1
|
Unal S, Beken S, Anuk Ince D, Turan O, Korkmaz Toygar A, Ecevit A, Akcan AB, Akın MA, Aktas S, Ciftdemir NA, Altuncu E, Altunhan H, Arcagok BC, Armangil D, Arun Ozer E, Aydın B, Bezirganoglu H, Bilgin L, Calısıcı E, Calkavur S, Celik K, Celik Y, Cetinkaya B, Cetinkaya M, Demirel A, Demirel G, Dogan NN, Doğan P, Durukan M, Engur D, Erener Ercan T, Gokmen Z, Guney Varal I, Gulası S, Gunlemez A, Gursoy T, Hakyemez Toptan H, Hamitoğlu S, Isleyen F, Iyigun I, Kader S, Kahvecioğlu D, Kaykı G, Kostu M, Kurnaz D, Mammadalıyev T, Mungan Akin I, Narlı N, Okulu E, Okur N, Olukman O, Ovalı F, Ozcan B, Ozdemir A, Ozdemir O, Ozkan H, Sandal G, Sarıcı D, Sivrikaya C, Siyah Bilgin B, Sundus S, Surmeli Onay O, Simsek H, Tandırcıoğlu UA, Tanrıverdi S, Tekgunduz KS, Terek D, Tunc G, Tunc T, Tutak E, Tufekcioğlu E, Tuzun Erdogan F, Ulu E, Ulubas Isik D, Uras N, Uslu SI, Unal I, Yılmaz FH, Moniri A. Caffeine use in preterm neonates: national insights into Turkish NICU practices. Front Pediatr 2025; 13:1492716. [PMID: 40083438 PMCID: PMC11905296 DOI: 10.3389/fped.2025.1492716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 01/28/2025] [Indexed: 03/16/2025] Open
Abstract
Objective Caffeine is a proven medication used for the prevention and treatment of apnea in premature infants, offering both short- and long-term benefits. International guidelines provide a range of recommendations regarding the preterm population eligible for caffeine prophylaxis, including the timing, dosage, and duration of treatment. Our national guidelines, published prior to the most recent updates of the international guidelines, recommend the use of caffeine citrate starting from the first day after delivery for preterm infants with a gestational age of <28 weeks. For infants up to 32 weeks, if positive pressure ventilation is required, the decision should be made on an individual basis. This study aims to describe the variability in caffeine usage across neonatal intensive care units in our country. Methods An online survey was sent to neonatologist who are members of the Turkish Neonatology Society to describe the variability in caffeine usage in neonatal intensive care units in our country. Results We collected responses from 74 units. Prophylactic caffeine usage was observed as; GA ≤276/7: 98.6%, GA 280/7-286/7: 89.0%, GA 290/7-296/7: 75.3%, GA 300/7-316/7: 53.4%. 62.2% of units reported administering loading dose within the first two hours. The initial maintenance dose was 5 mg/kg in 64.8% of units, 10 mg/kg in 32.4% of units, and intermediate dose in 5.3% of units. 47.3% of units reported no routine dose adjustment. The postmenstrual age that caffeine treatment was stopped was found to be 34 (min-max; 32-36) weeks for infants without apnea and respiratory support, 36 (min-max; 34-52) weeks for infants without apnea but any respiratory support. The time to discharge after treatment cessation was found as; 1-4 days: 37.8%, 5-7 days: 68.9%. Among the 56 units with multiple responsible physicians, 32.1% reported intra-unit variations. Conclusion The significant differences in caffeine usage characteristics between and within units highlight the need for clear recommendations provided by standardized guidelines.
Collapse
Affiliation(s)
- Sezin Unal
- Division of Neonatology, Department of Pediatrics, Baskent University Faculty of Medicine, Ankara, Türkiye
| | - Serdar Beken
- Division of Neonatology, Department of Pediatrics, Acıbadem Mehmet Ali Aydınlar University Faculty of Medicine, Atakent Hospital, İstanbul, Türkiye
| | - Deniz Anuk Ince
- Division of Neonatology, Department of Pediatrics, Baskent University Faculty of Medicine, Ankara, Türkiye
| | - Ozden Turan
- Division of Neonatology, Department of Pediatrics, Baskent University Faculty of Medicine, Ankara, Türkiye
| | - Ayse Korkmaz Toygar
- Division of Neonatology, Department of Pediatrics, Acıbadem Mehmet Ali Aydınlar University Faculty of Medicine, Atakent Hospital, İstanbul, Türkiye
| | - Ayse Ecevit
- Division of Neonatology, Department of Pediatrics, Baskent University Faculty of Medicine, Ankara, Türkiye
| | - Abdullah Baris Akcan
- Division of Neonatology, Department of Pediatrics, Aydın Adnan Menderes University Faculty of Medicine, Aydın, Türkiye
| | - Mustafa Ali Akın
- Division of Neonatology, Department of Pediatrics, On Dokuz Mayıs University Faculty of Medicine, Samsun, Türkiye
| | - Selma Aktas
- Division of Neonatology, Department of Pediatrics, Acıbadem Mehmet Ali Aydınlar University Faculty of Medicine, Maslak Hospital, İstanbul, Türkiye
| | - Nukhet Aladag Ciftdemir
- Division of Neonatology, Department of Pediatrics, Trakya University Faculty of Medicine, Edirne, Türkiye
| | - Emel Altuncu
- Division of Neonatology, Department of Pediatrics, University of Health Sciences, Kartal Dr Lutfi Kırdar City Hospital, İstanbul, Türkiye
| | - Huseyin Altunhan
- Division of Neonatology, Department of Pediatrics, Necmettin Erbakan University Faculty of Medicine, Konya, Türkiye
| | - Baran Cengiz Arcagok
- Division of Neonatology, Department of Pediatrics, Acıbadem Mehmet Ali Aydınlar University Faculty of Medicine, Altunizade Hospital, İstanbul, Türkiye
| | - Didem Armangil
- Division of Neonatology, Department of Pediatrics, Koru Ankara Hospital, Ankara, Türkiye
| | - Esra Arun Ozer
- Division of Neonatology, Department of Pediatrics, İzmir Tınaztepe University Faculty of Medicine, İzmir, Türkiye
| | - Banu Aydın
- Division of Neonatology, Department of Pediatrics, Lokman Hekim University Faculty of Medicine, Ankara, Türkiye
| | - Handan Bezirganoglu
- Division of Neonatology, Department of Pediatrics, Trabzon Kanuni Training and Research Hospital, Trabzon, Türkiye
| | - Leyla Bilgin
- Division of Neonatology, Department of Pediatrics, İstanbul University İstanbul Faculty of Medicine, İstanbul, Türkiye
| | - Erhan Calısıcı
- Division of Neonatology, Department of Pediatrics, University of Health Sciences Gulhane Faculty of Medicine, Gulhane Training and Research Hospital, Ankara, Türkiye
| | - Sebnem Calkavur
- Division of Neonatology, Department of Pediatrics, University of Health Sciences İzmir Dr. Behcet Uz Pediatric and Pediatric Surgery Training and Research Hospital, İzmir, Türkiye
| | - Kıymet Celik
- Division of Neonatology, Department of Pediatrics, Akdeniz University Faculty of Medicine, Antalya, Türkiye
| | - Yalcın Celik
- Division of Neonatology, Department of Pediatrics, Mersin University Faculty of Medicine, Mersin, Türkiye
| | - Bilin Cetinkaya
- Division of Neonatology, Department of Pediatrics, Baskent University Faculty of Medicine, Adana Dr. Turgut Noyan Training and Research Center, Adana, Türkiye
| | - Merih Cetinkaya
- Division of Neonatology, Department of Pediatrics, University of Health Sciences, Basaksehir Cam Sakura City Hospital, İstanbul, Türkiye
| | - Atalay Demirel
- Division of Neonatology, Department of Pediatrics, Acıbadem Mehmet Ali Aydınlar University Faculty of Medicine, Acıbadem Kadıkoy Sinasi Can Hospital, İstanbul, Türkiye
| | - Gamze Demirel
- Division of Neonatology, Department of Pediatrics, Koc University Faculty of Medicine, Amerikan Hospital, İstanbul, Türkiye
| | - Nazan Neslihan Dogan
- Division of Neonatology, Department of Pediatrics, University of Health Sciences Bakırkoy Dr. Sadi Konuk Training and Research Hospital, İstanbul, Türkiye
| | - Pelin Doğan
- Division of Neonatology, Department of Pediatrics, Acıbadem Mehmet Ali Aydınlar University Faculty of Medicine, Acıbadem Atasehir Hospital, İstanbul, Türkiye
| | - Mehtap Durukan
- Division of Neonatology, Department of Pediatrics, Mardin Training and Research Hospital, Mardin, Türkiye
| | - Defne Engur
- Division of Neonatology, Department of Pediatrics, University of Health Sciences Tepecik Training and Research Hospital, İzmir, Türkiye
| | - Tugba Erener Ercan
- Division of Neonatology, Department of Pediatrics, Maltepe University Faculty of Medicine, İstanbul, Türkiye
| | - Zeynel Gokmen
- Division of Neonatology, Department of Pediatrics, Baskent University Faculty of Medicine, Konya Training and Research Hospital, Konya, Türkiye
| | - Ipek Guney Varal
- Division of Neonatology, Department of Pediatrics, University of Health Sciences Bursa Yuksek İhtisas Training and Research Hospital, Bursa, Türkiye
| | - Selvi Gulası
- Division of Neonatology, Department of Pediatrics, University of Health Sciences Adana Sehir Hospital, Adana, Türkiye
| | - Ayla Gunlemez
- Division of Neonatology, Department of Pediatrics, Kocaeli University Faculty of Medicine, Kocaeli, Türkiye
| | - Tugba Gursoy
- Division of Neonatology, Department of Pediatrics, Koc University Faculty of Medicine, Koc University Hospital, İstanbul, Türkiye
| | - Handan Hakyemez Toptan
- Division of Neonatology, Department of Pediatrics, Marmara University Faculty of Medicine Pendik Training and Research Hospital, İstanbul, Türkiye
| | - Serif Hamitoğlu
- Division of Neonatology, Department of Pediatrics, Bursa Medicana Hospital, Bursa, Türkiye
| | - Fatih Isleyen
- Division of Neonatology, Department of Pediatrics, Sanlıurfa Training and Research Hospital, Urfa, Türkiye
| | - Irem Iyigun
- Division of Neonatology, Department of Pediatrics, Ordu University Training and Research Hospital, Ordu, Türkiye
| | - Sebnem Kader
- Division of Neonatology, Department of Pediatrics, Kırklareli Training and Research Hospital, Kırlareli, Türkiye
| | - Dilek Kahvecioğlu
- Division of Neonatology, Department of Pediatrics, University of Health Sciences Ankara Training and Research Hospital, Ankara, Türkiye
| | - Gozdem Kaykı
- Division of Neonatology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Türkiye
| | - Murat Kostu
- Division of Neonatology, Department of Pediatrics, University of Health Sciences İstanbul Kanuni Sultan Suleyman Training and Research Hospital, İstanbul, Türkiye
| | - Dilek Kurnaz
- Division of Neonatology, Department of Pediatrics, İstanbul Haseki Training and Research Hospital, İstanbul, Türkiye
| | - Tural Mammadalıyev
- Division of Neonatology, Department of Pediatrics, Gazı University Faculty of Medicine, Ankara, Türkiye
| | - Ilke Mungan Akin
- Division of Neonatology, Department of Pediatrics, University of Health Sciences Umraniye Training and Research Hospital, İstanbul, Türkiye
| | - Nejat Narlı
- Division of Neonatology, Department of Pediatrics, Cukurova University Faculty of Medicine, Adana, Türkiye
| | - Emel Okulu
- Division of Neonatology, Department of Pediatrics, Ankara University Faculty of Medicine, Ankara, Türkiye
| | - Nilufer Okur
- Division of Neonatology, Department of Pediatrics, University of Health Sciences Gazi Yasargil Training and Research Hospital, Diyarbakır, Türkiye
| | - Ozgur Olukman
- Division of Neonatology, Department of Pediatrics, İzmir BakırCay University Faculty of Medicine, Ciğli Training and Research Hospital, İzmir, Türkiye
| | - Fahri Ovalı
- Division of Neonatology, Department of Pediatrics, İstanbul Medeniyet University Goztepe Prof. Dr. Suleyman YalCın City Hospital, İstanbul, Türkiye
| | - Beyza Ozcan
- Division of Neonatology, Department of Pediatrics, University of Health Sciences Konya City Hospital, Konya, Türkiye
| | - Ahmet Ozdemir
- Division of Neonatology, Department of Pediatrics, University of Health Sciences Kayseri City Hospital, Kayseri, Türkiye
| | - Ozmert Ozdemir
- Division of Neonatology, Department of Pediatrics, Pamukkale University Faculty of Medicine, Denizli, Türkiye
| | - Hilal Ozkan
- Division of Neonatology, Department of Pediatrics, Uludağ University Faculty of Medicine, Bursa, Türkiye
| | - Gonca Sandal
- Division of Neonatology, Department of Pediatrics, Medipol University Faculty of Medicine, Medipol Mega Hospital, İstanbul, Türkiye
| | - Dilek Sarıcı
- Division of Neonatology, Department of Pediatrics, University of Health Sciences Ankara Ataturk Sanatoryum Training and Research Hospital, Ankara, Türkiye
| | - Cansu Sivrikaya
- Division of Neonatology, Department of Pediatrics, Hatay Training and Research Hospital, Hatay, Türkiye
| | - Betul Siyah Bilgin
- Division of Neonatology, Department of Pediatrics, University of Health Sciences Ankara Bilkent Sehir Hospital, Ankara, Türkiye
| | - Saime Sundus
- Division of Neonatology, Department of Pediatrics, SelCuk University Faculty of Medicine, Konya, Türkiye
| | - Ozge Surmeli Onay
- Division of Neonatology, Department of Pediatrics, Eskisehir Osmangazi University Faculty of Medicine, Eskisehir, Türkiye
| | - Huseyin Simsek
- Division of Neonatology, Department of Pediatrics, University of Health Sciences Mersin City Hospital, Mersin, Türkiye
| | - Umit Ayse Tandırcıoğlu
- Division of Neonatology, Department of Pediatrics, Kırıkkale University Faculty of Medicine, Kırıkkale, Türkiye
| | - Sema Tanrıverdi
- Division of Neonatology, Department of Pediatrics, Manisa Celal Bayar University Faculty of Medicine, Manisa, Türkiye
| | - Kadir Serafettin Tekgunduz
- Division of Neonatology, Department of Pediatrics, Ataturk University Faculty of Medicine, Erzurum, Türkiye
| | - Demet Terek
- Division of Neonatology, Department of Pediatrics, Ege University Faculty of Medicine, İzmir, Türkiye
| | - Gaffari Tunc
- Division of Neonatology, Department of Pediatrics, Sivas Cumhuriyet University Faculty of Medicine, Sivas, Türkiye
| | - Turan Tunc
- Division of Neonatology, Department of Pediatrics, Memorial Atasehir Hospital, İstanbul, Türkiye
| | - Ercan Tutak
- Division of Neonatology, Department of Pediatrics, University of Health Sciences Prof. Dr. Cemil Tascıoğlu City Hospital, İstanbul, Türkiye
| | - Eda Tufekcioğlu
- Division of Neonatology, Department of Pediatrics, University of Health Sciences Ankara Etlik City Hospital, Ankara, Türkiye
| | - Funda Tuzun Erdogan
- Division of Neonatology, Department of Pediatrics, Dokuz Eylul University Faculty of Medicine, İzmir, Türkiye
| | - Ersin Ulu
- Division of Neonatology, Department of Pediatrics, İstanbul Universitesi- Cerrahpasa, Cerrahpasa Faculty of Medicine, İstanbul, Türkiye
| | - Dilek Ulubas Isik
- Division of Neonatology, Department of Pediatrics, University of Health Sciences Etlik Zubeyde Hanım Maternal Health Training and Research Hospital, Ankara, Türkiye
| | - Nurdan Uras
- Division of Neonatology, Department of Pediatrics, İstinye University Bahcesehir Liv Hospital, İstanbul, Türkiye
| | - Sait Ilker Uslu
- Division of Neonatology, Department of Pediatrics, Demiroğlu Bilim University İstanbul Florence Nightingale Hospital, İstanbul, Türkiye
| | - Irem Unal
- Division of Neonatology, Department of Pediatrics, Sehit Prof. Dr. İlhan Varank Sancaktepe Training and Research Hospital, İstanbul, Türkiye
| | - Fatma Hilal Yılmaz
- Division of Neonatology, Department of Pediatrics, Dr. Ali Kemal Belviranli Obstetrics and Gynecology Hospital, Konya, Türkiye
| | - Ariorad Moniri
- Department of Biostatistics, Acıbadem Mehmet Ali Aydınlar University Faculty of Medicine, İstanbul, Türkiye
| |
Collapse
|
2
|
Ohkuchi A, Okazaki K, Iwamoto S, Sako M, Kobayashi T, Yanagihara I, Nomiyama M. A phase II, multicenter, nonblinded, randomized controlled trial for evaluating protective effects of ABPC/SBT plus, azithromycin versus erythromycin, in pregnant women with pPROM occurring at <28 weeks of gestation on the development of BPD in neonates: Study protocol. PLoS One 2024; 19:e0304705. [PMID: 38980858 PMCID: PMC11232965 DOI: 10.1371/journal.pone.0304705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 05/13/2024] [Indexed: 07/11/2024] Open
Abstract
This is a protocol for PPROM-AZM Study, phase II, nonblinded, randomized controlled trial. Bronchopulmonary dysplasia (BPD) at a postmenstrual age of 36 weeks (BPD36) is often observed in infants with preterm premature rupture of the membranes (pPROM). A regimen of ampicillin (ABPC) intravenous infusion for 2 days and subsequent amoxicillin (AMPC) oral administration for 5 days plus erythromycin (EM) intravenous infusion for 2 days followed by EM oral administration for 5 days is standard treatment for pPROM. However, the effect on the prevention of moderate/severe BPD36 using the standard treatment has not been confirmed. Recently, it is reported that ampicillin/sulbactam (ABPC/SBT) plus azithromycin (AZM) was effective for the prevention of moderate/severe BPD36 in pPROM patients with amniotic infection of Ureaplasma species. Therefore, our aim is to evaluate the occurrence rate of the composite outcome of "incidence rate of either moderate/severe BPD36 or intrauterine fetal death, and infantile death at or less than 36 weeks 0 days" comparing subjects to receive ABPC/SBT for 14 days plus AZM for 14 days (intervention group) and those to receive ABPC/SBT for 14 days plus EM for 14 days (control group), in a total of 100 subjects (women with pPROM occurring at 22-27 weeks of gestation) in Japan. The recruit of subjects was started on April 2022, and collection in on-going. We also investigate the association between the detection of Ureaplasma species and occurrence of BPD36. In addition, information on any adverse events for the mother and fetus and serious adverse events for infants are collected during the observation period. We allocate patients at a rate of 1:1 considering two stratification factors: onset of pPROM (22-23 or 24-27 weeks) and presence/absence of a hospital policy for early neonatal administration of caffeine. Trial registration: The trial number in the Japan Registry of Clinical Trials is jRCTs031210631.
Collapse
Affiliation(s)
- Akihide Ohkuchi
- Maternal and Fetal Neonatal Intensive Care Unit, Jichi Medical University Hospital, Tochigi, Japan
| | - Kaoru Okazaki
- Department of Neonatology, Tokyo Metropolitan Children's Medical Center, Fuchu-shi, Tokyo, Japan
| | - Shintaro Iwamoto
- Division of Biostatistics, Department of Data Science, Clinical Research Center, National Center for Child Health and Development, Setagaya-ku, Tokyo, Japan
| | - Mayumi Sako
- Department of Clinical Research Promotion, Clinical Research Center, National Center for Child Health and Development, Setagaya-ku, Tokyo, Japan
| | - Tohru Kobayashi
- Department of Data Science, Clinical Research Center, National Center for Child Health and Development, Setagaya-ku, Tokyo, Japan
| | - Itaru Yanagihara
- Department of Developmental Medicine, Research Institute, Osaka Women's and Children's Hospital, Izumi-shi, Osaka, Japan
| | - Makoto Nomiyama
- Department of Obstetrics and Gynecology, National Hospital Organization Saga Hospital, Saga-shi, Saga, Japan
| |
Collapse
|
3
|
Yang K, Liu J, He T, Dong W. Caffeine and neonatal acute kidney injury. Pediatr Nephrol 2024; 39:1355-1367. [PMID: 37665410 DOI: 10.1007/s00467-023-06122-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023]
Abstract
Acute kidney injury is one of the most threatening diseases in neonates, with complex pathogenesis and limited treatment options. Caffeine is a commonly used central nervous system stimulant for treating apnea in preterm infants. There is compelling evidence that caffeine may have potential benefits for preventing neonatal acute kidney injury, but comprehensive reports are lacking in this area. Hence, this review aims to provide a summary of clinical data on the potential benefits of caffeine in improving neonatal acute kidney injury. Additionally, it delves into the molecular mechanisms underlying caffeine's effects on acute kidney injury, with a focus on various aspects such as oxidative stress, adenosine receptors, mitochondrial dysfunction, endoplasmic reticulum stress, inflammasome, autophagy, p53, and gut microbiota. The ultimate goal of this review is to provide information for healthcare professionals regarding the link between caffeine and neonatal acute kidney injury and to identify gaps in our current understanding.
Collapse
Affiliation(s)
- Kun Yang
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, 646000, China
| | - Jinjing Liu
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, 646000, China
| | - Ting He
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, 646000, China
| | - Wenbin Dong
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
- Sichuan Clinical Research Center for Birth Defects, Luzhou, 646000, China.
| |
Collapse
|
4
|
Sanlorenzo LA, Hatch LD. Developing a Respiratory Quality Improvement Program to Prevent and Treat Bronchopulmonary Dysplasia in the Neonatal Intensive Care Unit. Clin Perinatol 2023; 50:363-380. [PMID: 37201986 DOI: 10.1016/j.clp.2023.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Improvements in respiratory care have resulted in improved outcomes for preterm infants over the past three decades. To target the multifactorial nature of neonatal lung diseases, neonatal intensive care units (NICUs) should consider developing comprehensive respiratory quality improvement programs that address all drivers of neonatal respiratory disease. This article presents a potential framework for developing a quality improvement program to prevent bronchopulmonary dysplasia in the NICU. Drawing on available research and quality improvement reports, the authors discuss key components, measures, drivers, and interventions that should be considered when building a respiratory quality improvement program devoted to preventing and treating bronchopulmonary dysplasia.
Collapse
Affiliation(s)
- Lauren A Sanlorenzo
- Department of Pediatrics, Division of Neonatology, Columbia University Medical Center, 3959 Broadway Avenue, New York, NY 10032, USA
| | - Leon Dupree Hatch
- Department of Pediatrics, Division of Neonatology, Vanderbilt University Medical Center, 4413 VCH, 2200 Children's Way, Nashville, TN 37232, USA; Center for Child Health Policy, Vanderbilt University Medical Center, Nashville, TN, USA; Critical Illness, Brain Dysfunction, and Survivorship Center, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
5
|
Bruschettini M, Brattström P, Russo C, Onland W, Davis PG, Soll R. Caffeine dosing regimens in preterm infants with or at risk for apnea of prematurity. Cochrane Database Syst Rev 2023; 4:CD013873. [PMID: 37040532 PMCID: PMC10089673 DOI: 10.1002/14651858.cd013873.pub2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
BACKGROUND Very preterm infants often require respiratory support and are therefore exposed to an increased risk of bronchopulmonary dysplasia (chronic lung disease) and later neurodevelopmental disability. Caffeine is widely used to prevent and treat apnea (temporal cessation of breathing) associated with prematurity and facilitate extubation. Though widely recognized dosage regimes have been used for decades, higher doses have been suggested to further improve neonatal outcomes. However, observational studies suggest that higher doses may be associated with harm. OBJECTIVES To determine the effects of higher versus standard doses of caffeine on mortality and major neurodevelopmental disability in preterm infants with (or at risk of) apnea, or peri-extubation. SEARCH METHODS We searched CENTRAL, MEDLINE, Embase, CINAHL, the World Health Organization (WHO) International Clinical Trials Registry Platform (ICTRP), and clinicaltrials.gov in May 2022. The reference lists of relevant articles were also checked to identify additional studies. SELECTION CRITERIA We included randomized (RCTs), quasi-RCTs and cluster-RCTs, comparing high-dose to standard-dose strategies in preterm infants. High-dose strategies were defined as a high-loading dose (more than 20 mg of caffeine citrate/kg) or a high-maintenance dose (more than 10 mg of caffeine citrate/kg/day). Standard-dose strategies were defined as a standard-loading dose (20 mg or less of caffeine citrate/kg) or a standard-maintenance dose (10 mg or less of caffeine citrate/kg/day). We specified three additional comparisons according to the indication for commencing caffeine: 1) prevention trials, i.e. preterm infants born at less than 34 weeks' gestation, who are at risk for apnea; 2) treatment trials, i.e. preterm infants born at less than 37 weeks' gestation, with signs of apnea; 3) extubation trials: preterm infants born at less than 34 weeks' gestation, prior to planned extubation. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane. We evaluated treatment effects using a fixed-effect model with risk ratio (RR) for categorical data and mean, standard deviation (SD), and mean difference (MD) for continuous data. MAIN RESULTS: We included seven trials enrolling 894 very preterm infants (reported in Comparison 1, i.e. any indication). Two studies included infants for apnea prevention (Comparison 2), four studies for apnea treatment (Comparison 3) and two for extubation management (Comparison 4); in one study, indication for caffeine administration was both apnea treatment and extubation management (reported in Comparison 1, Comparison 3 and Comparison 4). In the high-dose groups, loading and maintenance caffeine doses ranged from 30 mg/kg to 80 mg/kg, and 12 mg/kg to 30 mg/kg, respectively; in the standard-dose groups, loading and maintenance caffeine doses ranged from 6 mg/kg to 25 mg/kg, and 3 mg/kg to 10 mg/kg, respectively. Two studies had three study groups: infants were randomized in three different doses (two of them matched our definition of high dose and one matched our definition of standard dose); high-dose caffeine and standard-dose caffeine were compared to theophylline administration (the latter is included in a separate review). Six of the seven included studies compared high-loading and high-maintenance dose to standard-loading and standard-maintenance dose, whereas in one study standard-loading dose and high-maintenance dose was compared to standard-loading dose and standard-maintenance dose. High-dose caffeine strategies (administration for any indication) may have little or no effect on mortality prior to hospital discharge (risk ratio (RR) 0.86, 95% confidence of interval (CI) 0.53 to 1.38; risk difference (RD) -0.01, 95% CI -0.05 to 0.03; I² for RR and RD = 0%; 5 studies, 723 participants; low-certainty evidence). Only one study enrolling 74 infants reported major neurodevelopmental disability in children aged three to five years (RR 0.79, 95% CI 0.51 to 1.24; RD -0.15, 95% CI -0.42 to 0.13; 46 participants; very low-certainty evidence). No studies reported the outcome mortality or major neurodevelopmental disability in children aged 18 to 24 months and 3 to 5 years. Five studies reported bronchopulmonary dysplasia at 36 weeks' postmenstrual age (RR 0.75, 95% CI 0.60 to 0.94; RD -0.08, 95% CI -0.15 to -0.02; number needed to benefit (NNTB) = 13; I² for RR and RD = 0%; 723 participants; moderate-certainty evidence). High-dose caffeine strategies may have little or no effect on side effects (RR 1.66, 95% CI 0.86 to 3.23; RD 0.03, 95% CI -0.01 to 0.07; I² for RR and RD = 0%; 5 studies, 593 participants; low-certainty evidence). The evidence is very uncertain for duration of hospital stay (data reported in three studies could not be pooled in meta-analysis because outcomes were expressed as medians and interquartile ranges) and seizures (RR 1.42, 95% CI 0.79 to 2.53; RD 0.14, 95% CI -0.09 to 0.36; 1 study, 74 participants; very low-certainty evidence). We identified three ongoing trials conducted in China, Egypt, and New Zealand. AUTHORS' CONCLUSIONS High-dose caffeine strategies in preterm infants may have little or no effect on reducing mortality prior to hospital discharge or side effects. We are very uncertain whether high-dose caffeine strategies improves major neurodevelopmental disability, duration of hospital stay or seizures. No studies reported the outcome mortality or major neurodevelopmental disability in children aged 18 to 24 months and 3 to 5 years. High-dose caffeine strategies probably reduce the rate of bronchopulmonary dysplasia. Recently completed and future trials should report long-term neurodevelopmental outcome of children exposed to different caffeine dosing strategies in the neonatal period. Data from extremely preterm infants are needed, as this population is exposed to the highest risk for mortality and morbidity. However, caution is required when administering high doses in the first hours of life, when the risk for intracranial bleeding is highest. Observational studies might provide useful information regarding potential harms of the highest doses.
Collapse
Affiliation(s)
- Matteo Bruschettini
- Paediatrics, Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden
- Cochrane Sweden, Department of Research and Education, Lund University, Skåne University Hospital, Lund, Sweden
| | | | | | - Wes Onland
- Department of Neonatology, Amsterdam University Medical Centers, VU University Medical Center, Emma Children's Hospital, University of Amsterdam, Amsterdam, Netherlands
| | - Peter G Davis
- Newborn Research Centre and Neonatal Services, The Royal Women's Hospital, Melbourne, Australia
- Murdoch Children's Research Institute, Melbourne, Australia
- Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Australia
| | - Roger Soll
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Larner College of Medicine at the University of Vermont, Burlington, Vermont, USA
| |
Collapse
|
6
|
Sahoo S, Behura SS, Wasiq MA, Nayak MK, Mishra A, Panda SK. Respiratory Outcome of Delivery Room Caffeine in Preterm Neonates-A Pilot, Randomized, Controlled Trial. Indian J Pediatr 2023; 90:94. [PMID: 36367678 DOI: 10.1007/s12098-022-04398-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/31/2022] [Indexed: 11/13/2022]
Affiliation(s)
- Swaranjika Sahoo
- Department of Pediatrics, Kalinga Institute of Medical Sciences, KIIT DU, Bhubaneswar, Odisha, 751024, India
| | - Sushree Smita Behura
- Department of Pediatrics, Kalinga Institute of Medical Sciences, KIIT DU, Bhubaneswar, Odisha, 751024, India
| | - Mohammed Abdul Wasiq
- Department of Pediatrics, Kalinga Institute of Medical Sciences, KIIT DU, Bhubaneswar, Odisha, 751024, India
| | - Manas Kumar Nayak
- Department of Pediatrics, Kalinga Institute of Medical Sciences, KIIT DU, Bhubaneswar, Odisha, 751024, India
| | - Alpana Mishra
- Department of Community Medicine, Kalinga Institute of Medical Sciences, KIIT DU, Bhubaneswar, Odisha, India
| | - Santosh Kumar Panda
- Department of Pediatrics, Kalinga Institute of Medical Sciences, KIIT DU, Bhubaneswar, Odisha, 751024, India.
| |
Collapse
|
7
|
Spencer BL, Mychaliska GB. Milestones for clinical translation of the artificial placenta. Semin Fetal Neonatal Med 2022; 27:101408. [PMID: 36437184 DOI: 10.1016/j.siny.2022.101408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Despite significant advances in the treatment of prematurity, premature birth results in significant mortality and morbidity. In particular, extremely low gestational age newborns (ELGANs) defined as <28 weeks estimated gestational age (EGA) suffer from disproportionate mortality and morbidity. A radical paradigm shift in the treatment of prematurity is to recreate fetal physiology using an extracorporeal VV-ECLS artificial placenta (AP) or an AV-ECLS artificial womb (AW). Over the past 15 years, tremendous advances have been made in the laboratory confirming long-term support and organ protection and ongoing development. The major milestones to clinical application are miniaturization, anticoagulation, clinical risk stratification, specialized critical care protocols, a regulatory path and a strategy and platform to translate technology to the bedside. Currently, several groups are addressing the remaining milestones for clinical translation.
