Network


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

Hotspot


Dive into the research topics where Igor M. Belyakov is active.

Publication


Featured researches published by Igor M. Belyakov.


Nature Reviews Immunology | 2001

Strategies for designing and optimizing new generation vaccines

Jay A. Berzofsky; Jeffrey D. Ahlers; Igor M. Belyakov

Although the field of immunology developed in part from the early vaccine studies of Edward Jenner, Louis Pasteur and others, vaccine development had largely become the province of virologists and other microbiologists, because the model for classic vaccines was to isolate the pathogen and prepare a killed or attenuated pathogen vaccine. Only recently has vaccinology returned to the realm of immunology, because a new understanding of immune mechanisms has allowed translation of basic discoveries into vaccine strategies.


Journal of Clinical Investigation | 2004

Progress on new vaccine strategies for the immunotherapy and prevention of cancer

Jay A. Berzofsky; Masaki Terabe; SangKon Oh; Igor M. Belyakov; Jeffrey D. Ahlers; John E. Janik; John C. Morris

In recent years, great strides in understanding and regulating the immune system have led to new hope for harnessing its exquisite specificity to destroy cancer cells without affecting normal tissues. This review examines the fundamental immunologic advances and the novel vaccine strategies arising from these advances, as well as the early clinical trials studying new approaches to treat or prevent cancer.


Proceedings of the National Academy of Sciences of the United States of America | 2003

Shared modes of protection against poxvirus infection by attenuated and conventional smallpox vaccine viruses

Igor M. Belyakov; Patricia L. Earl; Amiran Dzutsev; Vladimir A. Kuznetsov; Michael Lemon; Linda S. Wyatt; James T. Snyder; Jeffrey D. Ahlers; Genoveffa Franchini; Bernard Moss; Jay A. Berzofsky

The concern about bioterrorism with smallpox has raised the possibility of widespread vaccination, but the greater prevalence of immunocompromised individuals today requires a safer vaccine, and the mechanisms of protection are not well understood. Here we show that, at sufficient doses, the protection provided by both modified vaccinia Ankara and NYVAC replication-deficient vaccinia viruses, safe in immunocompromised animals, was equivalent to that of the licensed Wyeth vaccine strain against a pathogenic vaccinia virus intranasal challenge of mice. A similar variety and pattern of immune responses were involved in protection induced by modified vaccinia Ankara and Wyeth viruses. For both, antibody was essential to protect against disease, whereas neither effector CD4+ nor CD8+ T cells were necessary or sufficient. However, in the absence of antibody, T cells were necessary and sufficient for survival and recovery. Also, T cells played a greater role in control of sublethal infection in unimmunized animals. These properties, shared with the existing smallpox vaccine, provide a basis for further evaluation of these replication-deficient vaccinia viruses as safer vaccines against smallpox or against complications from vaccinia virus.


Nature Medicine | 2001

Mucosal AIDS vaccine reduces disease and viral load in gut reservoir and blood after mucosal infection of macaques

Igor M. Belyakov; Zdenek Hel; Brian L. Kelsall; Vladimir A. Kuznetsov; Jeffrey D. Ahlers; Janos Nacsa; David I. Watkins; Todd M. Allen; Alessandro Sette; John D. Altman; Ruth Woodward; Phillip D. Markham; John D. Clements; Genoveffa Franchini; Warren Strober; Jay A. Berzofsky

Given the mucosal transmission of HIV-1, we compared whether a mucosal vaccine could induce mucosal cytotoxic T lymphocytes (CTLs) and protect rhesus macaques against mucosal infection with simian/human immunodeficiency virus (SHIV) more effectively than the same vaccine given subcutaneously. Here we show that mucosal CTLs specific for simian immunodeficiency virus can be induced by intrarectal immunization of macaques with a synthetic-peptide vaccine incorporating the LT(R192G) adjuvant. This response correlated with the level of T-helper response. After intrarectal challenge with pathogenic SHIV-Ku2, viral titers were eliminated more completely (to undetectable levels) both in blood and intestine, a major reservoir for virus replication, in intrarectally immunized animals than in subcutaneously immunized or control macaques. Moreover, CD4+ T cells were better preserved. Thus, induction of CTLs in the intestinal mucosa, a key site of virus replication, with a mucosal AIDS vaccine ameliorates infection by SHIV in non-human primates.


Journal of Clinical Investigation | 2004

Transcutaneous immunization induces mucosal CTLs and protective immunity by migration of primed skin dendritic cells

Igor M. Belyakov; Scott A. Hammond; Jeffrey D. Ahlers; Gregory M. Glenn; Jay A. Berzofsky

Transcutaneous immunization (TCI), the application of vaccines on the skin, induces robust systemic and mucosal antibodies in animal models and in humans. The means by which mucosal immune responses to vaccine antigens are elicited by TCI has not been well characterized. We examined the effect of TCI with an HIV peptide vaccine on the induction of mucosal and systemic CTL responses and protective immunity against mucosal challenge with live virus in mice. Robust HIV-specific CTL responses in the spleen and in the gut mucosa were detected after TCI. The responses were dependent upon the addition of an adjuvant and resulted in protection against mucosal challenge with recombinant vaccinia virus encoding HIV gp160. Although it is clear that adjuvant-activated DCs migrated mainly to draining lymph nodes, coculture with specific T cells and flow cytometry studies with DCs isolated from Peyers patches after TCI suggested that activated DCs carrying skin-derived antigen also migrated from the skin to immune-inductive sites in gut mucosa and presented antigen directly to resident lymphocytes. These results and previous clinical trial results support the observation that TCI is a safe and effective strategy for inducing strong mucosal antibody and CTL responses.


Journal of Clinical Investigation | 1998

The importance of local mucosal HIV-specific CD8(+) cytotoxic T lymphocytes for resistance to mucosal viral transmission in mice and enhancement of resistance by local administration of IL-12.

Igor M. Belyakov; Jeffrey D. Ahlers; Benjamin Y. Brandwein; Patricia L. Earl; Brian L. Kelsall; Bernard Moss; Warren Strober; Jay A. Berzofsky

Although crucial to mucosal vaccine development, the mechanisms of defense against mucosal viral infection are still poorly understood. Protection, cytotoxic T lymphocytes (CTL), and neutralizing antibodies have all been observed, but cause and effect have been difficult to determine. The ability of CTL in the mucosa to mediate protection against mucosal viral transmission has never been proven. Here, we use an HIV peptide immunogen and an HIV-1 gp160-expressing recombinant vaccinia viral intrarectal murine challenge system, in which neutralizing antibodies do not play a role, to demonstrate for the first time that long-lasting immune resistance to mucosal viral transmission can be accomplished by CD8(+) CTL that must be present in the mucosal site of exposure. The resistance is ablated by depleting CD8(+) cells in vivo and requires CTL in the mucosa, whereas systemic (splenic) CTL are shown to be unable to protect against mucosal challenge. Furthermore, the resistance as well as the CTL response can be increased by local mucosal delivery of IL-12 with the vaccine. These results imply that induction of local mucosal CTL may be critical for success of a vaccine against viruses transmitted through a mucosal route, such as HIV.


Journal of Immunology | 2009

What Role Does the Route of Immunization Play in the Generation of Protective Immunity against Mucosal Pathogens

Igor M. Belyakov; Jeffrey D. Ahlers

The route of vaccination is important in influencing immune responses at the initial site of pathogen invasion where protection is most effective. Immune responses required for mucosal protection can differ vastly depending on the individual pathogen. For some mucosal pathogens, including acute self-limiting infections, high-titer neutralizing Abs that enter tissue parenchyma or transude into the mucosal lumen are sufficient for clearing cell-free virus. However, for pathogens causing chronic infections such as HIV, hepatitis C virus, herpes viruses, mycobacteria, and fungal and parasitic infections, a single arm of the immune response generated by systemic vaccination may be insufficient for protection. Induction of the mucosal innate and adaptive immune systems, including CD4+ T help, Th17, high avidity CD8+ CTL, and secretory IgA and IgG1 neutralizing Abs, at the site of pathogen entry may be required for effective protection against highly invasive pathogens that lead to chronic infection and may be generated predominantly by mucosal vaccination.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Toll-like receptor ligands synergize through distinct dendritic cell pathways to induce T cell responses: Implications for vaccines

Qing Zhu; Colt Egelston; Aravindhan Vivekanandhan; Satoshi Uematsu; Shizuo Akira; Dennis M. Klinman; Igor M. Belyakov; Jay A. Berzofsky

Toll-like receptors (TLRs) may need to cooperate with each other to be effective in detecting imminent infection and trigger immune responses. Understanding is still limited about the intracellular mechanism of this cooperation. We found that when certain TLRs are involved, dendritic cells (DCs) establish unidirectional intracellular cross-talk, in which the MyD88-independent TRIF-dependent pathway amplifies the MyD88-dependent DC function through a JNK-dependent mechanism. The amplified MyD88-dependent DC function determines the induction of the T cell response to a given vaccine in vivo. Therefore, our study revealed an underlying TLR mechanism governing the functional, nonrandom interplay among TLRs for recognition of combinatorial ligands that may be dangerous to the host, providing important guidance for design of novel synergistic molecular vaccine adjuvants.


Journal of Clinical Investigation | 2004

Progress on new vaccine strategies against chronic viral infections

Jay A. Berzofsky; Jeffrey D. Ahlers; John E. Janik; John C. Morris; SangKon Oh; Masaki Terabe; Igor M. Belyakov

Among the most cost-effective strategies for preventing viral infections, vaccines have proven effective primarily against viruses causing acute, self-limited infections. For these it has been sufficient for the vaccine to mimic the natural virus. However, viruses causing chronic infection do not elicit an immune response sufficient to clear the infection and, as a result, vaccines for these viruses must elicit more effective responses--quantitative and qualitative--than does the natural virus. Here we examine the immunologic and virologic basis for vaccines against three such viruses, HIV, hepatitis C virus, and human papillomavirus, and review progress in clinical trials to date. We also explore novel strategies for increasing the immunogenicity and efficacy of vaccines.


Journal of Clinical Investigation | 2010

Using 3 TLR ligands as a combination adjuvant induces qualitative changes in T cell responses needed for antiviral protection in mice

Qing Zhu; Colt Egelston; Susan J. Gagnon; Yongjun Sui; Igor M. Belyakov; Dennis M. Klinman; Jay A. Berzofsky

TLR ligands are promising candidates for the development of novel vaccine adjuvants that can elicit protective immunity against emerging infectious diseases. Adjuvants have been used most frequently to increase the quantity of an immune response. However, the quality of a T cell response can be more important than its quantity. Stimulating certain pairs of TLRs induces a synergistic response in terms of activating dendritic cells and eliciting/enhancing T cell responses through clonal expansion, which increases the number of responding T cells. Here, we have found that utilizing ligands for 3 TLRs (TLR2/6, TLR3, and TLR9) greatly increased the protective efficacy of vaccination with an HIV envelope peptide in mice when compared with using ligands for only any 2 of these TLRs; surprisingly, increased protection was induced without a marked increase in the number of peptide-specific T cells. Rather, the combination of these 3 TLR ligands augmented the quality of the T cell responses primarily by amplifying their functional avidity for the antigen, which was necessary for clearance of virus. The triple combination increased production of DC IL-15 along with its receptor, IL-15Ralpha, which contributed to high avidity, and decreased expression of programmed death-ligand 1 and induction of Tregs. Therefore, selective TLR ligand combinations can increase protective efficacy by increasing the quality rather than the quantity of T cell responses.

Collaboration


Dive into the Igor M. Belyakov's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeffrey D. Ahlers

Government of the United States of America

View shared research outputs
Top Co-Authors

Avatar

Brian L. Kelsall

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Warren Strober

Government of the United States of America

View shared research outputs
Top Co-Authors

Avatar

Dennis M. Klinman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Amiran Dzutsev

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Genoveffa Franchini

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Qing Zhu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

David H. Margulies

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Susan J. Gagnon

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge