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

Publication year range
1.
Cell ; 186(6): 1115-1126.e8, 2023 03 16.
Article in English | MEDLINE | ID: mdl-36931242

ABSTRACT

Previously, two men were cured of HIV-1 through CCR5Δ32 homozygous (CCR5Δ32/Δ32) allogeneic adult stem cell transplant. We report the first remission and possible HIV-1 cure in a mixed-race woman who received a CCR5Δ32/Δ32 haplo-cord transplant (cord blood cells combined with haploidentical stem cells from an adult) to treat acute myeloid leukemia (AML). Peripheral blood chimerism was 100% CCR5Δ32/Δ32 cord blood by week 14 post-transplant and persisted through 4.8 years of follow-up. Immune reconstitution was associated with (1) loss of detectable replication-competent HIV-1 reservoirs, (2) loss of HIV-1-specific immune responses, (3) in vitro resistance to X4 and R5 laboratory variants, including pre-transplant autologous latent reservoir isolates, and (4) 18 months of HIV-1 control with aviremia, off antiretroviral therapy, starting at 37 months post-transplant. CCR5Δ32/Δ32 haplo-cord transplant achieved remission and a possible HIV-1 cure for a person of diverse ancestry, living with HIV-1, who required a stem cell transplant for acute leukemia.


Subject(s)
Cord Blood Stem Cell Transplantation , HIV Infections , HIV-1 , Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute , Male , Adult , Female , Humans , Fetal Blood , Leukemia, Myeloid, Acute/therapy
2.
Clin Infect Dis ; 76(3): e744-e747, 2023 02 08.
Article in English | MEDLINE | ID: mdl-36031390

ABSTRACT

We followed 54 infants with in utero HIV after initiating very early antiretroviral treatment. At weeks 24 and 48, ≥80% had CD4 ≥1500 cells/mm3 and CD4% ≥25%. Routine Pneumocystis jirovecii pneumonia prophylaxis in the first year of life may not be necessary for all very early treated infants. CLINICAL TRIALS REGISTRATION: NCT02140255.


Subject(s)
Anti-HIV Agents , HIV Infections , Pneumocystis carinii , Pneumonia, Pneumocystis , Humans , Infant , HIV Infections/drug therapy , Pneumonia, Pneumocystis/drug therapy , Antiretroviral Therapy, Highly Active , Anti-HIV Agents/therapeutic use , Anti-Retroviral Agents/therapeutic use , CD4 Lymphocyte Count
3.
Curr Opin Pediatr ; 31(1): 144-147, 2019 02.
Article in English | MEDLINE | ID: mdl-30531403

ABSTRACT

PURPOSE OF REVIEW: The purpose of this review is to describe some current challenges facing the field of pediatric infectious diseases and discuss strategies for enhancing recognition of the value of infectious disease services and for recruiting new talent to the field. RECENT FINDINGS: Pediatric infectious disease programs are currently filling approximately half of their fellowship positions, and salaries are among the lowest in medical subspecialties. Research-intensive careers in pediatric infectious diseases are threatened by low the National Institutes of Health paylines for career development awards. Despite this, there are new opportunities in pediatric infectious diseases in growing areas, such as transplant infectious diseases and antimicrobial stewardship. SUMMARY: Pediatric infectious disease practitioners are concerned that infectious disease services are often undervalued by the healthcare system. Some of the contributions made by this cognitive specialty to overall patient outcomes are difficult to quantify and are not fully reimbursed. Strategies to enhance value and program support are needed, including sharing individual success stories and collecting data from programs nationally to generate some standards for support of pediatric infectious disease programs in areas, such as antimicrobial stewardship, transplant infectious diseases, and infection prevention. Recruitment of top talent to the field can be enhanced by a number of initiatives that can be implemented at the local level with encouragement and leadership from the pediatric infectious diseases society.


Subject(s)
Communicable Diseases , National Institutes of Health (U.S.) , Pediatrics , Career Mobility , Child , Delivery of Health Care , Forecasting , Humans , National Institutes of Health (U.S.)/economics , Pediatrics/education , Pediatrics/trends , United States
4.
Clin Infect Dis ; 66(11): 1770-1777, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29272365

ABSTRACT

Background: The presence of antiretroviral drug-associated resistance mutations (DRMs) may be particularly problematic in human immunodeficiency virus (HIV)-infected pregnant women as it can lead to mother-to-child transmission (MTCT) of resistant HIV strains. This study evaluated the prevalence and the effect of antiretroviral DRMs in previously untreated mother-infant pairs. Methods: A case-control design of 1:4 (1 transmitter to 4 nontransmitters) was utilized to evaluate DRMs as a predictor of HIV MTCT in specimens obtained from mother-infant pairs. ViroSeq HIV-1 genotyping was performed on mother-infant specimens to assess for clinically relevant DRMs. Results: One hundred forty infants acquired HIV infection; of these, 123 mother-infant pairs (88%) had specimens successfully amplified using ViroSeq and assessed for drug resistance genotyping. Additionally, 483 of 560 (86%) women who did not transmit HIV to infants also had samples evaluated for DRMs. Sixty-three of 606 (10%) women had clinically relevant DRMs; 12 (2%) had DRMs against >1 drug class. Among 123 HIV-infected infants, 13 (11%) had clinically relevant DRMs, with 3 (2%) harboring DRMs against >1 drug class. In univariate and multivariate analyses, DRMs in mothers were not associated with increased HIV MTCT (adjusted odds ratio, 0.8 [95% confidence interval, .4-1.5]). Presence of DRMs in transmitting mothers was strongly associated with DRM presence in their infants (P < .001). Conclusions: Preexisting DRMs were common in untreated HIV-infected pregnant women, but did not increase the risk of HIV MTCT. However, if women with DRMs are not virologically suppressed, they may transmit resistant mutations, thus complicating infant management.


Subject(s)
Anti-HIV Agents/pharmacology , Drug Resistance, Multiple, Viral , HIV Infections/virology , HIV-1/drug effects , Infectious Disease Transmission, Vertical , Adolescent , Adult , Anti-HIV Agents/classification , Female , HIV Infections/transmission , HIV-1/genetics , Humans , Infant , Mutation , Pregnancy , Young Adult
5.
Clin Infect Dis ; 65(3): 405-413, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28369278

ABSTRACT

BACKGROUND: Cytomegalovirus (CMV) urinary shedding in pregnant women infected with human immunodeficiency virus (HIV) was evaluated to determine whether it poses an increased risk for congenital CMV infection (cCMV). METHODS: A subset of mother-infant pairs enrolled in the perinatal NICHD HPTN 040 study (distinguished by no antiretroviral use before labor) was evaluated. Maternal and infant urines were tested by qualitative real-time polymerase chain reaction (RT-PCR) for CMV DNA with quantitative RT-PCR performed on positive specimens. RESULTS: Urine specimens were available for 260 women with 85.4% from the Americas and 14.6% from South Africa. Twenty-four women (9.2%) had detectable CMV viruria by qualitative PCR. Maternal CMV viruria was not associated with mean CD4 cell counts or HIV viral load but was associated with younger maternal age (P = .02). Overall, 10 of 260 infants (3.8%) had cCMV. Women with detectable peripartum CMV viruria were more likely to have infants with cCMV than those without: 20.8% (5/24) versus 2.1% (5/236), (P = .0001). Women with CMV viruria had significantly higher rates of HIV perinatal transmission (29.2% vs. 8.1%, P = .002). They were 5 times (adjusted odds ratio [aOR] = 5.6, 95% confidence interval [CI] 1.9-16.8) and nearly 30 times (aOR, 29.7; 95% CI, 5.4-164.2) more likely to transmit HIV and CMV to their infants, respectively. Maternal gonorrhea (aOR, 19.5; 95% CI, 2.5-151.3) and higher maternal HIV log10 viral load (OR, 2.8; 95% CI, 1.3-6.3) were also significant risk factors for cCMV. CONCLUSION: In this cohort of HIV-infected pregnant women not on antiretrovirals, urinary CMV shedding was a significant risk factor for CMV and HIV transmission to infants. CLINICAL TRIALS REGISTRATION NUMBER: NCT00099359.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , DNA, Viral/urine , HIV Infections/complications , Pregnancy Complications, Infectious , Adolescent , Adult , Cytomegalovirus/genetics , Cytomegalovirus/isolation & purification , Cytomegalovirus Infections/complications , Cytomegalovirus Infections/congenital , Cytomegalovirus Infections/urine , Cytomegalovirus Infections/virology , DNA, Viral/genetics , Female , Humans , Infant, Newborn , Predictive Value of Tests , Pregnancy , Pregnancy Complications, Infectious/urine , Pregnancy Complications, Infectious/virology , Prevalence , Real-Time Polymerase Chain Reaction , Retrospective Studies , Risk Factors , Viral Load , Virus Shedding , Young Adult
6.
N Engl J Med ; 366(25): 2368-79, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22716975

