Network


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

Hotspot


Dive into the research topics where Silvia Cattani is active.

Publication


Featured researches published by Silvia Cattani.


JAMA | 2009

Bovine Lactoferrin Supplementation for Prevention of Late-Onset Sepsis in Very Low-Birth-Weight Neonates: A Randomized Trial

Paolo Manzoni; Matteo Rinaldi; Silvia Cattani; Lorenza Pugni; Mario Giovanni Romeo; Hubert Messner; Ilaria Stolfi; Lidia Decembrino; Nicola Laforgia; Federica Vagnarelli; Luigi Memo; Linda Bordignon; Onofrio Sergio Saia; Milena Maule; Elena Gallo; Michael Mostert; Cristiana Magnani; Michele Quercia; Lina Bollani; Roberto Pedicino; Livia Renzullo; Pasqua Betta; Fabio Mosca; Fabrizio Ferrari; Rosario Magaldi; Mauro Stronati; Daniele Farina

CONTEXT Sepsis is a common and severe complication in premature neonates, particularly those with very low birth weight (VLBW) (<1500 g). Whether lactoferrin, a mammalian milk glycoprotein involved in innate immune host defenses, can reduce the incidence of sepsis is unknown. In animal models, the probiotic Lactobacillus rhamnosus GG (LGG) enhances the activity of lactoferrin but has not been studied in human infants. OBJECTIVE To establish whether bovine lactoferrin (BLF), alone or in combination with LGG, reduces the incidence of late-onset sepsis in VLBW neonates. DESIGN, SETTING, AND PATIENTS Prospective, multicenter, double-blind, placebo-controlled, randomized trial conducted in 11 Italian tertiary neonatal intensive care units. Patients were 472 VLBW infants enrolled from October 1, 2007, through July 31, 2008, and assessed until discharge for development of sepsis. INTERVENTION Infants were randomly assigned to receive orally administered BLF (100 mg/d) alone (n = 153), BLF plus LGG (6 x 10(9) colony-forming units/d) (n = 151), or placebo (n = 168) from birth until day 30 of life (day 45 for neonates <1000 g at birth). MAIN OUTCOME MEASURE First episode of late-onset sepsis, ie, sepsis occurring more than 72 hours after birth with isolation of any pathogen from blood or from peritoneal or cerebrospinal fluid. RESULTS Demographic, clinical, and management characteristics of the 3 groups were similar, including type of feeding and intake of maternal milk. Incidence of late-onset sepsis was significantly lower in the BLF and BLF plus LGG groups (9/153 [5.9%] and 7/151 [4.6%], respectively) than in the control group receiving placebo (29/168 [17.3%]) (risk ratio, 0.34; 95% confidence interval, 0.17-0.70; P = .002 for BLF vs control and risk ratio, 0.27; 95% confidence interval, 0.12-0.60; P < .001 for BLF plus LGG vs control). The decrease occurred for both bacterial and fungal sepsis. No adverse effects or intolerances to treatment occurred. CONCLUSION Compared with placebo, BLF supplementation alone or in combination with LGG reduced the incidence of a first episode of late-onset sepsis in VLBW neonates. TRIAL REGISTRATION isrctn.org Identifier: ISRCTN53107700.


Pediatrics | 2012

Bovine Lactoferrin Prevents Invasive Fungal Infections in Very Low Birth Weight Infants: A Randomized Controlled Trial

Paolo Manzoni; Ilaria Stolfi; Hubert Messner; Silvia Cattani; Nicola Laforgia; Mario G. Romeo; Lina Bollani; Matteo Rinaldi; Elena Gallo; Michele Quercia; Milena Maule; Michael Mostert; Lidia Decembrino; Rosario Magaldi; Fabio Mosca; Federica Vagnarelli; Luigi Memo; Pasqua Betta; Mauro Stronati; Daniele Farina

Background: Lactoferrin is a mammalian milk glycoprotein involved in innate immunity. Recent data show that bovine lactoferrin (bLF) prevents late-onset sepsis in preterm very low birth weight (VLBW) neonates. Methods: This is a secondary analysis of data from a multicenter randomized controlled trial where preterm VLBW neonates randomly received bLF (100 mg/day; group A1), bLF + Lactobacillus rhamnosus GG (106 colony-forming units per day; group A2), or placebo (group B) for 6 weeks. Here we analyze the incidence rates of fungal colonization, invasive fungal infection (IFI), and rate of progression from colonization to infection in all groups. Results: This study included 472 neonates whose clinical, nutritional, and demographical characteristics were similar. Overall, the incidence of fungal colonization was comparable (17.6%, 16.6%, and 18.5% in A1, A2, and B, respectively; P = .89 [A1] and .77 [A2]). In contrast, IFIs were significantly decreased in A1 and A2 (0.7% and 2.0%, respectively) compared with B (7.7%; P = .002 [A1] and .02 [A2]), and this was significantly true both in <1000 g (0.9% [A1] and 5.6% [A2], vs 15.0%) and in 1001 to 1500 g infants (0% and 0% vs 3.7%). The progression rate colonization-infection was significantly lower in the bLF groups: 3.7% (A1) and 12% (A2), vs 41.9%; P < .001 (A1) and P = .02 (A2). No IFI-attributable deaths occurred in the treatment groups, versus 2 in placebo. No adverse effects or intolerances occurred. Conclusions: Prophylactic oral administration of bLF reduces the incidence of IFI in preterm VLBW neonates. No effect is seen on colonization. The protective effect on IFI is likely due to limitation of ability of fungal colonies to progress toward invasion and systemic disease in colonized infants.


Early Human Development | 2014

Bovine lactoferrin supplementation for prevention of necrotizing enterocolitis in very-low-birth-weight neonates: a randomized clinical trial

Paolo Manzoni; Michael P. Meyer; Ilaria Stolfi; Matteo Rinaldi; Silvia Cattani; Lorenza Pugni; Mario Giovanni Romeo; Hubert Messner; Lidia Decembrino; Nicola Laforgia; Federica Vagnarelli; Luigi Memo; Linda Bordignon; Milena Maule; Elena Gallo; Michael Mostert; Michele Quercia; Lina Bollani; Roberto Pedicino; Livia Renzullo; Pasqua Betta; Fabrizio Ferrari; Tanith Alexander; Rosario Magaldi; Daniele Farina; Fabio Mosca; Mauro Stronati

IMPORTANCE NEC is a common and severe complication in premature neonates, particularly those with very-low-birth-weight (VLBW, <1500 g at birth). Probiotics including lactobacillus rhamnosus GG (LGG) proved effective in preventing NEC in preterm infants in several RCTs. OBJECTIVE Lactoferrin, a mammalian milk glycoprotein involved in innate immune host defences, can reduce the incidence of NEC in animal models, and its action is enhanced by LGG. We tried to assess whether bovine lactoferrin (BLF), alone or with the probiotic LGG, has a similar effect in human infants, something that has not yet been studied. DESIGN An international, multicenter, randomized, double-blind, placebo-controlled trial conducted from October 1st, 2007 through July 31st, 2010. SETTING Thirteen Italian and New Zealand tertiary neonatal intensive care units. PARTICIPANTS 743 VLBW neonates were assessed until discharge for development of NEC. INTERVENTION Infants were randomly assigned to receive orally either BLF (100 mg/day) alone (group LF; n = 247) or with LGG (at 6×10(9) CFU/day; group BLF + LGG; n = 238), or placebo (Control group; n = 258) from birth until day 30 of life (45 for neonates <1000 g at birth). MAIN OUTCOME MEASURES ≥ stage 2 NEC; death-and/or-≥ stage 2 NEC prior to discharge. RESULTS Demographics, clinical and management characteristics of the 3 groups were similar, including type of feeding and maternal milk intakes. NEC incidence was significantly lower in groups BLF and BLF + LGG [5/247 (2.0%)] and 0/238 (0%), respectively] than in controls [14/258 (5.4%)] (RR = 0.37; 95% CI: 0.136-1.005; p = 0.055 for BLF vs. control; RR = 0.00; p < 0.001 for BLF + LGG vs. control). The incidence of death-and/or-NEC was significantly lower in both treatment groups (4.0% and 3.8% in BLF and BLF + LGG vs. 10.1% in control; RR = 0.39; 95% CI: 0.19-0.80; p = 0.008. RR = 0.37; 95% CI: 0.18-0.77; p = 0.006, respectively). No adverse effects or intolerances to treatment occurred. CONCLUSIONS AND RELEVANCE Compared with placebo, BLF supplementation alone or in combination with LGG reduced the incidence of ≥ stage 2 NEC and of death-and/or ≥ stage 2 NEC in VLBW neonates. BLF might be a promising strategy to prevent NEC in NICU settings. Further data on larger sample sizes are warranted before BLF can be widespreadly used in clinical settings. TRIAL REGISTRATION ISRCTN53107700-http://www.controlled-_trials.com/ISRCTN53107700.


Early Human Development | 2013

Human milk feeding prevents retinopathy of prematurity (ROP) in preterm VLBW neonates

Paolo Manzoni; Ilaria Stolfi; Roberto Pedicino; Federica Vagnarelli; Fabio Mosca; Lorenza Pugni; Lina Bollani; Margherita Pozzi; Kelly Gomez; Chryssoula Tzialla; Alessandro Borghesi; Lidia Decembrino; Michael Mostert; Maria Agnese Latino; Claudio Priolo; Paolo Galletto; Elena Gallo; Stefano Rizzollo; Elena Tavella; Martina Luparia; Giuseppina Corona; Ignazio Barberi; Elisabetta Tridapalli; Giacomo Faldella; Gennaro Vetrano; Luigi Memo; Onofrio Sergio Saia; Linda Bordignon; Hubert Messner; Silvia Cattani

BACKGROUND Retinopathy of prematurity (ROP) is a multifactorial disease, but little is known about its relationships with neonatal nutritional policies. Human, maternal milk is the best possible nutritional option for all premature infants, including those at high risk for severe complications of prematurity, such as ROP. OBJECTIVE This is a secondary analysis of data collected during two multicenter RCTs performed consecutively (years 2004 through 2008) by a network of eleven tertiary NICUs in Italy. The two trials aimed at assessing effectiveness of fluconazole prophylaxis (Manzoni et al., N Engl J Med 2007 Jun 14;356(24):2483-95), and of bovine lactoferrin supplementation (Manzoni et al., JAMA 2009 Oct 7;302(13):1421-8), in prevention of invasive fungal infection, and of late-onset sepsis in VLBW infants, respectively. We tested the hypothesis that exclusive feeding with fresh maternal milk may prevent ROP of any stage - as defined by the ETROP study - in VLBW neonates, compared to formula feeding. METHODS We analyzed the database from both trials. Systematic screening for detection of ROP was part of the protocol of both studies. The definition of threshold ROP was as defined by the ETROP study. Univariate analysis was performed to look for significant associations between ROP and several possible associated factors, and among them, the type of milk feeding (maternal milk or formula for preterms). When an association was indicated by p < 0.05, multiple logistic regression was used to determine the factors significantly associated with ROP. RESULTS In both trials combined, 314 infants received exclusively human maternal milk (group A), and 184 a preterm formula because their mothers were not expected to breastfeed. The clinical, demographical and management characteristics of the neonates did not differ between the two groups, particularly related to the presence of the known risk factors for ROP. Overall, ROP incidence (any stage) was significantly lower in infants fed maternal milk (11 of 314; 3.5%) as compared to formula-fed neonates (29 of 184; 15.8%) (RR 0.14; 95% CI 0.12-0.62; p = 0.004). The same occurred for threshold ROP (1.3% vs. 12.3%, respectively; RR 0.19; 95% CI 0.05-0.69; p = 0.009). At multivariate logistic regression controlling for potentially confounding factors that were significantly associated to ROP (any stage) at univariate analysis (birth weight, gestational age, days on supplemental oxygen, systemic fungal infection, outborn, hyperglycaemia), type of milk feeding retained significance, human maternal milk being protective with p = 0.01. CONCLUSIONS Exclusive human, maternal milk feeding since birth may prevent ROP of any stage in VLBW infants in the NICU.


International Journal of Pediatrics | 2012

Oral Feeding Competences of Healthy Preterm Infants: A Review

Natascia Bertoncelli; G. Cuomo; Silvia Cattani; C. Mazzi; Marisa Pugliese; Elena Coccolini; P. Zagni; B Mordini; Fabrizio Ferrari

Background. With increasing sophistication and technology, survival rates hugely improved among preterm infants admitted to the neonatal intensive care unit. Nutrition and feeding remain a challenge and preterm infants are at high risk of encountering oral feeding difficulties. Objective. To determine what facts may impact on oral feeding readiness and competence and which kind of interventions should enhance oral feeding performance in preterm infants. Search Strategy. MEDILINE database was explored and articles relevant to this topic were collected starting from 2009 up to 2011. Main Results. Increasingly robust alertness prior to and during feeding does positively impact the infants feeding Skills. The review found that oral and non-oral sensorimotor interventions, provided singly or in combination, shortened the transition time to independent oral feeding in preterm infants and that preterm infants who received a combined oral and sensorimotor intervention demonstrated more advanced nutritive sucking, suck-swallow and swallow-respiration coordination than those who received an oral or sensorimotor intervention singly.


Early Human Development | 2010

Lactoferrin and prevention of late-onset sepsis in the pre-term neonates.

Paolo Manzoni; Lidia Decembrino; Ilaria Stolfi; Lorenza Pugni; Matteo Rinaldi; Silvia Cattani; Mario Giovanni Romeo; Hubert Messner; Nicola Laforgia; Federica Vagnarelli; Luigi Memo; Linda Bordignon; Onofrio Sergio Saia; Milena Maule; Elena Gallo; Michael Mostert; Cristiana Magnani; Michele Quercia; Lina Bollani; Roberto Pedicino; Livia Renzullo; Pasqua Betta; Fabrizio Ferrari; Rosario Magaldi; Fabio Mosca; Mauro Stronati; Daniele Farina

Late-onset sepsis (LOS) affects a large proportion of pre-term neonates in neonatal intensive care units (NICUs) worldwide, with high morbidity and related mortality, and frequent occurrence of severe late neurodevelopmental impairment. Due to the frequency, severity and difficulties in early diagnosis and prompt therapy, prevention is crucial for decreasing the burden of infection-related complications in NICUs. It is well known that feeding with fresh maternal milk, hygiene measures and the cautious use of H2-blockers are related with a decreased risk of developing sepsis. However, evidence from randomised clinical trials exists only for fluconazole in the prevention of fungal infections in the NICU. Lactoferrin is the main whey protein in mammalian milk, and is involved in innate immune host defences. Notably, human lactoferrin can be found at increased concentrations in colostrum and in milk from mothers of premature neonates. Human (hLF) and bovine lactoferrin (bLF) share a high (77%) amino-acid homology, and the same N-terminal peptide responsible for antimicrobial activity, called lactoferricin. In vitro, bLF shows potent direct antimicrobial activity against all types of pathogens, which occurs via anti-cell wall actions and leads to disintegration of the micro-organisms membranes. bLF is also synergistic with many antimicrobials and antifungals, and promotes growth and differentiation of the immature gut. Based on this background data, a randomised clinical trial was recently conducted in very low birth weight pre-term neonates given bLF alone or with the probiotic Lactobacillus GG. The aim of the trial was to assess the ability of bLF to prevent late-onset sepsis of any origin in the studied infants during their stay in the NICU. This article discusses the preliminary data from this study, along with the proposed mechanisms of action of bLF in pre-term infants.


Journal of Hospital Infection | 2005

A simultaneous outbreak of Serratia marcescens and Klebsiella pneumoniae in a neonatal intensive care unit

Chiara Casolari; Monica Pecorari; Giuliana Fabio; Silvia Cattani; Claudia Venturelli; Piccinini L; Maria Grazia Tamassia; William Gennari; Anna Maria Teresa Sabbatini; G. Leporati; Patrizia Marchegiano; Fabio Rumpianesi; Fabrizio Ferrari


Archive | 2012

Bovine Lactoferrin Supplementation for Prevention of Late-Onset Sepsis in Very Low-Birth-Weight Neonates

Paolo Manzoni; Matteo Rinaldi; Silvia Cattani; Lorenza Pugni; Mario Giovanni Romeo; Hubert Messner; Ilaria Stolfi; Lidia Decembrino; Nicola Laforgia; Federica Vagnarelli; Luigi Memo; Linda Bordignon; Onofrio Sergio Saia; Milena Maule; Elena Gallo; Michael Mostert; Cristiana Magnani; Michele Quercia; Lina Bollani; Roberto Pedicino; Livia Renzullo; Pasqua Betta; Fabio Mosca; Fabrizio Ferrari


New Microbiologica | 2013

Serratia marcescens in a neonatal intensive care unit: two long-term multiclone outbreaks in a 10-year observational study.

Chiara Casolari; Monica Pecorari; Elisa Della Casa; Silvia Cattani; Claudia Venturelli; Giuliana Fabio; Sara Tagliazucchi; Giulia Fregni Serpini; Mario Migaldi; Patrizia Marchegiano; Fabio Rumpianesi; Fabrizio Ferrari


Journal of Hospital Infection | 2005

A simultaneous outbreak of and in a neonatal intensive care unit

Chiara Casolari; Monica Pecorari; Giuliana Fabio; Silvia Cattani; Claudia Venturelli; Lino Piccinini; Maria Grazia Tamassia; William Gennari; Anna Maria Teresa Sabbatini; Guido Leporati

Collaboration


Dive into the Silvia Cattani's collaboration.

Top Co-Authors

Avatar

Fabrizio Ferrari

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fabio Mosca

Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico

View shared research outputs
Top Co-Authors

Avatar

Ilaria Stolfi

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Matteo Rinaldi

Marche Polytechnic University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge