Network


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

Hotspot


Dive into the research topics where Yvonne Maldonado is active.

Publication


Featured researches published by Yvonne Maldonado.


Pediatrics | 2014

Updated Guidance for Palivizumab Prophylaxis Among Infants and Young Children at Increased Risk of Hospitalization for Respiratory Syncytial Virus Infection

Michael T. Brady; Carrie L. Byington; H. Dele Davies; Kathryn M. Edwards; Mary Anne Jackson; Yvonne Maldonado; Dennis L. Murray; Walter A. Orenstein; Mobeen H. Rathore; Mark H. Sawyer; Gordon E. Schutze; Rodney E. Willoughby; Theoklis E. Zaoutis; Henry H. Bernstein; David W. Kimberlin; Sarah S. Long; H. Cody Meissner; Marc A. Fischer; Bruce G. Gellin; Richard L. Gorman; Lucia H. Lee; R. Douglas Pratt; Jennifer S. Read; Joan Robinson; Marco Aurelio Palazzi Safadi; Jane F. Seward; Jeffrey R. Starke; Geoffrey R. Simon; Tina Q. Tan; Joseph A. Bocchini

Palivizumab was licensed in June 1998 by the Food and Drug Administration for the reduction of serious lower respiratory tract infection caused by respiratory syncytial virus (RSV) in children at increased risk of severe disease. Since that time, the American Academy of Pediatrics has updated its guidance for the use of palivizumab 4 times as additional data became available to provide a better understanding of infants and young children at greatest risk of hospitalization attributable to RSV infection. The updated recommendations in this policy statement reflect new information regarding the seasonality of RSV circulation, palivizumab pharmacokinetics, the changing incidence of bronchiolitis hospitalizations, the effect of gestational age and other risk factors on RSV hospitalization rates, the mortality of children hospitalized with RSV infection, the effect of prophylaxis on wheezing, and palivizumab-resistant RSV isolates. This policy statement updates and replaces the recommendations found in the 2012 Red Book.


The Journal of Infectious Diseases | 2001

Immune Responses to Measles and Mumps Vaccination of Infants at 6, 9, and 12 Months

Hayley A. Gans; Linda L. Yasukawa; Mary Rinki; Ross DeHovitz; Bagher Forghani; Judith Beeler; Susette Audet; Yvonne Maldonado; Ann M. Arvin

Immunizing infants against measles at the youngest age possible has the potential to reduce morbidity and mortality. The ability of infants at 6, 9, or 12 months to respond to measles and mumps vaccines was evaluated by measuring T cell proliferation, interferon-gamma production, and neutralizing antibody titers before and after vaccination. Infants in all age groups had equivalent cellular immune responses to measles or mumps viruses, with or without passive antibodies when immunized. In contrast, 6-month-old infants without passive antibodies had low geometric mean titers of antibody to measles or mumps viruses and low seroconversion rates. Geometric mean titers of antibody to measles virus increased if infants were revaccinated at 12 months. Six-month-old infants had limited humoral responses to paramyxovirus vaccines, whereas cellular immunity was equivalent to that of older infants. T cell responses can be established by immunization with these live attenuated virus vaccines during the first year, despite the presence of passive antibodies.


Journal of Clinical Investigation | 1995

Altered representation of naive and memory CD8 T cell subsets in HIV-infected children.

Ronald L. Rabin; Mario Roederer; Yvonne Maldonado; Ann Petru; Leonore A. Herzenberg

CD8 T cells are divided into naive and memory subsets according to both function and phenotype. In HIV-negative children, the naive subset is present at high frequencies, whereas memory cells are virtually absent. Previous studies have shown that the overall number of CD8 T cells does not decrease in HIV-infected children. In studies here, we use multiparameter flow cytometry to distinguish naive from memory CD8 T cells based on expression of CD11a, CD45RA, and CD62L. With this methodology, we show that within the CD8 T cell population, the naive subset decreases markedly (HIV+ vs. HIV-, 190 vs. 370 cells/microliter; P < or = 0.003), and that there is a reciprocal increase in memory cells, such that the total CD8 T cell counts remained unchanged (800 vs. 860 cells/microliter; P < or = 0.76). In addition, we show that for HIV-infected children, the naive CD8 T cell and total CD4 T cell counts correlate (chi 2 P < or = 0.001). This correlated loss suggests that the loss of naive CD8 T cells in HIV infection may contribute to the defects in cell-mediated immunity which become progressively worse as the HIV disease progresses and CD4 counts decrease.


Pediatrics | 2013

Clostridium difficile Infection in Infants and Children

Gordon E. Schutze; Rodney E. Willoughby; Michael T. Brady; Carrie L. Byington; H. Dele Davies; Kathryn M. Edwards; Mary P. Glode; Mary Anne Jackson; Harry L. Keyserling; Yvonne Maldonado; Dennis L. Murray; Walter A. Orenstein; Theoklis E. Zaoutis

Infections caused by Clostridium difficile in hospitalized children are increasing. The recent publication of clinical practice guidelines for C difficile infection in adults did not address issues that are specific to children. The purpose of this policy statement is to provide the pediatrician with updated information and recommendations about C difficile infections affecting pediatric patients.


Pediatrics | 2012

Recommendations for Prevention and Control of Influenza in Children, 2012–2013

Michael T. Brady; Carrie L. Byington; H. Dele Davies; Kathryn M. Edwards; Mary P. Glode; Mary Anne Jackson; Harry L. Keyserling; Yvonne Maldonado; Dennis L. Murray; Walter A. Orenstein; Gordon E. Schutze; Rodney E. Willoughby; Theoklis E. Zaoutis

The purpose of this statement is to update recommendations for routine use of trivalent seasonal influenza vaccine and antiviral medications for the prevention and treatment of influenza in children. The key points for the upcoming 2012–2013 season are: (1) this year’s trivalent influenza vaccine contains A/California/7/2009 (H1N1)–like antigen (derived from influenza A [H1N1] pdm09 [pH1N1] virus); A/Victoria/361/2011 (H3N2)–like antigen; and B/Wisconsin/1/2010–like antigen (the influenza A [H3N2] and B antigens differ from those contained in the 2010–2011 and 2011–2012 seasonal vaccines); (2) annual universal influenza immunization is indicated; and (3) an updated dosing algorithm for administration of influenza vaccine to children 6 months through 8 years of age has been created. Pediatricians, nurses, and all health care personnel should promote influenza vaccine use and infection control measures. In addition, pediatricians should promptly identify influenza infections to enable rapid treatment, when indicated, to reduce morbidity and mortality.


Vaccine | 2003

Measles and mumps vaccination as a model to investigate the developing immune system: passive and active immunity during the first year of life.

Hayley A. Gans; Ross DeHovitz; Bagher Forghani; Judith Beeler; Yvonne Maldonado; Ann M. Arvin

Evaluations of neutralizing antibody responses in 6-, 9- and 12-month-old infants given measles or mumps vaccine indicated that 6-month-old infants had diminished humoral immune responses associated with passive antibody effects, but also had an intrinsic deficiency in antiviral antibody production, which was independent of passive antibody effects. In contrast, lower neutralizing antibody titers in 9-month-olds were related only to passive antibody effects. Measles and mumps-specific T-cell proliferation and interferon-gamma (IFNgamma) production were induced by vaccination at 6, 9 or 12 months, regardless of passive neutralizing antibodies or age. These observations suggest a need to refine concepts about passive antibody interference and primary vaccine failure, taking into account the sensitization of antiviral T-cells, which occurs in the presence of passive antibodies and is observed in infants who do not develop active humoral immunity. A second dose of measles vaccine given at 12-15 months enhanced antiviral T-cell responses to measles in infants who were vaccinated at 6 or 9 months, and produced higher seroconversion rates. Since T-cell immunity is elicited under the cover of passive antibodies, the youngest infants benefit from the synergistic protection mediated by maternal antibodies and their own capacity to develop sensitized antiviral T-cells, which prime for subsequent exposures to the viral antigens. Conceptually, maternal immunization approaches with vaccines that can be given to women of child-bearing age before pregnancy, or that are safe for administration during pregnancy, should enhance passive antibody protection. Rather than being detrimental to infant adaptive immune responses, maternal vaccination can be coupled effectively with vaccine regimens that elicit priming of antiviral immune responses in infants during the first year of life.


The Journal of Infectious Diseases | 1998

Population-Based Prevalence of Symptomatic and Asymptomatic Astrovirus Infection in Rural Mayan Infants

Yvonne Maldonado; Michael F. Cantwell; Matthew O. Old; David Hill; Maria de la Luz Sanchez; Linda Logan; Francisco Millan-Velasco; José Luis Valdespino; Jaime Sepulveda; Suzanne M. Matsui

Symptomatic and asymptomatic astrovirus infection was prospectively determined in a 3-year birth cohort of Mayan infants. Stool samples from 271 infants and 268 older siblings were tested for astrovirus, adenovirus 40/41, rotavirus and Salmonella, Shigella and Campylobacter species. Concurrent diarrhea, vomiting, fever, or anorexia were noted. Astrovirus was detected in 164 infants (61%) and 20 siblings (7%). Rotavirus (4%) and adenovirus 40/41 (13%) were isolated less frequently. Of all diarrheal episodes reported at a visit, 26% (78/305) were associated with astrovirus; 17% (78/452) of astrovirus infections were associated with diarrhea and 9% with other symptoms. Only diarrhea was associated with astrovirus infection (odds ratio, 1.4; 95% confidence interval [CI], 1.07-1.92; P = .01). Of infants with astrovirus, 70% shed at multiple visits over a period of 2-17 weeks (median, 5). The point prevalence of astrovirus infection was significantly higher among infants than siblings (relative risk, 6.18; 95% CI, 3.93-9.72; P < .0001, chi2). Astrovirus was identified throughout the year, peaked in March and May, and decreased in September. In this population, astrovirus was the most common enteric pathogen isolated; symptomatic infection was prevalent among infants.


The Lancet | 2012

Efficacy and safety of an extended nevirapine regimen in infant children of breastfeeding mothers with HIV-1 infection for prevention of postnatal HIV-1 transmission (HPTN 046): a randomised, double-blind, placebo-controlled trial.

Hoosen M. Coovadia; Elizabeth R. Brown; Mary Glenn Fowler; Tsungai Chipato; Dhayendre Moodley; Karim Manji; Philippa Musoke; Lynda Stranix-Chibanda; Vani Chetty; Wafaie W. Fawzi; Clemensia Nakabiito; Lindiwe Msweli; Roderick R Kisenge; Laura A. Guay; Anthony Mwatha; Diana J. Lynn; Susan H. Eshleman; Paul G. Richardson; Kathleen George; Philip Andrew; Lynne M. Mofenson; Sheryl Zwerski; Yvonne Maldonado

BACKGROUND Nevirapine given once-daily for the first 6, 14, or 28 weeks of life to infants exposed to HIV-1 via breastfeeding reduces transmission through this route compared with single-dose nevirapine at birth or neonatally. We aimed to assess incremental safety and efficacy of extension of such prophylaxis to 6 months. METHODS In our phase 3, randomised, double-blind, placebo-controlled HPTN 046 trial, we assessed the incremental benefit of extension of once-daily infant nevirapine from age 6 weeks to 6 months. We enrolled breastfeeding infants born to mothers with HIV-1 in four African countries within 7 days of birth. Following receipt of nevirapine from birth to 6 weeks, infants without HIV infection were randomly allocated (by use of a computer-generated permuted block algorithm with random block sizes and stratified by site and maternal antiretroviral treatment status) to receive extended nevirapine prophylaxis or placebo until 6 months or until breastfeeding cessation, whichever came first. The primary efficacy endpoint was HIV-1 infection in infants at 6 months and safety endpoints were adverse reactions in both groups. We used Kaplan-Meier analyses to compare differences in the primary outcome between groups. This study is registered with ClinicalTrials.gov, number NCT00074412. FINDINGS Between June 19, 2008, and March 12, 2010, we randomly allocated 1527 infants (762 nevirapine and 765 placebo); five of whom had HIV-1 infection at randomisation and were excluded from the primary analyses. In Kaplan-Meier analysis, 1·1% (95% CI 0·3-1·8) of infants who received extended nevirapine developed HIV-1 between 6 weeks and 6 months compared with 2·4% (1·3-3·6) of controls (difference 1·3%, 95% CI 0-2·6), equating to a 54% reduction in transmission (p=0·049). However, mortality (1·2% for nevirapine vs 1·1% for placebo; p=0·81) and combined HIV infection and mortality rates (2·3%vs 3·2%; p=0·27) did not differ between groups at 6 months. 125 (16%) of 758 infants given extended nevirapine and 116 (15%) of 761 controls had serious adverse events, but frequency of adverse events, serious adverse events, and deaths did not differ significantly between treatment groups. INTERPRETATION Nevirapine prophylaxis can safely be used to provide protection from mother-to-child transmission of HIV-1 via breastfeeding for infants up to 6 months of age. FUNDING US National Institutes of Health.


Pediatrics | 2012

Strategies for Prevention of Health Care-Associated Infections in the NICU

Richard A. Polin; Susan E. Denson; Michael T. Brady; Lu Ann Papile; Jill E. Baley; Waldemar A. Carlo; James J. Cummings; Praveen Kumar; Rosemarie C. Tan; Kristi L. Watterberg; Carrie L. Byington; H. Dele Davies; Kathryn M. Edwards; Mary P. Glode; Mary Anne Jackson; Harry L. Keyserling; Yvonne Maldonado; Dennis L. Murray; Walter A. Orenstein; Gordon E. Schutze; Rodney E. Willoughby; Theoklis E. Zaoutis

Health care–associated infections in the NICU result in increased morbidity and mortality, prolonged lengths of stay, and increased medical costs. Neonates are at high risk of acquiring health care–associated infections because of impaired host-defense mechanisms, limited amounts of protective endogenous flora on skin and mucosal surfaces at time of birth, reduced barrier function of their skin, use of invasive procedures and devices, and frequent exposure to broad-spectrum antibiotic agents. This clinical report reviews management and prevention of health care–associated infections in newborn infants.


The Journal of Infectious Diseases | 2004

Humoral and Cell-Mediated Immune Responses to an Early 2-Dose Measles Vaccination Regimen in the United States

Hayley A. Gans; Junyuan Ren; Linda L. Yasukawa; Amanda Alderson; Mary Rinki; Ross DeHovitz; Judith Beeler; Susette Audet; Yvonne Maldonado; Ann M. Arvin

BACKGROUND Shifts in peak measles incidence to children <12 months old and the associated high mortality support the study of an early 2-dose measles vaccine regimen. METHODS Fifty-five infants were vaccinated with measles vaccine at age 6 (n=32) or 9 (n=23) months, followed by measles-mumps-rubella (MMR)-II vaccine at age 12 months. A control group received MMR-II only at age 12 months. Measles-specific humoral and cell-mediated immunity were evaluated before, 12 weeks after measles immunization, and 24 weeks after MMR-II. RESULTS Measles-specific T cell proliferation after both doses of vaccine was equivalent, regardless of age or the presence of passive antibodies. Seroconversion rates, geometric mean titers, and the percentage of infants with antibody titers >120 mIU after the first measles vaccine were lower in infants vaccinated at age 6 months, regardless of the presence of passive antibodies, but measles humoral responses increased after the administration of MMR-II vaccine in children initially vaccinated at age 6 or 9 months. CONCLUSION Measles vaccination elicits T cell responses in infants as young as 6 months old, which may prime the humoral response to the second dose. Initiating measles vaccination as an early 2-dose regimen results in an immunologic response that is likely to have clinical benefits in developed and developing countries.

Collaboration


Dive into the Yvonne Maldonado'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

Michael T. Brady

Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Gordon E. Schutze

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

H. Dele Davies

Michigan State University

View shared research outputs
Top Co-Authors

Avatar

Rodney E. Willoughby

Medical College of Wisconsin

View shared research outputs
Researchain Logo
Decentralizing Knowledge