AAC Accepts, published online ahead of print on 10 November 2014 Antimicrob. Agents Chemother. doi:10.1128/AAC.03951-14 Copyright © 2014, American Society for Microbiology. All Rights Reserved.

1

Pharmacokinetics, Microbial Response, and Pulmonary Outcomes of Multi-dose

2

Intravenous Azithromycin in Preterm Infants at Risk for Ureaplasma Respiratory

3

Colonization

4

Running Title: Azithromycin 20 mg/kg multi-dose in preterm infants

5

L. Marcela Merchan, MD1*; Hazem E. Hassan, PhD3*; Michael L. Terrin, MD2; Ken B.

6

Waites, MD4; David A. Kaufman, MD5; Namasivayam Ambalavanan, MD4; Pamela

7

Donohue, ScD6; Susan J. Dulkerian, MD1; Robert Schelonka, MD7, Laurence S.

8

Magder, PhD2, Sagar Shukla, PharmD3, Natalie D. Eddington, PhD3; and Rose M.

9

Viscardi, MD1#.

10 1

11

Departments of Pediatrics, and 2Epidemiology and Preventive Medicine, University of

12

Maryland, Baltimore School of Medicine, Baltimore, MD; 3University of Maryland,

13

Baltimore School of Pharmacy, Baltimore, MD; 4Departments of Pathology and

14

Pediatrics, University of Alabama at Birmingham School of Medicine, Birmingham, AL;

15

5

Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA; 6

16

Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore,

17

MD; and 7Department of Pediatrics, Oregon Health and Science University, Portland,

18

OR.

19 20

*

21

#

22

e-mail: [email protected]

23

L.L.M. and H.E.H. contributed equally to the work. Corresponding Author: Rose M. Viscardi, M.D

24

ABSTRACT

25

The study objective was to refine the population pharmacokinetics model, determine

26

microbial clearance, and assess short-term pulmonary outcomes of multiple dose

27

azithromycin treatment in preterm infants at risk for Ureaplasma respiratory colon-

28

ization. Fifteen subjects (Ureaplasma-positive=7) received intravenous azithromycin 20

29

mg/kg every 24h for 3 doses. Azithromycin concentrations were determined in plasma

30

samples obtained up to 168h post-first dose, by a validated LC/MS/MS method.

31

Respiratory samples were obtained pre-dose and at three time points post-last dose for

32

Ureaplasma culture, PCR, antibiotic susceptibility testing, and cytokine concentrations.

33

Pharmacokinetic data from these 15 subjects as well as 25 additional subjects [single

34

10 mg/kg dose (N=12), single 20 mg/kg dose (N=13)] were analyzed using a non-linear

35

mixed-effect population modeling (NONMEM) approach. Pulmonary outcomes were

36

assessed at 36 wk post-menstrual age and 6 months adjusted age. A 2-compartment

37

model with all PK parameters allometrically scaled on body weight best described the

38

azithromycin pharmacokinetics in preterm neonates. The population pharmacokinetic

39

parameters’ estimates for clearance, central volume of distribution, inter-compartmental

40

clearance, and peripheral volume of distribution were 0.15 L/h x WT(kg)0.75, 1.88 L x

41

WT(kg), 1.79 L/h x WT(kg)0.75 and 13 L x WT(kg), respectively. The estimated

42

AUC24/MIC90 was ~4 hr. All post-treatment cultures were negative and there were no

43

drug-related adverse events. One Ureaplasma-positive infant died at 4 months of age,

44

but no survivors were hospitalized for respiratory etiologies during the first 6 months

45

adjusted age. A 3 day course of 20 mg/kg/day intravenous azithromycin shows

46

preliminary efficacy in eradicating Ureaplasma spp. from the preterm respiratory tract.

2

47

INTRODUCTION

48

Bronchopulmonary dysplasia (BPD) is the major pulmonary morbidity in infants born

49

preterm characterized by arrested alveolar development and chronic inflammation.

50

Studies of human infants and experimental animal models indicate that the central

51

event in BPD pathogenesis is the interruption of normal developmental signaling during

52

early stages of lung development by lung injury that may be initiated in utero by

53

intrauterine infection with a subsequent dysregulated inflammatory response (1-3). A

54

recent meta-analysis of 39 studies confirmed that respiratory tract colonization with the

55

genital mycoplasma species Ureaplasma parvum and Ureaplasma urealyticum

56

increases the risk for development of BPD in extremely low gestation infants (4). It has

57

not been established whether eradicating Ureaplasma spp. from the respiratory tract of

58

preterm infants prevents or attenuates Ureaplasma infection-mediated lung injury.

59 60

Azithromycin, an azalide antibiotic, has anti-inflammatory properties and antimicrobial

61

activity against Ureaplasma spp. in in vitro (5, 6) and in in vivo experimental models (7-

62

9). Although the efficacies of azithromycin and a related macrolide, clarithromycin, to

63

prevent BPD have been assessed in single-center studies of preterm infants (10-13),

64

the optimal dosing regimens for these antibiotics have not been determined in

65

pharmacokinetic and pharmacodynamic studies and the impact on long term pulmonary

66

and neurologic outcomes are unknown. Our first steps to address these questions has

67

been to conduct studies in the at-risk population to determine the optimal dose, safety,

68

and in vivo anti-infective efficacy of azithromycin in preparation for future phase III

69

randomized, placebo-controlled trials (14, 15). We previously characterized the

3

70

pharmacokinetics (PK) of a single dose of intravenous azithromycin (10 mg/kg and 20-

71

mg/kg) in preterm infants (14). We demonstrated both doses were safe in 24 to 28

72

weeks gestation mechanically ventilated infants, but the 20 mg/kg dose was more

73

effective in eradicating Ureaplasma from the respiratory tract (14,15). Neither dose

74

reduced the pulmonary inflammatory response. Simulation analysis using our previously

75

developed PK model suggested that a multiple-dose regimen of 20 mg/kg intravenous

76

azithromycin may be efficacious for microbial clearance as it could mostly maintain

77

azithromycin plasma levels above the MIC50 for at least 120 hr post I.V. infusion.

78 79

The current study of preterm infants treated with 3 doses of 20 mg/kg intravenous

80

azithromycin was designed to 1) determine the microbiological and short term

81

pulmonary outcomes of the multiple dose; 2) assess the safety of this regimen, and 3)

82

collect additional PK data to further refine the population PK model. We hypothesized

83

that intravenous azithromycin therapy will prevent BPD in Ureaplasma-colonized

84

preterm infants by accelerating pathogen clearance or down-regulating the pulmonary

85

inflammatory response.

86 87

MATERIALS AND METHODS

88

Subject Enrollment

89

We conducted a Phase IIa non-randomized, open-label, PK and safety study of

90

intravenous azithromycin 20 mg/kg every 24h x 3d in preterm infants who were at high

91

risk for Ureaplasma respiratory tract colonization and the development of BPD (FDA

92

IND78990). Study subjects were recruited from six clinical sites from December 2011 to

4

93

June 2012. The institutional review board of each institution approved the study, and

94

parental informed consent was obtained. Inclusion criteria were: 1) gestational age 240-

95

286 weeks; 2) appropriate size for gestational age; 3) 1.5 mg/dL);

103

8) corrected QT interval (QTc) ≥450 ms; 9) neonatal exposure to any other systemic

104

macrolide antibiotic; 10) clinically suspected Ureaplasma CNS infection or other

105

confirmed bacterial or viral infection; and 11) participation in other clinical trials involving

106

investigational products.

107 108

Drug administration and blood sampling

109

After baseline laboratory tests and respiratory specimens were obtained, study infants

110

received azithromycin 20 mg/kg administered at a concentration of 2 mg/ml by

111

intravenous infusion over 60 min within 24 h of enrollment. A total of 3 doses were

112

administered at 24 ± 0.5h intervals. Six blood samples (0.25 ml each) were obtained

113

from each subject during specified time periods post- first dose infusion and processed

114

as previously described (15). Optimum sampling periods were predicted using the PK

115

software ADAPT5 (Biomedical Simulations Resource (BMSR), U. Southern California,

5

116

Los Angeles, CA). Samples were collected anywhere between 1-2, 2-4, 6-8, 25-48, 49-

117

96 and 120-168 hr post first dose. Specifying windows for withdrawing plasma samples

118

helps to better characterize the disposition profile and is a more clinically practical

119

approach (14-17). Levels of azithromycin in plasma were measured using a validated

120

high-performance liquid chromatography-tandem mass spectroscopy (HPLC/MS/MS)

121

detection method as previously described (14, 15).

122 123

Pharmacokinetic data analysis

124

The plasma concentration vs. time data from the a) single 10 mg/kg (14), b) single 20

125

mg/kg (15) and c) multiple 20 mg/kg studies were compiled and analyzed

126

simultaneously using the nonlinear mixed-effects modeling software, NONMEM 7.2

127

(ICON, MD). The first-order conditional estimation procedure (FOCE) with interaction

128

was used for the analysis. Upon development of the base model (two compartment with

129

proportional residual error model), covariate-parameter relationships were explored.

130

The available covariates were weight (WT), gestational age (GA), sex, height, and body

131

surface area.

132 133

The likelihood ratio test was applied to discriminate the alternative nested models where

134

a drop of the objective function value by at least 3.84 points was necessary to declare

135

model improvement, data significance level of P = 0.05, and one degree of freedom.

136

Inter-occasion variability was also estimated where study one (single 10 mg/kg

137

azithromycin), study two (single 20 mg/kg azithromycin) and study three (multiple 20

138

mg/kg azithromycin) were considered as first-, second- and third-occasion, respectively. 6

139

The final model that best fit the data was a two-compartment structural model (ADVAN3

140

TRANS4 NONMEM subroutine) with all parameters allometrically scaled on body weight

141

with a fixed exponent of 0.75 for both Cl and Q and a fixed exponent of 1 for both V1

142

and V2. Post-hoc Bayesian estimates of the individual subjects’ PK parameters were

143

calculated.

144

A proportional error model was used to describe the residual error as follows:

145

Yij obs = Yij pred*(1 + εij)

146

where Yij obs is the observed azithromycin plasma concentration j in subject I, Yij pred is

147

the model-predicted azithromycin plasma concentration, and ε is a residual random

148

error for individual i and measurement j and is assumed to be normally distributed

149

ε~N(0,σ2).

150

An exponential variance model was used to describe the variability of PK parameters

151

across individuals as follows:

152

Pi = θκ exp (ηκi)

153

where Pi is the estimated parameter value for subject i, θκ is the typical population value

154

of parameter k, ηki is subject specific deviation from population mean for individual i and

155

parameter k and assumed to follow normal distribution η~N(0, ω2).

156 157

Ureaplasma culture, antibiotic susceptibility testing, and real-time PCR

158

Upon enrollment, 2 tracheal aspirate samples at least 2h apart and one nasopharyngeal

159

sample from intubated infants, or 2 nasopharyngeal samples at least 2h apart from non-

160

intubated infants were obtained pre-dose for Ureaplasma culture and PCR. Subsequent

161

samples were obtained at 2 and 4 or 5 days post last dose and 21 days postnatal age.

7

162

Each specimen was directly inoculated in 10B broth and frozen at – 80 o C for later

163

shipment to University of Alabama at Birmingham Diagnostic Mycoplasma laboratory for

164

10B broth and A8 agar quantitative culture, azithromycin susceptibility testing by the

165

10B broth microdilution method (18-20), and real-time PCR for detection and species

166

determination using species-specific primers(14, 15, 21). A culture was considered

167

negative if no growth was detected after 7 days incubation. A confirmed positive culture

168

was defined as a positive broth culture from either tracheal aspirate or nasopharyngeal

169

specimen confirmed by typical morphology on agar plates. All isolates were confirmed

170

and speciated by PCR. Since Ureaplasma is only transmitted vertically, neonates who

171

were positive by confirmed culture or PCR at any time point were considered a

172

colonized neonate. Ureaplasma eradication was defined as 3 negative cultures post-last

173

dose.

174 175

Tracheal aspirate cytokine analysis

176

Tracheal aspirates for cytokines were obtained at the same time points as the culture

177

specimens pre- and post-treatment and were separated by centrifugation into cell pellet

178

and supernatant fractions as previously described (14). The cytokines interleukin (IL)-

179

1ß, IL-6, IL-8, and IL-17 were measured in the supernatants using LuminexTM multi-

180

analyte immunoassay with reagents from Upstate Biotechnology. Cell pellet lysates

181

were assayed for myeloperoxidase (MPO) activity (indicator of neutrophil activation) as

182

previously described (22, 23).

183 184

Whole blood cytokines

8

185

Whole blood spots were collected on filter paper (50 µl) pre-dose, 4-5 d post-dose and

186

21d of age and immediately frozen at -80°C. The stored blood spots were eluted and

187

analyzed by the multiplex LuminexTM immunoassay for the same cytokines as the

188

tracheal aspirates, as previously described (24-26).

189 190

Clinical outcomes

191

All infants were monitored for safety including vital signs, frequency of apnea and

192

bradycardia, cardiac rhythm, pulse oximetry up to 8 h post dose, clinical laboratory

193

testing pre-dose and up to study day 28, and adverse events until hospital discharge.

194

Infants were assessed for morbidities associated with prematurity, concomitant

195

medications, hearing screen results, and adverse events until discharge or transfer.

196 197

For the BPD endpoint, infants who were receiving supplemental oxygen or positive

198

pressure support at 36 weeks postmenstrual age (PMA) were considered to have BPD.

199

For infants receiving supplemental oxygen by nasal cannula, the delivered fraction of

200

inspired oxygen concentration or “effective FiO2” (27) was calculated by the technique

201

described by Benaron et al. (28) that was used in the STOP-ROP trial (29) which is

202

based on weight, oxygen liter flow, and oxygen concentration. Study subjects who were

203

transferred or discharged at ≤35 weeks PMA on supplemental oxygen were considered

204

to have BPD.

205 206

Pulmonary Outcomes at Six Months Adjusted Age

9

207

Data were collected by structured parental interviews before discharge and follow-up

208

phone interview at 6 months adjusted age utilizing the validated Tucson Children’s

209

Respiratory Study questionnaires that were designed to elicit a complete history of

210

possible covariates such as family history of asthma or atopy, and important

211

environmental (e.g., smoking) and infectious exposures and detailed interval respiratory

212

health history (30, 31). Recurrent wheezing and chronic cough were defined as

213

episodes occurring more than twice per week. The primary clinical outcome was

214

occurrence of hospitalization for respiratory illnesses and secondary outcomes included

215

the need for home supplemental oxygen, doctor and emergency room visits, parental

216

report of chronic cough and wheezing, and use of respiratory medications.

217 218

RESULTS

219

Subject Characteristics

220

As shown in Figure 1, seventy-one of 192 infants (63%) with a gestational age of 240 to

221

286 weeks gestation who were less than 72h age upon NICU admission were eligible for

222

the study. Parents of 43 infants were approached for consent, and 15 consented to the

223

study. Maternal macrolide exposure [N=36 (30%)] was the most common reason for

224

non-eligibility. All enrolled subjects received the 3 doses of intravenous 20 mg/kg

225

azithromycin and survived to BPD assessment at 36 wk PMA.

226 227

Seven subjects (47%) were culture and PCR positive for Ureaplasma spp. pre-dose. As

228

previously observed, U. parvum [N=5, (71%)] was more commonly isolated than U.

229

urealyticum [N=2, (29%)]. In 6 subjects with paired tracheal aspirate and

10

230

nasopharyngeal samples, 4 were culture-positive in both specimens and 2 were positive

231

in nasopharyngeal, but not tracheal aspirate specimens. In one Ureaplasma-positive

232

subject who was not intubated at the time of study entry, the NP sample was culture

233

and PCR positive. There were no positive cultures with 20 mg/kg dosing at any follow-

234

up time point, but one subject was PCR positive 4-5d post-last dose. The baseline

235

characteristics and clinical outcomes of the study sample stratified by Ureaplasma

236

status pre-treatment are presented in Tables 1 and 2 respectively. The Ureaplasma-

237

positive and negative subjects were similar for birthweight, gestational age, and

238

respiratory support at study entry.

239 240

Four of 15 (26%) infants developed BPD at 36 weeks PMA (Table 2). Three of seven

241

(43%) Ureaplasma-positive subjects developed BPD compared to one of eight (12.5%)

242

Ureaplasma-negative infants (p NS by Fisher Exact test). There was no difference by

243

Ureaplasma culture status in duration of mechanical ventilation support or supplemental

244

oxygen. One Ureaplasma-positive and one Ureaplasma-negative infant were

245

discharged home on supplemental oxygen. Fourteen subjects survived to discharge and

246

one Ureaplasma-positive subject died at four months of age due to persistent

247

pulmonary hypertension as a complication of BPD.

248 249 250 251

Pharmacokinetic analysis Plasma samples (N=239) from 40 subjects (including the 2 single dose studies and

252

current multiple dose study) were included in the PK analysis to further refine our

253

previously reported population model. The disposition of azithromycin was biphasic

254

(Figure 3A) suggesting that the pharmacokinetics of azithromycin could follow a two

11

255

compartment model and since the magnitude of many body processes [e.g., Clearance

256

(CL) or Volume of distribution (V)] may change in a regular fashion as the magnitude of

257

a certain covariate changes (e.g., body weight), we allometrically scaled PK parameters

258

on body weight during model development. Indeed, the inclusion of body weight as a

259

covariate improved the model fit to the data and explained some of the inter-subject

260

variability as was the case in our previous reports (14, 15) and the final model was a 2-

261

compartment model with all PK parameters allometrically scaled on body weight (Table

262

3). Inclusion of other covariates and accounting for inter-occasion variability did not

263

further reduce the inter-subject variability for any of the parameters and resulted in no

264

significant drop in the objective function value, therefore they were not included in the

265

final model. Goodness-of-fit plots for the final population PK model indicated that the

266

model described the data well without systematic bias (Figure 2). Based on the final

267

model parameters, the elimination half-life (t1/2) was estimated to be 69 hours in a

268

typical neonate weighing 1 kg.

269 270

Azithromycin MICs were determined using Clinical and Laboratory Standards Institute

271

(CLSI) guidelines (18) for 65 non-duplicate ureaplasma isolates (22 isolates from the

272

three azithromycin study cohorts and 43 isolates from banked specimens from preterm

273

infants (20)). The MIC50 and MIC90 were 2 and 8 µg/mL, respectively. The MIC range for

274

isolates from the current cohort was 1-8 µg/mL. No azithromycin resistance (MIC ≥16

275

µg/mL) as designated by the CLSI (18) was detected among the isolates from study

276

participants or banked specimens. Following three doses of 20 mg/kg intravenous

277

administration, azithromycin observed plasma concentrations were mostly maintained

12

278

between MIC50 (2 µg/ml) and MIC90 (8 µg/ml) for ~120 hr post first dose (Figure 3A),

279

consistent with our previously reported simulation analysis (15). The estimated

280

AUC24/MIC90 value was 4 hr. Simulation analysis indicated that the model generally

281

captured the trend in azithromycin disposition in preterm neonates, but it under-

282

predicted some concentrations (Figure 3B).

283 284

Azithromycin Effects on Indices of Inflammation

285

We analyzed 4 cytokines previously associated with Ureaplasma respiratory tract

286

colonization and BPD (IL-1ß, IL-6, and IL-8) (32, 33) and BPD (IL-17A)(26). In the 7

287

subjects (4 Ureaplasma-negative and 3 Ureaplasma-positive) with paired pre- and post-

288

dose tracheal aspirates, there was a decrease in IL-1ß, IL-8, IL-6, and IL-17A two days

289

post-last dose, but no difference between baseline and post-4 days concentrations

290

except for IL-17A (Tables 4 and 5). These differences were only significant for IL-17A.

291

There were no significant differences for cytokines measured in blood samples (data not

292

shown). Myeloperoxidase activity in tracheal aspirate cell pellets was also not

293

influenced by azithromycin treatment.

294 295

Safety and Serious Adverse Events

296

All subjects were monitored for adverse events until discharge from the NICU. There

297

were no abnormal vital signs or arrhythmias noted during or within 4 hours of drug

298

administration nor episodes of feeding intolerance. There were no abnormal laboratory

299

values attributable to study drug. A 25 week gestation Ureaplasma-negative infant

300

experienced a spontaneous intestinal perforation three days after the third study drug

13

301

dose and subsequently developed necrotizing enterocolitis Bell Stage 2. There were

302

three subjects with intraventricular hemorrhage Grade ≥3, one of whom also had

303

periventricular leukomalacia noted on a head ultrasound obtained on day 3 of life and

304

subsequently developed post-hemorrhagic hydrocephalus requiring ventriculoperitoneal

305

shunt placement.

306 307

Six Months Adjusted Age Pulmonary Outcomes. None of the 14 survivors were

308

hospitalized for respiratory illnesses between NICU discharge and six months adjusted

309

age (Table 6). One Ureaplasma negative subject remained on supplemental oxygen at

310

6 months adjusted age. There were no differences in reports of doctor/ER visits for

311

cough or wheezing, or respiratory medication use.

312 313

DISCUSSION

314

The PK properties of azithromycin support short courses of therapy. The drug has a

315

relatively long elimination half-life, is concentrated in phagocytic cells with good tissue

316

penetration, especially in the lung, and persists in tissues for much longer than in

317

plasma (34). Therefore, in this study we tested a 3 day course of 20 mg/kg intravenous

318

dose of azithromycin in preterm neonates to evaluate its effectiveness in eradicating

319

Ureaplasma, to determine its safety in this population, and to further refine the

320

population PK model of azithromycin in extremely preterm infants at risk for Ureaplasma

321

respiratory tract colonization and BPD. This dosage regimen showed preliminary

322

efficacy to eradicate ureaplasma and safety, and the model PK parameters were

323

estimated with high precision. The developed model was based on data from our

14

324

previously reported 2 single dose studies (14, 15) and the current multiple dose.

325

Allometric scaling of all four PK parameters (CL, V1, Q and V2) on body weight resulted

326

in significant model improvement and the PK parameter estimates were comparable to

327

our previously reported model (15). Overall, the model fit the data well as indicated by

328

the goodness-of-fit plots (Figures 2 and 3A), significant drop of the objective function

329

value and precision of parameter estimates (Table 3). To evaluate the final model and

330

determine its predictive accuracy we performed simulation analysis of the current study

331

with 200 replicates (n=15 subjects/each). Visual Predictive Check (Figure 3B) indicated

332

that the model is generally capable of predicting azithromycin concentrations in preterm

333

neonates, however, it under-predicted some concentrations that could be due to

334

random, unaccounted for inter-study variability. The model will be further refined and

335

inter-study variability can be better explained as data from more subjects become

336

available from current and future clinical studies.

337 338

Pharmacodynamic studies suggest that the AUC24/MIC90 ratio is most predictive

339

parameter of azithromycin efficacy (35). Results of experimental infections in animal

340

models suggest that increasing the dose early in the infectious process will result in

341

clearance of even marginally susceptible organisms (36). Although optimal AUC24/MIC90

342

ratios for azithromycin to eradicate other common organisms were reported to range

343

from 0.5 to 14.8 h (37), the optimal AUC24/MIC90 for azithromycin to effectively eradicate

344

Ureaplasma is unknown. In the prior 10 mg/kg and 20 mg/kg single dose studies, the

345

MIC90 of 4 µg/mL based on previously published reference data (38) was used to

346

calculate the AUC24/MIC90. For the current study, we based the MIC90 of 8 µg/ml on

15

347

azithromycin susceptibility testing of Ureaplasma isolates from study participants and

348

banked specimens from preterm infants. Our results indicated that an AUC24/MIC90 of >

349

4 hr would be effective to eradicate Ureaplasma. Since the activity of macrolides in vitro

350

is affected by pH of the medium (39, 40), the higher azithromycin MICs observed in

351

some isolates from the study cohorts as well as banked isolates may be due, in part, to

352

the acidic pH (6.0) of the 10B broth that is necessary for growth of the organisms in vitro

353

and measurement of the MIC endpoint. Therefore, at physiologic pH, the MIC may be 2-

354

4 dilutions lower, resulting in a higher AUC/MIC ratio. Furthermore, experimental

355

evidence in animal models and adult humans suggest that azithromycin concentrations

356

in alveolar macrophages in the lung will far exceed the Ureaplasma isolates MICs.

357

Following oral administration of 50 mg/kg azithormycin in rats, the AUC in alveolar

358

macrophages to AUC in plasma ratio was 648 (41). In human adults who received

359

either oral (42) or intravenous azithromcyin (43) for 5 days, the drug concentration was

360

increased more than 1500 fold in alveolar macrophages compared to plasma levels.

361 362

Based on the current study in preterm neonates, a dosage regimen of 3 intravenous

363

doses of 20 mg/kg/d azithromycin appeared to be safe and effectively eradicated

364

Ureaplasma from the respiratory tract in all culture-positive subjects. We suggest that

365

this azithromycin dosage regimen is appropriate for future randomized clinical trials to

366

eradicate Ureaplasma in preterm neonates.

367 368

Infants with BPD are at risk for adverse pulmonary outcomes during childhood. Up to

369

50% of BPD infants require re-hospitalization in the first year of life (44-47). Infants who

16

370

required home oxygen therapy are at highest risk for re-hospitalizations (45, 48).

371

Respiratory symptoms, primarily chronic cough and wheezing, and use of respiratory

372

medications (bronchodilators, steroids, oxygen) are more common in BPD than non-

373

BPD infants in early childhood. Maternal smoking during pregnancy and smoking in the

374

home also contribute to adverse respiratory outcomes in preterm infants (48).

375 376

Ureaplasma respiratory tract colonization has been proposed as an etiologic factor in

377

reactive airway disease in young infants. Wheezing in infants and children less than 3

378

years of age has been associated with isolation of Ureaplasma from the upper

379

respiratory tract (49). In a large study of almost 3000 women and their offspring in

380

Sweden, maternal vaginal colonization with Ureaplasma during pregnancy was

381

associated with a 2-fold increased risk for hospitalizations for asthma in the offspring

382

during the first 3 years of life (50). In preliminary data, we observed that 50% of

383

Ureaplasma-respiratory tract colonized infants compared to 27% of non-colonized

384

infants were re-hospitalized primarily for respiratory illnesses in the first year of life

385

(unpublished data). In the current study, none of the 14 survivors who had been treated

386

with the 3 day course of azithromycin were hospitalized during the first 6 months

387

(adjusted age) for respiratory illnesses.

388 389

Assessments of pulmonary outcomes beyond the neonatal period are important to

390

determine the larger impact of neonatal interventions to prevent BPD on lung health.

391

Treatment with intratracheal recombinant human CuZn superoxide dismutase at birth

392

did not reduce the rate of BPD at 28d, but did reduce the number of episodes of

17

393

wheezing, bronchodilator use, frequency of emergency room visits, and hospitalization

394

in treated infants in the first year of life compared to the placebo group (51). Exposure to

395

inhaled nitric oxide (iNO) between 7 and 21d of age in infants

Pharmacokinetics, microbial response, and pulmonary outcomes of multidose intravenous azithromycin in preterm infants at risk for Ureaplasma respiratory colonization.

The study objectives were to refine the population pharmacokinetics (PK) model, determine microbial clearance, and assess short-term pulmonary outcome...
890KB Sizes 2 Downloads 5 Views