Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
3.
Sci Transl Med ; 7(279): 279fs11, 2015 Mar 18.
Article in English | MEDLINE | ID: mdl-25787760

ABSTRACT

The Point-Person Project, a child-health research initiative, enables rapid response to opportunities for participation in multicenter pediatric clinical trials.


Subject(s)
Biomedical Research/trends , Pediatrics/methods , Chemistry, Pharmaceutical/methods , Child , Clinical Trials as Topic , Drug Design , Humans , National Institutes of Health (U.S.) , Research Design , United States
4.
Cell Mol Neurobiol ; 35(3): 377-387, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25392236

ABSTRACT

Multiple sclerosis is a chronic disease of the central nervous system characterized by an autoimmune inflammatory reaction that leads to axonal demyelination and tissue damage. Glucocorticoids, such as prednisolone, are effective in the treatment of multiple sclerosis in large part due to their ability to inhibit pro-inflammatory pathways (e.g., NFκB). However, despite their effectiveness, long-term treatment is limited by adverse side effects. VBP15 is a recently described compound synthesized based on the lazeroid steroidal backbone that shows activity in acute and chronic inflammatory conditions, yet displays a much-reduced side effect profile compared to traditional glucocorticoids. The purpose of this study was to determine the effectiveness of VBP15 in inhibiting inflammation and disease progression in experimental autoimmune encephalomyelitis (EAE), a widely used mouse model of multiple sclerosis. Our data show that VBP15 is effective at reducing both disease onset and severity. In parallel studies, we observed that VBP15 was able to inhibit the production of NFκB-regulated pro-inflammatory transcripts in human macrophages. Furthermore, treatment with prednisolone-but not VBP15-increased expression of genes associated with bone loss and muscle atrophy, suggesting lack of side effects of VBP15. These findings suggest that VBP15 may represent a potentially safer alternative to traditional glucocorticoids in the treatment of multiple sclerosis and other inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Pregnadienediols/therapeutic use , Severity of Illness Index , Animals , Anti-Inflammatory Agents/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/pathology , Pregnadienediols/pharmacology , Pregnancy , Treatment Outcome
5.
Sci Transl Med ; 6(227): 227fs11, 2014 Mar 12.
Article in English | MEDLINE | ID: mdl-24622511

ABSTRACT

High-quality, cost-effective pediatric clinical trials require a robust research and regulatory infrastructure and a properly trained workforce.


Subject(s)
Biomedical Research/standards , Clinical Trials as Topic , Health Services Needs and Demand , Pediatrics , Child , Child Welfare , Clinical Trials as Topic/economics , Clinical Trials as Topic/standards , Health Policy , Health Records, Personal , Humans , Research Support as Topic/economics
6.
Discov Med ; 16(89): 233-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24229740

ABSTRACT

Duchenne muscular dystrophy is a relatively common 'rare disorder,' with an incidence of about 1/5,000 males worldwide. The responsible gene and deficient protein (dystrophin) were identified in 1987, an early success of human molecular genetics and emerging genome projects. A rational approach to therapeutics is to replace dystrophin in patient muscle, thus addressing the primary biochemical defect. Fast forward 25 years, and two phase 2b/3 trials have been carried out with agents designed to induce de novo dystrophin production in DMD patient's muscle; ataluren (stop codon read through) with 174 patients, and drisapersen (exon skipping) with 186 patients. Both used a six minute walk test as the primary outcome measure. Neither drisapersen nor high dose ataluren showed any significant improvement in this outcome, whereas low dose ataluren is reported to show some possible improvement. Experience with ataluren and drisapersen has been disappointing and this is a good time to ask: What can we learn from these programs and how can this inform further drug development in DMD? At the times these two trials were started, there was a lack of existing data and infrastructure regarding both clinical and biochemical outcome measures. The recent publications of more extensive natural history data in multiple DMD cohorts, and ongoing efforts to define reliable and sensitive dystrophin assays are important. If the drisapersen and ataluren programs were instead begun today, new progress in biochemical and clinical endpoints may have triggered a re-design, with better de-risking in phase 2 studies prior to resource-intensive phase 3 trials.


