+ All Categories
Home > Documents > Phenotypically Deficient Urinary Elimination of ......accomplished using combined thin-layer...

Phenotypically Deficient Urinary Elimination of ......accomplished using combined thin-layer...

Date post: 01-Feb-2021
Category:
Upload: others
View: 1 times
Download: 0 times
Share this document with a friend
6
[CANCER RESEARCH 48, 5167-5171, September 15. 1988] Phenotypically Deficient Urinary Elimination of Carboxyphosphamide after Cyclophosphamide Administration to Cancer Patients1 Al-Hakam F. A. Hadidi,2 Carmel E. A. Coulter, and Jeffrey R. Idle3 Department of Pharmacology and Toxicology, St. Mary's Hospital Medical School [A. F. A. H., /. R. I], and Department of Radiotherapy, St. Mary's Hospital [C. E. A. C.I, London, United Kingdom ABSTRACT The 0-24-h urinary metabolic profile of cyclophosphamide was inves tigated in a series of 14 patients with various malignancies receiving combination chemotherapy including i.v. cyclophosphamide. This was accomplished using combined thin-layer chromatography-photography- densitometry, which can quantitate cyclophosphamide and its four prin cipal urinary metabolites (4-ketocyclophosphamide, nor-nitrogen mus tard, Carboxyphosphamide, and phosphoramide mustard). Recovery of drug-related metabolites was36.5 ±17.8% (SD) dose, the most abundant metabolites being phosphoramide mustard (18.5 ±16.1% dose) and unchanged cyclophosphamide (12.7 ±9.3% dose). The most variable metabolite was Carboxyphosphamide, with five patients excreting 0.3% dose or less. These patients were termed low carboxylators (LC) and could be distinguished from high carboxylators (HC) by a carboxylation index (relative percentage as Carboxyphosphamide multiplied by 10). Mean carboxylation indices for the LC and HC phenotypes were 3.4 ± 2.6 and 151 ±115, respectively. There were no associations between patient age, sex, body weight, tumor type, or concomitant drug therapy and carboxylation phenotype. Neither 4-ketocyclophosphamide nor nor- nitrogen mustard excretion differed between LC and HC phenotypes; however, HC patients had a greater excretion of cyclophosphamide (46.4 ±15.5 relative percentage) than LC patients (19.4 ±12.6%). The DNA cross-linking cytotoxic metabolite phosphoramide mustard was elevated more than 2-fold in the LC (76.5 ±13.9%) compared with the HC (33.0 ±12.2%) phenotype. It is concluded that these data represent the first evidence of a defect in cyclophosphamide metabolism, and it is proposed that this arises from a hitherto unrecognized aldehyde dehydrogenase genotype. INTRODUCTION Metabolic transformation is of paramount importance for the cytotoxic antineoplastic drug cyclophosphamide because it exerts both its beneficial DNA-alkylating effect and its un wanted clinical toxicity via metabolites. Variation between pa tients in the extent of this metabolic activation would not only be of interest but would also play a significant part in determin ing the outcome of therapy with cyclophosphamide. Neverthe less, it is the qualitative pattern of metabolism rather than detailed quantitative studies which have preoccupied investiga tors to date, primarii) as an attempt to understand its mode of action and favorable therapeutic index. As a consequence, the principal metabolic pathways under taken by cyclophosphamide are now well established (Fig. 1). Cyclophosphamide is first converted to 4-hydroxycyclophos- phamide (1-3) by hepatic cytochrome P-450 monooxygenases (4-6). Tautomerization of 4-hydroxycyclophosphamide yields the ring-opened aldehyde metabolite aldophosphamide (7-10) Received 2/10/88; revised 6/15/88; accepted 6/21/88. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. 1Supported by grants from the Cancer Research Campaign (UK) and The Wellcome Trust. 2 Supported by The British Council and Yarmouk University, Jordan. Present address: Faculty of Medicine, Jordan University of Science and Technology, Erbid, Jordan. 3 Wellcome Trust Senior Lecturer. To whom requests for reprints should be addressed. which acts as the branching point for either detoxication to Carboxyphosphamide (2, 11, 12) or further activation to the cytotoxic species phosphoramide mustard (13, 14), which can itself be further cleaved to bis(2-chloroethyl)amine (nor-nitro gen mustard) (7, 15, 16). Minor metabolites include 4-oxocy- clophosphamide (4-ketocyclophosphamide) (2, 12,14) together with the 4-hydroxycyclophosphamide dehydration product im- inocyclophosphamide (not shown in Fig. 1) (17). Finally and importantly, the production of phosphoramide mustard from aldophosphamide yields an equimolar quantity of acrolein (13, 14, 18) thought to be responsible for the urotoxicity of cyclo phosphamide (19-22). Considerable evidence has not been ac cumulated which implicates phosphoramide mustard rather than 4-hydroxycyclophosphamide, aldophosphamide, or nor- nitrogen mustard as the ultimate alkylating and DNA cross- linking metabolite (23). Although cyclophosphamide was orig inally synthesized (24) as a prodrug for nor-nitrogen mustard release (25) within the tumor, nor-nitrogen mustard is now known only to cause DNA-protein cross-links (26). Clearly, interindividual differences in the balance of aldo phosphamide metabolism to either phosphoramide mustard (activation) or Carboxyphosphamide (detoxication) would be of considerable clinical importance. Until now, however, no single and simple method has been available for the quantitative determination of cyclophosphamide metabolites in body fluids. We have recently described (27) a combined TLC-PD" method which can determine cyclophosphamide, 4-Ketocyclophos- phamide, Carboxyphosphamide, phosphoramide mustard, and nor-nitrogen mustard in biological samples and have applied this in a reappraisal of the quantitative nature of cyclophos phamide metabolism in cancer patients. This paper reports the finding of 5 cancer patients, from a series of 14 who were given i.v. cyclophosphamide, with a virtual complete absence in urine of the "major" metabolite Carboxyphosphamide with a conse quent amplification of the activation pathway that yields phos phoramide mustard. MATERIALS AND METHODS Authentic metabolites of cyclophosphamide were the gift of Asta- Werke AC, Bielefeld, Federal Republic of Germany, and of Boehringer Ingelheim Limited, Bracknell, United Kingdom. Each substance was authenticated by 'H-nuclear magnetic resonance and IR spectroscopy at Asta-Werke AG and by elemental analysis in London, details of which are reported elsewhere (27). The TLC-PD method for the deter mination of cyclophosphamide metabolites has been described in detail (27). Briefly, urinary metabolites are extracted onto a non-ionic poly meric adsorban), cltiled with methanol, and separated with TLC. Vis ualization of drug-related spots is accomplished by spraying the plates with 4-(4-nitrobenzyl)pyridine, an alkyl acceptor which gives a blue chromophore on alkalinization (28). Because the blue spots deteriorate on standing, the TLC plate is photographed within 10 s of spot development and the resultant black and white print is scanned in a densitometer. The method is sensitive down to 1 /ig/ml and linear up 4 The abbreviations used are: TLC-PD, combined thin-layer chromatography- photography-densitometry; LC, low carboxylator; HC, high carboxylator; TLC, thin-layer chromatography; ALDH, aldehyde dehydrogenase. 5167 on June 17, 2021. © 1988 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from
Transcript
  • [CANCER RESEARCH 48, 5167-5171, September 15. 1988]

    Phenotypically Deficient Urinary Elimination of Carboxyphosphamide afterCyclophosphamide Administration to Cancer Patients1

    Al-Hakam F. A. Hadidi,2 Carmel E. A. Coulter, and Jeffrey R. Idle3

    Department of Pharmacology and Toxicology, St. Mary's Hospital Medical School [A. F. A. H., /. R. I], and Department of Radiotherapy, St. Mary's Hospital

    [C. E. A. C.I, London, United Kingdom

    ABSTRACT

    The 0-24-h urinary metabolic profile of cyclophosphamide was investigated in a series of 14 patients with various malignancies receivingcombination chemotherapy including i.v. cyclophosphamide. This wasaccomplished using combined thin-layer chromatography-photography-densitometry, which can quantitate cyclophosphamide and its four principal urinary metabolites (4-ketocyclophosphamide, nor-nitrogen mustard, Carboxyphosphamide, and phosphoramide mustard). Recovery ofdrug-related metabolites was 36.5 ±17.8% (SD) dose, the most abundantmetabolites being phosphoramide mustard (18.5 ±16.1% dose) andunchanged cyclophosphamide (12.7 ±9.3% dose). The most variablemetabolite was Carboxyphosphamide, with five patients excreting 0.3%dose or less. These patients were termed low carboxylators (LC) andcould be distinguished from high carboxylators (HC) by a carboxylationindex (relative percentage as Carboxyphosphamide multiplied by 10).Mean carboxylation indices for the LC and HC phenotypes were 3.4 ±2.6 and 151 ±115, respectively. There were no associations betweenpatient age, sex, body weight, tumor type, or concomitant drug therapyand carboxylation phenotype. Neither 4-ketocyclophosphamide nor nor-nitrogen mustard excretion differed between LC and HC phenotypes;however, HC patients had a greater excretion of cyclophosphamide (46.4±15.5 relative percentage) than LC patients (19.4 ±12.6%). The DNAcross-linking cytotoxic metabolite phosphoramide mustard was elevatedmore than 2-fold in the LC (76.5 ±13.9%) compared with the HC (33.0±12.2%) phenotype. It is concluded that these data represent the firstevidence of a defect in cyclophosphamide metabolism, and it is proposedthat this arises from a hitherto unrecognized aldehyde dehydrogenasegenotype.

    INTRODUCTION

    Metabolic transformation is of paramount importance forthe cytotoxic antineoplastic drug cyclophosphamide because itexerts both its beneficial DNA-alkylating effect and its unwanted clinical toxicity via metabolites. Variation between patients in the extent of this metabolic activation would not onlybe of interest but would also play a significant part in determining the outcome of therapy with cyclophosphamide. Nevertheless, it is the qualitative pattern of metabolism rather thandetailed quantitative studies which have preoccupied investigators to date, primarii) as an attempt to understand its mode ofaction and favorable therapeutic index.

    As a consequence, the principal metabolic pathways undertaken by cyclophosphamide are now well established (Fig. 1).Cyclophosphamide is first converted to 4-hydroxycyclophos-phamide (1-3) by hepatic cytochrome P-450 monooxygenases(4-6). Tautomerization of 4-hydroxycyclophosphamide yieldsthe ring-opened aldehyde metabolite aldophosphamide (7-10)

    Received 2/10/88; revised 6/15/88; accepted 6/21/88.The costs of publication of this article were defrayed in part by the payment

    of page charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

    1Supported by grants from the Cancer Research Campaign (UK) and The

    Wellcome Trust.2Supported by The British Council and Yarmouk University, Jordan. Present

    address: Faculty of Medicine, Jordan University of Science and Technology,Erbid, Jordan.

    3Wellcome Trust Senior Lecturer. To whom requests for reprints should be

    addressed.

    which acts as the branching point for either detoxication toCarboxyphosphamide (2, 11, 12) or further activation to thecytotoxic species phosphoramide mustard (13, 14), which canitself be further cleaved to bis(2-chloroethyl)amine (nor-nitrogen mustard) (7, 15, 16). Minor metabolites include 4-oxocy-clophosphamide (4-ketocyclophosphamide) (2, 12,14) togetherwith the 4-hydroxycyclophosphamide dehydration product im-inocyclophosphamide (not shown in Fig. 1) (17). Finally andimportantly, the production of phosphoramide mustard fromaldophosphamide yields an equimolar quantity of acrolein (13,14, 18) thought to be responsible for the urotoxicity of cyclophosphamide (19-22). Considerable evidence has not been accumulated which implicates phosphoramide mustard ratherthan 4-hydroxycyclophosphamide, aldophosphamide, or nor-nitrogen mustard as the ultimate alkylating and DNA cross-linking metabolite (23). Although cyclophosphamide was originally synthesized (24) as a prodrug for nor-nitrogen mustardrelease (25) within the tumor, nor-nitrogen mustard is nowknown only to cause DNA-protein cross-links (26).

    Clearly, interindividual differences in the balance of aldophosphamide metabolism to either phosphoramide mustard(activation) or Carboxyphosphamide (detoxication) would be ofconsiderable clinical importance. Until now, however, no singleand simple method has been available for the quantitativedetermination of cyclophosphamide metabolites in body fluids.We have recently described (27) a combined TLC-PD" method

    which can determine cyclophosphamide, 4-Ketocyclophos-phamide, Carboxyphosphamide, phosphoramide mustard, andnor-nitrogen mustard in biological samples and have appliedthis in a reappraisal of the quantitative nature of cyclophosphamide metabolism in cancer patients. This paper reports thefinding of 5 cancer patients, from a series of 14 who were giveni.v. cyclophosphamide, with a virtual complete absence in urineof the "major" metabolite Carboxyphosphamide with a conse

    quent amplification of the activation pathway that yields phosphoramide mustard.

    MATERIALS AND METHODS

    Authentic metabolites of cyclophosphamide were the gift of Asta-Werke AC, Bielefeld, Federal Republic of Germany, and of BoehringerIngelheim Limited, Bracknell, United Kingdom. Each substance wasauthenticated by 'H-nuclear magnetic resonance and IR spectroscopyat Asta-Werke AG and by elemental analysis in London, details ofwhich are reported elsewhere (27). The TLC-PD method for the determination of cyclophosphamide metabolites has been described in detail(27). Briefly, urinary metabolites are extracted onto a non-ionic polymeric adsorban), cltiled with methanol, and separated with TLC. Visualization of drug-related spots is accomplished by spraying the plateswith 4-(4-nitrobenzyl)pyridine, an alkyl acceptor which gives a bluechromophore on alkalinization (28). Because the blue spots deteriorateon standing, the TLC plate is photographed within 10 s of spotdevelopment and the resultant black and white print is scanned in adensitometer. The method is sensitive down to 1 /ig/ml and linear up

    4The abbreviations used are: TLC-PD, combined thin-layer chromatography-photography-densitometry; LC, low carboxylator; HC, high carboxylator; TLC,thin-layer chromatography; ALDH, aldehyde dehydrogenase.

    5167

    on June 17, 2021. © 1988 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    http://cancerres.aacrjournals.org/

  • DEFICIENT URINARY CARBOXYPHOSPHAMIDE EXCRETION

    XCICH.jCH/

    CICH,CH

    OCH,CH;

    OCH;CH

    GKHfH

    CP

    P-450

    4HCP

    aCH,CH/PM +

    CICHjCHA>NH

    CICH,CH/COOH

    NNMFig. 1. Metabolic transformations of cyclophosphamide «I') Boldface ar

    rows, activation pathways; open arrows, detoxication pathways. For abbreviationssee Table 2. 4HCP, 4-hydroxycyclophosphamide; KP, 4-ketocyclophosphamide;AP, aldophosphamide; I'M, phosphoramide mustard; ( 'V. carboxyphosphamide;

    NNM, nor-nitrogen mustard.

    Table 1 Details of patients studied

    Patient1234567891011121314SexMFMMFMMMFMMFFFAge(yr)5350504864726437485549535652Wt(kg)6460924070707075607183738853TumorSCLC°SCLCSCLCSCLCSCLCNHLNHLNHLNHLMFHMFHBCBCUSCPdose(mg)1800100012001000120012001200120060010001200120010001200

    peridol, naproxen, allopurinol, cimetidine, digoxin, potassium chloride,cyclizine, and aluminum hydroxide.

    All patients remained in the hospital for at least 25 h post-cyclo-phosphamide administration. They collected their bulked urine forvarious periods as follows: 1 patient 0-2, 2-8, and 8-24 h; 5 patients0-8 and 8-24 h; 8 patients 0-24 h only. Accordingly, 21 urine samplesof recorded volume were frozen at —20°Cimmediately after collection

    and then analyzed for their content of cyclophosphamide, 4-ketocyclophosphamide, carboxyphosphamide, phosphoramide mustard, and nor-nitrogen mustard by TLC-PD.

    RESULTS

    The photographic record of a TLC plate which had beenvisualized with 4-(4-nitrobenzyl)pyridine as described (27) isshown in Fig. 2. The method clearly separates the principalurinary metabolites 4-ketocyclophosphamide, nor-nitrogenmustard, carboxyphosphamide, and phosphoramide mustardfrom unchanged cyclophosphamide and is specific for the derivatives of cyclophosphamide in the urine, blank urine producing no bands (27). Each urine (typical samples shown in lanes2-7) contained six bands of variable color intensity and Rf 0.67(4-ketocyclophosphamide), 0.61 (cyclophosphamide), 0.40(nor-nitrogen mustard), 0.30 (unidentified metabolite, probablythe des-2-chloroethyl compound by comparison with authenticstandard), 0.26 (carboxyphosphamide), and 0.02 (phosphor-amide mustard). Visual inspection of the photographs of theTLC plates, such as that shown in Fig. 2, revealed that therelative excretion of the metabolites, in particular carboxyphosphamide and phosphoramide mustard, was highly variable between patients. When the black and white photographs of theTLC plates were scanned using a densitometer, a techniquereferred to as TLC-PD (27), chromatograms were obtainedwhich could be quantitated for each urine specimen. From theconcentration of each metabolite in the appropriate urine samples, the percentage of the administered dose excreted as eachmetabolite was calculated. Table 2 gives these individual datafor each of the 14 patients together with the means and SDs.In the 0-24-h urine after i.v. doses of 600-1800 mg cyclophosphamide, the total recovery of drug-related material determinedby TLC-PD was 6.5-64.1% (36.2 ±17.8) dose, in good agreement with results of workers who used radioisotopically labeled

    * SCLC, small cell lung cancer; NHL, non-Hodgkin's lymphoma; MFH,

    malignant fibrous histiocytoma; BC, breast carcinoma; US, uterine sarcoma. t KPCPto 250 Mg/ml for cyclophosphamide, 4-ketocyclophosphamide, carboxyphosphamide, phosphoramide mustard, and nor-nitrogen mustardin human urine and has been termed TLC-PD (27).

    Fourteen patients with various malignancies were studied, each receiving i.v. cyclophosphamide as part of their combination chemotherapy. The patients comprised eight males and six females, mean age53.6 ±8.6 (SD) years, body weight 69.2 ±13.7 kg, five with small celllung cancer, four with non-Hodgkin's lymphoma, two with malignant

    fibrous histiocytoma, two with breast carcinoma, and one with uterinesarcoma (see Table 1). Patients received 600-1800 mg (1130 ±260)cyclophosphamide with the following adjuvant drugs (number of patients): promethazine (11), metoclopramide (10), chlorpromazine (10),Adriamycin (9), vincristine (9), paracetamol (8), dextropropoxyphene(8), lorazepam (8), prednisolone (6), etoposide (6), morphine (5), ami-nophylline (2), salbutamol (2), prochlorperazine (2), and the followingdrugs each received by one patient only: nitrazepam, phenytoin, halo-

    cx

    *§__«* —. — PM12345678

    Fig. 2. TLC plate which has been eluted from bottom to top and visualizedwith 4-(4-nitrobenzyl)pyridine. Lanes 1 and 8, authentic standards dissolved inblank urine; Lanes 2-7, urines from patients given cyclophosphamide (CP) i.v.KP, 4-ketophosphamide; NNM, nor-nitrogen mustard; CX, carboxyphosphamide;I'M. phosphoramide mustard.

    5168

    on June 17, 2021. © 1988 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    http://cancerres.aacrjournals.org/

  • DEFICIENT URINARY CARBOXYPHOSPHAMIDE EXCRETION

    Table 2 Percentage of dose excreted as cyclophosphamide and its various metabolites in the 0-24-h urine in 14 cancer patients

    Numbers in parentheses are relative percentage of each urinary metabolite.

    Patient1234567891011121314Mean

    ±SDCP°5.82.831.413.84.814.127.32.86.111.522.919.05.110.412.7±9.3(21.8)(20.4)(48.9)(40.1)(9.8)(46.4)(55.5)(43.9)(28.5)(41.0)(61.7)(71.6)(8.9)(18.1)(36.9±19.5)KP0.80.40.41.00.71.01.20.50.60.41.01.10.73.91.0+ 0.9(2.9)(3.1)(0.6)(3.0)(1.4)(3.4)(2.5)(7.4)(2.5)(1.3)(2.7)(4.1)(1.2)(6.8)(3.1±2.0)NNM0.50.10.30.60.80.42.10.10.90.31.20.7

  • DEFICIENT URINARY CARBOXYPHOSPHAMIDE EXCRETION

    may be of considerable clinical importance and will be returnedto later.

    DISCUSSION

    The recent development of a method which quantitates allthe principal metabolites of the cytotoxic antineoplastic drugcyclophosphamide has permitted a more detailed inspection ofthe metabolic balance sheet for cyclophosphamide than hasbeen possible hitherto. While several investigators have reported on the various metabolites in patients, studies that haveyielded good excretion data have invariably relied on the administration of radiolabeled drug. The vast majority of determinations of cyclophosphamide "metabolism" or "activation"have utilized the colorimetrie Epstein assay (28) of total alky I•ating activity. This method, which is usually calibrated with themost readily available metabolite bis(2-chloroethyl)amine (nor-nitrogen mustard), can only give the broadest of indications asto the quantitative aspects of cyclophosphamide metabolismand no clue whatsoever as to the qualitative pattern of metabolites in the sample analyzed, for the simple reason that thesine qua non that each metabolite has an identical molar extinction coefficient in the Epstein assay does not hold. In ourhands, the molar extinction coefficients for each authenticmetabolite differ by more than an order of magnitude.5 We

    therefore sought to develop and apply a method which wouldovercome the weaknesses of the Epstein assay to investigate indetail the complete metabolic profile of cyclophosphamide incancer patients.

    With the exception of one breast cancer patient who excretedno detectable nor-nitrogen mustard, all the principal metabolites of cyclophosphamide including 4-ketocyclophosphamide,nor-nitrogen mustard, carboxyphosphamide, phosphoramidemustard, and unchanged drug were found in the 0 24 h urineof all 14 patients studied. The percentage of administered doseexcreted in urine was 36.2 ±17.8% in 0-24 h, which comparesfavorably with the findings of others who have used radioiso-topic methods (29-31). Based upon the relative percentage ofeach urinary metabolite (Table 2), the excretion of each metabolite was highly variable. The two most abundant metabolicproducts carboxyphosphamide and phosphoramide mustardhad coefficients of variation of 118 and 51%, respectively. Alarge proportion of this observed variation seemed to stem fromfive of the patients who excreted 0.3% or less of the administered dose as carboxyphosphamide (Table 2). When this wasfurther examined by plotting the relative percentage of excretion as carboxyphosphamide (multiplied by 10 and called thecarboxylation index, to keep the logarithmic values positive foreach patient) as a logarithmic distribution (Fig. 3), patientswere classified as either LC or HC. Using the variable carboxylation index simply to phenotype patients as either LC or HC,the origins of the metabolic carboxylation defect were soughtfrom the data available. Firstly, the possibility that age, sex,body weight, tumor type, or concomitant drug treatment werethe source of the aberrant metabolism in 5 or 14 patients wasdiscounted.

    The observation of a common phenotype in patients treatedwith cyclophosphamide expressed as a marked deficiency in theexcretion of carboxyphosphamide requires further explanation.It is not possible that the defect lies in the initial cytochromeP-450 mediated production of the aldophosphamide/4-hy-droxycyclophosphamide tautomerie pair, since it would also be

    5 H. L. Roberts. A. F. A. Hadidi, and J. R. Idle, manuscript in preparation.

    reflected in the excretion of other metabolites. As can be seenfrom the metabolic scheme for cyclophosphamide given in Fig.1, the only plausible explanation is that the defect lies in theconversion of aldophosphamide to carboxyphosphamide byALDH (EC 1.2.1.3). Because a frank defect in this pathway hasbeen observed it is unlikely that several enzymes or isozymescontribute to the detoxication of aldophosphamide to carboxyphosphamide. It is probable that a single isozymic form ofALDH which metabolizes aldophosphamide is silent or absentin the low carboxylator phenotype.

    The formation of carboxyphosphamide from microsomalmetabolites of cyclophosphamide has been shown to be catalyzed in vitro by ALDH (32, 33). The selective toxicity ofcyclophosphamide against tumor cells in vivo has been attributed to the differential tissue distribution of ALDH (14, 33).Tissues which are high in ALDH such as liver and kidneydeactivate cyclophosphamide effectively under experimentalconditions (33) and in addition these organs do not showtoxicity when the drug is administered in vivo, whereas it is therelatively lower levels of ALDH in the tumor which permit theconversion of circulating aldophosphamide to the cytotoxicphosphoramide mustard by the cancer cells (34). Moreover,the sensitivity of cells such as murine pluripotential hemato-poietic stem cells and myeloid progenitor cells to oxazaphos-phorine compounds can be manipulated dramatically by thepretreatment of cells with ALDH inhibitors such as disulfiram,diethyldithiocarbamate, and cyanamide (35). Non-oxazaphos-phorines and acrolein showed no potentiation with these treatments. Additionally, murine LI210 cells which have been selected for cyclophosphamide resistance and have developed ahigh ALDH expression (36) will revert to the wild-type sensitivity in the presence of several ALDH inhibitors (37). Theforegoing highlight the essential role of ALDH in determiningtarget tissue sensitivity to cyclophosphamide.

    In spite of the growing interest in the biochemistry andgenetics of ALDH over the past decade, no deficiency in aldophosphamide to carboxyphosphamide conversion has hithertobeen reported. In the main, work has concentrated upon themetabolism of acetaldehyde to acetic acid by ALDH and itspart in alcoholic disease. Human tissues contain two mainisozymes, one with a low Km for acetaldehyde, located in mitochondria and insensitive to disulfiram (ALDH1), and onewith a high Km for acetaldehyde, cytosolic and inhibited bydisulfiram (ALDH2) (38, 39). Two additional forms with a highKm for acetaldehyde and sensitive to disulfiram (ALDH3 andALDH4) are found in certain tissues only (39, 40). A geneticpolymorphism of ALDH 1, associated with alcohol sensitivityand facial flushing, is now well established. In the "unusual"

    phenotype, found in 48% of Japanese, only ALDH2 is foundon electrophoresis and thus the high affinity mitochondrialisozyme is missing. This deficiency, found predominantly inoriental populations, has not been described in Europeans (41).Since cyclophosphamide metabolism is sensitive to disulfiramand other ALDH2 inhibitors, it is highly probable that it isALDH2, ALDH3, or ALDH4 which catalyzes the conversionof aldophosphamide to carboxyphosphamide and was deficientin a significant proportion of our patients. No informationregarding genetic polymorphism of ALDH2-4 is available. Thefindings which we describe in this paper are the first evidencefor such a polymorphism. Because of the cytotoxic nature ofcyclophosphamide it was not possible for us to perform familystudies to confirm the genetic transmission of LC/HC pheno-types. However, a number of strategies at both the protein andDNA level can be envisaged.

    5170

    on June 17, 2021. © 1988 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    http://cancerres.aacrjournals.org/

  • DEFICIENT URINARY CARBOXYPHOSPHAMIDE EXCRETION

    The detection of genetic polymorphism of the ALDH iso-

    zyme responsible for the detoxication of cyclophosphamidewould be of great clinical interest, from the point of view ofboth tolerability of the drug and acquired resistance to it. Thelow carboxylators who comprised 36% of our small patientcohort excreted over twice the relative amount of cytotoxicphosphoramide mustard and presumably therefore producedtwice the molar equivalent of the toxic aldehyde acrolein (seeFig. 1). The TLC-PD method provides a simple means forscreening patient populations for ascertainment of the completemetabolic picture after cyclophosphamide administration andthe ability to assign carboxylation phenotype. Parallel studiesto be reported elsewhere6 with the isomer of cyclophosphamide,ifosfamide, in non-small cell lung cancer patients reveal asimilar proportion of patients with defective excretion of thecarboxylic acid metabolite.

    ACKNOWLEDGMENTS

    The expert technical and photographic skills of Jane Baker aregratefully acknowledged.

    REFERENCES

    1. Hohorst, H. J., Ziemann, A., and Brock, N. 4-Ketocyclophosphamide, ametabolite of cyclophosphamide. Formation, chemical and biological properties. Arzneim. Forsch., 21: 1254-1257, 1971.

    2. Struck, R. F., Kirk, M. C, Mellett, L. B., El Dareer, S., and Hill, D. L.Urinary metabolites of the antitumour agent cyclophosphamide. Mol. Phar-macol., 7:519-529, 1971.

    3. Takamizawa, A., Tochino, Y.. Hamashima, Y., and Iwata, T. Studies oncyclophosphamide metabolites and their related compounds. Chem. Pharm.Bull. 20: 1612-1616, 1972.

    4. Cohen, J. L., and Jao, J. Y. Enzymatic basis of cyclophosphamide activationby hepatic microsomes of the rat. J. Pharmacol. Exp. Ther. 174: 206-210,1970.

    5. Connors, T. A., Grover, P. L., and McLoughlin, A. M. Microsomal activationof cyclophosphamide in vivo. Biochem. Pharmacol. 19: 1533-1535, 1970.

    6. Sladek, N. E. Metabolism of cyclophosphamide by rat hepatic microsomes.Cancer Res. 31:901-908, 1971.

    7. Struck, R. F. Isolation and identification of a stabilized derivative of aldo-phosphamide, a major metabolite of cyclophosphamide. Cancer Res. 3-1:2933-2935, 1974.

    8. Struck, R. F. Aldophosphamide; synthesis, characterization, and comparisonwith "Hohorst's aldophosphamide." Cancer Treat. Rep., 60:317-319,1976.

    9. Volker, G., Drager, U., Peter, G., and Hohorst, H. J. Studien zum Spontanzerfall von 4-Hydroxycyclophosphamid und 4-Hydroperoxycyclophos-phamid mit Hilfe der Dunnschichtchromatographie. Arzneim. Forsch. 24:1172-1176,1974.

    10. Myles, A., Fenselau, C., and Friedman, O. M. Synthesis of aldophosphamide,a key cyclophosphamide metabolite. Tetrahedron Lett. 29:2475-2478,1977.

    11. Bakke, J. E., Feil, V. J., and Zaylskie, R. G. Characterization of the majorsheep urinary metabolites of cyclophosphamide, a defleecing chemical. J.Agrie. Food Chem. 19: 788-790, 1971.

    12. Bakke, J. A., Feil, V. J., Fjelstul, C. E., and Thacker, E. J. Metabolism ofcyclophosphamide by sheep. J. Agrie. Food Chem. 20: 384-388, 1972.

    13. Colvin, M., Padgett, C. A., and Fenselau, C. A biologically active metaboliteof cyclophosphamide. Cancer Res. 33: 915-918, 1973.

    14. Connors, T. A., Cox, P. J., Farmer, P. B., Foster, A. B., and Jarman, M.Some studies on the active intermediates formed in the microsomal metabolism of cyclophosphamide and isophosphamide. Biochem. Pharmacol. 23:115-129, 1974.

    15. Struck, R. F., Kirk, M. C., Witt, M. H., and Laster, W. R., Jr. Isolation andmass spectral identification of blood metabolites of cyclophosphamide: evi-

    * H. L. Roberts, M. Lind, N. Thatcher, and J. R. Idle, manuscript in prepara

    tion.

    dence for phosphoramide mustard as the biologically active metabolite.Biomed. Mass Spectrom., 2: 46-52, 1975.

    16. Colvin, M., Brundrett, R. B., Kan, M. N. N., Jardine, I., and Fenselau, C.Alkylating properties of phosphoramide mustard. Cancer Res. 36: 1Hill 26, 1976.

    17. Fenselau, C., Lehman, J. P., Myles, A., Brandt, J., Yost, G. S., Friedman,O. M., and Colvin, M. Iminocyclophosphamide as a chemically reactivemetabolite of cyclophosphamide. Drug Metab. Dispos. 10: 636-640, 1982.

    18. Alarcon, R. A., and Meienhofer, J. Formation of the cytotoxic aldehydeacrolein during in vitro degradation of cyclophosphamide. Nat. (New Biol.),233: 250-252, 1971.

    19. Brock, N., Stekar, J., Pohl, J., Niemeyer, U., and Scheffler, G. Acrolein, thecausative factor of urotoxic side-effects of cyciophosphamide, ifosfamide,trofosfamide and sufosfamide. Arznein. Forsch. 29:659-661, 1979.

    20. Brock, N., Pohl, J., and Stekar, J. Studies on the urotoxicity of oxazaphos-phorine cytostatics and its prevention. I. Experimental studies on the urotoxicity of alkylating compounds. Eur. J. Cancer 17: 595-607, 1981.

    21. Cox, P. J. Cyclophosphamide cystitis—identification of acrolein as thecausative agent. Biochem. Pharmacol. 28: 2045-2049, 1979.

    22. Sladek, N. E., Smith, P. C., Bratt, P. M., Low, J. E., Powers, J. F., Borch,R. F., and Coveney, J. R. Influence of diuretics on urinary general baseactivity and cyclophosphamide-induced bladder toxicity. Cancer Treat. Rep.66: 1889-1900, 1982.

    23. Sladek, N. E. In: G. Powis and R. A. Prough (eds.). Metabolism and Actionof Anti-cancer Drugs, pp. 48-90. London: Taylor & Francis, 1987.

    24. Arnold, H., and Borseaux, F. Synthese und Abbau cytostatisch wirksamercyclischer yV-Phosphamidester des Bis-(/3-chlorathyl)-atnins. Angew. Chem.70:539-544, 1958.

    25. Friedman, O. M., and Seligman, A. M. Preparation of /V-phosphorylatedderivatives of bis-0-chloroethylaminc. J. Am. Chem. Soc. 76:655-658, 1954.

    26. Hilton, J. Deoxyribonucleic acid crosslinking by 4-hydroperoxycyclophos-phamide in cyclophosphamide-sensitive and -resistant L1210 cells. Biochem.Pharmacol. 33:1867-1872, 1984.

    27. Hadidi, A. F. A., and Idle, J. R. Combined thin-layer chromatography-photography-densitometry for the quantitation of cyclophosphamide and itsfour principal urinary metabolites. J. Chromatogr. 427: 121-130, 1988.

    28. Epstein, J., Rosenthal, R. W., and Ess, R. J. Use of 4-(4-nitrobenzyl)pyridineas an analytical reagent for ethylenimines and alkylating agents. Anal. Chem.27:1435-1439, 1955.

    29. De Vita, V. T., and Adamson, R. H. The metabolism of 14C-cyclophospha-

    mide: comparative drug metabolism studies on African and American patients with lymphosarcoma and Burkitt's tumor. Prog. Antimicrob. Antican-cer Chemother., 2: 218-225, 1970.

    30. Bagley, C. M., Jr., Bostick, F. W., and De Vita, V. T., Jr. Clinical pharmacology of cyclophosphamide. Cancer Res. 33: 226-233, 1973.

    31. Mouridsen, H. T., Faber, O., and Skovsted, L. The biotransformation ofcyclophosphamide in man: analysis of the variation in normal subjects. ActaPharmacol. Toxicol. 35: 98-106, 1974.

    32. Hill, D. L., Laster, W. R. Jr., and Struck, R. F. Enzymatic metabolism ofcyclophosphamide and nicotine and production of a toxic cyclophosphamidemetabolite. Cancer Res. 32: 658-665, 1972.

    33. Cox, P. J., Phillips, B. J., and Thomas, P. The enzymatic basis of the selectiveaction of cyclophosphamide. Cancer Res. 35: 3755-3761, 1975.

    34. Hipkens, J. H., Struck, R. F., and Gurtoo, H. L. Role of aldehyde dehydro-genase in the metabolism dependent biological activity of cyclophosphamide.Cancer Res. 41: 3571-3583, 1981.

    35. Kohn, F. R., and Sladek, N. E. Aldehyde dehydrogenase activity as the basisfor the relative insensitivity of murine pluripotem hematopoietic stem cellsto oxazaphosphorines. Biochem. Pharmacol. 34: 3465-3471, 1985.

    36. Hilton, J. Role of aldehyde dehydrogenase in cyclophosphamide-resistantL1210 leukemia. Cancer Res., 44: 5156-5160, 1984.

    37. Sladek, N. E., and Landkamer, G. L. Restoration of sensitivity to oxazaphosphorines by inhibitors of aldehyde dehydrogenase activity in culturedoxazaphosphorine-resistant I 1210 and cross-linking agent-resistant P388cell lines. Cancer Res., 45: 1549-1555, 1985.

    38. Greenfield, N. J., and Pietruszko, R. Two aldehyde dehydrogenases fromhuman liver. Isolation via affinity chromatography and characterisation ofthe isozymes. Biochim. Biophys. Acta 483: 35-45, 1977.

    39. Goedde. H. W., Meier-Tackmann, D., and Harada, S. Physiological role ofaldehyde dehydrogenase isozymes. In: H. Weiner and B. Wermuth (eds.),Enzymology of Carbonyl Metabolism, pp. 347-362. New York: Alan R.Liss, Inc., 1982.

    40. Harada, S., Agarwal, D. P., and Goedde, H. W. Electrophoretic and biochemical studies of human aldehyde dehydrogenase isozymes in various tissues.Life Sci. 26:1773-1780, 1980.

    41. Goedde, H. W., Agarwal, D. P.. Harada, S., Meier-Tackmann, D., Ruofu,D.. Bienzle, U., Kroeger, A., and Hussein, L. Population genetic studies onaldehyde dehydrogenase isozyme deficiency and alcohol sensitivity. Am. J.Hum. Genet. 35: 769-772, 1983.

    5171

    on June 17, 2021. © 1988 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    http://cancerres.aacrjournals.org/

  • 1988;48:5167-5171. Cancer Res Al-Hakam F. A. Hadidi, Carmel E. A. Coulter and Jeffrey R. Idle to Cancer PatientsCarboxyphosphamide after Cyclophosphamide Administration Phenotypically Deficient Urinary Elimination of

    Updated version

    http://cancerres.aacrjournals.org/content/48/18/5167

    Access the most recent version of this article at:

    E-mail alerts related to this article or journal.Sign up to receive free email-alerts

    Subscriptions

    Reprints and

    [email protected] at

    To order reprints of this article or to subscribe to the journal, contact the AACR Publications

    Permissions

    Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

    .http://cancerres.aacrjournals.org/content/48/18/5167To request permission to re-use all or part of this article, use this link

    on June 17, 2021. © 1988 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    http://cancerres.aacrjournals.org/content/48/18/5167http://cancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://cancerres.aacrjournals.org/content/48/18/5167http://cancerres.aacrjournals.org/

Recommended