Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunother Cancer ; 12(7)2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39032940

ABSTRACT

BACKGROUND: Human and mouse natural killer (NK) cells have been shown to develop memory-like function after short-term exposure to the cocktail of IL-12/15/18 or to overnight co-culture with some tumor cell lines. The resulting cells retain enhanced lytic ability for up to 7 days as well as after cryopreservation, and memory-like NK cells (mlNK) have been shown to induce complete remissions in patients with hematological malignancies. No single phenotype has been described for mlNK and the physiological changes induced by the short-term cytokine or tumor-priming which are responsible for these enhanced functions have not been fully characterized. Here, we have generated mlNK by cytokine and tumor-priming to find commonalities to better define the nature of NK cell "memory" in vitro and, for the first time, in vivo. METHODS: We initiated mlNK in vitro from healthy donors with cytokines (initiated cytokine-induced memory-like (iCIML)-NK) and by tumor priming (TpNK) overnight and compared them by high-dimensional flow cytometry, proteomic and metabolomic profiling. As a potential mechanism of enhanced cytolytic function, we analyzed the avidity of binding of the mlNK to NK-resistant tumors (z-Movi). We generated TpNK from healthy donors and from cancer patients to determine whether mlNK generated by interaction with a single tumor type could enhance lytic activity. Finally, we used a replication-incompetent tumor cell line (INKmune) to treat patients with myeloid leukaemias to potentiate NK cell function in vivo. RESULTS: Tumor-primed mlNK from healthy donors and patients with cancer showed increased cytotoxicity against multiple tumor cell lines in vitro, analogous to iCIML-NK cells. Multidimensional cytometry identified distinct memory-like profiles of subsets of cells with memory-like characteristics; upregulation of CD57, CD69, CD25 and ICAM1. Proteomic profiling identified 41 proteins restricted to mlNK cells and we identified candidate molecules for the basis of NK memory which can explain how mlNK overcome inhibition by resistant tumors. Finally, of five patients with myelodysplastic syndrome or refractory acute myeloid leukemia treated with INKmune, three responded to treatment with measurable increases in NK lytic function and systemic cytokines. CONCLUSIONS: NK cell "memory" is a physiological state associated with resistance to MHC-mediated inhibition, increased metabolic function, mitochondrial fitness and avidity to NK-resistant target cells.


Subject(s)
Immunologic Memory , Killer Cells, Natural , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Neoplasms/immunology , Neoplasms/therapy , Proteomics/methods , Cytokines/metabolism , Cell Line, Tumor , Immunotherapy/methods , Phenotype , Mice
2.
Cancers (Basel) ; 16(6)2024 Mar 18.
Article in English | MEDLINE | ID: mdl-38539519

ABSTRACT

BACKGROUND: Tissue-specificity for fimbrial fallopian tube ovarian carcinogenesis remains largely unknown in BRCA1 mutation carriers. We aimed to assess the cell autonomous and cell-nonautonomous implications of a germline BRCA1 mutation in the context of cancer immunosurveillance of CD3- CD56+ natural killer (NK) cells. METHODS: Premenopausal BRCA1 mutation carriers versus age-matched non-carriers were compared. Daily urinary 5ß-pregnanediol levels were used to determine progesterone metabolomics across an ovarian cycle. Using peripherally acquired NK cells the cell-mediated cytotoxicity of tumor targets (OVCAR-3, K-562) was determined using live cellular impedance (xCELLigence®) and multicolor flow cytometry. Hypoxia-inducible factor 1-alpha (HIF-1α) immunohistochemistry of cancer-free fallopian tube specimens allowed a comparison of proximal versus distal portions. Utilizing these findings the role of environmental factors relevant to the fimbrial fallopian tube (progesterone, hypoxia) on NK cell functional activity were studied in an ovarian phase-specific manner. RESULTS: BRCA1 mutation carriers demonstrate a differential progesterone metabolome with a phase-specific reduction of peripheral NK cell functional activity. Progesterone exposure further impairs NK cell-mediated cytotoxicity in a dose-dependent manner, which is reversed with the addition of mifepristone (1.25 µM). The fimbrial fallopian tube demonstrated significantly higher HIF-1α staining, particularly in BRCA1 mutation carriers, reflecting a site-specific 'hypoxic niche'. Exposure to hypoxic conditions (1% O2) can further impair tumor cytotoxicity in high-risk carriers. CONCLUSIONS: Phase-specific differential NK cell activity in BRCA1 mutation carriers, either systemically or locally, may favor site-specific pre-invasive carcinogenesis. These cumulative effects across a reproductive lifecycle in high-risk carriers can have a detrimental effect further supporting epidemiological evidence for ovulation inhibition.

3.
Cells ; 12(4)2023 02 16.
Article in English | MEDLINE | ID: mdl-36831300

ABSTRACT

Myelodysplastic syndrome (MDS) treatment remains a big challenge due to the heterogeneous nature of the disease and its ability to progress to acute myeloid leukemia (AML). The only curative option is allogeneic hematopoietic stem cell transplantation (HSCT), but most patients are unfit for this procedure and are left with only palliative treatment options, causing a big unmet need in the context of this disease. Natural killer (NK) cells are attractive candidates for MDS immunotherapy due to their ability to target myeloid leukemic cells without prior sensitization, and in recent years we have seen an arising number of clinical trials in AML and, recently, MDS. NK cells are reported to be highly dysfunctional in MDS patients, which can be overcome by adoptive NK cell immunotherapy or activation of endogenous NK cells. Here, we review the role of NK cells in MDS, the contribution of the tumor microenvironment (TME) to NK cell impairment, and the most recent data from NK cell-based clinical trials in MDS.


Subject(s)
Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Humans , Myelodysplastic Syndromes/pathology , Killer Cells, Natural , Leukemia, Myeloid, Acute/pathology , Immunotherapy, Adoptive , Transplantation, Homologous , Tumor Microenvironment
4.
PLoS Comput Biol ; 17(3): e1008789, 2021 03.
Article in English | MEDLINE | ID: mdl-33711017

ABSTRACT

We introduce poly-adenine CRISPR gRNA-based single-cell RNA-sequencing (pAC-Seq), a method that enables the direct observation of guide RNAs (gRNAs) in scRNA-seq. We use pAC-Seq to assess the phenotypic consequences of CRISPR/Cas9 based alterations of gene cis-regulatory regions. We show that pAC-Seq is able to detect cis-regulatory-induced alteration of target gene expression even when biallelic loss of target gene expression occurs in only ~5% of cells. This low rate of biallelic loss significantly increases the number of cells required to detect the consequences of changes to the regulatory genome, but can be ameliorated by transcript-targeted sequencing. Based on our experimental results we model the power to detect regulatory genome induced transcriptomic effects based on the rate of mono/biallelic loss, baseline gene expression, and the number of cells per target gRNA.


Subject(s)
CRISPR-Cas Systems/genetics , Regulatory Elements, Transcriptional/genetics , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Transcriptome/genetics , Algorithms , Animals , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Computational Biology , Databases, Factual , Humans , Mice , RNA, Guide, Kinetoplastida/genetics
5.
Stem Cells Transl Med ; 9(9): 974-984, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32416056

ABSTRACT

Adoptive cell therapy (ACT) is an approach to cancer treatment that involves the use of antitumor immune cells to target residual disease in patients after completion of chemo/radiotherapy. ACT has several advantages compared with other approaches in cancer immunotherapy, including the ability to specifically expand effector cells in vitro before selection for adoptive transfer, as well as the opportunity for host manipulation in order to enhance the ability of transferred cells to recognize and kill established tumors. One of the main challenges to the success of ACT in cancer clinical trials is the identification and generation of antitumor effector cells with high avidity for tumor recognition. Natural killer (NK) cells, cytokine-induced killers and natural killer T cells are key innate or innate-like effector cells in cancer immunosurveillance that act at the interface between innate and adaptive immunity, to have a greater influence over immune responses to cancer. In this review, we discuss recent studies that highlight their potential in cancer therapy and summarize clinical trials using these effector immune cells in adoptive cellular therapy for the treatment of cancer.


Subject(s)
Adoptive Transfer , Immunity, Innate , Neoplasms/immunology , Neoplasms/therapy , Animals , Clinical Trials as Topic , Genetic Techniques , Humans , Killer Cells, Natural/immunology
6.
PLoS One ; 14(6): e0218674, 2019.
Article in English | MEDLINE | ID: mdl-31242243

ABSTRACT

An emerging cellular immunotherapy for cancer is based on the cytolytic activity of natural killer (NK) cells against a wide range of tumors. Although in vitro activation, or "priming," of NK cells by exposure to pro-inflammatory cytokines, such as interleukin (IL)-2, has been extensively studied, the biological consequences of NK cell activation in response to target cell interactions have not been thoroughly characterized. We investigated the consequences of co-incubation with K562, CTV-1, Daudi RPMI-8226, and MCF-7 tumor cell lines on the phenotype, cytokine expression profile, and transcriptome of human NK cells. We observe the downregulation of several activation receptors including CD16, CD62L, C-X-C chemokine receptor (CXCR)-4, natural killer group 2 member D (NKG2D), DNAX accessory molecule (DNAM)-1, and NKp46 following tumor-priming. Although this NK cell phenotype is typically associated with NK cell dysfunction in cancer, we reveal the upregulation of NK cell activation markers, such as CD69 and CD25; secretion of pro-inflammatory cytokines, including macrophage inflammatory proteins (MIP-1) α /ß and IL-1ß/6/8; and overexpression of numerous genes associated with enhanced NK cell cytotoxicity and immunomodulatory functions, such as FAS, TNFSF10, MAPK11, TNF, and IFNG. Thus, it appears that tumor-mediated ligation of receptors on NK cells may induce a primed state which may or may not lead to full triggering of the lytic or cytokine secreting machinery. Key signaling molecules exclusively affected by tumor-priming include MAP2K3, MARCKSL1, STAT5A, and TNFAIP3, which are specifically associated with NK cell cytotoxicity against tumor targets. Collectively, these findings help define the phenotypic and transcriptional signature of NK cells following their encounters with tumor cells, independent of cytokine stimulation, and provide insight into tumor-specific NK cell responses to inform the transition toward harnessing the therapeutic potential of NK cells in cancer.


Subject(s)
Killer Cells, Natural/immunology , Neoplasms/immunology , Cell Line, Tumor , Cytokines/genetics , Cytokines/immunology , Cytotoxicity, Immunologic , Gene Regulatory Networks , Humans , Immunotherapy , Inflammation Mediators/metabolism , K562 Cells , Killer Cells, Natural/metabolism , Lymphocyte Activation , MCF-7 Cells , Neoplasms/genetics , Neoplasms/therapy , Phenotype , Transcriptome
7.
Front Immunol ; 4: 408, 2013 Nov 25.
Article in English | MEDLINE | ID: mdl-24324471

ABSTRACT

The functional impairment of natural killer (NK) cells has been frequently reported in cancer studies. As one of the central components of host anti-tumor immunity, NK cells exert cellular cytotoxicity against tumor cells, and secrete a cytokine milieu to inhibit tumor progression and enable the recruitment of other immune cells to the tumor site. The unlocking of the full functional potential of NK cells requires successful progression through discrete activation stages that are tightly regulated by a complex array of signaling molecules. Target cell susceptibility to NK cell-mediated killing is dependent on the intensity and specific combination of ligand expression for NK cell receptors. Tumor cells utilize numerous strategies for evading NK cells, including the downregulation of important NK cell-activating ligands. Here, we review key studies on NK cell activation requirements, and argue, based on our findings from NK cell-tumor interactions, that the altered characteristics of tumor-associated NK cells are indicative of unmet signaling requirements for full NK cell activation, rather than NK cell dysfunction in cancer.

8.
J Immunol ; 187(12): 6227-34, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22084431

ABSTRACT

Resting human NK cells require a two-stage activation process that we have previously described as "priming" and "triggering." NK-sensitive tumor cells provide both priming and triggering signals. NK-resistant tumors evade lysis, mostly by failure to prime; however, we recently reported a tumor cell line (CTV-1) that primes resting NK cells but fails to trigger lysis. In this article, we report two additional leukemia cell lines that prime NK cells but are resistant to lysis. Tumor-mediated NK priming is via CD2 binding to a ligand within CD15 on the tumor cell. NK-resistant RAJI cells became susceptible to NK lysis following transfection and expression of CD15. Blockade of CD15 on K562 cells or on CD15(+) RAJI cells significantly inhibited lysis, as did blockade of CD2 on resting NK cells. NK priming via CD2 induced CD16 shedding, releasing CD3ζ to the CD2, leading to its phosphorylation and the subsequent phosphorylation of linker for activation of T cells and STAT-5 and synthesis of IFN-γ. Blockade of C-type lectin receptors significantly suppressed the tumor-mediated priming of NK cells, whereas blockade of Ig-superfamily-like receptors had no effect at the NK-priming stage. Tumor priming of resting NK cells was irrespective of HLA expression, and blockade of HLA-killer Ig-like receptor interactions did not influence the incidence or degree of priming. However, CD15-CD2 interactions were critical for NK priming and were required, even in the absence of HLA-mediated NK inhibition. Tumor-mediated priming led to a sustained primed state, and the activated NK cells retained the ability to lyse NK-resistant tumors, even after cryopreservation.


Subject(s)
CD2 Antigens/metabolism , Cytotoxicity, Immunologic , Fucosyltransferases/physiology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lewis X Antigen/physiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Receptors, KIR/physiology , Resting Phase, Cell Cycle/immunology , Cell Differentiation/immunology , Cell Line, Tumor , Cell Lineage/immunology , Coculture Techniques , Disease Resistance , Fucosyltransferases/metabolism , Humans , Killer Cells, Natural/cytology , Leukemia, Monocytic, Acute/immunology , Leukemia, Monocytic, Acute/metabolism , Leukemia, Monocytic, Acute/pathology , Leukemia, Myelomonocytic, Acute/immunology , Leukemia, Myelomonocytic, Acute/metabolism , Leukemia, Myelomonocytic, Acute/pathology , Lewis X Antigen/metabolism , Ligands , Lymphocyte Activation/immunology , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/metabolism , Lymphoma, Non-Hodgkin/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Binding/immunology , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL