VEGF in ovarian cancer suppresses T cell activity by eliciting Myeloid Derived Suppressor Cells (MDSC) into tumor microenvironment

Author(s):  
Naoki Horikawa
2010 ◽  
Vol 207 (11) ◽  
pp. 2439-2453 ◽  
Author(s):  
Cesar A. Corzo ◽  
Thomas Condamine ◽  
Lily Lu ◽  
Matthew J. Cotter ◽  
Je-In Youn ◽  
...  

Myeloid-derived suppressor cells (MDSCs) are a major component of the immune-suppressive network described in cancer and many other pathological conditions. We demonstrate that although MDSCs from peripheral lymphoid organs and the tumor site share similar phenotype and morphology, these cells display profound functional differences. MDSC from peripheral lymphoid organs suppressed antigen-specific CD8+ T cells but failed to inhibit nonspecific T cell function. In sharp contrast, tumor MDSC suppressed both antigen-specific and nonspecific T cell activity. The tumor microenvironment caused rapid and dramatic up-regulation of arginase I and inducible nitric oxide synthase in MDSC, which was accompanied by down-regulation of nicotinamide adenine dinucleotide phosphate–oxidase and reactive oxygen species in these cells. In contrast to MDSC from the spleen, MDSC from the tumor site rapidly differentiated into macrophages. Exposure of spleen MDSC to hypoxia resulted in the conversion of these cells to nonspecific suppressors and their preferential differentiation to macrophages. Hypoxia-inducible factor (HIF) 1α was found to be primarily responsible for the observed effects of the tumor microenvironment on MDSC differentiation and function. Thus, hypoxia via HIF-1α dramatically alters the function of MDSC in the tumor microenvironment and redirects their differentiation toward tumor-associated macrophages, hence providing a mechanistic link between different myeloid suppressive cells in the tumor microenvironment.


2014 ◽  
Vol 16 (suppl 5) ◽  
pp. v121-v122
Author(s):  
N. Kamran ◽  
M. Ayala ◽  
Y. Li ◽  
H. Assi ◽  
M. Candolfi ◽  
...  

2021 ◽  
Vol Publish Ahead of Print ◽  
Author(s):  
Madhurima Sarkar ◽  
Avishek Bhuniya ◽  
Sarbari Ghosh ◽  
Anirban Sarkar ◽  
Akata Saha ◽  
...  

2019 ◽  
Vol 20 (20) ◽  
pp. 5079 ◽  
Author(s):  
Ching-Chuan Hsieh ◽  
Chien-Hui Hung ◽  
Meihua Chiang ◽  
Yu-Chin Tsai ◽  
Jie-Teng He

The tumor microenvironment, which consists of fibroblasts, smooth muscle cells, endothelial cells, immune cells, epithelial cells, and extracellular matrices, plays a crucial role in tumor progression. Hepatic stellate cells (HSCs), a class of unique liver stromal cells, participate in immunomodulatory activities by inducing the apoptosis of effector T-cells, generation of regulatory T-cells, and development of myeloid-derived suppressor cells (MDSCs) to achieve long-term survival of islet allografts. This study provides in vitro and in vivo evidences that HSCs induce the generation of MDSCs to promote hepatocellular carcinoma (HCC) progression through interleukin (IL)-6 secretion. HSC-induced MDSCs highly expressed inducible nitric oxide synthase (iNOS) and arginase 1 mRNA and presented potent inhibitory T-cell immune responses in the tumor environment. Wild-type HSC-induced MDSCs expressed lower levels of CD40, CD86, and MHC II, and a higher level of B7-H1 surface molecules, as well as increased the production of iNOS and arginase I compared with MDSCs induced by IL-6-deficient HSCs in vitro. A murine-transplanted model of the liver tumor showed that HCCs cotransplanted with HSCs could significantly enhance the tumor area and detect more MDSCs compared with HCCs alone or HCCs cotransplanted with HSCs lacking IL-6. In conclusion, the results indicated that MDSCs are induced mainly by HSCs through IL-6 signaling and produce inhibitory enzymes to reduce T-cell immunity and then promote HCC progression within the tumor microenvironment. Therapies targeting the pathway involved in MDSC production or its immune-modulating pathways can serve as an alternative immunotherapy for HCC.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A397-A397
Author(s):  
Sarina Piha-Paul ◽  
Alexander Starodub ◽  
Raghad Karim ◽  
Michael Shafique ◽  
Gabriel Tinoco Suarez ◽  
...  

BackgroundOvercoming the immune-suppressive tumor environment induced by myeloid-derived suppressor cells (MDSC) is a major challenge in immune therapy. AMV564 is a potent conditional agonist that engages T cells to selectively deplete target cells such as MDSC while promoting T cell polarization and activation. Whereas CD33 plays an insignificant role in differentiated myeloid cells, CD33 signaling in immature myeloid cells promotes expansion of MDSC and production of immune-suppressive factors. Preferential binding of AMV564 to areas of high CD33 density enables selective targeting of MDSC. Ex vivo data1 as well as data from a clinical trial in acute myeloid leukemia (NCT03144245) demonstrate the ability of AMV564 to selectively deplete MDSC while sparing monocytes and neutrophils.2 3MethodsNCT04128423 is a multi-center Phase 1 study to determine the safety and tolerability, define the maximum-tolerated or pharmacologically active dose, and assess the preliminary efficacy of AMV564. In this 3+3 dose escalation study, patients with advanced solid tumors receive AMV564 once daily via subcutaneous (SC) injection on Days 1–5 and 8–12 of a 21-day cycle. Primary endpoints include incidence, nature and severity of adverse events (AEs). Secondary endpoints include assessment of pharmacokinetics and pharmacodynamics.ResultsAs of June 30, 2020, 11 patients have been dosed across 3 dose cohorts (15 mcg – 75 mcg). The tumor types enrolled were: colorectal (n=2), GE junction (n=2), pancreatic (n=2), squamous cell carcinoma (n=2), small intestine, ovarian, and endometrial cancer. AMV564 has been well tolerated with no dose-limiting toxicities. The most common treatment-related AEs were fever/pyrexia (Grade 1: n=3; Grade 2: n=8) and injection site reactions (Grade 1: n=1; Grade 2: n=9). Preliminary estimate of median plasma half-life for AMV564 after SC injection was >48 hours, with dose-related increases in peak plasma concentration (Cmax). Tumor responses were evaluable in 9 patients; 1 patient had not reached their first assessment and 1 patient was not efficacy evaluable due to a non-treatment-related AE resulting in study discontinuation. Single-agent activity has been observed including a complete response by RECISTv1.1 criteria in 1 patient with ovarian cancer refractory to all standard therapies and anti-PD-1 therapy, and stable disease in 4 additional patients.ConclusionsAMV564 has been well tolerated across multiple dose levels, with good plasma exposure and evidence of anti-tumor activity when administered subcutaneously. Single-agent anti-tumor activity was observed in an ovarian cancer patient.AcknowledgementsWe would like to thank the patients and their families for participating in this clinical trial.Trial RegistrationNCT04128423Ethics ApprovalThe study was approved by the Institutional Review Board at each center where the study is being conducted.ReferencesCheng P, Eksioglu E, Chen X, et al. Immunodepletion of MDSC By AMV564, a novel Tetravalent bispecific CD33/CD3 T cell engager restores immune homeostasis in MDS in Vitro. Blood. 2017; 130:51 (abstract).Eckard S, Gehrs L, Smith V, et al. AMV564, a novel bivalent, bispecific T-cell engager, targets myeloid-derived suppressor cells. SITC Annual Meeting; 2019 Nov 6-10. Oral Presentation O71.Westervelt P, Roboz G, Cortes J, et al. Safety and Clinical Activity of AMV564, a CD33/CD3 T-cell Engager, in Patients with Relapsed/Refractory Acute Myeloid Leukemia (AML): Updated Results from the Phase 1 First-in-Human Trial. EHA Annual Congress; 2019 Jun 13-16. Abstract S877.


2017 ◽  
Vol 35 (7_suppl) ◽  
pp. 17-17
Author(s):  
Weiping Zou

17 Background: The tumor microenvironment is comprised of immune cells that have been reprogrammed by active tumor-mediated processes to defeat tumor-specific immunity and promote tumor growth in a highly effective manner. Previous studies have helped define the nature of immune responses in the tumor microenvironment, and provide new insights into designing novel immune therapies to target the immune suppressive mechanisms including Tregs and inhibitory B7 family members (e.g. PD-L1/PD-1) and treat patients with cancer. However, the mechanisms controlling tumor immune phenotype and biological phenotype, and their relevance in cancer therapy are not well understood. Methods: Using human colon and ovarian cancer as our models, we have studied the cross-talk between immune cell subsets and tumor/stem cells, and between immunogenic and oncogenic pathways in the tumor microenvironment, and its impact on tumor immunity and therapy. Results: In patients with colon and ovarian cancer as our models, we demonstrate that Th22 cells and myeloid derived suppressor cells (MDSCs) regulate tumor stemness via epigenetic mechanisms. Furthermore, we found that histone modification and DNA methylation repress the tumor production of Th1-type chemokines, and subsequently determine effector T cell trafficking to the tumor microenvironment, affect tumor progression, and therapeutic efficacy of checkpoint blockade and adoptive T cell transfusion, chemotherapy and patient outcome. Conclusions: Epigenetic silencing of Th1-type chemokine is a novel tumor immune evasion mechanism. Selective epigenetic reprogramming alters T cell landscape in cancer and may enhance clinical efficacy of cancer therapy.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1105-1105
Author(s):  
Jie Chen ◽  
Fengdong Cheng ◽  
Michael Wang ◽  
Eva Sahakian ◽  
John J. Powers ◽  
...  

Abstract Myeloid-derived suppressor cells constitute a heterogeneous population of immature myeloid cells derived from bone marrow that negatively regulate both innate and adaptive immunity in the tumor microenvironment. Previous work in our lab had demonstrated that MDSCs lacking histone deacetylase 11 (HDAC11) displayed an increase in suppressive activity against IFN-γ producing CD8+ T cells. The upregulated suppressive activity of HDAC11KO MDSCs was associated with a more aggressive tumor growth pattern when compared with wild type control mice. Mechanistically we have found that tumor infiltrated HDAC11KO MDSCs isolated from lymphoma-bearing mice displayed up-regulation of expression and enzymatic activity of arginase 1 and Nos2, two enzymes that are crucial in regulating MDSCs suppressive function, when compare with wild type MDSCs. However, both arginase activity and NO production were at a similar level in the wild type and HDAC11KO MDSCs isolated from spleens of the same lymphoma-bearing mice. This finding suggests that HDAC11KO MDSCs are more suppressive within the tumor microenvironment. Moreover, the aberrant enzymatic activities of Arg1 and Nos2 in HDAC11KO MDSCs correlate with over-expression of the lineage-specific transcription factor C/EBPβ, which has been previously shown to be essential for the differentiation of functional MDSCs. Furthermore, ChIP analysis confirmed that HDAC11 is recruited to the C/EBPβ gene promoter where exerts a negative regulatory effect upon gene transcription. Unlike MDSC's in which absence of HDAC11 is associated with a suppressive phenotype, T-cell lacking HDAC11 are hyper-reactive and endowed with strong antitumor activity. To assess which phenotype will be the dominant one in vivo, we performed adoptive immune cell transfer experiments of both MDSC and/or T-cells from either wild type or HDAC11 KO mice into C57BL/6 lymphoma-bearing animals. The transfer of HDAC11KO MDSCs was able to eliminate, at least partially, the anti-tumor effect elicited by the HDAC11KO T cells in the lymphoma microenvironment Taken together, we have uncovered a previously unknown role for HDAC11 as a transcriptional regulator of MDSCs phenotype and function in a murine lymphoma model. A better understanding of this novel role of HDAC11 in myeloid biology will lead to targeted epigenetic therapies to manipulate the suppressive effect of these immunoregulatory cells in vivo. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document