Filter by
-
Type
Scientific Publications
Evidence required to evaluate the use of bacteriologically confirmed asymptomatic tuberculosis disease as a primary endpoint in prevention of tuberculosis disease vaccine licensure trials
Richard G White, Gavin J Churchyard, Katherine C Horton, Andrew Fiore-Gartland, Marcel A Behr, Rebecca A Clark, Frank Cobelens, Joel D Ernst, Hanif Esmail, Alberto L Garcia-Basteiro, Sri Rezeki Hadinegoro, Willem A Hanekom, Mark Hatherill, Philip C Hill, Rudzani Muloiwa, Puck T Pelzer, Lele Rangaka, Helen Rees, Lewis Schrager, Margaret Stanley, Marta Tufet, Emily B Wong, Rein M G J Houben
Lancet Respir Med. 2025 Oct;13(10):933-942
Abstract
Current licensure trials of new vaccines to prevent tuberculosis disease use bacteriologically confirmed symptomatic tuberculosis disease as the primary endpoint. Globally, the incidence of symptomatic tuberculosis disease is low, making licensure trials large, long, and expensive. New data suggest that bacteriologically confirmed asymptomatic tuberculosis disease might occur more frequently than symptomatic tuberculosis disease. Therefore, if vaccines have efficacy against asymptomatic disease, tuberculosis vaccine licensure trials could include it in the primary endpoint, potentially leading to smaller or shorter trials. We describe the potential benefits and risks of this inclusion in the primary endpoint of tuberculosis vaccine licensure trials. We also simulate licensure trial endpoint accrual and summarise feedback from anonymous regulators and policy makers on the knowledge needed to consider this proposal and research studies needed to fill these evidence gaps. If bacteriologically confirmed asymptomatic tuberculosis disease could be included in the primary endpoint of tuberculosis disease licensure trials, it could lead to cheaper and more rapid tuberculosis vaccine development.
Scientific Publications
Augmented humoral responses to HIV Env trimers delivered as transmembrane immunogens by self-replicating RNA
Parisa Yousefpour, Amrit Raj Ghosh, Himanshi Chawla, Rachel Yeung, Justin Gregory, Kristen Si, Tanaka K Remba, Kristen A Rodrigues, Mariane B Melo, Jonathan Dye, Jon M Steichen, Yuebao Zhang, Yizhou Dong, Max Crispin, William R Schief, Facundo D Batista, Darrell J Irvine
Mol Ther. 2025 Oct 1;33(10):4858-4873
Abstract
mRNA vaccines have emerged as an important platform for vaccine development. Unlike protein subunit vaccines, mRNA-expressed antigens can be expressed in either secreted or transmembrane (TM) forms mimicking a viral envelope (Env) protein. Here, we investigated the impact of antigen expression format on the antigenicity profile, glycosylation, and immunogenicity of stabilized HIV Env trimer immunogens expressed from self-replicating RNA (replicon) vaccines. Replicon-encoded trimers in both forms exhibited proper folding, and replicon-expressed secreted trimers exhibited glycosylation patterns largely consistent with recombinant trimer protein, although with enrichment of complex glycans over high mannose at some sites. Both formats were highly immunogenic in mice, eliciting comparable serum antibody and T cell responses. Interestingly, the TM format initiated smaller germinal center (GC) responses, but these GCs were enriched for trimer-binding B cells compared to secreted trimer vaccines. In a B cell receptor knockin adoptive transfer model for assessing germline targeting, the replicon-encoded TM trimer elicited a greater frequency of epitope-targeting antibodies and recruited broadly neutralizing antibody precursor B cells to the GC response more efficiently compared to the replicon-encoded secreted trimer or protein trimer combined with adjuvant. These results indicate that the form of immunogen expression can impact key elements of immune responses to RNA vaccines.
Scientific Publications
Neutralizing and binding antibodies are a correlate of risk of COVID-19 in the CoVPN 3008 study in people with HIV
Nonhlanhla N Mkhize, Bo Zhang, Caroline Brackett, Peter James Elyanu, Asa Tapley, Sufia Dadabhai, Jiani Hu, Bich T N Do, Daniel J Schuster, Jack Heptinstall, Sheetal Sawant, Kelly Seaton, Marcella Sarzotti-Kelsoe, Aaron Hudson, Yutong Jin, Sinethemba Bhebhe, Haajira Kaldine, Prudence Kgagudi, Tandile Modise, Nyaradzo M Mgodi, Jessica Andriesen, April K Randhawa, Leigh H Fisher, Jia Jin Kee, Craig A Magaret, James Peng, Avi Kenny, Lindsay N Carpp, Zhe Chen, Siyu Heng, Manuel Villaran, Azwidihwi Takalani, Bert Le Roux, Eduan Wilkinson, Jackline Odhiambo, Parth Shah, Laura Polakowski, Margaret Yacovone, Taraz Samandari, Zvavahera Chirenje, Joseph Makhema, Ethel Kamuti, Katanekwa Njekwa, Harriet Nuwagaba-Biribonwoha, Allan Baguma, Sharlaa Badal-Faesen, William Brumskine, Soritha Coetzer, Rodney Dawson, Sinead Delany-Moretlwe, Andreas Henri Diacon, Samantha Fry, Katherine Gill, Anda Madikida, Zaheer Ahmed Ebrahim Hoosain, Mina C Hosseinipour, Mubiana Inambao, Craig Innes, Steve Innes, Dishiki Kalonji, Humphrey Mwape, Priya Kassim, Melvin C Kamanga, William Kilembe, Fatima Laher, Mookho Malahleha, Vongane Louisa Maluleke, Grace Mboya, Philister Adhiambo Madiega, Kirsten McHarry, Essack Mitha, Yajna Duki, Pamela Mda, Moroesi Moerane, Tumelo Moloantoa, Simpson Nuwamanya, Sharana Mahomed, Vimla Naicker, Anusha Nana, Annet Nanvubya, Barbarah Kawoozo, Maphoshane Nchabeleng, Walter Otieno, Elsje Louise Potgieter, Disebo Potloane, Zelda Punt, Jamil Said, Yashna Singh, Sheetal Kassim, Dorothie van der Vendt, Mohammed Siddique Tayob, Yacoob Vahed, Deo Ogema Wabwire, James G Kublin, Linda-Gail Bekker, Lawrence Corey, Glenda E Gray, Yunda Huang, Philip Kotze, Nigel Garrett, John Hural, Guido Ferrari, Erica Andersen-Nissen, David Montefiori, Penny L Moore, M Juliana McElrath, Georgia D Tomaras, Peter B Gilbert; CoVPN Study Team
Nat Commun. 2025 Oct 6;16(1):8876
Abstract
People with HIV (PWH) are understudied in COVID-19 vaccine trials, leaving knowledge gaps on whether the identified immune correlates of protection also hold in PWH. CoVPN 3008 (NCT05168813) enrolled predominantly PWH and reported lower COVID-19 incidence for a Hybrid vs. Vaccine Group (baseline SARS-CoV-2-positive and one mRNA-1273 dose vs. negative and two doses). Using case-cohort sampling, antibody markers at enrolment (M0) and four weeks post-final vaccination (Peak) are assessed as immune correlates of COVID-19. For the Hybrid Group [n = 287 (195 PWH)], all M0 markers inversely correlate with COVID-19 through 230 days post-Peak, with 50% inhibitory dilution BA.4/5 neutralizing antibody titer (nAb-ID50 BA.4/5) the strongest and only independent correlate (HR per 10-fold increase=0.46, 95% CI 0.28, 0.75; P = 0.002). For the Vaccine Group [n = 115 (86 PWH)], Peak nAb-ID50 BA.4/5 correlates with reduced COVID-19 risk (1.9%, 1.1%, and 0.3% at titers 10, 100, and 1000 AU/ml) through 92, but not 165, days post-Peak. Using multivariable Cox analysis of binding and nAb, nAb titers predict COVID-19 in PWH. Two doses of a 100-µg Ancestral strain mRNA vaccine in baseline-SARS-CoV-2-negative individuals elicit sufficient cross-reacting Omicron antibodies to reduce COVID-19 incidence for 90 days post-Peak, but viral evolution and waning antibodies abrogate this protection thereafter.
Scientific Publications
Common cold embecovirus imprinting primes broadly neutralizing antibody responses to SARS-CoV-2 S2
Siriruk Changrob, Atsuhiro Yasuhara, Suncheol Park, Sandhya Bangaru, Lei Li, Chloe A Troxell, Peter J Halfmann, Steven A Erickson, Nicholas J Catanzaro, Meng Yuan, Panpan Zhou, Min Huang, G Dewey Wilbanks, Joshua J C McGrath, Gagandeep Singh, Sean A Nelson, Yanbin Fu, Nai-Ying Zheng, Sofia M Carayannopoulos, Haley L Dugan, Dustin G Shaw, Christopher T Stamper, Maria Lucia L Madariaga, Florian Krammer, Raiees Andrabi, Dennis R Burton, Andrew B Ward, Ian A Wilson, Yoshihiro Kawaoka, Patrick C Wilson
J Exp Med. 2025 Dec 1;222(12):e20251146
doi: 10.1084/jem.20251146
Abstract
The S2 subunit of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike is highly conserved across coronavirus strains and therefore is a potential pan-coronavirus vaccine target. However, antibodies targeting this region are typically non-neutralizing. We report herein that S2-targeting antibodies from patients who recovered from SARS-CoV-2 infection bound only closely related sarbecovirus subgenus strains and, like most known S2 antibodies, none of these were neutralizing. In contrast, first-exposure, severe acutely infected COVID-19 patients predominantly induced back-boosted antibody-secreting cells imprinted against past common cold coronavirus strain OC43 that were cross-reactive to as many as five subgenera of betacoronavirus strains and gave rise to antibodies that were neutralizing and protective. The antibodies targeted two different sites: one defined by competition with stem helix antibodies, and the second to an underdescribed epitope at the apex of S2. These findings suggest that S2-targeted vaccines could strategically exploit controlled OC43 priming followed by SARS-CoV-2 boosting to enhance the breadth and quality of protective antibody responses.
Scientific Publications
Simultaneous priming of HIV broadly neutralizing antibody precursors to multiple epitopes by germline-targeting mRNA-LNP immunogens in mouse models
Zhenfei Xie, Xuesong Wang, Yu Yan, Jon M Steichen, Krystal M Ma, Christopher A Cottrell, Eleonora Melzi, Maria Bottermann, Paula Maldonado Villavicencio, Kimmo Rantalainen, Torben Schiffner, John E Warner, Stephanie R Weldon, Thavaleak Prum, Jordan R Ellis-Pugh, Jonathan L Torres, Abigail M Jackson, Claudia T Flynn, Gabriel Ozorowski, Sunny Himansu, Andrea Carfi, Andrew B Ward, Usha Nair, William R Schief, Facundo D Batista
Sci Immunol. 2025 Oct 17;10(112):eadu7961
Abstract
Germline-targeting is a promising approach to HIV vaccine development that begins with the elicitation of precursors to broadly neutralizing antibodies (bnAbs), but it remains unclear whether simultaneous elicitation of precursors to multiple epitopes on the HIV envelope (Env) would be inhibited by competition. This study used preclinical mouse models with physiologically relevant frequencies of bnAb precursor-bearing B cells to compare precursor elicitation by coadministration of multiple protein or mRNA lipid nanoparticle (mRNA-LNP) germline-targeting immunogens. These immunogens activate multiple bnAb precursor classes targeting distinct epitopes on Env but with evidence of potential competition. Simultaneous delivery of immunogens encoded by mRNA-LNPs, however, drove maturation across different precursor frequencies and immunogen doses. Furthermore, administration of a cocktail of mRNA-LNP immunogens (N332-GT5 gp151, ApexGT5 gp151, eOD-GT8 60mer, and 10E8-GT12 24mer) led to balanced activation of four distinct bnAb precursor classes, indicating that multiepitope HIV bnAb precursor priming might be successfully implemented in humans but might be immunogen dependent.
Scientific Publications
Simultaneous induction of multiple classes of broadly neutralizing antibody precursors by combination germline-targeting immunization in nonhuman primates
Henry J Sutton, Krystal M Ma, Jon M Steichen, Torben Schiffner, Tasha K Altheide, Alessia Liguori, Danny Lu, Michael Kubitz, Erik Georgeson, Nicole Phelps, Ryan Tingle, Nushin B Alavi, Elana Ben-Akiva, Xiaoya Zhou, Carolyne Kifude, Claudia T Flynn, Eva Rakasz, Darrell J Irvine, William R Schief, Shane Crotty
Sci Immunol. 2025 Oct 17;10(112):eadu8878
doi: 41105754/
Abstract
Inducing broadly neutralizing antibodies (bnAbs) against HIV remains a key challenge in vaccine development. Germline-targeting immunogens have effectively primed bnAb B cell lineages to individual HIV envelope epitopes in humans and nonhuman primates. However, eliciting consistent bnAb breadth requires the induction of multiple bnAb classes. We investigated whether immunization with a combination of germline-targeting immunogens could concurrently prime multiple bnAb lineages in nonhuman primates. Animals were immunized with three immunogens, targeting distinct epitopes: the V3-glycan/N332 supersite, the V2 Apex region, and the membrane-proximal external region (MPER), either individually or in combinations of two or all three. Triple combination immunization transiently reduced V2 Apex and V3-glycan responses, but by 8 weeks postboost, bnAb precursor lineages were observed to all three epitopes. Similar somatic hypermutation was observed across groups, indicative of permissive germinal center responses. These findings support combination germline-targeting immunization as a viable strategy to prime multiple bnAb lineages simultaneously.
Scientific Publications
Driving innovation from discovery to access: Meeting report of the 7th Global Forum on TB Vaccines (8-10 October 2024, Rio de Janeiro, Brazil)
Shaun Palmer, Rebecca A Clark, Bridgette J Connell, Vanessa Mwebaza Muwanga, Arthur Coelho, Paul Ogongo, Carly Young
Gates Open Res. 2025 Aug 27:9:65
Abstract
We urgently need novel, effective, and accessible vaccines to end tuberculosis (TB) as a public health crisis. The 7th Global Forum on TB Vaccines was convened from 8-10 October 2024 in Rio de Janeiro, Brazil. Under the theme of "Driving innovation from discovery to access," the program covered the breadth of TB vaccine research and development (R&D) through implementation, while underscoring the need for greater innovation and investments to advance development and ensure rapid, affordable, and equitable access. Participants shared the latest research on: approaches to diversify the TB vaccine pipeline, candidates advancing through late-stage trials toward licensure, and efforts to ensure new TB vaccines reach the populations that most need them. The forum provided a platform to learn from diverse experts across the field, including researchers, industry, funders, civil society, and affected communities. Participants examined cross-cutting enablers throughout, including opportunities to establish novel partnership and financing models, enhance open science, optimize R&D practices, and strengthen leadership and engagement with community members and high burden countries alike. In this report, we synthesize key themes and findings from the meeting, highlighting progress and priorities in the TB vaccine field.
Scientific Publications
HIV broadly neutralizing antibody precursors to the Apex epitope induced in nonhuman primates
Krystal M Ma, Henry J Sutton, Payal P Pratap, Jon M Steichen, Diane Carnathan, James Quinn, Oleksandr Kalyuzhniy, Alessia Liguori, Sashank Agrawal, Sabyasachi Baboo, Patrick Madden, Christopher A Cottrell, Jordan R Willis, Jeong-Hyun Lee, Elise Landais, Xiaozhen Hu, Parham Ramezani-Rad, Gabriel Ozorowski, Vanessa R Lewis, Jolene K Diedrich, Xiaoya Zhou, Tasha K Altheide, Nicole Phelps, Erik Georgeson, Nushin B Alavi, Danny Lu, Saman Eskandarzadeh, Michael Kubitz, Yumiko Adachi, Tina-Marie Mullen, Murillo Silva, Mariane B Melo, Sunny Himansu, Darrell J Irvine, Dennis R Burton, John R Yatesrd, James C Paulson, Devin Sok, Ian A Wilson, Guido Silvestri, Andrew B Ward, Shane Crotty, William R Schief
Sci Immunol. 2025 Aug 22;10(110):eadt6660
Abstract
An effective prophylactic HIV vaccine will likely need to induce broadly neutralizing antibodies (bnAbs). bnAbs to the Apex region of the HIV envelope glycoprotein (Env) are promising targets for vaccination because of their relatively low somatic hypermutation compared with other bnAbs. Most Apex bnAbs engage Env using an exceptionally long heavy-chain complementarity-determining region 3 (HCDR3) containing specific binding motifs, which reduces bnAb precursor frequency and makes priming of rare bnAb precursors a likely limiting step in the path to Apex bnAb induction. We found that adjuvanted protein or mRNA lipid nanoparticle (LNP) immunization of rhesus macaques with ApexGT6, an Env trimer engineered to bind Apex bnAb precursors, consistently induced Apex bnAb-related precursors with long HCDR3s bearing bnAb-like sequence motifs. Cryo-electron microscopy revealed that elicited Apex bnAb-related HCDR3s had structures combining elements of several prototype Apex bnAbs. These results achieve a critical HIV vaccine development milestone in outbred primates.
Scientific Publications
Structural insights into VRC01-class bnAb precursors with diverse light chains elicited in the IAVI G001 human vaccine trial
Xiaohe Lin, Christopher A Cottrell, Oleksandr Kalyuzhniy, Ryan Tingle, Michael Kubitz, Danny Lu, Meng Yuan, William R Schief, Ian A Wilson
Proc Natl Acad Sci U S A. 2025 Aug 19;122(33):e2510163122
Abstract
The development of germline-targeting vaccines represents a potentially transformative strategy to elicit broadly neutralizing antibodies (bnAbs) against HIV and other antigenically diverse pathogens. Here, we report on structural characterization of vaccine-elicited VRC01-class bnAb precursors in the IAVI G001 Phase 1 clinical trial with the eOD-GT8 60mer nanoparticle as immunogen. High-resolution X-ray structures of eOD-GT8 monomer complexed with Fabs of five VRC01-class bnAb precursors with >90% germline identity revealed a conserved mode of binding to the HIV CD4-binding site via IGHV1-2-encoded heavy chains, mirroring mature bnAb interactions. The light-chain V-gene diversity emulated VRC01 bnAbs and stabilized antigen engagement, while their conserved five-residue LCDR3 motifs prevented steric clashes. Notably, the VRC01-class bnAb precursors accommodated the N276 glycan, a key barrier in HIV Env recognition, through structural rearrangements in HCDR3 or LCDR1, despite its absence in the immunogen. Surface plasmon resonance analysis showed that 87% of elicited antibodies retained glycan binding capacity, albeit with reduced affinity. These findings validate the ability of eOD-GT8 60mer nanoparticles to prime VRC01-class bnAb precursors with native-like paratopes but with intrinsic glycan adaptability. Structural mimicry of mature bnAbs was observed even with limited somatic hypermutation, indicating that critical features are encoded in the germline repertoire. The structures highlight how germline-encoded features drive bnAb-like recognition at early stages. This work provides molecular evidence supporting germline targeting in humans and provides guidance for designing booster immunogens to shepherd affinity maturation toward broad neutralization.
Scientific Publications
Precise targeting of HIV broadly neutralizing antibody precursors in humans
Tom G Caniels, Madhu Prabhakaran, Gabriel Ozorowski, Kellie J MacPhee, Weiwei Wu, Karlijn van der Straten, Sashank Agrawal, Ronald Derking, Emma I M M Reiss, Katrina Millard, Martina Turroja, Aimee Desrosiers, Jeffrey Bethony, Elissa Malkin, Marinus H Liesdek, Annelou van der Veen, Michelle Klouwens, Jonne L Snitselaar, Joey H Bouhuijs, Rhianna Bronson, Jalen Jean-Baptiste, Suprabhath Gajjala, Zahra Rikhtegaran Tehrani, Alison Benner, Mukundhan Ramaswami, Michael O Duff, Yung-Wen Liu, Alicia H Sato, Ju Yeong Kim, Isabel J L Baken, Catarina Mendes Silva, Tom P L Bijl, Jacqueline van Rijswijk, Judith A Burger, Albert Cupo, Anila Yasmeen, Swastik Phulera, Wen-Hsin Lee, Kipchoge N Randall Jr, Shiyu Zhang, Martin M Corcoran, Isabel Regadas, Alex C Sullivan, David M Brown, Jennifer A Bohl, Kelli M Greene, Hongmei Gao, Nicole L Yates, Sheetal Sawant, Jan M Prins, Neeltje A Kootstra, Stephen M Kaminsky, Burc Barin, Farhad Rahaman, Margaret Meller, Vince Philiponis, Dagna S Laufer, Angela Lombardo, Lindsey Mwoga, Solmaz Shotorbani, Drienna Holman, Richard A Koup, Per Johan Klasse, Gunilla B Karlsson Hedestam, Georgia D Tomaras, Marit J van Gils, David C Montefiori, Adrian B McDermott, Ollivier Hyrien, John P Moore, Ian A Wilson, Andrew B Ward, David J Diemert, Godelieve J de Bree, Sarah F Andrews, Marina Caskey, Rogier W Sanders
Science. 2025 Jul 31;389(6759):eadv5572
Abstract
A protective HIV vaccine will need to induce broadly neutralizing antibodies (bnAbs) in humans, but priming rare bnAb precursor B cells has been challenging. In a double-blinded, placebo-controlled phase 1 human clinical trial, the recombinant, germline-targeting envelope glycoprotein (Env) trimer BG505 SOSIP.v4.1-GT1.1, adjuvanted with AS01B, induced bnAb precursors of the VRC01-class at a high frequency in the majority of vaccine recipients. These bnAb precursors, which target the CD4 receptor binding site, had undergone somatic hypermutation characteristic of the VRC01-class. A subset of isolated VRC01-class monoclonal antibodies neutralized wild-type pseudoviruses and was structurally extremely similar to bnAb VRC01. These results further support germline-targeting approaches for human HIV vaccine design and demonstrate atomic-level manipulation of B cell responses with rational vaccine design.
Scientific Publications
Vaccination with mRNA-encoded membrane-anchored HIV envelope trimers elicited tier 2 neutralizing antibodies in a phase 1 clinical trial
K Rachael Parks, Zoe Moodie, Mary A Allen, Catherine Yen, Briana D Furch, Kellie J MacPhee, Gabriel Ozorowski, Jack Heptinstall, William O Hahn, Zihan Zheng, Huiyin Lu, Shannon Grant, Elize Domin, Michael O Duff, Aaron Seese, Constanza Marini-Macouzet, Lamar Ballweber-Fleming, Wen-Hsin Lee, Christopher A Cottrell, Alessia Liguori, Erik Georgeson, Nushin Alavi, Michael Kubitz, Nicole Phelps, Kelly E Seaton, Kristen W Cohen, Maija A Anderson, Kajari Mondal, Dagna S Laufer, James G Kublin, Andrew B Ward, Ollivier Hyrien, Stephen C De Rosa, Sunny Himansu, Brett Leav, Caroline Reuter, Georgia D Tomaras, David Montefiori, Stephen R Walsh, Ian Frank, Magdalena E Sobieszczyk, Paul A Goepfert, Kathryn E Stephenson, Lindsey R Baden, Hong Van Tieu, Michael C Keefer, Jesse Clark, Sharon A Riddler, William R Schief, M Juliana McElrath
Sci Transl Med. 2025 Jul 30;17(809):eadw0721
Abstract
A protective vaccine against human immunodeficiency virus (HIV) will likely need to induce broadly neutralizing antibodies (bnAbs) that engage relatively conserved epitopes on the HIV envelope glycoprotein (Env) trimer. Nearly all vaccine strategies to induce bnAbs require the use of complex immunization regimens involving a series of different immunogens, most of which are Env trimers. Producing protein-based clinical material to evaluate such relatively complex regimens in humans presents major challenges in cost and time. Furthermore, immunization with HIV trimers as soluble proteins induces strong nonneutralizing responses to the trimer base, which is normally occluded on the virion. These base responses could potentially detract from the elicitation of nAbs and the eventual induction of bnAbs. mRNA vaccine platforms offer potential advantages over protein delivery for HIV vaccine development, including increased production speed, reduced cost, and the ability to deliver membrane-bound trimers that might facilitate improved immuno-focusing to nonbase epitopes. We report the design of mRNA-delivered soluble and membrane-bound forms of a stabilized native-like Env trimer (BG505 MD39.3); initial immunogenicity evaluation in rabbits that triggered clinical evaluation; and more comprehensive evaluation of B cell, T cell, and antibody responses in nonhuman primates. mRNA-encoded membrane-bound Env immunization elicited reduced off-target base-directed Env responses and stronger nAb responses compared with mRNA-encoded soluble Env. Overall, mRNA delivery of membrane-bound Env appears promising for enhancing B cell responses to subdominant epitopes and facilitating rapid translation to clinical testing, which should assist HIV vaccine development.
Scientific Publications
Vaccination with an mRNA-encoded membrane-bound HIV envelope trimer induces neutralizing antibodies in animal models
Parham Ramezani-Rad, Christopher A Cottrell, Ester Marina-Zárate, Alessia Liguori, Elise Landais, Jonathan L Torres, Amber Myers, Jeong Hyun Lee, Sabyasachi Baboo, Claudia Flynn, Katherine McKenney, Eugenia Salcedo, Xiaoya Zhou, Oleksandr Kalyuzhniy, Erik Georgeson, Nicole Phelps, Danny Lu, Saman Eskandarzadeh, Sergey Menis, Michael Kubitz, Bettina Groschel, Nushin Alavi, Abigail M Jackson, Wen-Hsin Lee, Andy S Tran, Elana Ben-Akiva, Katarzyna Kaczmarek Michaels, Jolene K Diedrich, Chiamaka A Enemuo, Vanessa Lewis, Arpan Pradhan, Sudhir Pai Kasturi, Torben Schiffner, Jon M Steichen, Diane G Carnathan, Sunny Himansu, John R Yatesrd, James C Paulson, Gabriel Ozorowski, Darrell J Irvine, Guido Silvestri, Devin Sok, Andrew B Ward, Shane Crotty, William R Schief
Sci Transl Med. 2025 Jul 30;17(809):eadw0721
Abstract
A protective vaccine against human immunodeficiency virus (HIV) will likely need to induce broadly neutralizing antibodies (bnAbs) that engage relatively conserved epitopes on the HIV envelope glycoprotein (Env) trimer. Nearly all vaccine strategies to induce bnAbs require the use of complex immunization regimens involving a series of different immunogens, most of which are Env trimers. Producing protein-based clinical material to evaluate such relatively complex regimens in humans presents major challenges in cost and time. Furthermore, immunization with HIV trimers as soluble proteins induces strong nonneutralizing responses to the trimer base, which is normally occluded on the virion. These base responses could potentially detract from the elicitation of nAbs and the eventual induction of bnAbs. mRNA vaccine platforms offer potential advantages over protein delivery for HIV vaccine development, including increased production speed, reduced cost, and the ability to deliver membrane-bound trimers that might facilitate improved immuno-focusing to nonbase epitopes. We report the design of mRNA-delivered soluble and membrane-bound forms of a stabilized native-like Env trimer (BG505 MD39.3); initial immunogenicity evaluation in rabbits that triggered clinical evaluation; and more comprehensive evaluation of B cell, T cell, and antibody responses in nonhuman primates. mRNA-encoded membrane-bound Env immunization elicited reduced off-target base-directed Env responses and stronger nAb responses compared with mRNA-encoded soluble Env. Overall, mRNA delivery of membrane-bound Env appears promising for enhancing B cell responses to subdominant epitopes and facilitating rapid translation to clinical testing, which should assist HIV vaccine development.
Scientific Publications
Optimizing human B cell repertoire analyses to interpret clinical data and design sequential HIV vaccines
Milton Maciel Jr, Lindsey R Baden, Brian Briney, Nicole A Doria-Rose, Kasalina N Kiwanuka, Shelly J Krebs, Angela Malaspina, Georgia D Tomaras, Andrew Ward, Gunilla B Karlsson Hedestam, Wilton B Williams, M Patricia D'Souza
NPJ Vaccines. 2025 Jul 17;10(1):157
Abstract
Advances in HIV vaccine development focus on eliciting broadly neutralizing antibodies (bNAbs) through next-generation immunogens. Human trials are testing these candidates for their ability to initiate B cell maturation. This workshop report reviews emerging data and methods for analyzing B cell repertoires, highlighting strategies to streamline these labor-intensive processes. The goal is to enable timely, cost-effective design of sequential immunization regimens that induce potent, protective bNAbs against HIV.