Vancomycin-resistant Enterococcus

Last updated
Vancomycin-resistant Enterococcus
Other namesVancomycin-resistant enterococci
Vancomycin-Resistant Enterococcus 01.jpg
SEM micrograph of vancomycin-resistant enterococci
Specialty Microbiology
PreventionScreen with peri-rectal swab [1]
TreatmentLinezolid [2]

Vancomycin-resistant Enterococcus, or vancomycin-resistant enterococci (VRE), are bacterial strains of the genus Enterococcus that are resistant to the antibiotic vancomycin. [3]

Contents

Mechanism of acquired resistance

Vancomycin Vancomycin-from-xtal-1996-3D-balls.png
Vancomycin

Six different types of vancomycin resistance are shown by enterococcus: Van-A, Van-B, Van-C, Van-D, Van-E and Van-G. [4] The significance is that Van-A VRE is resistant to both vancomycin and teicoplanin, [5] Van-B VRE is resistant to vancomycin but susceptible to teicoplanin, [6] [7] and Van-C is only partly resistant to vancomycin.

The mechanism of resistance to vancomycin found in enterococcus involves the alteration of the peptidoglycan synthesis pathway. [8]

The D-alanyl-D-lactate variation results in the loss of one hydrogen-bonding interaction (four, as opposed to five for D-alanyl-D-alanine) being possible between vancomycin and the peptide. The D-alanyl-D-serine variation causes a six-fold loss of affinity between vancomycin and the peptide, likely due to steric hindrance. [9] [10]

To become vancomycin-resistant, vancomycin-sensitive enterococci typically obtain new DNA in the form of plasmids or transposons which encode genes that confer vancomycin resistance. [11] This acquired vancomycin resistance is distinguished from the natural vancomycin resistance of certain enterococcal species including E. gallinarum and E. casseliflavus/flavescens. [12] [13]

Diagnosis

Once the individual has VRE, it is important to ascertain which strain. [14]

Screening

Screening for VRE can be accomplished in a number of ways. For inoculating peri-rectal/anal swabs or stool specimens directly, one method uses bile esculin azide agar plates containing 6 μg/ml of vancomycin. Black colonies should be identified as an enterococcus to species level and further confirmed as vancomycin resistant by an MIC method before reporting as VRE. [1]

Vancomycin resistance can be determined for enterococcal colonies available in pure culture by inoculating a suspension of the organism onto a commercially available brain heart infusion agar (BHIA) plate containing 6 μg/ml vancomycin. The Clinical and Laboratory Standards Institute (CLSI) recommends performing a vancomycin MIC test and also motility and pigment production tests to distinguish species with acquired resistance (vanA and vanB) from those with vanC intrinsic resistance. [1] Detection of vancomycin resistance by the use of PCR targeting vanA and vanB can also be performed. [15] [16]

Treatment of infection

Linezolid Linezolid-from-xtal-2008-3D-balls.png
Linezolid

Ceftriaxone (a third generation cephalosporin) use is a risk factor for colonization and infection by VRE, and restriction of cephalosporin usage has been associated with decreased VRE infection and transmission in hospitals. [17] Lactobacillus rhamnosus GG (LGG), a strain of L. rhamnosus, was used successfully for the first time to treat gastrointestinal carriage of VRE. [18] In the US, linezolid is commonly used to treat VRE. [2] The combination of daptomycin and ampicillin is another option to treat VRE infections, especially for bacteremia. [19] For invasive vancomycin-resistant E. faecalis infections, both ampicillin-ceftriaxone and ampicillin-gentamicin combinations have been used successfully, with the latter specifically showing success in treating endocarditis. [20] If the VRE strain is vanB, teicoplanin and dalbavancin are suitable therapeutic options. [21] Another antibiotic often used as off-label salvage therapy in systemic VRE infections is oritavancin, a semisynthetic glycopeptide that has demonstrated synergic activity with fosfomycin. [22]

History

High-level vancomycin-resistant E. faecalis and E. faecium are clinical isolates first documented in Europe in 1986 and the United States in 1987. [23] [24] In the United States, vancomycin-resistant E. faecium was associated with 4% of healthcare-associated infections reported to the Centers for Disease Control and Prevention National Healthcare Safety Network from January 2006 to October 2007. [25] VRE can be carried by healthy people who have come into contact with the bacteria, usually in a hospital [26] (nosocomial infection), [27] although it is thought that a significant percentage of intensively farmed chickens also carry VRE. [28] Other regions have noted a similar distribution, but with increased incidence of VRE. For example, a 2006 study of nosocomial VRE revealed a rapid spread of resistance among enterococci along with an emerging shift in VRE distribution in the Middle East region, such as Iran. Treatment failures in enterococcal infections result from inadequate information regarding glycopeptide resistance of endemic enterococci due to factors such as the presence of VanA and VanB. The study from Iran reported the first case of VRE isolates that carried VanB gene in enterococcal strains from Iran. This study also noted the first documented isolation of nosocomial E. raffinosus and E. mundtii in the Middle East region. [29]

See also

Related Research Articles

<span class="mw-page-title-main">Ampicillin</span> Antibiotic

Ampicillin is an antibiotic belonging to the aminopenicillin class of the penicillin family. The drug is used to prevent and treat a number of bacterial infections, such as respiratory tract infections, urinary tract infections, meningitis, salmonellosis, and endocarditis. It may also be used to prevent group B streptococcal infection in newborns. It is used by mouth, by injection into a muscle, or intravenously.

<span class="mw-page-title-main">Vancomycin</span> Antibiotic medication

Vancomycin is a glycopeptide antibiotic medication used to treat a number of bacterial infections. It is used intravenously as a treatment for complicated skin infections, bloodstream infections, endocarditis, bone and joint infections, and meningitis caused by methicillin-resistant Staphylococcus aureus. Blood levels may be measured to determine the correct dose. Vancomycin is also taken orally as a treatment for severe Clostridium difficile colitis. When taken orally it is poorly absorbed.

<i>Enterococcus</i> Genus of bacteria

Enterococcus is a large genus of lactic acid bacteria of the phylum Bacillota. Enterococci are gram-positive cocci that often occur in pairs (diplococci) or short chains, and are difficult to distinguish from streptococci on physical characteristics alone. Two species are common commensal organisms in the intestines of humans: E. faecalis (90–95%) and E. faecium (5–10%). Rare clusters of infections occur with other species, including E. casseliflavus, E. gallinarum, and E. raffinosus.

Methicillin-resistant <i>Staphylococcus aureus</i> Bacterium responsible for difficult-to-treat infections in humans

Methicillin-resistant Staphylococcus aureus (MRSA) is a group of gram-positive bacteria that are genetically distinct from other strains of Staphylococcus aureus. MRSA is responsible for several difficult-to-treat infections in humans. It caused more than 100,000 deaths worldwide attributable to antimicrobial resistance in 2019.

<span class="mw-page-title-main">Linezolid</span> Antibiotic medication

Linezolid is an antibiotic used for the treatment of infections caused by Gram-positive bacteria that are resistant to other antibiotics. Linezolid is active against most Gram-positive bacteria that cause disease, including streptococci, vancomycin-resistant enterococci (VRE), and methicillin-resistant Staphylococcus aureus (MRSA). The main uses are infections of the skin and pneumonia although it may be used for a variety of other infections including drug-resistant tuberculosis. It is used either by injection into a vein or by mouth.

Vancomycin-resistant <i>Staphylococcus aureus</i> Antibiotica resistant bacteria

Vancomycin-resistant Staphylococcus aureus (VRSA) are strains of Staphylococcus aureus that have acquired resistance to the glycopeptide antibiotic vancomycin. Bacteria can acquire resistant genes either by random mutation or through the transfer of DNA from one bacterium to another. Resistance genes interfere with the normal antibiotic function and allow a bacteria to grow in the presence of the antibiotic. Resistance in VRSA is conferred by the plasmid-mediated vanA gene and operon. Although VRSA infections are uncommon, VRSA is often resistant to other types of antibiotics and a potential threat to public health because treatment options are limited. VRSA is resistant to many of the standard drugs used to treat S. aureus infections. Furthermore, resistance can be transferred from one bacterium to another.

<i>Enterococcus faecalis</i> Species of bacterium

Enterococcus faecalis – formerly classified as part of the group D Streptococcus system – is a Gram-positive, commensal bacterium inhabiting the gastrointestinal tracts of humans. Like other species in the genus Enterococcus, E. faecalis is found in healthy humans and can be used as a probiotic. The probiotic strains such as Symbioflor1 and EF-2001 are characterized by the lack of specific genes related to drug resistance and pathogenesis. As an opportunistic pathogen, E. faecalis can cause life-threatening infections, especially in the nosocomial (hospital) environment, where the naturally high levels of antibiotic resistance found in E. faecalis contribute to its pathogenicity. E. faecalis has been frequently found in reinfected, root canal-treated teeth in prevalence values ranging from 30% to 90% of the cases. Re-infected root canal-treated teeth are about nine times more likely to harbor E. faecalis than cases of primary infections.

<span class="mw-page-title-main">Oritavancin</span> Pharmaceutical drug

Oritavancin, sold under the brand name Orbactiv among others, is a semisynthetic glycopeptide antibiotic medication for the treatment of serious Gram-positive bacterial infections. Its chemical structure as a lipoglycopeptide is similar to vancomycin.

Enterococcus faecium is a Gram-positive, gamma-hemolytic or non-hemolytic bacterium in the genus Enterococcus. It can be commensal in the gastrointestinal tract of humans and animals, but it may also be pathogenic, causing diseases such as neonatal meningitis or endocarditis.

<span class="mw-page-title-main">Ceftobiprole</span> Chemical compound

Ceftobiprole, sold under the brand name Zevtera among others, is a fifth-generation cephalosporin antibacterial used for the treatment of hospital-acquired pneumonia and community-acquired pneumonia. It is marketed by Basilea Pharmaceutica under the brand names Zevtera and Mabelio. Like other cephalosporins, ceftobiprole exerts its antibacterial activity by binding to important penicillin-binding proteins and inhibiting their transpeptidase activity which is essential for the synthesis of bacterial cell walls. Ceftobiprole has high affinity for penicillin-binding protein 2a of methicillin-resistant Staphylococcus aureus strains and retains its activity against strains that express divergent mecA gene homologues. Ceftobiprole also binds to penicillin-binding protein 2b in Streptococcus pneumoniae (penicillin-intermediate), to penicillin-binding protein 2x in Streptococcus pneumoniae (penicillin-resistant), and to penicillin-binding protein 5 in Enterococcus faecalis.

<span class="mw-page-title-main">Dalbavancin</span> Antibiotic used to treat MRSA

Dalbavancin, sold under the brand names Dalvance in the US and Xydalba in the EU among others, is a second-generation lipoglycopeptide antibiotic medication. It belongs to the same class as vancomycin, the most widely used and one of the treatments available to people infected with methicillin-resistant Staphylococcus aureus (MRSA).

Enterococcus gallinarum is a species of Enterococcus. E. gallinarum demonstrates an inherent, low-level resistance to vancomycin. Resistance is due to a chromosomal gene, vanC, which encodes for a terminal D-alanine-D-serine instead of the usual D-alanine-D-alanine in cell wall peptidoglycan precursor proteins. That is a separate mechanism than the vancomycin resistance seen in VRE isolates of E. faecium and E. faecalis which is mediated by vanA or vanB. This species is known to cause clusters of infection, although it considered very rare. It is the only other known enterococcal species besides E. faecium and E. faecalis known to cause outbreaks and spread in hospitals.

<span class="mw-page-title-main">Sophoraflavanone G</span> Chemical compound

Sophoraflavanone G is a volatile phytoncide, released into the atmosphere, soil and ground water, by members of the Sophora genus. Sophora pachycarpa, and Sophora exigua; all found to grow within the United States in a variety of soil types, within temperate conditions, no lower than 0 °F. Sophoraflavanone G is released in order to protect the plant against harmful protozoa, bacteria, and fungi. Sophoraflavanone G, also called kushenin, is a flavonoid compound.

In molecular biology, VanY are protein domains found in enzymes named metallopeptidases. They are vital to bacterial cell wall synthesis and antibiotic resistance.

Enterococcus malodoratus is a species of the genus Enterococcus and a gram positive bacteria capable of opportunistic pathogenic response. These microbes have a thick polypeptide layer. Enterococcus can be found in the gastrointestinal tracts of humans and other mammals. In a study on the enterococcal flora of swine, E. malodoratus was found in the intestines and feces. It was not identified within the tonsils of swine, nor within cats, calves, dogs, horse, or poultry. The name "malodoratus" translates to "ill smelling".

ESKAPE is an acronym comprising the scientific names of six highly virulent and antibiotic resistant bacterial pathogens including: Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp. The acronym is sometimes extended to ESKAPEE to include Escherichia coli. This group of Gram-positive and Gram-negative bacteria can evade or 'escape' commonly used antibiotics due to their increasing multi-drug resistance (MDR). As a result, throughout the world, they are the major cause of life-threatening nosocomial or hospital-acquired infections in immunocompromised and critically ill patients who are most at risk. P. aeruginosa and S. aureus are some of the most ubiquitous pathogens in biofilms found in healthcare. P. aeruginosa is a Gram-negative, rod-shaped bacterium, commonly found in the gut flora, soil, and water that can be spread directly or indirectly to patients in healthcare settings. The pathogen can also be spread in other locations through contamination, including surfaces, equipment, and hands. The opportunistic pathogen can cause hospitalized patients to have infections in the lungs, blood, urinary tract, and in other body regions after surgery. S. aureus is a Gram-positive, cocci-shaped bacterium, residing in the environment and on the skin and nose of many healthy individuals. The bacterium can cause skin and bone infections, pneumonia, and other types of potentially serious infections if it enters the body. S. aureus has also gained resistance to many antibiotic treatments, making healing difficult. Because of natural and unnatural selective pressures and factors, antibiotic resistance in bacteria usually emerges through genetic mutation or acquires antibiotic-resistant genes (ARGs) through horizontal gene transfer - a genetic exchange process by which antibiotic resistance can spread.

Kerry L. LaPlante is an American pharmacist, academic and researcher. She is the Dean at the University of Rhode Island College of Pharmacy. She is a Professor of Pharmacy and former department Chair of the Department of Pharmacy Practice at the University of Rhode Island, an Adjunct Professor of Medicine at Brown University, an Infectious Diseases Pharmacotherapy Specialist, and the Director of the Rhode Island Infectious Diseases Fellowship and Research Programs at the Veterans Affairs Medical Center in Providence, Rhode Island.

<span class="mw-page-title-main">Antimicrobial nanotechnology</span>

Antimicrobial nanotechnology is the study of using biofilms to disrupt a microbe's cell membrane, deliver an electric charge to the microbe, and cause immediate cellular death via a "mechanical kill" process, preventing the original microbe from mutating into a superbug.

References

  1. 1 2 3 "Vancomycin-resistant Enterococci (VRE) and the Clinical Laboratory". Centers for Disease Control and Prevention. Archived from the original on 14 April 2019. Retrieved 21 May 2017.PD-icon.svg This article incorporates text from this source, which is in the public domain .
  2. 1 2 Balli EP, Venetis CA, Miyakis S (2014). "Systematic Review and Meta-Analysis of Linezolid versus Daptomycin for Treatment of Vancomycin-Resistant Enterococcal Bacteremia". Antimicrobial Agents and Chemotherapy. 58 (2): 734–739. doi:10.1128/AAC.01289-13. ISSN   0066-4804. PMC   3910884 . PMID   24247127.
  3. "Vancomycin-resistant Enterococci (VRE) in Healthcare Settings". VRE in Healthcare Settings - HAI. CDC. Archived from the original on 2019-05-18. Retrieved 2015-06-09.
  4. Fong IW, Drlica K (2007-11-15). Antimicrobial Resistance and Implications for the 21st Century. Springer Science & Business Media. ISBN   9780387724188. Archived from the original on 2023-01-14. Retrieved 2020-12-17.
  5. Mendez-Vilas A (2011). Science and Technology Against Microbial Pathogens: Research, Development and Evaluation, Proceedings of the International Conference on Antimicrobial Research (ICAR2010), Valladolid, Spain 3 - 5 November 2010. World Scientific. ISBN   9789814354868.
  6. Ahmad I, Aqil F (2008-11-21). New Strategies Combating Bacterial Infection. John Wiley & Sons. ISBN   9783527622948.
  7. Grayson ML, Crowe SM, McCarthy JS, Mills J, Mouton JW, Norrby SR, Paterson DL, Pfaller MA (2010-10-29). Kucers' The Use of Antibiotics Sixth Edition: A Clinical Review of Antibacterial, Antifungal and Antiviral Drugs. CRC Press. ISBN   9781444147520. Archived from the original on 2023-01-14. Retrieved 2020-12-17.
  8. Arias CA, Murray BE (2012). "Figure 4 : The rise of the Enterococcus: beyond vancomycin resistance : Nature Reviews Microbiology". Nature Reviews Microbiology. 10 (4): 266–278. doi:10.1038/nrmicro2761. PMC   3621121 . PMID   22421879.
  9. Meziane-Cherif D, Saul FA, Haouz A, Courvalin P (2012). "Structural and Functional Characterization of VanG d-Ala:d-Ser Ligase Associated with Vancomycin Resistance in Enterococcus faecalis♦". The Journal of Biological Chemistry. 287 (45): 37583–37592. doi: 10.1074/jbc.M112.405522 . ISSN   0021-9258. PMC   3488035 . PMID   22969085.
  10. Coates AR (2012-08-31). Antibiotic Resistance. Springer Science & Business Media. ISBN   9783642289507. Archived from the original on 2023-01-14. Retrieved 2020-12-17.
  11. Gould D, Brooker C (2008-08-20). Infection Prevention and Control: Applied Microbiology for Healthcare. Palgrave Macmillan. ISBN   9781137045928.[ permanent dead link ]
  12. Monticelli J, et al. (2018). "Clinical management of non-faecium non-faecalis vancomycin-resistant enterococci infection. Focus on Enterococcus gallinarum and Enterococcus casseliflavus/flavescens". J Infect Chemother. 24 (4): 237–246. doi: 10.1016/j.jiac.2018.01.001 . PMID   29396199.
  13. "VRE and the Clinical Laboratory - HAI". Healthcare-associated Infections. CDC. Archived from the original on 2019-04-14. Retrieved 2015-06-09.
  14. Levitus M, Rewane A, Perera TB (2022). "Vancomycin-Resistant Enterococci". StatPearls. StatPearls Publishing. PMID   30020605. Archived from the original on 27 March 2022. Retrieved 27 March 2022.
  15. Zirakzadeh A, Patel R (April 2006). "Vancomycin-resistant enterococci: colonization, infection, detection, and treatment". Mayo Clin Proc. 81 (4): 529–36. doi:10.4065/81.4.529. PMID   16610573. Archived from the original on 2022-09-11. Retrieved 2022-09-11. The LightCycler instrument (Roche Diagnostics Corporation) is used to detect vanA and vanB using a rapid real-time PCR assay(Figure 3). This method is more sensitive and faster (~3.5 vs >72 hours) than culture for detecting VRE colonization.47 The assay detects the presence of genes associated with vancomycin resistance in enterococci, vanA and vanB.
  16. Holzknecht BJ, Hansen DS, Nielsen L, Kailow A, Jarløv JO (March 2017). "Screening for vancomycin-resistant enterococci with Xpert® vanA/vanB: diagnostic accuracy and impact on infection control decision making". New Microbes New Infect. 16: 54–59. doi:10.1016/j.nmni.2016.12.020. PMC   5295639 . PMID   28203378.
  17. McKinnell JA, Kunz DF, Chamot E, Patel M, Shirley RM, Moser SA, Baddley JW, Pappas PG, Miller LG (2012). "Association of Vancomycin-Resistant Enterococcus Bacteremia and Ceftriaxone Usage". Infection Control and Hospital Epidemiology. 33 (7): 718–724. doi:10.1086/666331. ISSN   0899-823X. PMC   3879097 . PMID   22669234.
  18. Phoenix DA, Harris F, Dennison SR (2014-08-25). Novel Antimicrobial Agents and Strategies. John Wiley & Sons. ISBN   9783527676156.
  19. Werth BJ, Barber KE, Tran N, Nonejuie P, Sakoulas G, Pogliano J, Rybak MJ (2015-02-01). "Ceftobiprole and ampicillin increase daptomycin susceptibility of daptomycin-susceptible and -resistant VRE". Journal of Antimicrobial Chemotherapy. 70 (2): 489–493. doi: 10.1093/jac/dku386 . ISSN   0305-7453. PMID   25304643. Archived from the original on 2022-05-08. Retrieved 2023-10-25.
  20. de Oliveira Santos JV, da Costa Júnior SD, de Fátima Ramos Dos Santos Medeiros SM, Cavalcanti ID, de Souza JB, Coriolano DL, da Silva WR, Alves MH, Cavalcanti IM (April 2022). "Panorama of Bacterial Infections Caused by Epidemic Resistant Strains". Current Microbiology. 79 (6): 175. doi:10.1007/s00284-022-02875-9. PMC   9055366 . PMID   35488983.
  21. Riccardi N, Monticelli J, Antonello RM, Di Lallo G, Frezza D, Luzzati R, Di Bella S (2021-04-01). "Therapeutic Options for Infections Due to vanB Genotype Vancomycin-Resistant Enterococci". Microbial Drug Resistance. 27 (4): 536–545. doi:10.1089/mdr.2020.0171. ISSN   1076-6294. PMID   32799629. S2CID   221144533.
  22. Di Cecco C, Monticelli J, Di Bella S, Di Maso V, Luzzati R (2023-08-21). "Vancomycin-resistant enterococcus bloodstream infection successfully managed with oritavancin and fosfomycin as sequential treatment". Journal of Chemotherapy. 36 (1): 31–34. doi:10.1080/1120009X.2023.2247205. ISSN   1120-009X. PMID   37602423. S2CID   261048377. Archived from the original on 2023-08-21. Retrieved 2023-10-25.
  23. Robinson DA, Feil EJ, Falush D (2010-03-16). Bacterial Population Genetics in Infectious Disease. John Wiley & Sons. ISBN   9780470600115.
  24. Murray BE (March 2000). "Vancomycin-resistant enterococcal infections". N Engl J Med. 342 (10): 710–21. doi:10.1056/NEJM200003093421007. PMID   10706902. Archived from the original on 2022-09-11. Retrieved 2022-09-11. The first reports of vancomycin-resistant enterococci (later classified as VanA type of resistance) involved strains of E. faecium that were resistant to vancomycin and teicoplanin (another glycopeptide) and that were isolated from patients in France and England in 1986. Vancomycin-resistant E. faecalis, subsequently classified as VanB type, was recovered from patients in Missouri in 1987.
  25. Hidron AI, Edwards JR, Patel J, et al. (November 2008). "NHSN annual update: antimicrobial-resistant pathogens associated with healthcare-associated infections: annual summary of data reported to the National Healthcare Safety Network at the Centers for Disease Control and Prevention, 2006-2007". Infect Control Hosp Epidemiol. 29 (11): 996–1011. doi:10.1086/591861. PMID   18947320. S2CID   205988392.
  26. "Diseases and Organisms in Healthcare Settings". Healthcare-associated Infections (HAIs). CDC. Archived from the original on 2015-07-08. Retrieved 2015-06-09.
  27. Kouchak F, Askarian M (2012). "Nosocomial Infections: The Definition Criteria". Iranian Journal of Medical Sciences. 37 (2): 72–73. PMC   3470069 . PMID   23115435.
  28. Nilsson O (2012). "Vancomycin resistant enterococci in farm animals – occurrence and importance". Infection Ecology & Epidemiology. 2 (1): 16959. Bibcode:2012InfEE...216959N. doi:10.3402/iee.v2i0.16959. PMC   3426332 . PMID   22957131.
  29. Fathollahzadeh B, Hashemi FB, Emaneini M, Aligholi M, Nakhjavani FA (2006). "Frequency of Vancomycin-Resistant Enterococci from Three Hospitals in Iran". Daru. 14 (3): 141–146. Archived from the original (PDF) on 2016-03-04. Retrieved 2015-12-30.

Further reading