Generic placeholder image

Recent Patents on Biotechnology

Editor-in-Chief

ISSN (Print): 1872-2083
ISSN (Online): 2212-4012

Review Article

Basic Guidelines for Bacteriophage Isolation and Characterization

Author(s): Safia Samir*

Volume 17, Issue 4, 2023

Published on: 28 October, 2022

Page: [312 - 331] Pages: 20

DOI: 10.2174/1872208317666221017094715

Price: $65

Abstract

The world is on the cusp of a post-antibiotic period. A century ago, before the advent of antibiotics, bacteriophage therapy was the treatment of choice for bacterial infections. Although bacteriophages have yet to be approved as a treatment in Western medicine, researchers and clinicians have begun to anticipate phage therapy. Bacteriophages are viruses that depend on bacterial cell metabolism to multiply. They offer a promising alternative to the use of antibiotics and an excellent antibacterial option for combating multidrug resistance in bacteria. However, not every phage is suitable for phage therapy. In particular, prophages should not be used because they can lysogenize host cells instead of lysing them. To offer adequate therapeutic options for patients suffering from various infectious diseases, a wide selection of different phages is needed. While there is no evidence of direct toxicity induced by phage particles, it is crucial to study mammalian cell–phage interactions. This requires phage preparations to be free of bacterial cells, toxins and other compounds to avoid skewing host responses. Negative staining of purified viruses and electron microscopy remain the gold standard in the identification of bacteriophages. Interestingly, genomics has greatly changed our understanding of phage biology. Bacteriophage genome sequencing is essential to obtain a complete understanding of the bacteriophages' biology and to obtain confirmation of their lifestyle. Full genetic sequencing of bacteriophage will enable a better understanding of the phage-encoded proteins and biomolecules (especially phage lytic enzymes) involved in the process of bacterial cell lysis and death. Mass spectrometry can be used for the identification of phage structural proteins. The use of lytic phages as biocontrol agents requires the most appropriate and standard methods to ensure application safety. This review pursues recent research and methods in molecular biology for the isolation and characterization of phages to facilitate follow-up and implementation of work for other researchers. Patents related to this topic have been mentioned in the text.

Keywords: Bacterial infection, antibiotics, antibiotic alternatives, bacteriophage therapy, bacteriophage isolation, electron microscopy, genome sequencing, mass spectrometry.

Graphical Abstract
[1]
El-Far A, Samir S, El-Gebaly E, et al. Assessment of eugenol inhibitory effect on biofilm formation and biofilm gene expression in methicillin resistant Staphylococcus aureus clinical isolates in Egypt. Infect Genet Evol 2021; 89: 104722.
[http://dx.doi.org/10.1016/j.meegid.2021.104722] [PMID: 33444856]
[2]
Rios AC, Moutinho CG, Pinto FC, et al. Alternatives to overcoming bacterial resistances: State-of-the-art. Microbiol Res 2016; 191: 51-80.
[http://dx.doi.org/10.1016/j.micres.2016.04.008] [PMID: 27524653]
[3]
Murugan N, Malathi J, Therese KL, Madhavan HN. Application of six multiplex PCR’s among 200 clinical isolates of Pseudomonas aeruginosa for the detection of 20 drug resistance encoding genes. Kaohsiung J Med Sci 2018; 34(2): 79-88.
[http://dx.doi.org/10.1016/j.kjms.2017.09.010] [PMID: 29413231]
[4]
El-Halfawy OM, Czarny TL, Flannagan RS, et al. Discovery of an antivirulence compound that reverses β-lactam resistance in MRSA. Nat Chem Biol 2020; 16(2): 143-9.
[http://dx.doi.org/10.1038/s41589-019-0401-8] [PMID: 31768032]
[5]
Nasir N, Rehman F, Omair SF. Risk factors for bacterial infections in patients with moderate to severe COVID‐19: A case‐control study. J Med Virol 2021; 93(7): 4564-9.
[http://dx.doi.org/10.1002/jmv.27000] [PMID: 33822390]
[6]
Tondi D. Novel targets and mechanisms in antimicrobial drug discovery. Antibiotics 2021; 10(2): 141.
[7]
Cheng G, Hao H, Xie S, Wang X, Dai M, Huang L. Antibiotic alternatives: the substitution of antibiotics in animal husbandry? Front Microbiol 2014; 5: 217.
[http://dx.doi.org/10.3389/fmicb.2014.00217]
[8]
Rima M, Rima M, Fajloun Z, Sabatier JM, Bechinger B, Naas T. Antimicrobial peptides: A potent alternative to antibiotics. Antibiotics 2021; 10(9): 1095.
[http://dx.doi.org/10.3390/antibiotics10091095] [PMID: 34572678]
[9]
Rosini R, Nicchi S, Pizza M, Rappuoli R. Vaccines against antimicrobial resistance. Front Immunol 2020; 11(6): 1048.
[http://dx.doi.org/10.3389/fimmu.2020.01048] [PMID: 32582169]
[10]
Rao MJ, Ahmed S, Rekha RS, et al. Immunomodulatory agents combat multidrug-resistant tuberculosis by improving antimicrobial immunity. J Infect Dis 2021; 224(2): 332-44.
[http://dx.doi.org/10.1093/infdis/jiab100] [PMID: 33606878]
[11]
Lener MS. Cardiac extraction from suckling rat HHS Public Access. Physiol Behav 2016; 176(1): 139-48.
[http://dx.doi.org/10.1016/j.clinthera.2020.06.011.The]
[12]
Imran M, Jha SK, Hasan N, et al. Overcoming multidrug resistance of antibiotics via nanodelivery systems. Pharmaceutics 2022; 14(3): 586.
[http://dx.doi.org/10.3390/pharmaceutics14030586] [PMID: 35335962]
[13]
Song M, Liu Y, Li T, et al. Plant natural flavonoids against multidrug resistant pathogens. Adv Sci 2021; 8(15): 2100749.
[http://dx.doi.org/10.1002/advs.202100749] [PMID: 34041861]
[14]
El-Shibiny A, El-Sahhar S. Bacteriophages: The possible solution to treat infections caused by pathogenic bacteria. Can J Microbiol 2017; 63(11): 865-79.
[http://dx.doi.org/10.1139/cjm-2017-0030] [PMID: 28863269]
[15]
Carmody CM, Goddard JM, Nugen SR. Bacteriophage capsid modification by genetic and chemical methods. Bioconjug Chem 2021; 32(3): 466-81.
[http://dx.doi.org/10.1021/acs.bioconjchem.1c00018] [PMID: 33661607]
[16]
Orlova EV. Bacteriophages and their structural organisation. In: Kurtboke Ipek, Ed. Bacteriophages. Australia 2012; p. 270.
[http://dx.doi.org/10.5772/34642]
[17]
Samir S. Bacteriophages as therapeutic agents: Alternatives to antibiotics. Recent Pat Biotechnol 2021; 15(1): 25-33.
[http://dx.doi.org/10.2174/1872208315666210121094311] [PMID: 33475081]
[18]
De Vos DA, Pirnay JP. Phage therapy: could viruses help resolve the worldwide antibiotic crisis? AMR Control 2015; 2015: 110.
[19]
Brives C, Pourraz J. Phage therapy as a potential solution in the fight against AMR: Obstacles and possible futures. Palgrave Commun 2020; 6(1): 100.
[http://dx.doi.org/10.1057/s41599-020-0478-4]
[20]
Andrzej G, Weberd B. Poland — Its history, milestones and international recognition. Viruses 2022; 14(6): 1170.
[21]
Trifonova A, Todorova I, Savov E. Bacteriophage therapy. Probl Infect Parasit Dis 2016; 34(1): 28-31.
[http://dx.doi.org/10.1071/MA13009]
[22]
Pirnay JP, Verbeken G, Ceyssens PJ, et al. The magistral phage. Viruses 2018; 10(2): 64.
[http://dx.doi.org/10.3390/v10020064] [PMID: 29415431]
[23]
Maddocks S, Fabijan AP, Ho J, et al. Bacteriophage therapy of ventilator-associated pneumonia and empyema caused by Pseudomonas aeruginosa. Am J Respir Crit Care Med 2019; 200(9): 1179-81.
[http://dx.doi.org/10.1164/rccm.201904-0839LE] [PMID: 31437402]
[24]
Aslam S, Lampley E, Wooten D, et al. Lessons learned from the first 10 consecutive cases of intravenous bacteriophage therapy to treat multidrug-resistant bacterial infections at a single center in the United States. Open Forum Infect Dis 2020; 7(9): ofaa389.
[http://dx.doi.org/10.1093/ofid/ofaa389] [PMID: 33005701]
[25]
Khalid A, Lin RCY, Iredell JR. A phage therapy guide for clinicians and basic scientists: Background and highlighting applications for developing countries. Front Microbiol 2021; 11(2): 599906.
[http://dx.doi.org/10.3389/fmicb.2020.599906] [PMID: 33643225]
[26]
Cano EJ, Caflisch KM, Bollyky PL, et al. Phage therapy for limb-threatening prosthetic knee klebsiella pneumoniae infection: Case report and in vitro characterization of anti-biofilm activity. Clin Infect Dis 2021; 73(1): e144-51.
[http://dx.doi.org/10.1093/cid/ciaa705] [PMID: 32699879]
[27]
Koh L. (12) United States Patent Figure 1 Thermolysin 2012; 2(2012): 2790-1.
[28]
World Health Organization. WHO | Health data and statistics. Available from: http://www.who.int/healthinfo/statistics/en/ [Accessed October 3, 2020].
[29]
Nagel TE, Chan BK, De Vos D, et al. The developing world urgently needs phages to combat pathogenic bacteria. Front Microbiol 2016; 7(6): 882.
[http://dx.doi.org/10.3389/fmicb.2016.00882] [PMID: 27375602]
[30]
Kassa T. Bacteriophages against pathogenic bacteria and possibilities for future application in Africa. Infect Drug Resist 2021; 14: 17-31.
[http://dx.doi.org/10.2147/IDR.S284331] [PMID: 33442273]
[31]
Międzybrodzki R, Harper DR, Abedon ST. Current updates from the long-standing phage research centers in Georgia, Poland, and Russia. Springer Int Publ 2018.
[32]
Schooley RT, Biswas B, Gill JJ, et al. Development and use of personalized bacteriophage-based therapeutic cocktails to treat a patient with a disseminated resistant Acinetobacter baumannii infection. Antimicrob Agents Chemother 2017; 61(10): e00954-17.
[http://dx.doi.org/10.1128/AAC.00954-17] [PMID: 28807909]
[33]
Anand T, Virmani N, Bera BC, et al. Phage Display technique as a tool for diagnosis and antibody selection for coronaviruses. Curr Microbiol 2021; 78(4): 1124-34.
[http://dx.doi.org/10.1007/s00284-021-02398-9] [PMID: 33687511]
[34]
Wang HL, Gao S, Lv J, Lin Y, Zhou L. Phage display technology and its applications in cancer immunotherapy. Anticancer Agents Med Chem 2019; 19: 2229-35.
[http://dx.doi.org/10.2174/1871520618666181029140814]
[35]
Lemire S, Yehl KM, Lu TK. Phage-based applications in synthetic biology. Annu Rev Virol 2018; 5(1): 453-76.
[http://dx.doi.org/10.1146/annurev-virology-092917-043544] [PMID: 30001182]
[36]
Jones HJ, Shield CG, Swift BMC. The application of bacteriophage diagnostics for bacterial pathogens in the agricultural supply chain: From farm-to-fork. Phage 2020; 1(4): 176-88.
[http://dx.doi.org/10.1089/phage.2020.0042]
[37]
Chevallereau A, Pons BJ, van Houte S, Westra ER. Interactions between bacterial and phage communities in natural environments. Nat Rev Microbiol 2021; 20: 49-62.
[http://dx.doi.org/10.1038/s41579-021-00602-y] [PMID: 34373631]
[38]
Kavagutti VS, Andrei AŞ, Mehrshad M, Salcher MM, Ghai R. Phage-centric ecological interactions in aquatic ecosys-tems revealed through ultra- deep metagenomics. Microbiome 2019; 7: 135.
[http://dx.doi.org/10.1186/s40168-019-0752-0]
[39]
Walker JM. P53 protocols. In: Sumitra D, Swati DP, Eds; Molecular Biology. 2nd eds.; Humana Press 2009.
[40]
Martha RJC, Andrew MK, Rob L, Eds. In: Bacteriophages: Methods and protocols: Advanced techniques. Humana Press 2018; vol 3: p. 1681.
[http://dx.doi.org/10.1007/978-1-4939-7343-9]
[41]
Rasool MH, Yousaf R, Siddique AB, Saqalein M, Khurshid M. Isolation, characterization, and antibacterial activity of bacteriophages against methicillin-resistant Staphylococcus aureus in Pakistan. Jundishapur J Microbiol 2016; 9(10): e36135.
[http://dx.doi.org/10.5812/jjm.36135] [PMID: 27942361]
[42]
James B, Alain S, Eds. Methods for Isolation, Purifi cation, and Propagation of Bacteriophages. In: Campylobacter jejuni. Humana, New York 2017; p. 274.
[http://dx.doi.org/10.1007/978-1-4939-6536-6]
[43]
Gamian A, Gorski A, Pan D. Purified bacteriophage, its preparation and application. Patent US 9255251B2, 2016. Available from: 1499091266716832064-09255251 (storage.googleapis.com)
[44]
Hoai TD, Yoshida T. Induction and characterization of a lysogenic bacteriophage of Lactococcus garvieae isolated from marine fish species. J Fish Dis 2016; 39(7): 799-808.
[http://dx.doi.org/10.1111/jfd.12410] [PMID: 26471724]
[45]
Induction of phage using mitomycin c to check for phage presence. pp.1-2. Availabale from: https://static.igem.org/mediawiki/2018/9/9a/T--CSU_Fort_Collins--MitomycinC_Phage_Induction_Protocol.pdf
[46]
Wright EE, Elliman JR, Owens L. Induction and characterization of lysogenic bacteriophages from Streptococcus iniae. J Appl Microbiol 2013; 114(6): 1616-24.
[http://dx.doi.org/10.1111/jam.12192] [PMID: 23490045]
[47]
Hyman P, Abedon ST. Bacteriophage host range and bacterial resistance. Adv Appl Microbiol 2010; 70: 217-48.
[http://dx.doi.org/10.1016/S0065-2164(10)70007-1] [PMID: 20359459]
[48]
de Melo ACC, da Mata Gomes A, Melo FL, et al. Characterization of a bacteriophage with broad host range against strains of Pseudomonas aeruginosa isolated from domestic animals. BMC Microbiol 2019; 19(1): 134.
