Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

[CANCER RESEARCH 44, 5638-5643, December 1984]

Role of NADPH:Cytochrome c ReducÃ-aseand DT-Diaphorase in the


Biotransformation of Mitomycin C1

Susan R. Keyes, Paula M. Fracasso, David C. Heimbrook,2 Sara Rockwell, Stephen G. Silgar,3 and
Alan C. Sai-torelli4

Departments of Pharmacology [S. R. K., P. M. F., A. C. S.] and Therapeutic Radiology [S. R.J and Developmental Therapeutics Program, Comprehensive Cancer Center,
Yale University School of Medicine, New Haven 06510, and Department of Molecular Biophysics and Biochemistry, [S. G. S., D. C. H.¡Yale University,
New Haven 06511, Connecticut

ABSTRACT rally occurring antibiolic, MC.5 This drug is aclive against a broad
range of transplantable animal (5) and human (2) tumors, is
Hypoxie cells of solid tumors are difficult to eradicate by X- preferentially cytotoxic to some hypoxic tumor cell lines in vitro
irradiation or chemotherapy; as an approach to this problem, our (11, 23, 27), and is also toxic to hypoxic tumor cells in vivo (25,
laboratories are investigating the effects of the bioreductive 26).
alkylating agent mitomycin C (MC) on hypoxic cells. This anti Early studies have demonslraled lhal enzymalic bioactivalion
biotic was preferentially toxic to EMT6 mouse mammary tumor of MC results in cyloloxicily associated wilh cross-linking of
cells and V79 Chinese hamster lung fibroblasts under hypoxic DNA (9). Using liver homogenales, the anaerobic production of
conditions, but it was equitoxic to Chinese hamster ovary cells reaclive species from MC has been shown lo require NADPH
in the presence and absence of oxygen. All cell lines catalyzed and to be catalyzed primarily by enzymes in the microsomal and
the formation of reactive metabolites under hypoxic conditions nuclear subcellular fractions (12, 28), and some alkylation prod
and contained NADPH:cytochrome c reducÃ-aseand DT-diaphor- ucts have been identified (31). Limited studies with purified
ase, two enzymes which may be responsible for the cellular enzymes have indicated that both NADPHxytochrome c reduc
activation of MC. Although a correlation existed between enzy Ã-aseand xanthine oxidase actÃ-valeMC under hypoxic conditions,
matic activities and the formation of reactive metabolites from wilh Ihe concomilanl generalion of a reaclive eleclrophile (14,
MC, there was no correspondence between these parameters 21 ). Studies in our laboratories have suggested lhal cytochrome
and the degree of cytotoxicity expressed by MC under hypoxic P-450 might be involved in the anaerobic metabolism of MC (11,
conditions. 12), and work by others on DT-diaphorase, an enzyme active in
Purified NADPHrcytochrome c reducÃ-ase reduced MC in the the reduction of quiñones (6, 30), suggests that this enzyme
absence of oxygen, with addition of cytochrome P-450 enhanc should also be considered as a possible activator of MC.
ing, but noi participating directly in, the reduction reaction. Ad Although the mechanism by which MC is metabolically acti
dition of NADP+ lo cell sonicates substantially reduced NADPH:- vated has been examined extensively in bacteria and liver, little
cylochrome c reducÃ-ase activity, while Ihe formation of reactive is known about the bioaclivation of MC by neoplastic cells.
metabolites was affected only slightly; converse results were Previous investigations in our laboratories have demonslraled
observed using mersalyl. Exposure of cell sonicates lo dicumarol that MC is more toxic to EMT6 and Sarcoma 180 tumor cells
inhibiled DT-diaphorase activity, while the rate of formation of under hypoxic conditions than in the presence of air, and lhal
reactive metabolites of MC was enhanced. The findings suggest sonicates of these cells are capable of generating a reactive
that NADPH:cytochrome c reducÃ-ase and some as yet to be species from MC (11, 27). However, neilher Ihe enzyme(s) in
identified enzyme(s) are important for the reductive activalion of lumor cells responsible for activating MC nor the exact biochem
MC. DT-diaphorase and cytochrome P-450 are not directly in ical basis of Ihe enhanced cyloloxicily in Ihe absence of oxygen
volved in the activalion of MC, bul they appear lo modulate the has yet been characterized. The studies described in Ihis paper
degree of activalion lo reaclive species, which are presumably were designed to address these issues.
responsible for Ihe observed cytotoxicity.

MATERIALS AND METHODS


INTRODUCTION
MC was supplied by the Bristol-Myers Co. (Syracuse, NY). Other
materials were obtained from the following sources: NADP*, NADH,
Solid neoplasms contain hypoxic tumor cell populations; since
oxygen-deficient cells are relatively refractory lo convenlional NADPH, glucose-6-phosphate dehydrogenase (type XII), glucose 6-phos-
phate, dicumarol, mersalyl, 2,6-dichlorophenolindophenol, dilaurylphos-
radiolherapy and mosl chemolherapy, hypoxic cells have Ihe phatidylcholine, DEAE-cellulose, and 2',5'-ADP agarose (Sigma Chemi
potential to limit curability (13, 19). As an approach to the cal Co., St. Louis, MO); fetal bovine serum and Waymouth's medium
circumvention of this problem, we have been studying the natu-
(Grand Island Biological Co., Grand Island, NY); DEAE-Trisacryl M and
1This work was supported in part by American Cancer Society Grants CH-211 HA-Utrogel (LKB Instruments, Gaithersburg, MD); gases (Presto Welding
and PDT 145, USPHS Grant CA-02817, NIH Grants GM-31756 and AM-01160, Service Center, North Haven, CT); and 4-<p-nitrobenzyl)pyridine (Aldrich
and American Heart Association Fellowship 11-104-812. Chemical Co., Milwaukee, Wl). Chinese hamster V79 cells (subline V79-
2 Present address: Department of Pharmacology, Yate University School of
8) and CHO cells (subline HA-1) were gifts of Dr. R. Michael Liskay and
Medicine, New Haven, CT 06510.
3 Present address: Department of Biochemistry, University of Illinois, Urbana, IL Dr. Daniel S. Kapp, Department of Therapeutic Radiology, Yale University
61801. School of Medicine.
4 To whom requests for reprints should be addressed.
Received May 21, 1984; accepted August 29, 1984. 5The abbreviations used are: MC, mitomycin C; CHO, Chinese hamster ovary.

CANCER RESEARCH VOL. 44 DECEMBER 1984


5638

Downloaded from cancerres.aacrjournals.org on April 10, 2019. © 1984 American Association for Cancer Research.
BIOTRANSFORMATION OF MITOMYCIN C

EMT6 mouse mammary tumor cells, Chinese hamster V79 cells, and previous studies have suggested their importance in the bioac-
CHO cells were grown at 37° in Waymouth's medium supplemented
tivation of MC (11,12,14, 21). Preliminary experiments showed
with 15% fetal bovine serum and antibiotics in a humidified atmosphere that an increase in the reactive reduction product(s) of MC
of 95% air:5% CO2. The characteristics of these 3 cell lines have been trapped with 4-(p-nitrobenzyl)pyridine correlated with the disap
described in detail previously (10,16, 24).
pearance of the quinone absorbance at 363 nm (data not shown);
For survival studies, cells were cultured in glass milk dilution bottles
and used in midexponential growth. Cells were exposed to 1.5 UM MC
therefore, the latter parameter was monitored in these experi
for various times after pregassing with 95% air:5% CO2 or 95% N2:5% ments. The anaerobic reduction of MC by NADPH :cytochrome
CO2 for 2 hr (26). The survival of cells was assayed by testing their ability c reductase or the reductase plus P-450LM2was a typical first-
to form macroscopic colonies (11, 24, 27). Both hypoxic and aerobic order reaction between 0 and 50 UM MC, as would be expected
vehicle (50 n\ of 0.9% NaCI solution or 70% ethanol) controls were for substrate concentrations significantly below the Km of the
included in each experiment; the surviving fractions for hypoxic cultures enzymes. Although MC was reduced under hypoxic conditions
were calculated using the hypoxic controls. by NADPHxytochrome c reductase alone, the rate of reduction
Cell sonicates and microsomes were prepared from cells grown as was enhanced by addition of P-450 and reached a maximum at
either suspensions or as monolayers. After being washed with cold
approximately a 1:1 molar ratio of NADPH:cytochrome c reduc
phosphate-buffered saline, the cells were resuspended in approximately
tase to P-45ÛLM2(Chart 1).
6 volumes of water. After a 10-min incubation on ice, an equal volume
of 1.8% NaCI solution was added, and the cells were disrupted by three
The rate of MC reduction catalyzed by purified NADPH:
6-sec bursts with a Branson Model 160 sonicator set at 25% of maximum cytochrome c reductase and P-450 was not decreased by carbon
intensity. Cell sonicates were used for enzyme assays and for preparation monoxide (data not shown). When the reoxidation rate of the
of subcellular fractions. Microsomes were prepared by the method of ferrous P-450:carbonyl complex after exhaustion of NADPH was
Cinti er al. (3) after cell sonicates were adjusted to a concentration of monitored at 450 nm in the presence of MC, the rate was
0.25 M sucrose by addition of 1.25 M sucrose. Microsomes were contam approximately 100 times slower than the initial velocity of MC
inated with <5% mitochondria as judged by succinatercytochrome c reduction after the addition of a second aliquot of NADPH (Chart
reductase activity (29) and <1 % cytosol as measured by lactate dehy-
2). These data support the concept that NADPH:cytochrome c
drogenase activity (8), and total protein recovery averaged 92% as reductase is the primary reductant of MC, while P-450 acts only
determined by the method of Lowry ef al. (17). as a modulator of enzymatic activity. Under oxygenated condi-
Reactive metabolites of MC were estimated by a modification of the
method of Wheeler ef al. (33) as described previously (11). The absorb-
ance of the adducts of MC and 4-(p-nitrobenzyl)pyridine was measured 0006
at 540 nm. The reaction rate was linear for at least 30 min, and the 5%
final concentration of acetone in which the trapping agent and MC were 7 0004 *•
dissolved did not affect the enzymatic activation of MC. O

Cellular oxidoreductase activities were measured by standard assays. 0.002


NADPH: and rotenone-insensitive NADH:cytochrome c reductases were
determined at 30°by monitoring the rate of reduction of cytochrome c
at 550 nm (29, 34), using an extinction coefficient of 27.7 rnw"1 cm"1. 0 0.2 0.4 06
P450 [//M]
NADHxytochrome bs reductase and DT-diaphorase activities were de
termined at 30°by the reduction of KaFefCNfe (420 nm, «= 1.02 mn/T1 Chart 1. Anaerobic reduction of MC by NADPHxytochrome c reductase and
cm~1) and dichlorophenolindophenol (600 nm, < = 21 rriM"1 cm"1), re cytochrome P-450. The reaction mixture contained 30 MM MC, 0.2 MM
NADPH:cytcchrome c reductase, dilaurylphosphatidylcholine(30 M9/mt), 100 MM
spectively (6,29). Cytochrome i>5and cytochrome P-450 were measured NADPH, 0 to 0.6 MMcytochrome P-450, and 100 mw Tris-HCI (pH 7.0) in a total
volume of 1 ml. The anaerobic reduction of MC was monitored at 25°by the loss
in microsomes by the method of Omura and Sato (20) using an Aminco
DW-2 UV/VIS spectrophotometer in the split-beam mode. The extinction of the quinone absorbance at 363 nm, and the results show the first-order rate
coefficients used were 185 rtiM~1 cm"1 for cytochrome to5for the wave constant of MC reduction in the absence of oxygen.
length pairs, 425 and 410 nm, and 91 mw"1 cm"1 for the cytochrome P-
450:carbon monoxide complex for the wavelength pairs, 450 and 495
nm.
Cytochrome P-450uiœ and NADPH:cytochrome c reductase were pu I.O IO
rified from the livers of phenobarbital-induced rabbits by standard pro
cedures (4,7). Both were homogeneous as judged by gel electrophoresis
(15). Enzymatic reduction of MC with purified enzymes was conducted
in a 1-ml solution of 100 mM Tris-HCI (pH 7.0) containing dilaurylphos- S 0.8
phatidylcholine (30 /¿g/ml),30 MM MC, 0.2 ^M NADPH:cytochrome c o
reductase, and varying concentrations of P-45ÛLM2.The reaction was ç

initiated by the addition of 100 nmol NADPH, and the time course of the
60.6 6 I
reduction was followed by measuring loss of the absorbance of the o 1
quinone moiety at 363 nm. Anaerobic experiments were performed under
an atmosphere of N2 in a Thunberg-type cuvet, after repeated degassing
and purging with N2. In some experiments, carbon monoxide replaced
the N2 atmosphere. All spectrophotometric observations were performed I.O 2.0 3.0
in a Gary 219 spectrophotometer at 25°.
Time (mm)
Chart 2. Comparison of the rate of cytochrome P-450u«reoxidation to the rate
RESULTS of MC reduction. The reaction conditions were as described in Chart 1, with 0.6
MMcytochrome P-450 and 30 nmol NADPH. For 0 to 1.5 min, the concentration of
the ferrous:carbonyl complex of P-450 was monitored at 450 nm, and for 1.5 to
Purified NADPH :cytochrome c reductase and cytochrome P-
3.0 min, the concentration of MC was monitored by the quinone absorbance at
45ÛLM2were used to reduce MC under hypoxic conditions, since 363 nm after a second addition of NADPH.

CANCER RESEARCH VOL. 44 DECEMBER 1984


5639

Downloaded from cancerres.aacrjournals.org on April 10, 2019. © 1984 American Association for Cancer Research.
BIOTRANSFORMATION OF MITOMYCIN C

lions, the quinone absorbance of MC did not decrease, even Table 1


though NADPH was still consumed (data not shown). This finding Formation of reactive metabolites from reduction of mitomycin C
The standard reaction mixture contained an NADPH-regenerating system (0.72
is consistent with the scavenging of the MC semiquinone radical m«NADP* :5 my MgCI2:5 mw glucose 6-phosphate: 1.25 unit glucose 6-phosphate
by oxygen, which results in oxygen-induced inhibition of the dehydrogenase), 470 MM4-(p-nilrobenzyl)pyridine, and 3 to 5 mg cell sonicate in a
activation of MC to a reactive metabolite. total volume of 1 ml 100 mm Tris-HCI (pH 7.4). The reaction was initiated by the
addition of 300 nmol mitomycin C.
These findings with purified enzymes were compared to the
AMO* lO^/min/mg protein
hypoxic activation of MC in whole cells. As model systems, we
chose EMT6 mouse mammary tumor cells, V79 Chinese hamster
lung fibroblasta, and CHO cells, since these cell lines are com systemAir
cofactorshypoxia*
monly used for investigations on the effects of radiosensitizing Cell +
hypoxia*NADH hypoxia"No
agents and cytotoxic drugs with specificity for hypoxic cells. The lineNADPH-regenerating HypoxiaNADPH

toxicity of MC to these cell lines under hypoxic and oxygenated CHO 0.03 ±0.0r0.33 ±0.06 0.18 ±0.03 0.15 ±0.03 0.11 ±0.02
V79 0.03 ±0.01 0.39 ±0.04 0.30 ±0.04 0.20 ±0.02 0.12 ±0.05
conditions is shown in Chart 3. The survival curves for EMT6 EMT6 0.05 ±0.01 1.04 ±0.05 0.77 ±0.13 0.36 ±0.11 0.17 ±0.02
and V79 cells were similar, with the survival of the hypoxic cells * The NADPH-regeneratingsystem was omitted or replaced with 1 mw reduced
being significantly lower than that of aerobic cells and with the pyridine nudeotjde.
slopes of the hypoxic curves being steeper than those of the Mean ±S.E. with n > 4.
aerobic curves. For CHO cells, there was no significant difference
between aerobic and hypoxic cell survival with short (<2.5 hr) The rate of generation of a reactive species in the absence of
exposure to MC and only insignificantly greater toxicity to hy cofactors was markedly faster than the rate in the presence of
poxic cells at prolonged (>2.5 hr) times of exposure. The data oxygen, indicating that there were significant levels of endoge
from 2 and 4 independent experiments are shown for V79 and nous reducing equivalents in the sonicates. These results quali
CHO cells, respectively; the experimental results for EMT6 cells tatively agree with earlier data from this laboratory using EMT6
were from a single determination, performed during the same and S180 cells (11, 27) and with data obtained using liver
time period with the same lot of MC. The findings with EMT6 homogenates (28).
cells agreed with those published previously (11), which were The presence of activities representative of enzymes impli
based on independent and more extensive data. cated in the bioactivation of MC was measured in the cell lines
Since most research on the anaerobic metabolism of MC has under investigation. Enzyme activities characteristic of 3 liver
been conducted with liver, we analyzed the basic requirements microsomal electron transport proteins and one liver cytosolic
needed for activation of MC in these cell systems under anaer enzyme were present in the cultured cell lines. In addition,
obic conditions with an NADPH-regenerating system; all 3 cell cytochrome os, but not cytochrome P-450, was detected in the
lines activated MC to a reactive species that alkylated the cellular microsomal fraction (Table 2). NADPHxytochrome c
nucleophile 4-(p-nitrobenzyl)pyridine (Table 1). The greatest reductase activity and DT-diaphorase activity were higher in
monoalkylating activity was obtained with EMT6 cell sonicates; EMT6 cells than in CHO and V79 cells. NADH:cytochrome c
substantially lower activities occurred with CHO and V79 cell reductase activity (a measurement of NADH:cytochrome i>5re
sonicates. The generation of reactive metabolites from MC was ductase plus cytochrome i>5), NADH:cytochrome u5 reductase
inhibited by oxygen and by the absence of exogenous reduced activity, and cytochrome 05 were not significantly different in the
pyridine nucleotides. Both NADPH and NADH supported the cell lines.
formation of reactive metabolites from MC, with greater rates of NADHxytochrome 65 reductase, NADHxytochrome c reduc
alkylation occurring in the presence of NADPH; however, neither tase, and DT-diaphorase activities were markedly variable, as
cofactor was as efficient as the NADPH-regenerating system. evidenced by the large standard errors. This variation reflected
increases in the enzyme activities in the cells as the cultures
entered plateau phase (data not shown). In contrast,
NADPHxytochrome c reductase activity was identical in expo
nentially growing and plateau-phase cells. All studies involving
cell survival, inhibitors, and reactive metabolites were performed
with cells in exponential growth.
Inhibitors of various enzymes implicated in the biotransforma-
tion of MC were used as probes to estimate the contribution of
specific oxidoreductases to the reductive activation of MC. Pre
vious papers from this laboratory (11, 12) reported inhibition of
reactive metabolite formation by CO, suggesting a role for cy
tochrome P-450 in the activation of MC. However, anaerobic
metabolism of MC with purified NADPHxytochrome c reductase
and cytochrome P-450 was unaffected by CO, and no cyto
chrome P-450 could be detected in the microsomal fraction of
Hours of
Charts. Survival of aerobic and hypoxic EMT6, V79, and CHO cells as a
the cultured cells.
function of time after treatment with MC. The cells, grown to midlog phase in glass In an attempt to resolve these discrepancies, EMT6 cells were
bottles, were treated with 1.5 ¡MMC as described in 'Materials and Methods."
treated with CO and MC. If P-450, which may have been present
Hypoxia was achieved by gassing with 95% N2:5% CO, for 2 hr before treatment
with MC for 1.0 to 3.5 hr, with continued gassing. O, O, A, D, aerobic survival; ». at levels undetectable by our procedures, were involved in the
»,A, •.
hypoxic survival. Each symbol represents a different experiment. activation of MC, CO should protect cells from MC toxicity by

CANCER RESEARCH VOL. 44 DECEMBER 1984


5640

Downloaded from cancerres.aacrjournals.org on April 10, 2019. © 1984 American Association for Cancer Research.
BIOTRANSFORMATION OF MITOMYCIN C

Table2
Oxidoreductaseactivities in cultured cell lines
All enzyme activities or concentrations were determined by standard assays described in "Materials and Methods.'
Cytochrome P-450 pathway Cytochrome b, pathway
NADPHicytochrome c reducÃ-ase one-insensit!ve
inCell activity (nmol/min/mg protein) reducÃ-ase NADHrcytochromec
P-450 (pmol/ activity OÃ-mol/min/ reducÃ-ase activity b, (pmol/mg activity
mg protein) in mg protein) in (nmol/min/mg protein) protein) in (nmol/min/mg protein)
microsomes*<0.5 microsomes*34.2
lineCHO sonicates1.6 sonicates42.6
in insonicates10.0±
±0.16 ±0.6 ±0.2" ±10.7* 3.2e
V79 1.1 ±0.1 3.6 ±1.0 <0.5 1.0 ±0.1 34.8 ± 5.6 37.5 13.7± 2.1
EMT6Sonicates0.9 14.0 ±1.6Cytochrome
4.6 ±0.6Microsomes*2.0 <0.5NADHrcytochromeus
1.5 ±0.3Roter 60.1 ±20.3Cytochrome 31.1OT-diaphorase 151.4 ±48.7
" Cytochrome P-450, Cytochromei>5,and NADPHicytochrome c reducÃ-ase
activities in microsomes were the average of one, 2, and 3 determinations, respectively.
Mean ±S.E. of 12 determinations.
Mean ±S.E. of 7 independent experiments.

Tables
I.CH
Effects of inhibitors of NADPH:cytochromec reductase activity and DT-
diaphorase activity on the formation of reactive metabolites of mitomycin C
The reaction mixtures were as described in Tables 1 and 2, with the addition of
inhibitors as indicated. NADP* was dissolved in 100 mw Tris-HCI (pH 7.4), and
IO"
mersalyl and dicumarol were added in 0.5 M NaOH. Neither vehicle affected the
reaction rates.
% of control activity*

Cytochromec
Inhibitor
(HIM)Dicumarol0.10.51.0NADP*1.05.010.0Mersalyl0.10.250.5NADPH:
reductase
activity100 activity2± metabolites98

1*10083
± 10018 ±13143±13185
IO"'-
±1021 ±2274

83±
± 94945
± ±1069
3088 ±1233±
O 1.0 2O 3O 271
Time of Exposure to CO (hr)
Chart 4. Effect of carbon monoxide on the survival of MC-treated cells. The ±1265 319± 922±
experimentalconditions were as described in Chart 3 and 'Materials and Methods.'
±1033±13DT-diaphorase
413±13Reactive
± ±106±
Ninety-five% CO:5% CO2replaced95% N2:5%C02 where indicated. MC or vehicle 3
treatments were for 1 hr. A, aerobic cell survival, vehicle; O, aerobic cell survival, * The effects of inhibitors are expressed as percentages of the control activities
1.5 IM MC; A, hypoxic cell survival, vehicle with CO; •, hypoxic ce«survival, 1.5
„MMC with CO. which are given in previous tables.
Mean ±S.E. of at least 3 determinations where applicable.

inhibiting the formation of cytotoxic species. As shown in Chart NADPH:cytochrome c reductase and DT-diaphorase activities
4, CO, which as a single agent was not toxic, failed to protect and decreased the rate of formation of reactive MC metabolites.
EMT6 cells against the lethal effects of MC but, instead, in Since studies with dicumarol showed that DT-diaphorase did not
creased hypoxic cell susceptibility to the antibiotic. In addition, catalyze the production of an electrophile(s) from MC, the results
measurement of reactive produces) from MC demonstrated that of studies with NADP* and mersalyl can be used to assess the
the formation of a reactive electrophile by cell sonicates was not role of NADPHrcytochrome c reductase in the formation of
inhibited by CO (data not shown). The contradiction between reactive products from MC. There was no quantitative relation
previous results (11,12) and the present studies may be attrib ship between the degree of inhibition of NADPH:cytochrome c
utable to an oxygen leak in the experimental apparatus used for reductase activity by NADP* or mersalyl and the rate of produc
the earlier investigations. These findings indicate that cyto-
tion of reactive metabolites over the entire range of inhibitor
chrome P-450 is not involved in bioactivation of MC in EMT6
concentrations used. NADPHxytochrome c reductase was more
cells and, furthermore, that Cytochrome P-450, if present, would sensitive to inhibition by NADP+ than was the generation of
only function to modify the activity of NADPHicytochrome c reactive metabolites; the reverse was true with mersalyl. Similar
reductase. results were found using V79 and CHO cell sonicates (data not
Additional experiments using inhibitors of NADPH:cytochrome shown). These results support the concept that NADPH:
c reductase and DT-diaphorase examined the role of these
Cytochrome c reductase is involved in the activation of MC under
enzymes in the production of a reactive product from MC by hypoxic conditions but indicate that other factors are also in
EMT6 cell sonicates (Table 3). Dicumarol effectively blocked the volved in this reaction.
activity of DT-diaphorase at 0.1 HIM but did not decrease
NADPHrcytochrome c reductase activity at concentrations up to DISCUSSION
1 (TIM. In addition, dicumarol did not decrease but instead en
hanced the formation of a reactive product from MC. The biochemical mechanism of MC transformation has been
Both mersalyl and NADP+ significantly lowered the widely investigated using bacterial and liver systems, but few

CANCER RESEARCH VOL. 44 DECEMBER 1984


5641

Downloaded from cancerres.aacrjournals.org on April 10, 2019. © 1984 American Association for Cancer Research.
BIOTRANSFORMATION OF MITOMYCIN C

studies have used tumors or other mammalian cell lines. Previous relative importance of NADPH:cytochrome c reductase and DT-
work in our laboratories demonstrated that EMT6 mouse mam diaphorase in the biotransformation of MC, the effects of NADP+,
mary tumor cells and Sarcoma 180 ascites cells are more sen mersalyl, and dicumarol on enzyme activities and reactive me
sitive to the cytotoxic action of MC under hypoxic conditions tabolites were examined. Dicumarol markedly inhibited DT-dia
than in air (11, 27), and that sonicates of these cell lines are phorase without decreasing either NADPH:cytochrome c reduc
capable of consuming MC to generate a reactive species. Exten tase activity or the generation of reactive metabolites. This
sions of these findings to other cell lines demonstrated that MC finding indicates that DT-diaphorase is not involved in the bioac
is essentially equitoxic to aerobic and hypoxic CHO cells under tivation of MC. Indeed, the reactive product(s) generated from
the experimental conditions used in these studies, whereas it is MC increased significantly in the presence of dicumarol, sug
preferentially cytotoxic to V79 cells under hypoxic conditions. In gesting that DT-diaphorase may metabolize MC to a nontoxic
addition, sonicates of EMT6, V79, and CHO cells are capable of substance, possibly by the pathways reported by Thor eÃ-al. (30)
generating reactive species from MC, with the greatest alkylating for the metabolism of menadione in rat liver hepatocytes. These
activity occurring in EMT6 sonicates (Table 1). NADPH is more results suggest that treatment of whole cells with both MC and
efficient than NADH as an electron donor for the generation of dicumarol in combination might be used to increase the toxicity
the reaction product(s). These findings are in agreement with of MC to hypoxic cells. In addition, they imply that the activation
data derived using liver homogenates (12, 28). Enzyme activities of MC under hypoxic conditions may proceed through an inter
characteristic of NADPH:cytochrome c reductase and DT-dia- mediate semiquinone radical, as well as through an intermediate
phorase were present in all 3 cell lines, but cytochrome P-450 hydroquinone (18). The semiquinone radical intermediate has
was undetectable. Both the measured amounts of enzyme activ been suggested by Bachur eÃ-al. (1, 21) and Tomasz eÃ-al. (32).
ity and the rate of formation of reactive metabolites were highest The nonspecific NADPHxytochrome c reductase inhibitors,
in the EMT6 cells. However, neither the measured oxidoreduc- NADP+ and mersalyl, also yielded interesting results. NADP+
tase activities (Table 2) nor the rate of formation of reactive completely inhibited NADPHxytochrome c activity but had less
metabolites (Table 1) in the different cell lines con-elated with the effect on the generation of reactive metabolites from MC. Con
extent of cytotoxicity under hypoxic conditions. These findings versely, mersalyl at 0.25 mw inhibited the generation of reactive
do not preclude bioactivation of MC as a critical event responsible metabolites of MC to a greater extent than it inhibited
for cytotoxicity, because studies with sonicates cannot include NADPHxytochrome c reductase activity. The inhibition of both
the effects of such factors as the rate of transport of drug into alkylating activity and NADPHxytochrome c reductase activity
cells, the extent of repair of cytotoxic lesions, and changes in by NADP+ and mersalyl implies that NADPHxytochrome c re
intracellular pH. However, the lack of a correlation between the ductase is responsible for at least some of the bioactivation of
rate at which sonicated cells generate reactive species and the MC in EMT6 cells, but the different degrees of inhibition of these
cytotoxicity of MC to intact cells indicates either that interaction processes suggest that at least one other enzyme may be
between activated MC and 4-(p-nitrobenzyl)pyridine does not involved.
reflect accurately the reactions leading to cytotoxicity or that the In conclusion, the data in this paper demonstrate that, under
rate of bioactivation of MC is not the sole determinant of cyto hypoxic conditions, MC is differentially toxic to EMT6 and V79
toxicity. cells but has less differential toxicity to CHO cells, and that all 3
In assessing the involvement of mammalian cell enzymes in cell types can generate an electrophile from MC which can be
the activation of MC, cytochrome P-450 did not appear to be a measured with 4-(p-nitrobenzyl)pyridine. Both NADPHxyto
primary reductant. This enzyme was not present in any of the chrome c reductase and DT-diaphorase were present in all 3 cell
cell lines at levels within the limits of detectability (<0.5 pmol/ lines. Although there was a qualitative relationship between
mg). Moreover, CO neither inhibited the formation of MC-derived enzyme activity and the generation of reactive metabolites from
reactive metabolites by either cell sonicates or purified MC, these parameters did not correlate quantitatively with the
NADPHxytochrome c reductase and P-450 (Chart 2) nor pro extent of MC-induced cytotoxicity to hypoxic cells. Experiments
tected EMT6 cells from MC-induced cytotoxicity (Chart 4). with enzyme inhibitors suggested that DT-diaphorase and cyto
When purified enzymes were used, the combination of ana- chrome P-450 are not involved in the bioactivation of MC,
erobiosis, NADPH:cytochrome c reductase, and NADPH was although they may modulate its activation. On the other hand,
sufficient for MC activation; however, addition of P-450 increased NADPHxytochrome c reductase was capable of activating MC
the reaction rate (Chart 1). The 100-fold difference between the but did not appear to be the sole enzyme involved in the reductive
rate of P-450 reoxidation and the rate of MC disappearance activation of the antibiotic.
(Chart 2) indicates, however, that P-450 does not participate
directly in the metabolism of MC to either a toxic or a nontoxic ACKNOWLEDGMENTS
metabolite. Instead, P-450 appears to modulate NADPH:
cytochrome c reductase activity by increasing the efficiency of The authors would like to thank Carmen Arroyo for her excellent technical
assistance.
electron transfer from NADPH :cytochrome c reductase to MC.
Potter and Reed (22) reported recently a similar phenomenon for
the anaerobic reduction of 1-(2-chloroethyl)-3-(cyclohexyl)-1-ni- REFERENCES
trosourea.
1. Andrews, P. A., Pan, S., Glover, C. J., and Bachur, N. R. Electrochemical
EMT6 cells contained more NADPHxytochrome c reductase reduction of mitomycin C to alkylating intermediates. Proc. Am. Assoc. Cancer.
and DT-diaphorase activity than CHO and V79 cells; EMT6 cells Res., 24:246,1983.
2. Carter, S. K. Mitomycin C (NSC-26980)—clinicalbrochure. CancerChemother.
also formed reactive metabolites from MC under anaerobic con Rep. Part III, 1:99-144,1968.
ditions faster than did either CHO or V79 cells. To determine the 3. Cinti, D. L., Moldeus. P., and Schenkman, J. B. Kinetic parameters of drug-

CANCER RESEARCH VOL. 44 DECEMBER 1984


5642

Downloaded from cancerres.aacrjournals.org on April 10, 2019. © 1984 American Association for Cancer Research.
BIOTRANSFORMATION OF MITOMYCIN C

metabolizing enzymes in Ca2* -sedimented microsomes from rat liver. Biochem. 20. Omura, T., and Sato, R. The carbon monoxide-bindingpigment of liver micro
Pharmacol., 21: 3249-3256,1972. somes. I. Evidence for its hemoprotein nature. J. Biol. Chem., 239: 2370-
4. Coon, M. S., van der Hoeven, T. A., Dahl, S. B., and Haugen, D. A. Two forms 2378,1964.
of liver microsomal cytochrome P-450. P-450LM2 and P-450tM4 (rabbit liver). 21. Pan, S.-S., Andrews, P. A., Glover, C. J., and Bachur. N. R. Reductive
Methods Enzymol., 52:109-117,1978. activation of mitomycin C and mitomycin C metabolites catalyzed by NADPH-
5. Crooke, S. T., and Bradner, W. T. Mitomycm C: a review. Cancer Treat. Rev., cytochrome P-450 reducÃ-aseand xanthine oxidase. J. Biol. Chem., 259:959-
3:121-139,1976. 966,1984.
6. Ernster, L. DT-diaphorase.Methods Enzymol., 10:309-317,1967. 22. Poner, D. W., and Reed, D. J. Involvement of FMN and phénobarbital cyto
7. French, J. S., and Coon, M. J. Properties of NADPH-cytochrome P-450 chrome P-450 in stimulating a one-electron reductive demtrosation of i-(2-
reducÃ-ase purified from rabbit liver microsomes. Arch. Biochem. Biophys., 795: chloroethyl)-3-(c>c!ohexyl)-1-nitrosoureacatalyzed by NADPH-cytochrome P-
565-577,1979. 450 reducÃ-ase. J. Biol. Chem., 258: 6906-6911,1983.
8. Henry, R. J., Chiamori, N., Golub, O. J., and Berkman, S. Revisedspectropho- 23. Rauth, A. M., Mohindra, J. K., and Tannock, I. F. Activity of mitomycin C for
tometric methods for the determination of glutamic-oxalacetic transaminase, aerobic and hypoxic cells in vitro and in vivo. Cancer Res., 43: 4154-4158,
glutarnic-pyruvic transaminase, and lactic acid dehydrogenase. Am. J. Clin. 1983.
Pathol., 34:381-398,1960. 24. Rockwell, S. In vivo-in vitro tumors systems: new models for studying the
response of tumors to therapy. Lab. Anim. Sci. 27 (Part 2).-831-851,1977.
9. Iyer, V. N., and Szybalski, W. A molecular mechanism of mitomyoin action:
linking of complementary DNA strands. Proc. Nati. Acad Sci. USA, 50: 355- 25. Rockwell, S. Effects of mitomycin C alone and in combination with X-rays on
EMT6 mouse mammary tumors in vivo. J. Nati. Cancer Inst., 71: 765-771,
362,1963.
10. Kapp, D. S., and Hahn, G. M. Thermosensitization by sulfhydryl compounds 1983.
of exponentially growing Chinese hamster cells. Cancer Res., 39:4630-4635, 26. Rockwell, S., and Kennedy, K. A. Combination therapy with radiation and
1979. mitomycin C: preliminary results with EMT6 tumor cells in vitro and in vivo.
Int. J. Radiât.Oncol. Biol. Phys., 5:1673-1676,1979.
11. Kennedy, K. A., Rockwell, S., and Sartorelli, A. C. A preferential activation of
mitomyan C to cytotoxic metabolites by hypoxic tumor cells. Cancer Res., 40: 27. Rockwell, S., Kennedy, K. A., and Sartorelli, A. C. Mitomycin C as a prototype
2356-2360,1980. bioreductive alkylating agent: in vitro studies of metabolism and cytotoxicity
Int. J. Radiât.Oncol. Biol. Phys., 8:753-755,1982.
12. Kennedy, K. A., Sugar, S. G., Polomski, L., and Sartorelli, A. C. Metabolic
28. Schwartz, H. S. Pharmacologyof mitomycin C. III. in vitro metabolism by rat
activation of mitomycin C by liver microsomes and nuclei. Biochem. Pharma liver. J. Pharmacol.Exp. Ther., 736:250-258,1962.
col., 37: 2011-2016,1982. 29. Sottocasa, G. L., Kuylenstiema,T., Ernster, L., and Bergstand, A. An electron-
13. Kennedy, K. A., Teicher, B. A., Rockwell, S., and Sartorelli, A. C. The hypoxic transport system associated with the outer membraneof liver mitochondria.A
tumor cell: a target for selective cancer chemotherapy. Biochem. Pharmacol., biochemicaland morphologicalstudy. J. Cell Biol., 32: 415-438,1967.
29: 1-8, 1980.
30. Thor, H., Smith, M. T., Hartzell, P., Bellomo, G., Jewell, S. A., and Orrenius,
14. Komiyama,T., Oki, T., and Inui, T. Activation of mitomycin C and quinone drug S. The metabolism of menadione (2-methyl-1,4-naphthoquinone)by isolated
metabolism by NADPH-cytochromeP-450 reducÃ-ase. J. Pharm. Dyn., 2:407-
hepatocytes. A study of the implications of oxidative stress in intact cells. J.
410,1979. Bid. Chem., 257:12419-12425,1982.
15. Laemmli.U. K. Cleavageof structural proteins during the assembly of the head 31. Tomasz, M., and LJpman,R. Reductive metabolism and alkylating activity of
of bactenophage T4. Nature (Lond.), 227: 680-685,1970. mitomycin C induced by rat liver microsomes. Biochemistry, 20: 5056-5061,
16. LJskay, R. M. Genetic analysis of a Chinese hamster cell line lacking a d 1981.
phase. Exp. Cell Res., 114: 69-77,1978. 32. Tomasz, M., Mercado, C. M., Olson, J., and Chatterjie, N. The mode of
17. Lowry, O. H., Rosebrough, N. J., Farr, A. L, and Randall, R. J. Protein interaction of mitomycin C with deoxyribonudeic acid and other poiynudeo-
measurements with the Folin phenol reagent. J. Biol. Chem., 193: 265-275, tides in vitro. Biochemistry, 73:4878-4887,1974.
1951. 33. Wheeler, G. P., Bowdon, B. J., Gremsley,J. A., and Lloyd. H. H. Interrelation
18. Moore, H. W., and Czemiak, R. Naturally-occurring quiñonesas potential ships of some chemical, physicochemicai.and biological activities of several
bioreductive alkylating agents. Med. Res. Rev., 1: 249-280,1981. H2-hatoethyl)-1-nitrosoureas. Cancer Res., 34:194-200,1974.
19. Moulder, J. E., and Rockwell, S. Hypoxic fractions of solid tumors: experimental 34. Yasukochi,Y.. and Masters, B. S. S. Someproperties of a detergent-solubilized
techniques, methods of analysis, and a survey of existing data. Int. J. Radiât. NADPH-cytochrome c (cytochrome P-450) reducÃ-asepurified by btospecific
Oncol. Biol. Phys., 10: 695-712,1984. affinity chromatography. J. Biol. Chem., 257: 5337-5344,1976.

CANCER RESEARCH VOL. 44 DECEMBER 1984


5643

Downloaded from cancerres.aacrjournals.org on April 10, 2019. © 1984 American Association for Cancer Research.
Role of NADPH:Cytochrome c Reductase and DT-Diaphorase in
the Biotransformation of Mitomycin C
Susan R. Keyes, Paula M. Fracasso, David C. Heimbrook, et al.

Cancer Res 1984;44:5638-5643.

Updated version Access the most recent version of this article at:
http://cancerres.aacrjournals.org/content/44/12_Part_1/5638

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

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Subscriptions Department at pubs@aacr.org.

Permissions To request permission to re-use all or part of this article, use this link
http://cancerres.aacrjournals.org/content/44/12_Part_1/5638.
Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)
Rightslink site.

Downloaded from cancerres.aacrjournals.org on April 10, 2019. © 1984 American Association for Cancer Research.

You might also like