Skip to main content
Top
Published in: Breast Cancer Research 1/2022

01-12-2022 | Solid Tumor | Short report

Inducible localized delivery of an anti-PD-1 scFv enhances anti-tumor activity of ROR1 CAR-T cells in TNBC

Authors: Micaela Harrasser, Satyen Harish Gohil, Hiu Lau, Marco Della Peruta, Vincent Muczynski, Dominic Patel, Elena Miranda, Kristiana Grigoriadis, Anita Grigoriadis, David Granger, Rachel Evans, Amit Chunilal Nathwani

Published in: Breast Cancer Research | Issue 1/2022

Login to get access

Abstract

Background

Chimeric antigen receptor (CAR)-T cells can induce powerful immune responses in patients with hematological malignancies but have had limited success against solid tumors. This is in part due to the immunosuppressive tumor microenvironment (TME) which limits the activity of tumor-infiltrating lymphocytes (TILs) including CAR-T cells. We have developed a next-generation armored CAR (F i-CAR) targeting receptor tyrosine kinase-like orphan receptor 1 (ROR1), which is expressed at high levels in a range of aggressive tumors including poorly prognostic triple-negative breast cancer (TNBC). The F i-CAR-T is designed to release an anti-PD-1 checkpoint inhibitor upon CAR-T cell activation within the TME, facilitating activation of CAR-T cells and TILs while limiting toxicity.

Methods

To bolster potency, we developed a F i-CAR construct capable of IL-2-mediated, NFAT-induced secretion of anti-PD-1 single-chain variable fragments (scFv) within the tumor microenvironment, following ROR1-mediated activation. Cytotoxic responses against TNBC cell lines as well as levels and binding functionality of released payload were analyzed in vitro by ELISA and flow cytometry. In vivo assessment of potency of F i-CAR-T cells was performed in a TNBC NSG mouse model.

Results

F i-CAR-T cells released measurable levels of anti-PD-1 payload with 5 h of binding to ROR1 on tumor and enhanced the cytotoxic effects at challenging 1:10 E:T ratios. Treatment of established PDL1 + TNBC xenograft model with F i-CAR-T cells resulted in significant abrogation in tumor growth and improved survival of mice (71 days), compared to non-armored CAR cells targeting ROR1 (F CAR-T) alone (49 days) or in combination with systemically administered anti-PD-1 antibody (57 days). Crucially, a threefold increase in tumor-infiltrating T cells was observed with F i-CAR-T cells and was associated with increased expression of genes related to cytotoxicity, migration and proliferation.