[http://dx.doi.org/10.1186/s12866-019-1481-z] [PMID: 31208333]
[49]
Bretaudeau L, Tremblais K, Aubrit F, Meichenin M, Arnaud I. Good Manufacturing Practice (GMP) compliance for phage therapy medicinal products. Front Microbiol 2020; 11(6): 1161.
[http://dx.doi.org/10.3389/fmicb.2020.01161] [PMID: 32582101]
[50]
Duyvejonck H, et al. Evaluation of the stability of bacteriophages in different solutions suitable for the production of magistral preparations in belgium. Viruses 2021; 13(5): 865.
[http://dx.doi.org/10.3390/v13050865]
[51]
Ács N, Gambino M, Brøndsted L. Bacteriophage Enumeration and Detection Methods. Front Microbiol 2020; 11(10): 594868.
[http://dx.doi.org/10.3389/fmicb.2020.594868] [PMID: 33193274]
[52]
Samir S, El-Far A, Okasha H, Mahdy R, Samir F, Nasr S. Isolation and characterization of lytic bacteriophages from sewage at an egyptian tertiary care hospital against methicillin-resistant Staphylococcus aureus clinical isolates. Saudi J Biol Sci 2022; 29(5): 3097-106.
[http://dx.doi.org/10.1016/j.sjbs.2022.03.019] [PMID: 35360502]
[53]
Anderson B, Rashid MH, Carter C, et al. Enumeration of bacteriophage particles. Bacteriophage 2011; 1(2): 86-93.
[http://dx.doi.org/10.4161/bact.1.2.15456] [PMID: 22334864]
[54]
Thermo Fisher Scientific. What is Transmission Electron Microscopy? Available from: https://www.news-medical.net/life-sciences/What-is-Transmission-Electron-Microscopy.aspx
[55]
Ackermann HW. Bacteriophage electron microscopy. Adv Virus Res 2012; 82: 1-32.
[http://dx.doi.org/10.1016/B978-0-12-394621-8.00017-0] [PMID: 22420849]
[56]
Beniac D, Siemens C, Wright C, Booth T. A filtration based technique for simultaneous SEM and TEM sample preparation for the rapid detection of pathogens. Viruses 2014; 6(9): 3458-71.
[http://dx.doi.org/10.3390/v6093458] [PMID: 25243370]
[57]
Picot J, Guerin CL, Le Van Kim C, Boulanger CM. Flow cytometry: Retrospective, fundamentals and recent instrumentation. Cytotechnology 2012; 64(2): 109-30.
[http://dx.doi.org/10.1007/s10616-011-9415-0] [PMID: 22271369]
[58]
Dlusskaya EA, Atrazhev AM, Ashbolt NJ. Colloid chemistry pitfall for flow cytometric enumeration of viruses in water. Water Res X 2019; 2: 100025.
[http://dx.doi.org/10.1016/j.wroa.2019.100025] [PMID: 31194069]
[59]
Elina BR, Susana GB. Random amplified polymorphic DNA PCR in the teaching of molecular epidemiology. IUBMB J 2016; 44(4): 391-6.
[http://dx.doi.org/10.1002/bmb.20957]
[60]
Kim S, Kim S. Bacterial pathogen detection by conventional culture-based and recent alternative (polymerase chain reaction, isothermal amplification, enzyme linked immunosorbent assay, bacteriophage amplification, and gold nanoparticle aggregation) methods in food samples: A review. J Food Safety 2021; 41(1): e12870.
[http://dx.doi.org/10.1111/jfs.12870]
[61]
Luo J, Liu M, Wang P, et al. Evaluation of a direct phage DNA detection-based taqman qPCR methodology for quantification of phage and its application in rapid ultrasensitive identification of Acinetobacter baumannii. BMC Infect Dis 2022; 22(1): 523.
[http://dx.doi.org/10.1186/s12879-022-07493-1] [PMID: 35672689]
[62]
Abril AG, Mónica C, Karola B, et al. proteomic characterization of bacteriophage peptides from the mastitis producer staphylococcus aureus by LC-ESI-MS/MS and the bacteriophage phylogenomic analysis. Foods 2021; 10(4): 799.
[http://dx.doi.org/10.3390/foods10040799]
[63]
Abril AG, Carrera M, Böhme K, et al. Characterization of bacteriophage peptides of pathogenic Streptococcus by LC-ESI-MS/MS: Bacteriophage phylogenomics and their relationship to their host. Front Microbiol 2020; 11(6): 1241.
[http://dx.doi.org/10.3389/fmicb.2020.01241] [PMID: 32582130]
[64]
Peng X, Leal J, Mohanty R, Soto M, Ghosh D. Quantitative PCR of T7 bacteriophage from biopanning. J Vis Exp 2018; 2018(139): 1-8.
[http://dx.doi.org/10.3791/58165] [PMID: 30320762]
[65]
Kłopot A, Zakrzewska A, Lecion D, et al. Real-time qPCR as a method for detection of antibody-neutralized phage particles. Front Microbiol 2017; 8(11): 2170.
[http://dx.doi.org/10.3389/fmicb.2017.02170] [PMID: 29163448]
[66]
Farkas K, Varsani A, Pang L. Adsorption of rotavirus, ms2 bacteriophage and surface-modified silica nanoparticles to hydrophobic matter. Food Environ Virol 2015; 7(3): 261-8.
[http://dx.doi.org/10.1007/s12560-014-9171-3] [PMID: 25342436]
[67]
Taylor SC, Laperriere G, Germain H. Droplet digital PCR versus qPCR for gene expression analysis with low abundant targets: from variable nonsense to publication quality data. Sci Rep 2017; 7(1): 2409.
[http://dx.doi.org/10.1038/s41598-017-02217-x] [PMID: 28546538]
[68]
Morella NM, Gomez AL, Wang G, Leung MS, Koskella B. The impact of bacteriophages on phyllosphere bacterial abundance and composition. Mol Ecol 2018; 27(8): 2025-38.
[http://dx.doi.org/10.1111/mec.14542] [PMID: 29457297]
[69]
Clokie MRJ, Kropinski AM, Eds. Bacteriophages, Methods and Protocols: Molecular and Applied Aspects. Humana Press 2009; p. 502.
[http://dx.doi.org/10.1007/978-1-60327-565-1]
[70]
Abril AG, Carrera M, Notario V, Sánchez-Pérez Á, Villa TG. The use of bacteriophages in biotechnology and recent insights into proteomics. Antibiotics 2022; 11(5): 653.
[http://dx.doi.org/10.3390/antibiotics11050653] [PMID: 35625297]
[71]
Al-Wrafy F, Brzozowska E, Górska S, Drab M, Strus M, Gamian A. Identification and characterization of phage protein and its activity against two strains of multidrug-resistant Pseudomonas aeruginosa. Sci Rep 2019; 9(1): 13487.
[http://dx.doi.org/10.1038/s41598-019-50030-5] [PMID: 31530875]
[72]
Braun R, Schönberger N, Vinke S, Lederer F, Jörn K, Katrin P. Application of Next Generation Sequencing (NGS) in phage displayed peptide selection to support the identification of arsenic-binding motifs. Viruses 2020; 12(12): 1360.
[http://dx.doi.org/10.3390/v12121360]
[73]
Marco TP, Laís S, Luiz AA, Maryoris E. Bacteriophage genome sequencing: A new alternative to understand biochemical interactions between prokaryotic cells and phages. J Microb Biochem Technol 2017; 9: 4.
[http://dx.doi.org/10.4172/1948-5948.1000362]
[74]
Ayalew N, Mebrat E. Review on bacteriophages and its antimicrobial uses. Amer-Eurournal Sci Res 2016; 11: 199-208.
[75]
Barreto K, Maruthachalam BV, Hill W, et al. Next-generation sequencing-guided identification and reconstruction of antibody CDR combinations from phage selection outputs. Nucleic Acids Res 2019; 47(9): e50.
[http://dx.doi.org/10.1093/nar/gkz131] [PMID: 30854567]
[76]
Sutton TDS, Hill C. Gut bacteriophage: Current understanding and challenges. Front Endocrinol 2019; 10(11): 784.
[http://dx.doi.org/10.3389/fendo.2019.00784] [PMID: 31849833]
[77]
Shkoporov AN, Ryan FJ, Draper LA, et al. Reproducible protocols for metagenomic analysis of human faecal phageomes. Microbiome 2018; 6(1): 68.
[http://dx.doi.org/10.1186/s40168-018-0446-z] [PMID: 29631623]
[78]
Callanan J, Stockdale SR, Shkoporov A, Draper LA, Ross RP, Hill C. Biases in viral metagenomics-based detection, cataloguing and quantification of bacteriophage genomes in human faeces, a review. Microorganisms 2021; 9(3): 524.
[http://dx.doi.org/10.3390/microorganisms9030524] [PMID: 33806607]
[79]
Hinkley TC, Singh S, Garing S, et al. A phage-based assay for the rapid, quantitative, and single CFU visualization of E. coli (ECOR #13) in drinking water. Sci Rep 2018; 8(1): 14630.
[http://dx.doi.org/10.1038/s41598-018-33097-4] [PMID: 30279488]
[80]
Brown M, Hahn W, Bailey B, et al. Development and evaluation of a sensitive bacteriophage-based MRSA diagnostic screen. Viruses 2020; 12(6): 631.
[http://dx.doi.org/10.3390/v12060631] [PMID: 32545159]
[81]
Alonzo LF, Jain P, Hinkley T, et al. Rapid, sensitive, and low-cost detection of Escherichia coli bacteria in contaminated water samples using a phage-based assay. Sci Rep 2022; 12(1): 7741.
[http://dx.doi.org/10.1038/s41598-022-11468-2] [PMID: 35562180]
[82]
Speck P, Smithyman A. Safety and efficacy of phage therapy via the intravenous route. FEMS Microbiol Lett 2016; 363(3): fnv242.
[http://dx.doi.org/10.1093/femsle/fnv242] [PMID: 26691737]
[83]
Hietala V, Horsma-Heikkinen J, Carron A, Skurnik M, Kiljunen S. The removal of endo- and enterotoxins from bacteriophage preparations. Front Microbiol 2019; 10(7): 1674.
[http://dx.doi.org/10.3389/fmicb.2019.01674] [PMID: 31396188]
[84]
Carroll-Portillo A, Coffman CN, Varga MG, Alcock J, Singh SB, Lin HC. Standard bacteriophage purification procedures cause loss in numbers and activity. Viruses 2021; 13(2): 328.
[http://dx.doi.org/10.3390/v13020328] [PMID: 33672780]
[85]
European Medicines Agency. ICH guideline Q3C (R6) on impurities: guideline for residual solvents. 2019. Available from: https://www.ema.europa.eu/en/documents/scientific-guideline/international-conferenceharmonisation-technical-requirements-registrationpharmaceuticals-human-use_en-33.pdf
[86]
Food and Drug Administration. FDA alerts health care professionals of significant safety risks associated with cesium chloride. 2018. Available from: https://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/ PharmacyCompounding/ucm614211
[87]
Szermer-Olearnik B, Boratyński J. Removal of endotoxins from bacteriophage preparations by extraction with organic solvents. PLoS One 2015; 10(3): e0122672.
[http://dx.doi.org/10.1371/journal.pone.0122672] [PMID: 25811193]
[88]
Bonilla B, Rojas N, Netto Flores Cruz MI, Hung G, Rohwer SH. J. J. Phage on tap-a quick and efficient protocol for the preparation of bacteriophage laboratory stocks. PeerJ 2016; 4: e2261.
[http://dx.doi.org/10.7717/peerj.2261]
[89]
Pires DP, Costa AR, Pinto G, Meneses L, Azeredo J. Current challenges and future opportunities of phage therapy. FEMS Microbiol Rev 2020; 44(6): 684-700.
[http://dx.doi.org/10.1093/femsre/fuaa017] [PMID: 32472938]
[90]
Mutti M, Corsini L. Robust approaches for the production of active ingredient and drug product for human phage therapy. Front Microbiol 2019; 10: 2289.
[http://dx.doi.org/10.3389/fmicb.2019.02289] [PMID: 31649636]
[91]
Pirnay JP, Blasdel BG, Bretaudeau L, et al. Quality and safety requirements for sustainable phage therapy products. Pharm Res 2015; 32(7): 2173-9.
[http://dx.doi.org/10.1007/s11095-014-1617-7] [PMID: 25585954]
[92]
Łobocka M, Dąbrowska K, Górski A. Engineered bacteriophage therapeutics: Rationale, challenges and future. BioDrugs 2021; 35: 255-80.
[93]
Jérôme G. Prophage in phage manufacturing: Is the risk overrated compared to other therapies or food? Antibiotics 2020; 9(8): 435.
[http://dx.doi.org/10.3390/antibiotics9080435] [PMID: 32707901]
[94]
Merabishvili M, Pirnay JP, De Vos D. Guidelines to Compose an Ideal Bacteriophage Cocktail. In: Azeredo J, Sil-lankorva S, Eds. Bacteriophage Therapy Methods in Molecular Biology. Humana Press, New York 2018; Vol. 1693.
[http://dx.doi.org/10.1007/978-1-4939-7395-8_9]
[95]
Jault P, Leclerc T, Jennes S, et al. Efficacy and tolerability of a cocktail of bacteriophages to treat burn wounds infected by Pseudomonas aeruginosa (PhagoBurn): A randomised, controlled, double-blind phase 1/2 trial. Lancet Infect Dis 2019; 19(1): 35-45.
[http://dx.doi.org/10.1016/S1473-3099(18)30482-1] [PMID: 30292481]
[96]
Cortes P, Cano-Sarabia M. Nano/Micro formulations for bacteriophage delivery. In: Bacteriophage Therapy: Methods in Molecular Biology. Humana Press, New York, 2018; Vol. 1693: pp. 271-83.
[97]
D. K. Phage therapy: What factors shape phage pharmacokinetics and bioavailability? Systematic and critical review. 2019. Med Res Rev 2019; 39(5): 2000-25.
[98]
Otero J, García-Rodríguez A, Cano-Sarabia M, et al. Biodistribution of liposome-encapsulated bacteriophages and their transcytosis during oral phage therapy. Front Microbiol 2019; 10: 689.
[http://dx.doi.org/10.3389/fmicb.2019.00689] [PMID: 31019499]
[99]
Vinner GK, Richards K, Leppanen M, Sagona AP, Malik JD. Microencapsulation of enteric bacteriophages in a pH-Responsive solid oral dosage formulation using a scalable membrane emulsification process. Pharmaceutics 2019; 11(9): 475.
[http://dx.doi.org/10.3390/pharmaceutics11090475]
[100]
David Y, Dunn LVA. Icemaker, process for controlling same and making ice. Patent US 10174981B2 2019. Available from: US00000010174981B220190108 (nist.gov)
[101]
Oechslin F. Resistance development to bacteriophages occurring during bacteriophage therapy. Viruses 2018; 10(7): 351.
[http://dx.doi.org/10.3390/v10070351] [PMID: 29966329]
[102]
Bernheim A, Sorek R. The pan-immune system of bacteria: antiviral defence as a community resource. Nat Rev Microbiol 2020; 18(2): 113-9.
[http://dx.doi.org/10.1038/s41579-019-0278-2] [PMID: 31695182]
[103]
Oechslin F, Piccardi P, Mancini S, et al. Synergistic interaction between phage therapy and antibiotics clears Pseudomonas aeruginosa infection in endocarditis and reduces virulence. J Infect Dis 2016; 215(5): jiw632.
[http://dx.doi.org/10.1093/infdis/jiw632] [PMID: 28007922]
[104]
Jansen M, Wahida A, Latz S, et al. Enhanced antibacterial effect of the novel T4-like bacteriophage KARL-1 in combination with antibiotics against multi-drug resistant Acinetobacter baumannii. Sci Rep 2018; 8(1): 14140.
[http://dx.doi.org/10.1038/s41598-018-32344-y] [PMID: 30237558]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy