin D, Walther T, Snyder A, Femia MR, Comen EA, Wen HY, Hellmann MD, Anandasabapathy N, Liu Y, Altorki NK, Lauer P, Levy O, Glickman MS, Kaye J, Betel D, Philip M, Schietinger A (2019) TOX is a critical regulator of tumour-specific T cell differentiation. Nature 571(7764):270–274. https://doi.org/10.1038/s41586-019-1324-yCrossRefPubMedPubMedCentral Scott AC, Dundar F, Zumbo P, Chandran SS, Klebanoff CA, Shakiba M, Trivedi P, Menocal L, Appleby H, Camara S, Zamarin D, Walther T, Snyder A, Femia MR, Comen EA, Wen HY, Hellmann MD, Anandasabapathy N, Liu Y, Altorki NK, Lauer P, Levy O, Glickman MS, Kaye J, Betel D, Philip M, Schietinger A (2019) TOX is a critical regulator of tumour-specific T cell differentiation. Nature 571(7764):270–274. https://​doi.​org/​10.​1038/​s41586-019-1324-yCrossRefPubMedPubMedCentral
27.
go back to reference Matsumura Y, Maeda H (1986) A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. Cancer Res 46(12 Pt 1):6387–6392PubMed Matsumura Y, Maeda H (1986) A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. Cancer Res 46(12 Pt 1):6387–6392PubMed
Metadata
Title
Mechanism of action of a T cell-dependent bispecific antibody as a breakthrough immunotherapy against refractory colorectal cancer with an oncogenic mutation
Authors
Daisuke Kamakura
Ryutaro Asano
Hiroki Kawai
Masahiro Yasunaga
Publication date
01-01-2021
Publisher
Springer Berlin Heidelberg
Published in
Cancer Immunology, Immunotherapy / Issue 1/2021
Print ISSN: 0340-7004
Electronic ISSN: 1432-0851
DOI
https://doi.org/10.1007/s00262-020-02667-9

Other articles of this Issue 1/2021

Cancer Immunology, Immunotherapy 1/2021 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine