Skip to main content

Lund University Publications

LUND UNIVERSITY LIBRARIES

Tasquinimod Modulates Suppressive Myeloid Cells and Enhances Cancer Immunotherapies in Murine Models

Shen, Li ; Sundstedt, Anette ; Ciesielski, Michael ; Miles, Kiersten Marie ; Celander, Mona ; Adelaiye, Remi ; Orillion, Ashley ; Ciamporcero, Eric ; Ramakrishnan, Swathi and Ellis, Leigh , et al. (2015) In Cancer immunology research 3(2). p.136-148
Abstract
A major barrier for cancer immunotherapy is the presence of suppressive cell populations in patients with cancer, such as myeloid-derived suppressor cells (MDSC) and tumor-associated macrophages (TAM), which contribute to the immunosuppressive microenvironment that promotes tumor growth and metastasis. Tasquinimod is a novel antitumor agent that is currently at an advanced stage of clinical development for treatment of castration-resistant prostate cancer. A target of tasquinimod is the inflammatory protein S100A9, which has been demonstrated to affect the accumulation and function of tumor-suppressive myeloid cells. Here, we report that tasquinimod provided a significant enhancement to the antitumor effects of two different... (More)
A major barrier for cancer immunotherapy is the presence of suppressive cell populations in patients with cancer, such as myeloid-derived suppressor cells (MDSC) and tumor-associated macrophages (TAM), which contribute to the immunosuppressive microenvironment that promotes tumor growth and metastasis. Tasquinimod is a novel antitumor agent that is currently at an advanced stage of clinical development for treatment of castration-resistant prostate cancer. A target of tasquinimod is the inflammatory protein S100A9, which has been demonstrated to affect the accumulation and function of tumor-suppressive myeloid cells. Here, we report that tasquinimod provided a significant enhancement to the antitumor effects of two different immunotherapeutics in mouse models of cancer: a tumor vaccine (SurVaxM) for prostate cancer and a tumor-targeted superantigen (TTS) for melanoma. In the combination strategies, tasquinimod inhibited distinct MDSC populations and TAMs of the M2-polarized phenotype (CD206(+)). CD11b(+) myeloid cells isolated from tumors of treated mice expressed lower levels of arginase-1 and higher levels of inducible nitric oxide synthase (iNOS), and were less immunosuppressive ex vivo, which translated into a significantly reduced tumor-promoting capacity in vivo when these cells were coinjected with tumor cells. Tumor-specific CD8(+) T cells were increased markedly in the circulation and in tumors. Furthermore, T-cell effector functions, including cell-mediated cytotoxicity and IFN gamma production, were potentiated. Taken together, these data suggest that pharmacologic targeting of suppressive myeloid cells by tasquinimod induces therapeutic benefit and provide the rationale for clinical testing of tasquinimod in combination with cancer immunotherapies. (C) 2014 AACR. (Less)
Please use this url to cite or link to this publication:
author
; ; ; ; ; ; ; ; and , et al. (More)
; ; ; ; ; ; ; ; ; ; ; ; ; ; and (Less)
organization
publishing date
type
Contribution to journal
publication status
published
subject
in
Cancer immunology research
volume
3
issue
2
pages
136 - 148
publisher
American Association for Cancer Research
external identifiers
  • wos:000349422600006
  • scopus:84962290319
  • pmid:25370534
ISSN
2326-6074
DOI
10.1158/2326-6066.CIR-14-0036
language
English
LU publication?
yes
id
1e9f5271-2f0e-4d04-a3c2-ceabe2fe396d (old id 5175957)
date added to LUP
2016-04-01 14:04:01
date last changed
2022-03-06 17:16:55
@article{1e9f5271-2f0e-4d04-a3c2-ceabe2fe396d,
  abstract     = {{A major barrier for cancer immunotherapy is the presence of suppressive cell populations in patients with cancer, such as myeloid-derived suppressor cells (MDSC) and tumor-associated macrophages (TAM), which contribute to the immunosuppressive microenvironment that promotes tumor growth and metastasis. Tasquinimod is a novel antitumor agent that is currently at an advanced stage of clinical development for treatment of castration-resistant prostate cancer. A target of tasquinimod is the inflammatory protein S100A9, which has been demonstrated to affect the accumulation and function of tumor-suppressive myeloid cells. Here, we report that tasquinimod provided a significant enhancement to the antitumor effects of two different immunotherapeutics in mouse models of cancer: a tumor vaccine (SurVaxM) for prostate cancer and a tumor-targeted superantigen (TTS) for melanoma. In the combination strategies, tasquinimod inhibited distinct MDSC populations and TAMs of the M2-polarized phenotype (CD206(+)). CD11b(+) myeloid cells isolated from tumors of treated mice expressed lower levels of arginase-1 and higher levels of inducible nitric oxide synthase (iNOS), and were less immunosuppressive ex vivo, which translated into a significantly reduced tumor-promoting capacity in vivo when these cells were coinjected with tumor cells. Tumor-specific CD8(+) T cells were increased markedly in the circulation and in tumors. Furthermore, T-cell effector functions, including cell-mediated cytotoxicity and IFN gamma production, were potentiated. Taken together, these data suggest that pharmacologic targeting of suppressive myeloid cells by tasquinimod induces therapeutic benefit and provide the rationale for clinical testing of tasquinimod in combination with cancer immunotherapies. (C) 2014 AACR.}},
  author       = {{Shen, Li and Sundstedt, Anette and Ciesielski, Michael and Miles, Kiersten Marie and Celander, Mona and Adelaiye, Remi and Orillion, Ashley and Ciamporcero, Eric and Ramakrishnan, Swathi and Ellis, Leigh and Fenstermaker, Robert and Abrams, Scott I. and Eriksson, Helena and Leanderson, Tomas and Olsson, Anders and Pili, Roberto}},
  issn         = {{2326-6074}},
  language     = {{eng}},
  number       = {{2}},
  pages        = {{136--148}},
  publisher    = {{American Association for Cancer Research}},
  series       = {{Cancer immunology research}},
  title        = {{Tasquinimod Modulates Suppressive Myeloid Cells and Enhances Cancer Immunotherapies in Murine Models}},
  url          = {{http://dx.doi.org/10.1158/2326-6066.CIR-14-0036}},
  doi          = {{10.1158/2326-6066.CIR-14-0036}},
  volume       = {{3}},
  year         = {{2015}},
}