Aminoglycoside antibiotics in infectious diseases. An overview..pdf

13
Arhnoglycoside Antibiotics in Infectious Diseases An Overview WALTER E. SIEGENTHALER, M.D. ANTONIO BONETTI, M.D. RUEDI LUTHY, M.D. Zurich, Switzerland This article presents an overview of the aminoglycoside antibiotics used in clinical practice. Facts concerning the discovery and prop- erties of the aminoglycosides are followed by information about spectrums of activity and mechanisms of action and resistance. Individual compounds are compared and proposals on the possibil- ities for their clinical use, both as single drugs and in combination with beta-lactam antibiotics, are made. The importance placed on measuring the serum concentrations of aminoglycoside antibiotics should serve as a reminder that this procedure is important, on one hand, to increase clinical efficacy and, on the other, to reduce the side effects of these antibiotics. Finally, the aminoglycosides are compared briefly with other antibacterial compounds, some of which are very new. There is no doubt that in the future the amino- glycosides will continue to occupy an important place in the treat- ment of severe infections, although newly developed agents appear to be effective complements. Since the discovery and clinical use of streptomycin by Waksman and co-workers [l] more than 40 years ago, various aminoglycosides have been developed and introduced in clinical medicine. This article provides an overview of the actual significance of aminoglycosides in infectious disease. From the Department of Medicine, University Hos- pital, Zurich, Switzerland. Requests for reprints should be addressed to Dr. Walter E. Siegenthaler, Department of Medicine, University Hospital, Ramistrasse 100, CH 8091 Zurich, Switzerland. The discovery of streptothricin in 1942 and the isolation of streptomycin from Streptomyces griseus by Waksman’s research group in 1943 ushered in an exploratory era for a new chemical class of antibiotics, the aminoglycosidic aminocyclitols, i.e., the aminoglycosides (Table I). The clinical application of streptomycin began in 1944, followed in 1949 by neomycin, isolated from Streptomyces fradiae; in 1957 by kanamycin, isolated from Streptomyces kanamyceticus; in 1963 by gentamicin, iso- lated from Micromonospora purpurea; in 1967 by tobramycin, isolated from Streptomyces tenebrarius; and in 1970 by sisomicin, isolated from Micromonospora inyoensis. Amikacin, introduced in 1972, is a semisyn- thetic derivative of kanamycin A, and netilmicin, introduced in 1975, is a semisynthetic analog of sisomicin. Kanamycin is composed of three frac- tions, 98 percent A and 2 percent 6 and C. Gentamicin is also composed of three fractions, 40 percent gentamicin Cl, 20 percent gentamicin Cl a, and 40 percent gentamicin C2. The newer aminoglycosides, however, are single substances. As shown in Table II, all aminoglycosides are obtained from microorganisms of the genus Streptomyces (e.g., strepto- mycin, neomycin, kanamycin, tobramycin, and amikacin) or from the genus Micromonospora (e.g., gentamicin, sisomicin, and netilmicin). Structurally, all representatives of the kanamycin and gentamicin fami- 2 June 30, 1988 The American Journal of Medicine Volume 80 (suppl 8B)

Transcript of Aminoglycoside antibiotics in infectious diseases. An overview..pdf

Arhnoglycoside Antibiotics in Infectious Diseases

An Overview

WALTER E. SIEGENTHALER, M.D. ANTONIO BONETTI, M.D. RUEDI LUTHY, M.D.

Zurich, Switzerland

This article presents an overview of the aminoglycoside antibiotics used in clinical practice. Facts concerning the discovery and prop- erties of the aminoglycosides are followed by information about spectrums of activity and mechanisms of action and resistance. Individual compounds are compared and proposals on the possibil- ities for their clinical use, both as single drugs and in combination with beta-lactam antibiotics, are made. The importance placed on measuring the serum concentrations of aminoglycoside antibiotics should serve as a reminder that this procedure is important, on one hand, to increase clinical efficacy and, on the other, to reduce the side effects of these antibiotics. Finally, the aminoglycosides are compared briefly with other antibacterial compounds, some of which are very new. There is no doubt that in the future the amino- glycosides will continue to occupy an important place in the treat- ment of severe infections, although newly developed agents appear to be effective complements.

Since the discovery and clinical use of streptomycin by Waksman and co-workers [l] more than 40 years ago, various aminoglycosides have been developed and introduced in clinical medicine. This article provides an overview of the actual significance of aminoglycosides in infectious disease.

From the Department of Medicine, University Hos- pital, Zurich, Switzerland. Requests for reprints should be addressed to Dr. Walter E. Siegenthaler, Department of Medicine, University Hospital, Ramistrasse 100, CH 8091 Zurich, Switzerland.

The discovery of streptothricin in 1942 and the isolation of streptomycin from Streptomyces griseus by Waksman’s research group in 1943 ushered in an exploratory era for a new chemical class of antibiotics, the aminoglycosidic aminocyclitols, i.e., the aminoglycosides (Table I). The clinical application of streptomycin began in 1944, followed in 1949 by neomycin, isolated from Streptomyces fradiae; in 1957 by kanamycin, isolated from Streptomyces kanamyceticus; in 1963 by gentamicin, iso- lated from Micromonospora purpurea; in 1967 by tobramycin, isolated from Streptomyces tenebrarius; and in 1970 by sisomicin, isolated from Micromonospora inyoensis. Amikacin, introduced in 1972, is a semisyn- thetic derivative of kanamycin A, and netilmicin, introduced in 1975, is a semisynthetic analog of sisomicin. Kanamycin is composed of three frac- tions, 98 percent A and 2 percent 6 and C. Gentamicin is also composed of three fractions, 40 percent gentamicin Cl, 20 percent gentamicin Cl a, and 40 percent gentamicin C2. The newer aminoglycosides, however, are single substances. As shown in Table II, all aminoglycosides are obtained from microorganisms of the genus Streptomyces (e.g., strepto- mycin, neomycin, kanamycin, tobramycin, and amikacin) or from the genus Micromonospora (e.g., gentamicin, sisomicin, and netilmicin).

Structurally, all representatives of the kanamycin and gentamicin fami-

2 June 30, 1988 The American Journal of Medicine Volume 80 (suppl 8B)

lies possess two amino sugars, whereas neomycin and paromomycin possess three. The amino sugars are gly- cosidically linked with a central hexose, aminocyclitol, a 2-deoxystreptamine. In streptomycin, this aminocyclitol is streptidine, and is not located in the center of the mole- cule. Various structural arrangements for these com- pounds are depicted in Figure 1.

The aminoglycosides are strongly polar cations, stable in the pH range of 6 to 8, and basic in character. Being water soluble, they are distributed throughout the extra- cellular space, i.e., about 25 percent of the lean body mass. Metabolically, they are highly stable; about 95 per- cent is excreted via the kidneys. Aminoglycosides are poorly absorbed from the intestinal tract, and because of their polarity, they inadequately penetrate the intracellular space and cerebrospinal fluid.

SPECTRUM OF ACTIVITY

The aminoglycosides have a very broad antimicrobial spectrum, extending from gram-positive cocci to gram- negative bacilli. They are ineffective against all anaerobic organisms, and their activity against the enterococci is inadequate when used as monotherapy [2]. Listeria mon- ocytogenes and Nocardia species are generally resistant to the aminoglycosides; however, amikacin is effective against Nocardia [3]. Streptomycin and kanamycin are used successfully against Mycobacterium tuberculosis [3], but their activity against Pseudomonas aeruginosa is limited. All aminoglycosides share similar antibacterial action against gram-negative aerobic pathogens, al- though their intrinsic activities and sensitivity patterns dif- fer. In addition, individual differences between agents are reported from hospital to hospital due to time- and indica- tion-related induction of resistance.

MECHANISM OF ACTION AND RESISTANCE

Although the mechanisms of aminoglycoside action are only partially understood, it is now known that the drugs bind to the surface of the bacteria and are transported through the cell wall. Once within the cell, they bind to the 30s ribosomal subunit, causing a misreading of messen- ger RNA during the translation process and producing “nonsense proteins.” The sum of the events from drug entry into the cell to interference with protein synthesis disturbs membrane function and causes potassium, so- dium, amino acids, and other ,essential constituents to leak out, resulting in bacterial death.

Almost all pathogens that are sensitive to aminoglyco- sides can develop resistance. Resistance develops most commonly in response to plasmid-mediated enzymes that can modify the aminoglycoside molecule in three different ways: by acetylation at an amino group, by phosphoryla- tion, or by adenylation at a hydroxyl group. The molecule becomes changed in such a way that it can no longer bind to the ribosome subunits. The enzymes involved include

SYMPOSIUM ON AMINOGLYCOSIDE THERAPY-SIEGENTHALER ET AL

TABLE I Discovery of the Aminoglycosides

Year Antibiotic Species

1944 Streptomycin From Streptomyces griseus 1949 Neomycin From Streptomyces fradiae 1957 Kanamycin From Streptomyces kanamyceticus 1963 Gentamicin From Micromonospora purpurea 1967 Tobramycin From Streptomyces tenebrarius 1970 Sisomicin From Micromonospora inyoensis 1972 Amikacin Semisynthetic derivative of kanamycin A 1975 Netilmicin Semisynthetic derivative of sisomicin

TABLE II Groups of Aminoglycosides

Streptomyces Group Micromonospora Group

Streptomycin Gentamicin Neomycin Sisomicin Kanamycin Netilmicin Tobramycin Amikacin

acetyltransferases, phosphotransferases, and adenyl- transferases, all of which have numerous subtypes (Fig- ure 1). Due to chemical differences, not all aminoglyco- sides are suitable substrates for all enzymes, and the binding sites for these enzymes even differ within the mol- ecule. Amikacin, the agent most resistant to inactivation, is inactivated principally by an acetyltransferase from gram-negative organisms, whereas an adenyltransferase from certain staphylococci and a phosphotransferase from various enterococci are not important quantitatively [4-71. This also explains why cross-resistance does not necessarily exist between aminoglycosides.

A second possibility for development of resistance is that the complex and energy-dependent transport system across the cytoplasmic membrane to the ribosomes be- comes changed in such a way that the target cannot be reached. P. aeruginosa, Serratia species, and Streptococ- cus faecalis can become resistant to aminoglycosides in this way, whereas the group resistance of the anaerobes results from the absence of an oxygen-dependent trans- port system across the cytoplasmic membrane [8].

A third possibility appears to be of importance only for streptomycin. In contrast to the other aminoglycosides, streptomycin binds only to a subgroup of the 30s ribo- somes [6]. These proteins can mutate in such a way that no binding takes place between streptomycin and the ri- bosomes. This phenomenon can be clinically important in combination therapy of enterococcal endocarditis, a situa- tion in which streptomycin resistance necessitates the use of gentamicin along with penicillin for adequate treatment.

June 30,1988 The American Journal of Medicine Volume 80 (suppl 86) 3

SYMPOSIUM ON AMINOGLYCOSIDE THERAPY-SIEGENTHALER ET AL

CH,NH, ZGip j-0.

Kanamycin A

Tobramycin

Amlkacin

b

0

OH

Gentamlcin C,a

Slsomicin

Netllmicln

OH

gure 1. Chemical structure of aminnoglycosides. AAC = site of action of acetylation enzymes; AAD = site of action of adeny- !ion enzymes; APH = site of action of phosphoryiation enzymes.

CdM~ARATlVE REVIEW OF THE AMINOGLYCOSIDES

Streptomycin. Although streptomycin. has been re- placed by newer aminoglycosides, it is still used today for special indications in tuberculosis therapy [9] and in enter- ococcal endocarditis. In the latter condition, it should be used only when the minimal inhibitory concentration is less than 2,000 mg/liter, and it should be combined with penicillin G or, in the case of penicillin allergy, with vanco- mycin [IO]. The same regimen is used under certain con- ditions to treat other streptococci implicated in endocardi-

tis. In Francisella tularensis infection, i.e., in tularemia, streptomycin ranks as the drug of first choice, as it does against Yersinia pestis, i.e., in bubonic plague.

Streptomycin is principally vestibulotoxic, but in 4 to 15 percent of treated patients, cochleotoxic side effects also appear after one week of therapy. They begin primarily in the high-tone range and are demonstrable only on the audiogram; the entire hearing range becomes affected in later stages [ll]. Certain side effects that are virtually never observed with the other aminoglycosides can de-

4 June 30,1988 The American Journal of Medicine Volume 80 (suppl 8B)

SYMPOSIUM ON AMINOGLYCOSIDE THERAPY-SIEGENTHALER ET AL

velop with streptomycin use, including neuromuscular block, peripheral neuritis, perioral paresthesia with flush- ing, exfoliative dermatitis, scotoma, and eosinophilia. In contrast, the nephrotoxic potency of streptomycin is very low [ll]. Neomycin. For years, neomycin has not been used par- enterally because of its marked ototoxicity and nephrotox- icity. In combination with bacitracin, which is bactericidal against gram-positive organisms, neomycin is an effective and safe topical agent. Occasionally, neomycin is admin- istered orally for preoperative sterilization of the gut and in portal encephalopathy. Despite the minimal intestinal ab- sorption (between 1 and 6 percent of the total dose), se- vere side effects such as tubular necrosis and deafness have occurred [12]. Malabsorption syndrome can also develop locally [I 31. Kanamycin. Kanamycin is another aminoglycoside that is rarely used today because of its considerable ototoxic- ity. Against P. aeruginosa, kanamycin is almost com- pletely inactive, and the Enterobacteriaceae become rap- idly resistant due to numerous modifying enzymes, i.e., two acetyltransferases, two phosphotransferases, and two adenyltransferases [4,14]. Only in the presence of multiple mycobacterial resistance to other drugs can kan- amycin still be used as a reserve tuberculostatic agent. Gentamicin. Since 1963, gentamicin has been used worldwide, and during its years of clinical application, re- sistance and sensitivity changes have appeared repeat- edly in Staphylococcus aureus [I!%171, P. aeruginosa, and other Enterobacteriaceae [18,19]. Here, too, the most important cause of resistance lies in the bacterial produc- tion of modifying enzymes coded by plasmids, trans- posons, episomes, and phages. In sensitive Enterobacte- riaceae, tobramycin and gentamicin are identical in effi- cacy, according to the findings of controlled clinical double-blind studies [20]. Against Escherichia coli and Serratia species, gentamicin appears to have greater effi- cacy [21]. Gentamicin can be modified primarily by four enzymes: two acetyltransferases, one adenyltransferase, and one phosphotransferase [4]. Cross-resistance with other aminoglycosides is common and, with netilmicin, almost complete [22]. In contrast, amikacin is still effective against gentamicin-resistant Enterobacteriaceae and P. aeruginosa strains, because amikacin is rarely inacti- vated by gentamicin-modifying enzymes.

Gentamicin appears to have the most definite nephro- toxic potency compared with tobramycin, netilmicin, and amikacin [23]. With regard to cochleotoxicity and ves- tibulotoxicity, the findings need to be interpreted with some caution. Audiograms and vestibular investigations are harder to perform and assess in a standardized man- ner than are creatinine clearance measurements or an excretion analysis of tubular enzymes. In part, the dis- crepancies in results from various investigators reflect

these technical difficulties. In a retrospective analysis, Lance and co-workers [24] found ototoxicity with amikacin in 2 to 4 percent of cases, whereas Black and associates [25], in prospective studies, found an ototoxicity incidence of 24 to 25 percent with amikacin. The comprehensive results of investigations by Kahlmeter and Dahlager [23], who reviewed comparative aminoglycoside toxicity stud- ies published between 1975 and 1982, showed that gen- tamicin, tobramycin, and amikacin were about equally vestibulotoxic, that amikacin was more cochleotoxic than gentamicin and tobramycin, and that netilmicin showed the lowest potency for cochlear and vestibular toxicity. In directly comparative studies between individual aminogly- cosides, these investigators obtained somewhat divergent results, due to the technical problems already mentioned. In seven comparative trials between gentamicin and netil- micin, the incidence of cochleotoxicity for gentamicin amounted to 2.1 percent; in contrast, in 10 comparative trials between gentamicin and amikacin, cochlear toxicity with gentamicin was 11.4 percent. Tobramycin. Tobramycin closely resembles gentamicin in terms of antimicrobial and pharmacokinetic properties. One recognized difference is that tobramycin has greater activity agains P. aeruginosa [26,27]. However, it should be noted that, depending on each individual situation, gentamicin-resistant strains are not always tobramycin- sensitive [28,29]. Tobramycin-resistant Pseudomonas strains are generally cross-resistant with streptomycin, kanamycin, gentamicin, and netilmicin [30]. Against gen- tamicin-resistant Klebsiella, Serratia, and Enterobacter species, tobramycin is also usually inactive. Sensitive Enterobacteriaceae, with the exception of Serratia and E. coli, are killed by tobramycin just as they would be by gen- tamicin [31]. Tobramycin is mainly modified by five en- zymes: two acetyltransferases, two adenyltransferases, and one phosphotransferase. Resistance associated with a second acetyltransferase is inconsistent [4].

Tobramycin ototoxicity appears to develop as com- monly as does gentamicin ototoxicity, whereas tobramy- tin is less nephrotoxic than gentamicin 1231. A large-scale, double-blind study in the United States showed that ele- vations of the serum creatinine level were recorded signif- icantly less frequently with tobramycin than with gentami- tin [32]. In the comparative assessment of ototoxicity, the investigators found no significant differences. Other stud- ies have also established a lower nephrotoxicity rate for tobramycin when compared with gentamicin [33,34]. Sisomicin. Introduced in 1970, sisomicin is chemically related to gentamicin and is described as 4,5 dehydro- gentamicin Cl a. It appears to be superior in efficacy to the other aminoglycosides, especially against Serratia and Proteus species, including some resistant strains [35-371. Sisomicin is more active than gentamicin against P. aeru- ginosa, and comparable in this respect to tobramycin, to

June 30,1988 The American Journal of Medicine Volume 80 (suppl 6B) 5

SYMPOSIUM ON AMINOGLYCOSIDE THERAPY-SIEGENTHALER ET AL

TABLE III Aminoglycosides

Advantages Disadvantages

Chemical stability Nephrotoxicity and ototoxicity Broad antibacterial spectrum Lack of activity against anaerobic Rapid bactericidal action organisms Experience over many years Low concentration in cerebrospinal Rare- allergic side effects fluid and bile Synergism with beta-lactam Variable pharmacokinetics

antibiotics Lack of correlation between administered dose and measured serum concentration

Inactivation of aminoglycosides

the extent that tobramycin-resistant strains of P. aerugin- osa are usually also resistant to sisomicin [30,38]. This agent, which seems to penetrate bacterial cells better than other aminoglycosides, is primarily modified by five enzymes: three acetyltransferases, one adenyltransfer- ase, and one phosphotransferase [4].

The factor limiting broad clinical application of sisomicin is its well-documented renal toxicity in animal studies. A comparison of so-called tubulotoxic threshold doses re- vealed that the dose of sisomicin that first leads to excre- tion of renal epithelial cells is one-fifth and one-tenth lower than the doses of tobramycin and gentamicin, respec- tively, that produce this effect [39]. Amikacin. A semisynthetic analog of kanamycin A, ami- kacin was introduced in 1972. The main advantage of this aminoglycoside lies in its extensive resistance to inacti- vating enzymes. Only an acetyltransferase from gram- negative pathogens, principally P. aeruginosa, is of quan- titative relevance, whereas both an adenyltransferase from certain staphylococci and a phosphotransferase from certain enterococci and staphylococci are of minor importance [4-71. Amikacin can, therefore, be used against gentamicin-resistant pathogens, for which it offers the greatest certainty of therapeutic response. It continues to be active against organisms resistant to tobramycin and netilmicin, including such resistant strains as P. aerugin- osa [40]. Cross-resistance between gentamicin and ami- kacin is observed less often than between gentamicin and tobramycin or sisomicin [41]. Certain “non-fermenters” such as Acinetobacter species, Flavobacter species, and P. aeruginosa show cross-resistance against amikacin, tobramycin, and gentamicin, caused by failure of amino- glycoside uptake into the cell [42,43].

Like the other aminoglycosides, amikacin also has a nephrotoxic potential, which, adjusted for the higher doses given, approximates that of netilmicin and must be judged more favorable than that of gentamicin [23,44]. The vestibulotoxic potency appears to equal that of genta- micin and tobramycin; however, cochleotoxicity is some- what greater [23].

Gerding and Larson [45] and Bet% and co-workers [46] reported the interesting and important observations that the resistance of gram-negative organisms to gentamicin and tobramycin declined significantly in numerous hospi- tals when amikacin was used alone, while amikacin resist- ance itself did not increase significantly. Although there are centers that regard amikacin as a reserve aminogly- coside, other centers employ it as first-line aminoglyco- side therapy and find no substantial increase in resistance [47-491. Netilmicin. A semisynthetic derivative of sisomicin, netil- micin was introduced in 1975. This aminoglycoside is pri- marily modified by three acetyltransferases, but not by adenyltransferases or phosphotransferases [4]. There- fore, sisomicin-, gentamicin-, and tobramycin-resistant E. coli, Klebsiella species, Serratia species, and even P. aeruginosa are often still eradicated by netilmicin. Nev- ertheless, cross-resistance with gentamicin is not uncom- mon in P. aeruginosa strains [50-521. In animal studies, netilmicin shows a lower nephrotoxicity than gentamicin and amikacin [53,54], and clinical trials indicate that the fewest cochlear and vestibular side effects occur with net- ilmicin [23].

INDICATIONS FOR AMINOGLYCOSIDES

Before discussing specific indications for aminoglycoside therapy, we should consider some of the advantages and disadvantages of these antibiotics (Table Ill). The advan- tages include chemical stability without metabolic changes, broad antibacterial spectrum, rapid bactericidal action, and comprehensive experience over many years. This experience has shown that the efficacy and success rate can be improved by the use of controlled, optimal serum concentrations. Two other advantages are the very rare occurrences of allergic side effects, and the syner- gism demonstrated when used along with beta-lactam antibiotics.

Some disadvantages include the potential for nephro- toxicity and ototoxicity and the associated narrow thera- peutic range between suboptimal serum concentrations and toxic values. Other disadvantages are the lack of ac- tivity against anaerobic organisms; the relatively low con- centrations in cerebrospinal fluid and bile; the markedly variable pharmacokinetics influenced by age, renal func- tion, fever, ascites, and obesity; the lack of correlation be- tween the administered dose and the measured serum concentration; and, finally, the inactivation of aminoglyco- sides by reversible binding to lysed granulocytes, low pH, anaerobic environment, high concentrations of calcium and magnesium ions, and beta-lactam antibiotics. The last factor, however, probably plays a role only in vitro, and is rarely of clinical importance except during concomi- tant administrations and when renal function is severely impaired [55,56].

6 June 30,1966 The American Journal of Medicine Volume 60 (suppl 6B)

SYMPOSIUM ON AMINOGLYCOSIDE THERAPY-SIEGENTHALER ET AL

l Severe gram-negative, hospital-acquired infections, especially in neutropenic patients

TABLE V Infections Treated with Aminoglycosides

Malignant otitis externa hospital-acquired pneumonias Urogenital infections Endocarditis Intra-abdominal infections Gram-negative meningitis Infectious arthritis or osteomyelitis Nosocomial septicemia Infections in immunocompromised patients

5 percent and appears to be increasing [64-701. Endocar- ditis exhibits special anatomic and functional features, such as an impaired local host-defense response with few phagocytes. In addition, large bacterial populations of IO* to 10” colony-forming units per gram of tissue, with re- duced metabolic activity, are protected from the antibiotics by a fibrin network.

Clinically, aminoglycosides are used in infections caused by pathogens resistant to other less toxic antibiot- its (Table IV). When used empirically or as specific ther- apy in severe gram-negative, hospital-acquired infections, especially in neutropenic patients, aminoglycosides are often combined with beta-lactam antibiotics because of the possible synergy. This article briefly discusses some infections in which aminoglycosides are frequently used (Table V).

Among the important infections of the upper respiratory tract is malignant otitis externa caused by P. aeruginosa. It can lead to severe complications, such as osteomyelitis, basal meningitis with cranial nerve involvement, cerebri- tis, and venous sinus thromboses, and therefore requires combined therapy with an antipseudomonal penicillin or a third-generation cephalosporin with an aminoglycoside.

Hospital-acquired bronchopneumonias are generally caused by Pseudomonas, E. coli, Klebsiella, Enterobac- ter, Serratia, Proteus, Providencia, and Acinetobacter species, and, rarely, S. aureus. Depending on the bacte- rial resistance pattern of the particular hospital and the status of host-defense mechanisms, an aminoglycoside must be added to an antipseudomonal penicillin or a third- generation cephalosporin.

TABLE IV Principal Indications for Aminoglycosides

l Infections caused by pathogens resistant to other less toxic antibiotics

The pathogenic spectrum of bacterial pneumonias ac- quired outside the hospital has clearly changed in recent years. The pneumococci that were predominant until about 1970 have been less commonly isolated in recent years. According to a study in Connecticut, the frequency of pneumococci isolated during 1980 to 1981 was 30 to 40 percent, while Legionella pneumophila, Hemophilus influ- enzae, P. aeruginosa, and other aerobic/anaerobic orga- nisms were isolated more frequently [57-601.

Severe pyelonephritis with septicemia that is due to P. aeruginosa or Enterobacteriaceae can develop in hos- pitalized patients, especially those undergoing urologic intervention or those with urogenital anomalies, including obstruction, malformation, or neurogenic bladder. In these cases, the newer and less toxic third-generation cephalo- sporins offer a valuable alternative to the aminoglycosides [61,62]. However, combination therapy with an aminogly- coside is advised when multiply resistant organisms are isolated. It should be remembered that aminoglycosides can be inactivated by high urine concentrations of calcium and magnesium ions and by a low urinary pH [63].

Endocarditis requires some special considerations. Approximately 80 to SO percent of the endocarditis cases are caused by gram-positive cocci, i.e., the enterococci, viridans streptococci, other streptococci, and coagulase- positive or coagulase-negative staphylococci. Although aminoglycosides are not intrinsically very active against these organisms, they are indicated in this situation be- cause of the severity of the infection. The frequency of endocarditis caused by gram-negative pathogens is about

For these reasons, corroborated by animal studies, endocarditis is generally treated with combined drugs: penicillin G plus streptomycin against enterococci, pro- vided that the minimal inhibitory concentration for strepto- mycin is less than 2,000 mg/liter. If the minimal inhibitory concentration is greater, penicillin G plus gentamicin is used. Penicillin G is combined with streptomycin against viridans streptococci when the minimal inhibitory concen- tration for penicillin exceeds 0.2 mgiliter, or when the streptococci are fully sensitive (i.e., the minimal inhibitory concentration for penicillin is below 0.2 mg/liter) and a short, 14-day course of therapy is planned. Gentamicin is combined with a penicillinase-stable penicillin for treat- ment of S. aureus endocarditis. Bacteremia is cleared more rapidly by drug combinations, but the clinical course is not substantially influenced. Gentamicin plus vancomy- tin plus rifampicin is used in patients with Staphylococcus epidermidis endocarditis involving prosthetic heart valves. Finally, an aminoglycoside is generally used with a beta- lactam antibiotic against gram-negative endocarditis path- ogens. In these cases, the best possible combination with regard to synergism should be sought in vitro [lo,71 -741.

Empiric therapy of intra-abdominal infections should be effective against a mixed flora, including enterococci, En- terobacteriaceae, P. aeruginosa, and strictly anaerobic organisms. An aminoglycoside in combination with clinda- mycin, a 5nitroimidazole, or cefoxitin has been effective

June 30, 1988 The American Journal of Medicine Volume 80 (suppl 86) 7

SYMPOSIUM ON AMINOGLYCOSIDE THERAPY-SIEGENTHALER ET AL

TABLE VI Combination Therapy with Aminoglycosides

Aminoglycosides are combined l to broaden the antimicrobial spectrum or l to enhance antibacterial activity

especially in situations in which other antibiotic groups alone or in combination are not as effective, i.e.,

l in life-threatening infections with unknown pathogens l in mixed aerobic/anaerobic infections l in bacterial endoctirditis l in systemic Pseudomonas infections l in neutropenic or immunodeficient patients

in these situations. Among drugs used as monotherapy, piperacillin and imipenem have the broadest spectrums of activity. According to a review by Kager and Nord [75], imipenem has already been proved very successful as monotherapy.

The third-generation cephalosporins are strong com- petitors of the aminoglycosides in the treatment of gram- negative meningitis in patients of all ages [76-781. How- ever, when combination therapy is needed to treat special pathogens, such as P. aeruginosa or Enterobacter spe- cies, or when ventriculitis is present, additional routes of administration may be necessary. For example, injection of the aminoglycoside directly into the ventricle via an Ommaya or Rickham reservoir may improve its efficacy [79-811. McCracken and co-workers [82] reported that intraventricular administration of gentamicin does not im- prove prognosis in newborns. Therefore, this form of ther- apy should not be used as routine treatment for neonatal meningitis caused by gram-negative enteric bacilli.

If infectious arthritis or acute or chronic osteomyelitis develops in infants under one month of age or in patients more than 50 years of age, the differential diagnosis should consider Enterobacteriaceae and P. aeruginosa in addition to staphylococci as possible pathogens [83-851. Empiric therapy should, therefore, consist of combining an aminoglycoside with a beta-lactamase-stable penicillin. After causative organisms are identified, therapy should be modified.

It is important to consider the spectrums of action of all available antibiotics-especially those of beta-lactam an- tibiotics and aminoglycosides-when choosing therapy for nosocomial septicemia. For newly developed sub- stances such as monobactams, carbapenems, and 5-quinolones, there is still insufficient experience in this respect. Moreover, the therapeutic gaps with individual substances are important to know. For example, the ami- noglycosides are inactive against anaerobes; the new beta-lactam antibiotics exhibit only limited activity against penicillinase-producing strains of S. aureus; and the third- generation cephalosporins are inactive against entero-

cocci and also show inadequate activity against Bacter- oides fragilis. Most penicillins are inactive against Klebsi- ella species. In addition, clinical trials have shown that during monotherapy with third-generation cephalosporins and antipseudomonal penicillins, Pseudomonas strains often become resistant, causing therapeutic failures [86- 911. Furthermore, the choice of substances has to be adapted based on the pathogens in an individual hospital. In general, a combination of a beta-lactam antibiotic with an aminoglycoside is used initially, then changed to mon- otherapy only after identification of the pathogen in a pa- tient with normal host-defense mechanisms. As men- tioned previously, the aminoglycosides have new compe- tition in the monobactams (e.g., aztreonam), the carba- penems (e.g., imipenem), and the 5-quinolones (e.g., ciprofloxacin, ofloxacin, and norfloxacin). Some of these agents possess even broader activity than the aminogly- cosides, primarily because they attack anaerobes. It may eventually be possible to use them with sufficient safety as monotherapy in immunocompetent patients with septi- cemia.

A review of the many studies in which aminoglycosides have been used as monotherapy in septicemia reveals success rates of 24 to 100 percent. In neutropenic pa- tients with septicemia, failures often occur. However, con- sistently positive results can be attained in patients with normal host defenses [92-981. Pathogens for which an aminoglycoside is the drug of choice are discussed later in this article, as are those compounds that are equally ef- fective [99]. In infections with Acinetobacter anitratus or Iwoffi, an aminoglycoside, an antipseudomonal penicillin, or one of the new 5-quinolone preparations can all proba- bly be considered equally effective as alternative therapy. Cefotaxime, ceftizoxime, ceftazidime, or imipenem (plus cilastatin) are the alternatives to an aminoglycoside in in- fections with Enterobacter species. In infections with Haf- nia alvei, an aminoglycoside is the drug of first choice, and chloramphenicol is only the drug of second choice. Against Morganella species, imipenem (plus cilastatin) will most likely be as effective as an aminoglycoside. Ami- kacin is the drug of first choice against Providencia spe- cies, with the possible alternatives being cefotaxime, moxalactam, ceftizoxime, or imipenem (plus cilastatin). Streptomycin is the drug of choice against F. tularensis, with chloramphenicol the alternative.

Amikacin is the drug of choice against Serratia marces- tens, but the new 5-quinolone preparations are, in all like- lihood, equally effective. Yersinia enterocolitica is most successfully treated with an aminoglycoside, but ceftizox- ime, ceftriaxone, and moxalactam are also effective. Streptomycin continues to be the first-line drug for treat- ment of Y. pestis infection, with no equally effective alter- native. Only prolonged clinical experience and controlled comparative studies will establish the relative reliability

8 June 30, 1986 The American Journal of Medicine Volume 80 (suppl 6B)

SYMPOSIUM ON AMINOGLYCOSIDE THERAPY-SIEGENTHALER ET AL

and value of a given drug against a specific pathogen. Combination Therapy with Aminoglycosides (Table VI). The need to broaden the spectrum of antibiotic ther- apy has led to the administration of antibiotic combina- tions. The rationale for combination therapy also includes the enhancement of antibacterial activity due to synergis- tic or additive interactions. Increased activity reduces the risk of therapeutic f&lure that might iesult when bacterial subpopulations develop resistance to one or both antibiot- ics. In addition, these synergistic or iddiiive effects often allow a reduction in dosage and in consequent tbxic side effects. The value of combining aminoglycosides with beta-lactam antibiotics is recognized worldwide. However, drug combinations may also hatie antagonistic interac- tions, increased side effects caused by both drugs, possi- pie provocation of multiply resistant organisms, misinter- pretation of therapeutic safety, and finally, higher costs.

Regardless of the potential disadvantages, in mavy clinical situations, the combination of an aminoglycoside with a beta-lactam antibiotic continues to be the optimal therapy. This is true in life-threatening infections with un- known pathogens, in mixed aerobic/anaerobic infections, in infections in neutropenic or immunodeficient patients, in bactbrial endocarditis, and in systemic Pseudomonas in- fections. Above all, clinical trials have shown that patients with cjranulocytopenia appear to benefit from combined antibiotic ttierapy. Young and co-workers [loo] and Love et al [IO11 reported a success rate of 80 p&cent in granu- locytopenic patients with septicemia when both antibiotics us&d in the combination, a beta-lactam, plus an aminogly- coside, were individually active against the causative or- ganisms. The success rate declined to about 60 percent when one of the antibiotics in the combination proved in- active icvhen tested alone. If both antibiotics were inactive in vitro, the success rate dropped below 20 percent. There are now numerous animal and clinical trials that confirm the superiority of combined antibiotic therapy in appropri- ate indications [102-l 071, a fact that had been recognized as early as 1971 by Schimpff and co-workers [108]. Today, however, in various infections, new antibiotic groups alone or in combination show similar results, a fact that has to be considered when evaluating a therapeutic regimen. Monitoring Serum Concentratiok of Aknoglyco- sides. Suggested peak lgvels to be achieved at the end of a 60-minute infusion for gentamicin, tobramycin, and netilmicin are 6 to 8 mg/liter; trough levels before the next infusion should be between 0.5 and 2 mg/liter. For ami- kacin, the corresponding peak and trough values are 20 to 30 mg/liter and between 2 and 10 mg/liter, respectively (Table VII). These values can be obtained with a mainte- nance dose of 3 to 6 mg/kg per day for gentamicin, tobra- mycin, and netilmicin. For amikacin, they can be achieved by administration of 15 to 25 mg/kg per day, divided in two

TABLE VII Suggested Peak and Trough Levels for Aminoglycosides

mg/liter

Levels

Gentamicin, Tobramycin, Netilmicin Amikacin

Peak (end of 60-minute infusion) 7 (6-8) 25 (20-30) Pseudomonas infections, pneumonia 8-12 PO-35 Trough (before next infusion) ?0.5- < 2 z2- < 10

TABLE VIII Recommended Dosages bf Aminoglycosides

Gentamicin, Tobramycin, Netilmicin Amikacin

Loading dose (mg/kg) 2 8 Maintenance dose (mg/kg per day) 3-6 15-25 Infusion period (minutes) 60 60

Dosage interval - 3 x t1/2 tlj2 = In 2 x At / In (C,&)

TABLE IX Monitorihg of Serum Concentrations of Aminoglycosides

Relationship between aminoglycoside serum concentration and clinical ~efficacy

Lack of reproducible correlation between serum level and administered dose

Correlation of serum doncentrations with nephrotoxicity and ototoxicity

to four doses and administered over a period of one hour (Table VIII). Results of clinical and microbiologic studies indi,cate that during aminoglyco$ide therapy, drug serum concentrations should be monitored (Table IX). In vitro and in vivo studies show that there is a quantitative rela- tionship between aminoglycoside serum concentrations and clinical efficacy.

In 68 patients with gram-negative infections, Noone and colleagues [log] showed thtit 46 (84 percent) of 55 pa- tients had cures with adequate gentamicin therapy, com- pared with only three (23 percent) of 13 patients who re- ceived inadequate therapy. If gram-negative Septicemia was assessed alone, 10 (91 percent) of 11 treatments were successful with adequate gentamicin therapy, whereas no success was recorded ih any of the four pa- tients with suboptimal gentamicin concentrations. From case reports of 530 patients, Moore and co-workers [l IO,1 1 I] also showed significailtly higher clire rates in patients with adequate serum levels of gentamicin, tobra- mycin, and amikacin than in patients with suboptimal drug concentrations. Finally, Anderson et al [112] demon-

June 30, 1986 The American Journal of Medicine Volume 60 (suppl 66) 9

SYMPOSIUM ON AMINOGLYCOSIDE THERAPY-SIEGENTHALER ET AL

TABLE X Lack of Reproducible Correlation between Serum Level and Administered Dose

Infusion with Amikacin (9 mg/kg bodyweight)

After After After 30 Minutes 60 Minutes 120 Minutes

Concentrations in the 30.8-50.8 17.6-30.8 10.9-18.9 serum (mgiliter)

strated “breakthrough bacteremia” in 52 (22 percent) of 237 patients undergoing antibiotic therapy for gram- negative bacteremia; of 42 samples evaiuated, 20 showed subinhibitory drug concentrations when the posi- tive blood cultures were takeri. Instead of measuring ami- noglycoside concentrations in serum, some investigators have correlated clinical outcome with serum bactericidal titers [113,114]. Klastersky and co-workers [114] reported a success rate of more than 80 percent among 317 pa- tients with tumors when the bacteriostatic activity in the serum equaled or exceeded 1:8.

Another reason for measuring serum concentrations is the lack of a reproducible correlation between the serum level and the administered dose. Barza and colleagues [I 151 measured peak serum concentrations in 23 patients after intravenous or intramuscular administration of genta- micin at 1.2 to 1.7 mgikg bodyweight, and found that lev- eis ranged from 1.7 to 7.4 mg/liter. Kaye and co-workers [116] studied 23 patients in whom a gentamicin serum concehtration of 5 mg/liter was attained one hour after intramuscular injection of doses ranging from 0.9 to 2.35 mg/kg bodyweight. A dose of 2.35 mg/kg bodyweight given to two different patients yielded extremely different peak concentrations of 5.2 mg/liter and 14 mg/liter. Good- man et al [117] measured trough and peak concentrations and found, in agreement with other authors, a high degree of variability among patients. Moreover, patients given identical doses showed different serum concentrations at different times. In our institution, a, study by Ltithy (Table X) confirmed this variability [118]: 30, 60, and 120 minutes after starting an infusion of amikacin at 9 mg/kg body- weight, the corresponding serum concentrations were 30.8 to 50.8 mg/liter, 17.6 to 30.8 mg/liter, and 10.9 to 18.9 mg/liter, respectively.

Nephrotoxicity and ototoxicity associated with the ami- noglycosides appear to be correlated with the area under the serum concentration-time curve, but there are also reports suggesting that ototoxicity depends more on peak concentrations. According to a major study by Kahlmeter and Dahlager [23], who reviewed data on about 10,000 patients over the period from 1975 to 1982, nephrotoxic side effects associated with gentamicin, tobramycin, netil- micin, and amikacin occurred at a rate of about 10 percent

(the differences among individual aminoglycosides dis- cussed earlier should be considered here). Cochleotoxic side effects are even less frequent, and the occurrence of vestibulotoxic side effects are rare.

Moore et al [119] analyzed the course of gram-negative infection and treatment iri 214 patients who had received either gentamicin or tobramycin in randomized prospec- tive clinical studies. In the contrql group without aminogly- coside therapy, a 50 percent reduction in creatinine clear- ance (the criterion of nephrotoxicity) was observed only once; however, this side effect occurred in 30 (14.1 per- cent) patients receiving gentamicin or tobramycin. After investigation and statistical evaluation of various co- factors, the following circumstances were found to be sig- nificantly associated with nephrotoxicity. In the group that showed toxicity, the peak serum level of 7.2 -t 0.4 mgiliter was higher than in the group without toxicity, which had a level of 5.3 +- 0.1 mgiliter. The trough level of 3.4 -t 0.3 mg/liter in the group with toxicity was also higher than the trough level of 2.6 + 0.1 mgiliter in the group without side effects. Patients who experienced toxicity had a higher creatinine clearance before therapy. It is possible tliat the higher initial drug “flooding” to the tubular cells contrib- uted to the development of nephrotoxicity. Some of the affected patients also had hepatic disorders, suggesting a connection between hepatic insufficiency, reduced renal blood flow, and activation of the renin-angiotensin mecha- nism. Shock states occurred more commonly in the group with toxicity and led to reduced organ perfusion. Finally, women were affected more often than men.

The data from 135 patients receiving gentamicin and tobramycin were also analyzed by Moore and co-workers [120] with reference to ototoxicity. The total dose of ami- noglycoside received was higher in, the group in which inipaired hearing developed. This group had received 3.06 ? 0.37 grams of aminoglycoside, compared with 2.01 -C 0.15 grams in the group without side effects. The duration of therapy also differed. The group with side ef- fects received aminoglycosides for 9.1 -I 0.8 days, com- pared with 6.6 + 0.1 days for the group without side ef- fects. Patients with ototoxicity also had higher fever ini- tially. It was suspected in these cases that the cytoprotec- tive prostaglandins of class E are produced in smaller quantities under the influence of fever and aminoglyco- sides. More patients with ototoxicity had an initial bacte- remia that could lead to direct cochlear injury by bacterial endotoxins and/or changes in the endolymph caused by these toxins. The peak and trough levels of aminoglyco- side were, in contrast to the situation with nephrotoxicity, not significantly associated with the development of oto- toxicity. This observation is in disagreement to some ex- tent with the studies of Wilson and Ramsden [12lj, who described a reversible cochlear damage with peak tobra- mycin concentrations above 8 to 10 mgiliter.

