Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Hematol Oncol ; 15(1): 164, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36348457

ABSTRACT

Natural killer (NK) cells are unique immune effectors able to kill cancer cells by direct recognition of surface ligands, without prior sensitization. Allogeneic NK transfer is a highly valuable treatment option for cancer and has recently emerged with hundreds of clinical trials paving the way to finally achieve market authorization. Advantages of NK cell therapies include the use of allogenic cell sources, off-the-shelf availability, and no risk of graft-versus-host disease (GvHD). Allogeneic NK cell therapies have reached the clinical stage as ex vivo expanded and differentiated non-engineered cells, as chimeric antigen receptor (CAR)-engineered or CD16-engineered products, or as combination therapies with antibodies, priming agents, and other drugs. This review summarizes the recent clinical status of allogeneic NK cell-based therapies for the treatment of hematological and solid tumors, discussing the main characteristics of the different cell sources used for NK product development, their use in cell manufacturing processes, the engineering methods and strategies adopted for genetically modified products, and the chosen approaches for combination therapies. A comparative analysis between NK-based non-engineered, engineered, and combination therapies is presented, examining the choices made by product developers regarding the NK cell source and the targeted tumor indications, for both solid and hematological cancers. Clinical trial outcomes are discussed and, when available, assessed in comparison with preclinical data. Regulatory challenges for product approval are reviewed, highlighting the lack of specificity of requirements and standardization between products. Additionally, the competitive landscape and business field is presented. This review offers a comprehensive overview of the effort driven by biotech and pharmaceutical companies and by academic centers to bring NK cell therapies to pivotal clinical trial stages and to market authorization.


Subject(s)
Hematologic Neoplasms , Neoplasms , Receptors, Chimeric Antigen , Humans , Killer Cells, Natural , Immunotherapy, Adoptive/methods
2.
J Neuroimmunol ; 347: 577353, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32745802

ABSTRACT

This study aims to compare NK cells obtained from multiple sclerosis (MS) patients receiving interferon-ß1 and fingolimod therapies. Fingolimod reduced the CD56bright NK cell subset. The remaining CD56dim NK cells displayed NKG2D, NKp46, CD107a, and IFN-γ levels similar to those from the patients under interferon-ß1 therapy. Alternatively, comparative transcriptomics and pathway analyses revealed significant distinctions between two therapy modalities. Molecular signature of the CD56dim NK cells from fingolimod-treated MS patients was closely associated to those from healthy subjects. The basic assets of NK cells were modestly influenced by interferon-ß1 and fingolimod, however transcriptomics showed profound alterations in NK responses.


Subject(s)
Fingolimod Hydrochloride/therapeutic use , Interferon-beta/therapeutic use , Killer Cells, Natural/immunology , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/immunology , Transcriptome/physiology , Adult , Female , Fingolimod Hydrochloride/pharmacology , Humans , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Interferon-beta/pharmacology , K562 Cells , Killer Cells, Natural/drug effects , Male , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/genetics , Transcriptome/drug effects , Young Adult
3.
Front Immunol ; 11: 40, 2020.
Article in English | MEDLINE | ID: mdl-32082316

ABSTRACT

Sarcomas are malignancies of mesenchymal origin that occur in bone and soft tissues. Many are chemo- and radiotherapy resistant, thus conventional treatments fail to increase overall survival. Natural Killer (NK) cells exert anti-tumor activity upon detection of a complex array of tumor ligands, but this has not been thoroughly explored in the context of sarcoma immunotherapy. In this study, we investigated the NK cell receptor/ligand immune profile of primary human sarcoma explants. Analysis of tumors from 32 sarcoma patients identified the proliferative marker PCNA and DNAM-1 ligands CD112 and/or CD155 as commonly expressed antigens that could be efficiently targeted by genetically modified (GM) NK cells. Despite the strong expression of CD112 and CD155 on sarcoma cells, characterization of freshly dissociated sarcomas revealed a general decrease in tumor-infiltrating NK cells compared to the periphery, suggesting a defect in the endogenous NK cell response. We also applied a functional screening approach to identify relevant NK cell receptor/ligand interactions that induce efficient anti-tumor responses using a panel NK-92 cell lines GM to over-express 12 different activating receptors. Using GM NK-92 cells against primary sarcoma explants (n = 12) revealed that DNAM-1 over-expression on NK-92 cells led to efficient degranulation against all tested explants (n = 12). Additionally, NKG2D over-expression showed enhanced responses against 10 out of 12 explants. These results show that DNAM-1+ or NKG2D+ GM NK-92 cells may be an efficient approach in targeting sarcomas. The degranulation capacity of GM NK-92 cell lines was also tested against various established tumor cell lines, including neuroblastoma, Schwannoma, melanoma, myeloma, leukemia, prostate, pancreatic, colon, and lung cancer. Enhanced degranulation of DNAM-1+ or NKG2D+ GM NK-92 cells was observed against the majority of tumor cell lines tested. In conclusion, DNAM-1 or NKG2D over-expression elicited a dynamic increase in NK cell degranulation against all sarcoma explants and cancer cell lines tested, including those that failed to induce a notable response in WT NK-92 cells. These results support the broad therapeutic potential of DNAM-1+ or NKG2D+ GM NK-92 cells and GM human NK cells for the treatment of sarcomas and other malignancies.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/metabolism , Killer Cells, Natural/immunology , Lymphocyte Activation/genetics , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Sarcoma/immunology , Transgenes , Adolescent , Adult , Aged , Aged, 80 and over , Cell Degranulation/genetics , Cell Degranulation/immunology , Cell Line, Tumor , Cell- and Tissue-Based Therapy/methods , Child , Child, Preschool , Cytotoxicity, Immunologic , Genetic Vectors , Humans , Immunotherapy, Adoptive/methods , Infant , Infant, Newborn , Ligands , Lymphocytes, Tumor-Infiltrating/immunology , Middle Aged , Receptors, Cell Surface/metabolism , Receptors, Virus/metabolism , Sarcoma/pathology , Young Adult
4.
Sci Rep ; 9(1): 11697, 2019 08 12.
Article in English | MEDLINE | ID: mdl-31406210

ABSTRACT

Interferon (IFN)-γ is the major mediator of anti-tumor immune responses; nevertheless, cancer cells use intrigue strategies to alter IFN-γ signaling and avoid elimination. Understanding the immune regulatory mechanisms employed by acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) cells upon exposure to IFN-γ is critical for development of immunotherapy and checkpoint blockade therapy approaches. This study aims to explore the influence of myeloid maturation on IFN-γ-induced PD-L1 and PD-L2 expression and on pro-leukemogenic transcription factor STAT3 signaling in AML and MDS. Stimulation of myeloid blasts' maturation by all-trans retinoic acid (ATRA) or 1α,25-dihydroxyvitamin D3 (vitamin D) increased the CD11b+ fraction that expressed PD-1 ligands in response to IFN-γ. Intriguingly, STAT3 pathway was potently induced by IFN-γ and strengthened upon prolonged exposure. Nonetheless, STAT3-mediated atypical IFN-γ signaling appeared as a negligible factor for PD-L1 and PD-L2 expression. These negative influences of IFN-γ could be alleviated by a small-molecule inhibitor of STAT3, stattic, which also inhibited the upregulation of PD-L1. In conclusion, induction of myeloid maturation enhances the responsiveness of AML and MDS cells to IFN-γ. However, these malignant myeloid cells can exploit both STAT3 pathway and PD-1 ligands to survive IFN-γ-mediated immunity and maintain secondary immune resistance.


Subject(s)
B7-H1 Antigen/immunology , Gene Expression Regulation, Leukemic , Interferon-gamma/immunology , Leukemia, Myeloid, Acute/immunology , Myelodysplastic Syndromes/immunology , Rho Guanine Nucleotide Exchange Factors/immunology , Aged , B7-H1 Antigen/genetics , CD11b Antigen/genetics , CD11b Antigen/immunology , Calcitriol/pharmacology , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , HL-60 Cells , Humans , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Programmed Cell Death 1 Ligand 2 Protein/genetics , Programmed Cell Death 1 Ligand 2 Protein/immunology , Rho Guanine Nucleotide Exchange Factors/genetics , Signal Transduction , THP-1 Cells , Tretinoin/pharmacology
5.
Eur J Immunol ; 49(8): 1278-1290, 2019 08.
Article in English | MEDLINE | ID: mdl-31054264

ABSTRACT

Introduction of Chimeric Antigen Receptors to NK cells has so far been the main practical method for targeting NK cells to specific surface antigens. In contrast, T cell receptor (TCR) gene delivery can supply large populations of cytotoxic T-lymphocytes (CTL) targeted against intracellular antigens. However, a major barrier in the development of safe CTL-TCR therapies exists, wherein the mispairing of endogenous and genetically transferred TCR subunits leads to formation of TCRs with off-target specificity. To overcome this and enable specific intracellular antigen targeting, we have tested the use of NK cells for TCR gene transfer to human cells. Our results show that ectopic expression of TCR α/ß chains, along with CD3 subunits, enables the functional expression of an antigen-specific TCR complex on NK cell lines NK-92 and YTS, demonstrated by using a TCR against the HLA-A2-restricted tyrosinase-derived melanoma epitope, Tyr368-377 . Most importantly, the introduction of a TCR complex to NK cell lines enables MHC-restricted, antigen-specific killing of tumor cells both in vitro and in vivo. Targeting of NK cells via TCR gene delivery stands out as a novel tool in the field of adoptive immunotherapy which can also overcome the major hurdle of "mispairing" in TCR gene therapy.


Subject(s)
Immunotherapy, Adoptive/methods , Killer Cells, Natural/physiology , Melanoma/therapy , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Chimeric Antigen/genetics , Antigens, Neoplasm/immunology , Cell Line , Cytotoxicity, Immunologic , HLA-A2 Antigen/metabolism , Humans , Killer Cells, Natural/transplantation , Melanoma/immunology , Monophenol Monooxygenase/immunology , Peptides/immunology , Protein Engineering
6.
Proc Natl Acad Sci U S A ; 116(11): 5055-5060, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30808808

ABSTRACT

MHC-I epitope presentation to CD8+ T cells is directly dependent on peptide loading and selection during antigen processing. However, the exact molecular bases underlying peptide selection and binding by MHC-I remain largely unknown. Within the peptide-loading complex, the peptide editor tapasin is key to the selection of MHC-I-bound peptides. Here, we have determined an ensemble of crystal structures of MHC-I in complex with the peptide exchange-associated dipeptide GL, as well as the tapasin-associated scoop loop, alone or in combination with candidate epitopes. These results combined with mutation analyses allow us to propose a molecular model underlying MHC-I peptide selection by tapasin. The N termini of bound peptides most probably bind first in the N-terminal and middle region of the MHC-I peptide binding cleft, upon which the peptide C termini are tested for their capacity to dislodge the tapasin scoop loop from the F pocket of the MHC-I cleft. Our results also indicate important differences in peptide selection between different MHC-I alleles.


Subject(s)
Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/metabolism , Membrane Transport Proteins/metabolism , Animals , Crystallography, X-Ray , HEK293 Cells , Humans , Leucine/genetics , Membrane Transport Proteins/chemistry , Mice, Knockout , Models, Molecular , Mutation/genetics , Protein Binding , Protein Structure, Secondary
7.
Electrophoresis ; 40(2): 315-321, 2019 01.
Article in English | MEDLINE | ID: mdl-30362576

ABSTRACT

Monocyte heterogeneity and its prevalence are revealed as indicator of several human diseases ranking from cardiovascular diseases to rheumatoid arthritis, chronic kidney diseases, autoimmune multiple sclerosis, and stroke injuries. When monocytes and macrophages are characterized and isolated with preserved genetic, phenotypic and functional properties, they can be used as label-free biomarkers for precise diagnostics and treatment of various diseases. Here, the dielectrophoretic responses of the monocytes and macrophages were examined. We present 3D carbon-electrode dielectrophoresis (carbon-DEP) as a separation tool for U937 monocytes and U937 monocyte-differentiated macrophages. The carbon-electrodes advanced the usability and throughput of DEP separation, presented wider electrochemical stability. Using the 3D carbon-DEP chip, we first identified the selective positive and negative DEP responses and specific crossover frequencies of monocytes and macrophages as their signatures for separation. The crossover frequency of monocytes and macrophages was 17 and 30 kHz, respectively. Next, we separated monocyte and macrophage subpopulations using their specific dielectrophoretic responses. Afterward, we used a fluorescence-activated cell sorter to confirm our results. Finally, we enriched 70% of monocyte cells from the mixed cell population, in other words, concentration of monocyte cells to macrophage cells was five times increased, using the 30-kHz, 10-Vpp electric field and 1 µL/min flow rate.


Subject(s)
Cell Separation/instrumentation , Electrophoresis/instrumentation , Macrophages/cytology , Monocytes/cytology , Carbon/chemistry , Cell Separation/methods , Electrodes , Electrophoresis/methods , Equipment Design , Humans , U937 Cells
8.
Mol Pharm ; 15(4): 1526-1533, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29481086

ABSTRACT

While reshaping their microenvironment, tumors are also capable of influencing systemic processes including myeloid cell production. Therefore, the tumor-induced myeloid cells, such as myeloid-derived suppressor cells (MDSCs), which are characterized with pro-cancer properties, became another target in order to increase the success of the therapy. This study evaluated the capacity of a novel dendrimeric drug delivery platform to eliminate tumor-induced myeloid cells. As described in a previous study by our research group, the anti-Flt1 antibody-conjugated polyethylene glycol (PEG)-cored poly(amidoamine) (PAMAM) dendrimers improved the efficacy of gemcitabine against pancreatic cancer. Here, the biodistribution studies showed that these dendrimeric structures accumulated in the compartments that became rich in myeloid cells in the pancreatic tumor-bearing mice. When gemcitabine was loaded into the dendrimer complexes, the number of myeloid cells was significantly reduced while the percentage distribution of granulocytic and monocytic myeloid cells was not always significantly altered. The CD11b+Ly6G-Ly6C+ monocytes were more severely affected by the treatments than CD11b+Ly6G+Ly6C+ granulocytes. Immune infiltration levels in the tumor tissue were also altered. Myeloid cells in the spleen and F4/80+ macrophages of the liver were protected. The compartments, such as the liver and the bone marrow, which are known to have high vascular endothelial growth factor (VEGF)-Flt1 pathway activity, were particularly targeted by gemcitabine when delivered through anti-Flt1 antibody-conjugated PAMAM dendrimers. In conclusion, chemotherapeutic agents complexed with dendrimers not only improve anticancer efficacy, but they also assist in the elimination of the tumor-induced myeloid cells.


Subject(s)
Dendrimers/chemistry , Deoxycytidine/analogs & derivatives , Myeloid Cells/drug effects , Pancreatic Neoplasms/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Animals , Cell Line, Tumor , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Drug Delivery Systems , Humans , Male , Mice , Mice, Nude , Polyethylene Glycols/chemistry , Tissue Distribution , Gemcitabine
9.
Immunology ; 149(4): 460-471, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27565576

ABSTRACT

To cope with immune responses, tumour cells implement elaborate strategies such as adaptive resistance and induction of T-cell exhaustion. T-cell exhaustion has been identified as a state of hyporesponsiveness that arises under continuous antigenic stimulus. Nevertheless, contribution of co-stimulatory molecules to T-cell exhaustion in cancer remains to be better defined. This study explores the role of myeloid leukaemia-derived co-stimulatory signals on CD4+ T helper (Th) cell exhaustion, which may limit anti-tumour immunity. Here, CD86 and inducible T-cell co-stimulator ligand (ICOS-LG) co-stimulatory molecules that are found on myeloid leukaemia cells supported Th cell activation and proliferation. However, under continuous stimulation, T cells co-cultured with leukaemia cells, but not with peripheral blood monocytes, became functionally exhausted. These in vitro-generated exhausted Th cells were defined by up-regulation of programmed cell death 1 (PD-1), cytotoxic T-lymphocyte antigen 4 (CTLA-4), lymphocyte activation gene 3 (LAG3) and T-cell immunoglobulin and mucin domain-containing protein 3 (TIM-3) inhibitory receptors. They were reluctant to proliferate upon re-stimulation and produced reduced amounts of interleukin-2 (IL-2), tumour necrosis factor-α (TNF-α) and interferon-γ (IFN-γ). Nonetheless, IL-2 supplementation restored the proliferation capacity of the exhausted Th cells. When the co-stimulation supplied by the myeloid leukaemia cells were blocked, the amount of exhausted Th cells was significantly decreased. Moreover, in the bone marrow aspirates from patients with acute myeloid leukaemia (AML) or myelodysplastic syndrome (MDS), a subpopulation of Th cells expressing PD-1, TIM-3 and/or LAG3 was identified together with CD86+ and/or ICOS-LG+ myeloid blasts. Collectively, co-stimulatory signals derived from myeloid leukaemia cells possess the capacity to facilitate functional exhaustion in Th cells.


Subject(s)
Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/physiology , Hepatitis A Virus Cellular Receptor 2/metabolism , Leukemia, Myeloid/immunology , Programmed Cell Death 1 Receptor/metabolism , Adult , Aged , Antigens, CD/genetics , Cell Line, Tumor , Coculture Techniques , Cytokines/metabolism , Female , Hepatitis A Virus Cellular Receptor 2/genetics , Humans , Immunosenescence , Male , Middle Aged , Programmed Cell Death 1 Receptor/genetics , Tumor Escape , Up-Regulation , Young Adult , Lymphocyte Activation Gene 3 Protein
10.
Anadolu Kardiyol Derg ; 5(1): 13-7, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15755695

ABSTRACT

OBJECTIVE: Because low density lipoprotein-cholesterol (LDL-C) is a modifiable risk factor for coronary artery disease (CAD), its routine measurement is recommended in the evaluation and management of hypercholesterolemia. Concentrations of LDL-C are commonly monitored by means of the Friedewald formula (FF), which provides a relative estimation of LDL-C concentration when the triglyceride (TGs) concentration is <200 mg/dl and there are no abnormal lipids. Because of the limitations of the Friedewald calculation, direct methods for an accurate quantification of LDL-C are needed. METHODS: We critically examined an immunoseparation method for direct assay of LDL-C in a comparison with FF. 1) We measured intraassay and interassay precision using quality-control sera and patient serum pools. Accuracy was evaluated from total error analyses. Sample stability was examined over 2 months. 2) The LDL-C levels obtained with direct assay were compared with those calculated by the FF in 47 randomly chosen patient samples. The samples were classified as group 1 (patients with TGs 60-308 mg/dl, n=25) and group 2 (patients with TGs 320-695 mg/dl, n=22). RESULTS: The direct immunoseparation assay displayed an excellent precision (total coefficient of variance (CV)<2.5%, intraassay CV<1.5% and interassay CV<1.5%). Mean total error was 4.34%. The direct assay met the current National Cholesterol Education Program (NCEP) requirements for LDL-C testing for precision and accuracy. The results of direct method (x) and the FF (y) were highly correlated (r=0.9908, y=1.030 x -0.289, n=25) in group 1, but the results of two methods disagreed (r=0.716, y=0.956 x -24.869, n=22) in group 2 (patients with TGs 320-695 mg/dl). CONCLUSION: The direct immunoseparation assay meets the currently established analytical performance goals and may be useful for the diagnosis and management of hyperlipidemic patients.


Subject(s)
Cholesterol, LDL/blood , Coronary Artery Disease/diagnosis , Hypercholesterolemia/diagnosis , Biomarkers/blood , Cholesterol, HDL/blood , Coronary Artery Disease/blood , Humans , Hypercholesterolemia/blood , Mathematics , Reagent Kits, Diagnostic/standards , Regression Analysis , Reproducibility of Results , Triglycerides/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...