Cargando…

Antitumor effects of NK cells expanded by activation pre‑processing of autologous feeder cells before irradiation in colorectal cancer

Natural killer (NK) cells play a crucial role in early immune defenses against transformed cells and are used in the therapeutic management of cancer. However, it is difficult to sufficiently obtain high purity activated NK cells for clinical application. The function of NK cells is dependent on the...

Descripción completa

Detalles Bibliográficos
Autores principales: Koh, Eun-Kyoung, Lee, Hong-Rae, Son, Woo-Chang, Park, Ga-Young, Bae, Jaeho, Park, You-Soo
Formato: Online Artículo Texto
Lenguaje:English
Publicado: D.A. Spandidos 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10157612/
https://www.ncbi.nlm.nih.gov/pubmed/37153058
http://dx.doi.org/10.3892/ol.2023.13818
_version_ 1785036791111221248
author Koh, Eun-Kyoung
Lee, Hong-Rae
Son, Woo-Chang
Park, Ga-Young
Bae, Jaeho
Park, You-Soo
author_facet Koh, Eun-Kyoung
Lee, Hong-Rae
Son, Woo-Chang
Park, Ga-Young
Bae, Jaeho
Park, You-Soo
author_sort Koh, Eun-Kyoung
collection PubMed
description Natural killer (NK) cells play a crucial role in early immune defenses against transformed cells and are used in the therapeutic management of cancer. However, it is difficult to sufficiently obtain high purity activated NK cells for clinical application. The function of NK cells is dependent on the balance of activating and inhibitory signals. Strong and diverse stimuli are required to increase the function of NK cells. Radiotherapy modulates the expression of various immunomodulatory molecules that recruit and activate NK cells. NK cell-mediated antibody-dependent cellular cytotoxicity is one of the most potent cytotoxic effects of NK cells against target cancer cells. To generate activated and irradiated autologous peripheral blood mononuclear cells (PBMCs), cytokine and monoclonal antibody stimulation followed by ionizing radiation was performed in the present study. The expanded NK cells were cultured for 21 days using activated/irradiated autologous PBMCs. Colorectal cancer cells (SW480 and HT-29) were used to analyze the expression of NK group 2D ligands and EGFR by radiation. The cytotoxicity of radiation plus NK cell-based targeted therapy against colorectal cancer cell lines was analyzed using flow cytometry. Activated and irradiated PBMCs exhibited significantly increased expression of various activating ligands that stimulated NK cells. In total, >10,000-fold high-purity activated NK cells were obtained, with negligible T-cell contamination. To confirm the antitumor activity of the NK cells expanded by this method, the expanded NK cells were treated with cetuximab, radiotherapy, or a combination of cetuximab and radiotherapy in the presence of human colorectal cancer cells. Expanded NK cells were effective at targeting human colorectal cancer cells, particularly when combined with cetuximab and radiotherapy. Thus, in the present study, a novel method for high-purity activated NK cell expansion was developed using activated and irradiated PBMCs. In addition, combined radiotherapy and antibody-based immunotherapy with expanded NK cells may be an effective strategy to enhance the efficiency of treatment against colorectal cancer.
format Online
Article
Text
id pubmed-10157612
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher D.A. Spandidos
record_format MEDLINE/PubMed
spelling pubmed-101576122023-05-05 Antitumor effects of NK cells expanded by activation pre‑processing of autologous feeder cells before irradiation in colorectal cancer Koh, Eun-Kyoung Lee, Hong-Rae Son, Woo-Chang Park, Ga-Young Bae, Jaeho Park, You-Soo Oncol Lett Articles Natural killer (NK) cells play a crucial role in early immune defenses against transformed cells and are used in the therapeutic management of cancer. However, it is difficult to sufficiently obtain high purity activated NK cells for clinical application. The function of NK cells is dependent on the balance of activating and inhibitory signals. Strong and diverse stimuli are required to increase the function of NK cells. Radiotherapy modulates the expression of various immunomodulatory molecules that recruit and activate NK cells. NK cell-mediated antibody-dependent cellular cytotoxicity is one of the most potent cytotoxic effects of NK cells against target cancer cells. To generate activated and irradiated autologous peripheral blood mononuclear cells (PBMCs), cytokine and monoclonal antibody stimulation followed by ionizing radiation was performed in the present study. The expanded NK cells were cultured for 21 days using activated/irradiated autologous PBMCs. Colorectal cancer cells (SW480 and HT-29) were used to analyze the expression of NK group 2D ligands and EGFR by radiation. The cytotoxicity of radiation plus NK cell-based targeted therapy against colorectal cancer cell lines was analyzed using flow cytometry. Activated and irradiated PBMCs exhibited significantly increased expression of various activating ligands that stimulated NK cells. In total, >10,000-fold high-purity activated NK cells were obtained, with negligible T-cell contamination. To confirm the antitumor activity of the NK cells expanded by this method, the expanded NK cells were treated with cetuximab, radiotherapy, or a combination of cetuximab and radiotherapy in the presence of human colorectal cancer cells. Expanded NK cells were effective at targeting human colorectal cancer cells, particularly when combined with cetuximab and radiotherapy. Thus, in the present study, a novel method for high-purity activated NK cell expansion was developed using activated and irradiated PBMCs. In addition, combined radiotherapy and antibody-based immunotherapy with expanded NK cells may be an effective strategy to enhance the efficiency of treatment against colorectal cancer. D.A. Spandidos 2023-04-18 /pmc/articles/PMC10157612/ /pubmed/37153058 http://dx.doi.org/10.3892/ol.2023.13818 Text en Copyright: © Koh et al. https://creativecommons.org/licenses/by-nc-nd/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License (https://creativecommons.org/licenses/by-nc-nd/4.0/) , which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.
spellingShingle Articles
Koh, Eun-Kyoung
Lee, Hong-Rae
Son, Woo-Chang
Park, Ga-Young
Bae, Jaeho
Park, You-Soo
Antitumor effects of NK cells expanded by activation pre‑processing of autologous feeder cells before irradiation in colorectal cancer
title Antitumor effects of NK cells expanded by activation pre‑processing of autologous feeder cells before irradiation in colorectal cancer
title_full Antitumor effects of NK cells expanded by activation pre‑processing of autologous feeder cells before irradiation in colorectal cancer
title_fullStr Antitumor effects of NK cells expanded by activation pre‑processing of autologous feeder cells before irradiation in colorectal cancer
title_full_unstemmed Antitumor effects of NK cells expanded by activation pre‑processing of autologous feeder cells before irradiation in colorectal cancer
title_short Antitumor effects of NK cells expanded by activation pre‑processing of autologous feeder cells before irradiation in colorectal cancer
title_sort antitumor effects of nk cells expanded by activation pre‑processing of autologous feeder cells before irradiation in colorectal cancer
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10157612/
https://www.ncbi.nlm.nih.gov/pubmed/37153058
http://dx.doi.org/10.3892/ol.2023.13818
work_keys_str_mv AT koheunkyoung antitumoreffectsofnkcellsexpandedbyactivationpreprocessingofautologousfeedercellsbeforeirradiationincolorectalcancer
AT leehongrae antitumoreffectsofnkcellsexpandedbyactivationpreprocessingofautologousfeedercellsbeforeirradiationincolorectalcancer
AT sonwoochang antitumoreffectsofnkcellsexpandedbyactivationpreprocessingofautologousfeedercellsbeforeirradiationincolorectalcancer
AT parkgayoung antitumoreffectsofnkcellsexpandedbyactivationpreprocessingofautologousfeedercellsbeforeirradiationincolorectalcancer
AT baejaeho antitumoreffectsofnkcellsexpandedbyactivationpreprocessingofautologousfeedercellsbeforeirradiationincolorectalcancer
AT parkyousoo antitumoreffectsofnkcellsexpandedbyactivationpreprocessingofautologousfeedercellsbeforeirradiationincolorectalcancer