10 June 30, 1986 The American Journal of Medicine Volume 80 (suppl 66)

SYMPOSIUM ON AMINOGLYCOSIDE THERAPY-SIEGENTHALER ET AL

It is clear from all these studies that regular measure- ment of aminoglycoside serum concentrations can opti- mize therapy in many ways.

AMINOGLYCOSIDES AND THE FUTURE

Results of numerous clinical studies with similar indica- tions for aminoglycoside therapy are already available on new antibiotics such as carbapenems, monobactams, and 5-quinolones. Although the results regarding efficacy in most types of bacterial infections are consistently positive, it is also true that resistance to P. aeruginosa developed

2.

3.

4.

5.

6.

7.

8.

9.

10.

11.

12.

13.

14.

15.

16.

regardless of which new agent was used, resulting in ther- apeutic failures [I 22-l 241.

COMMENTS

Aminoglycosides have retained their place in the treat- ment of various infectious diseases, particularly those acquired in the hospital. Newly developed agents from other classes of antibiotics undoubtedly represent an en- richment of the clinician’s therapeutic armamentarium and, at present, appear to be effective complements in different situations to the aminoglycosides.

REFERENCES Waksman SA, Bugie E, Schatz A: Isolation of antibiotic sub-

stances from soil microorganisms, with special reference to streptothricin and strepiomycin. Proc Staff Meet Mayo Clin 1944; 19: 537-548.

Moellering RC Jr, Weinberg AN: Studies on antibiotic syner- gism against enterococci. II. Effect of various antibiotics on the uptake of C-labeled streptomycin by enterococci. J Clin Invest 1971; 50: 2580-2584.

Moellering RC Jr: In vitro antibacterial activity of the aminogly- coside antibiotics. Rev Infect Dis 1983; 5 (suppl 2): 212- 231.

Davies JE: Resistance to aminoglycosides: mechanisms and frequency. Rev Infect Dis 1983; 5 (suppl 2): 261-266.

Krogstad DJ, Korfhagen TR, Moellering RC Jr, Wennersten C, Swartz MN, Perzynski S, Davies J: Aminoglycoside-inacti- vating enzymes in clinical isolates of Streptococcus faecalis. J Clin Invest 1978; 62: 480-486.

Davies J, Courvalin P: Mechanisms of resistance to aminogly- cosides. Am J Med 1977; 62: 868-872.

Santanam P, Kayser FH: Tobramycin adenylyltransferase: a new aminoglycoside-inactivating enzyme from Staphylococ- cus epidermidis. J Infect Dis 1976; 134 (suppl): 33-39.

Verklin RM, Mandell GL: Alteration of effectiveness of antibiot- ics by anaerobiosis. J Lab Clin Med 1977; 89: 65-71.

VanScoy RE: Antituberculosis agents isoniazid, rifampin, streptomycin, ethambutol. Mayo Clin Proc 1977; 52: 694- 700.

Sande MA, Scheld WM: Combination antibiotic therapy of bac- terial endocarditis. Ann Intern Med 1980; 92: 390-395.

Sande MA, Mandell GL: Antimicrobial agents: the aminoglyco- sides. In: Goodman and Gilman, eds. The pharmacological basis of therapeutics, ed 6. New York: MacMillan Publishing Co., Inc., 1980; 1171-1173.

Greenberg LH, Momary H: Audiotoxicity and nephrotoxicity due to orally administered neomycin. JAMA 1965; 194: 827- 828.

Achord JL: Neomycin-induced malabsorption in fasting pa- tients (abstr). Gastroenterology 1969; 56: 1244.

Murray BE, Moellering RC Jr: Patterns and mechanisms of antibiotic resistance. Med Clin North Am 1978; 62: 899-923.

Speller DCE, Stephens M, Reeves DS, et al: Epidemic infec- tion by a gentamicin-resistant Staphylococcus aureus in three hospitals. Lancet 1976; I: 464-466.

Mayhall GG, Medeff G, Marr JJ: Variation in the susceptibility of strains of Staphylococcus aureus to oxacillin, cephalothin, and gentamicin. Antimicrob Agents Chemother 1976; 10:

707-712. 17. Dowding JE: Mechanisms of gentamicin resistance in Staphy-

lococcus aureus. Antimicrob Agents Chemother 1977; 12: 47-50.

18. Kauffmann CA, Ramundo NC, Williams SG, Dev CR, Phair JP, Watanakunakorn C: Surveillance of gentamicin-resistant gram-negative bacilli in a general hospital. Antimicrob Agents Chemother 1978; 13: 918-923.

19. Keys TF, Washington JA: Gentamicin-resistant Pseudomonas aeruginosa. Mayo Clin Proc 1977; 52: 797-8Oi.

20. Walker BD, Gentry LO: A randomized, comparative study of tobramycin and gentamicin in treatment of acute urinary tract infections. J Infect Dis 1976; 134 (suppl): S146-S148.

21. Appel GB, Neu HC: Gentamicin in 1978. Ann Intern Med 1978; 89: 528-538.

22. Briedis DJ, Robson HG: Comparative activity of netilmicin, gentamicin, amikacin and tobramycin against Pseudomonas aeruginosa and Enterobacteriaceae. Antimicrob Agents Chemother 1976; 10: 592-597.

23. Kahlmeter G, Dahlager JI: Aminoglycoside toxicity-a review of clinical studies published between 1975 and 1982. J An- timicrob Chemother 1984; 13 (suppl A): 9-22.

24. Lance AZ, Wrigth GE, Blair DC: Ototoxicity and nephrotoxicity of amikacin. Am J Med 1977; 62: 911-918.

25. Black RE, Lau WK, Weinstein RJ, Young LS, Hewitt WL: Oto- toxicity of amikacin. Antimicrob Agents Chemother 1976; 9: 956-961.

26. Brogden RN, Pinder RM, Sawyer PR, Speight FM, Avery GS: Tobramycin: a review of its antibacterial and pharmacoki- netic properties and therapeutic use. Drugs 1976; 12: 166- 200.

27. Marks MI, Mammerberg S, Greenstone G, Silver B: Activity of newer aminoglycosides and carbenicillin, alone and in com- bination, against gentamicin-resistant Pseudomonas aeru- ginosa. Antimicrob Agents Chemother 1976; 10: 399-401.

28. Neu HC: Tobramycin: an overview. J Infect Dis 1976; 134 (suppl): s3-s19.

29. Liithy R: Aminoglykosidantibiotika aus klinischer Sicht. Int J Clin Pharmacol 1975; 11: 112-l 25.

30. Yu PKW, Washington JA: Antimicrobial susceptibility of genta- micin-resistant Pseudomonas aeruginosa. Mayo Clin Proc 1977; 52: 802-805.

31. Moellering RC Jr, Wennersten C, Kunz LJ, Poitras JW: Resist: ante to gentamicin, tobramycin and amikacin among clinical isolates of bacteria. Am J Med 1977; 62: 873-881.

32. Smith CR, Lipsky JJ, Laskin OL, et al: Double-blind compari-

June 30, 1988 The American Journal of Medicine Volume 80 (suppl 8B) 11

SYMPOSIUM ON AMINOGLYCOSIDE THERAPY-SIEGENTCiALER ET AL

33.

34.

35.

36.

37.

38.

39.

40.

41.

42.

43.

44.

45.

46.

47.

48.

49.

50.

51.

52.

53.

54.

son of the nephrotoxicity and auditory toxicity of gentamicin .and tobramycin. N Engl J Med 1980; 302: 1106-1109.

Kumin GD: Clinical nephrotoxicity of tobramycin ‘and gentami- tin: a prospective study. JAMA 1980; 244: 1808-1810.

Schentag JJ, Cerra FB, Plaut ME: Clinical and pharmacoki- netic characteristics of aminoglycoside hephrotoxicity in 201 ill patients: Antimicrob Agents Chemother 1982; 5: 721-726.

Mauff G, Schaal KP, Pulverer G: Vergleichehde Untersuchung zur in vitro Aktivit$t von Sisomicih, Gentamicin und Tobra- mycin. Infection 1976; 4 (sup@): S300-5304.

Knothe H: Die antibakterielle Aktivitat von Gentamicin, Sisbmi- tin und Tobramycih. Infection i976; 4 (suppl): S294-S299.

Shadomy s, Jutz C, Wagner.G: In vitro studies with sisomicin, gentamicin and tobramycin. Infection 1976; 4 (suppl): S305- 5308.

Sanders CC, Sanders, WE, Goering RV: In vitro studies with Sch.21420 and Sch 22591: activity in comparison with six other aminoglycdsides and synergy with penicillin against enterococci. Antimicrob Agents Chemother 1978; 14: 178- 184.

Sack K, Kayser FH, Zullich B, Beck H, Schulz E: Nebenwirkun- gen von Aminoglykosiden: Nephrotoxizitat. Infection 1976; 4 (suppl): 231-238. ’

Meyer RD, Lewis R, CarmaltED, Finegold SM: Amikacin ther- apy for serious gram-negative bacillaty infections. Ann In- tern Med 1975; 83: 790-800.

Jotzoff M: Zur antimikrobiellen Aktivitat von Amikacin im Vergleich mit drei anderen Aminoglykosiden. Med Klin 1978; 73: 914-917.

Schiffmann DO: Evaluation of amikacin sulfate. JAMA 1977; 238,: 1547-1550.

Price KE, DeFuria MD, Pursiano TA: pmikacin, an aminoglyco- side with marked activity against antibiotic-resistant clinical isolates. J Infect Dis 1976; 134 (suppl 1): 249-261.

Holm SE, Hill B, Ldwestad A, Maller R, Vikerfors I: A prospec- tive, randomized study of amikacin and gentamicin in seri- 011s infections with focus on efficacy, toxicity and duration of serum levels above the MIC. J Antimicrob Chemother 1983; 12: 393-402.

Gerding DN, Larson TA: Aminoglycoside resistance in gram- negative bacilli during increased amikacin use. Am J Med 1985; 79 (suppl.lA): l-7.

Betts RF, Valenti WM,, Chapman SW, et al: Five-year surveil- lance of aminoglycoside usage in a university hospital. Ann Intern Med 1984; 100: 219-222.

Lee JT: Three-year experience with amikacin sulfate as an exclusive surgical aminoglycoside in a large acute-care hos- pital. Am J Med. 1985; 79 (suppl 1A): 37-42.

Shulmann ST, Yogev R: Treatment of pediatric infections with amikacin as first-line aminoglycoside. Am J Med 1985; 79 (suppl 1A): 43-50.

Carson CC, Paulson DF; Rudd C: Overview of first-line ami- kacin therapy for urologic infections. Am J Med 1985; 79 (suppl 1A): 51-54.

Panwalker Al?, Malow JB, Zimelis VM, Jackson GG: Netilmicin: clinical efficacy, tolerance and toxicity. Antimicrob Agents Cherriother 1978; 13: 170-176.

Chadwick P, Salmon G, Taylor B: Activity of netilmtcin com- pared with those of gentamicin and tobramycin against En- terobacteria and Pseudomonas aeruginosa. Antimicrob Agents Chemother 1977; 12: 301-307.

Fu KP, Neu HC: In vitro study of netilmicin compared with other aminoglycosides. Antimicrob Agents Chemother 1976; 10: 526-534.

Chiu PJS, Miller GH, Brown ADi Long JF, Waitz JA: Renal pFiarmacology of netilmicin. Antimicrob Agents Chemother 1977; 11: 821-825.

Luft FC, Yum MN, Kleit SA: Comparative nephrotoxicities of netilmicin and gentamicin in rats. Antimicrob Agents Chemo-

55.

56.

57.

58.

59.

66.

61.

62.

63.

64.

65.

66.

67.

68.

69.

70.

71.

72.

73.

74.

75.

76.

77.

78.

ther 1976: 10.: 845-849. Riff, LJ, Jackson GG: Laboratory and clinical conditions for

gentamicin inactivation by carbenicillin. Arch Intern Med 1972; 130: 887-891.;

Laughlin JE, Reeves DS: Clinical and laboratory evidence for inactivation of gentamicin by carbenicillin: Lancet 1971; I: 261-264. :

Karnad A, Alvarez S, Berk SL: Pneumonia caused by gram- negative baciili. Am J Med 1985; 70 (suppl 1A): 61-67.

Tillotson JR, Lerner M: Pneumonias caused by gram-negative bacilli. Medicine (Baltimore) 1966; 45: 65-76.

Neu HC: Optimal antibiotic therapy in bronchopulmonary infec- tions Infection 1980; 8 (suppl 1): 62-69.

Donowitz GR, Mandell GL: Empiric therapy for pneumonia. Rev Infect Dis 1983; 5 (suppl): 46-51.

Preheim LC: Complicated urinary tract infections. Am J Med i985; 79 (SUPPI 2A): 62-66.

Horowitz EA, Preheim LC, Safranek TJ,.Pugsley MP, Sanders C, Bittner MJ: Randomized, double-blkrd comparison of cef- tazidime and moxalactam in complicated urinary tract infec- tions. Antimicrob Agents Chemothe? 1985; 28: 299-301.

Minuth JN, Musher DM, Thorsteinsson SR: Inhibition of the antibacterial activity of gentamicin by urine. J Infect Dis 1976; 133: 14-21.

Kaye D: Changing pattern of infective endocarditis. Am J Med 1985; 78 (Suppl 68): 157-162.

Wilson WR, Giuliani ER, Danielson GK, Geraci JE: General considerations in the diagnosis and treatment of infective endocarditis. Mayo Clin Proc 1982; 57: ,81-85.

Griffin MR, Wilson WR, Edwards D, O’Fallon WM, Kurland LT: Infective endocarditis. Olmsted County, Minnesota, 1950 through 1981. JAMA 1985; 254: 1199-1202.

Pelletier LL Jr, Petersdorf RG: infective endocarditis: a review of 125 cases from the University of Washington hospitals, 1963-1972. Medicine (Baltimore) 1977; 56: 287-313.

Geraci JE, Wilson WR: Endocarditis due to gram-negative bacteria, report of 56 cases. Mayo Clin Proc 1982; 57: 145- 148.

Finland M, Barnes MW: Changing etiology.of bacterial endo- carditis in the antibacterial era. Ann Intern Med 1970; 72: 341-348.

Nastro LJ, Finegold SM: .Endocarditis due to anaerobic gram- negative bacilli. Am J Med 1973; 54: 482-496.

Reyes MP, El-Khatib MR, Brown WJ, Smith F, Lerner AM:.Syn- ergy between carbenicillin and an aminoglycqside (gentami- tin or tobramycin) against Pseudomonas aeruginosa iso- lated from patients with endocarditis and sensitivity of iso- lates to normal human serum. J Infect Dis 1979; 146: 19% 202.

Drake TA, Bande MA: Studies of the chemotherapy of endo- carditis: correlation of in vitro, animal model, and clinical studies. Rev Infect Dis 1983; 5 (suppl 2): 345-354.

Wilson WR, Geraci JE: Treatment of streptococcal infective endocarditis. Am J Med 1985; 78 (suppl 6B): i28-137.

Karch,mer AW: Staphylococcal endocarditis: laboratory and clinical basis for antibiotic therapy. Am J Med 1985; 78 (suppl 6B): 116-l 27.

Kager L, Nord CE: Imipenem/cilastatin in the treatment of intra- abdominal infections: a review of worldwide experience. Rev Infect Dis 1985; 7 (suppl 3): 518-521.

Nelson JD: Emerging role of cephalosporins in bacterial men- ingitis. Am J Med 1985; 79 (suppl 2A): 47-51.

Cherubin CE, Corrado ML, Nair SR, Gombert ME, Landesman S, Humberg G: Treatment of gram-negative bacillary menin- gitis: role of the new cephalosporin antibiotics. Rev Infect Dis 1982; 4 (suppl): 453-464.

Landesman SH, Corrado ML, Shah PM, Armengaud M, Barza M, Cherubin CE: Past and current roles for cephalosporin antibiotics in treatment of meningitis. Emphasis on use in

12 June 30, 1986 ihe American Journal of Medicine Volume 80 (suppl 6B)

SYMPOSIUM ON AMINOGLYCOSIDE THERAPY-SIEGENTHALER ET AL

gram-negative bacillary meningitis. Am J Med 1981; 71: 693-703.

79. Eigler JOC, Wellman WE, Rooke ED, Keith HM: Bacterial men- ingitis. General review (294 cases). Mayo Clin Proc 1961; 36: 357-365.

80. Kaiser AB, McGee ZA: Aminoglycoside therapy of gram-nega- tive bacillary meningitis. N Engl J Med 1975; 293: 1215- 1220.

81. Wright PF, Kaiser AB, Bowman CM, McKee KT Jr, Trujillo H, McGee ZA: The pharmacokinetics and efficacy of an amino- glycoside administered into the cerebral ventricles in neo- nates: implications for further evaluation of this route of ther- apy in meningitis. J Infect Dis 1981; 143: 141-147.

82. McCracken GH Jr, Mize SG, Threlkeld N: lntraventricular gen- tamicin therapy in gram-negative bacillary meningitis of in- fancy. Lancet 1980; I: 787-791.

83. Pichichero ME, Friesen HA: Polymicrobial osteomyelitis: report of three cases and review of the literature. Rev Infect Dis 1982; 4: 86-96.

84. Goldenberg DL, Cohen AS: Actue infectious arthritis: a review of patients with nongonococcal joint infections (with empha- sis on therapy and prognosis). Am J Med 1976; 60: 369- 377.

85. Wheat J: Diagnostic strategies in osteomyelitis. Am J Med 1985; 78 (suppl 6B): 218-224.

86. Gribble MJ, Chow AW, Naiman SC, et al: Prospective random- ized trial of piperacillin monotherapy versus carboxypeni- cillin-aminoglycoside combination regimens in the empirical treatment of serious bacterial infections. Antimicrob Agents Chemother 1983; 24: 388-393.

87. Platt R, Ehrlich SL, Afarian J, O’Brien TF, Pennington JE, Kass EH: Moxalactam therapy of infections caused by cephalo- thin-resistant bacteria: influence of serum inhibitory activity on clinical response and acquisition of antibiotic resistance during therapy. Antimicrob Agents Chemother 1981; 20: 351-355.

88. Neu HC: Mechanisms of bacterial resistance to antimicrobial agents, with particular reference to cefotaxime and other beta-lactam compounds. Rev Infect Dis 1982; 4 (suppl 1): 288-299.

89. Sanders CC, Sanders WE Jr, Goering RV, Werner V: Selection of multiple antibiotic resistance by quinolones, beta-lactams, and aminoglycosides with special reference to cross-resist- ance between unrelated drug classes. Antimicrob Agents Chemother 1984; 26: 797-801.

90. Sanders CC, Sanders WE Jr: Microbial resistance to newer generation beta-lactam antibiotics: clinical and laboratory implications. J Infect Dis 1985; 151: 399-405.

91. Hoogkamp-Korstanje JAA, Erpecum KJ, van Kamp H: Ceftazi- dime in serious hospital-acquired infections. J Antimicrob Chemother 1985; 15: 743-749.

92. Feld R, Valdivieso M, Bodey GP, Rodriguez V: Comparison of amikacin and tobramycin in the treatment of infections in patients with cancer. J Infect Dis 1977; 135: 61-66.

93. Jackson GG, Riff LJ: Pseudomonas bacteremia: pharmaco- logic and other bases for failure of treatment with gentami- tin. J Infect Dis 1971; 124 (suppl): 185-191.

94. Jaffe G, Ravreby W, Meyers BR, Hirschman SZ: Clinical study of the use of the new aminoglycoside tobramycin for therapy of infections due to gram-negative bacteria. Antimicrob Agents Chemother 1974; 5: 75-81.

95. Bock BV, Edelstein PH, Meyer RD: Prospective comparative study of efficacy and toxicity of netilmicin and amikacin. An- timicrob Agents Chemother 1980; 17: 217-225.

96. Valdivieso M, Feld R, Rodriguez V, Bodey GP: Amikacin ther- apy of infections in neutropenic patients. Am J Med Sci 1975; 270: 453-463.

97. Tally FP, Louie TJ, O’Keefe P, Gorbach SL, Bartlett JG: Ami- kacin therapy for severe gram-negative sepsis: efficacy in

98.

99.

100.

101.

102.

103.

104.

105.

106.

107.

108.

109.

110.

111.

112.

113.

114.

115.

116.

117.

118.

119.

infections involving gentamicin-resistant organisms. J Infect Dis 1976; 134 (suppl): 428-432.

Yap BS, Bodey GP: Netilmicin in the treatment of infections in patients with cancer. Arch Intern Med 1979; 139: 1259- 1262.

Sanford JP: Antimicrobial agents of choice against selected organisms, In: Sanford JP, ed. Guide to antimicrobial ther- apy, 1985; 34-36.

Young LS, Meyer-Dudnik LV, Hindler J, Martin WJ: Aminogly- cosides in the treatment of bacteremic infections in the im- munocompromised host. J Antimicrob Chemother 1981; 8 (suppl A): 121-132.

Love LJ, Schimpff SC, Schiffer CA, Wiernik PH: Improved prognosis for granulocytopenic patients with gram-negative bacteremia. Am J Med 1980; 68: 643-648.

Klastersky J, Meunier-Carpentier F, Prevost JM: Significance of antimicrobial synergism for the outcome of gram-negative sepsis. Am J Med Sci 1977; 273: 157-167.

Scott RE, Robson HG: Synergistic activity of carbenicillin and gentamicin in experimental Pseudomonas bacteremia in neutropenic rats. Antimicrob Agents Chemother 1976; 10: 646-651.

Baltch AL, Smith RP: Combinations of antibiotics against Pseudomonas aeruginosa. Am J Med 1985; 79 (suppl 1A): 8-l 6.

Levin S: Antibiotics of choice in suspected serious sepsis. J Antimicrob Chemother 1981; 8 (suppl A): 133-142.

Young L: Aminoglycosides in combination therapy. Chemio- terapia 1984; 3 (suppl): 38-41.

Zinner SH: Review of amikacin usage in the EORTC trials. Am J Med 1985; 79 (suppl IA): 17-20.

Schimpff S, Satterlee W, Young VM, Serpick A: Empiric ther- apy with carbenicillin and gentamicin for febrile patients with cancer and granulocytopenia. N Engl J Med 1971; 284: 1061-1065.

Noone P, Parsons TMC, Pattison JR, Slack RCB, Garfield- Davies D, Hughes K: Experience in monitoring gentamicin therapy during treatment of serious gram-negative sepsis. Br Med J 1974; I: 477-481.

Moore RD, Smith CR, Lietman PS: Association of aminoglyco- side plasma levels with therapeutic outcome in gram-nega- tive pneumonia. Am J Med 1984; 77: 657-662.

Moore RD, Smith CR, Lietman PS: The association of amino- glycoside plasma levels with mortality in patients with gram- negative bacteremia. J Infect Dis 1984; 149: 443-448.

Anderson ET, Young LS, Hewitt WL: Simultaneous antibiotic levels in “breakthrough” gram-negative rod bacteremia. Am J Med 1976; 61: 493-497.

Sculier JP, Klastersky J: Significance of the serum bactericidal test in gram-negative bacillary bacteremia in patients with and without granulocytopenia. Am J Med 1984; 76: 429- 435.

Klastersky J, Daneau D, Swings G, Weerts D: Antibacterial activity in serum and urine as a therapeutic guide in bacterial infections. J Infect Dis 1974; 129: 187-193.

Barza M, Brown RB, Shen D, Gibaldi M, Weinstein L: Predicta- bility of blood levels of gentamicin in man. J Infect Dis 1975; 132: 165-i 74.

Kaye D, Levison ME, Labovitz ED: The unpredictability of serum concentrations of gentamicin. J Infect Dis 1974; 130: 150-154.

Goodman EL, van Gelder J, Holmes R, Hull AR: Prospective comparative study of variable dosage and variable fre- quency regimens for administration of gentamicin. An- timicrob Agents Chemother 1975; 4: 434-438.

Liithy R: Basis for an adequate dosage of aminoglycoside anti- biotics Infection 1980; 8 (suppl 1): 58-61.

Moore RD, Smith CR: Lipsky JJ, Mellits ED, Lietman PS: Risk factors for nephrotoxicity in patients treated with aminogly-

June 30, 1988 The American Journal of Medicine Volume 80 (suppl 8B) 13

SYMPOSIUM ON AMINOGLYCOSIDE THERAPY-SIEGENTHALER ET AL

cosides. Ann Intern Med 1984; 100: 352-357. 120. Moore RD, Smith CR, Lietman PS: Risk factors for the devel-

opment of auditory toxicity in patients receiving aminoglyco- sides. J Infect Dis 1984; 149: 23-30.

121. Wilson P, Ramsden RD: Immediate effects of tobramycin on human cochlea and correlation with serum tobramycin lev- els. Br Med J 1977; I: 259-261.

122. Vivas JR, Creixems MR, Bouza E, et al: Evaluation of aztre- onam in the treatment of severe bacterial infections. An- timicrob Agents Chemother 1985; 28: 222-226.

123. Chalkley LJ, Koornhof HJ: Antimicrobial activity of ci- profloxacin against Pseudomonas aeruginosa, Escherichia coli, and Staphylococcus aureus determined by the killing curve method: antibiotic comparisons and synergistic in- teractions. Antimicrob Agents Chemother 1985; 28: 331- 342.

124. Eron W, Harvey L, Hixon DL, Poretz DM: Ciprofloxacin therapy of infections caused by Pseudomonas aeruginosa and other resistant bacteria. Antimicrob Agents Chemother 1985; 27: 308-310.

14 June 30, 1988 The American Journal of Medicine Volume 80 (suppl 66)