scholarly journals Hypoxia-Driven HIF-1α Activation Reprograms Pre-Activated NK Cells towards Highly Potent Effector Phenotypes via ERK/STAT3 Pathways

Cancers ◽  
2021 ◽  
Vol 13 (8) ◽  
pp. 1904
Author(s):  
Seon Ah Lim ◽  
Yunwon Moon ◽  
Min Hwa Shin ◽  
Tae-Jin Kim ◽  
Sehyun Chae ◽  
...  

NK cells are the predominant innate lymphocyte subsets specialized to kill malignant tumor cells. In patients with advanced cancer, hypoxic stress shapes NK cells toward tumor-resistant and immunosuppressive phenotypes, hence a strategy to restore NK function is critical for successful tumor immunotherapy. Here, we present evidence that pre-activation and subsequent HIF-1α-dependent metabolic shift of NK cells from oxidative phosphorylation into glycolysis are keys to overcome hypoxia-mediated impairment in NK cell survival, proliferation, and tumor cytotoxicity. Specifically, exposing NK cells to 7–9 days of normoxic culture followed by a pO2 of 1.5% hypoxia led to a highly potent effector phenotype via HIF-1α stabilization and upregulation of its target genes, BNIP3, PDK1, VEGF, PKM2, and LDHA. RNA sequencing and network analyses revealed that concomitant reduction of p21/p53 apoptotic pathways along with upregulation of cell cycle-promoting genes, CCNE1, CDC6, CDC20, and downregulation of cell cycle-arrest genes, CDKN1A, GADD45A, and MDM2 were accountable for superior expansion of NK cells via ERK/STAT3 activation. Furthermore, HIF-1α-dependent upregulation of the NKp44 receptor in hypoxia-exposed NK cells resulted in increased killing against K562, CEM, and A375 tumor targets both in-vitro and in-vivo tumor clearance assays. Therefore, hypoxic exposure on pre-activated proliferating NK cells triggered HIF-1α-dependent pathways to initiate coordinated regulation of cell cycle, apoptosis, and cytotoxicity at the global gene transcription level. Our results uncover a previously unidentified role of HIF-1α-mediated metabolic reprogramming that can reverse impaired NK effector phenotypes to generate requisite numbers of functionally robust NK cells for adoptive cellular therapy for clinical evaluation.

2020 ◽  
Vol 6 (4) ◽  
pp. 231
Author(s):  
Win Mar Soe ◽  
Joan Hui Juan Lim ◽  
David L. Williams ◽  
Jessamine Geraldine Goh ◽  
Zhaohong Tan ◽  
...  

Invasive aspergillosis (IA) is a major opportunistic fungal infection in patients with haematological malignancies. Morbidity and mortality rates are high despite anti-fungal treatment, as the compromised status of immune system prevents the host from responding optimally to conventional therapy. This raises the consideration for immunotherapy as an adjunctive treatment. In this study, we evaluated the utility of expanded human NK cells as treatment against Aspergillus fumigatus infection in vitro and in vivo. The NK cells were expanded and activated by K562 cells genetically modified to express 4-1BB ligand and membrane-bound interleukin-15 (K562-41BBL-mbIL-15) as feeders. The efficacy of these cells was investigated in A. fumigatus killing assays in vitro and as adoptive cellular therapy in vivo. The expanded NK cells possessed potent killing activity at low effector-to-target ratio of 2:1. Fungicidal activity was morphotypal-dependent and most efficacious against A. fumigatus conidia. Fungicidal activity was mediated by dectin-1 receptors on the expanded NK cells leading to augmented release of perforin, resulting in enhanced direct cytolysis. In an immunocompromised mice pulmonary aspergillosis model, we showed that NK cell treatment significantly reduced fungal burden, hence demonstrating the translational potential of expanded NK cells as adjunctive therapy against IA in immunocompromised patients.


2019 ◽  
Vol 20 (3) ◽  
pp. 756 ◽  
Author(s):  
Gulnur Zakiryanova ◽  
Elena Kustova ◽  
Nataliya Urazalieva ◽  
Emile Baimuchametov ◽  
Narymzhan Nakisbekov ◽  
...  

Natural killer (NK) cells have received a lot of attention in recent years for the roles they play in immunity and particularly in antitumor immune responses. Although defects in NK cell functions are recognized as important mechanisms for immune evasion of malignant cells, molecular pathways regulating NK cell dysfunction and exhaustion in cancer are largely unknown. Here we tested whether the c-myc proto-oncogene, known to promote cell proliferation, growth, differentiation, and apoptosis by regulating the expression of numerous target genes, may be involved in the mechanism of NK cell abnormalities in patients with lung and gastric cancer. Analysis of c-myc mRNA and protein expression in peripheral blood NK cells, mitogen-activated protein kinase (MAPK) activity, cell cycle, and cell longevity revealed a significantly decreased expression of c-myc mRNA and protein and mitotic arrest of NK cells in different phases of cell cycle. In addition, a significant decrease of NK cell death was also detected. These data allow the suggestion that defects of NK cell-mediated tumor surveillance may be associated with disturbed c-myc expression in NK cells in cancer patients. A better understanding of the mechanisms of NK cell dysfunction in cancer will help in the NK cell-mediated therapeutic eradication of primary and metastatic cancer cells and prolong patient survival.


2018 ◽  
Vol 19 (12) ◽  
pp. 3837 ◽  
Author(s):  
Kevin Sek ◽  
Christina Mølck ◽  
Gregory Stewart ◽  
Lev Kats ◽  
Phillip Darcy ◽  
...  

The immune system plays a major role in the surveillance and control of malignant cells, with the presence of tumor infiltrating lymphocytes (TILs) correlating with better patient prognosis in multiple tumor types. The development of ‘checkpoint blockade’ and adoptive cellular therapy has revolutionized the landscape of cancer treatment and highlights the potential of utilizing the patient’s own immune system to eradicate cancer. One mechanism of tumor-mediated immunosuppression that has gained attention as a potential therapeutic target is the purinergic signaling axis, whereby the production of the purine nucleoside adenosine in the tumor microenvironment can potently suppress T and NK cell function. The production of extracellular adenosine is mediated by the cell surface ectoenzymes CD73, CD39, and CD38 and therapeutic agents have been developed to target these as well as the downstream adenosine receptors (A1R, A2AR, A2BR, A3R) to enhance anti-tumor immune responses. This review will discuss the role of adenosine and adenosine receptor signaling in tumor and immune cells with a focus on their cell-specific function and their potential as targets in cancer immunotherapy.


Blood ◽  
2020 ◽  
Vol 136 (21) ◽  
pp. 2416-2427 ◽  
Author(s):  
Meisam Naeimi Kararoudi ◽  
Yuya Nagai ◽  
Ezgi Elmas ◽  
Marcelo de Souza Fernandes Pereira ◽  
Syed Abbas Ali ◽  
...  

Abstract Multiple myeloma (MM) is a plasma cell neoplasm that commonly expresses CD38. Daratumumab (DARA), a human monoclonal antibody targeting CD38, has significantly improved the outcome of patients with relapsed or refractory MM, but the response is transient in most cases. Putative mechanisms of suboptimal efficacy of DARA include downregulation of CD38 expression and overexpression of complement inhibitory proteins on MM target cells as well as DARA-induced depletion of CD38high natural killer (NK) cells resulting in crippled antibody-dependent cellular cytotoxicity (ADCC). Here, we tested whether maintaining NK cell function during DARA therapy could maximize DARA-mediated ADCC against MM cells and deepen the response. We used the CRISPR/Cas9 system to delete CD38 (CD38KO) in ex vivo expanded peripheral blood NK cells. These CD38KO NK cells were completely resistant to DARA-induced fratricide, showed superior persistence in immune-deficient mice pretreated with DARA, and enhanced ADCC activity against CD38-expressing MM cell lines and primary MM cells. In addition, transcriptomic and cellular metabolic analysis demonstrated that CD38KO NK cells have unique metabolic reprogramming with higher mitochondrial respiratory capacity. Finally, we evaluated the impact of exposure to all-trans retinoic acid (ATRA) on wild-type NK and CD38KO NK cell function and highlighted potential benefits and drawbacks of combining ATRA with DARA in patients with MM. Taken together, these findings provide proof of concept that adoptive immunotherapy using ex vivo expanded CD38KO NK cells has the potential to boost DARA activity in MM.


Cancers ◽  
2020 ◽  
Vol 12 (8) ◽  
pp. 2131 ◽  
Author(s):  
S. Elizabeth Franks ◽  
Benjamin Wolfson ◽  
James W. Hodge

Cellular therapy has emerged as an attractive option for the treatment of cancer, and adoptive transfer of chimeric antigen receptor (CAR) expressing T cells has gained FDA approval in hematologic malignancy. However, limited efficacy was observed using CAR-T therapy in solid tumors. Natural killer (NK) cells are crucial for tumor surveillance and exhibit potent killing capacity of aberrant cells in an antigen-independent manner. Adoptive transfer of unmodified allogeneic or autologous NK cells has shown limited clinical benefit due to factors including low cell number, low cytotoxicity and failure to migrate to tumor sites. To address these problems, immortalized and autologous NK cells have been genetically engineered to express high affinity receptors (CD16), CARs directed against surface proteins (PD-L1, CD19, Her2, etc.) and endogenous cytokines (IL-2 and IL-15) that are crucial for NK cell survival and cytotoxicity, with positive outcomes reported by several groups both preclinically and clinically. With a multitude of NK cell-based therapies currently in clinic trials, it is likely they will play a crucial role in next-generation cell therapy-based treatment. In this review, we will highlight the recent advances and limitations of allogeneic, autologous and genetically enhanced NK cells used in adoptive cell therapy.


2020 ◽  
Vol 57 (4) ◽  
pp. 175-184
Author(s):  
Simona Caruso ◽  
Biagio De Angelis ◽  
Simona Carlomagno ◽  
Francesca Del Bufalo ◽  
Simona Sivori ◽  
...  

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2637-2637 ◽  
Author(s):  
Wee-Joo Chng ◽  
Junli Yan ◽  
Gaofeng Huang ◽  
Viknesvaran Selvarajan ◽  
Jim Tay ◽  
...  

Abstract Abstract 2637 Background: Extranodal nasal-type Natural Killer/T-cell lymphoma (NKTL) is a relatively rare but aggressive type of non-Hodgkins lymphoma that is more prevalent in Asia. The outcome of patients with disseminated stage is universally fatal. Progress in therapy has been slow and is based on combination of chemotherapy. MicroRNA are short non-coding RNA sequences that could regulate the expression of a large number of genes by inhibiting translation or leading to mRNA degradation. It has been implicated in tumorigenesis and has prognostic value across a wide range of malignancies including haematologic malignancies. We performed a comprehensive genome-wide miRNA expression profiling (MEP) of NKTL to identify deregulated miRNA and their potential role in NKTL biology. Method: MEP was performed using the Agilent human miRNA Microarray V2 (Agilent Technologies, Santa Clara, CA) on formalin fixed paraffin-embedded tissue (FFPE) (n=30) and NK cell lines (n=6) in comparison with normal NK cells. Differential expressed miRNA were identified using fold change and Significance Analysis of Microarray (SAM). Some of the differentially expressed miRNA were validated using quantitative polymerase chain reaction (q-PCR). The functional relevance of candidate miRNAs are assessed using miRNA mimics or inhibitors, and observing for apoptosis and growth arrest in the cell lines. Potential targets of candidate miRNAs are identified using predictive algorithms and significant negative correlation with gene expression data. The strongest candidate target genes are further verified using luciferase assay and q-PCR. miRNA and target gene relationship was further confirmed in the patients samples using immunohistochemistry for the protein expression on tissue microarray of NKTL. Results: Compared to normal NK cells, differentially expressed miRNAs in NKTL are predominantly downregulated. Re-expression of downregulated miRNAs, such as mir-101, mir-26a, mir26b, mir-28-5 and mir-363, reduced the growth of NK cell line and modulated the expression of their predicted target genes, suggesting the potential functional role of the deregulated miRNAs in the oncogenesis of NKTL. Taken together, the predicted targets whose expression are inversely correlated with the expression of deregulated miRNA in NKTL are significant enriched for genes involved in cell cycle-related, p53 and MAPK signaling pathways. We validated and confirmed the regulation of STMN1, and BLIMP1 by miR-101 and miR-30b respectively. In addition, miR-101, miR26a and miR-26b also affect the expression of BCL2 and IGF-1. We also performed immunohistochemical validation for selected target proteins and found over-expression of MUM1, BLIMP1 and STMN1 in NKTL, and notably, a corresponding increase in MYC expression. Conclusion: miRNA are dysregulated in NKTL. Since MYC is known to cause repression of miRNA expression, it is possible that MYC activation in NKTL as we have shown previously may contribute to the suppression of the miRNAs. These suppressed miRNA in turn lead to increase and aberrant expression of proteins and pathways of biological relevance to NKTL including cell cycle related genes, genes involved in p53 and MAPK signaling pathways as well as MUM1, BLIMP1 and STMN1. Reintroduction of these suppressed miRNA lead to death of NKTL cells and may be a potential therapeutic strategy. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2536-2536
Author(s):  
Xuxiang Liu ◽  
Yunfei Shi ◽  
Yuping Li ◽  
Ru Chen ◽  
Sheng Pan ◽  
...  

Natural killer (NK) cells are innate lymphocytes responsible for early defense against infections and malignant cells. Specific transcription factors crucial for NK cell development and function include PRDM1, which also regulates T cell homeostasis and is essential for the terminal differentiation of B cells into plasma cells. Importantly, PRDM1 deletion, methylation and loss-of-function mutations were commonly detected in NK cell malignancies. However, the detailed mechanisms through which PRDM1 regulates NK cell homeostasis are still largely undefined. Here, we employed an in vitro culture system of human NK cells isolated from healthy donors, in which the NK cells were cultured in the presence of IL-2 with or without an engineered feeder cell line, K562-Cl9-mb21, expressing membrane-bound IL-21, 4-1BBL and CD86. The NK cells were able to expand for months when co-cultured with feeder cells, whereas IL-2 alone could only maintain NK cell survival with limited proliferation for one week. We performed ChIP-seq to compare the genome-wide binding profiles of PRDM1 in NK cells grown with or without feeder cells. We found that PRDM1 bound much fewer target genes (802) in NK cells with feeder than in NK cells with IL-2 alone (2880), and 98.5% (790/802) of the binding sites in feeder-stimulated NK cells overlapped with those found in NK cells without feeder. The PRDM1 consensus motifs were almost identical in both conditions. MEME analyses also identified motifs of other transcription factors enriched in the PRDM1 binding sites, such as the RUNX and T-Box families. Interestingly, the RUNX and T-Box motifs, among others, were more enriched in the PRDM1 binding sites lacking the PRDM1 motif than in those with the PRDM1 motif, which indicates that PRDM1 may often bind to DNA indirectly through other transcription factors. PRDM1 bound a large number of genes in the NK or T cell receptor signaling pathway, which are employed during NK cell activation. In addition, several genes encoding immune checkpoints that may restrict NK cell activation such as TIGIT, HAVCR2 (TIM3), and IL-1R8 were targeted by PRDM1. Many genes encoding NK cell inhibitory and activating receptors were also bound by PRDM1. Moreover, PRDM1 was found to target transcription factors that are important for NK cell development and homeostasis, such as the RUNX family, TBX21, MAF, and PRDM1 itself. Despite an extensive overlap of PRDM1 binding sites detected in NK cells grown with or without feeder cells, the most enriched pathways were not exactly the same. Importantly, our RNA-seq data on PRDM1-knockout NK cells validated the regulatory role of PRDM1 on a fraction of these PRDM1-bound genes and interestingly, PRDM1 appeared to be an activator for some of the genes, including those encoding immune checkpoint molecules TIGIT, HAVCR2, and IL-1R8. We also utilized ATAC-seq to examine the chromatin accessibility of NK cells grown with feeder cells or with IL-2 alone. We identified differentially enriched pathways for the NK cells cultured under different conditions. When compared with the PRDM1 ChIP-seq, we found that those PRDM1 binding sites that contain a consensus PRDM1 motif were less likely to be accessible to transposase than those without the PRDM1 motif, thereby confirming the transcriptional repressor role of PRDM1. To further understand how PRDM1 regulates its target genes in NK cells, we performed mass spectrometric analysis on the protein complexes associated with PRDM1. We were able to identify the interaction of PRDM1 with the corepressor Groucho (TLE3) and components from the SIN3, NCoR, and NuRD complex, which have been previously reported in mouse plasma cells. Surprisingly, the protein compositions of the PRDM1-associated complexes from the two NK cell populations were very different. Although PRDM1 is generally associated with transcriptional repression, we also detected the association of PRDM1 with transcriptional activators or coactivators, such as the RUNX-binding protein CBFβ and the T-Box family member EOMES, which may thereby upregulate some of the PRDM1 target genes. In summary, we found that PRDM1 binds and regulates an extensive network of genes responsible for NK cell activation and function. Extrinsic stimuli, as provided by the feeder cells, can alter the extent and profile of PRDM1 binding as well as the associated protein complexes and hence alter its regulatory function in NK cells. Disclosures No relevant conflicts of interest to declare.


2018 ◽  
Author(s):  
Kristin M. Snyder ◽  
Robert Hullsiek ◽  
Hemant K Mishra ◽  
Daniel C. Mendez ◽  
Yunfang Li ◽  
...  

AbstractAnti-tumor mAbs are the most widely used and characterized cancer immunotherapy agents. Despite having a significant impact on some malignancies, most cancer patients respond poorly or develop resistance to this therapy. A known mechanism of action of these therapeutic mAbs is antibody-dependent cell-mediated cytotoxicity (ADCC), which is a primarily effector function of NK cells. CD16A on human NK cells has an exclusive role in binding to tumor-bound IgG antibodies. Though CD16A is a potent activating receptor, it is a low affinity FcγR and its cell surface levels can be rapidly downregulated by a proteolytic process involving ADAM17 upon NK cell activation, which are likely to limit the efficacy of tumor-targeting therapeutic mAbs in the tumor environment. We sought to enhance NK cell binding to anti-tumor mAbs by engineering these cells with a recombinant FcγR consisting of the extracellular region of CD64, the highest affinity IgG Fc receptor expressed by leukocytes, and the transmembrane and cytoplasmic regions of CD16A. This novel recombinant FcγR (CD64/16A) was expressed in the human NK cell line NK92 and in induced pluripotent stem cells from which primary NK cells were derived. CD64/16A also lacked the ADAM17 cleavage region in CD16A and it was not rapidly downregulated in expression following NK cell activation during ADCC. CD64/16A on NK cells facilitated conjugation to antibody-treated tumor cells, ADCC, and cytokine production, demonstrating functional activity by its two components. Unlike NK cells expressing CD16A, CD64/16A captured soluble therapeutic mAbs and the modified NK cells mediated tumor cell killing. Hence, CD64/16A could potentially be used as a docking platform on engineered NK cells for therapeutic mAbs and IgG Fc chimeric proteins, allowing for switchable targeting elements, and a novel cancer cellular therapy.


Sign in / Sign up

Export Citation Format

Share Document