Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Nature ; 584(7821): 437-442, 2020 08.
Article in English | MEDLINE | ID: mdl-32555388

ABSTRACT

During the coronavirus disease-2019 (COVID-19) pandemic, severe acute respiratory syndrome-related coronavirus-2 (SARS-CoV-2) has led to the infection of millions of people and has claimed hundreds of thousands of lives. The entry of the virus into cells depends on the receptor-binding domain (RBD) of the spike (S) protein of SARS-CoV-2. Although there is currently no vaccine, it is likely that antibodies will be essential for protection. However, little is known about the human antibody response to SARS-CoV-21-5. Here we report on 149 COVID-19-convalescent individuals. Plasma samples collected an average of 39 days after the onset of symptoms had variable half-maximal pseudovirus neutralizing titres; titres were less than 50 in 33% of samples, below 1,000 in 79% of samples and only 1% of samples had titres above 5,000. Antibody sequencing revealed the expansion of clones of RBD-specific memory B cells that expressed closely related antibodies in different individuals. Despite low plasma titres, antibodies to three distinct epitopes on the RBD neutralized the virus with half-maximal inhibitory concentrations (IC50 values) as low as 2 ng ml-1. In conclusion, most convalescent plasma samples obtained from individuals who recover from COVID-19 do not contain high levels of neutralizing activity. Nevertheless, rare but recurring RBD-specific antibodies with potent antiviral activity were found in all individuals tested, suggesting that a vaccine designed to elicit such antibodies could be broadly effective.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Betacoronavirus/immunology , Coronavirus Infections/immunology , Pneumonia, Viral/immunology , Adolescent , Adult , Aged , Antibodies, Monoclonal/analysis , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/analysis , Antibodies, Viral/analysis , Antibody Specificity , COVID-19 , COVID-19 Vaccines , Coronavirus Infections/prevention & control , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Neutralization Tests , Pandemics , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/immunology , Viral Vaccines/immunology , Young Adult
2.
Cell ; 143(3): 416-29, 2010 Oct 29.
Article in English | MEDLINE | ID: mdl-21029863

ABSTRACT

Dendritic cells (DCs), critical antigen-presenting cells for immune control, normally derive from bone marrow precursors distinct from monocytes. It is not yet established if the large reservoir of monocytes can develop into cells with critical features of DCs in vivo. We now show that fully differentiated monocyte-derived DCs (Mo-DCs) develop in mice and DC-SIGN/CD209a marks the cells. Mo-DCs are recruited from blood monocytes into lymph nodes by lipopolysaccharide and live or dead gram-negative bacteria. Mobilization requires TLR4 and its CD14 coreceptor and Trif. When tested for antigen-presenting function, Mo-DCs are as active as classical DCs, including cross-presentation of proteins and live gram-negative bacteria on MHC I in vivo. Fully differentiated Mo-DCs acquire DC morphology and localize to T cell areas via L-selectin and CCR7. Thus the blood monocyte reservoir becomes the dominant presenting cell in response to select microbes, yielding DC-SIGN(+) cells with critical functions of DCs.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Differentiation , Dendritic Cells/cytology , Escherichia coli/immunology , Lectins, C-Type/metabolism , Monocytes/cytology , Receptors, Cell Surface/metabolism , Animals , Antigen Presentation , Cell Adhesion Molecules/immunology , Dendritic Cells/immunology , L-Selectin/immunology , Lectins, C-Type/immunology , Lipopolysaccharide Receptors/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Monocytes/immunology , Receptors, CCR7/immunology , Receptors, Cell Surface/immunology , T-Lymphocytes/immunology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/immunology
3.
Nature ; 561(7724): 479-484, 2018 09.
Article in English | MEDLINE | ID: mdl-30258136

ABSTRACT

Individuals infected with HIV-1 require lifelong antiretroviral therapy, because interruption of treatment leads to rapid rebound viraemia. Here we report on a phase 1b clinical trial in which a combination of 3BNC117 and 10-1074, two potent monoclonal anti-HIV-1 broadly neutralizing antibodies that target independent sites on the HIV-1 envelope spike, was administered during analytical treatment interruption. Participants received three infusions of 30 mg kg-1 of each antibody at 0, 3 and 6 weeks. Infusions of the two antibodies were generally well-tolerated. The nine enrolled individuals with antibody-sensitive latent viral reservoirs maintained suppression for between 15 and more than 30 weeks (median of 21 weeks), and none developed viruses that were resistant to both antibodies. We conclude that the combination of the anti-HIV-1 monoclonal antibodies 3BNC117 and 10-1074 can maintain long-term suppression in the absence of antiretroviral therapy in individuals with antibody-sensitive viral reservoirs.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , HIV Antibodies/therapeutic use , HIV Infections/drug therapy , HIV Infections/immunology , HIV-1/immunology , Virus Latency/immunology , Adolescent , Adult , Aged , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/immunology , Anti-HIV Agents/therapeutic use , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/adverse effects , Antibodies, Neutralizing/immunology , Binding Sites, Antibody , Broadly Neutralizing Antibodies , Carrier State/drug therapy , Carrier State/immunology , Carrier State/virology , Drug Combinations , Drug Resistance, Viral , Female , HIV Antibodies/administration & dosage , HIV Antibodies/adverse effects , HIV Antibodies/immunology , HIV Envelope Protein gp160/immunology , HIV Infections/virology , HIV-1/isolation & purification , Historically Controlled Study , Humans , Infusions, Intravenous , Male , Middle Aged , Phylogeny , Viremia/drug therapy , Viremia/immunology , Viremia/prevention & control , Viremia/virology , Virus Activation/immunology , Young Adult
4.
Nature ; 535(7613): 556-60, 2016 07 28.
Article in English | MEDLINE | ID: mdl-27338952

ABSTRACT

Interruption of combination antiretroviral therapy in HIV-1-infected individuals leads to rapid viral rebound. Here we report the results of a phase IIa open label clinical trial evaluating 3BNC117,a broad and potent neutralizing antibody against the CD4 binding site of the HIV-1 Env protein, during analytical treatment interruption in 13 HIV-1-infected individuals. Participants with 3BNC117-sensitive virus outgrowth cultures were enrolled. Results show that two or four 30 mg kg(-1) 3BNC117 infusions,separated by 3 or 2 weeks, respectively, are generally well tolerated.Infusions are associated with a delay in viral rebound of 5-9 weeks after two infusions, and up to 19 weeks after four infusions, or an average of 6.7 and 9.9 weeks, respectively, compared with 2.6 weeks for historical controls (P < 0.00001). Rebound viruses arise predominantly from a single provirus. In most individuals,emerging viruses show increased resistance, indicating escape.However, 30% of participants remained suppressed until antibody concentrations waned below 20 µg ml(-1), and the viruses emerging in all but one of these individuals showed no apparent resistance to 3BCN117, suggesting failure to escape over a period of 9-19 weeks.We conclude that the administration of 3BNC117 exerts strong selective pressure on HIV-1 emerging from latent reservoirs during analytical treatment interruption in humans.


Subject(s)
Anti-HIV Agents/administration & dosage , Antibodies, Neutralizing/immunology , HIV Antibodies/immunology , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/growth & development , HIV-1/immunology , Adolescent , Adult , Aged , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/therapeutic use , Binding Sites/drug effects , Binding Sites/immunology , Broadly Neutralizing Antibodies , CD4 Antigens/metabolism , Disease Reservoirs/virology , Drug Administration Schedule , Female , HIV Antibodies/administration & dosage , HIV Antibodies/therapeutic use , HIV Envelope Protein gp160/antagonists & inhibitors , HIV Envelope Protein gp160/chemistry , HIV Envelope Protein gp160/immunology , HIV Envelope Protein gp160/metabolism , HIV Infections/immunology , HIV-1/drug effects , Historically Controlled Study , Humans , Male , Middle Aged , Proviruses/drug effects , Proviruses/growth & development , Proviruses/immunology , Time Factors , Tissue Distribution , Viral Load/drug effects , Viral Load/immunology , Young Adult
5.
Nature ; 522(7557): 487-91, 2015 06 25.
Article in English | MEDLINE | ID: mdl-25855300

ABSTRACT

HIV-1 immunotherapy with a combination of first generation monoclonal antibodies was largely ineffective in pre-clinical and clinical settings and was therefore abandoned. However, recently developed single-cell-based antibody cloning methods have uncovered a new generation of far more potent broadly neutralizing antibodies to HIV-1 (refs 4, 5). These antibodies can prevent infection and suppress viraemia in humanized mice and nonhuman primates, but their potential for human HIV-1 immunotherapy has not been evaluated. Here we report the results of a first-in-man dose escalation phase 1 clinical trial of 3BNC117, a potent human CD4 binding site antibody, in uninfected and HIV-1-infected individuals. 3BNC117 infusion was well tolerated and demonstrated favourable pharmacokinetics. A single 30 mg kg(-1) infusion of 3BNC117 reduced the viral load in HIV-1-infected individuals by 0.8-2.5 log10 and viraemia remained significantly reduced for 28 days. Emergence of resistant viral strains was variable, with some individuals remaining sensitive to 3BNC117 for a period of 28 days. We conclude that, as a single agent, 3BNC117 is safe and effective in reducing HIV-1 viraemia, and that immunotherapy should be explored as a new modality for HIV-1 prevention, therapy and cure.


Subject(s)
Antibodies, Neutralizing/immunology , HIV Antibodies/immunology , HIV Infections/therapy , HIV-1/immunology , Viral Load/immunology , Viremia/therapy , Adult , Amino Acid Sequence , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/adverse effects , Antibodies, Neutralizing/pharmacology , Antibodies, Neutralizing/therapeutic use , Binding Sites , Broadly Neutralizing Antibodies , CD4 Antigens/metabolism , Case-Control Studies , Evolution, Molecular , Female , HIV Antibodies/administration & dosage , HIV Antibodies/adverse effects , HIV Antibodies/pharmacology , HIV Antibodies/therapeutic use , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/immunology , HIV Infections/immunology , HIV Infections/virology , HIV-1/chemistry , HIV-1/drug effects , Humans , Immunization, Passive/methods , Male , Middle Aged , Molecular Sequence Data , Time Factors , Viral Load/drug effects , Viremia/immunology , Viremia/virology , Young Adult
7.
Blood ; 121(25): 5034-44, 2013 Jun 20.
Article in English | MEDLINE | ID: mdl-23482932

ABSTRACT

Functional differences between human dendritic cell (DC) subsets and the potential benefits of targeting them with vaccines remain poorly defined. Here we describe that mice with reconstituted human immune system components (huNSG mice) develop all human conventional and plasmacytoid DC compartments in lymphoid organs. Testing different Toll-like receptor agonists for DC maturation in vivo, we found that IL-12p70 and interferon (IFN)-α production correlated with the maturation of CD141+ (BDCA3+) conventional DCs in huNSG mice. Furthermore, depletion of CD141+ DCs before stimulation significantly reduced IFN-α levels in vivo. This DC subset produced similar total amounts but different subtypes of IFN-α in response to synthetic double-stranded RNA compared with plasmacytoid DCs in response to a single-stranded RNA equivalent. Moreover, synthetic double-stranded RNA as adjuvant and antigen targeting to the endocytic receptor DEC-205, a combination that focuses antigen presentation for T-cell priming on CD141+ DCs, stimulated antigen-specific human CD4+ T-cell responses. Thus, the human CD141+ DC subset is a prominent source of IFN-α and interleukin-12 production and should be further evaluated for vaccine development.


Subject(s)
Antigens, CD/immunology , Dendritic Cells/immunology , Interferon-alpha/biosynthesis , Lectins, C-Type/immunology , Lymphocyte Activation/immunology , RNA, Double-Stranded/immunology , Receptors, Cell Surface/immunology , Animals , Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/cytology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Interferon-alpha/immunology , Mice , Minor Histocompatibility Antigens
8.
Blood ; 116(19): 3828-38, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-20668230

ABSTRACT

Protein vaccines for T-cell immunity are not being prioritized because of poor immunogenicity. To overcome this hurdle, proteins are being targeted to maturing dendritic cells (DCs) within monoclonal antibodies (mAbs) to DC receptors. To extend the concept to humans, we immunized human immunoglobulin-expressing mice with human DEC205 (hDEC205) extracellular domain. 3D6 and 3G9 mAbs were selected for high-affinity binding to hDEC205. In addition, CD11c promoter hDEC205 transgenic mice were generated, and 3G9 was selectively targeted to DCs in these animals. When mAb heavy chain was engineered to express HIV Gag p24, the fusion mAb induced interferon-γ- and interleukin-2-producing CD4(+) T cells in hDEC205 transgenic mice, if polynocinic polycytidylic acid was coadministered as an adjuvant. The T-cell response was broad, recognizing at least 3 Gag peptides, and high titers of anti-human immunoglobulin G antibody were made. Anti-hDEC205 also improved the cross-presentation of Gag to primed CD8(+) T cells from HIV-infected individuals. In all tests, 3D6 and 3G9 targeting greatly enhanced immunization relative to nonbinding control mAb. These results provide preclinical evidence that in vivo hDEC205 targeting increases the efficiency with which proteins elicit specific immunity, setting the stage for proof-of-concept studies of these new protein vaccines in human subjects.


Subject(s)
Antigens, CD/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , HIV Core Protein p24/immunology , HIV-1/immunology , Lectins, C-Type/immunology , Receptors, Cell Surface/immunology , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Animals , Antibodies, Monoclonal , Antigens, CD/genetics , Base Sequence , Cross-Priming , DNA Primers/genetics , HIV Core Protein p24/genetics , HIV-1/genetics , Humans , Immunity, Cellular , Immunity, Humoral , Lectins, C-Type/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Minor Histocompatibility Antigens , Receptors, Cell Surface/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology
9.
J Exp Med ; 201(9): 1503-17, 2005 May 02.
Article in English | MEDLINE | ID: mdl-15867097

ABSTRACT

Natural killer T (NKT) cells are distinct glycolipid reactive innate lymphocytes that are implicated in the resistance to pathogens and tumors. Earlier attempts to mobilize NKT cells, specifically, in vivo in humans met with limited success. Here, we evaluated intravenous injection of monocyte-derived mature DCs that were loaded with a synthetic NKT cell ligand, alpha-galactosyl-ceramide (alpha-GalCer; KRN-7000) in five patients who had advanced cancer. Injection of alpha-GalCer-pulsed, but not unpulsed, dendritic cells (DCs) led to >100-fold expansion of several subsets of NKT cells in all patients; these could be detected for up to 6 mo after vaccination. NKT activation was associated with an increase in serum levels of interleukin-12 p40 and IFN-gamma inducible protein-10. In addition, there was an increase in memory CD8+ T cells specific for cytomegalovirus in vivo in response to alpha-GalCer-loaded DCs, but not unpulsed DCs. These data demonstrate the feasibility of sustained expansion of NKT cells in vivo in humans, including patients who have advanced cancer, and suggest that NKT activation might help to boost adaptive T cell immunity in vivo.


Subject(s)
Cell Proliferation , Dendritic Cells/metabolism , Galactosylceramides/therapeutic use , Immunotherapy , Killer Cells, Natural/immunology , Neoplasms/therapy , Vaccination , Adult , Blood Chemical Analysis , CD8-Positive T-Lymphocytes/immunology , Chemokine CXCL10 , Chemokines/metabolism , Chemokines, CXC/blood , Cytokines/metabolism , Cytomegalovirus/immunology , Dendritic Cells/immunology , Flow Cytometry , Galactosylceramides/metabolism , Humans , Interleukin-12/blood , Neoplasms/immunology , Reverse Transcriptase Polymerase Chain Reaction
10.
Blood ; 112(4): 1231-9, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18519810

ABSTRACT

Dendritic cells (DCs) express many endocytic receptors that deliver antigens for major histocompatibility class (MHC) I and II presentation to CD8(+) and CD4(+) T cells, respectively. Here, we show that targeting Epstein-Barr virus (EBV) nuclear antigen 1 (EBNA1) to one of them, the human multilectin DEC-205 receptor, in the presence of the DC maturation stimulus poly(I:C), expanded EBNA1-specific CD4(+) and CD8(+) memory T cells, and these lymphocytes could control the outgrowth of autologous EBV-infected B cells in vitro. In addition, using a novel mouse model with reconstituted human immune system components, we demonstrated that vaccination with alphaDEC-205-EBNA1 antibodies primed EBNA1-specific IFN-gamma-secreting T cells and also induced anti-EBNA1 antibodies in a subset of immunized mice. Because EBNA1 is the one EBV antigen that is expressed in all proliferating cells infected with this virus, our data suggest that DEC-205 targeting should be explored as a vaccination approach against symptomatic primary EBV infection and against EBV-associated malignancies.


Subject(s)
Antigens, CD/immunology , Cell Proliferation , Epstein-Barr Virus Nuclear Antigens/immunology , Herpesvirus 4, Human/immunology , Lectins, C-Type/immunology , Lymphocyte Activation , Receptors, Cell Surface/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Viral , B-Lymphocytes/virology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Humans , Immunologic Memory , Mice , Mice, Transgenic , Minor Histocompatibility Antigens , Vaccination
11.
bioRxiv ; 2020 May 22.
Article in English | MEDLINE | ID: mdl-32511384

ABSTRACT

During the COVID-19 pandemic, SARS-CoV-2 infected millions of people and claimed hundreds of thousands of lives. Virus entry into cells depends on the receptor binding domain (RBD) of the SARS-CoV-2 spike protein (S). Although there is no vaccine, it is likely that antibodies will be essential for protection. However, little is known about the human antibody response to SARS-CoV-21-5. Here we report on 149 COVID-19 convalescent individuals. Plasmas collected an average of 39 days after the onset of symptoms had variable half-maximal neutralizing titers ranging from undetectable in 33% to below 1:1000 in 79%, while only 1% showed titers >1:5000. Antibody cloning revealed expanded clones of RBD-specific memory B cells expressing closely related antibodies in different individuals. Despite low plasma titers, antibodies to three distinct epitopes on RBD neutralized at half-maximal inhibitory concentrations (IC50s) as low as single digit ng/mL. Thus, most convalescent plasmas obtained from individuals who recover from COVID-19 do not contain high levels of neutralizing activity. Nevertheless, rare but recurring RBD-specific antibodies with potent antiviral activity were found in all individuals tested, suggesting that a vaccine designed to elicit such antibodies could be broadly effective.

12.
PLoS One ; 14(8): e0219142, 2019.
Article in English | MEDLINE | ID: mdl-31393868

ABSTRACT

BACKGROUND: Additional forms of pre-exposure prophylaxis are needed to prevent HIV-1 infection. 3BNC117 and 10-1074 are broadly neutralizing anti-HIV-1 antibodies that target non-overlapping epitopes on the HIV-1 envelope. We investigated the safety, tolerability, pharmacokinetics, and immunogenicity of the intravenous administration of the combination of 3BNC117 and 10-1074 in healthy adults. METHODS: This randomized, double-blind, placebo-controlled, single center, phase 1 study enrolled healthy adults aged 18-65 years to receive one infusion of 3BNC117 immediately followed by 10-1074 at 10 mg/kg, three infusions of 3BNC117 followed by 10-1074 at 3 mg/kg or 10 mg/kg every 8 weeks, or placebo infusions. The primary outcomes were safety and pharmacokinetics. This trial is registered with ClinicalTrials.gov, number NCT02824536. FINDINGS: Twenty-four participants were enrolled in a 3:1 ratio to receive the study products or placebo. The combination of 3BNC117 and 10-1074 was safe and generally well tolerated. There were no serious adverse events considered related to the infusions. The mean elimination half-lives of 3BNC117 and 10-1074 were 16.4 ± 4.6 days and 23.0 ± 5.4 days, respectively, similar to what was observed in previous studies in which each antibody was administered alone. Anti-drug antibody responses were rare and without evidence of related adverse events or impact on elimination kinetics. INTERPRETATION: Single and repeated doses of the combination of 3BNC117 and 10-1074 were well tolerated in healthy adults. These data support the further development of the combination of 3BNC117 and 10-1074 as a long-acting injectable form of pre-exposure prophylaxis for the prevention of HIV-1 infection.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Broadly Neutralizing Antibodies/pharmacology , HIV Antibodies/pharmacology , HIV Infections/immunology , Administration, Intravenous/methods , Adult , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacokinetics , Broadly Neutralizing Antibodies/immunology , Double-Blind Method , Drug Therapy, Combination/methods , Female , HIV Antibodies/immunology , HIV Infections/prevention & control , HIV Seropositivity/drug therapy , HIV-1/immunology , HIV-1/pathogenicity , Healthy Volunteers , Humans , Male , Placebo Effect , Pre-Exposure Prophylaxis/methods
13.
Immunology ; 123(3): 438-46, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17944899

ABSTRACT

The distribution of dendritic cells (DCs) and macrophages in the human spleen has received less attention than that of lymphocytes. Here we have addressed this problem with the human DEC-205/CD205 marker ('DEC'), which is an endocytic receptor on DCs that mediates efficient presentation of antigens. DEC was abundant on dendritic profiles in the white pulp but absent from the red pulp, the latter defined with antibodies to two antigens, mannose receptor/CD206 on sinusoidal lining cells, and macrosialin/CD68 on macrophages. Double staining with anti-DEC and anti-CD3 showed the expected concentration of DEC+ cells in the relatively small T-cell areas of the human spleen. DEC+ cells were also found in other regions of the white pulp. In all regions, the DEC+ cells were positive for major histocompatibility complex (MHC) class II and the CD11c integrin but largely immature, with low expression of B7-2/CD86 costimulator and DC-lysosome-associated membrane protein (LAMP)/CD208. When we concentrated on the perifollicular region between the red pulp and the marginal zone, we found macrophages that stained with antibodies to sialoadhesin/CD169 and DC-specific ICAM-3 grabbing non-integrin (SIGN)/CD209, and just inside these cells were DEC+ profiles. The DEC+ DCs were intertwined with cells that stained for the vascular addressin mucosal addressin cell adhesion molecule (MAdCAM). Therefore, anti-DEC-205/CD205 antibodies are useful for identifying DCs in human splenic white pulp and its border region with the red pulp.


Subject(s)
Antigens, CD/analysis , Dendritic Cells/immunology , Lectins, C-Type/analysis , Receptors, Cell Surface/analysis , Spleen/immunology , Antigens, Differentiation, Myelomonocytic/analysis , CD3 Complex/analysis , Cell Adhesion Molecules , Cell Differentiation/immunology , Fluorescent Antibody Technique/methods , Humans , Immunoglobulins/metabolism , Macrophages/immunology , Minor Histocompatibility Antigens , Mucoproteins/metabolism , T-Lymphocytes/immunology
14.
J Exp Med ; 215(9): 2311-2324, 2018 09 03.
Article in English | MEDLINE | ID: mdl-30072495

ABSTRACT

A clinical trial was performed to evaluate 3BNC117, a potent anti-HIV-1 antibody, in infected individuals during suppressive antiretroviral therapy and subsequent analytical treatment interruption (ATI). The circulating reservoir was evaluated by quantitative and qualitative viral outgrowth assay (Q2VOA) at entry and after 6 mo. There were no significant quantitative changes in the size of the reservoir before ATI, and the composition of circulating reservoir clones varied in a manner that did not correlate with 3BNC117 sensitivity. 3BNC117 binding site amino acid variants found in rebound viruses preexisted in the latent reservoir. However, only 3 of 217 rebound viruses were identical to 868 latent viruses isolated by Q2VOA and near full-length sequencing. Instead, 63% of the rebound viruses appeared to be recombinants, even in individuals with 3BNC117-resistant reservoir viruses. In conclusion, viruses emerging during ATI in individuals treated with 3BNC117 are not the dominant species found in the circulating latent reservoir, but frequently appear to represent recombinants of latent viruses.


Subject(s)
Anti-Retroviral Agents/administration & dosage , HIV Antibodies/administration & dosage , HIV Infections , HIV-1 , Recombination, Genetic , Viral Load , Adolescent , Adult , Aged , Anti-Retroviral Agents/immunology , Female , Follow-Up Studies , HIV Antibodies/immunology , HIV Infections/blood , HIV Infections/drug therapy , HIV Infections/genetics , HIV Infections/immunology , HIV-1/genetics , HIV-1/immunology , Humans , Male , Middle Aged , Recombination, Genetic/drug effects , Recombination, Genetic/immunology , Viral Load/genetics , Viral Load/immunology
15.
Nat Med ; 24(11): 1701-1707, 2018 11.
Article in English | MEDLINE | ID: mdl-30258217

ABSTRACT

Monotherapy of HIV-1 infection with single antiretroviral agents is ineffective because error-prone HIV-1 replication leads to the production of drug-resistant viral variants1,2. Combinations of drugs can establish long-term control, however, antiretroviral therapy (ART) requires daily dosing, can cause side effects and does not eradicate the infection3,4. Although anti-HIV-1 antibodies constitute a potential alternative to ART5,6, treatment of viremic individuals with a single antibody also results in emergence of resistant viral variants7-9. Moreover, combinations of first-generation anti-HIV-1 broadly neutralizing antibodies (bNAbs) had little measurable effect on the infection10-12. Here we report on a phase 1b clinical trial ( NCT02825797 ) in which two potent bNAbs, 3BNC11713 and 10-107414, were administered in combination to seven HIV-1 viremic individuals. Infusions of 30 mg kg-1 of each of the antibodies were well-tolerated. In the four individuals with dual antibody-sensitive viruses, immunotherapy resulted in an average reduction in HIV-1 viral load of 2.05 log10 copies per ml that remained significantly reduced for three months following the first of up to three infusions. In addition, none of these individuals developed resistance to both antibodies. Larger studies will be necessary to confirm the efficacy of antibody combinations in reducing HIV-1 viremia and limiting the emergence of resistant viral variants.