Subject(s)
Dystrophin/metabolism , Muscular Dystrophies/drug therapy , Muscular Dystrophies/metabolism , Orphan Drug Production , Clinical Trials as Topic , Humans
7.
EMBO Mol Med ; 5(10): 1569-85, 2013 10.
Article in English | MEDLINE | ID: mdl-24014378

ABSTRACT

Absence of dystrophin makes skeletal muscle more susceptible to injury, resulting in breaches of the plasma membrane and chronic inflammation in Duchenne muscular dystrophy (DMD). Current management by glucocorticoids has unclear molecular benefits and harsh side effects. It is uncertain whether therapies that avoid hormonal stunting of growth and development, and/or immunosuppression, would be more or less beneficial. Here, we discover an oral drug with mechanisms that provide efficacy through anti-inflammatory signaling and membrane-stabilizing pathways, independent of hormonal or immunosuppressive effects. We find VBP15 protects and promotes efficient repair of skeletal muscle cells upon laser injury, in opposition to prednisolone. Potent inhibition of NF-κB is mediated through protein interactions of the glucocorticoid receptor, however VBP15 shows significantly reduced hormonal receptor transcriptional activity. The translation of these drug mechanisms into DMD model mice improves muscle strength, live-imaging and pathology through both preventive and post-onset intervention regimens. These data demonstrate successful improvement of dystrophy independent of hormonal, growth, or immunosuppressive effects, indicating VBP15 merits clinical investigation for DMD and would benefit other chronic inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Myoblasts/drug effects , Pregnadienediols/pharmacology , Animals , Anti-Inflammatory Agents/toxicity , Cell Line , Cell Membrane/drug effects , Cell Membrane/physiology , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/toxicity , Lasers , Mice , Mice, Inbred mdx , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Myoblasts/cytology , Myoblasts/radiation effects , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Necrosis/etiology , Phenotype , Prednisolone/pharmacology , Prednisolone/toxicity , Pregnadienediols/toxicity , Protein Interaction Maps/drug effects , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Signal Transduction/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Transcription, Genetic/drug effects
8.
Phys Med Rehabil Clin N Am ; 23(4): 821-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23137739

ABSTRACT

Although prednisone has never been formally approved for use in Duchenne muscular dystrophy (DMD) by regulatory agencies, its efficacy has been confirmed in trials dating from the 1980s. There is a strong need for optimization of both specific type of glucocorticoid (eg, prednisone, vs deflazacort or others) and the dosing regimen. Ideally an optimized regimen would maximize efficacy while minimizing side-effect profiles. A new trial, FOR-DMD, aims to address this gap in knowledge. In parallel, there has been progress in the area of "dissociative steroids," drugs that are able to better separate efficacy and side effects, providing a broader therapeutic window.


Subject(s)
Glucocorticoids/therapeutic use , Muscular Dystrophy, Duchenne/drug therapy , Drug Discovery , Glucocorticoids/chemistry , Glucocorticoids/pharmacology , Humans , Prednisone/therapeutic use , Pregnenediones/therapeutic use
10.
Am J Pathol ; 179(1): 12-22, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21703390

ABSTRACT

The identification of the Duchenne muscular dystrophy gene and protein in the late 1980s led to high hopes of rapid translation to molecular therapeutics. These hopes were fueled by early reports of delivering new functional genes to dystrophic muscle in mouse models using gene therapy and stem cell transplantation. However, significant barriers have thwarted translation of these approaches to true therapies, including insufficient therapeutic material (eg, cells and viral vectors), challenges in systemic delivery, and immunological hurdles. An alternative approach is to repair the patient's own gene. Two innovative small-molecule approaches have emerged as front-line molecular therapeutics: exon skipping and stop codon read through. Both approaches are in human clinical trials and aim to coax dystrophin protein production from otherwise inactive mutant genes. In the clinically severe dog model of Duchenne muscular dystrophy, the exon-skipping approach recently improved multiple functional outcomes. We discuss the status of these two methods aimed at inducing de novo dystrophin production from mutant genes and review implications for other disorders.


Subject(s)
Codon, Terminator/genetics , Dystrophin/metabolism , Exons/genetics , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/therapy , Mutant Proteins/metabolism , Animals , Dogs , Dystrophin/genetics , Humans , Mice , Muscle, Skeletal/cytology , Muscular Dystrophy, Duchenne/genetics , Mutant Proteins/genetics
11.
Pediatrics ; 125(1): e35-51, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20008423

ABSTRACT

OBJECTIVE: Palivizumab reduces respiratory syncytial virus (RSV) hospitalization in children at high risk by approximately 50% compared with placebo. We compared the efficacy and safety of motavizumab, an investigational monoclonal antibody with enhanced anti-RSV activity in preclinical studies, with palivizumab. METHODS: This randomized, double-blind, multinational, phase 3, noninferiority trial assessed safety and RSV hospitalization in 6635 preterm infants aged

Subject(s)
Antibodies, Monoclonal/therapeutic use , Antiviral Agents/therapeutic use , Hospitalization/statistics & numerical data , Infant, Premature , Respiratory Syncytial Virus Infections/prevention & control , Antibodies, Monoclonal, Humanized , Child, Preschool , Confidence Intervals , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Female , Follow-Up Studies , Humans , Infant , Infant, Newborn , Kaplan-Meier Estimate , Male , Palivizumab , Primary Prevention/methods , Probability , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/mortality , Risk Assessment , Survival Rate , Treatment Outcome
12.
Pediatr Infect Dis J ; 28(9): 835-7, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19636278

ABSTRACT

Previously healthy children hospitalized with respiratory syncytial virus (RSV) received motavizumab (3, 15, or 30 mg/kg intravenously), an RSV-specific monoclonal antibody, or placebo. Safety, tolerability, motavizumab concentrations, and immunogenicity were assessed. Cultivatable RSV in the upper respiratory tract was significantly reduced with motavizumab compared with placebo day 1 post-treatment. No adverse events were considered motavizumab-related by site investigators.


Subject(s)
Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacology , Antiviral Agents/adverse effects , Antiviral Agents/pharmacology , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus, Human/drug effects , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/adverse effects , Antibodies, Neutralizing/pharmacology , Antiviral Agents/administration & dosage , Female , Humans , Infant , Male , Placebos/administration & dosage , Respiratory Syncytial Virus, Human/immunology , Respiratory System/virology
13.
Pediatr Infect Dis J ; 28(4): 267-72, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19258920

ABSTRACT

BACKGROUND: : Respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infection in young children. Motavizumab is an investigational humanized monoclonal antibody for RSV prophylaxis. METHODS: : A dose-escalation study was conducted followed by assessment of safety, tolerability, serum concentrations, and immunogenicity during a second consecutive RSV season. In season 1, premature infants aged < or =6 months or children < or =24 months with chronic lung disease of prematurity received monthly motavizumab (3 or 15 mg/kg). In season 2, children who received > or =3 motavizumab doses in season 1 were randomized to receive monthly motavizumab or palivizumab 15 mg/kg. RESULTS: : Of 217 children enrolled in season 1, 211 (97.2%) received motavizumab 15 mg/kg and 205 (94.5%) patients completed the study through 90 days after the final dose. In season 2, 136 children were randomized to receive motavizumab (n = 66) or palivizumab (n = 70). The most commonly reported related adverse event was transient injection site erythema. In season 1, mean trough motavizumab concentrations were 7.9 and 50.2 microg/mL after the 3- and 15-mg/kg doses, respectively. Trough concentrations increased with repeated motavizumab dosing; a similar pattern was seen in season 2. Antimotavizumab reactivity occurred infrequently (3.3%) in season 1. In season 2, no treatment group-specific antidrug antibody was detected through 90 to 120 days after dosing with either product. CONCLUSIONS: : The pharmacokinetic profile of motavizumab was similar to that of other IgG1 antibodies. Increased adverse reactions or immunogenicity were not observed during and after a second season of treatment with motavizumab. Safety findings from these studies supported the continued development of motavizumab.


Subject(s)
Antibodies, Monoclonal , Antiviral Agents , Infant, Premature, Diseases/prevention & control , Respiratory Syncytial Virus Infections/prevention & control , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Antiviral Agents/immunology , Antiviral Agents/pharmacokinetics , Chronic Disease , Double-Blind Method , Humans , Infant , Infant, Newborn , Infant, Premature , Injections, Intramuscular , Lung Diseases , Male , Palivizumab , Risk Factors
14.
Pediatr Infect Dis J ; 27(8): 744-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18600188

ABSTRACT

BACKGROUND: Reliable availability of influenza vaccine before October could enable the vaccination of many children who might not otherwise be vaccinated. METHODS: Available data for children were analyzed to describe protection provided by live attenuated influenza vaccine (LAIV) for greater than 5 months postvaccination. RESULTS: Four studies conducted in children aged 6 months to 18 years were identified. Culture-confirmed efficacy against A/H1N1 and A/H3N2 strains at 9-12 months postvaccination was 77% [95% confidence interval (CI): 53-89%] to 100% (95% CI: 68-100%) and through a second influenza season without revaccination was 56% (95% CI: 31-73%) and 57% (95% CI: 6-82%), respectively. Against B strains, 1 study demonstrated 86% (95% CI: 59-95%) efficacy at 5-7 months. Another study demonstrated 27% (95% CI: -62% to 67%) efficacy at 9-12 months compared with 74% (95% CI: 39-89%) at 1 to <5 months during a period of antigenic drift for circulating B strains. A third study estimated 50% (95% CI: -49% to 83%) efficacy against influenza B strains through a second season without revaccination. CONCLUSIONS: In children, live attenuated influenza vaccine provided sustained protection against influenza illness caused by antigenically similar strains. Efficacy at 1 to <5 months postvaccination was comparable to that at 9-12 months for A/H1N1 and A/H3N2 strains and at 5-7 months for B strains. Meaningful efficacy was seen through a second season without revaccination, although at a lower level than during the first 12 months postvaccination.


Subject(s)
Influenza A virus/classification , Influenza A virus/drug effects , Influenza B virus/drug effects , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Adolescent , Child , Child, Preschool , Double-Blind Method , Humans , Infant , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/drug effects , Influenza, Human/immunology , Influenza, Human/virology , Randomized Controlled Trials as Topic , Time Factors , Treatment Outcome , Vaccination
15.
N Engl J Med ; 356(7): 685-96, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17301299

ABSTRACT

BACKGROUND: Universal vaccination of children 6 to 59 months of age with trivalent inactivated influenza vaccine has recently been recommended by U.S. advisory bodies. To evaluate alternative vaccine approaches, we compared the safety and efficacy of intranasally administered live attenuated influenza vaccine with those of inactivated vaccine in infants and young children. METHODS: Children 6 to 59 months of age, without a recent episode of wheezing illness or severe asthma, were randomly assigned in a 1:1 ratio to receive either cold-adapted trivalent live attenuated influenza vaccine (a refrigeration-stable formulation of live attenuated intranasally administered influenza vaccine) or trivalent inactivated vaccine in a double-blind manner. Influenza-like illness was monitored with cultures throughout the 2004-2005 influenza season. RESULTS: Safety data were available for 8352 children, and 7852 children completed the study according to the protocol. There were 54.9% fewer cases of cultured-confirmed influenza in the group that received live attenuated vaccine than in the group that received inactivated vaccine (153 vs. 338 cases, P<0.001). The superior efficacy of live attenuated vaccine, as compared with inactivated vaccine, was observed for both antigenically well-matched and drifted viruses. Among previously unvaccinated children, wheezing within 42 days after the administration of dose 1 was more common with live attenuated vaccine than with inactivated vaccine, primarily among children 6 to 11 months of age; in this age group, 12 more episodes of wheezing were noted within 42 days after receipt of dose 1 among recipients of live attenuated vaccine (3.8%) than among recipients of inactivated vaccine (2.1%, P=0.076). Rates of hospitalization for any cause during the 180 days after vaccination were higher among the recipients of live attenuated vaccine who were 6 to 11 months of age (6.1%) than among the recipients of inactivated vaccine in this age group (2.6%, P=0.002). CONCLUSIONS: Among young children, live attenuated vaccine had significantly better efficacy than inactivated vaccine. An evaluation of the risks and benefits indicates that live attenuated vaccine should be a highly effective, safe vaccine for children 12 to 59 months of age who do not have a history of asthma or wheezing. (ClinicalTrials.gov number, NCT00128167 [ClinicalTrials.gov].).


Subject(s)
Influenza Vaccines , Influenza, Human/prevention & control , Child, Preschool , Female , Follow-Up Studies , Hospitalization/statistics & numerical data , Humans , Infant , Influenza Vaccines/adverse effects , Influenza, Human/epidemiology , Kaplan-Meier Estimate , Male , Respiratory Sounds , Treatment Outcome , Vaccines, Attenuated/adverse effects
16.
Semin Pediatr Infect Dis ; 17(4): 206-12, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17055372

ABSTRACT

Live attenuated influenza vaccine (LAIV) offers a novel approach to influenza vaccination and is approved for healthy individuals 5 to 49 years of age. In placebo-controlled studies in children, LAIV was 73 to 93 percent efficacious, and protection lasted more than 12 months. In head-to-head studies in children, LAIV demonstrated a 35 to 53 percent reduction in influenza attack rates compared with injectable influenza vaccine (TIV) for matched strains. Compared with TIV, LAIV has demonstrated broader serum antibody responses, particularly against mismatched influenza A. The most common adverse events are runny nose and nasal congestion. Increased rates of asthma events were observed in young children. Additional large-scale safety and efficacy studies in young children, including a formal risk-benefit assessment, are ongoing. The results of these analyses will guide potential future use in young children.


Subject(s)
Immunization/methods , Influenza A virus/immunology , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Administration, Intranasal , Antibodies, Viral/blood , Child , Child, Preschool , Clinical Trials as Topic , Humans , Influenza Vaccines/adverse effects , Influenza, Human/immunology , Influenza, Human/virology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects
17.
Pediatr Infect Dis J ; 23(8): 707-12, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15295219

ABSTRACT

BACKGROUND: Respiratory syncytial virus (RSV) infection represents a major cause of pediatric respiratory hospitalizations. Limited treatment options exist. Palivizumab is a humanized monoclonal IgG1 antibody to the fusion protein of RSV that is highly active against RSV A and B strains. METHODS: A phase I/II, multicenter, randomized, double-blind, placebo-controlled, escalating dose clinical trial to describe the safety, tolerance, pharmacokinetics and clinical outcome of a single intravenous dose of palivizumab in previously healthy children hospitalized with acute RSV infection. RESULTS: Fifty-nine children < or =2 years of age received study drug. Sixteen children received 5 mg/kg of palivizumab (n = 8) or placebo (n = 8); 43 received 15 mg/kg of palivizumab (n = 22) or placebo (n = 21). Adverse events judged to be related to study drug were seen in one 5-mg/kg palivizumab patient and one 15-mg/kg palivizumab patient. These events were transient or consistent with progression of RSV disease. No discontinuations of study drug infusion because of adverse events occurred. Mean serum concentrations of palivizumab in the 5- and 15-mg/kg groups, respectively, were 61.2 and 303.4 microg/mL at 60 min and 11.2 and 38.4 microg/mL after 30 days. There were no significant differences in clinical outcomes between placebo and palivizumab groups for either dose. CONCLUSIONS: Intravenous palivizumab was safe and well-tolerated in children hospitalized with RSV disease. A single 15-mg/kg dose achieved serum palivizumab concentrations above the 25- to 30-microg/mL concentration associated with 2-log reduction of pulmonary RSV titer in the cotton rat model.


Subject(s)
Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Antiviral Agents/adverse effects , Antiviral Agents/pharmacokinetics , Respiratory Syncytial Virus Infections/drug therapy , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antiviral Agents/therapeutic use , Child , Child, Preschool , Double-Blind Method , Female , Humans , Infant , Infusions, Intravenous , Male , Palivizumab , Placebos , Treatment Outcome
18.
J Pediatr ; 143(4): 532-40, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14571236

ABSTRACT

OBJECTIVES: To evaluate the safety, tolerance, and efficacy of palivizumab in children with hemodynamically significant congenital heart disease (CHD). STUDY DESIGN: A randomized, double-blind, placebo-controlled trial included 1287 children with CHD randomly assigned 1:1 to receive 5 monthly intramuscular injections of 15 mg/kg palivizumab or placebo. Children were followed for 150 days. The primary efficacy end point was antigen-confirmed respiratory syncytial virus (RSV) hospitalization. RESULTS: Palivizumab recipients had a 45% relative reduction in RSV hospitalizations (P=.003), a 56% reduction in total days of RSV hospitalization per 100 children (P=.003), and a 73% reduction in total RSV hospital days with increased supplemental oxygen per 100 children (P=.014). Adverse events were similar in the treatment groups; no child had drug discontinued for a related adverse event. Serious adverse events occurred in 55.4% of palivizumab recipients and 63.1% of placebo recipients (P<.005); none were related to palivizumab. Twenty-one children (3.3%) in the palivizumab group and 27 (4.2%) in the placebo group died; no deaths were attributed to palivizumab. The rates of cardiac surgeries performed earlier than planned were similar in the treatment groups. CONCLUSIONS: Monthly palivizumab (15 mg/kg IM) was safe, well-tolerated, and effective for prophylaxis of serious RSV disease in young children with hemodynamically significant CHD.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antiviral Agents/therapeutic use , Heart Defects, Congenital/epidemiology , Hospitalization/statistics & numerical data , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus Infections/prevention & control , Antibodies, Monoclonal, Humanized , Comorbidity , Double-Blind Method , Heart Defects, Congenital/surgery , Humans , Palivizumab , Respiration, Artificial , Respiratory Syncytial Virus Infections/therapy , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...