Find information on thousands of medical conditions and prescription drugs.

Teicoplanin

Teicoplanin is an antibiotic used in the prophylaxis and treatment of serious infections caused by Gram-positive bacteria. It is a glycopeptide antiobiotic extracted from Actinoplanes teichomyceticus, with a similar spectrum of activity to vancomycin. Teicoplanin is marketed by Aventis under the trade name Targocid®.

Oral teicoplanin has been demonstrated to be effective in the treatment of pseudomembranous colitis and Clostridium difficile-associated diarrhoea, with comparable efficacy to vancomycin. (de Lalla, 1992)

Home
Diseases
Medicines
A
B
C
D
E
F
G
H
I
J
K
L
M
N
O
P
Q
R
S
T
Oxytetracycline
Phentermine
Tacrine
Tacrolimus
Tagamet
Talbutal
Talohexal
Talwin
Tambocor
Tamiflu
Tamoxifen
Tamsulosin
Tao
Tarka
Taurine
Taxol
Taxotere
Tazarotene
Tazobactam
Tazorac
Tegretol
Teicoplanin
Telmisartan
Temazepam
Temocillin
Temodar
Temodar
Temozolomide
Tenex
Teniposide
Tenoretic
Tenormin
Tenuate
Terazosin
Terbinafine
Terbutaline
Terconazole
Terfenadine
Teriparatide
Terlipressin
Tessalon
Testosterone
Tetrabenazine
Tetracaine
Tetracycline
Tetramethrin
Thalidomide
Theo-24
Theobid
Theochron
Theoclear
Theolair
Theophyl
Theophyl
Theostat 80
Theovent
Thiamine
Thiomersal
Thiopental sodium
Thioridazine
Thorazine
Thyroglobulin
Tiagabine
Tianeptine
Tiazac
Ticarcillin
Ticlopidine
Tikosyn
Tiletamine
Timolol
Timoptic
Tinidazole
Tioconazole
Tirapazamine
Tizanidine
TobraDex
Tobramycin
Tofranil
Tolazamide
Tolazoline
Tolbutamide
Tolcapone
Tolnaftate
Tolterodine
Tomoxetine
Topamax
Topicort
Topiramate
Tora
Toradol
Toremifene
Tracleer
Tramadol
Trandate
Tranexamic acid
Tranxene
Tranylcypromine
Trastuzumab
Trazodone
Trenbolone
Trental
Trest
Tretinoin
Triacetin
Triad
Triamcinolone
Triamcinolone hexacetonide
Triamterene
Triazolam
Triclabendazole
Triclosan
Tricor
Trifluoperazine
Trilafon
Trileptal
Trimetazidine
Trimethoprim
Trimipramine
Trimox
Triprolidine
Triptorelin
Tritec
Trizivir
Troglitazone
Tromantadine
Trovafloxacin
Tubocurarine chloride
Tussionex
Tylenol
Tyrosine
U
V
W
X
Y
Z

Read more at Wikipedia.org


[List your site here Free!]


Staphylococcus aureus pneumonia: a "superbug" infection in community and hospital settings
From CHEST, 9/1/05 by Marin H. Kollef

Pneumonia, an infection of the lungs usually due to bacterial, viral, or fungal pathogens, is classified according to the location of the patient at the time the infection occurs. Hospital-associated pneumonia (HAP) is defined as occurring > 48 h after hospital admission. HAP is the second most common hospital-acquired infection but leads to the greatest number of nosocomial-related deaths. (1,2) Treatment of HAP has become problematic in the face of escalating emergence of antibiotic-resistant bacteria. (3,4) Successful outcomes depend on the prevention of HAP when possible and the administration of appropriate antibiotics in a timely manner when infection develops. (5) The latter can be difficult to achieve when the etiology of HAP is initially unknown.

Ventilator-associated pneumonia (VAP), one form of HAP, specifically refers to pneumonia developing in a patient receiving mechanical ventilation > 48 h after tracheal intubation. (5,6) Although not included in this definition, some patients may require intubation after severe HAP develops and should be managed in a manner similar to patients with VAP. Pneumonia occurring within 48 h of hospital admission can be difficult to differentiate from community-associated pneumonia (CAP). These early onset pneumonias may have begun prior to hospital admission or possibly as a result of aspiration resulting from tracheal intubation at the time of hospital admission.

Health-care-associated pneumonia (HCAP) has been described as pneumonia developing in patients admitted to the hospital from high-risk environments. These high-risk environments include nursing homes and extended-care facilities or patients' homes if they are receiving long-term dialysis, home infusion therapies (antibiotics, chemotherapy, blood product transfusions), home wound therapy, or have had a recent hospitalization. (7) These risk factors increase the likelihood of infection with antibiotic-resistant bacteria that are more commonly seen in HAP and VAP as compared with CAP. (8-10) Patients with HCAP and HAP may go on to respiratory failure, further blurring the differences between these various classifications of pneumonia.

Infectious organisms resulting in HAP, as well as HCAP and VAP, have traditionally differed from those that are most commonly associated with CAP. Gram-negative aerobes have usually comprised the majority of hospital and health-care-associated infections while more antibiotic-sensitive pathogens (Streptococcus pneumoniae, Haemophilus influenza, Escherichia coli) have been associated with community infections. (2,11,12) The individual organisms most commonly reported to be associated with HAP and VAP, Staphylococcus aureus (18.1%), Pseudomonas aeruginosa (17.0%), and Enterobacter species (11.2%), are also the most likely to be resistant to prescribed antimicrobial regimens. (2,5,12)

The recent emergence of methicillin-resistant S aureus (MRSA) as an increasingly common pathogen in all forms of pneumonia (CAP, HCAP, HAP, and YAP) has weakened the overall clinical importance of this classification scheme. (13) Community-associated MRSA (CAMRSA) is the newest threat to patients hospitalized with pneumonia. The Centers for Disease Control and Prevention uses the following clinical criteria to differentiate CAMRSA from hospital strains: (1) diagnosis of MRSA made in the outpatient setting or by a culture finding positive for MRSA within 48 h after admission to the hospital; (2) the patient has no medical history of MRSA infection or colonization; (3) the patient has not been hospitalized, resided in a nursing home, received hospice care, required dialysis, or undergone surgery in the past year; and (4) the patient has no permanent indwelling catheters or medical devices that pass through the skin into the body. (14)

The virulence of the newly identified MRSA strains, including CAMRSA, has resulted in the designation of "superbug" to describe this pathogen.15 MRSA is considered to be a superbug due to the multiple features it can possess resulting in enhanced antibiotic resistance and greater mortality compared to methicillin-sensitive S aureus (MSSA) strains. (14-16) These features include the presence of the mecA gene that encodes a penicillin-binding protein, PBP2a, which is intrinsically insensitive to methicillin and all [beta]-lactam antibiotics that have been developed, including the isoxazolyl penicillins (eg, oxacillin) and broad-spectrum [beta]-lactams (third-generation cephalosporins, cefamycins, and carbapenems); the occurrence of surface molecules that promote greater adherence to living surfaces (endothelium, bone, cartilage), as well as to artificial surfaces (endotracheal tubes, medical devices, locker room floors); and the ability of S aureus to produce various toxins.

Two recent metaanalyses (17,18) of S aureus bacteremia have demonstrated that MRSA infections have an almost twofold greater mortality risk compared to MSSA infections. The greater intrinsic antibiotic resistance and virulence of the MRSA strains likely contributes to this difference in mortality. Similarly, MRSA pneumonia has been associated with a greater risk of death compared to MSSA pneumonia. (19) Delays in the administration of appropriate initial antibiotic treatment for MRSA infections has been identified as one potential explanation for the excess mortality observed with this organism. (20) Such delays have been attributed to clinicians' failure to recognize risk factors for infection due to MRSA, including prior antibiotic administration and exposure to high-risk environments such as the ICU setting. (21)

Toxin production is an important aspect of the virulence associated with MRSA and MSSA infections. Most clinicians are aware of the toxin associated with toxic shock syndrome, staphylococcal enterotoxins, and the dermolytic toxin resulting in the scalded skin syndrome. More recently the Panton-Valentine leukocidin (PVL) toxin has been described as an important promoter of virulence in many strains of MRSA most prominent in CAMRSA. (15,16,22) The PVL toxin is a potent mediator of inflammation and can also destroy leukocytes by creating lytic pores in their cell membranes, contributing to the tissue necrosis observed in patients with PVL-positive infections. (15,22) PVL has been associated with necrotizing pneumonia and severe skin infections originating in the community setting. (15,23) In a study (24) of S aureus pneumonia, the mortality rate was 32% in patients with infection with PVL-positive strains vs 6% mortality for infection with the PVL-negative strains.

In this issue of CHEST (see page 1414), DeRyke et al (25) describe their experience with bacteremic S aureus VAP. The main limitation of their study is the relatively small sample size examined predisposing to type II errors for the comparisons they report among patient subgroups. Nevertheless, their study confirms many of the earlier observations of S aureus VAP. DeRyke et al (25) found that patients with MRSA VAP were more likely to have had prior antibiotic exposure as a risk factor for infection with this specific antibiotic-resistant pathogen. Additionally, the high overall mortality they observed supports the importance of bacteremic S aureus pneumonia as a cause of patient morbidity and nosocomial-related death. Interestingly, these investigators observed a trend toward greater mortality among patients treated with vancomycin compared to [beta]-lactams for both MRSA and MSSA infections. Although not statistically significant, this observation suggests that vancomycin may have limitations in the treatment of serious S aureus infections. In vitro studies (26) have shown that vancomycin is a slowly cidal antibiotic that requires serum concentrations above the minimum inhibitory concentration (MIC) of MRSA throughout the dosing interval to achieve optimal killing of this pathogen. Clinical data and animal models (27,28) suggest that bacteremia is more prolonged with MSSA endocarditis treated with glycopeptide antibiotics (vancomycin, teicoplanin) compared to [beta]-lactam antibiotics. Similarly, MSSA endocarditis treated with vancomycin has been shown to have greater overall failure rates (37% to 50%) compared to similar patients treated with nafcillin (1.4% to 26%). (29-33) Comparable results have also been found for endocarditis due to MRSA, with even more prolonged bacteremia when rifampin is added to vancomycin suggesting clinical antagonism between these agents compared to treatment with vancomycin alone. (34,35)

Potential limitations of vancomycin for the treatment of S aureus pneumonia have also been observed. Gonzalez et al (36) reported that patients with bacteremic MRSA pneumonia had a greater mortality compared to MSSA pneumonia. Patients with MSSA infection treated with vancomycin were also found to have a significantly greater risk of hospital mortality compared to patients with MSSA pneumonia treated with oxacillin. (36) Available studies (26,37,38) examining the treatment of MRSA pneumonia with vancomycin have found treatment to be successful in only 35 to 57% of patients.

A metaanalysis (38) of two studies performed by the same group of investigators using the same study protocol found that patients with MRSA HAP treated with linezolid had a statistically greater survival compared to patients treated with vancomycin. When examining the cohort of patients with MRSA VAP, these investigators (39) also found a greater eradication of MRSA among the patients treated with linezolid. An important limitation of this analysis is that the mortality benefit for MRSA was determined from a subgroup analysis. A confirmatory study has been undertaken to validate these findings by comparing linezolid to high-dose vancomycin (30 mg/ kg/d) for microbiologically confirmed MRSA pneumonia.

Potential explanations for the clinical failures observed with vancomycin treatment of pneumonia, endocarditis, and bloodstream infections attributed to S aureus include the slow killing of vancomyin for this pathogen, the degree of protein binding of vancomycin relative to unbound or free drug concentrations at the infection site, the inability to achieve tissue concentrations of vancomycin above the MIC for MRSA throughout the dosing interval, the observed upward drift in the MICs for MRSA strains, and the large inoculum size associated with MRSA infections, especially in the lungs (pneumonia, empyema) and soft-tissue abscesses. Additionally, vancomycin, unlike clindamycin and linezolid, acts on the cell wall of S aureus, having no effect on toxin production at the ribosomal level.

Vancomycin epithelial lining fluid (ELF) concentrations may be increased in patients with lung injury due to leakage of vancomycin bound to albumin and other proteins into the alveolar compartment. However, the achievable free drug levels of vancomycin in the ELF appear to only reach concentrations > 4 [micro]g/mL, the susceptibility breakpoint of MRSA, when plasma levels are > 20 to 25 [micro]g/mL. Thus, plasma levels of vancomycin and "active" ELF concentrations of vancomycin are linearly related in patients with S aureus pneumonia, in approximately a 5:1 ratio. (40) Another study (41) of in vivo lung tissue concentration measurements of vancomycin observed mean vancomycin levels at or below the 4 [micro]g/mL susceptibility breakpoint of MRSA 4 to 6 h after administration of a 1-g dose of vancomycin in patients undergoing thoracotomy. The limited tissue penetration of vancomycin, resulting from protein binding of the drug in plasma, along with upward drift of the MICs of MRSA to vancomycin offers the best explanation for the clinical failures of vancomycin reported in patients with serious MRSA infections. (42-44)

The enhanced ability of linezolid, compared to vancomycin, to penetrate into lung tissue at therapeutic concentrations may explain the microbiologic and clinical differences observed in the available clinical trials (38,39,45,46) comparing these agents for MRSA pneumonia. Teicoplanin is another glycopeptide antibiotic available in Europe for the treatment of MRSA that is also highly protein bound. (47) A trial (48) comparing linezolid to teicoplanin for the treatment of suspected or proven Gram-positive infection found that clinical responses were greater in the linezolid-treated patients, including the patients with pneumonia, further supporting a potential role for linezolid in the treatment of this infection. CAMRSA isolates may retain susceptibility to a number of non-[beta]-lactam antibiotics including trimethoprimsulfamethoxazole, clindamycin, tetracyclines, and fluoroquinolones. However, despite this phenotypic expression, the efficacy of these treatment options has not been established clinically, and the potential for resistance development is currently unknown.

In summary, pneumonia due to MRSA does appear to be a superbug infection due to the antibiotic resistance and overall virulence of this pathogen. Fortunately, a number of new currently available (linezolid) and investigational antibiotics (glycylcycline, dalbavancin, cephalosporin antibiotics with MRSA activity) hold promise for improved treatment of MRSA pneumonia, as well as other infections caused by MRSA. However, clinicians must balance the use of antibiotics for the treatment of MRSA pneumonia with the need to minimize the emergence of antimicrobial resistance. Strategies have emerged that allow clinicians to treat patients with suspected or microbiologically proven nonbacteremic HAP and VAP with courses of therapy that are often < 10 days in duration. (49,50) At present, we need additional studies to define the optimal antibiotic choices for the treatment of MRSA pneumonia. The emergence of this superbug along with concerns for the future appearance of avian influenza, potentially further increasing outbreaks of MRSA pneumonia, (24) makes this an important health-care concern for the community as well as the hospital setting.

REFERENCES

(1) McEachern R, Campbell GD Jr. Hospital-acquired pneumonia: epidemiology, and treatment. Infect Dis Clin North Am 1998; 12:761-779

(2) Richards MJ, Edwards JR, Culver DH, et al. Nosocomial infections in medical intensive care units in the United States. Crit Care Med 1999; 27:887-892

(3) Fridkin SK. Increasing prevalence of antimicrobial resistance in intensive care units. Crit Care Med 2001; 29:N64-N68

(4) Lynch JP. Hospital-acquired pneumonia: risk factors, microbiology, and treatment. Chest 2001; 119(suppl):373S-384S

(5) Chastre J, Fagon JY. Ventilator-associated pneumonia. Am J Respir Crit Care Med 2002; 165:867-903

(6) Craven DE, Steger KA. Epidemiology of nosocomial pneumonia: new perspectives on an old disease. Chest 1995; 108(suppl): 1S-16S

(7) Gaynes R. Health-care associated bloodstream infections: a change in thinking. Ann Intern Med 2002; 137:850-851

(8) Tablan OC, Anderson LJ, Besser R, et al. Guidelines for preventing health-care-associated pneumonia, 2003: recommendations of the CDC and the Healthcare Infection Control Practices Advisory Committee. MMWR Morb Mortal Wkly Rep 2004; 53(RR-3):1-36

(9) Mylotte JM. Nursing-home acquired pneumonia. Clin Infect Dis 2002; 35:1205-1211

(10) Hutt E, Kramer AM. Evidence-based guidelines for management of nursing home-acquired pneumonia. J Fam Pract 2002; 51:709-716

(11) American Thoracic Society. Hospital-acquired pneumonia in adults: diagnosis, assessment of severity, initial antimicrobial therapy, and preventive strategies; a consensus statement, November 1995. Am J Respir Crit Care Med 1996; 153:1711-1725 (12) National Nosocomial Infection Surveillance (NNIS) System report, data summary from January 1990-May 1999, issued June 1999. Am J Infect Control 1999; 27:502-532

(13) Carleton HA, Diep BA, Charlebois ED, et al. Community-adapted methicillin-resistant Staphylococcus aureus (MRSA): population dynamics of an expanding community reservoir of MRSA. J Infect Dis 2004; 190:1730-1738

(14) Center for Disease Control and Prevention. Community-associated MRSA. Information from the US Centers for Disease Control and Prevention. August 2003. Available at: http://www. cdc.gov/ncidod/hip/ARESIST/mrsa_comm_faq.htm; accessed July 31, 2005

(15) Foster TJ. The Staphylococcus aureus"superbug". J Clin Invest 2004; 114:1693-1696

(16) Rybak MJ, LaPlante KL. Community-associated methicillin-resistant Staphylococcus aureus: a review. Pharmacotherapy 2005; 25:74-85

(17) Cosgrove SE, Sakoulas G, Perencevich EN, et al. Comparison of mortality associated with methicillin-resistant and methicillin-susceptible Staphylococcus aureus bacteremia: a meta-analysis. Clin Infect Dis 2003; 36:53-59

(18) Whitby M, McLaws ML, Berry G. Risk of death from methicillin-resistant Staphylococcus aureus bacteraemia: a meta-analysis. Med J Aust 2001; 175:264-267

(19) Rello J, Torres A, Ricart M, et al. Ventilator-associated pneumonia by Staphylococcus aureus: comparison of methicillin-resistant and methicillin-sensitive episodes. Am J Respir Crit Care Med 1994; 150:1545-1549

(20) Lodise TP, McKinnon PS, Swiderski L, et al. Outcomes analysis of delayed antibiotic treatment for hospital-acquired Staphylococcus aureus bacteremia, Clin Infect Dis 2003; 36:1418-1423

(21) Trouillet JL, Chastre J, Vuagnat A, et al. Ventilator-associated pneumonia caused by potentially drug-resistant bacteria. Am J Respir Crit Care Med 1998; 157:531-539

(22) Dinges MM, Orwin PM, Schlievert PM. Exotoxins of Staphylococcus aureus. Clin Microbiol Rev 2000; 13:16-34

(23) Naimi TS, LeDell KH, Como-Sabetti K, et al. Comparison of community- and health care-associated methicillin-resistant Staphylococcus aureus infection. JAMA 2003; 290:2976-2984

(24) Gillet Y, Issartel B, Vanhems P, et al. Association between Staphylococcus aureus strains carrying gene for Panton-Valentine leukocidin and highly lethal necrotising pneumonia in young immunocompetent patients. Lancet 2002; 359:753-759

(25) DeRyke CA, Lodise TP, Rybaek MJ, et al. Epidemiology, treatment, and outcomes of nosocomial bacteremic Staphylococcus aureus pneumonia. Chest 2005; 128:1414-1422

(26) Moise PA, Forrest A, Bhavnani SM, et al. Area under the inhibitory curve and a pneumonia scoring system for predicting outcomes of vancomycin therapy for respiratory infections by Staphylococcus aureus. Am J Health System Pharm 2000; 57:S4-S9

(27) Korzeniowski O, Sande MA. Combination antimicrobial therapy for Staphylococcus aureus endocarditis in patients addicted to parenteral drugs and in nonaddicts: a prospective study. Ann Intern Med 1982; 97:496-503

(28) Chambers HF, Kennedy S. Effects of dosage, peak and trough concentrations in serum, protein binding, and bactericidal rate on efficacy of teicoplanin in a rabbit model of endocarditis. Antimicrob Agents Chemother 1990; 34:510-514

(29) Esposito AL, Gleckman RA. Vancomycin: a second look. JAMA 1977; 238:1756-1757

(30) Geraci JE, Wilson WR. Vancomycin therapy for infective endocarditis. Rev Infect Dis 1981; 3(suppl):S250-S258

(31) Small PM, Chambers HF. Vancomycin for Staphylococcus aureus endocarditis in intravenous drug users. Antimicrob Agents Chemother 1990; 34:1227-1231

(32) Gentry CA, Rodvold KA, Novak RM, et al. Retrospective evaluation of therapies for Staphylococcus aureus endocarditis. Pharmacotherapy 1997; 17:990-997

(33) Chang FY, Peacock JE Jr, Musher DM, et al. Staphylococcus aureus bacteremia: recurrence and the impact of antibiotic treatment in a prospective multicenter study. Medicine (Baltimore) 2003; 82:333-339

(34) Levine DP, Fromm BS, Reddy BR. Slow response to vancomycin or vancomycin plus rifampin in methicillin-resistant Staphylococcus aureus endocarditis. Ann Intern Med 1991; 115:674-680

(35) Shelburne SA, Musher DM, Hulten K, et al. In vitro killing of community-associated methicillin-resistant Staphylococcus aureus with drug combinations. Antimicrob Agents Chemother 2004; 48:4016-4019

(36) Gonzalez C, Rubio M, Romero-Vivas J, et al. Bacteremic pneumonia due to Staphylococcus aureus: a comparison of disease caused by methicillin-resistant and methicillin-susceptible organisms. Clin Infect Dis 1999; 29:1171-1177

(37) Fagon J, Patrick H, Haas DW, et al. Treatment of gram-positive nosocomial pneumonia: prospective randomized comparison of quinupristin/dalfopristin versus vancomycin. Nosocomial Pneumonia Group. Am J Respir Crit Care Med 2000; 161:753-762

(38) Wunderink RG, Rello J, Cammarata SK, et al. Linezolid vs vancomycin: analysis of two double-blind studies of patients with methicillin-resistant Staphylococcus aureus nosocomial pneumonia. Chest 2003; 124:1789-1797

(39) Kollef MH, Rello J, Cammarata SK, et al. Clinical cure and survival in Gram-positive ventilator-associated pneumonia: retrospective analysis of two double-blind studies comparing linezolid with vancomycin. Intensive Care Med 2004; 30: 388-394

(40) Lamer C, de Beco V, Soler P, et al. Analysis of vancomycin entry into pulmonary lining fluid by bronchoalveolar lavage in critically ill patients. Antimicrob Agents Chemother 1993; 37:281-286

(41) Cruciani M, Gatti G, Lazzarini L, et al. Penetration of vancomycin into human lung tissue. J Antimicrob Chemother 1996; 38:865-869/

(42) Sakoulas G, Moise-Broder PA, Schentag J, et al. Relationship of MIC and bactericidal activity to efficacy of vancomycin for treatment of methicillin-resistant Staphylococcus aureus bacteremia. J Clin Microbiol 2004; 42:2398-2402

(43) Moise-Broder PA, Sakoulas G, Eliopoulos GM, et al. Accessory gene regulator group II polymorphism in methicillin-resistant Staphylococcus aureus is predictive of failure of vancomycin therapy. Clin Infect Dis 2004; 38:1700-1705

(44) Howden BP, Ward PB, Charles PG, et al. Treatment outcomes for serious infections caused by methicillin-resistant Staphylococcus aureus with reduced vancomycin susceptibility. Clin Infect Dis 2004; 38:521-528

(45) Conte JE Jr, Golden JA, Kipps J, et al. Intrapulmonary pharmacokinetics of linezolid. Antimicrob Agents Chemother 2002; 46:1475-1480

(46) Honeybourne D, Tobin C, Jevons G, et al. Intrapulmonary penetration of linezolid. J Antimierob Chemother 2003; 51:1431-1434

(47) Knudsen JD, Fuursted K, Espersen F, et al. Activities of vancomycin and teicoplanin against penicillin-resistant pneumococci in vitro and in vivo and correlation to pharmacokinetic parameters in the mouse peritonitis model. Antimicrob Agents Chemother 1997; 41:1910-1915

(48) Wilcox M, Nathwani D, Dryden M. Linezolid compared with teicoplanin for the treatment of suspected or proven Gram-positive infections. J Antimicrob Chemother 2005; 53:335-344

(49) Chastre J, Wolff M, Fagon JY, et al. Comparison of 8 vs 15 days of antibiotic therapy for ventilator-associated pneumonia in adults: a randomized trial. JAMA 2003; 290:2588-2598

(50) Micek ST, Ward S, Fraser VJ, et al. A randomized controlled trial of an antibiotic discontinuation policy for clinically suspected ventilator-associated pneumonia. Chest 2004; 125: 1791-1799

Marin H. Kollef MD

Washington University School of Medicine

Scott T. Micek, PharmD

Barnes-Jewish Hospital

St. Louis, MO

Dr. Kollef is Director, Medical Intensive Care Unit, Washington University School of Medicine. Dr. Micek is Pharmacy Director, Medical Intensive Care Unit, Barnes-Jewish Hospital.

Reproduction of this article is prohibited without written permission from the American College of Chest Physicians (e-mail: www.chestjournal.org/misc/reprints.shtml).

Correspondence to: Marin H. Kollef, MD, FCCP, Washington University School of Medicine, Campus Box 8052, 660 South Euclid Ave, St. Louis, MO 63110; e-mail: mkollef@im.wustl.edu

COPYRIGHT 2005 American College of Chest Physicians
COPYRIGHT 2005 Gale Group

Return to Teicoplanin
Home Contact Resources Exchange Links ebay