.Qnfitméa. . . THE“ ICHIGAN STATE UNIVERSITY LIBRAR S Ill ll!(ll/llllllll!!!llll’llllllllllll 3 1293 01029 6543 This is to certify that the dissertation entitled EFFECT OF DIETARY FAT ON THE GROWTH OF HUMAN BREAST CARCINOMA TRANSPLANTED TO THE ATHYMIC NUDE MICE: ROLE OF LIPID PEROXIDATION IN THIS GROWTH PROCESS presented by Michael J. Gonzalez has been accepted towards fulfillment of the requirements for Ph . D . degree in Human Nutrition I). COM ,a/Céfifi x/M Rachel A. Schemmel Major professor Date 10/7/93 MSU is an Aflirmatiw Action/Equal Opportunity Institution 0- 12771 LIBRARY Mlchlgan State Unlverslty PLACE IN RETURN 30X to roman thb chockout from your record. 1'0 AVOID FINES Mum on or before date duo. DATE DUE DATE DUE DATE DUE [Til f MSUIcAnAIflmatMActIuVEqudOppomnIIyIn-mwon , ,_,,h,,,_ # Wan-m EFFECT OF DIETARY FAT ON THE GROHTH OF HUMAN BREAST CARCINOMA TRANSPLANTED TO THE ATHYMIC NUDE MICE: ROLE OF LIPID PEROXIDATION IN THIS GROHTH PROCESS By Michael J. Gonzalez Advisors: Dr. Rachel A. Schemmel Dr. Clifford II. Helsch AN ABSTRACT OF A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Food Science and Human Nutrition 1993 ABSTRACT EFFECT OF DIETARY FAT ON THE GROWTH OF HUMAN BREAST CARCINOMA TRANSPLANTED TO THE ATHYMIC NUDE MICE: ROLE OF LIPID PEROXIDATION IN THIS GROHTH PROCESS BY Michael J. Gonzalez In athymic nude mice, altering the Ievels and/or types of dietary fat, markedly influences the deveIopment and growth of transplanted human breast carcinomas (MCF-7 and MDA-M823l cell lines). This phenomenon has also been demonstrated in an array of spontaneous, carcinogen-induced, transplantable benign and carcinomatous experimental mammary tumor systems. In general, diets high in unsaturated fatty acids (i.e., corn oil) were more effective enhancers of mammary tumorigenesis than diets high in saturated fatty acids (i.e., butter). In contrast high-fat diets with elevated amounts of long-chain polyunsaturated fatty acids, such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), which are found in certain fish oils (i.e. menhaden oil), did not enhance mammary tumorigenesis, and had a suppressive effect on tumor growth. Various mechanisms have been proposed for this fish oil-induced inhibitory action; we have concentrated our efforts on lipid peroxidation. Our results demonstrate that this neglected mechanism, at Ieast in part, is responsible for the suppressive action exerted by fish oils (menhaden) on the growth of human breast carcinomas (MDA- M8231) maintained in athymic nude mice. In addition, evidence is provided that in the preparation of fish oil diets, the addition of antioxidants at the amounts recommended by the American Institute of Nutrition, or even substantially higher levels, did not completely suppress oxidative deterioration of experimental diets. In terms of tumor growth kinetics, the question arises as to what aspect of tumor growth dynamics can be affected by dietary fish oils: tumor cell proliferation or tumor cell loss. Our preliminary results suggest that tumor cell loss is more significantly affected than tumor cell proliferation in human breast carcinoma transplanted to athymic nude mice fed a diet high in fish oil compared to a diet high in corn oil. This increase in cell loss may partly explain why tumor growth is suppressed in animals fed diets high in fish oil compared to diets high in corn oil. Dedication To the memory of my grandparents Juan and Luz Maria Guzman whom in earth and now in heaven will always guide my steps. (Both died victims of cancer) iv Sc; anc her exce than and did t that With a form, perOXli i”formal COnsjde’ Than attended like to I ADpre ACKNOWLEDGMENTS I would like to express my deepest gratitude to Dr. Rachel A. Schemmel, chairperson of my graduate committee, for her encouragement and needed support. In reality, there are not enough words to thank her kindness. She has a very special place in my heart. I would also like to thank Dr. Clifford W. Welsch, who gave me an excellent opportunity to do first-class research. I would also like to thank him for letting me know my strengths and weaknesses in research and for permitting me to try my ideas, some that worked and some that did not. In many instances I looked up to him as the father figure that was absent in my life. Another sincere thank you goes to Dr. J. Ian Gray, who provided me with a magnificent laboratory setting in which my ideas took reality form. Also I would like to thank him for the lessons on lipid peroxidation and for his positive encouragement. My thanks to Dr. LeRoy Dugan, Jr. for his time, encouragement and informal but excellent teachings of the lipid peroxidation process. I consider him my definite academic grandfather. Thanks to Dr. Maurice R. Bennink, without whom I would have not attended Michigan State and obtained this excellent education. I would like to thank him for being an important part of my guidance committee. Appreciation also goes to Dr. Nikolay Dimitrov for being part of this excellent graduate committee. an di prc bel hav who: help Syst myg Mari¢ Felic Santa Dr. E Dr- Al Whitin Dr, Cr: to nsu cracke] Rodrl9U< I would like to give a special thank you for my grandparents, Juan and Luz M. Guzman, who are no longer here but who encouraged me to get a quality education and guided my steps toward this goal. This dissertation is dedicated to their memory. I would also like to thank my dear mother, Daisy Guzman, who always provided continuous support for me in every way possible, for always believing in me. Without her help, love and support this goal would have been unattainable. I would also like to thank Enid J. Gonzalez, my dear wife, without whom it would have been nearly impossible to obtain this goal; her help, encouragement and companionship were the basis of my support system. I would also like to thank my uncle Adolfo Guzman, the memories of my greatgrandmother, Agustina Diaz, my uncle John Guzman, my friends Mario Garcia, Roberto Zayas and my adoptive grandmother, Monserrate Feliciano, for serving as inspiration for me. A special appreciation goes to Daisy Gonzalez, my sister, Dely Santana, my niece, and to my new motivator, my son Michael John, Jr. AIso thanks to my formal mentors: Dr. Alan M. Preston, Dr. Elizabeth Sanchez, Dr. Gabriel Infante, Prof. Juan L. R. Ricart, Dr. Alejandro Ruiz, Prof. Nivea Benitez, Dr. Dave Copson, Prof. William Whiting, Prof. Fernando Cofresi, Dr. Richard Squire, Dr. Robert Bard, Dr. Cruz Maria Nazario, Dr. Flavio Padovani, Prof. Virginia Baez, and to MSU faculty members Dr. Dale Romsos, Dr. Dana Ott and Prof. Rhonda Crackel for pulling out the best of me. I would also like to thank my extended family: Dr. Jose R. Rodriguez, Prof. CarIos and Jackie Ricart, Dr. Carlos Benitez, vi Ul Ma Ca Ro ant Roc Hec Fun and supp P.R. at M! Dr. C Munoz Ponce wito PEYO Marie Vande Ricky returi Ina"kit Dr. Larry G. Martin, Krystina Kras, Dr. Carol Friesen, Fernando and Mari Herrera, Dr. Carlos M. CeboIlero, Dr. Ramon D. Martinez, Prof. Carlos Ramos, Dr. Dave McConnell, Graciela CeboIlero, Rafael A. Rodriguez, Dr. Lilian Ramos, Pedro and Gladys Bauza, Agnes, Pepe, Quico and Javier Bauza, Violeta and Hernan Marin, Nando, Oscar and Charlie Rodriguez, Angel Melendez, Sister Nildred Rodriguez, Dr. Tirso Pena, Hector Rodriguez, Guille Feliciano, Chit Leong, Cheng Hsuing Chen, Fumin Chu, Liang-Ting Chang, Su-Fen Weng, my adoptive aunt Leo Colon and Alba Rivera. Appreciation goes to the people and agencies that gave me financial support: Administration of Economic Development of the Government of P.R., The University of P.R., Dept. of Food Science and Human Nutrition at MSU, the Office of Equal Opportunity of Urban Affairs at MSU, Dr. Clifford W. Welsch, Senators Miguel Hernandez Agosto, Victoria Munoz, Velda Gonzalez, former Governor Rafael Hernandez Colon, Mayor of Ponce Rafael Cordero Santiago, President of the Municipal Assembly Luis wito Morales, Francisco Negron, Jaime Fuster, Jose Tito Colorado and Peyo Tata. Last but not least a real special thanks to my East Lansing family: Marlene Green, Mary Schneider, Dr. Lois Lund, Murry Edwards, Vonnie Vanderploeg, Maggie Welsch, Cori and Jessica Oakley, Sandra, Rei and Ricky Torres, Alba and Lee Echeverry, Gilmartin and Yon Pares. God bless them all! Above all, thanks to God the Almighty, to whom I hope one day I can return this favor using this bountiful knowledge for the good of mankind. vii Cl Su Co. Rec TABLE OF CONTENTS Page List of Tables .......................... ix List of Figures ......................... xi Introduction ........................... 1 Review of Literature ....................... 9 Chapter One ........................... 48 Chapter Two ........................... 72 Chapter Three .......................... 93 Chapter Four ........................... 118 Summary ............................. 147 Conclusions ........................... 149 Recommendations ......................... 151 Appendix ............................. 157 viii Che Chapter 1. 2. 3. Chapter 1. 2. Chapter 1. 2. 3. LIST OF TABLES 1 Page Diet composition ..................... 67 Predominant fatty acids in oils and fats (percentage) . . 68 Effect of dietary fat on growth of MCF-7 and MDA-M8231 human breast carcinomas transplanted to athymic nude mice ........................... 69 Effect of dietary fat on MDA-MBZ31 human breast carcinoma growth and carcinoma lipid peroxidation product levels. . 70 2 Diet composition ..................... 86 Effect of supplemented dietary antioxidants on peroxide values of fish (menhaden) oil diets ........... 87 Effect of supplemented dietary antioxidants on TBARS levels of fish (menhaden) oil diets ........... 89 Effect of supplemented dietary antioxidants on TBARS levels of organs in mice fed fish (menhaden) oil diets for 4 weeks ....................... 91 3 Diet composition ..................... 111 Predominant fatty acids (1) ............... 112 Effect of different ratios of dietary corn oil and fish oil on the growth and lipid peroxidation product concentrations of MDA-MBZSI human breast carcinomas in athymic nude mice .................... 113 Effect of different ratios of dietary corn oil and fish oil and the influence of antioxidants on the growth and lipid peroxidation product concentrations of MDA-MBZBI human breast carcinomas in athymic nude mice ....... 114 ix Table Chapter 1. 2. 3. Effect of amount of time the fish oil diet remained in the food Jar under usuaI feeding conditions on the growth and lipid peroxidation product concentrations of MDA-M8231 human breast carcinomas in athymic nude mice. . 4 Diet composition ..................... Predominant fatty acids in oils and fats (percentage) . . Effect of dietary fat (corn oil and fish oil) on DNA synthesis (H ~thymidine autoradiography and Brdu) of human breast carcinoma MDA-MBZ31 maintained in athymic nude mice ........................ Effect of dietary fat (corn oiI and fish oil) on DNA synthesis (Brdu and PCNA) of human breast carcinoma MDA-MBZ31 maintained in athymic nude mice ........ Effect of different ratios of dietary fats (corn oil and fish oil) on DNA synthesis (Brdu) of human breast carcinoma MDA-MBZ31 maintained in athymic nude mice . . . Effect of dietary fat (corn oil and fish oil) on loss of cells in human breast carcinoma MDA-M8231 maintained in athymic nude mice .................... Page 115 139 140 141 142 143 144 LIST OF FIGURES Chapter 3 Page 1. Chapter 1. Relationship between mean carcinoma volume (cm3) and mean carcinoma TBARS concentration per individual MDA-MBZ31 human breast carcinomas in athymic nude mice fed a 1% corn oil/19% fish oil diet (N-24) ..... 116 4 Rate of 1125Urd Toss from human breast carcinomas in athymic nude mice fed corn oil (CO), fish oil (F0) and fish oil supplemented with antioxidants (FO+A) for a period of 6 weeks .................... 146 xi El vi st an. 90' tun INTRODUCTION Numerous authors have reported that there is a strong association between dietary fat intake and cancer prevalence (see 1 for review). Various epidemiological studies indicated that the consumption of high levels of dietary fat by humans is associated with increased incidence of cancer, particularly of the breast (2), colon (3) and prostate (4). In addition, in experimental animal studies, mammary tumorigenesis enhancement by high levels of dietary fat has been achieved in virtually every rodent model that has been examined (5-8). In these studies, the type and amount of fat ingested influences the occurrence and development of mammary tumors. High dietary levels of poTyunsaturated fats have been more effective in enhancing mammary tumorigenesis than high Ievels of saturated fats (9-11). It has been suggested that linoleic acid may be the fatty acid primarily responsibIe for the tumorigenesis enhancing effect of the polyunsaturated fatty acids (12-14). It seems that in order to achieve maximal tumor growth a requirement for essential fatty acids (EFA) must be met (12). This requirement appears to be in the range of'3% to 8% of the total kilocalorie intake for rodent mammary tumor production (12). It is believed that once the EPA requirement is met, further enhancement of tumorigenesis depends on the amount and not the type of fat, although evidence exists that may challenge these criteria (15). Recent evidence has pointed to linoleic acid and arachidonic acid as tL re rat eic abu: grog not 011‘ Door deficj effect like 5‘ DOWN.“- 2 the most necessary fatty acids for mammary tumor growth in rats (14). It is also important to mention that linoleic acid is often included in synthetic tissue culture media of normal and neoplastic cell lines in order to obtain rapid cell proliferation and optimal cell growth in vitro (16). Moreover, diets high in fat have been reported to have an immunosuppressive action, especially when high in polyunsaturated fats (l7). Diets high in polyunsaturated fatty acids (PUFA) enhance immunosuppressive therapy following renal transplantation (17). Additionally, there is evidence that feeding linoleic acid prolongs skin allograft survival in rodents (18). This immunosuppressive activity of PUFAs, primarily those high in linoleic acid, has been suggested as a possible mechanism by which polyunsaturated fats enhance tumorigenesis (19,20). Another type of polyunsaturated fat that has been studied in relation to mammary carcinogenesis is fish oil (11). Mammary tumors in rats fed diets rich in polyunsaturated long-chain fatty acids [such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)], which are abundantly found in fish oils, resulted in a marked inhibition of growth (21,22). The mechanism(s) for this tumor suppressive action has not been clearly elucidated. In most of the earlier studies, menhaden oil was used as the only source of dietary fat. Menhaden oil is a very poor source of linoleic acid and the possibility of a linoleic acid deficiency in the menhaden oil-fed animals cannot be ruled out. The effect of fish oils in the generation of different series of hormone- like substances (prostaglandins) as compared to corn oil has been a popular proposed mechanism for the suppressive action exerted by fish DI“ CO. hai sop Thu: high deri free to d1 dimet rats ‘ tUmorf cytOSt 3 oils (19-22). Although seemingly important, this extensively studied prostaglandin mechanism may not be the sole one operating in this complex process of tumor growth enhancement. It has been reported that prevention of prostaglandin synthesis has not suppressed the inhibitory effects of PUFAs (23). When inhibitors of cyclooxygenase (indomethacin) or lipoxygenase (nordihydroguaiaretic acid) were added to neoplastic cells along with the fish oil, there was no decrease in the inhibitory effects of the oil. This suggests that there is no major involvement of prostaglandins or leukotrienes in mediating fish oil inhibition of tumor growth. Another attractive mechanism that has not been adequately examined in relation to the fish oil exerted growth suppressive action on tumors is lipid peroxidation. Considering that the mammary tumor cell growth- promoting effects of fat have been closely associated with PUFAs, it is conceivable that certain PUFAs and/or their oxidative metabolites may have an important effect on mammary tumor cell proliferation (15,24). In relation to this, it is known that antioxidants are effective suppressors of lipid peroxidation radicals and/or oxygen radicals. Thus, various laboratories have added different antioxidants to diets high in PUFAs; for example, vitamin E plus selenium (25), vitamin A derivatives (26), and butylated hydroxytoluene (BHT) (27) to decrease free radical production. Interestingly, the addition of antioxidants to diets high in PUFAs has been reported to inhibit dimethylbenzanthracene (DMBA) induced mammary gland tumorigenesis in rats (25-27). This antioxidant-induced inhibitory action on mammary tumorigenesis is contrasted with reports providing evidence of a cytostatic/cytolytic action of certain PUFAs and/or their oxidative pi ti ir pr tu oi oi di tu to of co pr gr UTI- Ca fa in tin (Ml 911 3m 4 products in an in vitro (16) and in an in vivo (15,28) setting. In these experiments, PUFAs underwent lipid peroxidation, generating an increased amount of secondary products of oxidation restricting cell proliferation and/or increasing cell loss resulting in a diminished tumor growth. Given the nature of the fatty acid composition of fish oils, this mechanism seems a very attractive one to explain the fish oil exerted tumor growth suppressive action compared to rodents fed diets high in corn oil. Another important kinetic feature of tumorigenesis, in addition to tumor cell proliferation, which has not been adequately examined, is tumor cell loss. There is evidence that the increase in growth (size) of rodent mammary tumors as a result of feeding diets rich in PUFAs could not be solely accounted for by an increase in tumor cell proliferation, which suggests an important role of cell loss on tumor growth (29,30). The study of dietary fat effects on tumor growth dynamics (cell loss vs cell proliferation) has been neglected. An understanding of these kinetic phenomena might result in more effective cancer therapies. Most of the experiments designed to examine the effect of dietary fat on mammary tumorigenesis have been carried out using rodent models, in order to evaluate the effects of dietary fats on human tumorigenesis, studies utilizing human tissue are essential. Our experimental model consists of human breast tumor cell lines (MDA-M8231), an estrogen receptor negative cell line derived from pleural effusions of a patient with medullary carcinoma of the breast and (MCF-7), an estrogen and progesterone receptor positive cell line derived from a pleural effusion of a patient with an infiltrating 5 ductal breast carcinoma transplanted subcutaneously to female athymic nude mice. The nude mouse is a mutant deficient in T-lymphocytes and therefore suitable for foreign tissue grafts without rejection. This model facilitates the study of human breast cancer in an in vivo setting. It is also a means of providing important information on the influence of dietary manipulations on the in vivo growth of human breast cancer. According to the two-stage theory of carcinogenesis (31), tumors are initiated by a mutational change in the cellular genome. This stage can occur rapidly and is essentially irreversible. Subsequent proliferation of tumor cells is a slower process (promotion), and it is thought to depend considerably on local environmental conditions. This concept offers the chance of preventing or suppressing carcinogenesis by modifying the cellular environment during the promotional stage of carcinogenesis. This can occur by creating an unfavorable or even hostile environment for proliferation and growth of tumor cells. The understanding of the role of dietary fat in human tumor cell metabolism during tumorigenesis may be a very relevant step to accomplish the suppression of malignant growth. Experiments and studies conduscent to a better understanding of nutritional influences on kinetic parameters in tumor biology are of great importance. Research involving nutrition-mediated cellular changes may provide new and different avenues of effective treatment for cancer, and may serve as a pivotal stage for new approaches against other degenerative diseases. 10. 11. 12. 13. 14. 15. 6 References . Committee on Diet, Nutrition and Cancer. Assembly of Life Sciences, National Research Council: Diet, Nutrition, and Cancer. 1982, National Academy Press, Washington, D.C. . Carroll KK. Influence of diet on mammary cancer. Nutr Cancer 1981:2:232-236. . Enstrom JE. Colorectal cancer and consumption of beef and fat. Brit J Cancer 1975;32:432-439. . Blair A and Fraumeni JF, Jr. Geographical patterns of prostate cancer in the United States. J Natl Cancer Inst 1978:61zl379-1384. . Carroll KK and Khor HT. Effects of level and type of dietary fat on incidence of mammary tumors induced in female Sprague-Dawley rats by 7,12 dimethylbenz(a)anthracene. Lipids 1971:6:415-420. . Chan PC, Ferguson KA and Dao TL. Effect of different dietary fats on mammary carcinogenesis. Cancer Res 1983:43:1079-1083. . Rogers AE and Wetzel WC. Mammary carcinogenesis in rats fed different amounts and types of fat. Cancer Res 1987:41:3735-3737. . Welsch CW and Aylsworth CF. Enhancement of murine mammary tumorigenesis by feeding high levels of dietary fat: A hormonal mechanism? J Natl Cancer Inst 1983:70:215-221. . Carroll KK and Hopkins GJ. Dietary polyunsaturated fat versus saturated fat in relation to mammary carcinogenesis. Lipids 1977:14:155-158. Carroll KK. Lipid and carcinogenesis. J Environ Pathol Toxicol 1980:3:253-271. Selenskas SL, Ip MM and Ip C. Similarity between trans fat and saturated fat in the modification of rat mammary carcinogenesis. Cancer Res 1984:44zl321-1326. 1p C. Fat and essential fatty acid in mammary carcinogenesis. Am J Clin Nutr. 1987:45:218-224. Carrol] KK, Hopkins GJ, Kennedy TG and Davidson MB. Essential fatty acids in relation to mammary carcinogenesis. Prog Lipid Res. 1981:20:685-690. Sauer L and Dauchy R. Identification of linoleic and arachidonic acids as rate limiting nutrients for tumor growth in rats (abstracts). FASEB J 1988:2:(4) abstract # 3269. A858. Gonzalez MJ, Schemmel RA, Dugan L Jr, Gray J1 and Welsch CW. Dietary fish oil inhibition of human breast carcinoma growth: a function of increased lipid peroxidation. Lipids 1993:28z827-832. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. 7 Begin ME. Effects of polyunsaturated fatty acids and of their oxidation products on cell survival. Chem Phys Lipids 1987:45:269- 313. Vitale JJ and Broitman SA. Lipids and immune function. Cancer Res. 1981;41:3706-3710. Mertin J and Hunt R. Influence of polyunsaturated fatty acids in the survival of skin allografts and tumor incidence in mice. Proc Natl Acad Sci USA 1976:73:928-931. Welsch CW. Enhancement of mammary tumorigenesis by dietary fat:review of potential mechanisms. Am J Clin Nutr 1987;45:192- 202. Hopkins GJ and West CE. Effect of dietary polyunsaturated fat on the growth of a transplatable adenocarcinoma in C3H AFB mice. J Natl Cancer Inst 1977;58:753-756. Karmali RA, Marsh J and Fuchs C. Effect of omega-3-fatty acids on growth of a rat mammary tumor. J Natl Cancer Inst 1984:73:457-461. Jurkowski JJ and Cave WT. Dietary effects of menhaden oil on the growth and membrane lipid composition of rat mammary tumors. J Natl Cancer Inst 1985;74:1145—1150. Rice-Evans C and Burdon R. Free radical-lipid interactions and their pathological consequences. Prog. Lipid Res. 1993:32:71-110. Bull AW, Nigro ND, Goremoleski WA, Crissman JD and Marnett LJ. In vivo stimulation of DNA synthesis and induction of ornithine decarboxylase in rat on colon by fatty acid hydroperoxides, autoxidation products of unsaturated fatty acids. Cancer Res 1984;44:4924-4928. Horvath PM and 1p C. Synergistic effect of vitamin E and selenium in the chemoprevention of mammary carcinogenesis in rats. Cancer Res 1983:43:5335-5341. Aylsworth CF, Cullum ME, Zile MH and Welsch CW. Influence of dietary retinyl acetate on enhancement of 7,12 dimethylbenz(a)anthracene-induced rat mammary tumorigenesis by high levels of dietary fat. J Natl Cancer Inst 1986:75:339-345. King MM, Bailey DM, Gibson DD, Pitha JV and McCay PB. Incidence and growth of mammary tumors induced by 7,12 dimethylbenz(a)anthracene as related to the dietary content of fat and antioxidant. J Natl Cancer Inst 1979:53z657-663. Gonzalez MJ, Schemmel RA, Gray IJ, Dugan L, Jr., Sheffield LG and Welsch CW. Effect of dietary fat on growth of MCF-7 and MDA-MBZ31 human breast carcinomas in athymic nude mice: relationship between carcinoma growth and lipid peroxidation product levels. Carcinogenesis 1991:12:1231-1235. 29. 30. 31. Hillyard LA and Abraham S. Effect of dietary polyunsaturated fatty acids on growth of mammary adenocarcinomas in mice and rats. Cancer Res 1979:39:4430-4437. Gabor H, Hillyard LA and Abraham S. Effect of dietary fat on growth kinetics of transplantable mammary adenocarcinoma in Balb/C mice. J Natl Cancer Inst 1985:74:1299-1305. Iversen OH. Initiation, Promotion: critical remarks on the two stage theory Part 3, Chapter 10:119-126. In Theories of Carcinogenesis. Iversen OH. (ed) 1988: Hemisphere Publishing Co., Washington. REVIEW OF LITERATURE It is very well established that the level and type of dietary fat can significantly influence the development and/or growth of mammary gland tumors in experimental animals (see 1 for review). In addition the amount and/or type of dietary fat can markedly affect the growth of human breast carcinoma (cell lines) maintained in immune deficient mice (2-7). Increasing the level of fat in the diet results in an increase in mammary gland tumorigenesis (1). Dietary fats containing large quantities of omega 6-polyunsaturated fatty acids [i.e. corn oil z 50% linoleic acid (18:2n-6)] are the most effective enhancers of the tumorigenic process while fats containing high amounts of omega 3- polyunsaturated long-chain fatty acids [i.e. menhaden oil, which contain elevated concentrations of eicosapentaenoic acid (EPA, 20:5n-3) and docosahexaenoic acid (DHA, 22:6n-3)] are less stimulatory and often suppressive to mammary gland tumorigenesis. Since the mid-1930’s, fish oils have been reported to have a suppressive effect on tumor growth (8,9). Four decades later, in the early seventies, further evidence was reported on the reduction of the incidence of spontaneously-developing mammary carcinomas in female C3H mice fed diets high in fish oil (menhaden oil) when compared to mice fed diets high in corn oil or safflower oil (10). Since then, inhibition or suppression of mammary gland tumorigenesis has been achieved by feeding fish oil in carcinogen-induced rat mammary 9 10 carcinoma model (II-l7) and in a carcinogen-induced mouse mammary carcinoma model (18). In these studies corn oil or safflower oil containing diets were used as controls. Moreover, diets high in fish oil (menhaden oil) have been reported to suppress growth of BN472 transplantable rat mammary carcinoma when compared to tumor-bearing rats fed a sunflower seed/cacao butter/olive oil diet (19). Also supplementation of laboratory stock diet with fish oil (MaxEPA) has been shown to suppress growth of R323OAC transplantable rat mammary carcinoma (20). Furthermore, the growth of the IX transplantable mouse mammary carcinoma has been reported to be reduced in mice fed high dietary levels of fish oil (menhaden oil), compared to mice fed a diet high in corn oil (21). In three of these studies (ll-l3), mammary gland tumorigenesis in rodents consuming high dietary levels of fish oil (menhaden oil, 20% by weight) was substantiaIly less than that in animals consuming a diet very low in total fat (corn oil, 0.5-3.0% by weight). The fish oil diets used in these studies were rich in omega-3 long chain polyunsaturated fatty acids 20:5n-3 (EPA) and 22:6n-3 (DHA). Feeding rats a diet in which the fat content is virtually only EPA (4.7%.by weight, 20:5n-3/0.3%, 18:2n-6) substantially suppressed carcinogen-induced mammary gland tumorigenesis when compared to a diet in which the sole source of fat was linoleic acid (LA, 18:2n-6, 5% by weight) (22). ’o o - “-1.4. H 1' ‘0 I I‘ .0 - .0, ‘4 (“p o. - o. The modulatory activities of omega-3 PUFAs on eicosanoid metabolism have been the most extensively studied mechanism to explain the inhibitory or suppressive activities of omega-3 PUFAs on mammary gland tumorigenesis. PGEz, TXB2 and 6-keto-PGF1. are metabolic products of 11 the action of cycloxygenase on arachidonic acid (20:4n-6). The PGEz/PGE1 ratios in mammary tumors from fish oil (menhaden oil or Max EPA) fed rats have been reported to be lower than such ratios in mammary tumors obtained from control rats fed corn oil or sunflower seed oil diets (15,21,23). Concentrations of metabolites such as PGEz/TXBz and/or 6-keto-PGF1, have also been reported to be lower in mammary tumors of fish oil-fed rats than in tumors from control rats (15,16,21,23). These eicosanoid metabolites have also been shown to be lower in mammary tumors of rats fed a diet containing primarily EPA as its fat source compared to animals fed a diet consisting solely of linoleic acid as the source of fat (24). In addition plasma PGEz and TXBz levels have also been shown to be reduced in mammary tumor-bearing rats fed a fish oil diet compared to plasma levels in rats fed a sunflower seed oil diet (20). Therefore, these metabolites are lower in mammary tumors of rodents fed diets high in omega-3 long chain PUFAs (i.e. EPA and DHA). Despite these results, Rice-Evans and Burdon (25) reported that prevention of prostaglandin synthesis failed to suppress tumor growth inhibitory actions of fish oils. When inhibitors of cycloxygenase (indomethacin) or lipoxygenase (nordihydroguaiatetic acid) were provided along with the fish oil, no decrease in tumor growth inhibitory actions of the oil was observed. Other mechanisms have also been examined in relation to the inhibitory effects of fish oil on tumor growth. For example, rats fed a diet high in fish oil (type not specified), compared to those fed sunflower seed, had reduced liver microsomal fatty acid desaturase activities (As and A6 desaturase). No significant decrease in the 12 activity of these enzymes was observed when rats were fed a diet rich in linseed oil (26). Both A5 and .5 desaturase activities are important in the metabolism of linoleate to arachidonte, ‘5 desaturase is believed to be rate limiting for these sequential reactions. Dietary fish oil (menhaden oil) has been reported to reduce the activity of rat liver fatty acid synthase compared to a diet rich in safflower seed oil (27). Fatty acid synthase is an important lipogenic enzyme system involved in the synthesis of palmitic acid from acetyl CoA and malonyl CoA. Evidence has also been provided indicating that fish oil diets (menhaden oil), compared to corn oil diets, may decrease plasma triglycerides and free fatty acids in rats possibly by increasing the activity of skeletal muscle lipoprotein lipase (28). Lipoprotein lipase stimulates the hydrolysis of triglycerides to free fatty acids thus possibly increasing the availability of free fatty acid substrates for skeletal muscle oxidative processes. Diets high in fish oil (menhaden) have also been reported to influence hepatic phase I and phase II detoxifying enzyme systems (e.g. mixed function oxidases, glutathione-s-transferase, etc.) (29). In mammary gland chemical carcinogenesis, this modification of hepatic enzymes may be particularly relevant when fish oil diets are fed during the time period of carcinogen treatment. Modulation of hepatic enzymes can markedly influence carcinogen metabolism, thus, influencing the carcinogenesis process itself. Another mechanism that has been largely ignored is the effect of dietary fish oil on energy accumulation. In animals fed high levels of fish oil (menhaden) a reduced carcass energy and reduced adipose tissue was observed when compared to animals fed equal amounts of corn oil or 13 lard (30,31). In addition, Mermier and Baker (32) reported that fatty acids, utilized for tumor energy and growth, are derived from host tissue, and it is known that fatty acid composition of tissues is influenced by dietary fat (1). In relation to this aspect of energy efficiency, Pariza and Boutwell (33) introduced the terms metabolizable energy and recovered energy. These terms differ in that energy is lost during the conversion of metabolizable energy into recovered energy. These authors also mentioned that the extent of energy loss depends on the energy source. These terms were described studying different energy sources, for instance more energy from carbohydrate is lost during this conversion than is lost when fat is used as an energy source. Nevertheless, it seems likely that certain fats such as corn oil may have a more efficient use of energy than others (i.e. menhaden oil) hence contributing more to nourishment and growth of neoplasms than do fish oils. In accordance with this hypothetical concept, it has been reported that there is a reduced lipogenic activity plus an increased mitochondrial efficiency in rats fed menhaden oil compared to rats fed corn oil (34-36). These studies provide evidence of a menhaden oil effect on intermediary metabolism. Tisdale (37) reported that EPA can induce a blockage on the production of a metabolic factor at the level of the adipocyte involved in cancer cachexia. Fatty acids are important in the maintenance of cell membrane structure and are key determinants of membrane bound enzyme activity and receptor expression. It is recognized that an important role in the regulation of enzymatic activity and receptor expression by membranes is played by the constitutive lipids (38). In addition, the oxidation of membrane phospholipids in tissues can decrease their 14 fluidity, thus affecting membrane permeability, osmosis and activity of certain bound enzymes, receptors and transport systems (39-41). It has been reported that omega-3 fatty acids increase spontaneous release of cytosolic components from tumor cells (42). This probably occurs because of the unique shape of the omega-3 PUFAs (EPA, DHA), which when incorporated into cellular constituents of tumor cells, are able to change properties associated with the plasma membrane. The authors suggested that dietary manipulation may be used to enhance tumor cell permeability and contribute to tumor cell destruction. Tumors obtain a substantial amount of fatty acids from the host (43), which are primarily supplied from circulating free fatty acids (44), the mixture of fatty acids provided by the host (after only minor structural modifications) is incorporated into the complex lipids formed by the neoplastic cells (45). If the composition of circulating fatty acids can be sufficiently changed, the phospholipids formed will have a different fatty acid composition and possibly altered physical and functional properties. It is conceivable that the change in membrane fatty acids and its consequences can be sufficient to alter the tumor growth pattern or increase its sensitivity to certain forms of therapy. For exampIe, fatty acid modification might make the neoplastic cell more sensitive to adriamycin chemotherapy. Adriamycin has been reported to accumulate in neoplastic cells enriched with polyunsaturates (46). In addition, fatty acids (LA, EPA) have been reported to modulate gene expression in human breast cancer cells (47). Fatty acids can exert these functions directly and, therefore, may themselves be important regulators of another possible mechanism: the immune response (48). The concentration and the degree of unsaturation 15 of dietary fats have been reported to influence several indices of immune status in a number of animal models (49-56). Diets high in certain PUFAs have demonstrated immunosuppressive potential (51). Furthermore, diets high in PUFAs enhance immunosuppressive therapy following renal transplantation (57). In addition there is evidence that feeding PUFAs high in linoleic acid prolongs skin allograft survival in rodents (58). In contrast, conflicting results regarding the effects of dietary omega-3 PUFAs in animal models were reported and only limited human studies on this topic have been conducted (49-51). Nevertheless the omega-3 enhancement of certain immunological parameters has been suggested as a possible mechanism for the inhibitory action of dietary fish oil (1). W1 Yet another possible mechanism for the suppressive action exhibited by omega-3 fatty acids on tumor growth is lipid peroxidation (59). Lipid peroxidation, or more specifically, the oxidation of polyunsaturated fatty acids refers to the oxidative deterioration of unsaturated fatty acids. It is a catalytic chain reaction probably initiated and propagated by free radicals (60), aided by molecular oxygen and bivalent cations. Susceptible substrates for this radical attack in biomembranes are PUFAs containing two or more double bonds (50,61). The groups between these double bonds are particularly susceptible to hydrogen abstraction (62). The radicals formed react rapidly with oxygen to form oxy-free radicals which convert to hydroperoxides (primary products of oxidation) that can break down to a wide variety of carbonyl compounds and to other radicals capable of further perpetuating the process (60). Most primary products of 16 peroxidation are unstable in vivo (63). The high concentrations of PUFA’s in cell membranes make cells very susceptible to peroxidative damage (64,65) and ultimately the damage can be of such an extent that it can result in the death of cells, this accomplished by the interaction of peroxidation products with biomolecules of important and/or regulatory functions (39,66-69). Ingestion of a diet containing high levels of fish oil may increase the tendency for a tissue to undergo lipid peroxidation (70). Several laboratories have reported increased peroxidation in tissues of animals fed high levels of fish oil (71-77). Won Of special interest are various aldehydes which are secondary products of omega-3 fatty acid oxidation that are highly cytotoxic, capable of producing a wide variety of inhibitory effects on cell proliferation (78-88). These aldehydes are mainly formed by B-cleavage reactions of alkoxy radicals arising from hydroperoxides of PUFAs based in phospholipids, glycolipids, triglycerides and cholesteryl esters (84). Also these aldehydes have been detected in many foodstuffs (i.e. microwave food containing fat) (87). No method has been developed that completely separates and identifies all the components of the complex mixture of substances that can possibly arise of the interaction of lipid-aldehyde species. These secondary products of lipid peroxidation consist of high molecular weight polymers and decomposed products. These decomposed products are mainly low molecular weight aldehydes (83) which can be easily absorbed from the diet. Examples of these aldehydes are alkanals, alkadienals, 2-alkenals and 4-hydroxyalkanal (89,90). Other examples of cytotoxic aldehydes that can be formed in me th V3 17 relatively large quantities in vivo are 4-hydroxynonenal, hexanal and propanal (89). Of special interest is 4-hydroxynonenal, the major cytotoxic product of lipid peroxidation (84), which has been reported to suppress the expression of specific genes involved in the metabolic cascade that leads to cell proliferation (88). Although there are a variety of enzymes (i.e. aldehyde dehydrogenases) that evolved to metabolize aldehydes to less reactive forms (i.e. carboxylic acids), these species are still formed in vivo in enough quantities to modulate various physiological and/or biochemical functions (87). In addition to some of these mentioned, other aldehydes capable of producing a chromagen in reaction with 2-thiobarbituric acid (thiobarbituric reactive substances, TBARS) which can be measured by the 2- thiobarbituric acid assay for detection of lipid peroxidation (TBA assay) are 2-hexanal, 2,4-hexadienal and malondialdehyde (MDA). These products, especially MDA, have been reported to be cytotoxic to cell lines at physiological concentrations (90). MW; MDA is a secondary product of PUFA peroxidation and is most frequently measured by the TBA assay (91,92). The TBA assay is used for evaluating the degree of lipid peroxidation and is one of the most commonly used methods for detection of lipid peroxidation products (91,93). The TBA assay is a colorimetric test in which a pink chromagen is formed by the condensation of 2 molecules of TBA and 1 molecule of MDA (94). The MDA measurement is an indicator of the secondary products of peroxidation in the sample in question: the greater the number of double bonds in a PUFAs the greater the chances of forming more MDA molecules plus other secondary reactive substances. m al al as is rei com; qUan m6ta enzy, DTQSE COmpo DI enzflne comple 18 MDA can react with amino and sulfhydryl groups (SH) of RNA, DNA and proteins (39,95) causing inter and/or intramolecular linkages that can lead to inactivation or damage of these biomolecules. Also MDA has been reported to inactivate DNA polymerases directly by blocking SH groups (69). Furthermore, it has been documented that MDA can accumulate in sufficient quantity in cells to exert various biological negative effects as the ones mentioned above (95). During lipid peroxidation other compounds besides MDA (other aldehydes) are formed which may have a similar absorbance and which may also be capable of inhibiting cell proliferation by the same mechanisms as MDA (63,96). These products may be detected by the TBA assay. This is the main reason why secondary products of peroxidation are now referred to as TBARS instead of MDA as it was referred to in the past. In addition it should be mentioned that TBARS can also be formed from monoxidized PUFAs, carbohydrates and amino acids (97). L1n1d_Eeroxination_and_1umor_§routh Various factors may affect the oxidation of unsaturated lipids in complex biological tissues. Among these are pH, light, temperature, quantity of antioxidants present in the tissue, quantity and nature of metal ions present in the tissue, efficiency of oxidation inhibitory enzymes (glutathione peroxidase, catalase, superoxidase dismutase), presence of reducing agents in tissue membranes and fatty acid composition of the tissue (59). Despite the variety of protective mechanisms [such as inhibitory enzymes and antioxidants (98)] against peroxidation, it still occurs in complex biological systems. If lipid peroxidation overwhelms the normally efficient cellular protective mechanisms, it can then cause gen pru fac eff can: this impo of t deco: decon reduc 0f ce Prolii Va additi death PUFAs, ”Ode oi maJ'or r (41,120 is dual: active 5. IYdrogen IIdroper pr ”agate that II‘On 19 genotoxicity or cytotoxicity and subsequently cell death (99). It is prudent to state that the interplay between enhancing and inhibiting factors of peroxidation is what probably determines its extent and its effect on biological systems. It has been documented that various PUFAs are effective in killing cancer cells in vitro (69,81,100-118) but the specific mechanism of this action is yet to be determined (106). Peroxidation may be a very important factor in PUFA cytotoxicity and also in determining the rate of tumor cell proliferation (100). Peroxidation of lipids followed by decomposition of the peroxides into aldehydes or other secondary decomposition products may be a crucial step on the path leading to a reduction of the rate of cell proliferation and/or increasing the rate of cell loss of tumors. Such products have been shown to restrict cell proliferation and to cause cell death (119). Various investigators (81,104,105,116-118) reported that the addition of iron salts to cell cultures increases the rate of cell death and TBARS number in cultured tumor cells fed high levels of PUFAs, suggesting the involvement of lipid peroxidation products in the mode of action. It is well known that transition metals can exert a major role in oxidative damage occurring in biological systems (41,120,121). In the case of lipid peroxidation, the metal involvement is dual: it can first react with molecular oxygen generating oxygen active species able to initiate lipid peroxidation by abstracting a hydrogen atom from the PUFAs; second, it can react directly with the hydroperoxide generating organic radicals that can initiate and propagate the peroxidation reaction (41). Also it is well established that iron and copper salts are prooxidant catalysts (120,121). At 37'C and dec as bre. pH i ferr cont and inve: and a to ad additi et al. lipid et al. than in Teoorts utiliza' earlier 1 variet exerted I initiatin Contrary lallgnant e"hence p . 20 and a pH near 7.4, iron may become critical in promoting hydroperoxide decomposition to MDA. In this environment, iron serves the dual role as both initiator of lipid peroxidation and promoter of lipid peroxide breakdown (122). It is conceivable that in an environment with a lower pH (<6), as can possibly be found in tumors, iron may dissociate from ferritin and/or hemoglobin and increase aldehyde formation (123). In contrast, the addition of alpha-tocopherol reduces both cell killing and levels of TBARS in cell culture (101,105,125). Also, several investigators (126-128) reported that there is less lipid peroxidation and a higher level of antioxidants in the cancerous tissue as compared to adjacent normal tissue in breast cancer patients (113,128-130). In addition, a rather interesting observation was reported by L’Abbe’ et al. (131), in which feeding a high fish oil diet induced higher lipid peroxidation in female rats compared to male rats. Meydani et al. (132) reported higher concentrations of plasma TBARS in older than in younger women supplemented with omega-3 fatty acids. These reports suggest a sex-age associated difference in incorporation or utilization of omega-3 fatty acids. In relation to antioxidants, earlier studies found antioxidants inhibited chemical carcinogenesis in a variety of tissues (see 133 for review). This antitumor activity exerted by antioxidants was probably due to an interference with initiating or promoting events in chemical carcinogenesis (134,135). Contrary to this mechanism, the addition of antioxidants to already malignant cells in the presence of PUFAs seems to protect the cells and enhance proliferation (100,105,124). Furthermore, there is some evidence of antioxidants enhancing cell proliferation (136,137). CI ki in an le. (13 PUF per caus (118 evide cell non-p antio: neOpIa report Oti cell pr '“ich n This Can in the c< e'"bi‘yonic know, thal 21 Evidence from cell culture studies also indicates that certain PUFAs such as eicosapentaenoic, docosahexaenoic and arachidonic acid have been effective in killing cancer cells, and this effect is correlated with intracellular TBARS levels (81,100,101,105,106); this killing of malignant cells is accelerated by iron and copper while inhibited by antioxidants (105,122). The generation of oxygen radicals and the stimulation of lipid peroxidation, seem to be necessary steps leading to cell death, in these experiments. Also, Burns and Wagner (138) reported that there is a heightened susceptibility of fish oil PUFA-enriched neoplastic cells to generate ethane during lipid peroxidation (138). Does cell death cause lipid peroxidation or does lipid peroxidation cause cell death? This question has been addressed by Takeda et al. (118) and Benchekroun et al. (139). They reported that there was no evidence of any substantial rise in lipid peroxidation in response to cell killing by other mechanisms such as temperature, pH changes and non-peroxidation drug induced cell death. The addition of an antioxidant suppressed the formation of lipid peroxidation in neoplastic cells and the killing effect was inhibited (118). This report has been substantiated by results of our laboratory (158). Other clues of the importance of lipid peroxidation in relation to cell proliferation can be derived from data about regenerating liver in which the content of lipid peroxidation products are low (140-142). This can suggest a possible involvement of lipid peroxidation products in the control of cell division (126,143). It is documented that embryonic tissue has a very low peroxidation content and it is also known that certain tumor cells resemble embryonic cells; also highly unc whi 144 mar the mect cell cell inhil Tubul nitot which DNA 5) (149), Of per: the ca; suscept ildehyd. and atta Site. I Messengel cell inju single fa Match-c AlthOUgh 22 undifferentiated tumor cells have a very low degree of peroxidation which seems to be inversely related to the growth rate of tumors (113, 144-147). Moreover, rapidly growing normal tissue such as testis, bone marrow and intestinal epithelium are resistant to peroxidation and their lipid peroxidation product content is very low (120). Conceivably, lipid peroxidation can inhibit tumor growth by several mechanisms; these include (1) interference with various phases of the cell cycle by damaging important cell compounds that would enable the cell to proceed normally through its cycle. For example, the inhibition of tubulin production by secondary products of peroxidation. Tubulin is the main functional protein involved in the formation of the mitotic spindle (148). (2) Inhibition of polyamine synthesis (148) which can directly affect the cell division process. (3) Increasing DNA synthesis time and decreasing the growth fraction of the tumor (149), thereby increasing overall cell cycle time. Secondary products of peroxidation have been shown to be diffusible (150,151), possessing the capability of traveling along the cells producing damage to susceptible sites through their pathway. Nevertheless some of these aldehydes are fairly stable (151) which permit them to escape the cell and attack susceptible targets far away from its originating initial site. In this manner these aldehydes can be considered second toxic messengers of the initial free radical event (89). However, tissue or cell injury caused by lipid peroxidation cannot be attributed to a single factor, since various radical species are formed with different cytotoxic, mutagenic, genotoxic and carcinogenic potential (87). Although lipid aldehyde levels in neoplastic cells or tissue may equate increased levels of absorption or production of aldehydes, it may also lean an a There conpositi as compai low leve' initiate levels 0 conparat the low increasc of lipii related (18:1, chelati vivo 5; fatty ; effect. perOXli When f1 1% 23 mean an alteration in removal pathways (84). WW There is evidence that tumor cells have an abnormal lipid composition (i.e. lower phospholipid content and low levels of PUFAs) as compared to their normal counterparts (60). Tumor cells have also a low level of enzymes of the cytochrome P450 system. These enzymes can initiate and propagate lipid peroxidation (60). In addition, higher levels of antioxidants have been reported in tumors as compared to comparable normal tissue (60). The low rate of lipid peroxidation and the low cytochrome P450 mediated activity can be associated with an increased rate of cell division (144). Apparently, secondary products of lipid peroxidation are inversely related to tumor growth. In related studies (152-154), it was reported that LA and oleic acid (18:1, n-9), in vitro, partially inhibited lipid peroxidation by chelating available free iron. The importance of this report to an in vivo setting remains to be determined. The content of nonesterified fatty acids in vivo may be insufficient to obtain this interesting effect. Two different investigators (80,155) have shown that lipid peroxidation products were too low to be detected or decreased in vivo when feeding diets high in linoleic acid content. WWW Cancer is a multistep, polygenic process involving various events. The induction phase is composed of two steps, initiation and promotion. At the initiation step, a change occurs in the cell genes resulting in a deregulation of the normal pathways. The cell is then endowed with the capacity to proliferate without control. In other words, the cell is transformed or 'initiated'. This occurs after a mutation, an nplificati repressor g step and fi against Illi Generally l the pronot' apronoter inducing p. cancer pro experiment onto the c promotion tumor cell hypothesiz Peroxidati growth (12 the Proces addition, Conceivab] SDGcific p enhancers ; ””5 conce ‘5 formed dependent ‘ A Very in "hich tl 24 amplification, a translocation of a cellular oncogene or a loss of a repressor gene. Chemical or physical mutagens can be involved in this step and free radicals are a common link in this process. Protection against the initiation phase can be provided by antioxidants. Generally the transformation is completed by successive mitosis during the promotion step. Any mechanism inducing the cell cycle will act as a promoter. Certain reactive free radicals are known to play an inducing part in cell activation (127) and are expected to act as cancer promoters. However, to further complicate the picture, various experiments in order to avoid the toxic effect of the free radicals onto the cell, the use of antioxidants was necessary to ensure the promotion phase (156). In tumor progression (the phase during which tumor cells become organized as a growing mass), it might be hypothesized that a senescent environment characterized by a high lipid peroxidation and a low antioxidant level may negatively influence tumor growth (127). Thus, it seems that free radicals can differently affect the process of carcinogenesis depending on the phase considered. In addition, to even further increase the complexity of this issue, it is conceivable that different species of free radicals may influence a specific phase of carcinogenesis in different ways. Some may be enhancers of cell proliferation while others can be highly toxic. If this concept is correct, then the importance is based on which species is formed in larger amounts by which dietary fat (which might be dependent on the specific composition of fatty acids). ummnmmmm A very interesting finding was reported by Gabor and Abraham (157) in which they state that the cancer-promoting effect exhibited by corn pH is m the high fish oil years ti but alsc dietary and 159 growth I (6,7,59 and inc: dietary and doc these h their c respect in puri it has diets h (7.160, Rec importa Sensm SEC a Cance the C81 may be c“Donn 25 oil is mainly related to a decrease in tumor cell loss as compared with the higher cell loss resulting in tumor of rodents fed a diet high in fish oil. It has become increasingly evident over the past several years that tumors are not just a reflection of the rate of cell growth but also the rate of cell loss. Many reports utilizing high levels of dietary fish oils have demonstrated a decrease in tumor growth (see 1 and 159 for reviews). Dietary fish oils may decrease the rate of tumor growth by increasing lipid peroxidation products in tumor cells (6,7,59,158,159), which can decrease the growth fraction of the tumor and increase the percent of cell loss. Animals fed high levels of dietary fish oil will have a higher intake of eicosapentaenoic (EPA) and docosahexaenoic (DHA) acid that results in a higher content of these highly unsaturated fatty acids in the phospholipid fraction of their cell membrane (160). These fatty acids have 5 and 6 double bonds respectively. It has been documented that the autoxidation of fish oil in purified diets is rapid, within 24 hours (7,162-164). Furthermore, it has been suggested that the increase in tissue TBARS after feeding diets high in PUFAs are not formed in vivo but absorbed from the diet (7,160,161). Recently it has been reported that lipid peroxidation plays an important role in tumor cell eradication by hematoporphyrin derivative sensitized phototherapy (165-166). Secondary products of peroxidation when constantly in contact with a cancerous cell may induce profound damage, making it impossible for the cell to survive. The inhibitory or suppressive effect of fish oil may be due, at least in part, to lipid peroxidation formation of toxic compounds. Cell Ti kinet‘ neces: of th: loss these a suc T proli of ce impor Proli in no in no Which tissu resul repla deVel. exCES: 26 WWII The growth rate of tumors is influenced by diverse cell cycle kinetic factors (I for review), namely: 1) cell cycle time (time necessary for the tumor cells to reproduce); 2) growth fraction (part of the tumor in which tumor cells are actively proliferating); 3) cell loss (tumor cells that die or leave the tumor). The understanding of these kinetic factors and their interplay is of ultimate importance if a successful treatment approach against cancer is to be accomplished. To date, cancer therapy has focused mainly on targeting cell proliferation with limited results in most tumors, while the phenomenon of cell loss has been largely ignored. However, cell loss is an important factor in determining the dynamic state of cell proliferation. Cell loss is a biological phenomenon which takes place in normal and neoplastic tissues (167). Usually an equilibrium exists in normal tissue of adults between cell proliferation and cell loss which results in a steady state growth pattern without any increase in tissue size. However, when cell proliferation exceeds cell loss, the result is an increase in size (hyperplasia). The innate ability to replace cells in a tissue is a highly successful evolutionary development (168), but perturbations in these processes can cause excessive loss or over-production of cells. This review will focus on the importance of cell loss to solid (carcinomas, sarcomas, adenomas) tumor growth. Obviously, if cell loss (by means of cell death) can be enhanced in a solid tumor in such a way that it will overcome cell proliferation; solid tumor regression can be accomplished. Tumor exfoliati The relat site of t growing i vessel, e chance it organ suc oetastasi circulati is the 01 circulati (171). I transplai Primary 1 doubling by Ietasi growth. Viable pa SUCh as 1 sloughim 1169). 1 ea' Cell of the m death (a' spatial . 27 Tumor cell loss can occur through various mechanisms such as exfoliation, metastasis, differentiation and necrosis (cell death). The relative contribution of the various modes of loss depends on the site of the tumor and its histological type. Tumors that have surfaces growing into a lumen such as in the peritoneum, pleurage, major blood vessel, etc. or growing at the surface of the body have a greater chance for cell loss by exfoliation than a tumor enclosed within an organ such as liver or kidney (169). Another mechanism of cell loss, metastasis, effectively promotes overall cancer spread. The circulating cancer cells have several effects on the host, one of which is the overcoming of immune resistance (170). However, most circulating cancer cells die in their journey throughout the body (171). But, if the metastatic cells are established, they behave like transplants that have the capacity to grow faster than the original primary tumor because of a shorter or absent lag phase or a shorter doubling time (172). Nevertheless, there is no evidence that cell loss by metastasis is a predominant mechanism affecting primary tumor growth. Differentiation of cells within tumors can provide another viable pathway for cell loss. For example, well differentiated tumors such as those of the skin and colon lose cells by keratinization or sloughing them off to the lumen of crypts (exfoliation), respectively (169). W Cell death is encountered in early embryogenesis since it is part of the morphogenesis of several organs (168). The programmed cell death (apoptosis) in the embryo is a vital process providing a strict spacial and temporal control. Its failure may result in congenital abnormal In ti and/or 3 However. cell 105 mechaniSI tumor ma in cell . Counmn p associatu lysosoma‘ Various 1 the ChI‘Ol El 0 : Disc1 doubling focus On growth r; n“Ch $101 of the u been stag 0f cell 1 the follo grow; is "he” can; of "mm I'll, this 28 abnormalities of varying severity (168). In tumor growth, there appears to be a reduction of the death index and/or a prolongation of the average life span of the cells (172). However, cell death in tumors appears to be the most relevant form of cell loss. Cell death in solid tumors can be promoted by numerous mechanisms, for example: insufficient blood supply with increasing tumor mass (169), immune reactions against the tumor (173), anomalies in cell division (174), and/or local effects of toxic products (59). Common pathways for cell injury that may reach a lethal outcome are associated with respiratory failure, inhibition of ATP synthesis, lysosomal release and alteration of membrane permeability (175). Various of these events may have a genetic component which may affect the chromosomes and ultimately DNA. Management Discrepancies between tumor cell doubling time and the volumetric doubling time of the tumor containing those cells led investigators to focus on the possibility of a negative effect of cell loss on tumor growth rate. Specifically, tumors are able to double in volume at a much slower rate than one would expect by monitoring cell cycle times of the tumor cells. The importance of cell loss in tumor growth has been stagnated by its measurement difficulty. An accurate assessment of cell loss in tumors has proven to be a very difficult task due to the following: it can only be measured in situations in which a tumor grows as a well defined mass which can be measured (176). In general, when carcinomas reach a large size this is often associated with a core of necrosis (dead center) and tumor growth is restricted to the outer rim, this would underestimate cell death substantially; especially if cell loss ‘ inrdathv different limhatior tumors. loss fact cells as hmnr vol of growtl A ce neither lost, tl producec regress¢ affectii time. increas IHIlUEn be 9m factor dUE to tUmor ‘ undere: necrota 29 cell loss is equated to cell death. Additional sources of variability in relation to tumor growth are that tumors proliferate at very different rates in different areas of the same tumor. In spite of this limitation, there have been various attempts to measure cell loss from tumors. In order to estimate cell loss, Steel (176) developed a cell loss factor method. The cell loss factor measures the rate of loss of cells as a fraction of the rate at which cells are being added to the tumor volume by mitosis. Steel's cell loss factor expresses the loss of growth potential by the tumor on the basis of production. A cell loss factor of 100% describes a tumor in which there is neither growth nor regression. If more cells are being generated than lost, the cell loss factor is less 100% and if more cells are lost than produced the cell loss factor is more than 100% and the tumor regresses. Tumor growth is affected by small fluctuations in elements affecting growth as well as by overall changes in growth patterns with time. Estimates of cell loss factor usually differ over time as tumors increase in size. There are a number of elements capable of influencing the cell loss factor calculations which can only, at best, be guides to the degree of cell loss. The estimations of the cell loss factor is an approximation and must be interpreted with reservation due to the impreciseness of the method. The simple measurement of tumor volume doubling time in the calculation of cell loss factor will underestimate cell loss due to the inclusion of blood, tissue fluid and necrotic material in the measurement of tumor volume. Impressions of the magnitude of cell loss has been obtained by comparing potential doubling times of tumors (expected doubling time without cell loss) determined from H3-thymidine labelling indices (176-178). In other studies the exp factors These r methodo reutili prolife tumors. Cel loss of deoxyur princi; associa wide di means I these 1 l"f0rmi that f¢ Provide Contra: thymjd. the 11: Elimin; p°DUla1 are Bf1 30 studies using human carcinomas, there was a marked disparity between the expected and observed doubling times, which suggested cell loss factors of around 60% (179) and in the region of 90% or higher (180). These results again point out the imprecision in the cell loss methodology (utilization of cell loss factor and isotopes of high reutilization potential). Also affected by local variations in tumor proliferative activity and varying distribution of cell cycle times in tumors. Cell loss has also been studied with some success by following the loss of radioactivity from cells labelled with radioactive 1125- deoxyuridine (178-188). However, this procedure follows the same principle of the H3-thymidine and also had similar problems as those associated with H3-thymidine labelling technique. For example, still a wide dispersion of individual tumor doubling times among their group means providing an excessive variability. It has been argued that these two methods (H3-thymidine and Ile-deoxyuridine) can provide information about different aspects of cell loss. Begg (183) reported that for tumors with a high cell loss rate, the H3-thymidine method provides more meaningful data than the Ilzs-deoxyuridine technique. In contrast, Dethlefsen et al. (184) considered that the use of H3- thymidine was well suited to the study of intratumor events, whereas the Ile-deoxyuridine was considered better suited to investigate the elimination of intact cells and necrotic products from a tumor population regardless of rate. Both Ilzs-deoxyuridine and H3-thymidine are efficiently incorporated into DNA of tumor cells, but reutilization of I125_ After in cells ur deterior limiting equate 1 death tl deoxyur' cells); they ar. labelle« excrete. and 5) method Althoug 'noise' cItocid Suitab] Vlvo. Showing Hell Wi dUe to ridiQSQ Cells b, Cell 10 “"1919“, 31 of Ilzs-deoxyuridine is much lower than that of H3-thymidine (185-188). After incorporation into DNA, Ilzs-deoxyuridine remains bound within cells until they die (178,186). When a labelled cell dies, rapid deterioration and excretion of the DNA breakdown products occur limiting the reutilization by other cells to a minimum. In order to equate the loss of 1125 activity to naturally occurring tumor cell death the following assumptions must be accepted as true: 1) the 1125- deoxyuridine is associated exclusively with the DNA (mainly of tumor cells); 2) Ilzs-deoxyuridine is not released from labelled cells while they are alive; 3) Ilzs-deoxyuridine does not damage or kill the labelled tumor cells; 4) Ilzs-deoxyuridine is rapidly broken down and excreted (mostly in the urine) following the death of labelled cells; and 5) chemical effects of IlZS-deoxyuridine are minimal. Hence, this method has the advantage of not perturbing the tumor in question. Although this method, when used in vivo, possesses a high degree of 'noise' (interfering variables) which makes it unfit to measure any cytocidal effect quantitatively. Nevertheless, it seems to be the most suitable way of determining cell loss occurring in solid tumors in vivo. It has been reported (189) that the volumes of carcinomas showing shrinkage 24 hours after being irradiated, did not correlate well with their cell loss factor numbers. The discrepancy is probably due to the fact that tumor regression depends not only on the radiosensitivity of the tissue but also on the rate of removal of dead cells by autolysis and phagocytosis. In order to attain a significant cell loss to incur into tumor regression, cell death needs to be complemented by an effective removal of dead cells. Tumor regression also de brings are not mass of tumor v tumor v loss or tumor 9 provide it is ; measure Sol death. loss It cell an Chain I unfavov death 1 Previol oil su; deCTeas It kinetic 32 also depends on blood supply which reabsorbs products of autolysis and brings phagocytes into contact with dead cells. When cells in a tumor are not effectively removed, they remain to form a necrotic mass. The mass of necrotic tissue is included with the live tissue when measuring tumor volume; thereby overestimating tumor volume. This suggests that tumor volume is not a useful measurement in the determination of cell loss or cell loss factor. However, tumor volume as used in studies of tumor growth kinetics is a simple straightforward procedure which provides an insight into tumor growth despite its limitation. Because it is possible to measure tumor volume in vivo, it can be used to measure tumor growth over time which cannot be done by other means. Solid tumors have high rates of cell loss (14) especially cell death. It remains to be determined how we can take advantage of cell loss to be used in solid tumor eradication. The manipulation of tumor cell membrane with nutritional substances (for example, omega-3 long chain polyunsaturated fatty acids) which may provide a hostile or unfavorable environment for tumor cell growth thereby promoting cell death is an attractive approach to this problem. As mentioned previously (151,190), Abraham et al. have proposed that dietary fish oil suppression of mammary tumor growth in mice is not a function of decreased cell proliferation but a function of increased cell death. It is conceivable that a better understanding of tumor growth kinetic factors may enable us to shift the intricate balance from tumor growth stimulating factors to inhibiting ones. This action may prove to be a pivotal step in controlling the neoplastic expression and improving the response to therapy. This principle might be relevant in improving current therapeutic approaches and in developing or designing 33 newer and complementary therapeutic strategies against cancer. The biological modification of cell loss instead of cell proliferation might prove to be a valuable turn in the strategy used against cancer. In conclusion, a better understanding of tumor growth dynamics (cell loss and cell proliferation) and its dietary manipulation studied in an vivo model of human tumors may provide important clues for effective cancer therapy. 8919121153.: 1. Welsch CW. Relationship between dietary fat and experimental mammary tumorigenesis: A review and critique. Cancer Res. 1992, 52: 20405-20485. Pritchard GA, Jones DL and Mansel RE. Lipids in breast carcinogenesis. Brit. J. Surg. 1989, 76:1069-1073. Blank EW and Ceriani RL. Fish oil enhancement of 131I-conjugated anti-human milk fat globule monoclonal antibody experimental radioimmunotherapy of breast cancer. J. Steroid Biochem. 1989, 34:149-153. Borgeson CE, Pardini L, Pardini RS and Reitz RC. Effects of dietary fish oil on human mammary carcinoma and on lipid- metabolizing enzymes. Lipids 1989, 24:290-295. Gabor H, Blank EW and Ceriani RL. Effect of dietary fat and monoclonal antibody therapy on the growth of human mammary agengcarcinoma MX-l grafted in athymic mice. Cancer Lett. 1990, 5 :1 3-178. Gonzalez MJ, Schemmel RA, Dugan L, Gray J1 and Welsch CW. Dietary fish oil, inhibition of human breast carcinoma growth: a function of increased lipid peroxidation. Lipids 1993, 28:827- 832. Gonzalez MJ, Gray JI, Schemmel RA, Dugan L and Welsch CW. Lipid peroxidation products are elevated in fish oil diets even in the presence of added antioxidants. J. Nutr. 1992, 12:2190-2195. Haven FL. Effect of dietary fats on growth and composition of tumors. Proc. Soc. Exptl. Biol. Med. 1935, 32:134-137. Haven FL. The effect of cod liver oil on tumor growth. Am. J. Cancer 1936, 27:95-98. 10. 11. 12. 13. 14. 15. 16. 17. 18. ccnl 21. T39 22. 10. ll. 12. 13. 14. 15. 16. 17. l8. 19. 20. 21. 22. 34 Harmon D. Free radical theory of aging: effect of the amount and degree of unsaturation of dietary fat on mortality rate. J. Gerontol. 1971, 26:451-457. Jurkowski JJ and Cave WT. Dietary effects of menhaden oil on the growth and membrane lipid composition of rat mammary tumors. J. Natl. Cancer Inst. 1985, 74:1145-1150. Carroll KK and Braden LM. Dietary fat and mammary carcinogenesis. Nutr. Cancer 1985, 6:254-259. Braden LM and Carroll KK. Dietary polyunsaturated fat in relation to mammary carcinogenesis in rats. Lipids 1986, 21:285- 288. Abou-El-Ela SH, Prasse KW, Carroll R, Wade AE, Dharwadkar S and Bunce OR. Eicosanoid synthesis in 7,12-dimethylbenzanthracene- induced mammary carcinomas in Sprague-Dawley rats fed primrose oil, menhaden oil or corn oil diet. Lipids 1988, 23:948-954. Abou-El-Ela SH, Prasse KW, Farrell RL, Carroll RW, Wade AE and Bunce OR. Effects of D,L-2-difluoromethylornithine and indomethacin on mammary tumor promotion in rats fed high n-3 and/or n-6 fat diets. Cancer Res. 1989, 49:1434-1440. Karmali RA, Donner A, Gobel S and Shimamura T. I. Effect of n-3 and n-6 fatty acids on 7,lZ-dimethylbenzanthracene-induced mammary tumorigenesis. Anticancer Res. 1989, 9:1161-1168. Ronai Z, Lau Y and Cohen LA. Dietary n-3 fatty acids do not affect induction of Ha-RAS mutations in mammary glands of NMU- treated rats. Mol. Carcinogenesis 1991, 4:120-128. Cameron E, Bland J and Marcuson R. Divertent effects of omega-6 and omega-3 fatty acids on mammary tumor development in C3H/Heston mice treated with DMBA. Nutr. Res. 1989, 9:383-393. Kort WJ, Weijma 1M, Bijma AM, Van Schalkwijk WP, Vergroesen AJ and Westbroek DL. Omega-3 fatty acids inhibiting the growth of a transplantable rat mammary adenocarcinoma. J. Natl. Cancer Inst. 1987, 79:593-599. Karmali RA, Marsh J and Fuchs C. Effect of omega-3 fatty acids 9; growth of a rat mammary tumor. J. Natl. Cancer Inst. 1984, :457-461. Gabor H and Abraham 5. Effect of dietary menhaden oil on tumor cell loss and the accumulation of mass of a transplantable mammary adenocarcinoma in Balb/C mice. J. Natl. Cancer Inst. 1986, 76:1223-1229. Takata T, Minoura T, Takada H, Sakaguchi M, Yamamura M, Hioki K and Yamamoto M. Specific inhibitory effect of dietary eicosapentaenoic acid on N-nitroso-N-methylurea-induced mammary 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35 carcinogenesis in female Sprague-Dawley rats. Carcinogenesis 1990, 11:2015-2019. Christiansen EN, Lund JS, Rortveit T and Rustan AC. Effect of dietary n-3 and n-6 fatty acids on fatty acid desaturation in rat liver. Biochem. Biophys. Acta 1991, 1082:57-62. Kamano K, Okuyama H, Konishi R and Nagasawa H. Effects of a high-linoleate and a high-gamma-linoleate diet on spontaneous mammary tumourigenesis in mice. Anticancer Res. 1989, 9:1903- 1908. Rice-Evans, C and Burdon R. Free radical-lipid interactions and their pathological consequences. Prog. Lipid Res. 1993, 32:71- 110. Karmali RA, Chao CC, Basu A and Modak M. II. Effect of n-3 and n-6 fatty acids on mammary H-ras expression and PGE2 levels in DMBA-treated rats. Anticancer Res. 1989, 9:1169-1174. Baltzell JK, Wooten JT and Otto DA. Lipoprotein lipase in rats fed fish oil: Apparent relationship to plasma insulin levels. Lipids 1991, 26:289-294. Wade AE, Bellows J and Dharwadkar S. Influence of dietary menhaden oil on the enzymes metabolizing drugs and carcinogens. Drug-Nutrient Interact. 1986, 4:339-347. Jones PJH. Effect of fatty acid composition of dietary fat on energy balance and expenditure in hamsters. Can. J. Physiol. Pharm. 1989, 67:994-998. Jandacek RJ, Hollenbach EJ, Holcombe BN, Kuehlthau CM, Peters JC and Taulbee JD. Reduced storage of dietary eicosapentaenoic and docosahexaenoic acids in the weanling rat. J. Nutr. Biochem. 1991, 2:142-149. Parrish CC, Pathy DA, and Angel A. Dietary fish oils limit adipose tissue hypertrophy in rats. Metab. Clin. Exptl. 1990, 39:217-219. Mermier P and Baker N. Flux of free fatty acids among host tissues ascites fluid and Ehrlich ascites carcinoma cell. J. Lipid Res. 1974, 15:339-351. Pariza MW and Boutwell RK. Historical perspective: calories and energy expenditure in carcinogenesis. Am. J. Clin. Nutr. 1987, 45:151-156. Pan JS and Berdanier CD. Thyroxine counteracts the effect of menhaden oil on fatty acid synthesis in BHE rats. Nutr. Res. 1990, 10:461-466. 35. 36. 37. 38. 39. 4Q. 41. 42. 43. 44. 45. 46. 47 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. 47. 36 Kim MJ and Berdanier CD. Influence of menhaden oil on mitochondrial respiration in BHE rats. Proc. Soc. Exp. Biol. Med. 1990, 192:173-176. Bunce OR, Abou-El-Ela SH, Berdanier CD. Long term feeding of corn oil, beef tallow or menhaden oil and eicosanoid levels in BHE/cab rats. Nutrition 1992, 8:421-425. Tisdale MJ. Mechanism of lipid mobilization associated with cancer cachexia: Interaction between the polyunsaturated fatty acid, eocosapentaenoic acid and inhibitory guanine nucleotide- regulatory protein. Prost. Leuk. Ess. Fatty Acids 1993, 48:105- 109. Burlakova EB, Molochkina EM and Palmina NP. Role of membrane lipid oxidation in control of enzymatic activity in normal and cancer cells. In: Webber G (ed) Advances in enzyme regulation 1981, 18:163-179, Academic Press, London. Roubal WT and Tappel AL. Damage to proteins, enzymes and amino acids by peroxidizing lipids. Arch. Biochem. Biophys. 1966, 113:5-8. Murphy MG. Dietary fatty acids and membrane protein function. J. Nutr. Biochem. 1990, 1:68-77. Tadolini B, Fiorentini D, Motta P, Cabrini L and Sechi AM. The influence of phospholipid polar head on the lipid hydroperoxide dependent initiation of lipid peroxidation. Biochem. Int. 1992, 26:275-285. Jenski LJ, Sturdevant LK, Ehringer W0 and Stillwell W. W3 fatty acids increase spontaneous release of cytosolic components from tumor cells. Lipids 1991, 26:353-358. Spector AA. Fatty acid metabolism in tumors. Prog. Biochem. Pharmacol. 1975, 10:42-75. Spector AA. The importance of free fatty acid in tumor nutrition. Cancer Res. 1967, 27:1580-1586. Spector AA and Burns CP. Biological and therapeutic potential of membrane lipid modification in tumors. Cancer Res. 1987, 47:4529-4537. Burns CP and North JA. Adriamycin transport and sensitivity in fatty acid-modified leukemic cells. Biochem. Biophys. Acta 1986, 888:10-17. Tiwari RK, Mukhopadhyay B, Telang NT and Osborne MP. Modulation of gene expression by selected fatty acids in human breast cancer cells. Anticancer Res. 1991, 11:1383-1388. 48. 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. S9. 60. 61. 52. 63. 48. 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. 59. 60. 61. 62. 63. 37 Zurier RB. Fatty acids, inflamation and immune responses. Prost. Leuk. Ess. Fatty Acids 1993, 48:57-62. Meade CJ and Mertin J. Fatty acids and immunity. Adv Lipid Res. 1978, 16:127-165. Gross R1 and Newberne PM. Role of nutrition in immunologic function. Physiol Rev 1980, 60:188-288. Vitale JJ and Broitman SA. Lipids and immune function. Cancer Res. 1981, 41:3706-10. Beisel WR. Single nutrients and immunity. Am J. Clin. Nutr. 1982, 35(suppl):417-68. Burr MI. The role of lipids in the regulation of the immune system. Prog Lipid Res. 1983, 22:257-87. Johnston DV and Marshall LA. Dietary fat, prostaglandins and the immune response. Prog. Food Nutr. Sci. 1984, 8:3-25. Simopoulos AP. The role of W-3 fatty acids in health and disease: dietary implications. Nutr. Today 1988, 23:12-8. Hwang D. Essential fatty acids and immune response. FASEB J. 1989, 3:2052-61. Donadio JV Jr. Omega-3 polyunsaturated fatty acids: A potential new treatment of immune renal disease. Mayo Clin. Proc. 1991, 66:1018-1028. Mertin J and Hunt R. Influence of polyunsaturated fatty acids in the survival of skin allografts and tumor incidence in mice. Proc. Natl. Acad. Sci. USA. 1976, 73:928-931. Gonzalez MJ. Lipid peroxidation and tumor growth: an inverse relationship. Med. Hypotheses 1992, 38:106-110. Cheeseman KH, Burton GW, Ingold KU and Slater TF. Lipid peroxidation and lipid antioxidants in normal and tumor cells. Toxicol. Pathol. 1984, 12:235-239. Muggli R. Dietary fish oils increase the requirement for vitamin E in humans. In: Chandra RK (ed). Health effects of fish and fish oils. 1989, 201-210, Arts Biomedical Publishers, St. Johns, Newfoundland. Chan HWS, Coxon DT, Peers KE and Price KR. Oxidative reactions 4 of unsaturated lipids. Food Chem. 1982, 9:21-34. Kosugi H, Kato T and Kikugawa K. Formation of red pigment by a two step 2-thiobarbituric acid reaction of alka-2,4 dienals. {agential products of lipid peroxidation. Lipids 1988, 23:1024- 64. 65. 66. 67. 69. 70. HI): 76. 64. 65. 66. 67. 68. 69. 70. 71. 72. 73. 74. 75. 76. 38 Vaca CE, Wilhelm J and Harms-Ringdahl M. Studies on lipid peroxidation in rat liver nuclei and isolated nuclear membranes. Biochem. Biophys. Acta 1988, 958:375-387. Iritani N, Fukuda E and Kitamura Y. Effect of corn oil feeding on lipid peroxidation in rats. J. Nutr. 1980, 110:924-930. Brooks BR and Klamerth OL. Interaction of DNA with bifunctional aldehydes. Eur. J. Biochem. 1968, 5:178-182. Reiss U and Tappel AL. Flourescent formation and changes in structure of DNA reacted with peroxidizing arachidonic acid. Lipids 1973, 8:199-202. Summerfield FW and Tappel AL. Effects of dietary polyunsaturated fats and vitamin E on aging and peroxidative damage to DNA. Arch. Biochem. Biophys. 1984, 233:408-416. Kwei GY, DeLumen 80 and Quantanilha AT. Rat liver nuclei are more sensitive to lipid peroxidation than microsomes: Role of vitamin A and E. Nutr. Rept. Int. 1985, 32:179-185. Wills ED. The role of dietary components in oxidative stress in tissues. In: Sies H. (ed) Oxidative Stress 1985:197-218, Academic Press, London. Ershoff BH. Effect of diet on fish oil toxicity in the rat. J. Nutr. 1980, 71:45-53. Hammer CT and Wills ED. The role of lipid components of the diet in the regulation of the fatty acid composition of the rat liver end0plasmic reticulum and lipid peroxidation. J. Biochem. 1978, 174:585-593. Herbert KE and Wills ED. Platelet function and tissue lipid peroxidation in rats fed polyunsaturated fatty acids. Biochem. Soc. Trans. 1987, 15:410-411. Lamers JM, Hartog JM, Guarnieri C, Vaona I, Verdouw PD and Koster JF. Lipid peroxidation in normoxic and ischaemic-reperfused hearts of fish oil and lard fat fed pigs. J. Mol. Cell. Cardiol. 1988, 20:605-615. Hu ML, Frankel EN, Leibovitz BE and Tappel AL. Effect of dietary lipids and vitamin E on in vitro lipid peroxidation in rat liver and kidney homogenates. J. Nutr. 1989, 119:1574-1582. Pritchard GA and Mansel RE. The effects of essential fatty acids on the growth of breast cancer and melanoma. In: Horobin DF (Ed.) Omega-6 Essential Fatty Acids: Pathophysiology and Roles in Clinical Medicine 1990; 374-390. 77. 78. 79. 80. 81. 82. 83. 77. 78. 79. 80. 81. 82. 83. 84. 85. 86. 87. 39 Marangoni F, Angeli MT, Tarantino A, Borabello A and Galli C. Transcutaneous absorption and incorporation of [H]EPA (10:5 N-3) in lipids of subcutaneous fluid, plasma and liver in the rat. In: Devron CA, Baksaas I and Krokan HE (eds.). Omega-3 Fatty Acids: Metabolism and Biological Effects. 1993, 35-40. Birkhauser Verlag Basel, Switzerland. Shuster CW. Effects of oxidized fatty acids on ascites tumor metabolism. Proc. Soc. Exp. Biol. Med. 1955, 90:423-426. Esterbauer H. Aldehydic products of lipid peroxidation. In: McBrien DC, Slater TF (eds) Free radicals, lipid peroxidation and cancer. 1982, 101-128, Academic Press, London. Hauptolorenz S, Esterbauer H, Moll W, Pumpel R, Shauenstein E and Puschendorf 8. Effects of the lipid peroxidation product 4- hydroxynonenal and related aldehydes on proliferation and viability of cultured Erlich ascites tumor cells. Biochem. Pharmacol. 1985, 34:3803-3809. Begin ME. Effects of polyunsaturated fatty acids and their oxidative products on cell survival. Chem. Phys. Lipids 1987, 45:269-313. Barrera G, DiMauro C, Muraca R, Ferrero D, Cavalli G, Fazio VM, Paradisi L and Dianzani MU. Induction of differentiation in human HL-60 cells by 4-hydroxynonenal, a product of lipid peroxidation. Exp. Cell Res. 1991, 197:148-152. Kanazawa K. Hepatotoxicity caused by dietary secondary products originating from lipid peroxidation. In: Friedman M (ed). Nutritional and Toxicological Consequences of Food Processing, 1991, 237-253, Plenum Press, New York. Zollner H, Schaur RJ and Esterbauer H. Biological activities of 4-hydroxy alkenals. In: Wills ED (ed) Oxidative Stress: Oxidants and Antioxidants, 1991, 337-369, Academic Press, London. Koga S, Nakano M, Ito T and Tomita Y. Importance of iron in lipid peroxidation in the tyrosine 14-hydroxyaniside system: Possible mechanism of killing of malignant melanoma cells by 4- hydroxyanisole. Biochem. Int. 1992, 26:397-403. Benamira and Marrett L. The lipid peroxidation product 4- hydroxynonenal is a potent inducer of sos response. Mutation Res. DNA Repair 1992, 293:1-10. Lindahl R. Aldehyde dehydrogenases and their role in carcinogenesis. Crit. Rev. Biochem. Molec. Biol. 1992, 27:283- 335. Fazio VM, Barrera G. Markinotti S, Farace MG, Giglioni B, Frati L, Manzari V and Dionzani MU. 4-hydroxynonenal, a product of 89. 90. 91. 92. 93. 94. 95. 96. 97. Ehp 101. 89. 90. 91. 92. 93. 94. 95. 96. 97. 98. 99. 100. 101. 40 cellular lipid peroxidation, which modulates c-myc and globin gene expression in K562 erythroleukemic cells. Cancer Res. 1992, 52, 4866-4871. Esterbauer H and Zollner H. Methods for determination of aldehydic lipid peroxidation products. Free Rad. Biol. Med. 1989, 7:197-203. Kikugawa K, Kojima T and Kosugi H. Major thiobarbituric acid- reactive substances of liver homogenate are alkadienals. Free Rad. Res. Comm. 1990, 8:107-113. Gray JI. Measurement of lipid peroxidation: A review. J. Am. Oil Chem. Soc. 1978, 55:539-546. Kosugi H and Kikawa K. Potential thiobarbituric acid reactive substances in peroxidized lipids. Free Rad. Biol. Med. 1989, 7:205-207. Kanazawa K, Inoue N, Ashida H, Mizuro M and Natake M. What do thiobarbituric acid and hemoglobin-methylene blue test evaluate in the endogenous lipid peroxidation of rat liver. J. Clin. Biochem. Nutr. 1990, 7:69-79. Bernheim F, Bernheim MLC and Wilbur KM. The reaction between .thiobarbituric acid and the oxidation products of certain lipids. J. Biol. Chem. 1948, 174: 257-264. Bird RP and Draper HH. Uptake and oxidation of malondialdehyde by cultured mammalian cells. Lipids 1982, 27:419-423. Bonnes-Taourel D, Guerin MC and Torreilles J. Is malonaldehyde a valuable indicator of lipid peroxidation? Biochem. Pharmacol. 1992, 44:985-988. Ceconi C. Use and limitations of thiobarbituric reaction to detect lipid peroxidation. Am. J. Physiol. 1992, 263:H981-H983. Gamma] EB, Carroll KK and Plunkett ER. Effects of dietary fat on mammary carcinogenesis by 7,12 demethylbenz(a)anthracene in rats. Cancer Res. 1967, 27:1737-1742. Slater TF. Lipid peroxidation and cell decusion in normal and tumour tissue. In: Nigam SK, McBrien OCH and Slater TF (eds). Eicosanoids, lipid peroxidation and cancer. 1988, 137-151, Springer Verlag, London. Gavino VC, Miller JS, Ikharebha SO, Mibo GE and Cornwell 06. Effect of polyunsaturated fatty acids and antioxidants on lipid peroxidation in tissue cultures. J. Lipid Res. 1981, 22:763-769. Begin ME, Das UN, Ells G and Horrobin OF. Selective killing of human cancer cells by polyunsaturated fatty acids. Prostaglandins Leukotrienes Med. 1985, 19:177-186. 102. 103. 104. 105. 106. 107. 108. 109. 110. 111. 'l‘ 112. t 113. 102. 103. 104. 105. 106. 107. 108. 109. 110. 111. 112. 113. 41 Begin ME, Ells G, Das UN and Horrobin DF. Differential killing of human carcinoma cells supplemented with n-3 and n-6 polyunsaturated fatty acids. J. Natl. Cancer Inst. 1986, 77:1053-1062. Poot M, Esterbauer H, Rabinovitch PS and Hoehn H. Disturbance of cell proliferation by two model compounds of lipid peroxidation contradicts causative role in proliferative senescence. J. Cell. Physiol. 1988, 137:421-429. Hammer A, Tillian HM, Kink E, Sharaf El Din M, Schaur RJ and Schauenstein E. Iron-induced lipid peroxidation and inhibition of proliferation in animal tumour cells. In: Nigam SK, McBrien DCH and Slater TF. (eds). Eicosanoids, lipid peroxidation and cancer. 1988, 221-226, Springer-Verlag, London. Begin ME, Ells GW and Horrobin DF. Polyunsaturated FA induced cytotoxicity against tumor cells and its relationship to lipid peroxidation. J. Natl. Cancer Inst. 1988; 80, 188-194. Begin ME. Tumor cytotoxicity of essential fatty acids. Nutr. 1989, 5:79-83. Tillian HM, Hammer A, Kink E, Shaur RJ and Shauenstein E. Iron induced lipid peroxidation and inhibition of proliferation in Ehrlich ascites tumor cells. J. Cancer Res. Clin. Oncol. 1989, 115:79-83. Begin ME, Sircar S and Weber JM. Differential sensitivity of tumorigenic and genetically related non-tumorigenic cells to cells to cytotoxic polyusaturated fatty acids. Anticancer Res. 1989, 9:1049-1052. Takeda S, Ells G, Manku M, Horrobin DF and Huang VS. Cytotoxicity effect of polyunsaturated fatty acids against human oral cancer cell lines. FASEB J 1989, 3:A471. Rose DP and Connolly JM. Effects of fatty acids and inhibitors of eicosanoid synthesis on the growth of a human breast cancer cell line in culture. Cancer Res. 1990, 50:7139-7144. Sircar S, Cal F, Begin ME and Weber JM. Transformation renders MDR cells more sensitive to polyunsaturated fatty acids. Anticancer Res. 1990, 10:1783-1786. Horrobin DF. Essential fatty acids, lipid peroxidation and cancer. In: Horrobin DF (ed) Omega-G-Essential Fatty Acids: Pathophysiology and Roles in Clinical Medicine, 1990, 351-377, Alan R. Liss, Inc., New York. Canuto RA, Muzio G, Biocca ME and Dianzani MU. Lipid peroxidation in rat AH-13O hematoma cells enriched in vitro with arachidonic acid. Cancer Res. 1991, 51:4603-4608. 114. 115. 116. 117. 118. 119. 120. 121. 122. 123. 124. 125. 126. OGEL mn Mm 114. 115. 116. 117. 118. 119. 120. 121. 122. 123. 124. 125. 126. 42 Das UN. Tumoricidal action of cis-unsaturated fatty acids and their relationship to free radicals and lipid peroxidation. Cancer Lett. 1991, 56:235-243. DeBravo MG, deAntueno RJ, Toledo J, deTomas ME, Mercuri OF and Quantans C. Effects of an eicosapentaenoic and docosahexanoic acid concentrate on a human lung carcinoma grown in nude mice. Lipids 1991, 26:866-870. Begin ME and Ells G. Levels of thiobarbituric acid reactive substances and the cytocidal potential of gammalinoleic and docosahexaenoic acids on ZR-75-1 and CV-l cells. Lipids 1992, 27:147-149. Takeda S, Sim PG, Horrobin DF, Chisholm KA, Simmons VA, Ells GW, Jenkins OK and Fisher-Morse NL. Intracellular free fatty acid release and lipid peroxidation in cultured human breast cancer cells in response to gammalinoleic acid with iron (GLA+Fe). Int. J. Oncol. 1992, 1:759-763. Takeda S, Horrobin DF, Manku M, Sim PG, Ells G and Simmons V. Lipid peroxidation in human breast cancer cells in response to gamma-linoleic acid and iron. Anticancer Res. 1992, 12:329-334. Shauenstein E. Effects of low concentrations of aldehydes on tumor cells and tumor growth. In: McBrien DC, Slater TF (eds). Free radical, lipid peroxidation and cancer, 1982, 159-171, London, Academic Press. Bernheim F. Biochemical implications of pro-oxidants and antioxidants. Rad. Res. (suppl) 1963, 3:17-32. Miller DM, Buettner GR and Aust SD. Transition metals as catalysts of autoxidation reactions. Free Rad. Biol. Med. 1990, 8:95-108. Janero DR and Burghardt B. Thiobarbituric acid reactive malondialdehyde formation during superoxide dependent iron catalyzed lipid peroxidation: Influence of peroxidation conditions. Lipids 1989; 24:125-131. Bast A, Haenen GRMM and Doelman CJA. Oxidants and antioxidants: state of the art. Am. J. Med. 1991, 91(5): 35-135. Burton GW, Cheeseman KH, Ingold KU and Slater TF. Lipid antioxidants and products of lipid peroxidation as potential tumor protective agents. Biochem. Soc. Trans. 1982, 11:261-262. Gerber M, Richardson S, DePaulet PC, Pujol H and DePaulet AC. Relationship between vitamin E and polyunsaturated fatty acids in breast cancer. Cancer 1989, 64:2347-2353. Gerber M and Segala C. The relationship between breast cancer and polyunsaturated fatty acids. Int. J. Cancer 1991, 47:319. 127. 128. 129. 130. 131. 132. 133. 134. 135. 136. 137, 138. 139, 127. 128. 129. 130. 131. 132. 133. 134. 135. 136. 137. 138. 139. 43 Gerber M and Segala C. Aging and cancer: Plasma antioxidants and lipid peroxidation in young and aged breast cancer patients. In: Emerit I and Chance 8. (ed) Free Radicals and Aging 1992, 235- 246. Brinkhauser Verlag Basel, Switzerland. Burton GW, Cheeseman KH, Ooba T, Ingold KU and Slater TF. Vitamin E as an antioxidant in vitro and in vivo. In: Ciba Fdn. Symposium 101, Biology of Vitamin E. 1983, 4-14, Pitman, London. Cheeseman KH, Collins M, Proudfoot K, Slater TF, Burton GW, Webb AC, and Ingold KU. Studies on lipid peroxidation in normal and tumour tissues. Biochem. J. 1986, 235:507-514. Cheeseman KH, Emery S, Maddix SP, Slater TF, Burton GW and Ingold KU. Studies on lipid peroxidation in normal and tumour tissues 11. Biochem. J. 1988, 250:247-252. L'Abbe’ MR, Trick OK and Beare-Rogers JL Dietary (n-3) fatty acids affect rat heart, liver and aorta protective enzyme activities and lipid peroxidation. J. Nutr. 1991, 121:1331-1340. Meydani M, Natiello F, Goldin 8, Free N, Woods M, Shaefer E, Blumberg JB and Gorbach SL. Effect of long term fish oil supplementation of vitamin E status and lipid peroxidation in women. J. Nutr. 1991, 121:484-491. Wattenburg LW. Inhibitors of chemical carcinogenesis. Adv. Cancer Res. 1978, 26:197-226. Slaga TJ and Bracken WM. The effect of antioxidants on skin tumor imitation and aryl hydrocarbon hydrolase. Cancer Res. 1977, 37:1631-1635. Verma AK, Conrad FA and Boutwell RK. Differential effects of retinoic acid and 7,8-benzoflavone on the induction of mouse skin tumors by the complete carcinogenesis process and by the imitation-promotion regime. Cancer Res. 1982, 42:3519-3525. Toth S and Patil K. Enhancing effect of vitamin E on Murine intestinal tumorigenesis by 1,2 demethylhydrazine dehydrochloride. J. Natl. Cancer Inst. 1983, 70:1107-1111. Clayson OB, Iverson F, Nera EA and Lok E. The importance of cellular proliferation induced by BHA and BHT. Cell Prolif. 1991, 1:231-242. Burns CP and Wagner BA. Heightened susceptibility of fish oil polyunsaturated rate-enriched neoplastic cells to ethane generation during lipid peroxidation. J. Lipid Res. 1991, 32:79- Benchekroun MN and Robert J. Measurement of doxorubicin-induced lipid peroxidation under the conditions that determine cytotoxicity in cultured tumor cells. Analytical Biochem. 1992, 140. 141. 142. 143. 144. 145. 146. 147. 148. 149. 150. 151. 152. 140. 141. 142. 143. 144. 145. 146. 147. 148. 149. 150. 151. 152. 44 201:326-330. Wolfson N, Wilbur KM and Bernheim F. Lipid peroxide formation in regenerating rat liver. Exp. Cell Res. 1956, 10:556-558. Bartoli GM and Galeotti T. Growth related lipid peroxidation in tumor microsomal membranes and mitochondria. Biochim. Biophys. Acta 1979, 574: 537-541. Cheeseman KH, Collins M, Maddix S, Milian A, Proudfoot K, Slater TF, Burton GW, Webb A and Ingold KU. Lipid peroxidation in regenerating liver. FEBS 1986, 209,191-196. Slater TF, Benedetto C, Burton GW, Cheeseman KH, Ingold KU and Nodes JT. Lipid peroxidation in animal tumours: a disturbance in the control of cell division? In: Thaler-an H, Paoletti R and Crustes dePaulet A. (eds.), Icosanoids and Cancer 1984, 21-29, Raven, New York. Massotti L, Cusali E and Galeotti T. Lipid peroxidation in tumor cells. Free Rad. Biol. Med. 1988, 4:377-386. Poli G, Cecchini G, Biasi F, Chiarpotto E, Canuto RA, Biocca ME, Muzio G, Esterbauer H and Dianzani MU. Resistance to oxidative stress by hyperplastic rat liver tissue monitored in terms of unpolar and medium polar carbonyls. Biochim. Biophys. Acta 1986, 883:207-214. Dianzani MU, Poli G, Canuto RA, Rossi MA, Biocca ME, Biasi F, Cecchini G, Muzio G, Ferro M and Esterbauer H. New data on kinetics of lipid peroxidation in external hepatomas and preneoplastic nodules. Toxicol. Pathol. 1986, 14:404-410. Yu YY, Ogino T and Okada S. Histochemical detection of lipid peroxidation in human gastrointestinal, mammary, and renal carcinomas. Acta. Med. Okayama 1992, 46:233-239. Dianzani MU. Biochemical effects of saturated and unsaturated aldehydes. In: McBrien O and Slater TF (eds.) Free Radicals, Lipid Peroxidation and Cancer. 1982, 129-158, Academic Press, London. Goldshmidt BM. Role of aldehydes in carcinogenesis. J. Environ. Sci. Health 1984, C2, 231. Roderg MK, Geende EA Jr, and Recknagel RO. Prelytic damage of red cells in filtrates from peroxidizing microsomes. Science 1986, 196:1221-1222. Witz G. Biological interactions of alpha, beta unsaturated aldehydes. Free Rad. Biol. Med, 1989, 7:333-349. Nalini S, Balsubramarian KA. Albumin bound nonesterified fatty acids inhibit in vitro lipid peroxidation. Ind. J. Biochem. 153. 154. 155. 156. 157. 158. 159. 160. 161. 162. 153, 153. 154. 155. 156. 157. 158. 159. 160. 161. 162. 163. 45 Biophys. 1989, 26: 357-360. Balsubramarian KA, Nalini S, Cheeseman KH and Slater TF. Non esterified fatty acids inhibit iron dependent lipid peroxidation. Biochim. Biophys. Acta 1989, 1003:232-237. Balsubramarian KA, Nalini S and Manohar M. Nonesterified fatty acids and lipid peroxidation. Molec. Cell. Biochem. 1992, 111:131-135. Lane HW, Butel JS, Howard C, Shepherd F, Halligan R and Median O. The role of high levels of dietary fat in 7,12 dimethylbenz(a)anthracene-induced mouse mammary tumorigenesis: Lack of an effect on lipid peroxidation. Carcinogenesis 1985, 6:403-40 . Cerruti PA. Response modification in carcinogenesis. Env. Health Persp. 1989, 81:39-43. Gabor H and Abraham S. Effect of dietary menhaden oil on tumor cell loss and the accumulation of mass of a transplantable mammary adenocarcinoma in Balb/C mice. J. Natl. Cancer Inst. 1984, 73:457-461. Carroll KK. Fish Oils and Cancer. In: Chandra RK. (ed) Health effects of fish and fish oils. 1989, 395-407. Arts Biomedical Publishers, St. John’s Newfoundland. Gonzalez MJ, Schemmel RA, Gray JI, Dugan L Jr, Scheffield LG and Welsch CW. Effect of dietary fat on growth of MCF-7 and MDA- M8231 human breast carcinomas in athymic nude mice: Relationship between carcinoma growth and lipid peroxidation product levels. Carcinogenesis 1991, 12:1231-1235. Iritani N and Rajikawa S. Competitive incorporation of dietary W-3 and W-6 polyunsaturated fatty acids into the tissue phospholipids in rats. J. Nutr. Sci. Vitaminol. 1982, 28:621- 629. Gonzalez MJ and Welsch CW. Inhibition of Mammary Gland Tumorigenesis by Dietary Fish Oil: A phenomenon associated with increased levels of tumor lipid peroxidation products? In: Orevon CA, Baksaas I and Krokan HE (eds.). Omega-3 fatty acids: Metabolism and Biological Effects. 1993, 315-326. Birkhauser Verlag, Basel/Switzerland. Rasheed AA, Oldfield JE, Kaufmes J and Sinhuber R0. Nutritive value of marine oils: Menhaden oil at varying oxidative levels :gtgzgng3zithout antioxidants in rat diets. J. Nutr. 1963, Lytle JS, Lytle TF, Newmark HL and Oeschner EE. Stability of a commercially prepared fish oil (omega-3 fatty acid) laboratory rodent diet. Nutr. Cancer 1992, 17:187-194. 164 165 166. 167. 168. 169. 170. 171. 172. 173. 174. 175. 176. 177. 178. 164. 165. 166. 167. 168. 169. 170. 171. 172. 173. 174. 175. 176. 177. 178. 46 Fritsche KL and Johnston PV. Rapid auto-oxidation of fish oil in diets without added antioxidants. J. Nutr. 1988, 118:425-426. Thomas JP and Girotti AN. Role of lipid peroxidation in hematoporphyrin derivitive-sensitized photokilling of tumor cells: Protective effects of glutathione peroxidase. Cancer Res. 1989, 49:1682-1686. Pass HI. Photodynamic therapy in incology: Mechanism and clinical use. J. Natl. Cancer Inst. 1993, 85:443-456. Brunner N, Bronzert O, Vindelov LL et al. Effect on growth and cell cycle kinetics of estradiol and tamoxifen on MCF-7 human breast cancer cells grown in vitro and in nude mice. Cancer Res. 1989, 49:1515-1520. Zwilling E. Controlled development during development. In: Cellular Injury. 1964. (Ed. ARS de Reuk and J Knight) p. 352- 373. Churchill, London. Cooper EH. The biology of cell death in tumors. Cell Tissue Kinet. 1973, 6:87-95. Oevita Jr. VT, Hellman S, Rosenberg SA. Cancer: Principles and Practice of Oncology. 3rd ed. 1982. J.P. Lippincott, Philadelphia. Marx JL. How cancer cells spread in the body. Science 1989, 244:147-148. Sommers SC. Growth rates, cell kinetics and mathematical models of human cancers. 1973. In: Pathobiology Annual 1973. (Ed. H.L. Ioachim) pp. 304-340. Appleton-Century-Crofts, New York. Klein G. Tumor specific transplantation antigens. GHA Clowes Memorial lecture. Cancer Res. 1968, 28:625-635. Levan A. Chromosomes in cancer tissue. Ann. N.Y. Acad. Sci. 1965, 63:774-792. McLean AEM, McLean E and Judatt JO. Cellular necrosis in the liver induced and modified by drugs. Int. Rev. Exp. Pathol. 1965, 4:127-131. Steel G.G. Cell loss factor in the growth rate of human tumours. Eur. J. Cancer 1967, 3:381-387. Iversen OH. Kinetics of cellular proliferation and cell loss in human carcinomas. Eur. J. Cancer 1967, 3:389-394. Hofer KG, Presky W and Hughes WL. Death and metastaggc distribution of tumor cells in mice monitored with iodo deoxyuridine. J. Natl. Cancer Inst. 1969, 43:763-773. 16 182 184» 185. 186. 187. 189. De 190. Ga 179. 180. 181. 182. 183. 184. 185. 186. 187. 188. 189. 190. 47 Dethlefsen LA. An evaluation of radioiodine labelled 5-iodo-2 deoxyuridine as a tracer for measuring cell loss from solid tumors. Cell Tissue Kinet. 1971, 4:123-138. Charbit A, Malaise EP and Tubiana M. Relation between the pathological nature and the growth rate of human tumours. Eur. J. Cancer 1971, 7:307-315. Clifton KH and Yatvin MB. Cell proliferation and cell loss in the MTG-B mouse mammary carcinoma. Cancer Res. 1970, 30:658-664. Jenkinson IS, Shuter BJ and Wright PNM. Cell kinggic methods for clinical use: assessment of autoradiography and 1 I iodo- deoxyuridine with mouse Ehrlich tumour. Cell Tissue Kinet. 1975, 8:307-320. Begg AC. Cellllgss from several types of solid murine tumour: comparison of I-iodo deoxyuridine and tritiated thymidine methods. Cell Tissue Kinet. 1977, 10:409-427. Dethlefsen LA, Sorensen J and Snively J. Cell loss from three gigablished lines of C3H mouse mammary tumor: a comparison of the I-UOR and the 3H-TdR-autoradiographic methods. Cell Tissue Kinet. 1977, 10:447-459. Purschen W and Feinendegen L. In vivo bestimmun der zeilnerlustrate bei experimental - tumoren mit maikierten Joddeoxyuridin. Strahlentherapie 1969, 137:718-722. Commerford SL. Biological stability of 5-iodo-2’-deoxyuridine labelled with iodine-125 after its incorporation into the deoxyirbonucleic acid of the mouse. Nature 1965, 206:949-950. Dethlefsen LA. Reutilization of 125I-5-iodo-2'-deoxyuridine as compared to 3H-thymidine in mouse duodenum and mammary tumor. J. Natl. Cancer Inst. 1970, 44:827-840. Fidler IJ. Metastasis: quantitative gnalysis of distribution and fate of tumor emboli labelled with 2 I-5-iodo-2’-deoxyuridine. J. Natl. Cancer Inst. 1970, 45:773-782. Oenekamp J. The relationship between the ’cell loss factor’ and the immediate response to radiation in animal tumours. Eur. J. Cancer 1972, 8:335-340. Gabor H and Abraham S. Effect of dietary menhaden oil on tumor cell loss and the accumulation of mass of a transplantable mammary adenocarcinoma in Balb/c mice. J. Natl. Cancer Institute 1986, 76:1223-1229. Effect Chapter 1 Effect of dietary fat on growth of MCF-7 and MOA-M8231 human breast carcinomas in athymic nude mice: relationship between carcinoma growth and lipid peroxidation product levels. (Carcinogenesis 1991, 12:1231-1235) NflWl Lm Dapa Depa Hict East EFFECT OF DIETARY FAT ON GROWTH OF NCF-7 AND MDA-N823] HUMAN BREAST CARCINONAS IN ATHYMIC NUDE NICE: RELATIONSHIP BETWEEN CARCINOMA GROWTH AND LIPID PEROXIDATION PRODUCT LEVELS Michael J. Gonzalezl, Rachel A. Schemmell, J. Ian Grayl, Leroy Dugan, Jr.1, Lewis G. Sheffield2 and Clifford W. Welsch2 1Department of Food Science and Human Nutrition 2Department of Pharmacology and Toxicology Michigan State University East Lansing, MI 48824 U.S.A. 49 and . prepi resea Board Hichi Reprin De] Mic Ea: 50 WE: We acknowledge the technical assistance of Chris Heil, Deb O’Connor and Steve Wysong in the experimental studies and Marlene Green for the preparation of this manuscript. These studies were supported by research grants NIH-CA 42876, AICR-9OBW05, Wisconsin Milk Marketing Board 111-17, and Ladies Auxiliary to the Veterans of Foreign Wars - Michigan Division to C.W. Welsch. mm: Dietary Fat and Human Breast Carcinoma Growth musing: C.W. Welsch Department of Pharmacology and Toxicology Michigan State University East Lansing, MI 48824 Huma transpla Seven to groups a containi corn oil (Menhade diets we diet plt citrate b1 measc termi nai breast c fed the tallow c differer and tho: Compari, numerici H8231) . diffEPei Product peroxidi mice fec "‘1' ani tumor pG 51 mm Human breast carcinoma cell lines MCF-7 and MDA-M8231 were transplanted s.c. to female athymic nude mice at 3-4 weeks of age. Seven to 10 days after transplantation, the mice were divided into groups and fed for 6-8 weeks one of the following semi-purified diets containing different amounts and types of fat, i.e., 5% corn oil, 20% corn oil, 20% butter, 19% beef tallow/1% corn oil and 19% fish (Menhaden) oil/1% corn oil. In additional experiments, the fish oil diets were supplemented with antioxidants (vitamin E, 89 or 2000 IU/kg diet plus tertiary butyl hydroquinone-TBHQ, 4g/kg diet) or ferric citrate (3g/kg diet). Tumor peroxidation product levels were assessed by measuring 2-thiobarbituric acid reactants (TBA assay). At the termination of the studies (6 to 8 weeks of diet feeding) mean human breast carcinoma volume (MCF-7 and MDA-M8231) was the largest in mice fed the 20% corn oil diet, intermediate in mice fed the butter or beef tallow diets and the least in mice fed the fish oil diet. The difference in mean tumor volumes among mice fed the 20% corn oil diet and those fed the fish oil diet was significant (P < 0.01). When comparing low (5% corn oil) and high (20% corn oil) fat diets, numerical increases in human breast carcinoma volume (MCF-7 and MDA- M8231) were consistently observed in the high fat diet groups but these differences were not always significant. Tumor lipid peroxidation product levels were determined on the MDA-M8231 tumors; tumor lipid peroxidation levels were significantly (P < 0.01) increased only in mice fed the fish oil diets. Supplementation of the fish oil diets with antioxidants (vitamin E + TBHQ) significantly reduced the level of tumor peroxidation products and significantly increased tumor volume (P < 0.( oil plus observec differel contraS‘ signifii levels . clearly H8231) | inhibit appears peroxid In fat mar This ha carcinc exDerin SEVera] may inc is mai, CaHCEr: embarks fat 0" mice. COrn 0. 0f dlEi 52 (P < 0.05). When tumor lipid peroxidation product levels in the fish oil plus antioxidant fed mice were reduced to the level of that observed in the tumors of the corn oil fed mice, no significant differences in tumor volumes were observed in these two groups. In contrast, supplementation of the fish oil diets with ferric citrate, significantly (P < 0.05) increased tumor lipid peroxidation product levels and decreased tumor volume. Thus, the type of dietary fat can clearly affect the growth of human breast carcinomas (MCF-7 and MDA- MBZ31) maintained in athymic nude mice. The consistent and significant inhibition of dietary fish oil (Menhaden) on growth of these carcinomas appears to be due to an increased accumulation of tumor lipid peroxidation products. MIMI]!!! In experimental animals, altering the level and/or type of dietary fat markedly influences the development and growth of mammary tumors. This has been demonstrated in an impressive array of spontaneous, carcinogen-induced, transplantable, benign and carcinomatous experimental rodent mammary tumor systems (1). In human populations, several epidemiological studies have suggested that diets high in fat may increase the risk of breast cancer development (2). To date, there is mainly epidemiological data to link dietary fat and human breast cancerigenesis. In an effort to resolve this relationship, we have embarked upon a series of studies to assess the influence of dietary fat on growth of human breast carcinomas maintained in athymic nude mice. In this communication, we report the influence of the amount (5% corn oil/20% corn oil) and type (corn oil/butter/beef tallow/fish oil) of dietary fat on growth of hormone responsive (MCF-7) (3) and hormone non-res these ‘ of the relatil deterii oxidat replic. concen' fats, . oxidat the in of fat implic diets Tw Spragu studie owerhe temper Surgic aSEpti Chow ( Until Thetea a Cal: ”Iner; 53 non-responsive (MDA-M8231) (4) human breast carcinoma cell lines in these immune deficient animals. In addition, we assessed the influence of the level of lipid peroxidation products in these carcinomas in relation to carcinoma growth. Lipid peroxidation is an oxidative deterioration of unsaturated fatty acids that give rise to a variety of oxidation products with the potential of interfering with cell replication and/or cell survival. The presence of higher concentrations of highly unsaturated fatty acids in certain dietary fats, e.g., fish oil brings into the context the possibility that oxidation products of these fatty acids may play an important role in the inhibitory or growth rate limiting action exhibited by these types of fat. In studies reported herein, we are providing evidence implicating lipid peroxidation products in the inhibitory action of diets high in fish oil on human breast carcinoma growth. HAIEBIAL§_AND_NEIHQD§ Two hundred and fifty-eight female athymic nude mice (Harlan Sprague Dawley Inc., Madison, WI) 3-4 weeks old were used in these studies. The mice were housed under aseptic conditions (enclosed overhead laminar flow hood, sterilized cages, bedding and water) in a temperature controlled (24°C) and light controlled (14 hr/day) room. Surgical procedures for transplantation of tumors were performed under aseptic conditions in a laminar flow hood. Autoclaved laboratory mouse chow (Purina Mills Inc., St. Louis, MO) was fed ad_11b11um before and until 7 to 10 days after human breast carcinoma transplantation. Thereafter, mice were fed ad libitum semi-purified diets (Table 1). On a caloric basis, the diets contained the same amount of protein, minerals, vitamins and fiber. Energy calculations were based on 4 Kcal/g 0 ingredie except 5 Mesa, Cl Corp. (1 Univers predomi of the prepare packed consumv steril- 19/1it §gggr§ Pa Cultur athymi to 4 5 Nice ‘ transF condit IntegL dlStar breast Has ta Spec1" 54 Kcal/g of protein and carbohydrate and 9 Kcal/g of fat. All dietary ingredients were obtained from U.S. Biochemicals, Inc. (Cleveland, OH) except sucrose which was obtained from ICN Biochemicals, Inc. (Costa Mesa, CA), fish oil (Menhaden) which was obtained from Zapata Haynie Corp. (Reedville, VA) and butter which was obtained from Michigan State University Food Stores (East Lansing, MI). The percentage of the predominant fatty acids (1% or greater, manufacturers specifications) of the dietary oils and fats are shown in Table 2. The diets are prepared in our laboratory weekly and stored at -20°C, individually packed with nitrogen prior to feeding. Mice were fed daily and non- consumed food discarded daily. Since the semi-synthetic diets were not sterilized, antibiotics (bacitracin, streptomycin and neomycin, lg/liter) were added to the drinking water. IieneraLEmedure Palpable MCF-7 and MDA-M8231 human breast carcinomas (American Type Culture Collection, Rockville, MD) were surgically excised from female athymic nude mice and transplanted to 3-4 week old athymic nude mice, 3 to 4 slices (2x4 mm, 0.1 to 0.3 mm thick) of tumor tissue per mouse. Mice were anesthetized with sodium pentobarbital prior to transplantation; all surgical procedures were done under aseptic conditions in a laminar flow hood. An incision was made in the integument, the tumor slices were placed s.c. in the dorsum at distances from each other of at least 2 cm. One to 3 palpable human breast carcinomas provided the tissue slices for each experiment; care was taken to randomize the carcinoma slices from pooled tumor specimens. Sin! for groi implantl cholest Ins/Ll- describ indepen growth diets 7 the car the ons bearing for a p tumors vernier Stu 3. The amount fat (co growth mice. itu Study w fit aff 55 Since the MCF-7 human breast carcinoma is dependent upon estrogen for growth in athymic nude mice, mice bearing these tumors were implanted with a single estrogen pellet (2 mg 17 B-estradiol/38 mg cholesterol) s.c. and were given estrone in their drinking water (0.5 mg/L). The preparation of the hormone pellets is as previously described (5). The MDA-M8231 human breast carcinoma is hormone independent; such tumors do not require supplemented hormones for growth in athymic nude mice. Mice were placed on the experimental diets 7 to 10 days after carcinoma transplantation. This period allows the carcinoma grafts to become established in the host animals before the onset of experimental dietary treatments. Athymic nude mice bearing human breast carcinoma grafts were fed the experimental diets for a period of 6 to 8 weeks. At the termination of the studies, all tumors were excised and their volume determined (V - 4/3 TT r3) with a vernier caliper. Three separate studies were performed (Studies 1-3). Study_1 The experimental design of this study is provided in Table 3. The purpose of this study was to determine whether or not the amount of dietary fat (5%.corn oil/20% corn oil) or the type of dietary fat (corn oil/butter/beef tallow/fish oil) could differentially affect growth of MCF-7 and MDA-M8231 human breast carcinomas in athymic nude mice. Stugy_z (Experimental design - see Table 4). The purpose of this study was to determine whether or not the amount and/or type of dietary fat affects the level-of human breast carcinoma lipid peroxidation products. Excised tumors were assayed for lipid peroxidation products by measuring Z-thiobarbituric acid reactants using a modified TBA assay as described below. 51 study (vitat oil d‘ lipid peroxi Yitami 89 or Chemic oil di added carcin assay. 51 EXperii lice). feeding Peroxic Weight Val'l'anc '35 set % SUbl excised, at .200( tumor sa Samples 56 Study_3 (Experimental design - see Table 5). The purpose of this study was to determine whether or not the addition of antioxidants (vitamin E and TBHQ) or an oxidizing agent (ferric citrate) to the fish oil diets would decrease and increase, respectively, the level of tumor lipid peroxidation products and whether such changes in lipid peroxidation products could influence human breast carcinoma growth. Vitamin E (DL-tocopheryl acetate, U.S. Biochemical Co., Cleveland, OH, Bg or 2000 IU/kg diet) and tertiary butyl hydroquinone (TBHQ, Eastman Chemical Products, Kingsport, TN, 4g/kg diet) were added to the fish oil diets. Ferric citrate (Sigma Chemical Co., St. Louis, MO) was added to the fish oil diet at a level of 3g/kg diet. Excised breast carcinomas were assayed for lipid peroxidation products by the TBA assay. This study was performed twice, experiments #1 and 2. Studie§_1;3 Athymic nude mice that became ill and/or died prior to experiment termination were excluded from all studies (< 3% of the mice). All mice were weighed at the beginning and termination of diet feeding. Mean human breast carcinoma volume (cm3), mean lipid peroxidation product levels (ug TBA reactants/g tissue) and mean body weight gains were statistically analyzed by one way analysis of variance and the Newman-Keuls multiple comparison test. Significance was set at P < 0.05. 188.853! Subcutaneously growing human breast carcinomas were surgically excised, placed in a 0.9% NaCl solution, immediately frozen and stored at -20°C. Immediately before the TBA assay, the tumors were thawed, tumor samples were pooled yielding 109 of tumor tissue; the 109 tumor samples were used in the assay. An antioxidant was added (TBHO, 0.2% of tota homogen homogeh contair suffici The dis of fres solutic triplit pigment 2000, l were mi reports ffect H05;ggg MEI at term corn oi and the differ, oil dis butter l'nterme decreaS c°mDare Statlst 57 of total fat) and samples were homogenized (4°C) in a polytron homogenizer (Ultra Turrax SOT 1810, Tekmar Co., Cincinnati, OH). The homogenates were transferred into prepared distillation flasks containing 2.5 ml of 4N HCl plus 0.5 ml silicone antifoam with sufficient distilled water to yield a total volume of 100 ml/flask. The distillate was collected (50 ml); 5 ml of this distillate plus 5 ml of freshly prepared 2-thiobarbituric acid solution (TBA, 0.2 N aqueous solution) were used for the assay. Samples were prepared in triplicate, heated in boiling water for 35 minutes to develop the pigment, cooled and absorbance read on a spectrophotometer (Spectronic 2000, Bausch 8 Lomb, Rochester, NY). The absorbance values obtained were multiplied by an adjustment factor of 6.2 (6). Results were reported as ug of TBA reactive substances per g of sample. RESULIS . ,- .- 1 . .Ug_, . . - . , .I . . f, . I - . . u'A'w; 1-11s] o -. . In“. I ‘11-. - . IIII Im‘ 11 Mean human breast carcinoma volume of MCF-7 or MDA-M8231 carcinoma at termination of these studies was the largest in mice fed the 20% corn oil diet, intermediate in mice fed the butter or beef tallow diets and the least in mice fed the fish oil diet (Tables 3 & 4). The differences in final tumor volumes of mice fed the corn oil and fish oil diets were consistent and significant (P < 0.01). In mice fed the butter or beef tallow diets, final tumor volume was consistently intermediate to those fed the corn oil or fish oil diets. This decrease in tumor volume in mice fed the butter or beef tallow diets compared to corn oil, however, did not reach the 5% level of statistical probability. 58 When comparing a high fat diet (20% corn oil) to a low fat diet (5% corn oil), numerical increases in MCF-7 and MDA-M8231 carcinoma volumes at termination of the experiments were consistently seen in mice fed the higher level of fat (Tables 3 & 4). Such volume increases were significant (P < 0.01) in 1 of the 2 studies utilizing MDA-M8231 cells (Table 3). Thus, a trend toward increased human breast carcinoma volume is observed, albeit marginally significant, when these animals are fed the higher level of fat. I l' 0' :1. 1H' 1 a a ' . . oI o 0 .I0 a.o a‘ 0‘ a. oI o oo_ ‘ ‘ a u'i'w: I “.I l ‘. . Io“. I As in the preceding study, human breast carcinoma volume at termination of this study was largest in mice fed the 20% corn oil diet, intermediate in mice fed the butter diet, and the least in mice fed the fish oil diet (Table 4). The difference in tumor volumes of mice fed the 20% corn oil and fish oil diets was significant (P < 0.01). Numerical increases in tumor volumes were observed in mice fed the 20% corn oil diet compared to mice fed the 5% corn oil diet, however, these differences did not reach the 5% level of statistical probability. Mean lipid peroxidation product levels (ug TBA reactants/g tissue) in the human breast carcinomas derived from the fish oil fed mice were significantly higher when compared to tumor lipid peroxidation products in each of the other diet groups (P < 0.01). No significant difference in the level of mean lipid peroxidation products in carcinomas derived from animals fed the 5% corn oil, 20% corn oil or butter diets was observed (Table 4). 59 As in the preceding studies, human breast carcinoma volume at the termination of study was significantly (P < 0.05) reduced in mice fed the fish oil diet compared to mice fed the corn oil. The addition of antioxidants (vitamin E and TBHQ) to the fish oil diet caused a striking increase (6 fold, P < 0.05) in final tumor volume, a final tumor volume, however, that was still quantitatively less than that observed in the corn oil fed mice (P < 0.05). This increase in tumor volume in mice fed the fish oil diet supplemented with antioxidants was accompanied by a significant (P < 0.05) reduction in tumor lipid peroxidation products (ug TBA reactants/g tissue). The level of tumor lipid peroxidation products in the antioxidant supplemented fish oil diet fed mice was still significantly (P < 0.05) more than the level of lipid peroxidation products in the tumors of the corn oil fed mice. The addition of ferric citrate to the fish oil diet, in contrast, resulted in a further reduced final tumor volume (P < 0.05). Such a reduction in tumor volume was accompanied by a significant (P < 0.05) increase in the level of tumor lipid peroxidation products. Willem The study described above was repeated, the results are identical to those observed in experiment #1 with but one important exception. The addition of the antioxidants (vitamin E and TBHO) to the fish oil diet C not si oil fe produ< supplt level diet. EffeC‘ among to th citra throu less nice. 0f hu trans the t leVel fats (Henh lines °bSer Certa SQed 60 diet caused an increase in final human breast carcinoma volume that was not significantly different than the tumor volumes observed in the corn oil fed mice. Furthermore, the level of tumor lipid peroxidation products in the tumors derived from the mice fed the antioxidant supplemented fish oil diet was not significantly different than the level of tumor lipid peroxidation products in mice fed the corn oil diet. Effest_oIED1ets_on_flodx_fle19ht_§ains. There were no significant differences in mean body weight gains among any of the dietary groups (data not shown). The only exception to this are the mice of study 3 (Table 5) who were fed the ferric citrate supplemented fish oil diet. While these mice gained weight throughout the study, their rate of gain was significantly (P < 0.05) less than the corn oil, fish oil or the fish oil plus antioxidant fed mice. DISCUSSION The results of our study demonstrate very clearly that the growth of human breast carcinoma cell lines MCF-7 and MDA-M8231, when transplanted to female athymic nude mice, can be markedly influenced by the type of dietary fat. Tumor growth is greatest in mice fed high levels of a vegetable oil (corn oil), intermediate in mice fed animal fats (butter, beef tallow) and the least in mice fed fish oils (Menhaden oil). Thus, the growth of these human breast carcinoma cell lines, as a function of the type of dietary fat, mimics what has been observed in numerous rodent mammary tumor models, i.e., diets rich in certain vegetable oils (e.g., corn oil, sunflower seed oil, safflower- seed oil) are the most stimulating to mammary tumor growth, diets high in fish in satu stimula and the to alts import; human s Ous growth interes while s reports lines I levels process xenogr; laborat (breast focus s pr0005s oil is liberal eXplm.E fish 0i aCCUmuI l” the ObserVe 61 in fish oils (e.g., Menhaden oil) the least stimulatory and diets high in saturated animal fats are of intermediate potency in the growth stimulation of these tumors (1,7). That the rodent mammary tumor cells and the human breast carcinoma cells have similar growth responsiveness to alterations in dietary fat provides additional support for the important utility of rodent mammary tumor systems as models for the human disease. Our observation that high levels of dietary fish oil suppress the growth of human breast carcinoma cell lines in athymic nude mice is an interesting and potentially important observation. Very recently, while our studies were in progress, other laboratories (8-12) have reported similar observations utilizing human breast carcinoma cell lines (MDA-330, MX-I and MCF-7) in athymic nude mice. Clearly, high levels of dietary fish oil suppress mammary tumorigenic growth processes, not only in rodent mammary tumor models (13-15) but in xenografts of human breast carcinomas maintained in immune deficient laboratory mice as well. How dietary fish oil suppresses mammary (breast) tumorigenic growth processes is not known and is currently the focus of attention of a number of laboratories. Many laboratories have proposed that the inhibition of mammary tumorigenesis by dietary fish oil is via alterations in eicosanoid metabolism (13-15). Our laboratory, in contrast and as reported in this communication, has explored another potential mechanism, i.e., that high levels of dietary fish oil suppress mammary tumorigenic processes by causing an increased accumulation of cytotoxic and/or cytostatic lipid peroxidation products in the tumor tissue. In athymic nude mice fed the fish oil diets, we observed a substantial increase in the level of lipid peroxidation ps in ad ans prs grc the lev dif‘ Furi i.e. prod Thus. supp] part, Peroa fed a level butts PTOdL large (DHA) diet llpic dIrEC 62 products (TBA reactants) in the breast carcinomas; a level of peroxidation products approximately 4 times greater than that observed in the breast carcinomas of mice of the other dietary groups. The addition of high levels of antioxidants to the fish oil diet (vitamin E and TBHQ) substantially reduced the level of tumor lipid peroxidation products and simultaneously caused a substantial increase in tumor growth. Indeed, when tumor lipid peroxidation products were reduced in the fish oil fed mice by dietary antioxidant supplementation, to the level observed in the tumors of the corn oil fed mice, no significant difference in tumor growth in these two groups of mice was observed. Furthermore, the addition to the fish oil diet of a pro-oxidant cation, i.e., ferric citrate, caused an increase in tumor lipid peroxidation product levels and a simultaneous further decrease in tumor growth. Thus, these data provide compelling evidence that the tumor growth suppressing activities of a diet high in fish oil is due, at least in part, to the generation of fish oil derived products of lipid peroxidation. Of the dietary groups examined in these studies, only the fish oil fed mice had substantial increases in tumor peroxidation product levels. Animals fed low or high levels of corn oil or high levels of butter had approximately 1/4 the level of tumor lipid peroxidation products than that observed in the tumors of mice fed fish oil. The large amounts of eicosapentaenoic (EPA) (20:5) and docosahexaenoic acid (DHA) (22:6) in the fish oil diets, compared to the corn oil or butter diet groups, is most likely the major reason for this increase in tumor lipid peroxidation products. It is well established that there is a direct relationship between the number of double bonds in a fatty acid and the oxidat‘ Hos influe second report substa (17). amino inacti polyam DEFOXII cell p‘ Indeed S“Ppre decrea (15). (20.21 certas linoli the ge cancer stimu] Sisuit 63 and the potential formation of lipid peroxidation products; such oxidation can occur via a chemical or an enzymatic reaction (16). How the accumulation of lipid peroxidation products in tumor tissue influences tumor growth processes is not entirely certain. The secondary products of lipid peroxidation measured by the TBA assay are reportedly capable of affecting cell proliferation. For example, these substances can inactivate polymerase reactions by blocking SH group (17). They can cause inter- and/or intramolecular linkages between amino acid sulfhydryl groups of RNA, DNA and proteins leading to the inactivation or damage of these molecules (18). They can inhibit polyamine synthesis (19). Such biochemical interactions of lipid peroxidation products with cellular macromolecules can not only reduce cell proliferation processes but can directly lead to cell death. Indeed, Abraham and colleagues have proposed that dietary fish oil suppression of mammary tumor growth in mice is not a function of decreased cell proliferation but a function of increased cell death (15). In a series of interesting studies, Horrobin and colleagues (20,21) have reported that the addition of relatively large amounts of certain polyunsaturated fatty acids, e.g., EPA, arachidonate, linolineate, to cultures of malignant cells causes a sharp increase in the generation of TBA reactive products and a parallel increase in cancer cell death. The addition to the culture media of inhibitors or stimulators of lipid peroxidation, e.g., vitamin E or iron, caused a significant inhibition or enhancement, respectively, of cell killing. Interestingly and of potential pragmatic importance is their observation that the enhancement of cell death by supplementation of polyunsaturated fatty acids is repeatedly seen in an array of cancer cell ‘ currei repor‘ mammal level of can cells is an II can is nude i dietai 0f the to an 64 cell lines but is notably absent in normal cells (21). We are currently in the process of determining whether or not our previously reported observation of dietary fish oil induced inhibition of normal mammary gland growth processes (22) is a function of increasing the level of mammary gland lipid peroxidation products. The vulnerability of cancer cells to certain polyunsaturated fatty acids, while normal cells appear resistant as reported by Horrobin and colleagues (20,21), is an observation that merits further examination. In summary, we report in this communication that the type of fat can influence growth of human breast carcinomas maintained in athymic nude mice. Most impressive was the consistent inhibitory effect of dietary fish oil on growth of these carcinomas. The inhibitory effect of the fish oil diets on growth of these carcinomas appears to be due to an accumulation of lipid peroxidation products in the tumor tissues. 10. 11. 12. I ‘ 4“ ‘ 3111/1 10. ll. 12. 65 References . Welsch, C.W. (1987) Enhancement of mammary tumorigenesis by dietary fat: Review of potential mechanisms. Am. J. Clin. Nutr., 45, 192-202. . Armstrong 8. and Doll, R. (1975) Environmental factors and cancer incidence and mortality in different countries, with special reference to dietary practices. Int. J. Cancer, 15, 617-631. . Sheffield, L.G. and Welsch, C.W. (1985) Cholera-toxin-enhanced growth of human breast cancer cell lines in vitro and in vivo: Interaction with estrogen. Int. J. Cancer, 36, 479-483. . Arteaga, C.L., Tandon, A.K., Van Hoff, 0.0. and Osborne, C.K. (1988) Transforming growth factor Beta: Potential autocrine growth inhibitor of estrogen receptor-negative human breast cancer cells. Cancer Res., 48, 3898-3904. . Soule, H.D. and McGrawth, C.M. (1980) Estrogen responsible proliferation of clonal human breast carcinoma cells in athymic nude mice. Cancer Lett., 10, 177-189. . Crackel, R.L., Gray, J.I., and Pearson, A.M. (1988) Some further observations on the TBA test as an index of lipid oxidation in meats. Food Chem., 28, 187-196. . Carroll, K.K. and Hopkins, G.J. (1979) Dietary polyunsaturated fat versus saturated fat in relation to mammary carcinogenesis. Lipids, 14, 155-158. . Fernandes, G., Frieduchs, W., and Shultz, J. (1989) Modulation of MCF-7 tumor cell growth in nude mice by omega-3 fatty acid diet. Breast Cancer Res. Treat., 14, 179a. . Pritchard, G.A., Jones, O.L., and Mansel, R.E. (1989) Lipids in breast cancer. Brit. J. Surg., 76, 1069-1073. BIink, E.W. and Ceriani, R.L. (1989) Fish oil enhancement of I-conjugated anti-human milk fat globule monoclonal antibody experimental radio-immunotherapy of breast cancer. J. Steroid Biochem., 34, 149-153. Borgeson, C.E., Pardini, L., Pardini, R.S., and Reitz, C. (1989) Effects of dietary fish oil on human mammary carcinoma and on lipid-metabolizing enzymes. Lipids, 24, 290-295. Gabor, H., Blank, E.W., and Ceriani, R.L. (1990) Effect of dietary fat and monoclonal antibody therapy on the growth of human mammary adeno-carcinoma MX-I grafted in athymic mice. Cancer Lett., 52, 173-178. 13. 14. 15. 16. 13. 14. 15. 16. 17. l8. 19. 20. 21. 22. 66 Karmali, R.A., Marsh, J., and Fuchs, C. (1984) Effect of omega3 fatty acids on growth of a rat mammary tumor. J. Natl. Cancer Inst., 73, 457-461. Jurkowski, J.J. and Cave, W.T. (1985) Dietary effects of menhaden oil on the growth and membrane lipid composition of rat mammary tumors. J. Natl. Cancer Inst., 74, 1145-1150. Gabor, H. and Abraham, S. (1986) Effect of dietary menhaden oil on tumor cell loss and the accumulation of mass of a transplantable mammary adenocarcinoma in BALB/c mice. J. Natl. Cancer Inst., 76, 1223-1229. Cheeseman, K.H., Burton, G.W., and Ingold, K.U. (1984) Lipid peroxidation and lipid antioxidants in normal and tumor cells. Toxicol. Pathol., 12, 235-239. Roubal, W.T. and Tappel, A.L. (1966) Damage to proteins, enzymes and amino acids by peroxidizing lipids. Arch. Biochem. Biophys., 113, 5-8. Reiss, U., and Tappel, A.L. (1973) Fluorescent product formation and changes in structure of DNA reacted with peroxidiZing arachidonic acid. Lipids, 8, 199-202. Dianzani, M.U. (1982) Biochemical effects of saturated and unsaturated aldehydes. In: McBrien DC, Slater TF, eds. Free radicals, lipid peroxidation and cancer. Academic Press, London, NFCR. Cancer Symposium, pp 129-158. Begin, M.E., Ells, G., and Horrobin, D.F. (1988) Polyunsaturated fatty acid induced cytotoxicity against tumor cells and its relationship to lipid peroxidation. J. Natl. Cancer Inst., 80, 188-194. Horrobin, D.F. (1990) Essential fatty acids, lipid peroxidation, and cancer. In: Horrobin DF, ed. Omega-6 Essential Fatty Acids: Pathophysiology and Roles in Clinical Medicine. Wiley-Liss, NY, pp 351-377. Welsch, C.W., and O’Connor, D.H. (1989) Influence of the type of dietary fat on developmental growth of the mammary gland in immature and mature female BALB/c mice. Cancer Res., 49, 5999- 6007. Table 1 ———-——- Composi Fat Casein Dt-meth Dextrin Sucrose AIN Sal AlN Vit Cellulo *The ca' diet 1: “Al pha conten‘ ***The l tal l 0W, fit dis 67 Table 1. Diet composition Composition* Low fat (5%) High fat (20%) diet*** diet*** Fat 5.00% 20.00% Casein 17.10 20.17 OL-methionine 0.30 0.35 Dextrin 45.50 32.18 Sucrose 22.70 16.09 AIN Salt Mixture** 3.50 4.13 AIN Vitamins** 1.00 1.18 Cellulose __§,QQ __§,29 100.00 100.00 *The calculated energy value (metabolizable energy) for the low fat diet is 3.87 kcal/g, for the high fat diets, 4.55 kcal/g. **Alpha tocopherol level content of the diet is 0.2 g/100 g diet. Iron content of the diet is 0.004 g/100 g diet. ***The high fat diets consisted of 20% corn oil, 20% butter, 19% beef tallow/1% corn oil or 19% fish (Menhaden) oil/1%.corn oil; the low fat diet consisted of only 5% corn oil. 68 Table 2. Predominant fatty acids in oils and fats (percentage) Fatty acids* Corn Butter Beef Menhaden oil tallow oil Caprylic (8:0) -- -- -- -- Capric (10:0) -- -- -- -- Lauric (12:0) -- 2.5 -- -- Myristic (14:0) -- 10.5 3.4 8.0 Palmitic (16:0) 10.1 31.0 25.5 28.9 Palmitoleic (16:1) -- 2.5 2.8 7.9 Stearic (18:0) 1.6 13.6 24.9 4.0 Oleic (18:1) 31.4 29.9 35.7 13.4 Linoleic (18:2) 56.3 1.8 1.6 1.1 Linolenic (18:3) -- -- -- 1.0 Eicosapentaenoic (20:5) -- -- -- 10.2 Docosahexaenoic (22:6) -- -- -- 12.8 *Fatty acid concentrations less than 1% are not included. Ti 20% 20% +11 19% +1% Treat 5% Cs 20% Cs 20% BL 19% Be +11 Co 19% Me +11 Co *Female 'eeks tI'anSp i/b, p d/b, p 69 Table 3. Effect of dietary fat on growth of MCF-7 and MDA-M8231 human breast carcinomas transplanted to athymic nude mice. INZF-7 human breast carcinoma Treatment* No. of No. of Mean mice carcinomas carcigoma volume (cm ,1 S.E.) 5% Corn oil 9 36 3.40 i 0.28“ 20% Corn oil 8 28 4.02 i 0.58“ 20% Butter 9 35 2.36 i 0.32“ 19% Beef tallow 9 35 2.36 i 0.36“ +1% Corn oil 19% Menhaden oil 9 36 0.20 1 0.16” +1% Corn oil KIA-118231 human breast carcinoma Treatment* No. of No. of Mean mice carcinomas carcipoma volume (cm 1 S.E.) 5% Corn oil 8 24 0.52 1 0.12” 20% Corn oil 8 22 1.54 1 0.37“ 20% Butter 8 24 1.16 1 0.19cl 19% Beef tallow 6 19 0.84 1 0.23 +1% Corn oil 19% Menhaden oil 7 24 0.56 z 0.50” +1% Corn oil *Female athymic nude mice were fed diets for 8 weeks, from 5 to 13 weeks of age, commencing 7-10 days after human breast carcinoma transplantation. a/b, P < 0.01 d/b, P < 0.05 Tab Tre 5% 20% 20% +1% *Fen wee tra a/b, 70 Table 4. Effect of dietary fat on MDA-M8231 human breast carcinoma growth and carcinoma lipid peroxidation product levels. Mean Mean carcinoma lipid carcinoma peroxidation Treatment* No. of No. of Iolume products (ug/ ,1 S.E.) TBA reactants/g tissue i S.E.) mice carcinomas (cm 5% Corn oil 5 19 0.20 i 0.06 0.16 i 0.04“ 20% Corn 011 8 20 0.37 i 0.11a 0.22 i 0.04a 20% Butter 6 19 0.10 i 0.03 0.20 1 0.07a 19% Menhaden Oil 7 21 0.03 10.01b 0.82 i 0.01b +1% Corn oil *Female athymic nude mice were fed diets for 6 weeks, from 5 to 11 weeks of age, commencing 7 to 10 days after human breast carcinoma transplantation. a/b, P < 0.01 Tabl Tree Lane 20% 19% +1% 19% +1% 1 anti (Vit 19% | +1% I + Fe £102 202: 19% l +12 1 19% I +11 1 antis (Vit 191 I +1: I + Fe *Fems l"Eel tray *‘Uei 71 Table 5. Effect of dietary fish oil plus antioxidants or iron supplementation on MDA-M8231 human breast carcinoma growth and carcinoma lipid peroxidation product levels. Mean Mean carcinoma lipid carcinoma peroxidation Treatment* No. of No. of v lume products mice carcinomas (cm 1S.E)** (ug/TBA reactants/g tissuetS.E.)** Emenimentil 20%.Corn oil 15 40 2.8210.41“ 0.4410.01“ 19% Menhaden oil 16 50 0.24:0.04b 1.411007b +1% Corn oil 19% Menhaden oil 15 39 1.45:0.14c 0.701003c +1% Corn oil and antioxidants (Vit E & TBHO) 19% Menhaden oil 16 40 0.181003“ 2.13:0.09d +1% Corn oil + Fe citrate 20% Corn oil 22' 55 3.2610.82“ 0.39:0.03“ 19% Menhaden oil 24 60 0.34_+_0.04” 13010.03” +1% Corn oil 19% Menhaden oil 25 70 2.3910.35“ 0.4510.02“ +1% Corn oil and antioxidants (Vit E G TBHO) 19% Menhaden oil 18 57 0.15:0.04c 1.52:0.02b +1% Corn oil + Fe citrate *Female athymic nude mice were fed diets for 6 weeks, from 5 to 11 weeks of age, commencing 7 to 10 days after human breast carcinoma transplantation. **Means with different letters are significantly different at P < 0.05. Chapter 2 Lipid peroxidation products are elevated in fish oil diets even in the presence of added antioxidants (J. Nutr. 1992, 122:2190-2195) LIPID PEROXIDATION PRODUCTS ARE ELEVATED IN FISH OIL DIETS EVEN IN THE PRESENCE OF ADDED ANTIOXIDANTS1 Michael J. Gonzalez, J. Ian Gray, Rachel A. Schemmel, LeRoy Dugan, Jr. and Clifford W. Welsch* Departments of Food Science and Human Nutrition and *Pharmacology and Toxicology Michigan State University, East Lansing, MI 48824 Request for reprints: C.W. Welsch Dept. Pharmacology and Toxicology Michigan State University East Lansing, MI 48824 Telephone 4 (517) 353-4549 Fax 4 (517) 353-8195 73 C0 11 p1 di 74 ABSTRACT Purified corn and fish oil diets with different types and concentrations of antioxidants were evaluated for oxidation products. In addition, a determination of different organ and carcass oxidation product levels was performed. PV2 and TBA2 assays were done on the diets immediately after mixing (0 h) and 24, 48 and 72 h after being fed to mice. The AIN recommended level of antioxidant addition (BHTZ, 0.02%.w/w of oil) and even the addition of 100 times this level (BHT, 2.0% w/w of oil), although decreasing the level of oxidation products, failed to totally prevent oxidative deterioration in diets high in fish oil. Furthermore, other antioxidants added in excess to the fish oil diets also failed to completely suppress oxidative deterioration of the diet and, in addition, when fed daily to mice for a period of 4 weeks, caused an increased accumulation of lipid peroxidation products in certain organs (e.g., heart, skeletal muscle, mammary glands) and in the carcass. Thus, these results provide evidence that in the preparation of fish oil diets, the addition of the AIN recommended level of antioxidants, or levels even substantially higher than that recommended does not completely suppress oxidative deterioration of experimental diets. Indexing keywords: fish oil, lipid peroxidation, antioxidants, Balb/c mice. Pol: unambh problem packagi recogni synthei experis Howeve than d becaus acids, Johns‘ added (1988 also 0T or auto: diet DTOC r900 of a by a has auts I‘eS‘ 75 INTRODUCTION Polyunsaturated fats can quickly autoxidize at ambient or subambient temperatures (Chan 1987). In the food industry, this problem has been reduced by the addition of antioxidants and vacuum packaging of foods containing these fats (Buford-Coulter 1988). In recognition of this problem, the AIN has recommended the addition of a synthetic antioxidant at a level of 0.02%, (by weight of oil) to experimental animal diets containing polyunsaturated fats (AIN 1980). However, diets containing fish oil are more susceptible to autoxidation than diets containing other polyunsaturated fats (e.g., corn oil), because of their high concentration of long chain polyunsaturated fatty acids, e.g., EPA“ (20:5, n-3) and DHA“ (22:6, n-3). Fritsche and Johnston (1988) reported an extremely rapid autoxidation of diets with added fish oil as measured by PV. This report by Fritsche and Johnston (1988) plus an earlier publication by Rasheed et al., (1963), which also reported a high autoxidation in diets containing fish oil, are two of only a few publications dealing with the important issue of autoxidation in fish 011 containing diets. It is our opinion that in most studies dealing with the role of dietary fish oil in physiological or pathological processes, the process of dietary autoxidation has been largely ignored. In a recent report, our laboratory has provided evidence that the inhibitory effect of a fish oil diet on tumorigenic processes may be mediated primarily by a lipid peroxidation process (Gonzalez et al. 1991). Such evidence has provided the basis for our belief that the importance of autoxidation or lipid peroxidation has been underestimated by those researchers interested in the health impact of fish oil diets. With this in autoxid oxidati under I Diets Pus modifh of the used f arriva with n after (~20°c of pre Henhad was ad essent corn 0 "‘51 a in EXp V/w ( T or 3H the f TBHQ 76 this in mind, we felt it necessary to further evaluate the issue of autoxidation of fish oil containing diets and the accumulation of oxidation products in organs of mice given a diet rich in fish oil under normal feeding circumstances. MATERIALS AND METHODS 01:15 Purified diets (Table 1) were formulated according to a slight modification of the guidelines developed by AIN (1977). Dry components of the diet (Table 1) were mixed for 15 min and stored at -20°C until used for diet preparation. The fish (Menhaden) oil was used upon arrival with the remaining oil stored in small dark containers sealed with nitrogen gas. Mean PV of the Menhaden oil when received was 7.25, after 7 days it was 7.76 (-20°C) and after 14 days it was 10.45 (-20°C). The Menhaden oil diet was prepared the following way: 800 g of previously mixed dry ingredients were combined with 190 g of Menhaden oil and 10 g of corn oil (19% fish oil/1%»corn oil). Corn oil was added to fish oil to increase the amount of linoleic acid, an essential fatty acid for normal growth and development. The use of corn oil, a polyunsaturated vegetable fat (:60% linoleic acid, 18:2, n-6) as a control comparison for fish oil was based on its frequent use in experimental animal dietary studies. These diets, utilizing 20% fat w/w (200 g/kg diet), are considered high fat diets. The antioxidants TBHQ“ (Eastman Chemical Products, Kingsport, TN) or BHT (Sigma Chemical Co., St. Louis, M0) were immediately added to the fish oil at a level of 0.02%.(BHT) (w/w oil) or 2.0% BHT or 2.0% TBHO,1 8 g/kg dl-alpha tocopherol acetate (U.S. Biochemical, Cleveland, 0H). plasti nitrog signil storas Agaly D exper (Balb mice/ light the I Trip' afte for Peru (197 385a the 291 to l tre. eQLi 506: 77 OH). Sufficient diet for one day’s feeding was packed into small plastic zipper sealed bags. Each bag containing diet was flushed with nitrogen gas and stored at -20°C for no longer than 7 days (no significant increase in diet peroxidation is observed during the 7 day storage period). :1- 0 sons. I..I.'-.,.-.-I,.1- Diets were fed (free access) to mice to expose diets to actual experimental conditions. One hundred twenty mature female mice (Balb/c), from our breeding colony, were housed in 24 cages (5 mice/cage). Mice were housed in a temperature controlled (24°C) and light controlled (14 hr. light/day) room. Our protocol complied with the Guide for the Care and Use of Laboratory Animals (NRC 1985). Triplicate samples of each diet were obtained for analysis immediately after mixing (0 h) and 6 diet samples of each diet group were analyzed for each time period, i.e. 24, 48 and 72 h after being fed. Lipid peroxidation was determined by two methods, the PV (mmol of peroxides/kg oil) by the method of the American Oil Chemist's Society (1973) and by measuring TBARS“ (ug TBARS/g sample) using a modified TBA assay as previously described (Gonzalez et al. 1991). Modification of the TBA assay is as follows: to 10 g of diet, TBHO (0.2% of total fat, 2 g/kg sample) was added to diet samples before homogenization at 4°C to prevent further oxidation during the assay. The homogenates were treated with 4N HCl. A distillate was collected and combined with equal amounts of freshly prepared TBA solution. Samples were heated in boiling water, cooled and absorbance read on a Spectronic 2000 spectrophotometer (Bausch 8 Lamb, Rochester, NY). Once diet samples were ob were d0 map; Li; glands (10 an' at al. consum oil/1% and tr SigniI Al 01‘] (,4 E (Tat hlghei on d; thrOug 0f 81'1“. 78 were obtained, they were analyzed immediately for PV and TBARS which were done in duplicate and triplicate, respectively. Wham Lipid peroxidation was assessed in liver, heart, kidney, mammary glands and skeletal muscle (15 animals/group) and in whole body carcass (10 animals/group) of female mice by a modified TBA assay (Gonzalez et al. 1991). Mice were fed one of the following diets daily (non- consumed diet discarded daily) for 4 weeks: 20% corn oil, 19% fish oil/1%.corn oil or 19% fish oil/1% corn oil with added antioxidants, i.e., TBHO, 2.0%.w/w of oil and dl-alpha tocopherol acetate, 8 g/kg or 2000 IU/kg diet. At termination, organs were collected and stored in a NaCl solution (9g/L) at -20°C. These were analyzed for TBARS the following day. Animals used for whole body carcass TBARS were terminated by carbon dioxide inhalation and stored intact in a plastic zipper bag at -20°C and analyzed for TBARS the following day. WW Results were statistically analyzed by one-way analysis of variance and the Newman-Keuls multiple comparison test (Wilkinson 1989). Significance was set at p < 0.01. RESULTS At 0 h, the PV of the corn oil diet was lower than any of the fish oil diets except for the fish oil diets containing 2.0% TBHQ 1 vitamin E (Table 2). At 0 h, the PV of the non-supplemented fish oil diet was higher than the PV of the corn oil diet or the PV of each of the fish oil diets supplemented with antioxidants. This trend was observed throughout the other time periods (24, 48, 72 h). Increasing the level of BHT from 0.02% to 2.0% reduced the PV at all time points. TBHQ (2%) was sli of vita values. compare in the (0 h) a rate 0 diet 9 48 ans A1 highe 3). decre fish TBAR: BHT redu 'TBAJI that ‘lovn Sum finer i11c at, anc 3k. 79 was slightly more effective than BHT (2%) in reducing PV. The addition of vitamin E to the TBHO group further reduced, albeit slightly, PV values. PV at 0 h (immediately after diet preparation) was the lowest compared to all other time points (24, 48, 72 h) both in the corn and in the fish oil diets. Oxidation occurring between diet preparation (0 h) and after being fed for a day (24 h) was the period in which the rate of PV increase was at its highest. PV levels increased in all diet groups over time throughout the entire experimental period (0, 24, 48 and 72 h). At 0 hour, the dietary TBARS were lowest in the corn oil diet and highest in the fish oil diet not supplemented with antioxidants (Table 3). The addition of antioxidants (BHT or TBHO) to the fish oil diet decreased the levels of TBARS when compared to the non-supplemented fish oil diet. Increasing the level of BHT from 0.02% to 2.0% reduced TBARS at all time points. TBHO (2%) was slightly more effective than BHT (2%) in reducing TBARS. The addition of vitamin E to TBHO further reduced, albeit slightly, TBARS. However, at 0 and 24 h, the levels of TBARS were higher at all levels of antioxidant supplementation than that observed in the corn oil diet. Dietary TBARS values at 72 h were lower than those at 48 h except for the fish oil diet which was not supplemented with antioxidants. In all of the foregoing, the TBARS were measured at the standard wavelength of 532 nm. However, an increase in dietary TBARS was observed at wavelengths 455 nm and 495 nm at 72 h as compared to 455 nm and 495 nm wavelength readings at 0, 24 and 48 n time points (data not shown). TBARS values of organs (liver, heart, kidney, mammary gland, skeletal muscle) and whole bodies of mice fed a fish oil diet without antioxl whole I levels organs skelet fish 0 mice f Th air an to 0cc the oi radica higher Perios likely (0.02: Perox- Concel 0.021 TEduc the up NEVEr' “Yen s Synth. manif. Prod” 80 antioxidants were consistently higher than that observed in organs and whole bodies of corn oil fed mice (Table 4). The addition of high levels of antioxidants to the fish oil diet reduced TBARS values in all organs and in whole bodies (carcass). However, TBARS values in heart, skeletal muscle, mammary glands and whole bodies (carcass) of mice fed fish oil containing antioxidants were still higher than TBARS values of mice fed a corn oil diet. DISCUSSION The PV of fish oil diets constantly increased with time exposure to air and under normal feeding conditions. Peroxide formation is likely to occur as susceptible polyunsaturated fatty acids are available in the oil (Esterbauer et al. 1986). PY increases are catalyzed by free radical attacks at multiple sites. This might explain the observed higher rate of dietary peroxide formation attained during the 0 to 24 h period in which more susceptible sites for free radical attack are likely to be present. The addition of the synthetic antioxidant BHT (0.02%.w/w oil) to the fish oil diet significantly reduced lipid peroxidation when compared to the control fish oil diet. The higher concentration of BHT (2.0%.w/w of oil) was more effective than the 0.02%ilevel. TBHO, at the 2.0% level was slightly more effective in reducing peroxide formation. TBHO has been reported previously to be the most effective antioxidant for fish oil (Ke et al. 1977). Nevertheless, peroxide formation still occurred in the fish oil diet even when 100 times the AIN recommended level (0.02%.w/w of oil) for a synthetic antioxidant (2.0% TBHO or BHT) was added. Since peroxides manifest a transitory nature, over time conversion to secondary products of oxidation is favored. 81 Secondary products of lipid peroxidation (TBARS) measured by the TBA assay also displayed an increasing trend over the first 48 h. However, this trend was not apparent at 72 h where a decline in TBARS values was observed. In contrast to PV, TBARS formation depends on specific site oxidation accompanied by serial decomposition reactions (Dahle et al. 1962). Further examination of TBARS at shorter wavelengths (455 nm and 495 nm) suggest the possibility of a subsequent conversion of these secondary products of lipid peroxidation to other related compounds, most probably aldehydes such as alkanals and alk-2-enals (Kosugi et al. 1987) in the 72 h sample. Such TBARS determined at shorter wavelengths, should be expected to increase as secondary reactions proliferate with continuing oxidation. It is also possible that the observed reductions in TBARS values at 72 h may be due, at least in part, to possible reactions between aldehydes (i.e., malondialdehyde and/or alkanals) and proteins (Cuppett et al. 1989). The addition of synthetic antioxidants to the fish oil diets resulted in a substantial decrease in dietary TBARS, as was observed for PV, i.e., 2.0% TBHO was most effective, followed by 2.0% BHT, then 0.02% BHT. Once again, and importantly, the addition of synthetic antioxidants at a level 100 times greater than the AIN recommended level (0.02%.w/w of oil) did not totally prevent the formation of oxidation products in the fish oil diet. TBARS values in the antioxidant supplemented fish oil diets were still higher than that observed for corn oil. In regard to the oxidative susceptibility of the dietary oils used in this study, it is clear that corn oil (2 60% linoleic acid, 18:2,n-6) is considerably less prone to autoxidation than is fish oil. This difference is no doubt due to the high content 82 of long chain polyunsaturated fatty acids, i.e., EPA and DHA, in fish oil; such fatty acids are absent in corn oil (Table 2). Significant differences in lipid peroxidation were found among fish oil diets prepared from oils originating from separate batches from the same supplier (data not shown). The mean PV in freshly obtained fish oil in our studies was 7.25 (for comparison purposes, fresh stripped corn oil had a mean PV value of 0.97). This particular problem may prove to be very difficult to solve and may be due to an array of variables such as storage, processing, air and light exposure, temperature and seasonal variations (e.g. type of phytoplankton available for fish consumption). The rapid addition of a synthetic antioxidant to the oil as soon as the oil container is opened should deter or decrease further deterioration of the oil. Organs and whole body carcasses of mice fed the corn oil and fish oil diets (1 antioxidants) were also evaluated for TBARS. TBARS values obtained from certain organs and carcasses were directly related to the oxidation content of the ingested diet, although it is conceivable that this process can be further enhanced 1n_§11u. Even large doses of antioxidants (100x the AIN recommended level of a synthetic antioxidant and approximately 30x the amount of tocopherol acetate of that supplied by the vitamin mix) added to the fish oil diet failed to prevent the accumulation of secondary products of oxidation in certain organs (heart, skeletal muscle, mammary gland) and in whole bodies (carcass) of mice fed the supplemented fish oil diet. Lebovitz et al. (1990) reported similar results in tissue slices obtained from rats consuming a fish oil diet. They reported a decrease in secondary oxidation products in tissue slices of organs of rats receiving high levels of 83 dietary vitamin E, but also were unable to fully suppress the formation of these oxidation products. In contrast to what was observed in our study with heart, skeletal muscle and mammary glands, the livers and kidneys of mice fed fish oils containing antioxidants had significantly lower TBARS values than did the livers and kidneys of mice fed corn oil. Although the exact explanation is not certain, it is conceivable that at least for the liver, the capacity for storage of antioxidant lipid soluble components and/or the function of this organ in the production of lipoproteins may explain this phenomenon. In summary, based on the results presented in this communication, the AIN recommended level of addition of a synthetic antioxidant (0.02% w/w of oil) to diets containing high levels of polyunsaturated fatty acids appears to be insufficient to completely inhibit oxidative deterioration when such diets have fish oil as their fat base. Thus, a distinct recommendation for fish oil diets compared to other polyunsaturated fats seems appropriate. Meanwhile, in order to minimize oxidative deterioration in experimental diets containing high levels of fish oil, a synthetic antioxidant should be added in quantities substantially above the AIN recommendations. PV and TBARS values should be monitored throughout all feeding experiments involving dietary fish oil. Animals must be provided a fresh diet at least every 24 h in clean food jars; the remaining non-consumed diet should be discarded. In addition, the mixing of diets in an inert atmosphere (e.g. N2 or C02) should be mandatory. Freshly prepared diets should be stored in individual zipper bags in an amount sufficient for a single day’s feeding and kept at -20°C until fed. These recommendations will substantially reduce, albeit not completely eliminate, excessive 84 oxidative deterioration of experimental diets incorporating high levels of fish oil. The multitude of research laboratories studying the effects of dietary fish oil on physiological and/or pathological processes should be cognizant of these concerns. LITERATURE CITED American Institute of Nutrition (1977). Ad hoc committee on standards for nutritional studies. J. Nutr. 107: 1340-1348. American Institute of Nutrition (1980). Ad hoc committee on standards for nutritional studies. J. Nutr. 110: 1726. American Oil Chemist’s Society (1973). Official and recommended practices of the American Oil Chemist’s Society. Vol. 1, Methods: cd 8-53 and Ba 3-38, AOCS, Champaign, IL. Buford-Coulter, R. (1988). Extending shelf life by using traditional phenolic antioxidants. Cereal Foods World 33: 207-210. Chan, H.W.S. (1987). The mechanism of autoxidation: In Autoxidation of Unsaturated Lipids. (Chan, H.W.S., ed.) pp. 1-16, Academic Press, Lon on. Committee on Care and Use of Laboratory Animals, Institute of Laboratory Animal Resources Commission on Life Sciences, National Research Council (1985) Guide for the Care and Use of Laboratory Animals. National Institutes of Health publication No. 85-23 (rev.). Washington, D.C. Cuppett, S.L., Gray, J.I., Booren, A.M., Price, J.F. and Stachiw, M.A. (1989). Effect of processing variables on lipid stability in smoked Great Lakes Whitefish. J. Food Sci. 54: 52-54. Dahle, L.K., Hill, E.G. and Holman, R.T. (1962). The thiobarbituric acid reaction and the autoxidation of polyunsaturated fatty acid methyl esters. Arch. Biochem. a Biophys. 98: 253-255. Esterbauer, H., Benedetti, A., Lang, J., Fulceri, R., Fauler, G. and Comporti, M. (1986). Studies on the mechanism of formation of 4- hydroxy-nonenal during microsomal lipid peroxidation. Biochem. Biophys. Acta 876: 154-166. Fritsche, K.L. and Johnston, P.V. (1988). Rapid autoxidation of fish oil in diets without added antioxidants. J. Nutr. 118: 425-426. Gonzalez, M.J., Schemmel, R.A., Gray, J.I., Dugan, L., Jr., Sheffield, L.G. and Welsch, C.W. (1991). Effect of dietary fat on growth of MCF- 7 and MDA-MB 231 human breast carcinomas in athymic nude mice: Relationship between carcinoma growth and lipid peroxidation levels. Carcinogenesis 12: 1231-1235. 85 Ke, P.J., Nash, D.M. and Ackman, R.G. (1977). Mackerel skin lipids as an unsaturated fat model system for the determination of antioxidative potency of TBHO and other antioxidant compounds. J. Am. Oil Chem. Soc. 54: 417-420. Kosugi, H., Kato, T. and Kikugawa, K. (1987). Formation of yellow, orange and red pigments in the reaction of Alk-2-enals with 2- thiobarbituric acid. Analytical Biochem. 165: 456-464. Leibovitz, B.E., Hu, M.L. and Tappel, A.L. (1990). Lipid peroxidation in rat tissue slices: effect of dietary vitamin E, corn oil - lard and menhaden oil. Lipids 24: 125-129. Rasheed, A.A., Oldfield, J.E., Kaufmes, J. and Sinhuber, R.O. (1963). Nutritive value of marine oils: menhaden oil at varying oxidation levels with and without antioxidants in rat diets. J. Nutr. 79: 323- 332. Wilkinson, L. (1989). Systat: the system for statistics. Evanston, IL. Systat, Inc. 86 Table 1. Diet Composition Ingredients1 g/100 g diet“ Fat“ 20.00 Casein“ 20.17 dl-Methionine 0.35 Dextrose 32.18 Sucrose 16.09 AIN Mineral nix5 4.13 AIN Vitamin nixs5 1.18 Cellulose6 5.90 1All ingredients (dry components) were obtained from U.S. Biochemical Cos, Cleveland, OH except sucrose (ICN Biochemicals, Inc., Costa Mesa, CA . “High fat diets (20%iw/w) consisted of 20% corn oil or 19% fish oil (Menhaden)/1% corn oil. Menhaden oil contains 50 IU/kg of dl-alpha tocopherol. 3Corn oil (tocopherol stripped) was obtained from U.S. Biochemical Co., Cleveland OH. Fish oil (Menhaden) was obtained from Zapata Haynie Corp., Reedville, VA. “Vitamin free, high nitrogen (14.5%). ”J. Nutr. 107: 1340-1348, 1977. 6Celufil, non nutritive bulk. 87 Table 2. Effect of supplemented dietary antioxidants on peroxide values of fish (Menhaden) oil diets. Peroxide Yalu (mmol/kg oil) Time (hours) 0.1.9.1. 03 2.4 so 12 20%.corn 1.3310.17“ 8.67:0.34“ 11.67:0.88“ 19.67i1.46“ oil 19% fish 13.57¢1.2s 45.331220“l 712012.89“ 262.671;7.27ff oil/1% corn oil 19% fish 6.67:0.88c 25.671088c 39.6711.46“ 78.20¢2.09° oil/1% corn oil 3 0.02%.BHT 19% fish 4.01_+_0.se” 17.03¢0.se” 306712.48“ 48.66i4.42° oil/1% corn oil +2xesn3 19% fish 2.07:0.29a 14.0811.“b 22.011058c 38.67¢1.20c oil/1% corn oil 4 + 2% TBHO 19%.fish 1.50:0.29“ 11.671088a 17.051116b 30.151050b oil/1% corn oil +2%TBHO‘ + 2000 IU Vitamin E5 88 Footnotes for Table 2 1Statistical comparisons made at each time point between diets (values are means 1_SEM, n-6) determined by ANOVA and Neuman-Keuls multiple comparison test. “Value with different superscript letters in the same column are significantly different at p < 0.01. 3Butylated hydroxytoluene “Tertiary butyl-hydroxyquinone Sdl-alpha-tocopherol acetate 89 Table 3. Effect of supplemented dietary antioxidants on TBARS levels of fish (Menhaden) oil diets. Thiobarbituric acid reactive substances (ug TBARS/ g diet) Time (hours) miss 93 21 is 12 20% corn 0.3510.01“ 0.49:0.03“ 0.74:0.04“ 0.50:0.03b oil sex fish 2.931033f 6.53:0.15“ 13.95¢0.45° 19.06:0.67“ oil/1% corn oil 19%.fish 1.18:0.03“ 1.6810.14° 1.951009d 0.97:0.05d oil/1% corn oil 3 0.02% BHT 19% fish 0.70:0.03d 0.951004d 1.10:0.04c 0.80:0.05c oil/1% corn oil3 + 2% BHT 19% fish 0.63:0.03“ 0.84:0.05c o.91;0.03” 0.47:0.04” oil/1% corn oil ‘ + 2% TBHO 19% fish 0.49:0.03b 0.671004b 0.7910.05“ 0.3710.03“ oil/1% corn oil + 2% TBHQ‘ + 2000 IU Vitamin :5 90 Footnotes for Table 3 1Statistical comparisons made at each time point between diets (values are means 2 SEM, n-6) determine by ANOVA and Neuman-Keuls multiple comparison test. 2Value with different superscript letters in the same column are significantly different at p < 0.01. 3Butylated hydroxytoluene ‘Tertiary butyl-hydroxyquinone Sdl-alpha-tocopherol acetate 1 9 .Bflmoo aflflnooouuflnafev .QQEETHE gums” .8 .89 on gum? aficfiflfiafim an E38 88 «5 5 52 fig £385 5? §fi>~ .umopgumflao 393d! mggggmnuflngg “Egan Enhance—Hugu— Egon “.9983“: .vnm+§§§am>v agggflggguo gaggfim u 55»; :m .H 88+ awn. »~+ Hmo 05 “H amodfloé n8.oHR.o £86436 £8.3on nuodumnd “.8648... \do in «a HMO FH8 #H 08.c.+.mm.~ oB.ofl.o.~ 03636 ogéflmd 08.33... 08.386 \do in «3 «864%; «nodflmnd «noduond unaduned anodflnmd anoéwwoa $0 58 «8 a '1;an d3 g g «g qua g 383$ H 3.8.3 3% as 89536 9% 33 0353383 .amoaeuou Bofifio A8338 fiufiu8€5§mg~§5§§§§uouflufi $3an 92 Footnotes 1Supported by research grants AICR-9OBH05 and NIH CA-42876 to C.W. Welsch and by a Predoctoral mini grant of FSHN Department of MSU, an Equal Opportunity Fellowship and government of Puerto Rico, and University of Puerto Rico Fellowships to H.J. Gonzalez. Thanks to Marlene Green for the preparation of this manuscript. 2Abbreviations used: BHT, butylated hydroxytoluene; DHA, Docosahexaenoic acid; EPA, eicosapentaenoic acid; PV, Peroxide value; TBA, thiobarbituric acid; TBARS, thiobarbituric acid reactive substances; TBHQ, tertiary butylhydroxyquinone. Chapter 3 Dietary fish oil inhibits human breast carcinoma growth: A function of increased lipid peroxidation (Lipids 1993: 28,827-832) Dietary Fish Oil Inhibits Human Breast Carcinoma Growth: A Function of Increased Lipid Peroxidation Michael J.'Gonzaleza, Rachel A. Schemmel“, LeRoy Dugan Jr.a, J. Ian Gray‘, and Clifford H. Nelschb* aDepartment of Food Science and Human Nutrition and bDepartment of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan 48824 USA Running title: DIETARY FISH OIL AND HUMAN BREAST CARCINOMA GRONTH Key words: Fish oil, breast carcinoma, lipid peroxidation, linoleic acid, antioxidants Mailing Address:Dr. Clifford H. Helsch Department of Pharmacology and Toxicology Michigan State University East Lansing, MI 48824, USA Phone: 517-353-4549 Fax: 517-353-8915 94 95 *To whom correspondence should be addressed at Dept. of Pharmacology and Toxicology, 357 Giltner Hall, Michigan State University, East Lansing, Michigan, 48824, USA Abbreviations: CO, corn oil; EPA, eicosapentaenoic acid; FO, fish oil; DHA, docosahexaenoic acid; TBA, thiobarbituric acid assay; TBARS, thiobarbituric acid reactive substances; TBHO, tertiary butyl hydroquinone 96 ABSTRACT Female athymic nude mice were implanted 5,9, with human breast carcinoma MDA-M8231. Seven to 10 days later, the mice were divided into groups and fed a purified diet containing the following types of fat (% of diet): 1) 20% corn oil (CO); 2) 15% CO/5% fish (menhaden) oil (F0); 3) 10% CO/10% F0; 4) 5% CO/15% F0; 5) 1%»CO/19% F0 and 6) 1% CO/19% FO plus antioxidants [alpha tocopherol acetate, 2000 IU/kg diet and tertiary butyl-hydroquinone (TBHO), 2% of total fat]. The linoleic acid levels (% of diet) of the groups were 12.0%, 9.1%, 6.2%, 3.3%, 0.9% and 0.9%, respectively. After 6-8 weeks, the carcinomas were assessed for tumor volume (cm3) and assayed for thiobarbituric acid reactive substances (TBARS). Human breast carcinoma growth was suppressed in mice consuming FO diets without antioxidants compared to mice fed CO; the greater the amount of dietary FO fed, the greater the carcinoma growth suppression (p<0.05). The addition of antioxidants to the FO diet significantly (p<0.05) reversed the F0 induced carcinoma growth suppression. Concentrations of TBARS in the human breast carcinomas were increased in all the FO (without antioxidants) fed mice, compared to mice fed CO; the level of increase in TBARS was directly related to the increase in the level of F0 fed (p<0.05). The addition of antioxidants to the FO diet significantly (p<0.05) reduced the concentration of TBARS in the breast carcinomas. Thus, these results provide evidence that dietary FO can significantly suppress growth of human breast carcinoma MDA-MB231, even in the presence of substantial amounts of linoleic acid (3.3%.to 9.1%). The inhibitory effect of F0 on growth of these carcinomas was associated with an increased concentration of TBARS in the tumor tissue. In conclusion, 97 dietary F0 induced suppression of human breast carcinoma growth is a function, at least in part, of an accumulation of lipid peroxidation products in the tumor tissues. INTRODUCTION Fish oil (F0), which contains elevated levels of long chain omega-3 polyunsaturated fatty acids, in particular, eicosapentaenoic acid (EPA, 20:5) and docosahexaenoic acid (DHA, 22:6), has been shown to suppress mammary gland tumorigenesis in rodent models (1-4). In addition, F0 has been shown to inhibit the growth of human breast carcinomas maintained in immune deficient animals (5-9). To explain how F0 inhibits mammary tumorigenic processes, many investigators (2-4) have focused on EPA and/or BHA-induced alterations in eicosanoid metabolism. Other investigators (IO-11) suggest that dietary FO inhibits mammary tumorigenesis because it may not provide a sufficient amount of essential fatty acids such as linoleic acid (18:2). Indeed, a direct relationship between the amount of dietary linoleic acid and mammary tumor growth has been clearly shown (12). In contrast, we have explored yet another potential mechanism, i.e., that high levels of dietary F0 inhibit mammary tumorigenesis by causing an increased accumulation of cytostatic and/or cytotoxic lipid peroxidation products in the tumor tissue (9). In this communication, we further examine the relationship between dietary F0 and growth of human breast carcinomas in athymic nude mice by pursuing the following objectives: 1) to determine whether or not high levels of dietary F0, in the presence of substantial amounts of linoleic acid, can inhibit growth of these tumors and 2) whether or not there is a correlation between tumor growth and the quantity of lipid peroxidation products in the tumor 98 tissues of mice fed FO. MATERIALS AND METHODS An1m31§_3ng_d1gt‘ One hundred sixty-four female athymic nude mice (Harlan Sprague Dawley Inc. Madison, NI) 4 to 5 weeks old were used in these studies. The mice were housed under aseptic conditions (enclosed overhead laminar flow hood, sterilized cages, bedding and water) in a temperature (24°C) and light (14 h/day) controlled room. All experimental procedures pertaining to the athymic mice were performed under aseptic conditions. Autoclaved laboratory mouse chow (Purina Mills Inc., St. Louis, MO) was fed an 11bitum before and up to 7-10 days after human breast carcinoma transplantation. Thereafter, mice were fed purified diets (Table 1) 3d,11hitum. The percentages of the predominant fatty acids (1% or greater, manufacturer's specifications) of the dietary oils are shown in Table 2. The diets were prepared weekly in our laboratory, individually packed in small plastic zipper- sealed bags of sufficient size for one day’s feed, flushed with nitrogen, sealed and stored at -20°C. Mice were fed daily and non- consumed food discarded daily. Since the purified diets were not sterilized, antibiotics (bacitracin combined with streptomycin or neomycin, 1 g/L) were added to the distilled drinking water. flgman_brgast_carcingma_tran§plantatign, Palpable MDA-M8231 human breast carcinomas (American Type Culture Collection, Rockville, M0) were surgically excised from female athymic mice, cut into slices (2x4 mm, 0.1-0.3mm thick) and implanted into recipient mice. Mice were anesthetized with sodium pentobarbital (60 ug/g, 1,9,) prior to transplantation. An incision was made in the integument, the tumor slices were placed 5‘s. in the dorsum at distances from each other of 99 at least 2 cm, 3 to 4 slices/mouse. Between 1 and 3 palpable human breast carcinomas were pooled to provide the tissue slices. Mice were placed on the experimental diets 7-10 days after carcinoma transplantation. This period allows the carcinoma grafts to become established in the host animals before the onset of experimental dietary treatments. Athymic mice bearing human breast carcinoma grafts were fed the experimental diets for a period of 6-8 weeks. At the termination of the studies, all tumors were excised and their volume determined (V - 4/3 x r3) with a vernier caliper. Four separate studies (1-4) were carried out, each study was a complete individual experiment. Stydy_1, The experimental design of this study is provided in Table 3. The purpose of this study was to determine if varying levels of F0 mixed with varying levels of linoleic acid-rich corn oil (C0) are capable of modifying the human breast carcinoma growth suppressive effect of a F0 diet. In addition, we sought to determine if a relationship exists between human breast carcinoma growth and the level of lipid peroxidation products in the tumor tissue. The level of lipid peroxidation products of excised 10 g tumor composites in each group were determined by measuring 2-thiobarbituric acid reactive substances (TBARS) using a modified macro-thiobarbituric acid (TBA) assay (9). Magrg;IBA_a§§ay,__Subcutaneously growing human breast carcinomas were surgically excised, placed individually in a 0.9% NaCl solution, immediately frozen (-20°C) and stored for one day. Immediately before the TBA assay, the tumors were thawed and tumor samples pooled to obtain 10 9 composites. Once sufficient tumor homogenate was obtained from pooling (10 g composite), this constituted one sample. To obtain 100 other samples, other tumors within the same group were also pooled to form 10 9 samples. Three to 5 samples (10 9 composites) were done in triplicate for each group. An antioxidant [tertiary butyl-hydroquinone (TBHO), Eastman Chemical Products, Kingsport, TN], at 0.02% of total fat, as determined by a dry column extraction method (13) was added to the samples; such samples were homogenized (4°C) in a polytron homogenizer (Ultra Turrax SDT 1810, Tekmar Co., Cincinnati, OH). The homogenates were transferred into distillation flasks containing 2.5 mL 4N HCl plus 0.5 mL silicone antifoam (Thomas Scientific Co., Swedesboro, NJ) with sufficient distilled water to yield a total volume of 100 mL/flask and distilled to collect 50 mL. Five mL of this distillate plus 5 mL of freshly prepared TBA (0.2N aqueous solution, Sigma Chemical Co., St. Louis, M0) were used for the assay. Samples were prepared in triplicate, heated in a boiling water bath for 35 min to develop the pigment, cooled and absorbance read on a spectrophotometer (Spectronic 2000, Bausch G Lomb, Rochester, NY) at 532 nm. The absorbance values obtained were multiplied by an adjustment factor of 6.2 (14). Results were reported as ug of TBARS per g of sample. §tugy_z, The experimental design of this study is provided in Table 4. The purpose and experimental design of this study was similar to Study 1 but two additional dietary groups (1% CO/I9%.FO and 1% CO/19% F0 plus antioxidants) were added. A modified micro-TBA assay (15) was used in this study in order to individually assay each excised tumor for TBARS. 519:9;Ifia_gssay, Subcutaneously growing human breast carcinomas were surgically excised, frozen (-20°C) for one day, thawed and 101 individually homogenized (4°C) in the presence of an antioxidant (TBHO, 0.02% of total fat) in a polytron homogenizer. One mL of homogenate was combined with 2.0 mL of TBA (0.375% wt/vol), trichloroacetic acid (15.0% wt/vol), and HCl (0.25N) and mixed thoroughly. The solution was heated for 15 min in a boiling water bath. After cooling, the precipitate was removed by centrifugation (1,000 xg for 10 min) and the supernatant kept for analysis. The absorbance of the sample was determined at 535 nm against a blank that contains all reagents minus the homogenate. This TBA assay is a slightly modified version of the Buege and Aust assay (15). TBARS concentration of the sample was calculated using an extinction coefficient of 1.56 x 105 M’1 cm'1 (16) and reported as nmoles TBARS per mg of protein. The protein concentration of tumors was determined according to the method of Lowry et a1. (17). §1ndy_3. The experimental design of this study is shown in the legend of Figure 1. The purpose of this study was to determine if the concentration of TBARS in individual human breast carcinomas correlated with tumor size from a single F0 dietary group (1%1CO/19% F0). Excised tumors were individually assayed for TBARS by the modified micro TBA assay. §_1;_ugly__4__L The experimental design of this study is provided in Table 5. The purpose of this study was to determine if the length of time the F0 diet is allowed to remain in the food jar, under feeding conditions, 1ggg, 24, 48 and 72 h after feeding, influences human breast carcinoma growth and carcinoma concentration of TBARS. The jars with diet were changed every 24, 48 or 72 h. Excised tumors were individually assayed for TBARS by the modified micro-TBA assay. 102 Statist1cal_analy§g§, Data in Studies 1,2, and 4 were analyzed by one-way analysis of variance and the Newman-Keuls multiple comparison test (18). Significance was set at p<0.05. Data for Study 3 were examined by Pearson’s correlation coefficient (18). RESULTS flgman_brga§t_ga[§1ngma_grgwth, In Study 1, mean human breast carcinoma volumes (cm3) obtained from the 4 dietary groups at experiment termination ranked (from largest to smallest) as follows: 20% CO > 15% CO/5% FO > 10% CO/10% F0 > 5% CO/15% F0 (Table 3). Mean carcinoma volumes in mice fed each of the 3 F0 diets were significantly (p<0.05) smaller than the mean carcinoma volume of mice fed CO as their sole source of fat (20% CO). Body weight gains were not significantly different among the mice in the different dietary groups. In Study 2, mean human breast carcinoma volumes (cm3) obtained from the 6 dietary groups at termination of the experiment ranked (largest to smallest) as follows: 20% CO > 15% CO/5%.FO > 1% CO/I9% F0 plus antioxidants >10% CO/10% F0 > 5% CO/15% FO > 1%.CO/19% F0 (Table 4). Mean carcinoma volume in mice fed 20% CO was numerically higher than those of the 15% CO/5% F0 and 1% CO/19% F0 plus antioxidants fed mice, but this difference did not reach a level of significance. However, each of these 3 groups had mean carcinoma volumes which were significantly (p<0.05) larger than those produced by the 10%.C0/10% F0, 5% CO/15% F0, 1%.C0/19% F0 dietary groups. Body weight gains were not significantly different among the mice in the different dietary groups. In Study 4, mean human breast carcinoma volumes (cm3) obtained from the 3 dietary groups at termination of the experiment ranked (largest 103 to smallest) as follows: 24 h - 1% CO/19%.FO > 48 h - 1% C0/19% FO > 72 h - 1%.CO/19% F0 (Table 5). Mean carcinoma volumes in mice in which the diet (1%.CO/19% F0) was changed every 24 h was significantly larger (p<0.05) than in the 48 h or 72 h diet groups. The mean carcinoma volume of the 48 h diet group was significantly larger (p<0.05) than that of the 72 h diet group. Body weight gains were significantly lower (p<0.05) for the 72 h diet group compared to the 24 hour diet group. 1 Mil - '- a non. . - .- n. n: '1 . '9. ' ' =:: The ranking of mean human breast carcinoma TBARS concentrations in the 4 dietary groups of Study 1 at termination was: 5% CO/15% F0 > 10% CO/10% FO > 15‘% CO/5% F0 > 20% CO (Table 3). Mean human breast carcinoma TBARS concentrations in mice fed each of the 3 F0 diets were significantly (p<0.05) greater than the mean carcinoma TBARS value in mice fed CO as their sole source of fat (20% CO). In Study 2, the ranking of the mean human breast carcinoma TBARS concentrations in the 6 dietary groups at termination was: 1% CO/19% F0 > 5% CO/15% FO > 10% CO/10%FO > 15% CO/5% F0 > 20% CO > 1% CO/19% F0 plus antioxidants (Table 4). Mean tumor TBARS concentrations were virtually identical in 'the 1%.CO/19% F0 plus antioxidants and 20% C0 fed animals. Mean tumor “TBARS concentration in mice fed each of the four non-antioxidant {supplemented F0 diets were significantly (p<0.05) greater than the mean ‘tumor TBARS concentrations in mice fed the antioxidant supplemented F0 {and in the group fed only CD as their sources of fat. In Study 3, an inverse relationship is shown between volumes of individual human breast carcinomas and carcinoma TBARS concentrations in mice fed the 1% (IO/19% FD diet (p<0.05) (Figure 1), Le... the smaller the tumor 104 volumes, the greater the concentration of TBARS. In Study 4, mean carcinoma TBARS concentration in mice in which the diet (1%.CO/19% F0) was changed every 24 h was significantly lower (p<0.05) than in the 48 or 72 h diet groups (Table 5). The mean carcinoma TBARS of the 48 h diet group was significantly less (p<0.05) than that of the 72 h diet group. DISCUSSION This study provides evidence that supplementation of a high fat (CO) diet with even a modest amount of F0 (gygg, 15% CO/5% FO) can result in a suppression of growth of a human breast carcinoma (MDA- M8231) maintained in athymic nude mice. Other laboratories have provided evidence that FO containing diets inhibit mammary gland tumorigenesis in rodents (1-4) and suppress growth of human breast carcinomas in athymic nude mice (5-9). Such studies often utilize diets containing FO as their only source of fat. F0 is extremely low in the essential fatty acid linoleic acid (18:2). It is conceivable, therefore, that the suppression of mammary tumorigenesis exerted by diets high in F0 may be via essential fatty acid (linoleic acid) insufficiency. Indeed, supplementation of F0 diets with modest amounts of linoleic acid, in certain mammary tumor animal models, has been reported to block (4) or partially block (19) the tumorigenic inhibitory activities of a diet high in F0. In this study, we were still able to attain growth inhibition of MDA-M8231 human breast carcinomas in athymic nude mice by feeding FO diets containing substantial amounts of linoleic acid (3.3% -9.1%). Hhile the estimated nutrient requirement for adequate growth and reproduction of mice for linoleic acid is 0.3%.wt/wt of diet (20), the level required for 105 optimal tumorigenic processes may be substantially higher. For example, Ip gt al. (12) have reported that approximately 4.4% of linoleic acid in the diet is required for optimal (maximal) mammary tumor development in a rodent model. In our study, diets containing F0 and substantial amounts of linoleic acid (up to 9.1% of diet) still suppressed growth of this human breast carcinoma. Thus, it is likely that dietary FO-induced suppression of growth of human breast carcinoma MDA-M8231 is not due to insufficient dietary linoleic acid. Inhibition of growth of human breast carcinoma MDA-MB231 by dietary FO may be yin the accumulation within the tumor of cytostatic and/or cytolytic lipid peroxidation products. Evidence presented in our study which supports this conclusion are as follows. 1) As the FO content of the diet is increased, a concurrent significant increase in carcinoma TBARS concentrations is observed. 2) Increasing the FO content of the diet decreases carcinoma growth. 3) Those carcinomas that have the highest concentrations of TBARS have the least tumor growth. 4) Allowing the FO diet to remain in the food jar for as long as 72 h, a feeding protocol that increases substantially the level of TBARS and the peroxide values in the diet (21), results in an increased accumulation of TBARS in the carcinomas and a concordant suppression of carcinoma growth. 5) The addition of efficacious antioxidants to the F0 diet substantially decreases carcinoma TBARS concentrations and concurrently increases carcinoma growth. Collectively, these results, clearly support the concept that lipid peroxidation products are relevant to the process of dietary FO-induced inhibition of breast carcinoma growth. Recent evidence from our laboratory (21) indicates that dietary F0 derived lipid peroxidation products are formed 106 extensively in the food jar under normal feeding conditions. After feeding FO, such products are not only found in tumor tissue but are found in normal tissues as well. The mechanism by which lipid peroxidation products retard or inhibit tumor growth processes is not certain (22). Secondary products of lipid peroxidation (measured by the TBA assay) are capable of decreasing cell proliferation through damaging cell membranes, by changing cellular composition and/or cytoskeleton assembly. These modifications in the molecular architecture of the membrane can lead to the inactivation of membrane transport systems and/or membrane bound enzymes (23-25). This phenomenon may adversely affect the entering of cells into the cell cycle or it may accelerate their exit from it (1‘g, cell death). Furthermore, secondary products of lipid peroxidation can decrease tumor cell survival by inactivating polymerase reactions (26), forming inter- and/or intramolecular linkages between amino acid sulfhydryl groups and biomolecules (DNA, RNA and proteins) (27) and inhibiting polyamine synthesis (28). Such processes may not only result in inhibition of cell proliferation but may also lead to an increase in cell death. Indeed, Gabor and Abraham (4) reported that dietary F0 inhibits mammary tumor growth processes by increasing mammary tumor cell death rather than suppressing mammary tumor cell proliferation. Secondary products of lipid peroxidation, while under certain conditions may decrease tumor growth via a cytostatic/cytolytic mechanism, under other circumstances such products could stimulate tumor growth. For example, a peroxidation product of linoleic acid, 1yg‘, 13-hydroxy (hydroperoxy) octadecadienoate is more mitogenic to 107 rat colonic mucosa cells (29) or to hamster embryo fibroblasts (30) than is the parent compound. In addition, a peroxidation product of arachidonic acid, 1‘g; hydroxy (hydroperoxy) eicosatetraenoic acid stimulates rat colonic mucosa cell proliferation while the parent compound itself lacks stimulatory activities (29). Thus, peroxidation products derived from fatty acids in dietary F0 (particularly EPA, DHA and possibly linoleic acid) could be stimulatory to tumor growth processes. In the tumor model used in this study (MDA-M8231 human breast carcinoma maintained in athymic nude mice), however, we find no evidence of dietary FO induced stimulation of tumor growth. The results of our study clearly show that the addition to the FO diet of large amounts of antioxidants (Vit E and TBHO) significantly counteracted the inhibitory effects of dietary F0 on human breast carcinoma growth. In essence, high levels of antioxidants, in the presence of dietary FO, stimulated tumor growth. Other laboratories have provided evidence that supplementation of the diet with antioxidants inhibits tumorigenesis. Clearly dietary antioxidants can suppress carcinogen activation in experimental tumor models (31). Occasionally it is reported that antioxidant supplementation can suppress tumor growth (32,33), although there is considerable evidence to the contrary (34-36). Thus, it is likely that various antioxidants can suppress genotoxic induced initiation and/or progression events in tumorigenesis; evidence that antioxidants can effectively suppress tumor growth processes, however, has not been consistent or compelling. In conclusion, the results of this study provide evidence that dietary FO can significantly suppress the growth of MDA-MB231 human breast carcinomas maintained in athymic nude mice, even in the presence 108 of substantial amounts of dietary linoleic acid. The suppressive effect appears to be directly related to the accumulation of TBARS in the tumor tissues. ACKNONLEDGEMENTS This work was supported by research grants AICR-9OH05 and NIH-Ca42876 to C.M. Nelsch and an Equal Opportunity Fellowship from Michigan State University, a scholarship from the Administration of Economic Development of the Government of Puerto Rico and a Fellowship from the University of Puerto Rico (Medical Sciences) to M.J. Gonzalez. He also thank Mrs. Marlene Green for her excellent work in typing this manuscript. 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. 109 REFERENCES . Helsch, C.M. (1992) Cancer Res. 52, 2040s-20485. . Karmali, R.A., Marsh, J., and Fuchs, C. (1984) J. Natl. Cancer Inst. 73, 457-561. . Jurkowski, J.J., and Cave, Jr., N.T. (1985) J. Natl. Cancer Inst. 74, 1145-1150. . Gabor, H., and Abraham, S. (1986) J. Natl. Cancer Inst. 76, 1223- 1229. . Fernandes, G., Frieduchs, H., and Schultz, J. (1989) Breast Cancer Res. Treat. 14, 179A. . Pritchard, G.A., Jones, O.L., and Mansel, R.E. (1989) Brit. J. Surg. 76, 1069-1073. . Borgeson, C.E., Pardini, L., Pardini, R.S., and Reitz, C. (1989) Lipids 24, 290-295. . Gabor,H., Blank, E.H., and Ceriani, R.L. (1990) Cancer Lett. 52, 173-178. . Gonzalez, M. J, Schemmel, R. A, Gray, J. 1., Dugan, Jr. L., Sheffield, L. G. ., and Helsch, C. H. (1991) Carcinogenesis 12,1231- 1235. Braden, L.M., and Carroll, K.K. (1986) Lipids 21, 285-288. Abou-El-Ela, S.H., Prasse, K.H., Carroll, R., Nade, A.E., Dharwadkar, S., and Bunce, O.R. (1988) Lipids 23, 948-959. 1p, C., Carter, C.A., and 1p, M.M. (1985) Cancer Res. 45, 1997- 2001. Marmer, H.N., and Maxwell, R.J. (1981) Lipids 16, 365-371. Crackel, R.L., Gray, J.I., and Pearson, A.M. (1988) Food Chem. 28, 187-196. Buege, J.A., and Aust, S.D. (1978) Methods Enzymol. 11, 302-310. Hillis, E.D. (1969) Biochem. J. 113, 315-319. Lowry, D.H., Rosebrough, N.J., Farr, A.L., and Randall, R.J. (1951) J. Biol. Chem. 193, 265-275. Hilkinson, L. (1989) Systat: The system for statistics. SYSTAT, Inc. Evanston,1L. Abou-El-Ela, S.H., Prasse, K.H., Farrell, B.L., Carroll,R., Hade, A.E., and Bunce, O.R. (1989) Cancer Res. 49, 1434-1440. 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. 110 National Research Council (1978) Nutrient Requirements for Laboratory Animals. Nutrient Requirements of Domestic Animals. No. 10, p. 44. National Academy of Sciences, Washington, D.C. Gonzalez, M.J., Gray, J.I., Schemmel, R.A., Dugan, Jr., L., and Helsch, C.H. (1992) J. Nutr. 122, 2190-2195. Gonzalez, M.J. (1992) Med. Hypotheses 38, 106-110. Spector, A.A., and Yorek, M.A. (1985) J. Lipid Res. 26, 1015- 1035. Spector, A.A., and Burns, C.P. (1987) Cancer Res. 47, 4529-4537. Farber, J.L., Kyle, M.E., and Coleman, J.B. (1990) Lab. Invest. 62, 670-679. Roubal, N.T., and Tappel, A.L. (1966) Arch. Biochem. Biophys. 113, 5-8. Reiss, U., and Tappel, A.L. (1973) Lipids 8, 199-202. Dianzani, M.U. (1982) Free Radicals, Lipid Peroxidation and Cancer (McBrien, D.C., and Slater, T.F., eds.) pp. 129-158, Academic Press, London. Bull, A.M., Nigro, N.D., and Marnett, J.J. (1988) Cancer Res. 48, 1771-1776. Glasgow, R.C., Afshari, C.A., Barrett, J.C., and Eling, T.E. (1992) J. Biol. Chem. 267, 10771-10779. Hattenberg, L.M., Loub, H.D., Lam, L.K., and Speier, J.L. (1976) Fed. Proc. 35, 1327-1331. Hirose, M., Masuda, A., Inoue, T., Fukushima, S., and Ito, N. (1986) Carcinogenesis 7, 1155-1159. McCormick, D.L., Major, N., and Moon, R.C. (1984) Cancer Res. 44, 2858-2863. 1p, C. (1982) Carcinogenesis 3, 1453-1456. King, M.M., and McCay, P.B. (1983) Cancer Res. 43, 2485s-2490s. Beth, H., Berger, M.R., Aksoy, H., and Schmahl, D. (1987) Br. J. Cancer 56, 445-449. 111 Table 1. Diet Composition Ingredientsa Percentb fatc 20.00 caseind 20.17 dl-methionine 0.35 dextrose 32.18 sucrose 16.09 AIN mineral mix° 4.13 AIN vitamin nixf 1.13 celluloseg 5.90 aAll ingredients (dry components) were obtained from U.S. Biochemical Co. (Cleveland, OH) except sucrose (ICN Biochemicals, Inc., Costa Mesa, CA). bHigh fat diets (20% wt/wt) consisted of 20% corn oil or a fish oil (menhaden)/corn oil combination totaling 20%. cCorn oil (tocopherol stripped) was obtained from U.S. Biochemical Co. (Cleveland OH). Fish oil (menhaden) was obtained from Zapata Haynie Corp. (Reedville, VA). dVitamin free, high nitrogen (14.5%). °The mineral mixture supplied, per kg diet: CaHPO‘, 20.659; NaCl, 3.06 kg; potassium citrate, 9.09 g; KZSO‘, 2.159; MgO, 991.20 mg; manganous carbonate, 144.55 mg; ferric citrate, 247.8 mg; zinc carbonate, 66.08 mg; cupric carbonate, 12.39 mg; K103, 0.41 mg; sodium selenite, 0.41 mg; chromium potassium sulfate, 22.72 mg; sucrose, 4.87 g. 1!The vitamin mixture provided, per kg diet: thiamine HCl, 7.08 mg; riboflavin, 708 mg; pyridoxine HCl, 8.26 mg; nicotinic acid, 35.40 mg; D-calcium pantothenate, 18.88 mg; folic acid, 2.36 mg; DL-a-tocopheryl acetate, 236.00 mg; cholecalciferol, 0.03 mg; menaquionine, 0.06 mg; sucrose, 11.48 g. “Celufil, non nutritive bulk. 112 Table 2. Predominant Fatty Acids (%) Fatty acidsa Corn oil Fish (menhaden) oil Myristic (14:0) --- 6.7 Palmitic (16:0) 10.8 15.7 Palmitoleic (16:1) --- 8.7 Stearic (18:0) 2.1 2.7 Oleic (18:1) 26.5 14.3 Linoleic (18:2) 60.0 1.8 Linolenic (18:3) --- 1.5 Arachidonic (20:4) --- 2.2 Eicosapentaenoic (20:5) --- 15.5 Docosahexaenoic (22:6) --- 12.1 aFatty acids concentrations <1% are not included. and lipid permcidatim growth athymic rude mice. oilmthe minutes 'I‘able3. Effectofdifferattratinsofdietaxymoilardfish in mmmdmmalmm Mean tum lipid permddatim product. levels 1: (ug TBARS/g sanple):S.E. 2.55:9.31“ 20% mail 28.1:O.5 (12.0% linoleic acid) 15%camoil/ 5%fishoil 0.31:9.02d (9.1% linoleic acid) 10% com 011/ 10% fish oil 113 79 1.1610.61d 27.1¢O.7 (6.2% linoleic acid) o.sq:o.oa° O.8810.lld 27.x:o.3 5% com 011/ 15% fish oil (3.3% linoleic acid) bValues with different superscript letters in the same column are significantly different at p<0.05. M to 25 animals/gram. 114 .858 00 000888 aflnnodfiodn 08 538 38 05 5 000002 03008090 0500008 55 083x. .§\§ a 3 03mm 880 088:3 3.8 .000 H83 3 GE. 0:0 93H 88 a 05 8.500835 “03.0qu; 08.04004 ma 0.30.8 + do in «deo 880 an «300 303a 3.8 08.0H8.~ 08.0Hnmd om 0.04.0.8 do and «maxdo :80 3 Gang Odoafldd and; 08.00.84 08.386 mm 0.0Hm.o~ do and fixdo 500 an A300 088:: «0.0. 08.04004 03.04090 om Condom do and 3