You are on page 1of 13

ARTHRITIS & RHEUMATISM

Vol. 62, No. 1, January 2010, pp 9–21


DOI 10.1002/art.25061
© 2010, American College of Rheumatology

REVIEW

Incidence and Prevention of Bladder Toxicity From


Cyclophosphamide in the Treatment of Rheumatic Diseases

A Data-Driven Review

Paul A. Monach, Lindsay M. Arnold, and Peter A. Merkel

Introduction agents in carrying an increased risk of both common and


opportunistic infections, teratogenicity, sterility, and
Cyclophosphamide has been used since the 1960s
secondary hematologic malignancy. Cyclophosphamide,
to treat severe manifestations of autoimmune inflamma-
and the related agent ifosfamide, can also cause toxicity
tory diseases such as systemic lupus erythematosus,
to the urinary bladder, including hemorrhagic cystitis
systemic vasculitis, and systemic sclerosis (scleroderma)
(1–4). This alkylating agent, which is considered to be and bladder cancer (10,11). The prospect of preventing
the “strongest” medication commonly used by rheuma- these serious bladder toxicities with mesna (2-
tologists, has well-established efficacy in lupus nephritis mercaptoethanesulfonic acid) has led to the common
and systemic necrotizing vasculitis, conditions for which use of this agent, which is considered safe and is
it became regarded as the standard of care by the 1980s frequently recommended in textbooks and clinical
(1,2,5). Cyclophosphamide continues to be used regu- guidelines for routine use (5,12–14).
larly, in clinical practice and in clinical trials, to treat Thus, it is relevant and timely to review the data
other severe manifestations of lupus, lung disease in on the bladder toxicity of cyclophosphamide, with a
scleroderma, many types of vasculitis, and other systemic focus on the rheumatic diseases, in order to review the
inflammatory diseases (4,6–9). Despite the development rationale and evidence for prescribing mesna to prevent
of and interest in other agents to treat severe inflamma- such bladder toxicity and to outline practical recommen-
tory disease, cyclophosphamide is unlikely to be con- dations for the use of mesna in the rheumatology setting.
signed to the status “of historic interest only” any time
soon. Medical literature search strategy
Although the doses of cyclophosphamide pre-
scribed in autoimmune diseases are lower than the doses Articles related to the association of cyclo-
typically prescribed for cancer chemotherapy, in rheu- phosphamide with hemorrhagic cystitis and bladder can-
matic diseases the drug is often used for extended cer were initially identified using the following search
periods of time and, due to a high rate of clinical relapse, parameters in PubMed (www.ncbi.nlm.nih.gov): [cyclo-
treatment often requires repeated courses. Cyclophos- phosphamide AND (cystitis OR bladder cancer) AND
phamide resembles many other chemotherapeutic (vasculitis OR arteritis OR granulomatosis OR rheuma-
toid OR lupus OR scleroderma OR non-Hodgkin’s)].
This search yielded 157 articles, the abstracts of which
Dr. Monach’s work was supported in part by an Arthritis
Investigator Award from the Arthritis Foundation.
were reviewed, yielding 19 articles that either contained
Paul A. Monach, MD, PhD, Lindsay M. Arnold, PharmD, primary data on more than a few patients or reviewed
BCPS, Peter A. Merkel, MD, MPH: Boston University School of older literature. A review of the references in these 19
Medicine and Boston Medical Center, Boston, Massachusetts.
Address correspondence and reprint requests to Paul A. articles revealed only 2 other studies containing addi-
Monach, MD, PhD, Vasculitis Center, E5, Boston University School of tional primary data.
Medicine, 72 East Concord Street, Boston, MA 02118. E-mail: Articles related to the use of mesna to prevent
pmonach@bu.edu.
Submitted for publication February 24, 2009; accepted in cystitis were initially identified using the following
revised form September 25, 2009. search parameters in PubMed: [(cyclophosphamide OR
9
10 MONACH ET AL

ifosfamide) AND cystitis AND mesna]. This search


yielded 164 articles, the abstracts of which were re-
viewed, yielding 15 articles that included primary data or
reviewed the data (much of it unpublished) related to
US Food and Drug Administration (FDA) approval of
oral mesna (15). An evaluation of the articles referenced
in this latter review as well as in a clinical practice
guideline for the use of chemotherapy and radiotherapy
protectants (16) yielded only 2 articles containing addi-
tional primary data. A search of the Cochrane Database
of Systematic Reviews using “mesna” as a key word
yielded 4 articles, none of which was related to cyclo-
phosphamide or ifosfamide and bladder toxicity. A
search using “cyclophosphamide” as a key word yielded
101 articles, only 1 of which focused on adverse events
(secondary malignancies); a review of this article did not
identify additional studies relevant to the present topic.
Information pertaining to the pharmacology of
cyclophosphamide, its metabolites, and mesna was ob-
tained from textbooks and reviews and from references
obtained from these sources.

Cyclophosphamide pharmacology
Cyclophosphamide is an oxazophosphorine alky-
lating agent that causes crosslinking of DNA strands,
Figure 1. Chemical structures, metabolism, and interactions of cyclo-
thereby preventing cell division and causing cell death. phosphamide and mesna. Phosphoramide mustard is the main active
Cyclophosphamide has major activity against rapidly (alkylating) metabolite of cyclophosphamide, and acrolein is the
proliferating cells that is not cell cycle specific, and its inactive metabolite thought to be responsible for bladder toxicity.
mechanism in treating autoimmune disease is not well Conversion of aldophosphamide to phosphoramide mustard and ac-
understood. Mechanisms postulated include apoptosis rolein occurs in the plasma. Acrolein is then excreted into the urine,
and phosphoramide mustard undergoes spontaneous degradation.
induction, decreased IgG production due to B cell Both mesna and dimesna are excreted in the urine, and one-third of
suppression, and decreased production of adhesion mol- excreted dimesna is converted back to mesna in the renal tubule. The
ecules and cytokines (17). periods of time listed with mesna and dimesna indicate the length of
Cyclophosphamide is a prodrug metabolized via time after administration that these agents appear in the bladder.
the hepatic P450 enzymatic system to both active and
inactive metabolites, as outlined in Figure 1. Among
these, the inactive (nonalkylating) metabolite acrolein
(18) is presumed to be the cause of cystitis, based on its peak concentrations (25), with oral bioavailability of
subsequent excretion into and concentration in the urine 75–100%. Cyclophosphamide is primarily cleared
and on direct evidence of acute bladder toxicity in through the liver, with only 10–15% of the drug excreted
animals (19–21). Acrolein is considered likely to be the unchanged in the urine, although urine is the site of
cause of bladder cancer as well, although there is no excretion of numerous inactive metabolites (17,18). Tu-
direct evidence to support this. The great majority of bular reabsorption is high given its nonionized nature,
bladder cancers that arise following cyclophosphamide resulting in significant elimination via hepatic metabo-
treatment, as with idiopathic bladder cancers, are tran- lism. No change in the rate of toxicity among patients
sitional cell carcinomas, but rare cases of sarcomas and with liver disease has been reported (17). A low level of
other types of bladder carcinoma have been reported alkylating activity from unbound metabolites is typically
(22–24). observed in the plasma for at least 24 hours, but this may
Oral and intravenous (IV) administration of cyclo- be significantly prolonged in patients with severe renal
phosphamide have been reported to produce similar failure. However, the necessity for dose adjustments in
CYCLOPHOSPHAMIDE AND MESNA 11

Table 1. Incidence of hemorrhagic cystitis following treatment with daily oral cyclophosphamide (CYC)*
Total CYC CYC duration,
No. of patients dose, gm‡ months‡

Author, year (ref.) Disease(s) CYC dose Mesna Total Cystitis† Mean Range Mean Range
Aptekar et al, 1973 (37) SLE, WG, RA 1.3–1.5 mg/kg No 46 10 28 6–74 15 3–41
Townes et al, 1976 (39) RA 2 mg/kg No 24 4 26 19–34 7 5–9
Plotz et al, 1979 (35)§ SLE, RA 1–4 mg/kg No 54 7 48 2–152 28 1–91
Austin et al/Carette et al, SLE 1–4 mg/kg No 40 6 ND ND ND ND
1986/1983 (1/36)
Stillwell et al, 1988 (32) WG 1–2 mg/kg No 111 17/45¶ 101 5–531 38 4–144
Pederson-Bjergaard et al, 1988 (10) NHL 100 mg/m2 No 471 33# ND ND ND ND
Silver et al, 1993 (31) SSc 1–2 mg/kg No 14 2 ND ND 9 6–12
Radis et al, 1995 (38)§ RA 50–150 mg No 119 14 53 ND 32 ND
Talar-Williams et al, 1996 (33) WG 2 mg/kg No 145 42/51** 124 ND 37 ND
Reinhold-Keller et al, 2000 (34) WG 2 mg/kg Yes 142 17†† 129 42–350 29 9–77

* There was some overlap of patients in the studies by Plotz et al, Austin et al, and Carette et al. SLE ⫽ systemic lupus erythematosus; WG ⫽
Wegener’s granulomatosis; RA ⫽ rheumatoid arthritis; ND ⫽ no data; NHL ⫽ non-Hodgkin’s lymphoma; SSc ⫽ systemic sclerosis.
† The definition of cystitis differed between studies; cystoscopy was not required for the diagnosis, except as noted otherwise.
‡ Among the subset of patients with cystitis, unless noted otherwise.
§ Data for CYC dose and duration were based on all patients in the study; no data were available for the subset of patients with cystitis.
¶ In the smaller number of patients (n ⫽ 17), cystitis was defined by gross hematuria or was confirmed by cystoscopy; the larger number of patients
(n ⫽ 45) was calculated based on the percent of patients with hematuria in whom cystitis was confirmed by cystoscopy (65%).
# Defined by gross hematuria or confirmed by cystoscopy.
** In the smaller number of patients (n ⫽ 42), cystitis was confirmed by cystoscopy; the larger number of patients (n ⫽ 51) was calculated based
on the percent of patients with hematuria in whom cystitis was confirmed by cystoscopy (70%).
†† Confirmed by cystoscopy.

patients with renal dysfunction is not well documented granulomatosis (WG) and in additional cohorts of pa-
but is commonly advised (5,17,18,26). tients with systemic lupus erythematosus (SLE), rheu-
For the treatment of autoimmune diseases, cyclo- matoid arthritis (RA), or non-Hodgkin’s lymphoma
phosphamide dosing is typically 1.5–2 mg/kg/day when it (NHL) (1,10,31–39) (Table 1). The strategy of encour-
is provided orally or 500–1,000 mg/m2 IV in monthly or aging patients to drink large volumes of fluids was
biweekly pulse-dose regimens (3,5,7,8,27–30). For com- specifically mentioned in a few studies (32,35,37), but
parison, treatment of malignancies often involves doses compliance with these directions was not assessed. The
⬎1,000 mg/m2 administered on weekly or biweekly methods of diagnosing hemorrhagic cystitis differed
schedules in combination with other drugs or radiation somewhat among these studies, but in all cases the initial
therapy. It is thus difficult to extrapolate outcomes and screening involved routine urinalyses during the period
toxicities from the oncology literature to patients given of treatment. In the studies in which the diagnosis was
cyclophosphamide for the treatment of autoimmune
usually (32,33) or always (34) confirmed by cystoscopy,
diseases.
the incidence of hemorrhagic cystitis ranged from 12%
to 41%. The mean cyclophosphamide exposure of pa-
Association between treatment with daily oral tients who had hemorrhagic cystitis was ⬎100 gm over a
cyclophosphamide and hemorrhagic cystitis mean of ⬎30 months, with a wide range of both the total
The mode of cyclophosphamide administration dose and the duration of treatment (Table 1). Few
that has generated the most concern with regard to studies provide any information about the severity of
bladder toxicity is daily oral dosing, because the duration hemorrhagic cystitis, in terms of either the degree or
of treatment and the total cumulative exposure are duration of discomfort or the need for transfusion. From
generally higher compared with intermittent IV dosing, these limited data, cystitis severe enough to require
and hydration and bladder evacuation cannot be as- transfusion appears to be uncommon (occurring in 3 of
sured. 69 patients in 3 studies) (32,33,37). Although many
The development of hemorrhagic cystitis during patients with WG or SLE had renal involvement, no
treatment with oral cyclophosphamide has been evalu- data relating the risk of cystitis to renal dysfunction were
ated in 3 large cohorts of patients with Wegener’s reported in any study.
12 MONACH ET AL

Table 2. Incidence of bladder cancer following treatment with daily oral cyclophosphamide (CYC)*
No. of patients CYC treatment

Bladder Latency, Duration, Total Followup,


Author, year (ref.) Disease(s) CYC dose Total cancer years OR 95% CI years dose, gm† years‡
Aptekar et al, 1973 (37) SLE, WG, RA 1.3–1.5 mg/kg 46 0 NA NA NA NA ND
Plotz et al, 1979 (35) SLE, RA 1–4 mg/kg 54 2 6–7 100 ND 2–4 ND 4.5
Austin et al/Carette et al, SLE 1–4 mg/kg 40 1 4 ND 4 ND 7.1
1983/1986 (1/36)
Stillwell et al, 1988 (32) WG 1–2 mg/kg 111 3 1–14 ND 1–12 56–531 6.0
Pederson-Bjergaard et al, NHL 100 mg/m2 471 7 5–12 6.8 3.2–14.2 2.8–4.2 83–129 3.8
1988 (10)
Radis et al, 1995 (38) RA 50–150 mg 119 9 4–23 22 8.8–56 5§ 120 ⫾ 56 13.1
Talar-Williams et al, 1996 WG 2 mg/kg 145 7 0.6–15 31 13–65 0.6–5.1 19–251 8.5
(33)
Westman et al, 1998 (40) WG, MPA 2 mg/kg 123 3 ⬎5 4.8 1.0–13.9 ⬎1 ND 4.6
Reinhold-Keller et al, WG 2 mg/kg 142 1 7 ND 6 350 7.0
2000 (34)
Knight et al, 2004/2002 WG ND 1,065 11 4–15 4.8 2.6–8.1 7.2¶ 0–234 ND
(42,43)
Faurschou et al, 2008 (41) WG 1–2 mg/kg 293 5 7–18 3.6 1.2–8.3 ⬎1# ⬎36# 6.0

* There is some overlap of patients in the studies by Plotz et al, Austin et al, and Carette et al. Latency is defined as the time between beginning
cyclophosphamide treatment and the diagnosis of bladder cancer. In the study by Reinhold-Keller et al, oral mesna was also given routinely. Odds
ratios (ORs) and 95% confidence intervals (95% CIs) for the development of bladder cancer represent the comparison of CYC-treated patients with
control subjects. SLE ⫽ systemic lupus erythematosus; WG ⫽ Wegener’s granulomatosis; RA ⫽ rheumatoid arthritis; NA ⫽ not applicable; ND ⫽
no data; NHL ⫽ non-Hodgkin’s lymphoma; MPA ⫽ microscopic polyangiitis.
† Values are the range or mean ⫾ SD, for patients in whom bladder cancer developed.
‡ For the studies by Aptekar et al, Plotz et al, Austin et al, Carette et al, Stillwell et al, Pederson-Bjergaard et al, and Radis et al, values are the means;
for all other studies, values are the medians.
§ Value is the mean.
¶ Value is the median.
# Values are for 4 of 5 patients with bladder cancer.

Association of daily oral cyclophosphamide with an for WG, RA, and NHL was clearly observed; an analo-
increased risk of bladder cancer gous risk was not identified with certainty among pa-
tients with SLE, but a smaller number of patients with
The incidence of bladder cancer following treat-
ment with daily oral cyclophosphamide has been re- SLE was studied (Table 2). In 3 studies, 2 of which
ported for many of the same cohorts used to investigate involved WG (31,40) and 1 of which involved NHL (10),
hemorrhagic cystitis (1,10,32–38) and for several sepa- time-dependent analyses demonstrated that the preva-
rate cohorts that were used to measure the incidence of lence of bladder cancer reached 10% between 11 years
multiple types of cancer (40–43) (Table 2). Most of the and 16 years after the beginning of treatment with
analyses were based on single-center cohorts, with odds cyclophosphamide. The largest study of bladder cancer
ratios (ORs) for the development of bladder cancer following cyclophosphamide treatment described 31
usually calculated in comparison with registry data. cases among 6,171 patients with NHL (5.0%) (44).
Although the number of cases of cancer observed Results of this study are more difficult to compare with
in each cohort was small (range 0–11), resulting in large those of the other studies (and thus are not included in
confidence intervals (CIs) for the ORs for cancer risk, a Table 2) due to the variety of cyclophosphamide regi-
substantially elevated risk of bladder cancer associated mens used and the frequent concomitant use of radia-
with cyclophosphamide treatment was observed in all tion therapy (an independent risk factor for bladder
studies (OR range 3.6–100). The total cumulative doses cancer).
of cyclophosphamide given to patients in whom bladder The yearly incidence of bladder cancer among
cancer subsequently developed varied widely but were white persons in the US has been estimated at 40 per
⬎100 gm in the majority of cases and ⬎30 gm in the 100,000 men and 10 per 100,000 women (45). This
great majority (missing data preclude the calculation of incidence rises with age; therefore, the baseline risk for
percentages) (Table 2). An elevated risk of bladder patients with WG, as used to calculate ORs as described
cancer in association with cyclophosphamide treatment above (10,40,41), is somewhat higher: 58–67 per
CYCLOPHOSPHAMIDE AND MESNA 13

Table 3. Association of bladder cancer with a prior diagnosis of hemorrhagic cystitis in patients treated
with daily oral cyclophosphamide*
No bladder
Bladder cancer cancer

Author, year (ref.)/ No No


cystitis definition Disease Cystitis cystitis Cystitis cystitis P†
Stillwell et al, 1988 (32) WG
Cystoscopy 3 0 14 94 0.003
Pedersen-Bjergaard et al, 1988 (10) NHL
No data 2 5 31 433 0.08
Radis et al, 1995 (38) RA
Nonglomerular hematuria 5 4 13 97 0.004
Cystoscopy 3 6 11 99 0.07
Talar-Williams et al, 1996 (33) WG
Nonglomerular hematuria 6 1 45 93 0.008
Cystoscopy 5 2 46 92 0.10
Reinhold-Keller et al, 2000 (34) WG
Cystoscopy 1 0 16 125 0.12
Total
Nonglomerular hematuria 17 10 119 842 ⬍0.0001
Cystoscopy 14 13 118 843 ⬍0.0001

* In the study by Reinhold-Keller et al, oral mesna was also given routinely. Cystitis was defined by the
presence of nonglomerular hematuria or was proven by cystoscopy. Both definitions were used separately
to calculate statistics, and 2 separate totals were calculated and analyzed due to the use of both definitions
in 2 of the studies. WG ⫽ Wegener’s granulomatosis; NHL ⫽ non-Hodgkin’s lymphoma; RA ⫽
rheumatoid arthritis.
† For the association of cystitis with bladder cancer, using Fisher’s exact test for each study individually
and the Mantel-Haenszel test for the total, stratified by study.

100,000. Smoking is the best-documented environmental cancer are associated with the cumulative dose of cyclo-
risk factor for bladder cancer and increases the risk phosphamide and the duration of treatment, a cause-
⬃3-fold (46). Among the studies referenced above in and-effect relationship between cystitis and cancer can-
which tobacco use was reported, more than half of the not be firmly established.
patients in whom bladder cancer developed were smok- There are 2 caveats to this apparent association.
ers or former smokers (10,32,33,35,38). First, it is possible that selection bias influenced these
Thus, treatment with oral cyclophosphamide con- studies: patients in whom hematuria develops early in
fers an independent, substantial, and probably dose- the course of treatment are much more likely to be
related increased risk of bladder cancer, with an effect evaluated later—by history, urinalysis, urine cytology,
that may be delayed for many years after the medication and/or screening cystoscopy—than are patients who
has been discontinued. never had such abnormalities. Second, a registry-based
case–control study does not support the conclusion that
Probable association of hemorrhagic cystitis with bladder cancer is associated with a history of cystitis.
increased future risk of bladder cancer Knight et al (42) reported that only 1 of 11 patients with
In most studies examining the risk of bladder bladder cancer (all of whom had been treated for WG
cancer following treatment with daily oral cyclo- with oral cyclophosphamide) had a history of cystitis
phosphamide, bladder cancer was associated with previ- documented by cystoscopy, and that hematuria in the
ous hematuria during cyclophosphamide treatment other 10 patients was first noted shortly before the
and/or with documented cystitis (32–34) (Table 3). Sim- diagnosis of cancer. In contrast, 18 of 25 control subjects
ilarly, the available data appear to confirm a strong had a history of microscopic hematuria (42). However, it
association between prior cystitis and the subsequent is unclear whether a uniform screening strategy was
risk of developing bladder cancer, with an OR of 7.2 used, or whether a distinction between glomerular (i.e.,
(95% CI 1.9–27) obtained using the Mantel-Haenszel associated with red blood cell casts or declining renal
estimator. However, because both cystitis and bladder function) and nonglomerular hematuria could be made.
14 MONACH ET AL

Table 4. Incidence of hemorrhagic cystitis following treatment with intermittent IV cyclophosphamide (CYC) for rheumatic diseases*
Total CYC CYC duration,
No. of patients dose, gm years

Author, year (ref.) Disease(s) CYC dose Mesna Total Cystitis Mean Range Mean Range
2
Austin et al, 1986 (1) SLE 0.5–1.0 gm/m every 3 mos No 20 0 ND ND 4 ND
Hoffman et al, 1990 (48) WG 1.0 gm/m2 every 1–3 mos No 14 1† ND ND 0.8 0.1–1.8
Gourley et al, 1996 (3) SLE 2
0.75 gm/m every 1–3 mos Yes 55 0 ND ⬍15 ND ⬍3
Martin et al, 1997 (50) SLE, vasc 0.5–1.0 gm/m2 every mo No 75 0 5 ND 0.5 ND
Guillevin et al, 1997 (53) WG 0.7–1.0 gm/m2 every 3–6 wks No 27 1 28 ND ND ND
Adu et al, 1997 (54) WG, MPA, PAN 15 mg/kg every 2–6 wks Yes 24 0 ND ⬍30 ND ⬍1.5
Martin-Suarez et al, 1997 (55) SLE, vasc, myo 0.5 gm every 1–4 wks Yes/no 90 1‡ 3 1–25 0.3 0.0–3.7
Koldingsnes et al, 1998 (51) WG 15 mg/kg every 2–8 wks No 11 0 ND 12–88 3.3 1.0–8.2
Haubitz et al, 1998 (52) WG, MPA 0.75 gm/m2 every mo ⫻ 12 No 22 0 16 ⬍20 ND ⬍1
Ginzler et al, 2005 (6) SLE 0.5–1.0 gm/m every mo ⫻ 6
2
No 69 0 ND ⬍9 ND 0.1–0.5
Hoyles et al, 2006 (8) SSc 0.6 gm/m2 every mo ⫻ 6 No 22 0 6 ND ND 0.1–0.5
Goransson et al, 2008 (49) SLE, WG, myo 15 mg/kg every mo No 42 0 10 1–52 1.2 0.0–8.8

* The total cyclophosphamide dose and the duration of therapy were based on all patients in the study. IV ⫽ intravenous; SLE ⫽ systemic lupus
erythematosus; ND ⫽ no data; WG ⫽ Wegener’s granulomatosis; vasc ⫽ vasculitis; MPA ⫽ microscopic polyangiitis; PAN ⫽ polyarteritis nodosa;
myo ⫽ myositis; SSc ⫽ systemic sclerosis.
† Patient also had a history of cystitis with oral cyclophosphamide.
‡ Patient was 1 of 8% of patients in this case series who were not treated with mesna.

In summary, the available data indicate, with difference in bladder toxicity between daily oral and
some caveats, that antecedent hemorrhagic cystitis is periodic IV cyclophosphamide regimens is real, it is
associated with a future increased risk of bladder cancer. possible that this finding simply reflects the total dose of
cyclophosphamide rather than the timing or route of
Lack of evidence for a strong association of periodic administration. Few patients in studies of periodic IV
IV cyclophosphamide with hemorrhagic cystitis or cyclophosphamide have received more than 30 gm, in
bladder cancer marked contrast to studies of bladder toxicity in patients
given daily oral cyclophosphamide (compare the total
Periodic, usually IV, dosing of cyclophosphamide doses in Tables 1, 2, and 4). Regardless of the reason,
has been the standard of care for lupus nephritis for ⬎20
however, it is clear that the risk of hemorrhagic cystitis
years (1,3,6), and similar regimens have been used for
with periodic IV dosing of cyclophosphamide as used in
other severe rheumatic diseases (8,28,47). Mesna is used
rheumatology is quite low, in contrast to the experience
more frequently in this setting than is daily oral therapy,
with high-dose IV cyclophosphamide and ifosfamide as
likely due to convenience or by the adoption of a
practice that is common in oncology. However, even used in oncology (see discussion below).
when mesna has not been routinely used, few cases of Additional, although indirect, support for a low
hemorrhagic cystitis have been reported in clinical trials risk of bladder cancer with periodic IV cyclo-
or additional case series of patients treated with periodic phosphamide comes from observational studies. Un-
IV cyclophosphamide for rheumatic diseases. Three selected patients with lupus do not appear to be at
cases were identified among 471 treated patients in the elevated risk of bladder cancer (OR 1.23, 95% CI
studies listed in Table 4, and the great majority of these 0.66–2.11), based on a multicenter cohort study of 9,547
patients did not receive mesna (1,3,6,8,48–55). The patients followed up for an average of 8 years (59).
development of bladder cancer after pulse regimens has Exposure to cyclophosphamide was not reported in this
been described in only a few case reports (56–58). study; however, based on the frequency of severe nephri-
It is important to note that cystitis and bladder tis in SLE and the near-universal use of this treatment
cancer were not the primary topics of investigation in during the period of study (59), one can expect that at
these studies; thus, there are no descriptions of strate- least 30% of the patients received periodic IV cyclo-
gies for systematic screening for hematuria or subse- phosphamide for at least 6 months. Several smaller
quent cystoscopy, and the duration of followup was studies have also consistently failed to document an
likely too short (median ⬍5 years) to detect many excess risk of bladder cancer in patients with lupus
bladder cancers. Even if one assumes that the reported (60–65).
CYCLOPHOSPHAMIDE AND MESNA 15

Mesna pharmacology When mesna is given orally, the dose should be equal to
40% of the cyclophosphamide dose (oral or IV), based
Mesna is a thiol compound, completely lacking in
on the 50% oral bioavailability of mesna. For conve-
antineoplastic activity, that is used in conjunction with
nience, a combination of IV and oral doses can be given:
oxazophosphorine alkylating agents as a uroprotective
an initial IV dose (equal to 6.8% of the cyclo-
agent. Mesna is thought to decrease the incidence of
phosphamide dose) followed by 2 oral doses (each equal
cystitis and bladder cancer by counteracting the toxic
to 13.3% of the cyclophosphamide dose). If the first dose
effects of acrolein (16,66,67). The administration of
of mesna is administered orally, it should be given 2
mesna with cyclophosphamide for the prevention of
hours before cyclophosphamide (oral or IV), but the
bladder toxicity is a practice derived primarily from second and third oral doses can still be given 4 hours and
investigations with ifosfamide, a structural analog of 8 hours after cyclophosphamide, as with IV mesna
cyclophosphamide. dosing.
Following either oral or IV administration, Two forms of mesna are available for oral use.
mesna undergoes rapid dimerization within plasma to The IV solution, at a concentration of 10 mg/ml, can be
the inactive disulfide compounds dimesna, mesna- ordered by outpatient pharmacies in the US, at a
cysteine, mesna-glutathione, and others (67). Both wholesale cost of approximately $0.55 per typical daily
mesna and dimesna are cleared quickly by the kidneys. dose of 60 mg. A tablet is available but at a cost of
Dimesna undergoes filtration by the glomerulus, and approximately $4.50 per 60 mg, and the only strength
roughly one-third is then converted back to mesna in the sold is 400 mg, making dosing for patients with rheu-
renal tubules by glutathione reductase (66). Mesna then matic diseases both expensive and technically challeng-
binds to acrolein within the urine. The free sulfhydryl ing. The principal disadvantage of using the IV solution
group found in mesna combines directly with the double orally is the strong sulfurous odor and taste; mixing with
bond of acrolein as well as other urotoxic a carbonated beverage or juice drink is recommended.
4-hydroxyoxazaphosphorine metabolites (16,66) (Figure Beyond the issues of taste and inconvenience, mesna is
1). The conversion of 4-hydroxycyclophosphamide to generally well tolerated, with mild gastrointestinal toxi-
acrolein may also be inhibited by mesna (68). Both of city being the most common side effect. Allergic reac-
these factors contribute to the inhibition of acrolein tions to mesna have been reported but are considered
binding to cell-surface proteins in the bladder, thereby rare (69).
potentially limiting cyclophosphamide-associated toxi- Although renal dysfunction might theoretically
cities. Urinary mesna concentrations greatly exceed increase the risk of systemic cyclophosphamide toxicity
plasma concentrations, allowing for rapid clearance of due to a decreased rate of elimination of the drug and its
acrolein from the urinary system. However, mesna does metabolites, renal insufficiency seems unlikely to lead to
not protect against nonurologic toxicities associated with an increased rate of bladder toxicity, because the deliv-
the oxazaphosphorines. ery of acrolein to the bladder is also reduced. Similarly,
Mesna has 50–75% bioavailability after oral ad- because mesna requires active glomerular filtration and
ministration and is not affected by food intake (16,66). tubular secretion in order to reach the urine, use of
Urinary thiol concentrations following oral administra- mesna in patients with significant renal dysfunction is
tion are approximately one-half those observed after IV unlikely to be beneficial. However, the relative urinary
administration. Oral administration is associated with concentrations of mesna and acrolein have never been
delayed peak urinary concentrations (3 hours after measured in the setting of renal insufficiency, and
dosing compared with 1 hour following IV administra- oliguria could lead to prolonged exposure of the bladder
tion [67]) and prolonged excretion. When oral doses of to whatever amount of acrolein arrives there; thus, one
mesna are doubled, peak urinary thiol concentrations cannot regard use of mesna in patients with renal
are unchanged but are maintained for longer periods of insufficiency as having been addressed conclusively.
time (67).
The recommended dosing of IV mesna is a total
Efficacy of mesna for the prevention of hemorrhagic
dose equal to 20% (weight/weight) of the total cyclo-
cystitis
phosphamide dose, in the form of 3 equal doses of
mesna, with the first dose administered 15–30 minutes Data for the use of mesna to prevent hemor-
prior to cyclophosphamide and the others administered rhagic cystitis caused by cyclophosphamide in the treat-
4 hours and 8 hours following cyclophosphamide (67). ment of rheumatic diseases are not conclusive. This
16 MONACH ET AL

topic is best approached in a broader context that receiving daily oral cyclophosphamide, and interpreta-
includes strategies for hydration, data on the use of tion and comparison of these studies are the subject of
mesna in patients with cancer, and insights from animal debate (34,75), a detailed review of the controversy is
models. These data were obtained with the use of warranted.
periodic high-dose IV cyclophosphamide or ifosfamide, Reinhold-Keller et al (34) diagnosed all cases of
both of which are associated with high risks of cystitis. It cystitis by cystoscopy, and all cases of nonglomerular
is unclear whether these results are more applicable, in hematuria were investigated in this way; therefore, their
rheumatic diseases, to periodic IV dosing (in which the report of a 12% incidence appears accurate. Talar-
route and schedule of administration are similar but the Williams et al (33) reported nonglomerular hematuria in
baseline risk is much lower) or to daily oral dosing (in 73 of 145 patients (50%). Cystoscopy was performed in
which the route and schedule are very different, but the only 60 of these 73 patients, with confirmation of cystitis
baseline risk is similarly high). in 42 (i.e., 70% of those undergoing cystoscopy), giving
Hydration. Hydration with IV fluids immediately a minimum prevalence of cystitis in the cohort of 29%.
prior to and for several hours after IV administration of Both “50%” and “29%” have been used to describe the
cyclophosphamide, as well as oral hydration for 72 hours incidence of cystitis in this study (34,75). However, the
following the cyclophosphamide dose, is typically recom- most appropriate estimate is probably neither of these
mended to aid in the prevention of hemorrhagic cystitis. figures but instead is the percent of confirmed cases (42
When oral cyclophosphamide is given, patients are of 60 [70%]) multiplied by the percent with hematuria
instructed to drink fluids at the time of dosing and for (50%), resulting in an incidence of 35% (Table 1).
several hours thereafter and to take cyclophosphamide Stillwell et al (32) diagnosed cystitis either on the basis
in the morning to limit retention of metabolites in the of gross hematuria (11 patients) or by cystoscopy per-
urine overnight. It should be noted that since the time formed for the followup of new microscopic hematuria.
when bladder toxicities of oxazophosphorines were first However, only a minority of patients with microscopic
recognized, the effectiveness of hydration per se has hematuria were investigated by cystoscopy (20 of 52),
never been formally evaluated, having been regarded as and because the percent of patients in whom cystitis was
the standard of care against which all other strategies for confirmed by cystoscopy was 65% (which notably is
bladder protection are to be compared. similar to the incidence of 70% reported by Talar-
Mesna in animal models of cystitis. Studies in Williams), one could reasonably extrapolate that the
rodents support the effectiveness of mesna in preventing true incidence of cystitis in the study by Stillwell et al is
acute bladder toxicity from cyclophosphamide or acro- 41% (11 ⫹ [0.65 ⫻ 52] ⫽ 45 of 111 patients). Despite the
lein (21,70–74). These studies typically involved the brief difficulty of directly comparing these 3 studies, it is
use of high doses of cyclophosphamide and thus provide worth noting that the study showing the lowest incidence
better support for the use of mesna in patients with of cystitis (in the setting of routine mesna therapy) used
malignancies than in patients with rheumatic disease. the most thorough screening strategy (34), which should
Mesna and cystitis in rheumatic diseases. The have biased toward a higher rather than lower incidence.
prospect of preventing cystitis and/or bladder cancer by Although routine treatment with mesna has re-
coadministering mesna to patients receiving daily oral cently been incorporated into some protocols that have
cyclophosphamide has never been addressed in a sys- used periodic IV dosing of cyclophosphamide (28), data
tematic manner. Among the studies of patients with on the efficacy of mesna in preventing cystitis are
WG, the incidence of cystitis may have been lower in the lacking, because the baseline risk of cystitis with such
single cohort that routinely received oral mesna (12%) regimens appears to be quite low (Table 4).
(34) than in the 2 others that did not receive oral mesna Mesna and cystitis with high-dose cyclo-
(35–41%) (30,31) (Table 1). However, the possibility of phosphamide or ifosfamide. Mesna as prophylactic ther-
important differences in the dosing schedules, total apy against bladder toxicity has been tested only in a
cumulative doses, and clinical characteristics, particu- randomized, controlled manner among patients receiv-
larly renal dysfunction, makes a direct comparison prob- ing high-dose IV cyclophosphamide or ifosfamide as
lematic. Similarly, differing approaches to screening for part of bone marrow transplantation (BMT) protocols
and the diagnosis of cystitis among these 3 studies or as chemotherapy for solid malignancies. Mesna treat-
introduce another source of uncertainty. However, be- ment (with maintenance hydration of 1.5–3 liters/day)
cause these studies provide the only data relevant to the has been compared with IV hyperhydration (either 6
question of whether mesna prevents cystitis in patients liters/day with a diuretic or 3 liters/m2/day), with similar
CYCLOPHOSPHAMIDE AND MESNA 17

but incomplete effectiveness in preventing short-term perhaps the most comprehensive (or at least the best-
bladder toxicity caused by high-dose IV cyclo- documented) screening strategy (34). However, with
phosphamide in BMT (76,77). By pooling the results such small numbers of events, a comparison among
from these 2 studies, hematuria of any degree was studies or pooling of these data is even more problem-
observed in 41 (57%) of 72 patients receiving mesna and atic than it is for comparing the incidences of cystitis.
in 44 (67%) of 66 patients receiving hyperhydration; There are no published data regarding the effect of
macroscopic hematuria was observed in 17 (24%) of 72 mesna on the rate of secondary bladder cancer in
patients receiving mesna and in 17 (26%) of 66 patients patients treated for malignancies with high-dose IV
receiving hyperhydration. In patients also receiving hy- cyclophosphamide or ifosfamide; the prospect and need
dration during high-dose IV cyclophosphamide treat- for such studies have been noted (92).
ment, the addition of mesna was more effective than the A single study in rodents supports the effective-
addition of continuous bladder irrigation in preventing ness of mesna in preventing bladder cancer induced by
acute bladder toxicity (78). prolonged exposure to cyclophosphamide. Cyclophosph-
Mesna is routinely used (and is FDA-approved in amide administered orally at a dosage of 2.5 mg/kg/day
both IV and oral forms) as prophylaxis against cystitis in for 5 days per week produced bladder tumors in 24 of 80
ifosfamide-containing regimens (16), based on 4 small treated rats (30%); the incidence of tumors was reduced
controlled trials of IV mesna (79–82), 3 subsequent to 22% (8 of 36) in rats given low-dose mesna (5
trials showing equivalent rates of cystitis comparing IV mg/kg/day) and to 7% (3 of 41) in rats given high-dose
with oral mesna (15,83), and additional uncontrolled but mesna (15 mg/kg/day) (93).
often larger series (84–90) that made comparisons with
the high rate of hemorrhagic cystitis (⬃50%) noted in Evidence-based consensus guidelines on the use of
early studies of ifosfamide (11). Even with mesna pro- mesna
phylaxis, however, the rates of acute urotoxicity ranged
from 1% to 10% in these studies (16). In addition, Evidence-based consensus guidelines on the use
among 15 patients without clinical evidence of bladder of mesna have not addressed its value in patients with
toxicity 24 hours after receiving ifosfamide and mesna, rheumatic diseases. Given the low reported incidence of
the bladder mucosa was macroscopically (10 of 15 hemorrhagic cystitis with standard doses of IV cyclo-
patients) and microscopically (100%) abnormal, with phosphamide in patients with cancer, the American
mucosal alterations including edema, exocytosis, and Society of Clinical Oncology does not recommend the
hemorrhage (91). routine use of mesna for patients receiving cyclo-
Summary. Direct evidence for the effectiveness phosphamide and states that mesna should be used only
of mesna in preventing cystitis comes only from its use in conjunction with the administration of high-dose
with ifosfamide in patients with cancer and on data from cyclophosphamide, typically defined as 50 mg/kg or 2
animal models, both of which are of uncertain relevance gm/m2 (16,94).
to the use of cyclophosphamide in patients with rheu- In the rheumatology literature, guidelines for the
matic diseases. The data from rheumatology series are use of mesna vary and are not detailed. Guidelines from
consistent with a protective effect but are inadequate to the British Society for Rheumatology for the manage-
draw a firm conclusion. ment of antineutrophil cytoplasmic antibody–associated
vasculitis have recommended that mesna be “considered
for patients on IV cyclophosphamide therapy” (95).
Paucity of data on the efficacy of mesna for the
Similarly, guidelines on vasculitis from the European
prevention of bladder cancer in patients treated with
League Against Rheumatism recommend that “patients
cyclophosphamide or ifosfamide
receiving pulse cyclophosphamide should also be given
No study has compared the incidences of bladder oral or IV mesna” and add that “mesna may also be
cancer resulting from the use of cyclophosphamide with beneficial in patients receiving continuous oral cyclo-
or without mesna. Among studies describing long-term phosphamide” (5). In contrast, consensus guidelines for
followup after treatment with daily oral cyclo- management of lupus have made no mention of mesna
phosphamide (10,32–34,38,40–42), the only study in (30,96). Rheumatology textbooks and other resources
which mesna was routinely used revealed a low rate of regularly recommend use of mesna for patients receiving
bladder cancer (1 of 142 treated patients) despite one of IV cyclophosphamide, with little or no comment on the
the longest average followup times (see Table 2) and use of mesna with oral cyclophosphamide (12–14).
18 MONACH ET AL

Overall, the paucity of evidence supporting the ing regimen for mesna, particularly for daily
use of mesna is striking in light of recommendations for dosing;
its use and the common adoption of such guidelines in c. the ability to tolerate hydration during either
rheumatology practice. However, there is also no evi- daily oral or pulse IV therapy with cyclo-
dence against the effectiveness of mesna and little phosphamide;
prospect of the issue being definitively addressed in a d. the total cumulative dose of cyclophosphamide
trial. With uncertain and unquantifiable risks and ben- for patients requiring a repeat course.
efits, patient and physician preference can and should 4. Upon starting a second course of cyclo-
play important roles, and thus, recommendations to phosphamide (i.e., more than 4–6 months of total
“consider” use of mesna are fully consistent with the treatment), particularly with oral dosing, consider
evidence. However, the conclusions of this review di- recommending mesna.
verge from most recommendations in one important
Cyclophosphamide remains an important treat-
respect: on the basis of the much clearer risks of bladder
ment for patients with various rheumatic diseases. Al-
toxicity with daily oral cyclophosphamide, the inclina-
though additional study of the usefulness of mesna in
tion to use mesna should be much stronger with daily
patients with rheumatic diseases would be welcomed,
oral dosing compared with periodic IV dosing.
such research is unlikely to occur, especially given the
trend toward developing non–cyclophosphamide-based
Summary and recommendations therapies. Given the uncertainties outlined in this re-
The conclusions of this literature review are as view, decisions regarding the use of mesna will need to
follows: 1) daily oral cyclophosphamide is associated be made on an individual basis, taking into consideration
the varying attitudes of both physicians and patients
with an increased risk of both hemorrhagic cystitis and
toward risk reduction.
bladder cancer, in a dose-dependent and/or duration-
dependent manner; 2) hemorrhagic cystitis that occurs
during cyclophosphamide treatment is associated with AUTHOR CONTRIBUTIONS
an increased risk of bladder cancer years later; 3) IV All authors were involved in drafting the article or revising it
critically for important intellectual content, and all authors approved
cyclophosphamide therapy, as prescribed for rheumatic the final version to be published.
diseases, carries a low risk of cystitis and probably also of
bladder cancer; 4) evidence for the effectiveness of
mesna in preventing cystitis is based largely on its use REFERENCES
with ifosfamide in patients with cancer and on data from 1. Austin HA III, Klippel JH, Balow JE, le Riche NG, Steinberg AD,
animal models, both of which are of uncertain relevance Plotz PH, et al. Therapy of lupus nephritis: controlled trial of
prednisone and cytotoxic drugs. N Engl J Med 1986;314:614–9.
to the use of cyclophosphamide in patients with rheu- 2. Fauci AS, Katz P, Haynes BF, Wolff SM. Cyclophosphamide
matic diseases; 5) there is no direct evidence for the therapy of severe systemic necrotizing vasculitis. N Engl J Med
effectiveness of mesna in preventing bladder cancer in 1979;301:235–8.
3. Gourley MF, Austin HA III, Scott D, Yarboro CH, Vaughan EM,
humans. Muir J, et al. Methylprednisolone and cyclophosphamide, alone or
On the basis of the current evidence, the follow- in combination, in patients with lupus nephritis: a randomized,
ing recommendations for use of mesna in the rheuma- controlled trial. Ann Intern Med 1996;125:549–57.
4. Tashkin DP, Elashoff R, Clements PJ, Goldin J, Roth MD, Furst
tology setting are proposed: DE, et al. Cyclophosphamide versus placebo in scleroderma lung
disease. N Engl J Med 2006;354:2655–66.
1. Explain to patients that the evidence for the benefit 5. Mukhtyar C, Guillevin L, Cid MC, Dasgupta B, de Groot K, Gross
of mesna in rheumatologic diseases is not strong. W, et al. EULAR recommendations for the management of
2. Upon starting a first course of oral or IV cyclo- primary small and medium vessel vasculitis. Ann Rheum Dis
2009;68:310–7.
phosphamide, discuss the issues with the patient; if 6. Ginzler EM, Dooley MA, Aranow C, Kim MY, Buyon J, Merrill
the patient expresses no strong preference, do not JT, et al. Mycophenolate mofetil or intravenous cyclophosphamide
use mesna. for lupus nephritis. N Engl J Med 2005;353:2219–28.
7. Wegener’s Granulomatosis Etanercept Trial (WGET) Research
3. Additional factors to consider when deciding on Group. Etanercept plus standard therapy for Wegener’s granulo-
the use of mesna are as follows: matosis. N Engl J Med 2005;352:351–61.
a. the expense of mesna, especially oral tablet 8. Hoyles RK, Ellis RW, Wellsbury J, Lees B, Newlands P, Goh NS,
et al. A multicenter, prospective, randomized, double-blind, pla-
form; cebo-controlled trial of corticosteroids and intravenous cyclo-
b. the inconvenience of the relatively complex dos- phosphamide followed by oral azathioprine for the treatment of
CYCLOPHOSPHAMIDE AND MESNA 19

pulmonary fibrosis in scleroderma. Arthritis Rheum 2006;54: vasculitis associated with antineutrophil cytoplasmic autoantibod-
3962–70. ies. N Engl J Med 2003;349:36–44.
9. Jayne DR, Gaskin G, Rasmussen N, Abramowicz D, Ferrario F, 28. De Groot K, Jayne D, Tesar V, Savage C. Randomised controlled
Guillevin L, et al. Randomized trial of plasma exchange or trial of daily oral versus pulse cyclophosphamide for induction of
high-dosage methylprednisolone as adjunctive therapy for severe remission in ANCA-associated systemic vasculitis. Kidney Blood
renal vasculitis. J Am Soc Nephrol 2007;18:2180–8. Press Res 2005;28:195.
10. Pedersen-Bjergaard J, Ersboll J, Hansen VL, Sorensen BL, Christ- 29. Illei GG, Austin HA, Crane M, Collins L, Gourley MF, Yarboro
offersen K, Hou-Jensen K, et al. Carcinoma of the urinary bladder CH, et al. Combination therapy with pulse cyclophosphamide plus
after treatment with cyclophosphamide for non-Hodgkin’s lym- pulse methylprednisolone improves long-term renal outcome with-
phoma. N Engl J Med 1988;318:1028–32. out adding toxicity in patients with lupus nephritis. Ann Intern
11. Brade WP, Herdrich K, Varini M. Ifosfamide: pharmacology, Med 2001;135:248–57.
safety and therapeutic potential. Cancer Treat Rev 1985;12:1–47. 30. Bertsias G, Ioannidis JP, Boletis J, Bombardieri S, Cervera R,
12. Stone JH. General principles of the use of cyclophosphamide in Dostal C, et al. EULAR recommendations for the management of
rheumatic and renal diseases. UpToDate online 2008; version systemic lupus erythematosus. Report of a Task Force of the
16.3. Online at http://uptodateonline.com. EULAR Standing Committee for International Clinical Studies
13. Boumpas DT, Illei GG, Balow JE. Treatment: renal involvement. Including Therapeutics. Ann Rheum Dis 2008;67:195–205.
In: Hochberg MC, Silman AJ, Smolen JS, Weinblatt ME, Weis- 31. Silver RM, Warrick JH, Kinsella MB, Staudt LS, Baumann MH,
man MH, editors. Rheumatology. 3rd ed. Edinburgh: Mosby; Strange C. Cyclophosphamide and low-dose prednisone therapy in
2003. p. 1405–17. patients with systemic sclerosis (scleroderma) with interstitial lung
14. Wofsy D. Therapy of systemic lupus erythematosus. In: Koopman disease. J Rheumatol 1993;20:838–44.
WJ, Moreland LW, editors. Arthritis and allied conditions: a 32. Stillwell TJ, Benson RC Jr, DeRemee RA, McDonald TJ, Weiland
textbook of rheumatology. 15th ed. Philadelphia: Lippincott Wil- LH. Cyclophosphamide-induced bladder toxicity in Wegener’s
liams & Wilkins; 2005. p. 1561–74. granulomatosis. Arthritis Rheum 1988;31:465–70.
15. Cohen MH, Dagher R, Griebel DJ, Ibrahim A, Martin A, Scher 33. Talar-Williams C, Hijazi YM, Walther MM, Linehan WM, Halla-
NS, et al. U.S. Food and Drug Administration drug approval han CW, Lubensky I, et al. Cyclophosphamide-induced cystitis and
summaries: imatinib mesylate, mesna tablets, and zoledronic acid. bladder cancer in patients with Wegener granulomatosis. Ann
Oncologist 2002;7:393–400. Intern Med 1996;124:477–84.
16. Hensley ML, Schuchter LM, Lindley C, Meropol NJ, Cohen GI, 34. Reinhold-Keller E, Beuge N, Latza U, de Groot K, Rudert H,
Broder G, et al. American Society of Clinical Oncology clinical Nolle B, et al. An interdisciplinary approach to the care of patients
practice guidelines for the use of chemotherapy and radiotherapy with Wegener’s granulomatosis: long-term outcome in 155 pa-
protectants. J Clin Oncol 1999;17:3333–55. tients. Arthritis Rheum 2000;43:1021–32.
17. Richmond R, McMillan TW, Luqmani RA. Optimisation of 35. Plotz PH, Klippel JH, Decker JL, Grauman D, Wolff B, Brown
cyclophosphamide therapy in systemic vasculitis. Clin Pharmaco- BC, et al. Bladder complications in patients receiving cyclo-
kinet 1998;34:79–90. phosphamide for systemic lupus erythematosus or rheumatoid
18. Cyclophosphamide. In: Dorr RT, Von Hoff DD, editors. Cancer arthritis. Ann Intern Med 1979;91:221–3.
chemotherapy handbook. 2nd ed. Norwalk, Connecticut: Appleton 36. Carette S, Klippel JH, Decker JL, Austin HA, Plotz PH, Steinberg
& Lange; 1994. p. 319–32. AD, et al. Controlled studies of oral immunosuppressive drugs in
19. Brock N. Comparative pharmacologic study in vitro and in vivo lupus nephritis: a long-term follow-up. Ann Intern Med 1983;99:
with cyclophosphamide (NSC-26271), cyclophosphamide metabo- 1–8.
lites, and plain nitrogen mustard compounds. Cancer Treat Rep 37. Aptekar RG, Atkinson JP, Decker JL, Wolff SM, Chu EW.
1976;60:301–8. Bladder toxicity with chronic oral cyclophosphamide therapy in
20. Brock N, Stekar J, Pohl J, Niemeyer U, Scheffler G. Acrolein, the nonmalignant disease. Arthritis Rheum 1973;16:461–7.
causative factor of urotoxic side-effects of cyclophosphamide, 38. Radis CD, Kahl LE, Baker GL, Wasko MC, Cash JM, Gallatin A,
ifosfamide, trofosfamide and sufosfamide. Arzneimittelforschung et al. Effects of cyclophosphamide on the development of malig-
1979;29:659–61. nancy and on long-term survival of patients with rheumatoid
21. Batista CK, Brito GA, Souza ML, Leitao BT, Cunha FQ, Ribeiro arthritis: a 20-year followup study. Arthritis Rheum 1995;38:
RA. A model of hemorrhagic cystitis induced with acrolein in 1120–7.
mice. Braz J Med Biol Res 2006;39:1475–81. 39. Townes AS, Sowa JM, Shulman LE. Controlled trial of cyclo-
22. Sigal SH, Tomaszewski JE, Brooks JJ, Wein A, LiVolsi VA. phosphamide in rheumatoid arthritis. Arthritis Rheum 1976;19:
Carcinosarcoma of bladder following long-term cyclophosphamide 563–73.
therapy. Arch Pathol Lab Med 1991;115:1049–51. 40. Westman KW, Bygren PG, Olsson H, Ranstam J, Wieslander J.
23. Stein JP, Skinner EC, Boyd SD, Skinner DG. Squamous cell Relapse rate, renal survival, and cancer morbidity in patients with
carcinoma of the bladder associated with cyclophosphamide ther- Wegener’s granulomatosis or microscopic polyangiitis with renal
apy for Wegener’s granulomatosis: a report of 2 cases. J Urol involvement. J Am Soc Nephrol 1998;9:842–52.
1993;149:588–9. 41. Faurschou M, Sorensen IJ, Mellemkjaer L, Loft AG, Thomsen BS,
24. Chasko SB, Keuhnelian JG, Gutowski WT III, Gray GF. Spindle Tvede N, et al. Malignancies in Wegener’s granulomatosis: inci-
cell cancer of bladder during cyclophosphamide therapy for We- dence and relation to cyclophosphamide therapy in a cohort of 293
gener’s granulomatosis. Am J Surg Pathol 1980;4:191–6. patients. J Rheumatol 2008;35:100–5.
25. Struck RF, Alberts DS, Horne K, Phillips JG, Peng YM, Roe DJ. 42. Knight A, Askling J, Granath F, Sparen P, Ekbom A. Urinary
Plasma pharmacokinetics of cyclophosphamide and its cytotoxic bladder cancer in Wegener’s granulomatosis: risks and relation to
metabolites after intravenous versus oral administration in a cyclophosphamide. Ann Rheum Dis 2004;63:1307–11.
randomized, crossover trial. Cancer Res 1987;47:2723–6. 43. Knight A, Askling J, Ekbom A. Cancer incidence in a population-
26. Cytoxan prescribing information. Princeton (NJ): Bristol-Myers based cohort of patients with Wegener’s granulomatosis. Int J
Squibb; 2005. Cancer 2002;100:82–5.
27. Jayne D, Rasmussen N, Andrassy K, Bacon P, Tervaert JW, 44. Travis LB, Curtis RE, Glimelius B, Holowaty EJ, Van Leeuwen
Dadoniene J, et al. A randomized trial of maintenance therapy for FE, Lynch CF, et al. Bladder and kidney cancer following cyclo-
20 MONACH ET AL

