1
|
Briones-Claudett KH, Briones-Claudett MH, Bajaña Huilcapi CK, Tripul Villamar OE, Ochoa Vásquez R, Rivera Salas CDR, Briones-Zamora KH, Benites Solis J, Briones-Márquez DC, Freire AX, Grunauer M. Surfactant therapy using vibrating-mesh nebulizers in adults with COVID-19-induced ARDS: A case series. SAGE Open Med Case Rep 2024; 12:2050313X241236313. [PMID: 38444695 PMCID: PMC10913513 DOI: 10.1177/2050313x241236313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 02/14/2024] [Indexed: 03/07/2024] Open
Abstract
Coronavirus adult respiratory distress syndrome, characterized by decreased surfactant due to lysis of type II pneumocytes and hyaline membrane formation, contributes to severe hypoxemia. The administration of surfactant via high-flow nasal cannula (HFNC) may positively affect lung structure and function in this context. In this study, we report on five clinical cases, encompassing patients aged 40-60 years of both sexes, who tested positive for coronavirus disease 2019 via real-time polymerase chain reaction and exhibited significant pulmonary compromise with elevated inflammatory biomarkers. These patients were treated with aerosol therapy using surfactant delivered through vibrating-mesh nebulizers alongside HFNC. Of these patients, four demonstrated positive responses to the treatment, suggesting that aerosol therapy with surfactant through vibrating-mesh nebulizers could be a viable rescue therapy in adults receiving HFNC oxygen therapy for hypoxemic respiratory failure caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Unfortunately, one patient had a negative outcome and succumbed. The findings from these cases indicate that the use of aerosol therapy with vibrating-mesh nebulizers as rescue therapy might offer an alternative approach for managing adults with hypoxemic respiratory failure due to SARS-CoV-2, as evidenced by the positive outcomes in four out of the five cases presented.
Collapse
Affiliation(s)
- Killen H Briones-Claudett
- Facultad de Medicina, Universidad de Las Americas, Quito, Ecuador
- Intensive Care Unit, Ecuadorian Institute of Social Security, Babahoyo, Ecuador
| | | | | | | | | | | | | | | | | | - Amado X Freire
- Division of Pulmonary, Critical Care, and Sleep Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Michelle Grunauer
- School of Medicine, Universidad San Francisco de Quito, Quito, Ecuador
| |
Collapse
|
2
|
Budh HP, Nimbalkar S. Surfactant Replacement Therapy: What’s the New Future? JOURNAL OF NEONATOLOGY 2022; 36:331-347. [DOI: 10.1177/09732179221136963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
Surfactant replacement therapy (SRT) can be lifesaving for preterm babies with respiratory distress because of surfactant deficiency. Attempts have been made over the last two decades to make surfactant administration as smooth and as nontraumatic as possible. Lesser invasive techniques, such as less invasive surfactant administration, minimally invasive surfactant therapy, intrapartum pharyngeal surfactant therapy, and the laryngeal mask airway, are preferred over invasive techniques like intubate surfactant extubation to reduce trauma and peridosing adverse effects. However, at present, aerosolized surfactant (AS) via nebulization remains the only truly noninvasive method of SRT. Many animal and human studies have shown promising results with the use of AS with similar clinical effects to an instilled surfactant with greater safety potential. But still AS has not been adapted to routine neonatal care. There is still scope for studies to further strengthen the role of AS. Also, SRT is a constantly changing field with new innovations revolutionizing and replacing old techniques.
Collapse
Affiliation(s)
- Hetal Pramod Budh
- Department of Neonatology, Pramukhswami Medical College, Bhaikaka University, Karamsad, Gujarat, India
| | - Somashekhar Nimbalkar
- Department of Neonatology, Pramukhswami Medical College, Bhaikaka University, Karamsad, Gujarat, India
| |
Collapse
|
3
|
Dani C, Talosi G, Piccinno A, Ginocchio VM, Balla G, Lavizzari A, Stranak Z, Gitto E, Martinelli S, Plavka R, Krolak-Olejnik B, Lista G, Spedicato F, Ciurlia G, Santoro D, Sweet D. A Randomized, Controlled Trial to Investigate the Efficacy of Nebulized Poractant Alfa in Premature Babies with Respiratory Distress Syndrome. J Pediatr 2022; 246:40-47.e5. [PMID: 35257740 DOI: 10.1016/j.jpeds.2022.02.054] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 02/09/2022] [Accepted: 02/28/2022] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To investigate the efficacy and safety of nebulized poractant alfa (at 200 and 400 mg/kg doses) delivered in combination with nasal continuous positive airway pressure compared with nasal continuous positive airway pressure alone in premature infants with diagnosed respiratory distress syndrome. STUDY DESIGN This randomized, controlled, multinational study was conducted in infants at 280/7 to 326/7 weeks of gestation. The primary outcome was the incidence of respiratory failure in the first 72 hours of life, defined as needing endotracheal surfactant and/or mechanical ventilation owing to prespecified criteria. Secondary outcomes included the time to respiratory failure in the first 72 hours, duration of ventilation, mortality, incidence of bronchopulmonary dysplasia, and major associated neonatal comorbidities. In addition, the safety and tolerability of the treatments were assessed reporting the number and percentage of infants with treatment-emergent adverse events and adverse drug reactions during nebulization. RESULTS In total, 129 infants were randomized. No significant differences were observed for the primary outcome: 24 (57%), 20 (49%), and 25 (58%) infants received endotracheal surfactant and/or mechanical ventilation within 72 hours in the poractant alfa 200 mg/kg, poractant alfa 400 mg/kg, and nasal continuous positive airway pressure groups, respectively. Similarly, secondary respiratory outcomes did not differ among groups. Enrollment was halted early owing to a change in the benefit-risk balance of the intervention. Nebulized poractant alfa was well-tolerated and safe, and no serious adverse events were related to the study treatment. CONCLUSIONS The intervention did not decrease the likelihood of respiratory failure within the first 72 hours of life. TRIAL REGISTRATION ClinicalTrials.gov: NCT03235986.
Collapse
Affiliation(s)
- Carlo Dani
- Careggi University Hospital of Florence, Florence, Italy.
| | - Gyula Talosi
- Bács-Kiskun County Teaching Hospital, Kecskemét, Hungary
| | | | | | | | | | - Zbynek Stranak
- Institute for the Care of Mother and Child, Prague, Czech Republic
| | - Eloisa Gitto
- University Hospital Gaetano Martino, Messina, Italy
| | | | - Richard Plavka
- General University Hospital in Prague, Prague, Czech Republic
| | | | | | | | | | | | - David Sweet
- Royal Jubilee Maternity Hospital, Belfast, United Kingdom
| | | |
Collapse
|
4
|
CFD Study of Dry Pulmonary Surfactant Aerosols Deposition in Upper 17 Generations of Human Respiratory Tract. ATMOSPHERE 2022. [DOI: 10.3390/atmos13050726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The efficient generation of high concentrations of fine-particle, pure surfactant aerosols provides the possibility of new, rapid, and effective treatment modalities for Acute Respiratory Distress Syndrome (ARDS). SUPRAER-CATM is a patented technology by Kaer BiotherapeuticsTM, which is a new class of efficient aerosol drug generation and delivery system using Compressor Air (CA). SUPRAER-CA is capable of aerosolizing relatively viscous solutions or suspensions of proteins and surfactants and of delivering them as pure fine particle dry aerosols. In this Computational Fluid Dynamics (CFD) study, we select a number of sites within the upper 17 generations of the human respiratory tract for calculation of the deposition of dry pulmonary surfactant aerosol particles. We predict the percentage of inhaled dry pulmonary surfactant aerosol arriving from the respiratory bronchioles to the terminal alveolar sacs. The dry pulmonary surfactant aerosols, with a Mass Median Aerodynamic Diameter (MMAD) of 2.6 µm and standard deviation of 1.9 µm, are injected into the respiratory tract at a dry surfactant aerosol flow rate of 163 mg/min to be used in the CFD study at an air inhalation flow rate of 44 L/min. This CFD study in the upper 17th generation of a male adult lung has shown computationally that the penetration fraction (PF) is approximately 25% for the inhaled surfactant aerosols. In conclusion, an ARDS patient might receive approximately one gram of inspired dry surfactant aerosol during an administration period of one hour as a possible means of further inflating partly collapsed alveoli.
Collapse
|
5
|
Erdeve Ö, Okulu E, Roberts KD, Guthrie SO, Fort P, Kanmaz Kutman HG, Dargaville PA. Alternative Methods of Surfactant Administration in Preterm Infants with Respiratory Distress Syndrome: State of the Art. Turk Arch Pediatr 2022; 56:553-562. [PMID: 35110053 PMCID: PMC8849067 DOI: 10.5152/turkarchpediatr.2021.21240] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
For preterm infants with respiratory distress syndrome, delivery of surfactant via brief intubation (INtubate, SURfactant, Extubate; InSurE) has been the standard technique of surfactant administration. However, this method requires intubation and positive pressure ventilation. It is thought that even the short exposure to positive pressure inflations may be enough to initiate the cascade of events that lead to lung injury in the smallest neonates. In an effort to avoid tracheal intubation and positive pressure ventilation, several alternative and less invasive techniques of exogenous surfactant administration have been developed over the years. These have been investigated in clinical studies, including randomized clinical trials, and have demonstrated advantages such as a decrease in the need for mechanical ventilation and incidence of bronchopulmonary dysplasia. These newer techniques of surfactant delivery also have the benefit of being easier to perform. Surfactant delivery via pharyngeal instillation, laryngeal mask, aerosolization, and placement of a thin catheter are being actively pursued in research. We present a contemporary review of surfactant administration for respiratory distress syndrome via these alternative methods in the hope of guiding physicians in their choices for surfactant application in the neonatal intensive care unit.
Collapse
Affiliation(s)
- Ömer Erdeve
- Division of Neonatology, Department of Pediatrics, Ankara University, Faculty of Medicine, Ankara, Turkey
| | - Emel Okulu
- Division of Neonatology, Department of Pediatrics, Ankara University School of Medicine, Ankara, Turkey
| | - Kari D Roberts
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, USA
| | - Scott O Guthrie
- Division of Neonatology, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Prem Fort
- Division of Neonatology, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, USA; Johns Hopkins All Children's Maternal Fetal and Neonatal Institute, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - H Gözde Kanmaz Kutman
- Division of Neonatology, Department of Pediatrics, Health Sciences University, Ankara, Turkey
| | - Peter A Dargaville
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia; Department of Paediatrics, Royal Hobart Hospital, Hobart, Tasmania, Australia
| |
Collapse
|
6
|
Anderson S, Atkins P, Bäckman P, Cipolla D, Clark A, Daviskas E, Disse B, Entcheva-Dimitrov P, Fuller R, Gonda I, Lundbäck H, Olsson B, Weers J. Inhaled Medicines: Past, Present, and Future. Pharmacol Rev 2022; 74:48-118. [PMID: 34987088 DOI: 10.1124/pharmrev.120.000108] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/06/2021] [Indexed: 12/21/2022] Open
Abstract
The purpose of this review is to summarize essential pharmacological, pharmaceutical, and clinical aspects in the field of orally inhaled therapies that may help scientists seeking to develop new products. After general comments on the rationale for inhaled therapies for respiratory disease, the focus is on products approved approximately over the last half a century. The organization of these sections reflects the key pharmacological categories. Products for asthma and chronic obstructive pulmonary disease include β -2 receptor agonists, muscarinic acetylcholine receptor antagonists, glucocorticosteroids, and cromones as well as their combinations. The antiviral and antibacterial inhaled products to treat respiratory tract infections are then presented. Two "mucoactive" products-dornase α and mannitol, which are both approved for patients with cystic fibrosis-are reviewed. These are followed by sections on inhaled prostacyclins for pulmonary arterial hypertension and the challenging field of aerosol surfactant inhalation delivery, especially for prematurely born infants on ventilation support. The approved products for systemic delivery via the lungs for diseases of the central nervous system and insulin for diabetes are also discussed. New technologies for drug delivery by inhalation are analyzed, with the emphasis on those that would likely yield significant improvements over the technologies in current use or would expand the range of drugs and diseases treatable by this route of administration. SIGNIFICANCE STATEMENT: This review of the key aspects of approved orally inhaled drug products for a variety of respiratory diseases and for systemic administration should be helpful in making judicious decisions about the development of new or improved inhaled drugs. These aspects include the choices of the active ingredients, formulations, delivery systems suitable for the target patient populations, and, to some extent, meaningful safety and efficacy endpoints in clinical trials.
Collapse
Affiliation(s)
- Sandra Anderson
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| | - Paul Atkins
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| | - Per Bäckman
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| | - David Cipolla
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| | - Andrew Clark
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| | - Evangelia Daviskas
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| | - Bernd Disse
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| | - Plamena Entcheva-Dimitrov
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| | - Rick Fuller
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| | - Igor Gonda
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| | - Hans Lundbäck
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| | - Bo Olsson
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| | - Jeffry Weers
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| |
Collapse
|
7
|
Wiegandt FC, Froriep UP, Müller F, Doll T, Dietzel A, Pohlmann G. Breath-Triggered Drug Release System for Preterm Neonates. Pharmaceutics 2021; 13:pharmaceutics13050657. [PMID: 34064425 PMCID: PMC8147847 DOI: 10.3390/pharmaceutics13050657] [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: 03/31/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 11/24/2022] Open
Abstract
A major disadvantage of inhalation therapy with continuous drug delivery is the loss of medication during expiration. Developing a breath-triggered drug release system can highly decrease this loss. However, there is currently no breath-triggered drug release directly inside the patient interface (nasal prong) for preterm neonates available due to their high breathing frequency, short inspiration time and low tidal volume. Therefore, a nasal prong with an integrated valve releasing aerosol directly inside the patient interface increasing inhaled aerosol efficiency is desirable. We integrated a miniaturized aerosol valve into a nasal prong, controlled by a double-stroke cylinder. Breathing was simulated using a test lung for preterm neonates on CPAP respiratory support. The inhalation flow served as a trigger signal for the valve, releasing humidified surfactant. Particle detection was performed gravimetrically (filter) and optically (light extinction). The integrated miniaturized aerosol valve enabled breath-triggered drug release inside the patient interface with an aerosol valve response time of <25 ms. By breath-triggered release of the pharmaceutical aerosol as a bolus during inhalation, the inhaled aerosol efficiency was increased by a factor of >4 compared to non-triggered release. This novel nasal prong with integrated valve allows breath-triggered drug release directly inside the nasal prong with short response time.
Collapse
Affiliation(s)
- Felix C. Wiegandt
- Division of Translational Biomedical Engineering, Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, 30625 Hannover, Germany; (U.P.F.); (F.M.); (T.D.)
- Correspondence: (F.C.W.); (G.P.); Tel.: +49-511-5350-287 (F.C.W.); +49-511-5350-116 (G.P.)
| | - Ulrich P. Froriep
- Division of Translational Biomedical Engineering, Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, 30625 Hannover, Germany; (U.P.F.); (F.M.); (T.D.)
| | - Fabian Müller
- Division of Translational Biomedical Engineering, Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, 30625 Hannover, Germany; (U.P.F.); (F.M.); (T.D.)
| | - Theodor Doll
- Division of Translational Biomedical Engineering, Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, 30625 Hannover, Germany; (U.P.F.); (F.M.); (T.D.)
- Department of Otorhinolaryngology, Hannover Medical School, 30625 Hannover, Germany
| | - Andreas Dietzel
- Institute of Microtechnology, Technische Universität Braunschweig, 38124 Braunschweig, Germany;
| | - Gerhard Pohlmann
- Division of Translational Biomedical Engineering, Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, 30625 Hannover, Germany; (U.P.F.); (F.M.); (T.D.)
- Correspondence: (F.C.W.); (G.P.); Tel.: +49-511-5350-287 (F.C.W.); +49-511-5350-116 (G.P.)
| |
Collapse
|
8
|
Bianco F, Salomone F, Milesi I, Murgia X, Bonelli S, Pasini E, Dellacà R, Ventura ML, Pillow J. Aerosol drug delivery to spontaneously-breathing preterm neonates: lessons learned. Respir Res 2021; 22:71. [PMID: 33637075 PMCID: PMC7908012 DOI: 10.1186/s12931-020-01585-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023] Open
Abstract
Delivery of medications to preterm neonates receiving non-invasive ventilation (NIV) represents one of the most challenging scenarios for aerosol medicine. This challenge is highlighted by the undersized anatomy and the complex (patho)physiological characteristics of the lungs in such infants. Key physiological restraints include low lung volumes, low compliance, and irregular respiratory rates, which significantly reduce lung deposition. Such factors are inherent to premature birth and thus can be regarded to as the intrinsic factors that affect lung deposition. However, there are a number of extrinsic factors that also impact lung deposition: such factors include the choice of aerosol generator and its configuration within the ventilation circuit, the drug formulation, the aerosol particle size distribution, the choice of NIV type, and the patient interface between the delivery system and the patient. Together, these extrinsic factors provide an opportunity to optimize the lung deposition of therapeutic aerosols and, ultimately, the efficacy of the therapy.In this review, we first provide a comprehensive characterization of both the intrinsic and extrinsic factors affecting lung deposition in premature infants, followed by a revision of the clinical attempts to deliver therapeutic aerosols to premature neonates during NIV, which are almost exclusively related to the non-invasive delivery of surfactant aerosols. In this review, we provide clues to the interpretation of existing experimental and clinical data on neonatal aerosol delivery and we also describe a frame of measurable variables and available tools, including in vitro and in vivo models, that should be considered when developing a drug for inhalation in this important but under-served patient population.
Collapse
Affiliation(s)
- Federico Bianco
- Department of Preclinical Pharmacology, R&D, Chiesi Farmaceutici S.P.A., 43122 Parma, Italy
| | - Fabrizio Salomone
- Department of Preclinical Pharmacology, R&D, Chiesi Farmaceutici S.P.A., 43122 Parma, Italy
| | - Ilaria Milesi
- Department of Preclinical Pharmacology, R&D, Chiesi Farmaceutici S.P.A., 43122 Parma, Italy
| | | | - Sauro Bonelli
- Department of Preclinical Pharmacology, R&D, Chiesi Farmaceutici S.P.A., 43122 Parma, Italy
| | - Elena Pasini
- Department of Preclinical Pharmacology, R&D, Chiesi Farmaceutici S.P.A., 43122 Parma, Italy
| | - Raffaele Dellacà
- TechRes Lab, Dipartimento Di Elettronica, Informazione E Bioingegneria (DEIB), Politecnico Di Milano University, Milano, Italy
| | | | - Jane Pillow
- School of Human Sciences, University of Western Australia, Perth, Australia
| |
Collapse
|
9
|
Sood BG, Thomas R, Delaney-Black V, Xin Y, Sharma A, Chen X. Aerosolized Beractant in neonatal respiratory distress syndrome: A randomized fixed-dose parallel-arm phase II trial. Pulm Pharmacol Ther 2020; 66:101986. [PMID: 33338661 DOI: 10.1016/j.pupt.2020.101986] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 11/02/2020] [Accepted: 12/09/2020] [Indexed: 01/01/2023]
Abstract
PURPOSE There is increasing research into novel techniques of administering surfactant to preterm infants (PTIs) with respiratory distress syndrome (RDS) receiving non-invasive respiratory support (NIRS). Although aerosolized surfactant (AS) is promising in PTIs receiving NIRS, the optimal surfactant dose and formulation, drug-device combination and patient profile is not known. The objective of this randomized clinical trial was to investigate the feasibility, safety, efficacy and impact of four dosing schedules of AS using two nebulizers in PTIs with RDS stratified by gestational age (GA). METHODS PTIs with RDS receiving pre-defined NIRS for ≤8 h were assigned to 4 A S dosing schedules and 2 nebulizers within three GA strata (I = 240/7-286/7, II = 290/7-326/7, III = 330/7-366/7 weeks). There was no contemporaneous control group; at the recommendation of the Data Monitoring Committee, data was collected retrospectively for control infants. RESULTS Of 149 subjects that received AS, the median age at initiation of the 1st dose and duration was 5.5 and 2.4 h respectively. There were 29 infants in stratum I, and 60 each in strata II and III. Of infants <32 weeks GA, 94% received caffeine prior to AS. Fifteen infants (10%) required intubation within 72 h; the rates were not significantly different between GA strata, dosing schedules and nebulizers for infants who received aerosolized surfactant. Compared to retrospective controls, infants who received AS were less likely to need intubation within 72 h in both the intention-to-treat (32% vs. 11%) and the per-protocol (22% vs. 10%) analyses (p < 0.05) with GA stratum specific differences. AS was well tolerated by infants and clinical caregivers. Commonest adverse events included surfactant reflux from nose and mouth (18%), desaturations (11%), and increased secretions (7%). CONCLUSIONS We have demonstrated the feasibility, absence of serious adverse events and short-term efficacy of four dosing schedules of AS in the largest Phase II clinical trial of PTIs 24-36 weeks' GA with RDS receiving NIRS (ClinicalTrials.gov NCT02294630). The commonest adverse events noted were surfactant reflux and desaturations; no serious adverse effects were observed. Infants who received AS were less likely to receive intubation within 72 h compared to historical controls. AS is a promising new therapy for PTIs with RDS.
Collapse
Affiliation(s)
- Beena G Sood
- Department of Pediatrics, Wayne State University School of Medicine, 540 E Canfield St, Detroit, MI, 48201, USA.
| | - Ronald Thomas
- Department of Pediatrics, Wayne State University School of Medicine, 540 E Canfield St, Detroit, MI, 48201, USA
| | - Virginia Delaney-Black
- Department of Pediatrics, Wayne State University School of Medicine, 540 E Canfield St, Detroit, MI, 48201, USA
| | - Yuemin Xin
- Department of Pediatrics, Wayne State University School of Medicine, 540 E Canfield St, Detroit, MI, 48201, USA
| | - Amit Sharma
- Department of Pediatrics, Wayne State University School of Medicine, 540 E Canfield St, Detroit, MI, 48201, USA
| | - Xinguang Chen
- Department of Epidemiology, University of Florida College of Medicine, 665 W 8th Street, Jacksonville, FL, 32209, USA
| |
Collapse
|
10
|
Cummings JJ, Gerday E, Minton S, Katheria A, Albert G, Flores-Torres J, Famuyide M, Lampland A, Guthrie S, Kuehn D, Weitkamp JH, Fort P, Abu Jawdeh EG, Ryan RM, Martin GC, Swanson JR, Mulrooney N, Eyal F, Gerstmann D, Kumar P, Wilding GE, Egan EA. Aerosolized Calfactant for Newborns With Respiratory Distress: A Randomized Trial. Pediatrics 2020; 146:peds.2019-3967. [PMID: 33060258 DOI: 10.1542/peds.2019-3967] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/11/2020] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Exogenous surfactants to treat respiratory distress syndrome (RDS) are approved for tracheal instillation only; this requires intubation, often followed by positive pressure ventilation to promote distribution. Aerosol delivery offers a safer alternative, but clinical studies have had mixed results. We hypothesized that efficient aerosolization of a surfactant with low viscosity, early in the course of RDS, could reduce the need for intubation and instillation of liquid surfactant. METHODS A prospective, multicenter, randomized, unblinded comparison trial of aerosolized calfactant (Infasurf) in newborns with signs of RDS that required noninvasive respiratory support. Calfactant was aerosolized by using a Solarys nebulizer modified with a pacifier adapter; 6 mL/kg (210 mg phospholipid/kg body weight) were delivered directly into the mouth. Infants in the aerosol group received up to 3 treatments, at least 4 hours apart. Infants in the control group received usual care, determined by providers. Infants were intubated and given instilled surfactant for persistent or worsening respiratory distress, at their providers' discretion. RESULTS Among 22 NICUs, 457 infants were enrolled; gestation 23 to 41 (median 33) weeks and birth weight 595 to 4802 (median 1960) grams. In total, 230 infants were randomly assigned to aerosol; 225 received 334 treatments, starting at a median of 5 hours. The rates of intubation for surfactant instillation were 26% in the aerosol group and 50% in the usual care group (P < .0001). Respiratory outcomes up to 28 days of age were no different. CONCLUSIONS In newborns with early, mild to moderate respiratory distress, aerosolized calfactant at a dose of 210 mg phospholipid/kg body weight reduced intubation and surfactant instillation by nearly one-half.
Collapse
Affiliation(s)
| | - Erick Gerday
- Utah Valley Regional Medical Center, Provo, Utah
| | | | - Anup Katheria
- Sharp Mary Birch Hospital for Women and Newborns, San Diego, California
| | | | | | - Mobolaji Famuyide
- Department of Pediatrics, University of Mississippi, Oxford, Mississippi
| | | | - Scott Guthrie
- Jackson-Madison County General Hospital, Jackson, Tennessee
| | - Devon Kuehn
- East Carolina University and Vidant Medical Center, Greenville, North Carolina
| | | | - Prem Fort
- Johns Hopkins All Children's Hospital, St. Petersburg, Florida
| | | | - Rita M Ryan
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| | | | | | | | - Fabien Eyal
- University of South Alabama Children's and Women's Hospital, Mobile, Alabama
| | | | - Praveen Kumar
- Order of St. Francis Children's Hospital of Illinois, Peoria, Illinois
| | - Greg E Wilding
- Department of Biostatistics, University at Buffalo, Buffalo, New York; and
| | | | | |
Collapse
|
11
|
Guo X, Luo S, Amidani D, Rivetti C, Pieraccini G, Pioselli B, Catinella S, Murgia X, Salomone F, Xu Y, Dong Y, Sun B. In vitro characterization and in vivo comparison of the pulmonary outcomes of Poractant alfa and Calsurf in ventilated preterm rabbits. PLoS One 2020; 15:e0230229. [PMID: 32168331 PMCID: PMC7069639 DOI: 10.1371/journal.pone.0230229] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 02/12/2020] [Indexed: 11/18/2022] Open
Abstract
Poractant alfa and Calsurf are two natural surfactants widely used in China for the treatment of neonatal respiratory distress syndrome, which are extracted from porcine and calf lungs, respectively. The purpose of this experimental study was to compare their in vitro characteristics and in vivo effects in the improvement of pulmonary function and protection of lung injury. The biophysical properties, ultrastructure, and lipid composition of both surfactant preparations were respectively analysed in vitro by means of Langmuir-Blodgett trough (LBT), atomic force microscopy (AFM), and liquid-chromatography mass-spectrometry (LC-MS). Then, as core pharmacological activity, both head-to-head (100 and 200 mg/kg for both surfactants) and licensed dose comparisons (70 mg/kg Calsurf vs. 200 mg/kg Poractant alfa) between the two surfactants were conducted as prophylaxis in preterm rabbits with primary surfactant deficiency, assessing survival time and rate and dynamic compliance of the respiratory system (Cdyn). Intrapulmonary surfactant pools, morphometric volume density as alveolar expansion (Vv), and lung injury scores were determined post mortem. AFM and LC-MS analysis revealed qualitative differences in the ultrastructure as well as in the lipid composition of both preparations. Calsurf showed a longer plateau region of the LBT isotherm and lower film compressibility. In vivo, both surfactant preparations improved Cdyn at any dose, although maximum benefits in terms of Vv and intrapulmonary surfactant pools were seen with the 200 mg/kg dose in both surfactants. The group of animals treated with 200 mg/kg of Poractant alfa showed a prolonged survival time and rate compared to untreated but ventilated controls, and significantly ameliorated lung injury compared to Calsurf at any dose, including 200 mg/kg. The overall outcomes suggest the pulmonary effects to be dose dependent for both preparations. The group of animals treated with 200 mg/kg of Poractant alfa showed a significant reduction of mortality. Compared to Calsurf, Poractant alfa exerted better effects if licensed doses were compared, which requires further investigation.
Collapse
Affiliation(s)
- Xiaojing Guo
- Departments of Pediatrics and Neonatology, Children’s Hospital of Fudan University, Shanghai, China
| | - Siwei Luo
- Departments of Pediatrics and Neonatology, Children’s Hospital of Fudan University, Shanghai, China
| | - Davide Amidani
- Department of Research and Development, Chiesi Farmaceutici, Parma, Italy
| | - Claudio Rivetti
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Giuseppe Pieraccini
- CISM Mass Spectrometry Centre, Department of Health Sciences, University of Florence, Firenze, Italy
| | - Barbara Pioselli
- Department of Research and Development, Chiesi Farmaceutici, Parma, Italy
| | - Silvia Catinella
- Department of Research and Development, Chiesi Farmaceutici, Parma, Italy
| | - Xabi Murgia
- Department of Drug Delivery, Helmholtz Institute for Pharmaceutical Research, Saarbrücken, Saarland, Germany
| | - Fabrizio Salomone
- Department of Research and Development, Chiesi Farmaceutici, Parma, Italy
| | - Yaling Xu
- Departments of Pediatrics and Neonatology, Children’s Hospital of Fudan University, Shanghai, China
| | - Ying Dong
- Departments of Pediatrics and Neonatology, Children’s Hospital of Fudan University, Shanghai, China
| | - Bo Sun
- Departments of Pediatrics and Neonatology, Children’s Hospital of Fudan University, Shanghai, China
- * E-mail: ,
| |
Collapse
|
12
|
In Vitro Performance of an Investigational Vibrating-Membrane Nebulizer with Surfactant under Simulated, Non-Invasive Neonatal Ventilation Conditions: Influence of Continuous Positive Airway Pressure Interface and Nebulizer Positioning on the Lung Dose. Pharmaceutics 2020; 12:pharmaceutics12030257. [PMID: 32178276 PMCID: PMC7151046 DOI: 10.3390/pharmaceutics12030257] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 11/25/2022] Open
Abstract
Non-invasive delivery of nebulized surfactant has been a long-pursued goal in neonatology. Our aim was to evaluate the performance of an investigational vibrating-membrane nebulizer in a realistic non-invasive neonatal ventilation circuit with different configurations. Surfactant (aerosols were generated with a nebulizer in a set-up composed of a continuous positive airway pressure (CPAP) generator with a humidifier, a cast of the upper airway of a preterm infant (PrINT), and a breath simulator with a neonatal breathing pattern. The lung dose (LD), defined as the amount of surfactant collected in a filter placed at the distal end of the PrINT cast, was determined after placing the nebulizer at different locations of the circuit and using either infant nasal mask or nasal prongs as CPAP interfaces. The LD after delivering a range of nominal surfactant doses (100–600 mg/kg) was also investigated. Surfactant aerosol particle size distribution was determined by laser diffraction. Irrespective of the CPAP interface used, about 14% of the nominal dose (200 mg/kg) reached the LD filter. However, placing the nebulizer between the Y-piece and the CPAP interface significantly increased the LD compared with placing it 7 cm before the Y-piece, in the inspiratory limb. (14% ± 2.8 vs. 2.3% ± 0.8, nominal dose of 200 mg/kg). The customized eFlow Neos showed a constant aerosol generation rate and a mass median diameter of 2.7 μm after delivering high surfactant doses (600 mg/kg). The customized eFlow Neos nebulizer showed a constant performance even after nebulizing high doses of undiluted surfactant. Placing the nebulizer between the Y-piece and the CPAP interface achieves the highest LD under non-invasive ventilation conditions.
Collapse
|
13
|
Bianco F, Ricci F, Catozzi C, Murgia X, Schlun M, Bucholski A, Hetzer U, Bonelli S, Lombardini M, Pasini E, Nutini M, Pertile M, Minocchieri S, Simonato M, Rosa B, Pieraccini G, Moneti G, Lorenzini L, Catinella S, Villetti G, Civelli M, Pioselli B, Cogo P, Carnielli V, Dani C, Salomone F. From bench to bedside: in vitro and in vivo evaluation of a neonate-focused nebulized surfactant delivery strategy. Respir Res 2019; 20:134. [PMID: 31266508 PMCID: PMC6604359 DOI: 10.1186/s12931-019-1096-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 06/12/2019] [Indexed: 01/17/2023] Open
Abstract
Background Non-invasive delivery of nebulized surfactant has been a neonatology long-pursued goal. Nevertheless, the clinical efficacy of nebulized surfactant remains inconclusive, in part, due to the great technical challenges of depositing nebulized drugs in the lungs of preterm infants. The aim of this study was to investigate the feasibility of delivering nebulized surfactant (poractant alfa) in vitro and in vivo with an adapted, neonate-tailored aerosol delivery strategy. Methods Particle size distribution of undiluted poractant alfa aerosols generated by a customized eFlow-Neos nebulizer system was determined by laser diffraction. The theoretical nebulized surfactant lung dose was estimated in vitro in a clinical setting replica including a neonatal continuous positive airway pressure (CPAP) circuit, a cast of the upper airways of a preterm neonate, and a breath simulator programmed with the tidal breathing pattern of an infant with mild respiratory distress syndrome (RDS). A dose-response study with nebulized surfactant covering the 100–600 mg/kg nominal dose-range was conducted in RDS-modelling, lung-lavaged spontaneously-breathing rabbits managed with nasal CPAP. The effects of nebulized poractant alfa on arterial gas exchange and lung mechanics were assessed. Exogenous alveolar disaturated-phosphatidylcholine (DSPC) in the lungs was measured as a proxy of surfactant deposition efficacy. Results Laser diffraction studies demonstrated suitable aerosol characteristics for inhalation (mass median diameter, MMD = 3 μm). The mean surfactant lung dose determined in vitro was 13.7% ± 4.0 of the 200 mg/kg nominal dose. Nebulized surfactant delivered to spontaneously-breathing rabbits during nasal CPAP significantly improved arterial oxygenation compared to animals receiving CPAP only. Particularly, the groups of animals treated with 200 mg/kg and 400 mg/kg of nebulized poractant alfa achieved an equivalent pulmonary response in terms of oxygenation and lung mechanics as the group of animals treated with instilled surfactant (200 mg/kg). Conclusions The customized eFlow-Neos vibrating-membrane nebulizer system efficiently generated respirable aerosols of undiluted poractant alfa. Nebulized surfactant delivered at doses of 200 mg/kg and 400 mg/kg elicited a pulmonary response equivalent to that observed after treatment with an intratracheal surfactant bolus of 200 mg/kg. This bench-characterized nebulized surfactant delivery strategy is now under evaluation in Phase II clinical trial (EUDRACT No.:2016–004547-36). Electronic supplementary material The online version of this article (10.1186/s12931-019-1096-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- F Bianco
- Department of Preclinical Pharmacology, R&D, Chiesi Farmaceutici S.p.A, Parma, Italy
| | - F Ricci
- Department of Preclinical Pharmacology, R&D, Chiesi Farmaceutici S.p.A, Parma, Italy
| | - C Catozzi
- Department of Preclinical Pharmacology, R&D, Chiesi Farmaceutici S.p.A, Parma, Italy
| | - X Murgia
- Scientific Consultancy, Saarbrücken, Germany
| | - M Schlun
- PARI Pharma GmbH, Starnberg, Germany
| | | | - U Hetzer
- PARI Pharma GmbH, Starnberg, Germany
| | - S Bonelli
- Department of Preclinical Pharmacology, R&D, Chiesi Farmaceutici S.p.A, Parma, Italy
| | - M Lombardini
- Department of Preclinical Pharmacology, R&D, Chiesi Farmaceutici S.p.A, Parma, Italy
| | - E Pasini
- Department of Preclinical Pharmacology, R&D, Chiesi Farmaceutici S.p.A, Parma, Italy
| | - M Nutini
- Department of Preclinical Pharmacology, R&D, Chiesi Farmaceutici S.p.A, Parma, Italy
| | - M Pertile
- Department of Preclinical Pharmacology, R&D, Chiesi Farmaceutici S.p.A, Parma, Italy
| | - S Minocchieri
- Division of Neonatology, Cantonal Hospital Winterthur, Winterthur, Switzerland
| | - M Simonato
- Pediatric Research Institute "Città della Speranza", Padova, Italy
| | - B Rosa
- Department of Preclinical Pharmacology, R&D, Chiesi Farmaceutici S.p.A, Parma, Italy
| | - G Pieraccini
- Mass Spectrometry Center (CISM), Polo Biomedico, Careggi University Hospital of Florence, Florence, Italy
| | - G Moneti
- Mass Spectrometry Center (CISM), Polo Biomedico, Careggi University Hospital of Florence, Florence, Italy
| | - L Lorenzini
- Health Science and Technologies Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Bologna, Italy
| | - S Catinella
- Department of Preclinical Pharmacology, R&D, Chiesi Farmaceutici S.p.A, Parma, Italy
| | - G Villetti
- Department of Preclinical Pharmacology, R&D, Chiesi Farmaceutici S.p.A, Parma, Italy
| | - M Civelli
- Department of Preclinical Pharmacology, R&D, Chiesi Farmaceutici S.p.A, Parma, Italy
| | - B Pioselli
- Department of Preclinical Pharmacology, R&D, Chiesi Farmaceutici S.p.A, Parma, Italy
| | - P Cogo
- Division of Pediatrics, Department of Medicine, University of Udine, Udine, Italy
| | - V Carnielli
- Polytechnic University of Marche and Azienda Ospedaliero-Universitaria Ospedali Riuniti, Ancona, Italy
| | - C Dani
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence School of Medicine, Careggi University Hospital of Florence, Viale Morgagni, 85, Florence, Italy.
| | - F Salomone
- Department of Preclinical Pharmacology, R&D, Chiesi Farmaceutici S.p.A, Parma, Italy
| |
Collapse
|
14
|
Sood BG, Cortez J, Kolli M, Sharma A, Delaney-Black V, Chen X. Aerosolized surfactant in neonatal respiratory distress syndrome: Phase I study. Early Hum Dev 2019; 134:19-25. [PMID: 31121339 DOI: 10.1016/j.earlhumdev.2019.05.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/03/2019] [Accepted: 05/07/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Treating respiratory distress syndrome (RDS) with intratracheal surfactant requires endotracheal intubation and mechanical ventilation, (MV) with their attendant risks. Use of non-invasive respiratory support in the delivery room averts the need for MV but delays surfactant administration. OBJECTIVE We hypothesized that aerosolized surfactant is feasible and safe in infants 240/7-366/7 weeks gestational age (GA) with RDS, receiving non-invasive respiratory support. DESIGN/METHODS In an unblinded Phase I study, sequentially enrolled infants with RDS stratified by GA received increasing doses (100 or 200 mg/kg of phospholipid) and dilutions (12.5 or 8.3 mg/ml) of surfactant using a jet nebulizer. Infants were monitored clinically and with cerebral oximetry. RESULTS Seventeen infants were enrolled. Age at start of first dose and dose duration were 4.9 (3.4-10.1) and 2.1 (1.0-2.8) hours respectively. Two infants in the lowest GA stratum (240/7-286/7) required intubation within 2 h after the first dose. Fifteen infants completed the study; 13 received two doses. Infants tolerated the aerosol treatment well. No other significant adverse events were identified. Parental permission for cerebral oximetry was obtained in 16 infants. In the two infants who later exited the study, values prior to start of aerosolized surfactant were lower compared to 14 infants who completed the study (p = 0.0835), increased after start of study intervention (p = 0.0105) and decreased after intubation (p = 0.0003). CONCLUSIONS We have demonstrated the feasibility and safety of aerosolized surfactant in preterm infants receiving non-invasive respiratory support. The treatment was well tolerated by infants and clinical caregivers.
Collapse
Affiliation(s)
- Beena G Sood
- Children's Hospital of Michigan, 3901 Beaubien Blvd., Suite 3N027, Detroit, MI 48201, USA; Hutzel Women's Hospital, 3990 John R St, Detroit, MI 48201, USA; Department of Pediatrics, Wayne State University School of Medicine, 540 E Canfield St, Detroit, MI 48201, USA.
| | - Josef Cortez
- Department of Pediatrics, University of Florida College of Medicine, 665 W 8th Street, Jacksonville, FL 32209, USA.
| | - Madhuri Kolli
- Department of Pediatrics, Wayne State University School of Medicine, 540 E Canfield St, Detroit, MI 48201, USA
| | - Amit Sharma
- Children's Hospital of Michigan, 3901 Beaubien Blvd., Suite 3N027, Detroit, MI 48201, USA; Hutzel Women's Hospital, 3990 John R St, Detroit, MI 48201, USA; Department of Pediatrics, Wayne State University School of Medicine, 540 E Canfield St, Detroit, MI 48201, USA.
| | - Virginia Delaney-Black
- Department of Pediatrics, Wayne State University School of Medicine, 540 E Canfield St, Detroit, MI 48201, USA.
| | - Xinguang Chen
- University of Florida College of Medicine, College of Public Health, 2004 Mowray Road, Gainesville, FL 32610, USA.
| |
Collapse
|
15
|
Ricci F, Casiraghi C, Storti M, D’Alò F, Catozzi C, Ciccimarra R, Ravanetti F, Cacchioli A, Villetti G, Civelli M, Murgia X, Carnielli V, Salomone F. Surfactant replacement therapy in combination with different non-invasive ventilation techniques in spontaneously-breathing, surfactant-depleted adult rabbits. PLoS One 2018; 13:e0200542. [PMID: 30001410 PMCID: PMC6042776 DOI: 10.1371/journal.pone.0200542] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 06/28/2018] [Indexed: 12/14/2022] Open
Abstract
Nasal intermittent positive pressure ventilation (NIPPV) holds great potential as a primary ventilation support method for Respiratory Distress Syndrome (RDS). The use of NIPPV may also be of great value combined with minimally invasive surfactant delivery. Our aim was to implement an in vivo model of RDS, which can be managed with different non-invasive ventilation (NIV) strategies, including non-synchronized NIPPV, synchronized NIPPV (SNIPPV), and nasal continuous positive airway pressure (NCPAP). Forty-two surfactant-depleted adult rabbits were allocated in six different groups: three groups of animals were treated with only NIV for three hours (NIPPV, SNIPPV, and NCPAP groups), while three other groups were treated with surfactant (SF) followed by NIV (NIPPV+SF, SNIPPV+SF, and NCPAP+SF groups). Arterial gas exchange, ventilation indices, and dynamic compliance were assessed. Post-mortem the lungs were sampled for histological evaluation. Surfactant depletion was successfully achieved by repeated broncho-alveolar lavages (BALs). After BALs, all animals developed a moderate respiratory distress, which could not be reverted by merely applying NIV. Conversely, surfactant administration followed by NIV induced a rapid improvement of arterial oxygenation in all surfactant-treated groups. Breath synchronization was associated with a significantly better response in terms of gas exchange and dynamic compliance compared to non-synchronized NIPPV, showing also the lowest injury scores after histological assessment. The proposed in vivo model of surfactant deficiency was successfully managed with NCPAP, NIPPV, or SNIPPV; this model resembles a moderate respiratory distress and it is suitable for the preclinical testing of less invasive surfactant administration techniques.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Xabi Murgia
- Department of Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland, Saarbrücken, Germany
| | - Virgilio Carnielli
- Division of Neonatology, Polytechnic University of Marche and Salesi Children’s Hospital, Ancona, Italy
| | | |
Collapse
|
16
|
Ricci F, Catozzi C, Murgia X, Rosa B, Amidani D, Lorenzini L, Bianco F, Rivetti C, Catinella S, Villetti G, Civelli M, Pioselli B, Dani C, Salomone F. Physiological, Biochemical, and Biophysical Characterization of the Lung-Lavaged Spontaneously-Breathing Rabbit as a Model for Respiratory Distress Syndrome. PLoS One 2017; 12:e0169190. [PMID: 28060859 PMCID: PMC5217971 DOI: 10.1371/journal.pone.0169190] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 12/13/2016] [Indexed: 12/01/2022] Open
Abstract
Nasal continuous positive airway pressure (nCPAP) is a widely accepted technique of non-invasive respiratory support in spontaneously-breathing premature infants with respiratory distress syndrome (RDS). Surfactant administration techniques compatible with nCPAP ventilation strategy are actively investigated. Our aim is to set up and validate a respiratory distress animal model that can be managed on nCPAP suitable for surfactant administration techniques studies. Surfactant depletion was induced by bronchoalveolar lavages (BALs) on 18 adult rabbits. Full depletion was assessed by surfactant component analysis on the BALs samples. Animals were randomized into two groups: Control group (nCPAP only) and InSurE group, consisting of a bolus of surfactant (Poractant alfa, 200 mg/kg) followed by nCPAP. Arterial blood gases were monitored until animal sacrifice, 3 hours post treatment. Lung mechanics were evaluated just before and after BALs, at the time of treatment, and at the end of the procedure. Surfactant phospholipids and protein analysis as well as surface tension measurements on sequential BALs confirmed the efficacy of the surfactant depletion procedure. The InSurE group showed a significant improvement of blood oxygenation and lung mechanics. On the contrary, no signs of recovery were appreciated in animals treated with just nCPAP. The surfactant-depleted adult rabbit RDS model proved to be a valuable and efficient preclinical tool for mimicking the clinical scenario of preterm infants affected by mild/moderate RDS who spontaneously breathe and do not require mechanical ventilation. This population is of particular interest as potential target for the non-invasive administration of surfactant.
Collapse
Affiliation(s)
| | | | - Xabier Murgia
- Department of Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland, Saarbrücken, Germany
| | - Brenda Rosa
- Chiesi Farmaceutici, R&D Department, Parma, Italy
| | - Davide Amidani
- Department of Life Sciences, University of Parma, Parma, Italy
| | | | | | - Claudio Rivetti
- Department of Life Sciences, University of Parma, Parma, Italy
| | | | | | | | | | - Carlo Dani
- Department of Neurosciences, Psychology, Drug Research and Child Health, Careggi University Hospital of Florence, Florence, Italy
| | | |
Collapse
|
17
|
Syedain ZH, Naqwi AA, Dolovich M, Somani A. In Vitro Evaluation of a Device for Intra-Pulmonary Aerosol Generation and Delivery. AEROSOL SCIENCE AND TECHNOLOGY : THE JOURNAL OF THE AMERICAN ASSOCIATION FOR AEROSOL RESEARCH 2015; 49:747-752. [PMID: 26884641 PMCID: PMC4753072 DOI: 10.1080/02786826.2015.1067670] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
For infants born with respiratory distress syndrome (RDS), liquid bolus delivery of surfactant administered through an endotracheal tube is common practice. While this method is generally effective, complications such as transient hypoxia, hypercapnia, and altered cerebral blood flow may occur. Aerosolized surfactant therapy has been explored as an alternative. Unfortunately, past efforts have led to disappointing results as aerosols were generated outside the lungs with significant pharyngeal deposition and minimal intrapulmonary instillation. A novel aerosol generator (Microjet™) is evaluated herein for intrapulmonary aerosol generation within an endotracheal tube and tested with Curosurf and Infasurf surfactants. Compared with other aerosol delivery devices, this process utilizes low air flow (range 0.01-0.2 L/min) that is ideal for limiting potential barotrauma to the premature newborn lung. The mass mean diameter (MMD) of the particles for both tested surfactants was less than 4 μm, which is ideal for both uniform and distal lung delivery. As an indicator of phospholipid function, surfactant surface tension was measured before and after aerosol formation; with no significant difference. Moreover, this device has an outside diameter of <1mm, which permits insertion into an endotracheal tube (of even 2.0 mm). In the premature infant where intravenous access is either technically challenging or difficult, aerosol drug delivery may provide an alternative route in patient resuscitation, stabilization and care. Other potential applications of this type of device include the delivery of nutrients, antibiotics, and analgesics via the pulmonary route.
Collapse
Affiliation(s)
- Zeeshan H. Syedain
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
- Division of Critical Care, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Myrna Dolovich
- Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Arif Somani
- Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
- Division of Critical Care, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
18
|
Acute Neonatal Respiratory Failure. PEDIATRIC AND NEONATAL MECHANICAL VENTILATION 2015. [PMCID: PMC7193706 DOI: 10.1007/978-3-642-01219-8_47] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Acute respiratory failure requiring assisted ventilation is one of the most common reasons for admission to the neonatal intensive care unit. Respiratory failure is the inability to maintain either normal delivery of oxygen to the tissues or normal removal of carbon dioxide from the tissues. It occurs when there is an imbalance between the respiratory workload and ventilatory strength and endurance. Definitions are somewhat arbitrary but suggested laboratory criteria for respiratory failure include two or more of the following: PaCO2 > 60 mmHg, PaO2 < 50 mmHg or O2 saturation <80 % with an FiO2 of 1.0 and pH < 7.25 (Wen et al. 2004).
Collapse
|
19
|
Goikoetxea E, Murgia X, Serna-Grande P, Valls-i-Soler A, Rey-Santano C, Rivas A, Antón R, Basterretxea FJ, Miñambres L, Méndez E, Lopez-Arraiza A, Larrabe-Barrena JL, Gomez-Solaetxe MA. In vitro surfactant and perfluorocarbon aerosol deposition in a neonatal physical model of the upper conducting airways. PLoS One 2014; 9:e106835. [PMID: 25211475 PMCID: PMC4161382 DOI: 10.1371/journal.pone.0106835] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 08/02/2014] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE Aerosol delivery holds potential to release surfactant or perfluorocarbon (PFC) to the lungs of neonates with respiratory distress syndrome with minimal airway manipulation. Nevertheless, lung deposition in neonates tends to be very low due to extremely low lung volumes, narrow airways and high respiratory rates. In the present study, the feasibility of enhancing lung deposition by intracorporeal delivery of aerosols was investigated using a physical model of neonatal conducting airways. METHODS The main characteristics of the surfactant and PFC aerosols produced by a nebulization system, including the distal air pressure and air flow rate, liquid flow rate and mass median aerodynamic diameter (MMAD), were measured at different driving pressures (4-7 bar). Then, a three-dimensional model of the upper conducting airways of a neonate was manufactured by rapid prototyping and a deposition study was conducted. RESULTS The nebulization system produced relatively large amounts of aerosol ranging between 0.3±0.0 ml/min for surfactant at a driving pressure of 4 bar, and 2.0±0.1 ml/min for distilled water (H2Od) at 6 bar, with MMADs between 2.61±0.1 µm for PFD at 7 bar and 10.18±0.4 µm for FC-75 at 6 bar. The deposition study showed that for surfactant and H2Od aerosols, the highest percentage of the aerosolized mass (∼65%) was collected beyond the third generation of branching in the airway model. The use of this delivery system in combination with continuous positive airway pressure set at 5 cmH2O only increased total airway pressure by 1.59 cmH2O at the highest driving pressure (7 bar). CONCLUSION This aerosol generating system has the potential to deliver relatively large amounts of surfactant and PFC beyond the third generation of branching in a neonatal airway model with minimal alteration of pre-set respiratory support.
Collapse
Affiliation(s)
- Estibalitz Goikoetxea
- Research Unit for Experimental Neonatal Respiratory Physiology, Cruces University Hospital, Barakaldo, Bizkaia, Spain
- Thermal and Fluids Engineering Division, Mechanical Engineering Department, TECNUN, University of Navarra, San Sebastian, Gipuzkoa, Spain
| | - Xabier Murgia
- Research Unit for Experimental Neonatal Respiratory Physiology, Cruces University Hospital, Barakaldo, Bizkaia, Spain
| | - Pablo Serna-Grande
- Research Unit for Experimental Neonatal Respiratory Physiology, Cruces University Hospital, Barakaldo, Bizkaia, Spain
| | - Adolf Valls-i-Soler
- Neonatal Intensive Care Unit, Cruces University Hospital, Barakaldo, Bizkaia, Spain
| | - Carmen Rey-Santano
- Research Unit for Experimental Neonatal Respiratory Physiology, Cruces University Hospital, Barakaldo, Bizkaia, Spain
| | - Alejandro Rivas
- Thermal and Fluids Engineering Division, Mechanical Engineering Department, TECNUN, University of Navarra, San Sebastian, Gipuzkoa, Spain
| | - Raúl Antón
- Thermal and Fluids Engineering Division, Mechanical Engineering Department, TECNUN, University of Navarra, San Sebastian, Gipuzkoa, Spain
| | - Francisco J. Basterretxea
- Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country, Leioa, Bizkaia, Spain
| | - Lorena Miñambres
- Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country, Leioa, Bizkaia, Spain
| | - Estíbaliz Méndez
- Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country, Leioa, Bizkaia, Spain
| | - Alberto Lopez-Arraiza
- Department of Electronics and Electrotechnics, High Technical School of Maritime Studies, University of the Basque Country, Bilbao, Bizkaia, Spain
| | - Juan Luis Larrabe-Barrena
- Department of Electronics and Electrotechnics, High Technical School of Maritime Studies, University of the Basque Country, Bilbao, Bizkaia, Spain
| | - Miguel Angel Gomez-Solaetxe
- Department of Electronics and Electrotechnics, High Technical School of Maritime Studies, University of the Basque Country, Bilbao, Bizkaia, Spain
| |
Collapse
|
20
|
Lampland AL, Wolfson MR, Mazela J, Henderson C, Gregory TJ, Meyers P, Plumm B, Worwa C, Mammel MC. Aerosolized KL4 surfactant improves short-term survival and gas exchange in spontaneously breathing newborn pigs with hydrochloric acid-induced acute lung injury. Pediatr Pulmonol 2014; 49:482-9. [PMID: 24039229 DOI: 10.1002/ppul.22844] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Accepted: 05/10/2013] [Indexed: 11/06/2022]
Abstract
BACKGROUND Surfactant therapy may be beneficial in acute lung injury (ALI). In spontaneously breathing newborn pigs with ALI supported with continuous positive airway pressure (CPAP), we evaluated the hypothesis that aerosolized KL4 surfactant (AERO KL4 S) would provide a similar therapeutic effect as intratracheal KL4 surfactant (ETT KL4 S) when compared to controls. METHODS We randomized pigs with HCl-induced ALI to: (1) 175 mg/kg KL4 surfactant via endotracheal tube (ETT); (2) AERO KL4 S (22.5 mg/min phospholipid) for 60 min via continuous positive airway pressure (CPAP); or (3) sham procedure on CPAP. We obtained physiologic data and arterial blood gases throughout the 3-hr study. At study end, lungs were excised for analysis of interleukin-8 (IL-8), myeloperoxidase (MPO) levels and histomorphometric data. RESULTS Pigs treated with ETT KL4 S and AERO KL4 S had improved survival and sustained pO2 compared to controls. The AERO KL4 S group had higher pH compared to controls. Lung IL-8 levels were lower in the AERO KL4 S group compared to controls. Histomorphometric analysis showed less hemorrhage in the ETT and AERO KL4 S groups compared to controls. The AERO KL4 S group had more open lung units per fixed-field than the ETT KL4 S or controls. CONCLUSIONS AERO KL4 S produced similar improvements in survival, physiology, inflammatory markers, and morphology as ETT KL4 S in an ALI model.
Collapse
Affiliation(s)
- Andrea L Lampland
- Infant Diagnostic and Research Center, Children's Hospitals and Clinics of Minnesota, St. Paul, Minnesota; Department of Pediatrics-Neonatology, University of Minnesota, Minneapolis, Minnesota
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Minocchieri S, Knoch S, Schoel WM, Ochs M, Nelle M. Nebulizing poractant alfa versus conventional instillation: Ultrastructural appearance and preservation of surface activity. Pediatr Pulmonol 2014; 49:348-56. [PMID: 24039226 DOI: 10.1002/ppul.22838] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 04/23/2013] [Accepted: 05/20/2013] [Indexed: 11/06/2022]
Abstract
BACKGROUND Nebulized surfactant therapy has been proposed as an alternative method of surfactant administration. The use of a perforated vibrating membrane nebulizer provides a variety of advantages over conventional nebulizers. We investigated the molecular structure and integrity of poractant alfa pre- and post-nebulization. METHOD Curosurf® was nebulized using an Investigational eFlow® Nebulizer System. Non-nebulized surfactant ("NN"), recollected surfactant droplets from nebulization through an endotracheal tube ("NT") and nebulization of surfactant directly onto a surface ("ND") were investigated by transmission electron microscopy. Biophysical characteristics were assessed by the Langmuir-Wilhelmy balance and the Captive Bubble Surfactometer. RESULTS Volume densities of lamellar body-like forms (LBL) and multi-lamellar forms (ML) were high for "NN" and "NT" samples (38.8% vs. 47.7% for LBL and 58.2% vs. 47.8% for ML). In the "ND" sample, we found virtually no LBL's, ML's (72.6%) as well as uni-lamellar forms (16.4%) and a new structure, the "garland-like" forms (9.4%). Surface tension for "NN" and "NT" was 23.33 ± 0.29 and 25.77 ± 1.12 mN/m, respectively. Dynamic compression-expansion cycling minimum surface tensions were between 0.91 and 1.77 mN/m. CONCLUSION The similarity of surfactant characteristics of nebulized surfactant via a tube and the non-nebulized surfactant suggests that vibrating membrane nebulizers are suitable for surfactant nebulization. Alterations in surfactant morphology and characteristics after nebulization were transient. A new structural subtype of surfactant was identified.
Collapse
Affiliation(s)
- Stefan Minocchieri
- The University of Western Australia, Centre for Neonatal Research and Education, Perth, Australia; Department of Neonatology and Intensive Care, University Children's Hospital, Zurich, Switzerland; Department of Pediatrics, University Hospital Bern, Bern, Switzerland
| | | | | | | | | |
Collapse
|
22
|
Sinha S, Tin W. Adjunctive drug therapies for treatment of respiratory diseases in the newborn: based on evidence or habit? Ther Adv Respir Dis 2014; 8:53-62. [PMID: 24670391 DOI: 10.1177/1753465814526444] [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: 11/16/2022] Open
Abstract
Respiratory distress syndrome is a disease of prematurity and is caused by a relative deficiency of endogenous surfactant production. Respiratory distress syndrome is the most common cause of mortality and morbidity in the newborn population and the standard of care is to provide exogenous surfactant therapy. This saves lives and reduces respiratory complications but, despite treatment, a significant proportion of these infants go onto develop chronic lung disease, the severest form of which is bronchopulmonary dysplasia. Once developed, this is a multisystem disease and treatment is mostly supportive by using various therapeutic adjuncts. Some of these have been proven to be safe and effective in large randomized, controlled trials but similar evidence for other drugs is lacking. The aim of this paper is to provide an overview and critically appraise the available scientific evidence for or against their use in routine practice.
Collapse
Affiliation(s)
- Sunil Sinha
- University of Durham & The James Cook University Hospital, Department of Neonatal Medicine, Marton Road, Middlesbrough TS4 3BW, UK
| | | |
Collapse
|
23
|
Kültürsay N, Uygur Ö, Yalaz M. The use of surfactant in the neonatal period- the known aspects, those still under research and those which need to be investigated further. TURK PEDIATRI ARSIVI 2014; 49:1-12. [PMID: 26078625 PMCID: PMC4462258 DOI: 10.5152/tpa.2014.963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2013] [Accepted: 04/18/2013] [Indexed: 11/22/2022]
Abstract
Respiratory distress syndrome is pulmoner insufficiency caused by the lack of surfactant and the main reason of morbidity and mortality in preterm infants. Mothers at high risk of preterm birth should be transferred to perinatal centers with experience for respiratory distress syndrome and ante-natal steroids should be given before 35 weeks' of gestational age. Surfactant treatment should be applied to babies with or at high risk for respiratory distress syndrome. Prophylaxis should be given to infants of <26 weeks of gestational age and to infants requiring entubation in the delivery room. Nasal continuous positive airway pressure should be considered in infants with complete steroid treatment and without entubation need. Early surfactant may be given if entubation is performed during follow-up. Natural forms of surfactant should be preferred when needed. If the infant is stable, early extubation and non-invasive respiratory support should be considered. In this review, the recent studies' current data about surfactant treatment will be discussed.
Collapse
Affiliation(s)
- Nilgün Kültürsay
- Department of Pediatrics, Division of Neonatology, Ege University Faculty of Medicine, İzmir, Turkey
| | - Özgün Uygur
- Department of Pediatrics, Division of Neonatology, Ege University Faculty of Medicine, İzmir, Turkey
| | - Mehmet Yalaz
- Department of Pediatrics, Division of Neonatology, Ege University Faculty of Medicine, İzmir, Turkey
| |
Collapse
|
24
|
Mazela J, Chmura K, Kulza M, Henderson C, Gregory TJ, Moskal A, Sosnowski TR, Florek E, Kramer L, Keszler M. Aerosolized Albuterol Sulfate Delivery under Neonatal Ventilatory Conditions: In Vitro Evaluation of a Novel Ventilator Circuit Patient Interface Connector. J Aerosol Med Pulm Drug Deliv 2014; 27:58-65. [DOI: 10.1089/jamp.2012.0992] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Affiliation(s)
- Jan Mazela
- Department of Neonatology, Poznan University of Medical Sciences, Poznan, Poland
- Discovery Laboratories, Inc., Warrington, PA, USA
| | - Krzysztof Chmura
- Department of Neonatology, Poznan University of Medical Sciences, Poznan, Poland
| | - Maksymilian Kulza
- Laboratory of Environmental Research, Department of Toxicology, Poznan University of Medical Sciences, Poznan, Poland
| | | | | | - Arkadiusz Moskal
- Faculty of Chemical and Process Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Tomasz R. Sosnowski
- Faculty of Chemical and Process Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Ewa Florek
- Laboratory of Environmental Research, Department of Toxicology, Poznan University of Medical Sciences, Poznan, Poland
| | - Lucyna Kramer
- Department of Computer Science and Statistics, Poznan University of Medical Sciences, Poznan, Poland
| | - Martin Keszler
- Women and Infant Hospital, Brown University, Providence, RI, USA
| |
Collapse
|
25
|
Lopez E, Gascoin G, Flamant C, Merhi M, Tourneux P, Baud O. Exogenous surfactant therapy in 2013: what is next? Who, when and how should we treat newborn infants in the future? BMC Pediatr 2013; 13:165. [PMID: 24112693 PMCID: PMC3851818 DOI: 10.1186/1471-2431-13-165] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 09/19/2013] [Indexed: 11/10/2022] Open
Abstract
Background Surfactant therapy is one of the few treatments that have dramatically changed clinical practice in neonatology. In addition to respiratory distress syndrome (RDS), surfactant deficiency is observed in many other clinical situations in term and preterm infants, raising several questions regarding the use of surfactant therapy. Objectives This review focuses on several points of interest, including some controversial or confusing topics being faced by clinicians together with emerging or innovative concepts and techniques, according to the state of the art and the published literature as of 2013. Surfactant therapy has primarily focused on RDS in the preterm newborn. However, whether this treatment would be of benefit to a more heterogeneous population of infants with lung diseases other than RDS needs to be determined. Early trials have highlighted the benefits of prophylactic surfactant administration to newborns judged to be at risk of developing RDS. In preterm newborns that have undergone prenatal lung maturation with steroids and early treatment with continuous positive airway pressure (CPAP), the criteria for surfactant administration, including the optimal time and the severity of RDS, are still under discussion. Tracheal intubation is no longer systematically done for surfactant administration to newborns. Alternative modes of surfactant administration, including minimally-invasive and aerosolized delivery, could thus allow this treatment to be used in cases of RDS in unstable preterm newborns, in whom the tracheal intubation procedure still poses an ethical and medical challenge. Conclusion The optimization of the uses and methods of surfactant administration will be one of the most important challenges in neonatal intensive care in the years to come.
Collapse
Affiliation(s)
- Emmanuel Lopez
- Réanimation et Pédiatrie Néonatales, Groupe Hospitalier Robert Debré, APHP, 48 Bd Sérurier, Paris, 75019, France.
| | | | | | | | | | | | | |
Collapse
|
26
|
Pohlmann G, Iwatschenko P, Koch W, Windt H, Rast M, de Abreu MG, Taut FJH, De Muynck C. A novel continuous powder aerosolizer (CPA) for inhalative administration of highly concentrated recombinant surfactant protein-C (rSP-C) surfactant to preterm neonates. J Aerosol Med Pulm Drug Deliv 2013; 26:370-9. [PMID: 23421901 DOI: 10.1089/jamp.2012.0996] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND In pulmonary medicine, aerosolization of substances for continuous inhalation is confined to different classes of nebulizers with their inherent limitations. Among the unmet medical needs is the lack of an aerosolized surfactant preparation for inhalation by preterm neonates, to avoid the risks associated with endotracheal intubation and surfactant bolus instillation. In the present report, we describe a high-concentration continuous powder aerosolization system developed for delivery of inhalable surfactant to preterm neonates. METHODS The developed device uses a technique that allows efficient aerosolization of dry surfactant powder, generating a surfactant aerosol of high concentration. In a subsequent humidification step, the heated aerosol particles are covered with a surface layer of water. The wet surfactant aerosol is then delivered to the patient interface (e.g., nasal prongs) through a tube. RESULTS The performance characteristics of the system are given as mass concentration, dose rate, and size distribution of the generated aerosol. Continuous aerosol flows of about 0.84 L/min can be generated from dry recombinant surfactant protein-C surfactant, with concentrations of up to 12 g/m(3) and median particle sizes of the humidified particles in the range of 3 to 3.5 μm at the patient interface. The system has been successfully used in preclinical studies. CONCLUSION The device with its continuous high-concentration delivery is promising for noninvasive delivery of surfactant aerosol to neonates and has the potential for becoming a versatile disperser platform closing the gap between continuously operating nebulizers and discontinuously operating dry powder inhaler devices.
Collapse
Affiliation(s)
- G Pohlmann
- 1 Fraunhofer Institute for Toxicology and Experimental Medicine ITEM , D-30625 Hannover, Germany
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Kanmaz HG, Erdeve O, Canpolat FE, Mutlu B, Dilmen U. Surfactant administration via thin catheter during spontaneous breathing: randomized controlled trial. Pediatrics 2013; 131:e502-9. [PMID: 23359581 DOI: 10.1542/peds.2012-0603] [Citation(s) in RCA: 181] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND The primary aim of this randomized study was to describe the feasibility of early administration of surfactant via a thin catheter during spontaneous breathing (Take Care) and compare early mechanical ventilation (MV) requirement with the InSurE (Intubate, Surfactant, Extubate) procedure. METHODS Preterm infants, who were <32 weeks and stabilized with nasal continuous positive airway pressure (nCPAP) in the delivery room, were randomized to receive early surfactant treatment either by the Take Care or InSurE technique. Tracheal instillation of 100 mg/kg poractant α via 5-F catheter during spontaneous breathing under nCPAP was performed in the intervention group. In the InSurE procedure, infants were intubated, received positive pressure ventilation for 30 seconds after surfactant instillation, and placed on nCPAP immediately. RESULTS One hundred infants in each group were analyzed. The MV requirement in the first 72 hours of life was significantly lower in the Take Care group when compared with the InSurE group (30% vs 45%, P = .02, odds ratio -0.52, 95% confidence interval -0.94 to -0.29). Mean duration of both nCPAP and MV were significantly shorter in the Take Care group (P values .006 and .002, respectively). Bronchopulmonary dysplasia rate was significantly lower among the infants treated with the Take Care technique (relative risk -0.27, 95% confidence interval -0.1 to -0.72) CONCLUSIONS The Take Care technique is feasible for the treatment of respiratory distress syndrome in infants with very low birth weight. It significantly reduces both the need and duration of MV, and thus the bronchopulmonary dysplasia rate in preterm infants.
Collapse
Affiliation(s)
- H Gozde Kanmaz
- Zekai Tahir Burak Kadin Sagligi Egitim ve Arastirma Hastanesi, Talatpasa Bulvari, Samanpazari, Ankara; Turkey 06230.
| | | | | | | | | |
Collapse
|
28
|
Novel approaches to surfactant administration. Crit Care Res Pract 2012; 2012:278483. [PMID: 23243504 PMCID: PMC3518953 DOI: 10.1155/2012/278483] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 11/19/2012] [Indexed: 11/17/2022] Open
Abstract
Surfactant replacement therapy has been the mainstay of treatment for preterm infants with respiratory distress syndrome for more than twenty years. For the most part, surfactant is administered intratracheally, followed by mechanical ventilation. In recent years, the growing interest in noninvasive ventilation has led to novel approaches of administration. This paper will review these techniques and the associated clinical evidence.
Collapse
|
29
|
Abdel-Latif ME, Osborn DA. Nebulised surfactant in preterm infants with or at risk of respiratory distress syndrome. Cochrane Database Syst Rev 2012; 10:CD008310. [PMID: 23076945 DOI: 10.1002/14651858.cd008310.pub2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Nebulised surfactant has the potential to deliver surfactant to the infant lung with the goal of avoiding endotracheal intubation and ventilation, ventilator-induced lung injury and bronchopulmonary dysplasia (BPD). OBJECTIVES To determine the effect of nebulised surfactant administration either as prophylaxis or treatment compared to placebo, no treatment or intratracheal surfactant administration on morbidity and mortality in preterm infants with, or at risk of, respiratory distress syndrome (RDS). SEARCH METHODS Searches were performed of CENTRAL (The Cochrane Library, January 2012), MEDLINE and PREMEDLINE (1950 to January 2012), EMBASE (1980 to January 2012) and CINAHL (1982 to January 2012), as well as proceedings of scientific meetings, clinical trial registries, Google Scholar and reference lists of identified studies. Expert informants and surfactant manufacturers were contacted. SELECTION CRITERIA Randomised, cluster-randomised or quasi-randomised controlled trials of nebulised surfactant administration compared to placebo, no treatment, or other routes of administration (laryngeal, pharyngeal instillation of surfactant before the first breath, thin endotracheal catheter surfactant administration or intratracheal surfactant instillation) on morbidity and mortality in preterm infants at risk of RDS. We considered published, unpublished and ongoing trials. DATA COLLECTION AND ANALYSIS Two review authors independently assessed studies for eligibility and quality, and extracted data. MAIN RESULTS No studies of prophylactic or early nebulised surfactant administration were found. A single small study of late rescue nebulised surfactant was included. The study is of moderate risk of bias. The study enrolled 32 preterm infants born < 36 weeks' gestation with RDS on nasal continuous positive airway pressure (nCPAP). The study reported no significant difference between nebulised surfactant administration compared to no treatment groups in chronic lung disease (risk ratio (RR) 5.00; 95% confidence interval (CI) 0.26 to 96.59) or other outcomes (oxygenation 1 to 12 hours after randomisation, need for mechanical ventilation, days of mechanical ventilation or continuous positive airways pressure (CPAP) or days of supplemental oxygen). No side effects of the nebulised surfactant therapy or aerosol inhalation were reported. AUTHORS' CONCLUSIONS There are insufficient data to support or refute the use of nebulised surfactant in clinical practice. Adequately powered trials are required to determine the effect of nebulised surfactant administration for prevention or early treatment of RDS in preterm infants. Nebulised surfactant administration should be limited to clinical trials.
Collapse
Affiliation(s)
- Mohamed E Abdel-Latif
- Department of Neonatology, Australian National University Medical School, Woden, Australia.
| | | |
Collapse
|
30
|
Shaffer TH, Alapati D, Greenspan JS, Wolfson MR. Neonatal non-invasive respiratory support: physiological implications. Pediatr Pulmonol 2012; 47:837-47. [PMID: 22777738 PMCID: PMC3762325 DOI: 10.1002/ppul.22610] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 05/16/2012] [Indexed: 11/10/2022]
Abstract
The introduction of assisted ventilation for neonatal pulmonary insufficiency has resulted in the successful treatment of many previously fatal diseases. During the past three decades, refinement of invasive mechanical ventilation techniques has dramatically improved survival of many high-risk neonates. However, as with many advances in medicine, while mortality has been reduced, morbidity has increased in the surviving high-risk neonate. In this regard, introduction of assisted ventilation has been associated with chronic lung injury, also known as bronchopulmonary dysplasia. This disease, unknown prior to the appearance of mechanical ventilation, has produced a population of patients characterized by ventilator or oxygen dependence with serious accompanying pulmonary and neurodevelopmental morbidity. The purpose of this article is to review non-invasive respiratory support methodologies to address the physiologic mechanisms by which these methods may prevent the pathophysiologic effects of invasive mechanical ventilation.
Collapse
Affiliation(s)
- Thomas H Shaffer
- Nemours Center for Pediatric Lung Research, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA.
| | | | | | | |
Collapse
|
31
|
Pfister RH, Soll RF. Initial respiratory support of preterm infants: the role of CPAP, the INSURE method, and noninvasive ventilation. Clin Perinatol 2012; 39:459-81. [PMID: 22954263 DOI: 10.1016/j.clp.2012.06.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
This article explores the potential benefits and risks for the various approaches to the initial respiratory management of preterm infants. The authors focus on the evidence for the increasingly used strategies of initial respiratory support of preterm infants with continuous positive airway pressure (CPAP) beginning in the delivery room or very early in the hospital course and blended strategies involving the early administration of surfactant replacement followed by immediate extubation and stabilization on CPAP. Where possible, the evidence referenced in this review comes from individual randomized controlled trials or meta-analyses of those trials.
Collapse
Affiliation(s)
- Robert H Pfister
- Department of Pediatrics, University of Vermont, FAHC-Smith 556, Burlington, VT 05401, USA.
| | | |
Collapse
|
32
|
Jargin SV. Surfactant preparations for tuberculosis and other diseases beyond infancy: a letter from Russia. Tuberculosis (Edinb) 2012; 92:280-2. [PMID: 22410300 DOI: 10.1016/j.tube.2012.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 02/10/2012] [Indexed: 11/30/2022]
Affiliation(s)
- Sergei V Jargin
- Peoples' Friendship University of Russia Clementovski per 6-82, 115184 Moscow, Russia
| |
Collapse
|
33
|
Zierenberg JR, Halpern D, Filoche M, Sapoval B, Grotberg JB. An asymptotic model of particle deposition at an airway bifurcation. MATHEMATICAL MEDICINE AND BIOLOGY-A JOURNAL OF THE IMA 2012; 30:131-56. [PMID: 22378463 DOI: 10.1093/imammb/dqs002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Particle transport and deposition associated with flow over a wedge is investigated as a model for particle transport and flow at the carina of an airway bifurcation during inspiration. Using matched asymptotics, a uniformly valid solution is obtained to represent the high Reynolds number flow over a wedge that considers the viscous boundary layer near the wedge and the outer inviscid region and is then used to solve the particle transport equations. Sometimes particle impaction on the wedge is prevented due to the boundary layer. We call this boundary layer shielding (BLS). This effect can be broken down into different types: rejection, trapping and deflection that are described by what happens to the particle's initial negative velocity normal to the wall either changing sign, reaching zero, or remaining negative in the boundary layer region. The deposition efficiency depends on the critical Stokes number but exhibits a weak dependence on Reynolds number. Deposition efficiency for S(c) in the range 0 < S(c) < 0.4 yields the following relationship De ≈ (1.867S(c)¹·⁷⁸-0.016) sin(βπ/2) at large Reynolds numbers, where βπ is the wedge angle. For a specific deposition efficiency, S(c) decreases as βπ increases. The distribution of impacted particles was also computed and revealed that particles primarily impact within one airway diameter of the carina, consistent with computational fluid dynamics approaches. This work provides a new insight that the BLS inherent to the wedge component of the structure is the dominant reason for the particle distribution. This finding is important in linking aerosol deposition to the location of airway disease as well as target sites for therapeutic deposition.
Collapse
Affiliation(s)
- Jennifer R Zierenberg
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | |
Collapse
|
34
|
Finer NN, Merritt TA, Bernstein G, Job L, Mazela J, Segal R. An open label, pilot study of Aerosurf® combined with nCPAP to prevent RDS in preterm neonates. J Aerosol Med Pulm Drug Deliv 2011; 23:303-9. [PMID: 20455772 DOI: 10.1089/jamp.2009.0758] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Nasal continuous positive airway pressure (nCPAP) is an accepted mode of respiratory support for preterm infants with respiratory insufficiency. To avoid potential sequelae of endotracheal (ET) intubation and mechanical ventilation, prophylactic aerosolization of surfactant delivered via nCPAP has been attempted with limited success. METHODS To determine the feasibility and safety of prophylactic aerosolization of a peptide-containing synthetic surfactant, Aerosurf® (lucinactant for inhalation) was delivered by nCPAP to preterm infants at risk for respiratory distress syndrome (RDS). Neonates were enrolled into treatment group 1 (Aerosurf retreatment separated by at least 3 h) or treatment group 2 (Aerosurf retreatment separated by at least 1 h). A vibrating membrane nebulizer Aeroneb Pro® was used to aerosolize 20 mg/mL Aerosurf. All neonates received the initial 3-h treatment, and three retreatments were permitted within 48 h based on clinical response. RESULTS Seventeen infants were enrolled. Aerosurf was well tolerated, with transient desaturations observed during dosing without bradycardia or hypotension. Variability in output rates of the Aeroneb Pro was observed leading to different average dispensed drug volumes per treatment per patient. All infants survived; 29.4% required subsequent ET surfactant replacement therapy, 23.5% were diagnosed with RDS at 24 h, and 11.8% with bronchopulmonary dysplasia (BPD) at 28 days of life. Mean FiO₂ was 0.4 at baseline, and 0.32 at 4 h posttreatment. CONCLUSIONS Aerosurf can be safely administered via nCPAP in preterm infants at risk for RDS and may provide an alternative to surfactant administration via an ET tube. Further studies are required to evaluate this delivery approach.
Collapse
Affiliation(s)
- Neil N Finer
- Department of Pediatrics, University of California San Diego, San Diego, California 92103, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Shah S. Exogenous surfactant: intubated present, nebulized future? World J Pediatr 2011; 7:11-5. [PMID: 20549420 DOI: 10.1007/s12519-010-0201-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Accepted: 05/25/2009] [Indexed: 11/29/2022]
Abstract
BACKGROUND exogenous surfactant is currently administered via intra-tracheal instillation, a method which can increase the possibility of clinical instability in the peri-surfactant administration period. Since its introduction, there has been an increase in understanding of the pathology of respiratory distress syndrome and surfactant biology. This includes development of a potential nebulized surfactant which has the potential to increase the number, safety and timely administration of the medication in preterm infants. DATA SOURCES based on recent original publications in the field of surfactant biology, we reviewed our experience with surfactant administration and discussed the available evidence on nebulized surfactant and outlined potential barriers toward widespread introduction of this therapy. RESULTS surfactant has revolutionized modern neonatal management and nebulized surfactant is attractive and a vector for administration. However, issues regarding costeffectiveness, development of nebulizer devices capable of administration, deposition of medication in the airway and dosing strategies remain unresolved. CONCLUSIONS nebulized surfactant has the potential to be a therapeutic breakthrough by eliminating the potent volu-and-baro-traumatic effects of mechanical ventilation in the peri-surfactant period. Nebulization would likely lead to increased administration immediately after birth and more emphasis on noninvasive ventilator strategies. These features will aid clinical implementation of nebulized surfactant as a standard of treatment after introduction.
Collapse
Affiliation(s)
- Shetal Shah
- Department of Pediatrics, State University of New York at Stony Brook, Stony Brook, New York 11791, USA.
| |
Collapse
|
36
|
Mazela J, Polin RA. Aerosol delivery to ventilated newborn infants: historical challenges and new directions. Eur J Pediatr 2011; 170:433-44. [PMID: 20878336 PMCID: PMC3059826 DOI: 10.1007/s00431-010-1292-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Accepted: 09/06/2010] [Indexed: 11/30/2022]
Abstract
There are several aerosolized drugs which have been used in the treatment of neonatal respiratory illnesses, such as bronchodilators, diuretics, and surfactants. Preclinical in vitro and in vivo studies identified a number of variables that affect aerosol efficiency, including particle size, aerosol flows, nebulizer choice, and placement. Nevertheless, an optimized aerosol drug delivery system for mechanically ventilated infants still does not exist. Increasing interest in this form of drug delivery requires more controlled and focused research of drug/device combinations appropriate for the neonatal population. In the present article, we review the research that has been conducted thus far and discuss the next steps in developing the optimal aerosol delivery system for use in mechanically ventilated neonates.
Collapse
Affiliation(s)
- Jan Mazela
- Department of Neonatology, Poznan University of Medical Sciences, Poznan, Poland.
| | - Richard A. Polin
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY USA
| |
Collapse
|
37
|
Abstract
BACKGROUND Since the first successful report of surfactant replacement therapy (SRT) in infants with respiratory distress syndrome (RDS), numerous randomized clinical trials have shown that SRT reduces mortality and morbidity in RDS. Surfactant is now a standard therapy for RDS. However, the use of SRT in the developing world has been extremely slow. OBJECTIVE The objective of this paper is to review the published information regarding the usage and barriers encountered in the use of SRT in developing countries. METHODS We reviewed the available literature and also gathered information from countries with a high burden of prematurity and high infant mortality rate regarding replacement therapy and the barriers to use of SRT. RESULTS We reviewed the available literature and found that developing countries bear a high burden of prematurity and RDS that contribute to high neonatal and infant mortality rates. Based on the effectiveness of SRT in RDS, surfactant preparations were included in the Essential Drug List of WHO in 2008. However, the use of SRT in developing countries is still limited because of (1) high cost, (2) lack of skilled personnel to administer SRT, and (3) lack of support systems after the SRT. The cost of SRT may exceed the per-capita GNP (300-500 USD) in some countries. Data from India and South Africa suggests that SRT is limited to rescue therapy in babies with potential for better survival, usually >28 weeks' gestation. Recent studies show that infants with RDS respond well to initial continuous positive airway pressure (CPAP) followed by SRT for those who do not respond. CONCLUSIONS In developing countries, CPAP may be used as the primary mode of management of RDS. SRT may be reserved for non-responders to CPAP. Alternate simpler methods of delivery of surfactant (aerosol technique) are also being explored. There is a need for further studies to develop and assess efficient and less expensive methods of application of CPAP and SRT in developing countries.
Collapse
Affiliation(s)
- Dharmapuri Vidyasagar
- Division of Neonatology, Department of Pediatrics, University of Illinois at Chicago Medical Center, Chicago, IL 60612., USA.
| | | | | |
Collapse
|
38
|
Terry MH, Merritt TA, Harding B, Schroeder H, Merrill-Henry J, Mazela J, Gregory TJ, Segal R, Power GG, Blood AB. Pulmonary distribution of lucinactant and poractant alfa and their peridosing hemodynamic effects in a preterm lamb model of respiratory distress syndrome. Pediatr Res 2010; 68:193-8. [PMID: 20531255 DOI: 10.1203/pdr.0b013e3181eaff66] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Tracheal instillation of surfactant to premature newborns improves their survivability but may transiently obstruct airways resulting in undesirable acute effects on cerebral blood flow (CBF) and oxygenation. The acute peridosing hemodynamic effects of surfactant administration may be avoided by minimizing the volume of surfactant administered, but smaller surfactant volumes may also result in less even distribution of surfactant throughout the lung. These experiments were undertaken to compare responses to two surfactants with different dose volumes (porcine-derived poractant alfa, 2.5 mL/kg vs peptide-based synthetic lucinactant, 5.8 mL/kg) given to newly delivered lambs at 85% gestation. Both surfactants resulted in similar improvements in blood gas values, a doubling of dynamic compliance, increases in brain tissue oxygen tension, and stable blood pressure with no significant change in CBF. Distribution of surfactant throughout the lungs was more uniform with lucinactant than poractant alfa when assessed by labeled microspheres. We conclude that improvements in lung mechanics, gas exchange, and changes in CBF are comparable for a porcine-derived and peptide-containing synthetic surfactant, despite instilled volumes differing by 2-fold. Intrapulmonary distribution of surfactant is more uniform after a larger volume is instilled.
Collapse
Affiliation(s)
- Michael H Terry
- Department of Respiratory Care, Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Barros FC, Bhutta ZA, Batra M, Hansen TN, Victora CG, Rubens CE. Global report on preterm birth and stillbirth (3 of 7): evidence for effectiveness of interventions. BMC Pregnancy Childbirth 2010; 10 Suppl 1:S3. [PMID: 20233384 PMCID: PMC2841444 DOI: 10.1186/1471-2393-10-s1-s3] [Citation(s) in RCA: 171] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
INTRODUCTION Interventions directed toward mothers before and during pregnancy and childbirth may help reduce preterm births and stillbirths. Survival of preterm newborns may also be improved with interventions given during these times or soon after birth. This comprehensive review assesses existing interventions for low- and middle-income countries (LMICs). METHODS Approximately 2,000 intervention studies were systematically evaluated through December 31, 2008. They addressed preterm birth or low birth weight; stillbirth or perinatal mortality; and management of preterm newborns. Out of 82 identified interventions, 49 were relevant to LMICs and had reasonable amounts of evidence, and therefore selected for in-depth reviews. Each was classified and assessed by the quality of available evidence and its potential to treat or prevent preterm birth and stillbirth. Impacts on other maternal, fetal, newborn or child health outcomes were also considered. Assessments were based on an adaptation of the Grades of Recommendation Assessment, Development and Evaluation criteria. RESULTS Most interventions require additional research to improve the quality of evidence. Others had little evidence of benefit and should be discontinued. The following are supported by moderate- to high-quality evidence and strongly recommended for LMICs: Two interventions prevent preterm births--smoking cessation and progesterone. Eight interventions prevent stillbirths--balanced protein energy supplementation, screening and treatment of syphilis, intermittant presumptive treatment for malaria during pregnancy, insecticide-treated mosquito nets, birth preparedness, emergency obstetric care, cesarean section for breech presentation, and elective induction for post-term delivery. Eleven interventions improve survival of preterm newborns--prophylactic steroids in preterm labor, antibiotics for PROM, vitamin K supplementation at delivery, case management of neonatal sepsis and pneumonia, delayed cord clamping, room air (vs. 100% oxygen) for resuscitation, hospital-based kangaroo mother care, early breastfeeding, thermal care, and surfactant therapy and application of continued distending pressure to the lungs for respiratory distress syndrome CONCLUSION The research paradigm for discovery science and intervention development must be balanced to address prevention as well as improve morbidity and mortality in all settings. This review also reveals significant gaps in current knowledge of interventions spanning the continuum of maternal and fetal outcomes, and the critical need to generate further high-quality evidence for promising interventions.
Collapse
Affiliation(s)
- Fernando C Barros
- Post-Graduate Course in Health and Behaviour, Universidade Catolica de Pelotas, Brazil
| | | | - Maneesh Batra
- Divison of Neonatology, Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA
| | | | | | - Craig E Rubens
- Global Alliance to Prevent Prematurity and Stillbirth, an initiative of Seattle Children's, Seattle, Washington, USA
- Department of Pediatrics at University of Washington School of Medicine, Seattle, Washington, USA
| | | |
Collapse
|
40
|
Abdel-Latif ME, Osborn DA. Nebulised surfactant for prevention of morbidity and mortality in preterm infants with or at risk of respiratory distress syndrome. THE COCHRANE DATABASE OF SYSTEMATIC REVIEWS 2010. [DOI: 10.1002/14651858.cd008310] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
41
|
de Winter JP, de Vries MAG, Zimmermann LJI. Clinical practice : noninvasive respiratory support in newborns. Eur J Pediatr 2010; 169:777-82. [PMID: 20179966 PMCID: PMC2876262 DOI: 10.1007/s00431-010-1159-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2009] [Accepted: 01/27/2010] [Indexed: 11/04/2022]
Abstract
The most important goal of introducing noninvasive ventilation (NIV) has been to decrease the need for intubation and, therefore, mechanical ventilation in newborns. As a result, this technique may reduce the incidence of bronchopulmonary dysplasia (BPD). In addition to nasal CPAP, improvements in sensors and flow delivery systems have resulted in the introduction of a variety of other types of NIV. For the optimal application of these novelties, a thorough physiological knowledge of mechanics of the respiratory system is necessary. In this overview, the modern insights of noninvasive respiratory therapy in newborns are discussed. These aspects include respiratory support in the delivery room; conventional and modern nCPAP; humidified, heated, and high-flow nasal cannula ventilation; and nasal intermittent positive pressure ventilation. Finally, an algorithm is presented describing common practice in taking care of respiratory distress in prematurely born infants.
Collapse
Affiliation(s)
| | | | - Luc J. I. Zimmermann
- Department of Pediatrics, Research School Oncology and Developmental Biology-grow, Maastricht University Medical Hospital, Maastricht, The Netherlands
| |
Collapse
|
42
|
Sun Y, Yang R, Zhong JG, Fang F, Jiang JJ, Liu MY, Lu J. Aerosolised surfactant generated by a novel noninvasive apparatus reduced acute lung injury in rats. Crit Care 2009; 13:R31. [PMID: 19257907 PMCID: PMC2689462 DOI: 10.1186/cc7737] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2008] [Revised: 02/23/2009] [Accepted: 03/04/2009] [Indexed: 11/18/2022] Open
Abstract
Introduction Exogenous surfactant has been explored as a potential therapy for acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). In the present study, a nebuliser driven by oxygen lines found in the hospital was developed to deliver aerosolised porcine pulmonary surfactant (PPS). We hypothesised that aerosolised surfactant inhaled through spontaneous breathing may effectively reduce severe lung injury. Methods Rats were intravenously injected with oleic acid (OA) to induce ALI and 30 minutes later they were divided into five groups: model (injury only), PPS aerosol (PPS-aer), saline aerosol (saline-aer), PPS instillation (PPS-inst), and saline instillation (Saline-Inst). Blood gases, lung histology, and protein and TNF-α concentrations in the bronchoalveolar lavage fluid (BALF) were examined. Results The PPS aerosol particles were less than 2.0 μm in size as determined by a laser aerosol particle counter. Treatment of animals with a PPS aerosol significantly increased the phospholipid content in the BALF, improved lung function, reduced pulmonary oedema, decreased total protein and TNF-α concentrations in BALF, ameliorated lung injury and improved animal survival. These therapeutic effects are similar to those seen in the PPS-inst group. Conclusions This new method of PPS aerosolisation combines the therapeutic effects of a surfactant with partial oxygen inhalation under spontaneous breathing. It is an effective, simple and safe method of administering an exogenous surfactant.
Collapse
Affiliation(s)
- Yu Sun
- Department of Pathophysiology, College of Basic Medical Sciences, Second Military Medical University, 800 Xiangyin Road, Shanghai, 200433, China
| | | | | | | | | | | | | |
Collapse
|
43
|
Lal MK, Sinha SK. Review: Surfactant respiratory therapy using Surfaxin/sinapultide. Ther Adv Respir Dis 2008; 2:339-44. [DOI: 10.1177/1753465808097113] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Respiratory distress syndrome (RDS) is a leading cause of mortality and morbidity in preterm infants. Surfactant replacement therapy has been widely used to prevent and treat RDS in these newborns and has now become a standard of care. First-generation synthetic surfactants such as Exosurf did not contain any surfactant protein. This disadvantage was overcome with animal-derived surfactant preparations which contain specific proteins but has the limitation of being derived from animal sources. This has led to development of newer synthetic surfactants such as lucinactant (Surfaxin, Discovery Laboratories, Philadelphia) which contains the protein B mimic synthetic peptide, sinapultide. Recent phase 3 clinical trials with Surfaxin show promising results with similar efficacy as animal derived surfactants and yet avoiding the disadvantage associated with animal products. The purpose of this paper is to summarise results of recent clinical trials of Surfaxin use in newborns with RDS.
Collapse
Affiliation(s)
| | - Sunil K. Sinha
- The James Cook University Hospital, Marton Road, Middlesbrough,
| |
Collapse
|
44
|
Minocchieri S, Burren JM, Bachmann MA, Stern G, Wildhaber J, Buob S, Schindel R, Kraemer R, Frey UP, Nelle M. Development of the premature infant nose throat-model (PrINT-Model): an upper airway replica of a premature neonate for the study of aerosol delivery. Pediatr Res 2008; 64:141-6. [PMID: 18391845 DOI: 10.1203/pdr.0b013e318175dcfa] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Clinical efficacy of aerosol therapy in premature newborns depends on the efficiency of delivery of aerosolized drug to the bronchial tree. To study the influence of various anatomical, physical, and physiological factors on aerosol delivery in preterm newborns, it is crucial to have appropriate in vitro models, which are currently not available. We therefore constructed the premature infant nose throat-model (PrINT-Model), an upper airway model corresponding to a premature infant of 32-wk gestational age by three-dimensional (3D) reconstruction of a three-planar magnetic resonance imaging scan and subsequent 3D-printing. Validation was realized by visual comparison and comparison of total airway volume. To study the feasibility of measuring aerosol deposition, budesonide was aerosolized through the cast and lung dose was expressed as percentage of nominal dose. The airway volumes of the initial magnetic resonance imaging and validation computed tomography scan showed a relative deviation of 0.94%. Lung dose at low flow (1 L/min) was 61.84% and 9.00% at high flow (10 L/min), p < 0.0001. 3D-reconstruction provided an anatomically accurate surrogate of the upper airways of a 32-wk-old premature infant, making the model suitable for future in vitro testing.
Collapse
Affiliation(s)
- Stefan Minocchieri
- Department of Paediatrics, Division of Neonatology, University of Bern, Inselspital, Bern, BE, Switzerland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Affiliation(s)
- Vineet Bhandari
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
46
|
Abstract
PURPOSE OF REVIEW To present recent data on the role of noninvasive ventilation in the respiratory management of newborn infants. RECENT FINDINGS Noninvasive ventilation is growing in popularity but is applied using widely varying devices and settings. Although short-term physiological advantages were reported for bubble and variable-flow continuous positive airways pressure, neither has convincingly shown superior clinically important outcomes. Continuous positive airways pressure may be used as the initial mode of support for very preterm infants but increased rates of pneumothorax in infants not receiving surfactant are a concern. Methods of administering surfactant without endotracheal intubation deserve further study. Nasal intermittent positive-pressure ventilation shows promise as a primary treatment for respiratory distress syndrome. Optimal pressure settings for continuous positive airways pressure and nasal intermittent positive-pressure ventilation remain uncertain. SUMMARY Noninvasive ventilation has partially fulfilled its promise as a gentler alternative to ventilation via an endotracheal tube. Appropriately designed randomized clinical trials are required to determine the best nasal interfaces and pressure generators.
Collapse
|
47
|
Sinha SK, Gupta S, Donn SM. Immediate respiratory management of the preterm infant. Semin Fetal Neonatal Med 2008; 13:24-9. [PMID: 17981103 DOI: 10.1016/j.siny.2007.09.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Infants born prematurely have underdeveloped lungs characterised by both morphological and biochemical abnormalities. Respiratory distress syndrome (RDS) is the leading cause of morbidity and mortality in this population. Both surfactant replacement therapy with mechanical ventilation and continuous positive airway pressure (CPAP) have been shown to be of benefit. However, considerable controversy exists about how best to use these therapies. This paper will review the pathophysiology of RDS and the evidence supporting each of these treatments.
Collapse
Affiliation(s)
- Sunil K Sinha
- Paediatrics and Neonatal Medicine, University of Durham, Durham, UK.
| | | | | |
Collapse
|
48
|
Donn SM, Sinha SK. Aerosolized lucinactant: a potential alternative to intratracheal surfactant replacement therapy. Expert Opin Pharmacother 2008; 9:475-8. [DOI: 10.1517/14656566.9.3.475] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
49
|
Sinha SK. USE OF SURFACTANTS. J Pediatr Pharmacol Ther 2007; 12:266-8. [DOI: 10.5863/1551-6776-12.4.266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Sunil K. Sinha
- Professor of Paediatrics and Neonatal Medicine, University of Durham & The James Cook University Hospital, Middlesbrough, United Kingdom,
| |
Collapse
|