Open access peer-reviewed chapter

Molecular Pathogenesis and Clinical Impact of Biofilms in Surgery

Written By

Roger Bayston

Submitted: 25 February 2022 Reviewed: 16 March 2022 Published: 22 April 2022

DOI: 10.5772/intechopen.104526

From the Edited Volume

Focus on Bacterial Biofilms

Edited by Theerthankar Das

Chapter metrics overview

140 Chapter Downloads

View Full Metrics

Abstract

Biofilms are responsible for chronic persistent infections and are a major problem in implant surgery. The microbial pathogenesis, treatment and prevention of biofilm infections is reviewed.

Keywords

  • biofilm infections
  • biofilm phenotype
  • small colony variants
  • prevention of biofilm infections

1. Introduction

Though the “discovery” of biofilms is ascribed to Anton van Leeuwenhoek in 1676 using a novel magnifying device, and possibly to Robert Hooke two decades earlier, and biofilms were recognised in a marine setting about a century ago, they were of no medical interest until two studies described them in a medical device and in sputum in 1972 and 1974 respectively. The latter was a description of aggregates of Pseudomonas aeruginosa in secretions from the lungs of people with cystic fibrosis [1], and led to a burgeoning of research into Ps aeruginosa infection in that field. Through a meeting with Costerton, Højby studied these aggregates and the term “Biofilm” was made popular by Costerton in 1987 [2], though the term was originally used by Mack et al. [3] to describe “biofilm” on a water filter. However, many biofilm infections occur in association with implanted materials and devices, and their use has become much more common since the middle 1900’s. The first biofilm reported in a medical device was found in a shunt to treat hydrocephalus in 1972 [4]. This discovery explained the difficulty in successfully treating these infections non-surgically with antibiotics alone, and the report demonstrated the extracellular matrix of the biofilm in vitro and in vivo and carried out investigations to suggest that it was a glycosaminoglycan. This was later confirmed by important studies in 1996 [5]. Implantable biomaterials and devices are now widely used in modern surgery, and the list is extensive (Figures 1 and 2).

Figure 1.

Examples of implantable devices.

Figure 2.

Anatomical sites of common implantable devices.

Advertisement

2. Biofilm definitions

Many definitions of “biofilm” are found in the literature, and they can be based on either structure or function. Many of the definitions and their accompanying images are derived from in vitro models, and the appearance of mushroom-like structures and water-channels are not seen in biofilms occurring in vivo [6]. A definition based on functional aspects of biofilms is more useful in a medical context. This could be reduced to a population of bacteria or other micro-organisms, often associated with a surface, and enveloped in an extracellular matrix, showing insusceptibility to antimicrobials and to the host immune system, and ability to persist for long periods.

2.1 Biofilm phenotypes

The basis of this functional definition is the paucity of nutrients, including iron, and oxygen in the depths of the biofilm leading to a bacterial stress response caused by a crisis in energy generation and transport [7]. The bacterial stress response is mediated by the intracellular signal sigma-B. The bacterial response to this is to downregulate all synthetic functions not needed in biofilm mode, such as cell wall material, toxin and other non-essential protein synthesis, and DNA replication. These are the targets for common antibiotics, and beta-lactams, glycopeptides, aminoglycosides, macrolides and fluoroquinolones all become significantly less effective against biofilm bacteria. Other factors contribute to the lack of effect of antibiotics, including a slowing of their penetration into the biofilm, though this is rarely a major factor. The bacterial stress response results in significantly reduced cell metabolic activity and loss of some synthetic activities leading to auxotrophy for heme and menadione, and sometimes other substances such as thymidine. This biofilm phenotype is crucial to the clinical impact of biofilm infections; the colonies of biofilm bacteria when grown from clinical samples in the laboratory are typically less than ten times the size of their planktonic counterparts, and are known as small colony variants or SCV. The molecular control and regulation of biofilm phenotype has been described in detail by Proctor et al. [8]. SCV are important in biofilm infections not only because their metabolism leads to antibiotic insusceptibility, but because, though they can be internalised by professional and non-professional phagocytes, they are not killed and survive inside the phagocytic cells. Auxotrophic SCV of Staphylococcus aureus for heme and menadione, that do not produce alpha-toxin, are more able to survive intracellularly, and supplementation of intracellular populations of S aureus in vitro with menadione resulted in restoration of alpha-toxin production and reduced intracellular survival [9, 10]. SCV are not always auxotrophic and considerable variation occurs, but intracellular survival is a common feature. Many also show reduced susceptibility to aminoglycosides, and exposure to gentamicin can induce SCV formation [11]. Some SCV are the result of mutations in the genes concerned with electron transport, and these do not revert to parent forms whereas other forms of SCV appear to be phenotypic variants that revert to parent forms when the stress factor is withdrawn [8, 12]. SCV of gram negative bacteria have been known for decades, having been produced in the laboratory from exposure to antibacterial chemicals [13, 14]. However, more recently capnophilic (carbon dioxide—dependent) SCV of Escherichia coli have been isolated from a patient with a urinary tract infection, though no information on biofilm involvement was given [15]. A report of septic shock in a patient from whose urine capnophilic Proteus mirabilis SCV were isolated again did not state that biofilms were involved [16] but the patient had chronic renal stones, known to be associated with biofilms [17]. P mirabilis is an important uropathogen as it is highly motile and is capable of enzymatically hydrolysing urea into ammonia, thus being highly inflammatory as well as alkalinising the urine. The rising pH causes crystallisation of calcium and magnesium phosphates [18], and the P mirabilis biofilm typically consists of a mesh of bacteria, their extracellular matrix and phosphate crystals. These biofilms are obviously different in composition from those consisting mainly of bacteria and their products, and another example of such complex biofilms is the vegetations found in native valve endocarditis. Here the lesion consists largely of a matrix of platelets and fibrin, with bacteria, usually viridans streptococci, embedded in it. The lesion usually begins as a response to damage to the endocardium, which is then colonised by bacteria from the bloodstream, becoming progressively built up of fibrin and platelets with rafts of bacteria interspersed [19, 20]. A similar situation arises with prosthetic heart valves. In both cases, SCVs have been reported [21, 22] as well as other auxotrophic variants [23].

The biofilm phenotype, and SCV in particular, are important in treatment of biofilm infections. Surviving intracellular bacteria are protected from further immune assault and from most therapeutic antibiotics, which do not accumulate inside host cells sufficiently to kill SCV [24]. These factors mean that the amount of antibiotic required to kill bacteria in biofilm mode is typically 500–1000 times the minimum inhibitory concentration as measured in the clinical laboratory. Such concentrations are not achievable by intravenous or oral therapy, and eradication of biofilm infection usually requires extensive surgery to debride the site and to remove all surgical hardware.

2.2 Biofilm development

Development of biofilms in surgery depends on a sequence of events. Initially, the causative bacteria must be able to gain access to the site of biofilm formation, usually an implantable device. In modern surgery most device pathogens originate on the patient’s skin or mucous membranes, consisting mainly of coagulase-negative staphylococci (CoNS), typically Staphylococcus epidermidis, and Cutibacterium acnes. Conventional pre-operative skin preparation reduces but does not eradicate these bacteria, and the importance of relatively small numbers of bacteria in the operation field has been shown by an experiment in human volunteers, where various “doses” of S aureus were inoculated into incisions to determine how many bacteria were necessary to produce an abscess [25]. In one group, “foreign” material in the form of sutures were also introduced into the incision, and the number of bacteria required to form an abscess in those cases was 10,000 times fewer. This study, which is unlikely to be repeated in a modern setting, is extremely important in illustrating the role played by implantable materials and devices in infection in modern surgery.

The sequence of events involved in development of a biofilm infection involving a surgically implanted device are (Figure 3):

Access to the device from the source. Though heavy contamination of the air in the operating environment has historically been associated with surgical infection, modern operating room design and ventilation has meant that this source has declined in importance, and most surgical infections are caused by bacteria originating on the patient’s skin or mucous membranes. Bacteria reach the incision from the cut edges of the skin, or from contamination from surrounding skin surfaces, during surgery. The causative bacteria are therefore often present when the device is implanted.

Attachment to the device. Many bacteria possess adhesins on their surfaces that allow them to attach to biomaterials (vitronectin—binding protein etc) but more often they employ specific adhesins for the glycoproteins, platelets and other host-derived materials that rapidly coat all implanted materials [26, 27]. S aureus possesses specific adhesins for fibrinogen, fibronectin, laminin, thrombospondin, bone sialoprotein and other host-derived components of the conditioning film. These bacterial surface adhesins are known as MSCRAMMs (Microbial Surface Component Recognising Adhesive Matrix Molecules) [28] and they can be found in other organisms such as S epidermidis and enterococci [29]. Gram negative bacteria often attach by means of swarming or twitching motility over the new surface [30], some using twitching motility by Type IV pili [31, 32], and this might be particularly important in biofilm formation on urinary catheters. In addition, Ps aeruginosa uses a von Willebrand Factor-like surface factor in twitching motility over biomaterial surfaces [33].

Once bacteria have attached to the surface or conditioning film, they begin to proliferate and to develop intercellular adhesins such as polysaccharide intercellular adhesin (PIA) in staphylococci. This substance is integral to further development of biofilm, and is encoded by the ABDC operon, and regulated by icaR. At this stage, bacterial stress responses are operating in response to limitation of nutrients and oxygen and the biofilm phenotype is appearing [34]. It is important to note that the bacterial stress response, mediated by Sigma B, downregulates icaR and increases PIA production, and the stress response can be provoked by external factors such as antibiotics as well as nutrient starvation. Once the biofilm phenotype has developed, the biofilm is stable and is not susceptible to host immune activity or to antimicrobials. There is often a lag phase of about 14–28 days before the biofilm reaches functional maturity, during which it might be more susceptible to antimicrobials [35].

Figure 3.

Sequence of events in development of biofilm infection. Here implant has an antimicrobial coating, but within minutes this is covered by a glycoprotein conditioning film produced by the patient. This usually prevents the activity of the coating and bacteria now adhere to the conditioning film. Within a few hours the attached bacteria begin to produce an extracellular matrix and to multiply. Powerful antibacterial activity is essential now, as after this point, it is almost inevitable that a biofilm will develop, within a few weeks.

Clear understanding of the sequence of events and periods of risk is essential for effective planning of preventative measures.

Advertisement

3. Prevention of biofilm infections

3.1 Surgical considerations

Since the days of Semmelweis, Lister and others in the mid–to late 1800s, personal hygiene of the surgeon, aseptic technique and antisepsis have become accepted norms. Since the 1950s, when bacteria-laden operating room air was identified as a major factor in surgical infection [36], greatly improved practices and ventilation systems have made this a minor source. Two main forms of ventilation are in use in modern operating rooms: plenum, and laminar flow with high efficiency particulate air (HEPA) filtration. While it is clear that the numbers of airborne bacteria are significantly reduced when laminar flow is used [37] there has never been a clear causative link between either this reduction or the actual bacteria and surgical infection, leading the USA CDC to downgrade their initial recommendation [38]. More recently, reports have appeared of small but significantly increased infection rates when laminar flow is used [39, 40] and this appears to be due to flaws in its design and manner of use [41]. For most types of implant surgery, plenum (conventional) ventilation appears to be satisfactory so long as other precautions are taken (Figure 4).

Figure 4.

Sequence of surgical preventative events.

Care bundles have been proposed for infection reduction in various healthcare settings. A bundle is a collection of interventions that are expected to contribute to reduced risk of infection, but which singly might have weak or no evidence base. A measure such as ensuring that only three people are present in the operating room during a procedure is not supported by any clear evidence but it is intuitively likely to be beneficial if only in reinforcing operating room discipline. A bundle must be directed towards behaviour change on the part of relevant staff members, and it works best if they contribute to its content, and formally agree to abide by it. Some bundles insist on contents being evidence-based, but the quality of evidence is usually very weak for individual components. However, when bundles are properly applied, they are often very effective in reducing surgical infection [42, 43] and in any case they and their contents should form part of a well-managed surgical discipline. Usually no single component can be identified to explain their success, but clinical trial evidence has shown that violations of the bundle are associated with re-emergence of infection [43].

As the major source of pathogens is the patient’s skin, attention has been directed towards the effectiveness of preoperative skin preparation. Two main antiseptics are in use: chlorhexidine and povidone iodine. Each can be formulated in water or 70% alcohol. A report by the World Health Organisation (WHO) favouring chlorhexidine [44] has been called into question on the basis of quality of evidence [45]. However, sampling is usually by swabbing of the skin surface, and almost none of the many studies on surgical skin preparation explore the effectiveness of any agent on bacteria resident in the dermis, though an early study showed that full thickness skin biopsy was necessary [46]. This has since been confirmed [47, 48]. When skin biopsy is used, neither antiseptic in alcohol is able to eradicate resident skin bacteria, and though reduced, the remaining numbers are often sufficient to cause a biomaterial-associated infection [25]. Two studies on the penetration of both aqueous and alcoholic chlorhexidine into human skin using full thickness biopsy have found it to be minimal [49, 50]. Further measures are therefore necessary. Some researchers have investigated the effect of antiseptic-soaked material to protect the incision from the skin edges during surgery, and while this is commonly used, there have been no quantitative studies to show benefit. Intravenous antibiotics are almost universally used in surgery, ideally as a single dose 30–60 min before incision, but extra doses are commonly used postoperatively though they offer no benefit over that of the single pre-operative dose. Antibiotic prophylaxis is undoubtedly highly effective in reducing infection risk in many types of surgery, including colorectal surgery [51] and orthopaedic surgery [52] but probably less so in neurosurgery due to limited penetration of systemic antibiotics intracranially. However, it is probably inevitable that a small number of bacteria will reach the implant during operation, and further measures have been directed to attempts to eradicate these. As knowledge of attached bacteria and biofilms has shown that very high concentrations of antibiotics are necessary, some surgeons have used either antiseptic or antibiotic irrigation [53, 54], or have simply added antibiotic powder to the incision before closure [55, 56, 57] with successful reduction in infection rates and complications. This intervention gives extremely high local antibiotic levels not reachable by systemic administration, yet avoids most of the complications associated with the latter method.

3.2 Antimicrobial biomaterials

Other methods of prevention accept that despite efforts, bacteria will reach the implant, and aim to prevent their attachment or to kill them when attached. Various “anti-fouling” surfaces have been investigated with the aim of allowing host cell and tissue proliferation but preventing bacterial attachment [58, 59] but none of these has yet reached clinical application, largely because of the complex relationship between implant surface, host tissue environment, and bacterial surface adhesins. Biomaterials designed to kill bacteria that do attach to them have generally included coatings of silver, antiseptic or antibiotic and combinations of these, often with a vehicle to bind the antimicrobial to the biomaterial surface. Such coatings have several disadvantages. The normal host reaction to the implant of deposition of plasma proteins [26, 27] also obliterates the antimicrobial coating in many cases, making it ineffective. Silver is susceptible to this due its avidity for proteins [60], and it can also be inactivated by chloride [61] which is abundant in the human body. Silver ions have also been shown to be cytotoxic in certain conditions [62]. Clinical studies on silver-processed devices give very variable results, and there is doubt about their cost-effectiveness in wound dressings [63]. A recent randomised controlled trial of silver-containing catheters intended to reduce ventriculitis in people with hydrocephalus shunts found no difference from plain catheters [64]. Another randomised controlled trial of silver-processed urinary catheters again found no significant difference from plain catheters [65]. In both of these clinical settings, biofilms play a key role, and the goal is to prevent bacterial proliferation and biofilm development on the catheters. Both have fluid containing proteins and chloride flowing through them.

Another approach has been impregnation of catheter material with antimicrobials. Though the impregnation processes differ, two catheter types can be considered: those containing rifampicin and minocycline, and those containing rifampicin and clindamycin. The first type has been used in central venous catheters [66] and external ventricular drains [67]. The second type has been used in hydrocephalus shunts and external ventricular drains. In all cases they have shown effectiveness in reducing device -related infection. The advantage of impregnation over coatings is that they give a long duration of activity: coatings are usually washed away by fluid after a few days, whereas the surface of an impregnated material is continually replenished by migrating antimicrobials until the depot in the material is depleted, usually several weeks later (Figure 5). This is important when the implantable device is at risk of contamination for an extended period.

Figure 5.

Principle of impregnated biomaterial. Antimicrobial molecules are motile within the device matrix and can migrate to the surface to replace those removed by fluid flow.

3.3 Importance of source of infection and period of risk

In order to formulate an effective preventive strategy, knowledge of the source and nature of device pathogens and the period during which the device is at risk is essential (Table 1). As many biofilm infections are caused by micro-organisms originating in or on the patient, a knowledge of the distribution of these is useful. The normal bacterial flora of the skin differs according to age and sex, but particularly depending on the anatomical site. The most common bacteria found on the skin are staphylococci, particularly members of the CoNS. These are typified by S epidermidis which is broadly distributed over the body surfaces, but other species such as Staphylococcus capitis have preferred sites such as the head and neck. C acnes is an important pathogen in the context of implant infections, but it is a good example of the importance of specific topographical distribution in determining the important pathogens in particular implants. C acnes is found on the upper body and head (Figure 6) [68], and it is therefore not surprising that devices implanted in these areas show a significantly higher incidence of C acnes infection. Examples are neurosurgical shunts and drains [69, 70], spine instrumentation [71], breast implants [72] and shoulder arthroplasty [73, 74]. Implants in other sites such as urinary catheters are at risk from a different microbial profile, as the pathogens originate in the large intestine, and E coli, Klebsiella pneumoniae and P mirabilis are the most common.

Implant/deviceDuration of useMain source of pathogensPeriod of risk
At insertionDuring use
Hydrocephalus shuntindefinitePatient’s skin++
External ventricular drainFew days-weeksPatient’s skin/environment±++
Joint replacementIndefinitePatient’s skin++±
Urinary catheter 1<28 daysPatient/environment±++
Urinary catheter 2~90 daysPatient/environment±++
Peritoneal dialysis catheterIndefinitePatient/environment±++
Vascular graftIndefinitePatient+++
Prosthetic heart valveIndefinitePatient+++
Spinal instrumentationIndefinitePatient++±
Venous access deviceDays—monthsPatient/environment±++
SuturesDaysPatient/healthcare worker+±

Table 1.

Periods of risk of infection of common implantable devices.

Figure 6.

Topographical distribution of common biofilm pathogens (after Grice et al. [68]).

The time at which the implant is at risk of microbial contamination also varies. While there is always a risk at the time of implantation, in some implants this is the main time, and the risk of subsequent contamination is proportionally small. Examples of this are hydrocephalus shunts and joint replacements. In other implants the risk at insertion is significantly outweighed by that during use. Examples are external ventricular drains (EVD) for raised intracranial pressure, urinary catheters, venous access catheters and peritoneal dialysis catheters, all of which can be contaminated from environmental sources or from the hands of staff or users during use. Other examples are vascular grafts and prosthetic heart valves, which are at risk from hematogenous seeding from bacteria entering the bloodstream at a distant site.

When planning strategies for prevention of biofilm infections involving antimicrobials, it is therefore important to match the antimicrobial to the most likely pathogen(s). If systemic antimicrobial prophylaxis is contemplated, then the adverse effects of this must be taken into consideration if there is a need for prolonged administration due to extended period of risk. If antimicrobial materials or devices are to be used, these must address not only the likely pathogen(s) but also the duration of protective activity required.

International guidelines indicate that for most surgical procedures, any systemic antimicrobial prophylaxis should be administered as one dose 30–60 min before start of surgery [75, 76]. Extension of this prophylaxis beyond 24 hours does not reduce surgical infection further, but it does increase the incidence of acute kidney injury and Clostridioides difficile infection [77], which is a life-threatening colitis associated with over-use of antibiotics. Where the period of risk extends beyond the insertion procedure, such as in EVD, long courses of systemic antibiotics are often given until the drain is removed. This has been shown in some cases to reduce brain infections, but at a cost. A randomised study comparing the use of plain catheters and prolonged systemic antibiotics with antimicrobial-impregnated catheters and one dose of antibiotic at insertion found no difference in the brain infection rate, which was low in each group, but there were three cases of C difficile infection in the prolonged antibiotics group, one patient requiring total colectomy [78].

Advertisement

4. Treatment of biofilm infections

The difficulty in treating biofilm infections in surgery emphasises the importance of effective prevention. However, this is not always possible. The nature of the biofilm phenotype and its implications for antibiotic treatment mean that further surgery is almost inevitable, and this usually involves removal of the device. This might be relatively simple, as in the case of a venous access catheter or a urinary catheter, but it can be both surgically complicated and hazardous, as in the case of spinal instrumentation or prosthetic heart valves.

Attempts to eradicate established biofilm with antibiotics usually fail. A comparison of treatment regimens for hydrocephalus shunt infections showed that results with shunt removal and antibiotics were significantly superior to those with antibiotics alone [79]. Successful treatment of joint replacement infections relies on device removal and extensive debridement of infected tissue, with prolonged antibiotic therapy. However, understanding of biofilm biology has led to advances in this area. The biofilm phenotype takes a few weeks to “mature” to the point where full insusceptibility to antibiotics is expressed, and this has been exploited in development of a regimen for treatment of prosthetic joint infection when the diagnosis can be made within 3–4 weeks of insertion [80]. In this regimen, known as Debridement, Antibiotics and Implant Retention (DAIR), surgical treatment of the infected joint prosthesis is carried out on a planned basis after careful investigation to establish the causative micro-organism and its antimicrobial susceptibilities, to allow consultation with specialists including Microbiology/Infectious Diseases, and to determine that the implant is stable (Figure 7). Infections due to multi-drug-resistant bacteria, fungi or multiple bacteria are not suitable for this approach. During the operation, the prosthetic components are exposed and the acetabular module is removed, leaving the main metal prosthesis in place. All infected tissue is removed and samples are sent for microbiological examination. Copious irrigation with antiseptic is applied, and biodegradable antibiotic—eluting beads can be inserted to provide high local concentrations. The choice of antibiotic in the beads should be made in consultation with a microbiologist. The joint is then closed and a long postoperative course of suitable antibiotics is then started [81]. The success rate of DAIR compared to conventional full implant removal and replacement is slightly lower. Moreover, despite the very thorough surgical debridement and long courses of antibiotics, often for over a year, relapse can occur [82], illustrating the difficulty in eradication of biofilms. DAIR spares the patient the much more extensive surgical removal of the main implant components, and the second surgery to inert fresh implants a few weeks later.

Figure 7.

Possibility of retention of infected implant based on knowledge of biofilm phenotype maturation (based on Zimmerli and Trampuz, 2004) [80].

Advertisement

5. Diagnosis of biofilm infections

5.1 Clinical features

Most biofilm infections in surgery are chronic and persistent, sometimes for many years [83]. It is important to distinguish between “late infection,” implying an infection contracted long after surgery, such as hematogenously, and “delayed infection,” meaning that the infection appears long after surgery even though it was contracted at the operation. Delayed infection in spine instrumentation is usually due to infection with CoNS or C acnes [84, 85]. A similar situation is found in shoulder arthroplasty [86]. Generally, more virulent bacteria such as S aureus are associated with either early-presenting or with hematogenous infections. The delay of months or years between initial surgical implantation and appearance of symptoms [84] has led to doubt about the surgical origins of some infections but this has now been largely dispelled. However, the need for prolonged follow-up and vigilance must be emphasised.

Acute postoperative biofilm infections usually appear within days or weeks of surgery, with failure of wound healing, drainage of pus or other fluid from the wound, local pain and swelling, fever and general illness. Delayed or chronic infections of joint prostheses present with persistent pain and restricted mobility, local swelling and sometimes a sinus. In the absence of a sinus, diagnosis might be delayed as it is often difficult to distinguish infective from mechanical complications. Aspiration of synovial fluid often gives a diagnosis but sensitivity is low [87, 88]. Delayed infection in spine instrumentation similarly presents with persistent pain, tenderness and possibly a draining sinus. Delayed infections in hydrocephalus shunts are very uncommon now that the preferred route of drainage is to the abdomen (ventriculoperitoneal, VP), but the ventriculo-atrial (VA) route is still used in some cases. In VP shunts infection usually presents within a few months as it leads to obstruction, but this does not happen in VA shunts and symptoms might not appear, or at least become recognisable, for several years. During this time, bacteria are being discharged from the biofilm in the shunt into the bloodstream, and this might give rise to periods of ill-health or sporadic fevers. It also provokes production of antibodies to the bacteria, and eventually the concentrations of circulating antigen and antibody, and therefore immune complexes, become so high that they precipitate on basement membranes of joints, renal glomeruli, alveoli and microvascular system. The presenting clinical picture can therefore be a confusing array of disorders from hematuria, hemorrhagic skin rashes, arthropathy, and chronic cough [89, 90]. Clinical diagnosis can therefore be very difficult, and a high level of suspicion is needed. Aspiration of cerebrospinal fluid from the shunt often gives the diagnosis, but blood cultures can be negative in the later stages.

5.2 Laboratory methods

Depending on the site of the infection and presence of an implant, sometimes blood cultures are positive, indicating systemic spread of the infection, and risk of sepsis. Blood inflammatory markers such as erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP) levels are usually raised. Swab cultures from the wound might yield the infecting pathogen, but they might be misleading due to contamination [91]. Surgical exploration of the incision and deeper layers allows tissue samples to be taken and these are more likely to yield the pathogen(s). Such samples should always be taken during debridement surgery [92], using fresh instruments for each of up to six separate samples [81, 93]. In view of the anaerobic preference of C acnes and its slow growth, cultures should be incubated anaerobically for up to 10 days [94]. The way in which tissue samples are processed in the laboratory is important. Simply rubbing them on a culture plate or incubating them in a fluid culture is prone to contamination and gives poor yield, leading to under-diagnosis of infection. Tissue should be homogenised but the method of doing this is also important [95]. When hardware such as joint replacement or spinal instrumentation components are removed, these should be seen as valuable samples. Sonication to remove the biofilm has been shown to significantly increase the culture positivity rate [96, 97]. A further aid to laboratory diagnosis has been PCR [98] especially when applied to tissue homogenates or hardware sonicates. However, if PCR is used in an attempt to certify eradication of infection before re-insertion of a prosthesis, residual DNA from bacteria successfully killed by antibiotic therapy can give false positive results suggesting ongoing active infection. This can be overcome by use of a modified PCR method that detects DNA only from live bacteria [99].

Advertisement

6. Conclusions

The impact of biofilm infections in surgery on healthcare systems, economies and personal lives of patients is immense. The financial cost can only be estimated and published figures do not usually take into account “unseen” costs such as loss of earnings due to disability, increased dependency, and financial burden on carers.

The physical and mental trauma of surgery such as joint replacement, reconstructive breast implant or hydrocephalus treatment can be made unimaginably worse by postoperative biofilm infection.

The significant difficulty in successfully treating biofilm infections with antibiotics, due largely to the biofilm phenotype, is now well recognised, and the importance of commensal bacteria previously thought to be harmless, such as S epidermidis and C acnes, is becoming more widely known. However, surgical device removal remains the mainstay of treatment, and new approaches that allow implant retention are needed. Prevention of biofilm infections is crucial, and biomaterials that either reduce bacterial attachment, such as those coated with novel synthetic polymers [100] or those designed to kill bacteria on contact [66, 67] are now in clinical use. Many other biomaterials approaches are in development, and considerable strides have been made in this direction but further progress is being slowed by unrealistic commercial and regulatory barriers [101].

References

  1. 1. Høiby N. Pseudomonas aeruginosa infection in cystic fibrosis. Relationship between mucoid strains of Pseudomonas aeruginosa and the humoral immune response. Acta Pathologica et Microbiologica Scandinavica. 1974;82B:551-558
  2. 2. Costerton JW, Cheng K-J, Geesey GG, Ladd TI, Nickel JC, Dasgupta M, et al. Bacterial biofilms in nature and disease. Annual Reviews in Microbiology. 1987;41:435-464. DOI: 10.1146/annurev.mi.41.100187.002251
  3. 3. Mack WN, Mack JP, Ackerson AO. Microbial film development in a trickling filter. Microbial Ecology. 1975;2:215-226. DOI: 10.1007/BF02010441
  4. 4. Bayston R, Penny SR. Excessive production of mucoid substance in Staphylococcus SIIA: A possible factor in colonisation of Holter shunts. Developmental Medicine & Child Neurology. 1972;14(Suppl 27):25-28. DOI: 10.1111/j.1469-8749.1972.tb09769.x
  5. 5. Mack D, Fischer W, Krokotsch A, Leopold K, Hartmann R, Egge H, et al. The intercellular adhesin involved in biofilm accumulation of Staphylococcus epidermidis is a linear beta-1,6-linked glycosaminoglycan: Purification and structural analysis. Journal of Bacteriology. 1996;178:175-183. DOI: 10.1128/jb.178.1.175-183.1996
  6. 6. Bjarnsholt T, Alhede M, Alhede M, Eickhardt-Sørensen SR, Moser C, Kühl M, et al. The in vivo biofilm. Trends in Microbiology. 2013;21:466-474. DOI: 10.1016/j.tim.2013.06.002
  7. 7. Proctor R. Bacterial energetics and antimicrobial resistance. Drug Resistance Updates. 1998;1:227-235. DOI: 10.1016/s1368-7646(98)80003-4
  8. 8. Proctor RA, Kriegeskorte A, Kahl BC, Becker K, Löffler B, Peters G. Staphylococcus aureus small colony variants (SCVs): A road map for the metabolic pathways involved in persistent infections. Frontiers in Cellular and Infection Microbiology. 2014;4:1-8. DOI: 10.3389/fcimb.2014.00099
  9. 9. Vann JM, Proctor RA. Cytotoxic effects of ingested Staphylococcus aureus on bovine endothelial cells: Role of S aureus α-hemolysin. Microbial Pathogenesis. 1988;4:443-453. DOI: 10.1016/0882-4010(88)90029-0
  10. 10. Balwit JM, van Langevelde P, Vann JM, Proctor RA. Gentamicin-resistant menadione and hemin auxotrophic Staphylococcus aureus persist within cultured endothelial cells. The Journal of Infectious Diseases. 1994;170:1033-1037. DOI: 10.1093/infdis/170.4.1033
  11. 11. Pelletier LL, Richardson M, Feist M. Virulent gentamicin-induced small colony variants of Staphylococcus aureus. The Journal of Laboratory and Clinical Medicine. 1979;94:324-334
  12. 12. Vulin C, Leimer N, Huemer M, Ackerman M, Zinkernagel AS. Prolonged bacterial lag time results in small colony variants that represent a sub-population of persisters. Nature Communications. 2018;9:4074. DOI: 10.1038/s41467-018-06527-0
  13. 13. Bryan LE, Kwan S. Aminoglycoside-resistant mutants of Pseudomonas aeruginosa deficient in cytochrome d, nitrate reductase, and aerobic transport. Antimicrobial Agents and Chemotherapy. 1981;19:958-964. DOI: 10.1128/aac.19.6.958
  14. 14. Colwell CA. Small colony variants of Escherichia coli. Journal of Bacteriology. 1946;52:417-422. DOI: 10.1128/jb.52.4.417-422.1946
  15. 15. Park YJ, Phuong NL, Pinto NA, Kwon JM, D’Souza R, Byun J-H, et al. Urinary tract infection caused by a small colony variant form of capnophilic Escherichia coli leading to misidentification and non-reactions in antimicrobial susceptibility tests. Antimicrobial Resistance and Infection Control. 2018;7:139. DOI: 10.1186/s13756-018-0438-6
  16. 16. Kikuchi M, Suzuki Y, Okada S, Sato A, Kengo O, Matsumoto T. Septic shock caused by a carbon dioxide-dependent and extended spectrum lactamase-producing Proteus mirabilis small colony variant in a long -term bedridden patient. Journal of Infection and Chemotherapy. 2022;28:455-458. DOI: 10.1016/j.jiac.2021.12.013
  17. 17. Marcus RJ, Post JC, Stoodley P, Hall-Stoodley L, RL MG, Sureshkumar KK, et al. Biofilms in nephrology. Expert Opinion on Biological Therapy. 2008;8:1159-1166. DOI: 10.1517/14712598.8.8.1159
  18. 18. Stickler DJ. Clinical complications of urinary catheters caused by crystalline biofilms: Something needs to be done. Journal of Internal Medicine. 2014;276:120-129. DOI: 10.1111/joim.12220
  19. 19. Durack DT. Experimental bacterial endocarditis. IV. Structure and evolution of very early lesions. The Journal of Pathology. 1975;115:81-89. DOI: 10.1002/path.1711150204
  20. 20. Jung C-J, Yeh C-Y, Shun C-T, Hsu R-B, Cheng H-W, Lin C-S, et al. Platelets enhance biofilm formation and resistance of endocarditis-inducing streptococci on the injured heart valve. The Journal of Infectious Diseases. 2012;205:1066-1075. DOI: 10.1093/infdis/jis021
  21. 21. Maduka-Ezeh A, Seville MT, Kusne S, Vikram HR, Blair JE, Greenwood-Quaintance K, et al. Thymidine auxotrophic Staphylococcus aureus small-colony variant endocarditis and left ventricular assist device infection. Journal of Clinical Microbiology. 2012;50:1102-1105. DOI: 10.1128/JCM.01170-11
  22. 22. Benes J, Dzupova O, Setina M, Feuereisl R, Svec P, Pantucek R. Relapsing endocarditis caused by Enterococcus faecalis forming small colony variants. Scandinavian Journal of Infectious Diseases. 2013;45:800-803. DOI: 10.3109/00365548.2013.800227
  23. 23. Madison G, Golamari R, Bhattacharya P. Endocarditis caused by Abiotrophia and Granulicatella species. In: Firstenberg MS, editor. Advanced Concepts in Endocarditis. London: IntechOpen; 2018. DOI: 10.5772/intechopen.74252
  24. 24. Thwaites GE, Gant V. Are bloodstream leucocytes Trojan horses for the metastasis of Staphylococcus aureus? Nature Reviews. Microbiology. 2011;9:215-222. DOI: 10.1038/nrmicro2508
  25. 25. Elek SD, Conen PE. The virulence of Staphylococcus pyogenes for man. A study of the problems of wound infection. British Journal of Experimental Pathology. 1957;38:573-586
  26. 26. Gristina AG. Biomaterial-centered infection: Microbial adhesion versus tissue integration. Science. 1987;237:1588-1595. DOI: 10.1126/science.3629258
  27. 27. Vaudaux P, Pittet D, Haeberli A, Lerch PG, Morgenthaler JJ, Proctor RA, et al. Fibronectin is more active than fibrin of fibrinogen in promoting Staphylococcus aureus adherence to inserted intravascular catheters. The Journal of Infectious Diseases. 1993;167:633-641. DOI: 10.1093/infdis/167.3.633
  28. 28. Foster TJ. The MSCRAMM family of Cell-wall_-anchored surface proteins of Gram positive cocci. Trends in Microbiology. 2019;27:927-941. DOI: 10.1016/j.tim.2019.06.007
  29. 29. Arrecubieta C, Lee M-H, Macey A, Foster TJ, Lowy FD. SdrF, a Staphylococcus epidermidis surface protein, binds type I collagen. The Journal of Biological Chemistry. 2007;282:18767-18776. DOI: 10.1074/jbc.M610940200
  30. 30. Harshey RM. Bacterial motility on a surface: Many ways to a common goal. Annual Review of Microbiology. 2003;57:249-273. DOI: 10.1146/annurev.micro.57.030502.09101
  31. 31. Merz AJ, So M, Sheetz MP. Pilus retraction powers bacterial twitching motility. Nature. 2000;407:98-102
  32. 32. Jarrell KF, Stark M, Nair DB, Chong JP. Flagella and pili are both necessary for efficient attachment of Methanococcus maripaludis to surfaces. FEMS Microbiology Letters. 2011;319:44-50. DOI: 10.1111/j.1574-6968.2011.02264.x
  33. 33. Kuchma SL, Ballok AE, Merritt JH, Hammond JH, Lu W, Rabinowitz JD, et al. Cyclic-di-GMP-mediated repression of swarming motility by Pseudomonas aeruginosa: The pilY1 gene and its impact on surface-associated behaviors. Journal of Bacteriology. 2010;192:2950-2964. DOI: 10.1128/JB.01642-09
  34. 34. Costerton JW, Stewart PS, Greenberg EP. Bacterial biofilms: A common cause of persistent infections. Science. 1999;284:1318-1322. DOI: 10.1126/science.284.5418.1318
  35. 35. Widmer AF. New developments in diagnosis and treatment of infection in orthopedic implants. Clinical Infectious Diseases. 2001;33(Suppl 2):S94-S106. DOI: 10.1086/321863
  36. 36. Blowers R, Mason GA, Wallace KR, Walton M. Control of wound infection in a thoracic surgery unit. The Lancet. 1955;266:786-794. DOI: 10.1016/S0140-6736(55)92385-0
  37. 37. Babb JR, Lynam P, Ayliffe GAJ. Risk of airborne transmission in an operating theatre containing four ultraclean air units. Journal of Hospital Infection. 1995;31:159-168. DOI: 10.1016/0195-6701(95)90062-4
  38. 38. Sehulster L, Chinn RYW. Guidelines for environmental infection control in health-care facilities. MMWR Recommendations and Report. 2003;52(RR10):1-42
  39. 39. Brandt C, Hott U, Sohr D, Daschner F, Gastmeier P, Rüden H. Operating room ventilation with laminar airflow shows no protective effect on the surgical site infection rate in orthopedic and abdominal surgery. Annals of Surgery. 2008;248:695-700. DOI: 10.1097/SLA.0b013e31818b757d
  40. 40. Hooper GJ, Rothwell AG, Frampton C, Wyatt MC. Does the use of laminar flow and space suits reduce early deep infection after total hip and knee replacement? Journal of Bone and Joint Surgery. 2011;93-B:85-90. DOI: 10.1302/0301-620X.93B1
  41. 41. Jain S, Reed M. Laminar air flow handling systems in the operating room. Surgical Infections. 2019;20:1-8. DOI: 10.1089/sur.2018.258
  42. 42. Choksey MS, Malik IA. Zero tolerance to shunt infections: Can it be achieved? Journal of Neurology, Neurosurgery, and Psychiatry. 2004;75:87-91
  43. 43. Korinek A-M, Reina M, Boch AL, Rivera AO, de Bels D, Puybasset L. Prevention of external ventricular drain—Related ventriculitis. Acta Neurochirurgica (Wien). 2005;147:39-46. DOI: 10.1007/s00701-004-0416-z
  44. 44. Allegranzi B, Bischoff P, de Jonge S, Kubilay NZ, Zayed B, Gomes SM, et al. New WHO recommendations on preoperative measures for surgical site infection prevention: An evidence-based global perspective. The Lancet Infectious Diseases. 2016;16:e276-e287. DOI: 10.1016/S1473-3099(16)30398-X
  45. 45. Allegranzi B, Widmer A. WHO’s recommendation for surgical skin antisepsis is premature. The Lancet Infectious Diseases. 2017;17:1023-1024
  46. 46. Selwyn S, Ellis H. Skin bacteria and skin disinfection reconsidered. British Medical Journal. 1972;5792:136-140. DOI: 10.1136/bmj.1.5793.136
  47. 47. Sato S, Sakuragi T, Dan K. Human skin flora as a potential source of epidural abscess. Anesthesiology. 1996;85:1276-1282. DOI: 10.1097/00000542-199612000-00008
  48. 48. Lee MJ, Pottinger PS, Butler-Wu S, Bumgarner RE, Russ SM, Matsen FA. Propionibacterium persists in the skin despite standard surgical preparation. Journal of Bone and Joint Surgery. 2014;96:1447-1450. DOI: 10.2106/JBJS.M.01474
  49. 49. Karpanen TJ, Worthington T, Conway BR, Hilton AC, Elliott TSJ, Lambert PA. Penetration of chlorhexidine into human skin. Antimicrobial Agents and Chemotherapy. 2008;52:3633-3636. DOI: 10.1128/AAC.00637-08
  50. 50. Karpanen TJ, Worthington T, Conway BR, Hilton AC, Elliott TSJ, Lambert PA. Permeation of chlorhexidine from alcoholic and aqueous solutions within excised human skin. Antimicrobial Agents and Chemotherapy. 2009;53:1717-1719. DOI: 10.1128/AAC.01289-08
  51. 51. Nelson RL, Gladman E, Barbateskovic M. Antimicrobial prophylaxis for colorectal surgery. Cochrane Database of Systematic Reviews. 2014;5. DOI: 10.1002/14651858.CD001181.pub4
  52. 52. Prokuski L. Prophylactic antibiotics in orthopaedic surgery. The Journal of the American Academy of Orthopaedic Surgeons. 2008;16:283-293. DOI: 10.5435/00124635-200805000-00007
  53. 53. Norman G, Atkinson RA, Smith TA, Rowlands C, Rithalia AD, Crosbie EJ, et al. Intracavity lavage and wound irrigation for prevention of surgical site infection. Cochrane Database of Systematic Reviews. 2017;10:CD012234. DOI: 10.1002/14651858.CD012234.pub2
  54. 54. Papadakis M. Wound irrigation for preventing surgical site infections. World Journal of Methodology. 2021;11:222-227. DOI: 10.5662/wjm.v11.i4.222
  55. 55. Abdullah KG, Attiah MA, Olsen AS, Richardson A, Lucas TH. Reducing surgical site infections following craniotomy: Examination of the use of topical vancomycin. Journal of Neurosurgery. 2015;123:1600-1604. DOI: 10.3171/2014.12.JNS142092
  56. 56. Beckman JM, Amankwah EK, Tetreault LL, Perlman SA, Tuite GF. Complications associated with bacitracin powder in surgical wounds. Journal of Neurosurgery: Pediatrics. 2015;16:719-725. DOI: 10.3171/2015.4.PEDS14699
  57. 57. Haimoto S, Schar RT, Nishimura Y, Hara M, Wakabayashi T, Ginsberg HJ. Reduction in surgical site infection with suprafascial intrawound application of vancomycin powder in instrumented posterior spinal fusion: A retrospective case-control study. Journal of Neurosurgery. Spine. 2018;29:193-198. DOI: 10.3171/2017.12.SPINE17997
  58. 58. Francolini I, Vuotto C, Piozzi A, Donelli G. Antifouling and antimicrobial biomaterials: An overview. Acta Pathologica, Microbiologica et Immunologica Scandinavica. 2017;125:392-417. DOI: 10.1111/apm.12675
  59. 59. Ozcan E, Mondal A, Douglass M, Hopkins SP, Garren M, Devine R, et al. Bioinspired ultra-low fouling coatings on medical devices to prevent device-associated infections and thrombosis. Journal of Colloid and Interface Science. 2022;608:1015-1024. DOI: 10.1016/j.jcis.2021.09.183
  60. 60. Mulley G, Jenkins ATA, Waterfield NR. Inactivation of the antibacterial and cytotoxic properties of silver ions by biologically relevant compounds. PLoS One. 2014;9:e94409. DOI: 10.1371/journal.pone.0094409
  61. 61. Li Y, Zhao J, Shang E, Xia X, Niu J, Crittenden J. Effects of chloride ions on dissolution, ROS generation, and toxicity of silver nanoparticles under UN irradiation. Environmental Science & Technology. 2018;52:4842-4849. DOI: 10.1021/acs.est.7b04547
  62. 62. Hemmerlein JB, Treretola SO, Kraus MA, Mendonca MS, Desmond LA. In vitro cytotoxicity of silver-impregnated collagen cuffs designed to decrease infection in tunneled catheters. Radiology. 1997;204:363-367
  63. 63. Anon. Silver dressings-do they work? Drug and Therapeutics Bulletin. 2010;48:38-42
  64. 64. Mallucci CL, Jenkinson MD, Conroy EJ, Hartley JC, Brown M, Dalton J, et al. Antibiotic or silver versus standard ventriculoperitoneal shunts (BASICS: A multicentre, single-blinded, randomised trial and economic evaluation). Lancet. 2019;394:1530-1539. DOI: 10.1016/S0140-6736(19)31603-4
  65. 65. Pickard R, Lam T, Maclennan G, Starr K, Kilonzo M, McPherson G, et al. Antimicrobial catheters for reduction of symptomatic urinary tract infection in adults requiring short-term catheterisation in hospital: A multicentre randomised controlled trial. Lancet. 2012;380:1927-1935. DOI: 10.1016/ S0140-6736(12)61380-4
  66. 66. Darouiche RO, Berger DH, Khardori N, Robertson CS, Wall MJ, Metzler MH, et al. Comparison of antimicrobial impregnation with tunneling of longterm central venous catheters. A randomized controlled trial. Annals of Surgery. 2005;242:193-200. DOI: 10.1097/01.sla.0000171874.29934.61
  67. 67. Zabramski JM, Whiting D, Darouiche RO, Horner TG, Olson J, Robertson C, et al. Efficacy of antimicrobial -impregnated external ventricular drain catheters: A prospective randomized, controlled trial. Journal of Neurosurgery. 2003;98:725-730. DOI: 10.3171/jns.2003.98.4.0725
  68. 68. Grice EA, Kong HH, Conlan S, Deming CB, Davies J, Young AC, et al. Topographical and temporal diversity of the human skin microbiome. Science. 2009;324:1190-1192. DOI: 10.1126/science.1171700
  69. 69. Viravaghavan R, Jantausch B, Campos J. Late-onset central nervous system shunt infections with Propionibacterium acnes: Diagnosis and management. La Clinica Pediatrica. 2004;43:393-397
  70. 70. Carneiro IM, Pereira AS, Pinto S, Prata F, Faria CC, Marques JC. Propionibacterium acnes: Cause of cerebrospinal fluid shunt infection. Pediatric Infectious Disease. 2018;37:e168-e169. DOI: 10.1097/INF.0000000000001786
  71. 71. Shiono Y, Nagai S, Kakinuma H, Sasaki A, Funao H, Kuramoto T, et al. Delayed Propionibacterium acnes surgical site infections occur only in the presence of an implant. Scientific Reports. 2016;6:1-10. DOI: 10.1038/srep32758
  72. 72. Lee M, Ponjara G, McLeod K, Chong S. Breast implant illness: A biofilm hypothesis. Plastic and Reconstructive Surgery. Global Open. 2020;8:e2755. DOI: 10.1097/GOX.0000000000002755
  73. 73. Kadler BK, Mehta SS, Funk L. Propionibacterium acnes infection after shoulder surgery. International Journal of Shoulder Surgery. 2015;9:139-144. DOI: 10.4103/0973-6042.167957
  74. 74. Millet PJ, Yen Y-M, Price CS, Horan MP, van der Meijden OA, Elser F. Propionibacterium acnes infection as an occult cause of postoperative shoulder pain. Clinical Orthopaedics and Related Research. 2011;469:2824-2830. DOI: 10.1007/s11999-011-1767-4
  75. 75. McDonald M, Grabsch E, Marshall C, Forbes A. Single -versus multiple-dose antimicrobial prophylaxis for major surgery: A systematic review. ANZ Journal of Surgery. 1998;68:388-396. DOI: 10.1111/j. 1445-2197.1998.tb04785.x
  76. 76. Bratzler DW, Dellinger EP, Olsen KM, Perl TM, Auwaerter PG, Bolon MK, et al. Clinical Practice Guidelines for antimicrobial prophylaxis in surgery. American Journal of Health-System Pharmacy. 2013;70:195-283. DOI: 10.2146/ajhp120568
  77. 77. Branch-Elliman W, O’Brien W, Strymish J, Itani K, Wyatt C, Gupta K. Association of duration and type of surgical prophylaxis with antimicrobial-associated adverse events. JAMA Surgery. 2019;154:590-598. DOI: 10.1001/jamasurg.2019. 0569
  78. 78. Wong GKC, Ip M, Poon WS, Mak CWK, Ng RYT. Antibiotics-impregnated ventricular catheter versus systemic antibiotics for prevention of nosocomial CSF and non-CSF infections: A prospective randomised clinical trial. Journal of Neurology, Neurosurgery, and Psychiatry. 2010;81:1064-1067. DOI: 10.1136/jnnp.2009.198523
  79. 79. James HE, Walsh JW, Wilson HD, Connor JD, Bean JR, Tibbs PA. Prospective randomized study of therapy in cerebrospinal fluid shunt infection. Neurosurgery. 1980;7:459-463. DOI: 10.1227/00006123-198011000-00006
  80. 80. Zimmerli W, Trampuz A, Ochsner PE. Prosthetic-joint infections. The New England Journal of Medicine. 2004;351:1645-1654. DOI: 10.1056/nejmra040181
  81. 81. Vaz K, Taylor A, Kendrick B, Alvand A. A guide to debridement, antibiotics, and implant retention. Annals of Joint. 2022;7:1-5. DOI: 10.21037/aoj-20-89
  82. 82. Byren I, Bejon P, Atkins BL, Angus B, Masters S, McLardy-Smith P, et al. One hundred and twelve infected arthroplasties treated with “DAIR” (debridement, antibiotics and implant retention): Antibiotic duration and outcome. The Journal of Antimicrobial Chemotherapy. 2009;63:1264-1271. DOI: 10.1093/jac/dkp107
  83. 83. Bjarnsholt T. The role of biofilms in chronic infections. Acta Pathologica, Microbiologica et Immunologica Scandinavica. 2013;121:1-58. DOI: 10.1111/apm.12099
  84. 84. Richards BR, Emara KM. Delayed infections after posterior TSRH spinal instrumentation for idiopathic scoliosis: Revisited. Spine. 2001;26:1990-1996. DOI: 10.1097/00007632-200109150-00009
  85. 85. Hahn F, Zbinden R, Min K. Late implant infections caused by Propionibacterium acnes in scoliosis surgery. European Spine Journal. 2005;14:783-788. DOI: 10.1007/s00586-004-0854-6
  86. 86. Kanafani ZA, Sexton DJ, Pien BC, Varkey J, Basmania C, Kaye KS. Postoperative joint infections due to Propionibacterium species: A case control study. Clinical Infectious Diseases. 2009;49:1083-1085. DOI: 10.1086/605577
  87. 87. Yee DKH, Chiu KY, Yan CH, Ng FY. Review article: Joint aspiration for diagnosis of periprosthetic infection. Journal of Orthopaedic Surgery. 2013;21:236-240. DOI: 10.1177/230949901302100225
  88. 88. Chalmers PN, Sporer SM, Levine BR. Correlation of aspiration results with periprosthetic sepsis in revision total hip arthroplasty. The Journal of Arthroplasty. 2014;29:438-442. DOI: 10.1016/j.arth.2013.06.020
  89. 89. Bayston R, Swinden J. The aetiology and prevention of shunt nephritis. Zeitschrift für Kinderchirurgie. 1979;28:377-384
  90. 90. Legoupil N, Ronco P, Berenbaum F. Arthritis -related shunt nephritis in an adult. Rheumatology. 2003;42:698-699. DOI: 10.1093/rheumatology/keg151
  91. 91. Aggrawal VK. Swab cultures are not as effective as tissue cultures for diagnosis of periprosthetic joint infection. Clinical Orthopaedics and Related Research. 2013;471:3196-3203
  92. 92. Parvizi J, Gehrke T, Chen AF. Proceedings of the international consensus on periprosthetic joint infection. Bone and Joint Journal. 2013;95B:1450-1452
  93. 93. Atkins BL, Athanasou N, Deeks JJ, Crook DWM, Simpson H, Peto TEA, et al. Prospective evaluation of criteria for microbiological diagnosis of prosthetic-joint infection at revision arthroplasty. Journal of Clinical Microbiology. 1998;36:2932-2939
  94. 94. Butler-Wu SM, Burns EM, Pottinger PS, Magaret AS, Rakeman JL, Matsen FA, et al. Optimization of periprosthetic culture for diagnosis of Propionibacterium acnes prosthetic joint infection. Journal of Clinical Microbiology. 2011;49:2490-2495. DOI: 10.1128/JCM.00450-11
  95. 95. Askar M, Ashraf W, Scammell B, Bayston R. Comparison of different human tissue processing methods for maximization of bacterial recovery. European Journal of Clinical Microbiology & Infectious Diseases. 2019;38:149-155. DOI: 10.1007/s10096-018-3406-4
  96. 96. Trampuz A, Piper KE, Mj J, Hanssen AD, Unni KK, Osmon DR, et al. Sonication of removed hip and knee prostheses for diagnosis of infection. The New England Journal of Medicine. 2007;357:654-663
  97. 97. Yano MH, Klautau GB, da Silva CB, Nigro S, Avanzi O, Mercadente MT, et al. Improved diagnosis of infection associated with osteosynthesis by use of sonication of fracture fixation implants. Journal of Clinical Microbiology. 2014;52:4176-4182. DOI: 10.1128/JCM.02140-14
  98. 98. Qu X, Zhai Z, Li H, Liu X, Zhu Z, Wang Y, et al. PCR-based diagnosis of prosthetic joint infection. Journal of Clinical Microbiology. 2013;51:2742-2746. DOI: 10.1128/JCM.00657-13
  99. 99. Askar M, Sajid M, Nassif Y, Ashraf W, Scammell B, Bayston R. Propidium monoazide-polymerase chain reaction for detection of residual periprosthetic joint infection in two-stage revision. Molecular Biology Reports. 2019;46:6463-6470. DOI: 10.1007/s11033-019-05092-z
  100. 100. Hook AL, Chang C-Y, Yang J, Lucket J, Cockayne A, Atkinson S, et al. Combinatorial discovery of polymers resistant to bacterial attachment. Nature Biotechnology. 2012;30:868-875. DOI: 10.1038/nbt.2316
  101. 101. Busscher HJ, Alt V, van der Mei HC, Fagette PH, Zimmerli W, Moriarty TF, et al. A transatlantic perspective on stagnation in clinical translation of antimicrobial strategies for the control of biomaterial-implant-associated infection. ACS Biomaterials Science & Engineering. 2018;5:402-406. DOI: 10.1021/acsbiomaterials.8b01071

Written By

Roger Bayston

Submitted: 25 February 2022 Reviewed: 16 March 2022 Published: 22 April 2022