Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 189
Filter
Add more filters

Publication year range
1.
Nature ; 543(7646): 559-563, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28289286

ABSTRACT

Highly potent and broadly neutralizing anti-HIV-1 antibodies (bNAbs) have been used to prevent and treat lentivirus infections in humanized mice, macaques, and humans. In immunotherapy experiments, administration of bNAbs to chronically infected animals transiently suppresses virus replication, which invariably returns to pre-treatment levels and results in progression to clinical disease. Here we show that early administration of bNAbs in a macaque simian/human immunodeficiency virus (SHIV) model is associated with very low levels of persistent viraemia, which leads to the establishment of T-cell immunity and resultant long-term infection control. Animals challenged with SHIVAD8-EO by mucosal or intravenous routes received a single 2-week course of two potent passively transferred bNAbs (3BNC117 and 10-1074 (refs 13, 14)). Viraemia remained undetectable for 56-177 days, depending on bNAb half-life in vivo. Moreover, in the 13 treated monkeys, plasma virus loads subsequently declined to undetectable levels in 6 controller macaques. Four additional animals maintained their counts of T cells carrying the CD4 antigen (CD4+) and very low levels of viraemia persisted for over 2 years. The frequency of cells carrying replication-competent virus was less than 1 per 106 circulating CD4+ T cells in the six controller macaques. Infusion of a T-cell-depleting anti-CD8ß monoclonal antibody to the controller animals led to a specific decline in levels of CD8+ T cells and the rapid reappearance of plasma viraemia. In contrast, macaques treated for 15 weeks with combination anti-retroviral therapy, beginning on day 3 after infection, experienced sustained rebound plasma viraemia when treatment was interrupted. Our results show that passive immunotherapy during acute SHIV infection differs from combination anti-retroviral therapy in that it facilitates the emergence of potent CD8+ T-cell immunity able to durably suppress virus replication.


Subject(s)
HIV Infections/immunology , HIV Infections/therapy , HIV/immunology , Immunization, Passive , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/therapy , Simian Immunodeficiency Virus/immunology , Animals , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/therapeutic use , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/therapeutic use , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Combined Modality Therapy , Disease Models, Animal , Female , HIV/drug effects , HIV/isolation & purification , HIV Antibodies/administration & dosage , HIV Antibodies/immunology , HIV Antibodies/therapeutic use , HIV Infections/virology , Half-Life , Macaca mulatta , Male , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/isolation & purification , Viral Load/drug effects , Viral Load/immunology , Viremia/immunology , Viremia/therapy , Virus Replication/drug effects , Virus Replication/immunology
2.
J Virol ; 95(18): e0026821, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34190597

ABSTRACT

Preventing human immunodeficiency virus (HIV) infection in newborns by vertical transmission remains an important unmet medical need in resource-poor areas where antiretroviral therapy (ART) is not available and mothers and infants cannot be treated prepartum or during the breastfeeding period. In the present study, the protective efficacy of the potent HIV-neutralizing antibodies PGT121 and VRC07-523, both produced in plants, were assessed in a multiple-SHIV (simian-human immunodeficiency virus)-challenge breastfeeding macaque model. Newborn macaques received either six weekly subcutaneous injections with PGT121 alone or as a cocktail of PGT121-LS plus VRC07-523-LS injected three times every 2 weeks. Viral challenge with SHIVSF162P3 was twice weekly over 5.5 weeks using 11 exposures. Despite the transient presence of plasma viral RNA either immediately after the first challenge or as single-point blips, the antibodies prevented a productive infection in all babies with no sustained plasma viremia, compared to viral loads ranging from 103 to 5 × 108 virions/ml in four untreated controls. No virus was detected in peripheral blood mononuclear cells (PBMCs), and only 3 of 159 tissue samples were weakly positive in the treated babies. Newborn macaques proved to be immunocompetent, producing transient anti-Env antibodies and anti-drug antibody (ADA), which were maintained in the circulation after passive broadly neutralizing antibody clearance. ADA responses were directed to the IgG1 Fc CH2-CH3 domains, which has not been observed to date in adult monkeys passively treated with PGT121 or VRC01. In addition, high levels of VRC07-523 anti-idiotypic antibodies in the circulation of one newborn was concomitant with the rapid elimination of VRC07. Plant-expressed antibodies show promise as passive immunoprophylaxis in a breastfeeding model in newborns. IMPORTANCE Plant-produced human neutralizing antibody prophylaxis is highly effective in preventing infection in newborn monkeys during repeated oral exposure, modeling virus in breastmilk, and offers advantages in cost of production and safety. These findings raise the possibility that anti-Env antibodies may contribute to the control of viral replication in this newborn model and that the observed immune responsiveness may be driven by the long-lived presence of immune complexes.


Subject(s)
Breast Feeding , Broadly Neutralizing Antibodies/immunology , HIV-1/physiology , Immunization, Passive/methods , Nicotiana/immunology , Simian Acquired Immunodeficiency Syndrome/therapy , Simian Immunodeficiency Virus/immunology , Animals , Animals, Newborn , Female , HIV Infections/immunology , HIV Infections/therapy , HIV Infections/virology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Macaca mulatta , Male , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Nicotiana/virology , Viremia/immunology , Viremia/therapy , Viremia/virology
3.
Immunol Rev ; 285(1): 9-25, 2018 09.
Article in English | MEDLINE | ID: mdl-30129208

ABSTRACT

The odds of living a long and healthy life with HIV infection have dramatically improved with the advent of combination antiretroviral therapy. Along with the early development and clinical trials of these drugs, and new field of research emerged called viral dynamics, which uses mathematical models to interpret and predict the time-course of viral levels during infection and how they are altered by treatment. In this review, we summarize the contributions that virus dynamics models have made to understanding the pathophysiology of infection and to designing effective therapies. This includes studies of the multiphasic decay of viral load when antiretroviral therapy is given, the evolution of drug resistance, the long-term persistence latently infected cells, and the rebound of viremia when drugs are stopped. We additionally discuss new work applying viral dynamics models to new classes of investigational treatment for HIV, including latency-reversing agents and immunotherapy.


Subject(s)
HIV Infections/therapy , HIV/physiology , Immunotherapy/methods , Models, Biological , Viremia/therapy , Virus Latency , Animals , Anti-Retroviral Agents/therapeutic use , Drug Resistance , HIV Infections/immunology , Humans , Treatment Outcome , Viral Load , Viremia/immunology , Virus Activation , Withholding Treatment
4.
BMC Infect Dis ; 21(1): 716, 2021 Jul 30.
Article in English | MEDLINE | ID: mdl-34330230

ABSTRACT

BACKGROUND: Hepatitis B virus (HBV) remains a major global public health problem worldwide; in endemic areas, mother-to-child transmission (MTCT) of HBV is the most common transmission route. Previous studies have shown that amniocentesis for prenatal diagnosis increases the risk of MTCT of HBV among highly viraemic mothers. However, no data is available on MTCT related fetal blood sampling (FBS) because of the paucity of cases or lack of attention. We present a case series of HBV-infected women who underwent FBS with or without antiviral therapy during pregnancy and discuss the risk of MTCT after FBS. CASE PRESENTATION: Six hepatitis B surface antigen (HBsAg)-positive pregnant women who underwent FBS for prenatal diagnosis were retrospectively reviewed. Their infants were followed up with HBV serology parameters until at least 12 months of age. Among 6 cases, two hepatitis B e-antigen (HBeAg)-positive mothers had high viral loads > 7.0 log10 IU/mL, and one of them received antiviral therapy at 26+ 3 gestational weeks and achieved an anticipated level of 4.52 log10 IU/mL before FBS, while the other one did not receive any antiviral treatment. The other 4 cases were HBeAg-negative with low viral loads. Only a child born to the HBeAg-positive mother, who had no antiviral therapy with a viral load of 7.48 log10 IU/mL before FBS, was found to have MTCT with HBsAg persistently positive from birth to 12 months of age. The other 5 children were both HBsAg-negative and HBsAb-positive at the end of follow-up. CONCLUSIONS: FBS may increase the risk of MTCT of HBV in women with HBeAg-positive and high viral loads; therefore, FBS should be avoided in this high-risk population. Maternal HBV serologic testing and awareness of the potential risk of MTCT should be recommended before FBS. Antiviral therapy may be effective to decrease the risk of MTCT after FBS in highly viraemic women.


Subject(s)
Fetal Blood/virology , Hepatitis B/transmission , Infectious Disease Transmission, Vertical , Pregnancy Complications, Infectious , Adult , Female , Hepatitis B/diagnosis , Hepatitis B/therapy , Hepatitis B/virology , Hepatitis B Surface Antigens/blood , Hepatitis B e Antigens/blood , Hepatitis B virus/isolation & purification , Humans , Infant , Infant, Newborn , Male , Pregnancy , Pregnancy Complications, Infectious/diagnosis , Pregnancy Complications, Infectious/virology , Prenatal Diagnosis , Retrospective Studies , Risk Factors , Viral Load , Viremia/therapy , Viremia/transmission
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
6.
Curr HIV/AIDS Rep ; 17(5): 499-506, 2020 10.
Article in English | MEDLINE | ID: mdl-32671567

ABSTRACT

PURPOSE OF REVIEW: The application of immunotherapies to HIV presents a new horizon of treatment options, but little is known about what impact they may have on the central nervous system (CNS). Here we review the most promising immunotherapeutic strategies that can be used to target HIV in the CNS and focus on identifying their potential benefits while exploring the challenges that remain in their application. RECENT FINDINGS: We have identified five immunotherapeutic strategies that hold potential in managing CNS disease among HIV-infected patients. These include broadly neutralizing antibodies, multi-affinity antibodies, CAR-T cell therapy, checkpoint inhibitors, and therapeutic vaccines. Each class of immunotherapy presents unique mechanisms by which CNS viremia and latency may be addressed but are faced with several challenges. CAR-T cell therapy and multi-affinity antibodies seem to hold promise, but combination therapy is likely to be most effective. However, more human trials are needed before the clinical benefits of these therapies are realized.


Subject(s)
Antibodies, Neutralizing/therapeutic use , HIV Antibodies/therapeutic use , HIV Infections/therapy , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy, Adoptive/methods , Viral Vaccines/therapeutic use , Antibodies, Neutralizing/immunology , Central Nervous System/pathology , Central Nervous System/virology , Combined Modality Therapy , HIV Antibodies/immunology , HIV Infections/immunology , Humans , Viremia/therapy
7.
Clin Infect Dis ; 68(8): 1406-1409, 2019 04 08.
Article in English | MEDLINE | ID: mdl-30357314

ABSTRACT

Here, we report the case of severe adenoviremia in a 7-year-old boy with highly-resistant, acute leukemia. A combined approach of αßTCR-CD19-depleted stem cell transplantation, enabling immunosuppression-free post-transplant care, and early transfer of adenovirus-specific donor T cells during aplasia resulted in rapid and complete clearance of the treatment-refractory adenoviremia.


Subject(s)
Adenovirus Infections, Human/therapy , Hematopoietic Stem Cell Transplantation , T-Lymphocytes/transplantation , T-Lymphocytes/virology , Viremia/therapy , Adenovirus Infections, Human/etiology , Antigens, CD19 , Child , Combined Modality Therapy , Fatal Outcome , Humans , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/virology
8.
Nature ; 503(7475): 224-8, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24172905

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1)-specific monoclonal antibodies with extraordinary potency and breadth have recently been described. In humanized mice, combinations of monoclonal antibodies have been shown to suppress viraemia, but the therapeutic potential of these monoclonal antibodies has not yet been evaluated in primates with an intact immune system. Here we show that administration of a cocktail of HIV-1-specific monoclonal antibodies, as well as the single glycan-dependent monoclonal antibody PGT121, resulted in a rapid and precipitous decline of plasma viraemia to undetectable levels in rhesus monkeys chronically infected with the pathogenic simian-human immunodeficiency virus SHIV-SF162P3. A single monoclonal antibody infusion afforded up to a 3.1 log decline of plasma viral RNA in 7 days and also reduced proviral DNA in peripheral blood, gastrointestinal mucosa and lymph nodes without the development of viral resistance. Moreover, after monoclonal antibody administration, host Gag-specific T-lymphocyte responses showed improved functionality. Virus rebounded in most animals after a median of 56 days when serum monoclonal antibody titres had declined to undetectable levels, although, notably, a subset of animals maintained long-term virological control in the absence of further monoclonal antibody infusions. These data demonstrate a profound therapeutic effect of potent neutralizing HIV-1-specific monoclonal antibodies in SHIV-infected rhesus monkeys as well as an impact on host immune responses. Our findings strongly encourage the investigation of monoclonal antibody therapy for HIV-1 in humans.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , HIV-1/immunology , Simian Acquired Immunodeficiency Syndrome/therapy , Simian Immunodeficiency Virus/physiology , Animals , DNA, Viral/blood , HIV Antibodies/immunology , Macaca mulatta , T-Lymphocytes/immunology , Viremia/therapy
9.
Nature ; 503(7475): 277-80, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24172896

ABSTRACT

Neutralizing antibodies can confer immunity to primate lentiviruses by blocking infection in macaque models of AIDS. However, earlier studies of anti-human immunodeficiency virus type 1 (HIV-1) neutralizing antibodies administered to infected individuals or humanized mice reported poor control of virus replication and the rapid emergence of resistant variants. A new generation of anti-HIV-1 monoclonal antibodies, possessing extraordinary potency and breadth of neutralizing activity, has recently been isolated from infected individuals. These neutralizing antibodies target different regions of the HIV-1 envelope glycoprotein including the CD4-binding site, glycans located in the V1/V2, V3 and V4 regions, and the membrane proximal external region of gp41 (refs 9-14). Here we have examined two of the new antibodies, directed to the CD4-binding site and the V3 region (3BNC117 and 10-1074, respectively), for their ability to block infection and suppress viraemia in macaques infected with the R5 tropic simian-human immunodeficiency virus (SHIV)-AD8, which emulates many of the pathogenic and immunogenic properties of HIV-1 during infections of rhesus macaques. Either antibody alone can potently block virus acquisition. When administered individually to recently infected macaques, the 10-1074 antibody caused a rapid decline in virus load to undetectable levels for 4-7 days, followed by virus rebound during which neutralization-resistant variants became detectable. When administered together, a single treatment rapidly suppressed plasma viraemia for 3-5 weeks in some long-term chronically SHIV-infected animals with low CD4(+) T-cell levels. A second cycle of anti-HIV-1 monoclonal antibody therapy, administered to two previously treated animals, successfully controlled virus rebound. These results indicate that immunotherapy or a combination of immunotherapy plus conventional antiretroviral drugs might be useful as a treatment for chronically HIV-1-infected individuals experiencing immune dysfunction.


Subject(s)
Antibodies, Neutralizing/therapeutic use , HIV Antibodies/therapeutic use , HIV-1/immunology , Immunotherapy , Simian Acquired Immunodeficiency Syndrome/therapy , Simian Immunodeficiency Virus/physiology , Viremia/therapy , Animals , Binding Sites/immunology , CD4 Antigens/metabolism , HIV Envelope Protein gp120/immunology , Macaca/immunology , Molecular Sequence Data , Peptide Fragments/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Time Factors , Viral Load
10.
Retrovirology ; 15(1): 73, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30445968

ABSTRACT

Novel broadly neutralizing antibodies targeting HIV-1 hold promise for their use in the prevention and treatment of HIV-1 infection. Pre-clinical results have encouraged the evaluation of these antibodies in healthy and HIV-1-infected humans. In first clinical trials, highly potent broadly neutralizing antibodies have demonstrated their safety and significant antiviral activity by reducing viremia and delaying the time to viral rebound in individuals interrupting antiretroviral therapy. While emerging antibody-resistant viral variants have indicated limitations of antibody monotherapy, strategies to enhance the efficacy of broadly neutralizing antibodies in humans are under investigation. These include the use of antibody combinations to prevent viral escape, antibody modifications to increase the half-life and the co-administration of latency-reversing agents to target the cellular reservoir of HIV-1. We provide an overview of the results of pre-clinical and clinical studies of broadly HIV-1 neutralizing antibodies, discuss their implications and highlight approaches for the ongoing advancement into humans.


Subject(s)
Antibodies, Neutralizing/therapeutic use , HIV Antibodies/therapeutic use , HIV Infections/prevention & control , HIV Infections/therapy , Immunization, Passive , Animals , Antibodies, Monoclonal/therapeutic use , Antiviral Agents/therapeutic use , Clinical Trials as Topic , Disease Models, Animal , HIV-1 , Humans , Mice , Viremia/therapy
12.
Clin Transplant ; 31(8)2017 08.
Article in English | MEDLINE | ID: mdl-28509373

ABSTRACT

BACKGROUND: While screening for asymptomatic BK viremia (BKV) has been well studied in isolated kidney transplant recipients, there is a paucity of published outcomes in simultaneous pancreas-kidney (SPK) transplant recipients who underwent BKV screening followed by pre-emptive reduction in immunosuppression. METHODS: This is a single-center, retrospective review of 31 consecutive SPK recipients who were transplanted over a 5-year period following the initiation of a serum BKV screening protocol. RESULTS: BK viremia developed in 11 (35.5%) patients, and all patients achieved complete viral clearance following reduction in immunosuppression. Two patients (6.5%) developed BK virus nephropathy, but both had preserved allograft function. One patient developed mild rejection of the kidney allograft following clearance of BKV, and two patients developed mild rejection of the pancreas allograft after reduction in immunosuppression, but there were no kidney or pancreas allograft losses due to rejection. The development of BK viremia did not impact overall patient survival or kidney and pancreas allograft survival. CONCLUSION: Screening asymptomatic SPK recipients for BKV followed by reduction in maintenance immunosuppression appears to be an effective strategy to prevent kidney allograft dysfunction and graft loss due to BK virus nephropathy, without compromising pancreas allograft outcomes.


Subject(s)
BK Virus/isolation & purification , Kidney Transplantation , Pancreas Transplantation , Polyomavirus Infections/diagnosis , Postoperative Complications/diagnosis , Tumor Virus Infections/diagnosis , Viremia/diagnosis , Adult , Aged , Drug Administration Schedule , Female , Follow-Up Studies , Graft Rejection/prevention & control , Graft Survival , Humans , Immunocompromised Host , Immunosuppressive Agents/therapeutic use , Incidence , Kaplan-Meier Estimate , Kidney Transplantation/methods , Male , Middle Aged , Pancreas Transplantation/methods , Polyomavirus Infections/epidemiology , Polyomavirus Infections/immunology , Polyomavirus Infections/therapy , Postoperative Complications/epidemiology , Postoperative Complications/immunology , Postoperative Complications/therapy , Real-Time Polymerase Chain Reaction , Retrospective Studies , Treatment Outcome , Tumor Virus Infections/epidemiology , Tumor Virus Infections/immunology , Tumor Virus Infections/therapy , Viremia/epidemiology , Viremia/immunology , Viremia/therapy
13.
Rev Med Virol ; 26(5): 330-9, 2016 09.
Article in English | MEDLINE | ID: mdl-27185010

ABSTRACT

Blood-borne viruses, such as hepatitis B virus, hepatitis C virus, human immunodeficiency virus, and the facultative blood-borne hepatitis E virus, are considered a major public health problem given that they are accountable for millions of deaths each year. Treatment options, including effective vaccine design, development of antiviral strategies and the implementation of antiretroviral therapy have improved substantially over the last couple of years and contribute to successful treatment and prevention of these infectious diseases. In this review, we summarise the current knowledge and concepts in prevention of transmission of these blood-borne viruses.


Subject(s)
Antiviral Agents/therapeutic use , HIV Infections/prevention & control , HIV Infections/therapy , Hepatitis, Viral, Human/prevention & control , Hepatitis, Viral, Human/therapy , Viral Vaccines/therapeutic use , Viremia/prevention & control , Humans , Viremia/therapy
14.
AIDS Res Ther ; 14(1): 45, 2017 Sep 12.
Article in English | MEDLINE | ID: mdl-28893280

ABSTRACT

Despite the significant success of combination anti-retroviral therapy to reduce HIV viremia and save lives, HIV-1 infection remains a lifelong infection that must be appropriately managed. Advances in the understanding of the HIV infection process and insights from vaccine development in other biomedical fields such as cancer, imaging, and genetic engineering have fueled rapid advancements in HIV cure research. In the last few years, several studies have focused on the development of "Kick and Kill" therapies to reverse HIV latency and kick start viral translational activity. This has been done with the aim that concomitant anti-retroviral treatment and the elicited immune responses will prevent de novo infections while eradicating productively infected cells. In this review, we describe our perspective on HIV cure and the new approaches we are undertaking to eradicate the established pro-viral reservoir.


Subject(s)
AIDS Vaccines/therapeutic use , HIV Infections/therapy , HIV-1/immunology , HIV-1/physiology , Virus Latency , AIDS Vaccines/administration & dosage , Anti-HIV Agents/therapeutic use , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , HIV Infections/immunology , HIV Infections/virology , Humans , Vaccination , Vaccines, Virus-Like Particle/immunology , Viremia/immunology , Viremia/therapy
15.
Br J Haematol ; 195(1): 140-143, 2021 10.
Article in English | MEDLINE | ID: mdl-34180535
16.
Proc Natl Acad Sci U S A ; 110(37): 15001-6, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-23980172

ABSTRACT

Hepatitis C virus (HCV) persistence is facilitated by exhaustion of CD8+ T cells that express the inhibitory receptor programmed cell death 1 (PD-1). Blockade of PD-1 signaling improves in vitro proliferation of HCV-specific T lymphocytes, but whether antiviral function can be restored in infected individuals is unknown. To address this question, chimpanzees with persistent HCV infection were treated with anti-PD-1 antibodies. A significant reduction in HCV viremia was observed in one of three treated animals without apparent hepatocellular injury. Viremia rebounded in the responder animal when antibody treatment was discontinued. Control of HCV replication was associated with restoration of intrahepatic CD4+ and CD8+ T-cell immunity against multiple HCV proteins. The responder animal had a history of broader T-cell immunity to multiple HCV proteins than the two chimpanzees that did not respond to PD-1 therapy. The results suggest that successful PD-1 blockade likely requires a critical threshold of preexisting virus-specific T cells in liver and warrants consideration of therapeutic vaccination strategies in combination with PD-1 blockade to broaden narrow responses. Anti-PD-1 immunotherapy may also facilitate control of other persistent viruses, notably the hepatitis B virus where options for long-term control of virus replication are limited.


Subject(s)
Hepatitis C, Chronic/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Antibodies, Monoclonal/therapeutic use , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Hepacivirus/immunology , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/virology , Immunotherapy , Pan troglodytes , Programmed Cell Death 1 Receptor/immunology , Viremia/immunology , Viremia/therapy , Virus Replication
17.
Ann Intern Med ; 163(2): 81-90, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-25961438

ABSTRACT

BACKGROUND: More than 26,000 cases of Ebola virus disease (EVD) have been reported in western Africa, with high mortality. Several patients have been medically evacuated to hospitals in the United States and Europe. Detailed clinical data are limited on the clinical course and management of patients with EVD outside western Africa. OBJECTIVE: To describe the clinical characteristics and management of a cluster of patients with EVD, including the first cases of Ebola virus (EBOV) infection acquired in the United States. DESIGN: Retrospective clinical case series. SETTING: Three U.S. hospitals in September and October 2014. PATIENTS: First imported EVD case identified in the United States and 2 secondary EVD cases acquired in the United States in critical care nurses who cared for the index case patient. MEASUREMENTS: Clinical recovery, EBOV RNA level, resolution of Ebola viremia, survival with discharge from hospital, or death. RESULTS: The index patient had high EBOV RNA levels, developed respiratory and renal failure requiring critical care support, and died. Both patients with secondary EBOV infection had nonspecific signs and symptoms and developed moderate illness; EBOV RNA levels were moderate, and both patients recovered. LIMITATION: Both surviving patients received uncontrolled treatment with multiple investigational agents, including convalescent plasma, which limits generalizability of the results. CONCLUSION: Early diagnosis, prompt initiation of supportive medical care, and moderate clinical illness likely contributed to successful outcomes in both survivors. The inability to determine the potential benefit of investigational therapies and the effect of patient-specific factors that may have contributed to less severe illness highlight the need for controlled clinical studies of these interventions, especially in the setting of a high level of supportive medical care. PRIMARY FUNDING SOURCE: None.


Subject(s)
Critical Care/methods , Hemorrhagic Fever, Ebola/diagnosis , Hemorrhagic Fever, Ebola/therapy , Adult , Early Diagnosis , Ebolavirus/genetics , Ebolavirus/metabolism , Fatal Outcome , Female , Hemorrhagic Fever, Ebola/virology , Humans , Male , RNA, Viral/blood , Renal Insufficiency/etiology , Respiratory Insufficiency/etiology , Retrospective Studies , Texas , Viremia/diagnosis , Viremia/therapy
18.
Biol Blood Marrow Transplant ; 21(9): 1663-78, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26028505

ABSTRACT

We conducted a phase I trial of allogeneic T cells sensitized in vitro against a pool of pentadecapeptides (15-mer peptides) spanning the sequence of CMVpp65 for adoptive therapy of 17 allogeneic hematopoietic cell transplant recipients with cytomegalovirus (CMV) viremia or clinical infection persisting despite prolonged treatment with antiviral drugs. All but 3 of the patients had received T cell-depleted transplants without graft-versus-host disease (GVHD) prophylaxis with immunosuppressive drugs after transplantation. The CMVpp65-specific T cells (CMVpp65CTLs) generated were oligoclonal and specific for only 1 to 3 epitopes, presented by a limited set of HLA class I or II alleles. T cell infusions were well tolerated without toxicity or GVHD. Of 17 patients treated with transplant donor (n = 16) or third-party (n = 1) CMVpp65CTLs, 15 cleared viremia, including 3 of 5 with overt disease. In responding patients, the CMVpp65CTLs infused consistently proliferated and could be detected by T cell receptor Vß usage in CMVpp65/HLA tetramer + populations for period of 120 days to up to 2 years after infusion. Thus, CMVpp65CTLs generated in response to synthetic 15-mer peptides of CMVpp65 are safe and can clear persistent CMV infections in the post-transplantation period.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus/immunology , Hematopoietic Stem Cell Transplantation , Lymphocyte Transfusion , Peptides/immunology , Phosphoproteins/immunology , T-Lymphocytes , Tissue Donors , Viral Matrix Proteins/immunology , Viremia , Aged , Allografts , Child , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/pathology , Cytomegalovirus Infections/therapy , Female , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Graft vs Host Disease/prevention & control , Hematologic Neoplasms/immunology , Hematologic Neoplasms/pathology , Hematologic Neoplasms/therapy , Humans , Male , Middle Aged , Peptides/pharmacology , Phosphoproteins/pharmacology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , T-Lymphocytes/transplantation , Viral Matrix Proteins/pharmacology , Viremia/immunology , Viremia/pathology , Viremia/therapy
19.
J Viral Hepat ; 22 Suppl 4: 21-41, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26513446

ABSTRACT

The total number, morbidity and mortality attributed to viraemic hepatitis C virus (HCV) infections change over time making it difficult to compare reported estimates from different years. Models were developed for 15 countries to quantify and characterize the viraemic population and forecast the changes in the infected population and the corresponding disease burden from 2014 to 2030. With the exception of Iceland, Iran, Latvia and Pakistan, the total number of viraemic HCV infections is expected to decline from 2014 to 2030, but the associated morbidity and mortality are expected to increase in all countries except for Japan and South Korea. In the latter two countries, mortality due to an ageing population will drive down prevalence, morbidity and mortality. On the other hand, both countries have already experienced a rapid increase in HCV-related mortality and morbidity. HCV-related morbidity and mortality are projected to increase between 2014 and 2030 in all other countries as result of an ageing HCV-infected population. Thus, although the total number of HCV countries is expected to decline in most countries studied, the associated disease burden is expected to increase. The current treatment paradigm is inadequate if large reductions in HCV-related morbidity and mortality are to be achieved.


Subject(s)
Hepacivirus/isolation & purification , Hepatitis C, Chronic/epidemiology , Hepatitis C, Chronic/virology , Models, Statistical , Viremia/epidemiology , Viremia/virology , Adolescent , Adult , Aged , Aged, 80 and over , Cost of Illness , Female , Global Health , Hepatitis C, Chronic/mortality , Hepatitis C, Chronic/therapy , Humans , Incidence , Male , Middle Aged , Prevalence , Survival Analysis , Viremia/mortality , Viremia/therapy , Young Adult
20.
J Immunol ; 190(4): 1583-90, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23315078

ABSTRACT

Therapeutic administration of IFN-α in clinical trials significantly reduced HIV-1 plasma viral load and human T-lymphotropic virus type I proviral load in infected patients. The mechanism may involve the concerted action of multiple antiretroviral effectors collectively known as "restriction factors," which could vary in relative importance according to the magnitude of transcriptional induction. However, direct genetic approaches to identify the relevant IFN-α restriction factors will not be feasible in humans in vivo. Meanwhile, mice encode an analogous set of restriction factor genes and could be used to obtain insights on how IFN-α could inhibit retroviruses in vivo. As expected, IFN-α treatment of mice significantly upregulated the transcription of multiple restriction factors including Tetherin/BST2, SAMHD1, Viperin, ISG15, OAS1, and IFITM3. However, a dominant antiretroviral factor, Apobec3, was only minimally induced. To determine whether Apobec3 was necessary for direct IFN-α antiretroviral action in vivo, wild-type and Apobec3-deficient mice were infected with Friend retrovirus, then treated with IFN-α. Treatment of infected wild-type mice with IFN-α significantly reduced acute plasma viral load 28-fold, splenic proviral load 5-fold, bone marrow proviral load 14-fold, and infected bone marrow cells 7-fold, but no inhibition was observed in Apobec3-deficient mice. These findings reveal that IFN-α inhibits acute Friend retrovirus infection primarily through the antiviral effector Apobec3 in vivo, demonstrate that transcriptional induction levels did not predict the mechanism of IFN-α-mediated control, and highlight the potential of the human APOBEC3 proteins as therapeutic targets against pathogenic retrovirus infections.


Subject(s)
Antiviral Agents/administration & dosage , Cytidine Deaminase/physiology , Friend murine leukemia virus/immunology , Interferon-alpha/administration & dosage , Retroviridae Infections/therapy , Retroviridae Infections/virology , Virus Replication/immunology , Acute Disease , Animals , Antiviral Agents/therapeutic use , Cells, Cultured , Cytidine Deaminase/deficiency , Cytidine Deaminase/genetics , Friend murine leukemia virus/pathogenicity , Humans , Interferon-alpha/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Retroviridae Infections/immunology , Viremia/immunology , Viremia/therapy , Viremia/virology
SELECTION OF CITATIONS
SEARCH DETAIL