Stefania Bellino
Istituto Superiore di Sanità
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Stefania Bellino.
PLOS ONE | 2010
Barbara Ensoli; Stefania Bellino; Antonella Tripiciano; Olimpia Longo; Vittorio Francavilla; Simone Marcotullio; Aurelio Cafaro; Orietta Picconi; Giovanni Paniccia; Arianna Scoglio; Angela Arancio; Cristina Ariola; Maria Alvarez; Massimo Campagna; Donato Scaramuzzi; Cristina Iori; Roberto Esposito; Cristina Mussini; Florio Ghinelli; Laura Sighinolfi; Guido Palamara; Alessandra Latini; Gioacchino Angarano; Nicoletta Ladisa; Fabrizio Soscia; Vito S Mercurio; Adriano Lazzarin; Giuseppe Tambussi; Raffaele Visintini; Francesco Mazzotta
Although HAART suppresses HIV replication, it is often unable to restore immune homeostasis. Consequently, non-AIDS-defining diseases are increasingly seen in treated individuals. This is attributed to persistent virus expression in reservoirs and to cell activation. Of note, in CD4+ T cells and monocyte-macrophages of virologically-suppressed individuals, there is continued expression of multi-spliced transcripts encoding HIV regulatory proteins. Among them, Tat is essential for virus gene expression and replication, either in primary infection or for virus reactivation during HAART, when Tat is expressed, released extracellularly and exerts, on both the virus and the immune system, effects that contribute to disease maintenance. Here we report results of an ad hoc exploratory interim analysis (up to 48 weeks) on 87 virologically-suppressed HAART-treated individuals enrolled in a phase II randomized open-label multicentric clinical trial of therapeutic immunization with Tat (ISS T-002). Eighty-eight virologically-suppressed HAART-treated individuals, enrolled in a parallel prospective observational study at the same sites (ISS OBS T-002), served for intergroup comparison. Immunization with Tat was safe, induced durable immune responses, and modified the pattern of CD4+ and CD8+ cellular activation (CD38 and HLA-DR) together with reduction of biochemical activation markers and persistent increases of regulatory T cells. This was accompanied by a progressive increment of CD4+ T cells and B cells with reduction of CD8+ T cells and NK cells, which were independent from the type of antiretroviral regimen. Increase in central and effector memory and reduction in terminally-differentiated effector memory CD4+ and CD8+ T cells were accompanied by increases of CD4+ and CD8+ T cell responses against Env and recall antigens. Of note, more immune-compromised individuals experienced greater therapeutic effects. In contrast, these changes were opposite, absent or partial in the OBS population. These findings support the use of Tat immunization to intensify HAART efficacy and to restore immune homeostasis. Trial registration ClinicalTrials.gov NCT00751595
Journal of Immunology | 2009
Emanuele Fanales-Belasio; Sonia Moretti; Valeria Fiorelli; Antonella Tripiciano; Maria Rosaria Pavone Cossut; Arianna Scoglio; Barbara Collacchi; Filomena Nappi; Iole Macchia; Stefania Bellino; Vittorio Francavilla; Antonella Caputo; Giovanni Barillari; Mauro Magnani; Maria Elena Laguardia; Aurelio Cafaro; Fausto Titti; Paolo Monini; Fabrizio Ensoli; Barbara Ensoli
Tat is an early regulatory protein that plays a major role in human HIV-1 replication and AIDS pathogenesis, and therefore, it represents a key target for the host immune response. In natural infection, however, Abs against Tat are produced only by a small fraction (∼20%) of asymptomatic individuals and are rarely seen in progressors, suggesting that Tat may possess properties diverting the adaptive immunity from generating humoral responses. Here we show that a Th1-type T cell response against Tat is predominant over a Th2-type B cell response in natural HIV-1 infection. This is likely due to the capability of Tat to selectively target and very efficiently enter CD1a-expressing monocyte-derived dendritic cells (MDDC), which represent a primary target for the recognition and response to virus Ag. Upon cellular uptake, Tat induces MDDC maturation and Th1-associated cytokines and β-chemokines production and polarizes the immune response in vitro to the Th1 pattern through the transcriptional activation of TNF-α gene expression. This requires the full conservation of Tat transactivation activity since neither MDDC maturation nor TNF-α production are found with either an oxidized Tat, which does not enter MDDC, or with a Tat protein mutated in the cysteine-rich region (cys22 Tat), which enters MDDC as the wild-type Tat but is transactivation silent. Consistently with these data, inoculation of monkeys with the native wild-type Tat induced a predominant Th1 response, whereas cys22 Tat generated mostly Th2 responses, therefore providing evidence that Tat induces a predominant Th1 polarized adaptive immune response in the host.
Vaccine | 2009
Olimpia Longo; Antonella Tripiciano; Valeria Fiorelli; Stefania Bellino; Arianna Scoglio; Barbara Collacchi; Maria Alvarez; Vittorio Francavilla; Angela Arancio; Giovanni Paniccia; Adriano Lazzarin; Giuseppe Tambussi; Chiara Tassan Din; Raffaele Visintini; Pasquale Narciso; Andrea Antinori; Gianpiero D'Offizi; Marina Giulianelli; Maria Carta; Aldo Di Carlo; Guido Palamara; Massimo Giuliani; Maria Elena Laguardia; Paolo Monini; Mauro Magnani; Fabrizio Ensoli; Barbara Ensoli
A randomized, double blind, placebo-controlled phase I vaccine trial based on the native Tat protein was conducted in HIV-infected asymptomatic individuals. The vaccine was administered five times subcute with alum or intradermally without adjuvant at 7.5microg, 15microg or 30microg doses, respectively. The Tat vaccine was well tolerated both locally and systemically and induced and/or maintained Tat-specific T helper (Th)-1 T-cell responses and Th-2 responses in all subjects with a wide spectrum of functional anti-Tat antibodies, rarely seen in HIV-infected subjects. The data indicate the achievement of both the primary (safety) and secondary (immunogenicity) endpoints of the study.
Vaccine | 2009
Barbara Ensoli; Valeria Fiorelli; Fabrizio Ensoli; Adriano Lazzarin; Raffaele Visintini; Pasquale Narciso; Aldo Di Carlo; Antonella Tripiciano; Olimpia Longo; Stefania Bellino; Vittorio Francavilla; Giovanni Paniccia; Angela Arancio; Arianna Scoglio; Barbara Collacchi; Maria Alvarez; Giuseppe Tambussi; Chiara Tassan Din; Guido Palamara; Alessandra Latini; Andrea Antinori; Gianpiero D’Offizi; Massimo Giuliani; Marina Giulianelli; Maria Carta; Paolo Monini; Mauro Magnani; Enrico Garaci
The native HIV-1 Tat protein was chosen as vaccine candidate for phase I clinical trials based on its role in the natural infection and AIDS pathogenesis, on the association of Tat-specific immune response with the asymptomatic stage as well as on its sequence conservation among HIV clades. A randomized, double blind, placebo-controlled phase I study (ISS P-001) was conducted in healthy adult volunteers without identifiable risk of HIV infection. Tat was administered 5 times monthly, subcute in alum or intradermic alone at 7.5 microg, 15 microg or 30 microg, respectively (ClinicalTrials.gov identifier: NCT00529698). Vaccination with Tat resulted to be safe and well tolerated (primary endpoint) both locally and systemically. In addition, Tat induced both Th1 and Th2 type specific immune responses in all subjects (secondary endpoint) with a wide spectrum of functional antibodies that are rarely seen in natural infection, providing key information for further clinical development of the Tat vaccine candidate.
International Reviews of Immunology | 2009
Antonella Caputo; Riccardo Gavioli; Stefania Bellino; Olimpia Longo; Antonella Tripiciano; Vittorio Francavilla; Cecilia Sgadari; Giovanni Paniccia; Fausto Titti; Aurelio Cafaro; Flavia Ferrantelli; Paolo Monini; Fabrizio Ensoli; Barbara Ensoli
The HIV epidemic continues to represent one of the major problems worldwide, particularly in the Asia and Sub-Saharan regions of the world, with social and economical devastating effects. Although antiretroviral drugs have had a dramatically beneficial impact on HIV-infected individuals that have access to treatment, it has had a negligible impact on the global epidemic. Hence, the inexorable spreading of the HIV pandemic and the increasing deaths from AIDS, especially in developing countries, underscore the urgency for an effective vaccine against HIV/AIDS. However, the generation of such a vaccine has turned out to be extremely challenging. Here we provide an overview on the rationale for the use of non-structural HIV proteins, such as the Tat protein, alone or in combination with other HIV early and late structural HIV antigens, as novel, promising preventative and therapeutic HIV/AIDS vaccine strategies.
Reviews on Recent Clinical Trials | 2009
Stefania Bellino; Vittorio Francavilla; Olimpia Longo; Antonella Tripiciano; Giovanni Paniccia; Angela Arancio; Valeria Fiorelli; Arianna Scoglio; Barbara Collacchi; Marcello Campagna; Adriano Lazzarin; Giuseppe Tambussi; C. Tassan Din; Raffaele Visintini; Pasquale Narciso; Andrea Antinori; Gianpiero D'Offizi; Marinella Giulianelli; Maria Carta; A. Di Carlo; Guido Palamara; Massimo Giuliani; Maria Elena Laguardia; Paolo Monini; Mauro Magnani; Fabrizio Ensoli; Barbara Ensoli
The native HIV-1 Tat protein was chosen as vaccine candidate for phase I clinical trials in both uninfected (ClinicalTrials.gov identifier: NCT00529698) and infected volunteers (ClinicalTrials.gov identifier: NCT00505401). The rationale was based on the role of Tat in the natural infection and AIDS pathogenesis, on the association of Tat-specific immune responses with the asymptomatic stage and slow-progression rate as well as on its sequence conservation among HIV clades (http://www.hiv1tat-vaccines.info/). The parallel conduction in the same clinical centers of randomized, double blind, placebo-controlled phase I studies both in healthy, immunologically competent adults and in HIV-infected, clinically asymptomatic, individuals represents a unique occasion to compare the vaccine-induced immune response in both the preventive and therapeutic setting. In both studies, the same lot of the native Tat protein was administered 5 times, every four weeks, subcute (SC) with alum adjuvant or intradermic (ID), in the absence of adjuvant, at 7.5 microg, 15 microg or 30 microg doses, respectively. The primary and secondary endpoints of these studies were the safety and immunogenicity of the vaccine candidate, respectively. The study lasted 52 weeks and monitoring was conducted for on additional 3 years. The results of both studies indicated that the Tat vaccine is safe and well tolerated both locally and systemically and it is highly immunogenic at all the dosages and by both routes of administration. Vaccination with Tat induced a balanced immune response in uninfected and infected individuals. In particular, therapeutic immunization induced functional antibodies and partially reverted the marked Th1 polarization of anti-Tat immunity seen in natural infection, and elicited a more balanced Th1/Th2 immune response. Further, the number of CD4 T cells correlated positively with anti-Tat antibody titers. Based on these results, a phase II study is ongoing in infected drug-treated individuals (http://www.hiv1tat-vaccines.info/).
PLOS ONE | 2012
Paolo Monini; Aurelio Cafaro; Indresh K. Srivastava; Sonia Moretti; Victoria Sharma; Claudia Andreini; Chiara Chiozzini; Flavia Ferrantelli; Maria Rosaria Pavone Cossut; Antonella Tripiciano; Filomena Nappi; Olimpia Longo; Stefania Bellino; Orietta Picconi; Emanuele Fanales-Belasio; Alessandra Borsetti; Elena Toschi; Ilaria Schiavoni; Ilaria Bacigalupo; Elaine Kan; Leonardo Sernicola; Maria Teresa Maggiorella; Katy Montin; Marco Porcu; Patrizia Leone; Pasqualina Leone; Barbara Collacchi; Clelia Palladino; Barbara Ridolfi; Mario Falchi
Use of Env in HIV vaccine development has been disappointing. Here we show that, in the presence of a biologically active Tat subunit vaccine, a trimeric Env protein prevents in monkeys virus spread from the portal of entry to regional lymph nodes. This appears to be due to specific interactions between Tat and Env spikes that form a novel virus entry complex favoring R5 or X4 virus entry and productive infection of dendritic cells (DCs) via an integrin-mediated pathway. These Tat effects do not require Tat-transactivation activity and are blocked by anti-integrin antibodies (Abs). Productive DC infection promoted by Tat is associated with a highly efficient virus transmission to T cells. In the Tat/Env complex the cysteine-rich region of Tat engages the Env V3 loop, whereas the Tat RGD sequence remains free and directs the virus to integrins present on DCs. V2 loop deletion, which unshields the CCR5 binding region of Env, increases Tat/Env complex stability. Of note, binding of Tat to Env abolishes neutralization of Env entry or infection of DCs by anti-HIV sera lacking anti-Tat Abs, which are seldom present in natural infection. This is reversed, and neutralization further enhanced, by HIV sera containing anti-Tat Abs such as those from asymptomatic or Tat-vaccinated patients, or by sera from the Tat/Env vaccinated monkeys. Thus, both anti-Tat and anti-Env Abs are required for efficient HIV neutralization. These data suggest that the Tat/Env interaction increases HIV acquisition and spreading, as a mechanism evolved by the virus to escape anti-Env neutralizing Abs. This may explain the low effectiveness of Env-based vaccines, which are also unlikely to elicit Abs against new Env epitopes exposed by the Tat/Env interaction. As Tat also binds Envs from different clades, new vaccine strategies should exploit the Tat/Env interaction for both preventative and therapeutic interventions.
International Journal of Cancer | 2011
Elena Toschi; Cecilia Sgadari; Laura Malavasi; Ilaria Bacigalupo; Chiara Chiozzini; Davide Carlei; Daniela Compagnoni; Stefania Bellino; Roberto Bugarini; Mario Falchi; Clelia Palladino; Patrizia Leone; Giovanni Barillari; Paolo Monini; Barbara Ensoli
Human immunodeficiency virus protease inhibitors (HIV‐PIs), such as indinavir and saquinavir, have been shown to block angiogenesis and tumor cell invasion and to induce tumor cell apoptosis and growth arrest, respectively, both in vitro and in vivo. These findings have suggested that HIV‐PIs or their analogues can be used as antitumor drugs. To this regard, indinavir and saquinavir were assessed for their ability to inhibit in vivo the growth of highly prevalent human tumors, such as lung, breast, colon and hepatic adenocarcinomas. We show here that both HIV‐PIs significantly inhibited the growth of all adenocarcinomas tested in the mice model. This was not mediated by effects on proteasome‐dependent cell growth arrest or on apoptosis but by the block of angiogenesis and matrix metalloproteinase activity. Accordingly, therapeutic steadystate concentrations of indinavir or saquinavir were highly effective in inhibiting invasion of tumor cells in vitro. In contrast, growth arrest was induced only by high concentrations of saquinavir that are not reached or are only transiently present in plasma of treated patients, likely through a proteasome‐mediated mechanism. These data suggest that HIV‐PIs or their analogues, characterized by a better biodistribution and lower toxicity, may represent a new class of antitumor drugs capable of targeting both matrix metalloproteinases and the proteasome for a most effective antitumor therapy.
Retrovirology | 2014
Stefania Bellino; Antonella Tripiciano; Orietta Picconi; Vittorio Francavilla; Olimpia Longo; Cecilia Sgadari; Giovanni Paniccia; Angela Arancio; Gioacchino Angarano; Nicoletta Ladisa; Adriano Lazzarin; Giuseppe Tambussi; Silvia Nozza; Carlo Torti; Emanuele Focà; Guido Palamara; Alessandra Latini; Laura Sighinolfi; Francesco Mazzotta; Massimo Di Pietro; Giovanni Di Perri; Stefano Bonora; Vito S Mercurio; Cristina Mussini; Andrea Gori; Massimo Galli; Paolo Monini; Aurelio Cafaro; Fabrizio Ensoli; Barbara Ensoli
BackgroundTat is a key HIV-1 virulence factor, which plays pivotal roles in virus gene expression, replication, transmission and disease progression. After release, extracellular Tat accumulates in tissues and exerts effects on both the virus and the immune system, promoting immune activation and virus spreading while disabling the host immune defense. In particular, Tat binds Env spikes on virus particles forming a virus entry complex, which favors infection of dendritic cells and efficient transmission to T cells via RGD-binding integrins. Tat also shields the CCR5-binding sites of Env rendering ineffective virus neutralization by anti-Env antibodies (Abs). This is reversed by the anti-Tat Abs present in natural infection or induced by vaccination.FindingsHere we present the results of a cohort study, showing that the presence of anti-Tat Abs in asymptomatic and treatment-naïve HIV-infected subjects is associated with containment of CD4+ T-cell loss and viral load and with a delay of disease progression. In fact, no subjects with high anti-Tat Ab titers initiated antiretroviral therapy during the three years of follow-up. In contrast, no significant effects were seen for anti-Env and anti-Gag Abs. The increase of anti-Env Ab titers was associated with a reduced risk of starting therapy only in the presence of anti-Tat Abs, suggesting an effect of combined anti-Tat and anti-Env Abs on the Tat/Env virus entry complex and on virus neutralization.ConclusionsAnti-Tat immunity may help delay HIV disease progression, thus, targeting Tat may offer a novel therapeutic intervention to postpone antiretroviral treatment or to increase its efficacy.
Journal of Virology | 2010
Aurelio Cafaro; Stefania Bellino; Fausto Titti; Maria Teresa Maggiorella; Leonardo Sernicola; Roger W. Wiseman; David Venzon; Julie A. Karl; David H. O'Connor; Paolo Monini; Marjorie Robert-Guroff; Barbara Ensoli
ABSTRACT The effects of the challenge dose and major histocompatibility complex (MHC) class IB alleles were analyzed in 112 Mauritian cynomolgus monkeys vaccinated (n = 67) or not vaccinated (n = 45) with Tat and challenged with simian/human immunodeficiency virus (SHIV) 89.6Pcy243. In the controls, the challenge dose (10 to 20 50% monkey infectious doses [MID50]) or MHC did not affect susceptibility to infection, peak viral load, or acute CD4 T-cell loss, whereas in the chronic phase of infection, the H1 haplotype correlated with a high viral load (P = 0.0280) and CD4 loss (P = 0.0343). Vaccination reduced the rate of infection acquisition at 10 MID50 (P < 0.0001), and contained acute CD4 loss at 15 MID50 (P = 0.0099). Haplotypes H2 and H6 were correlated with increased susceptibility (P = 0.0199) and resistance (P = 0.0087) to infection, respectively. Vaccination also contained CD4 depletion (P = 0.0391) during chronic infection, independently of the challenge dose or haplotype.