Silk Route to the Acceptance and Re-Implementation of Bacteriophage Therapy—Part II
Abstract
:1. Introduction
1.1. Factors Impacting the Broad-Scale Application of the Bacteriophages
1.1.1. Quality and Quantity of Previously Conducted Study Set-Ups
1.1.2. Bacteriophage Production and Application Methods in Context of the Current Legal Framework
1.1.3. Lack of Awareness among (Para-) Medical Staff and the Public About Bacteriophage Therapy
1.1.4. Limitations in Intellectual Property Protection
2. The Nagoya Protocol and the Implications on Bacteriophage Therapy
3. The Future of Bacteriophage Therapy by the CRISPR/Cas System
4. Limitations and Concerns for CRISPR/Cas Gene Edited Bacteriophages
4.1. Efficacy
4.2. Legislative Hurdles
4.3. Safety and Environmental Risks
5. Discussion and Conclusions
Author Contributions
Conflicts of Interest
References
- Aminov, R. History of antimicrobial drug discovery: Major classes and health impact. Biochem. Pharmacol. 2017, 133, 4–19. [Google Scholar] [CrossRef] [PubMed]
- Aminov, R.I. A brief history of the antibiotic era: Lessons learned and challenges for the future. Front. Microbiol. 2010, 1, 134. [Google Scholar] [CrossRef] [PubMed]
- Centers for Disease Control and Prevention. Antibiotic Resistance Threats in the United States, 2013; Centers for Disease Control and Prevention: Atlanta, GA, USA, 2013.
- European Centre for Disease Prevention and Control; European Food Safety Authority; European Medicines Agency. Ecdc/efsa/ema second joint report on the integrated analysis of the consumption of antimicrobial agents and occurrence of antimicrobial resistance in bacteria from humans and food-producing animals. EFSA J. 2017, 15, e04872. [Google Scholar] [CrossRef]
- Organization, W.H. Global Action Plan on Antimicrobial Resistance; WHO: Geneva, Switzerland, 2015. [Google Scholar]
- O’Neill, J. Tackling Drug–Resistant Infections Globally: Final Report and Recommendations. Available online: http://www.iica.int/en/press/news/tackling-drug-resistant-infections-globally-final-report-and-recommendations (accessed on 18 April 2018).
- Expert round-table on acceptance and re–implementation of bacteriophage therapy. Silk route to the acceptance and re-implementation of bacteriophage therapy. Biotechnol. J. 2016, 11, 595–600. [Google Scholar] [Green Version]
- Servick, K. Beleaguered phage therapy trial presses on. Science 2016, 352, 1506. [Google Scholar] [CrossRef] [PubMed]
- PhagoBurn. Evaluation of Phage Therapy for the Treatment of Escherichia coli and Pseudomonas aeruginosa Burn Wound Infections. Available online: http://www.phagoburn.eu/ (accessed on 18 April 2018).
- Sarker, S.A.; Sultana, S.; Reuteler, G.; Moine, D.; Descombes, P.; Charton, F.; Bourdin, G.; McCallin, S.; Ngom-Bru, C.; Neville, T. Oral phage therapy of acute bacterial diarrhea with two coliphage preparations: A randomized trial in children from bangladesh. EBioMedicine 2016, 4, 124–137. [Google Scholar] [CrossRef] [PubMed]
- Międzybrodzki, R.; Borysowski, J.; Weber-Dąbrowska, B.; Fortuna, W.; Letkiewicz, S.; Szufnarowski, K.; Pawełczyk, Z.; Rogóż, P.; Kłak, M.; Wojtasik, E. Chapter 3–Clinical aspects of phage therapy. In Advances in Virus Research; Łobocka, M., Szybalski, W., Eds.; Academic Press: Cambridge, MA, USA, 2012; Volume 83, pp. 73–121. [Google Scholar]
- Abedon, S.T.; García, P.; Mullany, P.; Aminov, R. Editorial: Phage therapy: past, present and future. Front. Microbiol. 2017, 8, 981. [Google Scholar] [CrossRef] [PubMed]
- McCallin, S.; Alam Sarker, S.; Barretto, C.; Sultana, S.; Berger, B.; Huq, S.; Krause, L.; Bibiloni, R.; Schmitt, B.; Reuteler, G. Safety analysis of a russian phage cocktail: From metagenomic analysis to oral application in healthy human subjects. Virology 2013, 443, 187–196. [Google Scholar] [CrossRef] [PubMed]
- Leitner, L.; Sybesma, W.; Chanishvili, N.; Goderdzishvili, M.; Chkhotua, A.; Ujmajuridze, A.; Schneider, M.P.; Sartori, A.; Mehnert, U.; Bachmann, L.M. Bacteriophages for treating urinary tract infections in patients undergoing transurethral resection of the prostate: A randomized, placebo-controlled, double-blind clinical trial. BMC Urol. 2017, 17, 90. [Google Scholar] [CrossRef] [PubMed]
- Ujmajuridze, A.; Chanishvili, N.; Goderdzishvili, M.; Leitner, L.; Mehnert, U.; Chkhotua, A.; Kessler, T.; Sybesma, W. Adapted bacteriophages for treating urinary tract infections. 2018. Submitted for publication. [Google Scholar]
- Górski, A.; Międzybrodzki, R.; Weber-Dąbrowska, B.; Fortuna, W.; Letkiewicz, S.; Rogóż, P.; Jończyk-Matysiak, E.; Dąbrowska, K.; Majewska, J.; Borysowski, J. Phage therapy: Combating infections with potential for evolving from merely a treatment for complications to targeting diseases. Front. Microbiol. 2016, 7, 1515. [Google Scholar] [CrossRef] [PubMed]
- Saussereau, E.; Vachier, I.; Chiron, R.; Godbert, B.; Sermet, I.; Dufour, N.; Pirnay, J.P.; De Vos, D.; Carrié, F.; Molinari, N. Effectiveness of bacteriophages in the sputum of cystic fibrosis patients. Clin. Microbiol. Infect. 2014, 20, O983–O990. [Google Scholar] [CrossRef] [PubMed]
- Bernstein, L.J.; Ochs, H.D.; Wedgwood, R.J.; Rubinstein, A. Defective humoral immunity in pediatric acquired immune deficiency syndrome. J. Pediatr. 1985, 107, 352–357. [Google Scholar] [CrossRef]
- Rhoads, D.D.; Wolcott, R.D.; Kuskowski, M.A.; Wolcott, B.M.; Ward, L.S.; Sulakvelidze, A. Bacteriophage therapy of venous leg ulcers in humans: Results of a phase i safety trial. J. Wound Care 2009, 18, 237–243. [Google Scholar] [CrossRef] [PubMed]
- Duplessis, C.; Biswas, B.; Hanisch, B.; Perkins, M.; Henry, M.; Quinones, J.; Wolfe, D.; Estrella, L.; Hamilton, T. Refractory pseudomonas bacteremia in a 2-year-old sterilized by bacteriophage therapy. J. Pediatr. Infect. Dis. Soc. 2017. [Google Scholar] [CrossRef] [PubMed]
- Schooley, R.T.; Biswas, B.; Gill, J.J.; Hernandez-Morales, A.; Lancaster, J.; Lessor, L.; Barr, J.J.; Reed, S.L.; Rohwer, F.; Benler, S. 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, e00954-17. [Google Scholar] [CrossRef] [PubMed]
- Zhvania, P.; Hoyle, N.S.; Nadareishvili, L.; Nizharadze, D.; Kutateladze, M. Phage therapy in a 16-year-old boy with netherton syndrome. Front. Med. 2017, 4, 94. [Google Scholar] [CrossRef] [PubMed]
- Jennes, S.; Merabishvili, M.; Soentjens, P.; Pang, K.W.; Rose, T.; Keersebilck, E.; Soete, O.; François, P.-M.; Teodorescu, S.; Verween, G. Use of bacteriophages in the treatment of colistin-only-sensitive pseudomonas aeruginosa septicaemia in a patient with acute kidney injury—A case report. Crit. Care 2017, 21, 129. [Google Scholar] [CrossRef] [PubMed]
- Fish, R.; Kutter, E.; Wheat, G.; Blasdel, B.; Kutateladze, M.; Kuhl, S. Bacteriophage treatment of intransigent diabetic toe ulcers: A case series. J. Wound Care 2016, 25, S27–S33. [Google Scholar] [CrossRef] [PubMed]
- Soothill, J. Use of bacteriophages in the treatment of pseudomonas aeruginosa infections. Expert Rev. Anti-Infect. Ther. 2013, 11, 909–915. [Google Scholar] [CrossRef] [PubMed]
- Khawaldeh, A.; Morales, S.; Dillon, B.; Alavidze, Z.; Ginn, A.N.; Thomas, L.; Chapman, S.J.; Dublanchet, A.; Smithyman, A.; Iredell, J.R. Bacteriophage therapy for refractory pseudomonas aeruginosa urinary tract infection. J. Med. Microbiol. 2011, 60, 1697–1700. [Google Scholar] [CrossRef] [PubMed]
- Letkiewicz, S.; Międzybrodzki, R.; Fortuna, W.; Weber-Dąbrowska, B.; Górski, A. Eradication of enterococcus faecalis by phage therapy in chronic bacterial prostatitis—Case report. Folia Microbiol. 2009, 54, 457–461. [Google Scholar] [CrossRef] [PubMed]
- Fadlallah, A.; Chelala, E.; Legeais, J.-M. Corneal infection therapy with topical bacteriophage administration. Open Ophthalmol. J. 2015, 9, 167–168. [Google Scholar] [CrossRef] [PubMed]
- Jikia, D.; Chkhaidze, N.; Imedashvili, E.; Mgaloblishvili, I.; Tsitlanadze, G.; Katsarava, R.; Glenn Morris, J.; Sulakvelidze, A. The use of a novel biodegradable preparation capable of the sustained release of bacteriophages and ciprofloxacin, in the complex treatment of multidrug-resistant staphylococcus aureus-infected local radiation injuries caused by exposure to sr90. Clin. Exp. Dermatol. 2005, 30, 23–26. [Google Scholar] [CrossRef] [PubMed]
- Leszczyński, P.; Weber-Dabrowska, B.; Kohutnicka, M.; Łuczak, M.; Górecki, A.; Górski, A. Successful eradication of methicillin-resistantstaphylococcus aureus (MRSA) intestinal carrier status in a healthcare worker—Case report. Folia Microbiol. 2006, 51, 236–238. [Google Scholar] [CrossRef]
- Fevre, C.; Ferry, T.; Petitjean, C.; Leboucher, C.; L’hostis, G.; Laurent, F.; Regulski, K. Phage therapy: Compassionate use in france in 2017. In Phages-sur-Yvette; Gif-sur-Yvette: Essonne, France, 2017. [Google Scholar]
- Verbeken, G.; Pirnay, J.P.; De Vos, D.; Jennes, S.; Zizi, M.; Lavigne, R.; Casteels, M.; Huys, I. Optimizing the european regulatory framework for sustainable bacteriophage therapy in human medicine. Arch. Immunol. Ther. Exp. 2012, 60, 161–172. [Google Scholar] [CrossRef] [PubMed]
- Pirnay, J.-P.; Verbeken, G.; Ceyssens, P.-J.; Huys, I.; De Vos, D.; Ameloot, C.; Fauconnier, A. The magistral phage. Viruses 2018, 10, 64. [Google Scholar] [CrossRef] [PubMed]
- Young, R.; Gill, J.J. Microbiology. Phage therapy redux–What is to be done? Science 2015, 350, 1163–1164. [Google Scholar] [CrossRef] [PubMed]
- Debarbieux, L.; Pirnay, J.-P.; Verbeken, G.; De Vos, D.; Merabishvili, M.; Huys, I.; Patey, O.; Schoonjans, D.; Vaneechoutte, M.; Zizi, M. A bacteriophage journey at the european medicines agency. FEMS Microbiol. Lett. 2015, 363. [Google Scholar] [CrossRef] [PubMed]
- Nagel, T.E.; Chan, B.K.; De Vos, D.; El-Shibiny, A.; Kang’ethe, E.K.; Makumi, A.; Pirnay, J.-P. The developing world urgently needs phages to combat pathogenic bacteria. Front. Microbiol. 2016, 7, 882. [Google Scholar] [CrossRef] [PubMed]
- Van Zimmeren, E.; Vanneste, S.; Matthijs, G.; Vanhaverbeke, W.; Van Overwalle, G. Patent pools and clearinghouses in the life sciences. Trends Biotechnol. 2011, 29, 569–576. [Google Scholar] [CrossRef] [PubMed]
- Inoue, M.; Minghui, R. Antimicrobial resistance: Translating political commitment into national action. Bull. World Health Organ. 2017, 95. [Google Scholar] [CrossRef]
- Johansen, E. Future access and improvement of industrial lactic acid bacteria cultures. Microb. Cell Fact. 2017, 16, 230. [Google Scholar] [CrossRef] [PubMed]
- Overmann, J.; Scholz, A.H. Microbiological research under the nagoya protocol: Facts and fiction. Trends Microbiol. 2017, 25, 85–88. [Google Scholar] [CrossRef] [PubMed]
- Smith, D.; da Silva, M.; Jackson, J.; Lyal, C. Explanation of the nagoya protocol on access and benefit sharing and its implication for microbiology. Microbiology 2017, 163, 289–296. [Google Scholar] [CrossRef] [PubMed]
- McCluskey, K.; Barker, K.B.; Barton, H.A.; Boundy-Mills, K.; Brown, D.R.; Coddington, J.A.; Cook, K.; Desmeth, P.; Geiser, D.; Glaeser, J.A. The U.S. Culture collection network responding to the requirements of the nagoya protocol on access and benefit sharing. mBio 2017, 8. [Google Scholar] [CrossRef] [PubMed]
- Kiro, R.; Shitrit, D.; Qimron, U. Efficient engineering of a bacteriophage genome using the type i-e crispr-cas system. RNA Biol. 2014, 11, 42–44. [Google Scholar] [CrossRef] [PubMed]
- Bari, S.M.N.; Walker, F.C.; Cater, K.; Aslan, B.; Hatoum-Aslan, A. Strategies for editing virulent staphylococcal phages using crispr-cas10. ACS Synth. Biol. 2017, 6, 2316–2325. [Google Scholar] [CrossRef] [PubMed]
- Pouillot, F.; Blois, H.; Iris, F. Genetically engineered virulent phage banks in the detection and control of emergent pathogenic bacteria. Biosecur. Bioterror. 2010, 8, 155–169. [Google Scholar] [CrossRef] [PubMed]
- Oślizło, A.; Miernikiewicz, P.; Piotrowicz, A.; Owczarek, B.; Kopciuch, A.; Figura, G.; Dąbrowska, K. Purification of phage display-modified bacteriophage T4 by affinity chromatography. BMC Biotechnol. 2011, 11, 59. [Google Scholar] [CrossRef] [PubMed]
- Vitiello, C.L.; Merril, C.R.; Adhya, S. An amino acid substitution in a capsid protein enhances phage survival in mouse circulatory system more than a 1000-fold. Virus Res. 2005, 114, 101–103. [Google Scholar] [CrossRef] [PubMed]
- Fagen, J.R.; Collias, D.; Singh, A.K.; Beisel, C.L. Advancing the design and delivery of crispr antimicrobials. Curr. Opin. Biomed. Eng. 2017, 4, 57–64. [Google Scholar] [CrossRef]
- Cross, A. Endotoxin: Back to the future. Crit. Care Med. 2016, 44, 450–451. [Google Scholar] [CrossRef] [PubMed]
- Martel, B.; Moineau, S. Crispr-cas: An efficient tool for genome engineering of virulent bacteriophages. Nucleic Acids Res. 2014, 42, 9504–9513. [Google Scholar] [CrossRef] [PubMed]
- Box, A.M.; McGuffie, M.J.; O’Hara, B.J.; Seed, K.D. Functional analysis of bacteriophage immunity through a type i-e crispr-cas system in vibrio cholerae and its application in bacteriophage genome engineering. J. Bacteriol. 2016, 198, 578–590. [Google Scholar] [CrossRef] [PubMed]
- Lemay, M.-L.; Tremblay, D.M.; Moineau, S. Genome engineering of virulent lactococcal phages using crispr-cas9. ACS Synth. Biol. 2017, 6, 1351–1358. [Google Scholar] [CrossRef] [PubMed]
- Tao, P.; Wu, X.; Tang, W.-C.; Zhu, J.; Rao, V. Engineering of bacteriophage t4 genome using crispr-cas9. ACS Synth. Biol. 2017, 6, 1952–1961. [Google Scholar] [CrossRef] [PubMed]
- Luo, M.L.; Leenay, R.T.; Beisel, C.L. Current and future prospects for crispr-based tools in bacteria. Biotechnol. Bioeng. 2016, 113, 930–943. [Google Scholar] [CrossRef] [PubMed]
- Bardy, P.; Pantucek, R.; Benesik, M.; Doskar, J. Genetically modified bacteriophages in applied microbiology. J. Appl. Microbiol. 2016, 121, 618–633. [Google Scholar] [CrossRef] [PubMed]
- Golan, T.; Milella, M.; Ackerstein, A.; Berger, R. The changing face of clinical trials in the personalized medicine and immuno-oncology era: Report from The International Congress on Clinical Trials in Oncology & Hemato–Oncology (Icto 2017). J. Exp. Clin. Cancer Res. 2017, 36, 192. [Google Scholar] [PubMed]
Name of Study and Organizations Running the Study | Target Organism(s) | Description and Objectives | Outcome Measures | Additional Comments | Clinical Trials.gov Identifier/Reference |
---|---|---|---|---|---|
Bacteriophages for treating urinary tract infections in patients undergoing transurethral resection of the prostate: a randomized, placebo-controlled, double-blind clinical trial. Tzulukidze National Center of Urology, Tbilisi, Georgia; Eliava Institute of Bacteriophages, Microbiology, and Virology in Tbilisi, Georgia; Balgrist University Hospital, Zürich, Switzerland The study is run in The Republic of Georgia. | Enterococcus spp., Streptococcus spp., Escherichia coli, Proteus spp., P. aeruginosa | Randomized placebo-controlled double-blind clinical trial: Patients planned for transurethral resection of the prostate are screened for UTIs and enrolled if eligible microorganisms in urine culture are ≥104 cfu/mL. Patients are randomized in a double-blind fashion to the three study treatment arms of 27 people, each in a 1:1:1 ratio to receive either: (a) bacteriophage (b) placebo solution, or (c) antibiotic treatment according to the antibiotic sensitivity pattern. | Primary: Success of intravesical treatment, defined as normalization of urine culture (no evidence of bacteria, i.e., <104 colony forming units/mL) after 7 days of treatment. Secondary: Adverse events, in categorization according to the National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE) version four in grade one to five. Tertiary: Changes in bladder and pain diary assessment of number of voids, number of leakages, post void residual. | The study uses the commercially available Pyo-bacteriophage cocktail as produced by The Eliava institute in Tbilisi. 81 patients are involved. The Pyo-bacteriophage cocktail is subjected to continuous adaptation during the study. The study started in 2016, and is expected to end in 2018. | NCT03140085 [14] |
PHAGOPIED - Standard treatment associated with bacteriophage therapy vs. placebo for diabetic foot ulcers infected by Staphylococcus aureus. The study is run by the Centre Hospitalier Universitaire de Nīmes, France, with collaboration with Pherecydes Pharma, Romainville, France. | Staphylococcus aureus, MSSA and MRSA | This project utilizes anti-Staphylococcus bacteriophages, delivered topically, vs. a placebo control. The study uses random allocations in a parallel assignment intervention design. The main objective of this study is to compare the efficacy of standard treatment associated with a topical anti-staphylococcal bacteriophage cocktail versus standard treatment plus placebo for diabetic foot ulcers mono-infected by methicillin-resistant or susceptible S. aureus (MRSA or MSSA) as measured by the relative reduction in wound surface area (%) at 12 weeks. | Primary: The relative reduction in wound surface area over 12 weeks. Secondary: Safety effects, local side effects (rash onset or worsening of local inflammatory signs) and general symptoms (vital signs, fever, rash, arthralgia, gastrointestinal symptoms). | First posted online in 2016. This study is not yet recruiting, but 60 patients are expected to join. | NCT02664740 |
MUCOPHAGES - Bacteriophage effects on Pseudomonas aeruginosa. The study is run by the University Hospital Montpellier, France. | Pseudomonas aeruginosa | The study is designed to evaluate the efficacy of bacteriophages on P. aeruginosa isolates recovered from sputum. | The study utilizes a suspension of ten bacteriophages. These are tested against isolates recovered from cystic fibrosis patients, to determine their ability to infect these strains. | Completed in 2012. No results posted online. | NCT01818206 [17] |
Experimental bacteriophage therapy of bacterial infections. The study is led by the Polish Academy of Sciences. | Staphylococcus, Enterococcus, Pseudomonas, Escherichia, Klebsiella, Proteus, Citrobacter, Acinetobacter, Serratia, Morganella, Shigella, Salmonella, Enterobacter, Stenotrophomonas, Burkholderia | The study uses suspensions of lytic bacteriophages active against clinical isolates of the test species. The program determines to use bacteriophage treatment in a therapeutic role where no other viable treatment is available. For each patient only, specific formulations of single bacteriophage or a bacteriophage mixture that are active against the pathogenic bacterial strain or strains isolated from the patient are used for the treatment (oral, rectal and/or topical application). | The principle focus of the work is to use bacteriophage suspensions to treat the following conditions: bone, upper respiratory, genital and urinary tract infections, as well as post-operative non-healing wounds where antibiotic treatment has not produced positive results. | Start: 2005. Current status unknown. Last update posted in 2013. Number of persons involved has not been stated. | NCT00945087 [11] |
PhagoBurn. Phase I/II Clinical Trial. This Project is a European Research & Development (R&D) Project Funded by the European Commission Under the 7th Framework Program for Research and Development Involving seven Clinical Sites in France, Belgium & Switzerland. | Escherichia coli & Pseudomonas aeruginosa | Evaluation of bacteriophage therapy for the treatment of Escherichia coli and Pseudomonas aeruginosa wound infections in burned patients A randomized, parallel assignment study assessing tolerance and efficacy of local bacteriophage treatment of wound infections due to E. coli or Ps. aeruginosa in burned patients. | This study tests the efficacy of E. coli and Ps. aeruginosa bacteriophage cocktails against silver sulfadiazine to treat wound infections by those bacterial species. Primary: Time necessary for a persistent bacteria reduction of two modes or persistent bacteria eradication relative to D0 adjusted on antibiotic treatment (active on targeted strain) introduced between D1 to D7. Secondary: Assessment of tolerance of treatment over 21 days. Adverse events frequencies will be assessed in each treatment arm. bacteriophage therapy safety profile will be compared to safety profile of standard of care. Incidence and delay of infection reduction with different bacterial species from the targets over a period of seven days. Number of sites cured: The number of infected burns or infected wounds getting a clinical improvement will be described and compared between treatment group over a period of 7 day. | Launched in 2013 and achieved in 2017, PhagoBurn was the world first prospective multicentric, randomized, single-blind and controlled clinical trial of bacteriophage therapy ever performed according to both Good Manufacturing (GMP) and Good Clinical Practices (GCP). Only 27 patients between 11 centers were included, which is far from the pre-calculated 220 patients needed to provide statistically significant results for the study. See main text for more information and lessons learned. | NCT02116010 [8,9] |
Antibacterial treatment against diarrhea in oral rehydration solution. The study was run by Nestlé, Switzerland in collaboration with Dhaka Hospital of the International Centre for Diarrheal Disease Research, Bangladesh. | Escherichia. coli (T4 bacteriophage) | This randomized double-blind, placebo-controlled trial aims to demonstrate the potentials of a new form of therapy for childhood diarrhea, by measuring the effect of oral administered E. coli bacteriophage in children aged 4–60 months of age with proven ETEC and EPEC diarrha. | Primary outcome measures: Assessment of safety, tolerability and efficacy (reduce severity of diarrhea assessed by reduced stool volume and stool frequency) of oral administration of T4 bacteriophages in young children with diarrhea due to ETEC and/or EPEC infections. Time frame: five days. Secondary outcome measures: Clinical assessment, blood tests, morbidity, duration of hospitalization. Time frame: five days. | First posted online 2009; study ended in 2013. Oral coliphages showed a safe gut transit in children, but failed to achieve intestinal amplification and to improve diarrhea outcome, possibly due to insufficient bacteriophage coverage and too low E. coli pathogen titers requiring higher oral bacteriophage doses. | NCT00937274 [10] |
Existence in the human digestive flora of bacteriophages able to prevent the acquisition of multiresistant Enterobacteria (PHAGO-BMR). The study is led by Assistance Publique-Hôpitaux de Paris, France. | MDR- Enterobacteria | The study plans to recruit 460 people hospitalized in intensive care unit (resuscitation). The choice of this unit is linked to the fact that the monitoring of resistant bacteria is carried out regularly during the hospitalization. On stool samples collected at separate times of the stay (admission and then during the stay), the scientists look for 2 types of bacteria and viruses capable of destroying them. | Primary: Presence or absence of bacteriophages capable of lysing circulating Ec-ESBL/EPC or Kp-ESBE/EPC in resuscitation units in non-carriers having acquired carriers E. coli or K. pneumoniae producing ESBL or carbapenemases. Secondary: Presence or absence of bacteriophages in patients identified as carriers of Ec-ESBL/EPC or Kp-ESBL/EPC at entry to resuscitation (control population). Isolated bacteriophages will be characterized. | First posted online in 2017. The study is not yet recruiting. | NCT03231267 |
METAKIDS Phages dynamics and influences during human gut microbiome establishment. | Enteric microbial species | This project relies on the ability of Meta3C, a technique developed to identify the bacterial host genomes of the different bacteriophages the investigators will detect thanks to the physical collision these molecules experience. Given the role that human gut bacteriophages may play in shaping the development of host microbiomes, their potential for application is of great interest. | Primary outcome measures: Genomic reconstruction and characterization of the different genomes (phages, bacteria, yeast) present in the human gut during the three first years of life. These outcomes will provide a large catalog of DNA sequences. Characterization of the variation of the different species present in human gut during the three first years of life and characterization of bacteriophages-bacteria interactions. This outcome will provide access to the dynamics of the different species present during this period and possibility to correlate them with environmental variation (dietary, age). Secondary: Characterization of bacteriophages and bacteria variations in response to environmental perturbations during infant gut development. Time frame: two to three weeks. | First posted online in 2017; the study is currently recruiting. Estimated enrollment is 20 persons. | NCT03296631 |
Evaluate bacteriophage as a useful immunogen in patients with primary immune deficiency diseases (PIDD) The study is run by the University of South Florida, USA. | Escherihia. coli | This protocol is designed to ascertain whether the bacteriophage 0X174 neoantigen is safe and effective as an antigen used in the evaluation of primary and secondary immune responses. Bacteriophage 0X174 is given intravenously two billion PFU/Kg of body weight; small blood specimens of 3–5 mL (about 1 teaspoon) are collected after 15 min, 7 days, 14 days, and 28 days. | Primary Outcome Measures: Evidence of capacity of switch from IgM to IgG during 12 weeks of trial. Blood samples are obtained after each immunization of bacteriophages. | Current status unknown. First posted online in 2012. Last update 2012. All patients receive two doses of bacteriophages. Selected patients may receive a tertiary vaccine. | NCT01617122 |
Evaluation and detection of facial Propionibacterium acnes bacteria and bacteriophage The study is led by Maccabi Healthcare Services, Israel. | Propionibacterium acnes | This multi-center, outpatient study will extract and evaluate the presence of facial P. acnes bacteria and bacteriophage strains using pore strips on up to 400 human subjects. An additional P. acnes visual detection method (VISIOPOR® PP34N) will be used in this study as per PI decision to explore whether there is a correlation between P. acnes bacterial presence and fluorescent signal. | Primary: Detection and analysis of facial P. acnes presence. Time frame: Day 0 and week 8. Secondary: Assessing correlation between bacteriophage and P. acnes using a. Demographic Questionnaire b. Visual Supportive Methodology (VISIOPOR® PP34N) as per PI decision. Time frame: As above. | First posted online in 2017. Not currently recruiting, but 400 people are estimated to participate. | NCT03009903 |
Bacteriophages PreforPro cocktails as novel Prebiotics The study is led by Colorado State University, USA. PreforPro is commercialized by Deerland Enzymes, Kennesaw, GA, USA | Enteric bacteria | The bacteriophage Study is a randomized, double-blind, placebo-controlled crossover trial that investigates the utility of four supplemental bacteriophage strains (LH01-Myoviridae, LL5-Siphoviridae, T4D-Myoviridae, and LL12-Myoviridae) to modulate the gut microbiota, and therefore ameliorate common inflammation-related GI distress symptoms (e.g., gas, bloating, diarrhea, constipation, etc.) experienced by healthy individuals. The main goal of this study is to see if consumption of PreforPro, a commercially available prebiotic dietary supplement consisting of a mixture of bacteriophages, improves gut bacteria profiles in individuals relative to a placebo control. | Primary: Microbiota modulation. Time frame: Baseline visit prior to starting treatments, four weeks after starting treatment one, end of two-week washout period, 4-weeks after starting treatment two. Use of 16s rRNA sequencing of stool samples to determine whether the administered interventions resulted in changes to microbial composition. Secondary: Local inflammation Time frame: as above. Inflammation in the bowels will be assessed by use of ELISA test for fecal calprotectin. Systemic Inflammation. Time Frame: As above. Systemic inflammation will be assessed by an ELISA test for CRP and circulating cytokines and immune factors. | The study completed in 2017, but results have not yet been posted online. 43 persons enrolled in the study. | NCT03269617 |
The Use of Bacteriophage Phi X174 to Assess the Immune Competence of HIV-Infected Patients in vivo. The study is run by the National Institute of Allergy and Infectious Diseases, USA. | Escherichia. coli | The objective of this study is to evaluate the safety and utility of bacteriophage phi X174 immunization as a tool to assess the immune competence of HIV-infected patients at different stages of disease in vivo, and to assess the impact of viral load levels and therapy-induced changes in viral load levels on the response to immunization with the neo-antigen bacteriophage phi X174. | Primary. Immune parameters (not further published online. | Study started in 1996, and ended in 2000. 52 patients were involved in the study. | NCT00001540 [18] |
Randomized and double-blinded placebo-controlled study of topical application of AB-SA01 cocktail to intact skin of healthy adults. The study is run by AmpliPhi bacteriophage Ltd., the US Army, and the Walter Reed Army Institute of Research Clinical Trials Center, USA. | Staphylococcus aureus | The study aims to examine the safety of ascending doses of AB-SA01 when topically applied to intact skin of healthy adults. AB-SA01 consists of three bacteriophages (viruses) that target Staphylococcus aureus bacteria. The safety of AB-SA01 will be assessed when topically administered once daily to the volar aspect of the forearm at different doses for three consecutive days. | Primary: Occurrence, intensity, and relationship of adverse events (AEs) from first dose through the end of study visit (14 ± 2 days). Change from baseline in clinical laboratory tests. Time frame: Day 0 (pre-dose), Day 3, and Day 14 ± 2 days. Clinical laboratory tests (hematology, chemistry, and urinalysis). Skin Reaction change from Baseline. Time Frame. | First published online in 2016, and the last update was in 2016 as well. 12 persons recruited. | NCT02757755 |
A prospective, randomized, double-blind controlled study of WPP-201 for the safety and efficacy of treatment of venous leg ulcers. The study was run by Southwest Regional Wound Care Center, USA. | Pseudomonas aeruginosa | The study was designed to assess the safety of Ps. aeruginosa-specific bacteriophages for the treatment of leg ulcers in human patients. WPP-201 is a pH neutral, polyvalent bacteriophage preparation, which contains 8 bacteriophages lytic for Ps. aeruginosa, S. aureus, and E. coli. The cocktail contains a concentration of approximately 1 × 109 PFU/mL of each of the component monophages. | Primary: Evaluate the safety of the use of WPP-201. | Study started in 2008 and was completed in 2011. 64 patients were involved. This study found no safety concerns with the bacteriophage treatment. Efficacy of the preparation will need to be evaluated in a phase II efficacy study. | NCT00663091 [19] |
Case Study Title | Description | Outcomes | Comments | Reference |
---|---|---|---|---|
Refractory Pseudomonas Bacteremia in a Two-Year-Old sterilized by bacteriophage therapy | The authors report a complex case that involved a pediatric patient who experienced recalcitrant multidrug-resistant Pseudomonas aeruginosa infection complicated by bacteremia/sepsis; antibacterial options were limited because of resistance, allergies, and suboptimal source control. | A cocktail of 2 bacteriophages targeting the infectious organism introduced on 2 separate occasions sterilized the bacteremia. | USA | [20] |
Development and use of personalized bacteriophage-based therapeutic cocktails to treat a patient with a disseminated resistant Acinetobacter baumannii infection | The authors report on a method used to produce a personalized bacteriophage-based therapeutic treatment for a 68-year-old diabetic patient with necrotizing pancreatitis complicated by an MDR A. baumannii infection. Despite multiple antibiotic courses and efforts at percutaneous drainage of a pancreatic pseudocyst, the patient deteriorated over a 4-month period. In the absence of effective antibiotics, two laboratories identified nine different bacteriophages with lytic activity for an A. baumannii isolate from the patient. | Administration of bacteriophages intravenously and percutaneously into the abscess cavities was associated with reversal of the patient’s downward clinical trajectory, clearance of the A. baumannii infection, and a return to health. | USA | [21] |
Phage therapy in a 16-year-old boy with Netherton syndrome | The authors report on a 16-year-old male with all the typical manifestations of Netherton Syndrome, including atopic diathesis and ongoing serious staphylococcal infections and allergy to multiple antibiotics whose family sought help at the Eliava bacteriophage Therapy Center when all other treatment options were failing. | Treatment with several antistaphylococcal bacteriophage preparations led to significant improvement within seven days and very substantial changes in his symptoms and quality of life after treatment for six months, including return visits to the Eliava bacteriophage Therapy Center after three and six months of ongoing use of bacteriophage at home | Georgia. (Patient came from France) | [22] |
Use of bacteriophages in the treatment of colistin-only-sensitive Pseudomonas aeruginosa septicaemia in a patient with acute kidney injury—a case report | A 61-year-old man with gangrene of the peripheral extremities, resulting in the amputation of the lower limbs and the development of large necrotic pressure sores, developed septicaemia with colistin-only-sensitive P. aeruginosa. Intravenous colistin therapy was started. Ten days later, the patient developed acute kidney injury and antibiotic therapy was discontinued to prevent further kidney damage. P. aeruginosa septicaemia re-emerged and two bacteriophages, which showed in vitro activity against the patient’s P. aeruginosa isolates, were administered as a 6-h intravenous infusion for ten days. | Immediately upon bacteriophage application, blood cultures turned negative, CRP levels dropped and the fever disappeared. Kidney function recovered after a few days. Hemofiltration was avoided and no unexpected adverse events, clinical abnormalities or changes in laboratory test results that could be related to the application of bacteriophages were observed. | Belgium The patient died four months after bacteriophage therapy of sudden in-hospital refractory cardiac arrest due to blood culture-confirmed Klebsiella pneumoniae sepsis. | [23] |
Bacteriophage treatment of intransigent diabetic toe ulcers: a case series | The authors present a compassionate-use case series of nine patients with diabetes and poorly perfused toe ulcers containing culture-proven Staphylococcus aureus infected bone and soft tissue, who had responded poorly to recommended antibiotic therapy. | All infections responded to the bacteriophage applications and the ulcers healed in an average of seven weeks with infected bone debridement. One ulcer, where vascularity was extremely poor and bone was not removed to preserve hallux function, required 18 weeks of treatment. | USA | [24] |
Use of bacteriophages in the treatment of Pseudomonas aeruginosa infections | The author reports on bacteriophage treatment of Pseudomonas aeruginosa otitis in a pet dog and in a human burn wound patient. | Symptomatic improvement and bacteriophage multiplication were seen in the pet dog and in the human patient. | UK | [25] |
Clinical aspects of bacteriophage therapy | The authors present a detailed retrospective analysis of the results of bacteriophage therapy of 153 patients with a wide range of infections resistant to antibiotic therapy admitted for treatment at the bacteriophage therapy unit of the Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Wrocław, Poland, between January 2008 and December 2010. | Data suggest that bacteriophage therapy provided good clinical results in a significant cohort of patients with otherwise untreatable chronic bacterial infections and is essentially well tolerated. | Poland | [11] |
Bacteriophage therapy for refractory Pseudomonas aeruginosa urinary tract infection | The authors describe adjunctive bacteriophage therapy for refractory Pseudomonas aeruginosa urinary tract infection in the context of bilateral ureteric stents and bladder ulceration, after repeated failure of antibiotics alone. | Combined therapy was well-tolerated, apparently resulting in symptomatic relief and microbiological cure where repeated courses of antibiotics combined with stent removal had failed. Bacteriophage did not persist nor was any antibiotic- or bacteriophage resistant P. aeruginosa identified. | Australia Additive effort of combined bacteriophage-antibiotic treatment | [26] |
Eradication of Enterococcus faecalis by bacteriophage therapy in chronic bacterial prostatitis | The authors report on the treatment of three patients suffering from chronic bacterial prostatitis who were qualified for an experimental bacteriophage therapy protocol managed at the bacteriophage Therapy Unit in Wrocław. The patients had previously been treated unsuccessfully with long-term targeted antibiotics, autovaccines, and laser biostimulation. | Rectal application of bacteriophage lysates targeted against Enterococcus faecalis cultured from the prostatic fluid gave encouraging results regarding bacterial eradication, abatement of clinical symptoms of prostatitis, and lack of early disease recurrence. | Poland | [27] |
Corneal Infection Therapy with Topical Bacteriophage Administration | A 65-year-old woman suffering from MDR S. aureus infection of the cornea as a post-operative complication of a craniotomy. This patient suffered the chronic, persistent infection for years before seeking therapy in Tbilisi, Georgia. A single bacteriophage was administered both topically in the eye and nasal application and intravenous application for four weeks. | The patient’s regular physicians published that the patient’s ocular and nasal cultures after returning from Georgia were negative at three and six months post-treatment. | Georgia. (Patient came from France) | [28] |
The use of a novel biodegradable preparation capable of the sustained release of bacteriophages and ciprofloxacin, in the complex treatment of multi-drug resistant Staphylococcus Aureus-infected local radiation injuries caused by exposure to Sr90. | Authors report the topical use of PhagoBioDerm (phage + ciprofloxacin wound polymer) to treat two wounds infected with S. aureus after the failure of conventional treatment. The responsible pathogen was resistant to ciprofloxacin, but sensitive to the bacteriophage. | PhagobioDerm resulted in reduced purulent drainage and symptom amelioration. S. aureus was eliminated from the wound | Georgia | [29] |
Successful eradication of methicillin-resistant Staphylococcus aureus (MRSA) intestinal carrier status in a healthcare worker | Healthcare worker suffered urinary tract infections caused by MRSA that was carried in the GI tract. Authors report that bacteriophage was applied orally | Eradiation of carrier status | Poland | [30] |
Phage therapy compassionate use in France in 2017. | Abstract presented by Pherecydes at bacteriophages-sur-Yvette in November 2017 documenting the use of bacteriophage to treat two patients with severe bone and joint infections caused by MDR organisms with topical bacteriophage at a hospital in Lyon. | Symptom amelioration and no reported side effects. | France | [31] |
Open-label treatment of RCT with bacteriophages for treating urinary tract infections in patients undergoing transurethral resection of the prostate: a randomized, as mentioned in Table 1. | Prior to the start of the double-blind placebo-controlled trial, nine patients were treated with bacteriophage Pyo cocktail. | In six of nine patients, the titer of the pathogenic bacteria was decreased, varying between 1 log and 7 log (sterile). | Georgia | NCT03140085 [14]. |
Gene | Modification | Purpose | Advantage to Natural Bacteriophage Therapy | Related Reference |
---|---|---|---|---|
Antimicrobial protein * | Insertion/gene replacement | Phage killing other strains | Product for mixed infections | [7] |
Biofilm degrading enzyme * | Insertion/gene replacement | Degrading biofilm | More active against biofilm-producing strains | |
Virulence factor | Gene deletion | No virulence transfer | Novel therapeutic bacteriophage | |
Baseplate proteins/tail fibers | Gene replacement | Altered host-range | Novel therapeutic bacteriophage for mixed infections | |
Receptor-binding protein/structural proteins | Single gene mutations | Broader host-range | More effective, and faster to obtain than by natural selection | [45] |
Major capsid protein | Purification tags insertion | More efficient purification | Purer product | [46] |
Major capsid protein | Anti-immune tags insertion/single gene mutations | Longer circulation in bloodstream | More effective, and faster to obtain than by natural selection | [47] |
Various, e.g., lytic module | Gene knockout | Non-replicative bacteriophage | Replication control of a bacteriophage | [48] |
Endotoxin antibody * | Insertion/gene replacement | Endotoxin removal | Safer product | [49] |
Bacteriophage | Host | CRISPR/Cas Type Employed | Mutation | Purpose | Gene | Reference |
---|---|---|---|---|---|---|
T7 | E. coli | I-E | Gene deletion | PoC of CRISPR/Cas-editing in bacteriophage | gene 1.7 | [43] |
2972 | Streptococcus thermophilus | II-A/Cas9 | Gene replacement with methyltransferase/Gene deletion | PoC of native II-A editing, bacteriophage resistant to RM system | orf 33, 39 | [50] |
ICP1 | Vibrio cholera | I-E | Gene deletion/Gene replacement with GFP | PoC in Vibrio model | cas 1, cas 2–3 | [51] |
P2 | Lactococcus lactis | II-A/Cas9 | Deletion/insertion/substitution | PoC of heterologous II-A editing | orf 24, 42, 47, 49 | [52] |
T4 | E. coli | II-A/Cas9 | Substitutions, deletion | PoC of editing by two gRNA, functional study of gene knock-out | mcp, rnlB | [53] |
Andhra | Staphylococcus epidermidis | III-A/Cas10 | Substitutions | PoC of native III-A editing | orf 9, 10 | [44] |
ISP | Staphylococcus aureus | III-A/Cas10 | Substitutions | PoC of heterologous III-A editing | orf 61 | [44] |
© 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Expert round table on acceptance and re-implementation of bacteriophage therapy; Sybesma, W.; Rohde, C.; Bardy, P.; Pirnay, J.-P.; Cooper, I.; Caplin, J.; Chanishvili, N.; Coffey, A.; De Vos, D.; et al. Silk Route to the Acceptance and Re-Implementation of Bacteriophage Therapy—Part II. Antibiotics 2018, 7, 35. https://doi.org/10.3390/antibiotics7020035
Expert round table on acceptance and re-implementation of bacteriophage therapy, Sybesma W, Rohde C, Bardy P, Pirnay J-P, Cooper I, Caplin J, Chanishvili N, Coffey A, De Vos D, et al. Silk Route to the Acceptance and Re-Implementation of Bacteriophage Therapy—Part II. Antibiotics. 2018; 7(2):35. https://doi.org/10.3390/antibiotics7020035
Chicago/Turabian StyleExpert round table on acceptance and re-implementation of bacteriophage therapy, Wilbert Sybesma, Christine Rohde, Pavol Bardy, Jean-Paul Pirnay, Ian Cooper, Jonathan Caplin, Nina Chanishvili, Aidan Coffey, Daniel De Vos, and et al. 2018. "Silk Route to the Acceptance and Re-Implementation of Bacteriophage Therapy—Part II" Antibiotics 7, no. 2: 35. https://doi.org/10.3390/antibiotics7020035
APA StyleExpert round table on acceptance and re-implementation of bacteriophage therapy, Sybesma, W., Rohde, C., Bardy, P., Pirnay, J. -P., Cooper, I., Caplin, J., Chanishvili, N., Coffey, A., De Vos, D., Scholz, A. H., McCallin, S., Püschner, H. M., Pantucek, R., Aminov, R., Doškař, J., & Kurtbӧke, D. İ. (2018). Silk Route to the Acceptance and Re-Implementation of Bacteriophage Therapy—Part II. Antibiotics, 7(2), 35. https://doi.org/10.3390/antibiotics7020035