Network


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

Hotspot


Dive into the research topics where Dagna S. Laufer is active.

Publication


Featured researches published by Dagna S. Laufer.


Journal of Virology | 2009

Human Immunodeficiency Virus Type 1 Elite Neutralizers: Individuals with Broad and Potent Neutralizing Activity Identified by Using a High-Throughput Neutralization Assay together with an Analytical Selection Algorithm

Melissa Simek; Wasima Rida; Frances Priddy; Pham Pung; Emily Carrow; Dagna S. Laufer; Jennifer Lehrman; Mark Boaz; Tony Tarragona-Fiol; George Miiro; Josephine Birungi; Anton Pozniak; Dale A. McPhee; Olivier Manigart; Etienne Karita; André Inwoley; Walter Jaoko; Jack DeHovitz; Linda-Gail Bekker; Punnee Pitisuttithum; Robert Paris; Laura M. Walker; Pascal Poignard; Terri Wrin; Patricia Fast; Dennis R. Burton; Wayne C. Koff

ABSTRACT The development of a rapid and efficient system to identify human immunodeficiency virus type 1 (HIV-1)-infected individuals with broad and potent HIV-1-specific neutralizing antibody responses is an important step toward the discovery of critical neutralization targets for rational AIDS vaccine design. In this study, samples from HIV-1-infected volunteers from diverse epidemiological regions were screened for neutralization responses using pseudovirus panels composed of clades A, B, C, and D and circulating recombinant forms (CRFs). Initially, 463 serum and plasma samples from Australia, Rwanda, Uganda, the United Kingdom, and Zambia were screened to explore neutralization patterns and selection ranking algorithms. Samples were identified that neutralized representative isolates from at least four clade/CRF groups with titers above prespecified thresholds and ranked based on a weighted average of their log-transformed neutralization titers. Linear regression methods selected a five-pseudovirus subset, representing clades A, B, and C and one CRF01_AE, that could identify top-ranking samples with 50% inhibitory concentration (IC50) neutralization titers of ≥100 to multiple isolates within at least four clade groups. This reduced panel was then used to screen 1,234 new samples from the Ivory Coast, Kenya, South Africa, Thailand, and the United States, and 1% were identified as elite neutralizers. Elite activity is defined as the ability to neutralize, on average, more than one pseudovirus at an IC50 titer of 300 within a clade group and across at least four clade groups. These elite neutralizers provide promising starting material for the isolation of broadly neutralizing monoclonal antibodies to assist in HIV-1 vaccine design.


PLOS Pathogens | 2016

Antigen-Specific Antibody Glycosylation Is Regulated via Vaccination

Alison E. Mahan; Madeleine F. Jennewein; Todd J. Suscovich; Kendall Dionne; Jacquelynne Tedesco; Amy W. Chung; Hendrik Streeck; Maria Pau; Hanneke Schuitemaker; Donald P. Francis; Patricia Fast; Dagna S. Laufer; Bruce D. Walker; Lindsey R. Baden; Dan H. Barouch; Galit Alter

Antibody effector functions, such as antibody-dependent cellular cytotoxicity, complement deposition, and antibody-dependent phagocytosis, play a critical role in immunity against multiple pathogens, particularly in the absence of neutralizing activity. Two modifications to the IgG constant domain (Fc domain) regulate antibody functionality: changes in antibody subclass and changes in a single N-linked glycan located in the CH2 domain of the IgG Fc. Together, these modifications provide a specific set of instructions to the innate immune system to direct the elimination of antibody-bound antigens. While it is clear that subclass selection is actively regulated during the course of natural infection, it is unclear whether antibody glycosylation can be tuned, in a signal-specific or pathogen-specific manner. Here, we show that antibody glycosylation is determined in an antigen- and pathogen-specific manner during HIV infection. Moreover, while dramatic differences exist in bulk IgG glycosylation among individuals in distinct geographical locations, immunization is able to overcome these differences and elicit antigen-specific antibodies with similar antibody glycosylation patterns. Additionally, distinct vaccine regimens induced different antigen-specific IgG glycosylation profiles, suggesting that antibody glycosylation is not only programmable but can be manipulated via the delivery of distinct inflammatory signals during B cell priming. These data strongly suggest that the immune system naturally drives antibody glycosylation in an antigen-specific manner and highlights a promising means by which next-generation therapeutics and vaccines can harness the antiviral activity of the innate immune system via directed alterations in antibody glycosylation in vivo.  


Clinical and Vaccine Immunology | 2013

Safety and immunogenicity of DNA prime and Modified Vaccinia Ankara virus HIV subtype C vaccine boost in healthy adults

Peter Hayes; Jill Gilmour; Andrea von Lieven; Dilbinder K. Gill; Lorna Clark; Jakub Kopycinski; Hannah Cheeseman; Amy W. Chung; Galit Alter; Len Dally; Devika Zachariah; Angela Lombardo; James Ackland; Eddy Sayeed; Akil Jackson; Marta Boffito; Brian Gazzard; Patricia Fast; Josephine H. Cox; Dagna S. Laufer

ABSTRACT A randomized, double-blind, placebo-controlled phase I trial was conducted in 32 HIV-uninfected healthy volunteers to assess the safety and immunogenicity of 3 doses of DNA vaccine (Advax) plus 1 dose of recombinant modified vaccinia virus Ankara (MVA) (TBC-M4) or 3 doses of TBC-M4 alone (groups A and B, respectively). Both vaccine regimens were found to be safe and well tolerated. Gamma interferon (IFN-γ) enzyme-linked immunosorbent spot (ELISPOT) assay responses were detected in 1/10 (10%) individuals in group A after three Advax primes and in 9/9 individuals (100%) after the MVA boost. In group B, IFN-γ ELISPOT responses were detected in 6/12 (50%) and 7/11 (64%) individuals after the second and third MVA vaccinations, respectively. Responses to all vaccine components, but predominantly to Env, were seen. The breadth and magnitude of the T cell response and viral inhibition were greater in group A than in group B, indicating that the quality of the T-cell response was enhanced by the DNA prime. Intracellular cytokine staining indicated that the T-cell responses were polyfunctional but were skewed toward Env with a CD4+ phenotype. At 2 weeks after the last vaccination, HIV-specific antibody responses were detected in all (100%) group B and 1/11 (9.1%) group A vaccinees. Vaccinia virus-specific responses were detected in all (100%) group B and 2/11 (18.2%) group A vaccinees. In conclusion, HIV-specific T-cell responses were seen in the majority of volunteers in groups A and B but with a trend toward greater quality of the T-cell response in group A. Antibody responses were better in group B than in group A.


PLOS ONE | 2015

A Phase I Double Blind, Placebo-Controlled, Randomized Study of the Safety and Immunogenicity of an Adjuvanted HIV-1 Gag-Pol-Nef Fusion Protein and Adenovirus 35 Gag-RT-Int-Nef Vaccine in Healthy HIV-Uninfected African Adults.

Gloria Omosa-Manyonyi; Juliet Mpendo; Eugene Ruzagira; William Kilembe; Elwyn Chomba; François Roman; Patricia Bourguignon; Marguerite Koutsoukos; Alix Collard; Gerald Voss; Dagna S. Laufer; Gwynn Stevens; Peter Hayes; Lorna Clark; Emmanuel Cormier; Len Dally; Burc Barin; Jim Ackland; Kristen Syvertsen; Devika Zachariah; Kamaal Anas; Eddy Sayeed; Angela Lombardo; Jill Gilmour; Josephine H. Cox; Patricia Fast; Frances Priddy

Background Sequential prime-boost or co-administration of HIV vaccine candidates based on an adjuvanted clade B p24, RT, Nef, p17 fusion protein (F4/AS01) plus a non-replicating adenovirus 35 expressing clade A Gag, RT, Int and Nef (Ad35-GRIN) may lead to a unique immune profile, inducing both strong T-cell and antibody responses. Methods In a phase 1, double-blind, placebo-controlled trial, 146 healthy adult volunteers were randomized to one of four regimens: heterologous prime-boost with two doses of F4/AS01E or F4/AS01B followed by Ad35-GRIN; Ad35-GRIN followed by two doses of F4/AS01B; or three co-administrations of Ad35-GRIN and F4/AS01B. T cell and antibody responses were measured. Results The vaccines were generally well-tolerated, and did not cause serious adverse events. The response rate, by IFN-γ ELISPOT, was greater when Ad35-GRIN was the priming vaccine and in the co-administration groups. F4/AS01 induced CD4+ T-cells expressing primarily CD40L and IL2 +/- TNF-α, while Ad35-GRIN induced predominantly CD8+ T-cells expressing IFN-γ +/- IL2 or TNF-α. Viral inhibition was induced after Ad35-GRIN vaccination, regardless of the regimen. Strong F4-specific antibody responses were induced. Immune responses persisted at least a year after the last vaccination. The complementary response profiles, characteristic of each vaccine, were both expressed after co-administration. Conclusion Co-administration of an adjuvanted protein and an adenovirus vector showed an acceptable safety and reactogenicity profile and resulted in strong, multifunctional and complementary HIV-specific immune responses. Trial Registration ClinicalTrials.gov NCT01264445


The Journal of Infectious Diseases | 2017

First-in-Human Evaluation of the Safety and Immunogenicity of an Intranasally Administered Replication-Competent Sendai Virus–Vectored HIV Type 1 Gag Vaccine: Induction of Potent T-Cell or Antibody Responses in Prime-Boost Regimens

Julien Nyombayire; Omu Anzala; Brian Gazzard; Etienne Karita; Philip Bergin; Peter Hayes; Jakub Kopycinski; Gloria Omosa-Manyonyi; Akil Jackson; Jean Bizimana; Bashir Farah; Eddy Sayeed; Christopher L. Parks; Makoto Inoue; Takashi Hironaka; Hiroto Hara; Tsugumine Shu; Tetsuro Matano; Len Dally; Burc Barin; Harriet Park; Jill Gilmour; Angela Lombardo; Jean-Louis Excler; Patricia Fast; Dagna S. Laufer; Josephine H. Cox

Background. We report the first-in-human safety and immunogenicity assessment of a prototype intranasally administered, replication-competent Sendai virus (SeV)–vectored, human immunodeficiency virus type 1 (HIV-1) vaccine. Methods. Sixty-five HIV-1–uninfected adults in Kenya, Rwanda, and the United Kingdom were assigned to receive 1 of 4 prime-boost regimens (administered at 0 and 4 months, respectively; ratio of vaccine to placebo recipients, 12:4): priming with a lower-dose SeV-Gag given intranasally, followed by boosting with an adenovirus 35–vectored vaccine encoding HIV-1 Gag, reverse transcriptase, integrase, and Nef (Ad35-GRIN) given intramuscularly (SLA); priming with a higher-dose SeV-Gag given intranasally, followed by boosting with Ad35-GRIN given intramuscularly (SHA); priming with Ad35-GRIN given intramuscularly, followed by boosting with a higher-dose SeV-Gag given intranasally (ASH); and priming and boosting with a higher-dose SeV-Gag given intranasally (SHSH). Results. All vaccine regimens were well tolerated. Gag-specific IFN-&ggr; enzyme-linked immunospot–determined response rates and geometric mean responses were higher (96% and 248 spot-forming units, respectively) in groups primed with SeV-Gag and boosted with Ad35-GRIN (SLA and SHA) than those after a single dose of Ad35-GRIN (56% and 54 spot-forming units, respectively) or SeV-Gag (55% and 59 spot-forming units, respectively); responses persisted for ≥8 months after completion of the prime-boost regimen. Functional CD8+ T-cell responses with greater breadth, magnitude, and frequency in a viral inhibition assay were also seen in the SLA and SHA groups after Ad35-GRIN boost, compared with those who received either vaccine alone. SeV-Gag did not boost T-cell counts in the ASH group. In contrast, the highest Gag-specific antibody titers were seen in the ASH group. Mucosal antibody responses were sporadic. Conclusions. SeV-Gag primed functional, durable HIV-specific T-cell responses and boosted antibody responses. The prime-boost sequence appears to determine which arm of the immune response is stimulated. Clinical Trials Registration. NCT01705990.


PLOS ONE | 2014

Acceptability and Feasibility of Repeated Mucosal Specimen Collection in Clinical Trial Participants in Kenya

Gloria Omosa-Manyonyi; Harriet Park; Gaudensia Mutua; Bashir Farah; Philip Bergin; Dagna S. Laufer; Jennifer Lehrman; Kundai Chinyenze; Burc Barin; Pat Fast; Jill Gilmour; Omu Anzala

Background Mucosal specimens are essential to evaluate compartmentalized immune responses to HIV vaccine candidates and other mucosally targeted investigational products. We studied the acceptability and feasibility of repeated mucosal sampling in East African clinical trial participants at low risk of HIV and other sexually transmitted infections. Methods and Findings The Kenya AIDS Vaccine Initiative (KAVI) enrolled participants into three Phase 1 trials of preventive HIV candidate vaccines in 2011–2012 at two clinical research centers in Nairobi. After informed consent to a mucosal sub-study, participants were asked to undergo collection of mucosal secretions (saliva, oral fluids, semen, cervico-vaginal and rectal), but could opt out of any collection at any visit. Specimens were collected at baseline and two additional time points. A tolerability questionnaire was administered at the final sub-study visit. Of 105 trial participants, 27 of 34 women (79%) and 62 of 71 men (87%) enrolled in the mucosal sub-study. Nearly all sub-study participants gave saliva and oral fluids at all visits. Semen was collected from about half the participating men (47–48%) at all visits. Cervico-vaginal secretions were collected by Softcup from about two thirds of women (63%) at baseline, increasing to 78% at the following visits, with similar numbers for cervical secretion collection by Merocel sponge; about half of women (52%) gave cervico-vaginal samples at all visits. Rectal secretions were collected with Merocel sponge from about a quarter of both men and women (24%) at all 3 visits, with 16% of men and 19% of women giving rectal samples at all visits. Conclusions Repeated mucosal sampling in clinical trial participants in Kenya is feasible, with a good proportion of participants consenting to most sampling methods with the exception of rectal samples. Experienced staff members of both sexes and trained counselors with standardized messaging may improve acceptance of rectal sampling.


Journal of Acquired Immune Deficiency Syndromes | 2016

Assessment of Anti-HIV-1 Antibodies in Oral and Nasal Compartments of Volunteers From 3 Different Populations.

Philip Bergin; Robert Langat; Gloria Omosa-Manyonyi; Bashir Farah; Gina Ouattara; Harriet Park; Helen Coutinho; Dagna S. Laufer; Pat Fast; Carl Verlinde; Jean Bizimana; Gisele Umviligihozo; Julien Nyombayire; Rosine Ingabire; Kristin Kuldanek; Josephine H. Cox; Martin McMorrow; Sarah Fidler; Etienne Karita; Jill Gilmour; Omu Anzala

Abstract:In this study, we assessed the feasibility of collecting standardized nasal and salivary samples at centers in Nairobi (Kenya), Kigali (Rwanda), and London (United Kingdom) using different collection devices and media (synthetic absorptive matrices versus flocked swabs, and Salimetrics oral swabs versus whole oral fluid collection). We detected anti-Gag (p24) and envelope (gp140) antibodies in both nasal fluid and salivary collections from all HIV-infected individuals, and cross-reactive anti-p24 antibodies were detected in 10% of HIV-uninfected individuals enrolled at one site. Collections from the nasal turbinates were comparable with samples collected deeper in the nasopharyngeal tract, and the yield of anti-p24 IgA in the whole oral fluid samples was higher than in samples collected from the parotid gland. We noted a trend toward reduced levels of anti-HIV antibody in the volunteers receiving anti-retroviral therapy. Levels of antibodies were stable over multiple collection visits. Overall, this study shows that nasal and salivary samples can be collected in a standardized manner over repeated visits in both low- and high-resource settings. These methods may be used in support for future HIV vaccine clinical trials.


PLOS Pathogens | 2016

Correction: Antigen-Specific Antibody Glycosylation Is Regulated via Vaccination

Alison E. Mahan; Madeleine F. Jennewein; Todd J. Suscovich; Kendall Dionne; Jacquelynne Tedesco; Amy W. Chung; Hendrik Streeck; Maria Pau; Hanneke Schuitemaker; Donald P. Francis; Patricia Fast; Dagna S. Laufer; Bruce D. Walker; Lindsey R. Baden; Dan H. Barouch; Galit Alter

[This corrects the article DOI: 10.1371/journal.ppat.1005456.].


Retrovirology | 2012

Uptake and tolerability of repeated mucosal specimen collection in two Phase 1 AIDS preventive vaccine trials in Kenya

Gaudensia Mutua; G Omosa-Manyonyi; Harriet Park; Philip Bergin; Dagna S. Laufer; Pauli N. Amornkul; Jennifer Lehrman; Patricia Fast; Jill Gilmour; Omu Anzala; B Farah

Methods The Kenya AIDS Vaccine Initiative (KAVI) initiated two AIDS preventive vaccine trials in Nairobi in 2011. After informed consent for a mucosal substudy, participants were asked to provide any of several types of mucosal secretions: saliva, oral fluids, semen, cervico-vaginal and rectal. Specimens were collected at baseline, one month after final vaccination, and at the next scheduled trial visit. A tolerability questionnaire was administered at the final visit.


The Journal of Infectious Diseases | 1996

Safety and Immunogenicity of a Combined Live Attenuated Measles, Mumps, Rubella, and Varicella Vaccine (MMRIIV) in Healthy Children

Barbara M. Watson; Dagna S. Laufer; Barbara J. Kuter; Brenda Staehle; C. Jo White

Collaboration


Dive into the Dagna S. Laufer's collaboration.

Top Co-Authors

Avatar

Patricia Fast

International AIDS Vaccine Initiative

View shared research outputs
Top Co-Authors

Avatar

Jill Gilmour

International AIDS Vaccine Initiative

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Josephine H. Cox

International AIDS Vaccine Initiative

View shared research outputs
Top Co-Authors

Avatar

Angela Lombardo

International AIDS Vaccine Initiative

View shared research outputs
Top Co-Authors

Avatar

Etienne Karita

International AIDS Vaccine Initiative

View shared research outputs
Top Co-Authors

Avatar

Harriet Park

International AIDS Vaccine Initiative

View shared research outputs
Top Co-Authors

Avatar

Len Dally

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Peter Hayes

International AIDS Vaccine Initiative

View shared research outputs
Top Co-Authors

Avatar

Philip Bergin

International AIDS Vaccine Initiative

View shared research outputs
Researchain Logo
Decentralizing Knowledge