Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Julie E. Ledgerwood is active.

Publication


Featured researches published by Julie E. Ledgerwood.


Science | 2013

Protection Against Malaria by Intravenous Immunization with a Nonreplicating Sporozoite Vaccine

Robert A. Seder; Lee Jah Chang; Mary E. Enama; Kathryn L. Zephir; Uzma N. Sarwar; Ingelise J. Gordon; LaSonji A. Holman; Eric R. James; Peter F. Billingsley; Anusha Gunasekera; Adam Richman; Sumana Chakravarty; Anita Manoj; Soundarapandian Velmurugan; Minglin Li; Adam Ruben; Tao Li; Abraham G. Eappen; Richard E. Stafford; Sarah Plummer; Cynthia S. Hendel; Laura Novik; Pamela Costner; Floreliz Mendoza; Jamie G. Saunders; Martha Nason; Jason H. Richardson; Jittawadee Murphy; Silas A. Davidson; Thomas L. Richie

Malaria Sporozoite Vaccine Each year, hundreds of millions of people are infected with Plasmodium falciparum, the mosquito-borne parasite that causes malaria. A preventative vaccine is greatly needed. Seder et al. (p. 1359, published online 8 August; see the Perspective by Good) now report the results from a phase I clinical trial where subjects were immunized intravenously with a whole, attenuated sporozoite vaccine. Three of 9 subjects who received four doses and zero of 6 subjects who received five doses of the vaccine went on to develop malaria after controlled malaria infection. Both antibody titers and cellular immune responses correlated positively with the dose of vaccine received, suggesting that both arms of the adaptive immune response may have participated in the observed protection. Intravenous immunization with an attenuated whole malaria sporozoite vaccine protected volunteers in a phase I clinical trial. [Also see Perspective by Good] Consistent, high-level, vaccine-induced protection against human malaria has only been achieved by inoculation of Plasmodium falciparum (Pf) sporozoites (SPZ) by mosquito bites. We report that the PfSPZ Vaccine—composed of attenuated, aseptic, purified, cryopreserved PfSPZ—was safe and wel-tolerated when administered four to six times intravenously (IV) to 40 adults. Zero of six subjects receiving five doses and three of nine subjects receiving four doses of 1.35 × 105 PfSPZ Vaccine and five of six nonvaccinated controls developed malaria after controlled human malaria infection (P = 0.015 in the five-dose group and P = 0.028 for overall, both versus controls). PfSPZ-specific antibody and T cell responses were dose-dependent. These data indicate that there is a dose-dependent immunological threshold for establishing high-level protection against malaria that can be achieved with IV administration of a vaccine that is safe and meets regulatory standards.


The New England Journal of Medicine | 2017

Chimpanzee Adenovirus Vector Ebola Vaccine - Preliminary Report.

Julie E. Ledgerwood; Adam DeZure; Daphne Stanley; Laura Novik; Mary E. Enama; Nina M. Berkowitz; Zonghui Hu; Gyan Joshi; Aurélie Ploquin; Sandra Sitar; Ingelise J. Gordon; Sarah A. Plummer; LaSonji A. Holman; Cynthia S. Hendel; Galina Yamshchikov; François Roman; Alfredo Nicosia; Stefano Colloca; Riccardo Cortese; Robert T. Bailer; Richard M. Schwartz; Mario Roederer; John R. Mascola; Richard A. Koup; Nancy J. Sullivan; Barney S. Graham; Abstr Act

Background The unprecedented 2014 epidemic of Ebola virus disease (EVD) prompted an international response to accelerate the availability of a preventive vaccine. A replication‐defective recombinant chimpanzee adenovirus type 3–vectored ebolavirus vaccine (cAd3‐EBO), encoding the glycoprotein from Zaire and Sudan species, that offers protection in the nonhuman primate model, was rapidly advanced into phase 1 clinical evaluation. Methods We conducted a phase 1, dose‐escalation, open‐label trial of cAd3‐EBO. Twenty healthy adults, in sequentially enrolled groups of 10 each, received vaccination intramuscularly in doses of 2×1010 particle units or 2×1011 particle units. Primary and secondary end points related to safety and immunogenicity were assessed throughout the first 8 weeks after vaccination; in addition, longer‐term vaccine durability was assessed at 48 weeks after vaccination. Results In this small study, no safety concerns were identified; however, transient fever developed within 1 day after vaccination in two participants who had received the 2×1011 particle‐unit dose. Glycoprotein‐specific antibodies were induced in all 20 participants; the titers were of greater magnitude in the group that received the 2×1011 particle‐unit dose than in the group that received the 2×1010 particle‐unit dose (geometric mean titer against the Zaire antigen at week 4, 2037 vs. 331; P=0.001). Glycoprotein‐specific T‐cell responses were more frequent among those who received the 2×1011 particle‐unit dose than among those who received the 2×1010 particle‐unit dose, with a CD4 response in 10 of 10 participants versus 3 of 10 participants (P=0.004) and a CD8 response in 7 of 10 participants versus 2 of 10 participants (P=0.07) at week 4. Assessment of the durability of the antibody response showed that titers remained high at week 48, with the highest titers in those who received the 2×1011 particle‐unit dose. Conclusions Reactogenicity and immune responses to cAd3‐EBO vaccine were dose‐dependent. At the 2×1011 particle‐unit dose, glycoprotein Zaire–specific antibody responses were in the range reported to be associated with vaccine‐induced protective immunity in challenge studies involving nonhuman primates, and responses were sustained to week 48. Phase 2 studies and efficacy trials assessing cAd3‐EBO are in progress. (Funded by the Intramural Research Program of the National Institutes of Health; VRC 207 ClinicalTrials.gov number, NCT02231866.)


Journal of Virology | 2011

Mechanism of Neutralization by the Broadly Neutralizing HIV-1 Monoclonal Antibody VRC01

Yuxing Li; Sijy O'Dell; Laura M. Walker; Xueling Wu; Javier Guenaga; Yu Feng; Stephen D. Schmidt; Krisha McKee; Mark K. Louder; Julie E. Ledgerwood; Barney S. Graham; Barton F. Haynes; Dennis R. Burton; Richard T. Wyatt; John R. Mascola

ABSTRACT The structure of VRC01 in complex with the HIV-1 gp120 core reveals that this broadly neutralizing CD4 binding site (CD4bs) antibody partially mimics the interaction of the primary virus receptor, CD4, with gp120. Here, we extended the investigation of the VRC01-gp120 core interaction to the biologically relevant viral spike to better understand the mechanism of VRC01-mediated neutralization and to define viral elements associated with neutralization resistance. In contrast to the interaction of CD4 or the CD4bs monoclonal antibody (MAb) b12 with the HIV-1 envelope glycoprotein (Env), occlusion of the VRC01 epitope by quaternary constraints was not a major factor limiting neutralization. Mutagenesis studies indicated that VRC01 contacts within the gp120 loop D, the CD4 binding loop, and the V5 region were necessary for optimal VRC01 neutralization, as suggested by the crystal structure. In contrast to interactions with the soluble gp120 monomer, VRC01 interaction with the native viral spike did not occur in a CD4-like manner; VRC01 did not induce gp120 shedding from the Env spike or enhance gp41 membrane proximal external region (MPER)-directed antibody binding to the Env spike. Finally, VRC01 did not display significant reactivity with human antigens, boding well for potential in vivo applications. The data indicate that VRC01 interacts with gp120 in the context of the functional spike in a manner distinct from that of CD4. It achieves potent neutralization by precisely targeting the CD4bs without requiring alterations of Env spike configuration and by avoiding steric constraints imposed by the quaternary structure of the functional Env spike.


Science Translational Medicine | 2015

Virologic effects of broadly neutralizing antibody VRC01 administration during chronic HIV-1 infection

Rebecca M. Lynch; Eli Boritz; Emily E. Coates; Adam DeZure; Patrick Madden; Pamela Costner; Mary E. Enama; Sarah Plummer; LaSonji A. Holman; Cynthia S. Hendel; Ingelise J. Gordon; Joseph P. Casazza; Michelle Conan-Cibotti; Stephen A. Migueles; Randall Tressler; Robert T. Bailer; Adrian B. McDermott; Sandeep Narpala; Sijy O’Dell; Gideon Wolf; Jeffrey D. Lifson; Brandie A. Freemire; Robert J. Gorelick; Janardan P. Pandey; Sarumathi Mohan; Nicolas Chomont; Rémi Fromentin; Tae-Wook Chun; Anthony S. Fauci; Richard M. Schwartz