ABSTRACT

BACKGROUND: The safety and efficacy of adding antiretroviral drugs to standard zidovudine prophylaxis in infants of mothers with human immunodeficiency virus (HIV) infection who did not receive antenatal antiretroviral therapy (ART) because of late identification are unclear. We evaluated three ART regimens in such infants. METHODS: Within 48 hours after their birth, we randomly assigned formula-fed infants born to women with a peripartum diagnosis of HIV type 1 (HIV-1) infection to one of three regimens: zidovudine for 6 weeks (zidovudine-alone group), zidovudine for 6 weeks plus three doses of nevirapine during the first 8 days of life (two-drug group), or zidovudine for 6 weeks plus nelfinavir and lamivudine for 2 weeks (three-drug group). The primary outcome was HIV-1 infection at 3 months in infants uninfected at birth. RESULTS: A total of 1684 infants were enrolled in the Americas and South Africa (566 in the zidovudine-alone group, 562 in the two-drug group, and 556 in the three-drug group). The overall rate of in utero transmission of HIV-1 on the basis of Kaplan-Meier estimates was 5.7% (93 infants), with no significant differences among the groups. Intrapartum transmission occurred in 24 infants in the zidovudine-alone group (4.8%; 95% confidence interval [CI], 3.2 to 7.1), as compared with 11 infants in the two-drug group (2.2%; 95% CI, 1.2 to 3.9; P=0.046) and 12 in the three-drug group (2.4%; 95% CI, 1.4 to 4.3; P=0.046). The overall transmission rate was 8.5% (140 infants), with an increased rate in the zidovudine-alone group (P=0.03 for the comparisons with the two- and three-drug groups). On multivariate analysis, zidovudine monotherapy, a higher maternal viral load, and maternal use of illegal substances were significantly associated with transmission. The rate of neutropenia was significantly increased in the three-drug group (P<0.001 for both comparisons with the other groups). CONCLUSIONS: In neonates whose mothers did not receive ART during pregnancy, prophylaxis with a two- or three-drug ART regimen is superior to zidovudine alone for the prevention of intrapartum HIV transmission; the two-drug regimen has less toxicity than the three-drug regimen. (Funded by the Eunice Kennedy Shriver National Institute of Child Health and Human Development [NICHD] and others; ClinicalTrials.gov number, NCT00099359.).


Subject(s)
Anti-Retroviral Agents/therapeutic use , HIV Infections/prevention & control , HIV-1 , Infectious Disease Transmission, Vertical/prevention & control , Lamivudine/therapeutic use , Nelfinavir/therapeutic use , Nevirapine/therapeutic use , Zidovudine/therapeutic use , Anti-Retroviral Agents/adverse effects , Drug Resistance, Viral , Drug Therapy, Combination/adverse effects , Female , HIV Infections/mortality , HIV Infections/transmission , Humans , Infant Formula , Infant, Newborn , Kaplan-Meier Estimate , Lamivudine/adverse effects , Male , Nelfinavir/adverse effects , Nevirapine/adverse effects , Postpartum Period , Pregnancy , Pregnancy Complications, Infectious , Zidovudine/adverse effects
7.
J Adolesc Health ; 74(3): 573-581, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38043041

ABSTRACT

PURPOSE: Gay, bisexual, and other cisgender men who have sex with men, and racial minority youth are at elevated risk of acquiring HIV infection. The Adolescent Trials Network 147 recruited youth with acute/recent HIV-infection for early antiretroviral treatment. The cohort make-up is described here. METHODS: Treatment-naïve, recently identified HIV + youth, aged 12-24 years, from Los Angeles and New Orleans were recruited from community centers, clinics, social media, and a high-risk seronegative cohort (n = 1,727, the Adolescent Trials Network 149) using point-of-care assays. Acute HIV infection was determined by Fiebig staging. HIV RNA viral load (VL) and CD4 cell counts, along with demographic and behavioral data were assessed at enrollment. RESULTS: Between July 2017 and July 2021, 103 newly diagnosed youth were enrolled, initiating antiretroviral treatment within a week. Mean age was 20.8 years (standard deviation: 2.4); 90.3% identified as cis male, 83.5% were single or in casual relationships, 71.8% were gay, bisexual, and other cisgender men who have sex with men; 60.2% were Black. One-fourth (24.3%) reported homelessness ever; 10.7% within last 4 months. At enrollment, median plasma VL was 37,313 HIV RNA copies/ml (interquartile range: 5,849-126,162) and median CD4 count 445.5 cells/mm3 (interquartile range: 357-613). 40% of youth reported acute retroviral symptoms before or at enrollment. Acutely infected, seroconverting youth had the highest VL. Sexually transmitted coinfections were present at enrollment in 56% of the cohort, with syphilis being most frequent (39%). DISCUSSION: Early identification and treatment of HIV can increase positive HIV outcomes. A high sexually transmitted infection burden was present in recently HIV-infected youth. Acute retroviral symptoms were not reported by most participants, demonstrating that broad universal HIV screening is needed for identification of recent infection in youth.


Subject(s)
HIV Infections , Sexual and Gender Minorities , Male , Adolescent , Humans , Young Adult , Adult , HIV Infections/diagnosis , HIV Infections/drug therapy , HIV Infections/epidemiology , Homosexuality, Male , CD4 Lymphocyte Count , RNA , Demography , Viral Load
8.
Lancet HIV ; 11(1): e20-e30, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38061376

ABSTRACT

BACKGROUND: Infants born with HIV-1 require lifelong antiretroviral therapy (ART). We aimed to assess whether very early ART in neonates might restrict HIV-1 reservoirs, an important step towards ART-free remission. METHODS: IMPAACT P1115 is an ongoing, phase 1/2, proof-of-concept study in which infants were enrolled at 30 research clinics in 11 countries (Brazil, Haiti, Kenya, Malawi, South Africa, Tanzania, Thailand, Uganda, the USA, Zambia, and Zimbabwe) into two cohorts. Infants at least 34 weeks' gestational age at high risk for in-utero HIV-1 with either untreated maternal HIV-1 (cohort 1) or who were receiving pre-emptive triple antiretroviral prophylaxis outside of the study (maternal ART permissible; cohort 2) were included. All infants initiated treatment within 48 h of life. Cohort 1 initiated three-drug nevirapine-based ART, and cohort 2 initiated three-drug nevirapine-based prophylaxis then three-drug nevirapine-based ART following HIV diagnosis by age 10 days. We added twice-daily coformulated oral ritonavir 75 mg/m2 and lopinavir 300 mg/m2 from 14 days of life and 42 weeks postmenstrual age. We discontinued nevirapine 12 weeks after two consecutive plasma HIV-1 RNA levels below limit of detection. We tracked virological suppression, safety outcomes, and meeting a predetermined biomarker profile at age 2 years (undetectable RNA since week 48, HIV-1 antibody-negative, HIV-1 DNA not detected, and normal CD4 count and CD4 percentage) to assess qualification for analytical treatment interruption. This study is registered with ClinicalTrials.gov, NCT02140255. FINDINGS: Between Jan 23, 2015, and Dec 14, 2017, 440 infants were included in cohort 1 and 20 were included in cohort 2. 54 of these infants (34 from cohort 1 and 20 from cohort 2) had confirmed in-utero HIV-1 and were enrolled to receive study ART. 33 (61%) of 54 infants were female and 21 (39%) were male. The estimated probability of maintaining undetectable plasma RNA through to 2 years was 33% (95% CI 17-49) in cohort 1 and 57% (28-78) in cohort 2. Among infants maintaining protocol-defined virological control criteria through to study week 108, seven of 11 (64%, 95% CI 31-89) in cohort 1 and five of seven (71%, 29-96) in cohort 2 had no detected HIV-1 DNA. Ten of 12 (83%, 52-100) in cohort 1 and all seven (100%, 59-100) in cohort 2 tested HIV-1 antibody-negative at week 108. Among 54 infants initiated on very early ART, ten (19%; six in cohort 1 and four in cohort 2) met all criteria for possible analytical treatment interruption. Reversible grade 3 or 4 adverse events occurred in 15 (44%) of 34 infants in cohort 1 and seven (35%) of 20 infants in cohort 2. INTERPRETATION: Very early ART for in-utero HIV-1 can achieve sustained virological suppression in association with biomarkers indicating restricted HIV-1 reservoirs by age 2 years, which might enable potential ART-free remission. FUNDING: National Institute of Allergy and Infectious Diseases, the Eunice Kennedy Shriver National Institute of Child Health and Human Development, and the National Institute of Mental Health.


Subject(s)
Anti-HIV Agents , HIV Infections , HIV Seropositivity , HIV-1 , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Anti-Retroviral Agents/adverse effects , DNA/therapeutic use , HIV Infections/drug therapy , HIV Infections/prevention & control , HIV Seropositivity/drug therapy , HIV-1/genetics , Nevirapine/therapeutic use , RNA/therapeutic use , Proof of Concept Study
9.
Biol Blood Marrow Transplant ; 19(3): 393-7, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23089564

ABSTRACT

Hematopoietic cell transplantation (HCT) using CCR5-Δ32/Δ32 stem cells from an adult donor has resulted in the only known cure of human immunodeficiency virus (HIV) infection. However, it is not feasible to repeat this procedure except rarely because of the low incidence of the CCR5-Δ32 allele, the availability of only a small number of potential donors for most patients, and the need for a very close human leukocyte antigen (HLA) match between adult donors and recipients. In contrast, cord blood (CB) transplantations require significantly less stringent HLA matching. Therefore, our hypothesis is that cure of HIV infections by HCT can be accomplished much more readily using umbilical CB stem cells obtained from a modestly sized inventory of cryopreserved CCR5-Δ32/Δ32 CB units. To test this hypothesis, we developed a screening program for CB units and are developing an inventory of CCR5-Δ32/Δ32 cryopreserved units available for HCT. Three hundred such units are projected to provide for white pediatric patients a 73.6% probability of finding an adequately HLA matched unit with a cell dose of ≥2.5 × 10(7) total nucleated cells (TNCs)/kg and a 27.9% probability for white adults. With a cell dose of ≥1 × 10(7) TNCs/kg, the corresponding projected probabilities are 85.6% and 82.1%. The projected probabilities are lower for ethnic minorities. Impetus for using CB HCT was provided by a transplantation of an adult with acute myelogenous leukemia who was not HIV infected. The HCT was performed with a CCR5-Δ32/Δ32 CB unit, and posttransplantation in vitro studies indicated that the patient's peripheral blood mononuclear cells were resistant to HIV infection.


Subject(s)
Cord Blood Stem Cell Transplantation , Leukemia, Myeloid, Acute/therapy , Leukocytes, Mononuclear/immunology , Receptors, CCR5/genetics , Sequence Deletion , Adult , Blood Banks , Cells, Cultured , Child , Cryopreservation , HIV Infections/immunology , HIV Infections/pathology , HIV Infections/therapy , HIV Infections/virology , HLA Antigens/genetics , HLA Antigens/immunology , Histocompatibility Testing , Humans , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/virology , Probability , Receptors, CCR5/immunology , Transplantation Chimera/immunology , Unrelated Donors , White People
12.
Biologicals ; 40(1): 15-20, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22192456

ABSTRACT

Natural membrane-bound HIV-1 envelope proteins (mHIVenv) could be used to produce an effective subunit vaccine against HIV infection, akin to effective vaccination against HBV infection using the hepatitis B surface antigen. The quaternary structure of mHIVenv is postulated to elicit broadly neutralizing antibodies protective against HIV-1 transmission. The founder virus transmitted to infected individuals during acute HIV-1 infection is genetically homogeneous and restricted to CCR5-tropic phenotype. Therefore, isolates of plasma-derived HIV-1 (PHIV) from infected blood donors while negative for antibodies to HIV proteins were selected for expansion in primary lymphocytes as an optimized cell substrate (OCS). Virions in the culture supernatants were purified by removing contaminating microvesicles using immunomagnetic beads coated with anti-CD45. Membrane cholesterol was extracted from purified virions with beta-cyclodextrin to permeabilize them and expel p24, RT and viral RNA, and permit protease-free Benzonase to hydrolyze the residual viral/host DNA/RNA without loss of gp120. The resultant mHIVenv, containing gp120 bound to native gp41 in immunoreactive form, was free from infectivity in vitro in co-cultures with OCS and in vivo after inoculating SCID-hu Thy/Liv mice. These data should help development of mHIVenv as a virally safe immunogen and enable preparation of polyclonal hyper-immune globulins for immunoprophylaxis against HIV-1 infection.


Subject(s)
HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp41/immunology , HIV Infections/immunology , HIV-1/immunology , Virion/immunology , Animals , Antibodies, Viral/immunology , HIV Envelope Protein gp120/blood , HIV Envelope Protein gp120/pharmacology , HIV Envelope Protein gp41/blood , HIV Envelope Protein gp41/pharmacology , HIV Infections/blood , Humans , Mice , Mice, SCID , Virion/metabolism
13.
Viruses ; 14(11)2022 10 26.
Article in English | MEDLINE | ID: mdl-36366448

ABSTRACT

The extent to which perinatally HIV-infected children, following cART initiation, develop a low proviral reservoir burden over time, as measured by HIV DNA droplet-digital polymerase chain reaction (ddPCR) and the effect on HIV antibody is not well characterized. We measured proviral HIV DNA and plasma RNA virus load (VL) in 37 perinatally HIV-infected children at 6 months of age who initiated stable cART. At 6-11 years of age, HIV proviral DNA, HIV VL (RNA), and HIV antibody by Western Blot (WB) were assessed. CART was initiated before 6 months of age in 13 children and after 6 months in 24. At school age, the HIV DNA levels did not differ by the timing of cART, and the HIV DNA levels were lower in children with negative/indeterminate WB (p = 0.0256). Children with undetectable HIV RNA VL > 50% of the time since cART initiation had lower median DNA VL than children with undetectable VL < 50% of the time (p = 0.07). Long-term viral suppression in perinatally HIV-infected children is associated with a decrease in HIV antibodies and reduced HIV reservoirs.


Subject(s)
HIV Infections , HIV-1 , Child , Humans , Infant , Proviruses/genetics , HIV Antibodies , HIV-1/genetics , Viral Load , HIV Infections/drug therapy , DNA, Viral/analysis , RNA
15.
Cell Rep ; 34(1): 108573, 2021 01 05.
Article in English | MEDLINE | ID: mdl-33406429

ABSTRACT

Whereas the human fetal immune system is poised to generate immune tolerance and suppress inflammation in utero, an adult-like immune system emerges to orchestrate anti-pathogen immune responses in post-natal life. It has been posited that cells of the adult immune system arise as a discrete ontological "layer" of hematopoietic stem-progenitor cells (HSPCs) and their progeny; evidence supporting this model in humans has, however, been inconclusive. Here, we combine bulk and single-cell transcriptional profiling of lymphoid cells, myeloid cells, and HSPCs from fetal, perinatal, and adult developmental stages to demonstrate that the fetal-to-adult transition occurs progressively along a continuum of maturity-with a substantial degree of inter-individual variation at the time of birth-rather than via a transition between discrete waves. These findings have important implications for the design of strategies for prophylaxis against infection in the newborn and for the use of umbilical cord blood (UCB) in the setting of transplantation.


Subject(s)
Fetus/metabolism , Hematopoietic Stem Cells/metabolism , Lymphocytes/metabolism , Myeloid Cells/metabolism , Single-Cell Analysis , T-Lymphocytes/metabolism , Transcriptome , Bone Marrow/metabolism , Cell Culture Techniques , Female , Fetal Blood , Humans , Immunity , Pregnancy , Sequence Analysis, RNA
16.
Lancet HIV ; 8(3): e149-e157, 2021 03.
Article in English | MEDLINE | ID: mdl-33242457

ABSTRACT

BACKGROUND: With increasing intention to treat HIV as early as possible, evidence to confirm the safety and therapeutic drug concentrations of a nevirapine-based antiretroviral regimen in the early neonatal period is needed. This study aims to establish dosing of nevirapine for very early treatment of HIV-exposed neonates at high risk of HIV acquisition. METHODS: IMPAACT P1115 is a multinational phase 1/2 proof-of-concept study in which presumptive treatment for in-utero HIV infection is initiated within 48 h of birth in HIV-exposed neonates at high risk of HIV acquisition. Participants were neonates who were at least 34 weeks gestational age at birth and enrolled within 48 h of birth, born to women with presumed or confirmed HIV infection who had not received antiretrovirals during this pregnancy. The regimen consisted of two nucleoside reverse transcriptase inhibitors plus nevirapine dosed at 6 mg/kg twice daily for term neonates (≥37 weeks gestational age) or 4 mg/kg twice daily for 1 week and 6 mg/kg twice daily thereafter for preterm neonates (34 to <37 weeks gestational age). Here, we report the secondary outcomes of the study: nevirapine exposures in study weeks 1 and 2 and treatment-associated grade 3 or 4 adverse events at least possibly related to study treatment up to study week 4. A population pharmacokinetic model to assess nevirapine exposure was developed from dried blood spot and plasma nevirapine concentrations at study weeks 1 and 2. Nevirapine exposure was assessed in all patients with available blood samples and safety was assessed in all participants. This trial is registered at ClinicalTrials.gov (NCT02140255). FINDINGS: Between Jan 23, 2015, and Sept 4, 2017, 438 neonates were enrolled and included in analyses; 36 had in-utero HIV infection and 389 (89%) were born at term. Neonates without confirmed in-utero HIV infection received nevirapine for a median of 13 days (IQR 7-14). Measured dried blood spot nevirapine concentrations were higher than the minimum HIV treatment target (3 µg/mL) in 314 (90%, 95% CI 86-93) of 349 neonates at week 1 and 174 (87%, 81-91) of 201 at week 2. In Monte-Carlo simulations, week 1 nevirapine concentrations exceeded 3 µg/mL in 80% of term neonates and 82% of preterm neonates. DAIDS grade 3 or 4 adverse events at least possibly related to antiretrovirals occurred in 30 (7%, 95% CI 5-10) of 438 infants but did not lead to nevirapine cessation in any neonates; neutropenia (25 [6%] neonates) and anaemia (six [1%]) were most common. INTERPRETATION: Nevirapine at the dose studied was confirmed to be safe and provides therapeutic exposure concentrations. These data support nevirapine as a component of presumptive HIV treatment in high-risk neonates. FUNDING: National Institute of Allergy and Infectious Diseases, Eunice Kennedy Shriver National Institute of Child Health and Human Development, and the National Institute of Mental Health.


Subject(s)
Anti-HIV Agents/adverse effects , Anti-HIV Agents/pharmacokinetics , HIV Infections/drug therapy , Nevirapine/adverse effects , Nevirapine/pharmacokinetics , Reverse Transcriptase Inhibitors/therapeutic use , Anti-HIV Agents/therapeutic use , Female , Gestational Age , HIV Infections/transmission , Humans , Infant , Infant, Newborn , Infectious Disease Transmission, Vertical , Male , Nevirapine/therapeutic use , Pregnancy , Pregnancy Complications, Infectious/drug therapy , Pregnancy Complications, Infectious/virology , Proof of Concept Study , Prospective Studies , Reverse Transcriptase Inhibitors/adverse effects , Reverse Transcriptase Inhibitors/pharmacokinetics
17.
Cell Rep Med ; 2(11): 100453, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34723226

ABSTRACT

While pregnancy increases the risk for severe COVID-19, the clinical and immunological implications of COVID-19 on maternal-fetal health remain unknown. Here, we present the clinical and immunological landscapes of 93 COVID-19 mothers and 45 of their SARS-CoV-2-exposed infants through comprehensive serum proteomics profiling for >1,400 cytokines of their peripheral and cord blood specimens. Prenatal SARS-CoV-2 infection triggers NF-κB-dependent proinflammatory immune activation. Pregnant women with severe COVID-19 show increased inflammation and unique IFN-λ antiviral signaling, with elevated levels of IFNL1 and IFNLR1. Furthermore, SARS-CoV-2 infection re-shapes maternal immunity at delivery, altering the expression of pregnancy complication-associated cytokines, inducing MMP7, MDK, and ESM1 and reducing BGN and CD209. Finally, COVID-19-exposed infants exhibit induction of T cell-associated cytokines (IL33, NFATC3, and CCL21), while some undergo IL-1ß/IL-18/CASP1 axis-driven neonatal respiratory distress despite birth at term. Our findings demonstrate COVID-19-induced immune rewiring in both mothers and neonates, warranting long-term clinical follow-up to mitigate potential health risks.


Subject(s)
COVID-19/immunology , Cytokines/blood , Inflammation , Proteomics , Adolescent , Adult , COVID-19/blood , COVID-19/metabolism , Female , Humans , Infant, Newborn , Mothers , Pregnancy , Serum/metabolism , Young Adult
18.
J Virol ; 83(21): 11016-26, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19692480

ABSTRACT

The affinity of human immunodeficiency virus (HIV) envelope for CD4 and CCR5 appears to be associated with aspects of R5 virus (virus using the CCR5 coreceptor) pathogenicity. However, entry efficiency results from complex interactions between the viral envelope glycoprotein and both CD4 and CCR5, which limits attempts to correlate viral pathogenicity with surrogate measures of envelope CD4 and CCR5 affinities. Here, we present a system that provides a quantitative and comprehensive characterization of viral entry efficiency as a direct interdependent function of both CD4 and CCR5 levels. This receptor affinity profiling system also revealed heretofore unappreciated complexities underlying CD4/CCR5 usage. We first developed a dually inducible cell line in which CD4 and CCR5 could be simultaneously and independently regulated within a physiologic range of surface expression. Infection by multiple HIV type 1 (HIV-1) and simian immunodeficiency virus isolates could be examined simultaneously for up to 48 different combinations of CD4/CCR5 expression levels, resulting in a distinct usage pattern for each virus. Thus, each virus generated a unique three-dimensional surface plot in which viral infectivity varied as a function of both CD4 and CCR5 expression. From this functional form, we obtained a sensitivity vector along with corresponding metrics that quantified an isolate's overall efficiency of CD4/CCR5 usage. When applied to viral isolates with well-characterized sensitivities to entry/fusion inhibitors, the vector metrics were able to encapsulate their known biological phenotypes. The application of the vector metrics also indicated that envelopes derived from elite suppressors had overall-reduced entry efficiencies compared to those of envelopes derived from chronically infected viremic progressors. Our affinity-profiling system may help to refine studies of R5 virus tropism and pathogenesis.


Subject(s)
CD4 Antigens/physiology , HIV-1/physiology , Receptors, CCR5/physiology , Simian Immunodeficiency Virus/physiology , Virus Internalization , Affinity Labels , Animals , CD4 Antigens/genetics , Cell Line , Ecdysterone/analogs & derivatives , Ecdysterone/metabolism , Humans , Mathematical Concepts , Minocycline/metabolism , Receptors, CCR5/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
19.
J Acquir Immune Defic Syndr ; 84(1): 70-77, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31913995

ABSTRACT

BACKGROUND: Adequate pharmacokinetic and safety data in neonates are lacking for most antiretroviral agents. Raltegravir is a selective HIV-1 integrase strand transfer inhibitor available in a granule formulation suitable for use in neonates and young infants as prophylaxis or treatment of HIV infection. METHODS: IMPAACT P1110 is a phase 1, multicenter, noncomparative dose-finding study of raltegravir in infants exposed to HIV-1 infection. A 2-cohort adaptive design was utilized where pharmacokinetic data from infants in cohort 1 who received 2 single doses of raltegravir 3 mg/kg were included in population modeling and simulations to guide selection of a daily dose for infants in cohort 2. RESULTS: A total of 52 infants enrolled in IMPAACT 1110: cohort 1 (N = 16) and cohort 2 (N = 36). Using simulations based on population PK modeling incorporating cohort 1 data, the following daily dosing regimen was selected for study: 1.5 mg/kg daily from birth through day 7; 3 mg/kg twice daily from days 8-28 of life; and 6 mg/kg twice daily after 4 weeks of age through 6 weeks of age. The geometric mean protocol exposure targets for AUC, Ctrough, and Cmax were met or slightly exceeded in all infants. The chosen neonatal raltegravir dosing regimen was safe and well tolerated in full-term neonates during treatment over the first 6 weeks of life and follow-up to age 24 weeks. CONCLUSIONS: Raltegravir can be safely administered to full-term infants using the daily dosing regimen studied. This regimen is not recommended for use in premature infants in a new version of P1110.


Subject(s)
HIV Infections/drug therapy , HIV Integrase Inhibitors/administration & dosage , Infant, Newborn, Diseases/drug therapy , Raltegravir Potassium/administration & dosage , Area Under Curve , Cohort Studies , Dose-Response Relationship, Drug , Female , HIV Infections/prevention & control , HIV Integrase Inhibitors/adverse effects , HIV Integrase Inhibitors/pharmacokinetics , HIV-1 , Half-Life , Humans , Infant, Newborn , Infant, Newborn, Diseases/prevention & control , Male , Raltegravir Potassium/adverse effects , Raltegravir Potassium/pharmacokinetics
20.
AIDS Res Hum Retroviruses ; 36(2): 122-130, 2020 02.
Article in English | MEDLINE | ID: mdl-31571497

ABSTRACT

HIV-1-induced cytopathicity of thymocytes is a major cause of reduced peripheral T cells and rapid disease progression observed in HIV-1-infected infants. Understanding the virulence factors responsible for thymocyte depletion has paramount importance in addressing the pathogenesis of disease progression in children. In this study, thymocyte depletion was analyzed following infection with two primary CXCR4-tropic HIV-1 pediatric isolates (PI), PI-2 and PI-2.1, which were serially derived from an in utero-infected infant. Although highly similar to each other, PI-2 showed markedly decreased thymocyte depletion in vitro compared with PI-2.1. Further analysis showed a novel deletion in the Nef protein (NefΔK7S) of PI-2, which was absent in PI-2.1. This deletion inhibited Nef-mediated major histocompatibility complex class I (MHC-I) downregulation in infected thymocytes in vitro and in vivo; in contrast, the mutated Nef continued to downregulate CD4 surface expression in vitro. These results suggest that HIV-1 Nef contributes to thymic damage in infants through selective functions.


Subject(s)
HIV Infections/genetics , Histocompatibility Antigens Class I/genetics , Thymocytes/virology , nef Gene Products, Human Immunodeficiency Virus/genetics , Animals , Cells, Cultured , Child, Preschool , Cytopathogenic Effect, Viral , Down-Regulation , Gene Deletion , HIV Infections/virology , HIV-1/genetics , Humans , Infant, Newborn , Mice , Mice, SCID , Mutation , Thymocytes/pathology
SELECTION OF CITATIONS
SEARCH DETAIL