Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Haemophilia ; 27(3): e331-e339, 2021 May.
Article in English | MEDLINE | ID: mdl-33772963

ABSTRACT

INTRODUCTION: In patients with haemophilia A undergoing surgery, factor VIII (FVIII) replacement therapy by continuous infusion (CI) may offer an alternative to bolus infusion (BI). AIM: To compare the perioperative haemostatic efficacy and safety of antihaemophilic factor (recombinant) (ADVATE® ; Baxalta US Inc., a Takeda company, Lexington, MA, USA) CI or BI administration. METHODS: In this multicentre, phase III/IV, controlled study (NCT00357656), 60 previously treated adult patients with severe or moderately severe disease undergoing elective unilateral major orthopaedic surgery (knee replacement, n = 48; hip surgery, n = 4; other, n = 8) requiring drain placement were randomized to receive antihaemophilic factor (recombinant) CI (n = 29) or BI (n = 31) through postoperative day 7. Primary outcome measure was cumulative packed red blood cell (PRBC)/blood volume in the drainage fluid within 24 h after surgery, used to establish non-inferiority of CI to BI. RESULTS: CI:BI ratio of cumulative PRBC volume in the 24-h drainage fluid was 0.92 (p-value <.001 for non-inferiority; 95% confidence interval, 0.82-1.05). Total antihaemophilic factor (recombinant) dose per kg body weight received in the combined trans- and postoperative periods was similar with CI and BI to maintain targeted FVIII levels during/after surgery. Treatment-related adverse events (AEs) were reported in five patients treated by CI (eight events) and five treated by BI (six events), including two serious AEs in each arm. CONCLUSION: CI administration of antihaemophilic factor (recombinant) is a viable alternative to BI in patients with haemophilia A undergoing major orthopaedic surgery, providing comparable efficacy and safety.


Subject(s)
Hemophilia A , Orthopedic Procedures , Adult , Blood Coagulation Tests , Factor VIII/therapeutic use , Hemophilia A/drug therapy , Hemophilia A/surgery , Hemostasis , Humans , Recombinant Proteins
2.
Thromb Haemost ; 120(5): 728-736, 2020 May.
Article in English | MEDLINE | ID: mdl-32369844

ABSTRACT

BACKGROUND: Factor VIII (FVIII) trough levels > 1 IU/dL in patients with severe hemophilia A receiving regular prophylaxis may optimize bleed protection. OBJECTIVES: In this post hoc analysis of patients receiving tertiary prophylaxis for approximately 1 year, the relationship between estimated FVIII levels and reported bleeds was investigated to predict the potential for zero bleeds. METHODS: Sixty-three patients (median [range] age, 28 [7-59] years) with severe hemophilia A (229 bleeds) were included. FVIII levels at time of each bleed were estimated from single-dose individual pharmacokinetics. The highest estimated FVIII level at which patients experienced a bleed was considered the "potentially effective trough level" for that bleed type. Kaplan-Meier estimates of proportions of patients with no bleeds above certain estimated FVIII levels were determined. Those not experiencing a bleed in the trial were assumed to have a bleed at 0 IU/dL (pragmatic approach) or at their median trough level (conservative approach). RESULTS: Kaplan-Meier estimates based on pragmatic approach predicted zero all bleeds, joint bleeds, and spontaneous joint bleeds in 1 year in 40, 43, and 63% of patients, respectively, when the potentially effective trough FVIII level was set at 1 IU/dL. Between 1 and 10 IU/dL, every 1 IU/dL rise in estimated FVIII level was associated with an additional 2% of patients having zero all bleeds. CONCLUSION: This post hoc analysis confirms benefits with trough levels of approximately 1 to 3 IU/dL in most patients starting tertiary prophylaxis; prophylaxis with higher trough levels may help patients to achieve zero bleeds.


Subject(s)
Factor VIII/pharmacokinetics , Hemarthrosis/prevention & control , Hemophilia A/drug therapy , Hemostasis/drug effects , Hemostatics/pharmacokinetics , Models, Biological , Tertiary Prevention , Adolescent , Adult , Canada , Child , Europe , Factor VIII/administration & dosage , Hemarthrosis/blood , Hemarthrosis/diagnosis , Hemophilia A/blood , Hemophilia A/diagnosis , Hemostatics/administration & dosage , Hemostatics/blood , Humans , Middle Aged , Risk Factors , Severity of Illness Index , United States , Young Adult
3.
Haemophilia ; 26(3): 478-486, 2020 May.
Article in English | MEDLINE | ID: mdl-32338433

ABSTRACT

INTRODUCTION: Antihaemophilic factor (recombinant) (rAHF; ADVATE® ) is approved for prophylaxis and treatment of bleeding in children and adults with haemophilia A. Reconstitution in 2 mL sterile water for injection instead of 5 mL allows for a 60% reduction in infusion volume and administration time, but could increase the likelihood of hypersensitivity and infusion-related reactions, especially in children. AIM: To assess local tolerability, safety and effectiveness of rAHF 2 mL during routine clinical practice factor VIII (FVIII) replacement (on-demand and prophylaxis) in children with severe (FVIII < 1%) or moderately severe (FVIII 1%-2%) haemophilia A. METHODS: This was a prospective, non-interventional, postauthorization safety surveillance study (NCT02093741). Eligible patients were previously treated with rAHF and had a negative inhibitor test result during ≤10 exposure days prior to study entry. RESULTS: Of 65 patients enrolled (0-11 years of age), 54 and 11 had severe and moderately severe haemophilia A, respectively; 56 patients received prophylaxis, and 11 had ≤50 exposure days, of which 4 had ≤4 exposure days. No patients reported local hypersensitivity reactions, treatment-related adverse events or developed inhibitors. Investigators rated overall effectiveness of rAHF 2 mL prophylaxis as excellent or good. Ninety-four bleeding events in 34 patients were treated. Haemostatic effectiveness was rated as excellent or good for 75.8% of bleeds; 86.2% of bleeds required 1 or 2 infusions. CONCLUSION: In children with severe/moderately severe haemophilia A, no hypersensitivity reactions were reported with rAHF 2 mL treatment, and the safety and effectiveness are consistent with data previously reported for rAHF 5 mL.


Subject(s)
Factor VIII/therapeutic use , Hemophilia A/drug therapy , Water/chemistry , Child , Child, Preschool , Epidemiological Monitoring , Factor VIII/pharmacology , Female , Humans , Infant , Infant, Newborn , Injections , Male
5.
Biologics ; 8: 115-27, 2014.
Article in English | MEDLINE | ID: mdl-24741292

ABSTRACT

Detailed analysis of data from studies of recombinant antihemophilic factor produced using a plasma/albumin-free method (rAHF-PFM) in previously treated patients showed a substantial level of interpatient variation in pharmacokinetics (PKs), factor VIII dosing, and annualized bleed rate (ABR), suggesting that individual patient characteristics contributed to outcome. For example, plasma half-life (t1/2), recovery, and clearance appeared to differ between patients aged <6 years and 10-65 years. Prophylaxis resulted in lower ABRs than episodic treatment in both age groups; better adherence to the prophylactic regimen resulted in a lower ABR in patients aged 10-65 years. The weekly frequency of dosing and adherence to dosing were both significantly and inversely related to the rate of bleeding (young children, P<0.0001 for both all bleeds and joint bleeds; older patients, P<0.0001 for all bleeds and P<0.05 for joint bleeds), as was adherence to dosing frequency (P<0.0001 for all comparisons). A post-marketing randomized study of prophylaxis demonstrated that a PK-guided dosing regimen, based on an individual patient's rAHF-PFM PK (infusion interval, estimated t1/2, and recovery), was as effective as standard prophylaxis and that both prophylactic regimens were superior to episodic treatment with respect to ABR and quality of life measures. Thus, compared with standard prophylaxis, the PK-guided regimen achieved comparable efficacy with fewer weekly infusions. A two-compartment population PK model describes the PK data across the entire age range and forms the basis for future PK-guided therapy with rAHF-PFM. The model confirmed a shorter t1/2 and faster clearance of rAHF-PFM in children <6 years of age versus patients ≥10 years and predicted similar PK parameters with either a full or reduced blood sampling schedule, offering the potential for the use of PK-guided, individualized treatment in the routine clinical care setting.