Subject(s)
Antibodies, Neutralizing/administration & dosage , HIV Infections/drug therapy , Immunotherapy , Viremia/drug therapy , Adolescent , Adult , Aged , Antibodies, Neutralizing/adverse effects , Antiretroviral Therapy, Highly Active , Female , HIV Infections/virology , HIV Seropositivity , HIV-1/pathogenicity , Humans , Male , Middle Aged , Viral Load/drug effects , Viremia/virology , Young Adult
16.
J Immunol Methods ; 324(1-2): 48-62, 2007 Jul 31.
Article in English | MEDLINE | ID: mdl-17553520

ABSTRACT

Langerin CD207 is a type II transmembrane protein. It is responsible for the formation of Birbeck granules, which are intracellular organelles within Langerhans cells, the dendritic cells of stratified squamous epithelia like the epidermis. Because current anti-CD207 antibodies have limitations, we prepared new monoclonals by immunizing rats with the extracellular region of mouse Langerin followed by a boost with enriched Langerhans cells (LCs). We secured a large panel of mAbs, most of which reacted with the carboxy terminal carbohydrate recognition domain. These mAbs could be used to immunoblot and immunoprecipitate mouse Langerin and to stain the cell surface and intracellular pools of CD207 by FACS analysis. Labeling of Birbeck granules was also achieved by immunoelectron microscopy. Anti-CD207 identified LCs in the epidermis and skin draining lymph nodes of BALB/c and C57BL/6 mice, but BALB/c mice had an additional Langerin(+) population in spleen, thymus and mesenteric lymph node. This additional subset had higher levels of CD8 and CD205 than epidermal LCs, and also had a less mature phenotype, i.e., lower MHC II, CD40 and CD86. Subcutaneous injection of IgG but not IgM forms of these new anti-CD207 mAbs led to rapid and selective labeling of the Langerin(+) cells in skin draining lymph nodes as well as spleen. The new IgG anti-CD207 mAbs should be useful for further research on LCs and dendritic cells including an evaluation of the consequences of antigen delivery within anti-CD207 mAbs in vivo.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Antigens, Surface/immunology , Antigens, Surface/metabolism , Binding Sites, Antibody , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Mannose-Binding Lectins/immunology , Mannose-Binding Lectins/metabolism , Animals , Antibodies, Monoclonal/metabolism , Antigens, Surface/genetics , CHO Cells , Cell Line , Cricetinae , Cricetulus , Humans , Immunoglobulin M/biosynthesis , Immunoglobulin M/metabolism , Lectins, C-Type/deficiency , Lectins, C-Type/genetics , Mannose-Binding Lectins/deficiency , Mannose-Binding Lectins/genetics , Mice , Mice, Inbred BALB C , Organ Culture Techniques , Protein Binding/immunology , Protein Structure, Tertiary , Rats , Rats, Inbred WF
17.
Nat Med ; 23(2): 185-191, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28092665

ABSTRACT

Monoclonal antibody 10-1074 targets the V3 glycan supersite on the HIV-1 envelope (Env) protein. It is among the most potent anti-HIV-1 neutralizing antibodies isolated so far. Here we report on its safety and activity in 33 individuals who received a single intravenous infusion of the antibody. 10-1074 was well tolerated and had a half-life of 24.0 d in participants without HIV-1 infection and 12.8 d in individuals with HIV-1 infection. Thirteen individuals with viremia received the highest dose of 30 mg/kg 10-1074. Eleven of these participants were 10-1074-sensitive and showed a rapid decline in viremia by a mean of 1.52 log10 copies/ml. Virologic analysis revealed the emergence of multiple independent 10-1074-resistant viruses in the first weeks after infusion. Emerging escape variants were generally resistant to the related V3-specific antibody PGT121, but remained sensitive to antibodies targeting nonoverlapping epitopes, such as the anti-CD4-binding-site antibodies 3BNC117 and VRC01. The results demonstrate the safety and activity of 10-1074 in humans and support the idea that antibodies targeting the V3 glycan supersite might be useful for the treatment and prevention of HIV-1 infection.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Neutralizing/administration & dosage , HIV Antibodies/administration & dosage , HIV Infections/drug therapy , Viremia/drug therapy , Adult , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Neutralizing/pharmacology , CD4 Lymphocyte Count , CHO Cells , Cricetulus , Female , HIV Antibodies/pharmacology , HIV Envelope Protein gp120/immunology , HIV Infections/blood , HIV-1/genetics , HIV-1/immunology , Humans , Immunoglobulin G/administration & dosage , Immunoglobulin G/pharmacology , Male , Middle Aged , Peptide Fragments/immunology , RNA, Viral/blood , Recombinant Proteins , Viral Load , Viremia/blood , Young Adult , env Gene Products, Human Immunodeficiency Virus/genetics
18.
J Immunol Methods ; 377(1-2): 15-22, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22273672

ABSTRACT

DEC205/CD205 is a C-type multilectin receptor, expressed highly in dendritic cells (DCs). Previous efforts to generate anti-human DEC205 (anti-hDEC205) monoclonal antibodies (mAbs) from mice immunized with subdomain proteins of hDEC205 resulted in a few mAbs. Recently, we expressed and utilized a full-length extracellular domain protein of hDEC205 to successfully generate 5 strong anti-hDEC205 mAbs from mice. In this study, DEC205 knockout (KO) mice were immunized with this full-length extracellular domain protein of hDEC205. One of the 3 immunized DEC205 KO mice was chosen for the highest anti-hDEC205 titer by flow cytometric analysis of serum samples on CHO cells stably expressing hDEC205 (CHO/hDEC205 cells) and used for hybridoma fusion. From a single fusion, more than 400 anti-hDEC205 hybridomas were identified by flow cytometric screen with CHO/hDEC205 cells, and a total of 115 hybridomas secreting strong anti-hDEC205 mAb were saved and named HD1 through HD115. To characterize in detail, 10 HD mAbs were chosen for superior anti-hDEC205 reactivity and further subjected to cloning and purification. Interestingly, out of those 10 chosen anti-hDEC205 HD mAbs, 5 mAbs were also strongly reactive to mouse DEC205 while 8 mAbs were found to stain DEC205(+) DCs on monkey spleen sections. In addition, we also identified that HD83, one of the 10 chosen HD mAbs, stains DEC205(+) DCs in rat spleen and lymph node. Therefore, by immunizing DEC205 KO mice with a full-length extracellular domain protein of hDEC205, we generated a large number of strong anti-hDEC205 mAbs many of which are cross-species reactive and able to visualize DEC205(+) DCs in lymphoid tissues of other mammals.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Antigens, CD/immunology , Dendritic Cells/immunology , Lectins, C-Type/immunology , Receptors, Cell Surface/immunology , Animals , Antibodies, Monoclonal/immunology , CHO Cells , Cloning, Molecular , Cricetinae , Epitopes/immunology , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Minor Histocompatibility Antigens , Recombinant Fusion Proteins/immunology , Specific Pathogen-Free Organisms , Transfection
20.
J Immunol ; 176(2): 991-8, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16393985

ABSTRACT

The infection of cultured monocyte-derived dendritic cells (DCs) with HIV-1 involves CD4 and CCR5 receptors, while transmission to T cells is enhanced at least in part by the lectin DC-SIGN/CD209. In the present study, we studied BDCA-1+ myeloid DCs isolated directly from human blood. These cells express CD4 and low levels of CCR5 and CXCR4 coreceptors, but not DC-SIGN. The myeloid DCs replicate two R5 viruses, BaL and YU2, and transfer infection to activated T cells. The virus productively infects a small fraction of the blood DCs that fail to mature in culture, as indicated by the maturation markers CD83 and DC-LAMP/CD208, and the expression of high CD86 and MHC class II, in contrast to many noninfected DCs. A greater proportion of BDCA-1+ DCs are infected when the virus is pseudotyped with the vesicular stomatitis envelope VSV-G (5-15%), as compared with the R5 virus (0.3-3.5%), indicating that HIV-1 coreceptors may limit the susceptibility of DCs to become infected, or the endocytic route of viral entry used by HIV/vesicular stomatitis virus enhances infectivity. When infected and noninfected cells are purified by cell sorting, the former uniformly express HIV p24 gag and are virtually inactive as stimulators of the allogeneic MLR, in contrast to potent stimulation by noninfected DCs from the same cultures. These results point to two roles for a small fraction of blood DCs in HIV-1 pathogenesis: to support productive infection and to evade the direct induction of T cell-mediated immunity.


Subject(s)
Dendritic Cells/virology , HIV-1/pathogenicity , Antigens, Surface/metabolism , Cell Differentiation , Dendritic Cells/classification , Dendritic Cells/cytology , Dendritic Cells/immunology , HIV Infections/blood , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , HIV-1/physiology , Humans , In Vitro Techniques , Lymphocyte Culture Test, Mixed , T-Lymphocytes/virology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL