You are on page 1of 6

Downloaded from adc.bmj.com on May 27, 2012 - Published by group.bmj.

com

Review

Azithromycin, Ureaplasma and chronic lung disease of prematurity: a case study for neonatal drug development
Mark A Turner,1 Evelyne Jacqz-Aigrain,2 Sailesh Kotecha3
of Womens and Childrens Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK 2Pharmacologie Pdiatrique et Pharmacogntique, Hpital Robert Debr, Paris, France 3Department of Child Health, School of Medicine, Cardiff University, Cardiff, UK Correspondence to Sailesh Kotecha, Department of Child Health, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK; KotechaS@cardiff.ac.uk Accepted 23 May 2011 Published Online First 22 June 2011
1Department

ABSTRACT Chronic lung disease of prematurity (CLD) remains a major cause of morbidity and mortality in preterm infants. Ureaplasma has received intermittent attention over the last two decades as a possible contributory factor. In addition, pulmonary inammation is associated with the development of CLD. The macrolide azithromycin provides an attractive option to determine if it can decrease the development of CLD as it has both anti-inammatory and anti-infective properties. In this article, the authors review the evidence for the role of Ureaplasma in the development of CLD and the obstacles faced in the development of a drug before it reaches clinical practice. INTRODUCTION
Advances in neonatal care have improved the survival of extremely preterm infants, but morbidity1 including neurodevelopmental sequelae and chronic lung disease of prematurity (CLD), also known as bronchopulmonary dysplasia, remains signicant. CLD is associated with prolonged hospital stay and often a need for domiciliary oxygen, 2 and places a signicant burden on the family. Indeed, CLD was identied as having the largest therapeutic gap in a recent survey of UK neonatologists. 3 The Paediatric Regulation from the European Commission has stimulated work on systematic drug development for neonates. It is now feasible to obtain a marketing authorisation for off-patent medicines for use in children.4 5 The aims of this review are: 1. to outline the state-of-the-art for one potential intervention for CLD and 2. to use this intervention as a case study for the current approach to drug development for neonates in the context of developing a paediatric investigation plan with a view to applying for a marketing authorisation. Multiple factors appear to contribute to the development of CLD. In infants who are at risk of developing CLD, one frequent nding is colonisation of the preterm lung with the microbe Ureaplasma.6 7 Ureaplasma belongs to the class of Mollicutes which are thought to be the smallest free-living, self-replicating organisms. A s they do not have a cell wall, they are limited to a parasitic existence in eukaryotic cells. Ureaplasma urealyticum, the only species known to infect humans, was recently subdivided into two separate species, U urealyticum and U parvum, on the basis of their 16S ribosomal RNA gene sequences.8

Two meta-analyses have suggested an association between the presence of pulmonary Ureaplasma and the development of CLD.9 10 The more recent report by Schelonka et al which included 23 studies and 2216 babies,10 showed a relative risk of 2.83 (95% CI 2.29 to 3.51) for CLD diagnosed as oxygen dependency at 28 days of life. For the 751 infants who were oxygen-dependent at 36 weeks postconceptional age, the relative risk was 1.62 (1.13 to 2.33). The studies, however, are disparate with most calling for a de nitive randomised trial to assess if eradication of pulmonary Ureaplasma reduces the rates of CLD. In addition, lung in ammation during the rst week of life is the hallmark of preterm babies who subsequently develop CLD.11 12 Tafari and colleagues described the isolation of Ureaplasma from the lungs of stillborn infants with pneumonitis.13 Further support for the contributory role of Ureaplasma in human disease comes from demonstration of a specic IgM response in the neonate resulting in radiographic changes indicative of pneumonia in culture positive infants and demonstration of the organism in lung tissue by immunouorescence and electron microscopy.14 15 Animal models have contributed substantially to understanding of the in ammatory processes that are triggered by Ureaplasma in the mammalian respiratory tract, primarily in mouse, sheep and primate models.16 17 For example, Novy et al used a rhesus monkey model of antenatal Ureaplasma infection in the amniotic uid.17 They demonstrated histopathological ndings of chorioamnionitis, a systemic fetal in ammatory response and pneumonitis, which worsened as the duration of in utero infection increased. Initial clinical trials in preterm neonates used erythromycin, a macrolide antibiotic commonly used to treat Ureaplasma. Two randomised controlled trials were included in the Cochrane review by Mabanta et al which examined studies that treated Ureaplasma to decrease the rate of CLD.18 In the rst study, 75 ventilated infants of less than 30 weeks gestation whose Ureaplasma status was unknown when treatment commenced, were randomly assigned to receive erythromycin intravenously for 7 days or to no treatment.19 Nine infants (13%) were positive for Ureaplasma. Both groups had similar rates of CLD and erythromycin did not decrease CLD severity.19 In the second study, erythromycin was commenced once Ureaplasma status was known. Endotracheal aspirates and nasopharynx swabs from ventilated preterm infants (n=155; median gestational age
573

Drug therapy

Arch Dis Child 2012;97:573577. doi:10.1136/adc.2010.195180

Downloaded from adc.bmj.com on May 27, 2012 - Published by group.bmj.com

Review
(GA) 26 (range 2329) weeks) were cultured for Ureaplasma. 20 Colonised infants (29/155, 19%) were randomly assigned to treatment with intravenous or oral erythromycin 40 mg/kg/ day. Erythromycin treatment was effective in eradicating colonisation in 12/14 (86%) of the treated infants. Treatment with erythromycin reduced colonisation but did not signicantly alter the length of time with supplemental oxygen. Neither trial showed a statistically signicant effect of erythromycin on CLD, death or the combined outcome CLD or death. Since the two studies differed markedly in their design, the results were not combined in the meta-analyses. Neither study noted adverse effects for 710-day courses of erythromycin. While neither study supports a role for erythromycin in CLD, they provide evidence of tolerability. These studies are at best seen as early phase clinical trials rather than as underpowered tests of efcacy. Other approaches to this problem have been suggested. and reported an improvement in CLD rates in Ureaplasma colonised infants with wide condence intervals. Ballards work indicates that azithromycin may affect surrogate outcomes, provides proof-of-concept for the anti-in ammatory effects of azithromycin in preterm neonates and demonstrates the tolerability of azithromycin in the target population. 31 This extends the proof-of-concept to a group comprising Ureaplasma colonised and non-colonised neonates. Clearly, at least one sufciently powered randomised controlled trial is needed to address the role of azithromycin in preterm neonates and would have support from the neonatal community. 33 However, before a large randomised controlled trial can be conducted, a number of decisions need to be taken so that a large trial has the best chance of giving a clear answer.

WHICH FORMULATION SHOULD BE USED?


The development of an age-appropriate formulation is central to the paediatric investigation plans introduced under the EU regulation. The plans are guided by a reection paper: Formulations of Choice for the Paediatric Population. 34 To ensure an appropriate formulation, it is necessary to consider excipients, the optimal concentration of the formulation, the required characteristics of infusions pumps and procedures according to the concentration of the formulation and the local tolerance and toxicity of the presentation.

RATIONALE FOR USE OF AZITHROMYCIN IN NEONATES


Another potential drug, azithromycin, has anti-infective action against microbes including Ureaplasma in older age groups and also has potent anti-in ammatory actions utilised in many conditions including respiratory diseases such as cystic brosis. 2123 Azithromycin is actively concentrated within alveolar macrophages and thus is attractive to treat pulmonary conditions. 24 If Ureaplasma colonisation is on the causal pathway to the development of CLD, then azithromycin may reduce the incidence of CLD in combination with its anti-in ammatory effects. Proof-of-mechanism for azithromycin as a treatment for Ureaplasma in the newborn is suggested by work such as that of Sadowsky who injected U parvum into the amniotic uid of a rhesus monkey. 25 Azithromycin was administered to mothers at a dose of 12.5 mg/kg/12 hourly for 10 days. It eradicated the Ureaplasma from the amniotic uid with signicantly decreased pneumonitis. 25 However, macrolides in pregnant women have not shown an alteration in maternal outcomes. 26 27 Antenatal exposure to erythromycin close to birth improved some neonatal outcomes in the ORACLE trial. 28 29 Antenatal exposure to azithromycin in the second trimester was not associated with any differences in outcomes in the APPLE trial. 30 Postnatal azithromycin needs further evaluation in an adequately powered clinical trial. Since pulmonary inammation is thought to play an important role in the development of CLD and azithromycin is thought to modify inammation, Ballard et al performed a pilot double-blind, randomised, placebo controlled trial which specically excluded neonates colonised by Ureaplasma. 31 Infants (birth weight 1000 g) were randomised to treatment or placebo within 12 h of beginning intermittent mandatory ventilation and within 72 h of birth. The treatment group received azithromycin 10 mg/kg/day for 7 days followed by 5 mg/kg/day until the infant no longer required mechanical ventilation or supplemental oxygen, to a maximum of 6 weeks. Mortality, incidence of CLD and other morbidities were not signicantly different between groups. Postnatal steroid use was signicantly less in the treatment group (31%, 6/19) versus the placebo group (62%, 10/16) (p=0.05) as was the duration of mechanical ventilation (median 13 days (range 147 days) vs 35 days (range 1112 days)) (p=0.02). Importantly, the study did not report any signicant adverse effects of the use of azithromycin for up to 6 weeks. In a subsequent study, Ballard and colleagues randomised 211 neonates with birth weight <1250 g to azithromycin or placebo irrespective of colonisation status32
574

Drug therapy

WHAT IS THE ROLE OF ANIMAL WORK IN THE DEVELOPMENT PLAN?


Animal work, often labelled as preclinical work, needs to be considered for drug licensing in the EU. 35 Preclinical testing of drugs for paediatric indications is likely to focus on developmental toxicity and short- and long-term safety. This is different from studies designed to elucidate the mechanism and test proof-of-concept. 24 25 The tests performed often include physiological tests, psychomotor development, learning and memory, and a dose ranging pilot study. However, these may not be required for azithromycin as it is already on the market.

WHICH DOSAGE REGIMEN?


A wrong dose will not work! Before use in any trials, it is essential to establish an appropriate dose. Pharmacokinetics (PK; ie, how the body handles the drug) specically for neonates are essential. 36 The value of examining drug disposition in neonates is illustrated by a recent report of PK in neonates of metronidazole which found that differences in clearance according to GA indicated that different optimal dosage regimens are required at different GAs. 37 With respect to azithromycin, there are two steps to consider: delivery and efcacy. Delivery will depend on a similar timeconcentration curve as in older age groups. This is summarised by the area under the timeconcentration curve (AUC). For children aged 0.516 years, the AUC did not differ with age for a single dose of 10 mg/kg. 38 In contrast, the pilot PK data which are available in extremely preterm neonates, suggest that the AUC is markedly greater than in older age groups. This reects the less extensive clearance of azithromycin and also a smaller volume of distribution. 39 The net effect of these effects can only be determined empirically. In re ning the PK model before a large trial, a balance must be struck between safety and benet. Under the European regulation, the European Medicines Agency (EMEA) has set
Arch Dis Child 2012;97:573577. doi:10.1136/adc.2010.195180

Downloaded from adc.bmj.com on May 27, 2012 - Published by group.bmj.com

Review
up a Paediatric Committee to balance the scientic and patient welfare issues in paediatric drug development. On the other hand, death may be related to CLD, in which case a composite outcome would be a conservative approach. Secondary end points should include length of oxygen dependency, hospital stay and respiratory support with intubated or non-invasive mechanical ventilation (including continuous positive airway pressure), drug usage including postnatal corticosteroids and antibiotics, infection rates, intraventricular haemorrhage, necrotising enterocolitis and retinopathy of prematurity. For the diagnosis of CLD, it is important to use a standardised physiological end point, as recommended by the National Institutes of Health consensus47 or a physiological test.48 49 It is important to follow-up after discharge. CLD at 36 weeks is an important outcome that reects hospital costs and predicts subsequent events, so it is a reasonable outcome to use to assess efcacy. Outcomes to 2 years, for example, are important to assess safety and provide preliminary evidence of effectiveness.

SHOULD WE TREAT INFECTION OR INFLAMMATION?


Azithromycin has both anti-infective and anti-in ammatory effects. If a trial is chosen to effect both actions, then duration of treatment needs to be optimised to cover both. The data available to inform this decision are: 1. Pulmonary in ammation is present early in life and does not settle within 37 days of age.40 41 2. Most previous studies using macrolides to eradicate Ureaplasma have used 714 days of treatment. Ballards trials continued azithromycin therapy for up to 6 weeks. 31 32 3. Hassan et al found that a single dose of 10 mg/kg of azithromycin did not affect in ammatory markers. 39 4. Prolonged antibiotic exposure is likely to have adverse consequences. Many clinicians would nd it difcult to justify continuing antibiotics for the length of time used by Ballard et al because of the risk of selecting for resistant strains and of potential adverse effects. Caution is advised by the retrospective cohort study of 5693 neonates born at less than 1000 g42 which showed that increasing duration of empiric antibiotics, started within 3 days of birth among babies with negative blood cultures, was associated with increasing risk of necrotising enterocolitis and death.

WHAT SHOULD THE SAMPLE SIZE BE FOR LARGE CLINICAL TRIALS?


Issues to consider include rates of CLD/death. It may be advantageous to take account of the proportion of participants who have Ureaplasma colonisation but may not be available within the treatment window. The effect of colonisation may best be considered as a prespecied secondary analysis. Uncertainties in sample size often remain. As an estimate, between 650 and 700 preterm infants of 28 weeks or less gestation are likely to be required in an adequately powered randomised controlled trial, which clearly may need modication when colonisation by pulmonary Ureaplasma is factored into the calculations. Prior to protocol nalisation, sample size and power can be modelled in light of the PK study, a feasibility survey among participating centres and the available literature. A range of analyses is required. The fundamental interpretation of trials for superiority should be based on an intention-to-treat analysis. This evaluation will need to be supplemented by per protocol analyses and analyses of microbiologically evaluable participants.

WHAT INCLUSION CRITERIA SHOULD WE USE?


It is important to include preterm infants at high risk of developing CLD, particularly those who are receiving respiratory support at birth. It is difcult to recruit immediately after birth. Inclusion of infants at highest risk of developing CLD, that is, with a GA of 28 weeks or less, receiving respiratory support within 12 h of birth and recruited within 48 h of birth (with rst dose administered within 72 h of birth) would seem reasonable. Those infants who may develop respiratory disease due to underlying conditions such as congenital, surgical or cardiac anomalies may be reasonably excluded.

Drug therapy

WHICH MICROBIOLOGICAL ASSESSMENTS SHOULD BE DONE?


It would be crucial to account for pulmonary colonisation by Ureaplasma among the secondary, explanatory outcomes. A nasogastric tube is almost always inserted within the rst few hours of age in such babies and thus provides an ideal sample to reect the antenatal environment, with tracheal or nasopharyngeal aspirates for reection of the postnatal environment.43 44 This should permit secondary analysis of the effect of colonisation status on outcome and identify the prevalence of Ureaplasma antibiotic resistance.45 46 Follow-up sampling to test for treatment cure is essential but may be limited by sample availability. Standard practice for test-of-cure is to examine whether the antimicrobial has been effective three half-lives after treatment stops.

HOW SHOULD SAFETY BE ASSESSED?


Safety assessments are central to drug development. The choice of safety assessments and their timing should be based on risk assessment which includes the known safety issues with azithromycin and the characteristics of the target population (comorbidity is high among babies born at less than 29 weeks gestation and should not be ascribed to azithromycin unless there is clear evidence of this).

SHOULD WE AIM FOR A MARKETING AUTHORISATION?


A marketing authorisation provides assurance that pharmaceutical quality, safety and efcacy have been assessed by independent experts, and dosage and interactions considered. Conventional peer review is not sufcient to meet these needs. 50 Marketing authorisation holders have a range of duties to monitor the usage of each authorised medicine and maintain standards, including postmarketing surveillance. I n the absence of a marketing authorisation, a range of individual prescribers each has responsibility for off-label use, which would make it very difcult to develop and maintain a safety registry; pharmacists carry out a lot of work behind the scenes to support off-label medicines. A marketing authorisation provides an income stream that increases the justication for
575

WHICH END POINTS?


Any chosen end points should have clinical relevance, should be reproducible and include both respiratory and non-respiratory secondary end points. One option is to use a combined outcome (death or CLD), another option is to use a single outcome (CLD) and use survival analysis to adjust for death. Death may be unrelated to CLD, so that a combined outcome may underestimate the difference that is due to the trial medication.
Arch Dis Child 2012;97:573577. doi:10.1136/adc.2010.195180

Downloaded from adc.bmj.com on May 27, 2012 - Published by group.bmj.com

Review

Figure 1 Sample drug development plan. DSMB, data and safety monitoring board; PD, pharmacodynamics; PK, pharmacokinetics; SUSAR suspected unexpected serious adverse reaction.

Drug therapy

manufacturers to invest in a medicine and provides protection for that investment. If the market is large enough, a marketing authorisation will be the optimal way for neonatal drug development in the future.

curiosity driven but must be based on a careful line of thinking about formulation, optimised dosage regimen, clinical trials that explore the therapeutic possibility of the medicine and clinical trials that con rm the efcacy of the medicine.
Funding The authors are part of the EU funded FP7 programme TINN2. Competing interests None. Provenance and peer review Commissioned; externally peer reviewed.

DRUG DEVELOPMENT COSTS


The costs of developing a drug for use in children are signicant. The majority of the costs incurred in clinical drug development relate to management of clinical trials and to data collection in clinical trials. Estimates of the costs to address a written request leading to changes in the marketing authorisation relating to children by the American Food and Drugs Administration vary between US$5.3 million for a programme involving a single trial of efcacy in 151 children with refractory tumours and US$50 million for a programme involving two PK studies, seven efcacy studies and 926 children with depression. 51 Azithromycin to prevent CLD by treating Ureaplasma was placed on the EMEAs list of priority off-patent medicines and the authors have successfully secured funds from the European Commissions Framework Programme 7.

REFERENCES
1. 2. 3. Iacovidou N, Varsami M, Syggellou A. Neonatal outcome of preterm delivery. Ann N Y Acad Sci 2010;1205:130 4. Dunbar H, Kotecha S. Domiciliary oxygen for infants with chronic lung disease of prematurity. Care Crit Ill 2000;16:90 3. Turner MA, Lewis S, Hawcutt DB, et al. Prioritising neonatal medicines research: UK Medicines for Children Research Network scoping survey. BMC Pediatr 2009;9:50. Regulation on the Use of Medicinal Products for Paediatric Use, 2007. http:// ec.europa.eu/health/les/eudralex/vol-1/reg_2006_1901/reg_2006_1901_en.pdf (accessed 23 Dec 2010). Rocchi F, Paolucci P, Ceci A, et al. The European paediatric legislation: bene ts and perspectives. Ital J Pediatr 2010;36:56. Viscardi RM, Hasday JD. Role of Ureaplasma species in neonatal chronic lung disease: epidemiologic and experimental evidence. Pediatr Res 2009;65:84R 90R. Kotecha S, Hodge R, Schaber JA, et al. Pulmonary Ureaplasma urealyticum is associated with the development of acute lung inammation and chronic lung disease in preterm infants. Pediatr Res 2004;55:61 8. Robertson JA, Stemke GW, Davis JW, Jr, et al. Proposal of Ureaplasma parvum sp. nov. and emended description of Ureaplasma urealyticum (Shepard et al 1974) Robertson et al 2001. Int J Syst Evol Microbiol 2002;52:587 97. Wang EE, Ohlsson A, Kellner JD. Association of Ureaplasma urealyticum colonization with chronic lung disease of prematurity: results of a metaanalysis. J Pediatr 1995;127:640 4. Schelonka RL, Katz B, Waites KB, et al. Critical appraisal of the role of Ureaplasma in the development of bronchopulmonary dysplasia with metaanalytic techniques. Pediatr Infect Dis J 2005;24:1033 9. Kotecha S, Chan B, Azam N, et al. Increase in interleukin-8 and soluble intercellular adhesion molecule-1 in bronchoalveolar lavage uid from premature infants who develop chronic lung disease. Arch Dis Child Fetal Neonatal Ed 1995;72:F90 6.

4.

5. 6.

WHAT SHOULD THE DRUG DEVELOPMENT PLAN LOOK LIKE?


One approach is illustrated in gure 1. This represents a balance between timely research and lling in gaps in knowledge.

7.

8.

CONCLUSIONS
Azithromycin is an attractive medicine to evaluate for the prevention of CLD. Epidemiological work, research in animals and early phase clinical trials in humans support the need for further investigation of this possible use. To date, it has a favourable safety pro le and preliminary PK studies have been completed. Rational drug development will require an integrated consideration of pharmaceutical quality, dosage regimen, efcacy and safety. Medicines research can no longer be
576

9.

10.

11.

Arch Dis Child 2012;97:573577. doi:10.1136/adc.2010.195180

Downloaded from adc.bmj.com on May 27, 2012 - Published by group.bmj.com

Review
12. Kotecha S, Wilson L, Wangoo A, et al. Increase in interleukin (IL)-1 beta and IL-6 in bronchoalveolar lavage uid obtained from infants with chronic lung disease of prematurity. Pediatr Res 1996;40:250 6. Tafari N, Ross S, Naeye RL, et al. Mycoplasma T strains and perinatal death. Lancet 1976;1:108 9. Viscardi RM. Ureaplasma species: role in diseases of prematurity. Clin Perinatol 2010;37:393 409. Schelonka RL, Waites KB. Ureaplasma infection and neonatal lung disease. Semin Perinatol 2007;31:2 9. Normann E, Lacaze-Masmonteil T, Eaton F, et al. A novel mouse model of Ureaplasma-induced perinatal inammation: effects on lung and brain injury. Pediatr Res 2009;65:430 6. Novy MJ, Duffy L, Axthelm MK, et al. Ureaplasma parvum or Mycoplasma hominis as sole pathogens cause chorioamnionitis, preterm delivery, and fetal pneumonia in rhesus macaques. Reprod Sci 2009;16:56 70. Mabanta CG, Pryhuber GS, Weinberg GA, et al. Erythromycin for the prevention of chronic lung disease in intubated preterm infants at risk for, or colonized or infected with Ureaplasma urealyticum. Cochrane Database Syst Rev 2003;4:CD003744. Lyon AJ, McColm J, Middlemist L, et al. Randomised trial of erythromycin on the development of chronic lung disease in preterm infants. Arch Dis Child Fetal Neonatal Ed 1998;78:F10 14. Jnsson B, Rylander M, Faxelius G. Ureaplasma urealyticum, erythromycin and respiratory morbidity in high-risk preterm neonates. Acta Paediatr 1998;87:1079 84. Wilms EB, Touw DJ, Heijerman HG. Pharmacokinetics and sputum penetration of azithromycin during once weekly dosing in cystic brosis patients. J Cyst Fibros 2008;7:79 84. Van Bambeke F, Tulkens PM. Macrolides: pharmacokinetics and pharmacodynamics. Int J Antimicrob Agents 2001;18(Suppl 1):S1723. Blasi F, Cazzola M, Tarsia P, et al. Azithromycin and lower respiratory tract infections. Expert Opin Pharmacother 2005;6:2335 51. Hand WL, Hand DL. Characteristics and mechanisms of azithromycin accumulation and efux in human polymorphonuclear leukocytes. Int J Antimicrob Agents 2001;18:419 25. Sadowsky DW, Colgin LM, Duffy LB, et al. Azithromycin (AZI) and antiinammatory therapy for intra-amniotic infection (IAI) with Ureaplasma parvum in a non-human primate model. Reprod Sci 2007;14(Suppl 1):65A . Andrews WW, Hauth JC, Cliver SP, et al. Randomized clinical trial of extended spectrum antibiotic prophylaxis with coverage for Ureaplasma urealyticum to reduce post-cesarean delivery endometritis. Obstet Gynecol 2003;101:1183 9. Keith K, Ogasawara KK, Goodwin TM. Efcacy of azithromycin in reducing lower genital Ureaplasma urealyticum colonization in women at risk for preterm delivery. J Matern Fetal Med 1999;8:1216. Kenyon SL, Taylor DJ, Tarnow-Mordi W; ORACLE Collaborative Group. Broad-spectrum antibiotics for preterm, prelabour rupture of fetal membranes: the ORACLE I randomised trial. ORACLE Collaborative Group. Lancet 2001;357:979 88. Kenyon SL, Taylor DJ, Tarnow-Mordi W; ORACLE Collaborative Group. Broad-spectrum antibiotics for spontaneous preterm labour: the ORACLE II randomised trial. ORACLE Collaborative Group. Lancet 2001;357:989 94. van den Broek NR, White SA, Goodall M, et al. The APPLe study: a randomized, community-based, placebo-controlled trial of azithromycin for the prevention of preterm birth, with meta-analysis. PLoS Med 2009;6:e1000191. Ballard HO, Anstead MI, Shook LA. Azithromycin in the extremely low birth weight infant for the prevention of bronchopulmonary dysplasia: a pilot study. Respir Res 2007;8:41. 32. Ballard HO, Shook LA, Bernard P, et al. Use of azithromycin for the prevention of bronchopulmonary dysplasia in preterm infants: a randomized, double-blind, placebo controlled trial. Pediatr Pulmonol 2011;46:11118. Maxwell NC, Nuttall D, Kotecha S. Does Ureaplasma spp. cause chronic lung disease of prematurity: ask the audience? Early Hum Dev 2009;85:291 6. European Medicines Agency. Reection Paper: Formulations of Choice for the Paediatric Population, 2006. http://www.ema.europa.eu/docs/en_GB/document_ library/Scientic_guideline/2009/09/WC500003782.pdf (accessed 23 Dec 2010). European Medicines Agency. Guideline on the Need for Non-clinical Testing in Juvenile Animals of Pharmaceuticals for Paediatric Indications, 2008. http:// www.ema.europa.eu/docs/en_GB/document_library/Scientic_guideline/2009/09/ WC500003305.pdf (accessed 23 Dec 2010). Kearns GL, Abdel-Rahman SM, Alander SW, et al. Developmental pharmacologydrug disposition, action, and therapy in infants and children. N Engl J Med 2003;349:1157 67. Suyagh M, Collier PS, Millership JS, et al. Metronidazole population pharmacokinetics in preterm neonates using dried blood-spot sampling. Pediatrics 2011;127:e36774. Jacobs RF, Maples HD, Aranda JV, et al. Pharmacokinetics of intravenously administered azithromycin in pediatric patients. Pediatr Infect Dis J 2005;24:34 9. Hassan HE, Othman AA, Eddington ND, et al. Pharmacokinetics, safety, and biologic effects of azithromycin in extremely preterm infants at risk for ureaplasma colonization and bronchopulmonary dysplasia. J Clin Pharmacol 2010. Kotecha S. Cytokines in chronic lung disease of prematurity. Eur J Pediatr 1996;155(Suppl 2):S14 17. Watterberg KL, Demers LM, Scott SM, et al. Chorioamnionitis and early lung inammation in infants in whom bronchopulmonary dysplasia develops. Pediatrics 1997;99:144. Cotten CM, Taylor S, Stoll B, et al. Prolonged duration of initial empirical antibiotic treatment is associated with increased rates of necrotizing enterocolitis and death for extremely low birth weight infants. Pediatrics 2009;123:58 66. Oue S, Hiroi M, Ogawa S, et al. Association of gastric uid microbes at birth with severe bronchopulmonary dysplasia. Arch Dis Child Fetal Neonatal Ed 2009;94:F1722. Beeton ML, Maxwell NC, Davies PL, et al. Role of pulmonary infection in the development of chronic lung disease of prematurity. Eur Respir J 2011;37:1424 30. Beeton ML, Chalker VJ, Maxwell NC, et al. Concurrent titration and determination of antibiotic resistance in ureaplasma species with identication of novel point mutations in genes associated with resistance. Antimicrob Agents Chemother 2009;53:2020 7. Beeton ML, Chalker VJ, Kotecha S, et al. Comparison of full gyrA, gyrB, parC and parE gene sequences between all Ureaplasma parvum and U. urealyticum serovars to separate true uoroquinolone antibiotic resistance mutations from non-resistance polymorphism. J Antimicrob Chemother 2009;64:529 38. Ehrenkranz RA, Walsh MC, Vohr BR, et al. Validation of the National Institutes of Health consensus denition of bronchopulmonary dysplasia. Pediatrics 2005;116:1353 60. Walsh MC, Yao Q, Gettner P, et al. Impact of a physiologic denition on bronchopulmonary dysplasia rates. Pediatrics 2004;114:1305 11. Quine D, Wong CM, Boyle EM, et al. Non-invasive measurement of reduced ventilation:perfusion ratio and shunt in infants with bronchopulmonary dysplasia: a physiological denition of the disease. Arch Dis Child Fetal Neonatal Ed 2006;91:F409 14. Hamburg MA. Shattuck lecture. Innovation, regulation, and the FDA. N Engl J Med 2010;363:2228 32. Li JS, Eisenstein EL, Grabowski HG, et al. Economic return of clinical trials performed under the pediatric exclusivity program. JAMA 2007;297:480 8.

13. 14. 15. 16.

33. 34.

35.

17.

36.

18.

37.

38. 39.

19.

20.

40. 41.

21.

22. 23. 24.

42.

43.

44.

Drug therapy

25.

45.

26.

46.

27.

28.

47.

48. 49.

29.

30.

50. 51.

31.

Arch Dis Child 2012;97:573577. doi:10.1136/adc.2010.195180

577

Downloaded from adc.bmj.com on May 27, 2012 - Published by group.bmj.com

Azithromycin, Ureaplasma and chronic lung disease of prematurity: a case study for neonatal drug development
Mark A Turner, Evelyne Jacqz-Aigrain and Sailesh Kotecha Arch Dis Child 2012 97: 573-577 originally published online June 22, 2011

doi: 10.1136/adc.2010.195180

Updated information and services can be found at:


http://adc.bmj.com/content/97/6/573.full.html

These include:

References Email alerting service

This article cites 47 articles, 13 of which can be accessed free at:


http://adc.bmj.com/content/97/6/573.full.html#ref-list-1

Receive free email alerts when new articles cite this article. Sign up in the box at the top right corner of the online article.

Topic Collections

Articles on similar topics can be found in the following collections Drugs: infectious diseases (453 articles) Epidemiologic studies (881 articles) Child health (1924 articles) Immunology (including allergy) (1116 articles) Infant health (327 articles) Neonatal health (180 articles) Pneumonia (infectious disease) (107 articles) Pneumonia (respiratory medicine) (94 articles) TB and other respiratory infections (350 articles)

Notes

To request permissions go to:


http://group.bmj.com/group/rights-licensing/permissions

To order reprints go to:


http://journals.bmj.com/cgi/reprintform

To subscribe to BMJ go to:


http://group.bmj.com/subscribe/

You might also like