Induction of In Vitro and In Vivo NK Cell Cytotoxicity Using High-Avidity Immunoligands Targeting Prostate-Specific Membrane Antigen in Prostate Carcinoma

2011 ◽  
Vol 10 (6) ◽  
pp. 1036-1045 ◽  
Author(s):  
Ron D. Jachimowicz ◽  
Giulio Fracasso ◽  
Paul J. Yazaki ◽  
Barbara E. Power ◽  
Peter Borchmann ◽  
...  
Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 925-925 ◽  
Author(s):  
Andreas Lundqvist ◽  
Kristy Greeneltch ◽  
Maria Berg ◽  
Shivani Srivastava ◽  
Nanae Harashima ◽  
...  

Abstract Killer IgG like receptor (KIR) inactivation of NK cells by self HLA molecules has been proposed as a mechanism through which malignant cells evade host NK cell-mediated immunity. To overcome this limitation, we sought to develop a method to sensitize the patient’s tumor to autologous NK cell cytotoxicity. The proteasome inhibitor bortezomib has recently been shown to enhance the activity of tumor death receptors. We found that exposure of a variety of different leukemia, lymphoma and solid tumor cancer cell lines to sub-apoptotic doses of bortezomib sensitized tumor cells in vitro to lysis by allogeneic NK cells. Importantly, this sensitizing effect also occurs with autologous NK cells normally rendered inactive via tumor KIR ligands; NK cells expanded from patients with metastatic renal cell carcinoma were significantly more cytotoxic against the patient’s own autologous tumor cells when pretreated with bortezomib compared to untreated tumors. This sensitization to autologous NK cell killing was also observed in vivo in two different murine tumor models. A significant delay in tumor growth in C57BL/6 mice bearing LLC1 tumors (figure) and a delay in tumor growth and a significant prolongation (p<0.01) in survival were observed in RENCA tumor bearing Balb/c mice treated with bortezomib and syngeneic NK cell infusions compared to untreated mice or animals treated with bortezomib alone or NK cells alone. An investigation into the mechanism through which NK cell cytotoxicity was potentiated revealed bortezomib enhanced the activity of tumor death receptor-dependent and -independent apoptotic pathways. More specifically, bortezomib sensitized human and murine tumor cells to TRAIL and perforin/granzyme mediated NK cell cytotoxicity respectively. These observations suggest that pretreatment of malignant cells with bortezomib could be used as a strategy to override NK cell inhibition via tumor KIR ligands, thus potentiating the activity of adoptively infused autologous NK cells. A clinical trial evaluating the safety and anti-tumor efficacy of adoptively infused autologous NK cells in patients with advanced malignancies with and without tumor sensitization using bortezomib is currently being explored. Figure: Tumor growth in LLC1 bearing C57BL/6 mice. Fourteen days following s.c. injection of 3x105 LLC1 tumor cells, mice received 15μg (i.p) bortezomib and/or an adoptive infusion of 1x106 NK cells from C57BL/6 mice (i.v) given on day 15. Each dot represents the tumor volume of individual mice measured on day 28 post tumor injection. Tumors were significantly smaller in mice treated with bortezomib followed by NK cells compared to controls or mice that received either NK cells alone or bortezomib alone (p<0.04 for all groups). Figure:. Tumor growth in LLC1 bearing C57BL/6 mice. . / Fourteen days following s.c. injection of 3x105 LLC1 tumor cells, mice received 15μg (i.p) bortezomib and/or an adoptive infusion of 1x106 NK cells from C57BL/6 mice (i.v) given on day 15. Each dot represents the tumor volume of individual mice measured on day 28 post tumor injection. Tumors were significantly smaller in mice treated with bortezomib followed by NK cells compared to controls or mice that received either NK cells alone or bortezomib alone (p<0.04 for all groups).


2019 ◽  
Author(s):  
Li-Juan Chen ◽  
Bin Hu ◽  
Zhi-Qiang Han ◽  
Jian Ni ◽  
Yong-Ming Zhou ◽  
...  

Abstract Background: Intriguingly, microRNA-20a (miR-20a) has been recently witnessed to be involved in the pathogenesis of endometriosis (EMS) but the molecular mechanism controlled by miR-20a is to be undefined. The present study is designed to probe into how miR-20a acts to regulate the cytotoxicity of natural killer (NK) cells. Methods: Most of all, consistent with the clinical determination in EMS patients, miR-20a was determined to be down-regulated in NK cells isolated from nude mice. miR-20a could specifically bind to ERG and negatively regulates its expression in NK cells. Additionally, shRNA-mediated silencing of ERG decreased the expression of HLX. HLX up-regulated STAT4 by inducing proteasome degradation and inhibited NK cell cytotoxicity. Results: Of great importance, forced expression of miR-20a consequently induced NK cell cytotoxicity in vitro by increasing perforin expression via enhancement of STAT4 that was caused by impairing the binding of ERG to HLX enhancer. The in vivo experiments further confirmed the promoting role of miR-20a in the cytotoxicity of NK cells isolated from EMS nude mice and subsequent protective role of miR-20a against EMS-induced endometrial injury. Conclusion: The aforementioned data suggest that miR-20a potentiates the cytotoxicity of NK via up-regulating perforin mediated by ERG/HLX/STAT4, highlighting potential novel mechanisms associated with EMS progression.


2012 ◽  
Vol 32 (3) ◽  
pp. 632-646 ◽  
Author(s):  
Caroline Veuillen ◽  
Thérèse Aurran-Schleinitz ◽  
Rémy Castellano ◽  
Jérôme Rey ◽  
Françoise Mallet ◽  
...  

2007 ◽  
Vol 204 (4) ◽  
pp. 893-906 ◽  
Author(s):  
Ulrike Schleicher ◽  
Jan Liese ◽  
Ilka Knippertz ◽  
Claudia Kurzmann ◽  
Andrea Hesse ◽  
...  

Natural killer (NK) cells are sentinel components of the innate response to pathogens, but the cell types, pathogen recognition receptors, and cytokines required for their activation in vivo are poorly defined. Here, we investigated the role of plasmacytoid dendritic cells (pDCs), myeloid DCs (mDCs), Toll-like receptors (TLRs), and of NK cell stimulatory cytokines for the induction of an NK cell response to the protozoan parasite Leishmania infantum. In vitro, pDCs did not endocytose Leishmania promastigotes but nevertheless released interferon (IFN)-α/β and interleukin (IL)-12 in a TLR9-dependent manner. mDCs rapidly internalized Leishmania and, in the presence of TLR9, produced IL-12, but not IFN-α/β. Depletion of pDCs did not impair the activation of NK cells in L. infantum–infected mice. In contrast, L. infantum–induced NK cell cytotoxicity and IFN-γ production were abolished in mDC-depleted mice. The same phenotype was observed in TLR9−/− mice, which lacked IL-12 expression by mDCs, and in IL-12−/− mice, whereas IFN-α/β receptor−/− mice showed only a minor reduction of NK cell IFN-γ expression. This study provides the first direct evidence that mDCs are essential for eliciting NK cell cytotoxicity and IFN-γ release in vivo and demonstrates that TLR9, mDCs, and IL-12 are functionally linked to the activation of NK cells in visceral leishmaniasis.


2020 ◽  
Vol 22 (9) ◽  
pp. 1302-1314 ◽  
Author(s):  
Cavan P Bailey ◽  
Mary Figueroa ◽  
Achintyan Gangadharan ◽  
Yanwen Yang ◽  
Megan M Romero ◽  
...  

Abstract Background Diffuse midline gliomas (DMG), including brainstem diffuse intrinsic pontine glioma (DIPG), are incurable pediatric high-grade gliomas (pHGG). Mutations in the H3 histone tail (H3.1/3.3-K27M) are a feature of DIPG, rendering them therapeutically sensitive to small-molecule inhibition of chromatin modifiers. Pharmacological inhibition of lysine-specific demethylase 1 (LSD1) is clinically relevant but has not been carefully investigated in pHGG or DIPG. Methods Patient-derived DIPG cell lines, orthotopic mouse models, and pHGG datasets were used to evaluate effects of LSD1 inhibitors on cytotoxicity and immune gene expression. Immune cell cytotoxicity was assessed in DIPG cells pretreated with LSD1 inhibitors, and informatics platforms were used to determine immune infiltration of pHGG. Results Selective cytotoxicity and an immunogenic gene signature were established in DIPG cell lines using clinically relevant LSD1 inhibitors. Pediatric HGG patient sequencing data demonstrated survival benefit of this LSD1-dependent gene signature. Pretreatment of DIPG with these inhibitors increased lysis by natural killer (NK) cells. Catalytic LSD1 inhibitors induced tumor regression and augmented NK cell infusion in vivo to reduce tumor burden. CIBERSORT analysis of patient data confirmed NK infiltration is beneficial to patient survival, while CD8 T cells are negatively prognostic. Catalytic LSD1 inhibitors are nonperturbing to NK cells, while scaffolding LSD1 inhibitors are toxic to NK cells and do not induce the gene signature in DIPG cells. Conclusions LSD1 inhibition using catalytic inhibitors is selectively cytotoxic and promotes an immune gene signature that increases NK cell killing in vitro and in vivo, representing a therapeutic opportunity for pHGG. Key Points 1. LSD1 inhibition using several clinically relevant compounds is selectively cytotoxic in DIPG and shows in vivo efficacy as a single agent. 2. An LSD1-controlled gene signature predicts survival in pHGG patients and is seen in neural tissue from LSD1 inhibitor–treated mice. 3. LSD1 inhibition enhances NK cell cytotoxicity against DIPG in vivo and in vitro with correlative genetic biomarkers.


2017 ◽  
Vol 7 (1) ◽  
Author(s):  
Li Yang ◽  
MingJing Shen ◽  
Li Jun Xu ◽  
Xiaodong Yang ◽  
Ying Tsai ◽  
...  

Abstract Major progress has been made clinically in inhibiting the programmed death receptor 1 (PD-1)/PD-L1 interaction to enhance T cell-mediated immune function, yet the effectiveness of anti-PD-L1/PD-1 agents in enhancing natural killer (NK) cell’s function remains largely unknown. Susceptibilities of cisplatin-resistant A549CisR and H157CisR cells vs. parental cells to the cytotoxic action of NK cells were examined. We found cisplatin-resistant cells more resistant to NK cell cytotoxicity than parental cells. There were constitutively higher expressions of PD-L1 in A549CisR and H157CisR cells than in parental cells in vitro, as well as in H157CisR cell-derived tumors than H157P cell-derived tumors. In contrast, we observed that the expression of PD-1 in NK cells was induced after co-culture with cisplatin-resistant cells. We also observed increased susceptibility of cisplatin-resistant cells to NK cell cytotoxicity when neutralizing antibody of PD-1 or PD-L1 was added. Further, we found that the NK group 2, member D (NKG2D) ligand levels were lower in A549CisR and H157CisR cells than in parental cells. Meanwhile, we discovered that the MEK/Erk signaling pathway played a significant role in this regulation, and the addition of a MEK/Erk pathway inhibitor significantly enhanced the PD-L1 Ab effect in enhancing NK cell cytotoxicity to cisplatin-resistant cells.


2019 ◽  
Vol 225 ◽  
pp. 115223 ◽  
Author(s):  
Xin Xie ◽  
Lingman Ma ◽  
Yiran Zhou ◽  
Wen Shen ◽  
Duiyue Xu ◽  
...  

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1015-1015 ◽  
Author(s):  
Elissa Furutani ◽  
Su Su ◽  
Aleah Smith ◽  
Maria Berg ◽  
Richard Childs

Abstract Abstract 1015 Natural killer (NK) cells are a component of the innate immune system that target both tumors and virally infected cells. NK cell killing of tumors is regulated by a delicate balance of activating and inhibitory receptors. These inhibitory receptors bind HLA ligands which prevent NK cell targeting of normal “self” cells. Up regulation of surface expression of HLA molecules has been utilized by tumors as a method to evade NK cell cytotoxicity. Disrupting the function or expression of inhibitory receptors on NK cells could potentially be used as a method to overcome this effect. While most inhibitory receptors are present in only a subset of NK cells, NK cells universally express the HLA-E binding inhibitory receptor NKG2A. We hypothesized that siRNA inactivation of NK cell NKG2A would could be used as a therapeutic approach to enhance NK cell tumor cytotoxicity in vivo. The human natural killer cell line NKL was transduced with lentiviral vectors encoding shRNA targeting various regions of the NKG2A transcript. Following lentiviral transduction, knockdown of receptor expression was confirmed by flow cytometry and RT-qPCR. Compared to wild type (WT) and GFP-transduced NKL controls, NKG2A silenced NKL cells had increased secretion of IFN-gamma and Fas-L by ELISA and increased granzymes A and B and Nkp30 expression by flow cytometry. In contrast, expression of NKG2D, Nkp44, Nkp46, LFA-1, DNAM, and TRAIL was not altered by NKG2A silencing. Chromium-based cytotoxicity assays showed shRNA knockdown of NKG2A significantly enhanced NK cell cytotoxicity of tumor cells: at a 20:1 effector to target ratio, NKG2A knockdown NKLs, WT NKLs and GFP-transduced NKLs induced 68.9%, 8.2% and 8.3% lysis respectively of 721.221 EBV-LCL tumor targets (p=0.001). Remarkably, NKG2A silencing enhanced NKL killing of both HLA-E positive (721.221 EBV-LCL and 526 melanoma cells) and HLA-E negative (K562) tumor cell lines, suggesting NKG2A inactivation increased NK cell cytotoxicity through both HLA-E dependent and independent mechanisms. Using a xenogeneic model, we next explored the in vivo effects of transferring NKG2A silenced NK cells in tumor bearing mice. Immunodeficient NSG mice were injected with 1 million human luciferase transduced 721.221 HLA-E expressing EBV-LCL tumor cells. Twenty-four hours later, tumor-bearing mice were injected with 2–5 million WT NKL cells, GFP-control-transduced NKL, or NKG2A silenced NKL cells, then received IL-2 sq for 10 days to induce in vivo NK cell proliferation. NKL numbers in blood were subsequently analyzed by flow cytometry and tumor burden was assessed by luciferase-based bioluminescence imaging (BLI). At 16 and 21 days following adoptive NK cell transfer, BLI showed that recipients of NKG2A silenced NKL cells had slower tumor growth and significantly smaller tumor burden compared to NKL wt and NKL-GFP transduced controls (figure). Importantly, no toxicity related to infusing NKG2A inactivated NK cells was observed. These in vitro and in vivo data suggest shRNA knockdown of the NKG2A inhibitory receptor could be used as a method to augment NK cell tumor cytotoxicity in patients with hematological malignancies. Figure: Tumor burden in mice Luciferase-tagged 721.221 HLA-E EBV LCLs were injected into mice and imaged using a bioluminescence imager at days 10, 16, and 22 following NKL injection. 5 mice were followed in each group. Figure:. Tumor burden in mice . / Luciferase-tagged 721.221 HLA-E EBV LCLs were injected into mice and imaged using a bioluminescence imager at days 10, 16, and 22 following NKL injection. 5 mice were followed in each group. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document