36 results on '"Grunenberg, Nicole A."'
Search Results
2. Safety and immunogenicity of a polyvalent DNA–protein HIV vaccine with matched Env immunogens delivered as a prime–boost regimen or coadministered in HIV-uninfected adults in the USA (HVTN 124): a phase 1, placebo-controlled, double-blind randomised controlled trial
- Author
-
Frank, Ian, Li, Shuying S, Grunenberg, Nicole, Overton, Edgar T, Robinson, Samuel T, Zheng, Hua, Seaton, Kelly E, Heptinstall, Jack R, Allen, Mary A, Mayer, Kenneth H, Culver, Daniel A, Keefer, Michael C, Edupuganti, Sri, Pensiero, Michael N, Mehra, Vijay L, De Rosa, Stephen C, Morris, Daryl E, Wang, Shixia, Seaman, Michael S, Montefiori, David C, Ferrari, Guido, Tomaras, Georgia D, Kublin, James G, Corey, Lawrence, and Lu, Shan
- Published
- 2024
- Full Text
- View/download PDF
3. Polytopic fractional delivery of an HIV vaccine alters cellular responses and results in increased epitope breadth in a phase 1 randomized trial
- Author
-
Kalichman, Artur, Edlefsen, Paul, Enama, Mary, Hural, John, Holt, Renee, Dunbar, Debora, Crawford, Dave, Maki, Ian, Johannessen, Jan, Estep, Scharla, Grigoriev, Yevgeny, Madenwald, Tamra, Hansen, Marianne, Holman, Drienna, Fair, Ramey, Meyer, Genevieve, Luke-Kilolam, Anya, Miner, Maurine D., deCamp, Allan, Grunenberg, Nicole, De Rosa, Stephen C., Fiore-Gartland, Andrew, Bar, Katherine, Spearman, Paul, Allen, Mary, Yu, Pei-Chun, Manso, Bryce, Frahm, Nicole, Kalams, Spyros, Baden, Lindsey, Keefer, Michael C., Scott, Hyman M., Novak, Richard, Van Tieu, Hong, Tomaras, Georgia D., Kublin, James G., McElrath, M. Juliana, Corey, Lawrence, and Frank, Ian
- Published
- 2024
- Full Text
- View/download PDF
4. Safety and immunogenicity of a subtype C ALVAC-HIV (vCP2438) vaccine prime plus bivalent subtype C gp120 vaccine boost adjuvanted with MF59 or alum in healthy adults without HIV (HVTN 107): A phase 1/2a randomized trial
- Author
-
Moodie, Zoe, Andersen-Nissen, Erica, Grunenberg, Nicole, Dintwe, One B., Omar, Faatima Laher, Kee, Jia J., Bekker, Linda-Gail, Laher, Fatima, Naicker, Nivashnee, Jani, Ilesh, Mgodi, Nyaradzo M., Hunidzarira, Portia, Sebe, Modulakgota, Miner, Maurine D., Polakowski, Laura, Ramirez, Shelly, Nebergall, Michelle, Takuva, Simbarashe, Sikhosana, Lerato, Heptinstall, Jack, Seaton, Kelly E., De Rosa, Stephen, Diazgranados, Carlos A., Koutsoukos, Marguerite, Van Der Meeren, Olivier, Barnett, Susan W., Kanesa-thasan, Niranjan, Kublin, James G., Tomaras, Georgia D., McElrath, M. Juliana, Corey, Lawrence, Mngadi, Kathryn, and Goepfert, Paul
- Subjects
United States. National Institutes of Health ,HIV (Viruses) -- Comparative analysis -- Usage ,Vaccination -- Usage -- Comparative analysis ,Immune response -- Usage -- Comparative analysis ,Immunoglobulin A -- Comparative analysis -- Usage ,Clinical trials -- Usage -- Comparative analysis ,Adults -- Comparative analysis -- Usage ,Immunoglobulin G -- Usage -- Comparative analysis ,AIDS vaccines -- Usage -- Product development ,Biological sciences - Abstract
Background Adjuvants are widely used to enhance and/or direct vaccine-induced immune responses yet rarely evaluated head-to-head. Our trial directly compared immune responses elicited by MF59 versus alum adjuvants in the RV144-like HIV vaccine regimen modified for the Southern African region. The RV144 trial of a recombinant canarypox vaccine vector expressing HIV env subtype B (ALVAC-HIV) prime followed by ALVAC-HIV plus a bivalent gp120 protein vaccine boost adjuvanted with alum is the only trial to have shown modest HIV vaccine efficacy. Data generated after RV144 suggested that use of MF59 adjuvant might allow lower protein doses to be used while maintaining robust immune responses. We evaluated safety and immunogenicity of an HIV recombinant canarypox vaccine vector expressing HIV env subtype C (ALVAC-HIV) prime followed by ALVAC-HIV plus a bivalent gp120 protein vaccine boost (gp120) adjuvanted with alum (ALVAC-HIV+gp120/alum) or MF59 (ALVAC-HIV+gp120/MF59) or unadjuvanted (ALVAC-HIV+gp120/no-adjuvant) and a regimen where ALVAC-HIV+gp120 adjuvanted with MF59 was used for the prime and boost (ALVAC-HIV+gp120/MF59 coadministration). Methods and findings Between June 19, 2017 and June 14, 2018, 132 healthy adults without HIV in South Africa, Zimbabwe, and Mozambique were randomized to receive intramuscularly: (1) 2 priming doses of ALVAC-HIV (months 0 and 1) followed by 3 booster doses of ALVAC-HIV+gp120/MF59 (months 3, 6, and 12), n = 36; (2) 2 priming doses of ALVAC-HIV (months 0 and 1) followed by 3 booster doses of ALVAC-HIV+gp120/alum (months 3, 6, and 12), n = 36; (3) 4 doses of ALVAC-HIV+gp120/MF59 coadministered (months 0, 1, 6, and 12), n = 36; or (4) 2 priming doses of ALVAC-HIV (months 0 and 1) followed by 3 booster doses of ALVAC-HIV+gp120/no adjuvant (months 3, 6, and 12), n = 24. Primary outcomes were safety and occurrence and mean fluorescence intensity (MFI) of vaccine-induced gp120-specific IgG and IgA binding antibodies at month 6.5. All vaccinations were safe and well-tolerated; increased alanine aminotransferase was the most frequent related adverse event, occurring in 2 (1.5%) participants (1 severe, 1 mild). At month 6.5, vaccine-specific gp120 IgG binding antibodies were detected in 100% of vaccinees for all 4 vaccine groups. No significant differences were seen in the occurrence and net MFI of vaccine-specific IgA responses between the ALVAC-HIV+gp120/MF59-prime-boost and ALVAC-HIV+gp120/alum-prime-boost groups or between the ALVAC-HIV+gp120/MF59-prime-boost and ALVAC-HIV+gp120/MF59 coadministration groups. Limitations were the relatively small sample size per group and lack of evaluation of higher gp120 doses. Conclusions Although MF59 was expected to enhance immune responses, alum induced similar responses to MF59, suggesting that the choice between these adjuvants may not be critical for the ALVAC+gp120 regimen. Trial registration HVTN 107 was registered with the South African National Clinical Trials Registry (DOH-27-0715-4894) and ClinicalTrials.gov (NCT03284710)., Author(s): Zoe Moodie 1,*, Erica Andersen-Nissen 1,2, Nicole Grunenberg 1, One B. Dintwe 1,2, Faatima Laher Omar 2, Jia J. Kee 1, Linda-Gail Bekker 3, Fatima Laher 4, Nivashnee Naicker [...]
- Published
- 2024
- Full Text
- View/download PDF
5. Efficacy of a bivalent (D614 + B.1.351) SARS-CoV-2 recombinant protein vaccine with AS03 adjuvant in adults: a phase 3, parallel, randomised, modified double-blind, placebo-controlled trial
- Author
-
Abalos, Karina, Accini, Jose, Aloysia, Naveena, Amuasi, John Humphrey, Ansah, Nana Akosua, Benkeser, David, Berge, Aude, Beyko, Hanna, Bilotkach, Oleksandra, Breuer, Thomas, Bonfanti, Alberto Cadena, Bukusi, Elisabeth, Canter, Richard, Carrillo, Jaime Augusto, Chansinghakul, Danaya, Coux, Florence, Das, Chandan, Das, Santa Kumar, Devlin, Louis, Espinoza, Luis, Fay, Michael, Follmann, Dean, Frago, Carina, Garinga, Agnes, Gilbert, Peter B, Gonzalez, Claudia, Granados, Maria Angelica, Guillery, Lea, Huang, Ying, Hudzina, Kathy, Jain, Manish, Kanodia, Piush, Khandelwal, Nitin, Mutuluuza, Cissy Kityo, Kiweewa, Francis, Kiwanuka, Noah, Kosolsak, Chalit, Kukian, Darshna, Kushwaha, Jitendra Singh, Laot, Thelma, Lopez-Medina, Eduardo, Macareno Arroyo, Hugo, Mandaliya, Kishorchandra, Mamod, Stephanie, Mangarule, Somnath, Martínez, Javier, McClelland, Scott, Menard, Lisa, Mendoza, Sandra, Mohapatra, Satyajit, Moreau, Catherine, Mugo, Nelly, Nduba, Videlis, Noriega, Fernando, Ntege, Patricia Nahirya, Okech, Brenda, Otero, Maria, Ouma, Samuel Gurrion, Oyieko, Janet, Paredes, Mercedes, Pardo, Erwin, Postol, Svitlana, Pekala, David, Peng, Penny, Py, Marie-Laure, Rivas, Enrique, Rivero, Rafael, Rodriguez, Edith, Saleh, Mansoor, Sánchez, Pedro, Sater, Nessryne, Shah, Jinen, Shrestha, Rajeev, Siika, Abraham, Singh, Chandramani, Singh, Veer Bahadur, Tamrakar, Dipesh, Tavares Da-Silva, Fernanda, Otieno Tina, Lucas, Velasquez, Hector, Wabwire, Deo, Wajja, Anne, Zaworski, Elodie, Zhang, Nianxian, Dayan, Gustavo H, Rouphael, Nadine, Walsh, Stephen R, Chen, Aiying, Grunenberg, Nicole, Allen, Mary, Antony, Johannes, Asante, Kwaku Poku, Bhate, Amit Suresh, Beresnev, Tatiana, Bonaparte, Matthew I, Celle, Médéric, Ceregido, Maria Angeles, Corey, Lawrence, Dobrianskyi, Dmytro, Fu, Bo, Grillet, Marie-Helene, Keshtkar-Jahromi, Maryam, Juraska, Michal, Kee, Jia Jin, Kibuuka, Hannah, Koutsoukos, Marguerite, Masotti, Roger, Michael, Nelson L, Neuzil, Kathleen M, Reynales, Humberto, Robb, Merlin L, Villagómez Martínez, Sandra M, Sawe, Fredrick, Schuerman, Lode, Tong, Tina, Treanor, John, Wartel, T Anh, Diazgranados, Carlos A, Chicz, Roman M, Gurunathan, Sanjay, Savarino, Stephen, and Sridhar, Saranya
- Published
- 2023
- Full Text
- View/download PDF
6. Efficacy of a monovalent (D614) SARS-CoV-2 recombinant protein vaccine with AS03 adjuvant in adults: a phase 3, multi-country study
- Author
-
Abalos, Karina, Adams, Michael, Allaw, Mohamed, Aloysia, Naveena, Amuasi, John Humphrey, Ansah, Nana Akosua, Asante, Kwaku Poku, Benkeser, David, Berge, Aude, Breuer, Thomas, Briesemeister, Liz, Broder, Gail, Bonfanti, Alberto Cadena, Calinescu, Cornell, Canter, Richard, Carrillo, Jaime Augusto, Chansinghakul, Danaya, Coux, Florence, Das, Chandan, Davies, Matthew, Devlin, Louis, Fay, Michael, Follmann, Dean, Frago, Carina, Fukase, Hiroyuki, Garinga, Agnes, Gilbert, Peter B., Gonzalez, Claudia, Granados, Maria Angelica, Greiwe, Cathy, Guillery, Lea, Hall, Jessicalee, Henderson, Jeffrey, Huang, Ying, Hudzina, Kathy, Hural, John, Hutchens, Mark, Jain, Manish, Jennings, William, Kanodia, Piush, Kimmel, Murray, Kirby, William, Khandelwal, Nitin, Kopp, James, Kosolsak, Chalit, Kublin, Jim, Kukian, Darshna, Kushwaha, Jitendra Singh, Laot, Thelma, Lopez-Medina, Eduardo, Arroyo, Hugo Macareno, Mamod, Stephanie, Mangarule, Somnath, Martin, Troy, Menard, Lisa, Mendoza, Sandra, Meyer, Robert, Middleton, Randle, Miracle, Jill, Mizuyama, Kazuyuki, Mohapatra, Satyajit, Moreau, Catherine, Murray, Linda, Nagamatsu, Shinya, Newberg, Joseph, Noriega, Fernando, Nugent, Paul, Peake-Andrasik, Michele, Pekala, David, Peng, Penny, Py, Marie-Laure, Ramirez, Shelly, Reddy, Chinthaparthi Prabhakar, Reynolds, Michelle, Rivas, Enrique, Sater, Nessryne, Shah, Jinen, Sher, Lawrence, Sieger, Silva, Singh, Chandramani, Singh, Veer Bahadur, Sirisuphmitr, Nuchra, Starkey, Thomas, Suzuki, Kazuo, Tamrakar, Dipesh, Tangemen, Cayce, Da-Silva, Fernanda Tavares, Taylor, David, Tharenos, Leslie, Wartel, T. Anh, Zaworski, Elodie, Zhang, Nianxian, Dayan, Gustavo H., Rouphael, Nadine, Walsh, Stephen R., Chen, Aiying, Grunenberg, Nicole, Allen, Mary, Antony, Johannes, Bhate, Amit Suresh, Beresnev, Tatiana, Bonaparte, Matthew I., Celle, Médéric, Ceregido, Maria Angeles, Corey, Lawrence, Fu, Bo, Grillet, Marie-Helene, Keshtkar-Jahromi, Maryam, Juraska, Michal, Kee, Jia Jin, Kaali, Seyram, Koutsoukos, Marguerite, Masotti, Roger, Michael, Nelson L., Neuzil, Kathleen M., Reynales, Humberto, Robb, Merlin L., Uchiyama, Akiyoshi, Sawe, Fredrick, Schuerman, Lode, Shrestha, Rajeev, Tong, Tina, Treanor, John, Diazgranados, Carlos A., Chicz, Roman M., Gurunathan, Sanjay, Savarino, Stephen, and Sridhar, Saranya
- Published
- 2023
- Full Text
- View/download PDF
7. Safety and immunogenicity of an AS03-adjuvanted SARS-CoV-2 recombinant protein vaccine (CoV2 preS dTM) in healthy adults: interim findings from a phase 2, randomised, dose-finding, multicentre study
- Author
-
Sridhar, Saranya, Joaquin, Arnel, Bonaparte, Matthew I, Bueso, Agustin, Chabanon, Anne-Laure, Chen, Aiying, Chicz, Roman M, Diemert, David, Essink, Brandon J, Fu, Bo, Grunenberg, Nicole A, Janosczyk, Helene, Keefer, Michael C, Rivera M, Doris M, Meng, Ya, Michael, Nelson L, Munsiff, Sonal S, Ogbuagu, Onyema, Raabe, Vanessa N, Severance, Randall, Rivas, Enrique, Romanyak, Natalya, Rouphael, Nadine G, Schuerman, Lode, Sher, Lawrence D, Walsh, Stephen R, White, Judith, von Barbier, Dalia, de Bruyn, Guy, Canter, Richard, Grillet, Marie-Helene, Keshtkar-Jahromi, Maryam, Koutsoukos, Marguerite, Lopez, Denise, Masotti, Roger, Mendoza, Sandra, Moreau, Catherine, Ceregido, Maria Angeles, Ramirez, Shelly, Said, Ansoyta, Tavares-Da-Silva, Fernanda, Shi, Jiayuan, Tong, Tina, Treanor, John, Diazgranados, Carlos A, and Savarino, Stephen
- Published
- 2022
- Full Text
- View/download PDF
8. Non-HIV Vaccine-Induced Immune Responses as Potential Baseline Immunogenicity Predictors of ALVAC-HIV and AIDSVAX B/E-Induced Immune Responses.
- Author
-
Huang, Ying, Alam, Shomoita, Andersen-Nissen, Erica, Carpp, Lindsay N., Dintwe, One B., Flach, Britta S., Grunenberg, Nicole, Laher, Fatima, De Rosa, Stephen C., Ferrari, Guido, Innes, Craig, Bekker, Linda-Gail, Kublin, James G., McElrath, M. Juliana, Tomaras, Georgia D., Gray, Glenda E., and Gilbert, Peter B.
- Subjects
HEPATITIS associated antigen ,AIDS vaccines ,TETANUS vaccines ,HEPATITIS B vaccines ,VACCINE effectiveness ,HEPATITIS B virus - Abstract
Identifying correlations between immune responses elicited via HIV and non-HIV vaccines could aid the search for correlates of HIV protection and increase statistical power in HIV vaccine-efficacy trial designs. An exploratory objective of the HVTN 097 phase 1b trial was to assess whether immune responses [focusing on those supported as correlates of risk (CoR) of HIV acquisition] induced via the RV144 pox-prime HIV vaccine regimen correlated with those induced via tetanus toxoid (TT) and/or hepatitis B virus (HBV) vaccines. We measured TT-specific and HBV-specific IgG-binding antibody responses and TT-specific and HBV-specific CD4+ T-cell responses at multiple time points in HVTN 097 participants, and we assessed their correlations at peak time points with HIV vaccine (ALVAC-HIV and AIDSVAX B/E)-induced responses. Four correlations were significant [false discovery rate-adjusted p-value (FDR) ≤ 0.2]. Three of these four were with IgG-binding antibody responses to TT measured one month after TT receipt, with the strongest and most significant correlation [rho = 0.368 (95% CI: 0.096, 0.588; p = 0.008; FDR = 0.137)] being with IgG-binding antibody responses to MN gp120 gDneg (B protein boost) measured two weeks after the second ALVAC-HIV and AIDSVAX B/E boost. The fourth significant correlation [(rho = 0.361; 95% CI: 0.049, 0.609; p = 0.021; FDR = 0.137)] was between CD4+ T-cell responses to a hepatitis B surface antigen peptide pool, measured 2 weeks after the third HBV vaccination, and IgG-binding antibody responses to gp70BCaseAV1V2 (B V1V2 immune correlate), measured two weeks after the second ALVAC-HIV and AIDSVAX B/E boost. These moderate correlations imply that either vaccine, TT or HBV, could potentially provide a moderately useful immunogenicity predictor for the ALVAC-HIV and AIDSVAX B/E HIV vaccine regimen. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Protein Dose-Sparing Effect of AS01B Adjuvant in a Randomized Preventive HIV Vaccine Trial of ALVAC-HIV (vCP2438) and Adjuvanted Bivalent Subtype C gp120.
- Author
-
Chirenje, Zvavahera Mike, Laher, Fatima, Dintwe, One, Muyoyeta, Monde, deCamp, Allan C, He, Zonglin, Grunenberg, Nicole, Omar, Faatima Laher, Seaton, Kelly E, Polakowski, Laura, Davis, Amanda S Woodward, Maganga, Lucas, Baden, Lindsey R, Mayer, Kenneth, Kalams, Spyros, Keefer, Michael, Edupuganti, Srilatha, Rodriguez, Benigno, Frank, Ian, and Scott, Hyman
- Subjects
CLINICAL trial registries ,HIV-1 glycoprotein 120 ,VACCINE trials ,HIV ,AIDS vaccines - Abstract
Background HVTN 120 is a phase 1/2a randomized double-blind placebo-controlled human immunodeficiency virus (HIV) vaccine trial that evaluated the safety and immunogenicity of ALVAC-HIV (vCP2438) and MF59- or AS01
B -adjuvanted bivalent subtype C gp120 Env protein at 2 dose levels in healthy HIV-uninfected adults. Methods Participants received ALVAC-HIV (vCP2438) alone or placebo at months 0 and 1. At months 3 and 6, participants received either placebo, ALVAC-HIV (vCP2438) with 200 μg of bivalent subtype C gp120 adjuvanted with MF59 or AS01B , or ALVAC-HIV (vCP2438) with 40 μg of bivalent subtype C gp120 adjuvanted with AS01B . Primary outcomes were safety and immune responses. Results We enrolled 160 participants, 55% women, 18–40 years old (median age 24 years) of whom 150 received vaccine and 10 placebo. Vaccines were generally safe and well tolerated. At months 6.5 and 12, CD4+ T-cell response rates and magnitudes were higher in the AS01B -adjuvanted groups than in the MF59-adjuvanted group. At month 12, HIV-specific Env-gp120 binding antibody response magnitudes in the 40 μg gp120/AS01B group were higher than in either of the 200 μg gp120 groups. Conclusions The 40 μg dose gp120/AS01B regimen elicited the highest CD4+ T-cell and binding antibody responses. Clinical Trials Registration. NCT03122223. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
10. Safety and Immunogenicity of a DNA Vaccine With Subtype C gp120 Protein Adjuvanted With MF59 or AS01 B : A Phase 1/2a HIV-1 Vaccine Trial.
- Author
-
Garrett, Nigel, Dintwe, One, Monaco, Cynthia L., Jones, Megan, Seaton, Kelly E., Church, E. Chandler, Grunenberg, Nicole, Hutter, Julia, deCamp, Allan, Huang, Yunda, Lu, Huiyin, Mann, Philipp, Robinson, Samuel T., Heptinstall, Jack, Jensen, Ryan L., Pantaleo, Giuseppe, Ding, Song, Koutsoukos, Marguerite, Hosseinipour, Mina C., and Van Der Meeren, Olivier
- Published
- 2024
- Full Text
- View/download PDF
11. Subtype C ALVAC-HIV and bivalent subtype C gp120/MF59 HIV-1 vaccine in low-risk, HIV-uninfected, South African adults: a phase 1/2 trial
- Author
-
Bekker, Linda-Gail, Moodie, Zoe, Grunenberg, Nicole, Laher, Fatima, Tomaras, Georgia D, Cohen, Kristen W, Allen, Mary, Malahleha, Mookho, Mngadi, Kathryn, Daniels, Brodie, Innes, Craig, Bentley, Carter, Frahm, Nicole, Morris, Daryl E, Morris, Lynn, Mkhize, Nonhlanhla N, Montefiori, David C, Sarzotti-Kelsoe, Marcella, Grant, Shannon, Yu, Chenchen, Mehra, Vijay L, Pensiero, Michael N, Phogat, Sanjay, DiazGranados, Carlos A, Barnett, Susan W, Kanesa-thasan, Niranjan, Koutsoukos, Marguerite, Michael, Nelson L, Robb, Merlin L, Kublin, James G, Gilbert, Peter B, Corey, Lawrence, Gray, Glenda E, and McElrath, M Juliana
- Published
- 2018
- Full Text
- View/download PDF
12. HIV-1 Vaccine Sequences Impact V1V2 Antibody Responses: A Comparison of Two Poxvirus Prime gp120 Boost Vaccine Regimens
- Author
-
Shen, Xiaoying, Laher, Fatima, Moodie, Zoe, McMillan, Arthur S., Spreng, Rachel L., Gilbert, Peter B., Huang, Ying, Yates, Nicole L., Grunenberg, Nicole, Juliana McElrath, M., Allen, Mary, Pensiero, Michael, Mehra, Vijay L., Der Meeren, Olivier Van, Barnett, Susan W., Phogat, Sanjay, Gray, Glenda E., Bekker, Linda-Gail, Corey, Lawrence, and Tomaras, Georgia D.
- Published
- 2020
- Full Text
- View/download PDF
13. Brief Report: Prediction of Serum HIV-1 Neutralization Titers After Passive Administration of VRC01
- Author
-
Huang, Yunda, Zhang, Yuanyuan, Bailer, Robert, Grunenberg, Nicole, Carpp, Lindsay N., Seaton, Kelly, Mayer, Kenneth H., Ledgerwood, Julie, Corey, Lawrence, Mascola, John, Montefiori, David, and Gilbert, Peter B.
- Published
- 2020
- Full Text
- View/download PDF
14. Rectal tissue and vaginal tissue from intravenous VRC01 recipients show protection against ex vivo HIV-1 challenge
- Author
-
Astronomo, Rena D., Lemos, Maria P., Narpala, Sandeep R., Czartoski, Julie, Fleming, Lamar Ballweber, Seaton, Kelly E., Prabhakaran, Madhu, Huang, Yunda, Lu, Yiwen, Westerberg, Katharine, Zhang, Lily, Gross, Mary K., Hural, John, Tieu, Hong- Van, Baden, Lindsey R., Hammer, Scott, Frank, Ian, Ochsenbauer, Christina, Grunenberg, Nicole, Ledgerwood, Julie E., Mayer, Kenneth, Tomaras, Georgia, McDermott, Adrian B., and McElrath, M. Juliana
- Subjects
Antiviral agents -- Testing -- Dosage and administration ,Anti-HIV agents -- Testing -- Dosage and administration ,Vagina -- Health aspects -- Physiological aspects ,Rectum -- Health aspects -- Physiological aspects ,Monoclonal antibodies -- Testing -- Dosage and administration ,HIV infection -- Drug therapy -- Models ,Health care industry - Abstract
BACKGROUND. VRC01, a potent, broadly neutralizing monoclonal antibody, inhibits simian-HIV infection in animal models. The HVTN 104 study assessed the safety and pharmacokinetics of VRC01 in humans. We extend the clinical evaluation to determine intravenously infused VRC01 distribution and protective function at mucosal sites of HIV-1 entry. METHODS. Healthy, HIV-1-uninfected men (n = 7) and women (n = 5) receiving VRC01 every 2 months provided mucosal and serum samples once, 4-13 days after infusion. Eleven male and 8 female HIV-seronegative volunteers provided untreated control samples. VRC01 levels were measured in serum, secretions, and tissue, and HIV-1 inhibition was determined in tissue explants. RESULTS. Median VRC01 levels were quantifiable in serum (96.2 [micro]g/mL or 1.3 pg/ng protein), rectal tissue (0.11 pg/ng protein), rectal secretions (0.13 pg/ng protein), vaginal tissue (0.1 pg/ng protein), and cervical secretions (0.44 pg/ng protein) from all recipients. VRC01/IgG ratios in male serum correlated with those in paired rectal tissue (r = 0.893, P = 0.012) and rectal secretions (r = 0.9643, P = 0.003). Ex vivo [HIV-1.sub.Bal26] challenge infected 4 of 21 rectal explants from VRC01 recipients versus 20 of 22 from controls (P = 0.005); [HIV-1.sub.Du422.1] infected 20 of 21 rectal explants from VRC01 recipients and 12 of 12 from controls (P = 0.639). [HIV-1.sub.Bal26] infected 0 of 14 vaginal explants of VRC01 recipients compared with 23 of 28 control explants (P = 0.003). CONCLUSION. Intravenous VRC01 distributes into the female genital and male rectal mucosa and retains anti-HIV-1 functionality, inhibiting a highly neutralization-sensitive but not a highly resistant HIV-1 strain in mucosal tissue. These findings lend insight into VRC01 mucosal infiltration and provide perspective on in vivo protective efficacy. FUNDING. National Institute of Allergy and Infectious Diseases and Bill & Melinda Gates Foundation., Introduction In 2019, 1.7 million people were newly infected with HIV-1 (1), in nearly all regions of the world, even in countries with access to the latest prevention toolkit. Clearly, [...]
- Published
- 2021
- Full Text
- View/download PDF
15. Efficacy of a bivalent (D614 + B.1.351) SARS-CoV-2 recombinant protein vaccine with AS03 adjuvant in adults: a phase 3, parallel, randomised, modified double-blind, placebo-controlled trial.
- Author
-
Dayan, Gustavo H, Rouphael, Nadine, Walsh, Stephen R, Chen, Aiying, Grunenberg, Nicole, Allen, Mary, Antony, Johannes, Asante, Kwaku Poku, Bhate, Amit Suresh, Beresnev, Tatiana, Bonaparte, Matthew I, Celle, Médéric, Ceregido, Maria Angeles, Corey, Lawrence, Dobrianskyi, Dmytro, Fu, Bo, Grillet, Marie-Helene, Keshtkar-Jahromi, Maryam, Juraska, Michal, and Kee, Jia Jin
- Subjects
RECOMBINANT proteins ,NUCLEIC acid amplification techniques ,BELL'S palsy ,SARS-CoV-2 Omicron variant ,PERICARDITIS ,SARS-CoV-2 - Abstract
COVID-19 vaccines with alternative strain compositions are needed to provide broad protection against newly emergent SARS-CoV-2 variants of concern. This study aimed to describe the clinical efficacy and safety of a bivalent SARS-CoV-2 recombinant protein vaccine as a two-injection primary series during a period of circulation of the omicron (B.1.1.529) variant. We conducted a phase 3, parallel, randomised, modified double-blind, placebo-controlled trial in adults aged 18 years or older at 54 clinical research centres in eight countries (Colombia, Ghana, India, Kenya, Mexico, Nepal, Uganda, and Ukraine). Participants were recruited from the community and randomly assigned (1:1) by use of an interactive response technology system to receive two intramuscular 0·5 mL injections, 21 days apart, of the bivalent vaccine (5 μg of ancestral [D614] and 5 μg of beta [B.1.351] variant spike protein, with AS03 adjuvant) or placebo (0·9% normal saline). All participants, outcome assessors, and laboratory staff performing assays were masked to group assignments; those involved in the preparation and administration of the vaccines were unmasked. Participants were stratified by age (18–59 years and ≥60 years) and baseline SARS-CoV-2 rapid serodiagnostic test positivity. Symptomatic COVID-19 was defined as laboratory-confirmed (via nucleic acid amplification test or PCR test) COVID-19 with COVID-19-like illness symptoms. The primary efficacy endpoint was the clinical efficacy of the bivalent vaccine for prevention of symptomatic COVID-19 at least 14 days after the second injection (dose 2). Safety was assessed in all participants receiving at least one injection of the study vaccine or placebo. This trial is registered with ClinicalTrials.gov (NCT04904549) and is closed to recruitment. Between Oct 19, 2021, and Feb 15, 2022, 13 002 participants were enrolled and randomly assigned to receive the first dose of the study vaccine (n=6512) or placebo (n=6490). 12 924 participants (6472 in the vaccine group and 6452 in the placebo group) received at least one study injection, of whom 7542 (58·4%) were male and 9693 (75·0%) were SARS-CoV-2 non-naive. Of these 12 924 participants, 11 543 (89·3%) received both study injections (5788 in the vaccine group and 5755 in the placebo group). The efficacy-evaluable population after dose 2 comprised 11 416 participants (5736 in the vaccine group and 5680 in the placebo group). The median duration of follow-up was 85 days (IQR 50–95) after dose 1 and 58 days (29–70) after dose 2. 121 symptomatic COVID-19 cases were reported at least 14 days after dose 2 (32 in the vaccine group and 89 in the placebo group), with an overall vaccine efficacy of 64·7% (95% CI 46·6 to 77·2). Vaccine efficacy against symptomatic COVID-19 was 75·1% (95% CI 56·3 to 86·6) in SARS-CoV-2 non-naive participants and 30·9% (–39·3 to 66·7) in SARS-CoV-2-naive participants. Viral genome sequencing identified the infecting strain in 68 (56·2%) of 121 cases (omicron [BA.1 and BA.2] in 63; delta in four; and both omicron and delta in one). Immediate unsolicited adverse events were reported by four (<0·1%) participants in the vaccine group and seven (0·1%) participants in the placebo group. Immediate unsolicited adverse reactions within 30 min after any injection were reported by four (<0·1%) participants in the vaccine group and six (<0·1%) participants in the placebo group. In the reactogenicity subset with available data, solicited reactions (solicited injection-site reactions and solicited systemic reactions) within 7 days after any injection occurred in 1398 (57·8%) of 2420 vaccine recipients and 983 (40·9%) of 2403 placebo recipients. Grade 3 solicited reactions were reported by 196 (8·1%; 95% CI 7·0 to 9·3) of 2420 vaccine recipients and 118 (4·9%; 4·1 to 5·9) of 2403 placebo recipients within 7 days after any injection, with comparable frequencies after dose 1 and dose 2 in the vaccine group. At least one serious adverse event occurred in 30 (0·5%) participants in the vaccine group and 26 (0·4%) in the placebo group. The proportion of adverse events of special interest and deaths was less than 0·1% in both study groups. No adverse event of special interest, serious adverse event, or death was deemed to be treatment related. There were no reported cases of thrombosis with thrombocytopenia syndrome, myocarditis, pericarditis, Bell's Palsy, or Guillain–Barré syndrome, or other immune-mediated diseases. The bivalent variant vaccine conferred heterologous protection against symptomatic SARS-CoV-2 infection in the epidemiological context of the circulating contemporary omicron variant. These findings suggest that vaccines developed with an antigen from a non-predominant strain could confer cross-protection against newly emergent SARS-CoV-2 variants, although further investigation is warranted. Sanofi, US Biomedical Advanced Research and Development Authority, and the US National Institute of Allergy and Infectious Diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
16. Efficacy of Messenger RNA–1273 Against Severe Acute Respiratory Syndrome Coronavirus 2 Acquisition in Young Adults From March to December 2021.
- Author
-
Stephenson, Kathryn E, Marcelin, Jasmine R, Pettifor, Audrey E, Janes, Holly, Brown, Elizabeth, Neradilek, Moni, Yen, Catherine, Andriesen, Jessica, Grunenberg, Nicole, Espy, Nicole, Trahey, Meg, Fischer, Rebecca S B, DeSouza, Christopher A, Shisler, Joanna L, Connick, Elizabeth, Houpt, Eric R, Chu, Helen Y, McCulloh, Russel J, Becker-Dreps, Sylvia, and Vielot, Nadja A
- Subjects
SARS-CoV-2 ,CORONAVIRUS diseases ,YOUNG adults ,COVID-19 ,SARS-CoV-2 Delta variant ,CLINICAL trial registries - Abstract
Background The efficacy of messenger RNA (mRNA)–1273 against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is not well defined, particularly among young adults. Methods Adults aged 18–29 years with no known history of SARS-CoV-2 infection or prior vaccination for coronavirus disease 2019 (COVID-19) were recruited from 44 US sites from 24 March to 13 September 2021 and randomized 1:1 to immediate vaccination (receipt of 2 doses of mRNA-1273 vaccine at months 0 and 1) or the standard of care (receipt of COVID-19 vaccine). Randomized participants were followed up for SARS-CoV-2 infection measured by nasal swab testing and symptomatic COVID-19 measured by nasal swab testing plus symptom assessment and assessed for the primary efficacy outcome. A vaccine-declined observational group was also recruited from 16 June to 8 November 2021 and followed up for SARS-CoV-2 infection as specified for the randomized participants. Results The study enrolled 1149 in the randomized arms and 311 in the vaccine-declined group and collected >122 000 nasal swab samples. Based on randomized participants, the efficacy of 2 doses of mRNA-1273 vaccine against SARS-CoV-2 infection was 52.6% (95% confidence interval, −14.1% to 80.3%), with the majority of infections due to the Delta variant. Vaccine efficacy against symptomatic COVID-19 was 71.0% (95% confidence interval, −9.5% to 92.3%). Precision was limited owing to curtailed study enrollment and off-study vaccination censoring. The incidence of SARS-CoV-2 infection in the vaccine-declined group was 1.8 times higher than in the standard-of-care group. Conclusions mRNA-1273 vaccination reduced the incidence of SARS-CoV-2 infection from March to September 2021, but vaccination was only one factor influencing risk. Clinical Trials Registration NCT04811664. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
17. Diversion of HIV-1 vaccine–induced immunity by gp41-microbiota cross-reactive antibodies
- Author
-
Williams, Wilton B., Liao, Hua-Xin, Moody, M. Anthony, Kepler, Thomas B., Alam, S. Munir, Gao, Feng, Wiehe, Kevin, Trama, Ashley M., Jones, Kathryn, Zhang, Ruijun, Song, Hongshuo, Marshall, Dawn J., Whitesides, John F., Sawatzki, Kaitlin, Hua, Axin, Liu, Pinghuang, Tay, Matthew Z., Seaton, Kelly E., Shen, Xiaoying, Foulger, Andrew, Lloyd, Krissey E., Parks, Robert, Pollara, Justin, Ferrari, Guido, Yu, Jae-Sung, Vandergrift, Nathan, Montefiori, David C., Sobieszczyk, Magdalena E., Hammer, Scott, Karuna, Shelly, Gilbert, Peter, Grove, Doug, Grunenberg, Nicole, McElrath, M. Juliana, Mascola, John R., Koup, Richard A., Corey, Lawrence, Nabel, Gary J., Morgan, Cecilia, Churchyard, Gavin, Maenza, Janine, Keefer, Michael, Graham, Barney S., Baden, Lindsey R., Tomaras, Georgia D., and Haynes, Barton F.
- Published
- 2015
18. Antibody and cellular responses to HIV vaccine regimens with DNA plasmid as compared with ALVAC priming: An analysis of two randomized controlled trials
- Author
-
Moodie, Zoe, Walsh, Stephen R., Laher, Fatima, Maganga, Lucas, Herce, Michael E., Naidoo, Sarita, Hosseinipour, Mina C., Innes, Craig, Bekker, Linda-Gail, Grunenberg, Nicole, Mann, Philipp, Yu, Chenchen, deCamp, Allan C., Miner, Maurine D., Yates, Nicole L., Heptinstall, Jack, Mkhize, Nonhlanhla N., Dintwe, One, Frahm, Nicole, Cohen, Kristen W., Allen, Mary, Hutter, Julia, Wagner, Ralf, Pantaleo, Giuseppe, McElrath, M. Juliana, Tomaras, Georgia D., Morris, Lynn, Montefiori, David C., Andersen-Nissen, Erica, Gray, Glenda E., Gilbert, Peter B., and Kublin, James G.
- Subjects
Clinical trials ,AIDS vaccines ,Genetic research ,HIV antigens ,HIV -- Drug therapy ,DNA ,Vaccination ,T cells ,Biological sciences - Abstract
Background DNA plasmids promise a pragmatic alternative to viral vectors for prime-boost HIV-1 vaccines. We evaluated DNA plasmid versus canarypox virus (ALVAC) primes in 2 randomized, double-blind, placebo-controlled trials in southern Africa with harmonized trial designs. HIV Vaccine Trials Network (HVTN) 111 tested DNA plasmid prime by needle or needleless injection device (Biojector) and DNA plasmid plus gp120 protein plus MF59 adjuvant boost. HVTN 100 tested ALVAC prime and ALVAC plus gp120 protein plus MF59 adjuvant boost (same protein/adjuvant as HVTN 111) by needle. Methods and findings The primary endpoints for this analysis were binding antibody (bAb) responses to HIV antigens (gp120 from strains ZM96, 1086, and TV1; variable 1 and 2 [V1V2] regions of gp120 from strains TV1, 1086, and B.CaseA, as 1086 V1V2 and B.CaseA were correlates of risk in the RV144 efficacy trial), neutralizing antibody (nAb) responses to pseudoviruses TV1c8.2 and MW925.26, and cellular responses to vaccine-matched antigens (envelope [Env] from strains ZM96, 1086, and TV1; and Gag from strains LAI and ZM96) at month 6.5, two weeks after the fourth vaccination. Per-protocol cohorts included vaccine recipients from HVTN 100 (n = 186, 60% male, median age 23 years) enrolled between February 9, 2015, and May 26, 2015 and from HVTN 111 (n = 56, 48% male, median age 24 years) enrolled between June 21, 2016, and July 13, 2017. IgG bAb response rates were 100% to 3 Env gp120 antigens in both trials. Response rates to V1V2 were lower and similar in both trials except to vaccine-matched 1086 V1V2, with rates significantly higher for the DNA-primed regimen than the ALVAC-primed regimen: 96.6% versus 72.7% (difference = 23.9%, 95% CI 15.6%-32.2%, p < 0.001). Among positive responders, bAb net mean fluorescence intensity (MFI) was significantly higher with the DNA-primed regimen than ALVAC-primed for 1086 V1V2 (geometric mean [GM] 2,833.3 versus 1,200.9; ratio = 2.36, 95% CI 1.42-3.92, p 98% in both trials, with significantly higher 50% inhibitory dilution (ID.sub.50) among DNA-primed positive responders (n = 53) versus ALVAC-primed (n = 182) to tier 1A MW965.26 (GM 577.7 versus 265.7, ratio = 2.17, 95% CI 1.67-2.83, p < 0.001) and to TV1c8.2 (GM 187.3 versus 100.4, ratio = 1.87, 95% CI 1.48-2.35, p < 0.001). CD4+ T-cell response rates were significantly higher with DNA plasmid prime via Biojector than ALVAC prime (91.4% versus 52.8%, difference = 38.6%, 95% CI 20.5%-56.6%, p < 0.001 for ZM96.C; 88.0% versus 43.1%, difference = 44.9%, 95% CI 26.7%-63.1%, p < 0.001 for 1086.C; 55.5% versus 2.2%, difference = 53.3%, 95% CI 23.9%-82.7%, p < 0.001 for Gag LAI/ZM96). The study's main limitations include the nonrandomized comparison of vaccines from 2 different trials, the lack of data on immune responses to other non-vaccine-matched antigens, and the uncertain clinical significance of the observed immunological effects. Conclusions In this study, we found that further investigation of DNA/protein regimens is warranted given enhanced immunogenicity to the V1V2 correlates of decreased HIV-1 acquisition risk identified in RV144, the only HIV vaccine trial to date to show any efficacy., Author(s): Zoe Moodie 1,*, Stephen R. Walsh 2,3,4, Fatima Laher 5, Lucas Maganga 6, Michael E. Herce 7, Sarita Naidoo 8, Mina C. Hosseinipour 9, Craig Innes 10, Linda-Gail Bekker [...]
- Published
- 2020
- Full Text
- View/download PDF
19. Effects of Food Intake on the Pharmacokinetic Properties of Mirabegron Oral Controlled-Absorption System: A Single-Dose, Randomized, Crossover Study in Healthy Adults
- Author
-
Lee, Jennifer, Zhang, Wenhui, Moy, Selina, Kowalski, Donna, Kerbusch, Virginie, van Gelderen, Marcel, Sawamoto, Taiji, Grunenberg, Nicole, and Keirns, James
- Published
- 2013
- Full Text
- View/download PDF
20. Assessment of the Safety and Immunogenicity of 2 Novel Vaccine Platforms for HIV-1 Prevention: A Randomized Trial
- Author
-
Baden, Lindsey R., Karita, Etienne, Mutua, Gaudensia, Bekker, Linda-Gail, Gray, Glenda, Page-Shipp, Liesl, Walsh, Stephen R., Nyombayire, Julien, Anzala, Omu, Roux, Surita, Laher, Fatima, Innes, Craig, Seaman, Michael S., Cohen, Yehuda Z., Peter, Lauren, Frahm, Nicole, McElrath, M. Juliana, Hayes, Peter, Swann, Edith, Grunenberg, Nicole, Grazia-Pau, Maria, Weijtens, Mo, Sadoff, Jerry, Dally, Len, Lombardo, Angela, Gilmour, Jill, Cox, Josephine, Dolin, Raphael, Fast, Patricia, Barouch, Dan H., and Laufer, Dagna S.
- Published
- 2016
21. HIV-1 VACCINES: Diversion of HIV-1 vaccine-induced immunity by gp41-microbiota cross-reactive antibodies
- Author
-
Williams, Wilton B., Liao, Hua-Xin, Moody, Anthony M., Kepler, Thomas B., Alam, Munir S., Gao, Feng, Wiehe, Kevin, Trama, Ashley M., Jones, Kathryn, Zhang, Ruijun, Song, Hongshuo, Marshall, Dawn J., Whitesides, John F., Sawatzki, Kaitlin, Hua, Axin, Liu, Pinghuang, Tay, Matthew Z., Seaton, Kelly E., Shen, Xiaoying, Foulger, Andrew, Lloyd, Krissey E., Parks, Robert, Pollara, Justin, Ferrari, Guido, Yu, Jae-Sung, Vandergrift, Nathan, Montefiori, David C., Sobieszczyk, Magdalena E., Hammer, Scott, Karuna, Shelly, Gilbert, Peter, Grove, Doug, Grunenberg, Nicole, McElrath, Juliana M., Mascola, John R., Koup, Richard A., Corey, Lawrence, Nabel, Gary J., Morgan, Cecilia, Churchyard, Gavin, Maenza, Janine, Keefer, Michael, Graham, Barney S., Baden, Lindsey R., Tomaras, Georgia D., and Haynes, Barton F.
- Published
- 2015
- Full Text
- View/download PDF
22. Prediction of Serum HIV-1 Neutralization Titers After Passive Administration of VRC01
- Author
-
Huang, Yunda, Zhang, Yuanyuan, Bailer, Robert, Grunenberg, Nicole, Carpp, Lindsay N., Seaton, Kelly, Mayer, Kenneth H., Ledgerwood, Julie, Corey, Lawrence, Mascola, John, Montefiori, David, and Gilbert, Peter B.
- Subjects
AIDS Vaccines ,Adult ,Male ,Adolescent ,Immunization, Passive ,HIV Infections ,HIV Antibodies ,Middle Aged ,Antibodies, Neutralizing ,Article ,Young Adult ,HIV Seropositivity ,HIV-1 ,Humans ,Female ,Immunization Schedule - Abstract
BACKGROUND: VRC01 is a human IgG1 broadly neutralizing antibody (bnAb) that binds to the HIV-1 envelope glycoprotein. It is being evaluated in two ongoing efficacy trials, the first assessment of a passively-administered bnAb for HIV-1 prevention. HVTN 104 was a Phase 1 trial of VRC01. SETTING: We measured serum concentrations and serum neutralization of VRC01 in 1079 longitudinal samples collected after passive administration of VRC01 in 84 HVTN 104 participants. As assays for measuring VRC01 serum neutralization titers are resource-intensive, we investigated approaches to predicting such titers. METHODS: Serum concentration was measured using an anti-idiotypic ELISA assay. Serum neutralization ID50 titers and in vitro neutralization potency IC50 of the VRC01 clinical lot were measured against Env-pseudoviruses. Three approaches were used to predict serum neutralization ID50 titers based on 1) observed serum concentration divided by IC50, 2) pharmacokinetics model-predicted serum concentration divided by IC50, and, 3) joint modeling of the longitudinal serum concentrations and ID50 titers. RESULTS: All three approaches yielded satisfactory prediction of neutralization titers against viruses of varied sensitivities; the median fold-differences (FDs) of observed-over-predicted ID50 titers were between 0.95 and 1.37. Approach 3 generally performed the best with FDs between 0.95 and 0.99, and
- Published
- 2020
23. Safety, pharmacokinetics, and immunological activities of multiple intravenous or subcutaneous doses of an anti-HIV monoclonal antibody, VRC01, administered to HIV-uninfected adults: Results of a phase 1 randomized trial
- Author
-
Mayer, Kenneth H., Seaton, Kelly E., Huang, Yunda, Grunenberg, Nicole, Isaacs, Abby, Allen, Mary, Ledgerwood, Julie E., Frank, Ian, Sobieszczyk, Magdalena E., Baden, Lindsey R., Rodriguez, Benigno, Van Tieu, Hong, Tomaras, Georgia D., Deal, Aaron, Goodman, Derrick, Bailer, Robert T., Ferrari, Guido, Jensen, Ryan, Hural, John, Graham, Barney S., Mascola, John R., Corey, Lawrence, and Montefiori, David C.
- Subjects
AIDS (Disease) -- Research ,AIDS research ,Monoclonal antibodies -- Testing ,HIV infection -- Care and treatment ,Biological sciences - Abstract
Background VRC01 is an HIV-1 CD4 binding site broadly neutralizing antibody (bnAb) that is active against a broad range of HIV-1 primary isolates in vitro and protects against simian-human immunodeficiency virus (SHIV) when delivered parenterally to nonhuman primates. It has been shown to be safe and well tolerated after short-term administration in humans; however, its clinical and functional activity after longer-term administration has not been previously assessed. Methods and findings HIV Vaccine Trials Network (HVTN) 104 was designed to evaluate the safety and tolerability of multiple doses of VRC01 administered either subcutaneously or by intravenous (IV) infusion and to assess the pharmacokinetics and in vitro immunologic activity of the different dosing regimens. Additionally, this study aimed to assess the effect that the human body has on the functional activities of VRC01 as measured by several in vitro assays. Eighty-eight healthy, HIV-uninfected, low-risk participants were enrolled in 6 United States clinical research sites affiliated with the HVTN between September 9, 2014, and July 15, 2015. The median age of enrollees was 27 years (range, 18-50); 52% were White (non-Hispanic), 25% identified as Black (non-Hispanic), 11% were Hispanic, and 11% were non-Hispanic people of diverse origins. Participants were randomized to receive the following: a 40 mg/kg IV VRC01 loading dose followed by five 20 mg/kg IV VRC01 doses every 4 weeks (treatment group 1 [T1], n = 20); eleven 5 mg/kg subcutaneous (SC) VRC01 (treatment group 3 [T3], n = 20); placebo (placebo group 3 [P3], n = 4) doses every 2 weeks; or three 40 mg/kg IV VRC01 doses every 8 weeks (treatment group 2 [T2], n = 20). Treatment groups T4 and T5 (n = 12 each) received three 10 or 30 mg/kg IV VRC01 doses every 8 weeks, respectively. Participants were followed for 32 weeks after their first VRC01 administration and received a total of 249 IV infusions and 208 SC injections, with no serious adverse events, dose-limiting toxicities, nor evidence for anti-VRC01 antibodies observed. Serum VRC01 levels were detected through 12 weeks after final administration in all participants who received all scheduled doses. Mean peak serum VRC01 levels of 1,177 [mu]g/ml (95% CI: 1,033, 1,340) and 420 [mu]g/ml (95% CI: 356, 494) were achieved 1 hour after the IV infusion series of 30 mg/kg and 10 mg/kg doses, respectively. Mean trough levels at week 24 in the IV infusion series of 30 mg/kg and 10 mg/kg doses, respectively, were 16 [mu]g/ml (95% CI: 10, 27) and 6 [mu]g/ml (95% CI: 5, 9) levels, which neutralize a majority of circulating strains in vitro (50% inhibitory concentration [IC50] > 5 [mu]g/ml). Post-infusion/injection serum VRC01 retained expected functional activity (virus neutralization, antibody-dependent cellular cytotoxicity, phagocytosis, and virion capture). The limitations of this study include the relatively small sample size of each VRC01 administration regimen and missing data from participants who were unable to complete all study visits. Conclusions VRC01 administered as either an IV infusion (10-40 mg/kg) given monthly or bimonthly, or as an SC injection (5 mg/kg) every 2 weeks, was found to be safe and well tolerated. In addition to maintaining drug concentrations consistent with neutralization of the majority of tested HIV strains, VRC01 concentrations from participants' sera were found to avidly capture HIV virions and to mediate antibody-dependent cellular phagocytosis, suggesting a range of anti-HIV immunological activities, warranting further clinical trials. Trial registration Clinical Trials Registration: NCT02165267, Author(s): Kenneth H. Mayer 1,*, Kelly E. Seaton 2, Yunda Huang 3, Nicole Grunenberg 3, Abby Isaacs 3, Mary Allen 4, Julie E. Ledgerwood 5, Ian Frank 6, Magdalena E. [...]
- Published
- 2017
- Full Text
- View/download PDF
24. Innate immune signatures to a partially-efficacious HIV vaccine predict correlates of HIV-1 infection risk.
- Author
-
Andersen-Nissen, Erica, Fiore-Gartland, Andrew, Ballweber Fleming, Lamar, Carpp, Lindsay N., Naidoo, Anneta F., Harper, Michael S., Voillet, Valentin, Grunenberg, Nicole, Laher, Fatima, Innes, Craig, Bekker, Linda-Gail, Kublin, James G., Huang, Ying, Ferrari, Guido, Tomaras, Georgia D., Gray, Glenda, Gilbert, Peter B., and McElrath, M. Juliana
- Subjects
AIDS vaccines ,INTERFERON gamma ,ANTIBODY-dependent cell cytotoxicity ,TYPE I interferons ,VACCINE effectiveness ,INTERFERON receptors - Abstract
The pox-protein regimen tested in the RV144 trial is the only vaccine strategy demonstrated to prevent HIV-1 infection. Subsequent analyses identified antibody and cellular immune responses as correlates of risk (CoRs) for HIV infection. Early predictors of these CoRs could provide insight into vaccine-induced protection and guide efforts to enhance vaccine efficacy. Using specimens from a phase 1b trial of the RV144 regimen in HIV-1-uninfected South Africans (HVTN 097), we profiled innate responses to the first ALVAC-HIV immunization. PBMC transcriptional responses peaked 1 day post-vaccination. Type I and II interferon signaling pathways were activated, as were innate pathways critical for adaptive immune priming. We then identified two innate immune transcriptional signatures strongly associated with adaptive immune CoR after completion of the 4-dose regimen. Day 1 signatures were positively associated with antibody-dependent cellular cytotoxicity and phagocytosis activity at Month 6.5. Conversely, a signature present on Days 3 and 7 was inversely associated with Env-specific CD4+ T cell responses at Months 6.5 and 12; rapid resolution of this signature was associated with higher Env-specific CD4+ T-cell responses. These are the first-reported early immune biomarkers of vaccine-induced responses associated with HIV-1 acquisition risk in humans and suggest hypotheses to improve HIV-1 vaccine regimens. Author summary: The innate immune response is the body's initial defense against pathogens and is linked to and shapes the subsequent adaptive immune response, which can confer long-lasting protection. For a vaccine with partial efficacy, such as the RV144 HIV vaccine regimen, identifying early innate responses that are linked with adaptive responses—particularly those for which evidence has accumulated that they might be important for protection—could help a more efficacious version be developed. In the HVTN 097 study, the RV144 prime-boost (ALVAC-HIV and AIDSVAX B/E) vaccine regimen was given to South African participants. We characterized the innate response to the first dose of ALVAC-HIV in these participants and identified gene expression signatures present within the first few days that were associated with antibody and T-cell responses to the full vaccine regimen measured up to 1 year later. As these antibody and T-cell responses have previously been implicated in protection, our findings suggest ways of refining the RV144 regimen and also have broader applications to vaccine development. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
25. Prediction of Serum HIV-1 Neutralization Titers After Passive Administration of VRC01.
- Author
-
Yunda Huang, Yuanyuan Zhang, Bailer, Robert, Grunenberg, Nicole, Carpp, Lindsay N., Seaton, Kelly, Mayer, Kenneth H., Ledgerwood, Julie, Corey, Lawrence, Mascola, John, Montefiori, David, and Gilbert, Peter B.
- Published
- 2020
- Full Text
- View/download PDF
26. Safety and immune responses after a 12-month booster in healthy HIV-uninfected adults in HVTN 100 in South Africa: A randomized double-blind placebo-controlled trial of ALVAC-HIV (vCP2438) and bivalent subtype C gp120/MF59 vaccines.
- Author
-
Laher, Fatima, Moodie, Zoe, Cohen, Kristen W., Grunenberg, Nicole, Bekker, Linda-Gail, Allen, Mary, Frahm, Nicole, Yates, Nicole L., Morris, Lynn, Malahleha, Mookho, Mngadi, Kathryn, Daniels, Brodie, Innes, Craig, Saunders, Kevin, Grant, Shannon, Yu, Chenchen, Gilbert, Peter B., Phogat, Sanjay, DiazGranados, Carlos A., and Koutsoukos, Marguerite
- Subjects
IMMUNIZATION of children ,IMMUNE response ,VACCINES ,NEUTRALIZATION tests ,AIDS vaccines ,CLINICAL trial registries ,PHAGOCYTOSIS ,INTRAMUSCULAR injections - Abstract
Background: HVTN 100 evaluated the safety and immunogenicity of an HIV subtype C pox-protein vaccine regimen, investigating a 12-month booster to extend vaccine-induced immune responses.Methods and Findings: A phase 1-2 randomized double-blind placebo-controlled trial enrolled 252 participants (210 vaccine/42 placebo; median age 23 years; 43% female) between 9 February 2015 and 26 May 2015. Vaccine recipients received ALVAC-HIV (vCP2438) alone at months 0 and 1 and with bivalent subtype C gp120/MF59 at months 3, 6, and 12. Antibody (IgG, IgG3 binding, and neutralizing) and CD4+ T-cell (expressing interferon-gamma, interleukin-2, and CD40 ligand) responses were evaluated at month 6.5 for all participants and at months 12, 12.5, and 18 for a randomly selected subset. The primary analysis compared IgG binding antibody (bAb) responses and CD4+ T-cell responses to 3 vaccine-matched antigens at peak (month 6.5 versus 12.5) and durability (month 12 versus 18) timepoints; IgG responses to CaseA2_gp70_V1V2.B, a primary correlate of risk in RV144, were also compared at these same timepoints. Secondary and exploratory analyses compared IgG3 bAb responses, IgG bAb breadth scores, neutralizing antibody (nAb) responses, antibody-dependent cellular phagocytosis, CD4+ polyfunctionality responses, and CD4+ memory sub-population responses at the same timepoints. Vaccines were generally safe and well tolerated. During the study, there were 2 deaths (both in the vaccine group and both unrelated to study products). Ten participants became HIV-infected during the trial, 7% (3/42) of placebo recipients and 3% (7/210) of vaccine recipients. All 8 serious adverse events were unrelated to study products. Less waning of immune responses was seen after the fifth vaccination than after the fourth, with higher antibody and cellular response rates at month 18 than at month 12: IgG bAb response rates to 1086.C V1V2, 21.0% versus 9.7% (difference = 11.3%, 95% CI = 0.6%-22.0%, P = 0.039), and ZM96.C V1V2, 21.0% versus 6.5% (difference = 14.5%, 95% CI = 4.1%-24.9%, P = 0.004). IgG bAb response rates to all 4 primary V1V2 antigens were higher 2 weeks after the fifth vaccination than 2 weeks after the fourth vaccination: 87.7% versus 75.4% (difference = 12.3%, 95% CI = 1.7%-22.9%, P = 0.022) for 1086.C V1V2, 86.0% versus 63.2% (difference = 22.8%, 95% CI = 9.1%-36.5%, P = 0.001) for TV1c8.2.C V1V2, 67.7% versus 44.6% (difference = 23.1%, 95% CI = 10.4%-35.7%, P < 0.001) for ZM96.C V1V2, and 81.5% versus 60.0% (difference = 21.5%, 95% CI = 7.6%-35.5%, P = 0.002) for CaseA2_gp70_V1V2.B. IgG bAb response rates to the 3 primary vaccine-matched gp120 antigens were all above 90% at both peak timepoints, with no significant differences seen, except a higher response rate to ZM96.C gp120 at month 18 versus month 12: 64.5% versus 1.6% (difference = 62.9%, 95% CI = 49.3%-76.5%, P < 0.001). CD4+ T-cell response rates were higher at month 18 than month 12 for all 3 primary vaccine-matched antigens: 47.3% versus 29.1% (difference = 18.2%, 95% CI = 2.9%-33.4%, P = 0.021) for 1086.C, 61.8% versus 38.2% (difference = 23.6%, 95% CI = 9.5%-37.8%, P = 0.001) for TV1.C, and 63.6% versus 41.8% (difference = 21.8%, 95% CI = 5.1%-38.5%, P = 0.007) for ZM96.C, with no significant differences seen at the peak timepoints. Limitations were that higher doses of gp120 were not evaluated, this study was not designed to investigate HIV prevention efficacy, and the clinical significance of the observed immunological effects is uncertain.Conclusions: In this study, a 12-month booster of subtype C pox-protein vaccines restored immune responses, and slowed response decay compared to the 6-month vaccination.Trial Registration: ClinicalTrials.gov NCT02404311. South African National Clinical Trials Registry (SANCTR number: DOH--27-0215-4796). [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
27. Antigenic competition in CD4+ T cell responses in a randomized, multicenter, double-blind clinical HIV vaccine trial.
- Author
-
Kallas, Esper G., Grunenberg, Nicole A., Yu, Chenchen, Manso, Bryce, Pantaleo, Giuseppe, Casapia, Martin, Baden, Lindsey R., Valencia, Javier, Sobieszczyk, Magdalena, Van Tieu, Hong, Allen, Mary, Hural, John, Graham, Barney S., Kublin, James, Gilbert, Peter B., Corey, Lawrence, Goepfert, Paul A., McElrath, M. Juliana, Johnson, R. Paul, and Huang, Yunda
- Subjects
AIDS vaccines ,RANDOMIZED response ,VACCINE effectiveness ,INTRAMUSCULAR injections ,VIRAL replication ,T cells ,IMMUNE response ,INTERLEUKIN-21 - Abstract
Troublesome T cells: Many HIV vaccines aim to generate robust T cell responses, but two new studies show that this is not always straightforward. Chamcha et al. analyzed data from several nonhuman primate vaccine studies with SIV/SHIV challenges and discovered that achieving a certain threshold of vaccine-induced IFN-γ
+ CD4 T cells lowered vaccine efficacy, possibly by providing target cells for the virus. Kallas et al. vaccinated people with HIV Gag/Pol alone or with Env to see whether antigenic competition could interfere with CD4 T cell responses; lower responses to Gag/Pol were detected when Env was also administered, indicating that including multiple antigens in a vaccine may preclude maximal T cell responses. Together, these studies highlight how we must tread carefully on the path to an effective HIV vaccine. T cell responses have been implicated in reduced risk of HIV acquisition in uninfected persons and control of viral replication in HIV-infected individuals. HIV Gag-specific T cells have been predominantly associated with post-infection control, whereas Env antigens are the target for protective antibodies; therefore, inclusion of both antigens is common in HIV vaccine design. However, inclusion of multiple antigens may provoke antigenic competition, reducing the potential effectiveness of the vaccine. HVTN 084 was a randomized, multicenter, double-blind phase 1 trial to investigate whether adding Env to a Gag/Pol vaccine decreases the magnitude or breadth of Gag/Pol-specific T cell responses. Fifty volunteers each received one intramuscular injection of 1 × 1010 particle units (PU) of rAd5 Gag/Pol and EnvA/B/C (3:1:1:1 mixture) or 5 × 109 PU of rAd5 Gag/Pol. CD4+ T cell responses to Gag/Pol measured 4 weeks after vaccination by cytokine expression were significantly higher in the group vaccinated without Env, whereas CD8+ T cell responses did not differ significantly between the two groups. Mapping of individual epitopes revealed greater breadth of the Gag/Pol-specific T cell response in the absence of Env compared to Env coimmunization. Addition of an Env component to a Gag/Pol vaccine led to reduced Gag/Pol CD4+ T cell response rate and magnitude as well as reduced epitope breadth, confirming the presence of antigenic competition. Therefore, T cell–based vaccine strategies should aim at choosing a minimalist set of antigens to reduce interference of individual vaccine components with the induction of the maximally achievable immune response. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
28. Immune correlates of the Thai RV144 HIV vaccine regimen in South Africa.
- Author
-
Gray, Glenda E., Huang, Ying, Grunenberg, Nicole, Laher, Fatima, Roux, Surita, Andersen-Nissen, Erica, De Rosa, Stephen C., Flach, Britta, Randhawa, April K., Jensen, Ryan, Swann, Edith M., Bekker, Linda-Gail, Innes, Craig, Lazarus, Erica, Morris, Lynn, Mkhize, Nonhlanhla N., Ferrari, Guido, Montefiori, David C., Shen, Xiaoying, and Sawant, Sheetal
- Subjects
AIDS vaccines ,ANTIBODY-dependent cell cytotoxicity ,HUMORAL immunity ,T cells ,ANTIBODY formation ,HIV ,IMMUNOGLOBULIN G - Abstract
Taking RV144 beyond Thailand: The RV144 vaccine trial in Thailand is the only HIV vaccine to show efficacy against HIV infection to date. Gray et al. designed the HVTN 097 trial to test this regimen in South Africa, where clade C HIV circulates; this clade is heterologous to the vaccine antigens. They intently examined immune protective responses previously identified in the RV144 trial and found that the vaccine seemed to be even more immunogenic in South Africans. CD4
+ T cell responses were stronger and more common in HVTN 097, and the magnitude of protective antibody responses was greater compared to RV144. Their results indicate that the RV144 regimen or others like it could be protective in areas where HIV is endemic. One of the most successful HIV vaccines to date, the RV144 vaccine tested in Thailand, demonstrated correlates of protection including cross-clade V1V2 immunoglobulin G (IgG) breadth, Env-specific CD4+ T cell polyfunctionality, and antibody-dependent cellular cytotoxicity (ADCC) in vaccinees with low IgA binding. The HIV Vaccine Trials Network (HVTN) 097 trial evaluated this vaccine regimen in South Africa, where clade C HIV-1 predominates. We compared cellular and humoral responses at peak and durability immunogenicity time points in HVTN 097 and RV144 vaccinee samples, and evaluated vaccine-matched and cross-clade immune responses. At peak immunogenicity, HVTN 097 vaccinees exhibited significantly higher cellular and humoral immune responses than RV144 vaccinees. CD4+ T cell responses were more frequent in HVTN 097 irrespective of age and sex, and CD4+ T cell Env-specific functionality scores were higher in HVTN 097. Env-specific CD40L+ CD4+ T cells were more common in HVTN 097, with individuals having this pattern of expression demonstrating higher median antibody responses to HIV-1 Env. IgG and IgG3 binding antibody rates and response magnitude to gp120 vaccine– and V1V2 vaccine–matched antigens were higher or comparable in HVTN 097 than in RV144 ADCC, and ADCP functional antibody responses were elicited in HVTN 097. Env-specific IgG and CD4+ Env responses declined significantly over time in both trials. Overall, cross-clade immune responses associated with protection were better than expected in South Africa, suggesting wider applicability of this regimen. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
29. Immunogenicity of a novel Clade B HIV-1 vaccine combination: Results of phase 1 randomized placebo controlled trial of an HIV-1 GM-CSF-expressing DNA prime with a modified vaccinia Ankara vaccine boost in healthy HIV-1 uninfected adults.
- Author
-
Buchbinder, Susan P., Grunenberg, Nicole A., Sanchez, Brittany J., Seaton, Kelly E., Ferrari, Guido, Moody, M. Anthony, Frahm, Nicole, Montefiori, David C., Hay, Christine M., Goepfert, Paul A., Baden, Lindsey R., Robinson, Harriet L., Yu, Xuesong, Gilbert, Peter B., McElrath, M. Juliana, Huang, Yunda, Tomaras, Georgia D., and null, null
- Subjects
- *
AIDS vaccines , *IMMUNOGENETICS , *RANDOMIZED controlled trials , *GRANULOCYTE-macrophage colony-stimulating factor , *DNA primers - Abstract
Background: A phase 1 trial of a clade B HIV vaccine in HIV-uninfected adults evaluated the safety and immunogenicity of a DNA prime co-expressing GM-CSF (Dg) followed by different numbers and intervals of modified vaccinia Ankara Boosts (M). Both vaccines produce virus-like particles presenting membrane-bound Env. Methods: Four US sites randomized 48 participants to receiving 1/10th the DNA dose as DgDgMMM given at 0, 2, 4, 6 and 8 months, or full dose DgDgM_M or DgDgMM_M regimens, given at 0, 2, 4, and 8 months, and 0, 2, 4, 6, and 10 months, respectively. Peak immunogenicity was measured 2 weeks post-last vaccination. Results: All regimens were well tolerated and safe. Full dose DgDgM_M and DgDgMM_M regimens generated Env-specific IgG to HIV-1 Env in >90%, IgG3 in >80%, and IgA in <20% of participants. Responses to gp140 and gp41 targets were more common and of higher magnitude than to gp120 and V1V2. The gp41 antibody included reactivity to the conserved immunodominant region with specificities known to mediate virus capture and phagocytosis and did not cross-react with a panel of intestinal flora antigens. The 3rd dose of MVA increased the avidity of elicited antibody (7.5% to 39%), the ADCC response to Bal gp120 (14% to 64%), and the one-year durability of the IgG3 responses to gp41 by 4-fold (13% vs. 3.5% retention of peak response). The co-expressed GM-CSF did not enhance responses over those in trials testing this vaccine without GM-CSF. Conclusion: This DNA/MVA prime-boost regimen induced durable, functional humoral responses that included ADCC, high antibody avidity, and Env IgG1 and IgG3 binding responses to the immunodominant region of gp41. The third, spaced MVA boost improved the overall quality of the antibody response. These products without co-expressed GM-CSF but combined with protein boosts will be considered for efficacy evaluation. Trial registration: ClinicalTrials.gov [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
30. Population pharmacokinetics analysis of VRC01, an HIV-1 broadly neutralizing monoclonal antibody, in healthy adults.
- Author
-
Huang, Yunda, Zhang, Lily, Ledgerwood, Julie, Grunenberg, Nicole, Bailer, Robert, Isaacs, Abby, Seaton, Kelly, Mayer, Kenneth H., Capparelli, Edmund, Corey, Larry, and Gilbert, Peter B.
- Published
- 2017
- Full Text
- View/download PDF
31. Uptake of Genital Mucosal Sampling in HVTN 097, a Phase 1b HIV Vaccine Trial in South Africa.
- Author
-
Lazarus, Erica Maxine, Otwombe, Kennedy, Adonis, Tania, Sebastian, Elaine, Gray, Glenda, Grunenberg, Nicole, Roux, Surita, Churchyard, Gavin, Innes, Craig, and Laher, Fatima
- Subjects
HIV infection transmission ,AIDS vaccines ,MUCOUS membranes ,IMMUNOPATHOLOGY ,CLINICAL trials - Abstract
: Because sexual transmission of HIV occurs across mucosal membranes, understanding the immune responses of the genital mucosa to vaccines may contribute knowledge to finding an effective candidate HIV vaccine. We describe the uptake of rectal secretion, cervical secretion and seminal mucosal secretion sampling amongst volunteers in a Phase 1b HIV vaccine trial. Age at screening, gender, study site and the designation of the person conducting the informed consent procedure were collected for volunteers who screened for the HVTN 097 study. A total of 211 volunteers (54% female) were screened at three sites in South Africa: Soweto (n = 70, 33%), Cape Town (n = 68, 32%) and Klerksdorp (n = 73, 35%). Overall uptake of optional mucosal sampling amongst trial volunteers was 71% (n = 149). Compared to Cape Town, volunteers from Soweto and Klerksdorp were less likely to consent to sampling (Soweto OR 0.08 CI: 0.03–0.25 p<0.001 and Klerksdorp OR 0.13 CI: 0.04–0.41 p = 0.001). In contrast, volunteers over 25 years of age were 2.39 times more likely to consent than younger volunteers (CI: 1.13–5.08, p = 0.02). Further studies are required to better understand the cultural, demographic and sociobehavioral factors which influence willingness to participate in mucosal sampling in HIV prevention studies. Trial Registration: ClinicalTrials.gov: [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
32. Single dose pharmacokinetics and absolute bioavailability of mirabegron, a ß3-adrenoceptor agonist for treatment of overactive bladder.
- Author
-
Eltink, Charlotte, Lee, Jennifer, Schaddelee, Marloes, Wenhui Zhang, Kerbusch, Virginie, Meijer, John, van Marle, Sjoerd, Grunenberg, Nicole, Kowalski, Donna, Drogendijk, Ted, Moy, Selina, Iitsuka, Hiromi, van Gelderen, Marcel, Matsushima, Hiroshi, and Sawamoto, Taiji
- Published
- 2012
- Full Text
- View/download PDF
33. HVTN 097: Evaluation of the RV144 Vaccine Regimen in HIV Uninfected South African Adults.
- Author
-
Andersen-Nissen, Erica, Grunenberg, Nicole, Huang, Ying, Roux, Surita, Laher, Fatima, Innes, Craig, Gu, Niya, DiazGranados, Carlos, Phogat, Sanjay, Lee, Carter, Swann, Edith, Kim, Jerome, O'Connell, Robert, Michael, Nelson, Flach, Britta, DeRosa, Steve, Frahm, Nicole, Morris, Lynn, Montefiori, David, and Gilbert, Peter
- Abstract
An abstract of the article "HVTN 097: Evaluation of the RV144 Vaccine Regimen in HIV Uninfected South African Adults" by Glenda E. Gray, Erica Andersen-Nissen, Fatima Laher and colleagues is presented.
- Published
- 2014
- Full Text
- View/download PDF
34. VLP-expressing DNA/MVA Vaccines: The Effect of Schedule and Regimen on Antibody Magnitude and Avidity.
- Author
-
Hay, Christine, Grunenberg, Nicole, Goepfert, Paul, Georgia, Tomaras, Seaton, Kelly, Sato, Alicia, Elizaga, Marnie, Yu, Xuesong, Gilbert, Peter, Moss, Bernard, and Robinson, Harriet
- Abstract
An abstract of the article "VLP-expressing DNA/MVA Vaccines: The Effect of Schedule and Regimen on Antibody Magnitude and Avidity" by Susan P. Buchbinder, Christine Hay, Nicole Grunenberg and colleagues is presented.
- Published
- 2014
- Full Text
- View/download PDF
35. Factors associated with reactogenicity to an investigational HIV vaccine regimen in HIV vaccine trials network 702.
- Author
-
Chihana, Rachel, Jin Kee, Jia, Moodie, Zoe, Huang, Yunda, Janes, Holly, Dadabhai, Sufia, Roxby, Alison C., Allen, Mary, Kassim, Sheetal, Naicker, Vimla, Innes, Craig, Naicker, Nivashnee, Dubula, Thozama, Grunenberg, Nicole, Malahleha, Mookho, Kublin, James G., Bekker, Linda-Gail, Gray, Glenda, Kumwenda, Johnstone, and Laher, Fatima
- Abstract
• Those assigned female at birth experience more reactogenicity. • Most symptoms are mild. • Pain and/or tenderness symptom common in both vaccine and placebo groups. Reactogenicity informs vaccine safety, and may influence vaccine uptake. We evaluated factors associated with reactogenicity in HVTN 702, a typical HIV vaccine efficacy trial with multiple doses and products. HVTN 702, a phase 2b/3 double-blind placebo-controlled trial, randomized 5404 African participants aged 18–35 years without HIV to placebo, or ALVAC-HIV (vCP2438) at months 0, 1 and ALVAC-HIV (vCP2438) + Bivalent Subtype C gp120/MF59 at months 3, 6, 12 and 18. Using multivariate logistic regression, we evaluated associations between reactogenicity with clinical, sociodemographic and laboratory variables. More vaccine than placebo-recipients reported local symptoms (all p < 0.001), arthralgia (p = 0.008), chills (p = 0.012) and myalgia (p < 0.001). Reactogenicity was associated with female sex at birth (OR v = 2.50, OR p = 1.81, both p < 0.001) and geographic region. Amongst vaccine-recipients, each year of age was associated with 3 % increase in reactogenicity (OR = 1.03, p = 0.002). Vaccine receipt, female sex at birth, older age, and region may affect reactogenicity. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
36. Antigenic competition in CD4 + T cell responses in a randomized, multicenter, double-blind clinical HIV vaccine trial.
- Author
-
Kallas EG, Grunenberg NA, Yu C, Manso B, Pantaleo G, Casapia M, Baden LR, Valencia J, Sobieszczyk M, Van Tieu H, Allen M, Hural J, Graham BS, Kublin J, Gilbert PB, Corey L, Goepfert PA, McElrath MJ, Johnson RP, Huang Y, and Frahm N
- Subjects
- Adolescent, Adult, CD8-Positive T-Lymphocytes immunology, Double-Blind Method, Epitopes immunology, Female, Humans, Male, Middle Aged, Vaccination, Young Adult, env Gene Products, Human Immunodeficiency Virus immunology, gag Gene Products, Human Immunodeficiency Virus immunology, AIDS Vaccines immunology, CD4-Positive T-Lymphocytes immunology, HIV Antigens immunology
- Abstract
T cell responses have been implicated in reduced risk of HIV acquisition in uninfected persons and control of viral replication in HIV-infected individuals. HIV Gag-specific T cells have been predominantly associated with post-infection control, whereas Env antigens are the target for protective antibodies; therefore, inclusion of both antigens is common in HIV vaccine design. However, inclusion of multiple antigens may provoke antigenic competition, reducing the potential effectiveness of the vaccine. HVTN 084 was a randomized, multicenter, double-blind phase 1 trial to investigate whether adding Env to a Gag/Pol vaccine decreases the magnitude or breadth of Gag/Pol-specific T cell responses. Fifty volunteers each received one intramuscular injection of 1 × 10
10 particle units (PU) of rAd5 Gag/Pol and EnvA/B/C (3:1:1:1 mixture) or 5 × 109 PU of rAd5 Gag/Pol. CD4+ T cell responses to Gag/Pol measured 4 weeks after vaccination by cytokine expression were significantly higher in the group vaccinated without Env, whereas CD8+ T cell responses did not differ significantly between the two groups. Mapping of individual epitopes revealed greater breadth of the Gag/Pol-specific T cell response in the absence of Env compared to Env coimmunization. Addition of an Env component to a Gag/Pol vaccine led to reduced Gag/Pol CD4+ T cell response rate and magnitude as well as reduced epitope breadth, confirming the presence of antigenic competition. Therefore, T cell-based vaccine strategies should aim at choosing a minimalist set of antigens to reduce interference of individual vaccine components with the induction of the maximally achievable immune response., (Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.