Antimicrobial Use in the Critical Care Patient

Chapter 194 Antimicrobial use in the Critical Care Patient





INTRODUCTION


Antimicrobials are among the most common with most important drugs prescribed for the critical care patient (CCP).1 Timely, effective antimicrobial therapy is a crucial determinant of outcome in the CCP2; however, the advent of antimicrobial resistance has profoundly altered its use. The goal of antimicrobial therapy includes the safe eradication of infection while minimizing the advent of resistance.


The CCP is particularly at risk for infections with antimicrobial resistant bacteria. The risk of infection increases because of bacterial translocation from the gastrointestinal (GI) tract, the use of invasive procedures, and foreign surfaces conducive to bacterial colonization (e.g., catheters). Patients are often immunocompromised. Cardiovascular, renal, and hepatic dysfunction or responses alter all aspects of drug disposition, increasing the risk of either adverse drug events or therapeutic failure. Polypharmacy, or the use of multiple medications in an individual patient, increases the risk of adverse drug events and drug interactions. Finally, the sense of urgency accompanying therapeutic decision making generally leads to empiric antimicrobial use.


The principles guiding antimicrobial therapy are regularly reviewed.3 This chapter summarizes those principles, with a focus on their relevance to the CCP. Included is a description of antimicrobial resistance, including factors predisposing to its emergence, methods by which resistance might be avoided, and then a step-wise decision path that might be implemented as antimicrobial therapy is considered in the CCP. The Infectious Diseases Society of America (http://www.idsociety.org) offers guidelines for the use of antimicrobial agents, many of which are specific to conditions characterized as critical. These guidelines are reassessed and modified on a cyclical basis.



ANTIMICROBIAL RESISTANCE



Advent of Resistance


Previous antimicrobial exposure is among the most predictive factors for the development of antimicrobial resistance. The diversity of the GI flora includes E. coli as the major gram-negative and Enterococcus spp as the major gram-positive aerobes; however, the often-underestimated anaerobic flora predominates. These microbes maintain an ecologic niche by competing for nutrients and actively suppressing surrounding growth through secretion of antibiotics. Self-destruction does not occur because genes encoding antibiotic secretion generally accompany genes that impart resistance. Constant exposure to antibiotics also causes surrounding commensal organisms to be primed for resistance. Rapid microbial turnover ensures frequent DNA replication and thus the potential for mutation. Genes imparting resistance are shared among organisms via integrins, plasmids, and transposons that facilitate rapid transfer of multidrug resistance.4 Resistance to any antimicrobial drug should be anticipated when the population of bacteria reaches or exceeds 106 to 108 colony-forming units (CFU).5


The use of broad-spectrum antimicrobial drugs facilitates selection of resistant organisms,6 although the potential for resistance varies among organisms. More problematic organisms have emerged since the 1990s including, in order of appearance, methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant Enterococcus (VRE), fluoroquinolone-resistant Pseudomonas (FQRP) and, most recently, vancomycin-resistant Staphylococcus aureus (VRSA). Each has developed multidrug resistance, defined as resistance to three or more antimicrobial agents to which the organism is generally considered susceptible. Staphylococcus is particularly intrinsically virulent, able to adapt to many different environmental conditions, and is increasingly associated with life-threatening infections. Resistance to methicillin is at a high level, reflecting a gene coding for an altered penicillin-binding protein with a low affinity for all β-lactam antibiotics, including cephalosporins and carbapenems. The advent of MRSA can be associated with cephalosporin use.7 The gene imparting methicillin resistance has been detected in Staphylococcus-infected dogs.8,9


Escherichia coli is also emerging as a multidrug-resistant (MDR) organism, with resistance associated with fluoroquinolone treatment.2 Even a single dose of a fluoroquinolone has been associated with changes in the resistance pattern of commensal coliforms in humans.10 Fluoroquinolone-resistant E. coli has been documented in the urinary tract of dogs and other tissues11 and has been associated with nosocomial infections in a veterinary teaching hospital.12 E. coli is among the isolates that are able to produce extended-spectrum β-lactamases (ESBLs).


These enzymes, encoded by large plasmids, have emerged in concert with high-level use of cefotaxime and ceftazidime. They cause MDR coliforms and require special testing procedures for detection. The ESBLs are found most commonly in Klebsiella spp, E. coli, or Proteus mirabilis (3.1% to 9.5%), but they also have been detected in other members of the Enterobacteriaceae family and in Pseudomonas aeruginosa. The genetic information for ESBL is conferred between and within organisms. Newer cephalosporins are affected, including cefotaxime, ceftazidime, and ceftriaxone, as well as cefpodoxime13 and several fourth-generation drugs. The effect on cephamycins (e.g., cefoxitin, cefotetan) is less clear. Monobactams (i.e., aztreonam), but not carbapenems (e.g., imipenem or meropenem), also are targeted by ESBLs. The impact on clavulanic acid and sulbactam is not clear, although their use in place of cephalosporins appears to reduce the emergence of ESBLs and may reduce resistance in other pathogens such as Clostridium difficile and VRE.14 Decreased cephalosporin usage reduces the advent of ESBLs. Emergence of an ESBL in a patient may depend on the size of the inoculum, and detection requires special testing procedures generally not offered by diagnostic microbiology laboratories. An ESBL should be suspected with organisms resistant to cefotaxime but susceptible to β-lactam/β-lactamase combinations.



Nosocomial Infections


Nosocomial infections occur as a result of medical treatment, usually in a hospital or clinic setting. Nosocomial infections are formally defined as infections arising more than 48 hours after hospital admission. Nosocomial organisms are generally opportunists. The most important source probably is the environment, although transfer from caregivers or other patients is possible. The risk of nosocomial infection is 5- to 10-fold higher in the human critical care ward compared with the hospital-at-large population.1


Bacterial colonization by nosocomial isolates occurs in the upper respiratory tract, GI tract, and urogenital tract and on skin of hospitalized patients within a few days of hospitalization. Accordingly, nosocomial infections most commonly occur in the respiratory system or urinary tract or skin. Infection is frequently associated with invasive procedures. Nosocomial infection in veterinary CCPs has been reviewed.15 The organisms reportedly associated with these infections in dogs and cats have been many and diverse, varying with the report. Organisms include, but not exclusively, Serratia spp, Staphylococcus spp, Streptococcus spp, Klebsiella spp, Enterococcus spp, and E. coli. As in humans, predisposing factors have included presence of indwelling catheters16 and previous antimicrobial therapy.


Nosocomial organisms are generally characterized by complex resistance patterns; in some intensive care units (ICUs) that treat humans selected isolates are characterized by a resistance prevalence of 86%. Resistance results increased morbidity and mortality, and increased costs. Effective treatment generally requires more expensive and potentially toxic drugs; selection should be based on culture and susceptibility testing.17



Reducing Environmental Microbial Resistance


ICUs have implemented a number of techniques intended to reduce antimicrobial resistance. Each proposed or implemented strategy has theoretic benefits and limitations, but good data on their efficacy in controlling antimicrobial resistance are limited.18,19 Hospital strategies involve a multitiered approach.






Changing Antimicrobial Prescribing Behaviors


Antimicrobial drug prescribing behaviors are the most significant mechanism by which bacterial resistance is likely to be reduced in the critical care environment.20 Protocols should be designed and subsequently implemented with the intent to deescalate antimicrobial use. Deescalation is among the more rational paradigms for empiric antimicrobial use in hospitalized patients with serious bacterial infections.20 The goal of deescalation is to prescribe an initial antibacterial regimen that will cover the most likely bacterial pathogens associated with infection, thus balancing the need for appropriate therapy, while minimizing the risk of emerging antibacterial resistance.20 The three-pronged approach includes narrowing the antibacterial regimen through culture, assessing the susceptibility for dosage determination, and choosing the shortest course of therapy clinically acceptable.


Although preapproval of selected drug use (e.g., by a committee) and antibiotic restriction practices may be useful, recommended and more reasonable deescalation procedures include rotating the use of antimicrobial drugs on a regular schedule, and designing the dosing regimen such that therapeutic success is maximized, resistance is minimized, and duration of therapy can be reduced.18 Deescalation procedures in humans have been associated with a return to susceptibility for ceftazidime, piperacillin, imipenem, and fluoroquinolones.


The risk of antimicrobial resistance is associated with both dose and duration of therapy. Basing drug selection on culture and susceptibility information, with accompanying minimum inhibitory concentration (MIC) data, is critical both to identify current bacterial resistance in patients at risk and to best design an individual patient dosing regimen. Approaches to dosage adjustment are discussed next. However, increasingly in human medicine, dosages are being based on the mutant prevention concentration (MPC) rather than the MIC.5


The MPC is defined as the highest MIC identified in a patient population of susceptible isolates (≥107) of an organism, thus including those CFUs or isolates that have undergone the first-step mutation. The MIC obtained from culture and susceptibility testing most likely reflects the majority of CFUs causing infection in the patient. However, the MIC is less likely to reflect those CFUs that already have undergone the first-step mutation (i.e., are characterized by higher MIC). Should drug therapy target the MIC, competing isolates will be inhibited or killed, allowing the mutants to emerge. In healthy patients, this population can probably be controlled by host defenses. However, in less capable patients, the new emergent population will be characterized by a higher MIC that is potentially unattainable with a safe dosing regimen.


Unfortunately, determining the MPC of an isolate cultured from a patient requires culture based on 107 or more organisms; current techniques cannot achieve this large an inoculum. Experimentally, the ratio of MPC to MIC for various fluoroquinolones given for infection by human pathogens ranges from a low of 6 to 10 for E. coli but 23 to 50 (and as high as 125) for selected drugs given for infection by Staphylococcus aureus.


Rational combination antimicrobial therapy can be a powerful tool for enhancing efficacy while reducing resistance in the CCP. Combination therapy should be considered routinely for organisms often associated with MDR (e.g., P. aeruginosa, Enterococcus spp, and MRSA). Resistance to a combination of antimicrobial drugs should be anticipated when the population reaches 1014 or more CFUs. Drugs chosen for combination therapy should be selected rationally, based on target organisms. Mechanisms of action should complement, rather than antagonize, one another.21


In general, “bacteriostatic” drugs that inhibit ribosomes and thus microbial growth (e.g., chloramphenicol, tetracyclines, and erythromycin) should not be combined with drugs whose mechanism of action depends on protein synthesis, such as growth of the organism (e.g., β-lactams) or formation of a target protein. The bactericidal activity of β-lactams and fluoroquinolones depend on continued synthesis of bacterial proteins. Antagonistic effects have been well documented between β-lactam antimicrobials and inhibitors of ribosomal activity.21


Chemical antagonism is also possible among two or more antimicrobials; the prototypical example is chemical inactivation of aminoglycosides and quinolones by β-lactams. However, chemical antagonism is unlikely to occur at concentrations achieved systemically in the clinical patient. In contrast to antagonism, drugs that have the same mechanism of action may act in an additive or synergistic fashion. The prototypical example of synergism is the combination of β-lactams and aminoglycosides; aminoglycoside penetration is facilitated by penicillin-induced cell wall failure.21 Indeed, aminoglycoside activity against Enterococcus spp is adequate only when the agent is used synergistically with a cell wall–active antibiotic, such as a β-lactam or vancomycin. Synergism has also been demonstrated against some strains of Enterobacteriaceae, P. aeruginosa, staphylococci (including MRSA), and other microorganisms. Enhanced movement into the bacteria may occur with other drugs (e.g., potentiated sulfonamides, fluoroquinolones) when combined with a β-lactam.


Combination antimicrobial therapy may be selected for a polymicrobial infection. Aminoglycosides or fluoroquinolones are often combined with β-lactams, metronidazole, or clindamycin to target both aerobic gram-positive and gram-negative infections, or aerobic infections caused by both aerobes and anaerobes. The combined use of selected antibiotics may result in effective therapy against a given microbe, even when either drug alone would be ineffective.

Stay updated, free articles. Join our Telegram channel

Sep 10, 2016 | Posted by in SMALL ANIMAL | Comments Off on Antimicrobial Use in the Critical Care Patient

Full access? Get Clinical Tree

Get Clinical Tree app for offline access