Scolaris Content Display Scolaris Content Display

Inositol para lactantes prematuros con síndrome de dificultad respiratoria o con riesgo de presentarlo

Contraer todo Desplegar todo

Referencias

Referencias de los estudios incluidos en esta revisión

Friedman 1995 {published and unpublished data}

Friedman CA, McVey J, Borne MJ, James M, May WL, Temple DM, et al. Relationship between serum inositol concentration and development of retinopathy of prematurity: A prospective study. Journal of Pediatric Ophthalmology and Strabismus 2000;37(2):79‐86. [PUBMED: 10779265]CENTRAL
Friedman CA, Temple DM, Robbins KK, Miller CJ, Rawson JE. Randomized controlled trial of high inositol and calorie supplementation in preterm infants at risk for chronic lung and eye disease. American Academy of Pediatrics Annual Meeting; San Francisco, California, USA. 1995 October. CENTRAL

Hallman 1986 {published data only}

Hallman M, Jarvanpaa AL, Pohjavuori M. Respiratory distress syndrome and inositol supplementation in preterm infants. Archives of Disease in Childhood 1986;61(11):1076‐83. [PUBMED: 3539028]CENTRAL

Hallman 1992 {published data only}

Hallman M, Bry K, Hoppu K, Lappi M, Pohjavuori M. Inositol supplementation in premature infants with respiratory distress syndrome. New England Journal of Medicine 1992;326(19):1233‐9. [DOI: 10.1056/NEJM199205073261901; PUBMED: 1560798]CENTRAL
Hallman M, Pohjavuori M, Bry K. Inositol supplementation in respiratory distress syndrome. Lung 1990;168 Suppl:877‐82. [PUBMED: 2117207]CENTRAL

Phelps 2013 {published data only}

Phelps DL, Ward RM, Williams RL, Watterberg KL, Laptook AR, Wrage LA, et al. Pharmacokinetics and safety of a single intravenous dose of myo‐inositol in preterm infants of 23‐29 wk. Pediatric Research 2013;74(6):721‐9. [DOI: 10.1038/pr.2013.162; PUBMED: 24067395 ]CENTRAL
Phelps DL, Ward RM, Williams RL, Watterberg KL, Laptook AR, Wrage LA, et al. on behalf of the Inositol Subcommittee of the Neonatal Research Network. Pharmacokinetics and safety of a single dose of myo‐inositol in preterm infants of 23‐29 wk. E‐PAS2010. 2010:3737.387. CENTRAL

Phelps 2016 {published data only}

Phelps DL, Ward RM, Williams RL, Nolen TL, Watterberg KL, Oh W, et al. Safety and pharmacokinetics of multiple dose myo‐inositol in preterm infants. Pediatric Research 2016;80(2):209‐17. CENTRAL

Phelps 2018 {published data only}

Phelps DL, Watterberg KL, Nolen TL, Cole CA, Cotten M, Oh W, et al. Effects of myo‐inositol on type 1 retinopathy of prematurity among preterm infants <28 weeks’ gestational age: a randomized clinical trial. JAMA 2018;320(16):1649‐58. [DOI: 10.1001/jama.2018.14996]CENTRAL

Referencias adicionales

Bell 1978

Bell MJ, Ternberg KL, Feigin RD, Keating JP, Marshall R, Barton L, et al. Neonatal necrotising enterocolitis: therapeutic decisions based on clinical staging. Annals of Surgery 1978;187(1):1‐7. [PUBMED: 413500]

Bromberger 1986

Bromberger P, Hallman M. Myoinositol in small preterm infants: Relationship between intake and serum concentration. Journal of Pediatric Gastroenterology and Nutrition 1986;5(3):455‐8. [PUBMED: 3088251]

Burton 1974

Burton LE, Wells WW. Studies on the development pattern of the enzymes converting glucose‐6‐phosphate to myo‐inositol in the rat. Developmental Biology 1974;37(1):35‐42. [PUBMED: 4362962]

Dawson 1961

Dawson RM, Freinkel N. The distribution of free mesoinositol in mammalian tissues, including some observations on the lactating rat. Biochemical Journal 1961;78(3):606‐10. [PUBMED: 13720328]

Eagle 1957

Eagle H, Oyama VI, Levy M, Freeman AE. Myo‐inositol as an essential growth factor for normal and malignant human cells in tissue culture. Journal of Biological Chemistry 1957;266(1):191‐205. [PUBMED: 13428752]

Egberts 1986

Egberts J, Noort WA. Gestational age‐dependent changes in plasma inositol levels and surfactant composition in the fetal rat. Pediatric Research 1986;20(1):24‐7. [PUBMED: 3753753]

Guadagnino 2012

Guadagino E, Zuccato D. Delamination propensity of pharmaceutical glass containers by accelerated testing with different extraction media. PDA Journal of Pharmaceutical Science and Technology 2012;66(2):116‐25. [DOI: 10.5731/pdajpst.2012.00853; PUBMED: 22492597]

Guarner 1992

Guarner V, Tordet C, Bourbon JR. Effects of maternal protein‐calorie malnutrition on the phospholipid composition of surfactant isolated from the fetal and neonatal rat lungs. Compensation by inositol and lipid supplementation. Pediatric Research 1992;31(6):629‐35. [DOI: 10.1203/00006450‐199206000‐00018; PUBMED: 1635827]

Hallman 1980

Hallman M, Epstein BL. Role of myo‐inositol in the synthesis of phosphatidylglycerol and phosphatidylinositol in the lung. Biochemical and Biophysical Research Communications 1980;92(4):1151‐9. [PUBMED: 6245646]

Hallman 1984

Hallman M. Effect of intracellular myo‐inositol on the surfactant phospholipid synthesis in the fetal rabbit lung. Biochimica et Biophysica Acta 1984;795(1):67‐78. [PUBMED: 6547857]

Hallman 1985

Hallman M, Saugstad OD, Porreco RP, Epstein BL, Gluck L. Role of myo‐inositol in regulation of surfactant phospholipids in the newborn. Early Human Development 1985;10(3‐4):245‐54. [PUBMED: 3838720]

Hallman 1987

Hallman M, Arjomaa P, Hoppu K. Inositol supplementation in respiratory distress syndrome: Relationship between serum concentration, renal excretion, and lung effluent phospholipids. Journal of Pediatrics 1987;110(4):604‐10. [PUBMED: 3559811]

Hallman 1990

Hallman M, Pohjavuori M, Bry K. Inositol supplementation in respiratory distress syndrome. Lung 1990;168 Suppl:877‐82. [PUBMED: 2117207]

Hasan 1974

Hasan SH, Nishigaki I, Tsutsui Y, Yagi K. Studies on myoinositol IX. Morphological examination of the effect of massive doses of myoinositol on the liver and kidney of rat. Journal of Nutritional Science and Vitaminology 1974;20(1):55‐8. [PUBMED: 4836951]

Higgins 2003

Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta‐analyses. BMJ 2003;327(7414):557‐60. [PUBMED: 12958120]

Higgins 2011

Higgins JP, Green S, (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1. The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

ICROP 1984

Committee for the Classification of Retinopathy of Prematurity. An international classification of retinopathy of prematurity. The Committee for the Classification of Retinopathy of Prematurity. Archives of Ophthalmology 1984;102(8):1130‐4. [PUBMED: 6547831]

Lewin 1978

Lewin LM, Melmed S, Passwell JH, Yannai Y, Brish M, Orda S, et al. Myoinositol in human neonates: serum concentrations and renal handling. Pediatric Research 1978;12(1):3‐6. [DOI: 10.1203/00006450‐197801000‐00002; PUBMED: 643373]

Manske 2016

Mankse C, Schell U, Hibi H. Metabolism of myo‐inositol by Legionella pneumophilia promotes infection of amoebae and macrophages. Applied Environmental Microbiology 2016;82(16):5000‐14. [DOI: 10.1128/AEM.01018‐16; PUBMED: 27287324]

Papile 1978

Papile LA, Burstein J, Burstein R, Koffler H. Incidence and evolution of subependymal and intraventricular hemorrhage: a study of infants with birthweights less than 1,500 gm. Journal of Pediatrics 1978;92(4):529‐34. [PUBMED: 305471]

Pereira 1990

Pereira GR, Baker L, Egler J, Corcoran L, Chiavacci R. Serum myoinositol concentrations in premature infants fed human milk, formula for infants, and parenteral nutrition. American Journal of Clinical Nutrition 1990;51(4):589‐93. [DOI: 10.1093/ajcn/51.4.589; PUBMED: 2108579]

Review Manager 2014 [Computer program]

Nordic Cochrane Centre, The Cochrane Collaboration. Review Manager 5 (RevMan 5). Version 5.3. Copenhagen: Nordic Cochrane Centre, The Cochrane Collaboration, 2014.

Shennan 1988

Shennan AT, Dunn MS, Ohlsson A, Lennox K, Hoskins EM. Abnormal pulmonary outcomes in premature infants: prediction from oxygen requirement in the neonatal period. Pediatrics 1988;82(4):527‐32. [PUBMED: 3174313]

Soll 1992

Soll RF, McQueen MC. Respiratory distress syndrome. In: Sinclair JC, Bracken MB editor(s). Effective Care of the Newborn Infant. Oxford: Oxford University Press, 1992.

Zhao 2014

Zhao J, Lavalley V, Mangiagalli P, Wright JM, Bankston TE. Glass delamination: a comparison of the inner surface performance of vials and pre‐filled syringes. AAPS PharmSciTech 2014;15(6):1398‐409. [DOI: 10.1208/s12249‐014‐0167‐y; PUBMED: 24938618]

Referencias de otras versiones publicadas de esta revisión

Howlett 1997

Howlett A, Ohlsson A. Inositol for respiratory distress syndrome in preterm infants. Cochrane Database of Systematic Reviews 1997, Issue 4. [DOI: 10.1002/14651858.CD000366]

Howlett 2003

Howlett A, Ohlsson A. Inositol for respiratory distress syndrome in preterm infants. Cochrane Database of Systematic Reviews 2003, Issue 4. [DOI: 10.1002/14651858.CD000366]

Howlett 2012

Howlett A, Ohlsson A, Plakkal N. Inositol for respiratory distress syndrome in preterm infants. Cochrane Database of Systematic Reviews 2012, Issue 3. [DOI: 10.1002/14651858.CD000366.pub2]

Howlett 2015

Howlett A, Ohlsson A, Plakkal N. Inositol in preterm infants at risk for or having respiratory distress syndrome. Cochrane Database of Systematic Reviews 2015, Issue 2. [DOI: 10.1002/14651858.CD000366.pub3]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Friedman 1995

Methods

Randomised, placebo‐controlled trial

Participants

Preterm infants (birth weight < 1500 grams) with a diagnosis of RDS, requiring mechanical ventilation.

24 infants were randomised to high concentration inositol formula (SC 30) (estimated PMA 27.7, SD 1.9) and 24 infants were randomised to a low concentration of inositol formula (SC 24). Randomisation ended when the high‐inositol formula was no longer available.

Location: 2 NICUs in the US. Study period: October 1994 to June 1998.

Interventions

The study group was enterally fed high‐inositol formula (2500 µmol/L inositol), while the control group was given low‐inositol formula (242 µmol/L) enterally.

Outcomes

Neonatal deaths, infant deaths, infants with bacteraemia, necrotizing enterocolitis (radio graphically documented), IVH > grade 2, BPD (oxygen therapy > 30 days), duration of mechanical ventilation, ROP (reported in unpublished data from 1995).

Notes

The results of this study have been reported 3 times; in abstract form in 1995 after 37 infants were enrolled; in a personal communication report to us in 1995 when 41 infants had been enrolled; and in a final published report in 2000 when 48 infants had been entered.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No information provided.

Allocation concealment (selection bias)

Unclear risk

Infants were allocated to one of the two groups by sequential random card selection. No information provided whether the cards were enclosed in opaque and numbered envelopes.

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Infants were blinded but no information provided whether the clinical staff and the researchers were.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Outcomes provided for all 48 infants randomised.

Selective reporting (reporting bias)

Unclear risk

The protocol for the study was not available to us so we cannot judge if there were any deviations between the protocol and the final report.

Other bias

High risk

The results of this study have been reported 3 times: in abstract form in 1995 after 37 infants were enrolled; in a personal communication report to us in 1995 when 41 infants had been enrolled; and in a final published report in 2000 when 48 infants had been entered.

Hallman 1986

Methods

Randomised, placebo‐controlled, double‐blind trial. Enrolment from 1983 to 1985.

Participants

Preterm infants (birth weight < 2000 grams) (mean PMA 29.5, SD 2.0 in the inositol group and 29.5, SD 2.1 in the placebo group) with a diagnosis of RDS, requiring mechanical ventilation.
N = 74; placebo group = 37, inositol group = 37.

Location: 1 NICU in Helsinki, Finland. Study period: January 1983 to August 1985.

Interventions

IV or supplemental inositol (120 to 160 mg/kg/day) or placebo (5% glucose) given daily for 10 days.

Outcomes

Neonatal deaths, infant deaths, BPD (supplemental oxygen at 28 days and x‐ray findings compatible with BPD), IVH, ROP (ophthalmological exam at PMA of 9 and 13 months), NEC (clinical findings and abdominal x‐ray showing pneumatosis intestinalis and air in the portal circulation), and sepsis.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No information provided.

Allocation concealment (selection bias)

Low risk

"Infants were randomly and blindly assigned to be treated with inositol or placebo (glucose) after their parents had consented to their participation". For further details see "Blinding" below.

Blinding (performance bias and detection bias)
All outcomes

Low risk

Each set of solutions, containing either inositol or glucose (5% weight/volume each) had a code number. Only the pharmacist preparing the doses knew the contents of the drug packages.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Of the 83 infants who entered the trial, nine did not fulfil the entrance criteria and were excluded from the final analysis. An explanation was provided for each excluded infant.

Selective reporting (reporting bias)

Unclear risk

The protocol for the study was not available to us so we cannot judge if there were any deviations between the protocol and the final report.

Other bias

High risk

The present report represents the third interim analysis and the researchers may have been influenced by the results of the two previous interim analyses. The study was not registered in a trials registry.

Hallman 1992

Methods

Randomised, placebo‐controlled, double‐blind trial, occurring between 1985 and 1989.

Participants

Preterm infants (birth weight < 2000 grams and 24.0 to 31.9 weeks' PMA at birth) with evidence of RDS, requiring mechanical ventilation.
Total N = 233, placebo group = 114, inositol group = 119.
Age at enrolment 2 to 10 hours of life.

Interventions

The study group received IV inositol 80 mg/kg body weight daily for 5 days, with repeated courses at day 10 and day 20 if necessary (infant continued to require ventilation, required supplemental O₂ or did not tolerate enteral feeds). The control group received 5% glucose.

Outcomes

Neonatal death, infant death, BPD (supplemental oxygen at 28 days of age), BPD (supplemental oxygen at 38 weeks' PMA or the week of discharge from hospital), ROP (as per International Classification assessed from 4 to 6 weeks and ending at 12 months), IVH (all grades, grade > 2), NEC (no definition provided), and sepsis (no definition provided).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No information provided.

Allocation concealment (selection bias)

Unclear risk

Unclear.

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

5% glucose was given as placebo, but no information provided on whether staff was blinded to study drugs or not.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

4 infants in the placebo group and 3 in the inositol group died before receiving any treatment, 2 had lethal malformations (1 in each group), and 3 did not have RDS (2 in the placebo group and 1 in the inositol group). These 12 infants were included only in the safety analysis.

Selective reporting (reporting bias)

Unclear risk

This study was not registered in a trials registry so we cannot judge if there were any deviations between the protocol and the final report.

Other bias

High risk

Interim analyses were to be performed after enrolment of 100, 200, 300 patients. Early termination of the trial was recommended by the monitoring committee after the second interim analysis, when the Chi² test revealed a significant increase in neonatal survival without BPD and no trend towards serious morbidity in 1 study group. 1 interim analysis previously reported, Hallman 1992 (published in Lung 1990;168 Suppl: 877 to 82).

Phelps 2013

Methods

Randomised, double‐masked, placebo‐controlled pharmacokinetic (PK) study. Enrolment between June 2006 and December 2007. The trial was conducted by the National Institutes of Child Health and Human Development Neonatal Research Network. 10 of the Neonatal Research Network Centers participated.

Participants

Eligible subjects were of 23 0/7 to 29 6/7 weeks' PMA and ≥ 600 G BW, had no major congenital anomalies, were between 12 hours and 6 days of age at randomisation, and had received no human milk or formula feedings since birth.

Interventions

Inositol was given as a single low (60 mg/kg) (N = 25) or high (120 mg/kg) (N = 24) dose of 5% myo‐inositol IV over 20 min in a 1:1:1 randomisation with placebo delivered in 1 of 2 volumes to maintain masking (5% glucose) (N = 25). Drug or placebo was dispensed from the respective pharmacies in unit doses labelled as 'inositol study drug', and all clinical and research personnel except the pharmacist were masked to the study group.

Outcomes

Pharmacokinetic data for inositol (central volume of distribution, clearance, endogenous production, the half‐life, renal inositol excretion during the first 12 H and after 48 H and diuretic side effect.
In addition adverse events were reported for the first 7 days as well as neonatal morbidities from birth through hospital discharge (or 120 days if sooner).

Notes

Abbott Nutrition Division, Abbott Laboratories, supplied the inositol drug used in the study.

Portions of this study were presented at the 2010 Paediatric Academic Societies Annual Meeting, Vancouver, Canada, May 1–4, 2010 (Abstract 3737.387).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was performed centrally via computer within two pre‐specified PMA strata (23 0/7 to 26 6/7 weeks and 27 0/7 to 29 6/7 weeks).

Allocation concealment (selection bias)

Low risk

There was central allocation to study group.

Blinding (performance bias and detection bias)
All outcomes

Low risk

Drug or placebo was dispensed from the respective pharmacies in unit doses labelled as 'inositol study drug', and all clinical and research personnel except the pharmacist were masked to the study group.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Consent was obtained for 79 infants, 76 infants were randomised, and 74 infants received study drug. 2 infants did not complete the minimum of 4 specified blood samples (three post drug infusion), and their randomisation were replaced with 2 additional enrollees from the same centre and of the same gestational age (GA) stratum, per protocol. Available data from the 2 replaced infants were included in the PK and safety analyses. 1 infant received placebo instead of the assigned 120 mg/kg dose, and for the PK analysis, this infant’s serum and urine data were included in the placebo group. However, this subject’s data on adverse events and clinical outcomes were included as randomised (intention to treat).

Selective reporting (reporting bias)

Low risk

The study was registered with ClinicalTrials.gov (NCT00349726) and there do not appear to be any deviations from the protocol.

Other bias

Low risk

Appears free of other bias.

Phelps 2016

Methods

Prospective, parallel, randomised controlled trial. Infants enrolled in 14 centres in the Eunice Kennedy Shriver NICHD Neonatal Research Network.

Participants

Infants ≤ 29 weeks' PMA (23 0/7 to 29 6/7 weeks' PMA), who weighed at least 400 G, and could receive study drug by 72 H after birth.

Interventions

Myo‐inositol provided by Abbott Nutrition, Columbus, Ohio, USA as an isotonic, preservative and pyrogen‐free, sterile, 5% solution at 10, 40 or 80 mg/kg/day.

Intravenous administration converted to enteral when feedings were established, and continued to the first of 10 weeks, 34 weeks' PMA, death or discharge. Total number randomised: 10 mg/kg N = 29; 40 mg/kg N = 30; 80 mg/kg N = 28

Placebo: 5% glucose. Total number randomised: N = 35.

Outcomes

Adverse events were prospectively monitored from 24 hours prior to study drug until 7 days following the final dose (unless discharged sooner), and judged according to a neonatal toxicity table developed for the study. An unfavourable outcome was defined as either type 1 ROP or worse, in either eye, or surgical intervention for severe ROP in either eye. A favourable ROP outcome was assigned if the retinal vessels progressed to full vascularization in both eyes without meeting criteria for severe ROP, or if on 2 consecutive examinations the retinal vessels were in zone III. Infants who did not meet either criterion had all available examinations reviewed by an adjudication committee. Adjudication was conducted by a committee of 3 experienced ophthalmologists not involved with the study and masked to study group assignment. The final ROP status was judged separately in each eye as 'probably favourable', 'probably unfavourable' or 'cannot be determined', and the majority classification was assigned as the adjudicated outcome. At 18 to 22 months' corrected age, infants received a set of standardized examinations of neurologic function and development according to the NRN Follow‐Up Protocol (to be reported separately).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer generated.

Allocation concealment (selection bias)

Low risk

Computer generated and communicated to research pharmacist.

Blinding (performance bias and detection bias)
All outcomes

Low risk

Personal communication from the first author indicates blinded performance and detection bias for all outcomes. Ophthalmologists were blinded during the adjudication process.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Severe ROP data presented for 106 surviving infants and there were 15 deaths, which adds up to 121 infants not 122 infants, which was the number enrolled.

Selective reporting (reporting bias)

Low risk

The protocol was available to us and we did not notice any major deviations from the planned study. The study was registered as: NCT01030575.

Other bias

Low risk

Appears free of other bias.

Phelps 2018

Methods

Randomised clinical trial included infants enrolled from 18 neonatal intensive care centres throughout the USA from 17 April 2014 to 4 September 2015; final date of follow‐up was 12 February 2016.

Participants

638 infants < 28 weeks’ PMA, surviving for at least 12 hours, and admitted to 1 of the 18 Neonatal Research Network centres before 72 hours’ postnatal age.

Interventions

A 40 mg/kg dose of myo‐inositol was given every 12 hours (initially intravenously, then enterally when feeding; (N = 317) or for up to 10 weeks. The active drug was an isotonic, sterile, pyrogen‐ and preservative‐free aqueous solution of 5% myo‐inositol (50 mg/mL) at neutral pH and was provided by Abbott Laboratories. A dose of 40 mg/kg every 12 hours was selected to achieve serum concentrations similar to those previously reported. A therapeutic duration of up to 10 weeks was chosen to sustain serum myo‐inositol levels similar to those found in utero throughout the period of normal retinal vascular development and because of the reported benefits in the treatment of ROP and survival.

Placebo (N = 321) (5% glucose for IV use from pharmacy stock).

Outcomes

The unfavourable primary outcome was type 1 ROP, which was defined as meeting the criteria for ophthalmological intervention to prevent retinal detachment, a more severe ROP type than ROP type 1 (e.g. aggressive posterior ROP or Rush disease), or death before the ROP outcome could be determined.

Notes

The planned enrolment of 1760 participants would permit detection of an absolute reduction in death or type 1 ROP of 7% with 90% power. The trial was terminated early due to a statistically significantly higher mortality rate in the myo‐inositol group. The favourable primary outcome was survival with only milder ROP or no ROP. Infants were followed up as outpatients to determine the primary outcome up to a maximum of 55 weeks’ PMA.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated and centrally administered randomisation.

Allocation concealment (selection bias)

Low risk

With the exception of pharmacists, who prepared the daily unit doses of myo‐inositol or placebo according to randomisation assignment, all other clinical and research personnel and families were blind to group assignment.

Blinding (performance bias and detection bias)
All outcomes

Low risk

See above for allocation concealment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Outcome group: of the 317 infants randomised to receive myo‐inositol 313 received the intervention as randomised; 3 died prior to receiving the intervention and 1 was withdrawn prior to intervention. 313 included in primary analysis. Of the 321 infants randomised to placebo 315 received the intervention as randomised; 2 died prior to receiving intervention and 4 received < 2 doses of myo‐inositol. 320 included in the primary analysis of type 1 retinopathy of prematurity or death. 1 excluded from primary analysis.

Selective reporting (reporting bias)

Low risk

The protocol for the study was available to us and we did not identify any deviations except that the study was terminated prior to reaching the full sample size because of safety concerns. The authors state: “No changes to the protocol occurred during the trial”.

Registered as: NCT01954082.

Other bias

Low risk

Appears free of other bias.

BW = birth weight

PMA = postmenstrual age

PK = pharmacokinetics

Data and analyses

Open in table viewer
Comparison 1. Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Neonatal death (age < 28 days) Show forest plot

3

355

Risk Ratio (M‐H, Fixed, 95% CI)

0.53 [0.31, 0.91]

Analysis 1.1

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 1 Neonatal death (age < 28 days).

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 1 Neonatal death (age < 28 days).

2 Infant death (age < one year) Show forest plot

5

1115

Risk Ratio (M‐H, Fixed, 95% CI)

0.89 [0.71, 1.13]

Analysis 1.2

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 2 Infant death (age < one year).

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 2 Infant death (age < one year).

3 BPD (supplementary oxygen ar 36 weeks; PMA or death due to BPD)at 36 week's PMA Show forest plot

2

666

Risk Ratio (M‐H, Fixed, 95% CI)

1.00 [0.87, 1.14]

Analysis 1.3

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 3 BPD (supplementary oxygen ar 36 weeks; PMA or death due to BPD)at 36 week's PMA.

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 3 BPD (supplementary oxygen ar 36 weeks; PMA or death due to BPD)at 36 week's PMA.

4 Bronchopulmonary dysplasia (at 28 to 30 days of age) Show forest plot

3

343

Risk Ratio (M‐H, Fixed, 95% CI)

0.78 [0.54, 1.13]

Analysis 1.4

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 4 Bronchopulmonary dysplasia (at 28 to 30 days of age).

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 4 Bronchopulmonary dysplasia (at 28 to 30 days of age).

5 Bronchopulmonary dysplasia (at 36 to 38 weeks PMA) Show forest plot

2

737

Risk Ratio (M‐H, Fixed, 95% CI)

1.04 [0.90, 1.20]

Analysis 1.5

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 5 Bronchopulmonary dysplasia (at 36 to 38 weeks PMA).

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 5 Bronchopulmonary dysplasia (at 36 to 38 weeks PMA).

6 Retinopathy of prematurity, stage ≥ 3 or ≥ 2 Show forest plot

3

810

Risk Ratio (M‐H, Fixed, 95% CI)

0.89 [0.75, 1.06]

Analysis 1.6

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 6 Retinopathy of prematurity, stage ≥ 3 or ≥ 2.

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 6 Retinopathy of prematurity, stage ≥ 3 or ≥ 2.

6.1 ROP ≥ 3

2

262

Risk Ratio (M‐H, Fixed, 95% CI)

0.09 [0.01, 0.67]

6.2 Retinopathy of prematurity, stage ≥ 2

1

548

Risk Ratio (M‐H, Fixed, 95% CI)

0.95 [0.80, 1.13]

7 Retinopathy of prematurity, any stage Show forest plot

4

889

Risk Ratio (M‐H, Fixed, 95% CI)

0.94 [0.83, 1.07]

Analysis 1.7

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 7 Retinopathy of prematurity, any stage.

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 7 Retinopathy of prematurity, any stage.

8 Necrotizing enterocolitis (suspected or proven) Show forest plot

5

1115

Risk Ratio (M‐H, Fixed, 95% CI)

0.94 [0.64, 1.39]

Analysis 1.8

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 8 Necrotizing enterocolitis (suspected or proven).

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 8 Necrotizing enterocolitis (suspected or proven).

9 Sepsis (early and/or late onset) Show forest plot

4

1067

Risk Ratio (M‐H, Fixed, 95% CI)

1.21 [0.95, 1.54]

Analysis 1.9

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 9 Sepsis (early and/or late onset).

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 9 Sepsis (early and/or late onset).

10 Intraventricular haemorrhage, grade > 2 Show forest plot

5

1103

Risk Ratio (M‐H, Fixed, 95% CI)

0.77 [0.58, 1.01]

Analysis 1.10

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 10 Intraventricular haemorrhage, grade > 2.

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 10 Intraventricular haemorrhage, grade > 2.

11 Intraventricular haemorrhage, all grades Show forest plot

3

427

Risk Ratio (M‐H, Fixed, 95% CI)

0.77 [0.59, 1.00]

Analysis 1.11

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 11 Intraventricular haemorrhage, all grades.

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 11 Intraventricular haemorrhage, all grades.

12 Minor neural developmental impairment at one year corrected age Show forest plot

1

169

Risk Ratio (M‐H, Fixed, 95% CI)

0.84 [0.38, 1.86]

Analysis 1.12

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 12 Minor neural developmental impairment at one year corrected age.

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 12 Minor neural developmental impairment at one year corrected age.

13 Major neural developmental impairment at one year corrected age Show forest plot

1

169

Risk Ratio (M‐H, Fixed, 95% CI)

0.53 [0.24, 1.16]

Analysis 1.13

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 13 Major neural developmental impairment at one year corrected age.

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 13 Major neural developmental impairment at one year corrected age.

Open in table viewer
Comparison 2. Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Death during hospital stay Show forest plot

1

74

Risk Ratio (M‐H, Fixed, 95% CI)

1.19 [0.34, 4.21]

Analysis 2.1

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 1 Death during hospital stay.

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 1 Death during hospital stay.

2 Bronchopulmonary dysplasia at 36 weeks PMA Show forest plot

1

65

Risk Ratio (M‐H, Fixed, 95% CI)

2.74 [0.88, 8.48]

Analysis 2.2

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 2 Bronchopulmonary dysplasia at 36 weeks PMA.

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 2 Bronchopulmonary dysplasia at 36 weeks PMA.

3 Retinopathy of prematurity (infants who underwent surgery for ROP) Show forest plot

1

25

Risk Ratio (M‐H, Fixed, 95% CI)

0.35 [0.10, 1.22]

Analysis 2.3

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 3 Retinopathy of prematurity (infants who underwent surgery for ROP).

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 3 Retinopathy of prematurity (infants who underwent surgery for ROP).

4 Necrotizing enterocolitis (stage 2A or worse) Show forest plot

1

74

Risk Ratio (M‐H, Fixed, 95% CI)

0.41 [0.12, 1.39]

Analysis 2.4

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 4 Necrotizing enterocolitis (stage 2A or worse).

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 4 Necrotizing enterocolitis (stage 2A or worse).

5 Necrotizing enterocolitis (infants who underwent surgery for NEC) Show forest plot

1

74

Risk Ratio (M‐H, Fixed, 95% CI)

0.51 [0.08, 3.41]

Analysis 2.5

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 5 Necrotizing enterocolitis (infants who underwent surgery for NEC).

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 5 Necrotizing enterocolitis (infants who underwent surgery for NEC).

6 Sepsis (late onset) Show forest plot

1

74

Risk Ratio (M‐H, Fixed, 95% CI)

1.46 [0.71, 2.97]

Analysis 2.6

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 6 Sepsis (late onset).

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 6 Sepsis (late onset).

7 Intraventricular haemorrhage (grade 3 or 4) Show forest plot

1

72

Risk Ratio (M‐H, Fixed, 95% CI)

1.06 [0.29, 3.90]

Analysis 2.7

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 7 Intraventricular haemorrhage (grade 3 or 4).

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 7 Intraventricular haemorrhage (grade 3 or 4).

8 Hearing test (failed both ears) Show forest plot

1

57

Risk Ratio (M‐H, Fixed, 95% CI)

0.58 [0.09, 3.84]

Analysis 2.8

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 8 Hearing test (failed both ears).

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 8 Hearing test (failed both ears).

Open in table viewer
Comparison 3. Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Type 1 ROP or death before determination of ROP outcome using the adjudicated ROP outcome Show forest plot

2

679

Risk Ratio (M‐H, Fixed, 95% CI)

1.28 [0.99, 1.67]

Analysis 3.1

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 1 Type 1 ROP or death before determination of ROP outcome using the adjudicated ROP outcome.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 1 Type 1 ROP or death before determination of ROP outcome using the adjudicated ROP outcome.

2 Type 1 ROP Show forest plot

1

511

Risk Ratio (M‐H, Fixed, 95% CI)

1.41 [0.89, 2.24]

Analysis 3.2

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 2 Type 1 ROP.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 2 Type 1 ROP.

3 Death before determination of ROP outcome Show forest plot

1

638

Risk Ratio (M‐H, Fixed, 95% CI)

1.53 [1.02, 2.31]

Analysis 3.3

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 3 Death before determination of ROP outcome.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 3 Death before determination of ROP outcome.

4 Type 1 ROP including adjudicated ROP outcome Show forest plot

2

605

Risk Ratio (M‐H, Fixed, 95% CI)

1.24 [0.82, 1.86]

Analysis 3.4

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 4 Type 1 ROP including adjudicated ROP outcome.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 4 Type 1 ROP including adjudicated ROP outcome.

5 Any ROP Show forest plot

1

553

Risk Ratio (M‐H, Fixed, 95% CI)

1.00 [0.88, 1.13]

Analysis 3.5

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 5 Any ROP.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 5 Any ROP.

6 ROP ≥ 2 ROP Show forest plot

1

548

Risk Ratio (M‐H, Fixed, 95% CI)

0.95 [0.80, 1.13]

Analysis 3.6

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 6 ROP ≥ 2 ROP.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 6 ROP ≥ 2 ROP.

7 All cause infant mortality to 55 week's PMA Show forest plot

1

638

Risk Ratio (M‐H, Fixed, 95% CI)

1.67 [1.12, 2.48]

Analysis 3.7

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 7 All cause infant mortality to 55 week's PMA.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 7 All cause infant mortality to 55 week's PMA.

8 All cause mortality (outcome collected through first event: death, hospital discharge, hospital transfer, or 120 days after birth) Show forest plot

2

701

Risk Ratio (M‐H, Fixed, 95% CI)

1.35 [0.91, 2.00]

Analysis 3.8

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 8 All cause mortality (outcome collected through first event: death, hospital discharge, hospital transfer, or 120 days after birth).

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 8 All cause mortality (outcome collected through first event: death, hospital discharge, hospital transfer, or 120 days after birth).

9 BPD (requiring oxygen at 36 week's PMA for oxygen saturation > 90%) Show forest plot

1

560

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.89, 1.18]

Analysis 3.9

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 9 BPD (requiring oxygen at 36 week's PMA for oxygen saturation > 90%).

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 9 BPD (requiring oxygen at 36 week's PMA for oxygen saturation > 90%).

10 BPD or death by it prior to 37 weeks' PMA (outcomes collected through first event: death, hospital discharge, hospital transfer, or 120 days after birth) Show forest plot

2

616

Risk Ratio (M‐H, Fixed, 95% CI)

1.01 [0.87, 1.16]

Analysis 3.10

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 10 BPD or death by it prior to 37 weeks' PMA (outcomes collected through first event: death, hospital discharge, hospital transfer, or 120 days after birth).

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 10 BPD or death by it prior to 37 weeks' PMA (outcomes collected through first event: death, hospital discharge, hospital transfer, or 120 days after birth).

11 Severe IVH (grade 3 or 4) Show forest plot

2

690

Risk Ratio (M‐H, Fixed, 95% CI)

0.92 [0.65, 1.29]

Analysis 3.11

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 11 Severe IVH (grade 3 or 4).

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 11 Severe IVH (grade 3 or 4).

12 Cystic areas in the cerebral parenchyma measured through 28 d Show forest plot

2

225

Risk Ratio (M‐H, Fixed, 95% CI)

1.29 [0.58, 2.85]

Analysis 3.12

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 12 Cystic areas in the cerebral parenchyma measured through 28 d.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 12 Cystic areas in the cerebral parenchyma measured through 28 d.

13 Early onset sepsis Show forest plot

1

63

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Analysis 3.13

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 13 Early onset sepsis.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 13 Early onset sepsis.

14 Late onset sepsis (> 72 hrs of age) Show forest plot

2

701

Risk Ratio (M‐H, Fixed, 95% CI)

1.33 [1.00, 1.75]

Analysis 3.14

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 14 Late onset sepsis (> 72 hrs of age).

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 14 Late onset sepsis (> 72 hrs of age).

15 Suspected or proven NEC Show forest plot

2

701

Risk Ratio (M‐H, Fixed, 95% CI)

0.88 [0.55, 1.41]

Analysis 3.15

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 15 Suspected or proven NEC.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 15 Suspected or proven NEC.

16 Surgical NEC Show forest plot

2

701

Risk Ratio (M‐H, Fixed, 95% CI)

1.21 [0.57, 2.58]

Analysis 3.16

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 16 Surgical NEC.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 16 Surgical NEC.

17 Spontaneous gastro‐intestinal perforation Show forest plot

2

701

Risk Ratio (M‐H, Fixed, 95% CI)

0.86 [0.48, 1.52]

Analysis 3.17

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 17 Spontaneous gastro‐intestinal perforation.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 17 Spontaneous gastro‐intestinal perforation.

18 Pulmonary haemorrhage Show forest plot

1

638

Risk Ratio (M‐H, Fixed, 95% CI)

0.98 [0.59, 1.62]

Analysis 3.18

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 18 Pulmonary haemorrhage.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 18 Pulmonary haemorrhage.

19 PDA Show forest plot

2

700

Risk Ratio (M‐H, Fixed, 95% CI)

0.98 [0.85, 1.14]

Analysis 3.19

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 19 PDA.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 19 PDA.

20 PDA requiring indomethacin Show forest plot

1

637

Risk Ratio (M‐H, Fixed, 95% CI)

0.90 [0.67, 1.22]

Analysis 3.20

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 20 PDA requiring indomethacin.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 20 PDA requiring indomethacin.

21 PDA requiring surgery Show forest plot

2

700

Risk Ratio (M‐H, Fixed, 95% CI)

0.96 [0.65, 1.42]

Analysis 3.21

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 21 PDA requiring surgery.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 21 PDA requiring surgery.

22 Seizure treatment for ≥ 2 days Show forest plot

2

700

Risk Ratio (M‐H, Fixed, 95% CI)

1.04 [0.43, 2.56]

Analysis 3.22

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 22 Seizure treatment for ≥ 2 days.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 22 Seizure treatment for ≥ 2 days.

23 Negative hearing screening in either ear at discharge Show forest plot

2

472

Risk Ratio (M‐H, Fixed, 95% CI)

1.45 [0.92, 2.29]

Analysis 3.23

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 23 Negative hearing screening in either ear at discharge.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 23 Negative hearing screening in either ear at discharge.

24 Respiratory distress syndrome Show forest plot

1

63

Risk Ratio (M‐H, Fixed, 95% CI)

0.99 [0.91, 1.09]

Analysis 3.24

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 24 Respiratory distress syndrome.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 24 Respiratory distress syndrome.

25 Sepsis, necrotizing enterocolitis, pneumonia or other infection as a cause of death Show forest plot

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

1.36 [0.95, 1.93]

Analysis 3.25

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 25 Sepsis, necrotizing enterocolitis, pneumonia or other infection as a cause of death.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 25 Sepsis, necrotizing enterocolitis, pneumonia or other infection as a cause of death.

Study flow diagram: review update
Figuras y tablas -
Figure 1

Study flow diagram: review update

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figuras y tablas -
Figure 2

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figuras y tablas -
Figure 3

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Forest plot of comparison: 1 Inositol supplementation (repeat doses in any amount and any duration of treatment) versus control, outcome: 1.5 Retinopathy of prematurity, stage ≥ 3.
Figuras y tablas -
Figure 4

Forest plot of comparison: 1 Inositol supplementation (repeat doses in any amount and any duration of treatment) versus control, outcome: 1.5 Retinopathy of prematurity, stage ≥ 3.

Forest plot of comparison: 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, outcome: 3.4 Type 1 ROP including adjudicated ROP outcome.
Figuras y tablas -
Figure 5

Forest plot of comparison: 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, outcome: 3.4 Type 1 ROP including adjudicated ROP outcome.

Forest plot of comparison: 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, outcome: 3.14 Late onset sepsis (> 72 hours of age).
Figuras y tablas -
Figure 6

Forest plot of comparison: 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, outcome: 3.14 Late onset sepsis (> 72 hours of age).

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 1 Neonatal death (age < 28 days).
Figuras y tablas -
Analysis 1.1

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 1 Neonatal death (age < 28 days).

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 2 Infant death (age < one year).
Figuras y tablas -
Analysis 1.2

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 2 Infant death (age < one year).

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 3 BPD (supplementary oxygen ar 36 weeks; PMA or death due to BPD)at 36 week's PMA.
Figuras y tablas -
Analysis 1.3

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 3 BPD (supplementary oxygen ar 36 weeks; PMA or death due to BPD)at 36 week's PMA.

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 4 Bronchopulmonary dysplasia (at 28 to 30 days of age).
Figuras y tablas -
Analysis 1.4

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 4 Bronchopulmonary dysplasia (at 28 to 30 days of age).

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 5 Bronchopulmonary dysplasia (at 36 to 38 weeks PMA).
Figuras y tablas -
Analysis 1.5

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 5 Bronchopulmonary dysplasia (at 36 to 38 weeks PMA).

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 6 Retinopathy of prematurity, stage ≥ 3 or ≥ 2.
Figuras y tablas -
Analysis 1.6

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 6 Retinopathy of prematurity, stage ≥ 3 or ≥ 2.

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 7 Retinopathy of prematurity, any stage.
Figuras y tablas -
Analysis 1.7

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 7 Retinopathy of prematurity, any stage.

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 8 Necrotizing enterocolitis (suspected or proven).
Figuras y tablas -
Analysis 1.8

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 8 Necrotizing enterocolitis (suspected or proven).

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 9 Sepsis (early and/or late onset).
Figuras y tablas -
Analysis 1.9

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 9 Sepsis (early and/or late onset).

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 10 Intraventricular haemorrhage, grade > 2.
Figuras y tablas -
Analysis 1.10

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 10 Intraventricular haemorrhage, grade > 2.

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 11 Intraventricular haemorrhage, all grades.
Figuras y tablas -
Analysis 1.11

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 11 Intraventricular haemorrhage, all grades.

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 12 Minor neural developmental impairment at one year corrected age.
Figuras y tablas -
Analysis 1.12

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 12 Minor neural developmental impairment at one year corrected age.

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 13 Major neural developmental impairment at one year corrected age.
Figuras y tablas -
Analysis 1.13

Comparison 1 Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control, Outcome 13 Major neural developmental impairment at one year corrected age.

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 1 Death during hospital stay.
Figuras y tablas -
Analysis 2.1

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 1 Death during hospital stay.

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 2 Bronchopulmonary dysplasia at 36 weeks PMA.
Figuras y tablas -
Analysis 2.2

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 2 Bronchopulmonary dysplasia at 36 weeks PMA.

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 3 Retinopathy of prematurity (infants who underwent surgery for ROP).
Figuras y tablas -
Analysis 2.3

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 3 Retinopathy of prematurity (infants who underwent surgery for ROP).

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 4 Necrotizing enterocolitis (stage 2A or worse).
Figuras y tablas -
Analysis 2.4

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 4 Necrotizing enterocolitis (stage 2A or worse).

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 5 Necrotizing enterocolitis (infants who underwent surgery for NEC).
Figuras y tablas -
Analysis 2.5

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 5 Necrotizing enterocolitis (infants who underwent surgery for NEC).

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 6 Sepsis (late onset).
Figuras y tablas -
Analysis 2.6

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 6 Sepsis (late onset).

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 7 Intraventricular haemorrhage (grade 3 or 4).
Figuras y tablas -
Analysis 2.7

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 7 Intraventricular haemorrhage (grade 3 or 4).

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 8 Hearing test (failed both ears).
Figuras y tablas -
Analysis 2.8

Comparison 2 Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants, Outcome 8 Hearing test (failed both ears).

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 1 Type 1 ROP or death before determination of ROP outcome using the adjudicated ROP outcome.
Figuras y tablas -
Analysis 3.1

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 1 Type 1 ROP or death before determination of ROP outcome using the adjudicated ROP outcome.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 2 Type 1 ROP.
Figuras y tablas -
Analysis 3.2

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 2 Type 1 ROP.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 3 Death before determination of ROP outcome.
Figuras y tablas -
Analysis 3.3

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 3 Death before determination of ROP outcome.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 4 Type 1 ROP including adjudicated ROP outcome.
Figuras y tablas -
Analysis 3.4

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 4 Type 1 ROP including adjudicated ROP outcome.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 5 Any ROP.
Figuras y tablas -
Analysis 3.5

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 5 Any ROP.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 6 ROP ≥ 2 ROP.
Figuras y tablas -
Analysis 3.6

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 6 ROP ≥ 2 ROP.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 7 All cause infant mortality to 55 week's PMA.
Figuras y tablas -
Analysis 3.7

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 7 All cause infant mortality to 55 week's PMA.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 8 All cause mortality (outcome collected through first event: death, hospital discharge, hospital transfer, or 120 days after birth).
Figuras y tablas -
Analysis 3.8

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 8 All cause mortality (outcome collected through first event: death, hospital discharge, hospital transfer, or 120 days after birth).

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 9 BPD (requiring oxygen at 36 week's PMA for oxygen saturation > 90%).
Figuras y tablas -
Analysis 3.9

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 9 BPD (requiring oxygen at 36 week's PMA for oxygen saturation > 90%).

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 10 BPD or death by it prior to 37 weeks' PMA (outcomes collected through first event: death, hospital discharge, hospital transfer, or 120 days after birth).
Figuras y tablas -
Analysis 3.10

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 10 BPD or death by it prior to 37 weeks' PMA (outcomes collected through first event: death, hospital discharge, hospital transfer, or 120 days after birth).

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 11 Severe IVH (grade 3 or 4).
Figuras y tablas -
Analysis 3.11

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 11 Severe IVH (grade 3 or 4).

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 12 Cystic areas in the cerebral parenchyma measured through 28 d.
Figuras y tablas -
Analysis 3.12

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 12 Cystic areas in the cerebral parenchyma measured through 28 d.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 13 Early onset sepsis.
Figuras y tablas -
Analysis 3.13

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 13 Early onset sepsis.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 14 Late onset sepsis (> 72 hrs of age).
Figuras y tablas -
Analysis 3.14

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 14 Late onset sepsis (> 72 hrs of age).

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 15 Suspected or proven NEC.
Figuras y tablas -
Analysis 3.15

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 15 Suspected or proven NEC.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 16 Surgical NEC.
Figuras y tablas -
Analysis 3.16

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 16 Surgical NEC.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 17 Spontaneous gastro‐intestinal perforation.
Figuras y tablas -
Analysis 3.17

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 17 Spontaneous gastro‐intestinal perforation.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 18 Pulmonary haemorrhage.
Figuras y tablas -
Analysis 3.18

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 18 Pulmonary haemorrhage.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 19 PDA.
Figuras y tablas -
Analysis 3.19

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 19 PDA.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 20 PDA requiring indomethacin.
Figuras y tablas -
Analysis 3.20

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 20 PDA requiring indomethacin.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 21 PDA requiring surgery.
Figuras y tablas -
Analysis 3.21

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 21 PDA requiring surgery.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 22 Seizure treatment for ≥ 2 days.
Figuras y tablas -
Analysis 3.22

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 22 Seizure treatment for ≥ 2 days.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 23 Negative hearing screening in either ear at discharge.
Figuras y tablas -
Analysis 3.23

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 23 Negative hearing screening in either ear at discharge.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 24 Respiratory distress syndrome.
Figuras y tablas -
Analysis 3.24

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 24 Respiratory distress syndrome.

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 25 Sepsis, necrotizing enterocolitis, pneumonia or other infection as a cause of death.
Figuras y tablas -
Analysis 3.25

Comparison 3 Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA, Outcome 25 Sepsis, necrotizing enterocolitis, pneumonia or other infection as a cause of death.

Summary of findings for the main comparison. Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) compared to control for preterm infants at risk for or having respiratory distress syndrome (Comparison 1)

Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) compared to control for preterm infants at risk for or having respiratory distress syndrome

Patient or population: preterm infants at risk for or having respiratory distress syndrome
Setting: NICU
Intervention: Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment)
Comparison: control

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with control

Risk with Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment)

Infant death (age < 1 year)

Study population

RR 0.89
(0.71 to 1.13)

1115
(5 studies)

Low

Design (risk of bias): the risk of bias for random sequence generation was low in 2 studies and unclear in 3 studies; the risk of bias for allocation concealment was low in 3 studies and unclear in 2 studies; the risk of bias regarding performance bias and detection bias was low in 3 studies and unclear in 2 studies. We downgraded the quality of the evidence by 1 step

Heterogeneity/consistency across studies: there was high heterogeneity for RR (I² = 80 % ) and for RD (I² = 84%). We downgraded the quality of the evidence by 1 step

Directness of the evidence: Studies were conducted in the target population.

Precision of estimates: Results from 1115 infants have been reported in the studies to date and the confidence intervals around the point estimates for RR and RD were narrow.

Presence of publication bias: As only 5 studies were included in the analysis we did not perform a funnel plot.

207 per 1000

184 per 1000
(147 to 234)

Bronchopulmonary dysplasia (at 36 to 38 weeks' PMA)

Study population

RR 1.04
(0.90 to 1.20)

737
(2 studies)

Moderate

Design (risk of bias): the risk of bias for random sequence generation was low in 1 study and unclear in 1 study; The risk of bias for allocation concealment was low in 1 study and unclear in 1 study; the risk of bias regarding performance bias and detection bias was low in 1 study and unclear in 1 study. We downgraded the quality of the evidence by 1 step

Heterogeneity/consistency across studies: there was no heterogeneity for RR (I² = 0%) and for RD (I² = 0%)

Directness of the evidence: studies were conducted in the target population

Precision of estimates: to date the results from 737 infants have been reported in the studies and the confidence intervals around the point estimates for RR and RD were narrow

Presence of publication bias: as only 2 studies were included in the analysis we did not perform a funnel plot

459 per 1000

477 per 1000
(413 to 551)

ROP, stage ≥ 3 or ≥ 2

Study population

RR 0.89
(0.75 to 1.06)

810
(3 studies)

Moderate

Design (risk of bias): the risk of bias for random sequence generation was low in 1 study and unclear in 2 studies; the risk of bias for allocation concealment was low in 1 study and unclear in 2 studies; the risk of bias regarding performance bias and detection bias was low in 1 study and unclear in 2 studies. We downgraded the quality of the evidence by 1 step

Heterogeneity/consistency across studies: there was moderate heterogeneity for RR (I² = 63% ) and none for RD (I² = 23%)

Directness of the evidence: Studies were conducted in the target population

Precision of estimates: to date the results from 810 infants have been reported in the studies and the confidence intervals around the point estimates for RR and RD were narrow

Presence of publication bias: as only 3 studies were included in the analysis we did not perform a funnel plot

368 per 1000

328 per 1000
(276 to 390)

Sepsis (early or late onset)

Study population

RR 1.21
(0.95 to 1.54)

1067
(4 studies)

Moderate

Design (risk of bias): the risk of bias for random sequence generation was low in 2 studies and unclear in 2 studies; the risk of bias for allocation concealment was low in 3 studies and unclear in 1 study; the risk of bias regarding performance bias and detection bias was low in 3 studies and unclear in 1 study. We downgraded the quality of the evidence by 1 step

Heterogeneity/consistency: across studies: There was no heterogeneity for RR (I² = 24% ) and low for RD (I² = 34%)

Directness of the evidence: studies were conducted in the target population

Precision of estimates: to date results from 1067 infants have been reported in the studies and the confidence intervals around the point estimates for RR and RD were narrow

Presence of publication bias: as only 4 studies were included in the analysis we did not perform a funnel plot

189 per 1000

229 per 1000
(180 to 292)

Necrotizing enterocolitis (suspected or proven)

Study population

RR 0.94
(0.64 to 1.39)

1115
(5 studies)

Moderate

Design (risk of bias): the risk of bias for random sequence generation was low in 2 studies and unclear in 3 studies; the risk of bias for allocation concealment was low in 3 studies and unclear in 2 studies; the risk of bias regarding performance bias and detection bias was low in 3 studies and unclear in 2 studies. We downgraded the quality of the evidence by 1 step

Heterogeneity/consistency across studies: there was no heterogeneity for RR (I² = 0%) nor for RD (I² = 0%)

Directness of the evidence: studies were conducted in the target population

Precision of estimates: to date results from 1115 infants have been reported in the studies and the confidence intervals around the point estimates for RR and RD were narrow

Presence of publication bias: as only 5 studies were included in the analysis we did not perform a funnel plot

83 per 1000

78 per 1000
(53 to 115)

Intraventricular haemorrhage, grade > 2

Study population

RR 0.77
(0.58 to 1.01)

1103
(5 studies)

Moderate

Design (risk of bias): the risk of bias for random sequence generation was low in 2 studies and unclear in 3 studies; the risk of bias for allocation concealment was low in 3 studies and unclear in 2 studies; the risk of bias regarding performance bias and detection bias was low in 3 studies and unclear in 2 studies. We downgraded the quality of the evidence by 1 step

Heterogeneity/consistency across studies: there was low heterogeneity for RR (I² = 48% ) and for RD (I² = 42%)

Directness of the evidence: studies were conducted in the target population

Precision of estimates: to date the results from 1103 infants have been reported in the studies and the confidence intervals around the point estimates for RR and RD were narrow

Presence of publication bias: As only 5 studies were included in the analysis we did not perform a funnel plot

177 per 1000

136 per 1000
(103 to 179)

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: Confidence interval;PMA: Postmenstrual age; RD: Risk difference; ROP: Retinopathy of Prematurity; RR: Risk ratio;

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect.

Figuras y tablas -
Summary of findings for the main comparison. Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) compared to control for preterm infants at risk for or having respiratory distress syndrome (Comparison 1)
Summary of findings 2. Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at less than 30 weeks' PMA compared to placebo for preterm infants at risk for or having respiratory distress syndrome (Comparison 3)

Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA compared to placebo for preterm infants at risk for or having respiratory distress syndrome

Patient or population: preterm infants at risk for or having respiratory distress syndrome
Setting:
Intervention: Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA
Comparison: placebo

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with placebo

Risk with Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA

Type 1 ROP or death before determination of ROP outcome using the adjudicated ROP outcome

Study population

RR 1.28
(0.99 to 1.67)

679
(2 studies)

Moderate

Design (risk of bias): the risk of bias for random sequence generation, for allocation concealment, for performance bias and detection bias was low in both studies

Heterogeneity/consistency across studies: there was high heterogeneity for RR (I² = 79 %) and for RD (I² = 85%). We downgraded the quality of the evidence by 1 step

Directness of the evidence: studies were conducted in the target population

Precision of estimates: this outcome was reported for 679 infants and the confidence intervals around the point estimates for RR and RD were narrow

Presence of publication bias: As only 2 studies were included in the analysis we did not perform a funnel plot

222 per 1000

284 per 1000
(220 to 371)

Type 1 ROP including adjudicated ROP outcome

Study population

RR 1.24
(0.82 to

1.86)

605
(2 studies)

Moderate

Design (risk of bias): the risk of bias for random sequence generation, for allocation concealment, for performance bias and detection bias was low in both studies

Heterogeneity/consistency across studies: there was low heterogeneity for RR (I² = 46 %) and moderate for RD (I² = 54%). We downgraded the quality of the evidence by 1 step

Directness of the evidence: studies were conducted in the target population

Precision of estimates: this outcome was reported on for 605 infants and the confidence intervals around the point estimates for RR and RD were narrow

Presence of publication bias: as only 2 studies were included in the analysis we did not perform a funnel plot

120 per 1000

149 per 1000
(99 to 224)

All‐cause mortality (outcome collected through first event: death, hospital discharge, hospital transfer, or 120 days after birth)

Study population

RR 1.35
(0.91 to

2.00)

701
(2 studies)

Moderate

Design (risk of bias): the risk of bias for random sequence generation, for allocation concealment, for performance bias and detection bias was low in both studies

Heterogeneity/consistency across studies: there was moderate heterogeneity for RR (I² = 72%) and high for RD (I² = 84%). We downgraded the quality of the evidence by 1 step

Directness of the evidence: studies were conducted in the target population

Precision of estimates: this outcome was reported for 701 infants and the confidence intervals around the point estimates for RR and RD were narrow

Presence of publication bias: as only 2 studies were included in the analysis we did not perform a funnel plot

110 per 1000

148 per 1000
(100 to 219)

BPD or death by it prior to 37 weeks' PMA (outcomes collected through first event: death, hospital discharge, hospital transfer, or 120 days after birth)

Study population

RR 1.01
(0.87 to

1.16)

616
(2 studies)

High

Design (risk of bias): the risk of bias for random sequence generation, for allocation concealment, for performance bias and detection bias was low in both studies

Heterogeneity/consistency across studies: there was no heterogeneity for RR (I² = 0%) nor for RD (I² = 0%)

Directness of the evidence: studies were conducted in the target population.

Precision of estimates: this outcome was reported for 616 infants and the confidence intervals around the point estimates for RR and RD were narrow

Presence of publication bias: as only 2 studies were included in the analysis we did not perform a funnel plot

555 per 1000

561 per 1000
(483 to 644)

Severe IVH (grade 3 or 4)

Study population

RR 0.92
(0.65 to

1.29)

690
(2 studies)

Moderate

Design (risk of bias): The risk of bias for random sequence generation, for allocation concealment, for performance bias and detection bias was low in both studies

Heterogeneity/consistency across studies: there was moderate heterogeneity for RR (I² = 74%) and high for RD (I² = 82%)

Directness of the evidence: studies were conducted in the target population

Precision of estimates: this outcome was reported for 690 infants and the confidence intervals around the point estimates for RR and RD were narrow

Presence of publication bias: as only 2 studies were included in the analysis we did not perform a funnel plot.

171 per 1000

157 per 1000
(111 to 221)

Late‐onset sepsis (> 72 hours of age)

Study population

RR 1.33
(1.00 to

1.75)

701
(2 studies)

HIgh

Design (risk of bias): the risk of bias for random sequence generation, for allocation concealment, for performance bias and detection bias was low in both studies

Heterogeneity/consistency across studies: there was no heterogeneity for RR (I² = 0%) nor for RD (I² = 0%)

Directness of the evidence: studies were conducted in the target population

Precision of estimates: this outcome was reported for 701 infants and the confidence intervals around the point estimates for RR and RD were narrow

Presence of publication bias: as only 2 studies were included in the analysis we did not perform a funnel plot

191 per 1000

254 per 1000
(191 to 334)

Suspected or proven NEC

Study population

RR 0.88
(0.55 to

1.41)

701
(2 studies)

High

Design (risk of bias): the risk of bias for random sequence generation, for allocation concealment, for performance bias and detection bias was low in both studies

Heterogeneity/consistency across studies: there was low heterogeneity for RR (I² = 36%) and moderate for RD (I² = 53%).

Directness of the evidence: studies were conducted in the target population.

Precision of estimates: this outcome was reported on in 701 infants and the confidence intervals around the point estimates for RR and RD were narrow

Presence of publication bias: as only 2 studies were included in the analysis we did not perform a funnel plot.

98 per 1000

87 per 1000
(54 to 139)

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: Confidence interval; PMA: Postmenstrual age; RD: Risk difference; ROP: Retinopathy of prematurity; RR: Risk ratio

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: We are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low certainty: Our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

Figuras y tablas -
Summary of findings 2. Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at less than 30 weeks' PMA compared to placebo for preterm infants at risk for or having respiratory distress syndrome (Comparison 3)
Comparison 1. Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Neonatal death (age < 28 days) Show forest plot

3

355

Risk Ratio (M‐H, Fixed, 95% CI)

0.53 [0.31, 0.91]

2 Infant death (age < one year) Show forest plot

5

1115

Risk Ratio (M‐H, Fixed, 95% CI)

0.89 [0.71, 1.13]

3 BPD (supplementary oxygen ar 36 weeks; PMA or death due to BPD)at 36 week's PMA Show forest plot

2

666

Risk Ratio (M‐H, Fixed, 95% CI)

1.00 [0.87, 1.14]

4 Bronchopulmonary dysplasia (at 28 to 30 days of age) Show forest plot

3

343

Risk Ratio (M‐H, Fixed, 95% CI)

0.78 [0.54, 1.13]

5 Bronchopulmonary dysplasia (at 36 to 38 weeks PMA) Show forest plot

2

737

Risk Ratio (M‐H, Fixed, 95% CI)

1.04 [0.90, 1.20]

6 Retinopathy of prematurity, stage ≥ 3 or ≥ 2 Show forest plot

3

810

Risk Ratio (M‐H, Fixed, 95% CI)

0.89 [0.75, 1.06]

6.1 ROP ≥ 3

2

262

Risk Ratio (M‐H, Fixed, 95% CI)

0.09 [0.01, 0.67]

6.2 Retinopathy of prematurity, stage ≥ 2

1

548

Risk Ratio (M‐H, Fixed, 95% CI)

0.95 [0.80, 1.13]

7 Retinopathy of prematurity, any stage Show forest plot

4

889

Risk Ratio (M‐H, Fixed, 95% CI)

0.94 [0.83, 1.07]

8 Necrotizing enterocolitis (suspected or proven) Show forest plot

5

1115

Risk Ratio (M‐H, Fixed, 95% CI)

0.94 [0.64, 1.39]

9 Sepsis (early and/or late onset) Show forest plot

4

1067

Risk Ratio (M‐H, Fixed, 95% CI)

1.21 [0.95, 1.54]

10 Intraventricular haemorrhage, grade > 2 Show forest plot

5

1103

Risk Ratio (M‐H, Fixed, 95% CI)

0.77 [0.58, 1.01]

11 Intraventricular haemorrhage, all grades Show forest plot

3

427

Risk Ratio (M‐H, Fixed, 95% CI)

0.77 [0.59, 1.00]

12 Minor neural developmental impairment at one year corrected age Show forest plot

1

169

Risk Ratio (M‐H, Fixed, 95% CI)

0.84 [0.38, 1.86]

13 Major neural developmental impairment at one year corrected age Show forest plot

1

169

Risk Ratio (M‐H, Fixed, 95% CI)

0.53 [0.24, 1.16]

Figuras y tablas -
Comparison 1. Inositol supplementation to preterm infants (repeat doses in any amount and any duration of treatment) versus control
Comparison 2. Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Death during hospital stay Show forest plot

1

74

Risk Ratio (M‐H, Fixed, 95% CI)

1.19 [0.34, 4.21]

2 Bronchopulmonary dysplasia at 36 weeks PMA Show forest plot

1

65

Risk Ratio (M‐H, Fixed, 95% CI)

2.74 [0.88, 8.48]

3 Retinopathy of prematurity (infants who underwent surgery for ROP) Show forest plot

1

25

Risk Ratio (M‐H, Fixed, 95% CI)

0.35 [0.10, 1.22]

4 Necrotizing enterocolitis (stage 2A or worse) Show forest plot

1

74

Risk Ratio (M‐H, Fixed, 95% CI)

0.41 [0.12, 1.39]

5 Necrotizing enterocolitis (infants who underwent surgery for NEC) Show forest plot

1

74

Risk Ratio (M‐H, Fixed, 95% CI)

0.51 [0.08, 3.41]

6 Sepsis (late onset) Show forest plot

1

74

Risk Ratio (M‐H, Fixed, 95% CI)

1.46 [0.71, 2.97]

7 Intraventricular haemorrhage (grade 3 or 4) Show forest plot

1

72

Risk Ratio (M‐H, Fixed, 95% CI)

1.06 [0.29, 3.90]

8 Hearing test (failed both ears) Show forest plot

1

57

Risk Ratio (M‐H, Fixed, 95% CI)

0.58 [0.09, 3.84]

Figuras y tablas -
Comparison 2. Inositol supplementation (single dose of 60 mg/kg or 120 mg/kg) in preterm infants
Comparison 3. Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Type 1 ROP or death before determination of ROP outcome using the adjudicated ROP outcome Show forest plot

2

679

Risk Ratio (M‐H, Fixed, 95% CI)

1.28 [0.99, 1.67]

2 Type 1 ROP Show forest plot

1

511

Risk Ratio (M‐H, Fixed, 95% CI)

1.41 [0.89, 2.24]

3 Death before determination of ROP outcome Show forest plot

1

638

Risk Ratio (M‐H, Fixed, 95% CI)

1.53 [1.02, 2.31]

4 Type 1 ROP including adjudicated ROP outcome Show forest plot

2

605

Risk Ratio (M‐H, Fixed, 95% CI)

1.24 [0.82, 1.86]

5 Any ROP Show forest plot

1

553

Risk Ratio (M‐H, Fixed, 95% CI)

1.00 [0.88, 1.13]

6 ROP ≥ 2 ROP Show forest plot

1

548

Risk Ratio (M‐H, Fixed, 95% CI)

0.95 [0.80, 1.13]

7 All cause infant mortality to 55 week's PMA Show forest plot

1

638

Risk Ratio (M‐H, Fixed, 95% CI)

1.67 [1.12, 2.48]

8 All cause mortality (outcome collected through first event: death, hospital discharge, hospital transfer, or 120 days after birth) Show forest plot

2

701

Risk Ratio (M‐H, Fixed, 95% CI)

1.35 [0.91, 2.00]

9 BPD (requiring oxygen at 36 week's PMA for oxygen saturation > 90%) Show forest plot

1

560

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.89, 1.18]

10 BPD or death by it prior to 37 weeks' PMA (outcomes collected through first event: death, hospital discharge, hospital transfer, or 120 days after birth) Show forest plot

2

616

Risk Ratio (M‐H, Fixed, 95% CI)

1.01 [0.87, 1.16]

11 Severe IVH (grade 3 or 4) Show forest plot

2

690

Risk Ratio (M‐H, Fixed, 95% CI)

0.92 [0.65, 1.29]

12 Cystic areas in the cerebral parenchyma measured through 28 d Show forest plot

2

225

Risk Ratio (M‐H, Fixed, 95% CI)

1.29 [0.58, 2.85]

13 Early onset sepsis Show forest plot

1

63

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14 Late onset sepsis (> 72 hrs of age) Show forest plot

2

701

Risk Ratio (M‐H, Fixed, 95% CI)

1.33 [1.00, 1.75]

15 Suspected or proven NEC Show forest plot

2

701

Risk Ratio (M‐H, Fixed, 95% CI)

0.88 [0.55, 1.41]

16 Surgical NEC Show forest plot

2

701

Risk Ratio (M‐H, Fixed, 95% CI)

1.21 [0.57, 2.58]

17 Spontaneous gastro‐intestinal perforation Show forest plot

2

701

Risk Ratio (M‐H, Fixed, 95% CI)

0.86 [0.48, 1.52]

18 Pulmonary haemorrhage Show forest plot

1

638

Risk Ratio (M‐H, Fixed, 95% CI)

0.98 [0.59, 1.62]

19 PDA Show forest plot

2

700

Risk Ratio (M‐H, Fixed, 95% CI)

0.98 [0.85, 1.14]

20 PDA requiring indomethacin Show forest plot

1

637

Risk Ratio (M‐H, Fixed, 95% CI)

0.90 [0.67, 1.22]

21 PDA requiring surgery Show forest plot

2

700

Risk Ratio (M‐H, Fixed, 95% CI)

0.96 [0.65, 1.42]

22 Seizure treatment for ≥ 2 days Show forest plot

2

700

Risk Ratio (M‐H, Fixed, 95% CI)

1.04 [0.43, 2.56]

23 Negative hearing screening in either ear at discharge Show forest plot

2

472

Risk Ratio (M‐H, Fixed, 95% CI)

1.45 [0.92, 2.29]

24 Respiratory distress syndrome Show forest plot

1

63

Risk Ratio (M‐H, Fixed, 95% CI)

0.99 [0.91, 1.09]

25 Sepsis, necrotizing enterocolitis, pneumonia or other infection as a cause of death Show forest plot

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

1.36 [0.95, 1.93]

Figuras y tablas -
Comparison 3. Inositol supplementation IV initially followed by enteral administration (repeat doses of 80 mg/kg/day) in preterm infants born at < 30 weeks' PMA