Sei sulla pagina 1di 8

Current Pharmaceutical Design, 2003, 9, 975-981 975

Infections and Urinary Stone Disease

Nadeem U. Rahman, Maxwell V. Meng, and Marshall L. Stoller*

Department of Urology, University of California School of Medicine San Francisco, California

Abstract: The relationship between urinary infections and stone formation has been recognized since antiquity and it has
been over a century since bacterial degradation of urea was postulated to cause struvite stones. Specific therapy for
urease-producing bacteria, such as urease-inhibitors and antibiotics, has allowed for treatment for this subset of urinary
stones. Future directions for research include development of novel urease-inhibitors and chemicals to enhance the
protective glycosaminoglycan layer.

An improved understanding of the pathogenesis of calcium-based stones has led to the discovery of potential roles for
nanobacteria and Oxalobacter formingenes. Methods of altering intestinal regulation of oxalate by reintroduction of lactic
acid bacteria may significantly impact the treatment of calcium oxalate stones.

The use of catheters, both urethral and ureteral, is common in the urinary tract and is associated with significant
morbidity, primarily from associated infections. Catheters to prevent bacterial colonization and formation of biofilms have
been created using various coatings, including ciprofloxacin, hydrogel, and silver. Use of these types of catheters may
minimize infections and encrustation inherent with their placement in the urinary tract.

STRUVITE CALCULI 2NH3 + H2O 2NH4+ + 2OH- (increase pH>7.2) and

Urinary tract calculi have plagued humans since the CO2 + H2O H+ + HCO3- 2H+ + CO32-
beginning of civilization and associated infections have long
been thought to play a critical role in their lithogenesis. In The hydrolysis of urea increases the local concentration
387 B.C., Hippocrates first documented an association of ammonia and increases urinary pH. The alkaline urinary
between urinary tract infections, loin abscesses, and urinary environment favors the formation of trivalent phosphate
stones. Over two thousand years later, in 1817 Alexander (PO43-), normally present in uninfected, non-alkalotic urine
Marcet noted that there was a relationship between urinary as univalent phosphate (HPO42-). It is the presence of trivalent
infections and an alkalotic urine contributing to the phosphate along with the ammonium ion in an alkaline urine
formation of phosphate stones [1]. This was further eluciated (pH >7.2) which allows the formation of magnesium ammo-
in 1901 by T. R. Brown who suggested that alkalotic urine nium phosphate crystals (MgNH4PO4 .6H2O) [7-9]. The
and stone formation was a consequence of bacterial splitting mineral was termed struvite in 1920 by the Swedish
of urea and subsequent ammonia production. In addition, as geologist Ulex, who named it after his mentor, the Russian
three of the six patients in his case series were infected with diplomat H.C. von Struve. Typically these stones are
Proteus, this was the first time that Proteus species were admixed with carbonate apatite, Ca10(PO4CO3OH)6.(OH)2.
implicated in stone formation [2]. In 1925, T.B. Magath and Hence, the term triple phosphate comes from association of
B.H. Hagar suggested that a bacterial protein, urease, must the trivalent phosphate anion with the three cations, calcium
be responsible for the breakdown of urea [3]; this was from carbonate apatite and magnesium + ammonium from
confirmed one year later upon the isolation of urease by J. B. the magnesium ammonium phosphate crystal [10].
Sumner. For isolating the first enzyme in its pure form, he
was awarded the Nobel Prize in Chemistry in 1946 [4]. Under physiological conditions, urinary ammonia levels
are maintained at an approximate concentration of 100
Bacterial urease, which is normally intracellular, is mEq/L. However, as the chemistry of the urine is altered by
released into the urine after bacterial cell lysis [5]. infection, the urinary ammonia, hydroxide, and carbonate
Subsequently, it catalyzes the hydrolysis of urea at a rate 104 levels become significantly elevated. The pathological
times that of spontaneous urea hydrolysis [6]. The following supersaturation of these ions leads to particle formation
equations summarize the conversion of urea to ammonia, which aggregate to form crystals and eventually stones. This
ammonia to ammonium, and acidification from carbon stone propogation is enhanced because ammonia also
dioxide: damages the normally protective urothelial mucopoly-
saccharide (glycosaminoglycan) layer. The struvite crystals
H2NCONH2 + H20 2NH3 + CO2 are thus thought to adhere to the damaged glycosamino-
glycan layer, allowing for adhesion to the urothelium and
*Address correspondence to this author at the Department of Urology, U-
accelerated growth of the stone. Overall, it is a combination
575, University of California, San Francisco, San Francisco, CA 94143- of the supersaturation of the ions along with increased
0738, USA; Tel: (415) 476-1611; Fax: (415) 476-8849; crystal adhesion that accounts for the large sizes and rapid
E-mail: mmeng@urol.ucsf.edu growth rates of struvite stones [11].

1381-6128/03 $41.00+.00 2003 Bentham Science Publishers Ltd.


976 Current Pharmaceutical Design, 2003, Vol. 9, No. 12 Stoller et al.

Attempts have been made to develop inhibitors of urease saminoglycans are hypothesized to prevent crystal retention
to aid in the treatment of struvite calculi. Acetohydroxamic and thus may inhibit stone formation. In a study by Senthil et
acid (AHA) has been used as such an inhibitor. It has both a al., ammonium oxalate was used to induce stone formation
high renal clearance and is able to penetrate the bacterial cell in rats [19]. The glycosaminoglycan sodium pentosan
wall. Clinical studies have demonstrated that AHA decreases polysulphate (Elmiron) was administered and was shown
the urinary alkalinity and ammonia levels even in the to significantly reduce urinary stone forming constituents.
presence of infection. A double-blind study of AHA versus However, in a trial using another synthetic glycosamino-
placebo revealed that none of the 20 patients taking AHA glycan, pentosan polysulphate, the effects as a stone inhibitor
doubled the size of their stones, the predetermined endpoint were equivocal. In this study, 121 patients were followed
in the study, compared to 7 of 19 patients receiving placebo over a period of three years both before and after treatment.
[12]. Furthermore, when one of these patients previously The study demonstrated that there were no differences in the
taking placebo started taking AHA, the stone burden stone episode and stone operation rates [20]. Although the
dramatically decreased after one month of therapy [13]. use of glycosaminoglycan for stone inhibition seems feasible
Unfortunately, the side effects of AHA, including phlebitis, theoretically, restoration of the glycosaminoglycan layer has
deep venous thrombosis, and hemolytic anemia, limit its not yet been shown to be of use clinically.
clinical utility. In addition, many patients with struvite stones
have concomitant impairment in renal function, which not Bacteria that produce urease are usually in the
only increases AHA toxicity but makes the drug less Enterobacteriacae family, with the most common pathogen
effective. It is recommended that AHA not be given to being Proteus mirablis (Table 1). Another common pathogen
patients with a serum creatinine greater than 2.5 mg/dL. The includes Ureaplasma urealyticum, which requires urea as an
clinical utility of urease inhibitors will thus remain limited obligate growth factor. All these urease-producing organisms
until drugs with fewer side effects are developed [14]. derive their nitrogen requirements from the breakdown of
urea. It should be noted that Escherichia coli, the most
ubiquitous uropathogen, rarely produces urease.
ANTI-INFECTIVES
The gastrointestinal tract harbors the largest urease-
Antibiotics alone are unlikely to sterilize the urine of producing bacterial population, including Helicobacter
patients with struvite stones, as the interstices of the stone pylori in the stomach. The discovery of H. pylori has
harboring bacteria are not adequately penetrated by drugs. revolutionized the treatment of peptic ulcer disease [21].
Thus, treatment for struvite stones requires intervention to Implicating H. pylori in the etiology of peptic ulcer disease
reduce or remove the stones. Chronic suppressive antibiotic generated tremendous excitement, because for the first time
therapy is sub-optimal but may have a role in patients with it became possible to cure and prevent ulcers with the simple
residual stone fragments at risk for chronic urinary tract use of antimicrobial agents to eradicate the bacteria. It is now
infections. It must be kept in mind that certain anti-infectives believed that H. pylori is the cause of 80% of all gastric
themselves are considered lithogenic and may be responsible ulcers and 90% of all duodenal ulcers [22]. If we look to the
for as many as 2% of all patients with stones. For instance, current situation in kidney stones, release of bacterial urease
ceftriaxone may induce kidney stones in high dosages, and the subsequent formation of struvite stones has been
especially in children. The calculi may be composed of the thought to be the only mechanism for formation of infection-
drug itself or result from changes in the urinary mileau [15]. associated kidney stones. Yet struvite stones represent less
Indinavir, a human immunodeficiency viral protease inhibi- than 15% of all urinary stone disease. Could there be other
tor, is another common medication known to cause urinary pathogens involved in the formation of kidney stones and
calculi. It is suspected that indinavir stones are composed of what would be the role of anti-microbials in their
the un-metabolized drug, indinavir monohydrate. The prevention? An analogous change in the traditional
incidence of indinavir stones varies in the literature from 3 - paradigms of stone pathogenesis may be required to
13.1% [16]. Morbidity associated with indinavir therapy is significantly impact future work on urolithiasis.
not affected by treatment duration, as patients who do and do
not develop stones had similar mean treatment times [17].
Other protease inhibitors, such as saquinavir and ritonavir, NANOBACTERIA
have not been reported to result in urinary precipitation and
stones. Recently, medical treatment with another HIV Kajander et al. suggested that not only struvite stones,
protease inhibitor, nelfinavir, resulted in urinary stone but calcium based stones, may have an infectious origin
formation [18]. Awareness of the association between certain nanobacteria [23]. These unconventional, cytotoxic bacteria
medications and urinary stones is important for early were discovered within the past decade in human blood and
identification, cessation of the drug, and initiation of blood products, as well as in urine. They are thought to be
conservative management, as the majority of these calculi the smallest living organism, nearly 100-fold smaller than
will pass spontaneously. most common bacteria. Some forms of nanobacteria may
exist in a calcified state, with a spherical coating of the cell
wall made of carbonate apatite, similar to the common
GLYCOSAMINOGLYCANS component thought to be the nidus for most stones.
Nanobacteria have been shown to cause mineralization in
As mentioned previously, ammonia significantly vitro under physiologic concentrations of calcium and
damages the urothelium and enhances stone formation by phosphate. Furthermore, the kidney is thought to play a role
promoting crystal retention. Semi-synthetic sulfated glyco- in the clearance of nanobacteria. Akerman et al. published a
Infections and Urinary Stone Disease Current Pharmaceutical Design, 2003, Vol. 9, No. 12 977

Table 1. Common Urease Producing Organisms (Data from However, much of the data regarding nanobacteria have
[8-9]) been disputed. Cisar et al. have proposed that biominerali-
zation of calcium apatite, previously attributed to nanobac-
teria, may in fact have been a contaminant. In their study,
Urease Producing Organisms examination of the biomineralization culture failed to reveal
any nucleic acid or protein. Also, it was not inhibited by
Type of sodium azide. They have explained Kajanders results by
Organism
Organism suggesting that the DNA sequences attributed to the
nanabacterium species were in fact a contaminant from a
Gram Positive Staphylococcus aureus Phyllobacterium species present in the environment [28].
Bacteria Staphylococcus epidermidis Thus, the crystallization thought to result from nanobacteria
Corynebacterium species (C .ulcerans, C. renale, C. ovis, may in fact arise from the culture medium itself. More
C. hofmanii, C. murium, C. equi)) importantly, critics have pointed out that there have been no
Mycobacterium rhodochrous group studies which confirm the presence of nanobacteria in human
Micrococcus varians calculi or human urine. Chan et al. as well as Low et al. were
Bacillus species unable to duplicate the results of Kajandar and could not
Clostridium tetani detect nanobacterial antigens in human calculi [29-30].
Peptococcus asaccharolyticus Although the controversy remains and has become publicly
Gram Bacteroides corrodens
heated, it appears unlikely that nanobacteria represent a
Negative Helicobacter pylori
pathogen responsible for the development of human kidney
Bacteria Bordetella pertussis
stones [31].
Bordetella bronchiseptica
Haemophilius influenzae
OXALOBACTER FORMIGENES
Haemophilus parainfluenzae
Proteus species (P. mirabilils, P. morganii, P. rettgeri) Oxalobacter formigenes is known to affect oxalate
Providencia stuartii metabolism and has recently been implicated in calcium
Klebsiella species (K. penumoniae, K. oxytoca) stone formation. The majority of endogenous oxalate
Pasteurella species formation originates from the metabolism of glycine,
Peseudomonas aeruginosa glyoxylate and ascorbic acid. As much as 20% of oxalate is
Aeromonas hydrophilia derived from oral ingestion and colonic absorption.
Yersinia enterocolitica Increased absorption of oxalate can occur in patients with
Brucella species high oxalate intake or in those with intestinal disorders,
Flavobacterium species including small bowel resection, inflammatory bowel
Serratia marcescens disease, steatorrhea, jejeno-ileal bypass surgery, and cystic
Mycoplasma Ureaplasma urealyticum fibrosis [32-34]. The resultant hyperoxaluria has been shown
Mycoplasma T-strain to be a significant risk factor for calcium oxalate stone
disease. In 1985, Allison et al. first isolated Oxalobacter
Yeast Cryptococcus species formigenes from human feces, showing it to be an obligate
Rhodotorula species anaerobic gram negative rod that was able to degrade oxalic
Sporobolmyces species acid. Humans have no known endogenous enzyme capable
Trichosporon cutaneum of degrading oxalate. Thus, O. formigenes plays an
Candida humicola important symbiotic role with its vertebrate host by reducing
enteric absorption of oxalate and regulating oxalate
homeostasis [35]. Allison et al. later examined bacterial
report in which viable, radioactively labeled nanobacteria oxalate degradation rates in normal humans and compared
were injected into rabbits and appeared in urine within them to those who had undergone a jejuno-ileal bypass. The
fifteen minutes [24]. Hjelle et al . reported that nanobacteria latter group had a much slower rate of degradation, and thus
or its antigens were present in polycystic kidneys and in the it was postulated that the decreased rate of oxalate
urine, implicating them as a potential microbial pathogen breakdown resulted in greater oxalate absorption and
responsible for the disease [25]. In another study by associated hyperoxaluria. This hyperoxaluria is thought to be
Kajander et al., fibroblasts were infected with nanobacteria the main risk factor for the development of calcium oxalate
and later analyzed with electron microscopy, which revealed stones in these patients [36].
intra- and extracellular crystal deposits that resembled those
found in stone disease. Additional work demonstrated that Intestinal colonization by O. formigenes begins in
nanobacterial antigens were present in 70 of 72 human infancy and by age eight, nearly all children are colonized
kidney stones analyzed [26]. Also, injecting nanobacteria with the bacteria. However, antibiotic use affects the ability
into rat kidneys suggested that kidneys stones may develop of this bacteria to colonize the gut [37]. Prolonged use of
in a nanobacteria-inoculum dependent manner. Given this antibiotics by patients with cystic fibrosis may either
data, it was thought that nanobacteria acted as a nidus to preclude natural colonization of O. formigenes or destroy the
promote apatite nucleation and crystal growth, representing a existing colonies. Sidhu et al. examined O. formigenes in
novel mechanism by which bacterial infections relate to cystic fibrosis patients, another group of patients at risk for
stone formation [27]. hyperoxaluria and calcium oxalate stone formation. These
978 Current Pharmaceutical Design, 2003, Vol. 9, No. 12 Stoller et al.

