SciELO - Scientific Electronic Library Online

 
vol.27 número2Effects of Strength Training and Jatamansi on reducing Hand Tremor amongst archers índice de autoresíndice de materiabúsqueda de artículos
Home Pagelista alfabética de revistas  

Servicios Personalizados

Revista

Articulo

Indicadores

Links relacionados

  • No hay articulos similaresSimilares en SciELO

Compartir


Archivos Venezolanos de Farmacología y Terapéutica

versión impresa ISSN 0798-0264

AVFT v.27 n.2 Caracas dic. 2008

 

The recent evolution of therapeutic weapons against resistant Gram-positive microorganisms

Running head: Management of multiresistant Gram-positive pathogens

Roberto Manfredi, MD, Leonardo Calza, MD

Department of Clinical and Experimental Medicine, Division of Infectious Diseases, "Alma Mater Studiorum". University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy. Conflicts of interest, funding, sponsorhip, acknowledgements: none to be declared

Correspondence: Prof. Roberto Manfredi. Infectious Diseases, University of Bologna S. Orsola Hospital, Via Massarenti, 11 I-40138 Bologna (Italy). Telephone: +39-051-6363355 Telefax: +39-051-343500. E-mail: Roberto.manfredi@unibo.it

Abstract

Multiresistant Gram-positive cocci, including Staphylococcus aureus, the group of coagulase-negative staphylococci, Enterococcus faecalis and Enterococcus faecium, as well as Streptococcus pneumoniae and other streptococci, represent emerging pathogens. This issue is especially concerning in the setting of immunocompromised, hospitalized patients, in particular when surgery, invasive procedures, or prosthetic implants are carried out, patients are admitted in intensive care units, or underlying chronic disorders and immunodeficiency are of concern, and broad-spectrum antibiotics are widely used in the environment; moreover, a community spread of resistant Gram-positive cocci has been recognized during recent years. The spectrum of antimicrobials available for an effective management of these relevant infections is significantly threatened by the emerging of methicillin-resistant and more recently glycopeptide-resistant strains. The streptogramine association represented by quinupristin/dalfopristin, the oxazolidinone derivative linezolid, and the recently licensed daptomycin and tigecycline, together with a number of glycopeptides, fluoroquinolones, cephalosporins, and other experimental compounds, represent an effective response. It is due to the innovative mechanisms of action of these compounds, their maintained or enhanced activity against multiresistant pathogens, their effective pharmacokinetic/pharmacodynamic properties, their frequent possibility of synergistic activity with other compounds effective against Gram-positive pathogens, and a diffuse potential for a safe and easy administration, also to compromised patients. The main problems related to the epidemiology of multiresistant gram-positive infection, the potential clinical indications of all recently available compounds compared with the standard of care of treatment of resistant Gram-positive infections, and updated data on efficacy and tolerability of all these compounds, are updated and outlined on the ground of a review of recent literature evidences.

Key words: Enterococci, glycopeptides, Gram-positive organisms, novel antimicrobial agents, oxazolidinones, Pneumococci, resistance, Staphylococci, streptogramins

Recibido: 06/05/2008 Aceptacion: 12/05/2008

Introduction

Especially among nosocomial pathogens, since early eighties a significant reversal of tendency was observed, characterized by a predominance of Gram-positive over Gram-negative bacteria (which represented the most relevant concern in the two prior decades)1-5. The increased life expectancy of the general population, the extended survival of patients with underlying immunodeficiency and/or chronic disorders, the advances of surgical techniques and those of invasive diagnostic and therapeutic procedures and bone marrow and solid organ transplantation, the diffusion of prosthetic materials and other biocompatible materials, and the increased resort to endovascular catheters and devices, represent emerging risk factors for massive bacterial colonization, which turns into an increased risk of local and systemic infection. Moreover, the increased and prolonged administration of broad spectrum antimicrobial agents and their associations, and the wide employment of antufungal agents, all as therapeutic and/or prophylactic compounds in the immunocompromised to otherwise at risk host, extensively contribute to the re-emerging of Gram-positive pathogens, especially via a prolonged hospital admission. Among these microorganisms, both coagulasepositive and coagulase-negative Staphylococci and Enterococci are of particular concern in the hospital setting, because of their rising frequency, the severity of associated diseases, and the unpredictable spectrum of drug resistance1,2,4-8. This is also increasingly true in the pediatric population5,9,10.

On the other hand, in the community setting Pneumococci and other Streptococci remain the main respiratory pathogens, and their antibiotic resistance rate are progressively on the rise, although both macrolide- and beta-lactam resistant pathogens does not pose very striking problem in the majority of countries, until now 2-4, 8-12.

Finally, the progressive shortening of hospitalization favors the spread of hospital-acquired, nosocomial pathogens into the community, where there is increasing concern for the rising retrieval of microorganisms with a resistance pattern similar to that observed at the hospital, which may be responsible for an increasing failure of empiric therapeutic lines, when antimicrobial agents are administered before obtaining culture and in vitro resistance testing, or when cultures and sensitivity testing are not available or are still pending2,5,10,13.

Together with the ever-evolving modification of multiple environmental conditions, the changing features of both host and its microbial flora, and the broadened spectrum of currently available anti-infective compounds (Table 1), other emerging features become prominent, related to the pathomorphism of clinical features of a number of infections caused by Grampositive pathogens, often those which share an evident potential for the development and spread of antimicrobial resistance. While streptococcal scarlet fever, and post-streptococcal complications like rheumatic disease and acute glomerulonephritis virtually disappeared in the past two decades, a number of other disorders caused by highly pathogenic Streptococci gained increasing frequency and importance in the last decades, including severe cellulites, necrotizing fasciitis, and toxic shock syndrome (TSS). When considering Staphylococci, the major pathogen Staphylococcus aureus steadily ranks at the first place among the majority of nosocomial pathogens, and even in this case novel syndromes characterized by the predominant role of bacterial toxaemia are progressively emerging: the so-called staphylococcal scalded skin syndrome (or SSSS) is the paradigmatic clinical picture. Moreover, severe staphylococcal skin and soft tissue lesions have been attributed to the action of the Panton-Valentine leukocidin10. Remarkable levels of morbidity and mortality are also attributed to coagulase-negative cocci (i.e. Staphylococcus epidermidis and related organisms), especially when vascular or bone-joint prosthetic devices, central vascular lines, ventilator-associated pneumonia, and parenteral nutrition are of concern. Until a couple of decades ago, the same coagulase-negative staphylococci were mostly considered as trivial contaminants, or part of the normal saprophytic human flora.

In the developed countries of the world, the rate of methicillin (oxacillin) resistance of Staphylococci among hospitalized patients may overcome 20-25% of cases, with an extremely elevated frequency registered in specialized intensive care units and bone marrow and solid organ transplant units1,4,5,8,13,14. A relevant multicentre survey of nosocomial bacteremia carried out in 49 United States hospitals during a three-year period, allowed the authors to recognize a 64% prevalence of Grampositive pathogens, among over 10,000 identified microorganisms13. When analyzing the frequency of single microorganisms, coagulase-negative Staphylococci (32%) preceded S. aureus (16%), and Enterococci as a whole (11%). The overall level of methicillin resistance ranked around 29% of isolated organisms, with peaks reaching 80%, when coagulase-negative Staphylococci were specifically considered13. In a Spain nationwide S. aureus prevalence study, an overall increase in resistance to most antimicrobials was detected, mainly to oxacillin (from a frequency ranging from 1.5% and 32.5% in the year 1986, to 31.2%-61.3% in the year 2002), although all isolates remained susceptible to available glycopeptides, quinupristin/dalfopristin and linezolid, and a surprisingly low resistance rate remained towards cotrimoxazole (0.5-2.1%)15. The same phenomenon has been observed at a large teaching Hospital in Taiwan, where the noticeable rise of methicillin-resistant Staphylococci and vancomycinresistant Enterococci paralleled the increased prescription of glycopeptides, broad-spectrum beta-lactams, carbapenems, and fluoroquinolones16.

Among these microorganisms which share a predominant nosocomial isolation, the appearance of methicillin resistance is usually linked to an almost complete lack of in vitro susceptibility to all beta-lactam antibiotics, but usually it extends to macrolides, lincosamides, and a large part of aminoglycosides and fluoroquinolones, also, therefore leading to a very striking reduction of remaining therapeutic options1-8,17. Notwithstanding that the frequently concurrent pathophysiological conditions can also act unfavorably, by limiting both microbiological and clinical efficacy of an antimicrobial treatment in vivo (Table 2), however the selection and spread of pharmacoresistant bacterial strains represents the main feature responsible for their severely reduced activity in current clinical practice.

The guidelines for empiric antimicrobial chemotherapy of the immunocompromised and/or neutropenic patient took into careful account of the etiological shift from Gram-negative towards Gram-positive organisms, which occurred during the last two decades, as reminded above. In fact, in the past decade the recommendations for empiric therapy of at-risk patients strongly pointed out the inclusion of drugs which are highly active against methicillin-resistant Gram-positive cocci, but concomitantly contributed to the appearance and further spread of mutant strains, which test either "intermediate" or even "resistant" to both available glycopeptides (vancomycin and teicoplanin), which represented the "gold-standard" reference molecules for the management of multiresistant Gram-positive cocci, until a few years ago1-5,7,8,14,18. A relevant Italian survey referred to years 1997-199819, showed that Gram-positive microorganisms were responsible for slightly more than 50% of respiratory infection and septicemia identified in three different Italian intensive care units, with absolute predominance of S. aureus (29.2%), followed by coagulasenegative Staphylococci (9.5%), Streptococcus pneumoniae (4.1%), and Enterococcus faecalis (2.9%). Methicillin-resistance levels tested around 46% for S. aureus, but rose to 64% for coagulase-negative Staphylococci19. When evaluating the episodes of nosocomial sepsis, the international SCOPE study carried out in the year 199820 attributed the most elevated incidence to coagulase-negative Staphylococci, followed by S. aureus and Enterococcus spp., whose mortality rates proved 21%, 25%, and 32% of reported cases, respectively. Enterococcus spp. organisms showed an antibiotic susceptibility profile remarkably different between E. faecalis and E. faecium: this last pathogen was increasingly identified during recent years, in association with methicillin resistance levels often greater than 50% of tested strains. Concurrently, the incidence of methicillin resistance among coagulase-negative Staphylococci (with S. epidermidis as the leading organism) tested even greater in frequency (also over 60-80% in different clinical settings). Finally, an open debate is still ongoing regarding the role and frequency of staphylococcal strains which test "intermediate" to vancomycin and glycopeptides in general (the so-called "glycopeptide-intermediate S. aureus", or GISA), which were early identified in Japan since ten years ago (year 1996)21. The incidence of this last phenomenon is estimated to be still contained, although it is more commonly recognized in countries where the frequent resort to glycopeptide administration supported a non-specific selective pressure. The spread of glycopeptide resistance may occur through differentbacterial species especially in the gastrointestinal tract, which may act as a reservoir of genes conferring resistance to glycopeptides and concurrently to many other antimicrobial compounds2,3,8,22.

Quinupristin/dalfopristin: the streptogramin association

The streptogramin association quinupristin/dalfopristin is an antibiotic combination composed by two different molecules (in a 30%-70% proportion), which express a relevant synergistic activity against susceptible microbial pathogens, based on a double blockade of the polypeptide chain extension. Quinupristin/dalfopristin proves effective against a broad spectrum of Gram-positive organism, even when they become resistant to methicillin and also glycopeptides. Streptococci, pneumococci, and especially coagulase-positive and coagulase-negative Staphylococci, Clostridium spp. and Peptostreptococcus spp., and Enterococci, with the partial exception of multiresistant strains of E. faecalis (whose susceptibility index is however around 30% of tested strains)19, represent the target microorganisms of this novel compound. The in vitro sensitivity spectrum of quinupristin/dalfopristin is also extended towards multiple relevant Gram-negative pathogens, including Legionella pneumophila, Moraxella catarrhalis, and Mycoplasma pneumoniae7,17,23. The breakpoint values of quinupristin/dalfopristin recommended for in vitro microdilution techniques searching for minimum inhibitory concentrations (MIC) determination are <1 µg/mL for sensitive microorganisms, 2 µg/mL for moderately susceptible (or "intermediate") organisms, and >4 µg/mL per isolates defined as resistant17. Moreover, the development of acquired resistance against this streptogramin association is expected to represent a very rare event, as characterized by a frequency of mutations occurring in staphylococcal and enterococcal strains ranging from 10-9 and 10-11 17, while a confirmed in vivo resistance accounts for around 2% of clinical episodes24, and may rise to 8-20% is particular settings, where the incidence of vancomycin-resistant Enterococci is of elevated concern16, or among some coagulase-negative multiresistant Staphylococci25. However, this last microbiological resistance profile may be responsible for confirmed clinical failure7. Because of its prolonged post-antibiotic effect (ranging from 2-6 hours for methicillin-resistant S. aureus, to over 18 hours for Streptocococcus pyogenes)7,17, the quinupristin/dalfopristin association has a potent in vitro synergistic activity (often confirmed by in vivo experiences) with an elevated number of other antimicrobial drugs, including glycopeptides theirselves, but also rifampicin and derivatives, ciprofloxacin and derivatives, ampicillin, and some cephalosporins, directed especially against meticillin-resistant Staphylococci. When considering E. faecalis strains testing resistant to vancomycin and teicoplanin, again glycopeptides, tetracyclines, and penicillins protected by beta-lactamase inhibitors, are expected to show a synergistic activity with quinupristin/dalfopristin7,17,23.

Since the antimicrobial activity of quinupristin/dalfopristin is based on the synergistic action of both molecules, the pharmacokinetic and pharmacodynamic features of this fixed association are of striking importance: the rate of serum concentration of the two molecules is included in the range of antimicrobial activity of the quinupristin/dalfopristin association against the different susceptible microorganisms17,23. The in vivo half-life of biologically active compounds is 2-3 hours for quinupristin, and around one hour for dalfopristin. Therefore, the area under the curve (AUC)/minimal inhibitory concentration (MIC) ratio remains above the MIC of the target pathogens (i.e. 1 µg/mL), while the plasmatic coverage is enforced by the prolonged post-antibiotic effect. The initial, current dosage (7.5 mg per Kg of body weight, every 8 hours or 12 hours), needs i.v. administration through extensive dilution in glucose solution, and preferably by a central venous catheter and an infusion duration of at least 60 minutes, in order to prevent local toxicity, which can occur when peripheral veins are used for long-term administration.

Given its in vitro spectrum of antimicrobial activity, the clinical indications for quinupristin/dalfopristin administration presently include all "difficult" Gram-positive lower airways infections, infections of skin and soft tissues, and especially vancomycin-resistant E. faecium disease (regardless of the interested body site), due to the difficulty to have an effective treatment of these severe pathologies determined by the increasing occurrence of multiresistant strains7,19,22.

On the ground of the available clinical evidences reported by the international literature, severe infections of critically ill patient which deserve a quinupristin/dalfopristin treatment include those due to Gram-positive cocci testing in vitro resistant to glycopeptides, but also situations burdened by an elevated risk for multiresistant gram-positive agents, which failed to respond (clinically or microbiologically) to at least three days of a teicoplanin- or vancomycin-based antimicrobial chemotherapy. A combination treatment (including glycopeptides themselves, or rifampicin, aminoglycosides, cotrimoxazole), can be attempted on empiric basis or after in vitro susceptibility assays, in order to exploit the above-mentioned synergistic effects with quinupristin/dalfopristin7,17,20,23. The therapeutic choice may prefer quinupristin/dalfopristin also when risk factors which make other combination poorly tolerated or difficult to be delivered, because of expected toxicity, intolerance, or underlying systemic disorders (such as diabetes mellitus, kidney failure, or myelotoxicity).

While quinupristin/dalfopristin dosage does not need adjustment until renal insufficiency becomes severe, a reduced daily dosage and strict monitoring of hepatic function are recommended when liver failure is of concern. Should an extensive end-organ liver disease is present, the administration of quinupristin/dalfopristin becomes contraindicated.

From a clinical-therapeutic point of view, during the preregistration period the drug has been administered for the treatment of a high number of clinical episodes of severe diseases determined by multiresistant Gram-positive pathogens, often involving severely immunocompromised patients. Among disease localizations described in the earlier literature reports, we underline a wide spectrum of difficult-to-treat and life-threatening endocarditis, such as enterococcal endocarditis on a prosthetic valve26, multiresistant S. epidermidis endocarditis27, those due to multiresistant S. aureus occurring artificial valve and no chance of surgery, as well as E. faecium heart localization, where a combination with doxicyclin, rifampicin, and high-dose ampicillin was favorably used, and a demonstration of synergistic activity given28,29. E. faecium is infrequently responsible for bone and joint infection, although a progressive increase of frequency has been noticed among patients undergoing replacement of infected prosthetic devices, where the role of coagulase-positive and coagulase-negative Staphylococci, and that of Enterococci, are mounting. Also in these events, an interesting case report demonstrated the efficacy of quinupristin/dalfopristin in an inveterate vertebral osteomyelitis caused by vancomycinresistant bacterial pathogens30. Given the low cerebrospinal fluid concentration obtained after quinupristin/dalfopristin administration31, episodes of severe central nervous system infection caused by E. faecium (ventriculitis, ventricular drainage infection, meningitis, brain abscess), have been treated favorably after local drug administration (i.e. via intratecal or intraventricular route)31-33, with a mean dosage of 2 mg, and in absence of significant untoward events; in one case of meningitis quinupristin/dalfopristin was concurrently administered i.v. at full dosage33. Also in pediatric age, preliminary observations conducted on 11 overall children34,35, underlined the microbiological and clinical efficacy and the safe profile of quinupristin/dalfopristin in the management of vancomycinresistant intrabdominal E. faecium infection and septicemia, in patients who underwent bone marrow transplantation during treatment of hematological malignancies34, and in other young patients with an underlying severe immunosuppression35; the association with teicoplanin showed a synergistic effect also in these last episodes34.

Relevant multicentre randomized trials, and an extensive spectrum of clinical experiences and case series treated with quinupristin/dalfopristin, confirmed the elevated efficacy of this novel streptogramin combination in the treatment of pneumonia and severe skin and soft tissue infection, as well as infected surgical wound infection frequently observed in hospital settings7,17,23,36-38. Among randomized clinical trials, particular attention should be addressed to the comparison of quinupristin/dalfopristin with vancomycin in the management of gram-positive nosocomial pneumonia in critical care units36, the comparison with cefazolin, oxacillin, and vancomycin in the therapy of skin-soft tissue infection37, and the study especially devoted to E. faecium infections38. The very favorable results obtained in these experiences hypothesized the use of quinupristin/dalfopristin in experimental protocols of eradication of methicillin-resistant staphylococcal colonization, and as an empiric choice for neoplastic patients with febrile neutropenia. An underlying kidney failure followed by organ transplantation did not impair the effectiveness of this streptogramin combination in multivisceral and disseminated infections caused by multiresistant S. epidermidis strains, sometimes in combination with chloramphenicol, and after failure of multiple therapeutic attempts carried out with glycopeptides39. A more recent experience of the same research group, involving six patients who underwent hemodialysis (one submitted to renal transplantation, and four complicated be hepatic failure), confirmed the clinical efficacy and safety of quinupristin/dalfopristin even in patients with severe endorgan involvement, without need of drug dosage adjustment, and also determining the drug pharmacokinetic profile in these extreme conditions40. However, especially when transplanted patients undergoing an immunosuppressive therapy are of concern, the need to proceed to a repeated monitoring of cyclosporin serum levels and dosage during quinupristin/dalfopristin administration is confirmed, as already indicated in the early clinical experiences conducted with this streptogramine association41.

As we can deduct from the results of the above-mentioned controlled studies, and the numerous, intriguing case reports and case series, quinupristin/dalfopristin appears to be an effective and well tolerated agent in the management of septicemia, heart-thoracic, intrabdominal, bone and joint, and skin and soft tissue infections caused by methicillin-resistant Staphylococci, also in combination with a glycopeptide, notwithstanding that the previous administration of the sole glycopeptide agent resulted not effective42-45. The spectrum of activity of this dual streptogramin combination, since it remains restricted to gram-positive cocci, recommends the association with other antimicrobial agents with enlarged spectrum of action, when a polymicrobial infection is suspected, or a mixed flora containing Gram-positive and Gram-negative organisms is of concern7,17,23,29,44,46. Finally, the hospital and environmental spread of pathogenic multiresistant Gram-positive cocci as a result of the selective pressure determined by the increased and prolonged administration of other broad- and narrowspectrum antimicrobial agents, is probably responsible for the emerging of some streptococcal strains (i.e. Streptococcus mitis and Streptococcus pneumoniae in a broad surveillance study)47, as well as some E. faecium isolates48, which became intrinsically resistant to streptogramins, regardless of the prior use of antibiotics belonging to the same of similar classes47. In these last reports, the concomitant resistance to all available glycopeptides indicated the novel oxazolidinone linezolid as the only potentially effective alternative therapeutic choice23,42,43,46,48.

The adverse events registered upon administration of quinupristin/dalfopristin include predominantly gastrointestinal tract disturbances (nausea, vomiting, and diarrhea), followed by hyperbilirubinemia, cutaneous rash, and diffuse arthromyalgia7,17,43. These last signs and symptoms seem to be more frequent among hepatopathic and organ transplanted patients, and those treated with cyclosporin, although the mechanism of action of these adverse events still remains under investigation49. The administration of quinupristin/dalfopristin in hospital setting, and through central i.v. lines and appropriate fluid dilution, significantly reduces the risk of local thrombophlebitis. Notwithstanding the practical difficulties related to drug administration, in an United States pilot study a quinupristin/dalfopristin treatment has been administered to 37 patients suffering from osteomyelitis, bacteremia, abscess and cellulitis due to E. faecium, S. aureus, and coagulasenegative Staphylococci, all in outpatient setting, as a prosecution or completion of treatment schedules initiated at the hospital, relying on peripherally-inserted central catheters, or tunnellized catheters: 16 subjects out of 37 (43.2%) showed mild-to-moderate local intolerance, during or after i.v. drug infusion50. From a metabolic point of view, both quinupristin and dalfopristin are modified by the liver activity into different main derivatives, which contribute to its antimicrobial action, thanks to their intrinsic activities, and the maintained synergistic acitvity between themselves and the administered molecules7,17,23. Since the metabolism is principally carried out by the hepatic cytochrome system P450, pharmacological interactions with all drugs which interact with the same detoxification system are expected: in particular, quinidine, lidocaine, nifedipine, terfenadine, astemizole, cisapride, disopiramide, and midazolam, as well as drugs which might prompt a QT interval prolongation (antiarrhitmics, neuroleptics, antidepressive drugs, antimalaric compounds, fluoroquinolones, azole antifungals, and macrolides). A careful monitoring of serum levels (when possible) or clinical effect of these last drugs, and special clinical attention delivered to possible adverse events, and eventual need of dosage adjustment, are therefore warranted for patients who require a continued administration of the above-mentioned drugs, concurrently with that of quinupristin/dalfopristin7,17. In the event of critically ill or transplanted patients, preliminary demonstrations of possible interactions of quinupristin/dalfopristin and cyclosporin have been reported, so that serum cyclosporin levels and drug dosage adjustments deserve careful attention17,40.

Linezolid: the oxazolidinone derivative

Linezolid represent the first representative of a novel class of oxazolidinone derivatives, which encompasses an effective activity spectrum which covers all the most important Grampositive organisms, including those resistant to methicillin and glycopeptides. The oxazolidinones have a unique mode of action, which inhibits the start of bacterial protidosynthesis by preventing the formation of the ternary complex at 70S ribosomal subunit3, by an apparent double blockade of both the 50S and the 30S bacterial ribosomal subunits. The particular mechanisms of action of linezolid, which includes a blockade of ribosomal assemblation which occurs before the initiation of bacterial protein synthesis3, makes very improbable the emerging of cross resistance with other molecules. However, this phenomenon has been anecdotally reported until now, especially after long-term and low-dosage courses, and appears to be extremely rare among Staphylococci51, although linezolid-resistant Enterococci have been occasionally reported in intensive care units52.

From a pharmacokinetic point of view, linezolid is protein-bound for around 30%, and favorably penetrates into a broad variety of tissues, such as fat, bone, muscle, cerebrospinal fluid and wound sites, beyond the known, elevated penetration into the lungs and the entire respiratory tract3,46. The drug metabolism is not affected by the cytochrome P450 pathway, so that drug-drug interactions at this setting are not expected. On the other hand, urine concentrations is low (around 30% of plasma levels). I.v. formulation requires slow (30-120 minutes) infusion. Being formulated for both the i.v. and the oral route, linezolid retains a 100% bioavailability even after oral administration3,46,53, and regardless of meals, therefore making it easier to exploit the oral administration for switching therapies and early discharge from the hospital of patients who can be effectively followed on outpatient or Day-Hospital basis, and also to start therapy with oral route, whenever possible. These aspects have multiple favorable consequences, when comprehensive morbidity and mortality rates, overall length of hospitalization, and increased medical expenses are considered, even when compared with the proportionally elevated crude costs of linezolid therapy53. A European study54 considered 227 patients with serious Gram-positive infections, treated with linezolid compared with teicoplanin, and assessed hospital resource use and overall treatment costs. The enrolled patients were randomized according to a 50%-50% ratio to receive either linezolid for 7 to 28 consecutive days (initially by i.v. route, but with the possibility to shift to the oral administration as soon as possible), or teicoplanin (initially by i.v. administration, potentially followed by i.m. route)54. The mean i.v. treatment duration was 3.2-day shorter in the linezolid group (6.3 versus 9.5 days), tending to lead to a reduction of overall hospitalization costs, when comparing the novel oxazolidinone linezolid even against the most expensive glycopeptide teicoplanin53,54.

After expanded-access programs which included seriously ill patients suffering from multidrug-resistant, Gram-positive infections in situations also including bacteremia (46% of 796 cases), endocarditis, and line-related infection55 at its first approval in the United States, linezolid was initially registered for the treatment of both community-acquired and nosocomial pneumonia, uncomplicated and complicated skin and soft tissue infections (including diabetic foot infections and surgical site infection), and infectious caused by methicillin-and vancomycin-resistant Staphylococci and Enterococci, and penicillin- and macrolide-resistant Streptococci and Pnemococci, including episodes complicated by bacteremia53. Since ribosomal mutations have been detected that produce resistance against linezolid, longitudinal surveillance surveys remain needed to strictly monitor this phenomenon. The 2003 annual appraisal of potency and spectrum (ZAAPS) program compared MIC results of linezolid with 13-15 comparator agents in over 8,000 isolates56, and confirmed a maintained 99.93% linezolid susceptibility rate of tested Gram-positive organisms. The recently published continuation of this study until 200457 confirmed this figure on 20,158 overall tested isolates, pointing out that 99.5% of isolated S. aureus organisms had a MIC90 value of linezolid ranging from 0.5 to 2 mg/L, with only one isolate tested at 4 mg/L57.

In particular, when considering complicated skin and soft tissue infections, a randomized, open study has been conducted on 1,200 hospitalized patients with ascertained or suspected methicillin-resistant staphylococcal isolates58. The most common disorders were represented by severe cellulitis (46%), cutaneous abscess (26%), and surgical wound infection (11%). In the intention-to-treat comparison between linezolid (given at 600 mg i.v. twice daily) and vancomycin (at 1 g i.v. twice daily), the cure rate was 92.2% versus 88.5% respectively (p=0.057), while involved pathogens included methicillin-resistant Staphylococci in 42% of cases, followed by methicillin-sensitive Staphylococci (29%), and coagulasenegative Staphylococci (8%)58. When considering methicillinresistant staphylococcal infections only, a greater percentage of success rate was obtained with linezolid over vancomycin (88.6% versus 66.9% respectively; p<.001), paralleling the better bacteriological success rate of linezolid over vancomycin (p<.001)58. In a subset of the above-mentioned study including surgical site infections presumably due to methicillin-resistant Staphylococci58, although the clinical cure rate proved similar, linezolid obtained a greater bacteriological eradication rate (p<.008), and a higher rate of microbiological cure (p<.003)58. In an open study of complicated staphylococcal skin and soft tissue infections, oral linezolid and i.v. vancomycin were compared at the same dosage regimens for 7-21 days, and a higher clinical response was obtained with linezolid (p<.02), associated with a lower rate of failure requiring also surgical amputation (p<.02)59.

Pneumonia was assessed in two large, multicentre, randomized, double-blind trials conducted on 1,019 patient on the whole, affected by a nosocomial pneumonia presumably due to Gram-positive organisms. In these studies, an empirical linezolid treatment tested comparable to a vancomycin one, from an efficacy and safety point of view60,61. In a subgroup which included 123 patients with a confirmed hospital-acquired methicillin-resistant staphylococcal pneumonia, a greater survival rate (84% versus 62%; p=.02), and clinical cure (62% versus 21%; p<.001) were achieved with linezolid, compared with vancomycin. In another study62 which compared the efficacy of linezolid and teicoplanin in 430 patients with ascertained or presumed Gram-positive infections, linezolid proved as effective as teicoplanin in patients suffering from pneumonia (96% versus 93%), but infections complicated by bacteremia had a greater response rate when linezolid was administered (88.5% versus 56.7%; p<.001)62. The increasing recognition of the clinical potential of linezolid led to the inclusion of this novel agent in the treatment guidelines of the American Thoracic Society and those of the Infectious Disease Society of America, as empirical, initial choice for patients with a suspected nosocomial pneumonia caused by methicillin-resistant Staphylococci, or where the global prevalence of these organisms is elevated53,63.

Smaller patient series and anecdotal case reports of linezolid use in other relevant infectious processes are increasing day by day. The excellent tissue penetration makes this oxazolidinone drug extremely promising for the approach to difficult-to-treat endocarditis with or without bacteremia64, central nervous system infections65,66, and bone and joint infections67, caused by resistant Gram-positive cocci. Moreover, pediatric experiences are increasing10,46.

A combination therapy of linezolid with other antimicrobial compounds is indicated when the co-existence of Gramnegative pathogens is ascertained or suspected. Moreover, a very interesting activity of linezolid has been demonstrated both in vitro and in vivo against susceptible and especially multi-drug resistant Mycobacterium tuberculosis68-70, and a synergistic activity may be exploited with a broad spectrum of fluoroquinolones71, although the clinical significance of those associations needs to be clinically confirmed.

Interestingly, a recent study also pointed out a significant reduction of acute-phase proinflammatory cytokines from human peripheral mononuclear cells72 when linezolid was administered, thus confirming an immunomodulating response to linezolid use.

From a tolerability point of view, within the maximum allowed treatment duration of 28 days, linezolid showed a favorable safety profile, as showed in a very extensive methanalisis of 2.046 adult patients enrolled in seven different comparative, controlled studies53,73. The 85% of reported adverse events was mild-to-moderate in intensity: the most common clinical events were diarrhea (4.0% to 5.3%), nausea (3.3% to 3.5%), and headache (1.9% to 2.7%), while the most common laboratory disturbances included anemia and thrombocytopenia73. During the post-marketing surveillance, sparse cases of peripheral neuritis (also including optical neuritis), and lactic acidosis were anecdotally reported, especially when linezolid treatment was extended beyond four weeks74,75. Also myelotoxic effects (whose pathogenesis still remains unknown) were associated with prolonged treatment durations, although some reports showed a somewhat earlier appearance. However, the risk of clinically significant thrombocytopenia as assessed on 686 patients with nosocomial pneumonia treated with linezolid for at least five days was limited to 6.4% (compared with 7.7% observed in the comparative vancomycin group)76. When chronic or acute kidney insufficiency or hemodyalisis or hemofiltration are of concern, no correction of linezolid dosage are needed77, as opposed to the limitations occurring when vancomycin or teicoplanin are administered46,78. Therefore, the selection of linezolid appears indicated when a co-existing renal insufficiency may hamper the use of glycopeptides and other drugs with an increased renal toxicity. Finally, the well know activity exerted by linezolid on the inhibition of monoamine oxidase may prompt potential adverse drug-drug interactions with a broad range of antidepressant medications, which has to be taken in careful account when facing patients who receive complex, multiple pharmacological treatments79,80.

Although the efficacy and safety of linezolid have been well demonstrated in severe, high-risk infections where multiresistant Gram-positive cocci are involved, and multiple pharmacoeconomic appraisals show that the availability of a bioequivalent oral formulation and a rapid shift to an oral route of administration may effectively conterbalance the more elevated crude costs compared with older glycopeptides like vancomycin, however further controlled clinical trials are strongly needed to expand the indications of this promising antibiotic, and to check carefully its tolerability in more extensive patient populations and baseline conditions (i.e. extreme life ages, comorbidity including diabetes mellitus, drug-drug interactions, and so on)53. A ponderate prescription limited to selected cases of serious, resistant Gram-positive infections, associated with improved standards of control and monitoring of nosocomial infections, are expected to add significantly to the long-term planning of effective guidelines of prescription, and large-scale resource allocation, in the optimization process of the management of multiresistant Gram-positive infection in critical settings.

The lipopeptide daptomycin

The novel lipopeptide antibiotic daptomycin is a cyclic amino acid compound of particular interest, also due to its unique mechanism of action, encompassing a rapid concentration-dependent killing and an effective bactericidal activity, mainly exerted by its lipophilic tail, which inserts itself into the cytoplasmic membrane of Gram-positive pathogens, and dysrupts it through rapid a depolarization process81-84, which is responsible of a bactericidal activity faster than comparators belonging to glycopeptides, oxazolidinones, and streptogramins, and is also corroborated by an appreciable postantibiotic effect81.

Since the spectrum of activity of daptomycin is extended to include key Gram-positive pathogens (regardless of their resistance status to other compounds, and including all glycopeptide-resistant staphylococci and enterococci, and multiresistant pneumococci and streptococci)81-86, this novel compound adds significantly to the still reduced spectrum of antimicrobial weapons for the treatment of serious Gram-positive infections, and it is also effective against several anaerobes (Clostridium and Propionibacterium spp.), while no activity is expressed against Gram-negative pathogens81. The in vitro potency against multiresistant Gram-positive cocci is comparable to that of linezolid and dalfopristin/quinupristin, at the proposed breakpoint of <2 mg/L, but with a more rapid bactericidal activity85,87. Some synergistic activity has been shown against vancomycin-resistant Enterococci with the use of rifampicin, and at a lesser extent with ampicillin and tobramycin84,85,88. Although the possibility to generate in vitro resistant bacterial strains is apparently negligible, also due to the lack of transferable elements85, however anecdotal reports emerged since early experiences, after prolonged exposure of Staphylococci to the relevant drug89.

At the dosage of 4 mg/Kg i.v. once daily, daptomycin has been shown to be efficacious in two blinded trials of complicated skin and soft tissue infections conducted on even 1,092 patients, which led to a clinical success rate similar for daptomycin and comparators (either vancomycin or penicillinase-resistant penicillins), but with a rapid bactericidal activity82-84,86,87,90,91. Among patients successfully treated with i.v. daptomycin, 63% required only 4-7 days of therapy, as opposed to 33% of comparator-treated patients (p<.0001)90.

Until now, daptomycin is therefore approved for the management of complicated skin-skin structure infections, including infected surgical wounds, and complications of burns and diabetic foot81,84,86,87,90. The potential for shorter course regimens should demonstrate faster resolution rates, and may decrease the risk of resistance development, toxicity, and treatment-related direct and indirect costs. Unfortunately the modest daptomycin penetration into the lung tissue and the local drug competition with surfactant (which is implicated in sequestering this particular lipopeptide drug), poses this novel compound at an elevated risk of clinical failure in the treatment of respiratory infections81,86, so that no indication is expected to date in this last setting.

On the other hand, a daptomycin dose of 6 mg/Kg is under investigation for the management of endocardits and bacteremia81,84,87,92. In a recently published case series of 31 patients with bacteremia and endocarditis, i.v. daptomycin at 4-6 mg/day tested safe and effective, even when considering the elevated prevalence of vancomycin-resistant enterococci and methicillin-resistant Staphylococcus aureus (11 cases each), and taking into account of the proportionally prolonged duration of treatment (at least 14 days for bacteremia, 22-43 days for endocarditis)92.

Finally, a very recent experimental, animal model showed the superior efficacy of daptomycin over vancomycin in treating methicillin-susceptible staphylococcal meningitis93: the effect was attributed to the higher time-kiling assays and the better penetration of daptomycin into inflamed meninges (around 5%)93.

From a pharmacokinetic and pharmacodynamic point of view, both the peak concentration/MIC ratio and the 24-hour AUC/MIC ratio have a strong correlation with the in vivo efficacy of daptomycin. The protein binding of the drug is very elevated (90-94%), but a linear pharmacokinetic has been demonstrated. Once-daily administration optimizes the pharmacodynamics of daptomycin, also minimizing the initially observed musculoskeletal side effects. At the dose of 4 mg/kg, in adult subjects the reached Cmax is around 58 mg/L, and the half life lasts 8.1±1.0 hours. Daptomycin has no appreciable hepatic metabolism and does not act on the cytochrome P450 enzymatic cascade, so that relevant drug-drug interactions are not expected. Only temporary suspension of HMG-CoA reductase inhibitors is recommended during eventual daptomycin co-administration, in order to reduce the risk of increased serum creatin-phosphokinase (CPK), and the eventual skeletal muscle toxicity. Unchanged (active) drug may be recovered from urines over 24 hours at a rate ranging from 53% to 60%81, so that a dosage adjustement is required when kidney insufficiency is of concern, mainly by increasing the interval of administration. Safety and efficacy did not change significantly when moderate hepatic insufficiency or obesity were of concern81,84. In an extensive clinical trial that included patients treated for complicated skin-skin structure infections90, adverse events occurred in slightly more than 2% of subjects who received daptomycin or comparators: constipation, nausea, injection site reaction, and headache were the most frequently reported events, without any difference found with comparator drugs90. Hypersensitivity reactions and myopathy occurred very infrequently, but once-weekly serum CPK levels monitoring remains recommended84.

The glycylcline tigecyclin

The glycylcyclines (with tigecyclin as the first licensed compound of this class) are the representatives of a new class of antimicrobial compounds which may be considered as a long-term awaited evolution of the tetracycline class. Their expanded broad-spectrum activity against both Gram-negative and Gram-positive aerobes, anaerobes, and facultative organisms, as well as "atypical" bacteria, is of particular interest. Many pathogenic and clinically relevant organisms are susceptible to tigecyclin, including multiresistant Staphylococci, Streptococci, Pneumococci, and Enterococci. Its very extensive bacteriological spectrum also includes many beta-lactamase producing organisms, often retrieved when a Gram-positive and Gram-negative mixed flora is of concern (i.e. Enterobacteriaceae as a whole)94-97.

The large volume of distribution of tigecyclin leads to an extensive tissue penetration. In association with a very long terminal half-life (around 40 hours), these features allow twice-daily administration. The efficacy of tigecyclin seems to be best predicted by the ratio of the area under the concentration-time curve to the MIC, due to its linear pharmacokinetic profile94,97,98.

Tigecyclin may be administered by i.v. route (at 50 mg, twice daily, preceded by a loading dose of 100 mg, the first day), and until now it has been studied in the management of serious polymicrobial infections, i.e. complicated skin and skin structure infections, surgical wounds, and intrabdominal infections, where it resulted effective and well tolerated in phase III clinical studies, carried our in adults, and non-pregnant women94,96-98. Equivalence to imipenem in intrabdominal infections, and to vancomycin plus aztreonam in skin and soft tissue infections have been preliminarly achieved95,96.

The drug disposition is not affected by age, renal disease, or food, and the limited metabolism encompasses a reduced kidney and liver engagement94,97,98. Besides a low rate of gastrointestinal complaints (nausea, vomiting, and diarrhea), only mild and self-limiting adverse effects on blood chemistry or haematology have been observed98.

In conclusion, this novel drug is likely to find a key role also as a monotherapy in the treatment of mixed infections due to multiresistant pathogens, including beta-lactamase produ-cers and methicillin- and vancomycin-Gram-positive organisms95-97.

Novel glycopeptide antibiotics

This drug class still contains the reference standard antimicrobial choices for methicillin-resistant Gram-positive cocci, i.e. vancomycin and teicoplanin99. Among the novel compounds representing the evolution of this class, the new glycopeptides dalbavancin, oritavancin, telavancin99 and the glycolipodepsipeptide ramoplanin100, appear very promising for an upcoming commercial release in the next few months or years, although there are remaining concerns about the possible microbial resistance spread, based on existing antimicrobial compounds which belong to the same drug class99,101. Chemical modifications resulting in these secondgeneration glycopeptide analogues have however revealed new mechanisms of antibacterial action, and additional properties including pharmacokinetic ones99. In particular, oritavancin is a semi-synthetic glycopeptide with a long half-life (around 150-200 hours), and elevated intracellular penetration, as well as an effective diffusion into the brain. Its bacteriological activity includes vancomycin-resistant Enterococci, methicillin-resistant Staphylococci, vancomycin-intermediate and vancomycin-resistant S. aureus. Telavancin is another glycoptide compound with an half-life which allows once-daily administration. Its dual mechanisms of action (impairment of synthesis of peptidoglycan, and cell wall lipid structures) is a distinguishing property. Finally, ramoplanin is a glycolipopeptide, with an excellent activity against Gram-positive cocci and bacilli, including those which became resistant to currently glycopeptides. Its endothelial toxicity is a present limitation for i.v. delivery, so that ramoplanin is presently studies for the management of difficult-to-treat intestinal Clostridium difficile.

The first agent in the pipeline appears to be the semisynthet-ic lipoglycopeptide dalbavancin, developed for once-weekly i.v. treatment of serious Gram-positive infections, including a wide range of glycopeptide-resistant organisms. The mechanism of action replicates that of other glycopeptides, but dalbavancin resulted more potent in vitro102. It has a very elevated in vitro activity against a variety of Gram-positive organisms, save vancomycin-resistant Enterococci possessing the VanA gene101,102.

During Phase II-III clinical trials, dalbavancin proved effective and safe in the management of skin and skin-structure infections, and catheter-related bloodstream infections101. The only available double-blind randomized clinical trial compared i.v. dalbavancin (1 g at day 1, 500 mg at day 8), with linezolid (600 mg i.v. or orally twice daily for 14 days), in the treatment of complicated skin-skin structure infections caused by suspected methicillin-resistant S. aureus strains. Dalbavancin and linezolid proved comparable from a clinical point of view (leading to a 88.9% and 91.2% success rate, respectively), and microbiological success was attained in both arms in over 85% of cases, although the rate of methicillin resistance was retrospectively recognized around 51% of isolated strains.

Thanks to its prolonged half-life (6-10 days) a once-weekly i.v. administration becomes possible. Pharmacokinetic analyses conduced on 532 patients, the majority of them treated with 1000 mg-dose on day 1 and 500 mg-dose on day 8, showed a dual-compartment model, with a clearance influenced by body surface area and creatinine clearance. No evidence of metabolic substrates, inhibitors, or inducers of the liver cytochrome P450 was found101,103, thus eliminating the risk of competing drugs at the same hepatic site. At a preliminary assessment, adverse events were very mild and limited in frequency: pyrexia, headache, nausea, diarrhea and other gastrointestinal disturbances were the most commonly described adverse events101.

Among adverse events, gastrointestinal complaints seem to represent the most frequent occurrence. In the quoted randomized trial104, linezolid showed a slightly more elevated rate of overall adverse events compared with dalbavancin (32.2% versus 25.4% of enrolled subjects).

Novel cephalosporins, fluoroquinolones, and other agents under advanced development

Among the more promising agents under advanced development against multiresistant Gram-positive cocci, we can quote novel cephalosporins (i.e. BAL-9141 and RWJ-54428), which are expected to overcome methicillin resistance82,105.

Furthermore, among the topoisomerase inhibitors, several fluoroquinolones are awaited, including gemifloxacin (which recently entered commercialization), sitafloxacin, and especially garenoxacin82,105.

Finally, when considering the class of inhibitors of bacterial protein synthesis, the ketolides telitromycin and cetromycin, and the novel oxazolidinones (further to linezolid), are very promising agents for the fight against severe and life-threatening resistant Gram-positive infections18,23,83,100,105.

Conclusions and Outlook

The proportionally recent availability of quinupristin/dalfopristin and that of linezolid determined significant changes in the scenario of the management of severe infections due to multiresistant Gram- positive pathogens, usually acquired at the hospital and by a somewhat immunocompromised host8,18,83,105,106-108.

A recent, extensive survey conducted on 258 Gram-positive bacterial organisms isolated from blood cultures at a United States cancer reference centre allowed to compare the in vitro acitivity of daptomycin, linezolid, and quinupristin/dalfopristin107. Vancomycin-resistant Enterococci represented the largest proportion of tested organisms (32%), followed by methicillin-resistant coagulase-negative Staphylococci (23%), and vancomycin-sensitive Enterococci (14%). Through a detailed analysis of both MIC and MBC values, daptomycin showed a bactericidal activity against the majority of tested organisms, by killing almost 100% of bacteria within six hours. Quinupristin/dalfopristin was bactericidal against Staphylococci and bacteriostatic against the majority of Enterococci. Linezolid was bacteriostatic against all evaluated organisms, but a correlation between the in vitro features and the clinical outcome demonstrated an elevated potential of all these novel compounds107,108, which now deserve controlled studies in the setting of the management and prevention of serious infection in the immunocompromised host, including HIV/AIDS patients, subjects with hematologic malignancies or solid tumors, and those undergoing bone marrow or organ transplantation, or major surgery and hospitalization in intensive care units.

Highlights

- In the meantime, the overall understanding of the epidemiology and virulence of community-acquired multiresistant Gram-positive pathogens continued to grow, leading to major attention devoted to developing compounds, but also a re-examination of many older, but still active agents (including long-acting tetracyclynes, fluoroquinolones, rifampicin, cotrimoxazole, and clindamycin)14,106-108, which certainly retain some non-negligible therapeutic role, especially when a synergistic activity can be demonstrated18. Moreover, through novel laboratory assays like the so-called E-test synergy and time-kill methods will perhaps become possible to measure to extent of synergistic activity between differently combined molecules against glycopeptide-resistance gram-positive cocci (i.e. daptomycin and rifampicin against multiresistant Enterococci)81,105-108.

- As summarized above, during the next future the therapeutic research promises the development of novel compounds aimed at intervening favorably against the unavoidable increase of drug resistance frequency and levels against the present reference compounds (i.e. the glycopeptides vancomycin and teicoplanin)99, and later the two abovementioned recent molecules, i.e. quinupristin/dalfopristin and linezolid2,7,8,18,83. On the other hand, the clinical use of the streptogramin combination quinupristin/dalfopristin, which retains optimal activity against methicillin-resistant S. aureus and vancomycin-resistant E. faecium, is limited because its need to be administered in large volume of fluids82, while its activity in severe pneumonia is somewhat lower14. The oxazolidinone linezolid is active against methicillin-resistant Staphylococci and glycopeptide-resistant Enterococci, but resistant organisms and sparse treatment failures have been reported82, while unexpected tolerability issues are becoming of concern74.

- Furthermore, we have to remind that in many cases the most relevant therapeutic intervention for complicated Gram-positive abscesses, cellulitis, complicated skin and soft tissue diseases, but also osteomyelitis, infected bone and joint prostheses, and brain abscesses, remains an adequate surgical drainage and curettage of purulent fluid collections, and the elimination of affected, necrotic tissue2,9,82. Subsequently, the antimicrobial selection should be driven by disease severity, susceptibility patterns, clinical response to therapy, and also related costs (seen from a comprehensive point of view) (Table 3). Also special population such as the pediatric and neonatal ones9, are going to benefit from specifically-designed trials, which could address in the next future the major issues in the setting of epidemiology, mechanisms of virulence, continued changes in pathogenicity and antimicrobial susceptibility of involved organisms, and potential use of novel antimicrobial compounds, like daptomycin, glicylcyclines, newer glycopeptides, beta-lactamase-stable cephalosporins, and ketolides10,18 (Table 4).

- Finally, only randomized, comparative assessments of the novel molecules on the market will assist us to plan wellfounded recommendations for the treatment of serious Gram-positive infections, and parallel comprehensive pharmacoeconomic issues should be carefully deserved.

References

1. Karchmer AW. Nosocomial bloodstream infections: organisms, risk factors, and implications. Clin Infect Dis 2000; 31 (Suppl. 4): S139-S143.         [ Links ]

2. Menichetti F. Current and emerging serious Gram-positive infections. Clin Microbiol Infect 2005; 11 (Suppl. 3): 22-28.         [ Links ]

3. Stevens DL, Dotter B, Madaras-Kelly A. A review of linezolid: the first oxazolidinone antibiotic. Expert Rev Anti-Infective Ther 2004; 2: 51-59.         [ Links ]

4. Kollef MH. Antibiotic management of ventilator-associated pneumonia due to antibiotic-resistant gram-positive bacterial infection. Eur J Clin Microbiol Infect Dis 2005; 24: 794-803.         [ Links ]

5. Cunha BA. Methicillin-resistant Staphylococcus aureus: clinical manifestations and antimicrobial therapy. Clin Microbiol Infect 2005; 11 (Suppl. 4): 33-42.         [ Links ]

6. Wenzel RP, Edmond MB. Vancomycin-resistant Staphylococcus aureus: infection control considerations. Clin Infect Dis 1998; 27: 245-249.         [ Links ]

7. Linden PK. Treatment options for vancomycin-resistant enterococcal infections. Drugs 2002; 62: 425-41.         [ Links ]

8. Segreti J. Efficacy of current agents used in the treatment of Gram-positive infections and the consequences of resistance. Clin Microbiol Infect 2005; 11 (Suppl. 3): 29-35.         [ Links ]

9. Ladhani S, Garbash M. Staphylococcal skin infections in children: rational drug therapy recommendations. Paediatr Drugs 2005; 7: 77-102.         [ Links ]

10. Marcinak JF, Frank AL. Epidemiology and treatment of community-associated methicillin-resistant Staphylococcus aureus in children. Expert Rev Anti Infect Ther 2006; 4: 91-100.         [ Links ]

11. Pallares R, Fenoll A, Linares J. The epidemiology of antibiotic resistance in Streptococcus pneumoniae and the clinical relevance of resistance to cephalosporins, macrolides, and quinolones. Int J Antimicrob Agents 2003; 22 ( Suppl. 1): S15-S26.         [ Links ]

12. Lynch JP 3rd, Zhanel GG. Escalation of antimicrobial resistance among Streptococcus pneumoniae: implications for therapy. Semin Respir Crit Care Med 2005; 26: 575-616.         [ Links ]

13. Edmond MB, Wallace SE, McClish DK, et al. Nosocomial bloodstream infections in United States hospitals. A three-year analysis. Clin Infect Dis 1999; 29: 239-244.         [ Links ]

14. Manfredi R, Nanetti A, Morelli S, Valentini R, Calza L, Chiodo F. Sensitivity of methicillin-resistant Staphylococcus aureus (MRSA) to antimicrobial agents other than beta-lactams: profile and trend, 1998-2003. Isr Med Assoc J 2004; 6: 191-192.         [ Links ]

15. Cuevas O, Cercenado E, Vindel A, et al. Evolution of antimicrobial resistance of Staphylococcus spp. In Spain: five nationwide prevalence studies, 1986 to 2002. Antimicrob Agents Chemother 2004; 48: 4240-4245.         [ Links ]

16. Hsueh PR, Chen WH, Teng LJ, Luh KT. Nosocomial infections due to methicillin-resistant Staphylococcus aureus and vancomycin-resistant enterococci at a University Hospital in Taiwan from 1991 to 2003: resistance trends, antibiotic usage and in vitro activities of newer antimicrobial agents. Int J Antimicrob Agents 2005; 26: 43-49.         [ Links ]

17. Lamb HM, Figgitt DP, Faulds D. Quinupristin/dalfopristin: a review of its use in the management of serious gram-positive infections. Drugs 1999; 58: 1061-1097.         [ Links ]

18. Akins RL, Haase KK. Gram-positive resistance: pathogens, implications, and treatment options: insights from the Society of Infectious Diseases Pharmacists. Pharmacotherapy 2005; 25: 1001-1010.        [ Links ]

19. Nicoletti G, Bonfiglio G, Bartoloni A, et al. Distribution and antibiotic resistance of isolates from lower respiratory tract and blood cultures from patients in three Italian intensive care units: a 2-year comparison. Int J Antimicrob Agents 2000; 15: 265-269.         [ Links ]

20. Marshall SA, Wilke WW, Pfaller MA, Jones EN. Staphylococcus aureus and coagulase-negative staphylococci from blood stream infections: frequency of occurrence, antimicrobial susceptibility, and molecular (mecA) characterization of oxacillin resistance in the SCOPE program. Diagn Microbiol Infect Dis 1998; 30: 205-214.         [ Links ]

21. Anonymous. Reduced susceptibility of Staphylococcus aureus to vancomycin-Japan 1996. MMWR 1997; 46: 624-626.         [ Links ]

22. Salyers AA, Gupta A, Wang Y. Human intestinal bacteria as reservoirs for antibiotic resistance genes. Trends Microbiol 2004; 12: 412-416.         [ Links ]

23. Hershberger E, Donabedian S, Konstantinou K, Zervos MJ. Quinupristin-dalfopristin resistance in gram-positive bacteria: mechanism of resistance and epidemiology. Clin Infect Dis 2004; 38: 92-98.         [ Links ]

24. Kim HB, Lee B, Jang HC, et al. A high frequency of macrolide-lincosamidestreptogramin resistance determinants in Staphylococcus aureus isolated in South Korea. Microb Drug Resist 2004; 10: 248-254.         [ Links ]

25. Petinaki E, Spiliopoulou I, Maniati M, Maniatis AN. Emergence of Staphylococcus hominis strains expressing low-level resistance to quinupristin/dalfopristin in Greece. J Antimicrob Chemother 2005; 55: 811-812.         [ Links ]

26. Furlong WB, Rakowski TA. Therapy with RP 59500 (quinupristin/dalfopristin) for prosthetic valve endocarditis due to enterococci with VanA/VanB resistance patterns. Clin Infect Dis 1997; 25: 163-164.         [ Links ]

27. Larkin J, Busciglio L, Fontanet H, Gamouras G. Staphylococcus epidermidis endocarditis treated with RP 59500 (quinupristin/dalfopristin). Clin Infect Dis 1998; 26: 1239-1240.         [ Links ]

28. Matsumura S, Simor AE. Treatment of endocarditis due to vancomycin-resistant Enterococcus faecium with quinupristin/dalfopristin, doxycycline, and rifampin: a synergistic drug combination. Clin Infect Dis 1998; 27: 1554-1556.         [ Links ]

29. Thompson RL, Lavin B, Talbot GH. Endocarditis due to vancomycin-resistant Enterococcus faecium in an immunocompromised patient: cure by administering combination therapy with quinupristin/dalfopristin and high-dose ampicillin. South Med J 2003; 96: 818-820.         [ Links ]

30. Summers M, Misenhimer GR, Antony SJ. Vancomycin-resistant Enterococcus faecium osteomyelitis: successful treatment with quinupristin-dalfopristin. South Med J 2001; 94: 353-355.         [ Links ]

31. Garey KW, Tesoro E, Muggia V, Pasquier O, Rodvold KA. Cerebrospinal fluid concentrations of quinupristin/dalfopristin in a patient with vancomycin-resistant Enterococcus faecium ventriculitis. Pharmacotherapy 2001; 21: 748-750.         [ Links ]

32. Tan TY, Pitman I, Penrose-Stevens A, Simpson BA, Flanagan PG. Treatment of a vancomycin-resistant Enterococcus faecium ventricular drain infection with quinupristin/dalfopristin and review of the literature. J Infect 2000; 41: 95-97.         [ Links ]

33. Williamson JC, Glazier SS, Peacock JE Jr. Successful treatment of ventriculostomy-related meningitis caused by vancomycin-resistant Enterococcus with intravenous and intraventricular quinupristin/dalfopristin. Clin Neurol Neurosurg 2002; 104: 54-56.         [ Links ]

34. Carretto E, Barbarini D, Locatelli F, et al. Vancomycin-resistant Enterococcus faecium infection in three children given allogeneic hematopoietic stem cell transplantation: clinical and microbiological features. Haematologica 2000; 85: 1158-1164.         [ Links ]

35. Gray JW, Darbyshire PJ, Beath SV, Kelly D, Mann JR. Experience with quinupristin/dalfopristin in treating infections with vancomycin-resistant Enterococcus faecium in children. Pediatr Infect Dis J 2000; 19: 234-238.         [ Links ]

36. Fagon J, Patrick H, Haas DW, et al. Treatment of gram-positive nosocomial pneumonia. Prospective randomized comparison of quinupristin/dalfopristin versus vancomycin. Am J Respir Crit Care Med 2000; 161: 753-762.         [ Links ]

37. Nichols RL, Graham DR, Barriere SL, et al. Treatment of hospitalized patients with complicated Gram-positive skin and skin structure infections: two randomized, multicentre studies of quinupristin/dalfopristin versus cefazolin, oxacillin or vancomycin. J Antimicrob Chemother 1999; 44: 263-273.         [ Links ]

38. Moellering RC, Linden PK, Reinahrdt J, et al. The efficacy and safety of quinupristin/dalfopristin for the treatment of infections caused by vancomycin-resistant Enterococcus faecium. J Antimicrob Chemother 1999; 44: 251-261.         [ Links ]

39. Mundlein E, Von Baum H, Geiss HK, Springsklee M, Zeier M, Andrassy K. Life-threatening infection with multiresistant Staphylococcus epidermidis in a patient with end-stage renal disease: cure with chloramphenicol and quinupristin/dalfopristin (RP 59500). Infection 1997; 25: 252-254.        [ Links ]

40. Schwenger V, Mundlein E, Dagrosa EE, et al. Treatment of life-threatening multiresistant staphylococcal and enterococcal infections in patients with endstage renal failure with quinupristin/dalfopristin: preliminary report. Infection 2002; 30: 257-261.         [ Links ]

41. Stamatakis MK, Richards JG. Interaction between quinupristin/dalfopristin and cyclosporine. Ann Pharmacother 1997; 31: 576-578.         [ Links ]

42. Scotton PG, Rigoli R, Vaglia A. Combination of quinupristin/dalfopristin and glycopeptide in severe methicillin-resistant staphylococcal infections failing previous glycopeptide regimens. Infection 2002; 30: 161-163.         [ Links ]

43. Raad I, Hachem R, Hanna H, et al. Prospective, randomized study comparing quinupristin-dalfopristin with linezolid in the treatment of vancomycin-resistant Enterococcus faecium infections. J Antimicrob Chemother 2004; 53: 646-649.         [ Links ]

44. Brown J, Freeman BB. Combining quinupristin/dalfopristin with other agents for resistant infections. Ann Pharmacother 2004; 38: 677-685.         [ Links ]

45. Sgarabotto D, Cusinato R, Narne E, et al. Synercid plus vancomycin for the treatment of severe methicillin-resistant Staphylococcus aureus and coagulase-negative staphylococcal infections: evaluation of 5 cases. Scand J Infect Dis 2002; 34: 122-126.         [ Links ]

46. Eliopoulos GM. Quinupristin/dalfopristin and linezolid: evidence and opinions. Clin Infect Dis 2003; 36: 473-481.         [ Links ]

47. Kugler KC, Denys GA, Wilson ML, Jones RN. Serious streptococcal infections produced by isolates resistant to streptogramins (quinupristin/dalfopristin): case reports from the SENTRY antimicrobial surveillance program. Diagn Microbiol Infect Dis 2000; 36: 269-272.         [ Links ]

48. McNeil SA, Clark NM, Chandrasekar PH, Kauffman CA. Successful treatment of vancomycin-resistant Enterococcus faecium bacteremia with linezolid after failure of treatment with synercid (quinupristin/dalfopristin). Clin Infect Dis 2000; 30: 403-404.         [ Links ]

49. Carver PL, Whang E, VandenBussche HL, Kauffman CA, Malani P. Risk factors for arthralgias or myalgias associated with quinupristin/dalfopristin therapy. Pharmacotherapy 2003; 23: 159-164.         [ Links ]

50. Rehm SJ, Graham DR, Srinath L, Prokocimer P, Richard MP, Talbot GH. Successful administration of quinupristin/dalfopristin in the outpatient setting. J Antimicrob Chemother 2001; 47: 639-645. *Analysis of the possibility to administer quinupristin/dalfopristing in outpatient settings.         [ Links ]

51. Meka VG, Gold HS. Antimicrobial resistance to linezolid. Clin Infect Dis 2004; 39: 1010-1015.         [ Links ]

52. Herrero IA, Issa NC, Patel R. Nosocomial spread of linezolid-resistant, vancomycin-resistant Enterococcus faecium. N Engl J Med 2002; 346: 867-869.         [ Links ]

53. Moellering RC Jr. Linezolid: the first oxazolidinone antimicrobial. Ann Intern Med 2003; 138: 135-142.         [ Links ]

54. Nathwani D, Li JZ, Balan DA, et al. An economic evaluation of a European cohort from a multinational trial of linezolid versus teicoplanin in serious Grampositive bacterial infections: the importance of treatment setting in evaluating treatment effects. Int J Antimicrob Agents 2004; 23: 315-324.         [ Links ]

55. Birmingham MC, Rayner CR, Meagher AK, Flavin SM, Batts DH, Schentag JJ. Linezolid for the treatment of multidrug-resistant, Gram-positive infections: experience from a compassionate-use program. Clin Infect Dis 2003; 36: 159-168.         [ Links ]

56. Anderegg TR, Sader HS, Fritsche TR, Ross JE, Jones RN. Trends in linezolid susceptibility patterns: report from the 2002-2003 worldwide Zyvox annual appraisal of potency and spectrum (ZAAPS) program. Int J Antimicrob Agents 2005; 26: 13-21.         [ Links ]

57. Jones RN, Ross JE, Fritsche TR, Sader HS. Oxazolidinone susceptibility patterns in 2004: report from the Zyvox annual appraisal of potency and spectrum (ZAAPS) Program assessing isolates from 16 nations. J Antimicrob Chemother 2006; 57: 279-287.         [ Links ]

58. Weigelt J, Itani K, Stevens D, Lau W, Dryden M, Knirsch C. Linezolid versus vancomycin in treatment of complicated skin and soft tissue infections. Antimicrob Agents Chemother 2005; 49: 2260-2266.         [ Links ]

59. Sharpe JN, Shively EH, Polk HC Jr. Clinical and economic outcomes of oral linezolid versus intravenous vancomycin in the treatment of MRSA-complicated, lower extremity skin and soft tissue infections caused by methicillinresistant Staphyloccus aureus. Am J Surg 2005; 189: 425-428.         [ Links ]

60. Rubinstein E, Cammarata S, Oliphant T, Wunderink R. Linezolid (PNU-100766) versus vancomycin in the treatment of hospitalized patients with nosocomial pneumonia: a randomized, double-blind multicenter study. Clin Infect Dis 2001; 32: 402-412.         [ Links ]

61. Wunderink RG, Cammarata SK, Oliphant TH, Kollef MH. Continuation of a randomized, double-blind, multicenter study of linezolid versus vancomycin in the treatment of patients with nosocomial pneumonia. Clin Ther 2003; 25: 980-992.         [ Links ]

62. Wilcox M, Nathwani D, Dryden M. Linezolid compared with teicoplanin for the treatment of suspected or proven Gram-positive infections. J Antimicrob Chemother 2004; 53: 335-342.         [ Links ]

63. Craven DE, Palladino R, McQuillen DP. Healthcare-associated pneumonia in adults: management principles to improve outcomes. Infect Dis North Am 2004; 18: 939-962.         [ Links ]

64. Hill EE, Herijgers P, Herregods MC, Peetermans WE. Infective endocarditis treated with linezolid: case report and literature review. Eur J Clin Microbiol Infect Dis 2006; 25: 202-204.         [ Links ]

65. Sabbatani S, Manfredi R, Frank G, Chiodo F. Linezolid in the treatment of central nervous system infections resistant to recommended antimicrobial compounds. Infez Med 2005; 13: 112-119.         [ Links ]

66. Castro P, Soriano A, Eschrich C, Villalba G, Sarasa M, Mensa J. Linezolid treatment of ventriculoperitoneal shunt infection without implant removal. Eur J Clin Microbiol Infect Dis 2005; 24: 603-606.         [ Links ]

67. Manfredi R, Sabbatani S, Chiodo F. Severe staphylococcal knee arthritis responding favourably to linezolid, after glycopeptide-rifampicin failure: a case report and literature review. Scand J Infect Dis 2005; 37: 513-517.         [ Links ]

68. Alcala L, Ruiz-Serrano MJ, Pérez-Fernandez-Turégano C, et al. In vitro activities of linezolid against clinical isolates of Mycobacterium tuberculosis that are susceptible or resistant to first-line antituberculous drugs. Antimicrob Agents Chemother 2003; 47: 416-417.         [ Links ]

69. Erturan Z, Uzun M. In vitro activity of linezolid against multidrug-resistant Mycobacterium tuberculosis isolates. Int J Antimicrob Agents 2005; 26: 78-80.         [ Links ]

70. Fortun J, Martin-Davilla P, Navas E, et al. Linezolid for the treatment of multidrug-resistant tuberculosis. J Antimicrob Chemother 2005; 56: 180-185.         [ Links ]

71. Rodriguez Diaz JC, Ruiz M, Lopez M, Royo G. Synergistic activity of fluoroquinolones and linezolid against Mycobacterium tuberculosis. Int J Antimicrob Agents 2003; 21: 354-356.         [ Links ]

72. Garcia-Roca P, Mancilla-Ramirez J, Santos-Segura A, Fernandez-Avilés M, Calderon-Jaimes E. Linezolid diminishes inflammatory cytokine production from human peripheral blood mononuclear cells. Arch Med Res 2006; 37: 31-35.         [ Links ]

73. Rubinstein E, Isturiz R, Standiford HC, et al. Worldwide assessment of linezolid’s clinical safety and tolerability: comparator-controlled Phase III studies. Antimicrob Agents Chemother 2003; 47: 1824-1831.         [ Links ]

74. Bressler AM, Zimmer SM, Gilmore JL, Somani J. Peripheral neuropathy associated with prolonged use of linezolid. Lancet Infect Dis 2004; 4: 528-531.         [ Links ]

75. Kopterides P, Papadomichelakis E, Armaganidis A. Linezolid use associated with lactic acidosis. Scand J Infect Dis 2005; 37: 153-154.         [ Links ]

76. Nasraway SA, Shorr AF, Kuter DJ, O’Grady N, Le VH, Cammarata SK. Linezolid does not increase the risk of thrombocytopenia in patients with nosocomial pneumonia: comparative analysis of linezolid and vancomycin use. Clin Infect Dis 2003; 37: 1609-1616.         [ Links ]

77. Fiaccadori E, Maggiore U, Rotelli C, et al. Removal of linezolid by conventional intermittent hemodyalisis, sustained low-efficiency dyalisis, or continuous venovenous hemofiltration in patients with acute renal failure. Crit Care Med 2004; 32: 2437-2442.         [ Links ]

78. Lipsky BA, Itani K, Norden C. Treating foot infections in diabetic patients: a randomized, multicenter, open-label trial of linezolid versus ampicillin-sulbactam/amoxicillin-clavulanate. Clin Infect Dis 2004; 38: 17-24.         [ Links ]

79. Clark DB, Andrus MR, Byrd DC. Drug interactions between linezolid and selective serotonin reuptake inhibitors: case report involving sertraline and review of the literature. Pharmacotherapy 2006; 26: 269-276.         [ Links ]

80. Sola CL, Bostwick JM, Hart DA, Lineberry TW. Anticipating potential linezolid-SSRI interactions in the general hospital setting: an MAOI in disguise. Mayo Clin Proc 2006; 81: 330-334.        [ Links ]

81. LaPlante KL, Rybak MJ. Daptomycin-a novel antibiotic against Gram-positive pathogens. Exp Opin Pharmacother 2004; 5: 2321-2331.         [ Links ]

82. Shah PM. The need for new therapeutic agents: what is the pipeline?. Clin Microbiol Infect 2005; 11 (Suppl. 3): 36-42.         [ Links ]

83. Abbanat D, Macielag M, Bush K. Novel antibacterial agents for the treatment of serious Gram-positive infections. Expert Opin Investig Drugs 2003; 12: 379-399.         [ Links ]

84. Rybak MJ. The efficacy and safety of daptomycin: first in a new class of antibiotics for Gram-positive bacteria. Clin Microbiol Infect 2006; 12 (Suppl. 1): 24-32.         [ Links ]

85. Richter SS, Kealey DE, Murray CT, Heilmann KP, Coffman SL, Doern GV. The in vitro activity of daptomycin against Staphylococcus aureus and Enterococcus species. J Antimicrob Chemother 2003; 52: 123-127.         [ Links ]

86. Kern WV. Daptomycin: first in a new class of antibiotics for complicated skin and soft-tissue infections. Int J Clin Pract 2006; 60: 370-378.         [ Links ]

87. Steenbergen JN, Alder J, Thorne GM, Tally FP. Daptomycin: a lipopeptide antibiotic for the treatment of serious Gram-positive infections. J Antimicrob Chemother 2005; 55: 283-288.         [ Links ]

88. Scheetz M, Reddy P, Postelnick M, Flaherty J. In vivo synergy of daptomycin plus a penicillin agent for MRSA? J Antimicrob Chemother 2005; 55: 398-399.         [ Links ]

89. Skiest DJ. Treatment failure resulting from resistance of Staphylococcus aureus to daptomycin. J Clin Microbiol 2006; 44: 655-656.         [ Links ]

90. Arbeit RD, Maki D, Tally FP, Campanaro E, Eisenstein BI. The safety and efficacy of daptomycin for the treatment of complicated skin and skin-structure infections. Clin Infect Dis 2004; 38: 1673-1681.         [ Links ]

91. Lipski BA, Stoutenburgh U. Daptomycin for treating infected diabetic foot ulcers: evidence from a randomized, controlled trial comparing daptomycin with vancomycin or semi-synthetic penicillins for complicated skin and skin-structure infections. J Antimicrob Chemother 2005; 55: 240-245.         [ Links ]

92. Segreti JA, Crank CW, Finney MS. Daptomycin for the treatment of grampositive bacteremia and infective endocarditis: a retrospective case series of 31 patients. Pharmacotherapy 2006; 26: 347-352.         [ Links ]

93. Gerber P, Stucki A, Acosta F, Cottagnoud M, Cottagnoud P. Daptomycin is more efficacious than vancomycin against a mehicillin-susceptible Staphylococcus aureus in experimental meningitis. J Antimicrob Chemother 2006; 57: 720-723.         [ Links ]

94. Meagher AK, Ambrose PG, Grasela TH, Ellis-Grosse EJ. Pharmacokinetic/pharmacodynamic profile for tigecycline-a new glycylcycline antimicrobial agent. Diagn Microbiol Infect Dis 2005; 52: 165-171.         [ Links ]

95. Livermore DM. Tigecycline: what is it, and where should it be used? J Antimicrob Chemother 2005; 56: 611-614.         [ Links ]

96. Scheinfeld N. Tigecycline: a review of a new glycylcyline antibiotic. J Dermatol Treat 2005; 16: 207-212.         [ Links ]

97. Zhanel GG, Karlowsky JA, Rubinstein E, Hoban DJ. Tigecycline: a novel glycylcycline antibiotic. Expert Rev Anti Infect Ther 2006; 4: 9-25.         [ Links ]

98. Rello J. Pharmacokinetics, pharmacodynamics, safety and tolerability of tigecycline. J Chemother 2005; 17 (Suppl. 1): 12-22.         [ Links ]

99. Pace JL, Yang G. Glycopeptides: update on an old successful antibiotic class. Biochem Pharmacol 2006; 71: 968-980.         [ Links ]

100. Guay DR. Oritavancin and tigecycline: investigational antimicrobials for multidrug-resistant bacteria. Pharmacotherapy 2004; 24: 58-68.         [ Links ]

101. Lin SW, Carver PL, DePestel DD. Dalbavancin: a new option for the treatment of gram-positive infections. Ann Pharmacother 2006; 40: 449-460.         [ Links ]

102. Streit JM, Sader HS, Fritsche TR, Jones RN. Dalbavancin activity against selected populations of antimicrobial-resistant Gram-positive pathogens. Diagn Microbiol Infect Dis 2005; 53: 307-310.         [ Links ]

103. Buckwalter M, Dowell JA. Population pharmacokinetic analysis of dalbavancin, a novel lipoglycopeptide. J Clin Pharmacol 2005; 45: 1279-1287.         [ Links ]

104. Jauregui LE, Babazadech S, Seltzer E, et al. Randomized, double-blind comparison of once-weekly dalbavancin versus twice-daily linezolid therapy for the treatment of complicated skin and skin structure infections. Clin Infect Dis 2005; 41: 1407-1415.         [ Links ]

105. Hancock RE. Mechanism of action of newer antibiotics for Gram-positive pathogens. Lancet Infect Dis 2005; 5: 209-218.         [ Links ]

106. Schmidt-Ioanas M, De Roux A, Lode H. New antibiotics for the treatment of severe staphylococcal infection in the critically ill patient. Curr Opin Crit Care 2005; 11: 481-486.         [ Links ]

107. Tsuji BT, Rybak MJ. Etest synergy testing of clinical isolates of Staphylococcus aureus demonstrating heterogeneous resistance to vancomycin. Diagn Microbiol Infect Dis 2006; 54: 73-77.         [ Links ]

108. Smith PF, Booker BM, Ogundele AB, Kelchin P. Comparative in vitro activities of daptomycin, linezolid, and quinupristin/dalfopristin against Gram-positive bacterial isolates from a large cancer center. Diagn Microbiol Infect Dis 2005; 52: 255-259.         [ Links ]