Network


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

Hotspot


Dive into the research topics where Massimo Di Pietro is active.

Publication


Featured researches published by Massimo Di Pietro.


PLOS ONE | 2010

Therapeutic immunization with hiv-1 tat reduces immune activation and loss of regulatory t-cells and improves immune function in subjects on HAART

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


Clinical Infectious Diseases | 2000

Neurological Symptoms during Primary Human Immunodeficiency Virus (HIV) Infection Correlate with High Levels of HIV RNA in Cerebrospinal Fluid

Giuseppe Tambussi; Andrea Gori; Capiluppi B; Claudia Balotta; Laura Papagno; Barbara Morandini; Massimo Di Pietro; Donatella Ciuffreda; Alberto Saracco; Adriano Lazzarin

This analysis involves 22 patients with diagnosed symptomatic human immunodeficiency virus (HIV) infection. Neurologic symptoms were present in 11 patients, ranging from severe and persistent headache to clinical signs suggestive of meningitis. A strong correlation between neurological symptoms and cerebrospinal fluid (CSF) viral load was found. The mean CSF HIV ribonucleic acid (RNA) level was 4. 12 log for patients with neurological symptoms and 2.58 log for patients without neurological symptoms (P<.00001). Plasma viral load alone does not correlate or predict central nervous system (CNS) involvement. In our sample of patients, HIV RNA levels could be detected in most patients regardless of the presence of neurological symptoms. Moreover, early treatment including drugs with high levels of penetration in the CNS must be considered for patients with primary HIV infection.


AIDS | 2010

Emerging mutations at virological failure of HAART combinations containing tenofovir and lamivudine or emtricitabine.

Renato Maserati; Annalisa De Silvestri; Alessia Uglietti; Grazia Colao; Antonio Di Biagio; Bianca Bruzzone; Massimo Di Pietro; Maria Carla Re; Carmine Tinelli; Maurizio Zazzi

Objective:To compare the emergence of drug-resistant HIV variants at failure of lamivudine (3TC)/tenofovir (TDF)-containing or emtricitabine (FTC)/TDF-containing HAART as a consequence of the different 3TC and FTC intracellular half-lives. Design:Retrospective evaluation of 859 patients selected from an Italian HIV resistance database (Antiretroviral Resistance Cohort Analysis). Methods:Patients were selected for analysis if treated with a HAART whose nucleoside/nucleotide reverse transcriptase inhibitor backbone was either 3TC/TDF or FTC/TDF; if they experienced a virological failure after at least 6 months of plasma HIV-RNA undetectability; and if HIV genotypes before treatment and at failure were available. Univariate and multivariate logistic regression analyses were done to detect predictors of resistance mutations emerging at failure. Results:Of 714 patients failing with 3TC/TDF and 145 with FTC/TDF, 35.8 and 21.1% were in Centers for Disease Control and Prevention stage C, and 8.8 and 15.2% were on first-line HAART, respectively. At multivariate analysis, the emergence of K70R (P = 0.002), M184V (P = 0.031), T215F (P = 0.020) and Y181C (P = 0.005) was significantly more common in 3TC-treated than in FTC-treated patients, with an odds ratio of 4, 1.56, 1.89 and 3.84, respectively. Conclusion:Despite their close structural similarity, 3TC and FTC are associated with a significantly different rate of drug resistance at treatment failure when combined with TDF in HAART regimens independently of the third drug used.


PLOS ONE | 2013

Maraviroc as intensification strategy in HIV-1 positive patients with deficient immunological response: an Italian randomized clinical trial.

Stefano Rusconi; Paola Vitiello; Fulvio Adorni; Elisa Colella; Emanuele Focà; Amedeo Capetti; Paola Meraviglia; C. Abeli; Stefano Bonora; Marco D’Annunzio; Antonio Di Biagio; Massimo Di Pietro; Luca Butini; Giancarlo Orofino; Manuela Colafigli; Gabriella d’Ettorre; Daniela Francisci; Giustino Parruti; Alessandro Soria; Anna Rita Buonomini; Chiara Tommasi; Silvia Mosti; Francesca Bai; Silvia Di Nardo Stuppino; Manuela Morosi; Marco Montano; Pamela Tau; Esther Merlini; Giulia Marchetti

Background Immunological non-responders (INRs) lacked CD4 increase despite HIV-viremia suppression on HAART and had an increased risk of disease progression. We assessed immune reconstitution profile upon intensification with maraviroc in INRs. Methods We designed a multi-centric, randomized, parallel, open label, phase 4 superiority trial. We enrolled 97 patients on HAART with CD4+<200/µL and/or CD4+ recovery ≤25% and HIV-RNA<50 cp/mL. Patients were randomized 1:1 to HAART+maraviroc or continued HAART. CD4+ and CD8+ CD45+RA/RO, Ki67 expression and plasma IL-7 were quantified at W0, W12 and W48. Results By W48 both groups displayed a CD4 increase without a significant inter-group difference. A statistically significant change in CD8 favored patients in arm HAART+maraviroc versus HAART at W12 (p=.009) and W48 (p=.025). The CD4>200/µL and CD4>200/µL + CD4 gain ≥25% end-points were not satisfied at W12 (p=.24 and p=.619) nor at W48 (p=.076 and p=.236). Patients continuing HAART displayed no major changes in parameters of T-cell homeostasis and activation. Maraviroc-receiving patients experienced a significant rise in circulating IL-7 by W48 (p=.01), and a trend in temporary reduction in activated HLA-DR+CD38+CD4+ by W12 (p=.06) that was not maintained at W48. Conclusions Maraviroc intensification in INRs did not have a significant advantage in reconstituting CD4 T-cell pool, but did substantially expand CD8. It resulted in a low rate of treatment discontinuations. Trial Registration ClinicalTrials.gov NCT00884858 http://clinicaltrials.gov/show/NCT00884858


Retrovirology | 2014

The presence of anti-Tat antibodies in HIV-infected individuals is associated with containment of CD4+ T-cell decay and viral load, and with delay of disease progression: results of a 3-year cohort study

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.


The Journal of Infectious Diseases | 2006

Frequency and Treatment-Related Predictors of Thymidine-Analogue Mutation Patterns in HIV-1 Isolates after Unsuccessful Antiretroviral Therapy

Andrea De Luca; Simona Di Giambenedetto; Laura Romano; Angela Gonnelli; Paola Corsi; Marco Baldari; Massimo Di Pietro; Stefano Menzo; Daniela Francisci; Paolo Almi; Maurizio Zazzi

We investigated, in patients tested between 1991 and 2004, the patterns of mutually exclusive human immunodeficiency virus-1 thymidine-analogue mutations (TAMs) in 4039 reverse-transcriptase sequences with > or = 1 TAM. TAM pattern 1, which included M41L and L210W and excluded K70R and is coupled with more-extensive cross-resistance to drugs, became the most frequent pattern after 1996. In 1465 genotypes from 684 patients in whom highly active antiretroviral therapy (HAART) was unsuccessful, predictors of this pattern were the number of previous HAART regimens undergone (adjusted odds ratio [OR], 1.09 [95% confidence interval {CI}, 1.02-1.16]), use of stavudine/lamivudine (adjusted OR, 1.42 [95% CI, 1.05-1.99]), use of nevirapine (adjusted OR, 1.60 [95% CI, 1.14-2.24]), use of efavirenz (adjusted OR, 1.56 [95% CI, 1.08-2.27]), and use of ritonavir (adjusted OR, 1.35 [95% CI, 1.04-1.75]).


Hiv Clinical Trials | 2007

Predictors of AIDS-defining events among advanced naïve patients after HAART

Carlo Torti; Giuseppe Lapadula; Franco Maggiolo; Salvatore Casari; Fredy Suter; Lorenzo Minoli; Chiara Pezzoli; Massimo Di Pietro; Guglielmo Migliorino; Eugenia Quiros-Roldan; Nicoletta Ladisa; Laura Sighinolfi; Francesca Gatti; Giampiero Carosi

Abstract Background: Baseline and follow–up predictors of new AIDS-defining events or death (ADE/death) among patients who started HAART late in their disease history have rarely been assessed simultaneously. Method: ADE and mortality rates were assessed using Cox regression analyses. Variables were tested for prediction of ADE/death within the first 3 months of therapy and from month 3, thereafter. Results: 751 HIV–infected patients with <200 CD4+/mm3 before HAART were followed for a median of 49 months. 207 new ADE occurred (7.06 [6.16–8.10] per 100 patient-years). ADE/deaths clustered within the first 3 months of treatment (106/207, 51%). Higher CD4+ T-cell counts during the follow–up (per loge cells/mm3: hazard ratio [HR] 0.51; 0.41–0.64; p < .001) and use of antiretroviral therapy (HR 0.38; 95% CI 0.21–0.69; p = .001) appeared to protect from ADE/death after month 3. Conversely, increasing follow–up with HIV RNA >400 copies/mL correlated with ADE/death (per month: HR 1.09; 95% CI 1.06-1.12; p = .001). Use of boosted protease inhibitors as first-line HAART and HCV-seropositivity were additional risk factors. Baseline CD4+ T-cell count and HIV RNA had a predominant impact in the first 3 months after HAART initiation. Conclusion: A careful monitoring of patients with low CD4+ is particularly necessary during the first few months of HAART. Length and extent of viral replication during the follow-up appeared to induce a significantly higher risk of HIV disease progression afterwards, implying that new drugs and new strategies aimed at ensuring long-term suppression of HIV RNA are of outstanding importance.


Clinical and Vaccine Immunology | 2002

Modulation of Human Immunodeficiency Virus (HIV)-Specific Immune Response by Using Efavirenz, Nelfinavir, and Stavudine in a Rescue Therapy Regimen for HIV-Infected, Drug-Experienced Patients

Daria Trabattoni; Sergio Lo Caputo; Mara Biasin; Elena Seminari; Massimo Di Pietro; Giovanni Ravasi; Francesco Mazzotta; Renato Maserati; Mario Clerici

ABSTRACT Analysis of the virologic and immunomodulatory effects of an association of efavirenz (EFV), nelfinavir (NFV), and stavudine (d4T) was performed in 18 human immunodeficiency virus (HIV)-infected and highly active antiretroviral therapy (HAART)-experienced patients who failed multiple therapeutic protocols. Patients (<500 CD4+ cells/μl; >10,000 HIV copies/ml) were nonnucleoside reverse transcriptase inhibitor (NNRTI)-naive and were treated for 10 months with EFV (600 mg/day) in association with NFV (750 mg three times daily) and d4T (30 or 40 mg twice daily). Measurement of HIV peptide- and mitogen-stimulated production of interleukin-2 (IL-2), gamma interferon (IFN-γ), IL-4, and IL-10 as well as quantitation of mRNA for the same cytokines in unstimulated peripheral blood mononuclear cells were performed at baseline and 2 weeks (t1), 2 months (t2), and 10 months (t3) into therapy. The results showed that HIV-specific (but not mitogen-stimulated) IL-2 and IFN-γ production was augmented and IL-10 production was reduced in patients who received EFV, NFV, and d4T. Therapy was also associated with a reduction in HIV RNA in plasma and an increase in CD4+ cell count. These changes occurred in the first year of therapy (t2 and t3) and were confirmed by quantitation of cytokine-specific mRNA. Therapy with EFV, NFV, and d4T increases HIV-specific type 1 cytokine production as well as CD4 counts and reduces plasma viremia. This therapeutic regimen may be considered for use in cases of advanced HIV infection.


International Journal of Epidemiology | 2015

Cohort Profile: Standardized Management of Antiretroviral Therapy Cohort (MASTER Cohort)

Carlo Torti; Elena Raffetti; Francesco Donato; Francesco Castelli; Franco Maggiolo; Gioacchino Angarano; Francesco Mazzotta; Andrea Gori; Laura Sighinolfi; Angelo Pan; Roberto Cauda; Alfredo Scalzini; Eugenia Quiros-Roldan; Paola Nasta; Giampietro Gregis; Simone Benatti; Simona Digiambenedetto; Nicoletta Ladisa; Mariarosaria Giralda; Annalisa Saracino; Filippo Castelnuovo; Massimo Di Pietro; Sergio Lo Caputo; Giuseppe Lapadula; Silvia Costarelli; Silvia Lorenzotti; Nicola Mazzini; Giuseppe Paraninfo; Salvatore Casari; Emanuele Focà

Cohort Profile: Standardized Management of Antiretroviral Therapy Cohort (MASTER Cohort) Carlo Torti, Elena Raffetti,* Francesco Donato, Francesco Castelli, Franco Maggiolo, Gioacchino Angarano, Francesco Mazzotta, Andrea Gori, Laura Sighinolfi, Angelo Pan, Roberto Cauda, Alfredo Scalzini, Eugenia Quiros-Roldan, Paola Nasta, Giampietro Gregis, Simone Benatti, Simona Digiambenedetto, Nicoletta Ladisa, Mariarosaria Giralda, Annalisa Saracino, Filippo Castelnuovo, Massimo Di Pietro, Sergio Lo Caputo, Giuseppe Lapadula, Silvia Costarelli, Silvia Lorenzotti, Nicola Mazzini, Giuseppe Paraninfo, Salvatore Casari, Emanuele Focà, Chiara Pezzoli, Massimiliano Fabbiani, Laura Monno, Piera Pierotti, Claudio Ble, Sebastiano Leone, Maria Concetta Postorino, Chiara Fornabaio, Fabio Zacchi, Alessia Zoncada and Giampiero Carosi Unità di Malattie Infettive e Tropicali, Dipartimento di Scienze Mediche e Chirurgiche, Università Magna Grecia di Catanzaro, Catanzaro, Italia, Unità di Igiene, Epidemiologia e Sanità Pubblica, Università degli Studi di Brescia, Brescia, Italia, Divisione Universitaria di Malattie Infettive Spedali Civili di Brescia-Università degli Studi di Brescia, Brescia, Italia, Malattie Infettive Ospedale Papa Giovanni XXIII, Bergamo, Italia, Clinica di Malattie Infettive Policlinico di Bari, Bari, Italia, Malattie Infettive S.M. Annunziata, Firenze, Italia, Malattie Infettive Ospedale San Gerardo di Monza, Monza, Italia, Malattie Infettive Nuovo Polo Ospedaliero di Cona, Ferrara, Italia, Malattie Infettive Istituti Ospitalieri di Cremona, Cremona, Italia, Clinica di Malattie Infettive Policlinico A. Gemelli-Università Cattolica di Roma, Roma, Italia, Divisione Ospedaliera di Malattie Infettive Spedali Civili, Brescia, Italia and Fondazione Malattie Infettive e Salute Internazionale, Brescia, Italia


Journal of Acquired Immune Deficiency Syndromes | 2015

Risk of Liver Enzyme Elevation During Treatment With Ritonavir-Boosted Protease Inhibitors Among HIV-Monoinfected and HIV/HCV-Coinfected Patients

Giuseppe Lapadula; Silvia Costarelli; Liliane Chatenoud; Francesco Castelli; Noemi Astuti; Simona Di Giambenedetto; Eugenia Quiros-Roldan; Laura Sighinolfi; Nicoletta Ladisa; Massimo Di Pietro; Alessia Zoncada; Elisa Di Filippo; Andrea Gori; Paola Nasta; Carlo Torti

Background:The risk of liver enzyme elevation (LEE) after different ritonavir-boosted protease inhibitors (PI/r) has not been fully assessed in real-life settings and in populations with high rates of hepatitis C virus (HCV) coinfection. Methods:Patients introducing a new PI/r between 1998 and 2012 were included, if transaminases and HCV antibody (Ab) were assessed before treatment initiation. Time to grade 3 and 4 LEE were assessed using univariable and multivariable conditional Cox analyses, stratified by HCV serostatus. Results:A total of 6193 HIV-infected patients (3242 HCV-Ab negative and 2951 HCV-Ab positive) were included. Incidence of grade 3 LEE was 1.05, 7.66, and 8.08 per 100 patient-years of follow-up among HCV-Ab negative, HCV-Ab–positive and HCV-RNA–positive patients, respectively. Among HCV-Ab–negative patients, no differences were detected between different PI/r. Use of darunavir/ritonavir was not associated with LEE among HCV-coinfected patients. Atazanavir/ritonavir use was associated with grade 3 LEE but only among HCV-Ab–positive patients (versus LPV/r, hazard ratio: 1.39; 95% confidence interval: 1.1 to 1.75). This risk was not confirmed in a subanalysis restricted to HCV-RNA–positive patients (versus LPV/r, hazard ratio: 1.16; 95% confidence interval: 0.87 to 1.55). Other independent predictors of grade 3 LEE among HCV-Ab–positive patients were older age, male gender, being treatment naive, nonnucleoside reverse transcriptase inhibitor coadministration, increased aspartate aminotransferase at baseline, overweight, positive HCV-RNA, and advanced estimated liver fibrosis. Conclusions:Occurrence of hepatotoxicity was a rare finding among HCV-Ab–negative patients and was not influenced by the type of PI/r. In particular, the use of darunavir/ritonavir, previously linked with severe cases of hepatotoxicity, was not associated with a greater risk of LEE, irrespective from HCV serostatus.

Collaboration


Dive into the Massimo Di Pietro's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Simona Di Giambenedetto

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge