Cargando…

AntiCD3Fv fused to human interleukin-3 deletion variant redirected T cells against human acute myeloid leukemic stem cells

BACKGROUND: Leukemic stem cells (LSCs) are frequently seen as a cause of treatment failure and relapse in patients with acute myeloid leukemia (AML). Thus, successful new therapeutic strategies for the treatment of AML should aim at eradicating LSCs. The identification of targets on the cell surface...

Descripción completa

Detalles Bibliográficos
Autores principales: Fan, Dongmei, Li, Zhenzhen, Zhang, Xiaolong, Yang, Yuqi, Yuan, Xiangfei, Zhang, Xiuli, Yang, Ming, Zhang, Yizhi, Xiong, Dongsheng
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2015
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4389834/
https://www.ncbi.nlm.nih.gov/pubmed/25879549
http://dx.doi.org/10.1186/s13045-015-0109-5
_version_ 1782365620869791744
author Fan, Dongmei
Li, Zhenzhen
Zhang, Xiaolong
Yang, Yuqi
Yuan, Xiangfei
Zhang, Xiuli
Yang, Ming
Zhang, Yizhi
Xiong, Dongsheng
author_facet Fan, Dongmei
Li, Zhenzhen
Zhang, Xiaolong
Yang, Yuqi
Yuan, Xiangfei
Zhang, Xiuli
Yang, Ming
Zhang, Yizhi
Xiong, Dongsheng
author_sort Fan, Dongmei
collection PubMed
description BACKGROUND: Leukemic stem cells (LSCs) are frequently seen as a cause of treatment failure and relapse in patients with acute myeloid leukemia (AML). Thus, successful new therapeutic strategies for the treatment of AML should aim at eradicating LSCs. The identification of targets on the cell surface of LSCs is getting more and more attention. Among these, CD123, also known as the interleukin-3 (IL3)-receptor α chain, has been identified as a potential immunotherapeutic target due to its overexpression on LSCs in AML as well as on AML blasts, rather than normal hematopoietic stem cells. METHODS: We constructed a CD123-targeted fusion protein antiCD3Fv-⊿IL3, with one binding site for T cell antigen receptor (TCRCD3) and the other for CD123, by recombinant gene-engineering technology. Cysteine residues were introduced into the V domains of the antiCD3Fv segment to enhance its stability by locking the two chains of Fv together with disulfide covalent bonds. The stability and cytotoxicity of the two fusion proteins were detected in vitro and in vivo. RESULTS: Both fusion proteins were produced and purified from Escherichia coli 16C9 cells with excellent yields in fully active forms. High-binding capability was observed between these two fusion proteins and human IL3R, leading to the specific lysis of CD123-expressing cell lines KG1a; also, mononuclear cells from primary AML patients were inhibited in a colony forming assay in vitro, presumably by redirecting T lymphocytes in vitro. In addition, they displayed an antileukemic activity against KG1a xenografts in non-obese diabetic/severe combined immunodeficient (NOD/SCID) mice, especially disulfide-stabilized (ds)-antiCD3Fv-⊿IL3 for its improved stability. CONCLUSIONS: These results suggest that both fusion proteins display the antileukemic activity against CD123-expressing cell lines as well as leukemic progenitors in vitro and in vivo, especially ds-antiCD3Fv-⊿IL3. They could be the promising candidates for future immunotherapy of AML. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/s13045-015-0109-5) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-4389834
institution National Center for Biotechnology Information
language English
publishDate 2015
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-43898342015-04-09 AntiCD3Fv fused to human interleukin-3 deletion variant redirected T cells against human acute myeloid leukemic stem cells Fan, Dongmei Li, Zhenzhen Zhang, Xiaolong Yang, Yuqi Yuan, Xiangfei Zhang, Xiuli Yang, Ming Zhang, Yizhi Xiong, Dongsheng J Hematol Oncol Research BACKGROUND: Leukemic stem cells (LSCs) are frequently seen as a cause of treatment failure and relapse in patients with acute myeloid leukemia (AML). Thus, successful new therapeutic strategies for the treatment of AML should aim at eradicating LSCs. The identification of targets on the cell surface of LSCs is getting more and more attention. Among these, CD123, also known as the interleukin-3 (IL3)-receptor α chain, has been identified as a potential immunotherapeutic target due to its overexpression on LSCs in AML as well as on AML blasts, rather than normal hematopoietic stem cells. METHODS: We constructed a CD123-targeted fusion protein antiCD3Fv-⊿IL3, with one binding site for T cell antigen receptor (TCRCD3) and the other for CD123, by recombinant gene-engineering technology. Cysteine residues were introduced into the V domains of the antiCD3Fv segment to enhance its stability by locking the two chains of Fv together with disulfide covalent bonds. The stability and cytotoxicity of the two fusion proteins were detected in vitro and in vivo. RESULTS: Both fusion proteins were produced and purified from Escherichia coli 16C9 cells with excellent yields in fully active forms. High-binding capability was observed between these two fusion proteins and human IL3R, leading to the specific lysis of CD123-expressing cell lines KG1a; also, mononuclear cells from primary AML patients were inhibited in a colony forming assay in vitro, presumably by redirecting T lymphocytes in vitro. In addition, they displayed an antileukemic activity against KG1a xenografts in non-obese diabetic/severe combined immunodeficient (NOD/SCID) mice, especially disulfide-stabilized (ds)-antiCD3Fv-⊿IL3 for its improved stability. CONCLUSIONS: These results suggest that both fusion proteins display the antileukemic activity against CD123-expressing cell lines as well as leukemic progenitors in vitro and in vivo, especially ds-antiCD3Fv-⊿IL3. They could be the promising candidates for future immunotherapy of AML. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/s13045-015-0109-5) contains supplementary material, which is available to authorized users. BioMed Central 2015-02-28 /pmc/articles/PMC4389834/ /pubmed/25879549 http://dx.doi.org/10.1186/s13045-015-0109-5 Text en © Dongmei et al.; licensee BioMed Central. 2015 This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research
Fan, Dongmei
Li, Zhenzhen
Zhang, Xiaolong
Yang, Yuqi
Yuan, Xiangfei
Zhang, Xiuli
Yang, Ming
Zhang, Yizhi
Xiong, Dongsheng
AntiCD3Fv fused to human interleukin-3 deletion variant redirected T cells against human acute myeloid leukemic stem cells
title AntiCD3Fv fused to human interleukin-3 deletion variant redirected T cells against human acute myeloid leukemic stem cells
title_full AntiCD3Fv fused to human interleukin-3 deletion variant redirected T cells against human acute myeloid leukemic stem cells
title_fullStr AntiCD3Fv fused to human interleukin-3 deletion variant redirected T cells against human acute myeloid leukemic stem cells
title_full_unstemmed AntiCD3Fv fused to human interleukin-3 deletion variant redirected T cells against human acute myeloid leukemic stem cells
title_short AntiCD3Fv fused to human interleukin-3 deletion variant redirected T cells against human acute myeloid leukemic stem cells
title_sort anticd3fv fused to human interleukin-3 deletion variant redirected t cells against human acute myeloid leukemic stem cells
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4389834/
https://www.ncbi.nlm.nih.gov/pubmed/25879549
http://dx.doi.org/10.1186/s13045-015-0109-5
work_keys_str_mv AT fandongmei anticd3fvfusedtohumaninterleukin3deletionvariantredirectedtcellsagainsthumanacutemyeloidleukemicstemcells
AT lizhenzhen anticd3fvfusedtohumaninterleukin3deletionvariantredirectedtcellsagainsthumanacutemyeloidleukemicstemcells
AT zhangxiaolong anticd3fvfusedtohumaninterleukin3deletionvariantredirectedtcellsagainsthumanacutemyeloidleukemicstemcells
AT yangyuqi anticd3fvfusedtohumaninterleukin3deletionvariantredirectedtcellsagainsthumanacutemyeloidleukemicstemcells
AT yuanxiangfei anticd3fvfusedtohumaninterleukin3deletionvariantredirectedtcellsagainsthumanacutemyeloidleukemicstemcells
AT zhangxiuli anticd3fvfusedtohumaninterleukin3deletionvariantredirectedtcellsagainsthumanacutemyeloidleukemicstemcells
AT yangming anticd3fvfusedtohumaninterleukin3deletionvariantredirectedtcellsagainsthumanacutemyeloidleukemicstemcells
AT zhangyizhi anticd3fvfusedtohumaninterleukin3deletionvariantredirectedtcellsagainsthumanacutemyeloidleukemicstemcells
AT xiongdongsheng anticd3fvfusedtohumaninterleukin3deletionvariantredirectedtcellsagainsthumanacutemyeloidleukemicstemcells