The NMJI
VOLUME 17, NUMBER 5

SEPTEMBER/OCTOBER 2004

Review Article

Prostate cancer: Altering the natural history by dietary changes [PDF]

ASEEM R. SRIVASTAVA, D. DALELA

ABSTRACT
The importance of diet on the development and progression of prostate cancer was initially suggested by epidemiological studies. Since then, there has been a vast amount of research in this field. Compelling evidence now provides hope that evidence-based dietary alterations may markedly alter the natural history of this disease. Is there enough evidence for clinicians to be able to advise dietary modifications? The preliminary results no doubt are encouraging, but at present there seems to be no evidence to justify the widespread use of these proposed dietary interventions. However, as public awareness increases, all physicians involved with the care of patients with cancer of the prostate will need to be better armed with the current updates and advice on this issue.

Natl Med J India 2004;17:248–53

INTRODUCTION
The incidence and mortality rates of prostate cancer differ greatly among countries and ethnic groups. At any given age, men all over the world share the same risk for latent cancer, whereas the incidence of clinically manifest prostate cancer differs dramatically,
1 with mortality rates as low as 4 in 100 000 in Japan to about 20 per 100 000 in the USA. So, if American and Japanese men start with the same risk for latent prostate cancer, what accounts for the difference in the rates of clinical cancer? Why do these latent cancers progress to an aggressive form in so many American men and not in Japanese men? What are the factors that transform a latent tumour into a life-threatening one? Is it genetic susceptibility? Or is it because of environmental influences?
    Shimizu et al.
2 have shown that the incidence of prostate cancer in Japanese immigrants to the USA increases in one or two generations to about half that of the indigenous North American population. Migrants have the same genetic make-up as their families in their native countries, and thus this study suggests that there must be an environmental influence that causes the change in risk, possibly diet.

Prostate cancer prevention and tumour biology
Malignancies most likely to benefit from preventive measures must have an early event and a slow progression to malignant transformation. In these types of tumours, interactions between environmental and genetic influences are the greatest and, therefore, are potentially benefited by a preventive strategy. The development of invasive prostate cancer is a culmination of a complex series of initiating and promotional events, under the influence of many genetic, hormonal, dietary and environmental factors over a period of several decades. Prostate cancer may thus be an ideal solid tumour to evaluate preventive strategy.
3 The PTEN or MMAC1 gene located on chromosome 10 regulates cellular motility and matrix interactions and is associated with advanced prostate cancer.4 Other tumour suppressor genes implicated include p53 (present in 25%–75% of patients with prostate cancer)5 the retinoblastoma gene on chromosome 13, and KA 11 on chromosome 11.
    In untreated primary prostate cancer, expression of another tumour suppressor gene p
16 is reduced.6 Oncogenes that may be activated in prostate cancer include c-myc, bcl-2, c-met and ras. Because many risk factors such as age, race and family history are beyond an individual’s control, it is plausible that altering the diet (if it has a major influence) may minimize the risk of developing prostate cancer. One such plausible link (between diet and prostate cancer) seems to be oxidative stress,7 another could be the insulin-like growth factor (IGF-1) system. The ability of IGF-1 to activate androgen receptors even in the absence of androgens suggests its possible role in the progression of prostate cancer.8 IGF-1 also has mitogenic and anti-apoptotic effects on prostate epithelial cells.8,9 While high levels of IGF-1 are associated with prostate cancer, low levels are found in patients whose diets have been supplemented (see lycopene).
    A typical Asian diet is protective against prostate cancer (with India likely to be no exception). However, with a western diet gaining increasing acceptance, the incidence of clinical prostate cancer may begin to rise. Most studies in this field are preliminary and often conflicting, but since there is no curative therapy for advanced prostate cancer, identification of dietary risk factors may help in the development of possible preventive strategies in the future.

Role of serum biomarkers
In view of the long natural history of prostate cancer, if the incidence of cancer acts as the only end-point measured in trials, prolonged periods of observation will be necessary to obtain results. Biomarkers of carcinogenesis, tailored to the agent under investigation, are therefore essential. Examples of such biomarkers are prostate specific antigen (PSA) (serum levels of which often correlate with tumour volume), DNA adduct levels such as 8-oxo-dG or M1G, which can be measured in prostate tissue or in white blood cells as a surrogate for the prostate, to detect antioxidant changes elicited by dietary supplementation. Similarly, the soluble p105 component of p185erbB-2 proto-oncogene may also be monitored as a serum biomarker.

METHODS
Data were gathered from a wide range of sources, including a variety of electronic databases (Pubmed, Medscape), peer-reviewed publications and technical reports. Original publications were reviewed wherever possible to properly interpret the data and associated experimental methods. This was especially important where there was an apparent conflict in the published literature.

FATS
Studies demonstrate a direct relation between a country’s prostate cancer-specific mortality rate and average total calories from fat consumed by the country’s population,
10,11 possibly resulting from prolonged exposure to androgens under these circumstances. In the USA, where there is a high intake of fat, the mortality rate from prostate cancer is high, whereas in Japan, which has one of the lowest fat consumptions in the world, mortality from the disease is low.12
    Men with a high fat consumption are not only more likely to develop prostate cancer but are also more likely to develop a more aggressive form of the disease.
13 Also the risk of its progressing to an advanced stage is higher among men with a high fat intake.14
    Mice model studies show that a low fat diet is associated with slower tumour growth and this effect is probably mediated through the modulation of the IGF axis.
15 Omega-6 fatty acids (derived from linolenic acid) stimulate the growth of prostate cancer cell lines, whereas omega-3 fatty acids such as docosahexaenoic acid and eicosapentaenoic acid, inhibit the growth of these cells.16 A reduced risk of developing prostate cancer has also been seen among men with higher eicosapentaenoic acid levels in their erythrocytes,17 which is likely to be due to its effect on prostaglandin synthesis. Similarly, in a follow up of men in the Health Professionals Follow-up Study,18 a notable finding was that eating fish more than three times a week is associated with a decreased risk of prostate cancer, with the strongest association for metastatic cancer. Intake of marine fatty acids from other sources showed a similar but weaker association.
    However, not all investigators have uniformly reached these conclusions. A rat model study
19 of sex hormone-induced cancers, to examine the effects of high-fat diet on the incidence/latency of prostate cancer, revealed no difference in the pattern of carcinogenesis and weight of the prostate between rats on a high fat diet and controls on a standard, low fat diet. This study does not support the role of dietary fat in promoting sex hormone-induced prostate cancer. Similarly, in a randomized controlled trial,20 the intervention group received intensive counselling to consume a diet low in fat and high in fibre, fruits and vegetables, and the control group received a standard brochure on a healthy diet. There was no difference in the PSA slopes and in the frequency of elevated PSA values for those with an elevated PSA at baseline. The incidence of prostate cancer was also similar among the two groups. This study offers no evidence that dietary modifications over a 4-year period with reduced fat and increased fruits, vegetables and fibre affect the incidence of prostate cancer and that diet has an impact on serum PSA levels in men.
    There are suggestions that decreasing the saturated fat content in the diet of men with prostate cancer may reduce the chances of actually dying from the disease and potentially prolong life.
21 However, a review of published studies on the relationship between dietary fat and prostate cancer risk reveals that approximately half these studies found an increased risk with increased dietary fat while the remaining half found no association.22

VITAMIN D
Calcitriol is a steroid hormone obtained by dietary means or synthesized in the body on exposure to UV light. In 1990, Schwartz and Hulka
23 proposed that a low level of circulating calcitriol is a risk factor for prostate cancer. Since then, many investigators have attempted to explain the regional/racial risk factors of prostate cancer by this hypothesis. A higher mortality from prostate cancer in the northern latitudes of the USA has been linked with a lower exposure to UV rays and hence lower calcitriol levels.24 Similarly, racial pigmentation has been incriminated in the higher incidence of prostate cancer among people of African descent.25
    The normal prostate gland may be a vitamin D target organ;
26 receptors for vitamin D are widely expressed in the normal prostate tissue and more so in the peripheral zone.27 Calcitriol inhibits the growth of normal prostate epithelium28 and has an antiproliferative effect on human prostate cancer,29 initially presumed to be due to vitamin D-induced cell cycle arrest. However, studies now propose the induction of apoptosis as an additional mechanism.30
    To date, there have been two clinical trials on calcitriol in the treatment of human prostate cancer. In one, 7 patients of early recurrent prostate cancer were treated with calcitriol. The results showed a significant slowing of the rate of PSA increase in 6 patients.
31 Unfortunately, results from the other trial, in which 14 patients with hormone-refractory metastatic disease were treated, failed to demonstrate any response.32 More importantly, in both these studies, the dose of calcitriol was limited by hypercalcaemia and hypercalciuria.

SOY
The protective role of soy in prostate cancer was postulated from studies that indicated that men in Southeast Asia, who consume 20–50 times more soy daily than American men, have a 10-fold lower incidence of clinically significant prostate cancer and prostate cancer-related deaths.
33,34 Soy contains large amounts of isoflavones, genistein, daidzein and their glycosides, and has been implicated in the prevention of prostate cancer, possibly via a mild oestrogenic effect.
    The initial evidence that genistein inhibits human prostate growth was provided by studies on the histoculture of minced, surgically resected prostate tissue from men with benign prostatic hypertrophy (BPH).
35 In these studies, it caused inhibition of DNA synthesis. Genistein inhibits the growth of rat prostate adenocarcinoma cells in vitro as well as in Lobund–Wistar rats when injected subcutaneously.36 In these susceptible rats, soy proteins decrease the number of prostatic tumours induced by testosterone37 and protect against the development of chemically induced prostate cancer.38
    In contrast to genistein, daidzein (another major soy isoflavone) is only a weak inhibitor of tumour cell growth in vitro and has no inhibitory effect on the growth of DU-145 or LNCaP cells.
39 However, recent reports suggest that an active metabolite of daidzein (equol) may have potent antiproliferative effects on benign and malignant prostatic epithelial cells.40
    Although most studies suggest a protective role of soy proteins against prostate cancer, some show no effect and one suggests a paradoxically increased growth. Urban et al.
41 in a double-blind study found that short term exposure of elderly men with elevated serum PSA values to soy protein containing isoflavones decreases serum cholesterol but not the serum biomarkers PSA and p105erbB-2.
    Results from another study show that neither an isoflavone-rich soy protein isolate (SPI), nor conjugated linoleic acid (CLA) inhibit the in vivo growth and development of prostate tumour cells when administered in the diet of male Copenhagen rats inoculated with androgen independent R–3327–AT–1 rat prostate tunel cells. Moreover, at the highest concentrations of SPI and CLA there was a significant increase in tumour volume compared with controls. Administration of 10% SPI in the diet also enhanced tumour growth, whereas 5% SPI exerted no measurable effect.
42 Therefore, the authors have cautioned that isoflavone-rich soy protein isolates should not be used in human studies involving advanced prostate cancer until further data on their safety are available.

SELENIUM
There is increasing evidence that selenium, an essential trace element,
43,44 may have protective properties against prostate cancer. Selenium is a constituent of the enzyme glutathione peroxidase, one of the body’s antioxidants and, since oxidant damage has been linked to many cancers, investigators suggest that the anticancer benefits of selenium may arise from its antioxidant function. In cell culture, it reduces the effect of a number of described mutagens45–47 and may alter the metabolism of other carcinogens.48–50 Other potential actions suggested include effects on the immune and endocrine systems, production of cytotoxic selenium metabolites, initiation of apoptosis, inhibition of protein synthesis, as well as inhibition of specific enzymes.51–54 There are sufficient data to suggest that supplementation with selenium decreases the risk of many chemically induced cancers,55–61 spontaneous animal tumours,62 and transplanted animal tumour lines.63 Studies of geographical areas with varying dietary selenium content demonstrate an inverse relationship between selenium intake and cancer risk.64,65 Epidemiological studies have shown mixed results, with significant and inverse relationships encountered in some studies,66–77 while others have not encountered a higher risk in patients with low selenium levels or a low selenium intake.78–83 Salonen et al. studied serum samples collected from 111 subjects, obtained from the Hypertension Detection Follow up Programme, who developed cancer during the subsequent 5 years and compared them with serum samples from 210 cancer-free subjects matched for age, race, sex and smoking history. The mean serum selenium level was lower in cancer cases than in controls and an association between low selenium level and cancer was strongest for gastrointestinal and prostate cancer.66
    Similarly, a multi-institutional study designed to prevent skin cancer randomized a group of 1312 patients with a history of basal cell or squamous cell carcinoma of the skin to either 200 mg selenium supplementation per day or placebo.
84 The baseline serum PSA levels in both arms were also measured. After an average follow up of 6.4 years, selenium-treated patients developed only about one-third as many prostate tumours as patients receiving placebo. Importantly, no patient experienced toxicity due to selenosis, a side-effect that has been reported in association with chronic intake of selenium above 5 ppm.85 Although previous studies suggest that selenium and vitamin E (alone or in combination) may reduce the risk of developing prostate cancer, only a large clinical trial can confirm these initial findings.

LYCOPENE
Carotenoids are encountered in a number of vegetables, notably tomatoes, and are best absorbed if cooked with fats or oils. The most abundant carotenoid in tomato is lycopene, followed by phytoene, phytofluene, zeta-carotene, gamma-carotene, beta-carotene, neurosporene and lutein. These have been postulated to have anticancer properties, attributable to their antioxidant effect.
    Earlier studies evaluating the links between lycopene and prostate cancer risk generally did not find an association, with only one study showing a reduced risk.
86–89
    In 1995, an analysis of the Physicians’ Health Study
90 revealed a one-third reduction in prostate cancer risk in men with the highest consumption of tomato products, attributed to the lycopene in these vegetables. Several in vitro and in vivo studies since then suggest the protective effects of lycopene on specific cancers, including prostate cancer.91,92
    These reports prompted further studies, the results of which have been mixed.
93,94 A prospective study examining the relationship between the plasma concentration of several antioxidants and the risk for prostate cancer reveals that lycopene is the only antioxidant found in significantly lower levels in patients with prostate cancer than in matched controls.95 A similar study found that lycopene levels in prostatic tissue are significantly lower in patients with cancer than in controls. However, serum and tissue levels of beta-carotene and other major carotenoids were not different between the two groups.96 This is surprising, as dietary studies on rat models show a significant decrease in the risk of developing prostate cancer in rats fed on tomato powder compared with lycopene alone.97 Thus it is possible that the influence of tomato on the development of prostate cancer may relate not only to lycopene but to other phytochemicals also. However, in epidemiological studies, beta-carotene has not been found to be associated with prostate cancer risk, or is even associated with an increased risk.98
    From the available literature, it appears that the IGF-1 (a potent mitogen on prostate epithelial cells) axis may play a central role in the progression of prostate cancer. While high levels of IGF-1 have been associated with prostate cancer, low levels are found in those with increased tomato intake.
99 Furthermore, lycopene has been shown to inhibit IGF-1 induced proliferation of several tumour lines. Kucuk et al. randomized 33 men with prostate cancer and assigned them to consume lycopene or no supplement for 30 days before prostatectomy; postoperatively, in the lycopene-supplemented group, PSA levels fell by 20% and cancer had spread in only 33%. However, in 75% of the control group, the cancer had spread and their PSA levels remained unchanged.100 In a more recent study,101 32 patients with localized prostate cancer consumed tomato sauce-based pasta dishes before radical prostatectomy. The mean serum PSA concentrations and leucocyte 80HdG decreased after tomato sauce consumption, and the resected tissues had a lower prostate 80HdG and a higher apoptotic index compared with controls.
    A review of epidemiological studies of tomatoes, lycopene and prostate cancer risk
102 shows that of the 15 studies only 5 support a 30%–40% reduction in risk associated with high tomato or lycopene consumption, whereas 3 suggest a 30% reduction in risk, but the results were not statistically significant. Seven studies do not support any association. The largest relevant dietary study found that consumption of 2–4 servings of tomato sauce per week was associated with a 35% reduction in the risk of prostate cancer and a 50% reduction of advanced prostate cancer. In the largest serum sample-based study, a similar reduction in risk was observed for all and advanced prostate cancer for a high versus low concentration of lycopene.
    Most of these reports do suggest an association between a lower prostate cancer risk and high lycopene intake. However, this evidence is weak because previous studies were not controlled for total vegetable intake (i.e. separating the effect of tomatoes from vegetables in general), dietary intake instruments are poorly able to quantify lycopene intake, and other potential biases.
103
    More important, follow up for a sufficient period of time is required so that other end-points are also captured, as supplementation of the diet with higher than physiological doses of micro-nutrients has at times caused unexpected and unwanted results, e.g. an 18% increase in lung cancers observed in the beta-carotene arm of the ATBC trial.
54
    The final recommendations therefore await further research.

FLAXSEED
Flaxseed is the richest plant source of omega-3 fatty acids and is high in dietary fibre. As previously mentioned, omega-3 fatty acids and dietary fibre may protect against cancer. A diet supplemented with flaxseed has been shown to inhibit the growth and development of prostate cancer in a transgenic adenocarcinoma mouse prostate (TRAMP) model.
104 However, the quantity of flaxseed given to each mouse was 5% of its total food intake, which would probably be too high to be realistic for a human diet.

WINE
The finding that people in Mediterranean countries, with a high intake of legumes, olive oil, wine and vegetables, have a much lower incidence of prostate cancer and that polyphenols in green tea may induce apoptosis in human prostate cancer cell lines led to the hypothesis that polyphenols in red wine may have a similar effect. Romero et al.
105 studied 5 polyphenol constituents of red wine and found that they all inhibited the growth of LNCaP cells at different concentrations and induced apoptosis.

HERBAL THERAPIES
PC-SPES (PC stands for prostate cancer, SPES is Latin for hope), a mixture of 7 medicinal herbs and saw palmetto, was introduced in the USA as a dietary supplement in 1996. Each capsule contains 320 mg of the herbal combination powder with an unknown ratio of each herbal extract.
106Early studies suggested that PC-SPES was effective in reducing the levels of PSA in both hormone-responsive and -resistant prostate cancer patients. The clinical activity and adverse effects of PC-SPES are strikingly similar to high-dose oestrogen (diethyl stilboestrol)107,108 and it is not known if PC-SPES provides a significant additive benefit.
    A chemical analysis of PC-SPES has shown its contamination with warfarin, diethyl stilboestrol and indomethacin. Diethyl stilboestrol and indomethacin are known to have anticancer properties. Later preparations of PC-SPES, which contained less diethyl stilboestrol and indomethacin, showed a 6-fold lower anticancer potency.
109

SUMMARY
Although there are too many hypotheses on this subject, there are little hard and conclusive data. From the published literature, it is clear that diet does have a role to play in the development and progression of prostate cancer. However, clear and convincing recommendations will have to await the results of long term prospective clinical trials.
110 Presently, there seems to be little or no evidence to support an advise beyond a healthy diet that is low in fat, and rich in fruits, vegetables and fibre.
    However, continuing research in this field may lead to an improved understanding of the mechanism by which diet influences prostate glad oncogenesis and may provide means to lower the risk of the disease.

REFERENCES
  1. Wynder EL, Mabuchi K, Whitmore WF Jr. Epidemiology of cancer of the prostate. Cancer 1971;28:344–60.
  2. Shimizu H, Ross RK, Bernstein L, Yatani R, Henderson BE, Mack TM. Cancers of the prostate and breast among Japanese and white immigrants in Los Angeles county. Br J Cancer 1991;63:963–6.
  3. Uzzo RG, Bruner DW, Horwitz E. Prostate cancer prevention. Strategies and realities. In: Godec CJ (ed). Prostate cancer: Science and clinical practice. London:Academic Press; 2003:90–9.
  4. Suzuki H, Freije D, Nusskern DR, Okami K, Cairns P, Sidransky D, et al. Interfocal heterogeneity of PTEN/MMAC1 gene alterations in multiple metastatic prostate cancer tissues. Cancer Res 1998;58:204–9.
  5. Heidenberg HB, Sesterhenn IA, Gaddipati JP, Weghorst CM, Buzard GS, Moul JW, et al. Alteration of the tumor suppressor gene p53 in a high fraction of hormone refractory prostate cancer. J Urol 1995;154:414–21.
  6. Chi SG, de Vere White RW, Muenzer JT, Gumerlock PH. Frequent alteration of CDKNZ (p 16 [INK4A] MTS1) expression in human primary prostate carcinomas. Clin Cancer Res 1997;3:1889–97.
  7. Fleshner NE, Klotz LH. Diet, androgens, oxidative stress and prostate cancer susceptibility. Cancer Metastasis Rev 1998–99;17:325–30.
  8. Culig Z, Hobisch A, Cronauer MV, Hittmair A, Radmayr C, Bartsch G, et al. Activation of the androgen receptor by polypeptide growth factors and cellular regulators. World J Urol 1995;13:285–9.
  9. Iwamura M, Sluss PM, Casamento JB, Cockett AT. Insulin-like growth factor I: Action and receptor characterization in human prostate cancer cell lines. Prostate 1993;22: 243–52.
  10. Armstrong B, Doll R. Environmental factors and cancer incidence and mortality in different countries, with special reference to dietary practices. Int J Cancer 1975;15:617–31.
  11. Rose DP, Connolly JM. Dietary fat, fatty acids and prostate cancer. Lipids 1992;27: 798–803.
  12. Statland BE. Nutrition and cancer. Clin Chem 1992;38 (8B Pt 2):1587–94.
  13. Giovannucci E, Rimm EB, Colditz GA, Stamfer MJ, Ascherio A, Chute CC, et al. A prospective study of dietary fat and risk of prostate cancer. J Natl Cancer Inst 1993;85:1571–9.
  14. Bairati I, Meyer F, Fradet Y, Moore L. Dietary fat and advanced prostate cancer. J Urol 1998;159:1271–5.
  15. Ngo TH, Barnard RJ, Leung PS, Cohen P, Aronson WJ. Insulin-like growth factor 1 (IGF-1) and IGF binding protein-1 modulate prostate cancer cell growth and apoptosis: Possible mediators for the effects of diet and exercise on cancer cell survival. Endocrinology 2003;144:2319–24.
  16. Rose DP, Connolly JM. Effects of fatty acids and eicosanoid synthesis inhibitors on the growth of two human prostate cancer cell lines. Prostate 1991;18:243–54.
  17. Norrish AE, Skeaff CM, Arribas GL, Sharpe SJ, Jackson RT. Prostate cancer risk and consumption of fish oils: A dietary biomarker-based case–control study. Br J Cancer 1999;81:1238–42.
  18. Augustsson K, Michaud DS, Rimm EB, Leitzmann MF, Stampfer MJ, Willett WC, et al. A prospective study of intake of fish and marine fatty acids and prostate cancer. Cancer Epidemiol Biomarkers Prev 2003;12:64–7.
  19. Leung G, Benzie IF, Cheung A, Tsao SW, Wong YC. No effect of a high-fat diet on promotion of sex hormone-induced prostate and mammary carcinogenesis in the Noble rat model. Br J Nutr 2002;88:399–409.
  20. Shike M, Latkany L, Riedel E, Fleisher M, Schatzkin A, Lanza E, et al. Lack of effect of a low-fat, high-fruit, -vegetable, and -fiber diet on serum prostate-specific antigen of men without prostate cancer: Results from a randomized trial. J Clin Oncol 2002;20:3592–8.
  21. Weisburger JH. Nutritional approach to cancer prevention with emphasis on vitamins, antioxidants, and carotenoids. Am J Clin Nutr 1991;53 (1 Suppl):226S–237S.
  22. Zhou JR, Blackburn GL. Bridging animal and human studies: What are the missing segments in dietary fat and prostate cancer? Am J Clin Nutr 1997;66 (6 Suppl): 1572S–1580S.
  23. Schwartz GG, Hulka BS. Is vitamin D deficiency a risk factor for prostate cancer? (hypothesis). Anticancer Res 1990;10 (5A):1307–11.
  24. Hanchette CL, Schwartz GG. Geographic patterns of prostate cancer mortality. Evidence for a protective effect of ultraviolet radiation. Cancer 1992;70:2861–9.
  25. Matsuoka LY, Wortsman J, Haddad JG, Kolm P, Hollis BW. Racial pigmentation and the cutaneous synthesis of vitamin D. Arch Dermatol 1991;127:536–8.
  26. Konety BR, Schwartz GG, Acierno JS Jr, Becich MJ, Getzenberg RH. The role of vitamin D in normal prostate growth and differentiation. Cell Growth Differ 1996;7:1563–70.
  27. Krill D, DeFlavia P, Dhir R, Luo J, Becich MJ, Lehman E, et al. Expression patterns of vitamin D receptor in human prostate. J Cell Biochem 2001;82:566–72.
  28. Peehl DM, Skowronski RJ, Leung GK, Wong ST, Stamey TA, Feldman D. Antiproliferative effects of 1,25-dihydroxyvitamin D3 on primary cultures of human prostatic cells. Cancer Res 1994;54:805–10.
  29. Miller GJ, Stapleton GE, Ferrara JA, Lucia MS, Pfister S, Hedlund TE, et al. The human prostatic adenocarcinoma cell line LNCaP express biologically active, specific receptors for 1 alpha, 25-dihydroxyvitamin D3. Cancer Res 1992;52:515–20.
  30. Blutt SE, Polek TC, Stewart LV, Kattan MW, Weigel NL. A calcitriol analogue, EB1089, inhibits the growth of LNCaP tumors in nude mice. Cancer Res 2000;60: 779–82.
  31. Gross C, Stamey T, Hancock S, Feldman D. Treatment of early recurrent prostate cancer with 1,25-dihydroxyvitamin D3 (calcitriol). J Urol 1998;159:2035–9; discussion 2039–40. Erratum in: J Urol 1998;160 (3 Pt 1):840.
  32. Osborn JL, Schwartz GG, Smith DC, Bahnson R, Day R, Trump DL. Phase II trial of oral 1,25-dihydroxyvitamin D (calcitriol) in hormone refractory prostate cancer. Urol Oncol 1995;1:195–8.
  33. Stamey TA, McNeal JE. Adenocarcinoma of the prostate. In: Walsh PC, Retik AB, Stamey TA, Vaughan ED Jr (eds). Campbell’s Urology. 6th ed. Philadelphia:WB Saunders, 1992:1159–1221.
  34. Messina MJ, Persky V, Setchell KR, Barnes S. Soy intake and cancer risk: A review of the in vitro and in vivo data. Nutr Cancer 1994;21:113–31.
  35. Geller J, Sionit L, Partido C, Li L, Tan X, Youngkin T, et al. Genistein inhibits the growth of human-patient BPH and prostate cancer in histoculture. Prostate 1998;34: 75–9.
  36. Dalu A, Haskell JF, Coward L, Lamartiniere CA. Genistein, a component of soy, inhibits the expression of the EGF and ErbB2/Neu receptors in the rat dorsolateral prostate. Prostate 1998;37:36–43.
  37. Pollard M, Luckert PH. Influence of isoflavones in soy protein isolates on development of induced prostate-related cancers in L–W rats. Nutr Cancer 1997;28:41–5.
  38. Wang J, Eltoum IE, Lamartiniere CA. Dietary genistein suppresses chemically induced prostate cancer in Lobund–Wistar rats. Cancer Lett 2002;186:11–18.
  39. Peterson TG, Barnes S. Genistein and biochanin A inhibit the growth of human prostate cancer cells but not epidermal growth factor receptor tyrosine autophosphorylation. Prostate 1993;22:335–45.
  40. Hedlund TE, Johannes WU, Miller GJ. Soy isoflavonoid equol modulates the growth of benign and malignant prostatic epithelial cells in vitro. Prostate 2003;54:68–78.
  41. Urban D, Irwin W, Kirk M, Markiewicz MA, Myers R, Smith M, et al. The effect of isolated soy protein on plasma biomarkers in elderly men with elevated serum prostate specific antigen. J Urol 2001;165:294–300.
  42. Cohen LA, Zhao Z, Pittman B, Scimeca J. Effect of soy protein isolate and conjugated linoleic acid on the growth of Dunning R-3327-AT-1 rat prostate tumors. Prostate 2003;54:169–80.
  43. Muth OH, Weswig PH, Whanger PD, Oldfield JE. Effect of feeding selenium-deficient ration to the subhuman primate (Saimiri sciureus). Am J Vet Res 1971;32:1603–5.
  44. Young VR. Selenium: A case for its essentiality in man. N Engl J Med 1981;304: 1228–30.
  45. Norppa H, Westermarck T, Laasonen M, Knuutila L, Knuutila S. Chromosomal effects of sodium selenite in vivo. I. Aberrations and sister chromatid exchanges in human lymphocytes. Hereditas 1980;93:93–6.
  46. Shamberger RJ, Corlett CL, Beaman KD, Kasten BL. Effect of selenium and other antioxidants on the mutagenicity of malonaldehyde. Fed Proc 1978;37:A265, 261.
  47. Jacobs MM, Matney TS, Griffin AC. Inhibitory effects of selenium on the mutagenicity of 2-acetylaminofluorene (AAF) and AAF derivatives. Cancer Lett 1977;2:319–22.
  48. Griffin AC. Role of selenium in the chemoprevention of cancer. Adv Cancer Res 1979;29:419–42.
  49. Daoud AH, Griffin AC. Effects of selenium and retinoic acid on the metabolism of N-acetylaminofluorene and N-hydroxyacetylamino-fluorene. Cancer Lett 1978;5: 231–7.
  50. Marshall MV, Rasco MA, Griffin AC. Effects of selenium on benzo(a)pyrene metabolism. Fed Proc 1978;37:A628, 1383.
  51. Combs GF Jr, Scott ML. Nutritional interrelationships of vitamin E and selenium. BioScience 1977;27:467–73.
  52. Rotruck JT, Pope AL, Ganther HE, Swanson AB, Hafeman DG, Hoekstra WG. Selenium: Biochemical role as a component of glutathione peroxidase. Science 1973;179:588–90.
  53. Chow CK. Nutritional influence on cellular antioxidant defense systems. Am J Clin Nutr 1979;32:1066–81.
  54. The Alpha-Tocopherol, Beta Carotene Cancer Prevention Study Group. The effect of vitamin E and beta carotene on the incidence of lung cancer and other cancers in male smokers. N Engl J Med 1994;330:1029–35.
  55. Milner JA. Effect of selenium on virally induced and transplantable tumor models. Fed Proc 1985;44:2568–72.
  56. Thompson HJ, Wilson A, Lu J, Singh M, Jiang C, Upadhyaya P, et al. Comparison of the effects of an organic and an inorganic form of selenium on a mammary carcinoma cell line. Carcinogenesis 1994;15:183–6.
  57. Ip C, el-Bayoumy K, Upadhyaya P, Ganther H, Vadhanavikit S, Thompson H. Comparative effect of inorganic and organic selenocyanate derivatives in mammary cancer chemoprevention. Carcinogenesis 1994;15:187–92.
  58. Reddy BS, Rivenson A, Kulkarni N, Upadhyaya P, el-Bayoumy K. Chemoprevention of colon carcinogenesis by the synthetic organoselenium compound 1,4-phenylenebis (methylene) selenocyanate. Cancer Res 1992;52:5635–40.
  59. Reddy BS, Rivenson A, el-Bayoumy K, Upadhyaya P, Pittman B, Rao CV. Chemoprevention of colon cancer by organoselenium compounds and impact of high- or low-fat diets. J Natl Cancer Inst 1997;89:506–12.
  60. Shamberger RJ. Increase of peroxidation in carcinogenesis. J Natl Cancer Inst 1972;48:1491–7.
  61. Shamberger RJ. Relationship of selenium to cancer. I. Inhibitory effect of selenium on carcinogenesis. J Natl Cancer Inst 1970;44:931–6.
  62. Jacobs MM. Effects of selenium on chemical carcinogens. Prev Med 1980;9:362–7.
  63. Schrauzer GN, Ishmael D. Effects of selenium and of arsenic on the genesis of spontaneous mammary tumors in inbred C3H mice. Ann Clin Lab Sci 1974;4:441–7.
  64. Shamberger RJ, Tytko SA, Willis CE. Antioxidants and cancer. Part VI. Selenium and age-adjusted human cancer mortality. Arch Environ Health 1976;31:231–5.
  65. Schrauzer GN, White DA, Schneider CJ. Cancer mortality correlation studies—III: Statistical associations with dietary selenium intakes. Bioinorg Chem 1977;7:23–31.
  66. Salonen JT, Salonen R, Lappetelainen R, Maenpaa PH, Alfthan G, Puska P. Risk of cancer in relation to serum concentrations of selenium and vitamins A and E: Matched case–control analysis of prospective data. Br Med J (Clin Res Ed) 1985;290:417–20.
  67. Willett WC, Polk BF, Morris JS, Stampfer MJ, Pressel S, Rosner B, et al. Prediagnostic serum selenium and risk of cancer. Lancet 1983;2:130–4.
  68. Virtamo J, Valkeila E, Alfthan G, Punsar S, Huttunen JK, Karvonen MJ. Serum selenium and risk of cancer: A prospective follow-up of nine years. Cancer 1987;60: 145–8.
  69. van den Brandt PA, Goldbohm RA, van’t Veer P, Bode P, Dorant E, Hermus RJ, et al. A prospective cohort study on toenail selenium levels and risk of gastrointestinal cancer. J Natl Cancer Inst 1993;85:224–9.
  70. Peleg I, Morris S, Hames CG. Is serum selenium a risk factor for cancer? Med Oncol Tumor Pharmacother 1985;2:157–63.
  71. Broghamer WL, McConnell KP, Blotcky AL. Relationship between serum selenium levels and patients with carcinoma. Cancer 1976;37:1384–8.
  72. Shamberger RJ, Rukovena E, Longfield AK, Tytko SA, Deodhar S, Willis CE. Antioxidants and cancer. I. Selenium in the blood of normals and cancer patients. J Natl Cancer Inst 1973;50:863–70.
  73. McConnell KP, Broghamer WL Jr, Blotcky AJ, Hurt OJ. Selenium levels in human blood and tissues in health and in disease. J Nutr 1975;105:1026–31.
  74. Calautti P, Moschini G, Stievano BM, Tomio L, Calzavara F, Perona G. Serum selenium levels in malignant lymphoproliferative diseases. Scand J Haematol 1980;24:63–6.
  75. McConnell KP, Jager RM, Bland KI, Blotcky AJ. The relationship of dietary selenium and breast cancer. J Surg Oncol 1980;15:67–70.
  76. Clark LC, Graham GF, Crounse RG, Grimson R, Hulka B, Shy CM. Plasma selenium and skin neoplasms: A case–control study. Nutr Cancer 1984;6:13–21.
  77. Fex G, Pettersson B, Akesson B. Low plasma selenium as a risk factor for cancer death in middle-aged men. Nutr Cancer 1987;10:221–9.
  78. Robinson MF, Godfrey PJ, Thomson CD, Rea HM, van Rij AM. Blood selenium and glutathione peroxidase activity in normal subjects and in surgical patients with and without cancer in New Zealand. Am J Clin Nutr 1979;32:1477–85.
  79. Garland M, Morris JS, Stampfer MJ, Colditz GA, Spate VL, Baskett CK, et al. Prospective study of toenail selenium levels and cancer among women. J Natl Cancer Inst 1995;87:497–505.
  80. Nomura A, Heilbrun LK, Morris JS, Stemmermann GN. Serum selenium and the risk of cancer, by specific sites: Case–control analysis of prospective data. J Natl Cancer Inst 1987;79:103–8.
  81. Knekt P, Aromaa A, Maatela J, Alfthan G, Aaran RK, Teppo L, et al. Serum vitamin E, serum selenium and the risk of gastrointestinal cancer. Int J Cancer 1988;42:846–50.
  82. Ringstad J, Jacobsen BK, Tretli S, Thomassen Y. Serum selenium concentration associated with risk of cancer. J Clin Pathol 1988;41:454–7.
  83. Coates RJ, Weiss NS, Daling JR, Morris JS, Labbe RF. Serum levels of selenium and retinol and the subsequent risk of cancer. Am J Epidemiol 1988;128:515–23.
  84. Clark LC, Combs GF Jr, Turnbull BW, Slate EH, Chalker DK, Chow J, et al. Effects of selenium supplementation for cancer prevention in patients with carcinoma of the skin. A randomized controlled trial. Nutritional Prevention of Cancer Study Group. JAMA 1996;276:1957–63. Erratum in: JAMA 1997;277:1520.
  85. Fan AM, Kizer KW. Selenium: Nutritional, toxicologic, and clinical aspects. West J Med 1990;153:160–7.
  86. Hsing AW, Comstock GW, Abbey H, Polk BF. Serologic precursors of cancer: Retinol, carotenoids, and tocopherol and risk of prostate cancer. J Natl Cancer Inst 1990;82: 941–6.
  87. Mills PK, Beeson WL, Phillips RL, Fraser GE. Cohort study of diet, lifestyle, and prostate cancer in Adventist men. Cancer 1989;64:598–604.
  88. Schuman LM, Mandel JS, Radke A, Seal U, Halberg F. Some selected features of the epidemiology of prostatic cancer: Minneapolis–St Paul, Minnesota case–control study, 1976–1979. In: Magnus K (ed). Trends in cancer incidence: Causes and practical implications. Washington, DC:Hemisphere Publishing; 1982:345–54.
  89. Le Marchand L, Hankin JH, Kolonel LN, Wilken LR. Vegetable and fruit consumption in relation to prostate cancer risk in Hawaii: A reevaluation of the effect of dietary beta-carotene. Am J Epidemiol 1991;133:215–19.
  90. Giovannucci E, Ascherio A, Rimm EB, Stamfer MJ, Colditz GA, Willett WC. Intake of carotenoids and retinol in relation to risk of prostate cancer. J Natl Cancer Inst 1995;87:1767–76.
  91. Stahl W, Sies H. Lycopene: A biologically important carotenoid for humans? Arch Biochem Biophys 1996;336:1–9.
  92. Giovannucci E, Clinton SK. Tomatoes, lycopene, and prostate cancer. Proc Soc Exp Biol Med 1998;218:129–39.
  93. Jain MG, Hislop GT, Howe GR, Ghadirian P. Plant foods, antioxidants, and prostate cancer risk: Findings from case–control studies in Canada. Nutr Cancer 1999;34: 173–84.
  94. Key TJ, Silcocks PB, Davey GK, Appleby PN, Bishop DT. A case–control study of diet and prostate cancer. Br J Cancer 1997;76:678–87.
  95. Gann PH, Ma J, Giovannucci E, Willett W, Sacks FM, Hennekens CH, et al. Lower prostate cancer risk in men with elevated plasma lycopene levels: Results of a prospective analysis. Cancer Res 1999;59:1225–30.
  96. Rao AV, Fleshner N, Agarwal S. Serum and tissue lycopene and biomarkers of oxidation in prostate cancer patients: A case–control study. Nutr Cancer 1999;33: 159–64.
  97. Boileau TS, Clinton Z, Liao M, Monaca S, Donovan J, Erdman JW Jr. Lycopene, tomato powder and dietary restriction influence survival of rats with prostate cancer induced by NMU and testosterone. J Nutr 2001;131:191S.
  98. Schulman CC, Ekane S, Zlotta AR. Nutrition and prostate cancer: Evidence or suspicion? Urology 2001;58:318–34.
  99. Mucci LA, Tamimi R, Lagiou P, Trichopoulou A, Benetou V, Spanos E, et al. Are dietary influences on the risk of prostate cancer mediated through the insulin-like growth factor system? BJU Int 2001;87:814–20.
  100. Kucuk O, Sakr W, Sarkar FH, Djuric Z, Pollak MN, Khachik F, et al. Lycopene supplementation in men with localized prostate cancer (PCa) modulates grade and volume of prostatic intraepithelial neoplasia (PIN) and tumor, level of serum PSA and biomarkers of cell growth, differentiation and apoptosis. Proc Annu Meet Am Assoc Cancer Res 1999;40:409.
  101. Bowen P, Chen L, Stacewicz-Sapuntzakis M, Duncan C, Sharifi R, Ghosh L, et al. Tomato sauce supplementation and prostate cancer: Lycopene accumulation and modulation of biomarkers of carcinogenesis. Exp Biol Med (Maywood) 2002;227: 886–93.
  102. Giovannucci E. A review of epidemiologic studies of tomatoes, lycopene, and prostate cancer. Exp Biol Med (Maywood) 2002;227:852–9.
  103. Kristal AR, Cohen JH. Invited commentary: Tomatoes, lycopene, and prostate cancer. How strong is the evidence? Am J Epidemiol 2000;151:124–7.
  104. Lin X, Gingrich JR, Bao W, Li J, Haroon ZA, Demark-Wahnefried W. Effect of flaxseed supplementation on prostatic carcinoma in transgenic mice. Urology 2002;60:919–24.
  105. Romero I, Paez A, Ferruelo A, Lujan M, Berenguer A. Polyphenols in red wine inhibit the proliferation and induce apoptosis of LNCaP cells. BJU Int 2002;89: 950–4.
  106. Moyad MA, Pienta KJ, Montie JE. Use of PC-SPES, a commercially available supplement for prostate cancer, in a patient with hormone-naive disease. Urology 1999;54:319–23; discussion 323–4.
  107. The Leuprolide Study Group. Leuprolide versus diethylstilbestrol for metastatic prostate cancer. N Engl J Med 1984;311:1281–6.
  108. Citrin DL, Kies MS, Wallemark CB, Khandekar J, Kaplan E, Camacho F, et al. A phase II study of high-dose estrogens (diethylstilbestrol diphosphate) in prostate cancer. Cancer 1985;56:457–60.
  109. White J. PC-SPES—A lesson for future dietary supplement research. J Natl Cancer Inst 2002;94:1261–3.
  110. Ansari MS, Gupta NP, Hemal AK. Chemoprevention of carcinoma prostate: A review. Int Urol Nephrol 2002;34:207–14.

King George’s Medical University, Lucknow, Uttar Pradesh, India
ASEEM R. SRIVASTAVA Department of Surgery
D. DALELA Department of Urology
Correspondence to ASEEM R. SRIVASTAVA

 

 

 

 

 

 

 

 

 


Search
NMJI Web
 
Contact Us | Site Map | Feedback | Disclaimer