6.
J Virol ; 87(11): 6073-80, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23536679

ABSTRACT

APOBEC3 proteins mediate potent antiretroviral activity by hypermutating the retroviral genome during reverse transcription. To counteract APOBEC3 and gain a replicative advantage, lentiviruses such as human immunodeficiency virus type 1 (HIV-1) and simian immunodeficiency virus (SIV) have evolved the Vif protein, which targets APOBEC3 proteins for proteasomal degradation. However, the proteasome plays a critical role in the generation of T cell peptide epitopes. Whether Vif-mediated destruction of APOBEC3 proteins leads to the generation and presentation of APOBEC3-derived T cell epitopes on the surfaces of lentivirus-infected cells remains unknown. Here, using peptides derived from multiple Vif-sensitive APOBEC3 proteins, we identified APOBEC3-specific T cell responses in both HIV-1-infected patients and SIV-infected rhesus macaques. These results raise the possibility that these T cell responses may be part of the larger antiretroviral immune response.


Subject(s)
CD8-Positive T-Lymphocytes/virology , Cytidine Deaminase/immunology , Cytosine Deaminase/immunology , HIV Infections/enzymology , HIV-1/physiology , Simian Acquired Immunodeficiency Syndrome/enzymology , Simian Immunodeficiency Virus/physiology , APOBEC-3G Deaminase , Adult , Animals , CD8-Positive T-Lymphocytes/immunology , Cytidine Deaminase/genetics , Cytosine Deaminase/genetics , Female , Gene Products, vif/genetics , Gene Products, vif/immunology , HIV Infections/immunology , HIV Infections/virology , HIV-1/genetics , HIV-1/immunology , Humans , Macaca mulatta , Male , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology
7.
Ann Hematol ; 92(5): 689-98, 2013 May.
Article in English | MEDLINE | ID: mdl-23324986

ABSTRACT

Antihemophilic factor (recombinant), plasma/albumin-free method (rAHF-PFM) was evaluated during routine practice in Germany, among consenting subjects of any age with hemophilia A (HA) and no prior exposure to rAHF-PFM. The treating physician chose the dosing and inhibitor testing frequency. Data were captured for 12 months/subject from diaries and clinic records. Of 152 subjects, 69 % had severe HA, and 89 % had >150 exposure 6 days (ED) at baseline. The majority of subjects (63 %) were treated by continuous prophylaxis (CP). Assignment to CP was more likely for subjects ≥2 years of age and for those with FVIII ≤ 2 %. Median FVIII consumption was 3,548 IU/kg/year for CP and 999 IU/kg/year for continuous on-demand (OD) therapy. Median annual bleed rate was 0.82 for CP and 4.06 for OD. Of 1,218 bleeds, 97 % were home-treated and 68 % of evaluable bleeds involved joints. Based on evaluable subjects' worst ratings, 83/91 (91 %) on CP had a rating of excellent/good for all prophylactic assessments, 55/59 (93 %) on CP and 41/42 (98 %) on OD had a rating of excellent/good for all bleeding assessments. The de novo high-titer FVIII inhibitor rate in subjects with >50 ED at baseline was 1/144 (0.69 %; 95 % CI, 0.02 % to 3.81 %). No high-titer inhibitor occurred in patients with severe HA and >50 ED at baseline. Reduced HRQOL physical scores were predicted by older age (p < 0.0001), HIV positivity (p = 0.02), and presence of ≥1 target joint (p = 0.003). ADVATE rAHF-PFM is safe and efficacious for routine CP or OD management of patients with HA.


Subject(s)
Chemoprevention , Factor VIII/administration & dosage , Hemophilia A/drug therapy , Product Surveillance, Postmarketing , Quality of Life , Adolescent , Adult , Chemoprevention/methods , Child , Child, Preschool , Drug Approval , Germany , Humans , Infant , Infant, Newborn , Middle Aged , Recombinant Proteins/administration & dosage , Serum Albumin/administration & dosage , Young Adult
8.
PLoS One ; 7(6): e39311, 2012.
Article in English | MEDLINE | ID: mdl-22768072

ABSTRACT

BACKGROUND: Translational errors can result in bypassing of the main viral protein reading frames and the production of alternate reading frame (ARF) or cryptic peptides. Within HIV, there are many such ARFs in both sense and the antisense directions of transcription. These ARFs have the potential to generate immunogenic peptides called cryptic epitopes (CE). Both antiretroviral drug therapy and the immune system exert a mutational pressure on HIV-1. Immune pressure exerted by ARF CD8(+) T cells on the virus has already been observed in vitro. HAART has also been described to select HIV-1 variants for drug escape mutations. Since the mutational pressure exerted on one location of the HIV-1 genome can potentially affect the 3 reading frames, we hypothesized that ARF responses would be affected by this drug pressure in vivo. METHODOLOGY/PRINCIPAL FINDINGS: In this study we identified new ARFs derived from sense and antisense transcription of HIV-1. Many of these ARFs are detectable in circulating viral proteins. They are predominantly found in the HIV-1 env nucleotide region. We measured T cell responses to 199 HIV-1 CE encoded within 13 sense and 34 antisense HIV-1 ARFs. We were able to observe that these ARF responses are more frequent and of greater magnitude in chronically infected individuals compared to acutely infected patients, and in patients on HAART, the breadth of ARF responses increased. CONCLUSIONS/SIGNIFICANCE: These results have implications for vaccine design and unveil the existence of potential new epitopes that could be included as vaccine targets.


Subject(s)
Antiretroviral Therapy, Highly Active , Disease Progression , HIV Infections/drug therapy , HIV Infections/immunology , HIV-1/genetics , HIV-1/immunology , Reading Frames/immunology , Acute Disease , Adult , Amino Acid Sequence , Base Sequence , Chronic Disease , Genome, Viral/genetics , Geography , HIV Infections/genetics , Humans , Molecular Sequence Data , Nucleotides/genetics , Peptides/chemistry , Peptides/immunology , Reading Frames/genetics , Recombination, Genetic/genetics , Sequence Deletion , T-Lymphocytes/immunology , env Gene Products, Human Immunodeficiency Virus/chemistry , env Gene Products, Human Immunodeficiency Virus/genetics
9.
Blood ; 119(2): 612-8, 2012 Jan 12.
Article in English | MEDLINE | ID: mdl-22042695

ABSTRACT

Comparison of the pharmacokinetics (PK) of a coagulation factor between groups of patients can be biased by differences in study protocols, in particular between blood sampling schedules. This could affect clinical dose tailoring, especially in children. The aim of this study was to describe the relationships of the PK of factor VIII (FVIII) with age and body weight by a population PK model. The potential to reduce blood sampling was also explored. A model was built for FVIII PK from 236 infusions of recombinant FVIII in 152 patients (1-65 years of age) with severe hemophilia A. The PK of FVIII over the entire age range was well described by a 2-compartment model and a previously reported problem, resulting from differences in blood sampling, to compare findings from children and adults was practically abolished. The decline in FVIII clearance and increase in half-life with age could be described as continuous functions. Retrospective reduction of blood sampling from 11 to 5 samples made no important difference to the estimates of PK parameters. The obtained findings can be used as a basis for PK-based dose tailoring of FVIII in clinical practice, in all age groups, with minimal blood sampling.


Subject(s)
Factor VIII/administration & dosage , Factor VIII/pharmacokinetics , Hemophilia A/prevention & control , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Adolescent , Adult , Age Factors , Aged , Body Weight , Child , Child, Preschool , Dose-Response Relationship, Drug , Female , Hemophilia A/blood , Hemophilia A/epidemiology , Humans , Infant , Male , Middle Aged , Models, Theoretical , Prognosis , Retrospective Studies , Tissue Distribution , United States/epidemiology , Young Adult
10.
J Virol ; 85(14): 6977-85, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21525339

ABSTRACT

Eight percent of the human genome is composed of human endogenous retroviruses (HERVs), which are thought to be inactive remnants of ancient infections. Previously, we showed that individuals with early HIV-1 infection have stronger anti-HERV T cell responses than uninfected controls. In this study, we investigated whether these responses persist in chronic HIV-1 infection and whether they have a role in the control of HIV-1. Peripheral blood mononuclear cells (PBMCs) from 88 subjects diagnosed with HIV-1 infection for at least 1 year (median duration of diagnosis, 13 years) were tested for responses against HERV peptides in gamma interferon (IFN-γ) enzyme immunospot (ELISPOT) assays. Individuals who control HIV-1 viremia without highly active antiretroviral therapy (HAART) had stronger and broader HERV-specific T cell responses than HAART-suppressed patients, virologic noncontrollers, immunologic progressors, and uninfected controls (P < 0.05 for each pairwise comparison). In addition, the magnitude of the anti-HERV T cell response was inversely correlated with HIV-1 viral load (r(2) = 0.197, P = 0.0002) and associated with higher CD4(+) T cell counts (r(2) = 0.072, P = 0.027) in untreated patients. Flow cytometric analyses of an HLA-B51-restricted CD8(+) HERV response in one HIV-1-infected individual revealed a less activated and more differentiated phenotype than that stimulated by a homologous HIV-1 peptide. HLA-B51 tetramer dual staining within this individual confirmed two different T cell populations corresponding to these HERV and HIV-1 epitopes, ruling out cross-reactivity. These findings suggest a possible role for anti-HERV immunity in the control of chronic HIV-1 infection and provide support for a larger effort to design an HIV-1 vaccine that targets conserved antigens such as HERV.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Endogenous Retroviruses/immunology , HIV Infections/immunology , HIV-1/physiology , Antiretroviral Therapy, Highly Active , Chronic Disease , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Gene Products, env/immunology , Gene Products, gag/immunology , HIV Infections/drug therapy , HIV Infections/virology , Humans , Immunophenotyping , Male
11.
PLoS One ; 5(4): e10249, 2010 04 21.
Article in English | MEDLINE | ID: mdl-20422053

ABSTRACT

BACKGROUND: The HLA-B*35-Px allele has been associated with rapid disease progression in HIV-1 infection, in contrast to the HLA-B*35-Py allele. METHODOLOGY/PRINCIPAL FINDINGS: Immune responses to two HLA-B*35 restricted HIV-1 specific CTL epitopes and their variants were followed longitudinally during early HIV-1 infection in 16 HLA-B*35+ individuals. Subjects expressing HLA-B*35-Px alleles showed no difference in response to the consensus epitopes compared to individuals with HLA-B*35-Py alleles. Surprisingly, all the HLA-B*35-Px+ individuals responded to epitope-variants even in the absence of a consensus response. Sequencing of the viral population revealed no evidence of variant virus in any of the individuals. CONCLUSIONS/SIGNIFICANCE: This demonstrates a novel phenomenon that distinguishes individuals with the HLA-B*35-Px rapid progressing allele and those with the HLA-B*35-Py slower progressing allele.


Subject(s)
Alleles , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , HIV-1/immunology , HLA-B35 Antigen/genetics , T-Lymphocytes, Cytotoxic/immunology , CD4 Lymphocyte Count , Disease Progression , Epitopes, T-Lymphocyte/genetics , Genetic Variation , HIV Infections/blood , HIV Infections/diagnosis , HIV Infections/immunology , Humans , Longitudinal Studies , T-Cell Antigen Receptor Specificity , Viral Load
12.
J Acquir Immune Defic Syndr ; 53(1): 36-46, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19910798

ABSTRACT

We analyzed immune responses in chronically HIV-infected individuals who took part in a treatment interruption (TI) trial designed for patients who initiated antiretroviral therapy within 6 months of seroconversion. In the 2 subjects who exhibited the best viral control, we detected CD8(+) T-cell responses against 1-2 Gag epitopes during the early weeks of TI and a subsequent increase in the number of epitopes recognized by the later time points. Each of these subjects developed mutations within the epitopes targeted by the highest magnitude responses. In the subject with the worst viral control, we detected responses against 2 Gag epitopes throughout the entire TI and no Gag mutations. The magnitude of these responses increased dramatically with time, greatly exceeding those detected in the virologic controllers. The highest levels of contemporaneous autologous neutralizing antibody activity were detected in the virologic controllers, and a subsequent escape mutation developed within the envelope gene of one controller that abrogated the response. These data suggest that immune escape mutations are a sign of viral control during TI, and that the absence of immune escape mutations in the presence of high levels of viral replication indicates the lack of an effective host immune response.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes/immunology , HIV Infections/immunology , HIV-1/genetics , HIV-1/immunology , Withholding Treatment , Antibodies, Neutralizing/blood , Antiretroviral Therapy, Highly Active , CD4 Lymphocyte Count , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Epitopes/genetics , HIV Envelope Protein gp160/immunology , HIV Infections/drug therapy , HIV Infections/virology , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Longitudinal Studies , Mutation , Neutralization Tests , Viral Load , Virus Replication/genetics , Virus Replication/immunology , gag Gene Products, Human Immunodeficiency Virus/immunology
13.
J Exp Med ; 205(12): 2763-79, 2008 Nov 24.
Article in English | MEDLINE | ID: mdl-19001139

ABSTRACT

Progressive loss of T cell functionality is a hallmark of chronic infection with human immunodeficiency virus 1 (HIV-1). We have identified a novel population of dysfunctional T cells marked by surface expression of the glycoprotein Tim-3. The frequency of this population was increased in HIV-1-infected individuals to a mean of 49.4 +/- SD 12.9% of CD8(+) T cells expressing Tim-3 in HIV-1-infected chronic progressors versus 28.5 +/- 6.8% in HIV-1-uninfected individuals. Levels of Tim-3 expression on T cells from HIV-1-infected inviduals correlated positively with HIV-1 viral load and CD38 expression and inversely with CD4(+) T cell count. In progressive HIV-1 infection, Tim-3 expression was up-regulated on HIV-1-specific CD8(+) T cells. Tim-3-expressing T cells failed to produce cytokine or proliferate in response to antigen and exhibited impaired Stat5, Erk1/2, and p38 signaling. Blocking the Tim-3 signaling pathway restored proliferation and enhanced cytokine production in HIV-1-specific T cells. Thus, Tim-3 represents a novel target for the therapeutic reversal of HIV-1-associated T cell dysfunction.


Subject(s)
HIV Infections/immunology , HIV-1/immunology , Membrane Proteins/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antiretroviral Therapy, Highly Active , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , Disease Progression , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/immunology , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/immunology , HIV Infections/drug therapy , HIV Infections/pathology , HIV Infections/physiopathology , HIV-1/pathogenicity , HLA Antigens , Hepatitis A Virus Cellular Receptor 2 , Humans , Membrane Proteins/genetics , Phenotype , Programmed Cell Death 1 Receptor , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , Signal Transduction/immunology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocytes/cytology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/immunology
14.
J Infect Dis ; 197(1): 117-25, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18171294

ABSTRACT

Natural killer (NK) cells are believed to play a role in human immunodeficiency virus type 1 (HIV-1) disease progression, and NK cell levels are reduced in individuals with chronic HIV-1 infection. Interleukin (IL)-2 therapy results in an expansion of CD4(+) T cells as well as NK cells; however, little is known about the detailed effects of IL-2 therapy on NK cells in HIV-1 infection in general and in early infection in particular. Here, we investigated the effects of combined IL-2 therapy and antiretroviral therapy (ART) on the number, frequency, phenotype, and interferon (IFN)-gamma production of NK cells in individuals with early HIV-1 infection. Patients randomized to receive combined ART and IL-2 therapy predominantly expanded CD56(dim) NK cells, and the expansion was greater than in patients randomized to receive ART alone. Importantly, NK cell receptor expression and IFN-gamma production were maintained over time. This reconstitution of NK cells may be useful in helping contain viremia if patients discontinue therapy or develop drug resistance.


Subject(s)
Adjuvants, Immunologic/pharmacology , CD56 Antigen/immunology , HIV Infections/drug therapy , HIV-1/drug effects , Interleukin-2/pharmacology , Killer Cells, Natural/drug effects , Adult , Anti-Retroviral Agents/therapeutic use , Drug Therapy, Combination , Female , HIV Infections/immunology , HIV-1/immunology , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Male , Prospective Studies
15.
J Leukoc Biol ; 83(2): 254-62, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17982112

ABSTRACT

During the course of HIV-1 infection, the status of immune activation has been determined to be a powerful indicator of disease progression. The immune system has adopted self-regulatory mechanisms to counterbalance undesirable immune responses. CD25+CD4+ T regulatory (Treg) cells that express the transcription regulator, forkhead box P3 (FOXP3), play an important role in this immunosuppression. Using a combination of Treg cell discriminatory markers (FOXP3, CD25, CD127), we predicted that an expansion of Treg cell subsets would negatively correlate with immune activation during the early stages of HIV-1 infection. We report that FOXP3+CD127lo expressing CD4+ T cells increases in primary HIV-1 infection over time. Furthermore, the FOXP3+CD127lo CD4+ T cells may, in fact, reduce the levels of T cell activation following primary infection. It is interesting that the positive correlation between FOXP3+CD127lo CD4+ and CD25+CD127lo CD4+ T cells noted in HIV-uninfected persons is not only lost but may also be reversed in early, chronic HIV-1 infection. Unlike FOXP3+CD127lo CD4+, the level of FOXP3+CD25+CD127lo CD4+ T cells did not correlate with T cell activation, suggesting that these cells were not effective in reducing T cell activation. These observations suggest that different Treg populations may have different effects on reducing immune activation in HIV-1 infection and that the FOXP3+CD127lo CD4+ T cell population may be particularly important in limiting immune activation.


Subject(s)
ADP-ribosyl Cyclase 1/analysis , CD8-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/analysis , HIV Infections/immunology , HIV-1 , Interleukin-7 Receptor alpha Subunit/analysis , Membrane Glycoproteins/analysis , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , CD4 Lymphocyte Count , Cell Division , Disease Progression , Humans , Immunophenotyping , Interleukin-2 Receptor alpha Subunit/analysis , Lymphocyte Activation
16.
PLoS Pathog ; 3(11): e165, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17997601

ABSTRACT

Human endogenous retroviruses (HERVs) are remnants of ancient infectious agents that have integrated into the human genome. Under normal circumstances, HERVs are functionally defective or controlled by host factors. In HIV-1-infected individuals, intracellular defense mechanisms are compromised. We hypothesized that HIV-1 infection would remove or alter controls on HERV activity. Expression of HERV could potentially stimulate a T cell response to HERV antigens, and in regions of HIV-1/HERV similarity, these T cells could be cross-reactive. We determined that the levels of HERV production in HIV-1-positive individuals exceed those of HIV-1-negative controls. To investigate the impact of HERV activity on specific immunity, we examined T cell responses to HERV peptides in 29 HIV-1-positive and 13 HIV-1-negative study participants. We report T cell responses to peptides derived from regions of HERV detected by ELISPOT analysis in the HIV-1-positive study participants. We show an inverse correlation between anti-HERV T cell responses and HIV-1 plasma viral load. In HIV-1-positive individuals, we demonstrate that HERV-specific T cells are capable of killing cells presenting their cognate peptide. These data indicate that HIV-1 infection leads to HERV expression and stimulation of a HERV-specific CD8+ T cell response. HERV-specific CD8+ T cells have characteristics consistent with an important role in the response to HIV-1 infection: a phenotype similar to that of T cells responding to an effectively controlled virus (cytomegalovirus), an inverse correlation with HIV-1 plasma viral load, and the ability to lyse cells presenting their target peptide. These characteristics suggest that elicitation of anti-HERV-specific immune responses is a novel approach to immunotherapeutic vaccination. As endogenous retroviral sequences are fixed in the human genome, they provide a stable target, and HERV-specific T cells could recognize a cell infected by any HIV-1 viral variant. HERV-specific immunity is an important new avenue for investigation in HIV-1 pathogenesis and vaccine design.


Subject(s)
Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Endogenous Retroviruses/immunology , HIV Infections/virology , Cohort Studies , Cross-Sectional Studies , Flow Cytometry , HIV Infections/immunology , HIV-1 , Humans , RNA, Viral/blood , Reverse Transcriptase Polymerase Chain Reaction , Viral Load
17.
J Virol ; 81(11): 5759-65, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17376902

ABSTRACT

CD8(+) T cells are believed to play an important role in the control of human immunodeficiency virus type 1 (HIV-1) infection. However, despite intensive efforts, it has not been possible to consistently link the overall magnitude of the CD8(+) T-cell response with control of HIV-1. Here, we have investigated the association of different CD8(+) memory T-cell subsets responding to HIV-1 in early infection with future control of HIV-1 viremia. Our results demonstrate that both a larger proportion and an absolute number of HIV-1-specific CD8(+) CCR7(-) CD45RA(+) effector memory T cells (T(EMRA) cells) were associated with a lower future viral load set point. In contrast, a larger absolute number of HIV-1-specific CD8(+) CCR7(-) CD45RA(-) effector memory T cells (T(EM)) was not related to the viral load set point. Overall, the findings suggest that CD8(+) T(EMRA) cells have superior antiviral activity and indicate that both qualitative and quantitative aspects of the CD8(+) T-cell response need to be considered when defining the characteristics of protective immunity to HIV-1.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , HIV Infections/virology , HIV-1/immunology , T-Lymphocyte Subsets/immunology , Viral Load , Viremia/immunology , Viremia/virology , Adult , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Female , Gene Products, gag/immunology , HIV Infections/immunology , HIV Infections/pathology , Humans , Immunologic Memory , Immunophenotyping , Lymphocyte Count , Male , Middle Aged , Predictive Value of Tests , T-Lymphocyte Subsets/pathology , T-Lymphocyte Subsets/virology , Viremia/pathology
18.
PLoS One ; 2(2): e225, 2007 Feb 21.
Article in English | MEDLINE | ID: mdl-17311088

ABSTRACT

BACKGROUND: Antigen-specific CTL responses are thought to play a central role in containment of HIV-1 infection, but no consistent correlation has been found between the magnitude and/or breadth of response and viral load changes during disease progression. METHODS AND FINDINGS: We undertook a detailed investigation of longitudinal CTL responses and HIV-1 evolution beginning with primary infection in 11 untreated HLA-A2 positive individuals. A subset of patients developed broad responses, which selected for consensus B epitope variants in Gag, Pol, and Nef, suggesting CTL-induced adaptation of HIV-1 at the population level. The patients who developed viral escape mutations and broad autologous CTL responses over time had a significantly higher increase in viral load during the first year of infection compared to those who did not develop viral escape mutations. CONCLUSIONS: A continuous dynamic development of CTL responses was associated with viral escape from temporarily effective immune responses. Our results suggest that broad CTL responses often represent footprints left by viral CTL escape rather than effective immune control, and help explain earlier findings that fail to show an association between breadth of CTL responses and viral load. Our results also demonstrate that CTL pressures help to maintain certain elements of consensus viral sequence, which likely represent viral escape from common HLA-restricted CTL responses. The ability of HIV to evolve to escape CTL responses restricted by a common HLA type highlights the challenges posed to development of an effective CTL-based vaccine.


Subject(s)
HIV Antigens/immunology , HIV Infections/immunology , HIV-1/immunology , Immune Evasion , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Substitution , Base Sequence , Consensus Sequence , Disease Progression , Epitopes/genetics , Epitopes/immunology , Evolution, Molecular , Female , HIV Antigens/genetics , HIV-1/genetics , HLA-A2 Antigen/immunology , Humans , Immune Evasion/genetics , Interferon-gamma/blood , Male , Molecular Sequence Data , Phylogeny , Sequence Alignment , Sequence Homology, Nucleic Acid , Tumor Necrosis Factor-alpha/analysis , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/immunology , nef Gene Products, Human Immunodeficiency Virus/genetics , nef Gene Products, Human Immunodeficiency Virus/immunology , pol Gene Products, Human Immunodeficiency Virus/genetics , pol Gene Products, Human Immunodeficiency Virus/immunology
19.
AIDS ; 21(2): 239-41, 2007 Jan 11.
Article in English | MEDLINE | ID: mdl-17197817

ABSTRACT

We determined whether methamphetamine use is associated with the increased prevalence of primary HIV drug resistance among a cohort of men who have sex with men recently infected with HIV. In multivariate analysis, we found that frequent methamphetamine use was strongly associated with primary non-nucleoside reverse transcriptase inhibitor resistance, but not with protease inhibitor or nucleoside reverse transcriptase inhibitor resistance. We postulate that this association may be caused by methamphetamine-associated treatment interruptions among source partners.


Subject(s)
Anti-HIV Agents/pharmacology , Drug Resistance, Viral , HIV-1/drug effects , Methamphetamine , Substance-Related Disorders/complications , Adult , Cross-Sectional Studies , HIV Infections/complications , HIV Infections/drug therapy , HIV Infections/transmission , HIV Infections/virology , Homosexuality, Male , Humans , Male , Reverse Transcriptase Inhibitors/pharmacology
20.
AIDS Res Hum Retroviruses ; 22(8): 757-62, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16910831

ABSTRACT

Perturbations of plasma IL-10, IL-12, IL-13, TNF-alpha, and IFN-gamma were measured longitudinally in HIV-1 seroconverting plasma donors and were compared to subjects with symptomatic primary HIV-1 infection. Control groups included uninfected patients with symptoms and risks for primary HIV-1, healthy controls, and asymptomatic plasma donors with primary HCV. IL-10, TNF-alpha, and IFN-gamma rapidly rose in acute HIV-1 infection, while IL-13 predominated in acute HCV. Subjects with symptomatic primary HIV-1 had higher cytokine levels than asymptomatic subjects, statistically significant for TNF-alpha. Cytokine alterations occurred within 7 days of detectable HIV-1 viremia, emphasizing the need to study the earliest events of infection.


Subject(s)
Cytokines/blood , HIV Infections/immunology , HIV-1/immunology , Biomarkers/blood , Blood Donors , Female , HIV Infections/blood , Hepacivirus/immunology , Hepatitis C/blood , Hepatitis C/immunology , Humans , Interferon-alpha/blood , Male , Plasmapheresis , Tumor Necrosis Factor-alpha/metabolism , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL
...