Sunteți pe pagina 1din 7

Jpn J Clin Oncol 2004;34(7)379–385

Original Articles

Evaluation of Oxidative Stress and Nitric Oxide Levels in Patients


with Oral Cavity Cancer
S. Syed Sultan Beevi1, A. Muzib Hassanal Rasheed2 and A. Geetha3
1Department of Physiology and Biochemistry, Ragas Dental College and Hospital, Chennai, Tamil Nadu, India,
2Department of Surgical Oncology, Government Royapettah Hospital, Chennai, Tamil Nadu, India and 3Department
of Biochemistry, Bharathi Womens’ College, Chennai, Tamil Nadu, India
Received October 13, 2003; accepted March 31, 2004

Objective: The aim of this study was to evaluate the magnitude of oxidative stress and levels

Downloaded from http://jjco.oxfordjournals.org/ by guest on December 26, 2015


of nitric oxide in patients with oral cavity cancer by analyzing the levels of lipid peroxidation
products, antioxidants and nitric oxide products.
Methods: This prospective study was conducted on 15 patients with biopsy proven squamous
cell cancer of the oral cavity with clinical stage III/IV and an equal number of age and sex
matched healthy subjects. The levels of lipid peroxidation products, antioxidants and nitric
oxide products were determined by colorimetric methods.
Results: Lipid peroxidation products like lipid hydroperoxide (LHP) and malondialdehyde
(MDA) and nitric oxide products like nitrite (NO2–), nitrate (NO3–) and total nitrite (TNO2–) were
significantly elevated, whereas enzymatic and non-enzymatic antioxidants were significantly
lowered in oral cavity cancer patients when compared to normal healthy subjects.
Conclusions: Enhanced lipid peroxidation with concomitant decrease in antioxidants is indic-
ative of oxidative stress that provides evidence of the relationship between lipid peroxidation
and oral cavity cancer. Increased nitric oxide production represents a general mechanism in its
pathogenesis.

Key words: oral cavity cancer – nitric oxide – lipid peroxidation – antioxidants

INTRODUCTION It should be noted, however, that the development of human


cancer is multifactorial, depending on several factors including
Oral cavity cancer is an important cancer globally and is one of
the extent of DNA damage, effectiveness of antioxidant
the ten most frequent cancers worldwide (1). Tobacco is the
primary etiological factor in its development, other factors defense, DNA repair system and growth promoting effect of
being alcohol, genetic predisposition and a diet lacking in ROS (3).
micronutrients. The burst of ROS has been implicated in the development
The proposal that reactive oxygen species (ROS) such as of oral cavity cancer in tobacco chewers and smokers (4).
superoxide radicals (O2•), hydroxyl radicals (OH•) and hydro- Tobacco consumption in any form has been demonstrated to
gen peroxide (H2O2) play a key role in human cancer develop- have carcinogenic, teratogenic and genotoxic effects and is
ment has gained much support recently. They have been shown positively correlated with accumulation of DNA damage.
to possess several characteristics of carcinogens (2). ROS can Tobacco is therefore believed to directly induce cellular DNA
cause DNA base alterations, strand breaks, damage to tumor damages in the human oral cavity (5). Oxidative modification
suppressor genes and enhanced expression of protooncogenes of nucleic acids by ROS could result in the transformation of
(2). ROS-induced mutation could also arise from protein dam- normal cells into malignant cells (6). ROS induced lipid perox-
age and attack on lipids, which then initiate lipid peroxidation. idation has been implicated in malignant transformation (7).
The prime targets of peroxidation by ROS are the polyunsatu-
rated fatty acids (PUFA) in the membrane lipids. Furthermore,
the decomposition of these peroxidized lipids yields a variety
For reprints and all correspondence: S. Syed Sultan Beevi, Department of
Physiology and Biochemistry, Ragas Dental College and Hospital, 2/102, East of end products, including lipid hydroperoxides (LHP) and
Coast Road, Chennai, Tamil Nadu, India. E-mail: fathuus@rediffmail.com malondialdehyde (MDA). The levels of these lipid peroxides

© 2004 Foundation for Promotion of Cancer Research


380 Oxidative stress in oral cancer

indicate the extent of lipid peroxidation in general and serve as Table 1. Clinical characteristics of patients
markers of cellular damages due to free radicals.
Enzymatic and non-enzymatic antioxidant defense systems Characteristics No. of patients
include superoxide dismutase (SOD), catalase (CAT), gluta- Patients 15
thione peroxidase (GPx), reduced glutathione (GSH), vitamins Male/Female 12/3
E, C and A and β carotene. They protect cells against ROS pro-
Age (years) (range) 56.2 (33–72)
duced during normal metabolism and after an oxidative insult.
Stage
Antioxidant defense systems work cooperatively to alleviate
the oxidative stress caused by enhanced free radical produc- III 2
tion. Any changes in one of these systems may break this IV 13
equilibrium and cause cellular damages and ultimately malig- Grade
nant transformation. I (well differentiated SCC) 15
Nitric oxide (NO•) is a free radical, an uncharged molecule
Tumor location
with an unpaired electron. NO• plays multiple roles in both
Anterior 2/3rd of the tongue 4
intracellular and extracellular signaling mechanisms (8). This
highly reactive, yet simple molecule is produced in the body by Buccal mucosa 7
the isoenzyme nitric oxide synthase (NOS) using L-arginine as Soft palate 1

Downloaded from http://jjco.oxfordjournals.org/ by guest on December 26, 2015


a substrate. Three isoforms of NOS have been characterized. Alveolus 2
Two of them are constitutive NOS (cNOS) and the third is Retromolar area 1
inducible (iNOS) by endotoxins and cytokines (9).
Reaction of NO• with oxygen or other free radicals generates
reactive nitrogen species (RNS), which cause multiple bio- of the oral cavity with clinical stage III/IV, registered at the
logical effects (10). NO• is either cytostatic or cytotoxic, inter- Department of Oncology, Government Royapettah Hospital,
acting with a number of molecular targets within cells. Cells Chennai. All the patients included in the investigation were
within different tissues display varying responses to NO•, either smokers or tobacco chewers. The control groups
which may relate to the presence of cellular antioxidants such consisted of 15 age and sex matched non-smoking healthy
as GSH, GPx, CAT and SOD (11). volunteers (12 males, 3 females) from similar socioeconomic
Overexpression of NOS in chronic inflammation can lead to backgrounds. The age range was 33 to 72 years (mean ± SD
genotoxicity. NO• may mediate DNA damage through the for- 56.2 ± 11.8 years) for both patients and controls. The clinical
mation of carcinogenic nitrosamines, generation of RNS and characteristics of patients (sex, age, disease localization and
inhibition of DNA damage repair mechanism. It can thus be stage) are shown in Table 1. Tumors were classified according
considered as a tumor initiating agent (12). However, NO• may to the UICC criteria (13). Two patients were in stage III and 13
also have an impact on other stages of cancer development. patients in stage IV. All the carcinomas were graded as well-
These effects of NO• are broad, with its involvement ranging differentiated squamous cell carcinoma. The ethical committee
from cellular transformation and formation of neoplastic of the institution approved the study, and informed consent of
lesions to the regulation of various other aspects of tumor all the subjects was obtained. None of the patients and control
biology (11). subjects had concomitant diseases such as diabetes mellitus,
NO• plays an important role in host defense and homeostasis liver disease and rheumatoid arthritis. All the patients and the
when generated at a low level for a brief period of time, but control subjects included in this study were not on any medical
becomes genotoxic and mutagenic when generated at higher treatment including supplementation of antioxidants. All the
concentrations for prolonged periods of time. Thus, the bio- subjects underwent a thorough dental checkup. The subjects
logical outcome of the NO• mediated effects is complex and who had poor oral hygiene had dental scaling, removal of
depends on the internal and external environment of the target caries-affected teeth and were treated with antibiotics and
and generation sites of the cells as well as the concentration of mouth cleaning agents for a period of one week. In such cases,
NO• generated. blood samples were collected 15 days after the completion of
The aim of this study was to evaluate the extent of oxidative the dental treatment.
stress and the levels of nitric oxide in oral cavity cancer
patients by analyzing the levels of lipid peroxidation products, COLLECTION OF SAMPLES
antioxidants and nitric oxide products.
Taking aseptic precautions, blood samples (approximately 10
ml) were collected in appropriate sterile vials by venous arm
SUBJECTS AND METHODS puncture after overnight fasting. Five milliliters of blood was
collected with EDTA as an anticoagulant for erythrocyte
PATIENTS
preparation and plasma. Another 5 ml of blood was collected
This prospective study was conducted on 15 patients (12 without anticoagulant for the separation of serum. Plasma
males, 3 females) with biopsy proven squamous cell carcinoma and sera were separated by centrifugation at 1000 g for 15 min.
Jpn J Clin Oncol 2004;34(7) 381

Table 2. Levels (µmol/ml) of LHP and (nmol/ml) of MDA in plasma of oral Table 3. Activities (units/100 mg protein) of SOD, (units/mg protein) of GPx
cancer patients and control subjects (mean ± SD) and CAT in erythrocytes of oral cancer patients and control subjects (mean ±
SD)
LHP MDA
SOD GPx CAT
Control group (n = 15) 290.57 ± 48.70 2.02 ± 0.23
Control group (n = 15) 21.35 ± 2.80 13.80 ± 1.22 33.63 ± 2.59
Patient group (n = 15) 496.29 ± 19.51* 5.57 ± 0.97*
Patient group (n = 15) 10.07 ± 2.93* 3.33 ± 0.80* 14.44 ± 1.63*
P value 0.0000 0.0000
P value 0.0000 0.0000 0.0000
*P < 0.001, when compared with control group
*P < 0.001, when compared with control group

After the separation of plasma, the buffy coat was removed


and the packed cells were washed thrice with 0.89% saline. A 5,5′,dithiobisnitrobenzoic acid to form a complex that absorbs
known volume of erythrocytes was lysed with deionized water. at 412 nm. The results were expressed as mg/dl plasma.
The hemolysate was separated by centrifugation at 2500 g for Vitamin E was estimated in serum by the method of Baker
15 min at 2 °C. Biochemical estimations were carried out im- and Frank (20), which is based on the reduction of ferric to
mediately. ferrous ions by tocopherol, which then forms a red colored

Downloaded from http://jjco.oxfordjournals.org/ by guest on December 26, 2015


A portion of the oral mucosa was excised from oral cancer complex with 2,2′,bipyridyl that is read at 520 nm. The level
cavity patients by punch biopsy and washed immediately in of vitamin E was expressed as mg/l serum.
physiological saline. Tissues were preserved in 0.9% formalin Vitamin C in plasma was analyzed by the method of Jacob
saline. Thin tissue sections were fixed and stained with eosin– (21). This method is based on the oxidation of ascorbic acid to
hemotoxylin mixture. The stained tissue smear was observed dehydroascorbic acid, which when heated with dinitrophenyl-
through medical microscope under 100× magnification. hydrazine forms a colored complex with absorption maxima at
520 nm. The results were expressed as mg/dl plasma.
BIOCHEMICAL MEASUREMENTS Serum nitric oxide was measured in terms of its products,
The estimation of MDA in plasma was done by the method of nitrite and nitrate, by the method of Griess modified by Fiddler
Draper and Hadley (14). The color produced by the reaction of (22). This method is based on a two-step process. The first step
thiobarbituric acid with MDA was measured colorimetrically is the conversion of nitrate to nitrite using tin metal powder and
at 533 nm. The results were expressed as nmoles/ml plasma. the second is the addition of sulphanilamide and N (-naphthyl)
LHP in plasma was estimated by the method of Jiang et al. ethylenediamine (Griess reagent). This converts nitrite into a
(15). This method is based on the ability of H2O2 to oxidize deep purple azo compound, which was measured colorimetri-
ferrous ion under acidic condition in the presence of xylenol cally at 540 nm. Nitric oxide products were expressed as
orange. The resultant chromophore was measured colorimetri- µmoles/dl serum.
cally at 560 nm. The LHP level was expressed as µmoles/ml Protein in RBC hemolysate was estimated by the method of
plasma. Biuret (23). This method is based on the reaction of Biuret rea-
SOD was assayed in RBC hemolysate by the method of gent with peptide bonds of proteins. The violet color produced
Misra and Fridovich (16). This method is based on the ability was measured at 540 nm.
of SOD to inhibit autoxidation of epinephrine under specific
conditions. One unit of SOD is defined as the amount of STATISTICAL ANALYSIS
enzyme required to cause 50% inhibition of epinephrine
autoxidation. Enzyme activity was reported as units/100 mg The findings were expressed as the mean ± standard deviation.
protein in erythrocyte lysate. The data were analyzed with Student’s independent t test. All
CAT was assayed in RBC hemolysate by the method of statistical analyses were performed with the program Statisti-
Clairborne (17). The color produced by the reaction of H2O2 cal Package for the Social Science (SPSS for Windows, Ver-
with dichromate in acetic acid was measured colorimetrically sion 10.0). A P value of <0.05 was accepted as statistically
at 620 nm. Enzyme activity was expressed as µmoles of H2O2 significant.
decomposed/min/mg protein.
Glutathione peroxidase (GPx) was assayed in RBC hemo-
RESULTS
lysate by the method of Flohe and Gunzler (18) and based on
the reaction of the residual GSH with 5,5′,dithiobisnitroben- Our findings on the assessed parameters in oral cavity cancer
zoic acid to form a complex that absorbs at 412 nm. Enzyme patients and healthy control subjects are shown in Tables 2, 3,
activity was expressed as nmoles of GSH oxidized/min/mg 4 and 5.
protein. Table 2 shows the levels of plasma LHP and MDA in control
GSH was estimated in plasma by the method of Thomas and subjects and oral cavity cancer patients. The extent of lipid
Skrinska (19), which is based on the reaction of GSH with peroxidation as evidenced by plasma LHP and MDA was
382 Oxidative stress in oral cancer

Table 4. Levels (mg/dl) of GSH and vitamin C in plasma and (mg/l) of


vitamin E in serum of oral cancer patients and control subjects (mean ± SD)

GSH Vitamin C Vitamin E


Control group (n = 15) 10.02 ± 0.55 1.80 ± 0.32 7.71 ± 0.81
Patient group (n = 15) 3.09 ± 0.53* 0.51 ± 0.09* 1.40 ± 0.32*
P value 0.0000 0.0000 0.0000

*P < 0.001, when compared with control group

Table 5. Levels (µmol/dl) of NO2–, NO3– and TNO• in serum of oral cancer
patients and control subjects (mean ± SD)

NO2– NO3– TNO•


Control group (n = 15) 0.25 ± 0.04 0.27 ± 0.09 0.52 ± 0.13
Patient group (n = 15) 0.50 ± 0.12* 0.47 ± 0.11* 0.97 ± 0.23* Figure 2. Photomicrograph of the oral mucosa from oral cavity cancer patient
shows well-marked inflammatory regions, characterized by accumulation of

Downloaded from http://jjco.oxfordjournals.org/ by guest on December 26, 2015


P value 0.0000001 0.000007 0.000007 lymphocytes.

*P < 0.001, when compared with control group


The levels were found to be significantly elevated in the oral
significantly increased in the oral cavity cancer patients cancer patients group (P < 0.001), as compared to the control
(P < 0.001), compared to control subjects. subjects.
The enzymatic antioxidants profile in the circulation of oral Histopathology reports are shown in Figs 1, 2 and 3. Figure
cavity cancer patients and control subjects is presented in Table 1 shows oral mucosa from a control subject, which reveals
3. A decrease in the activities of SOD, GPx and CAT in the stratified squamous epithelium with normal cellular architec-
erythrocyte lysate was seen in the oral cavity cancer patients as ture. Histopathological studies of the oral mucosa excised from
compared to control subjects. The difference approached a the oral cavity cancer patients show the presence of well-
statistical significance of P < 0.001. marked inflammatory regions, characterized by the accumula-
Table 4 presents the non-enzymatic antioxidants in the con- tion of lymphocytes (Fig. 2) and several areas of necrosis (Fig.
trol subjects and oral cavity cancer patients. The levels of GSH 3) in the squamous epithelium.
and vitamin C in plasma and vitamin E in serum were signifi-
cantly lower in the oral cavity cancer patients (P < 0.001), when
compared to the control subjects.
DISCUSSION
Table 5 shows the levels of NO• in terms of NO2–, NO3– and Cancer is essentially an event occurring at the gene level and
TNO• in the control subjects and oral cavity cancer patients. the ultimate step resulting in carcinogenesis is DNA damage.
Multiple factors such as viruses, chemicals, irradiation and the

Figure 1. Photomicrograph of the oral mucosa from control shows stratified Figure 3. Photomicrograph of the oral mucosa from oral cavity cancer patient
squamous epithelium with normal cellular architecture. shows several areas of necrosis in the squamous epithelium.
Jpn J Clin Oncol 2004;34(7) 383

genetic makeup of the individual play a role in carcinogenesis. H2O2 and LHP using GSH. This prevents H2O2 mediated intra-
ROS and RNS are two important agents of DNA damage. cellular DNA damage, which is thought to be a prerequisite for
Damage to the DNA by ROS/RNS is believed to occur natu- carcinogenesis (33). Oxidative damage to the cell membrane
rally as well, since low steady-state levels of base damage has been reported to inactivate GPx (34). SOD metabolizes
products have been detected in human nuclear DNA. The free radicals and dismutates superoxide anions (O2•) to H2O2
magnitude of this damage (called oxidative stress or oxidative and protects the cells against O2• mediated lipid peroxidation.
damage) depends not only on ROS/RNS levels but also on the CAT acts on H2O2 by decomposing it, thereby neutralizing its
body’s defense mechanisms against them mediated by various toxicity. It has been reported that superoxide radicals inhibit
cellular antioxidants. Disruption of this delicate oxidant/anti- catalase activity and H2O2 suppresses SOD activity in the cell
oxidant balance in the body seems to play a causative role in (35). Gupta et al. (36) demonstrated that reduction in several
carcinogenesis. antioxidant defense mechanisms correlates with the emergence
There are increasing evidences to support the role of oxida- of the malignant phenotype. The low activities of these antioxi-
tive stress in several human pathological conditions such as dant enzymes observed in our study might be due to the deple-
rheumatoid arthritis, ischemic heart disease, several auto- tion of the antioxidant defense system. This could occur as a
immune disorders and cancer. consequence of overwhelming free radicals, as evidenced by
ROS/RNS are found to be involved in both initiation and pro- the elevated levels of lipid peroxides in the circulation of oral
motion of multistep carcinogenesis. They can cause DNA cavity cancer patients. Reports on CAT activity in cancer are

Downloaded from http://jjco.oxfordjournals.org/ by guest on December 26, 2015


damage, activate procarcinogens, initiate lipid peroxidation, contradictory. Both increase (37) and decrease (28,29,38) in
inactivate enzyme systems and alter the cellular antioxidant CAT activity have been reported previously. Reduction in CAT
defense system (24). activity as observed in this study might be due to increased
High levels of oxidative stress result in peroxidation of mem- endogenous production of the superoxide anion, as evidenced
brane lipids with the generation of peroxides that can decom- by increased LHP and MDA, or increased nitric oxide end
pose to multiple mutagenic carbonyl products. LHP and MDA products, or decreased activity of GPx and SOD or all of these
are well-characterized lipid peroxidation end products. They factors. Furthermore, it might also be due to a higher magni-
are considered to be mutagenic and carcinogenic (25). They tude of oxidative stress, since all our patients were in advanced
can also modulate the expression of genes related to tumor pro- clinical stages (stage III/IV) with a large tumor burden.
motion (26). The level of LHP and MDA reflect the extent of A strong synergism exists between GSH, vitamin E and
lipid peroxidation. vitamin C. Vitamin E is an important antioxidant in the
In our study, we observed increased systemic levels of LHP lipid domain. It is present in both erythrocyte membrane
and MDA in patients with oral cavity cancer, which could be and plasma. It is readily exchanged between erythrocytes and
attributed to increased formation or inadequate clearance of plasma, with a balance in favor of plasma. Vitamin C is an
free radicals by the cellular antioxidants. Elevated levels of important extracellular antioxidant that disappears faster than
lipid peroxidation products support the hypothesis that the other antioxidants when plasma is exposed to oxygen free
cancer cells produce large amount of free radicals (2) and that radicals. Vitamin C spares GSH and together with vitamin
there exists a relationship between free radical activity and E prevents the oxidation of GSH. Since regeneration of both
malignancy (27). Observations similar to our findings have vitamin E and vitamin C requires GSH, diminished level of
been reported in studies on various human cancers (28–30). plasma GSH in oral cavity cancer patients might be responsible
Antioxidants have been shown to inhibit both initiation and for the low levels of these antioxidants.
promotion in carcinogenesis and counteract cell immortaliza- The role of nitric oxide (NO•) is multidimensional. It func-
tion and transformation (31). Activities of different antioxi- tions as an intracellular messenger and is also implicated as a
dants show different patterns during neoplastic transformation deleterious agent in various pathophysiological conditions
and tumor cells exhibit abnormal activities of antioxidant including cancer, inflammatory conditions and autoimmune
enzymes, when compared to their appropriate normal cell diseases. Chronic inflammation can lead to the production of
counterparts (24). NO•, which in turn has the potential to mediate DNA damage
Cellular antioxidant enzymes and free radical scavengers directly, or indirectly through the generation of more persistent
protect a cell against toxic oxygen radicals. GSH, an important RNS (12).
non-protein thiol, in conjugation with glutathione transferase Serum levels of nitrite (NO2–) and nitrate (NO3–) are used to
(GST) and GPx, plays a significant role in protecting cells by estimate the level of NO• formation, since NO• is highly unsta-
scavenging ROS (32). A significant depletion of plasma GSH ble and has a very short half-life. Evidence of the role of NO•
observed in our study reflects enhanced pro-oxidant milieu of in carcinogenesis is provided by the fact that both cNOS and
the cells and correlates with the increased lipid peroxides in the iNOS are detected in various human cancers (39,40). Biopsy
circulation of oral cavity cancer patients. samples of human breast cancer show the presence of greater
Antioxidant enzymes such as SOD, CAT and GPx provide expression of iNOS in a high-grade tumor, which tends to be
the first line of cellular defense against toxic free radicals. more invasive (9). Our study is the first to undertake the evalu-
These enzymes react directly with oxygen free radicals to yield ation of serum nitric oxide levels in human oral cavity cancer.
non-radical products. Selenium dependent GPx removes both We observed significantly higher NO• end products in the
384 Oxidative stress in oral cancer

serum of oral cavity cancer patients. This could result from a 10. Patel RP, McAndrew J, Sellak H, White RC, Jo H, Freeman BA, et al. Bio-
logical aspects of reactive nitrogen species. Biochim Biophys Acta 1999;
generalized increased NO• synthesis throughout the body of 1411:385–400.
the cancer patient or reflect increased NO• degradation pro- 11. Hogg N, Singh RJ, Kalyanaraman B. The role of glutathione in the trans-
moted by oxidative stress. Observations similar to ours have port and catabolism of nitric oxide. FEBS Lett 1996;382:223–8.
12. Li RH, Hotchkiss JH. Potential genotoxicity of chronically elevated nitric
been reported previously in gastric, colorectal, hepatocellular oxide: a review. Mutat Res 1995;339:73–89.
and breast cancer (41–44). 13. Sobin LH, Wittekind CH. Head and neck tumors – lips and oral cavity. In:
Histopathological examinations of the oral mucosa revealed Wittekind CH, editor. TNM classification of malignant tumours, 5th ed.
International Union Against Cancer. New York: Wiley-Liss 1997:59–62.
necrosis and inflammatory changes. The oxidative stress might 14. Draper HH, Hadley M. Malondialdehyde determination as an index of
have resulted in the abnormal changes evidenced in the histo- lipid peroxidation. Methods Enzymol 1990;186:421–31.
logical examinations. Lipid peroxidation and oxidative stress 15. Jiang ZY, Hunt JV, Wolff SP. Ferrous ion oxidation in the presence of
xylenol orange for detection of lipid hydroperoxide in low-density lipo-
have been reported to be among the causes of tissue damage protein. Anal Biochem 1992;202:384–9.
and inflammation (30). 16. Misra HP, Fridovich I. The role of superoxide anion in the autoxidation of
From this study, it could be concluded that oxidative stress is epinephrine and a simple assay for superoxide dismutase. Biol Chem
1972;247:3170–5.
increased (as evidenced by elevated levels of lipid peroxidation 17. Clairborne A. Catalase activity. In: Greenwald, editor. CRS Handbook of
products and nitric oxide products) and antioxidant defenses methods in oxygen radical research, CRS Press, Boca Ratari 1985;283–4.
are compromised (as evidenced by depletion of enzymatic and 18. Flohe L, Gunzler WA. Assay of glutathione peroxidase. Methods Enzymol
1984;105:114–21.

Downloaded from http://jjco.oxfordjournals.org/ by guest on December 26, 2015


non-enzymatic antioxidants) in patients with oral cavity can- 19. Thomas G, Skrinska V. Determination of glutathione in human platelets.
cer. A weak antioxidant defense system makes the mucosal Clin Chem 1985;31:350–1.
cells more vulnerable to the genotoxic effect of ROS. This 20. Baker H, Frank O. Tocopherol. In: Clinical vitaminology, methods and
interpretation, New York Inter Science Publisher, John Wiley and Sons,
creates an intracellular environment more favorable for DNA Inc. 1968;172–3.
damage and disease progression. Studies have shown that a 21. Jacob RA. Assessment of human vitamin C status. J Nutr 1990;120:1480–
diet rich in antioxidants may alleviate oxidative stress to some 5.
22. Fiddler KM. Collaborative study of modified AOAC method of analysis of
extent (45). However, the degree of effectiveness with which nitrite in meat and meat products. J AOAC 1977;60:594–8.
the antioxidant system can be restored with dietary modifica- 23. Weischelbaum TE. Total serum or plasma protein concentration; Biuret
tions and nutritional supplements, so that cancer patients can method. Am J Clin Path 1946;16:40–3.
24. Sun Y. Free radicals, antioxidant enzymes and carcinogenesis. Free
actually be benefited remains to be elucidated. Since the role of Radical Biol Med 1990;8:583–99.
NO• in tumorigenesis is multidimensional, an elaborate study 25. Zhang Y, Chen SY, Hsu T, Santella RM. Immunohistochemical detection
with a larger sample size is needed to ascertain the actual role of malondialdehyde-DNA adducts in human oral mucosa cells. Carcino-
genesis 2002;23:207–11.
of NO• and RNS in the initiation and promotion of carcino- 26. Cerutti PA. Pro-oxidant status and tumor promotion. Science 1985;227:
genesis. 375–81.
27. Dormandy TI. An approach to free radicals. Lancet 1983;1:1010–4.
28. Ray G, Batra S, Shukla NK, Deo S, Raina V, Ashok S, Husain SK. Lipid
Acknowledgment peroxidation, free radical production and antioxidant status in breast can-
cer. Breast Cancer Res Treat 2000;59:163–70.
The authors wish to thank the Pathology Department of Ragas 29. Manju V, Kalaivani Sailaja J, Nalini N. Circulating lipid peroxidation and
antioxidant status in cervical cancer patients: a case-control study. Clin
Dental College and Hospital for evaluation of the histopathol- Biochem 2002;35:621–5.
ogy slides. 30. Geetha A, Karthiga S, Surendran G, Jayalakshmi G. Biochemical studies
on the level of lipid hydroperoxide and antioxidants in different types of
obstructive jaundice. J Lab Med 2001;2:20–7.
References 31. Oberley LW, Oberley TD. Role of antioxidant enzymes in cell immortali-
zation and transformation. Mol Cell Biochem 1988;84:147–53.
1. Parkin DM, Pisani P, Ferlay J. Estimates of the worldwide incidence of 32. Meister A. Glutathione metabolism and its selective modification. J Bio-
eighteen major cancers in 1985. Int J Cancer 1993;54:594–605. chem 1988;263:17205–8.
2. Cerutti PA. Oxy-radicals and cancer. Lancet 1994;344:862–3. 33. Mates JM, Perez-Gomez C, Nunez de Castro I. Antioxidant enzymes and
3. Burdon RH. Superoxide and hydrogen peroxide in relation to mammalian human disease. Clin Biochem 1999;32:595–603.
cell proliferation. Free Radic Biol Med 1995;18:775–94. 34. Stohs SJ, Al-Bayati ZF, Hassan MQ, Murray WJ, Mohammedpeur HA.
Glutathione peroxidase and reactive oxygen species in TCDD-induced
4. Stitch HF, Anders F. The involvement of reactive oxygen species in oral
lipid peroxidation. Adv Exp Med Biol 1986;197:357–65.
cancer of betel quid-tobacco chewers. Mutation Res 1989;214:47–61.
35. Hassan HM, Fridovich I. Superoxide radical and the oxygen enhancement
5. Singh A, Singh SP. Modulatory potential of smokeless tobacco on the gar- of the toxicity of paraquat in E. coli. J Biol Chem 1978;253:8143–8.
lic, mace or black mustard-altered hepatic detoxication system enzymes
36. Gupta A, Batts B, Kwei KA, Dvorakova K, Stratton SP, Briehl MM, et al.
sulphydryl contents and lipid peroxidation in murine system. Cancer Lett Attenuation of catalase activity in the malignant phenotype plays a func-
1997;118:109–14. tional role in an in vitro model for tumor progression. Cancer Lett 2001;
6. Guyton KZ, Kensler TW. Oxidative mechanisms in carcinogenesis. Br 73:115–25.
Med Bull 1993;49:523–44. 37. Hristozov D, Gadjeva V, Vlaykova T, Dimitrov G. Evaluation of oxidative
7. Gonzalez RA. Free radicals, oxidative stress and DNA metabolism in stress in patients with cancer. Arch Physiol Biochem 2001;109:331–6.
human cancer. Cancer Invest 1999;17:376–7. 38. Kumaraguruparan R, Subapriya R, Kabalimoorthy J, Nagini S. Antioxi-
8. Moncada S, Palmer RM, Higgs E. Nitric oxide: physiology, pathophysiol- dant profile in the circulation of patients with fibroadenoma and adenocar-
ogy and pharmacology. Pharmacol Rev 1991;43:109–42. cinoma of the breast. Clin Biochem 2002;35:275–9.
9. Jenkin DC, Charles IG, Thomson LL, Moss DW, Holmes LS, Baylis SA, 39. Thomson LL, Miles DW, Happerfield L, Bobrow LG, Knowles RG,
et al. Role of nitric oxide in tumor growth. Proc Natl Acad Sci USA 1995; Moncada S. Nitric oxide synthase activity in human breast cancer. Br J
92:4392–6. Cancer 1995;72:41–4.
Jpn J Clin Oncol 2004;34(7) 385

40. Prazma J, Pctrusz P, Mims W, Ball SS, Weissler MC. Immunohistochem- 43. Moriyama A, Tabaru A, Unoki H, Abe S, Masumota A, Otsuki M. Plasma
ical characterization of nitric oxide synthase in squamous cell carcinoma nitrite/nitrate concentrations as a tumor marker for hepatocellular carci-
of the head and neck. Otolaryngol Head Neck Surg 1995;113:541–9. noma. Clin Chim Acta 2000;296:181–91.
41. Bakan E, Taysi S, Polat FM, Dalga S, Umudum Z, Bakan N, Gumus M. 44. Alagol H, Erdem E, Sancak B, Turkmen G, Camlibel M, Bugdayli G.
Nitric oxide levels and lipid peroxidation in plasma of patients with gastric Nitric oxide biosynthesis and malondialdehyde level in advanced breast
cancer. Jpn J Clin Oncol 2002;32:162–6.
cancer. Aust NZ J Surg 1999;69:647–50.
42. Szaleczky E, Pronai L, Nakazawa H, Tulassay Z. Evidence of in vivo per-
oxynitrite formation in patients with colorectal carcinoma, higher plasma 45. Halliwell B. Free radical and antioxidants: a personal view. Nutr Rev
nitrite/nitrate level and lower protection against oxygen free radicals. 1994;52:253–65.
J Clin Gastroenterol 2000;30:47–51.

Downloaded from http://jjco.oxfordjournals.org/ by guest on December 26, 2015

S-ar putea să vă placă și