1
|
Dermitzaki N, Baltogianni M, Tsekoura E, Giapros V. Invasive Candida Infections in Neonatal Intensive Care Units: Risk Factors and New Insights in Prevention. Pathogens 2024; 13:660. [PMID: 39204260 PMCID: PMC11356907 DOI: 10.3390/pathogens13080660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/25/2024] [Accepted: 08/05/2024] [Indexed: 09/03/2024] Open
Abstract
Invasive Candida infections represent a significant cause of morbidity and mortality in neonatal intensive care units (NICUs), with a particular impact on preterm and low-birth-weight neonates. In addition to prematurity, several predisposing factors for Candida colonization and dissemination during NICU hospitalization have been identified, including prolonged exposure to broad-spectrum antibiotics, central venous catheters, parenteral nutrition, corticosteroids, H2 antagonist administration, and poor adherence to infection control measures. According to the literature, the implementation of antifungal prophylaxis, mainly fluconazole, in high-risk populations has proven to be an effective strategy in reducing the incidence of fungal infections. This review aims to provide an overview of risk factors for invasive Candida infections and current perspectives regarding antifungal prophylaxis use. Recognizing and reducing people's exposure to these modifiable risk factors, in conjunction with the administration of antifungal prophylaxis, has been demonstrated to be an effective method for preventing invasive candidiasis in susceptible neonatal populations.
Collapse
Affiliation(s)
- Niki Dermitzaki
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, 45500 Ioannina, Greece; (N.D.); (M.B.)
| | - Maria Baltogianni
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, 45500 Ioannina, Greece; (N.D.); (M.B.)
| | - Efrosini Tsekoura
- Paediatric Department, Asklepieion Voula’s General Hospital, 16673 Athens, Greece;
| | - Vasileios Giapros
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, 45500 Ioannina, Greece; (N.D.); (M.B.)
| |
Collapse
|
2
|
Marissen J, Reichert L, Härtel C, Fortmann MI, Faust K, Msanga D, Harder J, Zemlin M, Gomez de Agüero M, Masjosthusmann K, Humberg A. Antimicrobial Peptides (AMPs) and the Microbiome in Preterm Infants: Consequences and Opportunities for Future Therapeutics. Int J Mol Sci 2024; 25:6684. [PMID: 38928389 PMCID: PMC11203687 DOI: 10.3390/ijms25126684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/07/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Antimicrobial peptides (AMPs) are crucial components of the innate immune system in various organisms, including humans. Beyond their direct antimicrobial effects, AMPs play essential roles in various physiological processes. They induce angiogenesis, promote wound healing, modulate immune responses, and serve as chemoattractants for immune cells. AMPs regulate the microbiome and combat microbial infections on the skin, lungs, and gastrointestinal tract. Produced in response to microbial signals, AMPs help maintain a balanced microbial community and provide a first line of defense against infection. In preterm infants, alterations in microbiome composition have been linked to various health outcomes, including sepsis, necrotizing enterocolitis, atopic dermatitis, and respiratory infections. Dysbiosis, or an imbalance in the microbiome, can alter AMP profiles and potentially lead to inflammation-mediated diseases such as chronic lung disease and obesity. In the following review, we summarize what is known about the vital role of AMPs as multifunctional peptides in protecting newborn infants against infections and modulating the microbiome and immune response. Understanding their roles in preterm infants and high-risk populations offers the potential for innovative approaches to disease prevention and treatment.
Collapse
Affiliation(s)
- Janina Marissen
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; (J.M.); (L.R.)
- Würzburg Institute of Systems Immunology, Max-Planck Research Group, University of Würzburg, 97078 Würzburg, Germany;
| | - Lilith Reichert
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; (J.M.); (L.R.)
| | - Christoph Härtel
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; (J.M.); (L.R.)
- German Center for Infection Research, Site Hamburg-Lübeck-Borstel-Riems, 23538 Lübeck, Germany
| | - Mats Ingmar Fortmann
- Department of Pediatrics, University Hospital Schleswig-Holstein, 23538 Lübeck, Germany; (M.I.F.); (K.F.)
| | - Kirstin Faust
- Department of Pediatrics, University Hospital Schleswig-Holstein, 23538 Lübeck, Germany; (M.I.F.); (K.F.)
| | - Delfina Msanga
- Department of Pediatrics, Bugando Hospital, Catholic University of Health and Allied Sciences, Mwanza 33109, Tanzania;
| | - Jürgen Harder
- Department of Dermatology, Venerology and Allergology, Quincke Research Center, Kiel University, 24105 Kiel, Germany;
| | - Michael Zemlin
- Department of General Pediatrics and Neonatology, Saarland University Medical Center, 66421 Homburg, Germany;
| | - Mercedes Gomez de Agüero
- Würzburg Institute of Systems Immunology, Max-Planck Research Group, University of Würzburg, 97078 Würzburg, Germany;
| | - Katja Masjosthusmann
- Department of General Pediatrics, University Children’s Hospital Münster, 48149 Münster, Germany; (K.M.); (A.H.)
| | - Alexander Humberg
- Department of General Pediatrics, University Children’s Hospital Münster, 48149 Münster, Germany; (K.M.); (A.H.)
| |
Collapse
|
3
|
Eker F, Akdaşçi E, Duman H, Yalçıntaş YM, Canbolat AA, Kalkan AE, Karav S, Šamec D. Antimicrobial Properties of Colostrum and Milk. Antibiotics (Basel) 2024; 13:251. [PMID: 38534686 DOI: 10.3390/antibiotics13030251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024] Open
Abstract
The growing number of antibiotic resistance genes is putting a strain on the ecosystem and harming human health. In addition, consumers have developed a cautious attitude towards chemical preservatives. Colostrum and milk are excellent sources of antibacterial components that help to strengthen the immunity of the offspring and accelerate the maturation of the immune system. It is possible to study these important defenses of milk and colostrum, such as lactoferrin, lysozyme, immunoglobulins, oligosaccharides, etc., as biotherapeutic agents for the prevention and treatment of numerous infections caused by microbes. Each of these components has different mechanisms and interactions in various places. The compound's mechanisms of action determine where the antibacterial activity appears. The activation of the antibacterial activity of milk and colostrum compounds can start in the infant's mouth during lactation and continue in the gastrointestinal regions. These antibacterial properties possess potential for therapeutic uses. In order to discover new perspectives and methods for the treatment of bacterial infections, additional investigations of the mechanisms of action and potential complexes are required.
Collapse
Affiliation(s)
- Furkan Eker
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Turkey
| | - Emir Akdaşçi
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Turkey
| | - Hatice Duman
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Turkey
| | - Yalçın Mert Yalçıntaş
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Turkey
| | - Ahmet Alperen Canbolat
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Turkey
| | - Arda Erkan Kalkan
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Turkey
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Turkey
| | - Dunja Šamec
- Department of Food Technology, University North, Trg Dr. Žarka Dolinara 1, 48000 Koprivnica, Croatia
| |
Collapse
|
4
|
Mitchell M, Suh M, Hooda N, Bylsma LC, Cohen SS. The effect of bovine dairy products and their components on the incidence and natural history of infection: a systematic literature review. Nutr J 2024; 23:26. [PMID: 38413931 PMCID: PMC10898086 DOI: 10.1186/s12937-024-00923-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/30/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Dairy products and their components may impact immune function, although the current evidence base has some research gaps. As part of a larger systematic literature review of dairy products/components (including probiotics, dairy proteins, and dairy fats) and immune function, we identified the available epidemiologic research on the impact of dairy products/components on incidence and natural history of infectious diseases. METHODS PubMed and Embase databases were systematically searched through May 2022 to identify eligible studies using pre-defined Population, Intervention, Comparator, Outcomes, and Study design criteria. Herein, we focused on describing the impacts of dairy product/component on infectious disease outcomes, including the effect on leukocyte and cytokine response in humans. Risk of bias assessment was performed using the Academy of Nutrition and Dietetics Quality Criteria Checklist. The Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines were followed. RESULTS Among 9,832 studies identified from the larger literature search, 133 relevant publications from 128 studies reported on dairy product/component and infectious disease outcomes. Few studies are available on the impact of non-fermented milk and traditional yogurt on infectious disease. Evidence was identified to suggest milk and yogurt drinks fermented with Lactobacillus strains reduce the risk and burden of common infectious diseases (CIDs), although the findings are mixed and difficult to reconcile due to heterogenous study populations, bacterial strains, and study methods. Few studies are available on the impact of dairy products/components on the natural history of infection, with the available findings indicating probiotics may both improve gastrointestinal symptoms among HIV-infected persons and help eradicate and alleviate the symptoms of Heliobacter (H.) pylori. The available evidence also suggests lactoferrin may reduce the virological burden of COVID-19 and hepatitis C virus. No consistent changes in leukocytes or cytokine production were observed for any type of dairy product or their components, but probiotics appeared to enhance natural killer cell levels/activity and the phagocytic process. CONCLUSIONS Dairy products, particularly those with added probiotics, may represent an easily accessible nutritional intervention to prevent and improve the course of infectious diseases. This review highlights the need for additional research in this potentially impactful area. PROSPERO REGISTRATION CRD42022333780.
Collapse
Affiliation(s)
- Meghan Mitchell
- EpidStrategies, a division of ToxStrategies, LLC, 23501 Cinco Ranch Blvd, Suite B226, Katy, TX, 77494, USA.
| | - Mina Suh
- EpidStrategies, a division of ToxStrategies, LLC, 23501 Cinco Ranch Blvd, Suite B226, Katy, TX, 77494, USA
| | - Naushin Hooda
- EpidStrategies, a division of ToxStrategies, LLC, 23501 Cinco Ranch Blvd, Suite B226, Katy, TX, 77494, USA
| | - Lauren C Bylsma
- EpidStrategies, a division of ToxStrategies, LLC, 23501 Cinco Ranch Blvd, Suite B226, Katy, TX, 77494, USA
| | - Sarah S Cohen
- Independent contractor to ToxStrategies, LLC, Durham, NC, USA
| |
Collapse
|
5
|
Wilson A, Bogie B, Chaaban H, Burge K. The Nonbacterial Microbiome: Fungal and Viral Contributions to the Preterm Infant Gut in Health and Disease. Microorganisms 2023; 11:909. [PMID: 37110332 PMCID: PMC10144239 DOI: 10.3390/microorganisms11040909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/29/2023] Open
Abstract
The intestinal microbiome is frequently implicated in necrotizing enterocolitis (NEC) pathogenesis. While no particular organism has been associated with NEC development, a general reduction in bacterial diversity and increase in pathobiont abundance has been noted preceding disease onset. However, nearly all evaluations of the preterm infant microbiome focus exclusively on the bacterial constituents, completely ignoring any fungi, protozoa, archaea, and viruses present. The abundance, diversity, and function of these nonbacterial microbes within the preterm intestinal ecosystem are largely unknown. Here, we review findings on the role of fungi and viruses, including bacteriophages, in preterm intestinal development and neonatal intestinal inflammation, with potential roles in NEC pathogenesis yet to be determined. In addition, we highlight the importance of host and environmental influences, interkingdom interactions, and the role of human milk in shaping fungal and viral abundance, diversity, and function within the preterm intestinal ecosystem.
Collapse
Affiliation(s)
| | | | - Hala Chaaban
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Kathryn Burge
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
6
|
Acanthamoeba castellanii Genotype T4: Inhibition of Proteases Activity and Cytopathic Effect by Bovine Apo-Lactoferrin. Microorganisms 2023; 11:microorganisms11030708. [PMID: 36985284 PMCID: PMC10059889 DOI: 10.3390/microorganisms11030708] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Acanthamoeba castellanii genotype T4 is a clinically significant free-living amoeba that causes granulomatous amoebic encephalitis and amoebic keratitis in human beings. During the initial stages of infection, trophozoites interact with various host immune responses, such as lactoferrin (Lf), in the corneal epithelium, nasal mucosa, and blood. Lf plays an important role in the elimination of pathogenic microorganisms, and evasion of the innate immune response is crucial in the colonization process. In this study, we describe the resistance of A. castellanii to the microbicidal effect of bovine apo-lactoferrin (apo-bLf) at different concentrations (25, 50, 100, and 500 µM). Acanthamoeba castellanii trophozoites incubated with apo-bLf at 500 µM for 12 h maintained 98% viability. Interestingly, despite this lack of effect on viability, our results showed that the apo-bLf inhibited the cytopathic effect of A. castellanii in MDCK cells culture, and analysis of amoebic proteases by zymography showed significant inhibition of cysteine and serine proteases by interaction with the apo-bLf. From these results, we conclude that bovine apo-Lf influences the activity of A. castellanii secretion proteases, which in turn decreases amoebic cytopathic activity.
Collapse
|
7
|
Prophylaxis of Invasive Fungal Infection in Neonates: A Narrative Review for Practical Purposes. J Fungi (Basel) 2023; 9:jof9020164. [PMID: 36836279 PMCID: PMC9962596 DOI: 10.3390/jof9020164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/21/2023] [Accepted: 01/22/2023] [Indexed: 01/28/2023] Open
Abstract
Candida albicans is the most frequent cause of invasive fungal disease in preterm and/or low birth weight neonates, followed by Candida parapsilosis, whilst infections from other species are rare. Considering the severity of the disease, associated with poor clinical signs and diagnostic difficulties, primary prophylaxis becomes relevant. This paper summarizes the pathogenesis and clinical presentation of invasive candidiasis in neonates, focusing on prophylaxis. For late onset invasive disease, e.g., those occurring after the 3rd (or 7th according to some definitions) day of life possible approaches are the use of fluconazole, recommended in case of weight <1000 g or <1500 g if the local incidence of invasive candidiasis is higher than 2%, or the use of nystatin (for patients < 1500 g). Micafungin must be used in case of colonization by Candida auris, or in centers with a high prevalence of this pathogen. Concurrently, correct management of the central venous catheter and isolation procedures, with special regard to patients colonized by resistant strains, are fundamental. Other approaches such as reduced use of H2 blockers and broad-spectrum antibiotics (e.g., 3rd generation cephalosporins or carbapenems) and promotion of breast feeding proved useful. Reduction of early-onset infections (those occurring in the first 3 days of life) can also be obtained by treating maternal vulvo-vaginal candidiasis, which can represent a fastidious problem during pregnancy. In this case, topic azoles (the only recommendable treatment) can represent a kind of "prophylaxis" of early neonatal candidiasis. However, it must always be remembered that prophylaxis reduces the risk of invasive candidiasis but can not completely eliminate its occurrence, with the parallel risk of selecting for antifungal-resistant strains. Clinicians must maintain a high level of suspicion to start an appropriate therapy and strict epidemiological surveillance to identify the occurrence of clusters and the appearance of strains resistant to prophylaxis.
Collapse
|
8
|
Li W, Liu B, Lin Y, Xue P, Lu Y, Song S, Li Y, Szeto IMY, Ren F, Guo H. The application of lactoferrin in infant formula: The past, present and future. Crit Rev Food Sci Nutr 2022; 64:5748-5767. [PMID: 36533432 DOI: 10.1080/10408398.2022.2157792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Human milk is universally regarded as the gold standard to fulfill nutrition needs of infants. Lactoferrin (LF) is a major multiple bioactive glycoprotein in human milk but little is presented in infant formula. LF can resist digestion in the infant gastrointestinal tract and is absorbed into the bloodstream in an intact form to perform physiological functions. Evidence suggest that LF prevents pathogen infection, promotes immune system development, intestinal development, brain development and bone health, as well as ameliorates iron deficiency anemia. However, more clinical studies of LF need to be further elucidated to determine an appropriate dosage for application in infant formula. LF is sensitive to denaturation induced by processing of infant formula such as heat treatments and spay drying. Thus, further studies should be focus on maximizing the retention of LF activity in the infant formula process. This review summarizes the structural features of LF. Then the digestion, absorption and metabolism of LF in infants are discussed, followed by the function of LF for infants. Further, we summarize LF in infant formula and effects of processing of infant formula on bioactivities of LF, as well as future perspectives of LF research.
Collapse
Affiliation(s)
- Wusun Li
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
| | - Biao Liu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
- Inner Mongolia Yili Industrial Group Co., Ltd, Hohhot, PR China
| | - Yingying Lin
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
| | - Peng Xue
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
| | - Yao Lu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
| | - Sijia Song
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
| | - Yixuan Li
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
| | - Ignatius Man-Yau Szeto
- Inner Mongolia Yili Industrial Group Co., Ltd, Hohhot, PR China
- National Center of Technology Innovation for Dairy, Hohhot, PR China
| | - Fazheng Ren
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
| | - Huiyuan Guo
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
| |
Collapse
|
9
|
黄 方, 熊 涛, 唐 军. [Recent research on pharmacological prevention strategies for invasive fungal infection in preterm infants]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2022; 24:1171-1177. [PMID: 36305120 PMCID: PMC9627992 DOI: 10.7499/j.issn.1008-8830.2204158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 08/16/2022] [Indexed: 06/16/2023]
Abstract
There is a relatively high incidence rate of invasive fungal infection (IFI) in preterm infants admitted to the neonatal intensive care unit (NICU), and early diagnosis of IFI is difficult in clinical practice. The patients developing IFI tend to have severe conditions, a long course of treatment, high hospital costs, high mortality, and poor prognosis, and therefore, the prevention of IFI is of particular importance. At present, fluconazole is often used as the first-line drug for the prevention of IFI in preterm infants, but no consensus has been reached on the specific dose and course of treatment, and there are still controversies over the targeted population and prophylactic effect. This article reviews the recent research on the pharmacological prevention strategies for IFI in preterm infants in the NICU, so as to provide a reference for clinicians.
Collapse
Affiliation(s)
| | - 涛 熊
- 出生缺陷与相关妇儿疾病教育部重点实验室,四川成都610041
| | - 军 唐
- 出生缺陷与相关妇儿疾病教育部重点实验室,四川成都610041
| |
Collapse
|
10
|
Comparative Proteomic Analysis of Proteins in Breast Milk during Different Lactation Periods. Nutrients 2022; 14:nu14173648. [PMID: 36079904 PMCID: PMC9460426 DOI: 10.3390/nu14173648] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 08/30/2022] [Accepted: 08/31/2022] [Indexed: 11/17/2022] Open
Abstract
Breast milk is an unparalleled food for infants, as it can meet almost all of their nutritional needs. Breast milk in the first month is an important source of acquired immunity. However, breast milk protein may vary with the stage of lactation. Therefore, the aim of this study was to use a data-independent acquisition approach to determine the differences in the proteins of breast milk during different lactation periods. The study samples were colostrum (3-6 days), transitional milk (7-14 days), and mature milk (15-29 days). The results identified a total of 2085 different proteins, and colostrum contained the most characteristic proteins. Protein expression was affected by the lactation stage. The proteins expressed in breast milk changed greatly between day 3 and day 14 and gradually stabilized after 14 days. The expression levels of lactoferrin, immunoglobulin, and clusterin were the highest in colostrum. CTP synthase 1, C-type lectin domain family 19 member A, secretoglobin family 3A member 2, trefoil factor 3 (TFF3), and tenascin were also the highest in colostrum. This study provides further insights into the protein composition of breast milk and the necessary support for the design and production of infant formula.
Collapse
|
11
|
Kowalczyk P, Kaczyńska K, Kleczkowska P, Bukowska-Ośko I, Kramkowski K, Sulejczak D. The Lactoferrin Phenomenon-A Miracle Molecule. Molecules 2022; 27:2941. [PMID: 35566292 PMCID: PMC9104648 DOI: 10.3390/molecules27092941] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 04/29/2022] [Accepted: 05/03/2022] [Indexed: 01/11/2023] Open
Abstract
Numerous harmful factors that affect the human body from birth to old age cause many disturbances, e.g., in the structure of the genome, inducing cell apoptosis and their degeneration, which leads to the development of many diseases, including cancer. Among the factors leading to pathological processes, microbes, viruses, gene dysregulation and immune system disorders have been described. The function of a protective agent may be played by lactoferrin as a "miracle molecule", an endogenous protein with a number of favorable antimicrobial, antiviral, antioxidant, immunostimulatory and binding DNA properties. The purpose of this article is to present the broad spectrum of properties and the role that lactoferrin plays in protecting human cells at all stages of life.
Collapse
Affiliation(s)
- Paweł Kowalczyk
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jabłonna, Poland;
| | - Katarzyna Kaczyńska
- Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 St., 02-106 Warsaw, Poland;
| | - Patrycja Kleczkowska
- Maria Sklodowska-Curie, Medical Academy in Warsaw, Solidarności 12 St., 03-411 Warsaw, Poland
- Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163 Warsaw, Poland
| | - Iwona Bukowska-Ośko
- Department of Immunopathology of Infectious and Parasitic Diseases, Medical University of Warsaw, Pawinskiego 3c St., 02-106 Warsaw, Poland;
| | - Karol Kramkowski
- Department of Physical Chemistry, Medical University of Bialystok, Kilińskiego 1 St., 15-089 Bialystok, Poland;
| | - Dorota Sulejczak
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 St., 02-106 Warsaw, Poland
| |
Collapse
|
12
|
Malcangi G, Inchingolo AD, Inchingolo AM, Piras F, Settanni V, Garofoli G, Palmieri G, Ceci S, Patano A, Mancini A, Vimercati L, Nemore D, Scardapane A, Rapone B, Semjonova A, D’Oria MT, Macchia L, Bordea IR, Migliore G, Scarano A, Lorusso F, Tartaglia GM, Giovanniello D, Nucci L, Maggialetti N, Parisi A, Domenico MD, Brienza N, Tafuri S, Stefanizzi P, Curatoli L, Corriero A, Contaldo M, Inchingolo F, Dipalma G. COVID-19 Infection in Children and Infants: Current Status on Therapies and Vaccines. CHILDREN (BASEL, SWITZERLAND) 2022; 9:249. [PMID: 35204969 PMCID: PMC8870718 DOI: 10.3390/children9020249] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/21/2022] [Accepted: 02/10/2022] [Indexed: 02/06/2023]
Abstract
Since the beginning in December 2019, the SARS-CoV-2 outbreak appeared to affect mostly the adult population, sparing the vast majority of children who only showed mild symptoms. The purpose of this investigation is to assess the status on the mechanisms that give children and infants this variation in epidemiology compared to the adult population and its impact on therapies and vaccines that are aimed towards them. A literature review, including in vitro studies, reviews, published guidelines and clinical trials was performed. Clinical trials concerned topics that allowed a descriptive synthesis to be produced. Four underlying mechanisms were found that may play a key role in providing COVID-19 protection in babies. No guidelines are available yet for therapy due to insufficient data; support therapy remains the most used. Only two vaccines are approved by the World Health Organization to be used in children from 12 years of age, and there are currently no efficacy or safety data for children below the age of 12 years. The COVID-19 clinical frame infection is milder in children and adolescents. This section of the population can act as vectors and reservoirs and play a key role in the transmission of the infection; therefore, vaccines are paramount. More evidence is required to guide safely the vaccination campaign.
Collapse
Affiliation(s)
- Giuseppina Malcangi
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (G.M.); (A.D.I.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (A.M.); (L.V.); (D.N.); (A.S.); (B.R.); (A.S.); (M.T.D.); (G.D.)
| | - Alessio Danilo Inchingolo
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (G.M.); (A.D.I.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (A.M.); (L.V.); (D.N.); (A.S.); (B.R.); (A.S.); (M.T.D.); (G.D.)
| | - Angelo Michele Inchingolo
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (G.M.); (A.D.I.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (A.M.); (L.V.); (D.N.); (A.S.); (B.R.); (A.S.); (M.T.D.); (G.D.)
| | - Fabio Piras
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (G.M.); (A.D.I.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (A.M.); (L.V.); (D.N.); (A.S.); (B.R.); (A.S.); (M.T.D.); (G.D.)
| | - Vito Settanni
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (G.M.); (A.D.I.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (A.M.); (L.V.); (D.N.); (A.S.); (B.R.); (A.S.); (M.T.D.); (G.D.)
| | - Grazia Garofoli
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (G.M.); (A.D.I.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (A.M.); (L.V.); (D.N.); (A.S.); (B.R.); (A.S.); (M.T.D.); (G.D.)
| | - Giulia Palmieri
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (G.M.); (A.D.I.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (A.M.); (L.V.); (D.N.); (A.S.); (B.R.); (A.S.); (M.T.D.); (G.D.)
| | - Sabino Ceci
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (G.M.); (A.D.I.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (A.M.); (L.V.); (D.N.); (A.S.); (B.R.); (A.S.); (M.T.D.); (G.D.)
| | - Assunta Patano
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (G.M.); (A.D.I.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (A.M.); (L.V.); (D.N.); (A.S.); (B.R.); (A.S.); (M.T.D.); (G.D.)
| | - Antonio Mancini
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (G.M.); (A.D.I.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (A.M.); (L.V.); (D.N.); (A.S.); (B.R.); (A.S.); (M.T.D.); (G.D.)
| | - Luigi Vimercati
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (G.M.); (A.D.I.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (A.M.); (L.V.); (D.N.); (A.S.); (B.R.); (A.S.); (M.T.D.); (G.D.)
| | - Damiano Nemore
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (G.M.); (A.D.I.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (A.M.); (L.V.); (D.N.); (A.S.); (B.R.); (A.S.); (M.T.D.); (G.D.)
| | - Arnaldo Scardapane
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (G.M.); (A.D.I.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (A.M.); (L.V.); (D.N.); (A.S.); (B.R.); (A.S.); (M.T.D.); (G.D.)
| | - Biagio Rapone
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (G.M.); (A.D.I.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (A.M.); (L.V.); (D.N.); (A.S.); (B.R.); (A.S.); (M.T.D.); (G.D.)
| | - Alexandra Semjonova
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (G.M.); (A.D.I.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (A.M.); (L.V.); (D.N.); (A.S.); (B.R.); (A.S.); (M.T.D.); (G.D.)
| | - Maria Teresa D’Oria
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (G.M.); (A.D.I.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (A.M.); (L.V.); (D.N.); (A.S.); (B.R.); (A.S.); (M.T.D.); (G.D.)
- Department of Medical and Biological Sciences, University of Udine, Via delle Scienze, 206, 33100 Udine, Italy
| | - Luigi Macchia
- Department of Emergency and Organ Transplantation (D.E.T.O.), School and Chair of Allergology and Clinical Immunology, University of Bari Aldo Moro, 70121 Bari, Italy;
| | - Ioana Roxana Bordea
- Department of Oral Rehabilitation, Faculty of Dentistry, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | | | - Antonio Scarano
- Department of Innovative Technologies in Medicine and Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Felice Lorusso
- Department of Innovative Technologies in Medicine and Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Gianluca Martino Tartaglia
- UOC Maxillo-Facial Surgery and Dentistry, Department of Biomedical, Surgical and Dental Sciences, School of Dentistry, Fondazione IRCCS Ca Granda, Ospedale Maggiore Policlinico, University of Milan, 20100 Milan, Italy;
| | - Delia Giovanniello
- Department of Toracic Surgery, Hospital “San Camillo Forlanini”, 00152 Rome, Italy;
| | - Ludovica Nucci
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania Luigi Vanvitelli, Via Luigi de Crecchio, 6, 80138 Naples, Italy; (L.N.); (M.C.)
| | - Nicola Maggialetti
- Department of Medical Science, Neuroscience and Sensory Organs, University of Bari Aldo Moro, 70124 Bari, Italy;
| | - Antonio Parisi
- Istituto Zooprofilattico Sperimentale della Puglia e della Basilicata, 71121 Foggia, Italy;
| | - Marina Di Domenico
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy;
| | - Nicola Brienza
- Unit of Anesthesia and Resuscitation, Department of Emergencies and Organ Transplantations, Aldo Moro University, 70124 Bari, Italy; (N.B.); (A.C.)
| | - Silvio Tafuri
- Department of Biomedical Science and Human Oncology, University of Bari, 70124 Bari, Italy; (S.T.); (P.S.)
| | - Pasquale Stefanizzi
- Department of Biomedical Science and Human Oncology, University of Bari, 70124 Bari, Italy; (S.T.); (P.S.)
| | - Luigi Curatoli
- Department Neurosciences & Sensory Organs & Musculoskeletal System, University of Bari Aldo Moro, 70124 Bari, Italy;
| | - Alberto Corriero
- Unit of Anesthesia and Resuscitation, Department of Emergencies and Organ Transplantations, Aldo Moro University, 70124 Bari, Italy; (N.B.); (A.C.)
| | - Maria Contaldo
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania Luigi Vanvitelli, Via Luigi de Crecchio, 6, 80138 Naples, Italy; (L.N.); (M.C.)
| | - Francesco Inchingolo
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (G.M.); (A.D.I.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (A.M.); (L.V.); (D.N.); (A.S.); (B.R.); (A.S.); (M.T.D.); (G.D.)
| | - Gianna Dipalma
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (G.M.); (A.D.I.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (A.M.); (L.V.); (D.N.); (A.S.); (B.R.); (A.S.); (M.T.D.); (G.D.)
| |
Collapse
|
13
|
Zeinali LI, Giuliano S, Lakshminrusimha S, Underwood MA. Intestinal Dysbiosis in the Infant and the Future of Lacto-Engineering to Shape the Developing Intestinal Microbiome. Clin Ther 2021; 44:193-214.e1. [PMID: 34922744 DOI: 10.1016/j.clinthera.2021.11.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 11/06/2021] [Accepted: 11/12/2021] [Indexed: 12/16/2022]
Abstract
PURPOSE The goal of this study was to review the role of human milk in shaping the infant intestinal microbiota and the potential of human milk bioactive molecules to reverse trends of increasing intestinal dysbiosis and dysbiosis-associated diseases. METHODS This narrative review was based on recent and historic literature. FINDINGS Human milk immunoglobulins, oligosaccharides, lactoferrin, lysozyme, milk fat globule membranes, and bile salt-stimulating lipase are complex multifunctional bioactive molecules that, among other important functions, shape the composition of the infant intestinal microbiota. IMPLICATIONS The co-evolution of human milk components and human milk-consuming commensal anaerobes many thousands of years ago resulted in a stable low-diversity infant microbiota. Over the past century, the introduction of antibiotics and modern hygiene practices plus changes in the care of newborns have led to significant alterations in the intestinal microbiota, with associated increases in risk of dysbiosis-associated disease. A better understanding of mechanisms by which human milk shapes the intestinal microbiota of the infant during a vulnerable period of development of the immune system is needed to alter the current trajectory and decrease intestinal dysbiosis and associated diseases.
Collapse
Affiliation(s)
- Lida I Zeinali
- Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA, USA
| | | | | | - Mark A Underwood
- Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA, USA.
| |
Collapse
|
14
|
Kilpatrick R, Scarrow E, Hornik C, Greenberg RG. Neonatal invasive candidiasis: updates on clinical management and prevention. THE LANCET CHILD & ADOLESCENT HEALTH 2021; 6:60-70. [PMID: 34672994 DOI: 10.1016/s2352-4642(21)00272-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/30/2021] [Accepted: 08/13/2021] [Indexed: 01/15/2023]
Abstract
Neonatal invasive candidiasis is an important cause of morbidity and mortality in preterm infants. The incidence of invasive candidiasis in this population has been declining in high-income settings, largely due to preventive measures, although there are still considerable variations in incidence between health-care centres. Surveillance data and large, multicentre studies in lower-income settings are not available, although preventive measures in these settings have been shown to decrease the incidence of neonatal invasive candidiasis. Understanding risk factors and pathogenesis are key to the prevention of invasive candidiasis. The difficulty of a definitive diagnosis of invasive candidiasis and the high risk for death or substantial neurodevelopmental impairment, even with appropriate treatment, further increase the need for effective preventive measures. In this Review, we examine the pathogenesis, clinical presentation, and diagnosis of invasive candidiasis. We highlight commonly used and emerging preventive and prophylactic measures, including standardised central line care, antibiotic stewardship, antifungal prophylaxis, and probiotics. Finally, we provide updates on empirical treatment, clinical management in confirmed cases of invasive candidiasis, and antifungal pharmacotherapy.
Collapse
Affiliation(s)
- Ryan Kilpatrick
- Department of Pediatrics, Duke Clinical Research Institute, Durham, NC, USA
| | - Evelyn Scarrow
- Department of Pediatrics, Duke Clinical Research Institute, Durham, NC, USA
| | - Chi Hornik
- Department of Pediatrics, Duke Clinical Research Institute, Durham, NC, USA
| | - Rachel G Greenberg
- Department of Pediatrics, Duke Clinical Research Institute, Durham, NC, USA.
| |
Collapse
|
15
|
Malcangi G, Inchingolo AD, Inchingolo AM, Santacroce L, Marinelli G, Mancini A, Vimercati L, Maggiore ME, D’Oria MT, Hazballa D, Bordea IR, Xhajanka E, Scarano A, Farronato M, Tartaglia GM, Giovanniello D, Nucci L, Serpico R, Sammartino G, Capozzi L, Parisi A, Di Domenico M, Lorusso F, Contaldo M, Inchingolo F, Dipalma G. COVID-19 Infection in Children, Infants and Pregnant Subjects: An Overview of Recent Insights and Therapies. Microorganisms 2021; 9:1964. [PMID: 34576859 PMCID: PMC8469368 DOI: 10.3390/microorganisms9091964] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/07/2021] [Accepted: 09/09/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The SARS-CoV-2 pandemic has involved a severe increase of cases worldwide in a wide range of populations. The aim of the present investigation was to evaluate recent insights about COVID-19 infection in children, infants and pregnant subjects. METHODS a literature overview was performed including clinical trials, in vitro studies, reviews and published guidelines regarding the present paper topic. A descriptive synthesis was performed to evaluate recent insights and the effectiveness of therapies for SARS-CoV-2 infection in children, infants and pregnant subjects. RESULTS Insufficient data are available regarding the relationship between COVID-19 and the clinical risk of spontaneous abortion and premature foetus death. A decrease in the incidence of COVID-19 could be correlated to a minor expression of ACE2 in childrens' lungs. At present, a modulation of the dose-effect posology for children and infants is necessary. CONCLUSIONS Pregnant vertical transmission has been hypothesised for SARS-CoV-2 infection. Vaccines are necessary to achieve mass immunity for children and also pregnant subjects.
Collapse
Affiliation(s)
- Giuseppina Malcangi
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (L.S.); (G.M.); (A.M.); (L.V.); (M.E.M.); (M.T.D.); (D.H.); (F.I.); (G.D.)
| | - Alessio Danilo Inchingolo
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (L.S.); (G.M.); (A.M.); (L.V.); (M.E.M.); (M.T.D.); (D.H.); (F.I.); (G.D.)
| | - Angelo Michele Inchingolo
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (L.S.); (G.M.); (A.M.); (L.V.); (M.E.M.); (M.T.D.); (D.H.); (F.I.); (G.D.)
| | - Luigi Santacroce
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (L.S.); (G.M.); (A.M.); (L.V.); (M.E.M.); (M.T.D.); (D.H.); (F.I.); (G.D.)
| | - Grazia Marinelli
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (L.S.); (G.M.); (A.M.); (L.V.); (M.E.M.); (M.T.D.); (D.H.); (F.I.); (G.D.)
| | - Antonio Mancini
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (L.S.); (G.M.); (A.M.); (L.V.); (M.E.M.); (M.T.D.); (D.H.); (F.I.); (G.D.)
| | - Luigi Vimercati
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (L.S.); (G.M.); (A.M.); (L.V.); (M.E.M.); (M.T.D.); (D.H.); (F.I.); (G.D.)
| | - Maria Elena Maggiore
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (L.S.); (G.M.); (A.M.); (L.V.); (M.E.M.); (M.T.D.); (D.H.); (F.I.); (G.D.)
| | - Maria Teresa D’Oria
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (L.S.); (G.M.); (A.M.); (L.V.); (M.E.M.); (M.T.D.); (D.H.); (F.I.); (G.D.)
- Department of Medical and Biological Sciences, University of Udine, Via delle Scienze, 206, 33100 Udine, Italy
| | - Denisa Hazballa
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (L.S.); (G.M.); (A.M.); (L.V.); (M.E.M.); (M.T.D.); (D.H.); (F.I.); (G.D.)
- Kongresi Elbasanit, Rruga: Aqif Pasha, 3001 Elbasan, Albania
| | - Ioana Roxana Bordea
- Department of Oral Rehabilitation, Faculty of Dentistry, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Edit Xhajanka
- Department of Dental Prosthesis, Medical University of Tirana, Rruga e Dibrës, U.M.T., 1001 Tirana, Albania;
| | - Antonio Scarano
- Department of Innovative Technologies in Medicine and Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Marco Farronato
- UOC Maxillo-Facial Surgery and Dentistry, Department of Biomedical, Surgical and Dental Sciences, School of Dentistry, Fondazione IRCCS Ca Granda, Ospedale Maggiore Policlinico, University of Milan, 20100 Milan, Italy; (M.F.); (G.M.T.)
| | - Gianluca Martino Tartaglia
- UOC Maxillo-Facial Surgery and Dentistry, Department of Biomedical, Surgical and Dental Sciences, School of Dentistry, Fondazione IRCCS Ca Granda, Ospedale Maggiore Policlinico, University of Milan, 20100 Milan, Italy; (M.F.); (G.M.T.)
| | | | - Ludovica Nucci
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania Luigi Vanvitelli, Via Luigi de Crecchio, 6, 80138 Naples, Italy; (L.N.); (R.S.); (M.C.)
| | - Rosario Serpico
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania Luigi Vanvitelli, Via Luigi de Crecchio, 6, 80138 Naples, Italy; (L.N.); (R.S.); (M.C.)
| | - Gilberto Sammartino
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, 80131 Naples, Italy;
| | - Loredana Capozzi
- Istituto Zooprofilattico Sperimentale Della Puglia e Della Basilicata, 71121 Foggia, Italy; (L.C.); (A.P.)
| | - Antonio Parisi
- Istituto Zooprofilattico Sperimentale Della Puglia e Della Basilicata, 71121 Foggia, Italy; (L.C.); (A.P.)
| | - Marina Di Domenico
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy;
| | - Felice Lorusso
- Department of Innovative Technologies in Medicine and Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Maria Contaldo
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania Luigi Vanvitelli, Via Luigi de Crecchio, 6, 80138 Naples, Italy; (L.N.); (R.S.); (M.C.)
| | - Francesco Inchingolo
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (L.S.); (G.M.); (A.M.); (L.V.); (M.E.M.); (M.T.D.); (D.H.); (F.I.); (G.D.)
| | - Gianna Dipalma
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (L.S.); (G.M.); (A.M.); (L.V.); (M.E.M.); (M.T.D.); (D.H.); (F.I.); (G.D.)
| |
Collapse
|
16
|
Abstract
Necrotizing enterocolitis (NEC) is an inflammatory disease affecting premature infants. Intestinal microbial composition may play a key role in determining which infants are predisposed to NEC and when infants are at highest risk of developing NEC. It is unclear how to optimize antibiotic therapy in preterm infants to prevent NEC and how to optimize antibiotic regimens to treat neonates with NEC. This article discusses risk factors for NEC, how dysbiosis in preterm infants plays a role in the pathogenesis of NEC, and how probiotic and antibiotic therapy may be used to prevent and/or treat NEC and its sequelae.
Collapse
Affiliation(s)
- Jennifer Duchon
- Division of Newborn Medicine, Jack and Lucy Department of Pediatrics, Icahn School of Medicine at Mount Sinai, 1000 10th Avenue, New York, NY 10019, USA
| | - Maria E Barbian
- Division of Neonatal-Perinatal Medicine, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive Northeast, 3rd Floor, Atlanta, GA 30322, USA
| | - Patricia W Denning
- Division of Neonatal-Perinatal Medicine, Emory University School of Medicine, Children's Healthcare of Atlanta, Emory University Hospital Midtown, 550 Peachtree Street, 3rd Floor MOT, Atlanta, GA 30308, USA.
| |
Collapse
|
17
|
Morrin ST, Buck RH, Farrow M, Hickey RM. Milk-derived anti-infectives and their potential to combat bacterial and viral infection. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104442] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
18
|
Inchingolo AD, Dipalma G, Inchingolo AM, Malcangi G, Santacroce L, D’Oria MT, Isacco CG, Bordea IR, Candrea S, Scarano A, Morandi B, Del Fabbro M, Farronato M, Tartaglia GM, Balzanelli MG, Ballini A, Nucci L, Lorusso F, Taschieri S, Inchingolo F. The 15-Months Clinical Experience of SARS-CoV-2: A Literature Review of Therapies and Adjuvants. Antioxidants (Basel) 2021; 10:881. [PMID: 34072708 PMCID: PMC8226610 DOI: 10.3390/antiox10060881] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/23/2021] [Accepted: 05/26/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the virus responsible for the coronavirus disease of 2019 (COVID-19) that emerged in December 2019 in Wuhan, China, and rapidly spread worldwide, with a daily increase in confirmed cases and infection-related deaths. The World Health Organization declared a pandemic on the 11th of March 2020. COVID-19 presents flu-like symptoms that become severe in high-risk medically compromised subjects. The aim of this study was to perform an updated overview of the treatments and adjuvant protocols for COVID-19. METHODS A systematic literature search of databases was performed (MEDLINE PubMed, Google Scholar, UpToDate, Embase, and Web of Science) using the keywords: "COVID-19", "2019-nCoV", "coronavirus" and "SARS-CoV-2" (date range: 1 January 2019 to 31st October 2020), focused on clinical features and treatments. RESULTS The main treatments retrieved were antivirals, antimalarials, convalescent plasma, immunomodulators, corticosteroids, anticoagulants, and mesenchymal stem cells. Most of the described treatments may provide benefits to COVID-19 subjects, but no one protocol has definitively proven its efficacy. CONCLUSIONS While many efforts are being spent worldwide in research aimed at identifying early diagnostic methods and evidence-based effective treatments, mass vaccination is thought to be the best option against this disease in the near future.
Collapse
Affiliation(s)
- Alessio Danilo Inchingolo
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (G.D.); (A.M.I.); (L.S.); (M.T.D.); (C.G.I.); (F.I.)
| | - Gianna Dipalma
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (G.D.); (A.M.I.); (L.S.); (M.T.D.); (C.G.I.); (F.I.)
| | - Angelo Michele Inchingolo
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (G.D.); (A.M.I.); (L.S.); (M.T.D.); (C.G.I.); (F.I.)
| | - Giuseppina Malcangi
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (G.D.); (A.M.I.); (L.S.); (M.T.D.); (C.G.I.); (F.I.)
| | - Luigi Santacroce
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (G.D.); (A.M.I.); (L.S.); (M.T.D.); (C.G.I.); (F.I.)
| | - Maria Teresa D’Oria
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (G.D.); (A.M.I.); (L.S.); (M.T.D.); (C.G.I.); (F.I.)
- Department of Medical and Biological Sciences, University of Udine, Via delle Scienze, 206, 33100 Udine, Italy
| | - Ciro Gargiulo Isacco
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (G.D.); (A.M.I.); (L.S.); (M.T.D.); (C.G.I.); (F.I.)
- Research at Human Stem Cells Research Center HSC, Ho Chi Minh 70000, Vietnam
- Embryology and Regenerative Medicine and Immunology, Pham Chau Trinh University of Medicine Hoi An, Hoi An 70000, Vietnam
| | - Ioana Roxana Bordea
- Department of Oral Rehabilitation, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400012 Cluj-Napoca, Romania;
| | - Sebastian Candrea
- Department of Oral Rehabilitation, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400012 Cluj-Napoca, Romania;
- Department of Pedodontics, County Hospital Cluj-Napoca, 400000 Cluj-Napoca, Romania
| | - Antonio Scarano
- Department of Innovative Technologies in Medicine and Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Benedetta Morandi
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, 20122 Milan, Italy; (B.M.); (M.D.F.); (M.F.); (G.M.T.); (S.T.)
- Dental Clinic, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy
| | - Massimo Del Fabbro
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, 20122 Milan, Italy; (B.M.); (M.D.F.); (M.F.); (G.M.T.); (S.T.)
- Dental Clinic, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy
| | - Marco Farronato
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, 20122 Milan, Italy; (B.M.); (M.D.F.); (M.F.); (G.M.T.); (S.T.)
- UOC Maxillo-Facial Surgery and Dentistry, Fondazione IRCCS Ca Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Gianluca Martino Tartaglia
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, 20122 Milan, Italy; (B.M.); (M.D.F.); (M.F.); (G.M.T.); (S.T.)
- UOC Maxillo-Facial Surgery and Dentistry, Fondazione IRCCS Ca Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Mario Giosuè Balzanelli
- SET-118, Department of Pre-Hospital and Emergency-San Giuseppe Moscati Hospital, 74100 Taranto, Italy;
| | - Andrea Ballini
- Department of Biosciences, Biotechnologies and Biopharmaceutics, Campus Universitario, University of Bari, 70125 Bari, Italy;
- Department of Precision Medicine, University of Campania, 80138 Naples, Italy
| | - Ludovica Nucci
- Department of Medical-Surgical and Dental Specialties, University of Campania Luigi Vanvitelli, 80100 Naples, Italy;
| | - Felice Lorusso
- Department of Innovative Technologies in Medicine and Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Silvio Taschieri
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, 20122 Milan, Italy; (B.M.); (M.D.F.); (M.F.); (G.M.T.); (S.T.)
- Dental Clinic, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy
- Department of Oral Surgery, Institute of Dentistry, I. M. Sechenov First Moscow State Medical University, 119146 Moscow, Russia
| | - Francesco Inchingolo
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (G.D.); (A.M.I.); (L.S.); (M.T.D.); (C.G.I.); (F.I.)
| |
Collapse
|
19
|
Nayak A, Tiozzo C, Lin X, Mejia C, Gurzenda E, Kim M, Hanna N. Is Lactoferrin Supplementation Beneficial for All Preterm Infants? Am J Perinatol 2021; 40:680-687. [PMID: 34058763 DOI: 10.1055/s-0041-1730433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Human milk (HM) has antibacterial properties due to the presence of immune-modulators, including lactoferrin (LF). This study will determine effect(s) of HM maturation, fortification, and storage conditions on LF levels and its antibacterial properties. STUDY DESIGN HM samples (n = 30) were obtained from preterm and term mothers. The LF levels were analyzed by ELISA, and the antibacterial activity was measured after inoculation with Escherichia coli. RESULTS The highest level of LF in preterm HM was observed in the first week of lactation. However, storage of preterm HM at 4°C decreased LF levels significantly. Both LF levels and antibacterial activity in preterm HM was lower compared with term HM, but significantly higher than donor HM even after HM-based fortification. LF supplementation of donor HM improved its antibacterial activity. CONCLUSION Preterm infants fed donor HM, formula, or stored HM at 4°C may benefits from LF supplementation to improve HM antibacterial properties. KEY POINTS · Milk LF levels vary with storage and maturity.. · Donor milk is deficient in LF even after adding HM-based fortification.. · Donor HM and formula fed infants may benefit from LF..
Collapse
Affiliation(s)
- Amrita Nayak
- Department of Pediatrics, NYU Langone Hospital-Long Island, Mineola, New York
| | - Caterina Tiozzo
- Department of Pediatrics, NYU Langone Hospital-Long Island, Mineola, New York.,Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| | - Xinhua Lin
- Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| | - Claudia Mejia
- Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| | - Ellen Gurzenda
- Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| | - Maureen Kim
- Department of Pediatrics, NYU Langone Hospital-Long Island, Mineola, New York
| | - Nazeeh Hanna
- Department of Pediatrics, NYU Langone Hospital-Long Island, Mineola, New York.,Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| |
Collapse
|
20
|
Sánchez C, Franco L, Regal P, Lamas A, Cepeda A, Fente C. Breast Milk: A Source of Functional Compounds with Potential Application in Nutrition and Therapy. Nutrients 2021; 13:1026. [PMID: 33810073 PMCID: PMC8005182 DOI: 10.3390/nu13031026] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/09/2021] [Accepted: 03/18/2021] [Indexed: 12/12/2022] Open
Abstract
Breast milk is an unbeatable food that covers all the nutritional requirements of an infant in its different stages of growth up to six months after birth. In addition, breastfeeding benefits both maternal and child health. Increasing knowledge has been acquired regarding the composition of breast milk. Epidemiological studies and epigenetics allow us to understand the possible lifelong effects of breastfeeding. In this review we have compiled some of the components with clear functional activity that are present in human milk and the processes through which they promote infant development and maturation as well as modulate immunity. Milk fat globule membrane, proteins, oligosaccharides, growth factors, milk exosomes, or microorganisms are functional components to use in infant formulas, any other food products, nutritional supplements, nutraceuticals, or even for the development of new clinical therapies. The clinical evaluation of these compounds and their commercial exploitation are limited by the difficulty of isolating and producing them on an adequate scale. In this work we focus on the compounds produced using milk components from other species such as bovine, transgenic cattle capable of expressing components of human breast milk or microbial culture engineering.
Collapse
Affiliation(s)
- Cristina Sánchez
- Pharmacy Faculty, San Pablo-CEU University, 28003 Madrid, Spain;
| | - Luis Franco
- Medicine Faculty, Santiago de Compostela University, 15782 Santiago de Compostela, Spain;
| | - Patricia Regal
- Department of Analytical Chemistry, Nutrition and Bromatology, Santiago de Compostela University, 27002 Lugo, Spain; (P.R.); (A.L.); (A.C.)
| | - Alexandre Lamas
- Department of Analytical Chemistry, Nutrition and Bromatology, Santiago de Compostela University, 27002 Lugo, Spain; (P.R.); (A.L.); (A.C.)
| | - Alberto Cepeda
- Department of Analytical Chemistry, Nutrition and Bromatology, Santiago de Compostela University, 27002 Lugo, Spain; (P.R.); (A.L.); (A.C.)
| | - Cristina Fente
- Department of Analytical Chemistry, Nutrition and Bromatology, Santiago de Compostela University, 27002 Lugo, Spain; (P.R.); (A.L.); (A.C.)
| |
Collapse
|
21
|
Chen Y, Wang B, Yang C, Shi Y, Dong Z, Troy FA. Functional Correlates and Impact of Dietary Lactoferrin Intervention and its Concentration-dependence on Neurodevelopment and Cognition in Neonatal Piglets. Mol Nutr Food Res 2021; 65:e2001099. [PMID: 33641262 DOI: 10.1002/mnfr.202001099] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/16/2021] [Indexed: 01/26/2023]
Abstract
SCOPE Lactoferrin (Lf), a sialylated milk glycoprotein, promotes early neurodevelopment and cognition. Functional concentrations of Lf, however, remain unknown. Our objective is to determine the concentration-dependency of Lf on genes associated with neurodevelopment and cognition in neonatal piglets. METHODS AND RESULTS Piglets are given milk replacer with Lf at concentrations of 155 (low) or 285 mg kg-1 day-1 (high) from postnatal days 3 to 38. Gene expression associated with neurodevelopment, cognition, and cognate proteins were quantitated. This study found 1) The rate of learning and long-term memory was higher with 155 mg kg-1 day-1 assessed in an eight-arm radial maze; 2) Global gene transcription profiling showed this lower concentration upregulated genes and functions correlated with neurodevelopment and cognition, while the higher concentration regulated cellular processes for neuroprotection; 3) Expression of BDNF genes and proteins were higher with both concentrations, while genes regulating BDNF signaling, including SLC6A3, IGF-1 responded more to the lower concentration; 4) The lower concentration modulated genes in the five highest networks associated with cellularity and neurocognition, while the prevention of neurodevelopmental and neurological pathologies was associated with the higher concentration. CONCLUSION The lower concentrations of Lf enhanced neurodevelopment and cognition, while higher concentrations are greater neuroprotective, findings of potential novel clinical relevance.
Collapse
Affiliation(s)
- Yue Chen
- School of Medicine, Xiamen University, Xiamen, 361005, China
| | - Bing Wang
- School of Medicine, Xiamen University, Xiamen, 361005, China.,Graham Centre for Agricultural Innovation, Charles Sturt University, NSW 2678, Australia
| | - Changwei Yang
- School of Medicine, Xiamen University, Xiamen, 361005, China
| | - Yujie Shi
- Nestle Research Centre, Beijing, 100095, China
| | - Zhizhong Dong
- Nutrition & Health Research Institute, COFCO Corporation, Beijing, 102209, China
| | - Frederic A Troy
- School of Medicine, Xiamen University, Xiamen, 361005, China.,Department of Biochemistry and Molecular Medicine, University of California School of Medicine, Davis, CA, 95616, USA
| |
Collapse
|
22
|
Razak A, Hussain A. Lactoferrin Supplementation to Prevent Late-Onset Sepsis in Preterm Infants: A Meta-Analysis. Am J Perinatol 2021; 38:283-290. [PMID: 31529448 DOI: 10.1055/s-0039-1696676] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVE This study aimed to systematically review and meta-analyze the role of lactoferrin supplementation to prevent late-onset sepsis (LOS) in preterm infants. STUDY DESIGN Database search include PubMed, Web of Science, and Cochrane central for randomized clinical trial (RCTs). The Cochrane Grading of Recommendations Assessment, Development, and Evaluation methodology was used for summarizing the results. RESULTS Ten RCTs involving 3,679 infants were included. Lactoferrin supplementation with or without probiotics decreased all LOS (relative risk [RR]: 0.56; 95% confidence interval [CI]: 0.36-0.86; I 2 = 58%; 10 studies; 3,470 subjects; level of evidence [LOE]: low) significantly. Similarly, lactoferrin supplementation without probiotics decreased all LOS (RR: 0.43; 95% CI: 0.29-0.62; I 2 = 0%; 8 studies; 1,209 subjects; LOE: moderate) significantly. Lactoferrin supplementation did not significantly reduce necrotizing enterocolitis (RR: 0.62; 95% CI: 0.29-1.33; I 2 = 43%; 6 studies; 3,079 subjects; LOE: low), all-cause mortality (RR: 0.74; 95% CI: 0.36-1.53; I 2 = 53%; 8 studies; 3,395 subjects; LOE: very low), bronchopulmonary dysplasia (RR: 1; 95% CI: 0.90-1.13; I 2 = 0%; 4 studies; 2,570 subjects; LOE: moderate), and threshold retinopathy of prematurity eligible for surgical treatment (RR: 0.61; 95% CI: 0.25-1.51; I 2 = 74%; 2 studies; 2,481 subjects; LOE: very low). CONCLUSION Low to moderate quality evidence suggests that lactoferrin supplementation reduces LOS in preterm infants. Further research is needed to improve the certainty in the evidence.
Collapse
Affiliation(s)
- Abdul Razak
- Division of Neonatology, Department of Pediatrics, King Abdullah bin Abdulaziz University Hospital, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia.,Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Asif Hussain
- Department of Orthopedics, Bhimrao Ambedkar University, Agra, India
| |
Collapse
|
23
|
Wronowski MF, Kotowska M, Banasiuk M, Kotowski A, Kuzmicka W, Albrecht P. Bovine Lactoferrin in the Prevention of Antibiotic-Associated Diarrhea in Children: A Randomized Clinical Trial. Front Pediatr 2021; 9:675606. [PMID: 34164360 PMCID: PMC8215102 DOI: 10.3389/fped.2021.675606] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/27/2021] [Indexed: 11/30/2022] Open
Abstract
Introduction: Antibiotic-associated diarrhea (AAD) is a common adverse reaction to antibiotic treatment affecting up to 21% of children. The aim of the study is to evaluate whether bovine lactoferrin (bLf) might be used for AAD prevention. Materials and Methods: In this prospective, randomized, double-blind, placebo-controlled, single-center study, we enrolled 156 children aged between 1 and 18 years, treated with antibiotic due to acute respiratory or urinary tract infection. We randomly allocated children 1:1 to receive 100 mg of bLf or a placebo twice a day orally for the whole period of antibiotic therapy. The primary outcome was the occurrence of antibiotic-associated diarrhea during and up to 2 weeks after antibiotic therapy. The secondary endpoint was intravenous rehydration or antibiotic withdrawal due to diarrhea. We performed intention-to-treat analysis. Results: We included 150 patients in intention-to-treat analysis. AAD occurred in 16 of 75 (21.3%) patients in bLf group and in 7 of 75 (9.3%) individuals in placebo group [OR = 2.6, (95% CI: 1.01-6.84), p = 0.04]. Relative risk was 2.29 (95% CI: 0.89-5.88). The need for intravenous rehydration occurred in one patient in the placebo group (p = 0.3). We observed no adverse effects in neither of the groups. Discussion: The trial indicated that bLf is not effective in AAD prevention. The risk for AAD was higher in bovine lactoferrin group as compared with placebo. We registered the study protocol on ClinicalTrials.gov (NCT02626104).
Collapse
Affiliation(s)
- Michal F Wronowski
- Department of Pediatric Gastroenterology and Nutrition, Medical University of Warsaw, Warsaw, Poland.,Department of Pediatrics With Clinical Assessment Unit, Medical University of Warsaw, Warsaw, Poland
| | - Maria Kotowska
- Department of Pediatric Gastroenterology and Nutrition, Medical University of Warsaw, Warsaw, Poland
| | - Marcin Banasiuk
- Department of Pediatric Gastroenterology and Nutrition, Medical University of Warsaw, Warsaw, Poland
| | - Artur Kotowski
- Polish Association for Good Clinical Practice, Warsaw, Poland
| | - Weronika Kuzmicka
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Warsaw, Poland.,Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Piotr Albrecht
- Department of Pediatric Gastroenterology and Nutrition, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
24
|
van Esch BCAM, Porbahaie M, Abbring S, Garssen J, Potaczek DP, Savelkoul HFJ, van Neerven RJJ. The Impact of Milk and Its Components on Epigenetic Programming of Immune Function in Early Life and Beyond: Implications for Allergy and Asthma. Front Immunol 2020; 11:2141. [PMID: 33193294 PMCID: PMC7641638 DOI: 10.3389/fimmu.2020.02141] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/06/2020] [Indexed: 12/12/2022] Open
Abstract
Specific and adequate nutrition during pregnancy and early life is an important factor in avoiding non-communicable diseases such as obesity, type 2 diabetes, cardiovascular disease, cancers, and chronic allergic diseases. Although epidemiologic and experimental studies have shown that nutrition is important at all stages of life, it is especially important in prenatal and the first few years of life. During the last decade, there has been a growing interest in the potential role of epigenetic mechanisms in the increasing health problems associated with allergic disease. Epigenetics involves several mechanisms including DNA methylation, histone modifications, and microRNAs which can modify the expression of genes. In this study, we focus on the effects of maternal nutrition during pregnancy, the effects of the bioactive components in human and bovine milk, and the environmental factors that can affect early life (i.e., farming, milk processing, and bacterial exposure), and which contribute to the epigenetic mechanisms underlying the persistent programming of immune functions and allergic diseases. This knowledge will help to improve approaches to nutrition in early life and help prevent allergies in the future.
Collapse
Affiliation(s)
- Betty C. A. M. van Esch
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
- Danone Nutricia Research, Utrecht, Netherlands
| | - Mojtaba Porbahaie
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, Netherlands
| | - Suzanne Abbring
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
- Danone Nutricia Research, Utrecht, Netherlands
| | - Daniel P. Potaczek
- Institute of Laboratory Medicine, Member of the German Center for Lung Research (DZL), The Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University Marburg, Marburg, Germany
- John Paul II Hospital, Krakow, Poland
| | - Huub F. J. Savelkoul
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, Netherlands
| | - R. J. Joost van Neerven
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, Netherlands
- FrieslandCampina, Amersfoort, Netherlands
| |
Collapse
|
25
|
Ochoa T, Loli S, Mendoza K, Carcamo C, Bellomo S, Cam L, Castaneda A, Campos M, Jacobs J, Cossey V, Zegarra J. Effect of bovine lactoferrin on prevention of late-onset sepsis in infants <1500 g: a pooled analysis of individual patient data from two randomized controlled trials. Biochem Cell Biol 2020; 99:14-19. [PMID: 32931708 DOI: 10.1139/bcb-2020-0046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We previously conducted two randomized controlled trials with bovine lactoferrin (bLF) for the prevention of late-onset sepsis (LOS) in infants with a birth weight <2500 g (Study 1) and <2000 g (Study 2). The aim of this study was to determine the preventative effects of bLF on culture-proven or probable LOS in infants with a birth weight <1500 g from both studies, and to determine the effect of bLF in relation to intake of human milk. Both trial designs had similar inclusion and exclusion criteria, the same dose of bLF [200 mg·(kg body mass)-1·day-1], and used the same control (maltodextrin). We fitted multivariate Cox regression models to estimate the effect of bLF on the risk of development of the composite outcome, adjusting for covariates. We included 335 neonates with a mean birth weight of 1162 ± 244 g; 27.5% were <1000 g. There were 33 first episodes of LOS in the bLF treatment group and 48 in the control group (19.5% vs. 28.9%). bLF had a protective effect on the risk of development of LOS [hazard ratio (HR) = 0.64; %95 CI = 0.41-0.99; p = 0.048]; particularly among infants weighing <1000 g [HR = 0.46; %95 CI = 0.22-0.96; p = 0.039] and infants with a low intake of human milk [HR = 0.40; %95 CI = 0.19-0.84; p = 0.015]. Therefore, bLF supplementation protects infants <1500 g from LOS, particularly those infants not receiving human milk.
Collapse
Affiliation(s)
- Theresa Ochoa
- Facultad de Medicina, Universidad Peruana Cayetano Heredia, Lima, Peru.,School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA.,Doctoral School of Biomedial Sciences, KU Leuven, Belgium
| | - Sebastian Loli
- Facultad de Salud Pública y Administración, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Karina Mendoza
- Facultad de Salud Pública y Administración, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Cesar Carcamo
- Facultad de Salud Pública y Administración, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Sicilia Bellomo
- Facultad de Medicina, Universidad Peruana Cayetano Heredia, Lima, Peru.,Hospital Cayetano Heredia, Lima, Peru
| | - Luis Cam
- Hospital Nacional Alberto Sabogal, Lima, Peru
| | | | - Miguel Campos
- Facultad de Ciencias y Filosofia, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Jan Jacobs
- Department of Microbiology and Immunology, KU Leuven, Belgium.,Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Veerle Cossey
- Department of Development and Regeneration, KU Leuven, Belgium
| | - Jaime Zegarra
- Facultad de Medicina, Universidad Peruana Cayetano Heredia, Lima, Peru.,Hospital Cayetano Heredia, Lima, Peru
| |
Collapse
|
26
|
Elzoghby AO, Abdelmoneem MA, Hassanin IA, Abd Elwakil MM, Elnaggar MA, Mokhtar S, Fang JY, Elkhodairy KA. Lactoferrin, a multi-functional glycoprotein: Active therapeutic, drug nanocarrier & targeting ligand. Biomaterials 2020; 263:120355. [PMID: 32932142 PMCID: PMC7480805 DOI: 10.1016/j.biomaterials.2020.120355] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/18/2020] [Accepted: 08/31/2020] [Indexed: 12/21/2022]
Abstract
Recent progress in protein-based nanomedicine, inspired by the success of Abraxane® albumin-paclitaxel nanoparticles, have resulted in novel therapeutics used for treatment of challenging diseases like cancer and viral infections. However, absence of specific drug targeting, poor pharmacokinetics, premature drug release, and off-target toxicity are still formidable challenges in the clinic. Therefore, alternative protein-based nanomedicines were developed to overcome those challenges. In this regard, lactoferrin (Lf), a glycoprotein of transferrin family, offers a promising biodegradable well tolerated material that could be exploited both as an active therapeutic and drug nanocarrier. This review highlights the major pharmacological actions of Lf including anti-cancer, antiviral, and immunomodulatory actions. Delivery technologies of Lf to improve its pries and enhance its efficacy were also reviewed. Moreover, different nano-engineering strategies used for fabrication of drug-loaded Lf nanocarriers were discussed. In addition, the use of Lf for functionalization of drug nanocarriers with emphasis on tumor-targeted drug delivery was illustrated. Besides its wide application in oncology nano-therapeutics, we discussed the recent advances of Lf-based nanocarriers as efficient platforms for delivery of anti-parkinsonian, anti-Alzheimer, anti-viral drugs, immunomodulatory and bone engineering applications.
Collapse
Affiliation(s)
- Ahmed O Elzoghby
- Center for Engineered Therapeutics, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Harvard-MIT Division of Health Sciences & Technology (HST), Cambridge, MA, 02139, USA; Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt; Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt.
| | - Mona A Abdelmoneem
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt; Department of Pharmaceutics, Faculty of Pharmacy, Damanhur University, Damanhur, 22516, Egypt
| | - Islam A Hassanin
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt; Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, 21526, Egypt
| | - Mahmoud M Abd Elwakil
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt; Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, 060-0812, Japan
| | - Manar A Elnaggar
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt; Nanotechnology Program, School of Sciences & Engineering, The American University in Cairo (AUC), New Cairo, 11835, Egypt
| | - Sarah Mokhtar
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt; Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Taoyuan, 333, Taiwan; Research Center for Industry of Human Ecology, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Kweishan, Taoyuan, 333, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan, 333, Taiwan
| | - Kadria A Elkhodairy
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt; Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| |
Collapse
|
27
|
Pammi M, Preidis GA, Tarnow-Mordi WO. Evidence from systematic reviews of randomized trials on enteral lactoferrin supplementation in preterm neonates. Biochem Cell Biol 2020; 99:20-24. [PMID: 32721215 DOI: 10.1139/bcb-2020-0136] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In this commentary, we summarize the current evidence from randomized controlled trials on enteral lactoferrin supplementation in preterm neonates. Our recently completed systematic review includes 12 randomized controlled trials performed all over the world. Our meta-analysis suggests clinical benefit in decreasing late-onset sepsis, late-onset fungal sepsis, length of stay in the hospital and urinary tract infections. There were no adverse effects. There was no statistically significant decrease in necrotizing enterocolitis, mortality or neurodevelopmental impairment in lactoferrin supplemented preterm infants. There was significant statistical heterogeneity in the effects of lactoferrin on late-onset sepsis between larger and smaller studies, which may reflect either small study biases, differences in the effectiveness, dose or duration of supplemental lactoferrin products, or differences in underlying population risk, or any or all of these.
Collapse
Affiliation(s)
- Mohan Pammi
- Department of Pediatrics, Section of Neonatology, Baylor College of Medicine & Texas Children's Hospital, Houston, TX 77030, USA
| | - Geoffrey A Preidis
- Department of Pediatrics, Section of Gastroenterology, Hepatology & Nutrition, Baylor College of Medicine & Texas Children's Hospital, Houston, TX 77030, USA
| | - William O Tarnow-Mordi
- WINNER Centre for Newborn Research, NHMRC Clinical Trials Centre, University of Sydney, Camperdown, NSW 2050, Australia
| |
Collapse
|
28
|
Komine-Aizawa S, Ito S, Aizawa S, Namiki T, Hayakawa S. Cow milk exosomes activate NK cells and γδT cells in human PBMCs in vitro. Immunol Med 2020; 43:161-170. [PMID: 32649844 DOI: 10.1080/25785826.2020.1791400] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cow milk is a nourishing food containing numerous essential nutrients. In Japan, the consumption of cow milk is thought to enhance resistance to exhaustion-related diseases. Although several nutrients in cow milk, such as lactoferrin, are thought to modulate immune cells, the mechanisms remain unclear. Recently, the immunoregulatory functions of food-derived microRNAs or exosomes have been reported. Therefore, we studied the effects of exosomes derived from cow milk (CM-Exs) on immune cells in the present study. We obtained blood samples from healthy adult donors with the approval of the ethics committee. Peripheral blood mononuclear cells (PBMCs) were stimulated with CM-Exs in the absence or presence of interleukin-2 (IL-2) and IL-12. Cell surface markers and intracellular cytokine production were analysed by flow cytometry. CM-Ex stimulation enhanced the expression of CD69 on NK cells. Although CM-Ex stimulation alone did not induce interferon-γ (IFN-γ) production by NK cells or γδT cells, simultaneous stimulation with CM-Ex, IL-2 and IL-12 significantly enhanced IFN-γ production. In conclusion, cow milk consumption alone may not activate immune cells; however, CM-Exs could enhance immune cells under inflammatory conditions.
Collapse
Affiliation(s)
- Shihoko Komine-Aizawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Shun Ito
- Nihon University Itabashi Hospital, Tokyo, Japan
| | - Shu Aizawa
- Department of Animal Science, College of Bioresource Sciences, Nihon University, Kanagawa, Japan
| | | | - Satoshi Hayakawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
29
|
Jahan M, Francis N, Wang B. Milk lactoferrin concentration of primiparous and multiparous sows during lactation. J Dairy Sci 2020; 103:7521-7530. [PMID: 32448579 DOI: 10.3168/jds.2020-18148] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 03/18/2020] [Indexed: 01/21/2023]
Abstract
Lactoferrin (LF), a sialylated iron-binding glycoprotein, has numerous vital physiological functions including immunomodulation and protection against a large group of microorganisms, improving neurodevelopment, health, growth performance, and milk production. Lactoferrin occurs in human milk at a higher concentration compared with bovine milk, but little information is available on LF concentrations in porcine milk and the effects of sow parity on milk LF concentration. The objective of this study was to quantify the LF concentration in porcine milk and to compare that concentration between gilts and sows during lactation. We also investigated the effect of genetic background and litter size of the female pig on the LF concentration of porcine milk. The milk from 30 gilts and 35 sows was collected at 3 stages of lactation, namely colostrum, transition, and mature milk. Standard and experimental samples were analyzed by ultra-high performance liquid chromatography using a diode array UV detector. The following findings were reported: (1) porcine milk contained significant levels of LF with the highest concentration in colostrum, which decreased by ∼62% and ∼67% in transitional and mature milk, respectively; (2) mature gilt milk contained a 22% higher concentration of LF compared with sow milk, which was statistically significant; (3) breed line had an overall significant effect on the LF content of porcine milk; however, when the breed was considered, no significant difference was observed; and (4) LF concentration of porcine milk was not significantly influenced by the litter size. The presence of LF in a higher concentration in porcine milk suggests that LF is an important constituent of pig milk that might contribute to the optimum growth and development of piglets.
Collapse
Affiliation(s)
- M Jahan
- Graham Centre for Agricultural Innovation, Charles Sturt University, Wagga Wagga, NSW 2650, Australia; School of Animal and Veterinary Sciences, Charles Sturt University, Wagga Wagga, NSW 2678, Australia
| | - N Francis
- School of Animal and Veterinary Sciences, Charles Sturt University, Wagga Wagga, NSW 2678, Australia
| | - B Wang
- Graham Centre for Agricultural Innovation, Charles Sturt University, Wagga Wagga, NSW 2650, Australia; School of Animal and Veterinary Sciences, Charles Sturt University, Wagga Wagga, NSW 2678, Australia.
| |
Collapse
|
30
|
Early Effect of Supplemented Infant Formulae on Intestinal Biomarkers and Microbiota: A Randomized Clinical Trial. Nutrients 2020; 12:nu12051481. [PMID: 32443684 PMCID: PMC7284641 DOI: 10.3390/nu12051481] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/11/2020] [Accepted: 05/16/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Post-natal gut maturation in infants interrelates maturation of the morphology, digestive, and immunological functions and gut microbiota development. Here, we explored both microbiota development and markers of gut barrier and maturation in healthy term infants during their early life to assess the interconnection of gut functions during different infant formulae regimes. Methods: A total of 203 infants were enrolled in this randomized double-blind controlled trial including a breastfed reference group. Infants were fed starter formulae for the first four weeks of life, supplemented with different combination of nutrients (lactoferrin, probiotics (Bifidobacterium animal subsp. Lactis) and prebiotics (Bovine Milk-derived Oligosaccharides—BMOS)) and subsequently fed the control formula up to eight weeks of life. Stool microbiota profiles and biomarkers of early gut maturation, calprotectin (primary outcome), elastase, α-1 antitrypsin (AAT) and neopterin were measured in feces at one, two, four, and eight weeks. Results: Infants fed formula containing BMOS had lower mean calprotectin levels over the first two to four weeks compared to the other formula groups. Elastase and AAT levels were closer to levels observed in breastfed infants. No differences were observed for neopterin. Global differences between the bacterial communities of all groups were assessed by constrained multivariate analysis with hypothesis testing. The canonical correspondence analysis (CCA) at genus level showed overlap between microbiota profiles at one and four weeks of age in the BMOS supplemented formula group with the breastfed reference, dominated by bifidobacteria. Microbiota profiles of all groups at four weeks were significantly associated with the calprotectin levels at 4 (CCA, p = 0.018) and eight weeks of age (CCA, p = 0.026). Conclusion: A meaningful correlation was observed between changes in microbiota composition and gut maturation marker calprotectin. The supplementation with BMOS seems to favor gut maturation closer to that of breastfed infants.
Collapse
|
31
|
Tarnow-Mordi WO, Abdel-Latif ME, Martin A, Pammi M, Robledo K, Manzoni P, Osborn D, Lui K, Keech A, Hague W, Ghadge A, Travadi J, Brown R, Darlow BA, Liley H, Pritchard M, Kochar A, Isaacs D, Gordon A, Askie L, Cruz M, Schindler T, Dixon K, Deshpande G, Tracy M, Schofield D, Austin N, Sinn J, Simes RJ. The effect of lactoferrin supplementation on death or major morbidity in very low birthweight infants (LIFT): a multicentre, double-blind, randomised controlled trial. THE LANCET CHILD & ADOLESCENT HEALTH 2020; 4:444-454. [PMID: 32407710 DOI: 10.1016/s2352-4642(20)30093-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/06/2020] [Accepted: 03/23/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Very low birthweight or preterm infants are at increased risk of adverse outcomes including sepsis, necrotising enterocolitis, and death. We assessed whether supplementing the enteral diet of very low-birthweight infants with lactoferrin, an antimicrobial protein, reduces all-cause mortality or major morbidity. METHODS We did a multicentre, double-blind, pragmatic, randomised superiority trial in 14 Australian and two New Zealand neonatal intensive care units. Infants born weighing less than 1500 g and aged less than 8 days, were eligible and randomly assigned (1:1) using minimising web-based randomisation to receive once daily 200 mg/kg pasteurised bovine lactoferrin supplements or no lactoferrin supplement added to breast or formula milk until 34 weeks' post-menstrual age (or for 2 weeks, if longer), or until discharge from the study hospital if that occurred first. Designated nurses preparing the daily feeds were not masked to group assignment, but other nurses, doctors, parents, caregivers, and investigators were unaware. The primary outcome was survival to hospital discharge or major morbidity (defined as brain injury, necrotising enterocolitis, late-onset sepsis at 36 weeks' post-menstrual age, or retinopathy treated before discharge) assessed in the intention-to-treat population. Safety analyses were by treatment received. We also did a prespecified, PRISMA-compliant meta-analysis, which included this study and other relevant randomised controlled trials, to estimate more precisely the effects of lactoferrin supplementation on late-onset sepsis, necrotising enterocolitis, and survival. This trial is registered with the Australian and New Zealand Clinical Trials Registry, ACTRN12611000247976. FINDINGS Between June 27, 2014, and Sept 1, 2017, we recruited 1542 infants; 771 were assigned to the intervention group and 771 to the control group. One infant who had consent withdrawn before beginning lactoferrin treatment was excluded from analysis. In-hospital death or major morbidity occurred in 162 (21%) of 770 infants in the intervention group and in 170 (22%) of 771 infants in the control group (relative risk [RR] 0·95, 95% CI 0·79-1·14; p=0·60). Three suspected unexpected serious adverse reactions occurred; two in the lactoferrin group, namely unexplained late jaundice and inspissated milk syndrome, but were not attributed to the intervention and one in the control group had fatal inspissated milk syndrome. Our meta-analysis identified 13 trials completed before Feb 18, 2020, including this Article, in 5609 preterm infants. Lactoferrin supplements significantly reduced late-onset sepsis (RR 0·79, 95% CI 0·71-0·88; p<0·0001; I2=58%), but not necrotising enterocolitis or all-cause mortality. INTERPRETATION Lactoferrin supplementation did not improve death or major morbidity in this trial, but might reduce late-onset sepsis, as found in our meta-analysis of over 5000 infants. Future collaborative studies should use products with demonstrated biological activity, be large enough to detect moderate and clinically important effects reliably, and assess greater doses of lactoferrin in infants at increased risk, such as those not exclusively receiving breastmilk or infants of extremely low birthweight. FUNDING Australian National Health and Medical Research Council.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Kei Lui
- University of New South Wales, Kensington, NSW, Australia
| | | | - Wendy Hague
- University of Sydney, Sydney, NSW, Australia
| | | | | | | | | | - Helen Liley
- University of Queensland, Brisbane, QLD, Australia
| | | | - Anu Kochar
- University of Adelaide, Adelaide, SA, Australia
| | | | | | - Lisa Askie
- University of Sydney, Sydney, NSW, Australia
| | - Melinda Cruz
- Miracle Babies Foundation, Chipping Norton, NSW, Australia
| | - Tim Schindler
- University of New South Wales, Kensington, NSW, Australia
| | - Kelly Dixon
- University of Queensland, Brisbane, QLD, Australia
| | | | - Mark Tracy
- University of Sydney, Sydney, NSW, Australia
| | | | | | - John Sinn
- University of Sydney, Sydney, NSW, Australia
| | | | | |
Collapse
|
32
|
Ochoa TJ, Zegarra J, Bellomo S, Carcamo CP, Cam L, Castañeda A, Villavicencio A, Gonzales J, Rueda MS, Turin CG, Zea-Vera A, Guillen D, Campos M, Ewing-Cobbs L. Randomized Controlled Trial of Bovine Lactoferrin for Prevention of Sepsis and Neurodevelopment Impairment in Infants Weighing Less Than 2000 Grams. J Pediatr 2020; 219:118-125.e5. [PMID: 32037149 PMCID: PMC7096260 DOI: 10.1016/j.jpeds.2019.12.038] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 11/25/2019] [Accepted: 12/17/2019] [Indexed: 12/15/2022]
Abstract
OBJECTIVES To determine the effect of bovine lactoferrin on prevention of late-onset sepsis (LOS) and neurodevelopment delay. STUDY DESIGN Randomized, double-blind, controlled trial in neonates with a birth weight of 500-2000 g in 3 neonatal units in Lima, Peru, comparing bovine lactoferrin 200 mg/kg/day with placebo administered for 8 weeks. The primary outcome was the first episode of culture-proven LOS or sepsis-associated death. Neurodevelopment delay was assessed by the Mullen Scales at 24 months corrected age. RESULTS Of the 414 infants enrolled, 209 received bovine lactoferrin and 205 received placebo. LOS or sepsis-associated death occurred in 22 infants (10.5%) in the bovine lactoferrin group vs 30 (14.6%) in the placebo group; there was no difference after adjusting for hospital and birth weight; hazard ratio 0.73 (95% CI, 0.42-1.26). For infants with birth weights of <1500 g the hazard ratio was 0.69 (95% CI, 0.39-1.25). The mean age-adjusted normalized Mullen composite score at 24 months was 83.3 ± 13.6 in the bovine lactoferrin group vs 82.6 ± 13.1 in the placebo group. Growth outcomes and rehospitalization rates during the 2-year follow-up were similar in both groups, except for significantly less bronchiolitis in the bovine lactoferrin group (rate ratio, 0.34; 95% CI, 0.14-0.86). CONCLUSIONS Supplementation with bovine lactoferrin did not decrease the incidence of sepsis in infants with birth weights of <2000 g. Growth and neurodevelopment outcomes at 24 months of age were similar. Neonatal bovine lactoferrin supplementation had no adverse effects. TRIAL REGISTRATION ClinicalTrials.gov: NCT01525316.
Collapse
Affiliation(s)
- Theresa J. Ochoa
- Department of Pediatrics, School of Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru,Instituto de Medicina Tropical “Alexander von Humboldt”, Universidad Peruana Cayetano Heredia, Lima, Peru,Center for Infectious Diseases, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, United States
| | - Jaime Zegarra
- Department of Pediatrics, School of Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru,Hospital Cayetano Heredia, Lima, Peru
| | - Sicilia Bellomo
- Department of Pediatrics, School of Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru,Hospital Cayetano Heredia, Lima, Peru
| | - Cesar P. Carcamo
- School of Public Health and Administration, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Luis Cam
- Hospital Nacional Alberto Sabogal, Lima, Peru
| | | | - Aasith Villavicencio
- Department of Pediatrics, School of Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Jorge Gonzales
- Department of Pediatrics, School of Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Maria S. Rueda
- Department of Pediatrics, School of Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Christie G. Turin
- Department of Pediatrics, School of Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Alonso Zea-Vera
- Department of Pediatrics, School of Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Daniel Guillen
- Department of Pediatrics, School of Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru,Hospital Cayetano Heredia, Lima, Peru
| | - Miguel Campos
- Department of Mathematics, School of Science and Philosophy, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Linda Ewing-Cobbs
- Department of Pediatrics and Children’s Learning Institute, School of Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States
| | | |
Collapse
|
33
|
Pammi M, Suresh G. Enteral lactoferrin supplementation for prevention of sepsis and necrotizing enterocolitis in preterm infants. Cochrane Database Syst Rev 2020; 3:CD007137. [PMID: 32232984 PMCID: PMC7106972 DOI: 10.1002/14651858.cd007137.pub6] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Lactoferrin, a normal component of human colostrum and milk, can enhance host defenses and may be effective for prevention of sepsis and necrotizing enterocolitis (NEC) in preterm neonates. OBJECTIVES To assess the safety and effectiveness of lactoferrin supplementation to enteral feeds for prevention of sepsis and NEC in preterm neonates. Secondarily, we assessed the effects of lactoferrin supplementation to enteral feeds on the duration of positive-pressure ventilation, development of chronic lung disease (CLD) or periventricular leukomalacia (PVL), length of hospital stay to discharge among survivors, and adverse neurological outcomes at two years of age or later. SEARCH METHODS We used the standard search strategy of Cochrane Neonatal to update our search. We searched the Cochrane Central Register of Controlled Trials (CENTRAL 2019, Issue 9), MEDLINE via PubMed (1966 to 20 January 2020), PREMEDLINE (1996 to 20 January 2020), Embase (1980 to 20 January 2020), and CINAHL (1982 to 20 January 2020). We also searched clinical trials databases, conference proceedings, and the reference lists of retrieved articles for randomized controlled trials and quasi-randomized trials. SELECTION CRITERIA In our search, we included randomized controlled trials (RCTs) evaluating enteral lactoferrin supplementation at any dose or duration to prevent sepsis or NEC in preterm neonates. DATA COLLECTION AND ANALYSIS We used the standard methods of Cochrane Neonatal and the GRADE approach to assess the certainty of evidence. MAIN RESULTS Meta-analysis of data from twelve randomized controlled trials showed that lactoferrin supplementation to enteral feeds decreased late-onset sepsis (typical RR 0.82, 95% CI 0.74 to 0.91; typical RD -0.04, 95% CI, -0.06, -0.02; NNTB 25, 95% CI 17 to 50; 12 studies, 5425 participants, low-certainty evidence) and decreased length of hospital stay (MD -2.38, 95% CI, -4.67, -0.09; 3 studies, 1079 participants, low-certainty evidence). Sensitivity analysis including only good methodological certainty studies suggested a decrease in late-onset sepsis with enteral lactoferrin supplementation (typical RR 0.87, 95% CI, 0.78, 0.97; typical RD -0.03, 95% CI, -0.05, -0.0; 9 studies, 4702 participants, low-certainty evidence). There were no differences in NEC stage II or III (typical RR 1.10, 95% CI, 0.86, 1.41; typical RD -0.00, 95% CI, -0.02, 0.01; 7 studies, 4874 participants; low-certainty evidence) or 'all-cause mortality' (typical RR 0.90, 95% CI 0.69, 1.17; typical RD -0.00, 95% CI, -0.01, 0.01; 11 studies, 5510 participants; moderate-certainty evidence). One study reported no differences in neurodevelopmental testing by Mullen's or Bayley III at 24 months of age after enteral lactoferrin supplementation (one study, 292 participants, low-certainty evidence). Lactoferrin supplementation to enteral feeds with probiotics decreased late-onset sepsis (RR 0.25, 95% CI 0.14 to 0.46; RD -0.13, 95% CI -0.18 to -0.08; NNTB 8, 95% CI 6 to 13; 3 studies, 564 participants; low-certainty evidence) and NEC stage II or III (RR 0.04, 95% CI 0.00 to 0.62; RD -0.05, 95% CI -0.08 to -0.03; NNTB 20, 95% CI 12.5 to 33.3; 1 study, 496 participants; very low-certainty evidence), but not 'all-cause mortality' (very low-certainty evidence). Lactoferrin supplementation to enteral feeds with or without probiotics had no effect on CLD, duration of mechanical ventilation or threshold retinopathy of prematurity (low-certainty evidence). Investigators reported no adverse effects in the included studies. AUTHORS' CONCLUSIONS We found low-certainty evidence from studies of good methodological quality that lactoferrin supplementation of enteral feeds decreases late-onset sepsis but not NEC ≥ stage II or 'all cause mortality' or neurodevelopmental outcomes at 24 months of age in preterm infants without adverse effects. Low- to very low-certainty evidence suggests that lactoferrin supplementation of enteral feeds in combination with probiotics decreases late-onset sepsis and NEC ≥ stage II in preterm infants without adverse effects, however, there were few included studies of poor methodological quality. The presence of publication bias and small studies of poor methodology that may inflate the effect size make recommendations for clinical practice difficult.
Collapse
Affiliation(s)
- Mohan Pammi
- Baylor College of Medicine, Section of Neonatology, Department of Pediatrics, 6621, Fannin, MC.WT 6-104, Houston, Texas, USA, 77030
| | - Gautham Suresh
- Baylor College of Medicine, Section of Neonatology, Department of Pediatrics, 6621, Fannin, MC.WT 6-104, Houston, Texas, USA, 77030
| |
Collapse
|
34
|
Ochoa TJ, Mendoza K, Carcamo C, Zegarra J, Bellomo S, Jacobs J, Cossey V. Is Mother's Own Milk Lactoferrin Intake Associated with Reduced Neonatal Sepsis, Necrotizing Enterocolitis, and Death? Neonatology 2020; 117:167-174. [PMID: 32053823 PMCID: PMC7381382 DOI: 10.1159/000505663] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 12/28/2019] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Lactoferrin (LF) is a protective protein present in milk with anti-infective and immune-modulating properties. OBJECTIVES The aim of this study was to determine the association of maternal LF intake and mother's own milk intake in the first 10 days of life on the prevention of late-onset sepsis (LOS), necrotizing enterocolitis (NEC), or death in the first 8 weeks of life in newborns with a birth weight <2,000 g. METHODS A retrospective cohort study was conducted, with the exposure being the consumption of mother's own LF and mother's own milk in the first 10 days of life, and the outcome being LOS, NEC, or death during days 11 and 56 of life, analyzed by Cox regression. RESULTS Two hundred and ninety-nine infants were enrolled, including 240 with human LF intake information. The average daily human LF intake over days 4-10 of life was 283 mg/kg/day (IQR 114-606 mg/kg/day). The hazard ratio (HR) of mother's own milk LF intake ≥100 mg/kg/day in days 4-10 for LOS, NEC, or death was 0.297 (95% CI 0.156-0.568, p < 0.001); the adjusted HR was 0.752 (95% CI 0.301-1.877, p = 0.541). The adjusted HR of mother's own milk cumulative intake (days 4-10) of 54-344 mL/kg (25-75 quartiles) for LOS, NEC, or death was 0.414 (95% CI 0.196-0.873, p = 0.02). Infants who developed an event (LOS, NEC, or death) had significantly less median daily human LF intake than those that did not (89 vs. 334 mg/kg/day, respectively, p < 0.0001). CONCLUSION Consumption of higher amounts of mother's own milk in the first days of life is associated with less infection, NEC, and death. Early human milk intake should be strongly encouraged in all newborns.
Collapse
Affiliation(s)
- Theresa J Ochoa
- Facultad de Medicina, Universidad Peruana Cayetano Heredia, Lima, Peru, .,School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA, .,Doctoral School of Biomedical Sciences, KU Leuven, Leuven, Belgium,
| | - Karina Mendoza
- Facultad de Salud Pública y Administración, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Cesar Carcamo
- Facultad de Salud Pública y Administración, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Jaime Zegarra
- Facultad de Medicina, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Sicilia Bellomo
- Facultad de Medicina, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Jan Jacobs
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium.,Department of Clinical Science, Institute of Tropical Medicine, Antwerp, Belgium
| | - Veerle Cossey
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| |
Collapse
|
35
|
Li F, Wu SS, Berseth CL, Harris CL, Richards JD, Wampler JL, Zhuang W, Cleghorn G, Rudolph CD, Liu B, Shaddy DJ, Colombo J. Improved Neurodevelopmental Outcomes Associated with Bovine Milk Fat Globule Membrane and Lactoferrin in Infant Formula: A Randomized, Controlled Trial. J Pediatr 2019; 215:24-31.e8. [PMID: 31668885 DOI: 10.1016/j.jpeds.2019.08.030] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 08/02/2019] [Accepted: 08/14/2019] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To evaluate neurodevelopment, growth, and health outcomes in infants receiving bovine milk fat globule membrane (MFGM) and lactoferrin in infant formula. STUDY DESIGN Healthy term infants were randomized to a cow's milk-based infant formula or MFGM + LF (a similar infant formula, with an added source of bovine milk fat globule membrane [bMFGM; whey protein-lipid concentrate, 5 g/L] and bovine lactoferrin [0.6 g/L]) through 365 days of age. The Bayley Scales of Infant Development, 3rd edition cognitive composite score at day 365 was the primary outcome. Secondary outcomes included tolerance measures through day 365, additional neurodevelopmental and language outcomes, growth, and medically confirmed adverse events through day 545. RESULTS Of 451 infants enrolled (control, 228; MFGM + LF, 223), 291 completed study feeding and Bayley-III testing at day 365 (control, 148; MFGM + LF, 143). The mean cognitive (+8.7), language (+12.3), and motor (+12.6) scores were higher (P < .001) for the MFGM + LF group; no differences were observed at day 545. Global development scores from day 120 to day 275 and attention at day 365 were significantly improved. Few group differences in day 545 neurodevelopmental outcomes were detected, however scores of some subcategories of the MacArthur-Bates Communicative Development Inventories were higher (P < .05) in the MFGM + LF group. The overall incidence of respiratory-associated adverse events and diarrhea were significantly lower for the MFGM + LF group through day 545. CONCLUSIONS Infants receiving formula with added bovine MFGM and bovine lactoferrin had an accelerated neurodevelopmental profile at day 365 and improved language subcategories at day 545. Formulas were associated with age-appropriate growth and significantly fewer diarrhea and respiratory-associated adverse events through 545 days of age. TRIAL REGISTRATION CLINICALTRIALS.GOV:: NCT02274883.
Collapse
Affiliation(s)
- Fei Li
- Departments of Developmental and Behavioral Pediatrics & Child Primary Care, MOE-Shanghai Key Lab for Children's Environmental Health, Xinhua Hospital Affiliated To Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Steven S Wu
- Clinical Research, Department of Medical Affairs, Mead Johnson Nutrition, Evansville, IN.
| | - Carol Lynn Berseth
- Clinical Research, Department of Medical Affairs, Mead Johnson Nutrition, Evansville, IN; Medical and Scientific Affairs, Brightseed, San Francisco, California
| | - Cheryl L Harris
- Clinical Research, Department of Medical Affairs, Mead Johnson Nutrition, Evansville, IN
| | - James D Richards
- Nutrition Science, Department of Medical Affairs, Mead Johnson Nutrition, Evansville, IN; DSM Nutritional Products, Parsippany, New Jersey
| | - Jennifer L Wampler
- Clinical Research, Department of Medical Affairs, Mead Johnson Nutrition, Evansville, IN
| | - Weihong Zhuang
- Clinical Research, Department of Medical Affairs, Mead Johnson Nutrition, Evansville, IN
| | - Geoffrey Cleghorn
- Clinical Research, Department of Medical Affairs, Mead Johnson Nutrition, Evansville, IN; Faculty of Health, Queensland University of Technology, Brisbane, Australia
| | - Colin D Rudolph
- Clinical Research, Department of Medical Affairs, Mead Johnson Nutrition, Evansville, IN; Department of Pediatrics, University of California, San Francisco, CA
| | - Bryan Liu
- Clinical Research, Department of Medical Affairs, Mead Johnson Nutrition, Evansville, IN; College of Biotechnology, East China University of Science and Technology, Shanghai, China
| | - D Jill Shaddy
- Schiefelbusch Institute for Life Span Studies, University of Kansas, Lawrence, KS
| | - John Colombo
- Schiefelbusch Institute for Life Span Studies, University of Kansas, Lawrence, KS
| |
Collapse
|
36
|
Pempek JA, Watkins LR, Bruner CE, Habing GG. A multisite, randomized field trial to evaluate the influence of lactoferrin on the morbidity and mortality of dairy calves with diarrhea. J Dairy Sci 2019; 102:9259-9267. [PMID: 31400894 PMCID: PMC7094274 DOI: 10.3168/jds.2019-16476] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 05/20/2019] [Indexed: 11/25/2022]
Abstract
Diarrhea is one of the most common causes of antimicrobial use and mortality in young calves. To reduce antimicrobial use and resistance on dairy farms, research on alternative therapies for calf diarrhea is necessary. Our laboratory previously conducted a randomized clinical trial investigating the effectiveness of lactoferrin, an iron-binding protein found in colostrum, as a treatment for calf diarrhea. The trial showed significantly reduced calf mortality in diarrheic calves that were administered lactoferrin. Thus, the objective of this study was to corroborate the results of our prior clinical trial across multiple farms and to investigate the effect of lactoferrin on the morbidity and mortality of preweaned calves with naturally occurring cases of diarrhea. This randomized field trial was conducted on 5 commercial dairy farms in Ohio. In total, 485 calves (≤21 d of age) were enrolled at first diarrhea diagnosis (fecal score ≥2 defined as loose to watery) and randomly assigned to receive an oral dose of lactoferrin (3 g of lactoferrin powder dissolved in 30 mL of water) or 30 mL of water (control) once daily for 3 consecutive days. Health assessments were conducted on the day of diarrhea diagnosis (d 0) and 1, 2, 3, 7, 14, 21, 28, and 35 d following diagnosis. Producer records of disease treatment and mortality were collected 120 d following diagnosis. A Poisson regression model was used to test differences between treatments in disease frequency through 35 d post-diarrhea diagnosis and the incidence risk of treatment and mortality risk 120 d post-diarrhea diagnosis; the model controlled for calf age at enrollment, farm, and treatment. Median calf age at enrollment was 11 d and ranged from 1 to 26 d of age. At study enrollment, 51.3% (123/240) and 52.2% (128/245) of calves in the control and lactoferrin treatment groups, respectively, were diagnosed with severe diarrhea (fecal score = 3). The frequency of disease (diarrhea, dehydration, depression, signs of respiratory disease) through 35 d following diarrhea diagnosis was not significantly different for calves in the lactoferrin and control groups. Overall mortality risk for enrolled calves was 9.9%, and 10.7% (22/243) and 9.1% (26/242) of calves in the lactoferrin and control groups, respectively, died or were culled in the 120 d following diarrhea diagnosis. The relative risk of death or culling did not differ between treatment groups, however. Therefore, as performed in this study, lactoferrin as a treatment for calf diarrhea was not beneficial.
Collapse
Affiliation(s)
- J A Pempek
- Department of Veterinary Preventive Medicine, The Ohio State University, Columbus 43210
| | - L R Watkins
- Department of Veterinary Preventive Medicine, The Ohio State University, Columbus 43210
| | - C E Bruner
- Department of Veterinary Preventive Medicine, The Ohio State University, Columbus 43210
| | - G G Habing
- Department of Veterinary Preventive Medicine, The Ohio State University, Columbus 43210.
| |
Collapse
|
37
|
Hård A, Nilsson AK, Lund A, Hansen‐Pupp I, Smith LEH, Hellström A. Review shows that donor milk does not promote the growth and development of preterm infants as well as maternal milk. Acta Paediatr 2019; 108:998-1007. [PMID: 30565323 PMCID: PMC6520191 DOI: 10.1111/apa.14702] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 10/09/2018] [Accepted: 12/12/2018] [Indexed: 12/28/2022]
Abstract
Aim This nonsystematic review examined differences in the composition of raw maternal breastmilk and pasteurised donor milk and possible health effects on preterm infants. Methods We searched PubMed up to July 2018 for studies published in English that focused on four comparisons as follows: raw maternal milk versus donor milk, human milk before and after Holder pasteurisation, milk from mothers who delivered preterm and at term and milk collected during early and late lactation. We also searched for possible effects of the milk components, as well as the effects of maternal and donor milk on preterm infants’ health. Results Raw maternal milk contained factors involved in antioxidant and anti‐inflammatory defence, gut microbiome establishment and the maturation of immune defences, food tolerability and metabolism. Many of these factors were reduced or abolished in processed donor milk. Both maternal milk and donor milk have been associated with a reduced incidence of necrotising enterocolitis. High‐dose feeding with maternal milk during the neonatal period reportedly reduced the risk of other morbidities and promoted growth and neurodevelopment. Conclusion Many of the components in raw maternal breastmilk were lacking in pasteurised donor milk, which was inferior in promoting the growth and development of very preterm infants.
Collapse
Affiliation(s)
- Anna‐Lena Hård
- Department of Ophthalmology Institute of Neuroscience and Physiology Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| | - Anders K. Nilsson
- Department of Ophthalmology Institute of Neuroscience and Physiology Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| | - Anna‐My Lund
- Department of Pediatrics Institute of Clinical Sciences Lund Lund University and Skane University Hospital Lund Sweden
| | - Ingrid Hansen‐Pupp
- Department of Pediatrics Institute of Clinical Sciences Lund Lund University and Skane University Hospital Lund Sweden
| | - Lois E. H. Smith
- Department of Ophthalmology Boston Children's Hospital Harvard Medical School Boston MA USA
| | - Ann Hellström
- Department of Ophthalmology Institute of Neuroscience and Physiology Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| |
Collapse
|
38
|
Zingg W, Park BJ, Storr J, Ahmad R, Tarrant C, Castro-Sanchez E, Perencevich E, Widmer A, Krause KH, Kilpatrick C, Tomczyk S, Allegranzi B, Cardo D, Pittet D. Technology for the prevention of antimicrobial resistance and healthcare-associated infections; 2017 Geneva IPC-Think Tank (Part 2). Antimicrob Resist Infect Control 2019; 8:83. [PMID: 31139366 PMCID: PMC6530187 DOI: 10.1186/s13756-019-0538-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 05/13/2019] [Indexed: 11/10/2022] Open
Abstract
Background The high burden of healthcare-associated infections (HAIs) and antimicrobial resistance (AMR) is partially due to excessive antimicrobial use both in human and animal medicine worldwide. How can technology help to overcome challenges in infection prevention and control (IPC) and to prevent HAI and emerging AMR? Methods In June 2017, 42 international experts convened in Geneva, Switzerland to discuss four potential domains of technology in IPC and AMR: 1) role and potential contribution of microbiome research; 2) whole genome sequencing; 3) effectiveness and benefit of antimicrobial environmental surfaces; and 4) future research in hand hygiene. Results Research on the microbiome could expand understanding of antimicrobial use and also the role of probiotics or even faecal transplantation for therapeutic purposes. Whole genome sequencing will provide new insights in modes of transmission of infectious diseases. Although it is a powerful tool for public health epidemiology, some challenges with interpretation and costs still need to be addressed. The effectiveness and cost-effectiveness of antimicrobially coated or treated environmental high-touch surfaces requires further research before they can be recommended for routine use. Hand hygiene implementation can be advanced, where technological enhancement of surveillance, technique and compliance are coupled with reminders for healthcare professionals. Conclusions The four domains of technological innovation contribute to the prevention of HAI and AMR at different levels. Microbiome research may offer innovative concepts for future prevention, whole genome sequencing could detect new modes of transmission and become an additional tool for effective public health epidemiology, antimicrobial surfaces might help to decrease the environment as source of transmission but continue to raise more questions than answers, and technological innovation may have a role in improving surveillance approaches and supporting best practice in hand hygiene.
Collapse
Affiliation(s)
- Walter Zingg
- Infection control programme and WHO collaborating center, University of Geneva Hospitals and Faculty of Medicine, 4 Rue Gabrielle Perret-Gentil, 1211, Geneva 14, Switzerland
| | - Benjamin J Park
- 2US Centers for Disease Control and Prevention, Atlanta, GA USA
| | - Julie Storr
- 3Infection Prevention and Control Global Unit, World Health Organization, Geneva, Switzerland
| | - Raheelah Ahmad
- 4National Institute for Health Research in Healthcare Associated Infection and Antimicrobial Resistance, Imperial College London, London, UK
| | - Carolyn Tarrant
- 5Department of Health Sciences, University of Leicester, Leicester, UK
| | - Enrique Castro-Sanchez
- 4National Institute for Health Research in Healthcare Associated Infection and Antimicrobial Resistance, Imperial College London, London, UK
| | - Eli Perencevich
- 6Carver College of Medicine, University of Iowa, Iowa City, USA
| | - Andreas Widmer
- 7Infection Control Programme, University Hospitals of Basel, Basel, Switzerland
| | - Karl-Heinz Krause
- 8Institute of Genetics and Genomics, University of Geneva, Geneva, Switzerland
| | - Claire Kilpatrick
- 3Infection Prevention and Control Global Unit, World Health Organization, Geneva, Switzerland
| | - Sara Tomczyk
- 3Infection Prevention and Control Global Unit, World Health Organization, Geneva, Switzerland
| | - Benedetta Allegranzi
- 3Infection Prevention and Control Global Unit, World Health Organization, Geneva, Switzerland
| | - Denise Cardo
- 2US Centers for Disease Control and Prevention, Atlanta, GA USA
| | - Didier Pittet
- Infection control programme and WHO collaborating center, University of Geneva Hospitals and Faculty of Medicine, 4 Rue Gabrielle Perret-Gentil, 1211, Geneva 14, Switzerland
| | | |
Collapse
|
39
|
Griffiths J, Jenkins P, Vargova M, Bowler U, Juszczak E, King A, Linsell L, Murray D, Partlett C, Patel M, Berrington J, Embleton N, Dorling J, Heath PT, McGuire W, Oddie S. Enteral lactoferrin to prevent infection for very preterm infants: the ELFIN RCT. Health Technol Assess 2019; 22:1-60. [PMID: 30574860 DOI: 10.3310/hta22740] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Infections acquired in hospital are an important cause of morbidity and mortality in very preterm infants. Several small trials have suggested that supplementing the enteral diet of very preterm infants with lactoferrin, an antimicrobial protein processed from cow's milk, prevents infections and associated complications. OBJECTIVE To determine whether or not enteral supplementation with bovine lactoferrin (The Tatua Cooperative Dairy Company Ltd, Morrinsville, New Zealand) reduces the risk of late-onset infection (acquired > 72 hours after birth) and other morbidity and mortality in very preterm infants. DESIGN Randomised, placebo-controlled, parallel-group trial. Randomisation was via a web-based portal and used an algorithm that minimised for recruitment site, weeks of gestation, sex and single versus multiple births. SETTING UK neonatal units between May 2014 and September 2017. PARTICIPANTS Infants born at < 32 weeks' gestation and aged < 72 hours at trial enrolment. INTERVENTIONS Eligible infants were allocated individually (1 : 1 ratio) to receive enteral bovine lactoferrin (150 mg/kg/day; maximum 300 mg/day) or sucrose (British Sugar, Peterborough, UK) placebo (same dose) once daily from trial entry until a postmenstrual age of 34 weeks. Parents, caregivers and outcome assessors were unaware of group assignment. OUTCOMES Primary outcome - microbiologically confirmed or clinically suspected late-onset infection. Secondary outcomes - microbiologically confirmed infection; all-cause mortality; severe necrotising enterocolitis (NEC); retinopathy of prematurity (ROP); bronchopulmonary dysplasia (BPD); a composite of infection, NEC, ROP, BPD and mortality; days of receipt of antimicrobials until 34 weeks' postmenstrual age; length of stay in hospital; and length of stay in intensive care, high-dependency and special-care settings. RESULTS Of 2203 enrolled infants, primary outcome data were available for 2182 infants (99%). In the intervention group, 316 out of 1093 (28.9%) infants acquired a late-onset infection versus 334 out of 1089 (30.7%) infants in the control group [adjusted risk ratio (RR) 0.95, 95% confidence interval (CI) 0.86 to 1.04]. There were no significant differences in any secondary outcomes: microbiologically confirmed infection (RR 1.05, 99% CI 0.87 to 1.26), mortality (RR 1.05, 99% CI 0.66 to 1.68), NEC (RR 1.13, 99% CI 0.68 to 1.89), ROP (RR 0.89, 99% CI 0.62 to 1.28), BPD (RR 1.01, 99% CI 0.90 to 1.13), or a composite of infection, NEC, ROP, BPD and mortality (RR 1.01, 99% CI 0.94 to 1.08). There were no differences in the number of days of receipt of antimicrobials, length of stay in hospital, or length of stay in intensive care, high-dependency or special-care settings. There were 16 reports of serious adverse events for infants in the lactoferrin group and 10 for infants in the sucrose group. CONCLUSIONS Enteral supplementation with bovine lactoferrin does not reduce the incidence of infection, mortality or other morbidity in very preterm infants. FUTURE WORK Increase the precision of the estimates of effect on rarer secondary outcomes by combining the data in a meta-analysis with data from other trials. A mechanistic study is being conducted in a subgroup of trial participants to explore whether or not lactoferrin supplementation affects the intestinal microbiome and metabolite profile of very preterm infants. TRIAL REGISTRATION Current Controlled Trials ISRCTN88261002. FUNDING This project was funded by the National Institute for Health Research (NIHR) Health Technology Assessment programme and will be published in full in Health Technology Assessment; Vol. 22, No. 74. See the NIHR Journals Library website for further project information. This trial was also sponsored by the University of Oxford, Oxford, UK. The funder provided advice and support and monitored study progress but did not have a role in study design or data collection, analysis and interpretation.
Collapse
Affiliation(s)
- James Griffiths
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - Paula Jenkins
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - Monika Vargova
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - Ursula Bowler
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - Edmund Juszczak
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - Andrew King
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - Louise Linsell
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - David Murray
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | | | | | - Janet Berrington
- Newcastle Neonatal Service, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | - Nicholas Embleton
- Newcastle Neonatal Service, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | | | - Paul T Heath
- St George's, University of London and St George's University Hospitals NHS Trust, London, UK
| | - William McGuire
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Sam Oddie
- Bradford Institute for Health Research, Bradford, UK
| |
Collapse
|
40
|
Affiliation(s)
- Paolo Manzoni
- Division of Pediatrics and Neonatology, Degli Infermi Hospital, Biella, Italy
| |
Collapse
|
41
|
Porcine lactoferrin-derived peptide LFP-20 modulates immune homoeostasis to defend lipopolysaccharide-triggered intestinal inflammation in mice. Br J Nutr 2019; 121:1255-1263. [DOI: 10.1017/s0007114519000485] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
AbstractThe performance of immune system is vital for defending the body from pathogens, and it plays a crucial role in health homoeostasis. In a previous study, we have shown that LFP-20, a twenty-amino acid antimicrobial peptide in the N terminus of porcine lactoferrin, modulated inflammatory response in colitis. Here, we further investigated the effects of LFP-20 on immune homoeostasis to elucidate the mechanism of its anti-inflammation action. A lipopolysaccharide (LPS)-triggered systemic inflammatory response mice model was established. On the basis of observed mucosal lesions and apoptosis in small intestine, we found increased macrophage and neutrophil infiltration in ileum after LPS stimulation. Expectedly, LFP-20 pre-treatment attenuated the LPS-mediated immune disorders in ileum. Moreover, the flow cytometry results indicated pre-treatment with LFP-20 sustained the balance of CD3+CD8+ T cells, B cells and natural killer cells in LPS-triggered immune disturbance. Simultaneously, we demonstrated LFP-20 modulated the secretion of both activated Th1-related IL-12p70, interferon-γ, TNF-α and Th2-related IL-4, IL-5 and IL-6. Furthermore, we found LFP-20 facilitated a balanced Th1 and Th2 response, which triggered cellular defence mechanisms and induced B cells to produce opsonising antibodies belonging to certain IgG subclasses to defend against LPS stimulation. Collectively, our study indicated pre-treatment with LFP-20 could defend against LPS-triggered systemic inflammatory response in mice via modulating immune homoeostasis.
Collapse
|
42
|
Dietary Supplementation With Medium-Chain Triglycerides Reduces Candida Gastrointestinal Colonization in Preterm Infants. Pediatr Infect Dis J 2019; 38:164-168. [PMID: 29596218 PMCID: PMC6604858 DOI: 10.1097/inf.0000000000002042] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Candida is an important cause of infections in premature infants. Gastrointestinal colonization with Candida is a common site of entry for disseminated disease. The objective of this study was to determine whether a dietary supplement of medium-chain triglycerides (MCTs) reduces Candida colonization in preterm infants. METHODS Preterm infants with Candida colonization (n = 12) receiving enteral feedings of either infant formula (n = 5) or breast milk (n = 7) were randomized to MCT supplementation (n = 8) or no supplementation (n = 4). Daily stool samples were collected to determine fungal burden during a 3-week study period. Infants in the MCT group received supplementation during 1 week of the study period. The primary outcome was fungal burden during the supplementation period as compared with the periods before and after supplementation. RESULTS Supplementation of MCT led to a marked increase in MCT intake relative to unsupplemented breast milk or formula as measured by capric acid content. In the treatment group, there was a significant reduction in fungal burden during the supplementation period as compared with the period before supplementation (rate ratio, 0.15; P = 0.02), with a significant increase after supplementation was stopped (rate ratio, 61; P < 0.001). Fungal burden in the control group did not show similar changes. CONCLUSIONS Dietary supplementation with MCT may be an effective method to reduce Candida colonization in preterm infants.
Collapse
|
43
|
Liao H, Liu S, Wang H, Su H, Liu Z. Enhanced antifungal activity of bovine lactoferrin-producing probiotic Lactobacillus casei in the murine model of vulvovaginal candidiasis. BMC Microbiol 2019; 19:7. [PMID: 30621597 PMCID: PMC6325727 DOI: 10.1186/s12866-018-1370-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 12/10/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Vulvovaginal candidiasis (VVC) is a common vaginitis caused by Candida species,a frequently recurring condition. Fungal azole-resistant strains with azole-resistance have developed for long and wide explosion to the first-line antifungal azole agent. Bovine lactoferrin (BLF) is a protein from transferrin family secreted by the bovine mammary tissue. Its various biological functions are well known, especially the pronounced antifungal activity. RESULTS In the current study, we constructed a Lactobacillus casei strain (L.casei/pPG612.1-BLF), which secreted BLF encoded by a mature secretion vector plasmid pPG612.1, and evaluated its antifungal activity in vitro and in vivo. In a two-layer agar plate in vitro assay, the number of C. albicans CFUs decreased and the average colony size shrunk upon exposure to L. casei/pPG612.1-BLF. In a murine VVC model, the infection burden of mice intra-vaginally pre-inoculated with L. casei/pPG612.1-BLF was lower than in control groups. Furthermore, the infection burden in mice with VVC was reduced when the animals were continually given L. casei/pPG612.1-BLF as a topical treatment for 5 days. CONCLUSION Combined, these results suggested that the L. casei/pPG612.1-BLF strain is a promising preventative and therapeutic anti-VVC agent, highlighting the possibility of employing the probiotic L. casei as a vehicle for biotherapy in the female genital tract and exploiting the natural antibiotic antimicrobial peptides for other applications.
Collapse
Affiliation(s)
- Hong Liao
- Department of Obstetric & Gynecologic, West China Second University Hospital, Sichuan University, NO. 17.3rd Section, Renmin South Road, Chengdu, Sichuan 610041 China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Shanling Liu
- Department of Obstetric & Gynecologic, West China Second University Hospital, Sichuan University, NO. 17.3rd Section, Renmin South Road, Chengdu, Sichuan 610041 China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - He Wang
- Department of Obstetric & Gynecologic, West China Second University Hospital, Sichuan University, NO. 17.3rd Section, Renmin South Road, Chengdu, Sichuan 610041 China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Hang Su
- Department of Obstetric & Gynecologic, West China Second University Hospital, Sichuan University, NO. 17.3rd Section, Renmin South Road, Chengdu, Sichuan 610041 China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Zhenjun Liu
- Department of Obstetric & Gynecologic, West China Second University Hospital, Sichuan University, NO. 17.3rd Section, Renmin South Road, Chengdu, Sichuan 610041 China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| |
Collapse
|
44
|
van Splunter M, Perdijk O, Fick-Brinkhof H, Feitsma AL, Floris-Vollenbroek EG, Meijer B, Brugman S, Savelkoul HFJ, van Hoffen E, van Neerven RJJ. Bovine Lactoferrin Enhances TLR7-Mediated Responses in Plasmacytoid Dendritic Cells in Elderly Women: Results From a Nutritional Intervention Study With Bovine Lactoferrin, GOS and Vitamin D. Front Immunol 2018; 9:2677. [PMID: 30515164 PMCID: PMC6255898 DOI: 10.3389/fimmu.2018.02677] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 10/30/2018] [Indexed: 01/15/2023] Open
Abstract
During aging the immune system is dysregulated. Especially plasmacytoid dendritic cells (pDCs) and myeloid DCs (mDCs) have reduced Toll like receptor (TLR)-mediated responses resulting in increased susceptibility to infections. Consumption of bovine lactoferrin (bLF) has been shown to reduce infections with viruses. Galacto-oligosacharides (GOS) and vitamin D are associated with reduced pro-inflammatory cytokine levels in serum, and increased TLR7/8 responses, respectively. A double-blind placebo-controlled nutritional intervention study in elderly women was performed, to investigate the potential of bLF, GOS, and vitamin D to restore TLR responsiveness of pDCs and mDCs and to reduce inflammatory markers in serum. The nutritional intervention group (n = 15) received bLF for 3 weeks, followed by 3 weeks of bLF + GOS, and subsequently 3 weeks of bLF + GOS + vitamin D. The placebo group (n = 15) received maltodextrin for 9 weeks. Every 3 weeks, blood was collected and TLR responses of pDCs and mDCs, and inflammation-related markers in serum were measured. After 3 weeks of bLF supplementation, increased TLR7/8 and TLR1/2 responses were observed in pDCs of the nutritional intervention group compared to the placebo group. When the effects of the entire nutritional intervention were investigated, increased TLR1/2 mediated responses in mDCs were observed, and in serum sVCAM tended to decrease. Finally, based on the RAND-36 questionnaire physical function tended to improve in the intervention group. Since especially TLR7-mediated responses in pDCs were enhanced after bLF supplementation compared to placebo, this suggests that bLF may contribute to antiviral responses mediated by pDC in elderly women.Clinical trial registry number: NCT03026244, clinicaltrials.gov:
Collapse
Affiliation(s)
| | - Olaf Perdijk
- Cell Biology and Immunology, Wageningen University, Wageningen, Netherlands
| | | | | | | | - Ben Meijer
- Cell Biology and Immunology, Wageningen University, Wageningen, Netherlands
| | - Sylvia Brugman
- Cell Biology and Immunology, Wageningen University, Wageningen, Netherlands
| | - Huub F J Savelkoul
- Cell Biology and Immunology, Wageningen University, Wageningen, Netherlands
| | | | - R J Joost van Neerven
- Cell Biology and Immunology, Wageningen University, Wageningen, Netherlands.,FrieslandCampina, Amersfoort, Netherlands
| |
Collapse
|
45
|
Iosifidis E, Papachristou S, Roilides E. Advances in the Treatment of Mycoses in Pediatric Patients. J Fungi (Basel) 2018; 4:E115. [PMID: 30314389 PMCID: PMC6308938 DOI: 10.3390/jof4040115] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/05/2018] [Accepted: 10/07/2018] [Indexed: 12/16/2022] Open
Abstract
The main indications for antifungal drug administration in pediatrics are reviewed as well as an update of the data of antifungal agents and antifungal policies performed. Specifically, antifungal therapy in three main areas is updated as follows: a) Prophylaxis of premature neonates against invasive candidiasis; b) management of candidemia and meningoencephalitis in neonates; and c) prophylaxis, empiric therapy, and targeted antifungal therapy in children with primary or secondary immunodeficiencies. Fluconazole remains the most frequent antifungal prophylactic agent given to high-risk neonates and children. However, the emergence of fluconazole resistance, particularly in non-albicans Candida species, should be considered during preventive or empiric therapy. In very-low birth-weight neonates, although fluconazole is used as antifungal prophylaxis in neonatal intensive care units (NICU's) with relatively high incidence of invasive candidiasis (IC), its role is under continuous debate. Amphotericin B, primarily in its liposomal formulation, remains the mainstay of therapy for treating neonatal and pediatric yeast and mold infections. Voriconazole is indicated for mold infections except for mucormycosis in children >2 years. Newer triazoles-such as posaconazole and isavuconazole-as well as echinocandins, are either licensed or under study for first-line or salvage therapy, whereas combination therapy is kept for refractory cases.
Collapse
Affiliation(s)
- Elias Iosifidis
- Infectious Diseases Unit, 3rd Department of Pediatrics, Faculty of Medicine, Aristotle University School of Health Sciences, Konstantinoupoleos 49, 54642, Thessaloniki, Greece.
| | - Savvas Papachristou
- Infectious Diseases Unit, 3rd Department of Pediatrics, Faculty of Medicine, Aristotle University School of Health Sciences, Konstantinoupoleos 49, 54642, Thessaloniki, Greece.
| | - Emmanuel Roilides
- Infectious Diseases Unit, 3rd Department of Pediatrics, Faculty of Medicine, Aristotle University School of Health Sciences, Konstantinoupoleos 49, 54642, Thessaloniki, Greece.
| |
Collapse
|
46
|
He Y, Cao L, Yu J. Prophylactic lactoferrin for preventing late-onset sepsis and necrotizing enterocolitis in preterm infants: A PRISMA-compliant systematic review and meta-analysis. Medicine (Baltimore) 2018; 97:e11976. [PMID: 30170397 PMCID: PMC6392939 DOI: 10.1097/md.0000000000011976] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Currently, prophylactic use of drugs to promote a healthy gut microbiota and immune system in preterm infants is hot debated, among which lactoferrin is a promising supplementation. However, the effect and safety of lactoferrin to prevent late-onset sepsis (LOS) and necrotizing enterocolitis (NEC) in preterm infants remains controversial. METHODS Databases including Medline, Ovid-Embase, The Cochrane Library, CBM, CNKI, and VIP database of Chinese Journal were searched to collect randomized controlled trials (RCTs) about lactoferrin for preventing LOS and NEC in preterm infants. Languages of included RCTs were restricted to English and Chinese. Meta-analysis was conducted by Rev Man 5.3 software. The Mantel-Haenszel method with random-effects model was used to calculate pooled relative risks (RRs) and 95% confidence intervals (CIs). RESULTS A total of 9 RCTs, involving 1834 patients, were included. Pooled analysis showed that prophylactic lactoferrin could significantly reduce the incidence all culture-proven LOS (41/629 [6.5%] vs 96/659 [15.3%]; RR 0.47; 95% CI 0.33-0.67; P < .01) and NEC (stage II or more) (9/448 [2.0%] vs 26/462 [5.6%]; RR 0.40; 95% CI 0.18-0.86; P < .01). Lactoferrin was also associated with a significantly decreased hospital-acquired infection (16/139 [11.5%] vs 35/140 [25%]; RR 0.47; 95% CI 0.27-0.80; P < .01); and infection-related mortality (4/474 [0.8%] vs 25/505 [4.9%]; RR 0.24; 95% CI 0.04-1.32; P < .01, I = 53%). Lactoferrin could shorten time to reach full enteral feeding (weighted mean difference [WMD] = -2.11, 95% CI -3.12 to -1.10; P < .01) and showed a decreasing trend of duration of hospitalization (WMD = -1.69, 95% CI -6.87 to 3.50; P < .01; I = 95%). Lactoferrin did not have a significant effect on all-cause mortality (22/625 [3.5%] vs 35/647 [5.4%]; RR 0.70; 95% CI 0.38-1.30; P = .16; I = 13%). None of the included trials reported any confirmed adverse effects caused by the supplemented lactoferrin or probiotics. CONCLUSION Current evidence indicates that lactoferrin could significantly reduce the incidence of NEC and LOS, and decrease the risk of hospital-acquired infection and infection-related mortality in premature infants without obvious adverse effects.
Collapse
Affiliation(s)
- Yi He
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Yuzhong
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders
| | - Luying Cao
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Yuzhong
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders
| | - Jialin Yu
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Yuzhong
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
47
|
Garcia-Rodenas CL, De Castro CA, Jenni R, Thakkar SK, Beauport L, Tolsa JF, Fischer-Fumeaux CJ, Affolter M. Temporal changes of major protein concentrations in preterm and term human milk. A prospective cohort study. Clin Nutr 2018; 38:1844-1852. [PMID: 30093147 DOI: 10.1016/j.clnu.2018.07.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/24/2018] [Accepted: 07/11/2018] [Indexed: 11/29/2022]
Abstract
BACKGROUND Proteins are major contributors to the beneficial effects of human milk (HM) on preterm infant health and development. Alpha-lactalbumin, lactoferrin, serum albumin and caseins represent approximately 85% of the total HM protein. The temporal changes of these proteins in preterm (PT) HM and its comparison with term (T) HM is poorly characterized. AIMS To quantify and compare the temporal changes of the major proteins in PT HM and T HM. METHODS HM was collected for 4 months postpartum at 12 time points for PT HM (gestational age 28 0/7-32 6/7 weeks; 280 samples) and for 2 months postpartum at 8 time points for T HM (gestational age 37 0/7-41 6/7 weeks; 220 samples). Proteins were measured with a micro-fluidic LabChip system. RESULTS Casein, alpha-lactalbumin and lactoferrin decreased with advancing stages of lactation in PT and T HM, whereas serum albumin remained stable. Only marginal differences between PT and T HM were observed for alpha-lactalbumin during postpartum weeks 3-5 and for serum albumin at the first week. However, a comparison of HM provided to preterm and term infants at the same postmenstrual ages revealed that alpha-lactalbumin contents were significantly lower in PT HM than in T HM during the 39-48 postmenstrual weeks. CONCLUSIONS This study provides comprehensive information of the longitudinal changes of major proteins in PT and T HM, and suggests limited availability of alpha-lactalbumin, a nutritionally important protein, in breastfed PT infants after reaching the term corrected age. This information may be important to optimize HM protein fortification, although its biological relevance needs to be confirmed by intervention studies. CLINICAL TRIAL REGISTRY ClinicalTrials.gov (NCT02052245), https://clinicaltrials.gov/ct2/show/NCT02052245.
Collapse
Affiliation(s)
| | | | - Rosemarie Jenni
- Nestlé Institute of Food Safety & Analytical Science, Nestlé Research, Lausanne, Switzerland
| | - Sagar K Thakkar
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | - Lydie Beauport
- Clinic of Neonatology, Department Woman Mother Child, University Hospital of Lausanne, Switzerland
| | - Jean-François Tolsa
- Clinic of Neonatology, Department Woman Mother Child, University Hospital of Lausanne, Switzerland
| | - Céline J Fischer-Fumeaux
- Clinic of Neonatology, Department Woman Mother Child, University Hospital of Lausanne, Switzerland
| | - Michael Affolter
- Nestlé Institute of Food Safety & Analytical Science, Nestlé Research, Lausanne, Switzerland
| |
Collapse
|
48
|
Probiotics for Preterm Infants: A Strain-Specific Systematic Review and Network Meta-analysis. J Pediatr Gastroenterol Nutr 2018; 67:103-122. [PMID: 29384838 DOI: 10.1097/mpg.0000000000001897] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVES Several randomized controlled trials (RCTs) on the use of probiotics to reduce morbidity and mortality in preterm infants have provided inconsistent results. Although meta-analyses that group all of the used strains together are suggesting efficacy, it is not possible to determine the most effective strain that is more relevant to the clinician. We therefore used a network meta-analysis (NMA) approach to identify strains with greatest efficacy. METHODS A PubMed search identified placebo-controlled or head-to-head RCTs investigating probiotics in preterm infants. From trials that recorded mortality, necrotizing enterocolitis, late-onset sepsis, or time until full enteral feeding as outcomes, data were extracted and Bayesian hierarchical random-effects models were run to construct a NMA. RESULTS Fifty-one RCTs involving 11,231 preterm infants were included. Most strains or combinations of strains were only studied in one or a few RCTs. Only 3 of 25 studied probiotic treatment combinations showed significant reduction in mortality rates. Seven treatments reduced necrotizing enterocolitis incidence, 2 reduced late-onset sepsis, and 3 reduced time until full enteral feeding. There was no clear overlap of strains, which were effective on multiple outcome domains. CONCLUSIONS This NMA showed efficacy in reducing mortality and morbidity only in a minority of the studied strains or combinations. This may be due to an inadequate number, or size, of RCTs, or due to a true lack of effect for certain species. Further large and adequately powered RCTs using strains with the greatest apparent efficacy will be needed to more precisely define optimal treatment strategies.
Collapse
|
49
|
Bovine Lactoferrin Modulates Dendritic Cell Differentiation and Function. Nutrients 2018; 10:nu10070848. [PMID: 29966271 PMCID: PMC6073808 DOI: 10.3390/nu10070848] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/19/2018] [Accepted: 06/26/2018] [Indexed: 11/16/2022] Open
Abstract
Lactoferrin is an abundant glycoprotein in bovine milk that has immunomodulatory effects on human cells. Bovine lactoferrin (LF) binds lipopolysaccharides (LPS) with high affinity and is postulated to act via TLR4-dependent and -independent mechanisms. It has been shown that LF modulates differentiation of human monocytes into tolerogenic dendritic cells. However, in a previous study, we showed that LPS also mediates differentiation into tolerogenic dendritic cells (DC). Since LF binds LPS with high affinity, it remains to be investigated whether LF or LPS is mediating these effects. We, therefore, further investigated the LPS-independent effect of LF on differentiation of human monocytes into dendritic cells (DC). Human monocytes were isolated by magnetic cell sorting from freshly isolated PBMCs and cultured for six days in the presence of IL-4 and GM-CSF with or without LF or proteinase K treated LF to generate DC. These immature DC were stimulated for 48 h with LPS or Poly I:C + R848. Cell surface marker expression and cytokine production were measured by flow cytometry. DC differentiated in the presence of LF produced higher IL-6 and IL-8 levels during differentiation and showed a lower expression of CD1a and HLA-DR. These LFDCs showed to be hyporesponsive towards TLR ligands as shown by their semi-mature phenotype and reduced cytokine production. The effect of LF was abrogated by proteinase K treatment, showing that the functional effects of LF were not mediated by LPS contamination. Thus, LF alters DC differentiation and dampens responsiveness towards TLR ligands. This study indicates that LF can play a role in immune homeostasis in the human GI tract.
Collapse
|
50
|
Manzoni P, García Sánchez R, Meyer M, Stolfi I, Pugni L, Messner H, Cattani S, Betta PM, Memo L, Decembrino L, Bollani L, Rinaldi M, Fioretti M, Quercia M, Maule M, Tavella E, Mussa A, Tzialla C, Laforgia N, Mosca F, Magaldi R, Mostert M, Farina D. Exposure to Gastric Acid Inhibitors Increases the Risk of Infection in Preterm Very Low Birth Weight Infants but Concomitant Administration of Lactoferrin Counteracts This Effect. J Pediatr 2018; 193:62-67.e1. [PMID: 29198543 DOI: 10.1016/j.jpeds.2017.09.080] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 09/26/2017] [Accepted: 09/28/2017] [Indexed: 01/08/2023]
Abstract
OBJECTIVE To investigate whether exposure to inhibitors of gastric acidity, such as H2 blockers or proton pump inhibitors, can independently increase the risk of infections in very low birth weight (VLBW) preterm infants in the neonatal intensive care unit. STUDY DESIGN This is a secondary analysis of prospectively collected data from a multicenter, randomized controlled trial of bovine lactoferrin (BLF) supplementation (with or without the probiotic Lactobacillus rhamnosus GG) vs placebo in prevention of late-onset sepsis (LOS) and necrotizing enterocolitis (NEC) in preterm infants. Inhibitors of gastric acidity were used at the recommended dosages/schedules based on the clinical judgment of attending physicians. The distribution of days of inhibitors of gastric acidity exposure between infants with and without LOS/NEC was assessed. The mutually adjusted effects of birth weight, gestational age, duration of inhibitors of gastric acidity treatment, and exposure to BLF were controlled through multivariable logistic regression. Interaction between inhibitors of gastric acidity and BLF was tested; the effects of any day of inhibitors of gastric acidity exposure were then computed for BLF-treated vs -untreated infants. RESULTS Two hundred thirty-five of 743 infants underwent treatment with inhibitors of gastric acidity, and 86 LOS episodes occurred. After multivariate analysis, exposure to inhibitors of gastric acidity remained significantly and independently associated with LOS (OR, 1.03; 95% CI, 1.008-1.067; P = .01); each day of inhibitors of gastric acidity exposure conferred an additional 3.7% odds of developing LOS. Risk was significant for Gram-negative (P < .001) and fungal (P = .001) pathogens, but not for Gram-positive pathogens (P = .97). On the test for interaction, 1 additional day of exposure to inhibitors of gastric acidity conferred an additional 7.7% risk for LOS (P = .003) in BLF-untreated infants, compared with 1.2% (P = .58) in BLF-treated infants. CONCLUSION Exposure to inhibitors of gastric acidity is significantly associated with the occurrence of LOS in preterm VLBW infants. Concomitant administration of BLF counteracts this selective disadvantage. TRIAL REGISTRATION isrctn.org: ISRCTN53107700.
Collapse
Affiliation(s)
- Paolo Manzoni
- Neonatology and NICU, S Anna Hospital, AOU Città della Salute e della Scienza, Torino, Italy.
| | - Ruben García Sánchez
- Neonatology and NICU, Complejo Asistencial Universitario de Salamanca, Salamanca, Spain
| | - Michael Meyer
- Neonatology and NICU, Middlemore Hospital, Auckland, New Zealand
| | - Ilaria Stolfi
- Neonatology, Azienda Ospedaliera Universitaria Policlinico Umberto I, Rome, Italy
| | - Lorenza Pugni
- NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, Italy
| | - Hubert Messner
- Neonatology and NICU, Ospedale Regionale, Bolzano/Bozen, Italy
| | - Silvia Cattani
- NICU, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Luigi Memo
- UOC di Pediatria e Patologia Neonatale, Ospedale San Martino, Belluno, Italy
| | - Lidia Decembrino
- Patologia Neonatale, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Lina Bollani
- Patologia Neonatale, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Maria Fioretti
- Neonatology and NICU, S Anna Hospital, AOU Città della Salute e della Scienza, Torino, Italy
| | - Michele Quercia
- Cancer Epidemiology Unit, University of Turin, Department of Medical Sciences, Torino, Italy
| | - Milena Maule
- Cancer Epidemiology Unit, University of Turin, Torino, Italy
| | - Elena Tavella
- Neonatology and NICU, S Anna Hospital, AOU Città della Salute e della Scienza, Torino, Italy
| | - Alessandro Mussa
- Neonatology and NICU, S Anna Hospital, AOU Città della Salute e della Scienza, Torino, Italy
| | - Chryssoula Tzialla
- Patologia Neonatale, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Nicola Laforgia
- Cancer Epidemiology Unit, University of Turin, Department of Medical Sciences, Torino, Italy
| | - Fabio Mosca
- NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, Italy
| | | | - Michael Mostert
- Department of Pediatrics, University of Turin, Torino, Italy
| | - Daniele Farina
- Neonatology and NICU, S Anna Hospital, AOU Città della Salute e della Scienza, Torino, Italy
| | | |
Collapse
|