Collapse
Affiliation(s)
- Brianna L Spencer
- Department of Surgery, University of Michigan, 2101 Taubman Center 1500 E Medical Center Dr, Ann Arbor, MI, 48109, USA.
| | - George B Mychaliska
- Section of Pediatric Surgery, Department of Surgery, Fetal Diagnosis and Treatment Center, C.S. Mott Children's Hospital, 1540 E Hospital Dr, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
8
|
Long JY, Hu YH, Xia Y, Du FF, Dai HR, Tian M, Xu J, Cheng R, Ding XS, Guo HL, Chen F. Therapeutic drug monitoring of caffeine and its primary metabolites in plasma using LC-ESI-MS/MS for apnea of prematurity treatment: Evaluation of ultrapure water as a surrogate matrix. Biomed Chromatogr 2022; 36:e5462. [PMID: 35881540 DOI: 10.1002/bmc.5462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/18/2022] [Accepted: 07/22/2022] [Indexed: 11/05/2022]
Abstract
The growing evidence has endorsed the view that therapeutic drug monitoring (TDM) of caffeine for apnea of prematurity is helpful for dose tailoring when the therapeutic response is lacking or toxicity is suspected. However, the plasma without caffeine is difficult to obtain. Therefore, a method was developed and validated to measure caffeine and its three primary metabolites (paraxanthine, theobromine, and theophylline) using LC-ESI-MS/MS in human plasma and several surrogate matrices. The chromatographic separation of analytes was finally achieved on a Waters Symmetry C18 (4.6 × 75 mm, 3.5 μm) column. Several strategies were successfully applied to overcome the matrix effects: 1) appropriate dilution for sample cleanup; 2) a starting lower proportion of organic phase; 3) multiple individual stable-labeled isotopic internal standards. The parallelism between the authentic matrix and surrogate matrices was convincing. The recovery of the analytes in both human plasma and rat plasma was acceptable over the linear range (0.500 to 50.0 μg/mL for caffeine, and 0.0100 to 1.00 μg/mL for three metabolites). The method was successfully applied in 118 samples from 74 preterm infants with apnea of prematurity. The rat plasma or ultrapure water as surrogate matrix is worthy of being recommended for routine TDM of caffeine.
Collapse
Affiliation(s)
- Jia-Yi Long
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China.,School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ya-Hui Hu
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Ying Xia
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Fei-Fei Du
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Hao-Ran Dai
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Man Tian
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Xu
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Rui Cheng
- Neonatal Intensive Care Unit, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xuan-Sheng Ding
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hong-Li Guo
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Feng Chen
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
9
|
Yuan Y, Yang Y, Lei X, Dong W. Caffeine and bronchopulmonary dysplasia: Clinical benefits and the mechanisms involved. Pediatr Pulmonol 2022; 57:1392-1400. [PMID: 35318830 DOI: 10.1002/ppul.25898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/05/2022] [Accepted: 03/16/2022] [Indexed: 11/06/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic respiratory disease that occurs during the neonatal period and is commonly associated with prematurity. This condition results in a severe economic burden on society and the families involved. Caffeine is used not only for the treatment of apnea in prematurity, but also for the prevention of BPD. There are multiple clinical benefits of caffeine treatment, including improved extubation success, a reduced duration of mechanical ventilation, improved lung function, and a reduction of patent ductus arteriosus requiring treatment. These clinical benefits of caffeine for the treatment of BPD are supported by both clinical trials and evidence from animal models. However, the mechanism by which caffeine protects against BPD remains unclear. Here, we review the clinical value of caffeine in the prevention of BPD and its potential mechanisms of action, including anti-inflammatory, antioxidant, antifibrotic, and antiapoptotic properties, the regulation of angiogenesis, and diuretic effects. Our aim is to provide a new theoretical basis for the clinical treatment of BPD.
Collapse
Affiliation(s)
- Yuan Yuan
- Department of Pediatrics, Division of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yang Yang
- Department of Pediatrics, Division of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiaoping Lei
- Department of Pediatrics, Division of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wenbin Dong
- Department of Pediatrics, Division of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
10
|
Santoro KL, Yakah W, Singh P, Ramiro-Cortijo D, Medina-Morales E, Freedman SD, Martin CR. Acetaminophen and Xenobiotic Metabolites in Human Milk and the Development of Bronchopulmonary Dysplasia and Retinopathy of Prematurity in a Cohort of Extremely Preterm Infants. J Pediatr 2022; 244:224-229.e3. [PMID: 35093319 PMCID: PMC9152735 DOI: 10.1016/j.jpeds.2022.01.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/22/2021] [Accepted: 01/18/2022] [Indexed: 12/14/2022]
Abstract
This retrospective cohort study sought to identify the association between certain xenobiotic metabolites in maternal breast milk and the diagnoses of bronchopulmonary dysplasia and retinopathy of prematurity in extremely preterm infants. Several acetaminophen metabolites were associated with a 3- to 6-fold increased odds of these disorders, and metabolites of certain food products, benzoate, and caffeine were associated with decreased odds.
Collapse
Affiliation(s)
- Kristin L. Santoro
- Division of Newborn Medicine, Boston Children’s Hospital, Harvard Medical School, Boston MA 02115, USA
| | - William Yakah
- Department of Neonatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA 02215, USA
| | - Pratibha Singh
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA 02215, USA
| | - David Ramiro-Cortijo
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA 02215, USA
| | - Esli Medina-Morales
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA 02215, USA
| | - Steven D. Freedman
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA 02215, USA;,Division of Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA 02215, USA
| | - Camilia R. Martin
- Department of Neonatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA 02215, USA;,Division of Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA 02215, USA
| |
Collapse
|
11
|
Li J, Zhang J, Hao Q, Chen H, Cheng X. Erythropoietin for preventing bronchopulmonary dysplasia in preterm infants: A systematic review and meta-analysis. Pediatr Pulmonol 2022; 57:1051-1063. [PMID: 35043596 DOI: 10.1002/ppul.25837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 01/15/2022] [Accepted: 01/17/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND Recombinant erythropoietin (rEPO) has erythropoiesis and anti-inflammatory properties that might help reduce lung injury in preterm infants. OBJECTIVE To conduct a systematic review and meta-analysis to evaluate the possible role of rEPO in altering the risk of bronchopulmonary dysplasia (BPD). METHODS PubMed, EMBASE, Web of Science, and the Cochrane Library databases were searched to identify randomized controlled trials (RCTs) that evaluated the effects of rEPO for the prevention of BPD in preterm infants. RESULTS Fourteen studies (3199 infants) were included. Our results could not demonstrate a significant effect of rEPO on the incidence of BPD36 (risk ratio [RR]: 0.97, 95% confidence interval [CI]: 0.87-1.09, p = 0.63, I2 = 0, 12 RCTs, high-quality evidence), BPD28 (RR: 1.28, 95% CI: 0.91-1.79, p = 0.15, I2 = 17%, three RCTs, low-quality evidence) and oxygen dependence days. The test for subgroup analysis by administration route of rEPO showed similar outcomes above. Some of the included trials reported a significant effect of intravenous rEPO on reduction of sepsis (RR: 0.82, 95% CI: 0.70-0.96, p = 0.01, I2 = 0, high-quality evidence) and any stage necrotizing enterocolitis (NEC) (RR: 0.75, 95% CI: 0.59-0.94, p = 0.01, I2 = 0, moderate-quality evidence). The incidence of mortality and stage II or higher NEC was comparable in rEPO and control infants. CONCLUSION Our results suggest that rEPO does not affect the risk of developing BPD in preterm infants. Adequately powered RCTs are required to further confirm these findings.
Collapse
Affiliation(s)
- Jing Li
- Department of Neonatology, The First Affiliated Hospital of Zheng Zhou University, Zhengzhou, China
| | - Jing Zhang
- Department of Neonatology, The First Affiliated Hospital of Zheng Zhou University, Zhengzhou, China
| | - Qingfei Hao
- Department of Neonatology, The First Affiliated Hospital of Zheng Zhou University, Zhengzhou, China
| | - Haoming Chen
- Department of Neonatology, The First Affiliated Hospital of Zheng Zhou University, Zhengzhou, China
| | - Xiuyong Cheng
- Department of Neonatology, The First Affiliated Hospital of Zheng Zhou University, Zhengzhou, China
| |
Collapse
|
12
|
Zhang S, Luan X, Li H, Jin Z. Insulin-like growth factor-1: A potential target for bronchopulmonary dysplasia treatment (Review). Exp Ther Med 2022; 23:191. [PMID: 35126694 PMCID: PMC8794548 DOI: 10.3892/etm.2022.11114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 12/08/2021] [Indexed: 11/05/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a common respiratory disorder among preterm infants, particularly low-birth-weight infants (LBWIs) and very-low-birth-weight infants (VLBWIs). Although BPD was first reported 50 years ago, no specific drugs or efficient measures are yet available for prevention or treatment. Insulin-like growth factor-1 (IGF-1) belongs to the insulin family. It promotes mitosis and stimulates cell proliferation and DNA synthesis, the primary factors involved in pulmonary development during the fetal and postnatal periods. Several studies have reported that IGF-1 exerts certain effects on BPD genesis and progression by regulating BPD-related biological processes. In addition, exogenous addition of IGF-1 can alleviate lung inflammation, cell apoptosis and eliminate alveolar development disorders in children with BPD. These findings suggest that IGF-1 could be a new target for treating BPD. Here, we summarize and analyze the definition, pathogenesis, and research status of BPD, as well as the pathogenesis of IGF-1 in BPD and the latest findings in related biological processes.
Collapse
Affiliation(s)
- Shujian Zhang
- Department of Pediatrics, Affiliated Hospital of Yanbian University, Yanji, Jilin 133000, P.R. China
| | - Xue Luan
- Department of Pediatrics, First Hospital, Jilin University, Changchun, Jilin 130000, P.R. China
| | - Huiwen Li
- Department of Pediatrics, Affiliated Hospital of Yanbian University, Yanji, Jilin 133000, P.R. China
| | - Zhengyong Jin
- Department of Pediatrics, Affiliated Hospital of Yanbian University, Yanji, Jilin 133000, P.R. China
| |
Collapse
|
13
|
Qing C, Ziyun L, Xuefei Y, Xinyi Z, Xindong X, Jianhua F. Protective Effects of 18β-Glycyrrhetinic Acid on Neonatal Rats with Hyperoxia Exposure. Inflammation 2022; 45:1224-1238. [PMID: 34989920 DOI: 10.1007/s10753-021-01616-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/13/2021] [Accepted: 12/16/2021] [Indexed: 11/05/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a common devastating pulmonary complication in preterm infants. Supplemental oxygen is a lifesaving therapeutic measure used for premature infants with pulmonary insufficiency. However, oxygen toxicity is a significant trigger for BPD. Oxidative stress disrupts lung development, accompanied by increased pro-inflammatory cytokines and chemokines expression and immune cells infiltration in lung tissue. Licorice, a typical traditional herbal medicine, is commonly used in the medicine and food industries. 18β-Glycyrrhetinic acid (18β-GA), a primary active ingredient of licorice, has powerful anti-oxidative and anti-inflammatory effects. This study aimed to determine whether 18β-GA has a protective effect on neonatal rats with hyperoxia exposure. Newborn Sprague-Dawley rats were kept in either 21% (normoxia) or 80% O2 (hyperoxia) continuously from postnatal day (PN) 1 to 14. 18β-GA was injected intragastrically at 50 or 100 mg/kg body weight once a day from PN 1 to 14. We examined the body weight and alveolar development and measured ROS level and the markers of pulmonary inflammation. Mature-IL-1β and NF-κB pathway proteins, and the NLRP3 inflammasome, were assessed; concurrently, caspase-1 activity was measured. Our results indicated that hyperoxia resulted in alveolar simplification and decreased bodyweight of neonatal rats. Hyperoxia increased ROS level and pulmonary inflammation and activated NF-κB and the NLRP3 inflammasome. 18β-GA treatment inhibited the activation of NF-κB and the NLRP3 inflammasome, decreased ROS level and pulmonary inflammation, improved alveolar development, and increased the bodyweight of neonatal rats with hyperoxia exposure. Our study demonstrates that 18β-GA has a protective effect on neonatal rats with hyperoxia exposure.
Collapse
Affiliation(s)
- Cai Qing
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Liu Ziyun
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Yu Xuefei
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Zhao Xinyi
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Xue Xindong
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Fu Jianhua
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning, 110004, China.
| |
Collapse
|
14
|
Tian C, Li D, Fu J. Molecular Mechanism of Caffeine in Preventing Bronchopulmonary Dysplasia in Premature Infants. Front Pediatr 2022; 10:902437. [PMID: 35795332 PMCID: PMC9251307 DOI: 10.3389/fped.2022.902437] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/31/2022] [Indexed: 11/24/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic respiratory complication commonly seen in premature infants. Following continuous advances in neonatal intensive care diagnosis and treatment technology, an increasing number of premature babies are being treated successfully. Despite these remarkable improvements, there has been no significant decline in the incidence of BPD; in fact, its incidence has increased as more extremely preterm infants survive. Therefore, in view of the impact of BPD on the physical and mental health of children and the increased familial and social burden on these children, early prevention of BPD is emphasized. In recent decades, the clinical application of caffeine in treating primary apnea in premature infants was shown not only to stimulate the respiratory center but also to confer obvious protection to the nervous and respiratory systems. Numerous clinical cross-sectional and longitudinal studies have shown that caffeine plays a significant role in the prevention and treatment of BPD, but there is a lack of overall understanding of its potential molecular mechanisms. In this review, we summarize the possible molecular mechanisms of caffeine in the prevention or treatment of BPD, aiming to better guide its clinical application.
Collapse
Affiliation(s)
- Congliang Tian
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China.,Department of Pediatrics, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Danni Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
15
|
Tong Y, Zhang S, Riddle S, Zhang L, Song R, Yue D. Intrauterine Hypoxia and Epigenetic Programming in Lung Development and Disease. Biomedicines 2021; 9:944. [PMID: 34440150 PMCID: PMC8394854 DOI: 10.3390/biomedicines9080944] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 11/17/2022] Open
Abstract
Clinically, intrauterine hypoxia is the foremost cause of perinatal morbidity and developmental plasticity in the fetus and newborn infant. Under hypoxia, deviations occur in the lung cell epigenome. Epigenetic mechanisms (e.g., DNA methylation, histone modification, and miRNA expression) control phenotypic programming and are associated with physiological responses and the risk of developmental disorders, such as bronchopulmonary dysplasia. This developmental disorder is the most frequent chronic pulmonary complication in preterm labor. The pathogenesis of this disease involves many factors, including aberrant oxygen conditions and mechanical ventilation-mediated lung injury, infection/inflammation, and epigenetic/genetic risk factors. This review is focused on various aspects related to intrauterine hypoxia and epigenetic programming in lung development and disease, summarizes our current knowledge of hypoxia-induced epigenetic programming and discusses potential therapeutic interventions for lung disease.
Collapse
Affiliation(s)
- Yajie Tong
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China;
| | - Shuqing Zhang
- School of Pharmacy, China Medical University, Shenyang 110122, China;
| | - Suzette Riddle
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA;
| | - Rui Song
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA;
| | - Dongmei Yue
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China;
| |
Collapse
|
16
|
Long JY, Guo HL, He X, Hu YH, Xia Y, Cheng R, Ding XS, Chen F, Xu J. Caffeine for the Pharmacological Treatment of Apnea of Prematurity in the NICU: Dose Selection Conundrum, Therapeutic Drug Monitoring and Genetic Factors. Front Pharmacol 2021; 12:681842. [PMID: 34381359 PMCID: PMC8350115 DOI: 10.3389/fphar.2021.681842] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 07/12/2021] [Indexed: 12/20/2022] Open
Abstract
Caffeine citrate is the drug of choice for the pharmacological treatment of apnea of prematurity. Factors such as maturity and genetic variation contribute to the interindividual variability in the clinical response to caffeine therapy in preterm infants, making the optimal dose administered controversial. Moreover, the necessity for therapeutic drug monitoring (TDM) of caffeine is still worth discussing due to the need to achieve the desired target concentrations as well as concerns about the safety of higher doses. Therefore, we reviewed the pharmacokinetic profile of caffeine in preterm infants, evidence of the safety and efficacy of different doses of caffeine, therapeutic concentration ranges of caffeine and impact of genetic variability on caffeine therapy. Whereas the safety and efficacy of standard-dose caffeine have been demonstrated, evidence for the safety of higher administered doses is insufficient. Thus, preterm infants who lack clinical response to standard-dose caffeine therapy are of interest for TDM when dose optimization is performed. Polymorphisms in pharmacodynamics-related genes, but not in pharmacokinetics-related genes, have a significant impact on the interindividual variability in clinical response to caffeine therapy. For preterm infants lacking clinical response, how to develop individualized medication regimens for caffeine remains to be explored.
Collapse
Affiliation(s)
- Jia-Yi Long
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China.,School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hong-Li Guo
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xin He
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Ya-Hui Hu
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Ying Xia
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Rui Cheng
- Neonatal Intensive Care Unit, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xuan-Sheng Ding
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Feng Chen
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Xu
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
17
|
Villosis MFB, Barseghyan K, Ambat MT, Rezaie KK, Braun D. Rates of Bronchopulmonary Dysplasia Following Implementation of a Novel Prevention Bundle. JAMA Netw Open 2021; 4:e2114140. [PMID: 34181013 PMCID: PMC8239950 DOI: 10.1001/jamanetworkopen.2021.14140] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
IMPORTANCE Bronchopulmonary dysplasia (BPD) rates in the United States remain high and have changed little in the last decade. OBJECTIVE To develop a consistent BPD prevention bundle in a systematic approach to decrease BPD. DESIGN, SETTING, AND PARTICIPANTS This quality improvement study included 484 infants with birth weights from 501 to 1500 g admitted to a level 3 neonatal intensive care unit in the Kaiser Permanente Southern California system from 2009 through 2019. The study period was divided into 3 periods: 1, baseline (2009); 2, initial changes based on ongoing cycles of Plan-Do-Study-Act (2010-2014); and 3, full implementation of successive Plan-Do-Study-Act results (2015-2019). INTERVENTIONS A BPD prevention system of care bundle evolved with a shared mental model that BPD is avoidable. MAIN OUTCOMES AND MEASURES The primary outcome was BPD in infants with less than 33 weeks' gestational age (hereafter referred to as BPD <33). Other measures included adjusted BPD <33, BPD severity grade, and adjusted median postmenstrual age (PMA) at hospital discharge. Balancing measures were adjusted mortality and adjusted mortality or specified morbidities. RESULTS The study population included 484 infants with a mean (SD) birth weight of 1070 (277) g; a mean (SD) gestational age of 28.6 (2.9) weeks; 252 female infants (52.1%); and 61 Black infants (12.6%). During the 3 study periods, BPD <33 decreased from 9 of 29 patients (31.0%) to 3 of 184 patients (1.6%) (P < .001 for trend); special cause variation was observed. The standardized morbidity ratio for the adjusted BPD <33 decreased from 1.2 (95% CI, 0.7-1.9) in 2009 to 0.4 (95% CI, 0.2-0.8) in 2019. The rates of combined grades 1, 2, and 3 BPD decreased from 7 of 29 patients (24.1%) to 17 of 183 patients (9.3%) (P < .008 for trend). Grade 2 BPD rates decreased from 3 of 29 patients (10.3%) to 5 of 183 patients (2.7%) (P = .02 for trend). Adjusted median PMA at home discharge decreased by 2 weeks, from 38.2 (95% CI, 37.3-39.1) weeks in 2009 to 36.8 (95% CI, 36.6-37.1) weeks during the last 3 years (2017-2019) of the full implementation period. Adjusted mortality was unchanged, whereas adjusted mortality or specified morbidities decreased significantly. CONCLUSIONS AND RELEVANCE A sustained low rate of BPD was observed in infants after the implementation of a detailed BPD system of care.
Collapse
Affiliation(s)
- Maria Fe B. Villosis
- Department of Pediatrics (Neonatology), Kaiser Permanente Panorama City, Panorama City, California
| | - Karine Barseghyan
- Department of Pediatrics (Neonatology), Kaiser Permanente Panorama City, Panorama City, California
| | - Ma. Teresa Ambat
- Department of Pediatrics (Neonatology), Kaiser Permanente Panorama City, Panorama City, California
| | - Kambiz K. Rezaie
- Department of Pediatrics (Neonatology), Kaiser Permanente Panorama City, Panorama City, California
| | - David Braun
- Department of Pediatrics (Neonatology), Kaiser Permanente Panorama City, Panorama City, California
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena
| |
Collapse
|
18
|
Muehlbacher T, Bassler D, Bryant MB. Evidence for the Management of Bronchopulmonary Dysplasia in Very Preterm Infants. CHILDREN (BASEL, SWITZERLAND) 2021; 8:298. [PMID: 33924638 PMCID: PMC8069828 DOI: 10.3390/children8040298] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Very preterm birth often results in the development of bronchopulmonary dysplasia (BPD) with an inverse correlation of gestational age and birthweight. This very preterm population is especially exposed to interventions, which affect the development of BPD. OBJECTIVE The goal of our review is to summarize the evidence on these daily procedures and provide evidence-based recommendations for the management of BPD. METHODS We conducted a systematic literature research using MEDLINE/PubMed on antenatal corticosteroids, surfactant-replacement therapy, caffeine, ventilation strategies, postnatal corticosteroids, inhaled nitric oxide, inhaled bronchodilators, macrolides, patent ductus arteriosus, fluid management, vitamin A, treatment of pulmonary hypertension and stem cell therapy. RESULTS Evidence provided by meta-analyses, systematic reviews, randomized controlled trials (RCTs) and large observational studies are summarized as a narrative review. DISCUSSION There is strong evidence for the use of antenatal corticosteroids, surfactant-replacement therapy, especially in combination with noninvasive ventilation strategies, caffeine and lung-protective ventilation strategies. A more differentiated approach has to be applied to corticosteroid treatment, the management of patent ductus arteriosus (PDA), fluid-intake and vitamin A supplementation, as well as the treatment of BPD-associated pulmonary hypertension. There is no evidence for the routine use of inhaled bronchodilators and prophylactic inhaled nitric oxide. Stem cell therapy is promising, but should be used in RCTs only.
Collapse
Affiliation(s)
- Tobias Muehlbacher
- Department of Neonatology, University Hospital Zurich, 8091 Zurich, Switzerland; (D.B.); (M.B.B.)
| | | | | |
Collapse
|
19
|
Saroha V, Patel RM. Caffeine for preterm infants: Fixed standard dose, adjustments for age or high dose? Semin Fetal Neonatal Med 2020; 25:101178. [PMID: 33168464 PMCID: PMC8096613 DOI: 10.1016/j.siny.2020.101178] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Caffeine is an effective treatment for apnea of prematurity and has several important benefits, including decreasing respiratory morbidity and motor impairment. In this article, we focus on the dose of caffeine. We review the evidence regarding the efficacy and safety of standard caffeine dosing and alternative dosing approaches, including the use of high dose caffeine and routine dose adjustments for age. Current evidence suggests high dose caffeine may provide additional benefit in reducing the risk of bronchopulmonary dysplasia and extubation failure, but may also increase the risk of cerebellar hemorrhage and seizures. Increasing the standard caffeine citrate dose every 1-2 weeks to a goal dose of 8 mg per kilogram every 24 h may help maintain therapeutic effect. We conclude by highlighting the need for additional trials before high dose caffeine is routinely used.
Collapse
Affiliation(s)
- Vivek Saroha
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Dr. NE, Division of Neonatology, Atlanta, GA, 30322, USA.
| | - Ravi Mangal Patel
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Dr. NE, Division of Neonatology, Atlanta, GA, 30322, USA.
| |
Collapse
|
20
|
Abstract
Caffeine as tested in the CAP trial is safe and effective for preterm infants with birthweights less than 1250 g. Evidence for its use beyond the indications and timing used in this trial is of low quality and conflicting. Some studies suggest that earlier use of caffeine increases the risk of mortality while others suggest it has important benefits. It appears that infants with apnea of prematurity and those receiving assisted ventilation are the most likely to benefit from caffeine. Based on currently available evidence, routine early prescription of caffeine does not appear to be indicated. Infants continue to have potentially damaging episodes of hypoxia secondary to apnea beyond 34 weeks' postmenstrual age. It is unclear whether prolonged use of caffeine improves outcomes in these infants. Randomized trials to resolve these uncertainties are required. They need to be large, at least the size of the CAP trial, and include neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Peter G Davis
- Department of Newborn Research, The Royal Women's Hospital, The University of Melbourne, 20 Flemington Rd, Parkville, VIC, 3052, Australia.
| |
Collapse
|
21
|
Nylander Vujovic S, Nava C, Johansson M, Bruschettini M. Confounding biases in studies on early- versus late-caffeine in preterm infants: a systematic review. Pediatr Res 2020; 88:357-364. [PMID: 31931506 DOI: 10.1038/s41390-020-0757-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/31/2019] [Accepted: 01/01/2020] [Indexed: 01/14/2023]
Abstract
BACKGROUND Caffeine is indicated for the management of apnoea of prematurity and extubation in preterm infants. Early initiation of caffeine administration has increased in the past decades with the purpose of reducing respiratory morbidity. However, there might be harms associated with this approach. This systematic review aims to assess whether early administration of caffeine reduces morbidity and mortality in preterm infants. METHODS The methods were published in a preregistered protocol. The literature search was performed in February 2019 with no restrictions for language or publication date. Randomised controlled trials (RCTs) and cohort studies comparing early versus late caffeine administration to infants born before week 34 were included. RESULTS Two RCTs and 14 cohort studies were included. All studies but one had a serious/critical overall risk of bias. Few studies reported on long-term or patient-relevant outcomes. No meta-analysis could be performed. CONCLUSION Based on the available evidence, no conclusions about the optimal timing of caffeine administration can be drawn. There are inherent methodological problems in the cohort studies. RCTs are needed to answer the question of optimal timing for caffeine administration in neonatal care. Future trials should focus on outcomes relevant to patients and their families and include long-term outcomes.
Collapse
Affiliation(s)
| | | | | | - Matteo Bruschettini
- Cochrane Sweden, Skane University Hospital, Lund, Sweden. .,Department of Clinical Sciences Lund, Paediatrics, Lund University, Skane University Hospital, Lund, Sweden.
| |
Collapse
|
22
|
Prevention of Oxygen-Induced Inflammatory Lung Injury by Caffeine in Neonatal Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3840124. [PMID: 32831996 PMCID: PMC7429812 DOI: 10.1155/2020/3840124] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/15/2020] [Accepted: 07/18/2020] [Indexed: 12/26/2022]
Abstract
Background Preterm birth implies an array of respiratory diseases including apnea of prematurity and bronchopulmonary dysplasia (BPD). Caffeine has been introduced to treat apneas but also appears to reduce rates of BPD. Oxygen is essential when treating preterm infants with respiratory problems but high oxygen exposure aggravates BPD. This experimental study is aimed at investigating the action of caffeine on inflammatory response and cell death in pulmonary tissue in a hyperoxia-based model of BPD in the newborn rat. Material/Methods. Lung injury was induced by hyperoxic exposure with 80% oxygen for three (P3) or five (P5) postnatal days with or without recovery in ambient air until postnatal day 15 (P15). Newborn Wistar rats were treated with PBS or caffeine (10 mg/kg) every two days beginning at the day of birth. The effects of caffeine on hyperoxic-induced pulmonary inflammatory response were examined at P3 and P5 immediately after oxygen exposure or after recovery in ambient air (P15) by immunohistological staining and analysis of lung homogenates by ELISA and qPCR. Results Treatment with caffeine significantly attenuated changes in hyperoxia-induced cell death and apoptosis-associated factors. There was a significant decrease in proinflammatory mediators and redox-sensitive transcription factor NFκB in the hyperoxia-exposed lung tissue of the caffeine-treated group compared to the nontreated group. Moreover, treatment with caffeine under hyperoxia modulated the transcription of the adenosine receptor (Adora)1. Caffeine induced pulmonary chemokine and cytokine transcription followed by immune cell infiltration of alveolar macrophages as well as increased adenosine receptor (Adora1, 2a, and 2b) expression. Conclusions The present study investigating the impact of caffeine on the inflammatory response, pulmonary cell degeneration and modulation of adenosine receptor expression, provides further evidence that caffeine acts as an antioxidative and anti-inflammatory drug for experimental oxygen-mediated lung injury. Experimental studies may broaden the understanding of therapeutic use of caffeine in modulating detrimental mechanisms involved in BPD development.
Collapse
|
23
|
Chiu HY, Chu SM, Lin HY, Tsai ML, Chen YT, Lin HC. Evidence base multi-discipline critical strategies toward better tomorrow for very preterm infants. Pediatr Neonatol 2020; 61:371-377. [PMID: 32201157 DOI: 10.1016/j.pedneo.2020.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 08/30/2019] [Accepted: 01/21/2020] [Indexed: 12/15/2022] Open
Abstract
Despite advances in neonatal intensive care in the recent decade, a large number of very preterm infants (VPIs) remain at risk for significant neurodevelopmental impairment (NDI). Given that there are many interventions need to be implemented during the critical perinatal period so that complications of these vulnerable VPIs could be minimized, it is urgent to develop multi-discipline strategies based on evidence to be carried out. The objective of this new term evidence-based perinatal critical strategies (EBPCS), is to provide beneficial intervention towards better neurodevelopmental outcomes, specifically for preterm infants below 28 weeks gestational age. EBPCS is defined as the management of the VPIs during the perinatal period which would include antenatal counseling with team briefing and share decision making, treat the chorioamnionitis, antenatal MgS04, antenatal steroid, delayed cord clamping/milking, neonatal resuscitation team preparation, prevention of hypothermia, immediate respiratory support with continuous positive airway pressure at delivery room, less invasive surfactant administration, early surfactant with budesonide therapy, support of cardiovascular system, early initiate of probiotics administration, early caffeine, early parenteral and enteral nutrition, promptly initiating antibiotics. These critical strategies will be discussed detail in the text; nonetheless, standardized protocols, technical skills and repeated training are the cornerstones of successful of EBPCS. Further experience from different NICU is needed to prove whether these very complicate and comprehensive perinatal critical strategies could translate into daily practice to mitigate the incidence of NDI in high-risk VPIs.
Collapse
Affiliation(s)
- Hsiao-Yu Chiu
- Department of Neonatology, China Medical University Children's Hospital, Taichung, Taiwan; School of Medicine, China Medical University, Taichung, Taiwan.
| | - Shih-Ming Chu
- Division of Neonatology, Department of Pediatrics, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| | - Hsiang-Yu Lin
- Department of Neonatology, China Medical University Children's Hospital, Taichung, Taiwan; School of Medicine, China Medical University, Taichung, Taiwan.
| | - Ming-Luen Tsai
- Department of Neonatology, China Medical University Children's Hospital, Taichung, Taiwan; School of Medicine, China Medical University, Taichung, Taiwan.
| | - Yin-Ting Chen
- Department of Neonatology, China Medical University Children's Hospital, Taichung, Taiwan; School of Medicine, China Medical University, Taichung, Taiwan
| | - Hung-Chih Lin
- Department of Neonatology, China Medical University Children's Hospital, Taichung, Taiwan; School of Chinese Medicine, China Medical University, Taichung, Taiwan; Asia University Hospital, Asia University, Taichung, Taiwan.
| |
Collapse
|
24
|
Abstract
In the current era, the survival of extremely low-birth-weight infants has increased considerably because of new advances in technology; however, these infants often develop chronic dysfunction of the lung, which is called bronchopulmonary dysplasia (BPD). BPD remains an important cause of neonatal mortality and morbidity despite newer and gentler modes of ventilation. BPD results from the exposure of immature lungs to various antenatal and postnatal factors that lead to an impairment in lung development and aberrant growth of lung parenchyma and vasculature. However, we still struggle with a uniform definition for BPD that can help predict various short- and long-term pulmonary outcomes. With new research, our understanding of the pathobiology of this disease has evolved, and many new mechanisms of lung injury and repair are now known. By utilizing the novel ‘omic’ approaches in BPD, we have now identified various factors in the disease process that may act as novel therapeutic targets in the future. New investigational agents being explored for the management and prevention of BPD include mesenchymal stem cell therapy and insulin-like growth factor 1. Despite this, many questions remain unanswered and require further research to improve the outcomes of premature infants with BPD.
Collapse
Affiliation(s)
- Mitali Sahni
- Pediatrix Medical Group, Sunrise Children's Hospital, Las Vegas, NV, USA.,University of Nevada, Las Vegas, NV, USA
| | - Vineet Bhandari
- Neonatology Research Laboratory, Education and Research Building, Cooper University Hospital, Camden, NJ, USA
| |
Collapse
|
25
|
Mesenchymal stem/stromal cells stably transduced with an inhibitor of CC chemokine ligand 2 ameliorate bronchopulmonary dysplasia and pulmonary hypertension. Cytotherapy 2020; 22:180-192. [PMID: 32139242 DOI: 10.1016/j.jcyt.2020.01.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/21/2020] [Accepted: 01/22/2020] [Indexed: 12/20/2022]
Abstract
Perinatal bronchopulmonary dysplasia (BPD) is defined as lung injury in preterm infants caused by various factors, resulting in serious respiratory dysfunction and high mortality. The administration of mesenchymal stem/stromal cells (MSCs) to treat/prevent BPD has proven to have certain therapeutic effects. However, MSCs can only weakly regulate macrophage function, which is strongly involved in the development of BPD. 7ND-MSCs are MSCs transfected with 7ND, a truncated version of CC chemokine ligand 2 (CCL2) that promotes macrophage activation, using a lentiviral vector. In the present study, we show in a BPD rat model that 7ND-MSC administration, but not MSCs alone, ameliorated the impaired alveolarization evaluated by volume density and surface area in the lung tissue, as well as pulmonary artery remodeling and pulmonary hypertension induced by BPD. In addition, 7ND-MSCs, but not MSCs alone, reduced M1 macrophages and the messenger RNA expressions of interleukin-6 and CCL2 in the lung tissue. Thus, the present study showed the treatment effect of 7ND-MSCs in a BPD rat model, which was more effective than that of MSCs alone.
Collapse
|
26
|
Lin H, Wang X. The effects of gasotransmitters on bronchopulmonary dysplasia. Eur J Pharmacol 2020; 873:172983. [PMID: 32017936 DOI: 10.1016/j.ejphar.2020.172983] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/22/2020] [Accepted: 01/31/2020] [Indexed: 02/06/2023]
Abstract
Bronchopulmonary dysplasia (BPD), which remains a major clinical problem for preterm infants, is caused mainly by hyperoxia, mechanical ventilation and inflammation. Many approaches have been developed with the aim of decreasing the incidence of or alleviating BPD, but effective methods are still lacking. Gasotransmitters, a type of small gas molecule that can be generated endogenously, exert a protective effect against BPD-associated lung injury; nitric oxide (NO), carbon monoxide (CO) and hydrogen sulfide (H2S) are three such gasotransmitters. The protective effects of NO have been extensively studied in animal models of BPD, but the results of these studies are inconsistent with those of clinical trials. NO inhalation seems to have no effect on BPD, although side effects have been reported. NO inhalation is not recommended for BPD treatment in preterm infants, except those with severe pulmonary hypertension. Both CO and H2S decreased lung injury in BPD rodent models in preclinical studies. Another small gas molecule, hydrogen, exerts a protective effect against BPD. The nuclear factor erythroid-derived 2 (Nrf2)/heme oxygenase-1 (HO-1) axis seems to play a central role in the protective effect of these gasotransmitters on BPD. Gasotransmitters play important roles in mammals, but further clinical trials are needed to explore their effects on BPD.
Collapse
Affiliation(s)
- Hai Lin
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Xinbao Wang
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China.
| |
Collapse
|
27
|
Glaser K, Speer CP, Wright CJ. Fine Tuning Non-invasive Respiratory Support to Prevent Lung Injury in the Extremely Premature Infant. Front Pediatr 2020; 7:544. [PMID: 31998672 PMCID: PMC6966957 DOI: 10.3389/fped.2019.00544] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/13/2019] [Indexed: 12/31/2022] Open
Abstract
Within the last decades, therapeutic advances, such as antenatal corticosteroids, surfactant replacement, monitored administration of supplemental oxygen, and sophisticated ventilatory support have significantly improved the survival of extremely premature infants. In contrast, the incidence of some neonatal morbidities has not declined. Rates of bronchopulmonary dysplasia (BPD) remain high and have prompted neonatologists to seek effective strategies of non-invasive respiratory support in high risk infants in order to avoid harmful effects associated with invasive mechanical ventilation. There has been a stepwise replacement of invasive mechanical ventilation by early continuous positive airway pressure (CPAP) as the preferred strategy for initial stabilization and for early respiratory support of the premature infant and management of respiratory distress syndrome. However, the vast majority of high risk babies are mechanically ventilated at least once during their NICU stay. Adjunctive therapies aiming at the prevention of CPAP failure and the support of functional residual capacity have been introduced into clinical practice, including alternative techniques of administering surfactant as well as non-invasive ventilation approaches. In contrast, the strategy of applying sustained lung inflations in the delivery room has recently been abandoned due to evidence of higher rates of death within the first 48 h of life.
Collapse
Affiliation(s)
- Kirsten Glaser
- University Children's Hospital, University of Würzburg, Würzburg, Germany
| | - Christian P. Speer
- University Children's Hospital, University of Würzburg, Würzburg, Germany
| | - Clyde J. Wright
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora, CO, United States
| |
Collapse
|
28
|
Moschino L, Zivanovic S, Hartley C, Trevisanuto D, Baraldi E, Roehr CC. Caffeine in preterm infants: where are we in 2020? ERJ Open Res 2020; 6:00330-2019. [PMID: 32154294 PMCID: PMC7049734 DOI: 10.1183/23120541.00330-2019] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 12/04/2019] [Indexed: 12/16/2022] Open
Abstract
The incidence of preterm birth is increasing, leading to a growing population with potential long-term pulmonary complications. Apnoea of prematurity (AOP) is one of the major challenges when treating preterm infants; it can lead to respiratory failure and the need for mechanical ventilation. Ventilating preterm infants can be associated with severe negative pulmonary and extrapulmonary outcomes, such as bronchopulmonary dysplasia (BPD), severe neurological impairment and death. Therefore, international guidelines favour non-invasive respiratory support. Strategies to improve the success rate of non-invasive ventilation in preterm infants include pharmacological treatment of AOP. Among the different pharmacological options, caffeine citrate is the current drug of choice. Caffeine is effective in reducing AOP and mechanical ventilation and enhances extubation success; it decreases the risk of BPD; and is associated with improved cognitive outcome at 2 years of age, and pulmonary function up to 11 years of age. The commonly prescribed dose (20 mg·kg-1 loading dose, 5-10 mg·kg-1 per day maintenance dose) is considered safe and effective. However, to date there is no commonly agreed standardised protocol on the optimal dosing and timing of caffeine therapy. Furthermore, despite the wide pharmacological safety profile of caffeine, the role of therapeutic drug monitoring in caffeine-treated preterm infants is still debated. This state-of-the-art review summarises the current knowledge of caff-eine therapy in preterm infants and highlights some of the unresolved questions of AOP. We speculate that with increased understanding of caffeine and its metabolism, a more refined respiratory management of preterm infants is feasible, leading to an overall improvement in patient outcome.
Collapse
Affiliation(s)
- Laura Moschino
- Dept of Women's and Children's Health, University of Padua, Padua, Italy
| | - Sanja Zivanovic
- Newborn Services, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Dept of Paediatrics, University of Oxford, Oxford, UK
| | | | | | - Eugenio Baraldi
- Dept of Women's and Children's Health, University of Padua, Padua, Italy
| | - Charles Christoph Roehr
- Newborn Services, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Dept of Paediatrics, University of Oxford, Oxford, UK
| |
Collapse
|
29
|
Firman B, Molnar A, Gray PH. Early high-dose caffeine citrate for extremely preterm infants: Neonatal and neurodevelopmental outcomes. J Paediatr Child Health 2019; 55:1451-1457. [PMID: 30900326 DOI: 10.1111/jpc.14446] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/31/2019] [Accepted: 02/28/2019] [Indexed: 12/19/2022]
Abstract
AIM To examine neonatal morbidities, including the incidence of cerebellar haemorrhage (CBH), and neurodevelopmental outcomes following the administration of high loading dose caffeine citrate compared to standard loading dose caffeine citrate. METHODS This was a retrospective study of 218 preterm infants <28 weeks' gestation who received a loading dose of caffeine citrate within the first 36 h of life at the Mater Mothers' Hospital over a 3-year period (2011-2013). Two groups were compared, with 158 neonates in the high-dose cohort receiving a median dose of caffeine citrate of 80 mg/kg and 60 neonates in the standard dose cohort receiving a median dose of 20 mg/kg. Routine cranial ultrasound, including mastoid views, was performed during the neonatal period. At 2 years of age, infants presented for follow-up and were assessed with the Neurosensory Motor Developmental Assessment (NSMDA) and the Bayley Scales of Infant and Toddler Development-III (Bayley-III). RESULTS There was no difference in the incidence of neonatal morbidities, including CBH, between the two groups. The incidence of CBH in the high-dose group was 2.5% compared to 1.7% in the standard-dose group. There was no difference in the neurodevelopmental follow-up scores as evaluated with the NSMDA and the Bayley-III. CONCLUSIONS The use of early high loading dose caffeine citrate in extremely preterm infants was not shown to be associated with CBH or abnormal long-term neurodevelopmental outcomes. The overall incidence of CBH, however, was much lower than in studies using magnetic resonance imaging techniques. It is suggested that a large randomised clinical trial is needed to determine the optimal dose of caffeine citrate when given early to very preterm infants.
Collapse
Affiliation(s)
- Brooke Firman
- Newborn Services, Mater Mothers' Hospital, Brisbane, Queensland, Australia
| | - Attila Molnar
- Newborn Services, Mater Mothers' Hospital, Brisbane, Queensland, Australia
| | - Peter H Gray
- Newborn Services, Mater Mothers' Hospital, Brisbane, Queensland, Australia.,Mothers', Babies' and Women's Health Programme, Mater Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
30
|
Valenzuela-Stutman D, Marshall G, Tapia JL, Mariani G, Bancalari A, Gonzalez Á. Bronchopulmonary dysplasia: risk prediction models for very-low- birth-weight infants. J Perinatol 2019; 39:1275-1281. [PMID: 31337853 DOI: 10.1038/s41372-019-0430-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 05/03/2019] [Accepted: 05/23/2019] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Our objective is to develop risk prediction models for moderate/severe bronchopulmonary dysplasia (BPD) and BPD and/or death in very-low-birth-weight infants (VLBWI) at birth, 3, 7, and 14 postnatal days. STUDY DESIGN It is a multicenter study including 16,407 infants weighing 500-1500 g (2001-2015) from the Neocosur Network. BPD was defined as oxygen dependency at 36 weeks. Variables were selected using forward logistic regression models. Predictive values were evaluated using the ROC curve. RESULTS In total, 2580 (15.7%) presented BPD and 6121 (37.3%) BPD/death. The AUC values for the BPD models were 0.788, 0.818, 0.827, and 0.894 respectively. For BPD/death, the AUC values were 0.860, 0.869, 0.867, and 0.906. BW and gestational age had higher contribution at birth; at later ages, the length of oxygen therapy and ventilation had the highest contribution. All AUC values were statistically significant when compared with a neutral value of 0.5 (p-value < 0.001). CONCLUSIONS We developed high predictive power models for moderate/severe BPD and BPD/death at four postnatal ages.
Collapse
Affiliation(s)
- Daniela Valenzuela-Stutman
- Departamento de Neonatología, Facultad de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile.
| | - Guillermo Marshall
- Departamento de Neonatología, Facultad de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - José L Tapia
- Departamento de Neonatología, Facultad de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Gonzalo Mariani
- Unidad de Neonatologia, Instituto Universitario Hospital Italiano Buenos Aires, Potosí, Argentina
| | - Aldo Bancalari
- Servicio de Neonatología, Hospital Guillermo Grant Benavente y Departamento de Pediatría, Facultad de Medicina, Universidad de Concepción, Concepcion, Chile
| | - Álvaro Gonzalez
- Departamento de Neonatología, Facultad de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | | |
Collapse
|
31
|
Bovbjerg ML, Pillai S. Current Resources for Evidence-Based Practice, September 2019. J Obstet Gynecol Neonatal Nurs 2019; 48:568-582. [PMID: 31442383 DOI: 10.1016/j.jogn.2019.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
32
|
Is early caffeine therapy safe and effective for ventilated preterm infants? J Perinatol 2019; 39:754-757. [PMID: 30770880 DOI: 10.1038/s41372-019-0336-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 01/18/2019] [Indexed: 01/18/2023]
|
33
|
Greenberg RG, Gayam S, Savage D, Tong A, Gorham D, Sholomon A, Clark RH, Benjamin DK, Laughon M, Smith PB. Furosemide Exposure and Prevention of Bronchopulmonary Dysplasia in Premature Infants. J Pediatr 2019; 208:134-140.e2. [PMID: 30579586 PMCID: PMC6486845 DOI: 10.1016/j.jpeds.2018.11.043] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/31/2018] [Accepted: 11/26/2018] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To evaluate the association between furosemide exposure and risk of bronchopulmonary dysplasia (BPD). STUDY DESIGN This retrospective cohort study included infants (2004-2015) born at 23-29 weeks gestational age and 501-1249 g birth weight. We compared the demographic and clinical characteristics of infants exposed and not exposed to furosemide between postnatal day 7 and 36 weeks postmenstrual age. We examined the association between furosemide exposure and 2 outcomes: BPD and BPD or death. We performed multivariable probit regression models that included demographic and clinical variables in addition to 2 instrumental variables: furosemide exposure by discharge year, and furosemide exposure by site. RESULTS Of 37 693 included infants, 19 235 (51%) were exposed to furosemide; these infants were more premature and had higher respiratory support. Of 33 760 infants who survived to BPD evaluation, 15 954 (47%) had BPD. An increase in the proportion of furosemide exposure days by 10 percentage points was associated with a decrease in both the incidence of BPD (4.6 percentage points; P = .001), and BPD or death (3.7 percentage points; P = .01). CONCLUSIONS More days of furosemide exposure between postnatal day 7 and 36 weeks was associated with decreased risk of BPD and a combined outcome of BPD or death.
Collapse
Affiliation(s)
- Rachel G Greenberg
- Department of Pediatrics, Duke University School of Medicine, Durham, NC; Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC.
| | - Sreepriya Gayam
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | - Destiny Savage
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | - Andrew Tong
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | - Daniel Gorham
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | - Ari Sholomon
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | - Reese H Clark
- Pediatrix-Obstetrix Center for Research and Education, Sunrise, FL
| | | | - Matthew Laughon
- Department of Pediatrics, The University of North Carolina-Chapel Hill, Chapel Hill, NC
| | - P Brian Smith
- Department of Pediatrics, Duke University School of Medicine, Durham, NC; Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| |
Collapse
|
34
|
Brattström P, Russo C, Ley D, Bruschettini M. High-versus low-dose caffeine in preterm infants: a systematic review and meta-analysis. Acta Paediatr 2019; 108:401-410. [PMID: 30242903 DOI: 10.1111/apa.14586] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 06/26/2018] [Accepted: 09/18/2018] [Indexed: 12/13/2022]
Abstract
AIM Though caffeine is a consolidated treatment in preterm infants, the efficacy and safety of a higher dose have not been systematically appraised. METHODS A systematic review was conducted to compare high (loading dose >20 mg/kg and maintenance >10 mg/kg/day) versus low dose of caffeine. MEDLINE, EMBASE, Central and conference proceedings for randomised controlled trials (RCTs) and quasi-RCTs were searched. Two authors independently screened the records, extracted the data and assessed the risk of bias. RESULTS As only six RCTs enrolling a total of 816 preterm infants were included, the required information size was not reached. The loading and maintenance doses varied between 20 and 80 mg/kg/day and 3 and 20 mg/kg/day, respectively. The use of high dose had no impact on mortality (RR: 0.85; 95% CI: 0.53-1.38; RCTs = 4). However, it resulted in fewer cases of extubation failure, apnoeas and bronchopulmonary dysplasia (RR: 0.76; 95% CI: 0.60-0.96; studies = 4) and shorter duration of mechanical ventilation. The quality of the evidence was low due to imprecision of the estimates. CONCLUSION Due to imprecision, it is not possible to determine whether high-dose caffeine is more effective and safe than a low dose. High dose might improve short-term respiratory function and reduce bronchopulmonary dysplasia.
Collapse
Affiliation(s)
| | | | - David Ley
- Department of Paediatrics Lund University Skåne University Hospital Lund Sweden
| | - Matteo Bruschettini
- Department of Paediatrics Lund University Skåne University Hospital Lund Sweden
- Cochrane Sweden Lund Sweden
| |
Collapse
|
35
|
Eliminating Risk of Intubation in Very Preterm Infants with Noninvasive Cardiorespiratory Support in the Delivery Room and Neonatal Intensive Care Unit. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5984305. [PMID: 30733962 PMCID: PMC6348842 DOI: 10.1155/2019/5984305] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 11/09/2018] [Accepted: 12/20/2018] [Indexed: 11/18/2022]
Abstract
Introduction Avoiding intubation and promoting noninvasive modes of ventilator support including continuous positive airway pressure (CPAP) in preterm infants minimizes lung injury and optimizes neonatal outcomes. Discharge home on oxygen is an expensive morbidity in very preterm infants (VPI) with lung disease. In 2007 a standardized bundle was introduced for VPI admitted to the neonatal care unit (NICU) which included delayed cord clamping (DCC) at birth and noninvasive ventilation as first-line cardiorespiratory support in the delivery room (DR), followed by bubble CPAP upon NICU admission. Objective Our goal was to evaluate the risk of (1) intubation and (2) discharge home on oxygen after adopting this standardized DR bundle in VPI born at a regional perinatal center and treated in the NICU over a ten-year period (2008-2017). Materials and Methods We compared maternal and neonatal demographics, respiratory care processes and outcomes, as well as neonatal mortality and morbidity in VPI (< 33 weeks gestation) and extremely low birth weight (ELBW, < 1000 g) subgroup for three consecutive epochs: 2008-2010, 2011-2013, and 2014-2017. Results Of 640 consecutive inborn VPI, 55% were < 1500 g at birth and 23% were ELBW. Constant through all three epochs, DCC occurred in 83% of VPI at birth. There was progressive increase in maternal magnesium during the three epochs and decrease in maternal antibiotics during the last epoch. Over the three epochs, VPI had less risk of DR intubation (23% versus 15% versus 5%), NICU intubation (39% versus 31% versus 18%), and invasive ventilation (37% versus 30% versus 17%), as did ELBW infants. Decrease in postnatal steroid use, antibiotic exposure, and increase in early colostrum exposure occurred over the three epochs both in VPI and in ELBW infants. There was a sustained decrease in surfactant use in the second and third epochs. There was no significant change in mortality or any morbidity in VPI; however, there was a significant decrease in pneumothorax (17% versus 0%) and increase in survival without major morbidity (15% versus 41%) in ELBW infants between 2008-2010 and 2014-2017. Benchmarked risk-adjusted rate for oxygen at discharge in a subgroup of inborn VPI (401-1500 g or 22-31 weeks of gestation) is 2.5% (2013-2017) in our NICU compared with > 8% in all California NICUs and > 10% in all California regional NICUs (2014-2016). Conclusion Noninvasive strategies in DR and NICU minimize risk of intubation in VPI without adversely affecting other neonatal or respiratory outcomes. Risk-adjusted rates for discharge home on oxygen remained significantly lower for inborn VPI compared with rates at regional NICUs in California. Reducing intubation risk in ELBW infants may confer an advantage for survival without major morbidity. Prenatal magnesium may reduce intubation risk in ELBW infants.
Collapse
|
36
|
Selected Literature Watch. J Caffeine Adenosine Res 2018. [DOI: 10.1089/caff.2018.29009.slw] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
37
|
Carlson NS. Current Resources for Evidence-Based Practice, September 2018. J Obstet Gynecol Neonatal Nurs 2018; 47:673-683. [PMID: 30063896 DOI: 10.1016/j.jogn.2018.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|