Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Lancet HIV ; 11(5): e285-e299, 2024 May.
Article in English | MEDLINE | ID: mdl-38692824

ABSTRACT

BACKGROUND: An effective HIV vaccine will most likely need to have potent immunogenicity and broad cross-subtype coverage. The aim of the HIV Vaccine Trials Network (HVTN) 124 was to evaluate safety and immunogenicity of a unique polyvalent DNA-protein HIV vaccine with matching envelope (Env) immunogens. METHODS: HVTN 124 was a randomised, phase 1, placebo-controlled, double-blind study, including participants who were HIV seronegative and aged 18-50 years at low risk for infection. The DNA vaccine comprised five plasmids: four copies expressing Env gp120 (clades A, B, C, and AE) and one gag p55 (clade C). The protein vaccine included four DNA vaccine-matched GLA-SE-adjuvanted recombinant gp120 proteins. Participants were enrolled across six clinical sites in the USA and were randomly assigned to placebo or one of two vaccine groups (ie, prime-boost or coadministration) in a 5:1 ratio in part A and a 7:1 ratio in part B. Vaccines were delivered via intramuscular needle injection. The primary outcomes were safety and tolerability, assessed via frequency, severity, and attributability of local and systemic reactogenicity and adverse events, laboratory safety measures, and early discontinuations. Part A evaluated safety. Part B evaluated safety and immunogenicity of two regimens: DNA prime (administered at months 0, 1, and 3) with protein boost (months 6 and 8), and DNA-protein coadministration (months 0, 1, 3, 6, and 8). All randomly assigned participants who received at least one dose were included in the safety analysis. The study is registered with ClinicalTrials.gov (NCT03409276) and is closed to new participants. FINDINGS: Between April 19, 2018 and Feb 13, 2019, 60 participants (12 in part A [five men and seven women] and 48 in part B [21 men and 27 women]) were enrolled. All 60 participants received at least one dose, and 14 did not complete follow-up (six of 21 in the prime-boost group and eight of 21 in the coadminstration group). 11 clinical adverse events deemed by investigators as study-related occurred in seven of 48 participants in part B (eight of 21 in the prime-boost group and three of 21 in the coadministration group). Local reactogenicity in the vaccine groups was common, but the frequency and severity of reactogenicity signs or symptoms did not differ between the prime-boost and coadministration groups (eg, 20 [95%] of 21 in the prime-boost group vs 21 [100%] of 21 in the coadministration group had either local pain or tenderness of any severity [p=1·00], and seven [33%] vs nine [43%] had either erythema or induration [p=0·97]), nor did laboratory safety measures. There were no delayed-type hypersensitivity reactions or vasculitis or any severe clinical adverse events related to vaccination. The most frequently reported systemic reactogenicity symptoms in the active vaccine groups were malaise or fatigue (five [50%] of ten in part A and 17 [81%] of 21 in the prime-boost group vs 15 [71%] of 21 in the coadministration group in part B), headache (five [50%] and 18 [86%] vs 12 [57%]), and myalgia (four [40%] and 13 [62%] vs ten [48%]), mostly of mild or moderate severity. INTERPRETATION: Both vaccine regimens were safe, warranting evaluation in larger trials. FUNDING: US National Institutes of Health and US National Institute of Allergy and Infectious Diseases.


Subject(s)
AIDS Vaccines , HIV Antibodies , HIV Infections , HIV-1 , Vaccines, DNA , Humans , AIDS Vaccines/administration & dosage , AIDS Vaccines/immunology , AIDS Vaccines/adverse effects , Adult , Male , Female , Double-Blind Method , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Vaccines, DNA/adverse effects , HIV Infections/prevention & control , HIV Infections/immunology , Middle Aged , Young Adult , HIV Antibodies/blood , Adolescent , HIV-1/immunology , United States , Immunization, Secondary , Immunogenicity, Vaccine , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp120/genetics , Antibodies, Neutralizing/blood
2.
Vaccine ; 41(31): 4439-4446, 2023 07 12.
Article in English | MEDLINE | ID: mdl-37331838

ABSTRACT

This report summarizes the highlights of a workshop convened by the National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), on April 4-5, 2022, to provide a discussion forum for sharing insights on the current status, key challenges, and next steps to advance the current landscape of promising adjuvants in preclinical and clinical human immunodeficiency virus (HIV) vaccine studies. A key goal was to solicit and share recommendations on scientific, regulatory, and operational guidelines for bridging the gaps in rational selection, access, and formulation of clinically relevant adjuvants for HIV vaccine candidates. The NIAID Vaccine Adjuvant Program working group remains committed to accentuate promising adjuvants and nurturing collaborations between adjuvant and HIV vaccine developers.


Subject(s)
AIDS Vaccines , HIV Infections , United States , Humans , National Institute of Allergy and Infectious Diseases (U.S.) , HIV Infections/prevention & control , Adjuvants, Immunologic , National Institutes of Health (U.S.)
4.
PLoS Med ; 17(2): e1003038, 2020 02.
Article in English | MEDLINE | ID: mdl-32092060

ABSTRACT

BACKGROUND: HVTN 100 evaluated the safety and immunogenicity of an HIV subtype C pox-protein vaccine regimen, investigating a 12-month booster to extend vaccine-induced immune responses. METHODS AND FINDINGS: A phase 1-2 randomized double-blind placebo-controlled trial enrolled 252 participants (210 vaccine/42 placebo; median age 23 years; 43% female) between 9 February 2015 and 26 May 2015. Vaccine recipients received ALVAC-HIV (vCP2438) alone at months 0 and 1 and with bivalent subtype C gp120/MF59 at months 3, 6, and 12. Antibody (IgG, IgG3 binding, and neutralizing) and CD4+ T-cell (expressing interferon-gamma, interleukin-2, and CD40 ligand) responses were evaluated at month 6.5 for all participants and at months 12, 12.5, and 18 for a randomly selected subset. The primary analysis compared IgG binding antibody (bAb) responses and CD4+ T-cell responses to 3 vaccine-matched antigens at peak (month 6.5 versus 12.5) and durability (month 12 versus 18) timepoints; IgG responses to CaseA2_gp70_V1V2.B, a primary correlate of risk in RV144, were also compared at these same timepoints. Secondary and exploratory analyses compared IgG3 bAb responses, IgG bAb breadth scores, neutralizing antibody (nAb) responses, antibody-dependent cellular phagocytosis, CD4+ polyfunctionality responses, and CD4+ memory sub-population responses at the same timepoints. Vaccines were generally safe and well tolerated. During the study, there were 2 deaths (both in the vaccine group and both unrelated to study products). Ten participants became HIV-infected during the trial, 7% (3/42) of placebo recipients and 3% (7/210) of vaccine recipients. All 8 serious adverse events were unrelated to study products. Less waning of immune responses was seen after the fifth vaccination than after the fourth, with higher antibody and cellular response rates at month 18 than at month 12: IgG bAb response rates to 1086.C V1V2, 21.0% versus 9.7% (difference = 11.3%, 95% CI = 0.6%-22.0%, P = 0.039), and ZM96.C V1V2, 21.0% versus 6.5% (difference = 14.5%, 95% CI = 4.1%-24.9%, P = 0.004). IgG bAb response rates to all 4 primary V1V2 antigens were higher 2 weeks after the fifth vaccination than 2 weeks after the fourth vaccination: 87.7% versus 75.4% (difference = 12.3%, 95% CI = 1.7%-22.9%, P = 0.022) for 1086.C V1V2, 86.0% versus 63.2% (difference = 22.8%, 95% CI = 9.1%-36.5%, P = 0.001) for TV1c8.2.C V1V2, 67.7% versus 44.6% (difference = 23.1%, 95% CI = 10.4%-35.7%, P < 0.001) for ZM96.C V1V2, and 81.5% versus 60.0% (difference = 21.5%, 95% CI = 7.6%-35.5%, P = 0.002) for CaseA2_gp70_V1V2.B. IgG bAb response rates to the 3 primary vaccine-matched gp120 antigens were all above 90% at both peak timepoints, with no significant differences seen, except a higher response rate to ZM96.C gp120 at month 18 versus month 12: 64.5% versus 1.6% (difference = 62.9%, 95% CI = 49.3%-76.5%, P < 0.001). CD4+ T-cell response rates were higher at month 18 than month 12 for all 3 primary vaccine-matched antigens: 47.3% versus 29.1% (difference = 18.2%, 95% CI = 2.9%-33.4%, P = 0.021) for 1086.C, 61.8% versus 38.2% (difference = 23.6%, 95% CI = 9.5%-37.8%, P = 0.001) for TV1.C, and 63.6% versus 41.8% (difference = 21.8%, 95% CI = 5.1%-38.5%, P = 0.007) for ZM96.C, with no significant differences seen at the peak timepoints. Limitations were that higher doses of gp120 were not evaluated, this study was not designed to investigate HIV prevention efficacy, and the clinical significance of the observed immunological effects is uncertain. CONCLUSIONS: In this study, a 12-month booster of subtype C pox-protein vaccines restored immune responses, and slowed response decay compared to the 6-month vaccination. TRIAL REGISTRATION: ClinicalTrials.gov NCT02404311. South African National Clinical Trials Registry (SANCTR number: DOH--27-0215-4796).


Subject(s)
AIDS Vaccines/therapeutic use , Antibodies, Neutralizing/immunology , CD4-Positive T-Lymphocytes/immunology , HIV Infections/prevention & control , Human Immunodeficiency Virus Proteins/immunology , Immunization, Secondary , Immunoglobulin G/immunology , AIDS Vaccines/immunology , Adult , Arthralgia/chemically induced , Double-Blind Method , Female , Headache/chemically induced , Humans , Immunogenicity, Vaccine , Injection Site Reaction , Injections, Intramuscular , Male , South Africa , Young Adult
5.
Sci Rep ; 10(1): 2093, 2020 02 07.
Article in English | MEDLINE | ID: mdl-32034163

ABSTRACT

In the RV144 trial, vaccine-induced V1V2 IgG correlated with decreased HIV-1 risk. We investigated circulating antibody specificities in two phase 1 poxvirus prime-protein boost clinical trials conducted in South Africa: HVTN 097 (subtype B/E) and HVTN 100 (subtype C). With cross-subtype peptide microarrays and multiplex binding assays, we probed the magnitude and breadth of circulating antibody responses to linear variable loop 2 (V2) and conformational V1V2 specificities. Antibodies targeting the linear V2 epitope, a correlate of decreased HIV-1 risk in RV144, were elicited up to 100% and 61% in HVTN 097 and HVTN 100, respectively. Despite higher magnitude of envelope-specific responses in HVTN 100 compared to HVTN 097 (p's < 0.001), the magnitude and positivity for V2 linear epitope and V1V2 proteins were significantly lower in HVTN 100 compared to HVTN 097. Meanwhile, responses to other major linear epitopes including the variable 3 (V3) and constant 5 (C5) epitopes were higher in HVTN 100 compared to HVTN 097. Our data reveal substantial differences in the circulating antibody specificities induced by vaccination in these two canarypox prime-protein boost trials. Our findings suggest that the choice of viral sequences in prime-boost vaccine regimens, and potentially adjuvants and immunogen dose, influence the elicitation of V2-specific antibodies.


Subject(s)
AIDS Vaccines/immunology , Antibodies, Viral/immunology , Antibody Formation/immunology , HIV-1/immunology , Antibody Specificity/immunology , Canarypox virus/immunology , Epitopes/immunology , Female , HIV Envelope Protein gp120/immunology , Humans , Immunization, Secondary , Male
6.
Lancet HIV ; 5(7): e366-e378, 2018 07.
Article in English | MEDLINE | ID: mdl-29898870

ABSTRACT

BACKGROUND: Modest efficacy was reported for the HIV vaccine tested in the RV144 trial, which comprised a canarypox vector (ALVAC) and envelope (env) glycoprotein (gp120). These vaccine components were adapted to express HIV-1 antigens from strains circulating in South Africa, and the adjuvant was changed to increase immunogenicity. Furthermore, 12-month immunisation was added to improve durability. In the HIV Vaccine Trials Network (HVTN) 100 trial, we aimed to assess this new regionally adapted regimen for advancement to efficacy testing. METHODS: HVTN 100 is a phase 1/2, randomised controlled, double-blind trial at six community research sites in South Africa. We randomly allocated adults (aged 18-40 years) without HIV infection and at low risk of HIV infection to either the vaccine regimen (intramuscular injection of ALVAC-HIV vector [vCP2438] at 0, 1, 3, 6, and 12 months plus bivalent subtype C gp120 and MF59 adjuvant at 3, 6, and 12 months) or placebo, in a 5:1 ratio. Randomisation was done by computer-generated list. Participants, investigators, and those assessing outcomes were masked to random assignments. Primary outcomes included safety and immune responses associated with correlates of HIV risk in RV144, 2 weeks after vaccination at 6 months (month 6·5). We compared per-protocol participants (ie, those who completed the first four vaccinations and provided samples at month 6·5) from HVTN 100 with stored RV144 samples assayed contemporaneously. This trial is registered with the South African National Clinical Trials Registry (DOH-27-0215-4796) and ClinicalTrials.gov (NCT02404311). FINDINGS: Between Feb 9, 2015, and May 26, 2015, 252 participants were enrolled, of whom 210 were assigned vaccine and 42 placebo. 222 participants were included in the per-protocol analysis (185 vaccine and 37 placebo). 185 (100%) vaccine recipients developed IgG binding antibodies to all three vaccine-matched gp120 antigens with significantly higher titres (3·6-8·8 fold; all p<0·0001) than the corresponding vaccine-matched responses of RV144. The CD4+ T-cell response to the ZM96.C env protein in HVTN 100 was 56·4% (n=102 responders), compared with a response of 41·4% (n=79 responders) to 92TH023.AE in RV144 (p=0·0050). The IgG response to the 1086.C variable loops 1 and 2 (V1V2) env antigen in HVTN 100 was 70·5% (95% CI 63·5-76·6; n=129 responders), lower than the response to V1V2 in RV144 (99·0%, 95% CI 96·4-99·7; n=199 responders). INTERPRETATION: Although the IgG response to the HVTN 100 vaccine was lower than that reported in RV144, it exceeded the predicted 63% threshold needed for 50% vaccine efficacy using a V1V2 correlate of protection model. Thus, the subtype C HIV vaccine regimen qualified for phase 2b/3 efficacy testing, a critical next step of vaccine development. FUNDING: US National Institute of Allergy and Infectious Diseases (NIAID), and Bill & Melinda Gates Foundation.


Subject(s)
AIDS Vaccines/immunology , HIV Envelope Protein gp120/immunology , HIV Infections/prevention & control , HIV-1/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/adverse effects , Adjuvants, Immunologic/administration & dosage , Adolescent , Adult , Double-Blind Method , Female , Genetic Vectors , HIV Antibodies/blood , HIV Envelope Protein gp120/administration & dosage , HIV Envelope Protein gp120/genetics , HIV Infections/immunology , Humans , Immunoglobulin G/blood , Male , Polysorbates/administration & dosage , South Africa/epidemiology , Squalene/administration & dosage , Vaccination , Young Adult
7.
Vaccine ; 33(15): 1757-66, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25698494

ABSTRACT

The Division of AIDS Vaccine Research Program funds the discovery and development of HIV/AIDS vaccine candidates. Basic researchers, having discovered a potential vaccine in the laboratory, next want to take that candidate into the clinic to test the concept in humans, to see if it translates. Many of them have heard of "cGMP" and know that they are supposed to make a "GMP product" to take into the clinic, but often they are not very familiar with what "cGMP" means and why these good practices are so important. As members of the Vaccine Translational Research Branch, we frequently get asked "can't we use the material we made in the lab in the clinic?" or "aren't Phase 1 studies exempt from cGMP?" Over the years, we have had many experiences where researchers or their selected contract manufacturing organizations have not applied an appropriate degree of compliance with cGMP suitable for the clinical phase of development. We share some of these experiences and the lessons learned, along with explaining the importance of cGMP, just what cGMP means, and what they can assure, in an effort to de-mystify this subject and facilitate the rapid and safe translational development of HIV vaccines.


Subject(s)
AIDS Vaccines/standards , HIV Infections/prevention & control , Translational Research, Biomedical/standards , Clinical Trials as Topic , Government Regulation , Humans , Translational Research, Biomedical/legislation & jurisprudence
8.
Vaccine ; 26(32): 3969-77, 2008 Jul 29.
Article in English | MEDLINE | ID: mdl-18513838

ABSTRACT

In October 2007, a joint ANRS-NIH workshop was held on "Mucosal immunity and HIV/AIDS vaccines" in Veyrier-du-Lac, France. Goal of the meeting was to discuss recent developments in the understanding of viral entry and dissemination at mucosal surfaces, rationale for designing vaccines to elicit mucosal immune responses by various routes of immunization, and the types of immune responses elicited. Lessons were drawn from existing vaccines against viral mucosal infections, from the recent failure of the Merck Ad5/HIV vaccine and from attempts at mucosal immunization against SIV. This report summarizes the main concepts and conclusions that came out of the meeting.


Subject(s)
AIDS Vaccines/immunology , HIV Infections/immunology , HIV Infections/prevention & control , HIV/immunology , Animals , Drug Evaluation, Preclinical , HIV Antibodies , HIV Infections/virology , Humans , Immunity, Mucosal
10.
s.l; s.n; 1984. 24 p. tab, graf.
Non-conventional in English | Sec. Est. Saúde SP, HANSEN, Hanseníase Leprosy, SESSP-ILSLACERVO, Sec. Est. Saúde SP | ID: biblio-1234089

Subject(s)
Leprosy
11.
s.l; s.n; 1984. 5 p. ilus, tab.
Non-conventional in English | Sec. Est. Saúde SP, HANSEN, Hanseníase Leprosy, SESSP-ILSLACERVO, Sec. Est. Saúde SP | ID: biblio-1235876

Subject(s)
Leprosy
12.
s.l; s.n; nov. 1982. 6 p. tab, graf.
Non-conventional in English | Sec. Est. Saúde SP, HANSEN, Hanseníase Leprosy, SESSP-ILSLACERVO, Sec. Est. Saúde SP | ID: biblio-1240892

ABSTRACT

Leprosy is a spectral disease in which the patients at the lepromatous end display selective immunologic unresponsiveness to antigens of M. leprae. We have previously explored the possibility that the anergy in lepromatous leprosy mediated by suppressor cells. An in vitro system was developed in which lepromin induces suppression of the mitogenic response of lepromatous and borderline but not tuberculoid leprosy patients to concavalin A.


Subject(s)
Humans , Lepromin/immunology , Antigens, Bacterial/administration & dosage , Histocompatibility Antigens Class II/immunology , Leprosy/immunology , Leprosy/therapy , Immunotherapy , T-Lymphocytes, Regulatory/immunology , Receptors, Fc/genetics
13.
s.l; s.n; oct. 1979. 5 p. tab, graf.
Non-conventional in English | Sec. Est. Saúde SP, HANSEN, Hanseníase Leprosy, SESSP-ILSLACERVO, Sec. Est. Saúde SP | ID: biblio-1240785

ABSTRACT

The possibility of an active mechanism of immunologic suppression in leprosy was explored by assessing the in vitro lymphocyte responses of 61 leprosy patients and 30 normal individuals to the mitogen Con A in the presence or absence of Dharmendra lepromin. Lepromin-induced suppression of Con A stimulation was found in 32 of 35 lepromatous patients and 15 of 15 borderline patients, but only 2 of 15 tuberculoid patients and 2 of 30 normal controls. Cell fractionation studies indicated at least two cell populations involved in the in vitro lepromin-induced suppressor activity, adherent cells and T gamma-cells.


Subject(s)
Humans , Cell Adhesion , Lepromin/immunology , Lymphocyte Activation , Concanavalin A/pharmacology , Rosette Formation , Leprosy/immunology , Immunoglobulin G/immunology , T-Lymphocytes, Regulatory/immunology , Lymphocytes/classification , Receptors, Fc/immunology
14.
s.l; s.n; mar. 1979. 8 p. ilus, tab.
Non-conventional in English | Sec. Est. Saúde SP, HANSEN, Hanseníase Leprosy, SESSP-ILSLACERVO, Sec. Est. Saúde SP | ID: biblio-1240413

ABSTRACT

Guinea pigs immunized with intact or disrupted armadillo-grown human Mycobacterium leprae administered in aqueous or oil vehicles were tested with various dilutions of M. leprae suspended in saline, water-soluble M. leprae extract, purified protein derivative, and a water-soluble extract of normal armadillo tissue. The results demonstrated the following. (i) Under no conditions was any skin test reactivity found to normal armadillo tissue extract. (ii) Positive sensitization to both M. leprae and its water-soluble extract was achieved by sensitizing guinea pigs with M. leprae suspended in Hanks solution or saline. Autoclaved M. leprae in Hanks solution or saline inoculated intradermally was an effective immunogen. Oil suspensions or emulsions were effective at sensitization, but appeared to be no better and, in general, slightly weaker, than simple inoculation in aqueous suspension. (iii) Living BCG failed to reveal a significant adjuvant effect on sensitization to M. leprae. However, cord factor appeared to potentiate slightly the sensitization to M. leprae in aqueous suspension. (iv) The minimum dose required for sensitization with M. leprae in aqueous suspension was 55 micrograms of purified bacilli. (v) Animals inoculated with M. leprae in saline or with M. leprae together with BCG showed positive skin test reactivity to the first skin test application made fully 1 year after the initial sensitization. The efficacy of autoclaved, irradiated M. leprae in aqueous, oil-free medium suggests a relatively safe approach to human vaccination studies.


Subject(s)
Animals , Guinea Pigs , Leprosy/immunology , Hypersensitivity, Delayed/immunology , Immunity, Cellular , Mycobacterium bovis/immunology , Mycobacterium leprae/immunology , Cross Reactions , Dose-Response Relationship, Immunologic , Skin Tests , BCG Vaccine
SELECTION OF CITATIONS
SEARCH DETAIL
...