Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters

Publication year range
1.
Cell ; 175(7): 1744-1755.e15, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30503208

ABSTRACT

Tumor-infiltrating CD8 T cells were found to frequently express the inhibitory receptor NKG2A, particularly in immune-reactive environments and after therapeutic cancer vaccination. High-dimensional cluster analysis demonstrated that NKG2A marks a unique immune effector subset preferentially co-expressing the tissue-resident CD103 molecule, but not immune checkpoint inhibitors. To examine whether NKG2A represented an adaptive resistance mechanism to cancer vaccination, we blocked the receptor with an antibody and knocked out its ligand Qa-1b, the conserved ortholog of HLA-E, in four mouse tumor models. The impact of therapeutic vaccines was greatly potentiated by disruption of the NKG2A/Qa-1b axis even in a PD-1 refractory mouse model. NKG2A blockade therapy operated through CD8 T cells, but not NK cells. These findings indicate that NKG2A-blocking antibodies might improve clinical responses to therapeutic cancer vaccines.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines , Immunity, Cellular , NK Cell Lectin-Like Receptor Subfamily C , Neoplasm Proteins , Neoplasms, Experimental , Vaccination , Animals , Antibodies, Neoplasm/immunology , Antigens, CD/immunology , CD8-Positive T-Lymphocytes/pathology , Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Cell Line, Tumor , Histocompatibility Antigens Class I/immunology , Humans , Integrin alpha Chains/immunology , Mice , NK Cell Lectin-Like Receptor Subfamily C/antagonists & inhibitors , NK Cell Lectin-Like Receptor Subfamily C/immunology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/immunology , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , HLA-E Antigens
2.
Cell ; 175(7): 1731-1743.e13, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30503213

ABSTRACT

Checkpoint inhibitors have revolutionized cancer treatment. However, only a minority of patients respond to these immunotherapies. Here, we report that blocking the inhibitory NKG2A receptor enhances tumor immunity by promoting both natural killer (NK) and CD8+ T cell effector functions in mice and humans. Monalizumab, a humanized anti-NKG2A antibody, enhanced NK cell activity against various tumor cells and rescued CD8+ T cell function in combination with PD-x axis blockade. Monalizumab also stimulated NK cell activity against antibody-coated target cells. Interim results of a phase II trial of monalizumab plus cetuximab in previously treated squamous cell carcinoma of the head and neck showed a 31% objective response rate. Most common adverse events were fatigue (17%), pyrexia (13%), and headache (10%). NKG2A targeting with monalizumab is thus a novel checkpoint inhibitory mechanism promoting anti-tumor immunity by enhancing the activity of both T and NK cells, which may complement first-generation immunotherapies against cancer.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Carcinoma, Squamous Cell , Cetuximab/therapeutic use , Immunity, Cellular/drug effects , Immunotherapy , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily C , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/therapy , Clinical Trials, Phase II as Topic , Humans , Killer Cells, Natural/pathology , Mice , NK Cell Lectin-Like Receptor Subfamily C/antagonists & inhibitors , NK Cell Lectin-Like Receptor Subfamily C/immunology
3.
Nature ; 588(7836): 146-150, 2020 12.
Article in English | MEDLINE | ID: mdl-32726800

ABSTRACT

Coronavirus disease 2019 (COVID-19) is a disease caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and has resulted in a pandemic1. The C5a complement factor and its receptor C5aR1 (also known as CD88) have a key role in the initiation and maintenance of several inflammatory responses by recruiting and activating neutrophils and monocytes1. Here we provide a longitudinal analysis of immune responses, including phenotypic analyses of immune cells and assessments of the soluble factors that are present in the blood and bronchoalveolar lavage fluid of patients at various stages of COVID-19 severity, including those who were paucisymptomatic or had pneumonia or acute respiratory distress syndrome. The levels of soluble C5a were increased in proportion to the severity of COVID-19 and high expression levels of C5aR1 receptors were found in blood and pulmonary myeloid cells, which supports a role for the C5a-C5aR1 axis in the pathophysiology of acute respiratory distress syndrome. Anti-C5aR1 therapeutic monoclonal antibodies prevented the C5a-mediated recruitment and activation of human myeloid cells, and inhibited acute lung injury in human C5aR1 knock-in mice. These results suggest that blockade of the C5a-C5aR1 axis could be used to limit the infiltration of myeloid cells in damaged organs and prevent the excessive lung inflammation and endothelialitis that are associated with acute respiratory distress syndrome in patients with COVID-19.


Subject(s)
COVID-19/complications , COVID-19/immunology , Complement C5a/immunology , Inflammation/complications , Inflammation/immunology , Receptor, Anaphylatoxin C5a/immunology , Acute Lung Injury/drug therapy , Acute Lung Injury/immunology , Acute Lung Injury/prevention & control , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , CD11b Antigen/immunology , CD11b Antigen/metabolism , COVID-19/blood , COVID-19/pathology , Complement C5a/antagonists & inhibitors , Complement C5a/biosynthesis , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/prevention & control , Disease Models, Animal , Female , Humans , Inflammation/drug therapy , Inflammation/pathology , Lung/drug effects , Lung/immunology , Lung/pathology , Mice , Mice, Inbred C57BL , Myeloid Cells/drug effects , Myeloid Cells/immunology , Myeloid Cells/pathology , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/blood , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/prevention & control , SARS-CoV-2/drug effects , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity
4.
Blood ; 123(5): 678-86, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24326534

ABSTRACT

Natural killer (NK) cells mediate antilymphoma activity by spontaneous cytotoxicity and antibody-dependent cell-mediated cytotoxicity (ADCC) when triggered by rituximab, an anti-CD20 monoclonal antibody (mAb) used to treat patients with B-cell lymphomas. The balance of inhibitory and activating signals determines the magnitude of the efficacy of NK cells by spontaneous cytotoxicity. Here, using a killer-cell immunoglobulin-like receptor (KIR) transgenic murine model, we show that blockade of the interface of inhibitory KIRs with major histocompatibility complex (MHC) class I antigens on lymphoma cells by anti-KIR antibodies prevents a tolerogenic interaction and augments NK-cell spontaneous cytotoxicity. In combination with anti-CD20 mAbs, anti-KIR treatment induces enhanced NK-cell-mediated, rituximab-dependent cytotoxicity against lymphoma in vitro and in vivo in KIR transgenic and syngeneic murine lymphoma models. These results support a therapeutic strategy of combination rituximab and KIR blockade through lirilumab, illustrating the potential efficacy of combining a tumor-targeting therapy with an NK-cell agonist, thus stimulating the postrituximab antilymphoma immune response.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antigens, CD20/immunology , Killer Cells, Natural/immunology , Lymphoma/therapy , Receptors, KIR/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Murine-Derived/immunology , Antibody-Dependent Cell Cytotoxicity , Cell Line , Female , Histocompatibility Antigens Class I/immunology , Humans , Lymphoma/immunology , Male , Mice , Rituximab
5.
Haematologica ; 101(5): 626-33, 2016 05.
Article in English | MEDLINE | ID: mdl-26721894

ABSTRACT

Natural killer cells are key cells of the innate immune system. Natural killer cell receptor repertoires are diversified by a stochastic expression of killer-cell-immunoglobulin-like receptors and lectin-like receptors such as NKG2 receptors. All individuals harbor a subset of natural killer cells expressing NKG2A, the inhibitory checkpoint receptor for HLA-E. Most neoplastic and normal hematopoietic cells express HLA-E, the inhibitory ligand of NKG2A. A novel anti-human NKG2A antibody induced tumor cell death, suggesting that the antibody could be useful in the treatment of cancers expressing HLA-E. We found that immunodeficient mice, co-infused with human primary leukemia or Epstein-Barr virus cell lines and NKG2A(+) natural killer cells, pre-treated with anti-human NKG2A, were rescued from disease progression. Human NKG2A(+) natural killer cells reconstituted in immunodeficient mice after transplantation of human CD34(+) cells. These natural killer cells are able to kill engrafted human primary leukemia or Epstein-Barr virus cell lines by lysis after intraperitoneal administration of anti-human NKG2A. Thus, this anti-NKG2A may exploit the anti-leukemic action of the wave of NKG2A(+) natural killer cells recovering after hematopoietic stem cell transplants or adoptive therapy with natural killer cell infusions from matched or mismatched family donors after chemotherapy for acute leukemia, without the need to search for a natural killer cell alloreactive donor.


Subject(s)
Antibodies, Monoclonal/pharmacology , Bone Marrow Cells/drug effects , NK Cell Lectin-Like Receptor Subfamily C/antagonists & inhibitors , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Line, Transformed , Cytotoxicity, Immunologic , Disease Models, Animal , Female , Histocompatibility Antigens Class I/immunology , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukemia/drug therapy , Leukemia/metabolism , Leukemia/mortality , Leukemia/pathology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocyte Depletion/methods , Mice , HLA-E Antigens
6.
Haematologica ; 100(2): 263-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25510242

ABSTRACT

Despite recent treatment improvements, multiple myeloma remains an incurable disease. Since antibody-dependent cell-mediated cytotoxicity is an important effector mechanism of daratumumab, we explored the possibility of improving daratumumab-mediated cell-mediated cytotoxicity by blocking natural killer cell inhibitory receptors with the human monoclonal anti-KIR antibody IPH2102, next to activation of natural killer cells with the immune modulatory drug lenalidomide. In 4-hour antibody-dependent cell-mediated cytotoxicity assays, IPH2102 did not induce lysis of multiple myeloma cell lines, but it did significantly augment daratumumab-induced myeloma cell lysis. Also in an ex vivo setting, IPH2102 synergistically improved daratumumab-dependent lysis of primary myeloma cells in bone marrow mononuclear cells (n=21), especially in patients carrying the FcγRIIIa-158F allele or the FcγRIIa-131R allele, who bind IgG1 with lower affinity than patients carrying the FcγRIIIa-158V allele or the FcγRIIa-131H allele. Finally, a further synergistically improved myeloma cell lysis with the daratumumab-IPH2102 combination was observed by adding lenalidomide, which suggests that more effective treatment strategies can be designed for multiple myeloma by combining daratumumab with agents that independently modulate natural killer cell function.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cytotoxicity, Immunologic/immunology , Multiple Myeloma/pathology , Multiple Myeloma/therapy , Receptors, KIR/immunology , Blotting, Western , Cell Proliferation , Cells, Cultured , Drug Synergism , Flow Cytometry , Humans , Immunoglobulin G/immunology , Immunophenotyping , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lenalidomide , Multiple Myeloma/immunology , Polymorphism, Genetic/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, IgG/genetics , Receptors, KIR/genetics , Reverse Transcriptase Polymerase Chain Reaction , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives
7.
Blood ; 120(22): 4317-23, 2012 Nov 22.
Article in English | MEDLINE | ID: mdl-23002117

ABSTRACT

IPH2101 is an anti-killer inhibitory receptor (anti-KIR) mAb that can block KIR-mediated inhibition of natural killer (NK) cells to enhance cytotoxicity against acute myeloid leukemia blasts. We have conducted a phase 1 study of IPH2101 in elderly patients with acute myeloid leukemia in first complete remission. Patients received escalating doses (0.0003-3 mg/kg) of IPH2101 following a 3 + 3 design. Safety, toxicity (primary end points), pharmacokinetics, outcome, and immunologic correlates were evaluated. Twenty-three patients (median age, 71 years), were enrolled. Adverse events were mild and transient, consisting mainly of infusion syndrome and erythema. The maximum tolerated dose was not reached, although full KIR saturation (> 90%) was sustained for more than 2 weeks at 1 and 3 mg/kg. There was a clear correlation between mAb exposure and KIR occupancy. Neither hematologic toxicity nor significant changes in the numbers and distribution of lymphocyte subsets, NK cell receptor expression, or in vitro cytotoxicity were seen. At the highest dose levels (0.3, 1, and 3 mg/kg), transient increases in TNF-α and MIP-1ß serum concentrations and NK cell CD69 expression were observed. Overall and relapse-free survival in the present study compared favorably to reports in comparable patient populations. We conclude that IPH2101 administration is safe and can block KIR for prolonged periods of time with limited side effects. Registered with the European Union Drug Regulating Authorities Clinical Trials (EUDRACT) as 2005-005298-31.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Leukemia, Myeloid, Acute/therapy , Aged , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Cells, Cultured , Female , Humans , Immunotherapy/adverse effects , Immunotherapy/methods , K562 Cells , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Leukemia, Myeloid, Acute/immunology , Male , Middle Aged , Receptors, KIR/antagonists & inhibitors , Receptors, KIR/immunology , Remission Induction , Treatment Outcome
8.
Blood ; 120(22): 4324-33, 2012 Nov 22.
Article in English | MEDLINE | ID: mdl-23033266

ABSTRACT

Natural killer (NK) cells elicit cytotoxicity against multiple myeloma (MM); however, MM cells express HLA class I molecules as ligands to NK cell inhibitory killer immunoglobulin-like receptors (KIRs) as a means of immunoevasion. KIR-ligand mismatch may improve outcomes in allogeneic transplantation for MM. Extrapolating on this concept, we conducted a phase 1 trial of IPH2101, an anti-KIR antibody, in patients with relapsed/refractory MM. IPH2101 was administered intravenously every 28 days in 7 dose-escalated cohorts (0.0003-3 mg/kg) for up to 4 cycles. Pharmacokinetic, pharmacodynamic, and correlative immunologic studies were completed. A total of 32 patients were enrolled. The biologic endpoint of full KIR2D occupancy across the dosing cycle was achieved without dose-limiting toxicity or maximally tolerated dose. One severe adverse event was noted. Pharmacokinetic and pharmacodynamic findings approximated preclinical predictions, and IPH2101 enhanced ex vivo patient-derived NK cell cytotoxicity against MM. No objective responses were seen. No evidence of autoimmunity was observed. These findings suggest that IPH2101 is safe and tolerable at doses that achieve full inhibitory KIR saturation, and this approach warrants further development in MM. This trial was registered at www.clinicaltrials.gov as #NCT00552396.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Multiple Myeloma/therapy , Administration, Intravenous , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/immunology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/immunology , Female , Humans , Immunotherapy/methods , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Male , Middle Aged , Multiple Myeloma/immunology , Receptors, KIR/antagonists & inhibitors , Receptors, KIR/immunology , Recurrence
9.
Blood ; 118(24): 6387-91, 2011 Dec 08.
Article in English | MEDLINE | ID: mdl-22031859

ABSTRACT

Multiple myeloma (MM) patients who receive killer cell Ig-like receptor (KIR) ligand-mismatched, T cell-depleted, allogeneic transplantation may have a reduced risk of relapse compared with patients who receive KIR ligand-matched grafts, suggesting the importance of this signaling axis in the natural killer (NK) cell-versus-MM effect. Expanding on this concept, IPH2101 (1-7F9), an anti-inhibitory KIR mAb, enhances NK-cell function against autologous MM cells by blocking the engagement of inhibitory KIR with cognate ligands, promoting immune complex formation and NK-cell cytotoxicity specifically against MM cell targets but not normal cells. IPH2101 prevents negative regulatory signals by inhibitory KIR, whereas lenalidomide augments NK-cell function and also appears to up-regulate ligands for activating NK-cell receptors on MM cells. Lenalidomide and a murine anti-inhibitory NK-cell receptor Ab mediate in vivo rejection of a lenalidomide-resistant tumor. These mechanistic, preclinical data support the use of a combination of IPH2101 and lenalidomide in a phase 2 trial for MM.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Killer Cells, Natural/drug effects , Multiple Myeloma/drug therapy , Receptors, KIR/antagonists & inhibitors , Thalidomide/analogs & derivatives , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antigen-Antibody Complex , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Cytotoxicity, Immunologic/drug effects , Drug Evaluation, Preclinical , Humans , Immunomodulation/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lenalidomide , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Ligands , Mice , Mice, Inbred C57BL , Multiple Myeloma/blood , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , Thalidomide/pharmacology , Thalidomide/therapeutic use , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
10.
J Clin Pharmacol ; 63(7): 817-829, 2023 07.
Article in English | MEDLINE | ID: mdl-36852723

ABSTRACT

Monalizumab is a novel, first-in-class humanized immunoglobulin G4 monoclonal antibody immune checkpoint inhibitor that targets the inhibitory CD94/NKG2A receptors. The objectives of this analysis were to develop a population pharmacokinetic (PK) model of monalizumab, evaluate the impact of clinically relevant covariates on monalizumab PK, and provide dose justification for clinical trials. We developed a monalizumab population PK model to characterize the PK properties of monalizumab in patients with advanced solid tumors or head and neck squamous cell carcinoma. Data from clinical studies D419NC00001 (NCT02671435) and IPH2201-203 (NCT02643550) were pooled for the analysis, resulting in a data set of 3066 PK samples derived from 507 subjects. The PK of monalizumab were reasonably described by a 2-compartment model with first-order elimination. Monalizumab generally exhibited linear PK over a dose range of 22.5-750 mg or 10 mg/kg every 2 weeks. The estimate of clearance was ≈0.255 L/day, and apparent volume of distribution was 6.36 L for a typical individual, consistent with previous findings for endogenous immunoglobulin Gs and other therapeutic monoclonal antibodies. Baseline albumin and body weight were identified as significant covariates of clearance; body weight, sex, and smoking status had a significant impact on volume of distribution; and none of these covariates had impact on peripheral volume of distribution. Although these covariates were identified as statistically significant, they are considered to be not clinically meaningful, as changes in monalizumab exposure were <30%. Therefore, no dose adjustments of monalizumab based on patient or disease characteristics are recommended.


Subject(s)
Antibodies, Monoclonal, Humanized , Neoplasms , Humans , Antibodies, Monoclonal, Humanized/pharmacokinetics , Neoplasms/drug therapy , Antibodies, Monoclonal/pharmacokinetics , Body Weight , Models, Biological
11.
Proc Natl Acad Sci U S A ; 106(31): 12879-84, 2009 Aug 04.
Article in English | MEDLINE | ID: mdl-19561305

ABSTRACT

Natural killer (NK) cells are lymphocytes of the innate immune system able to recognize and kill tumors lacking self-MHC class I molecules. This "missing-self" recognition is mediated by the lack of engagement of MHC class I-specific inhibitory NK cell receptors that include the killer cell Ig-like receptors (KIR) in humans and Ly49 molecules in mice. A promising immunotherapeutic strategy against MHC class I(+) cancer cells is to block NK cell inhibitory receptors using monoclonal antibodies (mAb). However, interactions between MHC class I molecules and their inhibitory receptors are also required for the acquisition of NK cell functional competence, a process referred as to "education." In addition, inhibitory receptors are involved in self-tolerance on educated NK cells. Here, we developed a preclinical mouse model in which all NK cells are educated by a single transgenic inhibitory receptor, human KIR2DL3, through the engagement with its HLA-Cw3 ligand. This approach revealed that NK cells could be reprogrammed to control the development of mouse syngenic tumors in vivo. Moreover, in vivo anti-KIR mAb treatment induced the killing of HLA(+) target cells without breaking self-tolerance. Finally, the long-term infusion of anti-KIR mAb neither abolished NK cell education nor tumor cell recognition. Therefore, these results strongly support the use of inhibitory receptor blockade in cancer patients.


Subject(s)
Antibodies, Monoclonal/therapeutic use , HLA-C Antigens/physiology , Killer Cells, Natural/immunology , Neoplasms, Experimental/therapy , Receptors, KIR2DL3/physiology , Self Tolerance , Animals , Cell Line , Humans , Immunity, Innate , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Receptors, KIR2DL3/immunology
12.
Blood ; 114(13): 2667-77, 2009 Sep 24.
Article in English | MEDLINE | ID: mdl-19553639

ABSTRACT

Inhibitory-cell killer immunoglobulin-like receptors (KIR) negatively regulate natural killer (NK) cell-mediated killing of HLA class I-expressing tumors. Lack of KIR-HLA class I interactions has been associated with potent NK-mediated antitumor efficacy and increased survival in acute myeloid leukemia (AML) patients upon haploidentical stem cell transplantation from KIR-mismatched donors. To exploit this pathway pharmacologically, we generated a fully human monoclonal antibody, 1-7F9, which cross-reacts with KIR2DL1, -2, and -3 receptors, and prevents their inhibitory signaling. The 1-7F9 monoclonal antibody augmented NK cell-mediated lysis of HLA-C-expressing tumor cells, including autologous AML blasts, but did not induce killing of normal peripheral blood mononuclear cells, suggesting a therapeutic window for preferential enhancement of NK-cell cytotoxicity against malignant target cells. Administration of 1-7F9 to KIR2DL3-transgenic mice resulted in dose-dependent rejection of HLA-Cw3-positive target cells. In an immunodeficient mouse model in which inoculation of human NK cells alone was unable to protect against lethal, autologous AML, preadministration of 1-7F9 resulted in long-term survival. These data show that 1-7F9 confers specific, stable blockade of KIR, boosting NK-mediated killing of HLA-matched AML blasts in vitro and in vivo, providing a preclinical basis for initiating phase 1 clinical trials with this candidate therapeutic antibody.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Killer Cells, Natural/drug effects , Neoplasms/therapy , Receptors, KIR/immunology , Animals , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cells, Cultured , Cytotoxicity, Immunologic/drug effects , Humans , Immunotherapy/methods , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Neoplasms/immunology , Neoplasms/pathology , Receptors, KIR/antagonists & inhibitors , Receptors, KIR2DL1/chemistry , Receptors, KIR2DL1/genetics , Receptors, KIR2DL1/immunology , Receptors, KIR2DL2/chemistry , Receptors, KIR2DL2/genetics , Receptors, KIR2DL2/immunology , Receptors, KIR2DL3/genetics , Receptors, KIR2DL3/immunology , Up-Regulation/drug effects , Up-Regulation/immunology
13.
Cancers (Basel) ; 13(23)2021 Nov 23.
Article in English | MEDLINE | ID: mdl-34884993

ABSTRACT

BACKGROUND: Cancer-associated fibroblasts (CAF) are heterogeneous with multiple functions in breast cancer. Recently, we identified a specific CAF subpopulation (referred to as CAF-S1), which promotes immunosuppression and immunotherapy resistance. METHODS AND RESULTS: Here, by studying a large collection of human samples, we highlight the key function of CD73/NT5E in CAF-S1-mediated immunosuppression in breast cancer. We first reveal that CD73 protein level specifically accumulates in CAF-S1 in breast cancer patients. Interestingly, infiltration of regulatory T lymphocytes (Tregs) is significantly correlated with CD73 expression in stroma but not in epithelium, indicating that CD73 contributes to immunosuppression when expressed in CAF-S1 and not in tumor cells. By performing functional assays based on relevant systems using primary CAF-S1 isolated from patients, we demonstrate that CAF-S1 increase the content in both PD-1+ and CTLA-4+ Tregs. Importantly, the use of a blocking anti-CD73 antibody on CAF-S1 reduces CAF-S1-mediated immunosuppression by preventing expression of these immune checkpoints on Tregs. CONCLUSIONS: Our data support the potential clinical benefit of using both anti-CD73 and immune-checkpoint inhibitors in breast cancer patients for inhibiting CAF-S1-mediated immunosuppression and enhancing anti-tumor immune response.

14.
Clin Immunol ; 135(1): 26-32, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20093094

ABSTRACT

Killer Ig-like receptors (KIRs) are MHC class I-specific receptors expressed by Natural Killer (NK) and T cell subsets. KIRs either inhibit (KIR-L) or activate (KIR-S) lymphocyte functions. Inhibitory KIR2DL1 and activating KIR2DS1 share ligand specificity for the HLA-C2 group, consistent with their almost identical extracytoplasmic domain. This homology hampered the distinction between KIR2DL1 and KIR2DS1. We report here the characterization of the KIR2DS1(+) subsets among primary human NK and T cells. Regardless of the host HLA-C genotype, around 10% of circulating NK cells expressed KIR2DS1 in absence of KIR2DL1. In HLA-C2 individuals, KIR2DS1 was not able to induce NK cell education (i.e., the acquisition of NK cell competence) nor to interfere with KIR2DL1-induced NK cell education. KIR2DS1 was also present on rare oligoclonal TCRalphabeta(+)CD8alpha(+) and TCRalphabeta(+)CD4(-)CD8(-) subsets. As KIR2DS1 has been associated with autoimmunity and hematopoietic stem cell transplantation, these results pave the way to dissect the function of KIR2DS1 in these clinical conditions.


Subject(s)
HLA-C Antigens/immunology , Killer Cells, Natural/immunology , Receptors, KIR/biosynthesis , T-Lymphocytes/immunology , Animals , Cell Line , DNA/chemistry , DNA/genetics , Flow Cytometry , Genotype , Humans , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/immunology , Mice , Receptors, KIR/genetics , Receptors, KIR/immunology
15.
Cell Mol Immunol ; 16(5): 415-422, 2019 05.
Article in English | MEDLINE | ID: mdl-30911118

ABSTRACT

Natural killer (NK) cells are innate lymphoid cells endowed with cytolytic activity and a capacity to secrete cytokines and chemokines. Several lines of evidence suggest that NK cells play an important role in anti-tumor immunity. Some therapies against hematological malignacies make use of the immune properties of NK cells, such as their ability to kill residual leukemic blasts efficiently after conditioning during haploidentical hematopoietic stem cell transplantation. However, knowledge on NK cell infiltration and the status of NK cell responsiveness in solid tumors is limited so far. The pro-angiogenic role of the recently described NK cell-like type 1 innate lymphoid cells (ILC1s) and their phenotypic resemblance to NK cells are confounding factors that add a level of complexity, at least in mice. Here, we review the current knowledge on the presence and function of NK cells in solid tumors as well as the immunotherapeutic approaches designed to harness NK cell functions in these conditions, including those that aim to reinforce conventional anti-tumor therapies to increase the chances of successful treatment.


Subject(s)
Killer Cells, Natural/immunology , Neoplasms/immunology , Animals , Cytotoxicity, Immunologic , HLA Antigens/metabolism , Histocompatibility Antigens/metabolism , Homeostasis , Humans , Immunity, Innate , Mice , Receptor Cross-Talk , Th1-Th2 Balance
16.
J Immunother Cancer ; 7(1): 263, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31623687

ABSTRACT

The implementation of immune checkpoint inhibitors to the oncology clinic signified a new era in cancer treatment. After the first indication of melanoma, an increasing list of additional cancer types are now treated with immune system targeting antibodies to PD-1, PD-L1 and CTLA-4, alleviating inhibition signals on T cells. Recently, we published proof-of-concept results on a novel checkpoint inhibitor, NKG2A. This receptor is expressed on cytotoxic lymphocytes, including NK cells and subsets of activated CD8+ T cells. Blocking antibodies to NKG2A unleashed the reactivity of these effector cells resulting in tumor control in multiple mouse models and an early clinical trial. Monalizumab is inhibiting this checkpoint in human beings and future clinical trials will have to reveal its potency in combination with other cancer treatment options.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents, Immunological/pharmacology , NK Cell Lectin-Like Receptor Subfamily C/antagonists & inhibitors , Neoplasms/drug therapy , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Disease Models, Animal , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , NK Cell Lectin-Like Receptor Subfamily C/immunology , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , HLA-E Antigens
17.
Methods Mol Biol ; 415: 291-300, 2008.
Article in English | MEDLINE | ID: mdl-18370161

ABSTRACT

Natural killer (NK) cells are major actors of innate immune responses against viruses, bacteria, parasites, and other mediators of pathology such as malignant transformation. These cells are also directly implicated in the link between innate and adaptive immunity, shaping T-cell responses. It is now obvious that manipulation of this lymphocyte subset could be the basis of new therapeutic approaches for cancer and/or pathogen-driven pathology. Hence, techniques enabling the phenotypic and functional analysis of patient NK cells are of major importance. In this chapter, we present an extensive immunophenotyping of patient NK cells as well as a recently described method to assess NK cell functional activity at the single-cell level.


Subject(s)
Immunophenotyping/methods , Killer Cells, Natural/cytology , Humans , Interferon-gamma/biosynthesis , Lysosomal-Associated Membrane Protein 1/immunology
18.
Oncotarget ; 9(25): 17675-17688, 2018 Apr 03.
Article in English | MEDLINE | ID: mdl-29707140

ABSTRACT

PURPOSE: Anti-KIR monoclonal antibodies (mAbs) can enhance the antitumor responses of natural killer (NK) cells. We evaluated the safety of the anti-KIR2D mAb lirilumab in patients with various cancers. EXPERIMENTAL DESIGN: Thirty-seven patients with hematological malignancies (n = 22) or solid tumors (n = 15) were included in the study. Dose escalation (0.015 to 10 mg/kg) was conducted following a 3 + 3 design. Patients were scheduled to receive four cycles of treatment. In a second (extension) phase 17 patients were treated at 0.015 (n = 9) or 3 mg/kg (n = 8). RESULTS: No dose-limiting toxicity was recorded. The most frequent lirilumab-related adverse events were pruritus (19%), asthenia (16%), fatigue (14%), infusion-related reaction (14%), and headache (11%), mostly mild or moderate. Pharmacokinetics was dose-dependent and linear, with minimal accumulation resulting from the 4-weekly repeated administrations. Full KIR occupancy (>95%) was achieved with all dosages, and the duration of occupancy was dose-related. No significant changes were observed in the number or distribution of lymphocyte subpopulations, nor was any reduction in the distribution of KIR2D-positive NK cells. CONCLUSIONS: This phase 1 trial demonstrated the satisfactory safety profile of lirilumab up to doses that enable full and sustained blockade of KIR.

19.
PLoS One ; 12(12): e0189069, 2017.
Article in English | MEDLINE | ID: mdl-29216263

ABSTRACT

AIM: To examine the characteristics of community health workers (CHWs) involved in diabetes prevention programmes (DPPs) and their contributions to expected outcomes. METHODS: Electronic databases including PubMed-MEDLINE, EBSCOHost, and SCOPUS/EMBASE were searched for studies published between January 2000 and March 2016. All studies that used CHWs to implement DPP in ≥18-year-old participants without diabetes but at high risk for developing the condition, irrespective of the study design, setting or outcomes measured, were included. Results were synthesized narratively. RESULTS: Forty papers of 30 studies were identified. Studies were mainly community-based and conducted in minority populations in USA. Sample sizes ranged from 20 participants in a single community to 2369 participants in 46 communities. Although CHWs were generally from the local community, their qualifications, work experience and training received differed across studies. Overall the training was culturally sensitive and/or appropriate, covering topics such as the importance of good nutrition and the benefits of increased physical activity, communication and leadership. CHWs delivered a variety of interventions and also screened or recruited participants. The shared culture and language between CHWs and participants likely contributed to better programme implementation and successful outcomes. CONCLUSIONS: The complexity of DPPs and the diverse CHW roles preclude attributing specific outcomes to CHW involvement. Nevertheless, documenting potential CHW roles and the relevant training required may optimise CHW contributions and facilitate their involvement in DPPs in the future.


Subject(s)
Community Health Workers , Diabetes Mellitus/prevention & control , Professional Role , Humans
20.
Front Immunol ; 8: 64, 2017.
Article in English | MEDLINE | ID: mdl-28210257

ABSTRACT

NK cells are defective in acute myeloid leukemia (AML) at diagnosis. Here, we studied the kinetic of expression of the major activating and inhibitory receptors of NK, CD8 T, and γδ T cells in patients undergoing chemotherapy (CT) for the treatment of AML (n = 29). We showed that NK cells are the main affected population at diagnosis and that expression of activating receptors is partially restored within a few weeks after CT. CD8 T cells and γδ T cells are only weakly affected at diagnosis. Killer cell immunoglobulin-like receptor expression by NK cells, but not NKG2A and CD85j, was downregulated. Interestingly, the development of NK cells appeared altered as the most immature CD56bright NK cells were seriously underrepresented. Finally, we showed that NK cell functions were only partially restored 6 weeks after CT as degranulation capabilities of NK cells recovered, whereas cytokine production remained low. Our data point out NK cells as antitumor effectors peculiarly hampered by leukemic cells. This study may indicate a timeline when NK-mediated therapies or other immunotherapies could be performed, particularly for patients excluded of hematopoietic stem cell transplantation.

SELECTION OF CITATIONS
SEARCH DETAIL