Peptide Adjuvant to Invigorate Cytolytic Activity of NK Cells in an Obese Mouse Cancer Model

Cancer patients who are overweight compared to those with normal body weight have obesity-associated alterations of natural killer (NK) cells, characterized by poor cytotoxicity, slow proliferation, and inadequate anti-cancer activity. Concomitantly, prohibitin overexpressed by cancer cells elevates...

Full description

Bibliographic Details
Main Authors: Seungmin Han, Minjin Jung, Angela S. Kim, Daniel Y. Lee, Byung-Hyun Cha, Charles W. Putnam, Kwang Suk Lim, David A. Bull, Young-Wook Won
Format: Article
Language:English
Published: MDPI AG 2021-08-01
Series:Pharmaceutics
Subjects:
Online Access:https://www.mdpi.com/1999-4923/13/8/1279
_version_ 1797522345246588928
author Seungmin Han
Minjin Jung
Angela S. Kim
Daniel Y. Lee
Byung-Hyun Cha
Charles W. Putnam
Kwang Suk Lim
David A. Bull
Young-Wook Won
author_facet Seungmin Han
Minjin Jung
Angela S. Kim
Daniel Y. Lee
Byung-Hyun Cha
Charles W. Putnam
Kwang Suk Lim
David A. Bull
Young-Wook Won
author_sort Seungmin Han
collection DOAJ
description Cancer patients who are overweight compared to those with normal body weight have obesity-associated alterations of natural killer (NK) cells, characterized by poor cytotoxicity, slow proliferation, and inadequate anti-cancer activity. Concomitantly, prohibitin overexpressed by cancer cells elevates glucose metabolism, rendering the tumor microenvironment (TME) more tumor-favorable, and leading to malfunction of immune cells present in the TME. These changes cause vicious cycles of tumor growth. Adoptive immunotherapy has emerged as a promising option for cancer patients; however, obesity-related alterations in the TME allow the tumor to bypass immune surveillance and to down-regulate the activity of adoptively transferred NK cells. We hypothesized that inhibiting the prohibitin signaling pathway in an obese model would reduce glucose metabolism of cancer cells, thereby changing the TME to a pro-immune microenvironment and restoring the cytolytic activity of NK cells. Priming tumor cells with an inhibitory the prohibitin-binding peptide (PBP) enhances cytokine secretion and augments the cytolytic activity of adoptively transferred NK cells. NK cells harvested from the PBP-primed tumors exhibit multiple markers associated with the effector function of active NK cells. Our findings suggest that PBP has the potential as an adjuvant to enhance the cytolytic activity of adoptively transferred NK cells in cancer patients with obesity.
first_indexed 2024-03-10T08:28:06Z
format Article
id doaj.art-324329cacd0147d89a475316c5a68eaf
institution Directory Open Access Journal
issn 1999-4923
language English
last_indexed 2024-03-10T08:28:06Z
publishDate 2021-08-01
publisher MDPI AG
record_format Article
series Pharmaceutics
spelling doaj.art-324329cacd0147d89a475316c5a68eaf2023-11-22T09:15:24ZengMDPI AGPharmaceutics1999-49232021-08-01138127910.3390/pharmaceutics13081279Peptide Adjuvant to Invigorate Cytolytic Activity of NK Cells in an Obese Mouse Cancer ModelSeungmin Han0Minjin Jung1Angela S. Kim2Daniel Y. Lee3Byung-Hyun Cha4Charles W. Putnam5Kwang Suk Lim6David A. Bull7Young-Wook Won8Division of Cardiothoracic Surgery, Department of Surgery, University of Arizona College of Medicine—Tucson, Tucson, AZ 85724, USADivision of Cardiothoracic Surgery, Department of Surgery, University of Arizona College of Medicine—Tucson, Tucson, AZ 85724, USADivision of Cardiothoracic Surgery, Department of Surgery, University of Arizona College of Medicine—Tucson, Tucson, AZ 85724, USADivision of Cardiothoracic Surgery, Department of Surgery, University of Arizona College of Medicine—Tucson, Tucson, AZ 85724, USADivision of Cardiothoracic Surgery, Department of Surgery, University of Arizona College of Medicine—Tucson, Tucson, AZ 85724, USADivision of Cardiothoracic Surgery, Department of Surgery, University of Arizona College of Medicine—Tucson, Tucson, AZ 85724, USAInterdisciplinary Program in Biohealth-Machinery Convergence Engineering, Department of Biotechnology and Bioengineering, College of Art, Culture and Engineering, Kangwon National University, Chuncheon 24341, KoreaDivision of Cardiothoracic Surgery, Department of Surgery, University of Arizona College of Medicine—Tucson, Tucson, AZ 85724, USADivision of Cardiothoracic Surgery, Department of Surgery, University of Arizona College of Medicine—Tucson, Tucson, AZ 85724, USACancer patients who are overweight compared to those with normal body weight have obesity-associated alterations of natural killer (NK) cells, characterized by poor cytotoxicity, slow proliferation, and inadequate anti-cancer activity. Concomitantly, prohibitin overexpressed by cancer cells elevates glucose metabolism, rendering the tumor microenvironment (TME) more tumor-favorable, and leading to malfunction of immune cells present in the TME. These changes cause vicious cycles of tumor growth. Adoptive immunotherapy has emerged as a promising option for cancer patients; however, obesity-related alterations in the TME allow the tumor to bypass immune surveillance and to down-regulate the activity of adoptively transferred NK cells. We hypothesized that inhibiting the prohibitin signaling pathway in an obese model would reduce glucose metabolism of cancer cells, thereby changing the TME to a pro-immune microenvironment and restoring the cytolytic activity of NK cells. Priming tumor cells with an inhibitory the prohibitin-binding peptide (PBP) enhances cytokine secretion and augments the cytolytic activity of adoptively transferred NK cells. NK cells harvested from the PBP-primed tumors exhibit multiple markers associated with the effector function of active NK cells. Our findings suggest that PBP has the potential as an adjuvant to enhance the cytolytic activity of adoptively transferred NK cells in cancer patients with obesity.https://www.mdpi.com/1999-4923/13/8/1279prohibitin binding peptideNK cellimmunotherapyobesitytumor microenvironment
spellingShingle Seungmin Han
Minjin Jung
Angela S. Kim
Daniel Y. Lee
Byung-Hyun Cha
Charles W. Putnam
Kwang Suk Lim
David A. Bull
Young-Wook Won
Peptide Adjuvant to Invigorate Cytolytic Activity of NK Cells in an Obese Mouse Cancer Model
Pharmaceutics
prohibitin binding peptide
NK cell
immunotherapy
obesity
tumor microenvironment
title Peptide Adjuvant to Invigorate Cytolytic Activity of NK Cells in an Obese Mouse Cancer Model
title_full Peptide Adjuvant to Invigorate Cytolytic Activity of NK Cells in an Obese Mouse Cancer Model
title_fullStr Peptide Adjuvant to Invigorate Cytolytic Activity of NK Cells in an Obese Mouse Cancer Model
title_full_unstemmed Peptide Adjuvant to Invigorate Cytolytic Activity of NK Cells in an Obese Mouse Cancer Model
title_short Peptide Adjuvant to Invigorate Cytolytic Activity of NK Cells in an Obese Mouse Cancer Model
title_sort peptide adjuvant to invigorate cytolytic activity of nk cells in an obese mouse cancer model
topic prohibitin binding peptide
NK cell
immunotherapy
obesity
tumor microenvironment
url https://www.mdpi.com/1999-4923/13/8/1279
work_keys_str_mv AT seungminhan peptideadjuvanttoinvigoratecytolyticactivityofnkcellsinanobesemousecancermodel
AT minjinjung peptideadjuvanttoinvigoratecytolyticactivityofnkcellsinanobesemousecancermodel
AT angelaskim peptideadjuvanttoinvigoratecytolyticactivityofnkcellsinanobesemousecancermodel
AT danielylee peptideadjuvanttoinvigoratecytolyticactivityofnkcellsinanobesemousecancermodel
AT byunghyuncha peptideadjuvanttoinvigoratecytolyticactivityofnkcellsinanobesemousecancermodel
AT charleswputnam peptideadjuvanttoinvigoratecytolyticactivityofnkcellsinanobesemousecancermodel
AT kwangsuklim peptideadjuvanttoinvigoratecytolyticactivityofnkcellsinanobesemousecancermodel
AT davidabull peptideadjuvanttoinvigoratecytolyticactivityofnkcellsinanobesemousecancermodel
AT youngwookwon peptideadjuvanttoinvigoratecytolyticactivityofnkcellsinanobesemousecancermodel