Dendritic Cell-Derived Artificial Microvesicles Inhibit RLS<sub>40</sub> Lymphosarcoma Growth in Mice via Stimulation of Th1/Th17 Immune Response

Cell-free antitumor vaccines represent a promising approach to immunotherapy of cancer. Here, we compare the antitumor potential of cell-free vaccines based on microvesicles derived from dendritic cells (DCs) with DC- and cationic-liposome-based vaccines using a murine model of drug-resistant lympho...

Full description

Bibliographic Details
Main Authors: Oleg V. Markov, Aleksandra V. Sen’kova, Islam S. Mohamed, Elena V. Shmendel, Mikhail A. Maslov, Anastasiya L. Oshchepkova, Evgeniy V. Brenner, Nadezhda L. Mironova, Marina A. Zenkova
Format: Article
Language:English
Published: MDPI AG 2022-11-01
Series:Pharmaceutics
Subjects:
Online Access:https://www.mdpi.com/1999-4923/14/11/2542
_version_ 1797464257031307264
author Oleg V. Markov
Aleksandra V. Sen’kova
Islam S. Mohamed
Elena V. Shmendel
Mikhail A. Maslov
Anastasiya L. Oshchepkova
Evgeniy V. Brenner
Nadezhda L. Mironova
Marina A. Zenkova
author_facet Oleg V. Markov
Aleksandra V. Sen’kova
Islam S. Mohamed
Elena V. Shmendel
Mikhail A. Maslov
Anastasiya L. Oshchepkova
Evgeniy V. Brenner
Nadezhda L. Mironova
Marina A. Zenkova
author_sort Oleg V. Markov
collection DOAJ
description Cell-free antitumor vaccines represent a promising approach to immunotherapy of cancer. Here, we compare the antitumor potential of cell-free vaccines based on microvesicles derived from dendritic cells (DCs) with DC- and cationic-liposome-based vaccines using a murine model of drug-resistant lymphosarcoma RLS<sub>40</sub> in vivo. The vaccines were the following: microvesicle vaccines—cytochalasin B-induced membrane vesicles (CIMVs) obtained from DCs loaded with total tumor RNA using cholesterol/spermine-containing cationic liposomes L or mannosylated liposomes ML; DC vaccines—murine DCs loaded with total tumor-derived RNA using the same liposomes; and liposomal vaccines—lipoplexes of total tumor-derived RNA with liposomes L or ML. Being non-hepatotoxic, CIMV- and DC-based vaccines administered subcutaneously exhibited comparable potential to stimulate highly efficient antitumor CTLs in vivo, whereas liposomal vaccines were 25% weaker CTL inducers. Nevertheless, the antitumor efficiencies of the different types of the vaccines were similar: sizes of tumor nodes and the number of liver metastases were significantly decreased, regardless of the vaccine type. Notably, the booster vaccination did not improve the overall antitumor efficacy of the vaccines under the study. CIMV- and DC- based vaccines more efficiently than liposome-based ones decreased mitotic activity of tumor cells and induced their apoptosis, stimulated accumulation of neutrophil inflammatory infiltration in tumor tissue, and had a more pronounced immunomodulatory activity toward the spleen and thymus. Administration of CIMV-, DC-, and liposome-based vaccines resulted in activation of Th1/Th17 cells as well as the induction of positive immune checkpoint 4-1BBL and downregulation of suppressive immune checkpoints in a raw PD-1 >>> TIGIT > CTLA4 > TIM3. We demonstrated that cell-free CIMV-based vaccines exhibited superior antitumor and antimetastatic activity in a tumor model in vivo. The obtained results can be considered as the basis for developing novel strategies for oncoimmunotherapy.
first_indexed 2024-03-09T18:04:28Z
format Article
id doaj.art-eb3ad4ee0c254b07bea709de01437fb6
institution Directory Open Access Journal
issn 1999-4923
language English
last_indexed 2024-03-09T18:04:28Z
publishDate 2022-11-01
publisher MDPI AG
record_format Article
series Pharmaceutics
spelling doaj.art-eb3ad4ee0c254b07bea709de01437fb62023-11-24T09:37:30ZengMDPI AGPharmaceutics1999-49232022-11-011411254210.3390/pharmaceutics14112542Dendritic Cell-Derived Artificial Microvesicles Inhibit RLS<sub>40</sub> Lymphosarcoma Growth in Mice via Stimulation of Th1/Th17 Immune ResponseOleg V. Markov0Aleksandra V. Sen’kova1Islam S. Mohamed2Elena V. Shmendel3Mikhail A. Maslov4Anastasiya L. Oshchepkova5Evgeniy V. Brenner6Nadezhda L. Mironova7Marina A. Zenkova8Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentieva Ave. 8, 630090 Novosibirsk, RussiaInstitute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentieva Ave. 8, 630090 Novosibirsk, RussiaInstitute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentieva Ave. 8, 630090 Novosibirsk, RussiaM.V. Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Vernadskogo Ave. 86, 119571 Moscow, RussiaM.V. Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Vernadskogo Ave. 86, 119571 Moscow, RussiaInstitute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentieva Ave. 8, 630090 Novosibirsk, RussiaInstitute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentieva Ave. 8, 630090 Novosibirsk, RussiaInstitute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentieva Ave. 8, 630090 Novosibirsk, RussiaInstitute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentieva Ave. 8, 630090 Novosibirsk, RussiaCell-free antitumor vaccines represent a promising approach to immunotherapy of cancer. Here, we compare the antitumor potential of cell-free vaccines based on microvesicles derived from dendritic cells (DCs) with DC- and cationic-liposome-based vaccines using a murine model of drug-resistant lymphosarcoma RLS<sub>40</sub> in vivo. The vaccines were the following: microvesicle vaccines—cytochalasin B-induced membrane vesicles (CIMVs) obtained from DCs loaded with total tumor RNA using cholesterol/spermine-containing cationic liposomes L or mannosylated liposomes ML; DC vaccines—murine DCs loaded with total tumor-derived RNA using the same liposomes; and liposomal vaccines—lipoplexes of total tumor-derived RNA with liposomes L or ML. Being non-hepatotoxic, CIMV- and DC-based vaccines administered subcutaneously exhibited comparable potential to stimulate highly efficient antitumor CTLs in vivo, whereas liposomal vaccines were 25% weaker CTL inducers. Nevertheless, the antitumor efficiencies of the different types of the vaccines were similar: sizes of tumor nodes and the number of liver metastases were significantly decreased, regardless of the vaccine type. Notably, the booster vaccination did not improve the overall antitumor efficacy of the vaccines under the study. CIMV- and DC- based vaccines more efficiently than liposome-based ones decreased mitotic activity of tumor cells and induced their apoptosis, stimulated accumulation of neutrophil inflammatory infiltration in tumor tissue, and had a more pronounced immunomodulatory activity toward the spleen and thymus. Administration of CIMV-, DC-, and liposome-based vaccines resulted in activation of Th1/Th17 cells as well as the induction of positive immune checkpoint 4-1BBL and downregulation of suppressive immune checkpoints in a raw PD-1 >>> TIGIT > CTLA4 > TIM3. We demonstrated that cell-free CIMV-based vaccines exhibited superior antitumor and antimetastatic activity in a tumor model in vivo. The obtained results can be considered as the basis for developing novel strategies for oncoimmunotherapy.https://www.mdpi.com/1999-4923/14/11/2542dendritic cellscytochalasin B-induced membrane vesiclescationic liposomesmurine lymphosarcomaimmune checkpointsantitumor vaccines
spellingShingle Oleg V. Markov
Aleksandra V. Sen’kova
Islam S. Mohamed
Elena V. Shmendel
Mikhail A. Maslov
Anastasiya L. Oshchepkova
Evgeniy V. Brenner
Nadezhda L. Mironova
Marina A. Zenkova
Dendritic Cell-Derived Artificial Microvesicles Inhibit RLS<sub>40</sub> Lymphosarcoma Growth in Mice via Stimulation of Th1/Th17 Immune Response
Pharmaceutics
dendritic cells
cytochalasin B-induced membrane vesicles
cationic liposomes
murine lymphosarcoma
immune checkpoints
antitumor vaccines
title Dendritic Cell-Derived Artificial Microvesicles Inhibit RLS<sub>40</sub> Lymphosarcoma Growth in Mice via Stimulation of Th1/Th17 Immune Response
title_full Dendritic Cell-Derived Artificial Microvesicles Inhibit RLS<sub>40</sub> Lymphosarcoma Growth in Mice via Stimulation of Th1/Th17 Immune Response
title_fullStr Dendritic Cell-Derived Artificial Microvesicles Inhibit RLS<sub>40</sub> Lymphosarcoma Growth in Mice via Stimulation of Th1/Th17 Immune Response
title_full_unstemmed Dendritic Cell-Derived Artificial Microvesicles Inhibit RLS<sub>40</sub> Lymphosarcoma Growth in Mice via Stimulation of Th1/Th17 Immune Response
title_short Dendritic Cell-Derived Artificial Microvesicles Inhibit RLS<sub>40</sub> Lymphosarcoma Growth in Mice via Stimulation of Th1/Th17 Immune Response
title_sort dendritic cell derived artificial microvesicles inhibit rls sub 40 sub lymphosarcoma growth in mice via stimulation of th1 th17 immune response
topic dendritic cells
cytochalasin B-induced membrane vesicles
cationic liposomes
murine lymphosarcoma
immune checkpoints
antitumor vaccines
url https://www.mdpi.com/1999-4923/14/11/2542
work_keys_str_mv AT olegvmarkov dendriticcellderivedartificialmicrovesiclesinhibitrlssub40sublymphosarcomagrowthinmiceviastimulationofth1th17immuneresponse
AT aleksandravsenkova dendriticcellderivedartificialmicrovesiclesinhibitrlssub40sublymphosarcomagrowthinmiceviastimulationofth1th17immuneresponse
AT islamsmohamed dendriticcellderivedartificialmicrovesiclesinhibitrlssub40sublymphosarcomagrowthinmiceviastimulationofth1th17immuneresponse
AT elenavshmendel dendriticcellderivedartificialmicrovesiclesinhibitrlssub40sublymphosarcomagrowthinmiceviastimulationofth1th17immuneresponse
AT mikhailamaslov dendriticcellderivedartificialmicrovesiclesinhibitrlssub40sublymphosarcomagrowthinmiceviastimulationofth1th17immuneresponse
AT anastasiyaloshchepkova dendriticcellderivedartificialmicrovesiclesinhibitrlssub40sublymphosarcomagrowthinmiceviastimulationofth1th17immuneresponse
AT evgeniyvbrenner dendriticcellderivedartificialmicrovesiclesinhibitrlssub40sublymphosarcomagrowthinmiceviastimulationofth1th17immuneresponse
AT nadezhdalmironova dendriticcellderivedartificialmicrovesiclesinhibitrlssub40sublymphosarcomagrowthinmiceviastimulationofth1th17immuneresponse
AT marinaazenkova dendriticcellderivedartificialmicrovesiclesinhibitrlssub40sublymphosarcomagrowthinmiceviastimulationofth1th17immuneresponse