Network


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

Hotspot


Dive into the research topics where Jakub Kopycinski is active.

Publication


Featured researches published by Jakub Kopycinski.


PLOS ONE | 2011

In Vivo Electroporation Enhances the Immunogenicity of an HIV-1 DNA Vaccine Candidate in Healthy Volunteers

Sandhya Vasan; Arlene Hurley; Sarah J. Schlesinger; Drew Hannaman; David F. Gardiner; Daniel Dugin; Mar Boente-Carrera; Roselle Vittorino; Marina Caskey; Johanne Andersen; Yaoxing Huang; Josephine H. Cox; Tony Tarragona-Fiol; Dilbinder K. Gill; Hannah Cheeseman; Lorna Clark; Len Dally; Carol Smith; Claudia Schmidt; Harriet Park; Jakub Kopycinski; Jill Gilmour; Patricia Fast; Robert M. Bernard; David D. Ho

Background DNA-based vaccines have been safe but weakly immunogenic in humans to date. Methods and Findings We sought to determine the safety, tolerability, and immunogenicity of ADVAX, a multigenic HIV-1 DNA vaccine candidate, injected intramuscularly by in vivo electroporation (EP) in a Phase-1, double-blind, randomized placebo-controlled trial in healthy volunteers. Eight volunteers each received 0.2 mg, 1 mg, or 4 mg ADVAX or saline placebo via EP, or 4 mg ADVAX via standard intramuscular injection at weeks 0 and 8. A third vaccination was administered to eleven volunteers at week 36. EP was safe, well-tolerated and considered acceptable for a prophylactic vaccine. EP delivery of ADVAX increased the magnitude of HIV-1-specific cell mediated immunity by up to 70-fold over IM injection, as measured by gamma interferon ELISpot. The number of antigens to which the response was detected improved with EP and increasing dosage. Intracellular cytokine staining analysis of ELISpot responders revealed both CD4+ and CD8+ T cell responses, with co-secretion of multiple cytokines. Conclusions This is the first demonstration in healthy volunteers that EP is safe, tolerable, and effective in improving the magnitude, breadth and durability of cellular immune responses to a DNA vaccine candidate. Trial Registration ClinicalTrials.gov NCT00545987


Journal of Clinical Investigation | 2014

Immune activation alters cellular and humoral responses to yellow fever 17D vaccine

Enoch Muyanja; Aloysius Ssemaganda; Pearline Ngauv; Rafael Cubas; Hélène Perrin; Divya Srinivasan; Glenda Canderan; Benton Lawson; Jakub Kopycinski; Amanda S. Graham; Dawne K. Rowe; Michaela J. Smith; Denis Gaucher; Sharon Isern; Scott F. Michael; Guido Silvestri; Thomas H. Vanderford; Erika Castro; Giuseppe Pantaleo; Joel Singer; Jill Gillmour; Noah Kiwanuka; Annet Nanvubya; Claudia Schmidt; Josephine Birungi; Josephine H. Cox; Elias K. Haddad; Pontiano Kaleebu; Patricia Fast; Rafick-Pierre Sekaly

BACKGROUND Defining the parameters that modulate vaccine responses in African populations will be imperative to design effective vaccines for protection against HIV, malaria, tuberculosis, and dengue virus infections. This study aimed to evaluate the contribution of the patient-specific immune microenvironment to the response to the licensed yellow fever vaccine 17D (YF-17D) in an African cohort. METHODS We compared responses to YF-17D in 50 volunteers in Entebbe, Uganda, and 50 volunteers in Lausanne, Switzerland. We measured the CD8+ T cell and B cell responses induced by YF-17D and correlated them with immune parameters analyzed by flow cytometry prior to vaccination. RESULTS We showed that YF-17D-induced CD8+ T cell and B cell responses were substantially lower in immunized individuals from Entebbe compared with immunized individuals from Lausanne. The impaired vaccine response in the Entebbe cohort associated with reduced YF-17D replication. Prior to vaccination, we observed higher frequencies of exhausted and activated NK cells, differentiated T and B cell subsets and proinflammatory monocytes, suggesting an activated immune microenvironment in the Entebbe volunteers. Interestingly, activation of CD8+ T cells and B cells as well as proinflammatory monocytes at baseline negatively correlated with YF-17D-neutralizing antibody titers after vaccination. Additionally, memory T and B cell responses in preimmunized volunteers exhibited reduced persistence in the Entebbe cohort but were boosted by a second vaccination. CONCLUSION Together, these results demonstrate that an activated immune microenvironment prior to vaccination impedes efficacy of the YF-17D vaccine in an African cohort and suggest that vaccine regimens may need to be boosted in African populations to achieve efficient immunity. TRIAL REGISTRATION Registration is not required for observational studies. FUNDING This study was funded by Canadas Global Health Research Initiative, Defense Threat Reduction Agency, National Institute of Allergy and Infectious Diseases, Bill & Melinda Gates Foundation, and United States Agency for International Development.


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


Journal of Virology | 2011

Reappraisal of the Relationship between the HIV-1-Protective Single-Nucleotide Polymorphism 35 Kilobases Upstream of the HLA-C Gene and Surface HLA-C Expression

Tumena Wandifa Corrah; Nilu Goonetilleke; Jakub Kopycinski; Steven G. Deeks; Myron S. Cohen; Persephone Borrow; Andrew J. McMichael; Simon Brackenridge

ABSTRACT Previous studies have found an association between a single-nucleotide polymorphism 35 kb upstream of the HLA-C locus (−35 SNP), HLA-C expression, and HIV-1 set point viral loads. We show that the difference in HLA-C expression across −35 SNP genotypes can be attributed primarily to the very low expression of a single allelic product, HLA-Cw7, which is a common HLA type. We suggest that association of the −35 SNP and HIV-1 load manifests as a result of linkage disequilibrium of this polymorphism with both favorable and unfavorable HLA-C and -B alleles.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Replicative fitness of transmitted HIV-1 drives acute immune activation, proviral load in memory CD4+ T cells, and disease progression

Daniel T. Claiborne; Jessica L. Prince; Eileen Scully; Gladys Macharia; Luca Micci; Benton Lawson; Jakub Kopycinski; Martin J. Deymier; Thomas H. Vanderford; Krystelle Nganou-Makamdop; Zachary Ende; Kelsie Brooks; Jianming Tang; Tianwei Yu; Shabir Lakhi; William Kilembe; Guido Silvestri; Paul A. Goepfert; Matthew Price; Susan Allen; Mirko Paiardini; Marcus Altfeld; Jill Gilmour; Eric Hunter

Significance HIV infection is associated with elevated inflammation and aberrant cellular immune activation. Indeed, the activation status of an HIV-infected individual is often more predictive of disease trajectory than viral load. Here, we highlight the importance of the replicative fitness of the transmitted viral variant in driving an early inflammatory state, characterized by T-cell activation and immune dysfunction. This impact on T-cell homeostasis is independent of protective host immune response genes and viral load. Highly replicating transmitted variants were also significantly more efficient at infecting memory CD4+ T cells, a population important for maintaining the latent viral reservoir. Together, these data provide a mechanism whereby viral replicative fitness acts as a major determinant of disease progression and persistence. HIV-1 infection is characterized by varying degrees of chronic immune activation and disruption of T-cell homeostasis, which impact the rate of disease progression. A deeper understanding of the factors that influence HIV-1–induced immunopathology and subsequent CD4+ T-cell decline is critical to strategies aimed at controlling or eliminating the virus. In an analysis of 127 acutely infected Zambians, we demonstrate a dramatic and early impact of viral replicative capacity (vRC) on HIV-1 immunopathogenesis that is independent of viral load (VL). Individuals infected with high-RC viruses exhibit a distinct inflammatory cytokine profile as well as significantly elevated T-cell activation, proliferation, and CD8+ T-cell exhaustion, during the earliest months of infection. Moreover, the vRC of the transmitted virus is positively correlated with the magnitude of viral burden in naive and central memory CD4+ T-cell populations, raising the possibility that transmitted viral phenotypes may influence the size of the initial latent viral reservoir. Taken together, these findings support an unprecedented role for the replicative fitness of the founder virus, independent of host protective genes and VL, in influencing multiple facets of HIV-1–related immunopathology, and that a greater focus on this parameter could provide novel approaches to clinical interventions.


PLOS ONE | 2010

Equivalence of ELISpot Assays Demonstrated between Major HIV Network Laboratories

Dilbinder K. Gill; Yunda Huang; Gail Levine; Anna Sambor; Donald K. Carter; Alicia Sato; Jakub Kopycinski; Peter Hayes; Bridget Hahn; Josephine Birungi; Tony Tarragona-Fiol; Hong Wan; Mark Randles; Andrew Cooper; Aloysius Ssemaganda; Lorna Clark; Pontiano Kaleebu; Steven G. Self; Richard A. Koup; Blake Wood; M. Juliana McElrath; Josephine H. Cox; John Hural; Jill Gilmour

Background The Comprehensive T Cell Vaccine Immune Monitoring Consortium (CTC-VIMC) was created to provide standardized immunogenicity monitoring services for HIV vaccine trials. The ex vivo interferon-gamma (IFN-γ) ELISpot is used extensively as a primary immunogenicity assay to assess T cell-based vaccine candidates in trials for infectious diseases and cancer. Two independent, GCLP-accredited central laboratories of CTC-VIMC routinely use their own standard operating procedures (SOPs) for ELISpot within two major networks of HIV vaccine trials. Studies are imperatively needed to assess the comparability of ELISpot measurements across laboratories to benefit optimal advancement of vaccine candidates. Methods We describe an equivalence study of the two independently qualified IFN-g ELISpot SOPs. The study design, data collection and subsequent analysis were managed by independent statisticians to avoid subjectivity. The equivalence of both response rates and positivity calls to a given stimulus was assessed based on pre-specified acceptance criteria derived from a separate pilot study. Findings Detection of positive responses was found to be equivalent between both laboratories. The 95% C.I. on the difference in response rates, for CMV (−1.5%, 1.5%) and CEF (−0.4%, 7.8%) responses, were both contained in the pre-specified equivalence margin of interval [−15%, 15%]. The lower bound of the 95% C.I. on the proportion of concordant positivity calls for CMV (97.2%) and CEF (89.5%) were both greater than the pre-specified margin of 70%. A third CTC-VIMC central laboratory already using one of the two SOPs also showed comparability when tested in a smaller sub-study. Interpretation The described study procedure provides a prototypical example for the comparison of bioanalytical methods in HIV vaccine and other disease fields. This study also provides valuable and unprecedented information for future vaccine candidate evaluations on the comparison and pooling of ELISpot results generated by the CTC-VIMC central core laboratories.


Journal of Virology | 2011

Differences in HIV-Specific T Cell Responses between HIV-Exposed and -Unexposed HIV-Seronegative Individuals

Adam J. Ritchie; Suzanne Campion; Jakub Kopycinski; Zoe Moodie; Z. Maggie Wang; Kruti Pandya; Stephen Moore; Michael K. P. Liu; Simon Brackenridge; Kristin Kuldanek; Kenneth Legg; Myron S. Cohen; Eric Delwart; Barton F. Haynes; Sarah Fidler; Andrew J. McMichael; Nilu Goonetilleke

ABSTRACT HIV-1-specific T lymphocyte responses in individuals exposed to HIV-1 but who remain persistently seronegative (HESNs) have been reported in some but not all previous studies. This study was designed to resolve unequivocally the question of whether HESNs make HIV-1-specific T cell responses. We performed a blind investigation to measure HIV-1-specific T cell responses in both HIV-1-serodiscordant couples and HIV-1-unexposed seronegative controls (HUSNs). We found low-frequency HIV-1-specific T cells in both HESNs and HUSNs but show that the response rates were higher over time in the former (P = 0.01). Furthermore, the magnitudes of the HIV-1-specific T cell responses were significantly higher among responding HESNs than among HUSNs over time (P = 0.002). In both groups, responses were mediated by CD4 T cells. The responses were mapped to single peptides, which often corresponded to epitopes restricted by multiple HLA-DR types that have previously been detected in HIV-1-infected patients. HIV-1-specific T cell responses in HUSNs and some HESNs likely represent cross-reactivity to self or foreign non-HIV-1 antigens. The significantly greater T cell responses in HESNs, including in two who were homozygous for CCR5Δ32, demonstrates that HIV-1-specific T cell responses can be induced or augmented by exposure to HIV-1 without infection.


PLOS ONE | 2015

A Phase I Double Blind, Placebo-Controlled, Randomized Study of the Safety and Immunogenicity of Electroporated HIV DNA with or without Interleukin 12 in Prime-Boost Combinations with an Ad35 HIV Vaccine in Healthy HIV-Seronegative African Adults

Juliet Mpendo; Gaudensia Mutua; Julien Nyombayire; Rosine Ingabire; Annet Nanvubya; Omu Anzala; Etienne Karita; Peter Hayes; Jakub Kopycinski; Len Dally; Drew Hannaman; Michael A. Egan; John H. Eldridge; Kristen Syvertsen; Jennifer Lehrman; Beth Rasmussen; Jill Gilmour; Josephine H. Cox; Patricia Fast; Claudia Schmidt

Background Strategies to enhance the immunogenicity of DNA vaccines in humans include i) co-administration of molecular adjuvants, ii) intramuscular administration followed by in vivo electroporation (IM/EP) and/or iii) boosting with a different vaccine. Combining these strategies provided protection of macaques challenged with SIV; this clinical trial was designed to mimic the vaccine regimen in the SIV study. Methods Seventy five healthy, HIV-seronegative adults were enrolled into a phase 1, randomized, double-blind, placebo-controlled trial. Multi-antigenic HIV (HIVMAG) plasmid DNA (pDNA) vaccine alone or co-administered with pDNA encoding human Interleukin 12 (IL-12) (GENEVAX IL-12) given by IM/EP using the TriGrid Delivery System was tested in different prime-boost regimens with recombinant Ad35 HIV vaccine given IM. Results All local reactions but one were mild or moderate. Systemic reactions and unsolicited adverse events including laboratory abnormalities did not differ between vaccine and placebo recipients. No serious adverse events (SAEs) were reported. T cell and antibody response rates after HIVMAG (x3) prime—Ad35 (x1) boost were independent of IL-12, while the magnitude of interferon gamma (IFN-γ) ELISPOT responses was highest after HIVMAG (x3) without IL-12. The quality and phenotype of T cell responses shown by intracellular cytokine staining (ICS) were similar between groups. Inhibition of HIV replication by autologous T cells was demonstrated after HIVMAG (x3) prime and was boosted after Ad35. HIV specific antibodies were detected only after Ad35 boost, although there was a priming effect with 3 doses of HIVMAG with or without IL-12. No anti-IL-12 antibodies were detected. Conclusion The vaccines were safe, well tolerated and moderately immunogenic. Repeated administration IM/EP was well accepted. An adjuvant effect of co-administered plasmid IL-12 was not detected. Trial Registration ClinicalTrials.gov NCT01496989


Clinical and Vaccine Immunology | 2012

A DNA-Based Candidate HIV Vaccine Delivered via In Vivo Electroporation Induces CD4 Responses toward the α4β7-Binding V2 Loop of HIV gp120 in Healthy Volunteers

Jakub Kopycinski; Hannah Cheeseman; Ambreen Ashraf; Dilbinder K. Gill; Peter Hayes; Drew Hannaman; Jill Gilmour; Josephine H. Cox; Sandhya Vasan

ABSTRACT Administration of a clade C/B′ candidate HIV-1 DNA vaccine, ADVAX, by in vivo electroporation (EP) was safe and more immunogenic than intramuscular administration without EP. The breadth and specificity of T-cell responses to full-length Env were mapped. Responses to multiple Env regions were induced, with most focusing on V3/C4 and V2 regions, including the α4β7 integrin-binding domain. The breadth of responses induced by this DNA vaccine regimen was comparable to that of viral-vectored vaccine regimens.


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.

Collaboration


Dive into the Jakub Kopycinski's collaboration.

Top Co-Authors

Avatar

Jill Gilmour

International AIDS Vaccine Initiative

View shared research outputs
Top Co-Authors

Avatar

Peter Hayes

International AIDS Vaccine Initiative

View shared research outputs
Top Co-Authors

Avatar

Josephine H. Cox

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

Dilbinder K. Gill

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
Top Co-Authors

Avatar

Ambreen Ashraf

International AIDS Vaccine Initiative

View shared research outputs
Top Co-Authors

Avatar

Lorna Clark

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
Researchain Logo
Decentralizing Knowledge