Network


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

Hotspot


Dive into the research topics where Angela Lombardo is active.

Publication


Featured researches published by Angela Lombardo.


AIDS Research and Human Retroviruses | 2009

A Phase 1 study to evaluate the safety and immunogenicity of a recombinant HIV type 1 subtype C-modified vaccinia Ankara virus vaccine candidate in Indian volunteers.

Vadakkuppatu Devasenapathi Ramanathan; Makesh Kumar; Jayashri Mahalingam; Pattabiraman Sathyamoorthy; Paranji Ramaiyengar Narayanan; Suniti Solomon; Dennis Panicali; Sekhar Chakrabarty; Josephine H. Cox; Eddy Sayeed; James Ackland; Carl Verlinde; Dani Vooijs; Kelley Loughran; Burc Barin; Angela Lombardo; Jill Gilmour; Gwynneth Stevens; Michelle Seth Smith; Tony Tarragona-Fiol; Peter Hayes; Sonali Kochhar; Jean-Louis Excler; Patricia Fast

A recombinant modified vaccinia Ankara virus vaccine candidate (TBC-M4) expressing HIV-1 subtype C env, gag, tat-rev, and nef-RT genes was tested in a randomized, double-blind, dose escalation Phase I trial in 32 HIV-uninfected healthy volunteers who received three intramuscular injections of TBC-M4 at 0, 1, and 6 months of 5 x 10(7) plaque-forming units (pfu) (low dosage, LD) (n = 12) or 2.5 x 10(8) pfu (high dosage, HD) (n = 12) or placebo (n = 8). Local and systemic reactogenicity was experienced by approximately 67% and 83% of vaccine recipients, respectively. The reactogenicity events were mostly mild in severity. Severe but transient systemic reactogenicity was seen in one volunteer of the HD group. No vaccine-related serious adverse events or events suggesting perimyocarditis were seen. A higher frequency of local reactogenicity events was observed in the HD group. Cumulative HIV-specific IFN-gamma ELISPOT responses were detected in frozen PBMCs from 9/11 (82%), 12/12 (100%), and 1/8 (13%) volunteers after the third injection of the LD, HD, and placebo groups, respectively. Most of the responses were to gag and env proteins (maximum of 430 SFU/10(6) PBMCs) persisting across multiple time points. HIV-specific ELISA antibody responses were detected in 10/11, 12/12, and 0/8 volunteers post-third vaccination, in the LD, HD, and placebo groups, respectively. No neutralizing activity against HIV-1 subtype C isolates was detected. TBC-M4 appears to be generally safe and well-tolerated. The immune response detected was dose dependent, modest in magnitude, and directed mostly to env and gag proteins, suggesting further evaluation of this vaccine in a prime-boost regimen.


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.


Human Vaccines | 2011

Prevalence of specific neutralizing antibodies against Sendai virus in populations from different geographic areas: implications for AIDS vaccine development using Sendai virus vectors.

Hiroto Hara; Takashi Hironaka; Makoto Inoue; Akihiro Iida; Tsugumine Shu; Mamoru Hasegawa; Yoshiyuki Nagai; Ann R. Falsey; Anatoli Kamali; Omu Anzala; Eduard J. Sanders; Etienne Karita; Lawrence Mwananyanda; Sandhya Vasan; Angela Lombardo; Christopher L. Parks; Eddy Sayeed; Marietta Krebs; Emmanuel Cormier; James Ackland; Matthew Price; Jean-Louis Excler

A Sendai virus (SeV) vector is being developed for delivery of an HIV immunogen. SeV is not known to cause disease in humans. Because it is genetically and antigenically related to human parainfluenza virus type 1 (hPIV-1), it is important to determine whether pre-existing hPIV-1 antibodies will affect immune responses elicited by a SeV vector-based vaccine. To quantify SeV neutralizing antibodies (NAb) in human serum, a sensitive virus neutralization assay was developed using a SeV vector encoding green fluorescent protein. Samples from 255 HIV-uninfected subjects from Africa, Europe, United States, and Japan, as well as from 12 confirmed hPIV-1-infected patients, were analyzed. SeV NAb titers did not vary significantly after serum was treated with receptor-destroying enzyme, indicating that non-specific hemagglutination inhibitors did not affect the assay sensitivity. A significant correlation was observed between hPIV-1 ELISA and SeV NAb titers. SeV NAb were detected in 92.5% subjects with a median titer of 60.6 and values ranging from 5.9- 11,324. The majority had titers


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 | 2010

Phase 1 Safety and Immunogenicity Evaluation of ADMVA, a Multigenic, Modified Vaccinia Ankara-HIV-1 B'/C Candidate Vaccine

Sandhya Vasan; Sarah J. Schlesinger; Zhiwei Chen; Arlene Hurley; Angela Lombardo; Soe Than; Phumla Adesanya; Catherine Bunce; Mark Boaz; Rosanne Boyle; Eddy Sayeed; Lorna Clark; Daniel Dugin; Mar Boente-Carrera; Claudia Schmidt; Qing Fang; Ba Lei; Yaoxing Huang; Gerasimos J. Zaharatos; David F. Gardiner; Marina Caskey; Laura Seamons; Martin Ho; Len Dally; Carol Smith; Josephine H. Cox; Dilbinder K. Gill; Jill Gilmour; Michael C. Keefer; Patricia Fast


PLOS ONE | 2010

Phase 1 safety and immunogenicity evaluation of ADVAX, a multigenic, DNA-based clade C/B' HIV-1 candidate vaccine.

Sandhya Vasan; Sarah J. Schlesinger; Yaoxing Huang; Arlene Hurley; Angela Lombardo; Zhiwei Chen; Soe Than; Phumla Adesanya; Catherine Bunce; Mark Boaz; Rosanne Boyle; Eddy Sayeed; Lorna Clark; Daniel Dugin; Claudia Schmidt; Yang Song; Laura Seamons; Len Dally; Martin Ho; Carol Smith; Martin Markowitz; Josephine H. Cox; Dilbinder K. Gill; Jill Gilmour; Michael C. Keefer; Patricia Fast; David D. Ho


Annals of Internal Medicine | 2016

Assessment of the Safety and Immunogenicity of 2 Novel Vaccine Platforms for HIV-1 Prevention: A Randomized Trial

Lindsey R. Baden; Etienne Karita; Gaudensia Mutua; Linda-Gail Bekker; Glenda Gray; Liesl Page-Shipp; Stephen R. Walsh; Julien Nyombayire; Omu Anzala; Surita Roux; Fatima Laher; Craig Innes; Michael S. Seaman; Yehuda Z. Cohen; Lauren Peter; Nicole Frahm; M. Juliana McElrath; Peter Hayes; Edith Swann; Nicole Grunenberg; Maria Grazia-Pau; Mo Weijtens; Jerry Sadoff; Len Dally; Angela Lombardo; Jill Gilmour; Josephine H. Cox; Raphael Dolin; Patricia Fast; Dan H. Barouch


Archive | 2016

GENETICALLY STABLE REPLICATION COMPETENT SENDAI VIRUS VECTOR(S) CONTAINING AND EXPRESSING OPTIMIZED HIV GENES

John W. Coleman; Josephine H. Cox; Arban Domi; Hiroto Hara; Takashi Hironaka; Makoto Inoue; Dagna S. Laufer; Angela Lombardo; Christopher L. Parks; Eddy Sayeed; Maoli Yuan; Xinsheng Zhang


Annals of Internal Medicine | 2016

Assessment of the Safety and Immunogenicity of 2 Novel Vaccine Platforms for HIV-1 PreventionA Randomized TrialAssessment of 2 Novel Vaccine Platforms for HIV-1 Prevention

Lindsey R. Baden; Etienne Karita; Gaudensia Mutua; Linda-Gail Bekker; Glenda Gray; Liesl Page-Shipp; Stephen R. Walsh; Julien Nyombayire; Omu Anzala; Surita Roux; Fatima Laher; Craig Innes; Michael S. Seaman; Yehuda Z. Cohen; Lauren Peter; Nicole Frahm; M. Juliana McElrath; Peter Hayes; Edith Swann; Nicole Grunenberg; Maria Grazia-Pau; Mo Weijtens; Jerry Sadoff; Len Dally; Angela Lombardo; Jill Gilmour; Josephine H. Cox; Raphael Dolin; Patricia Fast; Dan H. Barouch

Collaboration


Dive into the Angela Lombardo's collaboration.

Top Co-Authors

Avatar

Josephine H. Cox

International AIDS Vaccine Initiative

View shared research outputs
Top Co-Authors

Avatar

Eddy Sayeed

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

Patricia Fast

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
Top Co-Authors

Avatar

Dagna S. Laufer

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

Lorna Clark

International AIDS Vaccine Initiative

View shared research outputs
Researchain Logo
Decentralizing Knowledge