patients were shown to have decreased levels of intestinal O. oxalyl-coenzyme A decarboxylase, reduced urinary oxalate
formigenes. Stool samples and 24-hour urine collections levels and, more importantly, decreased formation of urinary
from 43 patients with cystic fibrosis and 21 healthy controls tract stones compared to controls [42].
were analyzed for O. formigenes and oxalate, respectively.
Only seven of the 43 cystic fibrosis patients (16%) were In summary, O. formigenes is likely an important
colonized with O. formigenes , compared to 15 of 21 (71%) component in human oxalate degradation and urinary oxalate
of healthy controls. In addition, none of these seven levels. Its absence from the gut can increase the intestinal
colonized patients had elevated urinary oxalate levels, while absorption of oxalate, increasing the risk for hyperoxaluria
19 of 36 uncolonized cystic fibrosis patients (53%) had and subsequent kidney stone disease. Novel therapeutic
hyperoxaluria. Thus, the regulation of urinary oxalate strategies involving oxalobacteria, including recolonization
excretion was directly correlated with colonization of O. in the intestine, require further investigation and human
formigenes in the intestine [38]. Absence of this bacteria trials.
from the gut flora is associated with an increased suscep-
tibility to hyperoxaluria, which can lead to calcium oxalate
stones in the urinary tract. An epidemiological study LACTIC ACID BACTERIA
suggested an association between recurrent urinary lithiasis
and the lack of intestinal O. formigenes [39]. Thirty-three of Campierti et al. evaluated the role of other oxalate-
44 control patients (75%) had detectable O. formigenes in degrading bacteria in stone formation [43]. They postulated
stool samples. In contrast, in patients with calcium oxalate that oral supplementation with probiotic, freeze dried lactic
stones, there was a direct correlation with the number of acid bacteria may also affect the urinary oxalate excretion, as
stone episodes and the lack of intestinal O. formigenes many of these bacteria have oxalate consuming metabolic
colonization. Twelve of 15 (80%) with only one episode of pathways. Six patients with known mild hyperoxaluria (> 40
urolithiasis were colonized with O. formigenes, while 8 of 21 mg/24 hr) were administered a mixture of freeze dried lactic
(38%) with 2-episodes and 3 of 23 (13%) with more than 5 acid bacteria (4 gm slurry, with each gram containing 2 x
episodes had O. formigenes detected in their stool. These 1011 bacteria). The specific strains of bacteria are shown in
data have been confirmed by other investigators. Tunaguntla Table 2.
et al. also correlated urinary oxalate levels with O. formigenes
In addition to the probiotic treatment, each patient was
colonization in gut. Absence of O. formigenes was
encouraged to maintain a urine output of at least 1.5 L per
associated with hyperoxaluria and an increased risk of
recurrent oxalate stone disease [40]. Sidhu et al. further day and restrict consumption of high oxalate foods. In all six
patients, significant reductions in 24-hour urinary oxalate
documented this in an in vivo rat model. Sprague Dawley
were noted after 8 weeks of treatment. Fecal excretion of
rats not colonized with O. formigenes developed hyper-
oxalate was evaluated in two patients and both were found to
oxaluria when fed oxalate-rich diets. After introduction of O.
have a large reduction of fecal oxalate levels after 30 days of
formigenes into these rats in sufficient concentrations to
treatment. Other urinary paramaters, including 24-hour
cause bowel colonization, there were significant reductions
volume, phosphate, citrate, and calcium excretion, were not
in urinary oxalate levels. In 2001, Sidhu et al. elaborated on
significantly affected. The various bacterial strains differed
their previous study where male Sprague Dawley rats were
placed on a diet to induce a state of severe hyperoxaluria. in their abilities to degrade oxalate as well as to grow in
These rats were then treated with O. formigenes for a two- media containing different oxalate levels. For example, L.
acidophilus and S. thermophilus degraded oxalate but their
week period. The rats with hyperoxaluria showed decreased
growth was inhibited by the presence of oxalate; conversely,
levels of urinary oxalate within two days of initiating
L. brevis and L. plantarum had only a modest ability to
bacterial probiotic therapy; the amount of decrease was
degrade oxalate yet was able to grow in an oxalate medium.
proportional to the dose of bacteria. These rats tolerated the
therapy, without adverse affects on health [41]. Feeding rats Only B. infantis showed a significant ability to degrade
oxalate and grow rapidly in an oxalate rich medium (Table
other oxalate-degrading factors, including the enzyme

Table 2. In Vitro Growth and Degradation of Oxalate by Different Strains of Lactic Acid Bacteria

Lactic Acid Bacteria for Probiotic Treatment

Bacteria Degradation (%) Growth in low oxalate Degradation (%) Growth in high oxalate
in low oxalate (Time zero: end point) in high oxalate (Time zero: end point)

Lactobacillus acidophilus 11.8 25: 130 3.4 25: 52

Lactobacillus plantarum 1.4 32: 270 0.0 32: 230

Streptococcus thermophilus 2.3 2.5: 28 3.1 2.5: 9

Bifidobacterium infantis 5.3 36: 300 2.2 36: 230

Lactobacillus brevis 0.9 27: 130 0.7 27: 120


Infections and Urinary Stone Disease Current Pharmaceutical Design, 2003, Vol. 9, No. 12 979

2). Thus, manipulation of the endogenous bowel flora may where free-floating organisms can arise and spread. Biofilms
represent a novel mechanism to alter oxalate absorption and make it difficult for antimicrobial agents to be effective by
ultimately impact calcium oxalate stone formation. creating a bacterial reservoir and barrier to drug penetration.
In addition, studies have demonstrated that biofilms on
urethral catheters remain intact despite antibiotics. Bacterial
FOREIGN BODIES AND INFECTION species can survive within biofilms even in the presence of
antibiotic concentrations 1000 - 1500 times greater than the
Manipulation of the urinary tract with miniaturized minimal inhibitory concentration measured in cultured
instruments has become increasingly common with the bacterial species [49].
advent of endourologic techniques. As such, patients with
kidney stones undergoing surgical therapy are theoretically Biofilms form not only on urethral catheters but on any
at an increased risk of infection. Therefore, prevention of foreign body within the urinary tract, including indwelling
catheter-related infection are potentially important ureteral stents. The rates of ureteral stent colonization vary
considerations in the prevention of urinary stones. from 68-90%, with the incidence of clinical infections
ranging between 2730%, dependent upon the time the stent
remains in vivo. Chronic infection in the presence of a
ANTIBIOTIC COATED URETHRAL CATHETERS ureteral stent can lead to encrustation or stone formation,
urinary obstruction, and ultimately urosepsis [49-50].
Nosocomial urinary tract infections are extremely
common, with an estimated incidence of 4.35 cases per 1000 Preventing the formation of biofilms by altering the
hospitalized patients. Such infections represent a significant properties of catheter and stent biomaterials has been
source of morbidity, especially when associated with undertaken to potentially decrease morbidity. In a
indwelling urethral catheters and/or ureteral stents. With randomized study of 223 patients, Foley catheters coated
urethral catheterization alone, bacteriuria will develop at a with silver alloy on both inner and outer surfaces were
rate of 5% per day [44-45]. Reports in the literature compared to Teflonised latex Foley catheters. There was a
demonstrate that symptomatic bacteremia can cause a statistically significant reduction in the incidence of
mortality rate of up to 30%. In fact, patients with urethral bacteriuria in patients with the silver-coated catheter,
catheters who subsequently developed urinary tract suggesting that this modification may reduce the incidence
infections had as high as a threefold increase in mortality of catheter associated urinary tract infections [51]. Another
compared to those patients without urethral catheters [46]. study compared the ability of urethral catheters coated with
silver or hydrogel, or immersed in antibiotic solutions to
Table 3. Types of Urethral Catheters Under Investigation to decrease the bacterial adhesion to the catheter surface.
Reduce Infection Surprisingly, these new coating materials did not reduce
bacterial adhesion whereas immersion in antibiotic solutions
had a significantly lower adhesion rate [52]. Therefore, other
Types of Urethral Catheters antibiotic associated catheters have been developed. Cho et
al. developed a gentamicin-releasing urethral catheter. In a
Silver-coated rabbit model study comparing these catheters to untreated
Gentamicin-coated silicone catheters, the surface of the gentamicin-releasing
catheter decreased colonization. Moreover, there was a
Ciprofloxacin-coated decreased rate of bacteriuria after both 3 and 5 days of
Hydrogel-coated
catheterization with the gentamicin-releasing catheters.
These studies suggest a potential short-term benefit of these
treated urethral catheters in controlling infection [53].
Catheter-related bacteriuria may arise from several Several groups have described coating urethral catheters
sources - periurethral and/or perineal organisms, colonization with a ciprofloxacin liposome hydrogel. In theory, such a
of the collecting bag, or breaks in closed-drainage systems. hydrogel would prevent bacterial adhesion to the catheter
The presence of a urethral catheter may also compromise [54-55]. Pugach et al. evaluated this hydrogel in rabbit
immune function, with decreased ability of bladder models and compared these antiobiotic treated catheters to
antibacterial defense activity. Abrasion of the bladder placebo hydrogel-coated catheters. They report that
mucosa from the tip of the catheter disrupts the bacteriuria developed twice as rapidly in the non-antiobiotic
glycosaminoglycan layer, enhancing bacterial adherence. As coated catheters when compared to the ciprofloxacin coated
a result of these multiple insults, bacteria colonizing the catheters. This decreased bacteriuria translated to a
meatus which ascend to the bladder can more easily cause significant decrease in urinary tract infections.
symptomatic infections [47-48].
In addition to the direct coating of biomaterials, oral
Bacterial colonization allows the formation of biofilms administration of certain antibiotics may also disrupt the
on both the inner and outer catheter surfaces. Biofilms are biofilm. A study of oral fluroquinolones demonstrated drug
the accumulation of organisms and their by-products and adsorption on ureteral stents, sufficient to reduce biofilm
rapidly develop on surfaces. They are thought to be formation of certain uropathogens including Escherichia
comprised of three layers -- a film which attaches to tissue coli. Forty patients with indwelling ureteral stents received
surfaces; a film made of bacteria; and an outer surface film oral ciprofloxacin and ofloxacin; subsequently, measured
980 Current Pharmaceutical Design, 2003, Vol. 9, No. 12 Stoller et al.

antibiotic levels on stent surfaces were higher than the [18] Engeler DS, John H, Rentsch KM, Ruef C, Oertle D, Suter
minimum inhibitory concentration of many organisms S. J Urol 2002; 167: 1384.
including E. coli, Pseudomonas aeruginosa, Enterococcus [19] Senthil D, Malini MM, Varalakshmi P. Ren Fail 1998; 20:
facecalis and Staphlococcus aureus [56]. Antibiotics could 573.
potentially be designed to adsorb to stents or catheters,
[20] Fellstrom B, Backman U, DanielsonB, Wikstrom B. World
negating the requirements for precoating. Additional J Urol 1994; 12: 52.
research is necessary to confirm the clinical significance of
oral antibiotic manipulation of the biofilm. [21] Marshall BJ, Warren JR. Lancet 1984; 1 (8390) 1311.
[22] Mulvihill SJ, Debas HT. In Surgery: Basic Science and
Other approaches of drug design involve the detachment Clinical Evidence Norton JA, Bollinger RR, Chang AE,
of biofilms from biomaterials. Valraeds et al. demonstrated Lowry SF, Mulvihill SJ, Pass HI, Thompson RW. Ed,
that the non-pathogenic organism lactobacillus could alter Springer: New York 2001; pp 399-411.
uropathogen adhesion. In this study, biosurfactants from [23] Kajander EO, Ciftcioglu N. Proc Natl Acad Sci USA 1998;
lactobacilli inhibited adhesion to biomaterials, further 14: 8274.
supporting the use of certain lactobacilli as probiotic therapy
[24] Akerman KK, Kuikka JT, Ciftcioglu N, Parkkinen J,
as discussed above [57]. Alternatively, small electric currents Bergstrom KA, Kuronen I and Kajander EO. SPIE Proc
iontophoresis -- are able to eradicate biofilms in vitro when 1997; 3111 436.
combined with antibiotics. This may have clinical utility for
stone disease in the sterilization of instruments and tubing [25] Hjelle JT, Miller-Hjelle MA, Poxton IR, Kajander EO,
used in the urinary tract [58]. Finally, low intensity Ciftcioglu N, Jones ML, et al. Kidney Int 2000; 57: 2360.
ultrasound may enhance antibiotic therapy and alter biofilm [26] Ciftcioglu N, Bjorklund M, Kuorikoski K, Bergstrom K,
formation [59]. Kajander EO. Kidney Int 1999; 56: 1893.
[27] Kajander EO, Ciftcioglu N, Miller-Hjelle MA, Hjelle T.
Curr Opin Nephrol Hypertens 2001; 10: 445.
REFERENCES
[28] Cisar JO, Xu DQ, Thompson J, Swaim W, Hu L Kopecko
References 60-62 are related articles recently published in DJ. Proc Natl Acad Sci USA 2000; 21: 11511.
Current Pharmaceutical Design. [29] Chan D, Jarrett T, Kavoussi L, Nelson J. J Endourol 1999;
BRS2-11 A7.
[1] Griffith DP, Osborne CA. Miner Electrolyte Metab 1987;
13: 278. [30] Low R, Dallera M, Troxel SA, RaoHGS, Das S. J Endourol
1999; BRS2-12 A7.
[2] Brown TR. JAMA 1901; 36: 1395.
[31] Abbott A. Nature 1999; 401: 105.
[3] Hagar BH, Magath TB. JAMA 1925; 85: 352.
[32] James LF. Clin Toxicol 1972; 4: 231.
[4] Sumner JB. J Biol Chem 1926; 69: 435.
[33] Williams HE, Smith LH. Am J Med 1968; 45: 715.
[5] Griffith DP. Urol Res 1979; 7: 216.
[34] Hylander E, Jarnum SS, Jensen JH, Thale M. Scand J
[6] Kramer G, Klingler HC, Steiner GE. Curr Opin Urol 2000; Gastroentereol 1978; 13: 577.
10: 35.
[35] Allison M J, Dawson K A, Mayberry W R, Foss J G. Arch
[7] Griffith DP, Osborne CA. Miner Electrolyte Metab 1987; Microbiol 1985; 141: 1.
13: 278.
[36] Allison M J, Cook H M, Milne D B, Gallagher S, Clayman
[8] Goldstone L and Griffith DP. In Renal Tract Stones: R V. J Nutr 1986; 116: 455.
Metabolic Basis and Clinical Practice Wickham JE and
Buck AC, Ed, Churchill Livingstone: Edinburgh 1990; pp [37] Sidhu H, Yenatska L, Ogden S D, Allison M J, Peck A B.
367-385. Mol Diagn 1997; 2: 87.
[9] Wang LP, Wong HY, Griffith DP. Urol Clin North Am [38] Sidhu H, Hoppe B, Hesse A, Tenbrock K, Bromme S,
1997; 24: 149. Rietschel E, Peck AB. Lancet 1998; 352: 1026.
[10] Griffith DP, Osborne CA. Miner Electrolyte Metab 1987; [39] Sidhu H, Schmidt ME, Cornelius JG, Thamilselvan S, Khan
13: 278. S R, Hesse A, Peck AB. J Am Soc Nephrol 1999; 10: S334.
[11] Rodman JS. Nephron 1999; 81S 50. [40] Tunuguntla HSGR. J Urol, 2001; 165: 246A.
[12] Williams JJ, Rodman JS, Peterson CM. N Eng J Med 1984; [41] Sidhu H, Allison M J, Chow J M, Clark A, Peck A B. J
311: 760. Urol 2001; 166: 1487.
[13] Rodman JS, Williams JJ, Peterson CM. Urology 1983; 22: [42] Sidhu H, Schmidt ME, Cornelius JG, Thamilselvan S, Khan
410. S R, Hesse A, Peck AB. J Am Soc Nephrol 1999; 10: S334.
[14] Rodman JS. Nephron 1999; 81S 50. [43] Campieri C, Campieri M, BertuzziV, Swennen E, Matteuzzi
D, Stefoni S, et al. Kidney Int 2001; 60: 1097.
[15] deMoor RA, Egberts ACG, Schroder CH. Eur J Pediatr
1999; 158: 975. [44] SchaefferAJ. In Campbells Urology Walsh PC, Retik AB,
Vaughan ED, Wein AJ. Ed, WB Saunders: Philadelphia
[16] Reilly RF, Tray K, Perzaella MA. Am J Kidney Dis 2001; 1998; Vol1 pp 533-614.
38: E23.
[45] Bronsema DA, AdamsJR, Pallares R, Wenzel RP. J Urol
[17] Saltel E, Angel JP, Futter NG, Walsh WG, ORourke K, 1993; 150: 414.
Mahoney JE. J Urol 2000; 164: 1895.
Infections and Urinary Stone Disease Current Pharmaceutical Design, 2003, Vol. 9, No. 12 981

[46] Platt R, Polk BF, Murdock B, Rosner B. N Engl J Med [56] ReidG, Habash M, Vachon D, Denstedt J, Riddell J,
1982; 307: 637. Beheshti M. Int J Antimicrob Agents 2001; 17: 317.
[47] Schaeffer AJ, Chmiel J. J Urol 1983; 130: 1096. [57] Velraeds MC, van der Belt B, van der Mei HC, Reid G,
Buscher HJ. J Med Microbiol 1998; 49: 790.
[48] SchaefferAJ. In Campbells Urology Walsh PC, Retik AB,
Vaughan ED, Wein AJ. Ed, WB Saunders: Philadelphia [58] Blenkinsopp SA, Khoury A E, Costerton JW. Appl Environ
1998; Vol1 pp 533-614. Microbiol 1992; 48: 3770.
[49] Choong S, Whitfield H. Br J Urol 2000; 86: 935. [59] Rediske AM, Roeder BL, Brown MK, Nelson JL, Robison
RL, Draper DO, et al. Antimicrob Agent Chemother 1999;
[50] Denstedt JD, Reid G, Sofer M. World J Urol 2000; 18: 237.
43: 1211.
[51] Liedberg H, Lundeberg T. Br J Urol 1990; 4: 379.
[60] Anderson VE, Osheroff N. Curr Pharm Des 2001; 7(5):
[52] Cormio L, LaForgia P, LaForgia D, Sitonen A, Rutu M. Eur 337-53.
Urol 2001; 40: 354.
[61] Shafer WM, Katzif S, Bowers S, Fallon M, Hubalek M,
[53] Cho YH, Lee SJ, Lee JY, Kim SW, Kwon IC, Chung SY, Reed M, et al. Curr Pharm Des 2002; 8(9): 695-702.
Yoon MS. Br J Urol 2001; 87: 104.
[62] Vannier-Santos MA, Martiny A, de Souza W. Curr Pharm
[54] DiTizio V, Ferguson GW, Mittelman MW, Khoury AE, Des 2002; 8(4), 297-318.
Bruce AW, DiCosmo F. Biomaterials 1998; 20: 1877.
[55] Pugach JL, TiTizio V, MittelmanMW, Bruce AW,
DiCosmo F, Khoury AE. J Urol 1999; 162: 883.
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.

Potrebbero piacerti anche