Efficient elimination of primary B-ALL cells in vitro and in vivo using a novel 4-1BB-based CAR targeting a membrane-distal CD22 epitope. Velasco-Hernandez, T., Zanetti, S. R., Roca-Ho, H., Gutierrez-Aguera, F., Petazzi, P., Sánchez-Martínez, D., Molina, O., Baroni, M. L., Fuster, J. L., Ballerini, P., Bueno, C., Fernandez-Fuentes, N., Engel, P., & Menendez, P. Journal for ImmunoTherapy of Cancer, 8(2):e000896, August, 2020. Publisher: BMJ Specialist Journals Section: Immune cell therapies and immune cell engineering
Efficient elimination of primary B-ALL cells in vitro and in vivo using a novel 4-1BB-based CAR targeting a membrane-distal CD22 epitope [link]Paper  doi  abstract   bibtex   
Background There are few therapeutic options available for patients with B-cell acute lymphoblastic leukemia (B-ALL) relapsing as CD19– either after chemotherapy or CD19-targeted immunotherapies. CD22-chimeric antigen receptor (CAR) T cells represent an attractive addition to CD19-CAR T cell therapy because they will target both CD22+CD19– B-ALL relapses and CD19– preleukemic cells. However, the immune escape mechanisms from CD22-CAR T cells, and the potential contribution of the epitope binding of the anti-CD22 single-chain variable fragment (scFv) remain understudied. Methods Here, we have developed and comprehensively characterized a novel CD22-CAR (clone hCD22.7) targeting a membrane-distal CD22 epitope and tested its cytotoxic effects against B-ALL cells both in in vitro and in vivo assays. Results Conformational epitope mapping, cross-blocking, and molecular docking assays revealed that the hCD22.7 scFv is a high-affinity binding antibody which specifically binds to the ESTKDGKVP sequence, located in the Ig-like V-type domain, the most distal domain of CD22. We observed efficient killing of B-ALL cells in vitro, although the kinetics were dependent on the level of CD22 expression. Importantly, we show an efficient in vivo control of patients with B-ALL derived xenografts with diverse aggressiveness, coupled to long-term hCD22.7-CAR T cell persistence. Remaining leukemic cells at sacrifice maintained full expression of CD22, ruling out CAR pressure-mediated antigen loss. Finally, the immunogenicity capacity of this hCD22.7-scFv was very similar to that of other CD22 scFv previously used in adoptive T cell therapy. Conclusions We report a novel, high-affinity hCD22.7 scFv which targets a membrane-distal epitope of CD22. 4-1BB-based hCD22.7-CAR T cells efficiently eliminate clinically relevant B- CD22high and CD22low ALL primary samples in vitro and in vivo. Our study supports the clinical translation of this hCD22.7-CAR as either single or tandem CD22–CD19-CAR for both naive and anti-CD19-resistant patients with B-ALL.
@article{velasco-hernandez_efficient_2020,
	title = {Efficient elimination of primary {B}-{ALL} cells in vitro and in vivo using a novel 4-{1BB}-based {CAR} targeting a membrane-distal {CD22} epitope},
	volume = {8},
	copyright = {© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.. http://creativecommons.org/licenses/by-nc/4.0/This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See http://creativecommons.org/licenses/by-nc/4.0/.},
	issn = {2051-1426},
	url = {https://jitc.bmj.com/content/8/2/e000896},
	doi = {10.1136/jitc-2020-000896},
	abstract = {Background There are few therapeutic options available for patients with B-cell acute lymphoblastic leukemia (B-ALL) relapsing as CD19– either after chemotherapy or CD19-targeted immunotherapies. CD22-chimeric antigen receptor (CAR) T cells represent an attractive addition to CD19-CAR T cell therapy because they will target both CD22+CD19– B-ALL relapses and CD19– preleukemic cells. However, the immune escape mechanisms from CD22-CAR T cells, and the potential contribution of the epitope binding of the anti-CD22 single-chain variable fragment (scFv) remain understudied.
Methods Here, we have developed and comprehensively characterized a novel CD22-CAR (clone hCD22.7) targeting a membrane-distal CD22 epitope and tested its cytotoxic effects against B-ALL cells both in in vitro and in vivo assays.
Results Conformational epitope mapping, cross-blocking, and molecular docking assays revealed that the hCD22.7 scFv is a high-affinity binding antibody which specifically binds to the ESTKDGKVP sequence, located in the Ig-like V-type domain, the most distal domain of CD22. We observed efficient killing of B-ALL cells in vitro, although the kinetics were dependent on the level of CD22 expression. Importantly, we show an efficient in vivo control of patients with B-ALL derived xenografts with diverse aggressiveness, coupled to long-term hCD22.7-CAR T cell persistence. Remaining leukemic cells at sacrifice maintained full expression of CD22, ruling out CAR pressure-mediated antigen loss. Finally, the immunogenicity capacity of this hCD22.7-scFv was very similar to that of other CD22 scFv previously used in adoptive T cell therapy.
Conclusions We report a novel, high-affinity hCD22.7 scFv which targets a membrane-distal epitope of CD22. 4-1BB-based hCD22.7-CAR T cells efficiently eliminate clinically relevant B- CD22high and CD22low ALL primary samples in vitro and in vivo. Our study supports the clinical translation of this hCD22.7-CAR as either single or tandem CD22–CD19-CAR for both naive and anti-CD19-resistant patients with B-ALL.},
	language = {en},
	number = {2},
	urldate = {2020-09-01},
	journal = {Journal for ImmunoTherapy of Cancer},
	author = {Velasco-Hernandez, Talia and Zanetti, Samanta Romina and Roca-Ho, Heleia and Gutierrez-Aguera, Francisco and Petazzi, Paolo and Sánchez-Martínez, Diego and Molina, Oscar and Baroni, Matteo Libero and Fuster, Jose Luis and Ballerini, Paola and Bueno, Clara and Fernandez-Fuentes, Narcis and Engel, Pablo and Menendez, Pablo},
	month = aug,
	year = {2020},
	pmid = {32788237},
	note = {Publisher: BMJ Specialist Journals
Section: Immune cell therapies and immune cell engineering},
	keywords = {Application - Antibody Validation / Epitope Mapping, Application - Cancer Research, Country - France, Country - Spain, Human, PEPperCHIP - Customized - Cyclic constrained, PEPperMAP - Epitope Mapping - Conformational, Sample Type - Monoclonal/polyclonal antibody},
	pages = {e000896},
}

Downloads: 0