Network


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

Hotspot


Dive into the research topics where Eddy Sayeed is active.

Publication


Featured researches published by Eddy Sayeed.


PLOS ONE | 2012

A phase I double blind, placebo-controlled, randomized study of a multigenic HIV-1 adenovirus subtype 35 vector vaccine in healthy uninfected adults.

Michael C. Keefer; Jill Gilmour; Peter Hayes; Dilbinder K. Gill; Jakub Kopycinski; Hannah Cheeseman; Michelle Cashin-Cox; Marloes Naarding; Lorna Clark; Natalia Fernandez; Catherine Bunce; Christine M. Hay; Sabrina Welsh; Wendy Komaroff; Lottie Hachaambwa; Tony Tarragona-Fiol; Eddy Sayeed; Devika Zachariah; James Ackland; Kelley Loughran; Burc Barin; Emmanuel Cormier; Josephine H. Cox; Patricia Fast; Jean-Louis Excler

Background We conducted a phase I, randomized, double-blind, placebo-controlled trial to assess the safety and immunogenicity of escalating doses of two recombinant replication defective adenovirus serotype 35 (Ad35) vectors containing gag, reverse transcriptase, integrase and nef (Ad35-GRIN) and env (Ad35-ENV), both derived from HIV-1 subtype A isolates. The trial enrolled 56 healthy HIV-uninfected adults. Methods Ad35-GRIN/ENV (Ad35-GRIN and Ad35-ENV mixed in the same vial in equal proportions) or Ad35-GRIN was administered intramuscularly at 0 and 6 months. Participants were randomized to receive either vaccine or placebo (10/4 per group, respectively) within one of four dosage groups: Ad35-GRIN/ENV 2×109 (A), 2×1010 (B), 2×1011 (C), or Ad35-GRIN 1×1010 (D) viral particles. Results No vaccine-related serious adverse event was reported. Reactogenicity events reported were dose-dependent, mostly mild or moderate, some severe in Group C volunteers, all transient and resolving spontaneously. IFN-γ ELISPOT responses to any vaccine antigen were detected in 50, 56, 70 and 90% after the first vaccination, and in 75, 100, 88 and 86% of Groups A–D vaccine recipients after the second vaccination, respectively. The median spot forming cells (SFC) per 106 PBMC to any antigen was 78–139 across Groups A–C and 158–174 in Group D, after each of the vaccinations with a maximum of 2991 SFC. Four to five HIV proteins were commonly recognized across all the groups and over multiple timepoints. CD4+ and CD8+ T-cell responses were polyfunctional. Env antibodies were detected in all Group A–C vaccinees and Gag antibodies in most vaccinees after the second immunization. Ad35 neutralizing titers remained low after the second vaccination. Conclusion/Significance Ad35-GRIN/ENV reactogenicity was dose-related. HIV-specific cellular and humoral responses were seen in the majority of volunteers immunized with Ad35-GRIN/ENV or Ad35-GRIN and increased after the second vaccination. T-cell responses were broad and polyfunctional. Trial Registration ClinicalTrials.gov NCT00851383


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.


PLOS ONE | 2013

Safety and immunogenicity of DNA and MVA HIV-1 subtype C vaccine prime-boost regimens: a phase I randomised Trial in HIV-uninfected Indian volunteers.

Sanjay Mehendale; Madhuri Thakar; Seema Sahay; Makesh Kumar; Ashwini Shete; Pattabiraman Sathyamurthi; Amita Verma; Swarali Kurle; Aparna Shrotri; Jill Gilmour; Rajat Goyal; Len Dally; Eddy Sayeed; Devika Zachariah; James Ackland; Sonali Kochhar; Josephine H. Cox; Jean-Louis Excler; V. Kumaraswami; Ramesh Paranjape; Vadakkuppatu Devasenapathi Ramanathan

Study Design A randomized, double-blind, placebo controlled phase I trial. Methods The trial was conducted in 32 HIV-uninfected healthy volunteers to assess the safety and immunogenicity of prime-boost vaccination regimens with either 2 doses of ADVAX, a DNA vaccine containing Chinese HIV-1 subtype C env gp160, gag, pol and nef/tat genes, as a prime and 2 doses of TBC-M4, a recombinant MVA encoding Indian HIV-1 subtype C env gp160, gag, RT, rev, tat, and nef genes, as a boost in Group A or 3 doses of TBC-M4 alone in Group B participants. Out of 16 participants in each group, 12 received vaccine candidates and 4 received placebos. Results Both vaccine regimens were found to be generally safe and well tolerated. The breadth of anti-HIV binding antibodies and the titres of anti-HIV neutralizing antibodies were significantly higher (p<0.05) in Group B volunteers at 14 days post last vaccination. Neutralizing antibodies were detected mainly against Tier-1 subtype B and C viruses. HIV-specific IFN-γ ELISPOT responses were directed mostly to Env and Gag proteins. Although the IFN-γ ELISPOT responses were infrequent after ADVAX vaccinations, the response rate was significantly higher in group A after 1st and 2nd MVA doses as compared to the responses in group B volunteers. However, the priming effect was short lasting leading to no difference in the frequency, breadth and magnitude of IFN-γELISPOT responses between the groups at 3, 6 and 9 months post-last vaccination. Conclusions Although DNA priming resulted in enhancement of immune responses after 1st MVA boosting, the overall DNA prime MVA boost was not found to be immunologically superior to homologous MVA boosting. Trial Registration Clinical Trial Registry CTRI/2009/091/000051


PLOS ONE | 2012

Heterologous Prime-Boost Regimens Using rAd35 and rMVA Vectors Elicit Stronger Cellular Immune Responses to HIV Proteins Than Homologous Regimens

Silvia Ratto-Kim; Jeffrey R. Currier; Josephine H. Cox; Jean-Louis Excler; Anais Valencia-Micolta; Doris Thelian; Vicky Lo; Eddy Sayeed; Victoria R. Polonis; Patricia L. Earl; Bernard Moss; Merlin L. Robb; Nelson L. Michael; Jerome H. Kim; Mary Marovich

We characterized prime-boost vaccine regimens using heterologous and homologous vector and gene inserts. Heterologous regimens offer a promising approach that focuses the cell-mediated immune response on the insert and away from vector-dominated responses. Ad35-GRIN/ENV (Ad35-GE) vaccine is comprised of two vectors containing sequences from HIV-1 subtype A gag, rt, int, nef (Ad35-GRIN) and env (Ad35-ENV). MVA-CMDR (MVA-C), MVA-KEA (MVA-K) and MVA-TZC (MVA-T) vaccines contain gag, env and pol genes from HIV-1 subtypes CRF01_AE, A and C, respectively. Balb/c mice were immunized with different heterologous and homologous vector and insert prime-boost combinations. HIV and vector-specific immune responses were quantified post-boost vaccination. Gag-specific IFN-γ ELISPOT, intracellular cytokine staining (ICS) (CD107a, IFN-γ, TNF-α and IL-2), pentamer staining and T-cell phenotyping were used to differentiate responses to inserts and vectors. Ad35-GE prime followed by boost with any of the recombinant MVA constructs (rMVA) induced CD8+ Gag-specific responses superior to Ad35-GE-Ad35-GE or rMVA-rMVA prime-boost combinations. Notably, there was a shift toward insert-focus responses using heterologous vector prime-boost regimens. Gag-specific central and effector memory T cells were generated more rapidly and in greater numbers in the heterologous compared to the homologous prime-boost regimens. These results suggest that heterologous prime-boost vaccination regimens enhance immunity by increasing the magnitude, onset and multifunctionality of the insert-specific cell-mediated immune response compared to homologous vaccination regimens. This study supports the rationale for testing heterologous prime-boost regimens in humans.


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.


Biotechnology and Bioengineering | 2018

cGMP production and analysis of BG505 SOSIP.664, an extensively glycosylated, trimeric HIV-1 envelope glycoprotein vaccine candidate

Antu K. Dey; Albert Cupo; Gabriel Ozorowski; Vaneet Kumar Sharma; Anna-Janina Behrens; Eden P. Go; Thomas J. Ketas; Anila Yasmeen; P. J. Klasse; Eddy Sayeed; Heather Desaire; Max Crispin; Ian A. Wilson; Rogier W. Sanders; Thomas Hassell; Andrew B. Ward; John P. Moore

We describe the properties of BG505 SOSIP.664 HIV‐1 envelope glycoprotein trimers produced under current Good Manufacturing Practice (cGMP) conditions. These proteins are the first of a new generation of native‐like trimers that are the basis for many structure‐guided immunogen development programs aimed at devising how to induce broadly neutralizing antibodies (bNAbs) to HIV‐1 by vaccination. The successful translation of this prototype demonstrates the feasibility of producing similar immunogens on an appropriate scale and of an acceptable quality for Phase I experimental medicine clinical trials. BG505 SOSIP.664 trimers are extensively glycosylated, contain numerous disulfide bonds and require proteolytic cleavage, all properties that pose a substantial challenge to cGMP production. Our strategy involved creating a stable CHO cell line that was adapted to serum‐free culture conditions to produce envelope glycoproteins. The trimers were then purified by chromatographic methods using a 2G12 bNAb affinity column and size‐exclusion chromatography. The chosen procedures allowed any adventitious viruses to be cleared from the final product to the required extent of >12 log10. The final cGMP production run yielded 3.52 g (peptidic mass) of fully purified trimers (Drug Substance) from a 200 L bioreactor, a notable yield for such a complex glycoprotein. The purified trimers were fully native‐like as judged by negative‐stain electron microscopy, and were stable over a multi‐month period at room temperature or below and for at least 1 week at 50°C. Their antigenicity, disulfide bond patterns, and glycan composition were consistent with trimers produced on a research laboratory scale. The methods reported here should pave the way for the cGMP production of other native‐like Env glycoprotein trimers of various designs and genotypes.


Retrovirology | 2009

OA05-01. In vivo electroporation enhances the immunogenicity of ADVAX, a DNA-based HIV-1 vaccine candidate, in healthy volunteers

Sandhya Vasan; Arlene Hurley; Sj Schlesinger; D Hannaman; Df Gardiner; Daniel Dugin; Mar Boente-Carrera; Roselle Vittorino; M Caskey; J Andersen; Yaoxing Huang; Josephine H. Cox; Tony Tarragona; Dilbinder K. Gill; Hannah Cheeseman; Lorna Clark; L Dally; C Smith; Claudia Schmidt; Harriet Park; Eddy Sayeed; Jill Gilmour; Patricia Fast; R Bernard; David D. Ho

Open Access Oral presentation OA05-01. In vivo electroporation enhances the immunogenicity of ADVAX, a DNA-based HIV-1 vaccine candidate, in healthy volunteers S Vasan*1, A Hurley1, SJ Schlesinger1, D Hannaman2, DF Gardiner1, DP Dugin1, MM Boente-Carrera1, RM Vittorino1, M Caskey1, J Andersen1, Y Huang1, J Cox3, T Tarragona3, DK Gill3, H Cheeseman3, L Clark3, L Dally4, C Smith4, C Schmidt3, H Park3, E Sayeed3, J Gilmour3, P Fast3, R Bernard2 and DD Ho1

Collaboration


Dive into the Eddy Sayeed's collaboration.

Top Co-Authors

Avatar

Josephine H. Cox

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

Angela Lombardo

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

Jean-Louis Excler

Henry M. Jackson Foundation for the Advancement of Military Medicine

View shared research outputs
Top Co-Authors

Avatar

Len Dally

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Lorna Clark

International AIDS Vaccine Initiative

View shared research outputs
Top Co-Authors

Avatar

Dilbinder K. Gill

International AIDS Vaccine Initiative

View shared research outputs
Top Co-Authors

Avatar

James Ackland

International AIDS Vaccine Initiative

View shared research outputs
Top Co-Authors

Avatar

Peter Hayes

International AIDS Vaccine Initiative

View shared research outputs
Researchain Logo
Decentralizing Knowledge