Inhibition of IL-10 in the Tumor Microenvironment Potentiates Mesothelin-Chimeric Antigen Receptor NK-92MI-Mediated Killing of Ovarian Cancer Cells

2020 ◽  
Vol 231 (4) ◽  
pp. e54-e55
Author(s):  
Ramesh Babu Batchu ◽  
Oksana V. Gruzdyn ◽  
Bala K. Kolli ◽  
Pavan S. Tavva ◽  
Rajesh S. Dachepalli ◽  
...  
2017 ◽  
Vol 35 (7_suppl) ◽  
pp. 141-141
Author(s):  
Oladapo O. Yeku ◽  
Terence Purdon ◽  
David R. Spriggs ◽  
Renier J. Brentjens

141 Background: Chimeric antigen receptor (CAR) T cell therapy for solid tumor malignancies has not shown the same degree of clinical efficacy observed in hematologic malignancies such as B-ALL. The presence of an immunosuppressive cellular and cytokine microenvironment has been hypothesized as one reason for the failure of adoptive immunotherapy for solid tumors. In ovarian cancer, myeloid derived suppressor cells (MDSC) and immunosuppressive cytokines in the ascitic microenvironment have been reported. IL-12 is a proinflammatory cytokine produced by macrophages, dendritic cells (DC), and NK cells, and it has been shown to increase proliferation of T cells, induce differentiation of type 1 T helper cells, inhibit regulatory T cells, promote maturation of DCs, and enhance antigen presentation by macrophages. We hypothesize that CAR T cells genetically modified to constitutively secrete IL-12 will overcome a hostile tumor microenvironment in a peritoneal carcinomatosis model of ovarian cancer. Methods: CAR T cells were generated from retroviral transduction of second generation and IL-12 modified CAR’s directed to either an irrelevant CD-19 antigen or Muc16ecto. C57BL/6 mice were inoculated i.p with syngeneic ovarian cancer cells and treated with various CAR T cells. Results: Here we report increased production of IL-12, improved proliferation and cytotoxic activity of 4H1128ζ-IL12 CAR T cells. Further, we show increased levels of inflammatory cytokines at 24 and 48hrs after treatment of tumor-bearing mice, leading to increased survival at advanced stages of disease. Animals treated with 4H1128ζ-IL12 CAR T cells had decreased levels of F4/80+ CD11b+MDSCs. Genetic analysis of recovered MDSCs from the ascites of treated animals showed skewing towards an M1-phenotype via upregulation of cytokines, chemokines, MHC-II, and downregulation of Arg1. Furthermore, clodronate-mediated depletion of TAM’s further enhanced survival in mice treated with 4H1128ζ-IL12 CAR T cells. Conclusions: These results demonstrate the mechanisms of efficacy of localized delivery of IL-12 to the tumor microenvironment by 4H1128ζ-IL12 CAR T cells.


Biomedicines ◽  
2021 ◽  
Vol 9 (10) ◽  
pp. 1339
Author(s):  
Rüdiger Klapdor ◽  
Shuo Wang ◽  
Michael A. Morgan ◽  
Katharina Zimmermann ◽  
Jens Hachenberg ◽  
...  

Ovarian cancer is the most common cause of gynecological cancer-related death in the developed world. Disease recurrence and chemoresistance are major causes of poor survival rates in ovarian cancer patients. Ovarian cancer stem cells (CSCs) were shown to represent a source of tumor recurrence owing to the high resistance to chemotherapy and enhanced tumorigenicity. Chimeric antigen receptor (CAR)-based adoptive immunotherapy represents a promising strategy to reduce the risk for recurrent disease. In this study, we developed a codon-optimized third-generation CAR to specifically target CD44, a marker widely expressed on ovarian cancer cells and associated with CSC-like properties and intraperitoneal tumor spread. We equipped NK-92 cells with the anti-CD44 CAR (CD44NK) and an anti-CD19 control CAR (CD19NK) using lentiviral SIN vectors. Compared to CD19NK and untransduced NK-92 cells, CD44NK showed potent and specific cytotoxic activity against CD44-positive ovarian cancer cell lines (SKOV3 and OVCAR3) and primary ovarian cancer cells harvested from ascites. In contrast, CD44NK had less cytotoxic activity against CD44-negative A2780 cells. Specific activation of engineered NK cells was also demonstrated by interferon-γ (IFNγ) secretion assays. Furthermore, CD44NK cells still demonstrated cytotoxic activity under cisplatin treatment. Most importantly, the simultaneous treatment with CD44NK and cisplatin showed higher anti-tumor activity than sequential treatment.


Cells ◽  
2021 ◽  
Vol 10 (11) ◽  
pp. 3213
Author(s):  
Alessandra Ferraresi ◽  
Andrea Esposito ◽  
Carlo Girone ◽  
Letizia Vallino ◽  
Amreen Salwa ◽  
...  

Background Ovarian cancer progression and invasiveness are promoted by a range of soluble factors released by cancer cells and stromal cells within the tumor microenvironment. Our previous studies demonstrated that resveratrol (RV), a nutraceutical and caloric restriction mimetic with tumor-suppressive properties, counteracts cancer cell motility induced by stromal IL-6 by upregulating autophagy. Lysophosphatidic acid (LPA), a bioactive phospholipid that shows elevated levels in the tumor microenvironment and the ascites of ovarian cancers, stimulates the growth and tissue invasion of cancer cells. Whether LPA elicits these effects by inhibiting autophagy and through which pathway and whether RV can counteract the same remain obscure. Aims To investigate the molecular pathways involved in LPA-induced ovarian cancer malignancy, particularly focusing on the role of autophagy, and the ability of RV to counteract LPA activity. Results LPA stimulated while RV inhibited ovarian cancer cell migration. Transcriptomic and bioinformatic analyses showed an opposite regulation by LPA and RV of genes linked to epithelial-to-mesenchymal transition (EMT) and autophagy with involvement of the PI3K-AKT, JAK-STAT and Hedgehog (Hh) pathways. LPA upregulated the Hh and EMT members GLI1, BMI-1, SNAIL-1 and TWIST1 and inhibited autophagy, while RV did the opposite. Similar to the inhibitors of the Hh pathway, RV inhibited LPA-induced cancer cell migration and 3D growth of ovarian cancer cells. BMI-1 silencing prevented LPA-induced EMT, restored autophagy and hampered cell migration, resembling the effects of RV. TCGA data analyses indicated that patients with low expression of Hh/EMT-related genes together with active autophagy flux tended to have a better prognosis and this correlates with a more effective response to platinum therapy. In in vitro 3D spheroids, LPA upregulated BMI-1, downregulated autophagy and inhibited platinum toxicity while RV and Hh inhibitors restored autophagy and favored BAX-mediated cell death in response to platinum. Conclusions By inhibiting the Hh pathway and restoration of autophagy, RV counteracts LPA-induced malignancy, supporting its inclusion in the therapy of ovarian cancer for limiting metastasis and chemoresistance.


2021 ◽  
Vol 12 ◽  
Author(s):  
Xin Luo ◽  
Jing Xu ◽  
Jianhua Yu ◽  
Ping Yi

Reciprocal signaling between immune cells and ovarian cancer cells in the tumor microenvironment can alter immune responses and regulate disease progression. These signaling events are regulated by multiple factors, including genetic and epigenetic alterations in both the ovarian cancer cells and immune cells, as well as cytokine pathways. Multiple immune cell types are recruited to the ovarian cancer tumor microenvironment, and new insights about the complexity of their interactions have emerged in recent years. The growing understanding of immune cell function in the ovarian cancer tumor microenvironment has important implications for biomarker discovery and therapeutic development. This review aims to describe the factors that shape the phenotypes of immune cells in the tumor microenvironment of ovarian cancer and how these changes impact disease progression and therapy.


2019 ◽  
Vol 516 (1) ◽  
pp. 96-101
Author(s):  
Mariana Medeiros ◽  
Amanda Oliveira Ribeiro ◽  
Luiz Antônio Lupi ◽  
Guilherme Ribeiro Romualdo ◽  
Danillo Pinhal ◽  
...  

Cancers ◽  
2018 ◽  
Vol 10 (9) ◽  
pp. 295 ◽  
Author(s):  
Yuliya Klymenko ◽  
Kenneth P. Nephew

Metastatic dissemination of epithelial ovarian cancer (EOC) predominantly occurs through direct cell shedding from the primary tumor into the intra-abdominal cavity that is filled with malignant ascitic effusions. Facilitated by the fluid flow, cells distribute throughout the cavity, broadly seed and invade through peritoneal lining, and resume secondary tumor growth in abdominal and pelvic organs. At all steps of this unique metastatic process, cancer cells exist within a multidimensional tumor microenvironment consisting of intraperitoneally residing cancer-reprogramed fibroblasts, adipose, immune, mesenchymal stem, mesothelial, and vascular cells that exert miscellaneous bioactive molecules into malignant ascites and contribute to EOC progression and metastasis via distinct molecular mechanisms and epigenetic dysregulation. This review outlines basic epigenetic mechanisms, including DNA methylation, histone modifications, chromatin remodeling, and non-coding RNA regulators, and summarizes current knowledge on reciprocal interactions between each participant of the EOC cellular milieu and tumor cells in the context of aberrant epigenetic crosstalk. Promising research directions and potential therapeutic strategies that may encompass epigenetic tailoring as a component of complex EOC treatment are discussed.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. 3050-3050 ◽  
Author(s):  
Oladapo O. Yeku ◽  
Terence Purdon ◽  
David R. Spriggs ◽  
Renier J. Brentjens

3050 Background: Chimeric antigen receptor (CAR) T cell therapy for solid tumor malignancies has not shown the same degree of clinical efficacy observed in hematologic malignancies. The presence of an immunosuppressive cytokine and cellular microenvironment has been hypothesized as one reason for the failure of adoptive immunotherapy for solid tumors. In ovarian cancer, the presence of tumor associated macrophages (TAMs) and immunosuppressive cytokines in the ascitic microenvironment have been reported. IL-12 is a proinflammatory cytokine produced by macrophages, dendritic cells (DC) and NK cells, and has been shown to increase proliferation of T cells and enhance antigen presentation by macrophages. We hypothesized that CAR T cells genetically modified to constitutively secrete IL-12 would overcome a hostile tumor microenvironment in a peritoneal carcinomatosis model of ovarian cancer. Methods: CAR T cells were generated from retroviral transduction of second generation and IL-12 modified CAR’s directed to either an irrelevant CD-19 antigen or Muc16ecto. Results: Here we report increased production of IL-12, improved proliferation and cytotoxic activity of 4H1128-IL12 CAR T cells. We show increased levels of inflammatory cytokines at 24 and 48hrs after treatment of tumor-bearing mice, leading to increased survival at advanced stages of disease. Mice treated with 4H1128-IL12 CAR T cells had decreased levels of F4/80+ CD11b+TAM’s. Genetic analysis of recovered TAM’s from the ascites of treated animals showed skewing towards an M1-phenotype via upregulation of cytokines, chemokines, MHC-II and downregulation of Arg1. Recovered 4H1128-IL12 CAR T cells showed upregulation of FAS-L and recovered TAMs showed increased expression of FAS suggesting FAS/FAS-L engagement was responsible for decreased TAMs. Blocking the FAS/FAS-L pathway led to recovery of TAM populations in 4H1128-IL12 treated mice. Finally, clodronate-mediated depletion of TAM’s further enhanced survival in mice treated with 4H1128-IL12 CAR T cells. Conclusions: These results demonstrate the mechanisms of efficacy of localized delivery of IL-12 to the tumor microenvironment by 4H1128-IL12 CAR T cells.


2020 ◽  
Vol 302 (4) ◽  
pp. 1009-1017
Author(s):  
Jiang Yi ◽  
Yuan Lin ◽  
Wan Yicong ◽  
Luo Chengyan ◽  
Zhou Shulin ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document