A single infusion with broadly neutralizing antibody VRC01 resulted in lowered plasma virus load in HIV-1–infected subjects. Passive aggression for HIV Antibodies that neutralize HIV could add to the therapeutic arsenal to prevent and treat disease. Lynch et al. have now tested one such antibody—VRC01—in HIV-infected individuals. Although little difference was observed in viral reservoir in individuals on antiretroviral therapy, plasma viremia was reduced in untreated subjects with a single infusion of VRC01, preferentially suppressing neutralization-sensitive strains. Passive immunization with neutralizing antibodies could therefore aid in viral suppression in HIV-infected individuals. Passive immunization with HIV-1–neutralizing monoclonal antibodies (mAbs) is being considered for prevention and treatment of HIV-1 infection. As therapeutic agents, mAbs could be used to suppress active virus replication, maintain suppression induced by antiretroviral therapy (ART), and/or decrease the size of the persistent virus reservoir. We assessed the impact of VRC01, a potent human mAb targeting the HIV-1 CD4 binding site, on ART-treated and untreated HIV-1–infected subjects. Among six ART-treated individuals with undetectable plasma viremia, two infusions of VRC01 did not reduce the peripheral blood cell–associated virus reservoir measured 4 weeks after the second infusion. In contrast, six of eight ART-untreated, viremic subjects infused with a single dose of VRC01 experienced a 1.1 to 1.8 log10 reduction in plasma viremia. The two subjects with minimal responses to VRC01 were found to have predominantly VRC01-resistant virus before treatment. Notably, two subjects with plasma virus load <1000 copies/ml demonstrated virus suppression to undetectable levels for over 20 days until VRC01 levels declined. Among the remaining four subjects with baseline virus loads between 3000 and 30,000 copies, viremia was only partially suppressed by mAb infusion, and we observed strong selection pressure for the outgrowth of less neutralization-sensitive viruses. In summary, a single infusion of mAb VRC01 significantly decreased plasma viremia and preferentially suppressed neutralization-sensitive virus strains. These data demonstrate the virological effect of this neutralizing antibody and highlight the need for combination strategies to maintain virus suppression.


Lancet Infectious Diseases | 2011

DNA priming and influenza vaccine immunogenicity: two phase 1 open label randomised clinical trials

Julie E. Ledgerwood; Chih-Jen Wei; Zonghui Hu; Ingelise J. Gordon; Mary E. Enama; Cynthia S. Hendel; Patrick M. McTamney; Melissa B. Pearce; Hadi M. Yassine; Jeffrey C. Boyington; Robert T. Bailer; Terrence M. Tumpey; Richard A. Koup; John R. Mascola; Gary J. Nabel; Barney S. Graham

Summary Background Because the general population is largely naive to H5N1 influenza, antibodies generated to H5 allow analysis of novel influenza vaccines independent of background immunity from previous infection. We assessed the safety and immunogenicity of DNA encoding H5 as a priming vaccine to improve antibody responses to inactivated influenza vaccination. Methods In VRC 306 and VRC 310, two sequentially enrolled phase 1, open-label, randomised clinical trials, healthy adults (age 18–60 years) were randomly assigned to receive intramuscular H5 DNA (4 mg) at day 0 or twice, at day 0 and week 4, followed by H5N1 monovalent inactivated vaccine (MIV; 90 μg) at 4 or 24 weeks, and compared with a two-dose regimen of H5N1 MIV with either a 4 or 24 week interval. Antibody responses were assessed by haemagglutination inhibition (HAI), ELISA, neutralisation (ID80), and immunoassays for stem-directed antibodies. T cell responses were assessed by intracellular cytokine staining. After enrolment, investigators and individuals were not masked to group assignment. VRC 306 and VRC 310 are registered with ClinicalTrials.gov, numbers NCT00776711 and NCT01086657, respectively. Findings In VRC 306, 60 individuals were randomly assigned to the four groups (15 in each) and 59 received the vaccinations. In VRC 310, of the 21 individuals enrolled, 20 received the vaccinations (nine received a two-dose regimen of H5N1 MIV and 11 received H5 DNA at day 0 followed by H5N1 MIV at week 24). H5 DNA priming was safe and enhanced H5-specific antibody titres following an H5N1 MIV boost, especially when the interval between DNA prime and MIV boost was extended to 24 weeks. In the two studies, DNA priming with a 24-week MIV boost interval induced protective HAI titres in 21 (81%) of 26 of individuals, with an increase in geometric mean titre (GMT) of more than four times that of individuals given the MIV-MIV regimen at 4 or 24 weeks (GMT 103–206 vs GMT 27–33). Additionally, neutralising antibodies directed to the conserved stem region of H5 were induced by this prime-boost regimen in several individuals. No vaccine-related serious adverse events were recorded. Interpretation DNA priming 24 weeks in advance of influenza vaccine boosting increased the magnitude of protective antibody responses (HAI) and in some cases induced haemagglutinin-stem-specific neutralising antibodies. A DNA-MIV vaccine regimen could enhance the efficacy of H5 or other influenza vaccines and shows that anti-stem antibodies can be elicited by vaccination in man. Funding National Institutes of Health.


PLOS ONE | 2010

Priming Immunization with DNA Augments Immunogenicity of Recombinant Adenoviral Vectors for Both HIV-1 Specific Antibody and T-Cell Responses

Richard A. Koup; Mario Roederer; Laurie Lamoreaux; Jennifer Fischer; Laura Novik; Martha Nason; Brenda D. Larkin; Mary E. Enama; Julie E. Ledgerwood; Robert T. Bailer; John R. Mascola; Gary J. Nabel; Barney S. Graham; Vrc Study Teams

Background Induction of HIV-1-specific T-cell responses relevant to diverse subtypes is a major goal of HIV vaccine development. Prime-boost regimens using heterologous gene-based vaccine vectors have induced potent, polyfunctional T cell responses in preclinical studies. Methods The first opportunity to evaluate the immunogenicity of DNA priming followed by recombinant adenovirus serotype 5 (rAd5) boosting was as open-label rollover trials in subjects who had been enrolled in prior studies of HIV-1 specific DNA vaccines. All subjects underwent apheresis before and after rAd5 boosting to characterize in depth the T cell and antibody response induced by the heterologous DNA/rAd5 prime-boost combination. Results rAd5 boosting was well-tolerated with no serious adverse events. Compared to DNA or rAd5 vaccine alone, sequential DNA/rAd5 administration induced 7-fold higher magnitude Env-biased HIV-1-specific CD8+ T-cell responses and 100-fold greater antibody titers measured by ELISA. There was no significant neutralizing antibody activity against primary isolates. Vaccine-elicited CD4+ and CD8+ T-cells expressed multiple functions and were predominantly long-term (CD127+) central or effector memory T cells and that persisted in blood for >6 months. Epitopes mapped in Gag and Env demonstrated partial cross-clade recognition. Conclusion Heterologous prime-boost using vector-based gene delivery of vaccine antigens is a potent immunization strategy for inducing both antibody and T-cell responses. Trial Registration ClinicalTrails.gov NCT00102089, NCT00108654


The New England Journal of Medicine | 2017

A Recombinant Vesicular Stomatitis Virus Ebola Vaccine - Preliminary Report.

Jason Regules; John Beigel; Kristopher M. Paolino; Jocelyn Voell; Amy R. Castellano; Paula Muñoz; James E. Moon; Richard C. Ruck; Jason W. Bennett; Patrick S. Twomey; Ramiro L. Gutiérrez; Shon Remich; Holly R. Hack; Meagan L. Wisniewski; Matthew Josleyn; Steven A. Kwilas; Nicole M. Van Deusen; Olivier Tshiani Mbaya; Yan Zhou; Daphne Stanley; Robin L. Bliss; Deborah Cebrik; Kirsten S. Smith; Meng Shi; Julie E. Ledgerwood; Barney S. Graham; Nancy J. Sullivan; Linda L. Jagodzinski; Sheila A. Peel; Judie B. Alimonti

Background The worst Ebola virus disease (EVD) outbreak in history has resulted in more than 28,000 cases and 11,000 deaths. We present the final results of two phase 1 trials of an attenuated, replication‐competent, recombinant vesicular stomatitis virus (rVSV)–based vaccine candidate designed to prevent EVD. Methods We conducted two phase 1, placebo‐controlled, double‐blind, dose‐escalation trials of an rVSV‐based vaccine candidate expressing the glycoprotein of a Zaire strain of Ebola virus (ZEBOV). A total of 39 adults at each site (78 participants in all) were consecutively enrolled into groups of 13. At each site, volunteers received one of three doses of the rVSV‐ZEBOV vaccine (3 million plaque‐forming units [PFU], 20 million PFU, or 100 million PFU) or placebo. Volunteers at one of the sites received a second dose at day 28. Safety and immunogenicity were assessed. Results The most common adverse events were injection‐site pain, fatigue, myalgia, and headache. Transient rVSV viremia was noted in all the vaccine recipients after dose 1. The rates of adverse events and viremia were lower after the second dose than after the first dose. By day 28, all the vaccine recipients had seroconversion as assessed by an enzyme‐linked immunosorbent assay (ELISA) against the glycoprotein of the ZEBOV‐Kikwit strain. At day 28, geometric mean titers of antibodies against ZEBOV glycoprotein were higher in the groups that received 20 million PFU or 100 million PFU than in the group that received 3 million PFU, as assessed by ELISA and by pseudovirion neutralization assay. A second dose at 28 days after dose 1 significantly increased antibody titers at day 56, but the effect was diminished at 6 months. Conclusions This Ebola vaccine candidate elicited anti‐Ebola antibody responses. After vaccination, rVSV viremia occurred frequently but was transient. These results support further evaluation of the vaccine dose of 20 million PFU for preexposure prophylaxis and suggest that a second dose may boost antibody responses. (Funded by the National Institutes of Health and others; rVSV&Dgr;G‐ZEBOV‐GP ClinicalTrials.gov numbers, NCT02269423 and NCT02280408.)


Vaccine | 2010

A replication defective recombinant Ad5 vaccine expressing Ebola virus GP is safe and immunogenic in healthy adults

Julie E. Ledgerwood; Pamela Costner; N. Desai; LaSonji A. Holman; Mary E. Enama; Galina Yamshchikov; Sabue Mulangu; Zonghui Hu; Charla A. Andrews; R.A. Sheets; Richard A. Koup; Mario Roederer; Robert T. Bailer; John R. Mascola; Maria Grazia Pau; Nancy J. Sullivan; Jaap Goudsmit; Gary J. Nabel; Barney S. Graham

Ebola virus causes irregular outbreaks of severe hemorrhagic fever in equatorial Africa. Case mortality remains high; there is no effective treatment and outbreaks are sporadic and unpredictable. Studies of Ebola virus vaccine platforms in non-human primates have established that the induction of protective immunity is possible and safety and human immunogenicity has been demonstrated in a previous Phase I clinical trial of a 1st generation Ebola DNA vaccine. We now report the safety and immunogenicity of a recombinant adenovirus serotype 5 (rAd5) vaccine encoding the envelope glycoprotein (GP) from the Zaire and Sudan Ebola virus species, in a randomized, placebo-controlled, double-blinded, dose escalation, Phase I human study. Thirty-one healthy adults received vaccine at 2×10(9) (n=12), or 2×10(10) (n=11) viral particles or placebo (n=8) as an intramuscular injection. Antibody responses were assessed by ELISA and neutralizing assays; and T cell responses were assessed by ELISpot and intracellular cytokine staining assays. This recombinant Ebola virus vaccine was safe and subjects developed antigen specific humoral and cellular immune responses.


The New England Journal of Medicine | 2016

A Monovalent Chimpanzee Adenovirus Ebola Vaccine Boosted with MVA

Katie Ewer; Tommy Rampling; Navin Venkatraman; Georgina Bowyer; Danny Wright; Teresa Lambe; Egeruan B. Imoukhuede; Ruth O. Payne; Sarah Katharina Fehling; Thomas Strecker; Nadine Biedenkopf; Verena Krähling; Claire M. Tully; Nick J. Edwards; Emma Bentley; Dhan Samuel; Geneviève M. Labbé; Jing Jin; Malick Gibani; A. Minhinnick; M. Wilkie; Ian D. Poulton; N. Lella; Rachel Roberts; Felicity Hartnell; Carly M. Bliss; Kailan Sierra-Davidson; Jonathan Powlson; Eleanor Berrie; Richard S Tedder

BACKGROUND The West African outbreak of Ebola virus disease that peaked in 2014 has caused more than 11,000 deaths. The development of an effective Ebola vaccine is a priority for control of a future outbreak. METHODS In this phase 1 study, we administered a single dose of the chimpanzee adenovirus 3 (ChAd3) vaccine encoding the surface glycoprotein of Zaire ebolavirus (ZEBOV) to 60 healthy adult volunteers in Oxford, United Kingdom. The vaccine was administered in three dose levels--1×10(10) viral particles, 2.5×10(10) viral particles, and 5×10(10) viral particles--with 20 participants in each group. We then assessed the effect of adding a booster dose of a modified vaccinia Ankara (MVA) strain, encoding the same Ebola virus glycoprotein, in 30 of the 60 participants and evaluated a reduced prime-boost interval in another 16 participants. We also compared antibody responses to inactivated whole Ebola virus virions and neutralizing antibody activity with those observed in phase 1 studies of a recombinant vesicular stomatitis virus-based vaccine expressing a ZEBOV glycoprotein (rVSV-ZEBOV) to determine relative potency and assess durability. RESULTS No safety concerns were identified at any of the dose levels studied. Four weeks after immunization with the ChAd3 vaccine, ZEBOV-specific antibody responses were similar to those induced by rVSV-ZEBOV vaccination, with a geometric mean titer of 752 and 921, respectively. ZEBOV neutralization activity was also similar with the two vaccines (geometric mean titer, 14.9 and 22.2, respectively). Boosting with the MVA vector increased virus-specific antibodies by a factor of 12 (geometric mean titer, 9007) and increased glycoprotein-specific CD8+ T cells by a factor of 5. Significant increases in neutralizing antibodies were seen after boosting in all 30 participants (geometric mean titer, 139; P<0.001). Virus-specific antibody responses in participants primed with ChAd3 remained positive 6 months after vaccination (geometric mean titer, 758) but were significantly higher in those who had received the MVA booster (geometric mean titer, 1750; P<0.001). CONCLUSIONS The ChAd3 vaccine boosted with MVA elicited B-cell and T-cell immune responses to ZEBOV that were superior to those induced by the ChAd3 vaccine alone. (Funded by the Wellcome Trust and others; ClinicalTrials.gov number, NCT02240875.).


Journal of Virology | 2014

Enhanced Potency of a Broadly Neutralizing HIV-1 Antibody In Vitro Improves Protection against Lentiviral Infection In Vivo

Rebecca S. Rudicell; Young Do Kwon; Sung Youl Ko; Amarendra Pegu; Mark K. Louder; Ivelin S. Georgiev; Xueling Wu; Jiang Zhu; Jeffrey C. Boyington; Xuejun Chen; Wei Shi; Zhi Yong Yang; Nicole A. Doria-Rose; Krisha McKee; Sijy O'Dell; Stephen D. Schmidt; Gwo Yu Chuang; Aliaksandr Druz; Cinque Soto; Yongping Yang; Baoshan Zhang; Tongqing Zhou; John Paul Todd; Krissey E. Lloyd; Joshua Eudailey; Kyle E. Roberts; Bruce Randall Donald; Robert T. Bailer; Julie E. Ledgerwood; James C. Mullikin

ABSTRACT Over the past 5 years, a new generation of highly potent and broadly neutralizing HIV-1 antibodies has been identified. These antibodies can protect against lentiviral infection in nonhuman primates (NHPs), suggesting that passive antibody transfer would prevent HIV-1 transmission in humans. To increase the protective efficacy of such monoclonal antibodies, we employed next-generation sequencing, computational bioinformatics, and structure-guided design to enhance the neutralization potency and breadth of VRC01, an antibody that targets the CD4 binding site of the HIV-1 envelope. One variant, VRC07-523, was 5- to 8-fold more potent than VRC01, neutralized 96% of viruses tested, and displayed minimal autoreactivity. To compare its protective efficacy to that of VRC01 in vivo, we performed a series of simian-human immunodeficiency virus (SHIV) challenge experiments in nonhuman primates and calculated the doses of VRC07-523 and VRC01 that provide 50% protection (EC50). VRC07-523 prevented infection in NHPs at a 5-fold lower concentration than VRC01. These results suggest that increased neutralization potency in vitro correlates with improved protection against infection in vivo, documenting the improved functional efficacy of VRC07-523 and its potential clinical relevance for protecting against HIV-1 infection in humans. IMPORTANCE In the absence of an effective HIV-1 vaccine, alternative strategies are needed to block HIV-1 transmission. Direct administration of HIV-1-neutralizing antibodies may be able to prevent HIV-1 infections in humans. This approach could be especially useful in individuals at high risk for contracting HIV-1 and could be used together with antiretroviral drugs to prevent infection. To optimize the chance of success, such antibodies can be modified to improve their potency, breadth, and in vivo half-life. Here, knowledge of the structure of a potent neutralizing antibody, VRC01, that targets the CD4-binding site of the HIV-1 envelope protein was used to engineer a next-generation antibody with 5- to 8-fold increased potency in vitro. When administered to nonhuman primates, this antibody conferred protection at a 5-fold lower concentration than the original antibody. Our studies demonstrate an important correlation between in vitro assays used to evaluate the therapeutic potential of antibodies and their in vivo effectiveness.

Collaboration


Dive into the Julie E. Ledgerwood's collaboration.

Top Co-Authors

Avatar

Barney S. Graham

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Robert T. Bailer

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Richard A. Koup

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

John R. Mascola

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Mary E. Enama

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ingelise J. Gordon

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

LaSonji A. Holman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Cynthia S. Hendel

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Laura Novik

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Zonghui Hu

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge