Network


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

Hotspot


Dive into the research topics where Iole Macchia is active.

Publication


Featured researches published by Iole Macchia.


Vaccine | 2001

Vaccination with DNA containing tat coding sequences and unmethylated CpG motifs protects cynomolgus monkeys upon infection with simian/human immunodeficiency virus (SHIV89.6P)

Aurelio Cafaro; Fausto Titti; Claudio Fracasso; M.Teresa Maggiorella; Silvia Baroncelli; Antonella Caputo; Delia Goletti; Alessandra Borsetti; Monica Pace; Emanuele Fanales-Belasio; Barbara Ridolfi; Donatella R.M. Negri; Leonardo Sernicola; Roberto Belli; Franco Corrias; Iole Macchia; Pasqualina Leone; Zuleika Michelini; Peter ten Haaft; Stefano Buttò; Paola Verani; Barbara Ensoli

Recent evidence suggests that a CD8-mediated cytotoxic T cell response against the Tat protein of human immunodeficiency virus (HIV)/simian immunodeficiency virus (SIV) controls primary infection after pathogenic virus challenge, and correlates with the status of long-term nonprogressor in humans. Due to the presence of unmethylated CpG sequences, DNA vaccination can boost the innate immunity driving more potent T cell-mediated immune responses. Therefore, cynomolgus monkeys were vaccinated with a tat-expressing vector containing defined unmethylated CpG sequences (pCV-tat). Here it is shown that the intramuscular inoculation of the pCV-tat contained primary infection with the highly pathogenic SHIV89.6P virus preventing the CD4(+) T cell decline in all the vaccinated monkeys. Undetectable virus replication and negative virus isolation correlated in all cases with the presence of anti-Tat CTLs. However, a CD8-mediated non cytolytic antiviral activity was also present in all protected animals. Of note, this activity was absent in the controls but was present in the monkey inoculated with the CpG-rich vector alone that was partially protected against viral challenge (i.e. no virus replication but positive virus isolation). These results suggest that a CTL response against Tat protects against primary infection by blocking virus replication at its early stage, in the absence of sterilizing immunity. Nevertheless, the boost of the innate immunity by CpG sequences can contribute to this protection both by driving more potent CTL responses and by inducing other CD8-mediated antiviral activities. Thus, the CpG-rich tat DNA vaccine may represent a promising candidate for preventive and therapeutic vaccination against AIDS.


Journal of Medical Primatology | 2003

SHIV89.6P pathogenicity in cynomolgus monkeys and control of viral replication and disease onset by human immunodeficiency virus type 1 Tat vaccine

Aurelio Cafaro; Antonella Caputo; Maria Teresa Maggiorella; Silvia Baroncelli; Claudio Fracasso; Monica Pace; Alessandra Borsetti; Leonardo Sernicola; Donatella R.M. Negri; Peter ten Haaft; Monica Betti; Zuleika Michelini; Iole Macchia; Emanuele Fanales-Belasio; Roberto Belli; Franco Corrias; Stefano Buttò; Paola Verani; Fausto Titti; Barbara Ensoli

The Tat protein of human immunodeficiency virus (HIV) is produced very early after infection, plays a key role in the virus life cycle and in acquired immunodeficiency syndrome (AIDS) pathogenesis, is immunogenic and well conserved among all virus clades. Notably, a Tat‐specific immune response correlates with non‐progression to AIDS. Here, we show that a vaccine based on the Tat protein of HIV blocks primary infection with the simian/human immunodeficiency virus (SHIV)89.6P and prevents the CD4 T cell decline and disease onset in cynomolgus monkeys. No signs of virus replication were found in five out of seven vaccinated macaques for almost 1 year of follow‐up. Since the inoculated virus (derived from rhesus or from cynomolgus macaques) is shown to be highly pathogenic in cynomolgus macaques, the results indicate efficacy of Tat vaccination in protection against highly pathogenic virus challenge. Finally, the studies of the Tat‐specific immunological responses indicate a correlation of protection with a cytotoxic T cell response. Thus, a Tat‐based vaccine is a promising candidate for preventive and therapeutic vaccination in humans.


Journal of Immunology | 2009

HIV-1 Tat addresses dendritic cells to induce a predominant Th1-type adaptive immune response that appears prevalent in the asymptomatic stage of infection

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.


DNA and Cell Biology | 2002

HIV-1 Tat-Based Vaccines: From Basic Science to Clinical Trials

Emanuele Fanales-Belasio; Aurelio Cafaro; Andrea Cara; Donatella R.M. Negri; Valeria Fiorelli; Stefano Buttò; Sonia Moretti; Maria Teresa Maggiorella; Silvia Baroncelli; Zuleika Michelini; Antonella Tripiciano; Leonardo Sernicola; Arianna Scoglio; Alessandra Borsetti; Barbara Ridolfi; Roberta Bona; Peter ten Haaft; Iole Macchia; Pasqualina Leone; Maria Rosaria Pavone-Cossut; Filomena Nappi; Eftyhia Vardas; Mauro Magnani; Elena Laguardia; Antonella Caputo; Fausto Titti; Barbara Ensoli

Vaccination against human immunodeficiency virus (HIV)-1 infection requires candidate antigen(s) (Ag) capable of inducing an effective, broad, and long-lasting immune response against HIV-1 despite mutation events leading to differences in virus clades. The HIV-1 Tat protein is more conserved than envelope proteins, is essential in the virus life cycle and is expressed very early upon virus entry. In addition, both humoral and cellular responses to Tat have been reported to correlate with a delayed progression to disease in both humans and monkeys. This suggested that Tat is an optimal target for vaccine development aimed at controlling virus replication and blocking disease onset. Here are reviewed the results of our studies including the effects of the Tat protein on monocyte-derived dendritic cells (MDDCs) that are key antigen-presenting cells (APCs), and the results from vaccination trials with both the Tat protein or tat DNA in monkeys. We provide evidence that the HIV-1 Tat protein is very efficiently taken up by MDDCs and promotes T helper (Th)-1 type immune responses against itself as well as other Ag. In addition, a Tat-based vaccine elicits an immune response capable of controlling primary infection of monkeys with the pathogenic SHIV89.6P at its early stages allowing the containment of virus spread. Based on these results and on data of Tat conservation and immune cross-recognition in field isolates from different clades, phase I clinical trials are being initiated in Italy for both preventive and therapeutic vaccination.


PLOS ONE | 2012

HIV-1 Tat Promotes Integrin-Mediated HIV Transmission to Dendritic Cells by Binding Env Spikes and Competes Neutralization by Anti-HIV Antibodies

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.


Human Gene Therapy | 2010

Transduction of Human Antigen-Presenting Cells with Integrase-Defective Lentiviral Vector Enables Functional Expansion of Primed Antigen-Specific CD8+ T Cells

Donatella R.M. Negri; Roberta Bona; Zuleika Michelini; Pasqualina Leone; Iole Macchia; Mary E. Klotman; Mirella Salvatore; Andrea Cara

Nonintegrating lentiviral vectors are being developed as a efficient and safe delivery system for both gene therapy and vaccine purposes. Several reports have demonstrated that a single immunization with integration-defective lentiviral vectors (IDLVs) delivering viral or tumor model antigens in mice was able to elicit broad and long-lasting specific immune responses in the absence of vector integration. At present, no evidence has been reported showing that IDLVs are able to expand preexisting immune responses in the human context. In the present study, we demonstrate that infection of human antigen-presenting cells (APCs), such as monocyte-derived dendritic cells (DCs) and macrophages with IDLVs expressing influenza matrix M1 protein resulted in effective induction of in vitro expansion of M1-primed CD8(+) T cells, as evaluated by both pentamer staining and cytokine production. This is the first demonstration that IDLVs represent an efficient delivery system for gene transfer and expression in human APCs, useful for immunotherapeutic applications.


Journal of Medical Primatology | 2007

Multiprotein genetic vaccine in the SIV-Macaca animal model: a promising approach to generate sterilizing immunity to HIV infection.

Maria Teresa Maggiorella; Leonardo Sernicola; Federica Crostarosa; Roberto Belli; Maria Rosaria Pavone-Cossut; Iole Macchia; Stefania Farcomeni; Klara Tenner-Racz; Paul Racz; Barbara Ensoli; Fausto Titti

Background  Vaccine combining structural and regulatory proteins is an emerging approach to develop an HIV/AIDS vaccine and therefore, the immunogenicity and efficacy of two regimens of immunization combining structural (Gag/Pol, Env) and regulatory (Rev, Tat, Nef) Simian immunodeficiency virus (SIV) proteins were compared in cynomolgus monkeys.


BioMed Research International | 2013

Immune Monitoring in Cancer Vaccine Clinical Trials: Critical Issues of Functional Flow Cytometry-Based Assays

Iole Macchia; Francesca Urbani; Enrico Proietti

The development of immune monitoring assays is essential to determine the immune responses against tumor-specific antigens (TSAs) and tumor-associated antigens (TAAs) and their possible correlation with clinical outcome in cancer patients receiving immunotherapies. Despite the wide range of techniques used, to date these assays have not shown consistent results among clinical trials and failed to define surrogate markers of clinical efficacy to antitumor vaccines. Multiparameter flow cytometry- (FCM-) based assays combining different phenotypic and functional markers have been developed in the past decade for informative and longitudinal analysis of polyfunctional T-cells. These technologies were designed to address the complexity and functional heterogeneity of cancer biology and cellular immunity and to define biomarkers predicting clinical response to anticancer treatment. So far, there is still a lack of standardization of some of these immunological tests. The aim of this review is to overview the latest technologies for immune monitoring and to highlight critical steps involved in some of the FCM-based cellular immune assays. In particular, our laboratory is focused on melanoma vaccine research and thus our main goal was the validation of a functional multiparameter test (FMT) combining different functional and lineage markers to be applied in clinical trials involving patients with melanoma.


Advances in Experimental Medicine and Biology | 2009

Innovative approaches to develop prophylactic and therapeutic vaccines against HIV/AIDS.

Aurelio Cafaro; Iole Macchia; Maria Teresa Maggiorella; Fausto Titti; Barbara Ensoli

The acquired immunodeficiency syndrome (AIDS) emerged in the human population in the summer of 1981. According to the latest United Nations estimates, worldwide over 33 million people are infected with human immunodeficiency virus (HIV) and the prevalence rates continue to rise globally. To control the alarming spread of HIV, an urgent need exists for developing a safe and effective vaccine that prevents individuals from becoming infected or progressing to disease. To be effective, an HIV/AIDS vaccine should induce broad and long-lasting humoral and cellular immune responses, at both mucosal and systemic level. However, the nature of protective immune responses remains largely elusive and this represents one of the major roadblocks preventing the development of an effective vaccine. Here we summarize our present understanding of the factors responsible for resistance to infection or control of progression to disease in human and monkey that may be relevant to vaccine development and briefly review recent approaches which are currently being tested in clinical trials. Finally, the rationale and the current status of novel strategies based on nonstructural HIV-1 proteins, such as Tat, Nef and Rev, used alone or in combination with modified structural HIV-1 Env proteins are discussed.


Viral Immunology | 2009

Containment of Infection in Tat Vaccinated Monkeys After Rechallenge with a Higher Dose of SHIV89.6Pcy243

Alessandra Borsetti; Silvia Baroncelli; Maria Teresa Maggiorella; Sonia Moretti; Emanuele Fanales-Belasio; Leonardo Sernicola; Antonella Tripiciano; Iole Macchia; Zuleika Michelini; Roberto Belli; Stefania Farcomeni; Maria Rosaria Pavone-Cossut; Donatella R.M. Negri; Antonella Caputo; Stefania Bellino; Stefano Buttò; Fausto Titti; Aurelio Cafaro; Barbara Ensoli

We previously reported that cynomolgus monkeys vaccinated with the human immunodeficiency virus (HIV)-1 Tat protein controlled infection after challenge with the simian human immunodeficiency virus (SHIV) 89.6P(cy243) for up to 2 y of follow-up. To evaluate the breadth of the protective immunity elicited by the Tat protein, the vaccines along with the naïve monkeys were intravenously rechallenged with a fivefold higher dose (50 MID(50)) of the same SHIV-89.6P(cy243). The vaccinated monkeys exhibited a statistically significant and long-lasting reduction of viral replication compared to control monkeys. This effect was associated with a strong anamnestic response to Tat, while responses to Gag and Env were nearly undetectable. Taken together, these data provide further evidence for the usefulness of Tat-based vaccines.

Collaboration


Dive into the Iole Macchia's collaboration.

Top Co-Authors

Avatar

Fausto Titti

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Leonardo Sernicola

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Aurelio Cafaro

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Barbara Ensoli

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Donatella R.M. Negri

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Silvia Baroncelli

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Pasqualina Leone

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Zuleika Michelini

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge