Cellular based cancer vaccines: type 1 polarization of dendritic cells

Research output: Contribution to journalReviewResearchpeer-review

Standard

Cellular based cancer vaccines : type 1 polarization of dendritic cells. / Hansen, M; Met, Ö; Svane, I M; Andersen, M H.

In: Current Medicinal Chemistry, Vol. 19, No. 25, 2012, p. 4239-46.

Research output: Contribution to journalReviewResearchpeer-review

Harvard

Hansen, M, Met, Ö, Svane, IM & Andersen, MH 2012, 'Cellular based cancer vaccines: type 1 polarization of dendritic cells', Current Medicinal Chemistry, vol. 19, no. 25, pp. 4239-46.

APA

Hansen, M., Met, Ö., Svane, I. M., & Andersen, M. H. (2012). Cellular based cancer vaccines: type 1 polarization of dendritic cells. Current Medicinal Chemistry, 19(25), 4239-46.

Vancouver

Hansen M, Met Ö, Svane IM, Andersen MH. Cellular based cancer vaccines: type 1 polarization of dendritic cells. Current Medicinal Chemistry. 2012;19(25):4239-46.

Author

Hansen, M ; Met, Ö ; Svane, I M ; Andersen, M H. / Cellular based cancer vaccines : type 1 polarization of dendritic cells. In: Current Medicinal Chemistry. 2012 ; Vol. 19, No. 25. pp. 4239-46.

Bibtex

@article{7bcab4044c334e13ac01d4a490629952,
title = "Cellular based cancer vaccines: type 1 polarization of dendritic cells",
abstract = "Cancer vaccines designed to re-calibrate the existing host-tumour interaction, tipping the balance from tumor acceptance towards tumor control holds huge potential to complement traditional cancer therapies. In general, limited success has been achieved with vaccines composed of tumor-associated antigens introduced to dendritic cells (DCs) generated in vitro. This may in part result from suboptimal maturation of DCs leading to insufficient production of IL-12, a key driver of cellular immunity. Therefore, tremendous efforts have been put into the design of maturation cocktails that are able to induce IL-12 secreting type 1 polarized DCs mimicing pathogen-derived molecular activation of DCs. Correct timing and potential synergisms of clinical-grade toll-like receptor ligands, interferons (IFN) and CD40L enhance IL-12 production in DCs. However, cytokine exhaustion, predominant expression of tolerogenic molecules and activation-induced dendritic cell death should be avoided. Thus, compounds such as IFN-γ may initially induce immunity but later on tolerance. Maturation with PGE(2) obviously promotes migration via expression of CCR7 but on the down side PGE(2) limits the production of IL-12 especially following encounter with CD40L-expressing cells and furthermore, PGE(2) imprints DCs for preferential interaction with tolerogenic T cells. In addition, type 1 polarized DCs matured without PGE(2) also seem to be capable of migrating in vivo, although concomitant production of CCL19 seems to transiently affect in vitro migration via autocrine receptor-mediated endocytosis of CCR7. In the current review, we discuss optimal design of DC maturation focused on pre-clinical as well as clinical results from standard and polarized dendritic cell based cancer vaccines.",
keywords = "Animals, Cancer Vaccines, Dendritic Cells, Dinoprostone, Humans, Interferon-gamma, Interleukin-12, Neoplasms, Journal Article, Research Support, Non-U.S. Gov't, Review",
author = "M Hansen and {\"O} Met and Svane, {I M} and Andersen, {M H}",
year = "2012",
language = "English",
volume = "19",
pages = "4239--46",
journal = "Current Medicinal Chemistry",
issn = "0929-8673",
publisher = "Bentham Science Publishers",
number = "25",

}

RIS

TY - JOUR

T1 - Cellular based cancer vaccines

T2 - type 1 polarization of dendritic cells

AU - Hansen, M

AU - Met, Ö

AU - Svane, I M

AU - Andersen, M H

PY - 2012

Y1 - 2012

N2 - Cancer vaccines designed to re-calibrate the existing host-tumour interaction, tipping the balance from tumor acceptance towards tumor control holds huge potential to complement traditional cancer therapies. In general, limited success has been achieved with vaccines composed of tumor-associated antigens introduced to dendritic cells (DCs) generated in vitro. This may in part result from suboptimal maturation of DCs leading to insufficient production of IL-12, a key driver of cellular immunity. Therefore, tremendous efforts have been put into the design of maturation cocktails that are able to induce IL-12 secreting type 1 polarized DCs mimicing pathogen-derived molecular activation of DCs. Correct timing and potential synergisms of clinical-grade toll-like receptor ligands, interferons (IFN) and CD40L enhance IL-12 production in DCs. However, cytokine exhaustion, predominant expression of tolerogenic molecules and activation-induced dendritic cell death should be avoided. Thus, compounds such as IFN-γ may initially induce immunity but later on tolerance. Maturation with PGE(2) obviously promotes migration via expression of CCR7 but on the down side PGE(2) limits the production of IL-12 especially following encounter with CD40L-expressing cells and furthermore, PGE(2) imprints DCs for preferential interaction with tolerogenic T cells. In addition, type 1 polarized DCs matured without PGE(2) also seem to be capable of migrating in vivo, although concomitant production of CCL19 seems to transiently affect in vitro migration via autocrine receptor-mediated endocytosis of CCR7. In the current review, we discuss optimal design of DC maturation focused on pre-clinical as well as clinical results from standard and polarized dendritic cell based cancer vaccines.

AB - Cancer vaccines designed to re-calibrate the existing host-tumour interaction, tipping the balance from tumor acceptance towards tumor control holds huge potential to complement traditional cancer therapies. In general, limited success has been achieved with vaccines composed of tumor-associated antigens introduced to dendritic cells (DCs) generated in vitro. This may in part result from suboptimal maturation of DCs leading to insufficient production of IL-12, a key driver of cellular immunity. Therefore, tremendous efforts have been put into the design of maturation cocktails that are able to induce IL-12 secreting type 1 polarized DCs mimicing pathogen-derived molecular activation of DCs. Correct timing and potential synergisms of clinical-grade toll-like receptor ligands, interferons (IFN) and CD40L enhance IL-12 production in DCs. However, cytokine exhaustion, predominant expression of tolerogenic molecules and activation-induced dendritic cell death should be avoided. Thus, compounds such as IFN-γ may initially induce immunity but later on tolerance. Maturation with PGE(2) obviously promotes migration via expression of CCR7 but on the down side PGE(2) limits the production of IL-12 especially following encounter with CD40L-expressing cells and furthermore, PGE(2) imprints DCs for preferential interaction with tolerogenic T cells. In addition, type 1 polarized DCs matured without PGE(2) also seem to be capable of migrating in vivo, although concomitant production of CCL19 seems to transiently affect in vitro migration via autocrine receptor-mediated endocytosis of CCR7. In the current review, we discuss optimal design of DC maturation focused on pre-clinical as well as clinical results from standard and polarized dendritic cell based cancer vaccines.

KW - Animals

KW - Cancer Vaccines

KW - Dendritic Cells

KW - Dinoprostone

KW - Humans

KW - Interferon-gamma

KW - Interleukin-12

KW - Neoplasms

KW - Journal Article

KW - Research Support, Non-U.S. Gov't

KW - Review

M3 - Review

C2 - 22834814

VL - 19

SP - 4239

EP - 4246

JO - Current Medicinal Chemistry

JF - Current Medicinal Chemistry

SN - 0929-8673

IS - 25

ER -

ID: 184774413