Conclusions

Our next-generation of ROR1-targeting inducible armored CAR platform enables the release of an immune stimulating payload only in the presence of target tumor cells, enhancing the therapeutic activity of the CAR-T cells. This technology provided a significant survival advantage in TNBC xenograft models. This coupled with its potential safety attributes merits further clinical evaluation of this approach in TNBC patients.
Appendix
Available only for authorised users
Literature
1.
go back to reference Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Miklos DB, Jacobson CA, Braunschweig I, Oluwole OO, Siddiqi T, Lin Y, et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 2017;377(26):2531–44.CrossRef Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Miklos DB, Jacobson CA, Braunschweig I, Oluwole OO, Siddiqi T, Lin Y, et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 2017;377(26):2531–44.CrossRef
2.
go back to reference Fucà G, Reppel L, Landoni E, Savoldo B, Dotti G. Enhancing chimeric antigen receptor T-cell efficacy in solid tumors. Clin Cancer Res. 2020;26:2444.CrossRef Fucà G, Reppel L, Landoni E, Savoldo B, Dotti G. Enhancing chimeric antigen receptor T-cell efficacy in solid tumors. Clin Cancer Res. 2020;26:2444.CrossRef
3.
go back to reference Balakrishnan A, Goodpaster T, Randolph-Habecker J, Hoffstrom BG, Jalikis FG, Koch LK, Berger C, Kosasih PL, Rajan A, Sommermeyer D, et al. Analysis of ROR1 protein expression in human cancer and normal tissues. Clin Cancer Res Off J Am Assoc Cancer Res. 2017;23(12):3061–71.CrossRef Balakrishnan A, Goodpaster T, Randolph-Habecker J, Hoffstrom BG, Jalikis FG, Koch LK, Berger C, Kosasih PL, Rajan A, Sommermeyer D, et al. Analysis of ROR1 protein expression in human cancer and normal tissues. Clin Cancer Res Off J Am Assoc Cancer Res. 2017;23(12):3061–71.CrossRef
4.
go back to reference Keenan TE, Tolaney SM. Role of immunotherapy in triple-negative breast cancer. J Natl Compr Canc Netw. 2020;18(4):479–89.CrossRef Keenan TE, Tolaney SM. Role of immunotherapy in triple-negative breast cancer. J Natl Compr Canc Netw. 2020;18(4):479–89.CrossRef
5.
go back to reference Wei J, Luo C, Wang Y, Guo Y, Dai H, Tong C, Ti D, Wu Z, Han W. PD-1 silencing impairs the anti-tumor function of chimeric antigen receptor modified T cells by inhibiting proliferation activity. J Immunother Cancer. 2019;7(1):209.CrossRef Wei J, Luo C, Wang Y, Guo Y, Dai H, Tong C, Ti D, Wu Z, Han W. PD-1 silencing impairs the anti-tumor function of chimeric antigen receptor modified T cells by inhibiting proliferation activity. J Immunother Cancer. 2019;7(1):209.CrossRef
6.
go back to reference Grosser R, Cherkassky L, Chintala N, Adusumilli PS. Combination immunotherapy with CAR T cells and checkpoint blockade for the treatment of solid tumors. Cancer Cell. 2019;36(5):471–82.CrossRef Grosser R, Cherkassky L, Chintala N, Adusumilli PS. Combination immunotherapy with CAR T cells and checkpoint blockade for the treatment of solid tumors. Cancer Cell. 2019;36(5):471–82.CrossRef
7.
go back to reference Rafiq S, Yeku OO, Jackson HJ, Purdon TJ, van Leeuwen DG, Drakes DJ, Song M, Miele MM, Li Z, Wang P, et al. Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo. Nat Biotechnol. 2018;36(9):847–56.CrossRef Rafiq S, Yeku OO, Jackson HJ, Purdon TJ, van Leeuwen DG, Drakes DJ, Song M, Miele MM, Li Z, Wang P, et al. Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo. Nat Biotechnol. 2018;36(9):847–56.CrossRef
8.
go back to reference Gohil SH, Paredes-Moscosso SR, Harrasser M, Vezzalini M, Scarpa A, Morris E, Davidoff AM, Sorio C, Nathwani AC, Della Peruta M. An ROR1 bi-specific T-cell engager provides effective targeting and cytotoxicity against a range of solid tumors. Oncoimmunology. 2017;6(7):e1326437.CrossRef Gohil SH, Paredes-Moscosso SR, Harrasser M, Vezzalini M, Scarpa A, Morris E, Davidoff AM, Sorio C, Nathwani AC, Della Peruta M. An ROR1 bi-specific T-cell engager provides effective targeting and cytotoxicity against a range of solid tumors. Oncoimmunology. 2017;6(7):e1326437.CrossRef
9.
go back to reference Gohil SH, Della Peruta M, Paredes-Moscosso SR, Harrasser M, Cheung GWK, Davies DM, Pule MA, Nathwani AC. Novel humanised ROR1 chimeric antigen receptors for the treatment of haematological malignancies. Am Soc Hematol. 2016;128:3361. Gohil SH, Della Peruta M, Paredes-Moscosso SR, Harrasser M, Cheung GWK, Davies DM, Pule MA, Nathwani AC. Novel humanised ROR1 chimeric antigen receptors for the treatment of haematological malignancies. Am Soc Hematol. 2016;128:3361.
10.
go back to reference Hooijberg E, Bakker AQ, Ruizendaal JJ, Spits HJB. NFAT-controlled expression of GFP permits visualization and isolation of antigen-stimulated primary human T cells. J Am Soc Hematol. 2000;96(2):459–66. Hooijberg E, Bakker AQ, Ruizendaal JJ, Spits HJB. NFAT-controlled expression of GFP permits visualization and isolation of antigen-stimulated primary human T cells. J Am Soc Hematol. 2000;96(2):459–66.
11.
go back to reference Specht JM. (2018) Phase I study of immunotherapy for advanced ROR1+ malignancies with autologous ROR1-specific chimeric antigen receptor-modified (CAR)-T cells. J Clin Oncol. 2018;36:TPS79–TPS79.CrossRef Specht JM. (2018) Phase I study of immunotherapy for advanced ROR1+ malignancies with autologous ROR1-specific chimeric antigen receptor-modified (CAR)-T cells. J Clin Oncol. 2018;36:TPS79–TPS79.CrossRef
12.
go back to reference Nanda R, Chow LQ, Dees EC, Berger R, Gupta S, Geva R, Pusztai L, Pathiraja K, Aktan G, Cheng JD, et al. Pembrolizumab in patients with advanced triple-negative breast cancer: phase Ib KEYNOTE-012 study. J Clin Oncol Off J Am Soc Clin Oncol. 2016;34(21):2460–7.CrossRef Nanda R, Chow LQ, Dees EC, Berger R, Gupta S, Geva R, Pusztai L, Pathiraja K, Aktan G, Cheng JD, et al. Pembrolizumab in patients with advanced triple-negative breast cancer: phase Ib KEYNOTE-012 study. J Clin Oncol Off J Am Soc Clin Oncol. 2016;34(21):2460–7.CrossRef
13.
go back to reference Arlauckas SP, Garris CS, Kohler RH, Kitaoka M, Cuccarese MF, Yang KS, Miller MA, Carlson JC, Freeman GJ, Anthony RM, et al. In vivo imaging reveals a tumor-associated macrophage-mediated resistance pathway in anti-PD-1 therapy. Sci Transl Med. 2017;9:389.CrossRef Arlauckas SP, Garris CS, Kohler RH, Kitaoka M, Cuccarese MF, Yang KS, Miller MA, Carlson JC, Freeman GJ, Anthony RM, et al. In vivo imaging reveals a tumor-associated macrophage-mediated resistance pathway in anti-PD-1 therapy. Sci Transl Med. 2017;9:389.CrossRef
Metadata
Title
Inducible localized delivery of an anti-PD-1 scFv enhances anti-tumor activity of ROR1 CAR-T cells in TNBC
Authors
Micaela Harrasser
Satyen Harish Gohil
Hiu Lau
Marco Della Peruta
Vincent Muczynski
Dominic Patel
Elena Miranda
Kristiana Grigoriadis
Anita Grigoriadis
David Granger
Rachel Evans
Amit Chunilal Nathwani
Publication date
01-12-2022
Publisher
BioMed Central
Keyword
Solid Tumor
Published in
Breast Cancer Research / Issue 1/2022
Electronic ISSN: 1465-542X
DOI
https://doi.org/10.1186/s13058-022-01531-1

Other articles of this Issue 1/2022

Breast Cancer Research 1/2022 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