phosphamide therapy for non-Hodgkin’s lymphoma. J Natl Cancer patients under long-term review. Rheumatology (Oxford) 2000;39:
Inst 1995;87:524–30. 1147–52.
45. Cancer statistics, cancer fact sheets, urinary bladder. Surveillance, 63. Abu-Shakra M, Gladman DD, Urowitz MB. Malignancy in sys-
Epidemiology and End Results (SEER) online 2008. http://seer. temic lupus erythematosus. Arthritis Rheum 1996;39:1050–4.
cancer.gov/statfacts/html/urinb. 64. Sweeney DM, Manzi S, Janosky J, Selvaggi KJ, Ferri W, Medsger
46. Zeegers MP, Kellen E, Buntinx F, van den Brandt PA. The TA Jr, et al. Risk of malignancy in women with systemic lupus
association between smoking, beverage consumption, diet and erythematosus. J Rheumatol 1995;22:1478–82.
bladder cancer: a systematic literature review. World J Urol 65. Nived O, Bengtsson A, Jonsen A, Sturfelt G, Olsson H. Malignan-
2004;21:392–401. cies during follow-up in an epidemiologically defined systemic
47. De Groot K, Adu D, Savage CO. The value of pulse cyclo- lupus erythematosus inception cohort in southern Sweden. Lupus
phosphamide in ANCA-associated vasculitis: meta-analysis and 2001;10:500–4.
critical review. Nephrol Dial Transplant 2001;16:2018–27. 66. Dechant KL, Brogden RN, Pilkington T, Faulds D. Ifosfamide/
48. Hoffman GS, Leavitt RY, Fleisher TA, Minor JR, Fauci AS. mesna: a review of its antineoplastic activity, pharmacokinetic
Treatment of Wegener’s granulomatosis with intermittent high- properties and therapeutic efficacy in cancer. Drugs 1991;42:
dose intravenous cyclophosphamide. Am J Med 1990;89:403–10. 428–67.
49. Goransson LG, Brodin C, Ogreid P, Janssen EA, Romundstad PR, 67. Mesna. In: Dorr RT, Von Hoff DD, editors. Cancer chemotherapy
Vatten L, et al. Intravenous cyclophosphamide in patients with handbook. 2nd ed. Norwalk (CT): Appleton & Lange; 1994. p.
chronic systemic inflammatory diseases: morbidity and mortality. 685–91.
Scand J Rheumatol 2008;37:130–4. 68. Dorr RT. Chemoprotectants for cancer chemotherapy. Semin
50. Martin F, Lauwerys B, Lefebvre C, Devogelaer JP, Houssiau FA. Oncol 1991;18(1 Suppl 2):48–58.
Side-effects of intravenous cyclophosphamide pulse therapy. Lu- 69. Reinhold-Keller E, Mohr J, Christophers E, Nordmann K, Gross
pus 1997;6:254–7. WL. Mesna side effects which imitate vasculitis. Clin Investig
51. Koldingsnes W, Gran JT, Omdal R, Husby G. Wegener’s granu- 1992;70:698–704.
lomatosis: long-term follow-up of patients treated with pulse 70. Freedman A, Ehrlich RM, Ljung BM. Prevention of cyclo-
cyclophosphamide. Br J Rheumatol 1998;37:659–64. phosphamide cystitis with 2-mercaptoethane sodium sulfonate: a
52. Haubitz M, Schellong S, Gobel U, Schurek HJ, Schaumann D, histologic study. J Urol 1984;132:580–2.
Koch KM, et al. Intravenous pulse administration of cyclo- 71. Themann H, Oberdorf E, Brock N, Pohl J. Electron microscopic
phosphamide versus daily oral treatment in patients with antineu- investigations of the cyclophosphamide-induced lesions of the
trophil cytoplasmic antibody–associated vasculitis and renal in- urinary bladder of the rat and their prevention by mesna. Urol Int
volvement: a prospective, randomized study. Arthritis Rheum 1987;42:37–43.
1998;41:1835–44. 72. Habs M, Hebebrand J, Schmahl D. Influence of sulfur-containing
53. Guillevin L, Cordier JF, Lhote F, Cohen P, Jarrousse B, Royer I, compounds on the acute toxicity of cyclophosphamide in male
et al. A prospective, multicenter, randomized trial comparing Sprague-Dawley rats. Arzneimittelforschung 1984;34:792–3.
steroids and pulse cyclophosphamide versus steroids and oral 73. Kedar A, Simpson CL, Williams P, Moore R, Tritsch G, Murphy
cyclophosphamide in the treatment of generalized Wegener’s GP. The prevention of cyclophosphamide-induced bladder swell-
granulomatosis. Arthritis Rheum 1997;40:2187–98. ing in the rat by I.V. administration of sodium-2-mercaptoethane
54. Adu D, Pall A, Luqmani RA, Richards NT, Howie AJ, Emery P, sulfonate. Res Commun Chem Pathol Pharmacol 1980;29:339–48.
et al. Controlled trial of pulse versus continuous prednisolone and 74. Cavalletti E, Tofanetti O, Zunino F. Comparison of reduced
cyclophosphamide in the treatment of systemic vasculitis. QJM glutathione with 2-mercaptoethane sulfonate to prevent cyclo-
1997;90:401–9. phosphamide-induced urotoxicity. Cancer Lett 1986;32:1–6.
55. Martin-Suarez I, D’Cruz D, Mansoor M, Fernandes AP, Kha- 75. Sneller MC. Cystitis, bladder cancer, and myelodysplasia in pa-
mashta MA, Hughes GR. Immunosuppressive treatment in severe tients with Wegener’s granulomatosis: comment on the article by
connective tissue diseases: effects of low dose intravenous cyclo- Reinhold-Keller et al [letter]. Arthritis Rheum 2000;43:2853–5.
phosphamide. Ann Rheum Dis 1997;56:481–7. 76. Hows JM, Mehta A, Ward L, Woods K, Perez R, Gordon MY, et
56. Ortiz A, Gonzalez-Parra E, Alvarez-Costa G, Egido J. Bladder al. Comparison of mesna with forced diuresis to prevent cyclo-
cancer after cyclophosphamide therapy for lupus nephritis. phosphamide induced haemorrhagic cystitis in marrow transplan-
tation: a prospective randomised study. Br J Cancer 1984;50:
Nephron 1992;60:378–9.
753–6.
57. Chow SK, Looi LM, Loh CS, Yeap SS. Cyclophosphamide-
77. Shepherd JD, Pringle LE, Barnett MJ, Klingemann HG, Reece
induced transitional cell carcinoma of bladder in lupus nephritis.
DE, Phillips GL. Mesna versus hyperhydration for the prevention
Intern Med J 2002;32:114–6.
of cyclophosphamide-induced hemorrhagic cystitis in bone mar-
58. Alivizatos G, Dimopoulou I, Mitropoulos D, Dimopoulos AM, row transplantation. J Clin Oncol 1991;9:2016–20.
Koufakis I, Lykourinas M. Bladder cancer in a young girl with 78. Vose JM, Reed EC, Pippert GC, Anderson JR, Bierman PJ,
systemic lupus erythematosus treated with cyclophosphamide. Kessinger A, et al. Mesna compared with continuous bladder
Acta Urol Belg 1991;59:133–7. irrigation as uroprotection during high-dose chemotherapy and
59. Bernatsky S, Boivin JF, Joseph L, Rajan R, Zoma A, Manzi S, et transplantation: a randomized trial. J Clin Oncol 1993;11:1306–10.
al. An international cohort study of cancer in systemic lupus 79. Sakurai M, Saijo N, Shinkai T, Eguchi K, Sasaki Y, Tamura T, et
erythematosus. Arthritis Rheum 2005;52:1481–90. al. The protective effect of 2-mercapto-ethane sulfonate
60. Tarr T, Gyorfy B, Szekanecz E, Bhattoa HP, Zeher M, Szegedi G, (MESNA) on hemorrhagic cystitis induced by high-dose ifosf-
et al. Occurrence of malignancies in Hungarian patients with amide treatment tested by a randomized crossover trial. Jpn J Clin
systemic lupus erythematosus: results from a single center. Ann N Oncol 1986;16:153–6.
Y Acad Sci 2007;1108:76–82. 80. Fukuoka M, Negoro S, Masuda N, Furuse K, Kawahara M,
61. Ragnarsson O, Grondal G, Steinsson K. Risk of malignancy in an Kodama N, et al. Placebo-controlled double-blind comparative
unselected cohort of Icelandic patients with systemic lupus ery- study on the preventive efficacy of mesna against ifosfamide-
thematosus. Lupus 2003;12:687–91. induced urinary disorders. J Cancer Res Clin Oncol 1991;117:
62. Sultan SM, Ioannou Y, Isenberg DA. Is there an association of 473–8.
malignancy with systemic lupus erythematosus? An analysis of 276 81. Bryant BM, Jarman M, Ford HT, Smith IE. Prevention of
CYCLOPHOSPHAMIDE AND MESNA 21

isophosphamide-induced urothelial toxicity with 2-mercaptoeth- Phase II trial of ifosfamide and mesna in advanced colorectal
ane sulphonate sodium (mesnum) in patients with advanced cancer. Cancer Treat Rep 1987;71:663–4.
carcinoma. Lancet 1980;2:657–9. 90. Scheef W, Soemer G. The treatment of solid malignant tumours
82. Scheef W, Klein HO, Brock N, Burkert H, Gunther U, Hoefer- with holoxan and uromitexan. In: Burkert N, Nagel GA, editors.
Janker H, et al. Controlled clinical studies with an antidote against Contributions to oncology. Basel (Switzerland): Karger; 1980. p.
the urotoxicity of oxazaphosphorines: preliminary results. Cancer 21–4.
Treat Rep 1979;63:501–5. 91. Lima MV, Ferreira FV, Macedo FY, de Castro Brito GA, Ribeiro
83. Mace JR, Keohan ML, Bernardy H, Junge K, Niebch G, Romeis RA. Histological changes in bladders of patients submitted to
P, et al. Crossover randomized comparison of intravenous versus ifosfamide chemotherapy even with mesna prophylaxis. Cancer
Chemother Pharmacol 2007;59:643–50.
intravenous/oral mesna in soft tissue sarcoma treated with high-
92. Pratt CB. Current studies of ifosfamide for pediatric solid tumors
dose ifosfamide. Clin Cancer Res 2003;9(16 Pt 1):5829–34.
and leukemia in the United States. Semin Oncol 1992;19(6 Suppl
84. Scheulen ME, Niederle N, Bremer K, Schutte J, Seeber S. Efficacy
12):43–9.
of ifosfamide in refractory malignant diseases and uroprotection
93. Habs MR, Schmahl D. Prevention of urinary bladder tumors in
by mesna: results of a clinical phase II-study with 151 patients. cyclophosphamide-treated rats by additional medication with the
Cancer Treat Rev 1983;10 Suppl A:93–101. uroprotectors sodium 2-mercaptoethane sulfonate (mesna) and
85. Pratt CB, Goren MP. Ifosfamide/mesna and hematuria. Cancer disodium 2,2’-dithio-bis-ethane sulfonate (dimesna). Cancer 1983;
Treat Rep 1987;71:1124–5. 51:606–9.
86. Antman KH, Montella D, Rosenbaum C, Schwen M. Phase II trial 94. Schuchter LM, Hensley ML, Meropol NJ, Winer EP. 2002 update
of ifosfamide with mesna in previously treated metastatic sarcoma. of recommendations for the use of chemotherapy and radiother-
Cancer Treat Rep 1985;69:499–504. apy protectants: clinical practice guidelines of the American
87. Niederle N, Scheulen ME, Cremer M, Schutte J, Schmidt CG, Society of Clinical Oncology. J Clin Oncol 2002;20:2895–903.
Seeber S. Ifosfamide in combination chemotherapy for sarcomas 95. Lapraik C, Watts R, Bacon P, Carruthers D, Chakravarty K,
and testicular carcinomas. Cancer Treat Rev 1983;10 Suppl D’Cruz D, et al. BSR and BHPR guidelines for the management
A:129–35. of adults with ANCA associated vasculitis. Rheumatology (Ox-
88. Stuart-Harris RC, Harper PG, Parsons CA, Kaye SB, Mooney CA, ford) 2007;46:1615–6.
Gowing NF, et al. High-dose alkylation therapy using ifosfamide 96. American College of Rheumatology Ad Hoc Committee on
infusion with mesna in the treatment of adult advanced soft-tissue Systemic Lupus Erythematosus Guidelines. Guidelines for referral
sarcoma. Cancer Chemother Pharmacol 1983;11:69–72. and management of systemic lupus erythematosus in adults.
89. Kemeny N, Reichman B, Dougherty J, Lipperman R, Cheng E. Arthritis Rheum 1999;42:1785–96.

You might also like