Вы находитесь на странице: 1из 7

Kidney International, Vol. 61 (2002), pp.

1495–1501

Cyclophosphamide pharmacokinetics and dose requirements in


patients with renal insufficiency
MARION HAUBITZ, FRANK BOHNENSTENGEL, REINHARD BRUNKHORST, MATTHIAS SCHWAB,
UTE HOFMANN, and DAGMAR BUSSE
Department of Nephrology, University Medical School Hannover, Hannover, and Dr. Margarete Fischer-Bosch-Institute of
Clinical Pharmacology, Stuttgart, Germany

Cyclophosphamide pharmacokinetics and dose requirements account. For optimal dosing of CYC in patients with renal
in patients with renal insufficiency. insufficiency, the severity of renal impairment and the use and
Background: Intravenous pulse administration of cyclophos- timing of hemodialysis have to be considered.
phamide (CYC) has been successfully used for the treatment of
various autoimmune diseases. These patients often present with
impaired renal function or even end-stage renal failure. Never-
theless, data concerning pharmacokinetics of CYC in renal in-
sufficiency (RI) and on hemodialysis (HD) are rare and con- Intravenous pulse therapy with cyclophosphamide
tradictory. (CYC), an alkylating agent with cytotoxic and immuno-
Methods: The pharmacokinetics of CYC (0.5 to 1 g/m2 as a suppressive properties, is an established treatment for
one-hour infusion) were determined in patients with renal in- systemic lupus erythematosus and has also been used suc-
volvement of autoimmune diseases. Group A (N ⫽ 6) patients
had a creatinine clearance (CCr) of 25 to 50 mL/min, group B cessfully in Wegener’s granulomatosis, microscopic poly-
patients’ (N ⫽ 5) CCr was 10 to 24 mL/min, and group C (N ⫽ angiitis and other autoimmune diseases [1, 2]. Renal in-
6) patients had CCr values ⬍10 mL/min and HD. Concentrations volvement is frequent in these diseases and therefore
of CYC in serum, dialysate and urine were measured by HPLC.
patients often present with renal insufficiency or even
Twelve previously investigated patients with normal renal func-
tion served as controls. terminal renal failure. CYC is a prodrug that undergoes
Results: Mean clearance (CL) of CYC was significantly re- extensive metabolism to form active (alkylating) and
duced with decreased renal function (79 vs. 57 and 47 mL/min, inactive products [3–6]. Both the metabolites and a frac-
controls vs. A and B, respectively, P ⬍ 0.05), but only moderately
lower in the patients who received a three-hour HD during the tion (up to 25%) [3, 5] of unchanged parent compound
study period (group C, 64 mL/min, NS). This resulted in re- are ultimately eliminated from the body by the kidneys
ciprocal increases in systemic drug exposure (dose corrected [7]. Despite this fact and the frequent use of CYC in the
AUC was 216, 298, 382 and 266 ␮g · h/mL · g, controls, A, B presence of impaired renal function, data regarding the
and C, respectively). Urinary excretion of CYC was markedly
reduced in all patients with RI (renal CL was 14.9 vs. 3.4, 2.4 clinical pharmacokinetics of CYC in patients with renal
and 2.1 mL/min, controls vs. A, B and C, respectively, P ⬍ insufficiency are limited and controversial. Some studies
0.001). However, in patient group C, a mean of 22% of adminis- report no changes in the pharmacokinetics and toxicity
tered CYC dose was eliminated by a three hour HD starting profile of CYC in the presence of reduced renal function,
seven hours after CYC administration. Individual CCr values
were significantly (P ⬍ 0.001) correlated with renal and sys- leading the authors to suggest that dosage adjustment is
temic CL of CYC, respectively, and negatively correlated with not recommended [3, 4, 8]. In contrast, other investiga-
dose corrected AUC. tors found a decreased clearance of both CYC [9] and its
Conclusions: Clearance of CYC is decreased in patients with
alkylating metabolites [7, 10] in association with an en-
reduced renal function, thereby resulting in an increased sys-
temic drug exposure. However, in hemodialysis-dependent pa- hanced toxicity [7]. Furthermore, few data exist regard-
tients, removal of CYC into the dialysate has to be taken into ing the impact of hemodialysis on the pharmacokinetics
of CYC, and the optimal timing of dialysis in uremic pa-
Key words: renal insufficiency, hemodialysis, intravenous pulse ther-
tients receiving intravenous CYC therapy still remains
apy, immunosuppression, cytotoxicity, systemic lupus erythematosus. controversial [11–13]. We therefore investigated the im-
pact of renal insufficiency and hemodialysis on the phar-
Received for publication August 29, 2001
and in revised form November 12, 2001 macokinetics of CYC, in order to provide additional data
Accepted for publication November 15, 2001 as a rational base for the establishment of treatment guide-
 2002 by the International Society of Nephrology lines for CYC in patients with impaired renal function.

