4.6 Article

TGF-β1 programmed myeloid-derived suppressor cells (MDSC) acquire immune-stimulating and tumor killing activity capable of rejecting established tumors in combination with radiotherapy

Journal

ONCOIMMUNOLOGY
Volume 7, Issue 10, Pages -

Publisher

TAYLOR & FRANCIS INC
DOI: 10.1080/2162402X.2018.1490853

Keywords

MDSC; TGF-beta; CD86; tumor killing; radiotherapy; caspase-3; myeloid-derived suppressor cells (written out); SMAD2; adoptive cellular therapy

Funding

  1. Cancer Research Institute Clinical strategy Team Grant - Targeting the Tumor Immune Microenvironment to Enhance Immune-stimulating Effects of Chemoradiotherapy (PI Sikora)
  2. Cytometry and Cell Sorting Core at Baylor College of Medicine
  3. NIH [P30 AI036211, P30 CA125123, S10 RR024574]
  4. CPRIT Core Facility Award [RP170005]
  5. NCI [P30 CA123125]
  6. National Institute of General Medical Sciences T32 predoctoral training grant of the National Institutes of Health [T32GM088129]
  7. National Institute of Dental & Craniofacial Research F31 NRSA training grant of the National Institutes of Health [F31DE026682]

Ask authors/readers for more resources

Cancer-induced myeloid-derived suppressor cells (MDSC) play an important role in tumor immune evasion. MDSC programming or polarization has been proposed as a strategy for leveraging the developmental plasticity of myeloid cells to reverse MDSC immune suppressive functions, or cause them to acquire anti-tumor activity. While MDSC derived ex vivo from murine bone marrow precursor cells with tumor-conditioned medium efficiently suppressed T cell proliferation, MDSC derived from conditioned medium in presence of TGF-beta 1 (TGF beta-MDSC) acquired a novel immune-stimulatory phenotype, losing the ability to inhibit T cell proliferation and acquiring enhanced antigen-presenting capability. Altered immune function was associated with SMAD-2 dependent upregulation of maturation and costimulatory molecules, and downregulation of inducible nitric oxide synthase (iNOS), an effector mechanism of immunosuppression. TGF beta-MDSC also upregulated FAS-ligand expression, leading to FAS-dependent killing of murine human papillomavirus (HPV)-associated head and neck cancer cells and tumor spheroids in vitro and anti-tumor activity in vivo. Radiation upregulated FAS expression on tumor cells, and the combination of radiotherapy and intratumoral injection of TGF beta-MDSC strongly enhanced class I expression on tumor cells and induction of HPV E7 tetramer-positive CD8+T cells, leading to clearance of established tumors and long-term survival. TGF beta-MDSC derived from human PBMC with tumor conditioned medium also lost immunosuppressive function and acquired tumor-killing activity. Thus, TGF beta 1 mediated programming of nascent MDSC leads to a potent anti-tumor phenotype potentially suitable for adoptive immunotherapy.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available