95 results on '"Marovich M"'
Search Results
2. Early immune events during acute HIV infection
- Author
-
Slike BM, Tassaneetrithep B, Nitayaphan S, Rono K, Sanga E, Sekiziyivu A, Stablein D, Eller L, Eller MA, Kim J, Michael N, Robb M, and Marovich M
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2012
- Full Text
- View/download PDF
3. The impact of differences in viral entry and trafficking on the mechanism of HIV-1 neutralization
- Author
-
Cincotta CD, Wieczorek LM, Barrows B, Brown B, Asher L, Michael N, Marovich M, and Polonis VR
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2012
- Full Text
- View/download PDF
4. Distinct gene expression profiles associated with the susceptibility of pathogen-specific CD4+ T cells to HIV-1 infection
- Author
-
Hu H, Nau M, Ehrenberg P, Chenine A, Daye Z, Wei Z, Michael N, Vahey M, Kim J, Marovich M, and Ratto-Kim S
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2012
- Full Text
- View/download PDF
5. Antigenicity and immunogenicity of a novel, acute HIV-1 Tanzanian subtype C gp145 envelope protein for clinical development
- Author
-
Polonis V, Wieczorek L, Kalyanaraman V, Matyas G, Whitney S, Williams C, Tovanabutra S, Sanders-Buell E, Wesberry M, Ochsenbauer C, Chenine A, Rao M, Tong T, Alving C, Cheng H, Zolla-Pazner S, Michael N, VanCott T, and Marovich M
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2012
- Full Text
- View/download PDF
6. Antibody-mediated inhibition of HIV-1 elicited by HIV-I DNA priming and boosting with heterologous HIV-1 recombinant MVA in healthy Tanzanian adults
- Author
-
Joachim A, Nilsson C, Aboud S, Lyamuya EF, Robb M, Marovich M, Ochsenbauer C, Wahren B, Sandström E, Biberfeld G, Ferrari G, and Polonis VR
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2012
- Full Text
- View/download PDF
7. P04-22. Multispecific monoclonal antibodies bind to primary human macrophages and induce the production of protective chemokines, MIP-1α and MIP-1β
- Author
-
Alving CR, Marovich M, Polonis VR, Asher LM, Matyas GR, Wieczorek L, Boonnak K, Peachman KK, Weaver KL, and Rao M
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2009
- Full Text
- View/download PDF
8. OA04-02. Strong HIV-specific CD4 and CD8 T-lymphocyte proliferation in HIV-1 DNA prime/modified vaccinia virus Ankara (MVA) heterologous boost vaccinees
- Author
-
Biberfeld G, Sandström E, Gaines H, Marovich M, Wahren B, Karlén K, Aboud S, Nilsson C, and Godoy-Ramirez K
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2009
- Full Text
- View/download PDF
9. P14-14 LB. A low dose of multigene, multiclade HIV DNA given intradermally induces strong and broad immune responses after boosting with heterologous HIV MVA
- Author
-
Wahren B, Moss B, Earl P, Michael N, Marovich M, Robb M, Brave A, Hejdeman B, Stout R, Mwanyika L, Mbwana J, Janabi M, Lyamuya E, Aris EA, Moshiro C, Buma D, Francis J, Nilsson C, Aboud S, Bakari M, Biberfeld G, Pallangyo K, Mhalu F, and Sandstrom E
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2009
- Full Text
- View/download PDF
10. False positive endotoxin results in a DC product caused by (1→3)-β–d-glucans acquired from a sterilizing cellulose filter
- Author
-
Anderson, J., Eller, M., Finkelman, M., Birx, D., Schlesinger-Frankel, S., and Marovich, M.
- Published
- 2002
- Full Text
- View/download PDF
11. Change in Brain Magnetic Resonance Spectroscopy after Treatment during Acute HIV Infection
- Author
-
Sailasuta, N, Ross, W, Ananworanich, J, Chalermchai, T, DeGruttola, V, Lerdlum, S, Pothisri, M, Busovaca, E, Ratto-Kim, S, Jagodzinski, L, Spudich, S, Michael, N, Kim, JH, Valcour, V, Phanuphak, N, Teeratakulpisarn, N, Fletcher, JLK, Suttichom, D, Pinyakorn, S, Rattanamanee, S, Chomchey, N, Mangum, P, Ubolyam, S, Suwanwela, NC, Chaisinanunkul, N, Suthiponpaisan, U, Sutthapas, C, deSouza, M, Ngauy, V, Trichavaroj, R, Akapirat, S, Marovich, M, Wendelken, L, Liu, C, Mun, E, and Miller, B
- Abstract
Objective: Single voxel proton magnetic resonance spectroscopy (MRS) can be used to monitor changes in brain inflammation and neuronal integrity associated with HIV infection and its treatments. We used MRS to measure brain changes during the first weeks following HIV infection and in response to antiretroviral therapy (ART). Methods: Brain metabolite levels of N-acetyl aspartate (NAA), choline (tCHO), creatine (CR), myoinositol (MI), and glutamate and glutamine (GLX) were measured in acute HIV subjects (n = 31) and compared to chronic HIV+individuals (n = 26) and HIV negative control subjects (n = 10) from Bangkok, Thailand. Metabolites were measured in frontal gray matter (FGM), frontal white matter (FWM), occipital gray matter (OGM), and basal ganglia (BG). Repeat measures were obtained in 17 acute subjects 1, 3 and 6 months following initiation of ART. Results: After adjustment for age we identified elevated BG tCHO/CR in acute HIV cases at baseline (median 14 days after HIV infection) compared to control (p = 0.0014), as well as chronic subjects (p = 0.0023). A similar tCHO/CR elevation was noted in OGM; no other metabolite abnormalities were seen between acute and control subjects. Mixed longitudinal models revealed resolution of BG tCHO/CR elevation after ART (p = 0.022) with tCHO/CR similar to control subjects at 6 months. Interpretation: We detected cellular inflammation in the absence of measurable neuronal injury within the first month of HIV infection, and normalization of this inflammation following acutely administered ART. Our findings suggest that early ART may be neuroprotective in HIV infection by mitigating processes leading to CNS injury. © 2012 Sailasuta et al.
- Published
- 2012
- Full Text
- View/download PDF
12. Central nervous system viral invasion and inflammation during acute HIV infection
- Author
-
Valcour, V, Chalermchai, T, Sailasuta, N, Marovich, M, Lerdlum, S, Suttichom, D, Suwanwela, NC, Jagodzinski, L, Michael, N, Spudich, S, van Griensven, F, de Souza, M, Kim, J, Ananworanich, J, and RV254/SEARCH 010 Study Group
- Subjects
Inflammation ,Adult ,Male ,Young Adult ,Central Nervous System Infections ,RV254/SEARCH 010 Study Group ,Acute Disease ,Humans ,RNA, Viral ,HIV Infections ,Female ,Middle Aged ,Magnetic Resonance Imaging - Abstract
Understanding the earliest central nervous system (CNS) events during human immunodeficiency virus (HIV) infection is crucial to knowledge of neuropathogenesis, but these have not previously been described in humans.Twenty individuals who had acute HIV infection (Fiebig stages I-IV), with average 15 days after exposure, underwent clinical neurological, cerebrospinal fluid (CSF), magnetic resonance imaging, and magnetic resonance spectroscopy (MRS) characterization.HIV RNA was detected in the CSF from 15 of 18 subjects as early as 8 days after estimated HIV transmission. Undetectable CSF levels of HIV (in 3 of 18) was noted during Fiebig stages I, II, and III, with plasma HIV RNA levels of 285651, 2321, and 81978 copies/mL, respectively. On average, the CSF HIV RNA level was 2.42 log(10) copies/mL lower than that in plasma. There were no cases in which the CSF HIV RNA level exceeded that in plasma. Headache was common during the acute retroviral syndrome (in 11 of 20 subjects), but no other neurological signs or symptoms were seen. Intrathecal immune activation was identified in some subjects with elevated CSF neopterin, monocyte chemotactic protein/CCL2, and interferon γ-induced protein 10/CXCL-10 levels. Brain inflammation was suggested by MRS.CSF HIV RNA was detectable in humans as early as 8 days after exposure. CNS inflammation was apparent by CSF analysis and MRS in some individuals during acute HIV infection.
- Published
- 2012
- Full Text
- View/download PDF
13. The Meaning of Soul: Black Music and Resilience Since the 1960s.
- Author
-
Marovich, M.
- Subjects
- *
BLACK music , *SOUL music , *AFRICAN American music , *NINETEEN sixties , *MUSICAL perception ,IRISH music - Published
- 2021
14. P10-12. Altered NK cell phenotype and function in Ugandans with chronic HIV-1 infection
- Author
-
Robb ML, Wabwire-Mangen F, de Souza MS, Michael NL, Marovich MA, Guwatudde D, Kijak GH, Koehler RN, Eller L, Eller MA, Currier JR, and Sandberg JK
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2009
- Full Text
- View/download PDF
15. Update: Cutaneous Leishmaniasis in U.S. Military Personnel -- Southwest/Central Asia, 2002--2004.
- Author
-
Aronson, N., Ananthakrishnan, M., Berstein, W., Hochberg, L., Marovich, M., Ockenhouse, C., Yoon, I., Weina, P., Benson, P., Fischer, J., Hack, D., Hawkes, C., Polhemus, M., Wortmann, G., McEvoy, P., Neafie, R., Defraites, R., and Herwaldt, B. L.
- Subjects
CUTANEOUS leishmaniasis ,MILITARY hygiene ,AMERICAN military personnel ,DEPLOYMENT (Military strategy) ,HISPANIC American soldiers - Abstract
Presents information on cases of cutaneous leishmaniasis (CL) in U.S. military personnel deployed to Afghanistan, Iraq and Kuwait from August 2002 to February 2004. Percentage of CL military patients who were non-Hispanic white; Measures implemented by the Department of Defense to decrease the risk of CL.
- Published
- 2004
16. Defining epitope coverage requirements for T cell-based HIV vaccines: Theoretical considerations and practical applications
- Author
-
Currier Jeffrey R, Robb Merlin L, Michael Nelson L, and Marovich Mary A
- Subjects
Medicine - Abstract
Abstract Background HIV vaccine development must address the genetic diversity and plasticity of the virus that permits the presentation of diverse genetic forms to the immune system and subsequent escape from immune pressure. Assessment of potential HIV strain coverage by candidate T cell-based vaccines (whether natural sequence or computationally optimized products) is now a critical component in interpreting candidate vaccine suitability. Methods We have utilized an N-mer identity algorithm to represent T cell epitopes and explore potential coverage of the global HIV pandemic using natural sequences derived from candidate HIV vaccines. Breadth (the number of T cell epitopes generated) and depth (the variant coverage within a T cell epitope) analyses have been incorporated into the model to explore vaccine coverage requirements in terms of the number of discrete T cell epitopes generated. Results We show that when multiple epitope generation by a vaccine product is considered a far more nuanced appraisal of the potential HIV strain coverage of the vaccine product emerges. By considering epitope breadth and depth several important observations were made: (1) epitope breadth requirements to reach particular levels of vaccine coverage, even for natural sequence-based vaccine products is not necessarily an intractable problem for the immune system; (2) increasing the valency (number of T cell epitope variants present) of vaccine products dramatically decreases the epitope requirements to reach particular coverage levels for any epidemic; (3) considering multiple-hit models (more than one exact epitope match with an incoming HIV strain) places a significantly higher requirement upon epitope breadth in order to reach a given level of coverage, to the point where low valency natural sequence based products would not practically be able to generate sufficient epitopes. Conclusions When HIV vaccine sequences are compared against datasets of potential incoming viruses important metrics such as the minimum epitope count required to reach a desired level of coverage can be easily calculated. We propose that such analyses can be applied early in the planning stages and during the execution phase of a vaccine trial to explore theoretical and empirical suitability of a vaccine product to a particular epidemic setting.
- Published
- 2011
- Full Text
- View/download PDF
17. Immunogenic selection of HIV-1 MPER epitopes for improved vaccine design.
- Author
-
Wieczorek, L., Rao, M., Peachman, K., Steers, N., Marovich, M., Michael, N., Polonis, V., and Rao, V.
- Subjects
EPITOPES - Abstract
An abstract of the research paper on immunogenic selection of HIV-1 MPER epitopes that helps in improving vaccine design by L. Wieczorek colleagues at AIDS Vaccine 2012 in Boston, Massachusetts is presented.
- Published
- 2012
- Full Text
- View/download PDF
18. Pre-existing humoral immunity in military smallpox vaccinees temporally effects MVA-vectored transgene expression in dendritic cells.
- Author
-
Ngauy, V., Slike, B., and Marovich, M.
- Subjects
SMALLPOX vaccines ,TRANSGENE expression - Abstract
An abstract of the research paper "Pre-existing humoral immunity in military smallpox vaccinees temporally effects MVA-vectored transgene expression in dendritic cells," by B. Slike and colleagues is presented.
- Published
- 2012
- Full Text
- View/download PDF
19. The impact of differences in viral entry and trafficking on the mechanism of HIV-1 neutralization.
- Author
-
Macedo Cincotta, C. D., Wieczorek, L. M., Barrows, B., Brown, B., Asher, L., Michael, N., Marovich, M., and Polonis, V. R.
- Subjects
HIV infections - Abstract
An abstract of the conference paper "The Impact of Differences in Viral Entry and Trafficking on the Mechanism of HIV-1 Neutralization," by C.D. Macedo Cincotta and colleagues is presented.
- Published
- 2012
- Full Text
- View/download PDF
20. Distinct gene expression profiles associated with the susceptibility of pathogen-specific CD4+ T cells to HIV-1 infection.
- Author
-
H. Hu, M. Nau, Ehrenberg, P., Chenine, A., Daye, Z., Z. Wei, Michael, N., Vahey, M., J. Kim, Marovich, M., and Ratto-Kim, S.
- Subjects
GENE expression - Abstract
An abstract of the conference paper "Distinct gene expression profiles associated with the susceptibility of pathogen-specific CD4+ T cells to HIV-1 infection," by H. Hu and colleagues are presented.
- Published
- 2012
- Full Text
- View/download PDF
21. 146 Strong and Broad Immunogenicity of a Multigene, Multiclade HIV-1 DNA Prime MVA Boost Vaccine Regimen Among Healthy Tanzanian Volunteers.
- Author
-
Biberfeld, Gunnel, Aboud, S, Bakari, M, Nilsson, C, Moshiro, C, Aris, E, Lyamuya, E, Janabi, M, Godoy-Ramirez, K, Earl, P, Robb, M, Marovich, M, Michael, N, Wahren, B, Pallangyo, K, Mhalu, F, and Sandström, E
- Published
- 2011
- Full Text
- View/download PDF
22. Final Analysis of Efficacy and Safety of Single-Dose Ad26.COV2.S.
- Author
-
Sadoff, J., Gray, G., Vandebosch, A., Cardenas, V., Shukarev, G., Grinsztejn, B., Goepfert, P. A., Truyers, C., Van Dromme, I., Spiessens, B., Vingerhoets, J., Custers, J., Scheper, G., Robb, M. L., Treanor, J., Ryser, M. F., Barouch, D. H., Swann, E., Marovich, M. A., and Neuzil, K. M.
- Subjects
- *
CORONAVIRUS diseases , *SARS-CoV-2 , *COVID-19 , *VACCINE effectiveness - Abstract
Background: The Ad26.COV2.S vaccine was highly effective against severe-critical coronavirus disease 2019 (Covid-19), hospitalization, and death in the primary phase 3 efficacy analysis.Methods: We conducted the final analysis in the double-blind phase of our multinational, randomized, placebo-controlled trial, in which adults were assigned in a 1:1 ratio to receive single-dose Ad26.COV2.S (5×1010 viral particles) or placebo. The primary end points were vaccine efficacy against moderate to severe-critical Covid-19 with onset at least 14 days after administration and at least 28 days after administration in the per-protocol population. Safety and key secondary and exploratory end points were also assessed.Results: Median follow-up in this analysis was 4 months; 8940 participants had at least 6 months of follow-up. In the per-protocol population (39,185 participants), vaccine efficacy against moderate to severe-critical Covid-19 at least 14 days after administration was 56.3% (95% confidence interval [CI], 51.3 to 60.8; 484 cases in the vaccine group vs. 1067 in the placebo group); at least 28 days after administration, vaccine efficacy was 52.9% (95% CI, 47.1 to 58.1; 433 cases in the vaccine group vs. 883 in the placebo group). Efficacy in the United States, primarily against the reference strain (B.1.D614G) and the B.1.1.7 (alpha) variant, was 69.7% (95% CI, 60.7 to 76.9); efficacy was reduced elsewhere against the P.1 (gamma), C.37 (lambda), and B.1.621 (mu) variants. Efficacy was 74.6% (95% CI, 64.7 to 82.1) against severe-critical Covid-19 (with only 4 severe-critical cases caused by the B.1.617.2 [delta] variant), 75.6% (95% CI, 54.3 to 88.0) against Covid-19 leading to medical intervention (including hospitalization), and 82.8% (95% CI, 40.5 to 96.8) against Covid-19-related death, with protection lasting 6 months or longer. Efficacy against any severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection was 41.7% (95% CI, 36.3 to 46.7). Ad26.COV2.S was associated with mainly mild-to-moderate adverse events, and no new safety concerns were identified.Conclusions: A single dose of Ad26.COV2.S provided 52.9% protection against moderate to severe-critical Covid-19. Protection varied according to variant; higher protection was observed against severe Covid-19, medical intervention, and death than against other end points and lasted for 6 months or longer. (Funded by Janssen Research and Development and others; ENSEMBLE ClinicalTrials.gov number, NCT04505722.). [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
23. Subcutaneous REGEN-COV Antibody Combination to Prevent Covid-19.
- Author
-
O'Brien, M. P., Forleo-Neto, E., Musser, B. J., Isa, F., Chan, K.-C., Sarkar, N., Bar, K. J., Barnabas, R. V., Barouch, D. H., Cohen, M. S., Hurt, C. B., Burwen, D. R., Marovich, M. A., Hou, P., Heirman, I., Davis, J. D., Turner, K. C., Ramesh, D., Mahmood, A., and Hooper, A. T.
- Abstract
Background: REGEN-COV (previously known as REGN-COV2), a combination of the monoclonal antibodies casirivimab and imdevimab, has been shown to markedly reduce the risk of hospitalization or death among high-risk persons with coronavirus disease 2019 (Covid-19). Whether subcutaneous REGEN-COV prevents severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and subsequent Covid-19 in persons at high risk for infection because of household exposure to a person with SARS-CoV-2 infection is unknown.Methods: We randomly assigned, in a 1:1 ratio, participants (≥12 years of age) who were enrolled within 96 hours after a household contact received a diagnosis of SARS-CoV-2 infection to receive a total dose of 1200 mg of REGEN-COV or matching placebo administered by means of subcutaneous injection. At the time of randomization, participants were stratified according to the results of the local diagnostic assay for SARS-CoV-2 and according to age. The primary efficacy end point was the development of symptomatic SARS-CoV-2 infection through day 28 in participants who did not have SARS-COV-2 infection (as measured by reverse-transcriptase-quantitative polymerase-chain-reaction assay) or previous immunity (seronegativity).Results: Symptomatic SARS-CoV-2 infection developed in 11 of 753 participants in the REGEN-COV group (1.5%) and in 59 of 752 participants in the placebo group (7.8%) (relative risk reduction [1 minus the relative risk], 81.4%; P<0.001). In weeks 2 to 4, a total of 2 of 753 participants in the REGEN-COV group (0.3%) and 27 of 752 participants in the placebo group (3.6%) had symptomatic SARS-CoV-2 infection (relative risk reduction, 92.6%). REGEN-COV also prevented symptomatic and asymptomatic infections overall (relative risk reduction, 66.4%). Among symptomatic infected participants, the median time to resolution of symptoms was 2 weeks shorter with REGEN-COV than with placebo (1.2 weeks and 3.2 weeks, respectively), and the duration of a high viral load (>104 copies per milliliter) was shorter (0.4 weeks and 1.3 weeks, respectively). No dose-limiting toxic effects of REGEN-COV were noted.Conclusions: Subcutaneous REGEN-COV prevented symptomatic Covid-19 and asymptomatic SARS-CoV-2 infection in previously uninfected household contacts of infected persons. Among the participants who became infected, REGEN-COV reduced the duration of symptomatic disease and the duration of a high viral load. (Funded by Regeneron Pharmaceuticals and others; ClinicalTrials.gov number, NCT04452318.). [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
24. Safety and Efficacy of Single-Dose Ad26.COV2.S Vaccine against Covid-19.
- Author
-
Sadoff, J., Gray, G., Vandebosch, A., Cárdenas, V., Shukarev, G., Grinsztejn, B., Goepfert, P. A., Truyers, C., Fennema, H., Spiessens, B., Offergeld, K., Scheper, G., Taylor, K. L., Robb, M. L., Treanor, J., Barouch, D. H., Stoddard, J., Ryser, M. F., Marovich, M. A., and Neuzil, K. M.
- Subjects
- *
COVID-19 , *COVID-19 vaccines , *VACCINE effectiveness , *SARS-CoV-2 - Abstract
Background: The Ad26.COV2.S vaccine is a recombinant, replication-incompetent human adenovirus type 26 vector encoding full-length severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein in a prefusion-stabilized conformation.Methods: In an international, randomized, double-blind, placebo-controlled, phase 3 trial, we randomly assigned adult participants in a 1:1 ratio to receive a single dose of Ad26.COV2.S (5×1010 viral particles) or placebo. The primary end points were vaccine efficacy against moderate to severe-critical coronavirus disease 2019 (Covid-19) with an onset at least 14 days and at least 28 days after administration among participants in the per-protocol population who had tested negative for SARS-CoV-2. Safety was also assessed.Results: The per-protocol population included 19,630 SARS-CoV-2-negative participants who received Ad26.COV2.S and 19,691 who received placebo. Ad26.COV2.S protected against moderate to severe-critical Covid-19 with onset at least 14 days after administration (116 cases in the vaccine group vs. 348 in the placebo group; efficacy, 66.9%; adjusted 95% confidence interval [CI], 59.0 to 73.4) and at least 28 days after administration (66 vs. 193 cases; efficacy, 66.1%; adjusted 95% CI, 55.0 to 74.8). Vaccine efficacy was higher against severe-critical Covid-19 (76.7% [adjusted 95% CI, 54.6 to 89.1] for onset at ≥14 days and 85.4% [adjusted 95% CI, 54.2 to 96.9] for onset at ≥28 days). Despite 86 of 91 cases (94.5%) in South Africa with sequenced virus having the 20H/501Y.V2 variant, vaccine efficacy was 52.0% and 64.0% against moderate to severe-critical Covid-19 with onset at least 14 days and at least 28 days after administration, respectively, and efficacy against severe-critical Covid-19 was 73.1% and 81.7%, respectively. Reactogenicity was higher with Ad26.COV2.S than with placebo but was generally mild to moderate and transient. The incidence of serious adverse events was balanced between the two groups. Three deaths occurred in the vaccine group (none were Covid-19-related), and 16 in the placebo group (5 were Covid-19-related).Conclusions: A single dose of Ad26.COV2.S protected against symptomatic Covid-19 and asymptomatic SARS-CoV-2 infection and was effective against severe-critical disease, including hospitalization and death. Safety appeared to be similar to that in other phase 3 trials of Covid-19 vaccines. (Funded by Janssen Research and Development and others; ENSEMBLE ClinicalTrials.gov number, NCT04505722.). [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
25. Effect of timing of casirivimab and imdevimab administration relative to mRNA-1273 COVID-19 vaccination on vaccine-induced SARS-CoV-2 neutralising antibody responses: a prospective, open-label, phase 2, randomised controlled trial.
- Author
-
Isa F, Gonzalez Ortiz AM, Meyer J, Hamilton JD, Olenchock BA, Brackin T, Ganguly S, Forleo-Neto E, Faria L, Heirman I, Marovich M, Hutter J, Polakowski L, Irvin SC, Thakur M, Hooper AT, Baum A, Petro CD, Fakih FA, McElrath MJ, De Rosa SC, Cohen KW, Williams LD, Hellman CA, Odeh AJ, Patel AH, Tomaras GD, Geba GP, Kyratsous CA, Musser B, Yancopoulos GD, and Herman GA
- Abstract
Background: Deeper insight is needed on how monoclonal antibodies (mAbs) affect vaccine-mediated immune responses when targeting the same protein. We describe the first prospective randomised trial designed to understand mAb-mediated alterations in vaccine-induced immune responses to SARS-CoV-2 spike protein epitopes., Methods: This randomised, open-label, parallel-group study assessed the potential interaction of a mAb combination, casirivimab and imdevimab, with a vaccine, Moderna's mRNA-1273, in healthy SARS-CoV-2 immunologically naive, seronegative adults at six centres in the USA. Participants were randomly assigned (per prespecified randomisation ratios within enrolment waves) according to a computer-generated randomisation scheme, stratified by age (<65 years and ≥65 years), to various intravenous or subcutaneous doses of casirivimab and imdevimab before, after, or at the same time as mRNA-1273 or to mRNA-1273 only. The doses of casirivimab and imdevimab were chosen to mimic various time intervals between receipt of 1200 mg of the mAb and the first dose of a primary series with mRNA-1273. The primary endpoint was vaccine-induced 50% inhibitory dilution neutralising antibody titres to SARS-CoV-2 spike protein, 56 days after the first vaccination. Secondary endpoints included vaccine-induced total antibodies to SARS-CoV-2 antigens and incidence of treatment-emergent adverse events. Exploratory endpoints included blood-derived T-cell and B-cell responses. The per-protocol set was used for the analysis of the primary endpoint and included all randomly assigned participants who received both doses of the vaccine and completed the injection or infusion of casirivimab and imdevimab per protocol, had no evidence of SARS-CoV-2 infection in the past or in the 56 days after the first dose of vaccine, and did not receive any intervention outside of the study that could alter the immune response. Safety was assessed in the safety analysis set, which included all randomly assigned participants who had received one or more doses of mRNA-1273 or any study drug, and analysed based on treatment received. The study is registered with ClinicalTrials.gov, NCT04852978, and is complete., Findings: Between April 29, 2021, and Nov 21, 2022, 807 participants were assessed for eligibility and 295 were randomly assigned. 293 participants were included in the safety analysis set and 260 were included in the per-protocol set. All vaccinated participants developed neutralising antibodies to SARS-CoV-2, with median titres above the published protective threshold (100 IU/mL) against the SARS-CoV-2 D614G variant (considered a reference strain at the time the initial COVID-19 vaccines were developed). Titres were decreased up to 4-fold (median titres 280-450 IU/mL for casirivimab and imdevimab vs 1160 IU/mL for vaccine only on day 56) when casirivimab and imdevimab was given 85 days or less before vaccination (150-1200 mg intravenously) or co-administered subcutaneously (600 mg or 1200 mg) with vaccination. Minimal reduction in neutralisation titres was observed in the 48 mg and 12 mg intravenous groups, corresponding to receipt of casirivimab and imdevimab 113 days and 169 days, respectively, before vaccination, and when administering the vaccine 6 days before the mAb. Across all groups, mAbs had a minimal effect on vaccine-induced total antibodies and T-cell responses to the spike protein. Casirivimab and imdevimab plus mRNA-1273 was generally well tolerated; a slight increase in treatment-emergent adverse events was observed in the casirivimab and imdevimab plus vaccine groups versus the vaccine-only group., Interpretation: Casirivimab and imdevimab administration before or at the time of COVID-19 vaccination reduced the elicitation of SARS-CoV-2 neutralising antibodies, but minimal effect was observed when vaccination occurred before mAb administration. Although the clinical significance of this decrease in neutralisation is unclear, this evidence suggests that further investigation of potential interactions could be warranted before concurrent clinical use of mAbs and vaccines targeting the same viral proteins as their main modes of action for the prevention or treatment of infectious diseases., Funding: Regeneron Pharmaceuticals and F Hoffmann-La Roche., Competing Interests: Declaration of interests FI and JDH are employees and stockholders of Regeneron Pharmaceuticals, and report having patents pending, which have been licensed and receiving royalties, with Regeneron Pharmaceuticals. AMGO, BAO, TB, SG, LF, SCI, MT, GPG, and BM are employees and stockholders of Regeneron Pharmaceuticals. JM is an employee of Regeneron Pharmaceuticals. EF-N is a former employee and current stockholder of Regeneron Pharmaceuticals, and reports patents pending, which have been licensed and receiving royalties, with Regeneron Pharmaceuticals. IH is an employee and stockholder of Regeneron Pharmaceuticals, and a stockholder of Merck & Co. ATH is an employee and stockholder of Regeneron Pharmaceuticals, is a stockholder of Pfizer, and reports a patent pending, which has been licensed and receiving royalties, with Regeneron Pharmaceuticals. AB, CAK, and GDY are employees and stockholders of Regeneron Pharmaceuticals and report having issued patents (US patent numbers 10 787 501, 10 954 289, and 10 975 139) and pending patents, which have been licensed and receiving royalties, with Regeneron Pharmaceuticals. CDP is an employee and stockholder of Regeneron Pharmaceuticals and reports a patent pending with Regeneron Pharmaceuticals. MJM reports funding from Regeneron Pharmaceuticals, paid to her institution, and grants from the US National Institute of Allergy and Infectious Diseases, the Biomedical Advanced Research and Development Authority, Moderna, Sanofi Pasteur, and Janssen, paid to her institution. SCDR reports grants from the National Institutes of Health and the Bill & Melinda Gates Foundation awarded to his institution, and contracts from Regeneron Pharmaceuticals, The Henry M Jackson Foundation, Johnson & Johnson Innovative Medicine (formerly Janssen Pharmaceuticals), Gates Medical Research Institute, Paul G Allen Family Foundation, and Battelle, awarded to his institution. KWC reports funding from Regeneron Pharmaceuticals, paid to her institution, and is an employee and stockholder of Moderna. LDW CAH, AJO, AHP, and GDT report funding and provision of study materials from Regeneron Pharmaceuticals, paid to their institution. GAH is an employee and stockholder of Regeneron Pharmaceuticals and reports having a patent pending, which has been licensed and receiving royalties, with Regeneron Pharmaceuticals, as well as a pending patent application. MM, JH, LP, and FAF declare no competing interests., (Copyright © 2024. Elsevier Ltd. All rights reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
26. Examining protective effects of SARS-CoV-2 neutralizing antibodies after vaccination or monoclonal antibody administration.
- Author
-
Follmann D, O'Brien MP, Fintzi J, Fay MP, Montefiori D, Mateja A, Herman GA, Hooper AT, Turner KC, Chan KC, Forleo-Neto E, Isa F, Baden LR, El Sahly HM, Janes H, Doria-Rose N, Miller J, Zhou H, Dang W, Benkeser D, Fong Y, Gilbert PB, Marovich M, and Cohen MS
- Subjects
- Humans, 2019-nCoV Vaccine mRNA-1273, Antibodies, Neutralizing, Antibodies, Viral, COVID-19 Vaccines, SARS-CoV-2, Vaccination, Antibodies, Monoclonal, COVID-19 prevention & control
- Abstract
While new vaccines for SARS-CoV-2 are authorized based on neutralizing antibody (nAb) titer against emerging variants of concern, an analogous pathway does not exist for preventative monoclonal antibodies. In this work, nAb titers were assessed as correlates of protection against COVID-19 in the casirivimab + imdevimab monoclonal antibody (mAb) prevention trial (ClinicalTrials.gov #NCT4452318) and in the mRNA-1273 vaccine trial (ClinicalTrials.gov #NCT04470427). In the mAb trial, protective efficacy of 92% (95% confidence interval (CI): 84%, 98%) is associated with a nAb titer of 1000 IU50/ml, with lower efficacy at lower nAb titers. In the vaccine trial, protective efficacies of 93% [95% CI: 91%, 95%] and 97% (95% CI: 95%, 98%) are associated with nAb titers of 100 and 1000 IU50/ml, respectively. These data quantitate a nAb titer correlate of protection for mAbs benchmarked alongside vaccine induced nAb titers and support nAb titer as a surrogate endpoint for authorizing new mAbs., (© 2023. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2023
- Full Text
- View/download PDF
27. Quantifying how single dose Ad26.COV2.S vaccine efficacy depends on Spike sequence features.
- Author
-
Magaret C, Li L, deCamp A, Rolland M, Juraska M, Williamson B, Ludwig J, Molitor C, Benkeser D, Luedtke A, Simpkins B, Carpp L, Bai H, Deariove B, Greninger A, Roychoudhury P, Sadoff J, Gray G, Roels S, Vandebosch A, Stieh D, Le Gars M, Vingerhoets J, Grinsztejn B, Goepfert P, Truyers C, Van Dromme I, Swann E, Marovich M, Follmann D, Neuzil K, Corey L, Hyrien O, Paiva de Sousa L, Casapia M, Losso M, Little S, Gaur A, Bekker LG, Garrett N, Heng F, Sun Y, and Gilbert P
- Abstract
It is of interest to pinpoint SARS-CoV-2 sequence features defining vaccine resistance. In the ENSEMBLE randomized, placebo-controlled phase 3 trial, estimated single-dose Ad26.COV2.S vaccine efficacy (VE) was 56% against moderate to severe-critical COVID-19. SARS-CoV-2 Spike sequences were measured from 484 vaccine and 1,067 placebo recipients who acquired COVID-19 during the trial. In Latin America, where Spike diversity was greatest, VE was significantly lower against Lambda than against Reference and against all non-Lambda variants [family-wise error rate (FWER) p < 0.05]. VE also differed by residue match vs. mismatch to the vaccine-strain residue at 16 amino acid positions (4 FWER p < 0.05; 12 q-value ≤ 0.20). VE significantly decreased with physicochemical-weighted Hamming distance to the vaccine-strain sequence for Spike, receptor-binding domain, N-terminal domain, and S1 (FWER p < 0.001); differed (FWER ≤ 0.05) by distance to the vaccine strain measured by 9 different antibody-epitope escape scores and by 4 NTD neutralization-impacting features; and decreased (p = 0.011) with neutralization resistance level to vaccine recipient sera. VE against severe-critical COVID-19 was stable across most sequence features but lower against viruses with greatest distances. These results help map antigenic specificity of in vivo vaccine protection., Competing Interests: Competing interests: ALG reports contract testing from Abbott, Cepheid, Novavax, Pfizer, Janssen, and Hologic and research support from Gilead and Merck. JS declares support for the submitted work from the Janssen Pharmaceutical Companies of Johnson & Johnson and partial support (in the form of funding to his institution) from BARDA for the submitted work, declares support within the past 36 months from the Janssen Pharmaceutical Companies of Johnson & Johnson and BARDA funding for part of this work, has patents on invention of the Janssen COVID-19 vaccine, and has Johnson & Johnson stock and stock options. SR had partial support from the Department of Health and Human Services BARDA (in the form of contract payments to his institution) for the submitted work, has stock and/or stock options in Johnson & Johnson, and is an employee of Janssen Pharmaceutica NV. AV had partial support from BARDA (in the form of contract payments to her institution) for the submitted work, had all patent rights transferred to Johnson & Johnson, has stock and/or stock options in Johnson & Johnson, and is an employee of Janssen Pharmaceutica NV. DJS had partial support from the Department of Health and Human Services BARDA (in the form of contract payments to his institution) for the submitted work, has stock and/or stock options in Johnson & Johnson, and is an employee of Janssen. MLG had partial support from BARDA (in the form of contract payments to his institution) for the submitted work, has patents on invention of the Janssen COVID-19 vaccine, has shares in Johnson & Johnson, and is an employee of Johnson & Johnson. JV has stock and stock options in Johnson and Johnson and is an employee of Janssen Pharmaceutica NV. CT and IVD both had partial support from BARDA (in the form of contract payments to their institution) for the submitted work, hold stock in Janssen Pharmaceuticals, and are employees of Janssen Pharmaceutica NV. The views expressed by MR, HB and BLD are those of the authors and should not be construed to represent the positions of the U.S. Army, the Department of Defense, or the Department of Health and Human Services.
- Published
- 2023
- Full Text
- View/download PDF
28. Rapid Development of an Integrated Network Infrastructure to Conduct Phase 3 COVID-19 Vaccine Trials.
- Author
-
Mena Lora AJ, Long JE, Huang Y, Baden LR, El Sahly HM, Follmann D, Goepfert P, Gray G, Grinsztejn B, Kotloff K, Rouphael N, Sobieszczyk M, Walsh SR, Andriesen J, Shah KA, Zhang Y, Gilbert P, Janes H, Gay CL, Falsey AR, Tripp RL, Gorman RL, Tong T, Marovich M, Neuzil K, Corey L, and Kublin JG
- Subjects
- Humans, COVID-19 Vaccines, SARS-CoV-2, Pandemics prevention & control, COVID-19, Vaccines
- Abstract
Importance: The COVID-19 pandemic has caused millions of infections and deaths and resulted in unprecedented international public health social and economic crises. As SARS-CoV-2 spread across the globe and its impact became evident, the development of safe and effective vaccines became a priority. Outlining the processes used to establish and support the conduct of the phase 3 randomized clinical trials that led to the rapid emergency use authorization and approval of several COVID-19 vaccines is of major significance for current and future pandemic response efforts., Observations: To support the rapid development of vaccines for the US population and the rest of the world, the National Institute of Allergy and Infectious Diseases established the COVID-19 Prevention Network (CoVPN) to assist in the coordination and implementation of phase 3 efficacy trials for COVID-19 vaccine candidates and monoclonal antibodies. By bringing together multiple networks, CoVPN was able to draw on existing clinical and laboratory infrastructure, community partnerships, and research expertise to quickly pivot clinical trial sites to conduct COVID-19 vaccine trials as soon as the investigational products were ready for phase 3 testing. The mission of CoVPN was to operationalize phase 3 vaccine trials using harmonized protocols, laboratory assays, and a single data and safety monitoring board to oversee the various studies. These trials, while staggered in time of initiation, overlapped in time and course of conduct and ultimately led to the successful completion of multiple studies and US Food and Drug Administration-licensed or -authorized vaccines, the first of which was available to the public less than 1 year from the discovery of the virus., Conclusions and Relevance: This Special Communication describes the design, geographic distribution, and underlying principles of conduct of these efficacy trials and summarizes data from 136 382 prospectively followed-up participants, including more than 2500 with documented COVID-19. These successful efforts can be replicated for other important research initiatives and point to the importance of investments in clinical trial infrastructure integral to pandemic preparedness.
- Published
- 2023
- Full Text
- View/download PDF
29. Forty years of investment in HIV research: progress towards ending the HIV pandemic and preparation for future pandemics.
- Author
-
Read SW, Kim P, Marovich M, Dieffenbach CW, and Fauci AS
- Subjects
- Humans, Pandemics prevention & control, HIV Infections drug therapy, HIV Infections epidemiology, HIV Infections prevention & control
- Published
- 2022
- Full Text
- View/download PDF
30. The anti-SARS-CoV-2 monoclonal antibody bamlanivimab minimally affects the endogenous immune response to COVID-19 vaccination.
- Author
-
Benschop RJ, Tuttle JL, Zhang L, Poorbaugh J, Kallewaard NL, Vaillancourt P, Crisp M, Trinh TNV, Freitas JJ, Beasley S, Daniels M, Haustrup N, Higgs RE, Nirula A, Cohen MS, and Marovich M
- Subjects
- Antibodies, Monoclonal, Humanized therapeutic use, Antibodies, Neutralizing, Antibodies, Viral, Antibody Formation, COVID-19 Vaccines, Humans, SARS-CoV-2, Vaccination, COVID-19, Viral Vaccines
- Abstract
As the coronavirus disease 2019 (COVID-19) pandemic evolves and vaccine rollout progresses, the availability and demand for monoclonal antibodies for the prevention and treatment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are also accelerating. This longitudinal serological study evaluated the magnitude and potency of the endogenous antibody response to COVID-19 vaccination in participants who first received a COVID-19 monoclonal antibody in a prevention study. Over the course of 6 months, serum samples were collected from a population of nursing home residents and staff enrolled in a clinical trial who were randomized to either bamlanivimab treatment or placebo. In an unplanned component of this trial, a subset of these participants was subsequently fully vaccinated with two doses of either SpikeVax (Moderna) or Comirnaty (BioNTech/Pfizer) COVID-19 mRNA vaccines. This post hoc analysis assessed the immune response to vaccination for 135 participants without prior SARS-CoV-2 infection. Antibody titers and potency were assessed using three assays against SARS-CoV-2 proteins that bamlanivimab does not efficiently bind to, thereby reflecting the endogenous antibody response. All bamlanivimab and placebo recipients mounted a robust immune response to full COVID-19 vaccination, irrespective of age, risk category, and vaccine type with any observed differences of uncertain clinical importance. These findings are pertinent for informing public health policy with results that suggest that the benefit of receiving COVID-19 vaccination at the earliest opportunity outweighs the minimal effect on the endogenous immune response due to prior prophylactic COVID-19 monoclonal antibody infusion.
- Published
- 2022
- Full Text
- View/download PDF
31. Efficacy of the mRNA-1273 SARS-CoV-2 Vaccine at Completion of Blinded Phase.
- Author
-
El Sahly HM, Baden LR, Essink B, Doblecki-Lewis S, Martin JM, Anderson EJ, Campbell TB, Clark J, Jackson LA, Fichtenbaum CJ, Zervos M, Rankin B, Eder F, Feldman G, Kennelly C, Han-Conrad L, Levin M, Neuzil KM, Corey L, Gilbert P, Janes H, Follmann D, Marovich M, Polakowski L, Mascola JR, Ledgerwood JE, Graham BS, August A, Clouting H, Deng W, Han S, Leav B, Manzo D, Pajon R, Schödel F, Tomassini JE, Zhou H, and Miller J
- Subjects
- 2019-nCoV Vaccine mRNA-1273, Adolescent, Adult, Aged, COVID-19 epidemiology, COVID-19 Vaccines adverse effects, Follow-Up Studies, Humans, Immunization, Secondary, Incidence, Intention to Treat Analysis, Male, Middle Aged, Patient Acuity, Single-Blind Method, Treatment Outcome, Young Adult, COVID-19 prevention & control, COVID-19 Vaccines immunology, Immunogenicity, Vaccine
- Abstract
Background: At interim analysis in a phase 3, observer-blinded, placebo-controlled clinical trial, the mRNA-1273 vaccine showed 94.1% efficacy in preventing coronavirus disease 2019 (Covid-19). After emergency use of the vaccine was authorized, the protocol was amended to include an open-label phase. Final analyses of efficacy and safety data from the blinded phase of the trial are reported., Methods: We enrolled volunteers who were at high risk for Covid-19 or its complications; participants were randomly assigned in a 1:1 ratio to receive two intramuscular injections of mRNA-1273 (100 μg) or placebo, 28 days apart, at 99 centers across the United States. The primary end point was prevention of Covid-19 illness with onset at least 14 days after the second injection in participants who had not previously been infected with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The data cutoff date was March 26, 2021., Results: The trial enrolled 30,415 participants; 15,209 were assigned to receive the mRNA-1273 vaccine, and 15,206 to receive placebo. More than 96% of participants received both injections, 2.3% had evidence of SARS-CoV-2 infection at baseline, and the median follow-up was 5.3 months in the blinded phase. Vaccine efficacy in preventing Covid-19 illness was 93.2% (95% confidence interval [CI], 91.0 to 94.8), with 55 confirmed cases in the mRNA-1273 group (9.6 per 1000 person-years; 95% CI, 7.2 to 12.5) and 744 in the placebo group (136.6 per 1000 person-years; 95% CI, 127.0 to 146.8). The efficacy in preventing severe disease was 98.2% (95% CI, 92.8 to 99.6), with 2 cases in the mRNA-1273 group and 106 in the placebo group, and the efficacy in preventing asymptomatic infection starting 14 days after the second injection was 63.0% (95% CI, 56.6 to 68.5), with 214 cases in the mRNA-1273 group and 498 in the placebo group. Vaccine efficacy was consistent across ethnic and racial groups, age groups, and participants with coexisting conditions. No safety concerns were identified., Conclusions: The mRNA-1273 vaccine continued to be efficacious in preventing Covid-19 illness and severe disease at more than 5 months, with an acceptable safety profile, and protection against asymptomatic infection was observed. (Funded by the Biomedical Advanced Research and Development Authority and the National Institute of Allergy and Infectious Diseases; COVE ClinicalTrials.gov number, NCT04470427.)., (Copyright © 2021 Massachusetts Medical Society.)
- Published
- 2021
- Full Text
- View/download PDF
32. Effect of Bamlanivimab vs Placebo on Incidence of COVID-19 Among Residents and Staff of Skilled Nursing and Assisted Living Facilities: A Randomized Clinical Trial.
- Author
-
Cohen MS, Nirula A, Mulligan MJ, Novak RM, Marovich M, Yen C, Stemer A, Mayer SM, Wohl D, Brengle B, Montague BT, Frank I, McCulloh RJ, Fichtenbaum CJ, Lipson B, Gabra N, Ramirez JA, Thai C, Chege W, Gomez Lorenzo MM, Sista N, Farrior J, Clement ME, Brown ER, Custer KL, Van Naarden J, Adams AC, Schade AE, Dabora MC, Knorr J, Price KL, Sabo J, Tuttle JL, Klekotka P, Shen L, and Skovronsky DM
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal, Humanized adverse effects, Antibodies, Monoclonal, Humanized immunology, Antiviral Agents adverse effects, Antiviral Agents immunology, Assisted Living Facilities, COVID-19 epidemiology, Double-Blind Method, Drug Approval, Female, Health Personnel, Humans, Immunization, Passive, Incidence, Infusions, Intravenous, Male, Middle Aged, SARS-CoV-2 isolation & purification, Severity of Illness Index, Skilled Nursing Facilities, Young Adult, Antibodies, Monoclonal, Humanized therapeutic use, Antibodies, Neutralizing therapeutic use, Antiviral Agents therapeutic use, COVID-19 prevention & control
- Abstract
Importance: Preventive interventions are needed to protect residents and staff of skilled nursing and assisted living facilities from COVID-19 during outbreaks in their facilities. Bamlanivimab, a neutralizing monoclonal antibody against SARS-CoV-2, may confer rapid protection from SARS-CoV-2 infection and COVID-19., Objective: To determine the effect of bamlanivimab on the incidence of COVID-19 among residents and staff of skilled nursing and assisted living facilities., Design, Setting, and Participants: Randomized, double-blind, single-dose, phase 3 trial that enrolled residents and staff of 74 skilled nursing and assisted living facilities in the United States with at least 1 confirmed SARS-CoV-2 index case. A total of 1175 participants enrolled in the study from August 2 to November 20, 2020. Database lock was triggered on January 13, 2021, when all participants reached study day 57., Interventions: Participants were randomized to receive a single intravenous infusion of bamlanivimab, 4200 mg (n = 588), or placebo (n = 587)., Main Outcomes and Measures: The primary outcome was incidence of COVID-19, defined as the detection of SARS-CoV-2 by reverse transcriptase-polymerase chain reaction and mild or worse disease severity within 21 days of detection, within 8 weeks of randomization. Key secondary outcomes included incidence of moderate or worse COVID-19 severity and incidence of SARS-CoV-2 infection., Results: The prevention population comprised a total of 966 participants (666 staff and 300 residents) who were negative at baseline for SARS-CoV-2 infection and serology (mean age, 53.0 [range, 18-104] years; 722 [74.7%] women). Bamlanivimab significantly reduced the incidence of COVID-19 in the prevention population compared with placebo (8.5% vs 15.2%; odds ratio, 0.43 [95% CI, 0.28-0.68]; P < .001; absolute risk difference, -6.6 [95% CI, -10.7 to -2.6] percentage points). Five deaths attributed to COVID-19 were reported by day 57; all occurred in the placebo group. Among 1175 participants who received study product (safety population), the rate of participants with adverse events was 20.1% in the bamlanivimab group and 18.9% in the placebo group. The most common adverse events were urinary tract infection (reported by 12 participants [2%] who received bamlanivimab and 14 [2.4%] who received placebo) and hypertension (reported by 7 participants [1.2%] who received bamlanivimab and 10 [1.7%] who received placebo)., Conclusions and Relevance: Among residents and staff in skilled nursing and assisted living facilities, treatment during August-November 2020 with bamlanivimab monotherapy reduced the incidence of COVID-19 infection. Further research is needed to assess preventive efficacy with current patterns of viral strains with combination monoclonal antibody therapy., Trial Registration: ClinicalTrials.gov Identifier: NCT04497987.
- Published
- 2021
- Full Text
- View/download PDF
33. Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine.
- Author
-
Baden LR, El Sahly HM, Essink B, Kotloff K, Frey S, Novak R, Diemert D, Spector SA, Rouphael N, Creech CB, McGettigan J, Khetan S, Segall N, Solis J, Brosz A, Fierro C, Schwartz H, Neuzil K, Corey L, Gilbert P, Janes H, Follmann D, Marovich M, Mascola J, Polakowski L, Ledgerwood J, Graham BS, Bennett H, Pajon R, Knightly C, Leav B, Deng W, Zhou H, Han S, Ivarsson M, Miller J, and Zaks T
- Subjects
- 2019-nCoV Vaccine mRNA-1273, Adolescent, Adult, Aged, COVID-19 diagnosis, COVID-19 immunology, Female, Humans, Incidence, Male, Middle Aged, Patient Acuity, Single-Blind Method, Spike Glycoprotein, Coronavirus, Treatment Outcome, Young Adult, COVID-19 prevention & control, COVID-19 Vaccines adverse effects, COVID-19 Vaccines immunology, SARS-CoV-2
- Abstract
Background: Vaccines are needed to prevent coronavirus disease 2019 (Covid-19) and to protect persons who are at high risk for complications. The mRNA-1273 vaccine is a lipid nanoparticle-encapsulated mRNA-based vaccine that encodes the prefusion stabilized full-length spike protein of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes Covid-19., Methods: This phase 3 randomized, observer-blinded, placebo-controlled trial was conducted at 99 centers across the United States. Persons at high risk for SARS-CoV-2 infection or its complications were randomly assigned in a 1:1 ratio to receive two intramuscular injections of mRNA-1273 (100 μg) or placebo 28 days apart. The primary end point was prevention of Covid-19 illness with onset at least 14 days after the second injection in participants who had not previously been infected with SARS-CoV-2., Results: The trial enrolled 30,420 volunteers who were randomly assigned in a 1:1 ratio to receive either vaccine or placebo (15,210 participants in each group). More than 96% of participants received both injections, and 2.2% had evidence (serologic, virologic, or both) of SARS-CoV-2 infection at baseline. Symptomatic Covid-19 illness was confirmed in 185 participants in the placebo group (56.5 per 1000 person-years; 95% confidence interval [CI], 48.7 to 65.3) and in 11 participants in the mRNA-1273 group (3.3 per 1000 person-years; 95% CI, 1.7 to 6.0); vaccine efficacy was 94.1% (95% CI, 89.3 to 96.8%; P<0.001). Efficacy was similar across key secondary analyses, including assessment 14 days after the first dose, analyses that included participants who had evidence of SARS-CoV-2 infection at baseline, and analyses in participants 65 years of age or older. Severe Covid-19 occurred in 30 participants, with one fatality; all 30 were in the placebo group. Moderate, transient reactogenicity after vaccination occurred more frequently in the mRNA-1273 group. Serious adverse events were rare, and the incidence was similar in the two groups., Conclusions: The mRNA-1273 vaccine showed 94.1% efficacy at preventing Covid-19 illness, including severe disease. Aside from transient local and systemic reactions, no safety concerns were identified. (Funded by the Biomedical Advanced Research and Development Authority and the National Institute of Allergy and Infectious Diseases; COVE ClinicalTrials.gov number, NCT04470427.)., (Copyright © 2020 Massachusetts Medical Society.)
- Published
- 2021
- Full Text
- View/download PDF
34. Monoclonal Antibodies for Prevention and Treatment of COVID-19.
- Author
-
Marovich M, Mascola JR, and Cohen MS
- Subjects
- Antibodies, Neutralizing therapeutic use, Antibodies, Viral therapeutic use, Betacoronavirus, COVID-19, Humans, SARS-CoV-2, Spike Glycoprotein, Coronavirus immunology, Antibodies, Monoclonal therapeutic use, Coronavirus Infections prevention & control, Coronavirus Infections therapy, Pandemics prevention & control, Pneumonia, Viral prevention & control, Pneumonia, Viral therapy
- Published
- 2020
- Full Text
- View/download PDF
35. The complex challenges of HIV vaccine development require renewed and expanded global commitment.
- Author
-
Bekker LG, Tatoud R, Dabis F, Feinberg M, Kaleebu P, Marovich M, Ndung'u T, Russell N, Johnson J, Luba M, Fauci AS, Morris L, Pantaleo G, Buchbinder S, Gray G, Vekemans J, Kim JH, Levy Y, Corey L, Shattock R, Makanga M, Williamson C, Dieffenbach C, Goodenow MM, Shao Y, Staprans S, Warren M, and Johnston MI
- Subjects
- Drug Development methods, Epidemics prevention & control, Health Services Accessibility organization & administration, Humans, International Cooperation, Marketing of Health Services, AIDS Vaccines, HIV Infections prevention & control
- Published
- 2020
- Full Text
- View/download PDF
36. Immunological mechanisms of inducing HIV immunity in infants.
- Author
-
Fouda GG, De Paris K, Levy O, Marchant A, Gray G, Permar S, Marovich M, and Singh A
- Subjects
- AIDS Vaccines administration & dosage, AIDS Vaccines blood, Antibodies, Neutralizing blood, Education methods, HIV Antibodies blood, HIV Infections blood, HIV Infections prevention & control, HIV-1 drug effects, HIV-1 immunology, Humans, Immunization methods, Immunization trends, Infant, Infant, Newborn, AIDS Vaccines immunology, Antibodies, Neutralizing immunology, Education trends, HIV Antibodies immunology, HIV Infections immunology
- Abstract
The potential advantages and unique challenges of the early life immune system for the development of HIV-specific broadly neutralizing antibodies were discussed during a workshop entitled "Immunological Mechanisms of Inducing HIV Immunity in Infants" sponsored by the National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH) in conjunction with the 2018 HIVR4P Conference held in Madrid, Spain. A safe and effective HIV vaccine remains a critical need in the fight against the HIV pandemic, especially to prevent emerging infections in infants, adolescents, and young adults. To successfully target these populations, a vaccine should ideally induce protective immune responses during childhood. Interestingly, several recent studies highlighting differences in immune responses between adults and children have suggested that the early life immune system could present advantages for the elicitation of broadly neutralizing antibodies (bnAbs), a response highly desired for an HIV vaccine. Notably, HIV-infected children develop bnAbs responses earlier and more frequently than infected adults; with emerging evidence that the pathways of elicitation of bnAb lineages may differ between adults and children. Moreover, there is precedent for the prevention of lifelong infections with pediatric immunization, and early life provides a unique window of opportunity for the administration of a multi-dose HIV vaccine that will likely be needed to achieve protective immunity. Further understanding of how the distinct early life immune system can be harnessed to trigger bnAb lineages for induction of durable and polyfunctional HIV-specific immunity is warranted. This strategy will include testing promising HIV vaccine candidates in pediatric populations in preclinical and clinical studies. Novel approaches to identify molecular markers of protection are also key to guide and accelerate pediatric HIV vaccine development., (Copyright © 2019.)
- Published
- 2020
- Full Text
- View/download PDF
37. AIDS Vaccine Research Subcommittee (AVRS) Consultation: Early-Life Immunization Strategies against HIV Acquisition.
- Author
-
Singh A, Permar S, Kollmann TR, Levy O, Marovich M, and De Paris K
- Subjects
- Adjuvants, Immunologic, Adolescent, Child, Clinical Trials as Topic, Congresses as Topic, Female, HIV Infections immunology, HIV-1 immunology, Humans, Infant, Referral and Consultation, United States, AIDS Vaccines, HIV Infections prevention & control, Immunization Schedule
- Abstract
This report summarizes a consultation meeting convened by the National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), on 12 September 2017 to discuss the scientific rationale for selectively testing relevant HIV vaccine candidates in early life that are designed to initiate immune responses for lifelong protective immunity. The urgent need to develop interventions providing durable protective immunity to HIV before sexual debut coupled with the practicality of infant vaccine schedules supports optimizing infant HIV vaccines as a high priority. The panelists discussed the unique opportunities and challenges of testing candidate HIV vaccines in the context of distinct early-life immunity. Key developments providing rationale and grounds for cautious optimism regarding evaluation of early-life HIV vaccines include recent studies of early-life immune ontogeny, studies of HIV-infected infants demonstrating relatively rapid generation of broadly neutralizing antibodies (bNAbs), discovery of novel adjuvants active in early life, and cutting-edge sample-sparing systems biology and immunologic assays promising deep insight into vaccine action in infants. Multidisciplinary efforts toward the goal of an infant HIV vaccine are under way and should be nurtured and amplified. IMPORTANCE Young adults represent one of the highest-risk groups for new HIV infections and the only group in which morbidity continues to increase. Therefore, an HIV vaccine to prevent HIV acquisition in adolescence is a top priority. The introduction of any vaccine during adolescence is challenging. This meeting discussed the opportunities and challenges of testing HIV vaccine candidates in the context of the infant immune system given recent advances in our knowledge of immune ontogeny and adjuvant design and studies demonstrating that HIV-infected infants generate broadly neutralizing antibodies, a main target of HIV vaccines, more rapidly than adults. Considering the global success of pediatric vaccines, the concept of an HIV vaccine introduced in early life holds merit and warrants testing., (Copyright © 2019 Singh et al.)
- Published
- 2019
- Full Text
- View/download PDF
38. Modulation of Vaccine-Induced CD4 T Cell Functional Profiles by Changes in Components of HIV Vaccine Regimens in Humans.
- Author
-
Pissani F, Schulte B, Eller MA, Schultz BT, Ratto-Kim S, Marovich M, Thongcharoen P, Sriplienchan S, Rerks-Ngarm S, Pitisuttithum P, Esser S, Alter G, Robb ML, Kim JH, Michael NL, and Streeck H
- Subjects
- AIDS Vaccines genetics, Antibodies, Neutralizing immunology, CD8-Positive T-Lymphocytes drug effects, CD8-Positive T-Lymphocytes virology, Cells, Cultured, HIV Infections immunology, HIV Infections virology, HIV-1 drug effects, HIV-1 genetics, Humans, Vaccination, AIDS Vaccines classification, AIDS Vaccines immunology, CD8-Positive T-Lymphocytes immunology, HIV Antibodies immunology, HIV Envelope Protein gp120 immunology, HIV Infections prevention & control, HIV-1 immunology
- Abstract
To date, six vaccine strategies have been evaluated in clinical trials for their efficacy at inducing protective immune responses against HIV infection. However, only the ALVAC-HIV/AIDSVAX B/E vaccine (RV144 trial) has demonstrated protection, albeit modestly (31%; P = 0.03). One potential correlate of protection was a low-frequency HIV-specific CD4 T cell population with diverse functionality. Although CD4 T cells, particularly T follicular helper (Tfh) cells, are critical for effective antibody responses, most studies involving HIV vaccines have focused on humoral immunity or CD8 T cell effector responses, and little is known about the functionality and frequency of vaccine-induced CD4 T cells. We therefore assessed responses from several phase I/II clinical trials and compared them to responses to natural HIV-1 infection. We found that all vaccines induced a lower magnitude of HIV-specific CD4 T cell responses than that observed for chronic infection. Responses differed in functionality, with a CD40 ligand (CD40L)-dominated response and more Tfh cells after vaccination, whereas chronic HIV infection provoked tumor necrosis factor alpha (TNF-α)-dominated responses. The vaccine delivery route further impacted CD4 T cells, showing a stronger Th1 polarization after dendritic cell delivery than after intramuscular vaccination. In prime/boost regimens, the choice of prime and boost influenced the functional profile of CD4 T cells to induce more or less polyfunctionality. In summary, vaccine-induced CD4 T cell responses differ remarkably between vaccination strategies, modes of delivery, and boosts and do not resemble those induced by chronic HIV infection. Understanding the functional profiles of CD4 T cells that best facilitate protective antibody responses will be critical if CD4 T cell responses are to be considered a clinical trial go/no-go criterion. IMPORTANCE Only one HIV-1 candidate vaccine strategy has shown protection, albeit marginally (31%), against HIV-1 acquisition, and correlates of protection suggested that a multifunctional CD4 T cell immune response may be important for this protective effect. Therefore, the functional phenotypes of HIV-specific CD4 T cell responses induced by different phase I and phase II clinical trials were assessed to better show how different vaccine strategies influence the phenotype and function of HIV-specific CD4 T cell immune responses. The significance of this research lies in our comprehensive comparison of the compositions of the T cell immune responses to different HIV vaccine modalities. Specifically, our work allows for the evaluation of vaccination strategies in terms of their success at inducing Tfh cell populations., (Copyright © 2018 American Society for Microbiology.)
- Published
- 2018
- Full Text
- View/download PDF
39. Safety and Immunogenicity of PENNVAX-G DNA Prime Administered by Biojector 2000 or CELLECTRA Electroporation Device With Modified Vaccinia Ankara-CMDR Boost.
- Author
-
Ake JA, Schuetz A, Pegu P, Wieczorek L, Eller MA, Kibuuka H, Sawe F, Maboko L, Polonis V, Karasavva N, Weiner D, Sekiziyivu A, Kosgei J, Missanga M, Kroidl A, Mann P, Ratto-Kim S, Anne Eller L, Earl P, Moss B, Dorsey-Spitz J, Milazzo M, Laissa Ouedraogo G, Rizvi F, Yan J, Khan AS, Peel S, Sardesai NY, Michael NL, Ngauy V, Marovich M, and Robb ML
- Subjects
- Adult, Africa, Eastern, Double-Blind Method, Electroporation, Female, Humans, Male, Middle Aged, United States, Vaccination, AIDS Vaccines administration & dosage, AIDS Vaccines immunology, HIV Antibodies blood, HIV Infections prevention & control, Immunity, Cellular drug effects, Vaccinia virus immunology
- Abstract
Background: We report the first-in-human safety and immunogenicity evaluation of PENNVAX-G DNA/modified vaccinia Ankara-Chiang Mai double recombinant (MVA-CMDR) prime-boost human immuonodeficiency virus (HIV) vaccine, with intramuscular DNA delivery by either Biojector 2000 needle-free injection system (Biojector) or CELLECTRA electroporation device., Methods: Healthy, HIV-uninfected adults were randomized to receive 4 mg of PENNVAX-G DNA delivered intramuscularly by Biojector or electroporation at baseline and week 4 followed by intramuscular injection of 108 plaque forming units of MVA-CMDR at weeks 12 and 24. The open-label part A was conducted in the United States, followed by a double-blind, placebo-controlled part B in East Africa. Solicited and unsolicited adverse events were recorded, and immune responses were measured., Results: Eighty-eight of 100 enrolled participants completed all study injections, which were generally safe and well tolerated, with more immediate, but transient, pain in the electroporation group. Cellular responses were observed in 57% of vaccine recipients tested and were CD4 predominant. High rates of binding antibody responses to CRF01_AE antigens, including gp70 V1V2 scaffold, were observed. Neutralizing antibodies were detected in a peripheral blood mononuclear cell assay, and moderate antibody-dependent, cell-mediated cytotoxicity activity was demonstrated., Discussion: The PVG/MVA-CMDR HIV-1 vaccine regimen is safe and immunogenic. Substantial differences in safety or immunogenicity between modes of DNA delivery were not observed., Clinical Trials Registration: NCT01260727., (Published by Oxford University Press for the Infectious Diseases Society of America 2017. This work is written by (a) US Government employee(s) and is in the public domain in the US.)
- Published
- 2017
- Full Text
- View/download PDF
40. Differential Inhibitory Receptor Expression on T Cells Delineates Functional Capacities in Chronic Viral Infection.
- Author
-
Teigler JE, Zelinskyy G, Eller MA, Bolton DL, Marovich M, Gordon AD, Alrubayyi A, Alter G, Robb ML, Martin JN, Deeks SG, Michael NL, Dittmer U, and Streeck H
- Subjects
- Animals, Antibodies administration & dosage, Antibodies pharmacology, CD4-Positive T-Lymphocytes drug effects, CD4-Positive T-Lymphocytes virology, CTLA-4 Antigen antagonists & inhibitors, CTLA-4 Antigen genetics, CTLA-4 Antigen immunology, Costimulatory and Inhibitory T-Cell Receptors drug effects, Cytokines biosynthesis, Cytokines drug effects, Enterotoxins pharmacology, Friend murine leukemia virus physiology, HIV Infections virology, Humans, Interferon-gamma biosynthesis, Mice, Programmed Cell Death 1 Receptor antagonists & inhibitors, Programmed Cell Death 1 Receptor genetics, Programmed Cell Death 1 Receptor immunology, Retroviridae Infections immunology, Tumor Necrosis Factor-alpha biosynthesis, CD4-Positive T-Lymphocytes immunology, Costimulatory and Inhibitory T-Cell Receptors antagonists & inhibitors, Costimulatory and Inhibitory T-Cell Receptors genetics, Gene Expression, HIV Infections immunology
- Abstract
Inhibitory receptors have been extensively described for their importance in regulating immune responses in chronic infections and cancers. Blocking the function of inhibitory receptors such as PD-1, CTLA-4, 2B4, Tim-3, and LAG-3 has shown promise for augmenting CD8 T cell activity and boosting pathogen-specific immunity. However, the prevalence of inhibitory receptors on CD4 T cells and their relative influence on CD4 T cell functionality in chronic HIV infection remains poorly described. We therefore determined and compared inhibitory receptor expression patterns of 2B4, CTLA-4, LAG-3, PD-1, and Tim-3 on virus-specific CD4 and CD8 T cells in relation to their functional T cell profile. In chronic HIV infection, inhibitory receptor distribution differed markedly between cytokine-producing T cell subsets with, gamma interferon (IFN-γ)- and tumor necrosis factor alpha (TNF-α)-producing cells displaying the highest and lowest prevalence of inhibitory receptors, respectively. Blockade of inhibitory receptors differentially affected cytokine production by cells in response to staphylococcal enterotoxin B stimulation. CTLA-4 blockade increased IFN-γ and CD40L production, while PD-1 blockade strongly augmented IFN-γ, interleukin-2 (IL-2), and TNF-α production. In a Friend retrovirus infection model, CTLA-4 blockade in particular was able to improve control of viral replication. Together, these results show that inhibitory receptor distribution on HIV-specific CD4 T cells varies markedly with respect to the functional subset of CD4 T cells being analyzed. Furthermore, the differential effects of receptor blockade suggest novel methods of immune response modulation, which could be important in the context of HIV vaccination or therapeutic strategies. IMPORTANCE Inhibitory receptors are important for limiting damage by the immune system during acute infections. In chronic infections, however, their expression limits immune system responsiveness. Studies have shown that blocking inhibitory receptors augments CD8 T cell functionality in HIV infection, but their influence on CD4 T cells remains unclear. We assessed the expression of inhibitory receptors on HIV-specific CD4 T cells and their relationship with T cell functionality. We uncovered differences in inhibitory receptor expression depending on the CD4 T cell function. We also found differences in functionality of CD4 T cells following blocking of different inhibitory receptors, and we confirmed our results in a Friend virus retroviral model of infection in mice. Our results show that inhibitory receptor expression on CD4 T cells is linked to CD4 T cell functionality and could be sculpted by blockade of specific inhibitory receptors. These data reveal exciting possibilities for the development of novel treatments and immunotherapeutics., (Copyright © 2017 American Society for Microbiology.)
- Published
- 2017
- Full Text
- View/download PDF
41. Preferential Targeting of Conserved Gag Regions after Vaccination with a Heterologous DNA Prime-Modified Vaccinia Virus Ankara Boost HIV-1 Vaccine Regimen.
- Author
-
Bauer A, Podola L, Mann P, Missanga M, Haule A, Sudi L, Nilsson C, Kaluwa B, Lueer C, Mwakatima M, Munseri PJ, Maboko L, Robb ML, Tovanabutra S, Kijak G, Marovich M, McCormack S, Joseph S, Lyamuya E, Wahren B, Sandström E, Biberfeld G, Hoelscher M, Bakari M, Kroidl A, and Geldmacher C
- Subjects
- AIDS Vaccines administration & dosage, Drug Carriers, Healthy Volunteers, Humans, Interferon-gamma metabolism, Interleukin-2 metabolism, T-Lymphocyte Subsets immunology, Tanzania, Tumor Necrosis Factor-alpha metabolism, Vaccines, DNA administration & dosage, Vaccines, Subunit administration & dosage, Vaccines, Subunit immunology, Vaccines, Synthetic administration & dosage, Vaccines, Synthetic immunology, Vaccinia virus genetics, AIDS Vaccines immunology, HIV-1 immunology, T-Lymphocytes immunology, Vaccination methods, Vaccines, DNA immunology, gag Gene Products, Human Immunodeficiency Virus immunology
- Abstract
Prime-boost vaccination strategies against HIV-1 often include multiple variants for a given immunogen for better coverage of the extensive viral diversity. To study the immunologic effects of this approach, we characterized breadth, phenotype, function, and specificity of Gag-specific T cells induced by a DNA-prime modified vaccinia virus Ankara (MVA)-boost vaccination strategy, which uses mismatched Gag immunogens in the TamoVac 01 phase IIa trial. Healthy Tanzanian volunteers received three injections of the DNA-SMI vaccine encoding a subtype B and AB-recombinant Gag
p37 and two vaccinations with MVA-CMDR encoding subtype A Gagp55 Gag-specific T-cell responses were studied in 42 vaccinees using fresh peripheral blood mononuclear cells. After the first MVA-CMDR boost, vaccine-induced gamma interferon-positive (IFN-γ+ ) Gag-specific T-cell responses were dominated by CD4+ T cells ( P < 0.001 compared to CD8+ T cells) that coexpressed interleukin-2 (IL-2) (66.4%) and/or tumor necrosis factor alpha (TNF-α) (63.7%). A median of 3 antigenic regions were targeted with a higher-magnitude median response to Gagp24 regions, more conserved between prime and boost, compared to those of regions within Gagp15 (not primed) and Gagp17 (less conserved; P < 0.0001 for both). Four regions within Gagp24 each were targeted by 45% to 74% of vaccinees upon restimulation with DNA-SMI-Gag matched peptides. The response rate to individual antigenic regions correlated with the sequence homology between the MVA- and DNA Gag-encoded immunogens ( P = 0.04, r2 = 0.47). In summary, after the first MVA-CMDR boost, the sequence-mismatched DNA-prime MVA-boost vaccine strategy induced a Gag-specific T-cell response that was dominated by polyfunctional CD4+ T cells and that targeted multiple antigenic regions within the conserved Gagp24 protein. IMPORTANCE Genetic diversity is a major challenge for the design of vaccines against variable viruses. While including multiple variants for a given immunogen in prime-boost vaccination strategies is one approach that aims to improve coverage for global virus variants, the immunologic consequences of this strategy have been poorly defined so far. It is unclear whether inclusion of multiple variants in prime-boost vaccination strategies improves recognition of variant viruses by T cells and by which mechanisms this would be achieved, either by improved cross-recognition of multiple variants for a given antigenic region or through preferential targeting of antigenic regions more conserved between prime and boost. Engineering vaccines to induce adaptive immune responses that preferentially target conserved antigenic regions of viral vulnerability might facilitate better immune control after preventive and therapeutic vaccination for HIV and for other variable viruses., (Copyright © 2017 American Society for Microbiology.)- Published
- 2017
- Full Text
- View/download PDF
42. Preservation of Peripheral T Follicular Helper Cell Function in HIV Controllers.
- Author
-
Buranapraditkun S, Pissani F, Teigler JE, Schultz BT, Alter G, Marovich M, Robb ML, Eller MA, Martin J, Deeks S, Michael NL, and Streeck H
- Subjects
- Adult, CD4-Positive T-Lymphocytes chemistry, CD4-Positive T-Lymphocytes immunology, Female, Humans, Immunophenotyping, Interleukins metabolism, Male, Middle Aged, Receptors, CXCR5 analysis, B-Lymphocytes immunology, Germinal Center immunology, HIV Envelope Protein gp120 immunology, HIV Infections immunology, HIV Long-Term Survivors, T-Lymphocytes, Helper-Inducer immunology
- Abstract
The maturation process of high-affinity antibodies is a result of intricate interactions between B cells and follicular helper T (Tfh) cells occurring in lymphoid germinal centers. HIV infection induces significant chronic immune activation, phenotypic skewing, and inflammation driven by years of continuous viral replication. High levels of viremia as well as immune activation and dysfunction have been demonstrated to have a perturbing impact on the B cell memory compartment and contribute to B cell exhaustion. Counterintuitively, the factors associated with perturbation of the B cell compartment seem to be favorable for the generation of highly affinity-matured Env-specific antibodies in a minority of HIV-infected individuals. Thus, the impact of HIV antigenemia on B cells and Tfh cell interactions warrants further exploration. We therefore studied immunophenotypes of HIV-specific B cells in individuals with differing levels of viral control using HIV Env gp120 probes and characterized the functionality of matched T cells in peripheral blood. While CXCR5
+ CD4+ T cells were significantly diminished in HIV progressors, we found that a small subset of gp120-specific interleukin-21 (IL-21)-secreting CXCR5+ CD4+ T cells were significantly associated with gp120-specific B cell frequencies. In contrast, neither bulk CXCR5+ CD4+ T cells nor other HIV antigen specificities were associated with gp120-specific B cell levels. HIV-specific B cells derived from elite controllers displayed greater amounts of gp120-specific B cells in the resting memory subset, whereas HIV-specific B cells in progressors accumulated in tissue-like and activated memory subsets. Furthermore, CXCR5+ CD4+ T cells from elite controllers showed a stronger ex vivo capacity to induce B cell maturation and immunoglobulin class switching than cells from HIV progressors. IMPORTANCE Dissecting the factors that are involved in B cell maturation and antibody development is important for HIV vaccine design. In this study, we found that HIV Env-specific CXCR5+ CD4+ T cells that secrete interleukin-21 are strongly associated with B cell memory phenotypes and function. Moreover, we found that the immune responses of HIV controllers showed intrinsically better helper activity than those of HIV progressors., (Copyright © 2017 American Society for Microbiology.)- Published
- 2017
- Full Text
- View/download PDF
43. Humoral Immunity to Primary Smallpox Vaccination: Impact of Childhood versus Adult Immunization on Vaccinia Vector Vaccine Development in Military Populations.
- Author
-
Slike BM, Creegan M, Marovich M, and Ngauy V
- Subjects
- Adult, Child, Dendritic Cells immunology, Enzyme-Linked Immunosorbent Assay, Green Fluorescent Proteins metabolism, HIV genetics, Humans, Immunization, Neutralization Tests, Transgenes, Immunity, Humoral, Military Personnel, Smallpox immunology, Smallpox Vaccine immunology, Vaccination, Vaccinia virus immunology
- Abstract
Modified Vaccinia virus has been shown to be a safe and immunogenic vector platform for delivery of HIV vaccines. Use of this vector is of particular importance to the military, with the implementation of a large scale smallpox vaccination campaign in 2002 in active duty and key civilian personnel in response to potential bioterrorist activities. Humoral immunity to smallpox vaccination was previously shown to be long lasting (up to 75 years) and protective. However, using vaccinia-vectored vaccine delivery for other diseases on a background of anti-vector antibodies (i.e. pre-existing immunity) may limit their use as a vaccine platform, especially in the military. In this pilot study, we examined the durability of vaccinia antibody responses in adult primary vaccinees in a healthy military population using a standard ELISA assay and a novel dendritic cell neutralization assay. We found binding and neutralizing antibody (NAb) responses to vaccinia waned after 5-10 years in a group of 475 active duty military, born after 1972, who were vaccinated as adults with Dryvax®. These responses decreased from a geometric mean titer (GMT) of 250 to baseline (<20) after 10-20 years post vaccination. This contrasted with a comparator group of adults, ages 35-49, who were vaccinated with Dryvax® as children. In the childhood vaccinees, titers persisted for >30 years with a GMT of 210 (range 112-3234). This data suggests limited durability of antibody responses in adult vaccinees compared to those vaccinated in childhood and further that adult vaccinia recipients may benefit similarly from receipt of a vaccinia based vaccine as those who are vaccinia naïve. Our findings may have implications for the smallpox vaccination schedule and support the ongoing development of this promising viral vector in a military vaccination program., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2017
- Full Text
- View/download PDF
44. Circulating HIV-Specific Interleukin-21(+)CD4(+) T Cells Represent Peripheral Tfh Cells with Antigen-Dependent Helper Functions.
- Author
-
Schultz BT, Teigler JE, Pissani F, Oster AF, Kranias G, Alter G, Marovich M, Eller MA, Dittmer U, Robb ML, Kim JH, Michael NL, Bolton D, and Streeck H
- Subjects
- Cell Separation, Enzyme-Linked Immunospot Assay, Flow Cytometry, HIV-1 immunology, Humans, Real-Time Polymerase Chain Reaction, HIV Infections immunology, Interleukins immunology, T-Lymphocyte Subsets immunology, T-Lymphocytes, Helper-Inducer immunology
- Abstract
A central effort in HIV vaccine development is to generate protective broadly neutralizing antibodies, a process dependent on T follicular helper (Tfh) cells. The feasibility of using peripheral blood counterparts of lymph node Tfh cells to assess the immune response and the influence of viral and vaccine antigens on their helper functions remain obscure. We assessed circulating HIV-specific IL-21(+)CD4(+) T cells and showed transcriptional and phenotypic similarities to lymphoid Tfh cells, and hence representing peripheral Tfh (pTfh) cells. pTfh cells were functionally active and B cell helper quality differed depending on antigen specificity. Furthermore, we found higher frequency of pTfh cells in peripheral blood mononuclear cell specimens from the ALVAC+AIDSVAX (RV144) HIV vaccine trial associated with protective antibody responses compared to the non-protective DNA+Ad5 vaccine trial. Together, we identify IL-21(+)CD4(+) T cells as pTfh cells, implicating them as key populations in the generation of vaccine-evoked antibody responses., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
45. Cooperativity of HIV-Specific Cytolytic CD4 T Cells and CD8 T Cells in Control of HIV Viremia.
- Author
-
Johnson S, Eller M, Teigler JE, Maloveste SM, Schultz BT, Soghoian DZ, Lu R, Oster AF, Chenine AL, Alter G, Dittmer U, Marovich M, Robb ML, Michael NL, Bolton D, and Streeck H
- Subjects
- Cells, Cultured, HIV Infections virology, HIV-1 genetics, HIV-1 physiology, Humans, Viral Load, Viremia virology, CD4-Positive T-Lymphocytes immunology, HIV Infections immunology, HIV-1 immunology, T-Lymphocytes, Cytotoxic immunology, Viremia immunology
- Abstract
Unlabelled: CD4+ T cells play a pivotal role in the control of chronic viral infections. Recently, nontraditional CD4+ T cell functions beyond helper effects have been described, and a role for cytolytic CD4+ T cells in the control of HIV infection has been suggested. We define here the transcriptional, phenotypic, and functional profiles of HIV-specific cytolytic CD4+ T cells. Fluidigm BioMark and multiparameter flow cytometric analysis of HIV-specific cytolytic CD4+ T cells revealed a distinct transcriptional signature compared to Th1 CD4+ cells but shared similar features with HIV-specific cytolytic CD8+ T cells. Furthermore, HIV-specific cytolytic CD4+ T cells showed comparable killing activity relative to HIV-specific CD8+ T cells and worked cooperatively in the elimination of virally infected cells. Interestingly, we found that cytolytic CD4+ T cells emerge early during acute HIV infection and tightly follow acute viral load trajectory. This emergence was associated to the early viral set point, suggesting an involvement in early control, in spite of CD4 T cell susceptibility to HIV infection. Our data suggest cytolytic CD4+ T cells as an independent subset distinct from Th1 cells that show combined activity with CD8+ T cells in the long-term control of HIV infection., Importance: The ability of the immune system to control chronic HIV infection is of critical interest to both vaccine design and therapeutic approaches. Much research has focused on the effect of the ability of CD8+ T cells to control the virus, while CD4+ T cells have been overlooked as effectors in HIV control due to the fact that they are preferentially infected. We show here that a subset of HIV-specific CD4+ T cells cooperate in the cytolytic control of HIV replication. Moreover, these cells represent a distinct subset of CD4+ T cells showing significant transcriptional and phenotypic differences compared to HIV-specific Th1 cells but with similarities to CD8+ T cells. These findings are important for our understanding of HIV immunopathology., (Copyright © 2015, American Society for Microbiology. All Rights Reserved.)
- Published
- 2015
- Full Text
- View/download PDF
46. Priming with a simplified intradermal HIV-1 DNA vaccine regimen followed by boosting with recombinant HIV-1 MVA vaccine is safe and immunogenic: a phase IIa randomized clinical trial.
- Author
-
Munseri PJ, Kroidl A, Nilsson C, Joachim A, Geldmacher C, Mann P, Moshiro C, Aboud S, Lyamuya E, Maboko L, Missanga M, Kaluwa B, Mfinanga S, Podola L, Bauer A, Godoy-Ramirez K, Marovich M, Moss B, Hoelscher M, Gotch F, Stöhr W, Stout R, McCormack S, Wahren B, Mhalu F, Robb ML, Biberfeld G, Sandström E, and Bakari M
- Subjects
- Adult, DNA, Viral administration & dosage, Drug-Related Side Effects and Adverse Reactions immunology, Drug-Related Side Effects and Adverse Reactions pathology, Female, HIV Infections immunology, HIV Infections virology, HIV-1 immunology, HIV-1 pathogenicity, Humans, Immunity, Humoral drug effects, Immunity, Humoral immunology, Male, T-Lymphocytes immunology, Tanzania, AIDS Vaccines administration & dosage, HIV Infections prevention & control, Vaccines, DNA administration & dosage, Viral Vaccines administration & dosage
- Abstract
Background: Intradermal priming with HIV-1 DNA plasmids followed by HIV-1MVA boosting induces strong and broad cellular and humoral immune responses. In our previous HIVIS-03 trial, we used 5 injections with 2 pools of HIV-DNA at separate sites for each priming immunization. The present study explores whether HIV-DNA priming can be simplified by reducing the number of DNA injections and administration of combined versus separated plasmid pools., Methods: In this phase IIa, randomized trial, priming was performed using 5 injections of HIV-DNA, 1000 μg total dose, (3 Env and 2 Gag encoding plasmids) compared to two "simplified" regimens of 2 injections of HIV-DNA, 600 μg total dose, of Env- and Gag-encoding plasmid pools with each pool either administered separately or combined. HIV-DNA immunizations were given intradermally at weeks 0, 4, and 12. Boosting was performed intramuscularly with 108 pfu HIV-MVA at weeks 30 and 46., Results: 129 healthy Tanzanian participants were enrolled. There were no differences in adverse events between the groups. The proportion of IFN-γ ELISpot responders to Gag and/or Env peptides after the second HIV-MVA boost did not differ significantly between the groups primed with 2 injections of combined HIV-DNA pools, 2 injections with separated pools, and 5 injections with separated pools (90%, 97% and 97%). There were no significant differences in the magnitude of Gag and/or Env IFN-γ ELISpot responses, in CD4+ and CD8+ T cell responses measured as IFN-γ/IL-2 production by intracellular cytokine staining (ICS) or in response rates and median titers for binding antibodies to Env gp160 between study groups., Conclusions: A simplified intradermal vaccination regimen with 2 injections of a total of 600 μg with combined HIV-DNA plasmids primed cellular responses as efficiently as the standard regimen of 5 injections of a total of 1000 μg with separated plasmid pools after boosting twice with HIV-MVA., Trial Registration: World Health Organization International Clinical Trials Registry Platform PACTR2010050002122368.
- Published
- 2015
- Full Text
- View/download PDF
47. Identification of immunodominant CD4-restricted epitopes co-located with antibody binding sites in individuals vaccinated with ALVAC-HIV and AIDSVAX B/E.
- Author
-
Ratto-Kim S, de Souza MS, Currier JR, Karasavvas N, Sidney J, Rolland M, Valencia-Micolta A, Madnote S, Sette A, Nitayaphan S, Pitisuttuthum P, Kaewkungwal J, Rerks-Ngarm S, O'Connell R, Michael N, Robb ML, Marovich M, and Kim JH
- Subjects
- Adolescent, Adult, Cell Line, Epitope Mapping, Female, Humans, Male, Young Adult, AIDS Vaccines immunology, Binding Sites, Antibody immunology, CD4-Positive T-Lymphocytes immunology, HIV Antibodies immunology, HIV-1 immunology, Immunodominant Epitopes immunology
- Abstract
We performed fine epitope mapping of the CD4+ responses in the ALVAC-HIV-AIDSVAX B/E prime-boost regimen in the Thai Phase III trial (RV144). Non-transformed Env-specific T cell lines established from RV144 vaccinees were used to determine the fine epitope mapping of the V2 and C1 responses and the HLA class II restriction. Data showed that there are two CD4+ epitopes contained within the V2 loop: one encompassing the α4β7 integrin binding site (AA179-181) and the other nested between two previously described genetic sieve signatures (AA169, AA181). There was no correlation between the frequencies of CD4+ fine epitope responses and binding antibody.
- Published
- 2015
- Full Text
- View/download PDF
48. Initiation of ART during early acute HIV infection preserves mucosal Th17 function and reverses HIV-related immune activation.
- Author
-
Schuetz A, Deleage C, Sereti I, Rerknimitr R, Phanuphak N, Phuang-Ngern Y, Estes JD, Sandler NG, Sukhumvittaya S, Marovich M, Jongrakthaitae S, Akapirat S, Fletscher JL, Kroon E, Dewar R, Trichavaroj R, Chomchey N, Douek DC, O Connell RJ, Ngauy V, Robb ML, Phanuphak P, Michael NL, Excler JL, Kim JH, de Souza MS, and Ananworanich J
- Subjects
- Acute Disease, Adult, Anti-Retroviral Agents pharmacology, Biomarkers blood, Biopsy, Colon, Sigmoid pathology, Cytokines blood, Female, HIV Infections pathology, HIV Infections physiopathology, Humans, Immunity, Mucosal drug effects, Intestinal Mucosa pathology, Male, Middle Aged, Prospective Studies, Th17 Cells pathology, Time Factors, Treatment Outcome, Anti-Retroviral Agents therapeutic use, HIV Infections drug therapy, Immunity, Mucosal physiology, Intestinal Mucosa physiology, Th17 Cells physiology
- Abstract
Mucosal Th17 cells play an important role in maintaining gut epithelium integrity and thus prevent microbial translocation. Chronic HIV infection is characterized by mucosal Th17 cell depletion, microbial translocation and subsequent immune-activation, which remain elevated despite antiretroviral therapy (ART) correlating with increased mortality. However, when Th17 depletion occurs following HIV infection is unknown. We analyzed mucosal Th17 cells in 42 acute HIV infection (AHI) subjects (Fiebig (F) stage I-V) with a median duration of infection of 16 days and the short-term impact of early initiation of ART. Th17 cells were defined as IL-17+ CD4+ T cells and their function was assessed by the co-expression of IL-22, IL-2 and IFNγ. While intact during FI/II, depletion of mucosal Th17 cell numbers and function was observed during FIII correlating with local and systemic markers of immune-activation. ART initiated at FI/II prevented loss of Th17 cell numbers and function, while initiation at FIII restored Th17 cell numbers but not their polyfunctionality. Furthermore, early initiation of ART in FI/II fully reversed the initially observed mucosal and systemic immune-activation. In contrast, patients treated later during AHI maintained elevated mucosal and systemic CD8+ T-cell activation post initiation of ART. These data support a loss of Th17 cells at early stages of acute HIV infection, and highlight that studies of ART initiation during early AHI should be further explored to assess the underlying mechanism of mucosal Th17 function preservation.
- Published
- 2014
- Full Text
- View/download PDF
49. Cryptic determinant of α4β7 binding in the V2 loop of HIV-1 gp120.
- Author
-
Tassaneetrithep B, Tivon D, Swetnam J, Karasavvas N, Michael NL, Kim JH, Marovich M, and Cardozo T
- Subjects
- Amino Acid Sequence, Antigens, Viral immunology, Binding Sites, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Cells, Cultured, HIV Infections, Humans, Protein Binding, Protein Folding, Protein Structure, Quaternary, Protein Structure, Tertiary, Sequence Alignment, Structure-Activity Relationship, Viral Load, AIDS Vaccines immunology, HIV Antibodies immunology, HIV Envelope Protein gp120 immunology, HIV-1 immunology, Integrins immunology
- Abstract
The peptide segment of the second variable loop of HIV-1 spanning positions 166-181 harbors two functionally important sites. The first, spanning positions 179-181, engages the human α4β7 integrin receptor which is involved in T-cell gut-homing and may play a role in human immunodeficiency virus (HIV)-host cell interactions. The second, at positions 166-178, is a major target of anti-V2 antibodies elicited by the ALVAC/AIDSVAX vaccine used in the RV144 clinical trial. Notably, these two sites are directly adjacent, but do not overlap. Here, we report the identity of a second determinant of α4β7 binding located at positions 170-172 of the V2 loop. This segment - tripeptide QRV170-172- is located within the second site, yet functionally affects the first site. The absence of this segment abrogates α4β7 binding in peptides bearing the same sequence from position 173-185 as the V2 loops of the RV144 vaccines. However, peptides exhibiting V2 loop sequences from heterologous HIV-1 strains that include this QRV170-172 motif bind the α4β7 receptor on cells. Therefore, the peptide segment at positions 166-178 of the V2 loop of HIV-1 viruses appears to harbor a cryptic determinant of α4β7 binding. Prior studies show that the anti-V2 antibody response elicited by the RV144 vaccine, along with immune pressure inferred from a sieve analysis, is directed to this same region of the V2 loop. Accordingly, the anti-V2 antibodies that apparently reduced the risk of infection in the RV144 trial may have functioned by blocking α4β7-mediated HIV-host cell interactions via this cryptic determinant.
- Published
- 2014
- Full Text
- View/download PDF
50. Distinct gene-expression profiles associated with the susceptibility of pathogen-specific CD4 T cells to HIV-1 infection.
- Author
-
Hu H, Nau M, Ehrenberg P, Chenine AL, Macedo C, Zhou Y, Daye ZJ, Wei Z, Vahey M, Michael NL, Kim JH, Marovich M, and Ratto-Kim S
- Subjects
- Candida albicans immunology, Candida albicans pathogenicity, Cytomegalovirus immunology, Cytomegalovirus pathogenicity, HIV Infections virology, Host-Pathogen Interactions genetics, Host-Pathogen Interactions immunology, Humans, Immunity, Innate genetics, Immunity, Mucosal genetics, Tetanus Toxoid immunology, Th17 Cells immunology, Th17 Cells virology, Transcriptome, Virus Internalization, Virus Replication, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes virology, HIV Infections genetics, HIV Infections immunology, HIV-1 immunology, HIV-1 pathogenicity, HIV-1 physiology
- Abstract
In HIV infection, CD4 responses to opportunistic pathogens such as Candida albicans are lost early, but CMV-specific CD4 response persists. Little is currently known about HIV infection of CD4 T cells of different pathogen/antigen specificities. CFSE-labeled PBMCs were stimulated with CMV, tetanus toxoid (TT), and C albicans antigens and subsequently exposed to HIV. HIV infection was monitored by intracellular p24 in CFSE(low) population. We found that although TT- and C albicans-specific CD4 T cells were permissive, CMV-specific CD4 T cells were highly resistant to both R5 and X4 HIV. Quantification of HIV DNA in CFSE(low) cells showed a reduction of strong-stop and full-length DNA in CMV-specific cells compared with TT- and C albicans-specific cells. β-Chemokine neutralization enhanced HIV infection in TT- and C albicans-specific cells, whereas HIV infection in CMV-specific cells remained low despite increased entry by β-chemokine neutralization, suggesting postentry HIV restriction by CMV-specific cells. Microarray analysis (Gene Expression Omnibus accession number: GSE42853) revealed distinct transcriptional profiles that involved selective up-regulation of comprehensive innate antiviral genes in CMV-specific cells, whereas TT- and C albicans-specific cells mainly up-regulated Th17 inflammatory response. Our data suggest a mechanism for the persistence of CMV-specific CD4 response and earlier loss of mucosal Th17-associated TT- and C albicans-specific CD4 response in AIDS.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.