1495
1496 Haubitz et al: Cyclophosphamide in renal insufficiency

Table 1. Patient characteristics

Controls Group A Group B Group C


Characteristics CCr ⱖ80 mL/min CCr 25–50 mL/min CCr 10–24 mL/min CCr ⬍10 mL/min
N 12 6 5 6
CCr mL/min
Mean ⫾ SD 122 ⫾ 35 33 ⫾ 9 20 ⫾ 4 4⫾4
Range 80–196 25–49 14–24 0–9
Hemodialysis No No No Yes
Gender females/males 12/0 4/2 2/3 2/4
Age years
Mean ⫾ SD 43 ⫾ 8 52 ⫾ 20 61 ⫾ 18 54 ⫾ 20
Weight kg
Mean ⫾ SD 63 ⫾ 11 79 ⫾ 15 69 ⫾ 6 66 ⫾ 8
Daily prednisolone dose mg
Mean ⫾ SD — 16 ⫾ 7 25 ⫾ 19 34 ⫾ 26

METHODS similar doses of CYC (as a one hour infusion) for the
Patients treatment of breast cancer served as the reference [5].
Fifteen patients with autoimmune diseases and im- CYC administration and sample collection
paired renal function [creatinine clearance (CCr) ⬍50 mL/
Pulse therapy with CYC consisted in monthly infusions
min] who were scheduled for a pulse therapy with CYC
with 500 to 1000 mg/m2 CYC (Endoxan威; Asta Medica,
were included in this study. The underlying diseases were
Frankfurt, Germany), administered as a one-hour infu-
histologically proven ANCA-associated vasculitis [Weg-
sion in physiological sodium chloride. The individual
ener’s granulomatosis (WG), N ⫽ 5; microscopic polyan-
dose was determined independently of the study, ac-
giitis (MPA), N ⫽ 5], systemic lupus erythematosus
cording to the clinical judgment of the treating physician.
(SLE) with lupus nephritis (N ⫽ 3), ankylosing spondy-
To avoid any time conflict between the organizational
larthritis with amyloidosis (N ⫽ 1) and rapidly progres-
requirements of the present pharmacokinetic investiga-
sive IgA nephropathy with multiple crescents in the kid-
tion and the clinical handling of the patients, the patients
ney biopsy (N ⫽ 1). The diagnosis MPA or WG was made were enrolled in the study only during their second
according to the definition of the International Consen- course of CYC pulse therapy. In addition, two patients
sus Conference at Chapel Hill and criteria described were re-investigated at a later time-point (see last sec-
previously [2]. SLE was diagnosed according to the re- tion). Although CYC is known to induce its own metabo-
vised criteria of the American College of Rheumatology lism (most likely by induction of the cytochrome P450
[14]. Patients had normal liver function as assessed by enzymes involved in its biotransformation [15, 16]), a
standard laboratory parameters. During the study course, persistence of the effects of induction generated by one
all patients were on maintenance treatment with predni- CYC infusion over the dose-interval of four weeks ap-
solone (Table 1) in addition to individual co-medication pears extremely unlikely. For example, it has been shown
(most frequently loop diuretics, ACE inhibitors, calcium for cytochrome P4503A4 that the half-life of decrease
channel antagonists, antacids and vitamin D). Further in enzyme activity after discontinuation of the enzyme
medication on the day of CYC administration included inducing agent rifampin averaged 1.5 to 2.1 days [17]. In
ondansetron and mesna. The study was approved a priori addition, pharmacokinetics of CYC in patients during
by the local ethics committee and written informed con- four consecutive treatment cycles showed no trend for
sent was obtained from each patient. an enhanced CYC elimination with increasing number
Patient characteristics are summarized in Table 1. Pa- of courses [5].
tients were divided into three groups according to renal Blood without anticoagulant was drawn before and
function, as assessed by the measurement of creatinine 1, 2, 4, 8, 14 and 24 hours after the start of the CYC-
clearance following a 24-hour urine collection. In group A infusion, centrifuged immediately and separated serum
(N ⫽ 6), CCr was between 25 and 50 mL/min; in group B stored at ⫺70⬚C until analysis. In patients with end-stage
(N ⫽ 5) the CCr values were between 10 and 24 mL/min; renal failure (group C), a three-hour bicarbonate dialysis
and group C (N ⫽ 6) had CCr levels below 10 mL/min plus using a polysulfone membrane (F6HPS 1.3 m2 surface;
hemodialysis treatment. Group C included two patients Fresenius, Bad Homburg, Germany) was initiated 7 hours
who were re-investigated with recovered renal function after the end of the CYC infusion. Dialysate flow was
in groups A and B, respectively. Twelve previously re- 500 mL/min and a blood flow of 200 mL/min was chosen.
ported patients with normal kidney function receiving Blood samples and dialysate samples were drawn after
Haubitz et al: Cyclophosphamide in renal insufficiency 1497

0, 1, 2 and 3 hours of dialysis, respectively, and stored


at ⫺70⬚C until analysis.

Measurement of CYC
Concentrations of cyclophosphamide in serum, urine
and dialysate were determined by high-pressure liquid
chromatography (HPLC) as described previously [5],
with the following modifications. Samples were spiked
with the internal standard ifosfamide and extracted using
200 mg Bond Elut C8 solid-phase extraction columns
(Baker, Gro␤-Gerau, Germany). After conditioning and
sample load the columns were washed with 4 mL of
water, then dried by applying vacuum and eluted with
1 mL of methanol. The eluate was evaporated to dryness
Fig. 1. Serum concentration-time curves of cyclophosphamide (CYC)
under a stream of nitrogen, redissolved in 1 mL of water following a one-hour infusion. Symbols are: (䉱) controls (CCr ⬎80
and extracted with 4 mL of ethyl acetate before centrifu- mL/min; CYC dose 0.8 ⫾ 0.1 g); (䊉) group A (CCr 25 to 50 mL/min;
gation, evaporation and redissolving of the organic phase CYC dose 1.4 ⫾ 0.1 g); (䊐) group B (CCr 10 to 25 mL/min; CYC dose
1.1 ⫾ 0.2 g); (䊊) group C (CCr ⬍10 mL/min, treated with a 3-hour
as previously described [5]. The column was maintained hemodialysis during the study period, CYC dose: 1.2 ⫾ 0.4g). Data are
at 37⬚C and consisted of a Prontosil 120 C18 AQ column mean ⫾ SD.
(150 ⫻ 3 mm ID, 3 ␮m particle size) with a Hypersil
C18 precolumn.
(Fig. 1) suggests that serum levels of CYC gradually
Pharmacokinetic analysis
increase as renal function decreases. Moreover, it shows
The area-under-the-curve (AUC, calculated from 0 to that a rapid fall of CYC serum concentrations occurs
infinity), systemic clearance (CL), renal clearance (Cren), during dialysis, thereby indicating effective elimination
volume of distribution (Vss), elimination half-life (t1/2), of the drug from the systemic circulation. In fact, a total
and amount of CYC excreted unchanged in urine (Ae), amount of 317 ⫾ 43 mg CYC (corresponding to 22 ⫾ 3%
were calculated by standard procedures as previously of administered dose) were recovered in the dialysate
described [5]. Total amount of CYC cleared by the three- following a three-hour dialysis in the patients with end-
hour dialysis was calculated by ⌺hour 1-3 [CYC in dialysate stage renal disease (group C). In line with these findings,
(ng/mL) · dialysate volume (mL)]. Coefficients of varia- systemic drug exposure was increased in the presence of
tion were calculated as the percentage of standard devia- renal insufficiency, the dose corrected AUC being 42%
tion to mean value. (NS) and 77% (P ⬍ 0.01) higher in groups A and B, re-
spectively, than in controls (Fig. 2). The difference was
Statistical analysis less pronounced in the patients with terminal renal fail-
ure who received a three hour dialysis during the study
All data are presented as mean ⫾ standard deviation
period (⫹23%, NS). Systemic clearance of CYC was sig-
(SD). Multiple comparisons were analyzed by ANOVA
nificantly reduced in relation to renal impairment (⫺28%,
with subsequent Tukey-Kramer tests or, if required, by
P ⬍ 0.01 and ⫺41%, P ⬍ 0.001, for groups A and B vs.
the Kruskal Wallis test (nonparametric ANOVA) with
controls, respectively), and only moderately lower in the
subsequent Dunn’s test. The correlation of CCr with dif-
HD-treated group C (⫺19%, NS; Fig. 2). Body weight
ferent pharmacokinetic parameters of CYC (dose cor- correction of systemic clearance generated similar results
rected AUC, CL, Cren) was tested by use of the Spearman (data not shown). Renal clearance of CYC was almost
Rank correlation. A value of P ⱕ 0.05 was required for negligible in patients with reduced renal function and
statistical significance. reduced by 76 to 86% when compared to controls (14.9 ⫾
3.7 vs. 3.4 ⫾ 1.6, 2.4 ⫾ 1.2 and 2.1 ⫾ 1.1 mL/min; for
controls vs. group A, group B and group C, respectively,
RESULTS
P ⬍ 0.001). Accordingly, the percentage of administered
Mean administered CYC dose was 1.4 ⫾ 0.1 g dose excreted unchanged into urine was significantly re-
(group A), 1.1 ⫾ 0.2 g (group B), 1.2 ⫾ 0.4 g (group C), duced (Fig. 2). To further substantiate a possible rela-
and 0.83 ⫾ 0.05 g (controls). Even when taking into tionship between renal function and pharmacokinetics
consideration the differences in administered doses, the of CYC, we found a significant correlation between CCr
course of the serum-concentration time curves of CYC and the renal and systemic clearance of CYC, respec-
1498 Haubitz et al: Cyclophosphamide in renal insufficiency

Fig. 2. Pharmacokinetic parameters of CYC


in patients with normal renal function (con-
trols, N ⫽ 12), patients with moderate renal
insufficiency (group A, CCr 25 to 50 mL/min,
N ⫽ 6), severe renal insufficiency (group B,
CCr 10 to 25 mL/min: N ⫽ 5) and hemodialysis-
dependent end-stage renal disease (group C,
CCr ⬍10 mL/min and treated with a 3-hour
hemodialysis during the study period, N ⫽ 6).
Data are mean ⫾ SD. *P ⬍ 0.05, †P ⬍ 0.01
and ‡P ⬍ 0.001 vs. controls; 䊊P ⬍ 0.05 vs.
patient group A.

tively (Fig. 3 A, B). Moreover, there was a significant cokinetic results, as no effect of dose on the main phar-
inverse correlation between CCr and dose corrected AUC macokinetic parameters of unchanged CYC was ob-
of CYC (Fig. 3C). served over the standard dose ranges used clinically [4, 7].
Compared to controls, terminal elimination half-life of Moreover, no significant age- and gender-related differ-
CYC was prolonged in the presence of renal insufficiency ences in pharmacokinetics of CYC have been docu-
(Fig. 2). Finally, the volume of distribution of CYC was mented in adults so far [3, 4, 7]. Modification of CYC
in the same range in the controls and the dialysis-treated metabolism by prednisolone has been reported in the
patients, but significantly lower in the patients with renal literature, although with few studies and contradictory
insufficiency who were not under hemodialysis (controls, results [7, 18–21]. In six patients, maintenance treatment
0.49 ⫾ 0.1; group A, 0.36 ⫾ 0.1; group B, 0.36 ⫾ 0.1; of 12 to 14 days with oral doses of 50 mg prednisolone
group C, 0.49 ⫾ 0.1 L/kg, P ⬍ 0.05 for group A and reduced the terminal elimination half-life (mean ⫺19%)
group B vs. controls or group C, respectively). and increased the biotransformation rate of CYC [18].
Assuming CYC metabolism is in fact induced by predni-
solone, the steroid co-medication (which was given to the
DISCUSSION patients with renal insufficiency but not to the “control”
Our results clearly indicate that pharmacokinetics of subjects) may even lead to an underestimation of the re-
CYC are altered in patients with renal insufficiency. duction of CYC elimination in our patients with reduced
Compared to patients with normal renal function, we renal function. Ondansetron, which was given as anti-
found an increased systemic drug exposure associated emetic, has been reported to decrease the AUC of cyclo-
with a reduced systemic clearance and a prolonged elimi- phosphamide by yet not completely identified mecha-
nation half-life, the magnitude of the kinetic alterations nisms [22, 23]. However, as ondansetron was given in
being related to the degree of renal insufficiency. Of similar doses to all the patients of the study (including
note, the difference in mean administered CYC doses the 12 patients with normal renal function who served
between our patient groups is unlikely to affect pharma- as controls), any effect of ondansetron on CYC pharma-
Haubitz et al: Cyclophosphamide in renal insufficiency 1499

of CYC in patients with renal insufficiency. In addition,


the smaller volume of distribution found in patients with
renal insufficiency represents a further factor contribut-
ing to an increased plasma availability of CYC.
As indicated by the results of patient group C, the
hemodialysis partly compensated the pharmacokinetic
changes induced by the reduced kidney function. Based
on the serum concentration levels before hemodialysis,
the almost completely reduced renal elimination and the
prolonged half-life of CYC, one would have expected
the highest systemic drug exposure in the patients with
terminal renal failure (group C). However, the increase
in dose corrected AUC and decrease in clearance were
less pronounced than in those patients with residual renal
function (groups A and B). This is explained by the
fact that up to 25% of administered CYC dose were
recovered in the dialysate and thus removed from the
body during the three-hour dialysis, with a consequent
reduction in overall drug exposure. If one considers that
the time-lack between the end of the CYC infusion and
the start of the hemodialysis (that is, 7 hours) equals one
half-life of CYC (which means that only about half of
the administered CYC dose can be assumed to still have
been in the patient’s body at the time of hemodialysis),
the amount of CYC removed by the dialyzer corresponds
to approximately 50% of currently present drug. Similar
to our data, Wang et al reported a drug removal of 37%
during a four-hour dialysis started directly at the end of
CYC administration in four patients with chronic renal
failure, and estimated an average recovery of 74% for
a six-hour dialysis period [11]. The fact that CYC is
dialyzable is not unexpected as it is a small un-ionized
Fig. 3. Correlation between individual creatinine clearance (CCr) val- molecule with low protein binding and a distribution
ues and renal clearance (A), systemic clearance (B) and dose corrected
area-under-the-curve (AUC; C) of CYC (0.5 to 1.0 g/m2 as a 1-hour volume that equals total body water.
infusion) in 12 patients with normal renal function (CCr ⬎80 mL/min), The establishment of a dose-effect relationship for
6 patients with moderate renal insufficiency (CCr 25 to 50 mL/min), 5 CYC is not only hampered by the general multifactorial
patients with severe renal insufficiency (CCr 10 to 25 mL/min) and (only
in panel A) 6 patients with end-stage renal disease receiving a 3-hour problem of inter-patient variability in pharmacokinetics
hemodialysis during the study period (CCr 0 to 10 mL/min). and pharmacodynamics, but also by the fact that the com-
plex metabolism and precise mechanism of action of the
drug itself are not yet completely understood [3, 4, 6, 7].
Thus, few data are available relating plasma levels of
cokinetics would be systematic and therefore would not CYC to therapeutic efficacy or toxicity in humans and,
bias the reported differences in CYC pharmacokinetics similarly, studies correlating the clinical effects of impaired
between the patient groups. renal function and CYC dosage are sparse. Whereas some
Our data are in agreement with the data of Juma, Rog- report no association between renal insufficiency and
ers and Trounce, who reported a decrease in clearance toxicity of CYC [reviewed in 4], Bagley, Bostick and De
(⫺17%) and an increase in half-life (⫹24%) of CYC in Vita observed appreciable hematological toxicity and
six patients with a CCr of 18 to 51 mL/min in comparison prolonged retention of alkylating materials in one pa-
to eight matched controls with normal renal function [8]. tient with renal failure (CCr 18 mL/min) [7], and a recent
Both the significant reduction in renal clearance of CYC case report describes unexpected severe toxicity (includ-
found in our study and the fact that the biotransforma- ing myocarditis) in a myeloma patient with renal failure
tion rate has been shown to be unaffected in patients following treatment with CYC [24]. From clinical experi-
with renal impairment [10] suggest a decreased renal ence, the use of standard CYC doses in patients with
excretion of intact parent compound to be a relevant renal insufficiency is associated with increased toxicity,
factor responsible for the decreased systemic clearance mainly leukopenia, anemia and infection. However, it is
1500 Haubitz et al: Cyclophosphamide in renal insufficiency

not clear whether this is due to an increased drug effect on the disposition of CYC and thus emphasize the impor-
or to a reduced bone marrow capacity caused by the tance of individualized dosage in this group of patients.
uremia and the underlying disease. Our results indicate For optimal dosing of CYC in patients with reduced
that the use of standard dosage in patients with severely kidney function, the degree of renal impairment and the
impaired renal function will result in an increased sys- use and timing of hemodialysis should be considered.
temic drug exposure. Based on Figure 3C, for a given dose
of CYC one would predict a 20 to 30% higher AUC in ACKNOWLEDGMENTS
patients with a CCr of ⱕ30 mL/min as compared to pa- This study was supported by the Robert Bosch Foundation, Stutt-
tients with a CCr in the range of 80 to 120 mL/min. Al- gart, Germany. The authors thank the clinical fellows, clinical center
though the clinical effects of CYC are mediated by me- nursing staff and the Zentralapotheke for their help during the study,
and Martina Flechsig, Monika Seiler, Anja Ziegler and Susanne John-
tabolites, the reduced renal loss of intact drug represents son for excellent technical assistance.
a net increase in available parent compound to be poten-
tially bioactivated. Our assumption is supported by the Reprint requests to Dr. Marion Haubitz, M.D., Department of Ne-
phrology, Medical School Hannover, 30623 Hannover, Germany.
study of Chen et al, who found that a higher CYC expo- E-mail: Haubitz.Marion@Mh-Hannover.de
sure is associated with a higher exposure toward its bio-
activated metabolites, 4-hydroxycyclophosphamide/aldo- REFERENCES
phosphamide, and which could demonstrate in a computer
1. Austin HA, Klippel JH, Bolow HE: Therapy of lupus nephritis:
simulation that a 90% reduction of the renal CL of CYC Controlled trial of prednisone and cytotoxic drugs. N Engl J Med
may be associated with a 30% increase in the AUC of 314:614–618, 1986
4-hydroxycyclophosphamide/aldophosphamide [25]. De- 2. Haubitz M, Schellong S, Göbel U, et al: Intravenous pulse admin-
istration of cyclophosphamide versus daily oral treatment in pa-
spite its clinical efficacy, CYC treatment of autoimmune tients with antineutrophil cytoplasmic antibody-associated vasculi-
disorders remains a therapy that is accompanied by seri- tis and renal involvement. Arthritis Rheum 41:1835–1844, 1998
ous side effects, and current trials are still aimed at de- 3. Grochow LB, Colvin M: Clinical pharmacokinetics of cyclophos-
phamide. Clin Pharmacokin 4:380–394, 1979
termining the most effective and least toxic regimen. The 4. Moore MJ: Clinical pharmacokinetics of cyclophosphamide. Clin
fact that, in terms of overall toxicity, a reduction of the Pharmacokin 20:194–208, 1991
total dose required is thought to be the advantage of 5. Busse D, Busch FW, Bohnenstengel F, et al: Dose escalation of
cyclophosphamide in patients with breast cancer: Consequences for
pulse therapy when compared to continuous oral regi- pharmacokinetics and metabolism. J Clin Oncol 15:1885–1896, 1997
men [2, 26] emphasizes the critical role of CYC dosage. 6. Boddy AV, Yule SM: Metabolism and pharmacokinetics of oxaza-
Based on our results, a dose reduction of 20 to 30% phosphorines. Clin Pharmacokinet 38:291–304, 2000
7. Bagley CM, Bostick FW, De Vita VT: Clinical pharmacology of
(depending on the degree of renal insufficiency) is sug- cyclophosphamide. Cancer Res 33:226–233, 1973
gested, which would still ensure adequate systemic expo- 8. Bramwell V, Calvert RT, Edwards G, et al: The disposition of
sure of parent compound by normalizing the AUC to- cyclophosphamide in a group of myeloma patients. Cancer Chemo-
ther Pharmacol 3:253–259, 1979
ward the ranges as present in patients with normal kidney 9. Juma FD, Rogers HJ, Trounce JR: Effect of renal insufficiency
function but potentially prevent additional therapy- on the pharmacokinetics of cyclophosphamide and some of its
related toxicity. metabolites. Eur J Clin Pharmacol 19:443–451, 1981
10. Mouridsen HT, Jacobsen E: Pharmacokinetics of cyclophospha-
In patients on chronic dialysis treatment CYC elimina- mide in renal failure. Acta Pharmacol Toxicol 36:409–414, 1975
tion by the dialysis procedure has to be taken into ac- 11. Wang LH, Lee CS, Majeske BL, Marbury TC: Clearance and
count. Given the high dialysis clearance of CYC, the recovery calculations in hemodialysis: Application to plasma, red
blood cells, and dialysate measurements for cyclophosphamide.
suggestion of Wagner et al to start dialysis as early as Clin Pharmacol Ther 29:365–372, 1981
two hours after CYC [13] and the recommendation of 12. Milsted RAV, Jarman M: Hemodialysis during cyclophospha-
Milsted and Jarman that no interruption of hemodialysis mide treatment. BMJ 1:820–821, 1978
13. Wagner T, Heydrich D, Bartels H, Hohorst HJ: Einflu␤ von
is required during treatment with CYC [12] implicate Leberparenchymschäden, Niereninsuffiienz und Hämodialyse auf
the risk of inadequate low immunosuppressive effect. die Pharmakokinetik von Cyclophosphamid und seinen aktivierten
As the prevention of chronic organ damage depends on Metaboliten. Arzneimittel-Forsch. Drug Res 30:1588–1592, 1980
14. Tan EM, Cohen AS, Fries JF, et al: The 1982 revised criteria for
an early effective treatment [27], this should be avoided. the classification of systemic lupus erythematosus. Arthritis Rheum
Based on our results, dialysis should not be initiated 25:1271–1277, 1982
earlier than 12 hours after CYC infusion (most easily 15. Busse D, Busch FW, Schweizer E, et al: Fractionated administra-
tion of high-dose cyclophosphamide: Influence on dose-dependent
the day after CYC administration), which would prevent changes in pharmacokinetics and metabolism. Cancer Chemother
drug removal in the early distribution phase but would Pharmacol 43:263–268, 1999
still correct for the prolonged terminal elimination phase. 16. Chang TK, Yu L, Maurel P, Waxman DJ: Enhanced cyclophos-
phamide and ifosfamide activation in primary human hepatocyte
For decades, it has been widely accepted that impaired cultures: Response to cytochrome P-450 inducers and autoinduc-
renal function would not alter the pharmacokinetic be- tion by oxazaphosphorines. Cancer Res 57:1946–1954, 1997
havior of CYC because its clearance is primarily via 17. Fromm MF, Busse D, Kroemer HK, Eichelbaum M: Differential
induction of prehepatic and hepatic metabolism of verapamil by
nonrenal mechanisms. The present data clearly demon- rifampin. Hepatology 24:796–801, 1996
strate the impact of renal insufficiency and hemodialysis 18. Faber OK, Mouridsen HAT, Skovsted L: The biotransformation
Haubitz et al: Cyclophosphamide in renal insufficiency 1501

of cyclophosphamide in man: Influence of prednisone. Acta Phar- cokinetics of high-dose cyclophosphamide and cisplatin by anti-
macol Toxicol 35:195–200, 1974 emetics. Bone Marrow Transplant 24:1–4, 1999
19. Hayakawa T, Kanai N, Yamada R, et al: Effect of steroid hormone 24. Yamamoto R, Kanda Y, Matsuyama T, et al: Myopericarditis
on activation of endoxan (cyclophosphamide). Biochem Pharmacol caused by cyclophosphamide used to mobilize peripheral blood
18:129–135, 1969 stem cells in a myeloma patient with renal failure. Bone Marrow
20. Hill DL, Laster WR Jr, Struck RF: Enzymatic metabolism of Transplant 26:685–688, 2000
cyclophosphamide and nicotine and production of a toxic cyclo- 25. Chen TL, Kennedy MJ, Anderson LW, et al: Nonlinear pharma-
phosphamide metabolite. Cancer Res 32:658–665, 1972 cokinetics of cyclophosphamide and 4-hydroxycyclophosphamide/
aldophosphamide in patients with metastatic breast cancer receiv-
21. Hanasono GK, Fischer LJ: Plasma levels and urinary excretion
ing high-dose chemotherapy followed by autologous bone marrow
of (14C)cyclophosphamide and its radioactive metabolites in rats transplantation. Drug Metab Dispos 25:544–551, 1997
pretreated with prednisolone. Biochem Pharmacol 21:272–276, 1972 26. Richmond R, McMillam TW, Luqmani RA: Optimization of cy-
22. Gilbert CJ, Petros WP, Vredenburgh J, et al: Pharmacokinetic clophosphamide therapy in systemic vasculitis. Clin Pharmacokinet
interaction between ondansetron and cyclophosphamide during 34:79–90, 1998
high-dose chemotherapy for breast cancer. Cancer Chemother Phar- 27. Exley AR, Carruthers DM, Luqmani RA, et al: Damage occurs
macol 42:497–503, 1998 early in systemic vasculitis and is an index of outcome. Quart J
23. Cagnoni PJ, Matthes S, Day TC, et al: Modification of the pharma- Med 90:391–399, 1997

Вам также может понравиться