SlideShare ist ein Scribd-Unternehmen logo
1 von 47
CANCER TREATMENT
     _CHEMOTHERAPY_ALTERNATIVES
                                    Compiled and Edited By
                     G Vijaya Raghaqvan, CEO, DVS BioLife Ltd, HYDERABAD.

                                       CONTENTS:
   1.   INTRODUCTION
   2.   CHEMOTHERAPY
   3.   COMMON CANCER DRUGS
   4.   ROLE OF UNUSUAL PLANT DERIVATIVES INCLUDING ALKALOIDS FROM POMEGRANATE
   5.   ROLE OF MICROBES LIKE E COLI IN DIAGNOSIS AND IN TREATMENT
   6.   COMPLEMENTARY APPROACHES

                              1. INTRODUCTION:
In the year 2000, malignant tumours were responsible for 12 per cent of the nearly 56
million deaths worldwide from all causes. In many countries, more than a quarter of
deaths are attributable to cancer. In 2000, 5.3 million men and 4.7 million women
developed a malignant tumour and altogether 6.2 million died from the disease. The
report also reveals that cancer has emerged as a major public health problem in
developing countries, matching its effect in industrialized nations.

Every year about 85,0000 new cancer cases are diagnosed in India resulting in about
58,0000 cancer related death every year.

Bladder Cancer, Melanoma, Breast Cancer, Non-Hodgkin Lymphoma, Colon and Rectal
Cancer, Pancreatic Cancer, Endometrial Cancer, Prostate Cancer, Kidney (Renal Cell)
Cancer, Skin Cancer (Nonmelanoma), Leukemia, Thyroid Cancer and Lung Cancer
are the common types of Cancer found worldwide.

Cancer management practices involve in screening, diagnostics, surgery,
chemotherapy, radiation, alternative and complimentary therapies.

Cancer management is partly based on weighing risk factors attributed to noninfectious
agents, human genes and epigenetic factors. Infectious disease causation has largely
been restricted to genes directly responsible for causing cancer after sustaining damage
i.e. oncogenes. Lately, evidence has emerged linking infectious agents to a number of
chronic diseases. These studies have recognized the influence that acute, atypical,
latent and chronic infections may play in tricking the immune system and affecting
disease etiology. Similar evidence is emerging in model systems with respect to the role
of infectious agents in gastrointestinal, liver and lung cancers. Although viruses have
been found in association with breast cancer, skepticism remains about a role for other
infectious agents, notably microbes in the disease etiology. Improved experimental
designs employed in different cancer studies and a less rigid definition of infectious
causation may aid in confirming or refuting a microbe-breast cancer connection. Cancer
recurrence could potentially be minimized and treatment options further tailored on a
case by case basis if microbes/microbial components/strain variants associated with
breast cancer are identified; probiotics are employed to reduce treatment side-effects
and if microbes could effectively be harnessed in immunotherapy.


                                 2. Chemotherapy
Chemotherapy is a cancer treatment that uses drugs to destroy cancer cells.

Chemotherapy can be used to:
   • Destroy cancer cells
   • Stop cancer cells from spreading
   • Slow the growth of cancer cells

Chemotherapy can be given alone or with other treatments. It can help other treatments
work better.

Chemotherapy can be given in these forms:
   • An IV (intravenously)
   • A shot (injection) into a muscle or other part of the body
   • A pill or a liquid that can be swallowed
   • A cream that is rubbed on the skin
   • Other ways:


Chemotherapy is achieved by one or more of the following.
   • Alkylating Agents
   • Antimetabolites
   • Cytotoxic Agents
   • Plant Derivatives
   • Microbial

The following are the common drugs used in Cancer.
   1. Acarbose
   2. Acivicin
   3. Aclarubicin
   4. Acodazole
   5. Acronine
   6. Actinomycin D
   7. Adenine Phosphate
   8. Adenosine
   9. Adozelesin
   10. Adriamycin
   11. Adrucil
   12. Alanosine
   13. Aldesleukin
   14. Alemtuzumab
   15. Alestramustine
   16. Alfacalcidol
   17. Alitretinoin
18. Alosetron Hcl
19. Alprostadil
20. Altretamine
21. Ambamustine
22. Ametantrone
23. Amifostine
24. Aminoglutethimide
25. Aminopterin Sodium
26. Amonafide
27. Amphotericin B
28. Amrubicin
29. Amsacrine Hcl
30. Amygdalin
31. Anastrozole
32. Anaxirone
33. Ancitabine
34. Angoroside C
35. Apixaban
36. Arecoline hydrobromide
37. Argatroban Anhydrous
38. Arsenic Trioxide
39. Ascomycin
40. Asparaginase
41. Atevirdine
42. Atosiban
43. Atrimustine
44. Axitinib
45. 5-Azacytidine
46. Azacitidine
47. Azasetron Hcl
48. Azatepa
49. Azathioprine
50. Azetepa
51. Batimastat
52. Bavituximab
53. Benaxibine
54. Bendamustine Hcl
55. Benexate
56. Benzodepa
57. Betamerphalan
58. Betulinic acid
59. Bevacizumab
60. Bicalutamide
61. Bisantrene
62. Bisnafide
63. Bizelesin
64. Bleomycin
65. Bofumustine
66. Bortezomib
67. Bosutinib
68. Brefeldin A
69. Brequinar
70. Bromacrylide
71. Bromocriptine
72. Bropirimine
73. Broxuridine
74. Budotitane
75. Busulfan
76. Calcifediol
77. Calcipotriol
78. Calcitriol
79. Calcium Folinate
80. Calcium Levofolinate
81. Campotothecine
82. Canertinib
83. Cantharidin
84. Capecitabine
85. Caracemide
86. Carbetimer
87. Carboplatin
88. Carboquone
89. Carmofur
90. carmustine
91. Carzelesin
92. Catharanthine Tartrate
93. Celastrol
94. Cemadotin
95. Cephalotaxin
96. Cetuximab
97. Chlorambucil
98. Chlorethylaminouracil
99. Chlormethine
100.           Chlornaphazine
101.           Cisplatin
102.           Cladribine
103.           Cladribine
104.           Clanfenur
105.           Clarithromycin
106.           Clevudine
107.           Clodronate Disodium
108.           Clofarabine
109.           Clomifene Citrate
110.   Colchiceinamide
111.   Colchicine
112.   Cordycepin
113.   Curcumin
114.   Cyclocytidine
115.   Cyclophosphamide
116.   Cyclosprin
117.   Cytarabine
118.   Cytidine
119.   Dacarbazine
120.   Dactinomycin
121.   Daniquidone
122.   Dapoxetine Hcl
123.   Dasatinib
124.   Datelliptium Chloride
125.   Daunoblastin
126.   Daunorubicin
127.   D-Bicuculline
128.   Decarbazine
129.   Decitabine
130.   Deferasirox
131.   Deflazacort
132.   Defosfamide
133.   Demecolcine
134.   Desonide
135.   Desoximetasone
136.   Desoxycorticosterone
137.   Desoxycortone
138.   Dexamethasone
139.   Dexasone
140.   Dexniguldipine
141.   Dexormaplatin
142.   Dexrazoxane
143.   Dezaguanine
144.   Diacetoxysciroenol
145.   Dianhydrodulcitol
146.   Dianhydrodulcitolum
147.   Diaziquone
148.   Dibrospidium Chloride
149.   Dinaline
150.   Dirithromycin
151.   Ditercalinium Chloride
152.   Ditiomustine
153.   Docetaxel
154.   Dolasetron mesylate
155.   Dopan
156.   Doxifluridine
157.   Doxorubicin
158.   D-tetrahydropalmatine
159.   Ecomustine
160.   Edatrexate
161.   Edelfosine
162.   Eflornithine
163.   Egenine
164.   Elinafide
165.   Elliptinium Acetate
166.   Elmustine
167.   Elsamitrucin
168.   Emitefur
169.   Enloplatin
170.   Enocitabine
171.   Enpromate
172.   Entecavir
173.   Entricitabine
174.   Enzastaurin
175.   Epipropidine
176.   Epirubicin
177.   Eptaplatin
178.   Erlotinib
179.   Esorubicin
180.   Estramustine
181.   Ethoglucid
182.   7-Ethyl-10-Hydroxycamptothecin Etoglucid
183.   Etoposide
184.   Exemestane
185.   Exenatide Acetate
186.   Ezetimibe
187.   Fadrozole
188.   Fazarabine
189.   Fenclonine
190.   Fenretinide
191.   Filgrastim
192.   Finasteride
193.   Fingolimod HCL(FTY720)
194.   Floxuridine
195.   Fludarabine
196.   Flumazenil
197.   Flumetasone
198.   Fluoracil
199.   5-Fluorouracil
200.   Fluorouracil
201.   Fluracil
202.   Flurocitabine
203.   Flutamide
204.   Folic Acid
205.   Formestane
206.   Formylmerphalan
207.   Fosquidone
208.   Fotemustine
209.   Fotretamine
210.   Ftorafur
211.   Fulvestrant
212.   Galamustine
213.   Galanthamine HBr
214.   Galocitabine
215.   Gefitinib
216.   Gemcitabine
217.   Gimeracil
218.   Giracodazole
219.   Glyfosfin
220.   Goserelin
221.   Granisetron Hcl
222.   Harpagoside
223.   Heparin sodium
224.   Hexarelin
225.   Homoharringtonine
226.   Hydrocamptothecine
227.   10-Hydroxycamptothecin
228.   Hydroxycamptothecin
229.   Hydroxycarbamide
230.   Hydroxyurea
231.   Ibacitabine
232.   Ibandronate Sodium
233.   Ibandronic Acid
234.   Idarubicin Hcl
235.   Idoxifene
236.   Ifosfamide
237.   Ilmofosine
238.   Ilomastat
239.   Imatinib
240.   Improsulfan
241.   Indirubin
242.   Intoplicine
243.   Iproplatin
244.   Irinotecan
245.   Ivabradine HCL
246.   Ixabepilone
247.   Ketotrexate
248.   Lanreotide
249.   Lapatinib
250.   Latanoprost
251.   L-biopterin
252.   Lenalidomide
253.   Lentinan
254.   Letrozole
255.   Leucovorin calcium
256.   Leuprolide Acetate
257.   Leuprorelin
258.   Leurubicin
259.   Leustatin
260.   Levothyroxine
261.   Lobaplatin
262.   Lofexidine HCL
263.   Loganin
264.   Lometrexol
265.   Lomustine
266.   Lonidamine
267.   Losoxantrone
268.   Lupeol
269.   Lurtotecan
270.   Lycopene
271.   Lysipressin
272.   Mafosfamide
273.   Mannomustine
274.   Mannosulfan
275.   Maraviroc
276.   Marimastat
277.   Masoprocol
278.   Mechlorethamine
279.   Mechlorethaminoxide
280.   Medrysone
281.   Megastrol acetate
282.   Melphalan
283.   Menogaril
284.   Mepitiostane
285.   Meprednisone
286.   6-Mercaptopurine
287.   Mercaptopurine
288.   Mesna
289.   Metamelfalan
290.   Methotrexate
291.   Methoxymerphalan
292.   Methylprednisolone
293.   Methylprednisolone Aceponate
294.   Methylprednisolone Suleptanate
295.   Meturedepa
296.   Metyrosine
297.   Miboplatin
298.   Miltefosine
299.   Minamestane
300.   Miproxifene
301.   Mitindomide
302.   Mitobronitol
303.   Mitoclomine
304.   Mitoflaxone
305.   Mitoguazone
306.   Mitolactol
307.   Mitomycin
308.   Mitonafide
309.   Mitopodozide
310.   Mitoquidone
311.   Mitosper
312.   Mitotane
313.   Mitotenamine
314.   Mitoxantrone
315.   Mitozolomide
316.   Mizoribine
317.   Mofarotene
318.   Monocrotaline
319.   Mubritinib
320.   Mustine
321.   Mycophenolate Mofetil
322.   Mycophenolic Acid
323.   Myricetin
324.   Myricitrin
325.   Nebivolol HCL
326.   Nedaplatin
327.   Nemorubicin
328.   Neptamustine
329.   Nigericin
330.   Nilotinib
331.   Nilutamide
332.   Nimustine
333.   Nitrocaphane
334.   Nocodazole
335.   Nolatrexed 2HCL
336.   Norcantharidin
337.   Ocaphane
338.   Octreotide
339.   Ondansetron
340.   Ormaplatin
341.   Oseltamivir Phosphate
342.   Oteracil potassium
343.   Oxaliplatin
344.   Oxipurinol
345.   Oxylycorium Acetate
346.   Paclitaxel
347.   Paliperidone
348.   palonosetron
349.   Pamidronate Disodium
350.   Pamidronic Acid
351.   Panitimumab
352.   Pazelliptine
353.   Pegaspargase
354.   Pemetrexed
355.   Pentamustine
356.   Pentetreotide
357.   Pentostatin
358.   Perfosfamide
359.   Phenoxybenzamine HCl
360.   Pincristine
361.   Pipobroman
362.   Piposulfan
363.   Pirarubicin
364.   Pirazofurin
365.   Piritrexim
366.   Pirlimycin
367.   Piroxantrone
368.   Plomestane
369.   Plusonermin
370.   Podophyllotoxin
371.   Polymyxin E
372.   Prednimustine
373.   Prednisolone
374.   Procarbazine
375.   Procodazole
376.   Prospidium Chloride
377.   Psoralen
378.   Pteropterin
379.   Pumitepa
380.   Ractopamine HCL
381.   Raloxifene Hydrochloride
382.   Raltegravir
383.   Raltitrexed
384.   Ramosetron Hcl
385.   Ranimustine
386.       Rapamycin
   387.       Razoxane
   388.       Resveratrol
   389.       Retigabine
   390.       Riboprine
   391.       Risedronic acid
   392.       Ristocetin A
   393.       Ritrosulfan
   394.       Rituximab
   395.       Rocuronium Bromide
   396.       Roflumilast
   397.       Rogletimide
   398.       Ropinirole HCL
   399.       Rosuvastatin Calcium
   400.       Rubitecan
   401.       Sarcolysin
   402.       Sargramostim
   403.       Sebriplatin
   404.       Semustine
   405.       Sermorelin
   406.       Simtrazene
   407.       Sitagliptin Phosphate
   408.       Sizofiran
   409.       Sobuzoxane
   410.       Sodium Borocaptate[10B]
   411.       Solaziquone
   412.       Sonermin
   413.       Sorafenib
   414.       Spiclomazine
   415.       Spirogermanium
   416.       Spiromustine

Following are some excerpts from public domain.

Conventional Alkylating Agents
Alkylating agents are one of the earliest and most commonly used chemotherapy agents
used for cancer treatments. Their use in cancer treatments started in early 1940s.
Majority of alkaline agents are active or dormant nitrogen mustards, which are poisonous
compound initially used for certain military purposes. Chlorambucil, Cyclophosphamide,
CCNU, Melphalan, Procarbazine, Thiotepa, BCNU, and Busulfan are some of the
commonly used alkylating agents.
They are more effective in treating slow-growing cancers such as solid tumors and
leukemia

Traditional Antimetabolites
Structure of antimetabolites (antineoplastic agents) is similar to certain compounds such
as vitamins, amino acids, and precursors of DNA or RNA, found naturally in human
body. Antimetabolites help in treatment cancer by inhibiting cell division thereby
hindering the growth of tumor cells. These agents get incorporated in the DNA or RNA to
interfere with the process of division of cancer cells. They are commonly used to treat
gastrointestinal tract, breast, and ovary tumors.
Methotraxate, which is a commonly used antimetabolites chemotherapy agent, is
effective in the S-phase of the cell cycle. It works by inhibiting an enzyme that is
essential for DNA synthesis.
6-mercaptopurine and 5-fluorouracil (5FU) are two other commonly used
antimetabolites. 5-Fluorouracil (5-FU) works by interfering with the DNA components,
nucleotide, to stop DNA synthesis. This drug is used to treat many different types of
cancers including breast, esophageal, head, neck, and gastric cancers. 6-
mercaptopurine is an analogue of hypoxanthine and is commonly used to treat Acute
Lymphoblastic Leukemia (ALL).
Other popular antimetabolite chemotherapy drugs are Thioguanine, Cytarabine,
Cladribine. Gemcitabine, and Fludarabine.

Anthracyclines
Anthracyclines are daunosamine and tetra-hydronaphthacenedione-based
chemotherapy agents. These compounds are cell-cycle nonspecific and are used to
treat a large number of cancers including lymphomas, leukemia, and uterine, ovarian,
lung and breast cancers.
Anthracyclines drugs are developed from natural resources.
Daunorubicin is developed by isolating it from soil-dwelling fungus Streptomyces.
Doxorubicin, which is another commonly used anthracycline chemotherapy agent, is
isolated from mutated strain of Streptomyces.
Doxorubicin is more effective in treating solid tumors.
Idarubicin, Epirubicin, and Mitoxantrone are few of the other commonly used
anthracycline chemotherapy drugs.
Anthracyclines work by forming free oxygen radicals that breaks DNA strands thereby
inhibiting DNA synthesis and function.
These chemotherapeutic agents form a complex with DNA and enzyme to inhibit the
topoisomerase enzyme.
Topoisomerase is an enzyme class that causes the supercoiling of DNA, allowing DNA
repair, transcription, and replication.

Antitumor antibiotics
Antitumor antibiotics are also developed from the soil fungus Streptomyces. These
drugs are widely used to treat and suppress development of tumors in the body. Similar
to anthracyclines, antitumor antibiotics drugs also form free oxygen radicals that result in
DNA strand breaks, killing the growth of cancer cells. In most of the cases, these drugs
are used in combination with other chemotherapy agents.
Bleomycin is one of the commonly used antitumor antibiotic used to treat testicular
cancer and hodgkin’s lymphoma.

Monoclonal antibodies
The treatment is known to be useful in treating colon, lung, head, neck, and breast
cancers. Some of the monoclonal drugs are used to treat chronic lymphocytic leukemia,
acute myelogenous leukemia, and non-Hodgkin's lymphoma.
Monoclonal antibodies work by attaching to certain parts of the tumor-specific antigens
and make them easily recognizable by the host’s immune system. They also prevent
growth of cancer cells by blocking the cell receptors to which chemicals called ‘growth
factors’ attach promoting cell growth.
Monoclonal antibodies can be combined with radioactive particles and other powerful
anticancer drugs to deliver them directly to cancer cells. Using this method, long term
radioactive treatment and anticancer drugs can be given to patients without causing any
serious harm to other healthy cells of the body.

Platinum
Platinum-based chemotherapy agents work by cross-linking subunits of DNA. These
agents act during any part of cell cycle and help in treating cancer by impairing DNA
synthesis, transcription, and function.
Cisplatin, although found to be useful in treating testicular and lung cancer, is highly
toxic and can severely damage the kidneys of the patient. Second generation platinum-
complex carboplatin is found to be much less toxic in comparison to cisplatin and has
fewer kidney-related side effects. Oxaliplatin, which is third generation platinum-based
complex, is found to be helpful in treating colon cancer. Although, oxaliplatin does not
cause any toxicity in kidney it can lead to severe neuropathies. Platinum-based drugs
are often used for treatment of mesothelioma.


                      Role of UNUSUAL Plant Derivatives
They are primarily categorized into four groups: topoisomerase inhibitors, vinca
alkaloids, taxanes, and epipodophyllotoxins.

Topoisomerase inhibitors are chemotherapy agents are categorized into Type I and
Type II Topoisomerases inhibitors and they work by interfering with DNA transcription,
replication, and function to prevent DNA supercoiling.
        Type I Topoisomerase inhibitors: These chemotherapy agents are extracted from
        the bark and wood of the Camptotheca accuminata.
    • Type II Topoisomerase inhibitors: These are extracted from the alkaloids found in
        the roots of May Apple plants.
    • Amsacrine, etoposide, etoposide phosphate, and teniposide are some of the
        examples of type II topoisomerase inhibitors.

Vinca alkaloids
Vinca alkaloids are derived from the periwinkle plant, Vinca rosea (Catharanthus
roseus) and are useful in treating leukemias. They are effective in the M phase of the
cell cycle and work by inhibiting tubulin assembly in microtubules.
Vincristine, Vinblastine, Vinorelbine, and Vindesine are some of the popularly used vinca
alkaloid chemotherapy agents used today. Major side effect of vinca alkaloids is that
they can cause neurotoxicity in patients.

Taxanes
Taxanes are plant alkaloids that are isolated from the bark of the Pacific yew tree,
Taxus brevifolia.. Paclitaxel and docetaxel are commonly used taxanes. Taxanes work
in the M-phase of the cell cycle and inhibit the function of microtubules by binding with
them. Taxanes are used to treat a large array of cancers including breast, ovarian, lung,
head and neck, gastric, esophageal, prostrate and gastric cancers. The main side effect
of taxanes is that they lower the blood counts in patients.
Epipodophyllotoxins
Epipodophyllotoxins chemotherapy agents are extracted from the American May Apple
tree (Podophyllum peltatum).
Etoposide and Teniposide are commonly used epipodophyllotoxins chemotherapy
agents which are effective in the G1 and S phases of the cell cycle. They prevent DNA
replication by stopping the cell from entering the G1 phase and stop DNA replication in
the S phase.

Phytoestrogens and Cancer Treatment
Phytoestrogens are polyphenol compounds of plant origin that exhibit a structural
similarity to the mammalian steroid hormone 17β-oestradiol. In Asian nations the staple
consumption of phyto-oestrogen-rich foodstuffs correlates with a reduced incidence of
breast cancer. Human dietary intervention trials have noted a direct relationship between
phyto-oestrogen ingestion and a favourable hormonal profile associated with decreased
breast cancer risk. However, these studies failed to ascertain the precise effect of dietary
phyto-oestrogens on the proliferation of mammary tissue. Epidemiological and rodent
studies crucially suggest that breast cancer chemoprevention by dietary phyto-oestrogen
compounds is dependent on ingestion before puberty, when the mammary gland is
relatively immature. Phyto-oestrogen supplements are commercially marketed for use by
postmenopausal women as natural and safe alternatives to hormone replacement
therapy. Of current concern is the effect of phyto-oestrogen compounds on the growth of
pre-existing breast tumours. Data are contradictory, with cell culture studies reporting
both the oestrogenic stimulation of oestrogen receptor-positive breast cancer cell lines
and the antagonism of tamoxifen activity at physiological phyto-oestrogen
concentrations. Conversely, phyto-oestrogen ingestion by rodents is associated with the
development of less aggressive breast tumours with reduced metastatic potential.
Despite the present ambiguity, current data do suggest a potential benefit from use of
phyto-oestrogens in breast cancer chemoprevention and therapy.

Phyto-oestrogens may be classified into a number of principal groups [2,7-9]: the
isoflavones (genistein, daidzein, biochanin A), the lignans (enterolactone, enterodiol),
the coumestans (coumestrol) and the stilbenes (resveratrol). As illustrated in Fig. 1, all
are polyphenols sharing structural similarity with the principal mammalian estrogen 17β-
oestradiol. Shared features include the presence of a pair of hydroxyl groups and a
phenolic ring, which is required for binding to the oestrogen receptor (ER) subtypes α
and β. The position of the hydroxyl groups appears to be important in determining ER
binding ability and transcriptional activation, with maximal potency achieved at positions
four, six and seven [10-12]. The isoflavones are naturally found in soybeans and soy-
based food products, including tofu, soy milk, textured soy protein and miso. Lignans are
present in flaxseed and most fruit and vegetables, and the predominant dietary source of
stilbenes is peanuts, grapes and red wine [7,13]. The coumestans are much less
frequently consumed within the human diet, but they are more potent activators of ER
signalling pathways than are the isoflavones genistein and daidzein [10,14]. By contrast,
the stilbene resveratrol is the least potent activator of ER signalling [11].
The isoflavones are present in soy as β-glucosides. Metabolism by the gastrointestinal
microflora yields a number of metabolites including equol and O-desmethyl-angolensin.
Parental compounds and their metabolites are absorbed into the bloodstream, becoming
rapidly detectable in the plasma and urine [15-19]. Plasma isoflavone concentrations are
considerably elevated in Asian populations as compared with in western ones. A recent
comparison of Japanese and UK females revealed an almost 20-fold increase in plasma
genistein levels in the Japanese cohort, and daidzein concentrations were similarly
elevated by 18-fold [20]. Plasma isoflavone concentrations may accumulate to
approximately 100- to 1000-fold higher than endogenous oestradiol levels following the
ingestion of soy-rich meal. However, research suggests a decreased ER binding affinity
of isoflavone compounds as compared with the mammalian oestrogens [9,10,21,22].
Competition binding assays revealed a 50-fold lower binding affinity of genistein for
cytoplasmic ER sites as compared to 17β-oestradiol [23].
The complete metabolic activation of soy isoflavones is proposed to occur locally within
target tissue. In support of this hypothesis, the analysis of tissue culture supernatants
from genistein and biochanin A treated MCF-7 and T47-D cells revealed the presence of
hydroxylated and methylated isoflavone metabolites [24]. Current research suggests a
role for the CYP family of cytochrome P450 enzymes in the intratumour metabolism of
phyto-oestrogen compounds [25,26]. The CYP1B1 enzyme is expressed in a wide range
of human tumour types, including breast [27]; however, expression is absent within
normal tissue. CYP1B1 is proposed to catalyze the hydroxylation of resveratrol to yield
the related stilbene piceatannol [26]. Piceatannol is a tyrosine kinase inhibitor with
antileukaemic properties, which differs in structure from resveratrol by the presence of
an additional hydroxyl group. A number of plant flavonoids are also putative substrates
for the CYP family of enzymes [25]. Maubach and coworkers [28] recently reported the
use of high-performance liquid chromatography to quantify isoflavones in normal breast
biopsy tissue following consumption of soy for 5 consecutive days. Equol concentrations
were approximately fivefold higher in the breast tissue homogenates than in serum,
providing further evidence for the metabolism of phyto-oestrogen compounds within
mammary tissue.

Role of phyto-oestrogens in breast cancer
Serum concentrations of 17β-estradiol are approximately 40% lower in Asian women
than in their Caucasian counterparts [29]. A low lifetime exposure to oestrogen is
associated with a reduced risk for breast cancer. Human dietary intervention studies
revealed a direct association between the modest consumption of soy products and a
reduction in circulating steroid hormone levels. Daily consumption of 154 mg isoflavones
for the duration of a single menstrual cycle correlated with substantially decreased
plasma concentrations of 17β-oestradiol and progesterone in a cohort of premenopausal
women [18]. A longer-term study conducted by Kumar and coworkers [30] similarly
reported a moderate decrease in serum oestradiol and oestrone levels following daily
ingestion of 40 mg isoflavones for 3 months. Menstrual cycle length was increased by
3.52 days, and the follicular phase of the cycle was extended by 1.46 days. Increased
menstrual cycle length may serve to reduce the total number of cycles per lifetime,
therefore decreasing the total exposure of breast epithelia to endogenous oestrogens.
Conversely, a year-long dietary intervention trial involving 34 premenopausal women
failed to reveal a significant effect of 100 mg/day isoflavone consumption either on
menstrual cycle length or on serum levels of various steroid hormones, including
oestrone, oestradiol and progesterone [19].
As a possible explanation for these contradictory data, a study conducted by Duncan
and coworkers [31] revealed differential hormonal effects of soy isoflavones depending
on the ability to excrete the daidzein metabolite equol. Daily ingestion of 10 mg soy
protein by premenopausal equol excretors resulted in a hormonal profile associated with
reduced breast cancer risk, characterized by lowered plasma levels of oestrone,
oestrone–sulphate and testosterone. Hormone levels however remained unchanged in
the equol nonexcretors after soy ingestion.
The reduction in steroid hormone levels by phyto-oestrogens is proposed to occur via
the direct regulation of 17β-oestradiol biosynthesis and metabolism. Phytochemicals
isolated from vegetable extracts effectively suppress the activity of the aromatase
enzymes, which are responsible for conversion of androgens to oestrogens [32].
Isoflavone concentrations of 1–10 μmol/l similarly reduced by 50% the activity of the
estradiol biosynthetic enzymes 3β-hydroxysteroid dehydrogenase and 17β-
hydroxysteroid dehydrogenase [10]. The daily ingestion of 113–202 mg isoflavones by
premenopausal women correlated with a 40% increase in the urinary excretion of 2-
hydroxyestrone, a putative anticancer metabolite of 17β-oestradiol [33]. The above
studies thus suggest a dual chemoprotectant mechanism of soy, in which the
isoflavones suppress steroid hormone biosynthesis while promoting the metabolism of
oestradiol to the protective 2-hydroxylated metabolites.
Despite their apparent effect on endogenous hormone levels, the role of phyto-
oestrogens in breast cancer initiation and development is unclear. Few studies to date
have addressed the effects of long-term phyto-oestrogen exposure in humans. Daily
dietary supplementation with 45 mg soy isoflavone for 14 days correlated with increased
proliferation of normal breast epithelia in a group of 48 premenopausal women [16].
Expression of the ER target protein progesterone receptor was upregulated, suggesting
an oestrogenic effect. An identical trial using a larger cohort of 84 premenopausal
women conversely found no significant effect of soy consumption on the proliferation of
normal breast tissue [17]. A number of recent epidemiological studies similarly failed to
correlate soyfood consumption with reduced breast cancer risk. A Japanese prospective
study conducted in a cohort of approximately 35,000 women [34] revealed no significant
association between soy consumption during adulthood and breast cancer incidence.
The retrospective analysis of soy food intake in a multiethnic cohort of non-Asian breast
cancer patients and control individuals residing in the USA similarly failed to correlate
soy intake with breast cancer risk [35].
Increasing epidemiological evidence suggests that the chemoprotectant effects of phyto-
oestrogens are dependent on a lifelong exposure from childhood. A retrospective study
revealed decreased soyfood intake during adolescence in a cohort of 1459 Chinese
breast cancer patients, as compared with age-matched control individuals [36]. Daily soy
consumption between the ages of 13 and 15 was estimated at 6.45 g in the patient
group, increasing to 7.23 g in the control cohort. A potential flaw in the study, however,
concerns the ability of women up to the age of 64 years to recall accurately the precise
soyfood quantities consumed many years earlier during adolescence. These
observations may nonetheless explain the apparent lack of a growth inhibitory effect of
soy isoflavones in the adult dietary intervention studies discussed above. Isoflavones are
detectable in breast milk following soy consumption [15], implying that the lower breast
cancer incidence in Asian countries may be attributable to phyto-oestrogen exposure
from birth via breast-feeding. Rodent studies have accordingly revealed the effective
transfer of genistein from maternal milk to offspring [37].

Rodent breast cancer models
Most available information regarding the effects of phyto-oestrogens on tumour initiation
and the growth of pre-existing tumours is derived from rodent studies. A number of
similarities do exist between mammary gland development in rodents and humans. In
both species the differentiation of breast tissue to form lobules and terminal end-bud
structures occurs prepubertally. Further maturation does take place throughout
adulthood, giving rise to alveolar buds, which become alveoli during pregnancy and
lactation [38].
Rodent dietary intervention studies using phyto-oestrogens have reported
chemopreventive activities when feeding is initiated before puberty, at a time when the
mammary gland is undergoing development [3,38-40]. The consumption of a resveratrol-
supplemented diet by adolescent rats served to decrease sensitivity to the chemical
carcinogens 7,12-dimethylbenz(a)anthracene (DMBA) and N-methyl-N-nitrosaurea
[39,40]. DMBA treatment induced mammary tumours in 45% of the resveratrol-treated
rodents, increasing to 75% in the group receiving a control diet. An extended tumour
latency period in excess of 3 weeks was observed in the resveratrol treatment groups,
with the resultant tumours retaining a more differentiated morphology as compared with
control animals [18,39]. Resveratrol consumption was also associated with the reduced
mammary expression of a number of proteins that are putatively involved in malignant
progression, including cyclo-oxygenase-2, matrix metalloprotease-9 and nuclear factor-
κB. Similar findings have been noted in prepubertal rats fed an isoflavone-containing diet
before tumour initiation using DMBA. Despite having no effect on mammary tumour
incidence, soy isoflavone consumption was associated with an increased tumour latency
period. The resultant tumours excised from the soy-fed animals were smaller in size and
exhibited a more differentiated phenotype compared with control animals [42].
It has been proposed that phyto-oestrogens protect against cancer development in
adolescent rodents by promoting maturation of the mammary gland. Analysis of breast
tissue from prepubertal rats injected with genistein revealed a decrease in the number of
immature terminal end-buds, together with an increase in the more differentiated lobules
type II [38]. Genistein treatment of human breast cancer cell lines has similarly been
found to induce the expression of a number of maturation markers, including casein, lipid
droplets and intercellular adhesion molecule-1 [43].
The effect of soy isoflavones on spontaneous tumour development was recently
investigated using neu-ErbB2 over-expressing transgenic mice, which characteristically
develop multiple mammary tumours during adulthood [40]. Tumour initiation was
temporarily delayed following the consumption of an isoflavone mix, but no
chemoprotective effects were observed in mice consuming either genistein or daidzein in
isolation. An equal rate of tumour growth was noted in the control and treatment groups,
although the isoflavone group exhibited a lower incidence of lung metastases [40].
Antimetastatic activities of the isoflavones were similarly revealed in a study in which
mice were fed an isoflavone-supplemented diet before injection with the metastatic 4526
mammary carcinoma cell line [44]. The isoflavone diet was continued following surgical
excision of the resultant mammary tumour at a size of 1.0 cm diameter. Both the
incidence and size of macroscopically detectable lung metastases were significantly
reduced in the soy-fed mice, suggesting a potential clinical application of soy isoflavones
in the prevention of metastasis.

Phyto-oestrogens and tamoxifen
The selective ER modulator tamoxifen is used clinically in the adjuvant treatment of
oestrogen-dependent breast cancer. The drug is also administered as a prophylactic to
individuals who are at high risk for developing the disease [45,46]. Side effects
associated with tamoxifen therapy include menopause-like symptoms such as hot
flushes, joint pain, sleep disorders and depression, which may be reduced by the use of
hormone replacement therapy (HRT) [47,48]. Long-term HRT is associated with an
increased risk for mammary carcinogenesis, and its use by breast cancer patients is
therefore discouraged. As a natural alternative, patients may self-medicate with soy
isoflavone supplements to alleviate the tamoxifen-induced menopausal symptoms [49].
Published literature regarding the ingestion of dietary phyto-oestrogens by breast cancer
patients and survivors is, however, controversial [50,51].
The consumption of genistein by athymic mice antagonized the ability of tamoxifen to
inhibit the proliferation of oestrogen-dependent mammary tumours [52]. Tumour
suppression by tamoxifen correlated with decreased expression of the ER-inducible
genes presenelin-2 (pS2) and cyclin D1. Tumour growth was significantly enhanced in
mice simultaneously exposed to tamoxifen and genistein, whereas levels of pS2 and
cyclin D1 expression were increased. Physiological concentrations of genistein were
similarly found to reverse the antagonistic effects of 4-hydroxytamoxifen on ER signalling
pathways [53], promoting the binding of ER-α to the positively acting steroid receptor
coactivator (SRC)-1. A recent tissue culture study conversely reported a synergistic
antiproliferative effect of tamoxifen and genistein [54]. The proliferation of a panel of
dysplastic and cancerous breast cell lines was inhibited by tamoxifen in a dose-
dependent manner, and growth was more potently suppressed by combined treatment
with tamoxifen and genistein.

Hormone-dependent mechanisms of phyto-oestrogen action
Oestrogen signalling typically involves the diffusion of ligand through the cell cytoplasm
and subsequent binding to the nuclear receptor subtypes ER-α and ER-β. Ligand-bound
receptors dimerise and associate with oestrogen response element (ERE) and activator
protein-1 element located in the promoter region of target genes, thereby activating
transcription. The association between receptor dimers and DNA response elements is
enhanced by the binding of cofactor proteins, such as amplified in breast cancer-1,
thyroid hormone receptor-associated protein, SRC-1, glutamate receptor interacting
protein-1 and translation initiation factor 2 [55]. Examples of ERE-induced genes include
PR, c-fos, bcl-2 and cathepsin D, whereas pS2 and cyclin D1 are transcribed via the
activator protein-1 response element.
In breast carcinoma cell lines containing functional ER subtypes the isoflavones exert a
biphasic growth effect, stimulating cellular proliferation at concentrations below 5 μmol/l
and inhibiting growth in a dose-dependent manner at elevated doses [23,43,56,57].
Growth inhibition correlates with decreased DNA synthesis and cell cycle arrest at the
G2/M checkpoint [23,54,56,58-60]. Current research suggests a principal signalling role
of ER-β in response to isoflavone exposure [61]. Whereas 17β-oestradiol binds to ER-α
and ER-β with equal affinity, the soy isoflavones selectively associate with ER-β [62].
Receptor binding assays revealed an eightfold to 16-fold increase in the affinity of
genistein, daidzein and biochanin A for ER-β as compared with ER-α [63]. In ER-
negative breast cancer cells transfected to express ER-α alone, genistein was only
weakly able to stimulate gene transcription through the ERE. By contrast, genistein
effectively bound to ER-β and promoted the association of cofactor proteins, thereby
regulating downstream ER-β-mediated gene transcription [62]. The preferential binding
affinity of genistein for ER-β similarly resulted in a respective 12,000-fold and 33-fold
increase in the recruitment of translation initiation factor-2 and SRC-1a to ER-β as
compared with ER-α [22]. An enhanced transcriptional activity in response to genistein
was, however, noted in cells transfected to express both receptor subtypes as compared
with cells solely expressing ER-β [64]. Although ER-α is itself unable to mediate
isoflavone signal transduction, it was postulated that the presence of the receptor
subtype may enhance ER-β signalling via the formation of ER-α/β heterodimers. These
observations imply that the precise tissue-specific effects of the soy isoflavones are
dependent on the expression levels and ratios of ER-α and ERβ. The various cofactors
are similarly expressed in a tissue-specific manner, therefore further influencing the
cellular response to dietary phyto-oestrogens.
The stilbene resveratrol is structurally similar to the synthetic oestrogen diethylstilbestrol.
Treatment of breast cancer cell lines with resveratrol represses proliferation in a dose-
dependent manner, inducing G2/M phase cell cycle arrest [39]. Resveratrol exhibits a
relatively weak ER-binding affinity as compared with oestradiol [65]; however, unlike the
soy isoflavones, it is able to bind to both ER-α and ER-β with equal affinity. In cells
transfected to express either ER-α or ER-β resveratrol was found to act as an agonist for
both receptor subtypes, stimulating ERE transcriptional activity through either ER-α or
ER-β alone [21]. Similar agonist activity was observed in MCF-7 breast cancer cells,
which predominantly express the ER-α isoform. Resveratrol induced the dose-
dependent activation of ERE-mediated transcription, also upregulating the expression of
the ER target genes pS2 and PR [65]. Recent studies proposed that the cell cycle
inhibitor protein p21WAF1 is a potential downstream target of resveratrol-induced ER
signalling pathways [66]. The treatment of ER-α-expressing breast cancer cells with
resveratrol resulted in a 23-fold increase in p21WAF1 gene expression, as determined by
cDNA microarray analysis. The resveratrol-mediated induction of p21WAF1 was blocked by
treatment with the pure anti-oestrogen ICI 182,780, confirming p21WAF1 gene regulation
as an ER-mediated event.

Hormone-independent mechanisms
At concentrations in excess of 25 μmol/l, the soy isoflavones are capable of inducing
apoptosis in human breast cancer cells [23,67-69]. ER-negative cell lines retain
sensitivity to the apoptotic effects of soy isoflavones, thereby confirming that apoptosis
occurs in a hormone-independent manner. Apoptosis was effectively induced in the ER-
α-negative MDA-MB-231 breast cancer cell line by genistein and daidzein
concentrations of 50–100 μmol/l [59,60]. In MCF-7 cell cultures the induction of cell
death by treatment with genistein coincided with the increased expression of the
proapoptotic proteins Bax and p53 [67]. Breast cancer cell lines expressing mutant p53
also undergo apoptosis in response to phyto-oestrogen treatment, thereby implying
apoptosis induction by both p53-dependent and p53-independent mechanisms [67,70].
The polyphenol epigallocatechin (EGC) is principally found in green tea and is proposed
to have anticancer properties. Treatment of p53-mutant breast cancer cells with 100
μmol/l EGC induced a 40% increase in apoptosis, correlating with increased Bax
expression and reduced levels of the antiapoptotic protein Bcl-2 [70]. EGC-induced
apoptosis was abolished following treatment with anti-Fas neutralizing antibodies or
caspase inhibitors, suggesting the involvement of Fas signalling pathways. Although
phyto-oestrogen compounds are effective inducers of apoptosis in cell culture models, it
is unlikely that plasma isoflavone concentrations would accumulate to the required levels
for the activation of apoptotic pathways in vivo. It is estimated that plasma phyto-
oestrogen concentrations may reach a maximum of 2–4 μmol/l following the moderate
consumption of soy products [15,52], although it is possible that higher levels may be
present in target tissues. In a recently reported study, equol concentrations within breast
tissue were found to exceed serum levels; however, the reverse was true for genistein
and daidzein [28]. A recent in vitro study [68] revealed the flavone baicalein to be a more
potent inducer of apoptosis than genistein. Baicalein is isolated from the plant
Scutellariae radix and is a common ingredient in herbal tea preparations. A
concentration of 10 μmol/l baicalein induced significant cell death in MCF-7 cell cultures,
suggesting baicalein as a potentially useful pharmacological agent in breast cancer
therapy.
Dietary phyto-oestrogens are capable of inhibiting the proliferation of hormone-
independent breast cell lines [43,54,58,69]. It has been proposed that growth inhibition in
the absence of functional ER occurs via the inhibition of tyrosine kinase activity. The
protein tyrosine kinases are involved in a number of growth factor signalling pathways,
including transforming growth factor (TGF)-α, insulin-like growth factor (IGF)-I, IGF-II and
epidermal growth factor (EGF). In ER-negative breast cancer cultures 5 μmol/l genistein
negated the stimulatory effects of TGF-α, IGF-I and IGF-II, implying the inhibition of
tyrosine kinase activity [23]. The human EGF receptor-2 oncogene (Her-2) is
constitutively overexpressed in approximately 30% of breast cancers and is associated
with a poor patient prognosis [71]. Research using breast cancer cell lines suggests that
dietary phyto-oestrogens are capable of repressing EGF receptor activity. The inhibition
of tyrosine kinase activity by 5 μmol/l genistein in MCF-7 cells correlated with the
repression of EGF receptor tyrosine phosphorylation in response to EGF stimulation
[23]. Similar findings were reported in a recent study investigating the chemoprotective
effects of the green tea polyphenol epigallocatechin-3 gallate (EGCG). The treatment of
Her-2/neu over-expressing mouse mammary cells with 20–80 μg/ml EGCG inhibited
proliferation in a dose-dependent manner, correlating with a reduction in Her-2/neu
signalling activity [72]. The basal tyrosine phosphorylation of Her-2/neu was decreased
by approximately 96% following treatment with 80 μg/ml EGCG. Downstream activities
of the signalling proteins phosphoinositide 3-kinase, Akt and nuclear factor-κB were
similarly repressed, suggesting a potential clinical application of EGCG in breast cancer
therapy.
The soy isoflavones have additionally been proposed to regulate the proliferation of
breast epithelia via an alternative mechanism involving the modulation of TGF-β
synthesis [73]. In normal mammary tissue TGF-β maintains proliferative homeostasis by
inhibiting the growth of epithelial cells [74,75]. The incubation of human mammary
epithelial cells with 5 μmol/l genistein induced a fivefold increase in the level of TGF-β
secretion [76]. The further analysis of media conditioned with human mammary epithelial
cells revealed the presence of the active as opposed to latent form of TGF-β, thus
implying a direct link between soy isoflavones and the TGF-β signalling pathway.

Abbreviations
DMBA = 7,12-dimethylbenz(a)anthracene;
EGC = epigallocatechin;
EGF = epidermal growth factor;
EGCG = epigallocatechin-3 gallate;
ER = oestrogen receptor;
ERE = oestrogen response element;
HRT = hormone replacement therapy;
IGF = insulin-like growth factor;
SRC = steroid receptor coactivator;
TGF = transforming growth factor.

References:
    Messina MJ: Legumes and soybeans: overview of their nutritional profiles and health effects.
    Am J Clin Nutr 1999, Suppl:439S-450S.
    Barnes S: Phytoestrogens and cancer.
    Baillières Clin Endocrinol Metab 1998, 12:559-579.
    Lamartiniere CA: Protection against breast cancer with genistein: a component of soy.
    Am J Clin Nutr 2000, Suppl:1705S-1707S.
    Dai Q, Shu X-O, Jin F, Potter JD, Kushi LH, Teas J, Gao Y-T, Zheng W: Population-based case-
    control study of soyfood intake and breast cancer risk in Shanghai.
    Br J Cancer 2001, 85:372-387.
    Wu AH, Ziegler RG, Nomura AMY, West DW, Kolonel LN, Horn-Ross PL, Hoover RN, Pike MC: Soy
    intake and risk of breast cancer in Asians and Asian Americans.
    Am J Clin Nutr 1998, Suppl:1437S-1443S.
    Adlercreutz H: Phyto-oestrogens and cancer.
    Lancet Oncol 2002, 3:364-373.
Cassidy A, Hanley B, Lamuela-Raventos RM: Isoflavones, lignans and stilbenes – origins,
metabolism and potential importance to human health.
J Sci Food Agric 2000, 80:1044-1062.
Setchell KDR, Radd S: Soy and other legumes: 'bean' around for a long time but are they the
'superfoods' of the millennium and what are the safety issues for their constituent
phytoestrogens?
Asia Pacific J Clin Nutri 2000, Suppl:S13-S22.
Benassayag C, Perrot-Applanat M, Ferre F: Phytoestrogens as modulators of steroid action in
target cells.
J Chromatogr B Analyt Technol Biomed Life Sci 2002, 777:233-248. Le Bail J-C, Champavier Y, Chulia
A-J, Habrioux G: Effects of phytoestrogens on aromatase, 3β and 17β-hydroxysteroid
dehydrogenase activities and human breast cancer cells.
Life Sci 2001, 66:1281-1291. Rosenberg Zand RS, Jenkins DJA, Diamandis EP: Steroid hormone
activity of flavonoids and related compounds.
Breast Cancer Res Treat 2000, 62:35-49.
Tamir S, Eizenberg M, Somjen D, Stern N, Shelach R, Kaye A, Vaya J: Estrogenic and
antiproliferative properties from licorice in human breast cancer cells.
Cancer Res 2000, 60:5704-5709.
Tham DM, Gardner CD, Haskell WL: Potential health benefits of dietary phytoestrogens: a review
of the clinical, epidemiological and mechanistic evidence.
J Clin Endocrinol Metab 1998, 83:2223-2235.
Burow ME, Boue SM, Collins-Burow BM, Melnik LI, Duong BN, Carter-Wientjes CH, Li S, Wiese TE,
Cleveland TE, McLachlan JA: Phytochemical glyceollins, isolated from soy, mediate hormonal
effects through estrogen receptor α and β.
J Clin Endocrinol Metab 2001, 86:1750-1758.
Franke AA, Custer LJ, Tanaka Y: Isoflavones in human breast milk and other biological fluids.
Am J Clin Nutr 1998, Suppl:1466S-1473S.
McMichael-Philips DF, Harding C, Morton M, Roberts SA, Howell A, Potten CS, Bundred NJ: Effects of
soy-protein supplementation on epithelial proliferation in the histologically normal human
breast.
Am J Clin Nutr 1998, Suppl:1431S-1436S.
Hargreaves DF, Potten CS, Harding C, Shaw LE, Morton MS, Roberts SA, Howell A, Bundred NJ: Two-
week dietary soy supplementation has an estrogenic effect on normal pre-menopausal breast.
J Clin Endocrinol Metab 1999, 84:4017-4024.
Lu L-JW, Anderson KE, Grady JJ, Kohen F, Nagamani M: Decreased ovarian hormones during a
soya diet: implications for breast cancer prevention.
Cancer Res 2000, 60:4112-4121.
Maskarinec G, Williams AE, Inouye JS, Stanczyk FZ, Franke AA: A randomised isoflavone
intervention among premenopausal women.
Cancer Epidemiol Biomarkers Prev 2002, 11:195-201.
Morton MS, Arisaka O, Miyake N, Morgan LD, Evans AJ: Phytoestrogen concentrations in serum
from Japanese men and women over forty years of age.
J Nutr 2002, 132:3168-3171.
Bowers JL, Tyulmenkov VV, Jernigan SC, Klinge CM: Resveratrol acts as a mixed
agonist/antagonist for estrogen receptors α and β.
Endocrinology 2000, 141:3657-3667.
Routledge EJ, White R, Parker MG, Sumpter JP: Differential effects of xenoestrogens on
coactivator recruitment by estrogen receptor (ER) α and ERβ.
J Biol Chem 2000, 275:35986-35993.
Fioravanti L, Cappelletti V, Miodini P, Ronchi E, Brivio M, Di Fronzo G: Genistein in the control of
breast cancer cell growth: insights into the mechanism of action in vitro.
Cancer Lett 1998, 130:143-152.
Peterson TG, Ji G-P, Kirk M, Coward L, Falany CN, Barnes S: Metabolism of the isoflavones
genistein and biochanin A in human breast cancer cell lines.
Am J Clin Nutr 1998, Suppl:1505S-1511S.
Doostdar H, Burke MD, Mayer RT: Bioflavonoids: selective substrates and inhibitors for
cytochrome P450 CYP1A and CYP1B1.
Toxicology 2000, 144:31-38.
Potter GA, Patterson LH, Wanogho E, Perry PJ, Butler PC, Ijaz T, Ruparelia KC, Lamb JH, Farmer PB,
Stanley LA, Burke MD: The cancer preventative agent resveratrol is converted to the anti-cancer
agent piceatannol by the cytochrome P450 enzyme CYP1B1.
Br J Cancer 2002, 86:774-778.
McFadyen MCE, Breeman S, Payne S, Stirk C, Miller ID, Melvin WT, Murray GI:
Immunohistochemical localisation of cytochrome P450 CYP1B1 in breast cancer with
monoclonal antibodies specific for CYP1B1.
J Histochem Cytochem 1999, 47:1457-1464.
Maubach J, Bracke ME, Heyerich A, Depypere HT, Serreyn RF, Mareel MM, de Keukeleire D:
Quantitation of soy-derived phytoestrogens in human breast tissue and biological fluids by
high-performance liquid chromatography.
J Chromatogr B Analyt Technol Biomed Life Sci 2003, 784:137-144.
Peeters PHM, Keinen-Boker L, van der Schouw YT, Grobbee DE: Phytoestrogens and breast cancer
risk.
Breast Cancer Res Treat 2003, 77:171-183.
Kumar NB, Cantor A, Allen K, Riccardi D, Cox CE: The specific role of isoflavones on estrogen
metabolism in pre-menopausal women.
Cancer 2002, 94:1166-1174.
Duncan AM, Merz-Demlow BE, Xu X, Phipps WR, Kurzer MS: Premenopausal equol excretors show
plasma hormone profiles associated with lowered risk of breast cancer.
Cancer Epidemiol Biomarkers Prev 2000, 9:581-586.
Grube BJ, Eng ET, Kao Y-C, Kwon A, Chen S: White button mushroom phytochemicals inhibit
aromatase activity and breast cancer cell proliferation.
J Nutri 2001, 131:3288-3293.
Lu L-JW, Cree M, Josyula S, Nagamani M, Grady JJ, Anderson KE: Increased urinary excretion of 2-
hydroxyestrone but not 16α-hydroxyestrone in premenopausal women during a soya diet
containing isoflavones.
Cancer Res 2000, 60:1299-1305.
Key TJ, Sharp GB, Appleby PN, Beral V, Goodman MT, Soda M, Mabuchi K: Soya foods and breast
cancer risk: a prospective study in Hiroshima and Nagasaki, Japan.
Br J Cancer 1999, 81:1248-1256.
Horn-Ross PL, John EM, Lee M, Stewart SL, Koo J, Sakoda LC, Shiau AC, Goldstein J, Davis P,
Perez-Stable EL: Phytoestrogen consumption and breast cancer risk in a multiethnic population.
Am J Epidemiol 2001, 154:434-441.
Shu XO, Jin F, Dai Q, Wen W, Potter JD, Kushi LH, Ruan Z, Gao Y-T, Zheng W: Soyfood intake
during adolescence and subsequent risk of breast cancer among Chinese women.
Cancer Epidemiol Biomark Prev 2001, 10:483-488.
Lamartiniere CA: Timing of exposure and mammary cancer risk.
J Mammary Gland Biol Neoplasia 2002, 7:67-76.
Lamartiniere CA, Zhang J-X, Cotroneo MS: Genistein studies in rats: potential for breast cancer
prevention and reproductive and developmental toxicity.
Am J Clin Nutr 1998, Suppl:1400S-1405S.
Banerjee S, Bueso-Ramos C, Aggarwal BB: Suppression of 7,12-dimethyl(a)anthracene-induced
mammary carcinogenesis in rats by resveratrol: role of nuclear factor-κB, cyclooxygenase 2 and
matrix metalloprotease 9.
Cancer Res 2002, 62:4945-4954.
Jin Z, MacDonald RS: Soy isoflavones increase latency of spontaneous mammary tumours in
mice.
J Nutr 2002, 132:3186-3190.
Bhat KPL, Lantvit D, Christov K, Mehta RG, Moon RC, Pezzuto JM: Estrogenic and antiestrogenic
properties of resveratrol in mammary tumour models.
Cancer Res 2001, 61:7456-7463.
Gallo D, Giacomelli S, Cantelmo F, Zannoni GF, Ferrandina G, Fruscella E, Riva A, Morazzoni P,
Bombardelli E, Mancuso S, Scambia G: Chemoprevention of DMBA-induced mammary cancer in
rats by dietary soy.
Breast Cancer Res Treat 2001, 69:153-164.
Constantinou AI, Krygier AE, Mehta RR: Genistein induces maturation of cultured human breast
cancer cells and prevents tumour growth in nude mice.
Am J Clin Nutr 1998, Suppl:1426S-1430S.
Yan L, Li D, Yee JA: Dietary supplementation with isolated soy protein reduces metastasis of
mammary carcinoma cells in mice.
Clin Exp Metastas 2002, 19:535-540.
Jordan VC: Long-term adjuvant tamoxifen therapy for breast cancer.
Breast Cancer Res Treat 1990, 15:126-136.
Morrow M, Jordan VC: Tamoxifen for the prevention of breast cancer in high-risk women.
Ann Surg Oncol 2000, 7:67-71.
Marsden J: The menopause, hormone replacement therapy and breast cancer.
J Steroid Biochem Mol Biol 2002, 83:123-132.
Chiechi LM: Hormone replacement therapy and health protection.
Curr Opin Investig Drugs 2003, 4:439-443.
Wuttke W, Jarry H, Westphalen S, Christoffel V, Seidlova-Wuttke D: Phytoestrogens for hormone
replacement therapy?
J Steroid Biochem Mol Biol 2002, 83:133-147.
Messina MJ, Loprinzi CL: Soy for breast cancer survivors: a critical review of the literature.
J Nutr 2001, Suppl:3095S-3108S.
This P, De la Rochefordière A, Clough K, Fourquet A, Magdelenat H: Phytoestrogens after breast
cancer.
Endocr Relat Cancer 2001, 8:129-134.
Ju YH, Doerge DR, Allred KF, Allred CD, Helferich WG: Dietary genistein negates the inhibitory
effect of tamoxifen on growth of estrogen-dependent human breast cancer (MCF-7) cells
implanted in athymic mice.
Cancer Res 2002, 62:2474-2477.
Schwartz JA, Liu G, Brooks SC: Genistein-mediated attenuation of tamoxifen-induced antagonism
from estrogen receptor-induced genes.
Biochem Biophy Res Commun 1998, 253:38-43.
Tanos V, Brzezinski A, Drize O, Strauss N, Peretz T: Synergistic inhibitory effects of genistein and
tamoxifen on human dysplastic and malignant epithelial breast cells in vitro.
Eur J Obstet Gynecol Reprod Biol 2002, 102:188-194.
Hall JM, Couse JF, Korach KS: The multifaceted mechanisms of estradiol and estrogen receptor
signaling.
J Biol Chem 2001, 276:36869-36872.
Hsu J-T, Hung H-C, Chen C-J, Hsu W-L, Ying C: Effects of the dietary phytoestrogen biochanin A
on cell growth in the mammary carcinoma cell line MCF-7.
J Nutr Biochem 1999, 10:510-517.
Maggiolini M, Bonofiglio D, Marsico S, Panno ML, Cenni B, Picard D, Ando S: Estrogen receptor α
mediates the proliferative but not the cytotoxic dose-dependent effects of two major
phytoestrogens on human breast cancer cells.
Mol Pharmacol 2001, 60:595-602.
Dampier K, Hudson EA, Howells LM, Manson MM, Walker RA, Gescher A: Differences between
human breast cell lines in susceptibility towards growth inhibition by genistein.
Br J Cancer 2001, 85:618-624.
Upadhyay S, Neburi M, Chinni SR, Alhasan S, Miller F, Sarkar FH: Differential sensitivity of normal
and malignant breast epithelial cells to genistein is partly mediated by p21WAF1.
Clin Cancer Res 2001, 7:1782-1789.
Nomoto S, Arao Y, Horiguchi H, Ikeda K, Kayama F: Oestrogen causes G2/M arrest and apoptosis in
breast cancer MDA-MB-231.
Oncol Rep 2002, 9:773-776.
Morito K, Aomori T, Hirose T, Kinjo J, Hasegawa J, Ogawa S, Inoue S, Muramatsu M, Masamune Y:
Interaction of phytoestrogens with estrogen receptors α and β (II).
An J, Tzagarakis-Foster C, Scharschmidt TC, Lomri N, Leitman DC: Estrogen receptor β-selective
transcriptional activity and recruitment of coregulators by phytoestrogens.
J Biol Chem 2001, 276:17808-17814.
Liu J, Burdette JE, Xu H, Gu C, van Breemen RB, Bhat KPL, Booth A, Constantinou AI, Pezzuto JM,
Fong HHS, Farnsworth NR, Bolton JL: Evaluation of estrogenic activity of plant extracts for the
potential treatment of menopausal symptoms.
J Agric Food Chem 2001, 49:2472-2479.
Petterson K, Delaunay F, Gustafsson J-A: Estrogen receptor β acts as a dominant regulator of
estrogen signaling.
Oncogene 2000, 19:4970-4978.
Gehm BD, McAndrews JM, Chien P-Y, Jameson JL: Resveratrol, a polyphenolic compound found
in grapes and wine, is an agonist for the estrogen receptor.
Proc Natl Acad Sci USA 1997, 94:14138-14143.
Levenson AS, Gehm BD, Pearce ST, Horiguchi J, Simons LA, Ward JE, Jameson JL, Jordan VC:
Resveratrol acts as an estrogen receptor (ER) agonist in breast cancer cells stably transfected
with ER-α.
Int J Cancer 2003, 104:587-596.
Leung LK, Wang TT: Bcl-2 is not reduced in the death of MCF-7 cells at low genistein
concentration.
J Nutri 2000, 130:2922-2926.
Po LS, Chen Z-Y, Tsang DSC, Leung LK: Baicalein and genistein display differential actions on
estrogen receptor (ER) transactivation and apoptosis in MCF-7 cells.
Cancer Lett 2002, 187:33-34.
Pozo-Guisado E, Alvarez-Barrientos A, Mulero-Navarro S, Santiago-Josefat B, Fernandez-Salguero
PM: The antiproliferative activity of resveratrol results in apoptosis in MCF-7 but not in MDA-MB-
231 human breast cancer cells: cell-specific alterations of the cell cycle.
Biochem Pharmacol 2002, 64:1375-1386.
Vergote D, Cren-Olive C, Chopin V, Toillon R-A, Rolando C, Hondermarck H, Le Bourhis X: (-)-
Epigallocatechin (EGC) of green tea induces apoptosis of human breast cancer cells but not of
their normal counterparts.
Breast Cancer Res Treat 2002, 76:195-201.
Masood S, Bui MM: Prognostic and predictive value of HER2/neu oncogene in breast cancer.
Microsc Res Tech 2002, 59:102-108.
Pianetti S, Guo S, Kavanagh KT, Sonenshein GE: Green tea polyphenol epigallocatechin-3 gallate
inhibits Her-2/Neu signalling, proliferation and transformed phenotype of breast cancer cells.
Cancer Res 2002, 62:652-655.
   Kim H, Peterson TG, Barnes S: Mechanisms of action of the soy isoflavone genistein: emerging
   role for its effects via transforming growth factor β signalling pathways.
   Am J Clin Nutr 1998, Suppl:1418S-1425S.
   Sovak MA, Arsura M, Zanieski G, Kavanagh KT, Sonenshein GE: The inhibitory effects of
   transforming growth factor-β1 on breast cancer cell proliferation are mediated through
   regulation of aberrant nuclear factor-κB/rel expression.
   Cell Growth Differ 1999, 10:537-544.
   Knabbe C, Lippman ME, Wakefield LM, Flanders KC, Kasid A, Derynck R, Dickson RB: Evidence that
   transforming growth factor-β is a hormonally regulated negative growth factor in human breast
   cancer cells.
   Cell 1987, 48:417-428.
   Kim H, Xu J, Su Y, Xia H, Li L, Peterson G, Murphy-Ullrich J, Barnes S: Actions of the soy
   phytoestrogen genistein in models of human chronic disease: potential involvement of
   transforming growth factor-β.
   Biochem Soc Trans 2001, 29:216-222.
   Holmberg L, Anderson H: HABITS (hormonal replacement therapy after breast cancer – is it
   safe?), a randomised comparison: trial stopped.


Effects of Pomegranate Chemical Constituents/Intestinal Microbial
    Metabolites on CYP1B1 in 22Rv1 Prostate Cancer Cells

Sashi G. Kasimsetty†, Dobroslawa Bialonska† , Muntha K. Reddy†, Cammi Thornton‡,
Kristine L. Willett‡§ and Daneel Ferreira*†§
†
  Department of Pharmacognosy
‡
  Department of Pharmacology
§
  National Center for Natural Product Research, Research Institute for Pharmaceutical
Sciences
School of Pharmacy, The University of Mississippi, University, Mississippi 38677
  Institute of Environmental Sciences, Jagiellonian University, Gronostajowa 7, 30-387
Krakow, Poland
J. Agric. Food Chem., 2009, 57 (22), pp 10636–10644
DOI: 10.1021/jf902716r
Publication Date (Web): October 26, 2009
Copyright © 2009 American Chemical Society
*To whom correspondence should be addressed. Tel: 662-915-7026. Fax: 662-915-
6975. E-mail: dferreir@olemiss.edu.

Abstract
The cytochrome P450 enzyme, CYP1B1, is an established target in prostate cancer
chemoprevention. Compounds inhibiting CYP1B1 activity are contemplated to exert
beneficial effects at three stages of prostate cancer development, that is, initiation,
progression, and development of drug resistance. Pomegranate ellagitannins/microbial
metabolites were examined for their CYP1B1 inhibitory activity in a recombinant
CYP1B1-mediated ethoxyresorufin-O- deethylase (EROD) assay. Urolithin A, a microbial
metabolite, was the most potent uncompetitive inhibitor of CYP1B1-mediated EROD
activity, exhibiting 2-fold selectivity over CYP1A1, while urolithin B was a noncompetitive
inhibitor with 3-fold selectivity. The punicalins and punicalagins exhibited potent CYP1A1
inhibition with 5−10-fold selectivity over CYP1B1. Urolithins, punicalins, and punicalagins
were tested for their 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced CYP1
inhibitory activity in the 22Rv1 prostate cancer cell line. Urolithins A and B showed a
decrease in their CYP1-mediated EROD inhibitory IC50 values upon increasing their
treatment times from 30 min to 24 h. Urolithin C, 8-O-methylurolithin A, and 8,9-di-O-
methylurolithin C caused a potent CYP1-mediated EROD inhibition in 22Rv1 cells upon
24 h of incubation. Neutral red uptake assay results indicated that urolithin C, 8-O-
methylurolithin A, and 8,9-di-O-methylurolithin C induced profound cytotoxicity in the
proximity of their CYP1 inhibitory IC50 values. Urolithins A and B were studied for their
cellular uptake and inhibition of TCDD-induced CYP1B1 expression. Cellular uptake
experiments demonstrated a 5-fold increase in urolithin uptake by 22Rv1 cells. Western
blots of the CYP1B1 protein indicated that the urolithins interfered with the expression of
CYP1B1 protein. Thus, urolithins were found to display a dual mode mechanism by
decreasing CYP1B1 activity and expression.

Introduction
Dietary intervention to prevent carcinogenesis has been well-established in
epidemiological studies. The consumption of fruits and vegetables is considered to be a
safeguard against various forms of cancers. Polyphenols are the major constituents of
fruit and vegetable diets and are believed to elicit a number of biological properties due
to their antioxidant and anticarcinogenic activities (1). A number of flavonoids such as
quercetin, chrysin, apigenin, and luteolin have been investigated for their cytochrome
P450 1 (CYP1) enzyme inhibition activities, indicating that flavonoid-related
anticarcinogenesis is mediated in part by CYP1 inhibition (2). Ellagic acid, the hydrolysis
product of ellagitannins, exhibits anticarcinogenic effects by inhibition of CYP1A1-
dependent activation of procarcinogens. A number of ellagic acid analogues showed
similar inhibitory activities against CYP1-mediated benzo[a]pyrene activation (3). Ellagic
acid is a major polyphenol in pomegranate juice. Pomegranate juice polyphenols
showed a strong inhibitory activity against estrogen-dependent MCF-7 cell lines. In in
vivo studies, pomegranate juice exhibited 47% inhibition of cancerous lesion formation
induced by the carcinogen, 7,12-dimethylbenz[a]anthracene (4), indicating its potential
use as an adjuvant therapeutic in human breast cancer treatment. Pomegranate juice
components are also believed to exert cancer chemopreventive activity against skin and
colon cancer. Importantly, the consumption of pomegranate juice decreased the clinical
reemergence of prostate cancer-specific antigen in prostate cancer patients after
primary therapy (5, 6). Pomegranate ellagitannins are transformed by human colonic
flora into bioavailable organic molecules called urolithins (7). The pomegranate microbial
metabolites preferentially accumulate in prostate, colon, and intestinal tissues relative to
other organs in a mouse model and exert their beneficial effects to a greater extent in
those tissues (5). Pomegranate constituents inhibited prostate cancer cell growth by
affecting their proliferation, gene expression, invasion, and apoptosis but had a less
profound effect on normal prostate epithelial cells. However, the concentrations at which
they exhibited antiproliferation activity against human prostate cancer cell lines were
higher than the physiologically available concentrations (18.6 μM when 1 L of juice is
consumed for 5 days) (5, 8). These results suggest that there might be some other
pathways through which pomegranate constituents exert cancer chemoprevention. To
explore the other possible prostate cancer chemopreventive pathways, we studied the
effects of pomegranate juice ellagitannins and their microbial metabolites on CYP1B1-
induced carcinogenesis. Previously, it was shown that pomegranate juice consumption
resulted in lowered total hepatic CYP content and also decreased CYP1A2 and CYP3A.
Therefore, the anticarcinogenic effects of pomegranate juice could be partly attributed to
their ability to inhibit CYP activity/expression (9). Our work represents the first report
concerning the effects of pomegranate ellagitannins on CYP1B1 inhibition as a means
for cancer chemoprevention.
CYPs are responsible for the bioactivation of endogenous compounds, drugs, dietary
chemicals, and xenobiotics. The CYP1 isoforms, CYP1A1, CYP1A2, and CYP1B1, are
of major importance because they activate a number of polycyclic aromatic
hydrocarbons (PAHs) to genotoxic compounds leading to tumorogenesis (10). CYP1B1
is abundantly expressed extrahepatically in steroidogenic (ovaries, testes, and adrenal
glands) and steroid-responsive (breast, uterus, and prostate) tissues. The CYP1B1
enzyme plays an important role not only in the initiation and promotion of cancer but also
in the development of drug resistance. The CYP1B1 enzyme alone accounts for
activation of 15 PAHs, six heterocyclic amines, and two nitropolycyclic hydrocarbons into
mutagenic and carcinogenic compounds, which cause DNA damage and initiate cancer
formation. CYP1B1 is also involved in the metabolism of endogenous compounds such
as 17β-estradiol to an active metabolite, 4-hydroxyestradiol (4-OH-E2), which has been
implicated in breast cancer initiation. In comparison, CYP1A1 converts 17β-estradiol into
2-hydroxyestradiol (2-OH-E2), which is relatively noncarcinogenic as compared to 4-OH-
E2 and plays no role in cancer (11). CYP1B1 levels are overexpressed in prostate, lung,
esophageal, oral, and colon cancers but not in the corresponding normal tissues. The
increased expression of CYP1B1 could generate an excessive number of genotoxic
metabolites, which may attack the DNA of normal cells, thus allowing for cancer
promotion. Although the augmented CYP1B1 expression does not cause tumor invasion
or metastasis, it leads to deactivation of anticancer drugs such as flutamide in prostate
cancer treatment and docetaxel in breast cancer treatment (12). Considering the crucial
role played by CYP1B1 in cancer inititation, promotion, and resistance development, it is
an attractive molecular target for cancer chemoprevention. The expression of the CYP1
family is regulated by the aryl hydrocarbon receptor (AhR). The ligands of AhR range
from environmental contaminants to plant- or diet-derived constituents such as curcumin
and carotenoids (13). Because CYP1B1 is a therapeutic target in prostate cancer, we
hypothesized that pomegranate constituents/metabolites might exert prostate cancer
chemoprevention through CYP1B1 inhibition as one of the plausible mechanisms. Our
results indicate a previously unexplored pathway through which pomegranate juice
constituents may contribute to prostate cancer chemoprevention.

Materials and Methods
Isolation and Identification of Pomegranate Juice Ellagitannins
The extraction of ellagitannins was performed by a procedure described previously, by
use of a step gradient consisting of an increasing amount of methanol in water. The
commercial POMx (100 mL) was diluted to 500 mL with Millipore purified water and
successively partitioned with EtOAc (3 × 200 mL) and n-BuOH (3 × 200 mL).
The n-BuOH extract (2.0 g) was concentrated and subjected to Amberlite XAD-16
column chromatography (500 g, 6 cm × 35 cm) and eluted with H2O (2.0 L) and MeOH
(2.0 L) successively. The MeOH fraction on removal of solvent under reduced pressure
afforded a tannin fraction (XAD-n-BuOH) (1.3 g). This was further purified on Sephadex
LH-20 CC (6 cm × 55 cm) and eluted with H2O:MeOH (2:8, 350 mL), H2O:MeOH (1:9,
500 mL), MeOH (450 mL), and MeOH:Me2CO (1:1, 600 mL) to give nine fractions. A
follow-up of fractionation and further purification of all of the fractions on Sephadex LH-
20 column chromatography using H2O:MeOH gradient, MeOH, and MeOH:Me2CO
gradient system afforded the compounds gallic acid, hexahydroxydiphenic acid (HHDP),
gallagic acid, punicalins, and punicalagins. The latter compounds (punicalins and
punicalagins) exist in solution as the α- and β-anomers as well as acyclic
hydroxyaldehyde analogues (14) (Figure 1). The compounds were identified using LC-
MS retention time, UV absorption pattern, molecular mass, and 1H NMR spectra. The
LC-MS system consisted of Waters Micromass ZMTQ mass spectrophotometer, Waters
2695 Separation Module, and Waters 996 Photodiode Array Detector. Mass spectra
were recorded in negative mode, using a capillary voltage of 4000/3500 V and a gas
temperature of 300 °C. The column used was a 150 mm × 3.0 mm i.d., 5 μm, Luna C18
100 Å (Phenomenex, Torrance, CA). The analysis was performed using a 2.5% acetic
acid in water (solvent A) and 2.5% acetic acid in methanol (solvent B), starting from
100% A for 5 min, 0−60% B for 15 min, and 60−100% B for the next 15 min. The flow
rate was 0.3 mL/min with the pressure set at 900−1500 mmHg.




Figure 1. Structures of ellagitannins and urolithins.
Synthesis of Urolithins

Chemicals
Resorcinol, ReagentPlus (99%), 2-bromobenzoic acid (97%), 2-bromo-4,5-
dimethoxybenzoic acid (98%), and chlorobenzene were purchased from Sigma Aldrich
(St. Louis, MO). 2-Bromo-5-methoxybenzoic acid (98%) was purchased from Alfa Aesar
(Ward Hill, MA). Pyrogallol (ACS grade) was purchased from Acros Organics. CuSO4,
NaOH, and AlCl3 were purchased from Fisher Scientific (Pittsburgh, PA).
Purification of Compounds
The high-performance liquid chromatography (HPLC) system consisted of a Waters
Delta 600, a Waters 600 controller, a Waters 996 Photodiode Array Detector, and a 3.0
mm × 150 mm column (Phenomenex, ODS 5 μm C18 100 Å). Analyses were performed
in the gradient system A, 2.5% aqueous acetic acid, and B, 2.5% acetic acid in
methanol, starting from 100% A for 5 min, 0−60% B for 15 min, and 60−100% B for 15
min. The flow rate was 1 mL/min, and the pressure was 600−800 mmHg. The elution of
metabolites was monitored at 254 nm.
Urolithins (urolithin B, 8-O-methylurolithin A, urolithin A, 8,9-di-O-methylurolithin C,
urolithin C, 8,9-di-O-methylurolithin D, and urolithin D) were synthesized by the
condensation of resorcinol or pyrogallol with an appropriately substituted benzoic acid by
the modified protocols described by Ito et al. (15). The structures of urolithins were
confirmed by their molecular mass and comparison of observed and reported 1H NMR
data with reported data (Figure 1).
Recombinant CYP1 Ethoxyresorufin-O-deethylase (EROD) Assay and Inhibition Kinetics
To study the effects of pomegranate chemical constituents and their microbial
metabolites on recombinant CYP1A1 and CYP1B1, a 96-well plate EROD assay was
used (16). Concentrations of the test compounds ranged from 0.5 to 30 μM. Inhibition
kinetics of CYP1B1-mediated EROD activity was determined similarly. Concentrations of
0.5 and 1 μM were used for urolithins A and B in triplicate.
22Rv1 Prostate Cell EROD Assay
To evaluate the effects of pomegranate constituents and microbial metabolites in a cell-
based CYP1 activity, an EROD assay was conducted using 22Rv1 cells in a 48-well
plate format (32). The test compounds were studied for their effects on cell-based
2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced CYP1-mediated EROD activity. The
cells were treated with TCDD for 24 h to induce CYP1 expression. The cells were also
cotreated with compounds, for either 30 min or 24 h, at concentrations ranging from 6.75
to 50 μM. The cells were treated with DMSO, ellagitannins, and urolithins alone to
evaluate their ability to induce CYP1 expression in the absence of TCDD.
Microsome Preparation
Cells were seeded in 150 cm2 culture plates and exposed to various treatments for 24 h.
The cells were harvested, washed, and spun down (250g/5 min/22 °C). An appropriate
amount of the lysis buffer [10 mM Tris, pH 7.5, 10 mM KCl, and 0.5 mM
ethylenediaminetetraacetic acid (EDTA)] was added, and the cells were transferred to
the glass tube of a Teflon homogenizer and kept on ice for 10 min. This was followed by
the addition of an appropriate amount of homogenization buffer [0.25 M KH2PO4, 0.15 M
KCl, 10 mM EDTA, and 0.25 mM phenylmethanesulfonyl fluoride (PMSF)]. The cells
were then broken by 12 manual strokes on a tight-fitting Teflon homogenizer. After
centrifugation at 15000g/20 min/4 °C, the supernatant was again centrifuged at
105000g/90 min/4 °C. Microsomal pellets were resuspended in microsomal dilution
buffer [0.1 M KH2PO4, 20% glycerol, 10 mM EDTA, 0.25 mM PMSF, and 0.1 mM
dithiothreitol (DTT)] and stored in aliquots at −80 °C.

Western Blotting
Microsomal protein (5−10 μg) samples were prepared by heating at 95 °C for 5 min in
the sample buffer comprising 0.5 μM Tris HCl (pH 6.8), 10% glycerol, 2% sodium
dodecyl sulfate (SDS), 5% mercaptoethanol, and 0.001% bromophenol blue. The
samples were resolved by precast criterion SDS-polyacrylamide gel electrophoresis
(PAGE) gel (10%) at 200 V for 45−50 min (Bio-Rad Laboratories, Hercules, CA). The
proteins were then transferred to a PVDF membrane (Bio-Rad Laboratories) at 100 V for
90 min. Following transfer, the membranes were blocked in blocking buffer for 1 h
followed by incubation in CYP1B1 primary antibody (1:1000, antirat CYP1B1 polyclonal
antibody, kindly donated by Dr. Thomas R. Sutter). After they were washed, the
membranes were incubated in buffer containing the horseradish peroxidase-conjugated
secondary antibody (1:30000, antigoat IgG peroxidase conjugate, Sigma Chemicals) for
2 h. The membrane was washed and developed using LumiGLO Reserve
chemiluminescent substrate (KPL, Inc., Gaithersburg, MD). The signals were detected
using a CCD camera (VersaDoc Imaging System, Bio-Rad Laboratories). Human
CYP1B1 supersomes were used to make standard curves of known protein
concentrations (0.25−2 pmol). Standard curves were used to quantitate the CYP1
protein amounts in the samples using the Quantity One quantitation software (Bio-Rad
Laboratories). Statistical differences between the control and the treated samples were
determined using one-way analysis of variance (ANOVA) followed by Newman−Keuls
posthoc (p < 0.05) using GraphPad Prism software.
Neutral Red Cytotoxicity Assay
The assay was performed in 96-well microplates. Cells were seeded at a density of
10000 cells/well and allowed to settle for 30 min at 37 °C. The compounds, diluted
appropriately in RPMI-1640 medium, were added to the cells and again incubated for 48
h. The number of viable cells was determined using the neutral red assay procedure
(17).
Induction of Phase II Conjugating Enzymes Assay
The assays were performed according to standard procedures described by Kirlin et al.
(18)
Cellular Uptake of Urolithins A and B
22Rv1 cells were incubated with 20 μM urolithins A and B in the RPMI-1640 media for
0.5, 6, 12, and 24 h time periods. After incubation, the cells were washed and harvested
by trypsinization. The cells were extracted with acidified MeOH. The samples were
analyzed by reverse phase HPLC at 288 nm detection. The experiment was done in
triplicate. A standard curve of urolithins A and B was prepared from which the amount of
urolithin uptake was determined. The amount of urolithin uptake was adjusted to the
amount of protein.

Results and Discussion
Pomegranate fruit consists of two major classes of polyphenols, flavonoids and
ellagitannins. The flavonoids include quercetin, kaempferol, and myricetin (19). These
flavonoids have been reported to exhibit CYP1 inhibitory activities (16). Pomegranate
juice consumption has been found to decrease the expression/activity of total hepatic
CYP content (9). Pomegranate juice obtained by hydrostatic pressing of whole fruit
predominantly contains ellagitannins. Ellagitannins, with the exception of ellagic acid,
have not been previously studied for their CYP1 inhibitory activities. Ellagic acid has
been shown to inhibit CYP2A2, 3A1, 2C11, 2B1, 2B2, and 2C6 in rat liver microsomes
(20) and also inhibited the CYP1A1-dependent activation of benzo[a]pyrene (BaP) (3).
Inhibition of CYP1 protein expression and activity by some flavonoids, such as diosmin,
diosmetin, quercetin, kaempferol, and myricetin, was believed to be through AhR
antagonism or their effects on the downstream products of AhR signal transduction
pathways (21). However, ellagic acid decreased CYP1A1-dependent BaP activity
independent of the AhR-responsive element (3). The proposed mechanism of inhibition
of CYP1A1-dependent BaP activation involved scavenging of the carcinogen by ellagic
acid through chemical binding (22). Therefore, our objective was to study the effects of a
selection of pomegranate ellagitannins and urolithins on the inhibition of CYP1-
dependent carcinogen activation.
The ability of ellagitannins and urolithins to inhibit CYP1 activity was tested in a
recombinant CYP1A1- and CYP1B1-dependent EROD assay. The IC50 values for
CYP1B1 inhibition ranged from 1.15 ± 0.65 μM for urolithin A to 137 ± 11.08 μM for
urolithin D. CYP1A1 IC50 values ranged from 1.5 ± 0.32 μM for punicalins to 2907 ± 168
μM for urolithin D (Table 1, section A). Urolithins exhibited higher selectivity toward
CYP1B1 EROD inhibition as compared to CYP1A1, although the selectivity was not
significant. The Ki values of CYP1B1 and CYP1A1 depicted in Table 1, section A,
indicate that 8-O-methylurolithin A exhibited a 7.5-fold selectivity toward CYP1B1
inhibition. Punicalins and punicalgins were 10- and 5-fold more selective toward
CYP1A1 inhibition.

Table 1. Results of Recombinant CYP1-Mediated EROD Assay and Kinetic Parametersa
Section A
                   CYP1B1                         CYP1A1
         IC50 ± SEM       Ki ± SEM       IC50 ± SEM       Ki ± SEM Ki(CYP1A1:CYP1B1)
             (μM)           (μM)            (μM)             (μM)
UA     1.15 ± 0.65     0.25 ± 0.14     12.4 ± 4.7      0.51 ± 0.18    2.05
UB     1.55 ± 0.49     0.34 ± 0.17     26.8 ± 12.9     1.11 ± 0.53    3.28
UC     39.9 ± 29       8.74 ± 0.65 790 ± 75            32.7 ± 3.11 3.74
UD     137 ± 11.08     30.10 ± 25      2907 ± 168      120.3 ± 69.6 3.99
MUA 1.49 ± 0.39        0.327 ± 0.08 59.8 ± 8.7         2.47 ± 0.36 7.57
DMUC 89.6 ± 9.7        19.6 ± 2.13     657 ± 74.3      27.2 ± 3.00    1.39
PL     2.82 ± 0.33     0.618 ± 0.07 1.5 ± 0.32         0.062 ± 0.00 0.10
PG     2.6 ± 0.79      0.58 ± 0.02     2.67 ± 0.48     0.109 ± 0.20 0.19
                                Section B
               urolithin A                         urolithin B
      DMSO        0.5 μM      1 μM      DMSO         0.5 μM       1 μM
Vmax 339 ± 148 97.3 ± 14.6 66 ± 6.02 355 ± 103 150.9 ± 14.9 99.5 ± 8.53
Km 3.414 ± 2 2 ± 0.47      1.6 ± 0.25 4.78 ± 1.81 2.10 ± 0.33 1.9 ± 0.26

IC50 and Ki values (±SEM) and the ratio of CYP1A1 to CYP1B1 Ki for urolithins A (UA), B
(UB), C (UC), D (UD), 8-O-methylurolithin A (MUA), 8,9-di-O-methylurolithin C (DMUC),
punicalins (PL), and punicalagins (PG) mediated inhibition of EROD acitivity using
recombinant human CYP1B1 and CYP1A1 enzymes. For section B, kinetic parameters,
Vmax (pmol/mg/min), and Km (μM) ± standard errors (n = 3) for the inhibition of
recombinant human CYP1B1 by urolithin A and B (0.5 and 1 μM), determined by
nonlinear regression curve fit using the Michaelis−Menten equation ([S] vs V) plot using
GraphPad Prism.

Urolithins A and B are the major microbial metabolites of pomegranate chemical
constituents detected in human systemic circulation. These metabolites exhibited lower
IC50 values in recombinant CYP1 inhibition as compared to all other tested compounds.
Therefore, a study was conducted to investigate the mechanism of action of CYP1B1
inhibition by urolithins A and B. The concentrations of inhibitors used were in the vicinity
of their calculated IC50 values, that is, 0.5 and 1 μM. EROD activities were determined
with substrate concentrations ranging from 0.1 to 2.0 μM. Kinetic parameters, Vmax and
Km, were calculated using the Michaelis−Menten equation ([S] vs V curve). Double
reciprocal plots were plotted using 1/[S] and 1/V (Figure 2) from which Ki values were
calculated (Table 1, section B). The calculated Ki values for urolihins A and B (1.51 ±
0.91 and 1.33 ± 0.08 μM) were not statistically different from those calculated by using
Cheng−Prusoff equations (16). The calculated Vmax and Km for urolithin A changed
significantly with an increasing concentration of inhibitor, suggesting an uncompetitive
type of inhibition. However, the Vmax and Km of urolithin B did not differ significantly upon
increase of inhibitor concentration, suggesting a noncompetitive type of inhibition.
Figure 2. Double reciprocal plots for the inhibition of in vitro EROD activity of CYP1B1 by
urolithins A and B at 0.5 and 1 μM. EROD substrate concentrations used were 0.1, 0.2,
0.3, 0.4, 0.6, 1.0, and 2.0 μM. Recombinant CYP1B1 was preincubated with the inhibitor
or DMSO prior to initiation of reaction. Each experiment was done three times in
duplicate.
In our study, urolithins A and B, structural analogues of ellagic acid, exhibited a
significant inhibition of CYP1-dependent EROD activity. The results suggested that a
hydroxy group at C-8 and C-3 (urolithins A and B), corresponding to the C-4 and C-4′
position of ellagic acid, were required for full CYP1-dependent EROD activity inhibition in
accordance with a previous study by Barch et al. (7). However, in our study, additional
hydroxy groups at C-4 and C-9 of the urolithin pharmacophore (urolithins C and D)
resulted in decreased CYP1-dependent EROD inhibitory activity. Methylation of the
hydroxy groups to give 8-O-methylurolithin A and 8,9-di-O-methylurolithin C decreased
the activity, suggesting that the phenolic hydroxy groups are important for CYP1
inhibitory activity. To probe the importance of the lactone group for CYP1-dependent
EROD activity inhibition, HHDP and gallic acid were tested for their CYP1-dependent
EROD activity. The results showed that hydrolysis of the lactone functionality did not
result in CYP1 inhibition (data not shown).
The punicalins and punicalagins were potent inhibitors of CYP1-dependent EROD
activity with Ki values (Table 1, section A) comparable to the dietary flavonoids. The
dietary flavonoids, inhibiting CYP1-dependent metabolic activation of procarcinogens,
include quercetin, kaempferol, apigenin, myricetin, and rutin. Quercetin and kaempferol,
which are the predominant flavonoids in the human diet, inhibited CYP1-mediated
EROD activities. The apparent Ki values of inhibition of human recombinant CYP1B1
and CYP1A1 were 14 ± 3 and 52 ± 2 nM for kaempferol, whereas they were 23 ± 2 and
77 ± 5 nM for quercetin (23). In a different study, quercetin inhibited epoxidation of 7,8-
dihydro-7,8-benzo[a]pyrene-7,8-diol by CYP1A1 allelic variants with Ki values ranging
from 2.0 to 9.3 μM with mixed type inhibition (24). In another study, quercetin inhibited
recombinant CYP1A1 and CYP1B1 activities with Ki values of 0.25 ± 0.04 and 0.12 ±
0.02 μM with a mixed type inhibition (16). The bioavailability of the flavonoids depends
upon the source of food; for example, quercetin absorption from tomato puree, apples,
and onions was 0.082, 0.34, and 0.74 μM, respectively (25, 26). Kaempferol plasma
concentrations ranged from 0.01, 0.05, and 0.1 μM upon consumption of onion, tea, and
endive, respectively (27, 28). Bioavailable concentrations of quercetin and kaempferol
from some food sources were less than their reported Ki values of CYP1 inhibition.
Apigenin, a flavone present in parsley, exhibits CYP1B1 and CYP1A1 inhibition with Ki
values ranging from 60 nM to 0.2 μM. These concentrations are bioavailable (127 ± 81
nM) upon consumption of 2 g of blanched parsley (14). Bioavailability of rutin from
tomato puree as detected in plasma was calculated to be 0.1 μM (25), which was around
60-fold lower than the reported Ki of CYP1B1 inhibition. The bioavailability of many other
flavonoids is still unclear. However, dietary flavonoids could inhibit CYP1-mediated
bioactivation of environmental and dietary carcinogens into genotoxic compounds and
prevent cancer initiation in alimentary canal-related cancers because they come into
direct contact with the digestive epithelium of the digestive system (29). However, the
prostate cancer chemopreventive effects of flavonoids (30) depending on the
bioavailability are still debated. It is therefore important to choose an appropriate dietary
supplement that can release adequate amounts of cancer chemopreventive compounds
into plasma, whenever it is consumed, with an intended pharmacological activity.
Pomegranate juice ellagitannins have been extensively studied for their bioavailability
and their biological effects. In one study, it was established that the punicalagins
hydrolyze into ellagic acid and other smaller polyphenols that are responsible for the
bioactivity of ellagitannins. In a study performed on human subjects, consumption of 180
mL of pomegranate juice (equivalent to 25 mg of ellagic acid and 319 mg of
punicalagins) resulted in detection of ellagic acid in plasma with a maximum
concentration of 31.9 ng/mL (0.1 μM) (31). Previous studies also indicated that the
bioavailability of ellagic acid from pomegranate juice, pomegranate liquid concentrate
extract, and pomegranate powder extract was not statistically different (32). In our study,
punicalins and punicalagins inhibited CYP1A1 with Ki values of 0.062−0.109 μM and
CYP1B1 with Ki values 0.618 and 0.58 μM, respectively. Punicalins and punicalagins
exhibited approximately a 10- and 5-fold selectivity for CYP1A1 over CYP1B1. Thus,
consumption of pomegranate juice could be beneficial in decreasing CYP1-mediated
oral, esophageal, and colon cancers. However, these ellagitannins cannot exhibit a
systemic CYP1 inhibition activity because they are metabolized in the colon by
microflora into smaller organic molecules called urolithins.
Bioavailability studies indicate that maximum plasma concentrations of urolithins A and
B reach concentrations of 4−18 μM in human subjects (8, 32). Urolithins A and B
inhibited human recombinant CYP1A1 and CYP1B1 with Ki values in their bioavailable
concentration range. Our particular interest was to explore the beneficial effects of
urolithins in prostate cancer. Studies indicate that 15% of prostate cancer patients, who
have undergone a radical prostectomy, had a biochemical recurrence of prostate-
specific antigen (PSA). Among them, 34% of patients developed distant metastases
within 15 years (33). It was evident that consumption of pomegranate juice delayed the
doubling time of the PSA by 39 months after primary therapy (7). The effects were
ascribed to the antiproliferative, apoptotic, and antioxidant effects of pomegranate
constituents, observed in LNCaP, PC-3, 22Rv1, and DU 145 prostate cancer cell lines
(8, 34). A study about gene polymorphisms and risk of prostate cancer showed that
polymorphisms in CYP1B1 and PSA genes increased the risk and aggressiveness of
prostate cancer (35). Any dietary constituent with CYP1B1 inhibitory activity could
potentially lead to prostate cancer chemoprevention. There were no previous reports
about the ability of pomegranate constituents/metabolites to inhibit CYP1B1-dependent
carcinogenesis.

Therefore, we studied the capability of pomegranate constituents to inhibit CYP1B1-
induced metabolic activation in a prostate cancer cell line, 22Rv1. The cells were treated
with TCDD for 24 h to induce CYP1B1 protein expression. Then, the cells were treated
with punicalins, punicalagins, or urolithins for 30 min. Following 30 min of incubation,
urolithins A and B significantly decreased TCDD-induced EROD activity at the highest
concentration used (Figure 3A). The IC50 values calculated for cell-based CYP1
inhibition by urolithins A and B were 32 ± 8.9 and 38.2 ± 3.94 μM, respectively (Table 2).
The results indicate a 28- and 26-fold increase in IC50 values for cell-based CYP1-
mediated EROD activity of urolithins A and B, respectively, as compared to their in vitro
recombinant CYP1-mediated EROD inhibitory IC50. Punicalins and punicalagins did not
inhibit cell-based CYP1-mediated EROD activity at the highest concentration used (50
μM). To ascertain whether a decrease in IC50 occurred upon longer incubation, the cells
were allowed to grow in the presence of compounds and TCDD for 24 h (Figure 3B).
The EROD activity results indicated that the compounds more effectively inhibited
CYP1-mediated EROD activity and had IC50 values lower than those following 30 min of
incubation. After 24 h of cotreatment, urolithins A and B inhibited TCDD-induced EROD
activity in prostate cells with IC50 values of 13.3 ± 1.32 and 17.9 ± 1.8 μM, respectively,
which were in the vicinity of their bioavailability (8, 32). Punicalins and punicalagins did
not exhibit EROD inhibition even upon 24 h of incubation. Urolithin C, 8-O-
methylurolithin A, and 8,9-di-O-methyl urolithin C demonstrated IC50 values of 26.8 ± 2.5,
14.8 ± 2.24, and 11.5 ± 2.8 μM, respectively, which were lower than those of urolithins A
and B (Table 2). The compounds alone did not induce EROD activity after 24 h of
incubation. Because the prostate cells were treated with the test compounds for 24 h, it
was imperative to investigate if the compounds exhibited any cytotoxicity. A neutral red
dye uptake assay was used to measure the cytotoxicity of the test compounds. The data
were probit transformed followed by linear regression and IC50 calculation. The cytotoxic
IC50 values ranged from 20.6 ± 4.58 μM for 8,9-di-O-methylurolithin C to 108 ± 3.994 μM
for urolithin B (Table 3). The results indicate that the cytotoxicities of urolithin A and B
had no contribution toward decreased CYP1-mediated EROD activity. The results also
indicate that urolithin C, 8,9-di-O-methylurolithin C, and 8-O-methylurolithin A were false
positives in the prostate cell EROD assay. The activity was not because of CYP1
inhibition but due to cytotoxicity. This conclusion was verified based on four facts: (1)
These compounds inhibited recombinant CYP1-mediated EROD activity at higher
concentrations as compared to urolithins A and B; (2) these compounds did not inhibit
prostate cell EROD activity upon 30 min of incubation; (3) they inhibited prostate cell
EROD activity upon 24 h of treatment, with IC50 values lower than those exhibited in the
recombinant CYP1-mediated EROD assay; and (4) they exhibited cytotoxicity in the
vicinity of their prostate cell EROD inhibition IC50 values.




Figure 3. Effects of increasing concentration (6.75, 12.5, 25, and 50 μM) of urolithin A
(UA), urolithin B (UB), urolithin C (UC), 8-O-methylurolithin A (MUA), 8,9-di-O-
methylurolithin C (DMUC), punicalins (PL), and punicalagins (PG) on 50 nM TCDD-
induced EROD activity in intact 22Rv1 human prostate cancer cells. The cells were
exposed to the compounds for (A) 30 min or (B) 24 h prior to the EROD measurement.
Each experiment was done three times in triplicate. Global one-way (ANOVA) with a
Student−Newman−Keuls posthoc test was used to determine treatment effects. In the
same treatment groups, bars with different letters are statistically different.
Table 2. IC50 Values for Pomegranate Chemical Constituents/Microbial Metabolite
Mediated Inhibition of EROD Activity in TCDD-Induced 22Rv1 Prostate Cancer Cells

                           IC50 ± SEM (μM)
  chemical constituent 30 min         24 h
urolithin A               32 ± 8.9 13.3 ± 1.3
urolithin B               38 ± 3.9 17.9 ± 1.8
urolithin C                 NA 26.89 ± 2.5
8-O-methylurolithin A       NA 14.8 ± 2.2
8,9-di-O-methylurolithin C NA 11.5 ± 2.8
punicalins                  NA NA
punicalagins                NA NA

Table 3. IC50 Values for Urolithin-Mediated Cytotoxicity of 22Rv1 Prostate Cells
 chemical constituents IC50 ± SEM (μM)
urolithin A                98.7 ± 4.2 (a)
urolithin B                108 ± 3.9 (a)
urolithin C                36 ± 3.5 (b)
8-O-methylurolithin A      23.3 ± 3.9 (c)
8,9-di-O-methylurolithin C 20.6 ± 4.6 (c)

Following cell-based EROD assays, cellular uptake experiments were performed for
urolithins A and B. These experiments were performed to determine if the decrease in
IC50 values for CYP1-mediated EROD inhibition in 22Rv1 cells was related to increased
uptake over a 24 h time period. The experiments indicated that there was a 4.5-fold
increase in urolithin uptake upon 24 h of incubation as compared to 30 min of incubation.
The results also indicated that there was a dramatic increase in urolithin uptake between
6 and 12 h, beyond which the uptake was constant. The results suggested that
increased availability of urolithins (from 30 min to 24 h) could have contributed to the
decrease in IC50 of CYP1 inhibition (Table 4). The results also indicated that urolithins
were metabolically stable in 22Rv1 cells for up to 24 h.

Table 4. Uptake of Urolithins A and B by 22Rv1 Cells over a Period of 0.5−24 h
         uptake in μmol/mg protein
time (h) urolithin A urolithin B
0.5      5.2 ± 0.4     5.7 ± 0.8
6        9.6 ± 0.4     8.0 ± 1.1
12       20.1 ± 0.5    21.9 ± 1.1
24       23.9 ± 0.6    25.2 ± 1.6

The decrease in IC50 values could also be attributed to the decrease in the TCDD-
induced CYP1B1 protein expression. To examine if the treatment affected CYP1 protein
expression levels, Western blots were performed (Figure 4). Cell treatments were
DMSO, TCDD (50 nM), UA (50 μM), UB (50 μM), TCDD + UA (50 μM), TCDD + UB (50
μM), TCDD + UA (25 μM), and TCDD + UB (25 μM). CYP1B1 protein levels were
significantly increased ( 4.5-fold) by TCDD (2.8 ± 0.6 pmol/mg) as compared to DMSO
(0.62 ± 0.28 pmol/mg). Cotreatment of TCDD with urolithin A (50 μM) significantly
decreased CYP1B1 protein (1.26 ± 0.38 pmol/mg) production by 54%, while urolithin A
(25 μM) decreased CYP1B1 protein (2.00 ± 0.5 pmol/mg) production by 28% as
compared to TCDD-induced cells. Cotreatment of TCDD with urolithin B (50 μM)
decreased CYP1B1 protein (0.96 ± 0.4 pmol/mg) production by 65%, and urolithin B (25
μM) treatment decreased protein (1.9 ± 0.5 pmol/mg) production by 29% as compared to
TCDD-induced levels. Western blot analyses suggest that none of the urolithins induce
CYP1B1 basal levels significantly as compared to DMSO. However, cotreatments with
urolithins A and B 50 μM and TCDD decreased CYP1B1 protein expression levels as
compared to TCDD alone (Figure 4).




Figure 4.
(A) Western blot showing the effect of urolithins A and B and their cotreatment with
TCDD on CYP1B1 protein expression. Microsomes were prepared after treating cells for
24 h with different treatments (DMSO, 50 nM TCDD, UA 50 μM, UB 50 μM, TCDD + UA
50 μM, TCDD + UA 25 μM, TCDD + UB 50 μM, and TCDD + UB 25 μM), along with
standards, were loaded on 10% SDS-PAGE. Protein levels were expressed in pmol/mg.

(B) Effect of urolithin A and B on the TCDD-induced CYP1B1 protein expression in
22Rv1 prostate cancer cells. Each experiment was repeated three times. Global one-
way ANOVA with a Student−Newman−Keuls posthoc test was used to determine
treatment effects. Bars with different letters are statistically different.

Urolithins A and B inhibited CYP1 EROD activity by inhibiting both the protein
expression and the activity of CYP1B1. While the influence of urolithin C, 8,9-di-O-
methylurolithin C, and 8-O-methylurolithin A on CYP1B1 activity/expression was not
clear, they are believed to exert an antiproliferative activity on prostate cancer cells, in
accordance with previous studies (32, 34). An ideal anticarcinogenic agent would inhibit
phase I enzymes, involved in carcinogen activation while inducing the phase II enzymes,
responsible for the deactivation of carcinogens by assisting their excretion via increased
water solubility. The pomegranate ellagitannins and urolithins were tested for their
capacity to induce glutathione S-transferase and quinone O-reductase enzymes. The
basal levels of quinone O-reductase and glutathione S-transferase enzymes in 22Rv1
cells were determined to be 1.2 ± 0.32 and 0.51 ± 0.17 μmol min−1 mg protein−1,
respectively. However, none of the compounds exhibited (data not shown) induction of
quinone O-reductase or glutathione S-transferase as compared to normal proliferating
cells.
Cancer treatment
Cancer treatment
Cancer treatment
Cancer treatment
Cancer treatment
Cancer treatment
Cancer treatment
Cancer treatment
Cancer treatment
Cancer treatment

Weitere ähnliche Inhalte

Ähnlich wie Cancer treatment

Cancer and anti neoplastic agents
Cancer and anti neoplastic agentsCancer and anti neoplastic agents
Cancer and anti neoplastic agentsvarshitha Nakka
 
IMMUNOSUPREESSIVE_DRUGS_2[1].pptx
IMMUNOSUPREESSIVE_DRUGS_2[1].pptxIMMUNOSUPREESSIVE_DRUGS_2[1].pptx
IMMUNOSUPREESSIVE_DRUGS_2[1].pptxdevijit
 
Anticancer drugs 1 introduction and classification
Anticancer drugs 1 introduction and classificationAnticancer drugs 1 introduction and classification
Anticancer drugs 1 introduction and classificationSubramani Parasuraman
 
Anticancer agents in medicinal chemistry
Anticancer agents in medicinal chemistryAnticancer agents in medicinal chemistry
Anticancer agents in medicinal chemistrymuthulakshmi623285
 
Drug used in skin and mucus membrane
Drug used in skin and mucus membraneDrug used in skin and mucus membrane
Drug used in skin and mucus membraneMr. Dipti sorte
 
Antineoplastic Agents by Gauri Gurawa-1.pptx
Antineoplastic Agents by Gauri Gurawa-1.pptxAntineoplastic Agents by Gauri Gurawa-1.pptx
Antineoplastic Agents by Gauri Gurawa-1.pptxSVP college of pharmacy
 
Drug used in skin and mucus membrane
Drug used in skin and mucus membraneDrug used in skin and mucus membrane
Drug used in skin and mucus membraneAbhay Rajpoot
 
READING ASSIGNMENT.docx
READING ASSIGNMENT.docxREADING ASSIGNMENT.docx
READING ASSIGNMENT.docxMISS DANI
 
Cytotoxic Activity of Linum usitatissimum L. Essential oil against Lung Adeno...
Cytotoxic Activity of Linum usitatissimum L. Essential oil against Lung Adeno...Cytotoxic Activity of Linum usitatissimum L. Essential oil against Lung Adeno...
Cytotoxic Activity of Linum usitatissimum L. Essential oil against Lung Adeno...AmalDhivaharS
 
Anticancer Activity of Some Medicinal Plants A Review
Anticancer Activity of Some Medicinal Plants A ReviewAnticancer Activity of Some Medicinal Plants A Review
Anticancer Activity of Some Medicinal Plants A Reviewijtsrd
 
Principles Of Chemotherapy
Principles Of ChemotherapyPrinciples Of Chemotherapy
Principles Of ChemotherapyFrank Bonilla
 
Apicoplast: an excellent target for antimalarial drug development
Apicoplast: an excellent target for antimalarial drug developmentApicoplast: an excellent target for antimalarial drug development
Apicoplast: an excellent target for antimalarial drug developmentSuman Das
 
Anticancer drugs persentation
Anticancer drugs persentationAnticancer drugs persentation
Anticancer drugs persentationROHIT PAL
 

Ähnlich wie Cancer treatment (20)

Cancer chemotherapy
Cancer  chemotherapyCancer  chemotherapy
Cancer chemotherapy
 
NTCC PPT.pptx
NTCC PPT.pptxNTCC PPT.pptx
NTCC PPT.pptx
 
Cancer and anti neoplastic agents
Cancer and anti neoplastic agentsCancer and anti neoplastic agents
Cancer and anti neoplastic agents
 
IMMUNOSUPREESSIVE_DRUGS_2[1].pptx
IMMUNOSUPREESSIVE_DRUGS_2[1].pptxIMMUNOSUPREESSIVE_DRUGS_2[1].pptx
IMMUNOSUPREESSIVE_DRUGS_2[1].pptx
 
Anticancer drugs 1 introduction and classification
Anticancer drugs 1 introduction and classificationAnticancer drugs 1 introduction and classification
Anticancer drugs 1 introduction and classification
 
Anticancer agents
Anticancer agents Anticancer agents
Anticancer agents
 
Anticancer agents in medicinal chemistry
Anticancer agents in medicinal chemistryAnticancer agents in medicinal chemistry
Anticancer agents in medicinal chemistry
 
Himanshu
HimanshuHimanshu
Himanshu
 
Drug used in skin and mucus membrane
Drug used in skin and mucus membraneDrug used in skin and mucus membrane
Drug used in skin and mucus membrane
 
Anticancer drugs
Anticancer drugsAnticancer drugs
Anticancer drugs
 
Antineoplastic Agents by Gauri Gurawa-1.pptx
Antineoplastic Agents by Gauri Gurawa-1.pptxAntineoplastic Agents by Gauri Gurawa-1.pptx
Antineoplastic Agents by Gauri Gurawa-1.pptx
 
Drug used in skin and mucus membrane
Drug used in skin and mucus membraneDrug used in skin and mucus membrane
Drug used in skin and mucus membrane
 
READING ASSIGNMENT.docx
READING ASSIGNMENT.docxREADING ASSIGNMENT.docx
READING ASSIGNMENT.docx
 
Introduction to chemotherapy
Introduction to chemotherapyIntroduction to chemotherapy
Introduction to chemotherapy
 
Cytotoxic Activity of Linum usitatissimum L. Essential oil against Lung Adeno...
Cytotoxic Activity of Linum usitatissimum L. Essential oil against Lung Adeno...Cytotoxic Activity of Linum usitatissimum L. Essential oil against Lung Adeno...
Cytotoxic Activity of Linum usitatissimum L. Essential oil against Lung Adeno...
 
Anticancer Activity of Some Medicinal Plants A Review
Anticancer Activity of Some Medicinal Plants A ReviewAnticancer Activity of Some Medicinal Plants A Review
Anticancer Activity of Some Medicinal Plants A Review
 
Principles Of Chemotherapy
Principles Of ChemotherapyPrinciples Of Chemotherapy
Principles Of Chemotherapy
 
Apicoplast: an excellent target for antimalarial drug development
Apicoplast: an excellent target for antimalarial drug developmentApicoplast: an excellent target for antimalarial drug development
Apicoplast: an excellent target for antimalarial drug development
 
ANTICANCER NEW.pptx
ANTICANCER  NEW.pptxANTICANCER  NEW.pptx
ANTICANCER NEW.pptx
 
Anticancer drugs persentation
Anticancer drugs persentationAnticancer drugs persentation
Anticancer drugs persentation
 

Mehr von DVS BioLife Ltd

RELATION BETWEEN EARTH, MOON, MARS AND A GIANT IMPACT
RELATION BETWEEN EARTH, MOON, MARS AND A GIANT IMPACT RELATION BETWEEN EARTH, MOON, MARS AND A GIANT IMPACT
RELATION BETWEEN EARTH, MOON, MARS AND A GIANT IMPACT DVS BioLife Ltd
 
A review on the Role of Beneficial microorganisms and Prebiotics in Human Nut...
A review on the Role of Beneficial microorganisms and Prebiotics in Human Nut...A review on the Role of Beneficial microorganisms and Prebiotics in Human Nut...
A review on the Role of Beneficial microorganisms and Prebiotics in Human Nut...DVS BioLife Ltd
 
Effluent treatment employing Beneficial Algae
Effluent treatment employing Beneficial Algae Effluent treatment employing Beneficial Algae
Effluent treatment employing Beneficial Algae DVS BioLife Ltd
 
IMPROVING BUFFALO MILK FOR FEEDING NEONATALS AS A REPLACEMENT OF MOTHER'S MILK
IMPROVING BUFFALO MILK FOR FEEDING NEONATALS AS A REPLACEMENT OF MOTHER'S MILKIMPROVING BUFFALO MILK FOR FEEDING NEONATALS AS A REPLACEMENT OF MOTHER'S MILK
IMPROVING BUFFALO MILK FOR FEEDING NEONATALS AS A REPLACEMENT OF MOTHER'S MILKDVS BioLife Ltd
 
Protease for use in poultry
Protease for use in poultryProtease for use in poultry
Protease for use in poultryDVS BioLife Ltd
 
Littertreat litter2 feed
Littertreat litter2 feedLittertreat litter2 feed
Littertreat litter2 feedDVS BioLife Ltd
 
TREATING Cattle dung for use as manure or poultry feeding stuff
TREATING Cattle dung for use as manure or poultry feeding stuffTREATING Cattle dung for use as manure or poultry feeding stuff
TREATING Cattle dung for use as manure or poultry feeding stuffDVS BioLife Ltd
 
How to Make Plants tolerate Frost
How to Make Plants tolerate FrostHow to Make Plants tolerate Frost
How to Make Plants tolerate FrostDVS BioLife Ltd
 

Mehr von DVS BioLife Ltd (19)

Msw 3650-ppt-1 kv
Msw 3650-ppt-1 kvMsw 3650-ppt-1 kv
Msw 3650-ppt-1 kv
 
RELATION BETWEEN EARTH, MOON, MARS AND A GIANT IMPACT
RELATION BETWEEN EARTH, MOON, MARS AND A GIANT IMPACT RELATION BETWEEN EARTH, MOON, MARS AND A GIANT IMPACT
RELATION BETWEEN EARTH, MOON, MARS AND A GIANT IMPACT
 
Colion p
Colion pColion p
Colion p
 
A review on the Role of Beneficial microorganisms and Prebiotics in Human Nut...
A review on the Role of Beneficial microorganisms and Prebiotics in Human Nut...A review on the Role of Beneficial microorganisms and Prebiotics in Human Nut...
A review on the Role of Beneficial microorganisms and Prebiotics in Human Nut...
 
Human probiotics
Human probioticsHuman probiotics
Human probiotics
 
Somalatha
SomalathaSomalatha
Somalatha
 
Biopreserve meat
Biopreserve meatBiopreserve meat
Biopreserve meat
 
Effluent treatment employing Beneficial Algae
Effluent treatment employing Beneficial Algae Effluent treatment employing Beneficial Algae
Effluent treatment employing Beneficial Algae
 
Profiles coccicare
Profiles coccicareProfiles coccicare
Profiles coccicare
 
IMPROVING BUFFALO MILK FOR FEEDING NEONATALS AS A REPLACEMENT OF MOTHER'S MILK
IMPROVING BUFFALO MILK FOR FEEDING NEONATALS AS A REPLACEMENT OF MOTHER'S MILKIMPROVING BUFFALO MILK FOR FEEDING NEONATALS AS A REPLACEMENT OF MOTHER'S MILK
IMPROVING BUFFALO MILK FOR FEEDING NEONATALS AS A REPLACEMENT OF MOTHER'S MILK
 
Protease for use in poultry
Protease for use in poultryProtease for use in poultry
Protease for use in poultry
 
Profiles enzymix
Profiles enzymixProfiles enzymix
Profiles enzymix
 
Littertreat litter2 feed
Littertreat litter2 feedLittertreat litter2 feed
Littertreat litter2 feed
 
Ovofert
OvofertOvofert
Ovofert
 
TREATING Cattle dung for use as manure or poultry feeding stuff
TREATING Cattle dung for use as manure or poultry feeding stuffTREATING Cattle dung for use as manure or poultry feeding stuff
TREATING Cattle dung for use as manure or poultry feeding stuff
 
Mastitiscare
MastitiscareMastitiscare
Mastitiscare
 
2014 demand probiotics
2014 demand probiotics2014 demand probiotics
2014 demand probiotics
 
How to Make Plants tolerate Frost
How to Make Plants tolerate FrostHow to Make Plants tolerate Frost
How to Make Plants tolerate Frost
 
Profiles saltex revised
Profiles saltex revisedProfiles saltex revised
Profiles saltex revised
 

Kürzlich hochgeladen

Top Rated Bangalore Call Girls Richmond Circle ⟟ 8250192130 ⟟ Call Me For Gen...
Top Rated Bangalore Call Girls Richmond Circle ⟟ 8250192130 ⟟ Call Me For Gen...Top Rated Bangalore Call Girls Richmond Circle ⟟ 8250192130 ⟟ Call Me For Gen...
Top Rated Bangalore Call Girls Richmond Circle ⟟ 8250192130 ⟟ Call Me For Gen...narwatsonia7
 
Lucknow Call girls - 8800925952 - 24x7 service with hotel room
Lucknow Call girls - 8800925952 - 24x7 service with hotel roomLucknow Call girls - 8800925952 - 24x7 service with hotel room
Lucknow Call girls - 8800925952 - 24x7 service with hotel roomdiscovermytutordmt
 
Night 7k to 12k Navi Mumbai Call Girl Photo 👉 BOOK NOW 9833363713 👈 ♀️ night ...
Night 7k to 12k Navi Mumbai Call Girl Photo 👉 BOOK NOW 9833363713 👈 ♀️ night ...Night 7k to 12k Navi Mumbai Call Girl Photo 👉 BOOK NOW 9833363713 👈 ♀️ night ...
Night 7k to 12k Navi Mumbai Call Girl Photo 👉 BOOK NOW 9833363713 👈 ♀️ night ...aartirawatdelhi
 
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...Taniya Sharma
 
Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...
Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...
Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...Call Girls in Nagpur High Profile
 
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore EscortsCall Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escortsvidya singh
 
Call Girls Bangalore Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Bangalore Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Bangalore Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Bangalore Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Siliguri Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Siliguri Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Siliguri Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Siliguri Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Bangalore Call Girls Nelamangala Number 7001035870 Meetin With Bangalore Esc...
Bangalore Call Girls Nelamangala Number 7001035870  Meetin With Bangalore Esc...Bangalore Call Girls Nelamangala Number 7001035870  Meetin With Bangalore Esc...
Bangalore Call Girls Nelamangala Number 7001035870 Meetin With Bangalore Esc...narwatsonia7
 
VIP Hyderabad Call Girls Bahadurpally 7877925207 ₹5000 To 25K With AC Room 💚😋
VIP Hyderabad Call Girls Bahadurpally 7877925207 ₹5000 To 25K With AC Room 💚😋VIP Hyderabad Call Girls Bahadurpally 7877925207 ₹5000 To 25K With AC Room 💚😋
VIP Hyderabad Call Girls Bahadurpally 7877925207 ₹5000 To 25K With AC Room 💚😋TANUJA PANDEY
 
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Bareilly Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Bareilly Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Bareilly Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Bareilly Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Bangalore Call Girl Whatsapp Number 100% Complete Your Sexual Needs
Bangalore Call Girl Whatsapp Number 100% Complete Your Sexual NeedsBangalore Call Girl Whatsapp Number 100% Complete Your Sexual Needs
Bangalore Call Girl Whatsapp Number 100% Complete Your Sexual NeedsGfnyt
 
Call Girls Varanasi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Varanasi Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Varanasi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Varanasi Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Chandrapur Call girls 8617370543 Provides all area service COD available
Chandrapur Call girls 8617370543 Provides all area service COD availableChandrapur Call girls 8617370543 Provides all area service COD available
Chandrapur Call girls 8617370543 Provides all area service COD availableDipal Arora
 
(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...
(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...
(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...indiancallgirl4rent
 
VIP Call Girls Indore Kirti 💚😋 9256729539 🚀 Indore Escorts
VIP Call Girls Indore Kirti 💚😋  9256729539 🚀 Indore EscortsVIP Call Girls Indore Kirti 💚😋  9256729539 🚀 Indore Escorts
VIP Call Girls Indore Kirti 💚😋 9256729539 🚀 Indore Escortsaditipandeya
 
Call Girls Coimbatore Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Coimbatore Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Coimbatore Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Coimbatore Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...
Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...
Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...CALL GIRLS
 

Kürzlich hochgeladen (20)

Top Rated Bangalore Call Girls Richmond Circle ⟟ 8250192130 ⟟ Call Me For Gen...
Top Rated Bangalore Call Girls Richmond Circle ⟟ 8250192130 ⟟ Call Me For Gen...Top Rated Bangalore Call Girls Richmond Circle ⟟ 8250192130 ⟟ Call Me For Gen...
Top Rated Bangalore Call Girls Richmond Circle ⟟ 8250192130 ⟟ Call Me For Gen...
 
Lucknow Call girls - 8800925952 - 24x7 service with hotel room
Lucknow Call girls - 8800925952 - 24x7 service with hotel roomLucknow Call girls - 8800925952 - 24x7 service with hotel room
Lucknow Call girls - 8800925952 - 24x7 service with hotel room
 
Night 7k to 12k Navi Mumbai Call Girl Photo 👉 BOOK NOW 9833363713 👈 ♀️ night ...
Night 7k to 12k Navi Mumbai Call Girl Photo 👉 BOOK NOW 9833363713 👈 ♀️ night ...Night 7k to 12k Navi Mumbai Call Girl Photo 👉 BOOK NOW 9833363713 👈 ♀️ night ...
Night 7k to 12k Navi Mumbai Call Girl Photo 👉 BOOK NOW 9833363713 👈 ♀️ night ...
 
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
 
Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...
Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...
Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...
 
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore EscortsCall Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
 
Call Girls Bangalore Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Bangalore Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Bangalore Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Bangalore Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girls Siliguri Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Siliguri Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Siliguri Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Siliguri Just Call 9907093804 Top Class Call Girl Service Available
 
Bangalore Call Girls Nelamangala Number 7001035870 Meetin With Bangalore Esc...
Bangalore Call Girls Nelamangala Number 7001035870  Meetin With Bangalore Esc...Bangalore Call Girls Nelamangala Number 7001035870  Meetin With Bangalore Esc...
Bangalore Call Girls Nelamangala Number 7001035870 Meetin With Bangalore Esc...
 
VIP Hyderabad Call Girls Bahadurpally 7877925207 ₹5000 To 25K With AC Room 💚😋
VIP Hyderabad Call Girls Bahadurpally 7877925207 ₹5000 To 25K With AC Room 💚😋VIP Hyderabad Call Girls Bahadurpally 7877925207 ₹5000 To 25K With AC Room 💚😋
VIP Hyderabad Call Girls Bahadurpally 7877925207 ₹5000 To 25K With AC Room 💚😋
 
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girls Bareilly Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Bareilly Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Bareilly Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Bareilly Just Call 9907093804 Top Class Call Girl Service Available
 
Bangalore Call Girl Whatsapp Number 100% Complete Your Sexual Needs
Bangalore Call Girl Whatsapp Number 100% Complete Your Sexual NeedsBangalore Call Girl Whatsapp Number 100% Complete Your Sexual Needs
Bangalore Call Girl Whatsapp Number 100% Complete Your Sexual Needs
 
Call Girls Varanasi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Varanasi Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Varanasi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Varanasi Just Call 9907093804 Top Class Call Girl Service Available
 
Chandrapur Call girls 8617370543 Provides all area service COD available
Chandrapur Call girls 8617370543 Provides all area service COD availableChandrapur Call girls 8617370543 Provides all area service COD available
Chandrapur Call girls 8617370543 Provides all area service COD available
 
(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...
(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...
(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...
 
VIP Call Girls Indore Kirti 💚😋 9256729539 🚀 Indore Escorts
VIP Call Girls Indore Kirti 💚😋  9256729539 🚀 Indore EscortsVIP Call Girls Indore Kirti 💚😋  9256729539 🚀 Indore Escorts
VIP Call Girls Indore Kirti 💚😋 9256729539 🚀 Indore Escorts
 
Call Girls Coimbatore Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Coimbatore Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Coimbatore Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Coimbatore Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...
Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...
Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...
 

Cancer treatment

  • 1. CANCER TREATMENT _CHEMOTHERAPY_ALTERNATIVES Compiled and Edited By G Vijaya Raghaqvan, CEO, DVS BioLife Ltd, HYDERABAD. CONTENTS: 1. INTRODUCTION 2. CHEMOTHERAPY 3. COMMON CANCER DRUGS 4. ROLE OF UNUSUAL PLANT DERIVATIVES INCLUDING ALKALOIDS FROM POMEGRANATE 5. ROLE OF MICROBES LIKE E COLI IN DIAGNOSIS AND IN TREATMENT 6. COMPLEMENTARY APPROACHES 1. INTRODUCTION: In the year 2000, malignant tumours were responsible for 12 per cent of the nearly 56 million deaths worldwide from all causes. In many countries, more than a quarter of deaths are attributable to cancer. In 2000, 5.3 million men and 4.7 million women developed a malignant tumour and altogether 6.2 million died from the disease. The report also reveals that cancer has emerged as a major public health problem in developing countries, matching its effect in industrialized nations. Every year about 85,0000 new cancer cases are diagnosed in India resulting in about 58,0000 cancer related death every year. Bladder Cancer, Melanoma, Breast Cancer, Non-Hodgkin Lymphoma, Colon and Rectal Cancer, Pancreatic Cancer, Endometrial Cancer, Prostate Cancer, Kidney (Renal Cell) Cancer, Skin Cancer (Nonmelanoma), Leukemia, Thyroid Cancer and Lung Cancer are the common types of Cancer found worldwide. Cancer management practices involve in screening, diagnostics, surgery, chemotherapy, radiation, alternative and complimentary therapies. Cancer management is partly based on weighing risk factors attributed to noninfectious agents, human genes and epigenetic factors. Infectious disease causation has largely been restricted to genes directly responsible for causing cancer after sustaining damage i.e. oncogenes. Lately, evidence has emerged linking infectious agents to a number of chronic diseases. These studies have recognized the influence that acute, atypical, latent and chronic infections may play in tricking the immune system and affecting disease etiology. Similar evidence is emerging in model systems with respect to the role of infectious agents in gastrointestinal, liver and lung cancers. Although viruses have been found in association with breast cancer, skepticism remains about a role for other infectious agents, notably microbes in the disease etiology. Improved experimental designs employed in different cancer studies and a less rigid definition of infectious causation may aid in confirming or refuting a microbe-breast cancer connection. Cancer recurrence could potentially be minimized and treatment options further tailored on a case by case basis if microbes/microbial components/strain variants associated with
  • 2. breast cancer are identified; probiotics are employed to reduce treatment side-effects and if microbes could effectively be harnessed in immunotherapy. 2. Chemotherapy Chemotherapy is a cancer treatment that uses drugs to destroy cancer cells. Chemotherapy can be used to: • Destroy cancer cells • Stop cancer cells from spreading • Slow the growth of cancer cells Chemotherapy can be given alone or with other treatments. It can help other treatments work better. Chemotherapy can be given in these forms: • An IV (intravenously) • A shot (injection) into a muscle or other part of the body • A pill or a liquid that can be swallowed • A cream that is rubbed on the skin • Other ways: Chemotherapy is achieved by one or more of the following. • Alkylating Agents • Antimetabolites • Cytotoxic Agents • Plant Derivatives • Microbial The following are the common drugs used in Cancer. 1. Acarbose 2. Acivicin 3. Aclarubicin 4. Acodazole 5. Acronine 6. Actinomycin D 7. Adenine Phosphate 8. Adenosine 9. Adozelesin 10. Adriamycin 11. Adrucil 12. Alanosine 13. Aldesleukin 14. Alemtuzumab 15. Alestramustine 16. Alfacalcidol 17. Alitretinoin
  • 3. 18. Alosetron Hcl 19. Alprostadil 20. Altretamine 21. Ambamustine 22. Ametantrone 23. Amifostine 24. Aminoglutethimide 25. Aminopterin Sodium 26. Amonafide 27. Amphotericin B 28. Amrubicin 29. Amsacrine Hcl 30. Amygdalin 31. Anastrozole 32. Anaxirone 33. Ancitabine 34. Angoroside C 35. Apixaban 36. Arecoline hydrobromide 37. Argatroban Anhydrous 38. Arsenic Trioxide 39. Ascomycin 40. Asparaginase 41. Atevirdine 42. Atosiban 43. Atrimustine 44. Axitinib 45. 5-Azacytidine 46. Azacitidine 47. Azasetron Hcl 48. Azatepa 49. Azathioprine 50. Azetepa 51. Batimastat 52. Bavituximab 53. Benaxibine 54. Bendamustine Hcl 55. Benexate 56. Benzodepa 57. Betamerphalan 58. Betulinic acid 59. Bevacizumab 60. Bicalutamide 61. Bisantrene 62. Bisnafide 63. Bizelesin
  • 4. 64. Bleomycin 65. Bofumustine 66. Bortezomib 67. Bosutinib 68. Brefeldin A 69. Brequinar 70. Bromacrylide 71. Bromocriptine 72. Bropirimine 73. Broxuridine 74. Budotitane 75. Busulfan 76. Calcifediol 77. Calcipotriol 78. Calcitriol 79. Calcium Folinate 80. Calcium Levofolinate 81. Campotothecine 82. Canertinib 83. Cantharidin 84. Capecitabine 85. Caracemide 86. Carbetimer 87. Carboplatin 88. Carboquone 89. Carmofur 90. carmustine 91. Carzelesin 92. Catharanthine Tartrate 93. Celastrol 94. Cemadotin 95. Cephalotaxin 96. Cetuximab 97. Chlorambucil 98. Chlorethylaminouracil 99. Chlormethine 100. Chlornaphazine 101. Cisplatin 102. Cladribine 103. Cladribine 104. Clanfenur 105. Clarithromycin 106. Clevudine 107. Clodronate Disodium 108. Clofarabine 109. Clomifene Citrate
  • 5. 110. Colchiceinamide 111. Colchicine 112. Cordycepin 113. Curcumin 114. Cyclocytidine 115. Cyclophosphamide 116. Cyclosprin 117. Cytarabine 118. Cytidine 119. Dacarbazine 120. Dactinomycin 121. Daniquidone 122. Dapoxetine Hcl 123. Dasatinib 124. Datelliptium Chloride 125. Daunoblastin 126. Daunorubicin 127. D-Bicuculline 128. Decarbazine 129. Decitabine 130. Deferasirox 131. Deflazacort 132. Defosfamide 133. Demecolcine 134. Desonide 135. Desoximetasone 136. Desoxycorticosterone 137. Desoxycortone 138. Dexamethasone 139. Dexasone 140. Dexniguldipine 141. Dexormaplatin 142. Dexrazoxane 143. Dezaguanine 144. Diacetoxysciroenol 145. Dianhydrodulcitol 146. Dianhydrodulcitolum 147. Diaziquone 148. Dibrospidium Chloride 149. Dinaline 150. Dirithromycin 151. Ditercalinium Chloride 152. Ditiomustine 153. Docetaxel 154. Dolasetron mesylate 155. Dopan
  • 6. 156. Doxifluridine 157. Doxorubicin 158. D-tetrahydropalmatine 159. Ecomustine 160. Edatrexate 161. Edelfosine 162. Eflornithine 163. Egenine 164. Elinafide 165. Elliptinium Acetate 166. Elmustine 167. Elsamitrucin 168. Emitefur 169. Enloplatin 170. Enocitabine 171. Enpromate 172. Entecavir 173. Entricitabine 174. Enzastaurin 175. Epipropidine 176. Epirubicin 177. Eptaplatin 178. Erlotinib 179. Esorubicin 180. Estramustine 181. Ethoglucid 182. 7-Ethyl-10-Hydroxycamptothecin Etoglucid 183. Etoposide 184. Exemestane 185. Exenatide Acetate 186. Ezetimibe 187. Fadrozole 188. Fazarabine 189. Fenclonine 190. Fenretinide 191. Filgrastim 192. Finasteride 193. Fingolimod HCL(FTY720) 194. Floxuridine 195. Fludarabine 196. Flumazenil 197. Flumetasone 198. Fluoracil 199. 5-Fluorouracil 200. Fluorouracil 201. Fluracil
  • 7. 202. Flurocitabine 203. Flutamide 204. Folic Acid 205. Formestane 206. Formylmerphalan 207. Fosquidone 208. Fotemustine 209. Fotretamine 210. Ftorafur 211. Fulvestrant 212. Galamustine 213. Galanthamine HBr 214. Galocitabine 215. Gefitinib 216. Gemcitabine 217. Gimeracil 218. Giracodazole 219. Glyfosfin 220. Goserelin 221. Granisetron Hcl 222. Harpagoside 223. Heparin sodium 224. Hexarelin 225. Homoharringtonine 226. Hydrocamptothecine 227. 10-Hydroxycamptothecin 228. Hydroxycamptothecin 229. Hydroxycarbamide 230. Hydroxyurea 231. Ibacitabine 232. Ibandronate Sodium 233. Ibandronic Acid 234. Idarubicin Hcl 235. Idoxifene 236. Ifosfamide 237. Ilmofosine 238. Ilomastat 239. Imatinib 240. Improsulfan 241. Indirubin 242. Intoplicine 243. Iproplatin 244. Irinotecan 245. Ivabradine HCL 246. Ixabepilone 247. Ketotrexate
  • 8. 248. Lanreotide 249. Lapatinib 250. Latanoprost 251. L-biopterin 252. Lenalidomide 253. Lentinan 254. Letrozole 255. Leucovorin calcium 256. Leuprolide Acetate 257. Leuprorelin 258. Leurubicin 259. Leustatin 260. Levothyroxine 261. Lobaplatin 262. Lofexidine HCL 263. Loganin 264. Lometrexol 265. Lomustine 266. Lonidamine 267. Losoxantrone 268. Lupeol 269. Lurtotecan 270. Lycopene 271. Lysipressin 272. Mafosfamide 273. Mannomustine 274. Mannosulfan 275. Maraviroc 276. Marimastat 277. Masoprocol 278. Mechlorethamine 279. Mechlorethaminoxide 280. Medrysone 281. Megastrol acetate 282. Melphalan 283. Menogaril 284. Mepitiostane 285. Meprednisone 286. 6-Mercaptopurine 287. Mercaptopurine 288. Mesna 289. Metamelfalan 290. Methotrexate 291. Methoxymerphalan 292. Methylprednisolone 293. Methylprednisolone Aceponate
  • 9. 294. Methylprednisolone Suleptanate 295. Meturedepa 296. Metyrosine 297. Miboplatin 298. Miltefosine 299. Minamestane 300. Miproxifene 301. Mitindomide 302. Mitobronitol 303. Mitoclomine 304. Mitoflaxone 305. Mitoguazone 306. Mitolactol 307. Mitomycin 308. Mitonafide 309. Mitopodozide 310. Mitoquidone 311. Mitosper 312. Mitotane 313. Mitotenamine 314. Mitoxantrone 315. Mitozolomide 316. Mizoribine 317. Mofarotene 318. Monocrotaline 319. Mubritinib 320. Mustine 321. Mycophenolate Mofetil 322. Mycophenolic Acid 323. Myricetin 324. Myricitrin 325. Nebivolol HCL 326. Nedaplatin 327. Nemorubicin 328. Neptamustine 329. Nigericin 330. Nilotinib 331. Nilutamide 332. Nimustine 333. Nitrocaphane 334. Nocodazole 335. Nolatrexed 2HCL 336. Norcantharidin 337. Ocaphane 338. Octreotide 339. Ondansetron
  • 10. 340. Ormaplatin 341. Oseltamivir Phosphate 342. Oteracil potassium 343. Oxaliplatin 344. Oxipurinol 345. Oxylycorium Acetate 346. Paclitaxel 347. Paliperidone 348. palonosetron 349. Pamidronate Disodium 350. Pamidronic Acid 351. Panitimumab 352. Pazelliptine 353. Pegaspargase 354. Pemetrexed 355. Pentamustine 356. Pentetreotide 357. Pentostatin 358. Perfosfamide 359. Phenoxybenzamine HCl 360. Pincristine 361. Pipobroman 362. Piposulfan 363. Pirarubicin 364. Pirazofurin 365. Piritrexim 366. Pirlimycin 367. Piroxantrone 368. Plomestane 369. Plusonermin 370. Podophyllotoxin 371. Polymyxin E 372. Prednimustine 373. Prednisolone 374. Procarbazine 375. Procodazole 376. Prospidium Chloride 377. Psoralen 378. Pteropterin 379. Pumitepa 380. Ractopamine HCL 381. Raloxifene Hydrochloride 382. Raltegravir 383. Raltitrexed 384. Ramosetron Hcl 385. Ranimustine
  • 11. 386. Rapamycin 387. Razoxane 388. Resveratrol 389. Retigabine 390. Riboprine 391. Risedronic acid 392. Ristocetin A 393. Ritrosulfan 394. Rituximab 395. Rocuronium Bromide 396. Roflumilast 397. Rogletimide 398. Ropinirole HCL 399. Rosuvastatin Calcium 400. Rubitecan 401. Sarcolysin 402. Sargramostim 403. Sebriplatin 404. Semustine 405. Sermorelin 406. Simtrazene 407. Sitagliptin Phosphate 408. Sizofiran 409. Sobuzoxane 410. Sodium Borocaptate[10B] 411. Solaziquone 412. Sonermin 413. Sorafenib 414. Spiclomazine 415. Spirogermanium 416. Spiromustine Following are some excerpts from public domain. Conventional Alkylating Agents Alkylating agents are one of the earliest and most commonly used chemotherapy agents used for cancer treatments. Their use in cancer treatments started in early 1940s. Majority of alkaline agents are active or dormant nitrogen mustards, which are poisonous compound initially used for certain military purposes. Chlorambucil, Cyclophosphamide, CCNU, Melphalan, Procarbazine, Thiotepa, BCNU, and Busulfan are some of the commonly used alkylating agents. They are more effective in treating slow-growing cancers such as solid tumors and leukemia Traditional Antimetabolites Structure of antimetabolites (antineoplastic agents) is similar to certain compounds such as vitamins, amino acids, and precursors of DNA or RNA, found naturally in human body. Antimetabolites help in treatment cancer by inhibiting cell division thereby
  • 12. hindering the growth of tumor cells. These agents get incorporated in the DNA or RNA to interfere with the process of division of cancer cells. They are commonly used to treat gastrointestinal tract, breast, and ovary tumors. Methotraxate, which is a commonly used antimetabolites chemotherapy agent, is effective in the S-phase of the cell cycle. It works by inhibiting an enzyme that is essential for DNA synthesis. 6-mercaptopurine and 5-fluorouracil (5FU) are two other commonly used antimetabolites. 5-Fluorouracil (5-FU) works by interfering with the DNA components, nucleotide, to stop DNA synthesis. This drug is used to treat many different types of cancers including breast, esophageal, head, neck, and gastric cancers. 6- mercaptopurine is an analogue of hypoxanthine and is commonly used to treat Acute Lymphoblastic Leukemia (ALL). Other popular antimetabolite chemotherapy drugs are Thioguanine, Cytarabine, Cladribine. Gemcitabine, and Fludarabine. Anthracyclines Anthracyclines are daunosamine and tetra-hydronaphthacenedione-based chemotherapy agents. These compounds are cell-cycle nonspecific and are used to treat a large number of cancers including lymphomas, leukemia, and uterine, ovarian, lung and breast cancers. Anthracyclines drugs are developed from natural resources. Daunorubicin is developed by isolating it from soil-dwelling fungus Streptomyces. Doxorubicin, which is another commonly used anthracycline chemotherapy agent, is isolated from mutated strain of Streptomyces. Doxorubicin is more effective in treating solid tumors. Idarubicin, Epirubicin, and Mitoxantrone are few of the other commonly used anthracycline chemotherapy drugs. Anthracyclines work by forming free oxygen radicals that breaks DNA strands thereby inhibiting DNA synthesis and function. These chemotherapeutic agents form a complex with DNA and enzyme to inhibit the topoisomerase enzyme. Topoisomerase is an enzyme class that causes the supercoiling of DNA, allowing DNA repair, transcription, and replication. Antitumor antibiotics Antitumor antibiotics are also developed from the soil fungus Streptomyces. These drugs are widely used to treat and suppress development of tumors in the body. Similar to anthracyclines, antitumor antibiotics drugs also form free oxygen radicals that result in DNA strand breaks, killing the growth of cancer cells. In most of the cases, these drugs are used in combination with other chemotherapy agents. Bleomycin is one of the commonly used antitumor antibiotic used to treat testicular cancer and hodgkin’s lymphoma. Monoclonal antibodies The treatment is known to be useful in treating colon, lung, head, neck, and breast cancers. Some of the monoclonal drugs are used to treat chronic lymphocytic leukemia, acute myelogenous leukemia, and non-Hodgkin's lymphoma. Monoclonal antibodies work by attaching to certain parts of the tumor-specific antigens and make them easily recognizable by the host’s immune system. They also prevent growth of cancer cells by blocking the cell receptors to which chemicals called ‘growth factors’ attach promoting cell growth.
  • 13. Monoclonal antibodies can be combined with radioactive particles and other powerful anticancer drugs to deliver them directly to cancer cells. Using this method, long term radioactive treatment and anticancer drugs can be given to patients without causing any serious harm to other healthy cells of the body. Platinum Platinum-based chemotherapy agents work by cross-linking subunits of DNA. These agents act during any part of cell cycle and help in treating cancer by impairing DNA synthesis, transcription, and function. Cisplatin, although found to be useful in treating testicular and lung cancer, is highly toxic and can severely damage the kidneys of the patient. Second generation platinum- complex carboplatin is found to be much less toxic in comparison to cisplatin and has fewer kidney-related side effects. Oxaliplatin, which is third generation platinum-based complex, is found to be helpful in treating colon cancer. Although, oxaliplatin does not cause any toxicity in kidney it can lead to severe neuropathies. Platinum-based drugs are often used for treatment of mesothelioma. Role of UNUSUAL Plant Derivatives They are primarily categorized into four groups: topoisomerase inhibitors, vinca alkaloids, taxanes, and epipodophyllotoxins. Topoisomerase inhibitors are chemotherapy agents are categorized into Type I and Type II Topoisomerases inhibitors and they work by interfering with DNA transcription, replication, and function to prevent DNA supercoiling. Type I Topoisomerase inhibitors: These chemotherapy agents are extracted from the bark and wood of the Camptotheca accuminata. • Type II Topoisomerase inhibitors: These are extracted from the alkaloids found in the roots of May Apple plants. • Amsacrine, etoposide, etoposide phosphate, and teniposide are some of the examples of type II topoisomerase inhibitors. Vinca alkaloids Vinca alkaloids are derived from the periwinkle plant, Vinca rosea (Catharanthus roseus) and are useful in treating leukemias. They are effective in the M phase of the cell cycle and work by inhibiting tubulin assembly in microtubules. Vincristine, Vinblastine, Vinorelbine, and Vindesine are some of the popularly used vinca alkaloid chemotherapy agents used today. Major side effect of vinca alkaloids is that they can cause neurotoxicity in patients. Taxanes Taxanes are plant alkaloids that are isolated from the bark of the Pacific yew tree, Taxus brevifolia.. Paclitaxel and docetaxel are commonly used taxanes. Taxanes work in the M-phase of the cell cycle and inhibit the function of microtubules by binding with them. Taxanes are used to treat a large array of cancers including breast, ovarian, lung, head and neck, gastric, esophageal, prostrate and gastric cancers. The main side effect of taxanes is that they lower the blood counts in patients.
  • 14. Epipodophyllotoxins Epipodophyllotoxins chemotherapy agents are extracted from the American May Apple tree (Podophyllum peltatum). Etoposide and Teniposide are commonly used epipodophyllotoxins chemotherapy agents which are effective in the G1 and S phases of the cell cycle. They prevent DNA replication by stopping the cell from entering the G1 phase and stop DNA replication in the S phase. Phytoestrogens and Cancer Treatment Phytoestrogens are polyphenol compounds of plant origin that exhibit a structural similarity to the mammalian steroid hormone 17β-oestradiol. In Asian nations the staple consumption of phyto-oestrogen-rich foodstuffs correlates with a reduced incidence of breast cancer. Human dietary intervention trials have noted a direct relationship between phyto-oestrogen ingestion and a favourable hormonal profile associated with decreased breast cancer risk. However, these studies failed to ascertain the precise effect of dietary phyto-oestrogens on the proliferation of mammary tissue. Epidemiological and rodent studies crucially suggest that breast cancer chemoprevention by dietary phyto-oestrogen compounds is dependent on ingestion before puberty, when the mammary gland is relatively immature. Phyto-oestrogen supplements are commercially marketed for use by postmenopausal women as natural and safe alternatives to hormone replacement therapy. Of current concern is the effect of phyto-oestrogen compounds on the growth of pre-existing breast tumours. Data are contradictory, with cell culture studies reporting both the oestrogenic stimulation of oestrogen receptor-positive breast cancer cell lines and the antagonism of tamoxifen activity at physiological phyto-oestrogen concentrations. Conversely, phyto-oestrogen ingestion by rodents is associated with the development of less aggressive breast tumours with reduced metastatic potential. Despite the present ambiguity, current data do suggest a potential benefit from use of phyto-oestrogens in breast cancer chemoprevention and therapy. Phyto-oestrogens may be classified into a number of principal groups [2,7-9]: the isoflavones (genistein, daidzein, biochanin A), the lignans (enterolactone, enterodiol), the coumestans (coumestrol) and the stilbenes (resveratrol). As illustrated in Fig. 1, all are polyphenols sharing structural similarity with the principal mammalian estrogen 17β- oestradiol. Shared features include the presence of a pair of hydroxyl groups and a phenolic ring, which is required for binding to the oestrogen receptor (ER) subtypes α and β. The position of the hydroxyl groups appears to be important in determining ER binding ability and transcriptional activation, with maximal potency achieved at positions four, six and seven [10-12]. The isoflavones are naturally found in soybeans and soy- based food products, including tofu, soy milk, textured soy protein and miso. Lignans are present in flaxseed and most fruit and vegetables, and the predominant dietary source of stilbenes is peanuts, grapes and red wine [7,13]. The coumestans are much less frequently consumed within the human diet, but they are more potent activators of ER signalling pathways than are the isoflavones genistein and daidzein [10,14]. By contrast, the stilbene resveratrol is the least potent activator of ER signalling [11]. The isoflavones are present in soy as β-glucosides. Metabolism by the gastrointestinal microflora yields a number of metabolites including equol and O-desmethyl-angolensin. Parental compounds and their metabolites are absorbed into the bloodstream, becoming rapidly detectable in the plasma and urine [15-19]. Plasma isoflavone concentrations are considerably elevated in Asian populations as compared with in western ones. A recent comparison of Japanese and UK females revealed an almost 20-fold increase in plasma genistein levels in the Japanese cohort, and daidzein concentrations were similarly
  • 15. elevated by 18-fold [20]. Plasma isoflavone concentrations may accumulate to approximately 100- to 1000-fold higher than endogenous oestradiol levels following the ingestion of soy-rich meal. However, research suggests a decreased ER binding affinity of isoflavone compounds as compared with the mammalian oestrogens [9,10,21,22]. Competition binding assays revealed a 50-fold lower binding affinity of genistein for cytoplasmic ER sites as compared to 17β-oestradiol [23]. The complete metabolic activation of soy isoflavones is proposed to occur locally within target tissue. In support of this hypothesis, the analysis of tissue culture supernatants from genistein and biochanin A treated MCF-7 and T47-D cells revealed the presence of hydroxylated and methylated isoflavone metabolites [24]. Current research suggests a role for the CYP family of cytochrome P450 enzymes in the intratumour metabolism of phyto-oestrogen compounds [25,26]. The CYP1B1 enzyme is expressed in a wide range of human tumour types, including breast [27]; however, expression is absent within normal tissue. CYP1B1 is proposed to catalyze the hydroxylation of resveratrol to yield the related stilbene piceatannol [26]. Piceatannol is a tyrosine kinase inhibitor with antileukaemic properties, which differs in structure from resveratrol by the presence of an additional hydroxyl group. A number of plant flavonoids are also putative substrates for the CYP family of enzymes [25]. Maubach and coworkers [28] recently reported the use of high-performance liquid chromatography to quantify isoflavones in normal breast biopsy tissue following consumption of soy for 5 consecutive days. Equol concentrations were approximately fivefold higher in the breast tissue homogenates than in serum, providing further evidence for the metabolism of phyto-oestrogen compounds within mammary tissue. Role of phyto-oestrogens in breast cancer Serum concentrations of 17β-estradiol are approximately 40% lower in Asian women than in their Caucasian counterparts [29]. A low lifetime exposure to oestrogen is associated with a reduced risk for breast cancer. Human dietary intervention studies revealed a direct association between the modest consumption of soy products and a reduction in circulating steroid hormone levels. Daily consumption of 154 mg isoflavones for the duration of a single menstrual cycle correlated with substantially decreased plasma concentrations of 17β-oestradiol and progesterone in a cohort of premenopausal women [18]. A longer-term study conducted by Kumar and coworkers [30] similarly reported a moderate decrease in serum oestradiol and oestrone levels following daily ingestion of 40 mg isoflavones for 3 months. Menstrual cycle length was increased by 3.52 days, and the follicular phase of the cycle was extended by 1.46 days. Increased menstrual cycle length may serve to reduce the total number of cycles per lifetime, therefore decreasing the total exposure of breast epithelia to endogenous oestrogens. Conversely, a year-long dietary intervention trial involving 34 premenopausal women failed to reveal a significant effect of 100 mg/day isoflavone consumption either on menstrual cycle length or on serum levels of various steroid hormones, including oestrone, oestradiol and progesterone [19]. As a possible explanation for these contradictory data, a study conducted by Duncan and coworkers [31] revealed differential hormonal effects of soy isoflavones depending on the ability to excrete the daidzein metabolite equol. Daily ingestion of 10 mg soy protein by premenopausal equol excretors resulted in a hormonal profile associated with reduced breast cancer risk, characterized by lowered plasma levels of oestrone, oestrone–sulphate and testosterone. Hormone levels however remained unchanged in the equol nonexcretors after soy ingestion. The reduction in steroid hormone levels by phyto-oestrogens is proposed to occur via the direct regulation of 17β-oestradiol biosynthesis and metabolism. Phytochemicals
  • 16. isolated from vegetable extracts effectively suppress the activity of the aromatase enzymes, which are responsible for conversion of androgens to oestrogens [32]. Isoflavone concentrations of 1–10 μmol/l similarly reduced by 50% the activity of the estradiol biosynthetic enzymes 3β-hydroxysteroid dehydrogenase and 17β- hydroxysteroid dehydrogenase [10]. The daily ingestion of 113–202 mg isoflavones by premenopausal women correlated with a 40% increase in the urinary excretion of 2- hydroxyestrone, a putative anticancer metabolite of 17β-oestradiol [33]. The above studies thus suggest a dual chemoprotectant mechanism of soy, in which the isoflavones suppress steroid hormone biosynthesis while promoting the metabolism of oestradiol to the protective 2-hydroxylated metabolites. Despite their apparent effect on endogenous hormone levels, the role of phyto- oestrogens in breast cancer initiation and development is unclear. Few studies to date have addressed the effects of long-term phyto-oestrogen exposure in humans. Daily dietary supplementation with 45 mg soy isoflavone for 14 days correlated with increased proliferation of normal breast epithelia in a group of 48 premenopausal women [16]. Expression of the ER target protein progesterone receptor was upregulated, suggesting an oestrogenic effect. An identical trial using a larger cohort of 84 premenopausal women conversely found no significant effect of soy consumption on the proliferation of normal breast tissue [17]. A number of recent epidemiological studies similarly failed to correlate soyfood consumption with reduced breast cancer risk. A Japanese prospective study conducted in a cohort of approximately 35,000 women [34] revealed no significant association between soy consumption during adulthood and breast cancer incidence. The retrospective analysis of soy food intake in a multiethnic cohort of non-Asian breast cancer patients and control individuals residing in the USA similarly failed to correlate soy intake with breast cancer risk [35]. Increasing epidemiological evidence suggests that the chemoprotectant effects of phyto- oestrogens are dependent on a lifelong exposure from childhood. A retrospective study revealed decreased soyfood intake during adolescence in a cohort of 1459 Chinese breast cancer patients, as compared with age-matched control individuals [36]. Daily soy consumption between the ages of 13 and 15 was estimated at 6.45 g in the patient group, increasing to 7.23 g in the control cohort. A potential flaw in the study, however, concerns the ability of women up to the age of 64 years to recall accurately the precise soyfood quantities consumed many years earlier during adolescence. These observations may nonetheless explain the apparent lack of a growth inhibitory effect of soy isoflavones in the adult dietary intervention studies discussed above. Isoflavones are detectable in breast milk following soy consumption [15], implying that the lower breast cancer incidence in Asian countries may be attributable to phyto-oestrogen exposure from birth via breast-feeding. Rodent studies have accordingly revealed the effective transfer of genistein from maternal milk to offspring [37]. Rodent breast cancer models Most available information regarding the effects of phyto-oestrogens on tumour initiation and the growth of pre-existing tumours is derived from rodent studies. A number of similarities do exist between mammary gland development in rodents and humans. In both species the differentiation of breast tissue to form lobules and terminal end-bud structures occurs prepubertally. Further maturation does take place throughout adulthood, giving rise to alveolar buds, which become alveoli during pregnancy and lactation [38]. Rodent dietary intervention studies using phyto-oestrogens have reported chemopreventive activities when feeding is initiated before puberty, at a time when the mammary gland is undergoing development [3,38-40]. The consumption of a resveratrol-
  • 17. supplemented diet by adolescent rats served to decrease sensitivity to the chemical carcinogens 7,12-dimethylbenz(a)anthracene (DMBA) and N-methyl-N-nitrosaurea [39,40]. DMBA treatment induced mammary tumours in 45% of the resveratrol-treated rodents, increasing to 75% in the group receiving a control diet. An extended tumour latency period in excess of 3 weeks was observed in the resveratrol treatment groups, with the resultant tumours retaining a more differentiated morphology as compared with control animals [18,39]. Resveratrol consumption was also associated with the reduced mammary expression of a number of proteins that are putatively involved in malignant progression, including cyclo-oxygenase-2, matrix metalloprotease-9 and nuclear factor- κB. Similar findings have been noted in prepubertal rats fed an isoflavone-containing diet before tumour initiation using DMBA. Despite having no effect on mammary tumour incidence, soy isoflavone consumption was associated with an increased tumour latency period. The resultant tumours excised from the soy-fed animals were smaller in size and exhibited a more differentiated phenotype compared with control animals [42]. It has been proposed that phyto-oestrogens protect against cancer development in adolescent rodents by promoting maturation of the mammary gland. Analysis of breast tissue from prepubertal rats injected with genistein revealed a decrease in the number of immature terminal end-buds, together with an increase in the more differentiated lobules type II [38]. Genistein treatment of human breast cancer cell lines has similarly been found to induce the expression of a number of maturation markers, including casein, lipid droplets and intercellular adhesion molecule-1 [43]. The effect of soy isoflavones on spontaneous tumour development was recently investigated using neu-ErbB2 over-expressing transgenic mice, which characteristically develop multiple mammary tumours during adulthood [40]. Tumour initiation was temporarily delayed following the consumption of an isoflavone mix, but no chemoprotective effects were observed in mice consuming either genistein or daidzein in isolation. An equal rate of tumour growth was noted in the control and treatment groups, although the isoflavone group exhibited a lower incidence of lung metastases [40]. Antimetastatic activities of the isoflavones were similarly revealed in a study in which mice were fed an isoflavone-supplemented diet before injection with the metastatic 4526 mammary carcinoma cell line [44]. The isoflavone diet was continued following surgical excision of the resultant mammary tumour at a size of 1.0 cm diameter. Both the incidence and size of macroscopically detectable lung metastases were significantly reduced in the soy-fed mice, suggesting a potential clinical application of soy isoflavones in the prevention of metastasis. Phyto-oestrogens and tamoxifen The selective ER modulator tamoxifen is used clinically in the adjuvant treatment of oestrogen-dependent breast cancer. The drug is also administered as a prophylactic to individuals who are at high risk for developing the disease [45,46]. Side effects associated with tamoxifen therapy include menopause-like symptoms such as hot flushes, joint pain, sleep disorders and depression, which may be reduced by the use of hormone replacement therapy (HRT) [47,48]. Long-term HRT is associated with an increased risk for mammary carcinogenesis, and its use by breast cancer patients is therefore discouraged. As a natural alternative, patients may self-medicate with soy isoflavone supplements to alleviate the tamoxifen-induced menopausal symptoms [49]. Published literature regarding the ingestion of dietary phyto-oestrogens by breast cancer patients and survivors is, however, controversial [50,51]. The consumption of genistein by athymic mice antagonized the ability of tamoxifen to inhibit the proliferation of oestrogen-dependent mammary tumours [52]. Tumour suppression by tamoxifen correlated with decreased expression of the ER-inducible
  • 18. genes presenelin-2 (pS2) and cyclin D1. Tumour growth was significantly enhanced in mice simultaneously exposed to tamoxifen and genistein, whereas levels of pS2 and cyclin D1 expression were increased. Physiological concentrations of genistein were similarly found to reverse the antagonistic effects of 4-hydroxytamoxifen on ER signalling pathways [53], promoting the binding of ER-α to the positively acting steroid receptor coactivator (SRC)-1. A recent tissue culture study conversely reported a synergistic antiproliferative effect of tamoxifen and genistein [54]. The proliferation of a panel of dysplastic and cancerous breast cell lines was inhibited by tamoxifen in a dose- dependent manner, and growth was more potently suppressed by combined treatment with tamoxifen and genistein. Hormone-dependent mechanisms of phyto-oestrogen action Oestrogen signalling typically involves the diffusion of ligand through the cell cytoplasm and subsequent binding to the nuclear receptor subtypes ER-α and ER-β. Ligand-bound receptors dimerise and associate with oestrogen response element (ERE) and activator protein-1 element located in the promoter region of target genes, thereby activating transcription. The association between receptor dimers and DNA response elements is enhanced by the binding of cofactor proteins, such as amplified in breast cancer-1, thyroid hormone receptor-associated protein, SRC-1, glutamate receptor interacting protein-1 and translation initiation factor 2 [55]. Examples of ERE-induced genes include PR, c-fos, bcl-2 and cathepsin D, whereas pS2 and cyclin D1 are transcribed via the activator protein-1 response element. In breast carcinoma cell lines containing functional ER subtypes the isoflavones exert a biphasic growth effect, stimulating cellular proliferation at concentrations below 5 μmol/l and inhibiting growth in a dose-dependent manner at elevated doses [23,43,56,57]. Growth inhibition correlates with decreased DNA synthesis and cell cycle arrest at the G2/M checkpoint [23,54,56,58-60]. Current research suggests a principal signalling role of ER-β in response to isoflavone exposure [61]. Whereas 17β-oestradiol binds to ER-α and ER-β with equal affinity, the soy isoflavones selectively associate with ER-β [62]. Receptor binding assays revealed an eightfold to 16-fold increase in the affinity of genistein, daidzein and biochanin A for ER-β as compared with ER-α [63]. In ER- negative breast cancer cells transfected to express ER-α alone, genistein was only weakly able to stimulate gene transcription through the ERE. By contrast, genistein effectively bound to ER-β and promoted the association of cofactor proteins, thereby regulating downstream ER-β-mediated gene transcription [62]. The preferential binding affinity of genistein for ER-β similarly resulted in a respective 12,000-fold and 33-fold increase in the recruitment of translation initiation factor-2 and SRC-1a to ER-β as compared with ER-α [22]. An enhanced transcriptional activity in response to genistein was, however, noted in cells transfected to express both receptor subtypes as compared with cells solely expressing ER-β [64]. Although ER-α is itself unable to mediate isoflavone signal transduction, it was postulated that the presence of the receptor subtype may enhance ER-β signalling via the formation of ER-α/β heterodimers. These observations imply that the precise tissue-specific effects of the soy isoflavones are dependent on the expression levels and ratios of ER-α and ERβ. The various cofactors are similarly expressed in a tissue-specific manner, therefore further influencing the cellular response to dietary phyto-oestrogens. The stilbene resveratrol is structurally similar to the synthetic oestrogen diethylstilbestrol. Treatment of breast cancer cell lines with resveratrol represses proliferation in a dose- dependent manner, inducing G2/M phase cell cycle arrest [39]. Resveratrol exhibits a relatively weak ER-binding affinity as compared with oestradiol [65]; however, unlike the soy isoflavones, it is able to bind to both ER-α and ER-β with equal affinity. In cells
  • 19. transfected to express either ER-α or ER-β resveratrol was found to act as an agonist for both receptor subtypes, stimulating ERE transcriptional activity through either ER-α or ER-β alone [21]. Similar agonist activity was observed in MCF-7 breast cancer cells, which predominantly express the ER-α isoform. Resveratrol induced the dose- dependent activation of ERE-mediated transcription, also upregulating the expression of the ER target genes pS2 and PR [65]. Recent studies proposed that the cell cycle inhibitor protein p21WAF1 is a potential downstream target of resveratrol-induced ER signalling pathways [66]. The treatment of ER-α-expressing breast cancer cells with resveratrol resulted in a 23-fold increase in p21WAF1 gene expression, as determined by cDNA microarray analysis. The resveratrol-mediated induction of p21WAF1 was blocked by treatment with the pure anti-oestrogen ICI 182,780, confirming p21WAF1 gene regulation as an ER-mediated event. Hormone-independent mechanisms At concentrations in excess of 25 μmol/l, the soy isoflavones are capable of inducing apoptosis in human breast cancer cells [23,67-69]. ER-negative cell lines retain sensitivity to the apoptotic effects of soy isoflavones, thereby confirming that apoptosis occurs in a hormone-independent manner. Apoptosis was effectively induced in the ER- α-negative MDA-MB-231 breast cancer cell line by genistein and daidzein concentrations of 50–100 μmol/l [59,60]. In MCF-7 cell cultures the induction of cell death by treatment with genistein coincided with the increased expression of the proapoptotic proteins Bax and p53 [67]. Breast cancer cell lines expressing mutant p53 also undergo apoptosis in response to phyto-oestrogen treatment, thereby implying apoptosis induction by both p53-dependent and p53-independent mechanisms [67,70]. The polyphenol epigallocatechin (EGC) is principally found in green tea and is proposed to have anticancer properties. Treatment of p53-mutant breast cancer cells with 100 μmol/l EGC induced a 40% increase in apoptosis, correlating with increased Bax expression and reduced levels of the antiapoptotic protein Bcl-2 [70]. EGC-induced apoptosis was abolished following treatment with anti-Fas neutralizing antibodies or caspase inhibitors, suggesting the involvement of Fas signalling pathways. Although phyto-oestrogen compounds are effective inducers of apoptosis in cell culture models, it is unlikely that plasma isoflavone concentrations would accumulate to the required levels for the activation of apoptotic pathways in vivo. It is estimated that plasma phyto- oestrogen concentrations may reach a maximum of 2–4 μmol/l following the moderate consumption of soy products [15,52], although it is possible that higher levels may be present in target tissues. In a recently reported study, equol concentrations within breast tissue were found to exceed serum levels; however, the reverse was true for genistein and daidzein [28]. A recent in vitro study [68] revealed the flavone baicalein to be a more potent inducer of apoptosis than genistein. Baicalein is isolated from the plant Scutellariae radix and is a common ingredient in herbal tea preparations. A concentration of 10 μmol/l baicalein induced significant cell death in MCF-7 cell cultures, suggesting baicalein as a potentially useful pharmacological agent in breast cancer therapy. Dietary phyto-oestrogens are capable of inhibiting the proliferation of hormone- independent breast cell lines [43,54,58,69]. It has been proposed that growth inhibition in the absence of functional ER occurs via the inhibition of tyrosine kinase activity. The protein tyrosine kinases are involved in a number of growth factor signalling pathways, including transforming growth factor (TGF)-α, insulin-like growth factor (IGF)-I, IGF-II and epidermal growth factor (EGF). In ER-negative breast cancer cultures 5 μmol/l genistein negated the stimulatory effects of TGF-α, IGF-I and IGF-II, implying the inhibition of tyrosine kinase activity [23]. The human EGF receptor-2 oncogene (Her-2) is
  • 20. constitutively overexpressed in approximately 30% of breast cancers and is associated with a poor patient prognosis [71]. Research using breast cancer cell lines suggests that dietary phyto-oestrogens are capable of repressing EGF receptor activity. The inhibition of tyrosine kinase activity by 5 μmol/l genistein in MCF-7 cells correlated with the repression of EGF receptor tyrosine phosphorylation in response to EGF stimulation [23]. Similar findings were reported in a recent study investigating the chemoprotective effects of the green tea polyphenol epigallocatechin-3 gallate (EGCG). The treatment of Her-2/neu over-expressing mouse mammary cells with 20–80 μg/ml EGCG inhibited proliferation in a dose-dependent manner, correlating with a reduction in Her-2/neu signalling activity [72]. The basal tyrosine phosphorylation of Her-2/neu was decreased by approximately 96% following treatment with 80 μg/ml EGCG. Downstream activities of the signalling proteins phosphoinositide 3-kinase, Akt and nuclear factor-κB were similarly repressed, suggesting a potential clinical application of EGCG in breast cancer therapy. The soy isoflavones have additionally been proposed to regulate the proliferation of breast epithelia via an alternative mechanism involving the modulation of TGF-β synthesis [73]. In normal mammary tissue TGF-β maintains proliferative homeostasis by inhibiting the growth of epithelial cells [74,75]. The incubation of human mammary epithelial cells with 5 μmol/l genistein induced a fivefold increase in the level of TGF-β secretion [76]. The further analysis of media conditioned with human mammary epithelial cells revealed the presence of the active as opposed to latent form of TGF-β, thus implying a direct link between soy isoflavones and the TGF-β signalling pathway. Abbreviations DMBA = 7,12-dimethylbenz(a)anthracene; EGC = epigallocatechin; EGF = epidermal growth factor; EGCG = epigallocatechin-3 gallate; ER = oestrogen receptor; ERE = oestrogen response element; HRT = hormone replacement therapy; IGF = insulin-like growth factor; SRC = steroid receptor coactivator; TGF = transforming growth factor. References: Messina MJ: Legumes and soybeans: overview of their nutritional profiles and health effects. Am J Clin Nutr 1999, Suppl:439S-450S. Barnes S: Phytoestrogens and cancer. Baillières Clin Endocrinol Metab 1998, 12:559-579. Lamartiniere CA: Protection against breast cancer with genistein: a component of soy. Am J Clin Nutr 2000, Suppl:1705S-1707S. Dai Q, Shu X-O, Jin F, Potter JD, Kushi LH, Teas J, Gao Y-T, Zheng W: Population-based case- control study of soyfood intake and breast cancer risk in Shanghai. Br J Cancer 2001, 85:372-387. Wu AH, Ziegler RG, Nomura AMY, West DW, Kolonel LN, Horn-Ross PL, Hoover RN, Pike MC: Soy intake and risk of breast cancer in Asians and Asian Americans. Am J Clin Nutr 1998, Suppl:1437S-1443S. Adlercreutz H: Phyto-oestrogens and cancer. Lancet Oncol 2002, 3:364-373.
  • 21. Cassidy A, Hanley B, Lamuela-Raventos RM: Isoflavones, lignans and stilbenes – origins, metabolism and potential importance to human health. J Sci Food Agric 2000, 80:1044-1062. Setchell KDR, Radd S: Soy and other legumes: 'bean' around for a long time but are they the 'superfoods' of the millennium and what are the safety issues for their constituent phytoestrogens? Asia Pacific J Clin Nutri 2000, Suppl:S13-S22. Benassayag C, Perrot-Applanat M, Ferre F: Phytoestrogens as modulators of steroid action in target cells. J Chromatogr B Analyt Technol Biomed Life Sci 2002, 777:233-248. Le Bail J-C, Champavier Y, Chulia A-J, Habrioux G: Effects of phytoestrogens on aromatase, 3β and 17β-hydroxysteroid dehydrogenase activities and human breast cancer cells. Life Sci 2001, 66:1281-1291. Rosenberg Zand RS, Jenkins DJA, Diamandis EP: Steroid hormone activity of flavonoids and related compounds. Breast Cancer Res Treat 2000, 62:35-49. Tamir S, Eizenberg M, Somjen D, Stern N, Shelach R, Kaye A, Vaya J: Estrogenic and antiproliferative properties from licorice in human breast cancer cells. Cancer Res 2000, 60:5704-5709. Tham DM, Gardner CD, Haskell WL: Potential health benefits of dietary phytoestrogens: a review of the clinical, epidemiological and mechanistic evidence. J Clin Endocrinol Metab 1998, 83:2223-2235. Burow ME, Boue SM, Collins-Burow BM, Melnik LI, Duong BN, Carter-Wientjes CH, Li S, Wiese TE, Cleveland TE, McLachlan JA: Phytochemical glyceollins, isolated from soy, mediate hormonal effects through estrogen receptor α and β. J Clin Endocrinol Metab 2001, 86:1750-1758. Franke AA, Custer LJ, Tanaka Y: Isoflavones in human breast milk and other biological fluids. Am J Clin Nutr 1998, Suppl:1466S-1473S. McMichael-Philips DF, Harding C, Morton M, Roberts SA, Howell A, Potten CS, Bundred NJ: Effects of soy-protein supplementation on epithelial proliferation in the histologically normal human breast. Am J Clin Nutr 1998, Suppl:1431S-1436S. Hargreaves DF, Potten CS, Harding C, Shaw LE, Morton MS, Roberts SA, Howell A, Bundred NJ: Two- week dietary soy supplementation has an estrogenic effect on normal pre-menopausal breast. J Clin Endocrinol Metab 1999, 84:4017-4024. Lu L-JW, Anderson KE, Grady JJ, Kohen F, Nagamani M: Decreased ovarian hormones during a soya diet: implications for breast cancer prevention. Cancer Res 2000, 60:4112-4121. Maskarinec G, Williams AE, Inouye JS, Stanczyk FZ, Franke AA: A randomised isoflavone intervention among premenopausal women. Cancer Epidemiol Biomarkers Prev 2002, 11:195-201. Morton MS, Arisaka O, Miyake N, Morgan LD, Evans AJ: Phytoestrogen concentrations in serum from Japanese men and women over forty years of age. J Nutr 2002, 132:3168-3171. Bowers JL, Tyulmenkov VV, Jernigan SC, Klinge CM: Resveratrol acts as a mixed agonist/antagonist for estrogen receptors α and β. Endocrinology 2000, 141:3657-3667. Routledge EJ, White R, Parker MG, Sumpter JP: Differential effects of xenoestrogens on coactivator recruitment by estrogen receptor (ER) α and ERβ. J Biol Chem 2000, 275:35986-35993. Fioravanti L, Cappelletti V, Miodini P, Ronchi E, Brivio M, Di Fronzo G: Genistein in the control of breast cancer cell growth: insights into the mechanism of action in vitro.
  • 22. Cancer Lett 1998, 130:143-152. Peterson TG, Ji G-P, Kirk M, Coward L, Falany CN, Barnes S: Metabolism of the isoflavones genistein and biochanin A in human breast cancer cell lines. Am J Clin Nutr 1998, Suppl:1505S-1511S. Doostdar H, Burke MD, Mayer RT: Bioflavonoids: selective substrates and inhibitors for cytochrome P450 CYP1A and CYP1B1. Toxicology 2000, 144:31-38. Potter GA, Patterson LH, Wanogho E, Perry PJ, Butler PC, Ijaz T, Ruparelia KC, Lamb JH, Farmer PB, Stanley LA, Burke MD: The cancer preventative agent resveratrol is converted to the anti-cancer agent piceatannol by the cytochrome P450 enzyme CYP1B1. Br J Cancer 2002, 86:774-778. McFadyen MCE, Breeman S, Payne S, Stirk C, Miller ID, Melvin WT, Murray GI: Immunohistochemical localisation of cytochrome P450 CYP1B1 in breast cancer with monoclonal antibodies specific for CYP1B1. J Histochem Cytochem 1999, 47:1457-1464. Maubach J, Bracke ME, Heyerich A, Depypere HT, Serreyn RF, Mareel MM, de Keukeleire D: Quantitation of soy-derived phytoestrogens in human breast tissue and biological fluids by high-performance liquid chromatography. J Chromatogr B Analyt Technol Biomed Life Sci 2003, 784:137-144. Peeters PHM, Keinen-Boker L, van der Schouw YT, Grobbee DE: Phytoestrogens and breast cancer risk. Breast Cancer Res Treat 2003, 77:171-183. Kumar NB, Cantor A, Allen K, Riccardi D, Cox CE: The specific role of isoflavones on estrogen metabolism in pre-menopausal women. Cancer 2002, 94:1166-1174. Duncan AM, Merz-Demlow BE, Xu X, Phipps WR, Kurzer MS: Premenopausal equol excretors show plasma hormone profiles associated with lowered risk of breast cancer. Cancer Epidemiol Biomarkers Prev 2000, 9:581-586. Grube BJ, Eng ET, Kao Y-C, Kwon A, Chen S: White button mushroom phytochemicals inhibit aromatase activity and breast cancer cell proliferation. J Nutri 2001, 131:3288-3293. Lu L-JW, Cree M, Josyula S, Nagamani M, Grady JJ, Anderson KE: Increased urinary excretion of 2- hydroxyestrone but not 16α-hydroxyestrone in premenopausal women during a soya diet containing isoflavones. Cancer Res 2000, 60:1299-1305. Key TJ, Sharp GB, Appleby PN, Beral V, Goodman MT, Soda M, Mabuchi K: Soya foods and breast cancer risk: a prospective study in Hiroshima and Nagasaki, Japan. Br J Cancer 1999, 81:1248-1256. Horn-Ross PL, John EM, Lee M, Stewart SL, Koo J, Sakoda LC, Shiau AC, Goldstein J, Davis P, Perez-Stable EL: Phytoestrogen consumption and breast cancer risk in a multiethnic population. Am J Epidemiol 2001, 154:434-441. Shu XO, Jin F, Dai Q, Wen W, Potter JD, Kushi LH, Ruan Z, Gao Y-T, Zheng W: Soyfood intake during adolescence and subsequent risk of breast cancer among Chinese women. Cancer Epidemiol Biomark Prev 2001, 10:483-488. Lamartiniere CA: Timing of exposure and mammary cancer risk. J Mammary Gland Biol Neoplasia 2002, 7:67-76. Lamartiniere CA, Zhang J-X, Cotroneo MS: Genistein studies in rats: potential for breast cancer prevention and reproductive and developmental toxicity. Am J Clin Nutr 1998, Suppl:1400S-1405S.
  • 23. Banerjee S, Bueso-Ramos C, Aggarwal BB: Suppression of 7,12-dimethyl(a)anthracene-induced mammary carcinogenesis in rats by resveratrol: role of nuclear factor-κB, cyclooxygenase 2 and matrix metalloprotease 9. Cancer Res 2002, 62:4945-4954. Jin Z, MacDonald RS: Soy isoflavones increase latency of spontaneous mammary tumours in mice. J Nutr 2002, 132:3186-3190. Bhat KPL, Lantvit D, Christov K, Mehta RG, Moon RC, Pezzuto JM: Estrogenic and antiestrogenic properties of resveratrol in mammary tumour models. Cancer Res 2001, 61:7456-7463. Gallo D, Giacomelli S, Cantelmo F, Zannoni GF, Ferrandina G, Fruscella E, Riva A, Morazzoni P, Bombardelli E, Mancuso S, Scambia G: Chemoprevention of DMBA-induced mammary cancer in rats by dietary soy. Breast Cancer Res Treat 2001, 69:153-164. Constantinou AI, Krygier AE, Mehta RR: Genistein induces maturation of cultured human breast cancer cells and prevents tumour growth in nude mice. Am J Clin Nutr 1998, Suppl:1426S-1430S. Yan L, Li D, Yee JA: Dietary supplementation with isolated soy protein reduces metastasis of mammary carcinoma cells in mice. Clin Exp Metastas 2002, 19:535-540. Jordan VC: Long-term adjuvant tamoxifen therapy for breast cancer. Breast Cancer Res Treat 1990, 15:126-136. Morrow M, Jordan VC: Tamoxifen for the prevention of breast cancer in high-risk women. Ann Surg Oncol 2000, 7:67-71. Marsden J: The menopause, hormone replacement therapy and breast cancer. J Steroid Biochem Mol Biol 2002, 83:123-132. Chiechi LM: Hormone replacement therapy and health protection. Curr Opin Investig Drugs 2003, 4:439-443. Wuttke W, Jarry H, Westphalen S, Christoffel V, Seidlova-Wuttke D: Phytoestrogens for hormone replacement therapy? J Steroid Biochem Mol Biol 2002, 83:133-147. Messina MJ, Loprinzi CL: Soy for breast cancer survivors: a critical review of the literature. J Nutr 2001, Suppl:3095S-3108S. This P, De la Rochefordière A, Clough K, Fourquet A, Magdelenat H: Phytoestrogens after breast cancer. Endocr Relat Cancer 2001, 8:129-134. Ju YH, Doerge DR, Allred KF, Allred CD, Helferich WG: Dietary genistein negates the inhibitory effect of tamoxifen on growth of estrogen-dependent human breast cancer (MCF-7) cells implanted in athymic mice. Cancer Res 2002, 62:2474-2477. Schwartz JA, Liu G, Brooks SC: Genistein-mediated attenuation of tamoxifen-induced antagonism from estrogen receptor-induced genes. Biochem Biophy Res Commun 1998, 253:38-43. Tanos V, Brzezinski A, Drize O, Strauss N, Peretz T: Synergistic inhibitory effects of genistein and tamoxifen on human dysplastic and malignant epithelial breast cells in vitro. Eur J Obstet Gynecol Reprod Biol 2002, 102:188-194. Hall JM, Couse JF, Korach KS: The multifaceted mechanisms of estradiol and estrogen receptor signaling. J Biol Chem 2001, 276:36869-36872. Hsu J-T, Hung H-C, Chen C-J, Hsu W-L, Ying C: Effects of the dietary phytoestrogen biochanin A on cell growth in the mammary carcinoma cell line MCF-7.
  • 24. J Nutr Biochem 1999, 10:510-517. Maggiolini M, Bonofiglio D, Marsico S, Panno ML, Cenni B, Picard D, Ando S: Estrogen receptor α mediates the proliferative but not the cytotoxic dose-dependent effects of two major phytoestrogens on human breast cancer cells. Mol Pharmacol 2001, 60:595-602. Dampier K, Hudson EA, Howells LM, Manson MM, Walker RA, Gescher A: Differences between human breast cell lines in susceptibility towards growth inhibition by genistein. Br J Cancer 2001, 85:618-624. Upadhyay S, Neburi M, Chinni SR, Alhasan S, Miller F, Sarkar FH: Differential sensitivity of normal and malignant breast epithelial cells to genistein is partly mediated by p21WAF1. Clin Cancer Res 2001, 7:1782-1789. Nomoto S, Arao Y, Horiguchi H, Ikeda K, Kayama F: Oestrogen causes G2/M arrest and apoptosis in breast cancer MDA-MB-231. Oncol Rep 2002, 9:773-776. Morito K, Aomori T, Hirose T, Kinjo J, Hasegawa J, Ogawa S, Inoue S, Muramatsu M, Masamune Y: Interaction of phytoestrogens with estrogen receptors α and β (II). An J, Tzagarakis-Foster C, Scharschmidt TC, Lomri N, Leitman DC: Estrogen receptor β-selective transcriptional activity and recruitment of coregulators by phytoestrogens. J Biol Chem 2001, 276:17808-17814. Liu J, Burdette JE, Xu H, Gu C, van Breemen RB, Bhat KPL, Booth A, Constantinou AI, Pezzuto JM, Fong HHS, Farnsworth NR, Bolton JL: Evaluation of estrogenic activity of plant extracts for the potential treatment of menopausal symptoms. J Agric Food Chem 2001, 49:2472-2479. Petterson K, Delaunay F, Gustafsson J-A: Estrogen receptor β acts as a dominant regulator of estrogen signaling. Oncogene 2000, 19:4970-4978. Gehm BD, McAndrews JM, Chien P-Y, Jameson JL: Resveratrol, a polyphenolic compound found in grapes and wine, is an agonist for the estrogen receptor. Proc Natl Acad Sci USA 1997, 94:14138-14143. Levenson AS, Gehm BD, Pearce ST, Horiguchi J, Simons LA, Ward JE, Jameson JL, Jordan VC: Resveratrol acts as an estrogen receptor (ER) agonist in breast cancer cells stably transfected with ER-α. Int J Cancer 2003, 104:587-596. Leung LK, Wang TT: Bcl-2 is not reduced in the death of MCF-7 cells at low genistein concentration. J Nutri 2000, 130:2922-2926. Po LS, Chen Z-Y, Tsang DSC, Leung LK: Baicalein and genistein display differential actions on estrogen receptor (ER) transactivation and apoptosis in MCF-7 cells. Cancer Lett 2002, 187:33-34. Pozo-Guisado E, Alvarez-Barrientos A, Mulero-Navarro S, Santiago-Josefat B, Fernandez-Salguero PM: The antiproliferative activity of resveratrol results in apoptosis in MCF-7 but not in MDA-MB- 231 human breast cancer cells: cell-specific alterations of the cell cycle. Biochem Pharmacol 2002, 64:1375-1386. Vergote D, Cren-Olive C, Chopin V, Toillon R-A, Rolando C, Hondermarck H, Le Bourhis X: (-)- Epigallocatechin (EGC) of green tea induces apoptosis of human breast cancer cells but not of their normal counterparts. Breast Cancer Res Treat 2002, 76:195-201. Masood S, Bui MM: Prognostic and predictive value of HER2/neu oncogene in breast cancer. Microsc Res Tech 2002, 59:102-108. Pianetti S, Guo S, Kavanagh KT, Sonenshein GE: Green tea polyphenol epigallocatechin-3 gallate inhibits Her-2/Neu signalling, proliferation and transformed phenotype of breast cancer cells.
  • 25. Cancer Res 2002, 62:652-655. Kim H, Peterson TG, Barnes S: Mechanisms of action of the soy isoflavone genistein: emerging role for its effects via transforming growth factor β signalling pathways. Am J Clin Nutr 1998, Suppl:1418S-1425S. Sovak MA, Arsura M, Zanieski G, Kavanagh KT, Sonenshein GE: The inhibitory effects of transforming growth factor-β1 on breast cancer cell proliferation are mediated through regulation of aberrant nuclear factor-κB/rel expression. Cell Growth Differ 1999, 10:537-544. Knabbe C, Lippman ME, Wakefield LM, Flanders KC, Kasid A, Derynck R, Dickson RB: Evidence that transforming growth factor-β is a hormonally regulated negative growth factor in human breast cancer cells. Cell 1987, 48:417-428. Kim H, Xu J, Su Y, Xia H, Li L, Peterson G, Murphy-Ullrich J, Barnes S: Actions of the soy phytoestrogen genistein in models of human chronic disease: potential involvement of transforming growth factor-β. Biochem Soc Trans 2001, 29:216-222. Holmberg L, Anderson H: HABITS (hormonal replacement therapy after breast cancer – is it safe?), a randomised comparison: trial stopped. Effects of Pomegranate Chemical Constituents/Intestinal Microbial Metabolites on CYP1B1 in 22Rv1 Prostate Cancer Cells Sashi G. Kasimsetty†, Dobroslawa Bialonska† , Muntha K. Reddy†, Cammi Thornton‡, Kristine L. Willett‡§ and Daneel Ferreira*†§ † Department of Pharmacognosy ‡ Department of Pharmacology § National Center for Natural Product Research, Research Institute for Pharmaceutical Sciences School of Pharmacy, The University of Mississippi, University, Mississippi 38677 Institute of Environmental Sciences, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland J. Agric. Food Chem., 2009, 57 (22), pp 10636–10644 DOI: 10.1021/jf902716r Publication Date (Web): October 26, 2009 Copyright © 2009 American Chemical Society *To whom correspondence should be addressed. Tel: 662-915-7026. Fax: 662-915- 6975. E-mail: dferreir@olemiss.edu. Abstract The cytochrome P450 enzyme, CYP1B1, is an established target in prostate cancer chemoprevention. Compounds inhibiting CYP1B1 activity are contemplated to exert beneficial effects at three stages of prostate cancer development, that is, initiation, progression, and development of drug resistance. Pomegranate ellagitannins/microbial metabolites were examined for their CYP1B1 inhibitory activity in a recombinant CYP1B1-mediated ethoxyresorufin-O- deethylase (EROD) assay. Urolithin A, a microbial metabolite, was the most potent uncompetitive inhibitor of CYP1B1-mediated EROD activity, exhibiting 2-fold selectivity over CYP1A1, while urolithin B was a noncompetitive inhibitor with 3-fold selectivity. The punicalins and punicalagins exhibited potent CYP1A1 inhibition with 5−10-fold selectivity over CYP1B1. Urolithins, punicalins, and punicalagins were tested for their 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced CYP1
  • 26. inhibitory activity in the 22Rv1 prostate cancer cell line. Urolithins A and B showed a decrease in their CYP1-mediated EROD inhibitory IC50 values upon increasing their treatment times from 30 min to 24 h. Urolithin C, 8-O-methylurolithin A, and 8,9-di-O- methylurolithin C caused a potent CYP1-mediated EROD inhibition in 22Rv1 cells upon 24 h of incubation. Neutral red uptake assay results indicated that urolithin C, 8-O- methylurolithin A, and 8,9-di-O-methylurolithin C induced profound cytotoxicity in the proximity of their CYP1 inhibitory IC50 values. Urolithins A and B were studied for their cellular uptake and inhibition of TCDD-induced CYP1B1 expression. Cellular uptake experiments demonstrated a 5-fold increase in urolithin uptake by 22Rv1 cells. Western blots of the CYP1B1 protein indicated that the urolithins interfered with the expression of CYP1B1 protein. Thus, urolithins were found to display a dual mode mechanism by decreasing CYP1B1 activity and expression. Introduction Dietary intervention to prevent carcinogenesis has been well-established in epidemiological studies. The consumption of fruits and vegetables is considered to be a safeguard against various forms of cancers. Polyphenols are the major constituents of fruit and vegetable diets and are believed to elicit a number of biological properties due to their antioxidant and anticarcinogenic activities (1). A number of flavonoids such as quercetin, chrysin, apigenin, and luteolin have been investigated for their cytochrome P450 1 (CYP1) enzyme inhibition activities, indicating that flavonoid-related anticarcinogenesis is mediated in part by CYP1 inhibition (2). Ellagic acid, the hydrolysis product of ellagitannins, exhibits anticarcinogenic effects by inhibition of CYP1A1- dependent activation of procarcinogens. A number of ellagic acid analogues showed similar inhibitory activities against CYP1-mediated benzo[a]pyrene activation (3). Ellagic acid is a major polyphenol in pomegranate juice. Pomegranate juice polyphenols showed a strong inhibitory activity against estrogen-dependent MCF-7 cell lines. In in vivo studies, pomegranate juice exhibited 47% inhibition of cancerous lesion formation induced by the carcinogen, 7,12-dimethylbenz[a]anthracene (4), indicating its potential use as an adjuvant therapeutic in human breast cancer treatment. Pomegranate juice components are also believed to exert cancer chemopreventive activity against skin and colon cancer. Importantly, the consumption of pomegranate juice decreased the clinical reemergence of prostate cancer-specific antigen in prostate cancer patients after primary therapy (5, 6). Pomegranate ellagitannins are transformed by human colonic flora into bioavailable organic molecules called urolithins (7). The pomegranate microbial metabolites preferentially accumulate in prostate, colon, and intestinal tissues relative to other organs in a mouse model and exert their beneficial effects to a greater extent in those tissues (5). Pomegranate constituents inhibited prostate cancer cell growth by affecting their proliferation, gene expression, invasion, and apoptosis but had a less profound effect on normal prostate epithelial cells. However, the concentrations at which they exhibited antiproliferation activity against human prostate cancer cell lines were higher than the physiologically available concentrations (18.6 μM when 1 L of juice is consumed for 5 days) (5, 8). These results suggest that there might be some other pathways through which pomegranate constituents exert cancer chemoprevention. To explore the other possible prostate cancer chemopreventive pathways, we studied the effects of pomegranate juice ellagitannins and their microbial metabolites on CYP1B1- induced carcinogenesis. Previously, it was shown that pomegranate juice consumption resulted in lowered total hepatic CYP content and also decreased CYP1A2 and CYP3A. Therefore, the anticarcinogenic effects of pomegranate juice could be partly attributed to their ability to inhibit CYP activity/expression (9). Our work represents the first report
  • 27. concerning the effects of pomegranate ellagitannins on CYP1B1 inhibition as a means for cancer chemoprevention. CYPs are responsible for the bioactivation of endogenous compounds, drugs, dietary chemicals, and xenobiotics. The CYP1 isoforms, CYP1A1, CYP1A2, and CYP1B1, are of major importance because they activate a number of polycyclic aromatic hydrocarbons (PAHs) to genotoxic compounds leading to tumorogenesis (10). CYP1B1 is abundantly expressed extrahepatically in steroidogenic (ovaries, testes, and adrenal glands) and steroid-responsive (breast, uterus, and prostate) tissues. The CYP1B1 enzyme plays an important role not only in the initiation and promotion of cancer but also in the development of drug resistance. The CYP1B1 enzyme alone accounts for activation of 15 PAHs, six heterocyclic amines, and two nitropolycyclic hydrocarbons into mutagenic and carcinogenic compounds, which cause DNA damage and initiate cancer formation. CYP1B1 is also involved in the metabolism of endogenous compounds such as 17β-estradiol to an active metabolite, 4-hydroxyestradiol (4-OH-E2), which has been implicated in breast cancer initiation. In comparison, CYP1A1 converts 17β-estradiol into 2-hydroxyestradiol (2-OH-E2), which is relatively noncarcinogenic as compared to 4-OH- E2 and plays no role in cancer (11). CYP1B1 levels are overexpressed in prostate, lung, esophageal, oral, and colon cancers but not in the corresponding normal tissues. The increased expression of CYP1B1 could generate an excessive number of genotoxic metabolites, which may attack the DNA of normal cells, thus allowing for cancer promotion. Although the augmented CYP1B1 expression does not cause tumor invasion or metastasis, it leads to deactivation of anticancer drugs such as flutamide in prostate cancer treatment and docetaxel in breast cancer treatment (12). Considering the crucial role played by CYP1B1 in cancer inititation, promotion, and resistance development, it is an attractive molecular target for cancer chemoprevention. The expression of the CYP1 family is regulated by the aryl hydrocarbon receptor (AhR). The ligands of AhR range from environmental contaminants to plant- or diet-derived constituents such as curcumin and carotenoids (13). Because CYP1B1 is a therapeutic target in prostate cancer, we hypothesized that pomegranate constituents/metabolites might exert prostate cancer chemoprevention through CYP1B1 inhibition as one of the plausible mechanisms. Our results indicate a previously unexplored pathway through which pomegranate juice constituents may contribute to prostate cancer chemoprevention. Materials and Methods Isolation and Identification of Pomegranate Juice Ellagitannins The extraction of ellagitannins was performed by a procedure described previously, by use of a step gradient consisting of an increasing amount of methanol in water. The commercial POMx (100 mL) was diluted to 500 mL with Millipore purified water and successively partitioned with EtOAc (3 × 200 mL) and n-BuOH (3 × 200 mL). The n-BuOH extract (2.0 g) was concentrated and subjected to Amberlite XAD-16 column chromatography (500 g, 6 cm × 35 cm) and eluted with H2O (2.0 L) and MeOH (2.0 L) successively. The MeOH fraction on removal of solvent under reduced pressure afforded a tannin fraction (XAD-n-BuOH) (1.3 g). This was further purified on Sephadex LH-20 CC (6 cm × 55 cm) and eluted with H2O:MeOH (2:8, 350 mL), H2O:MeOH (1:9, 500 mL), MeOH (450 mL), and MeOH:Me2CO (1:1, 600 mL) to give nine fractions. A follow-up of fractionation and further purification of all of the fractions on Sephadex LH- 20 column chromatography using H2O:MeOH gradient, MeOH, and MeOH:Me2CO gradient system afforded the compounds gallic acid, hexahydroxydiphenic acid (HHDP), gallagic acid, punicalins, and punicalagins. The latter compounds (punicalins and punicalagins) exist in solution as the α- and β-anomers as well as acyclic hydroxyaldehyde analogues (14) (Figure 1). The compounds were identified using LC-
  • 28. MS retention time, UV absorption pattern, molecular mass, and 1H NMR spectra. The LC-MS system consisted of Waters Micromass ZMTQ mass spectrophotometer, Waters 2695 Separation Module, and Waters 996 Photodiode Array Detector. Mass spectra were recorded in negative mode, using a capillary voltage of 4000/3500 V and a gas temperature of 300 °C. The column used was a 150 mm × 3.0 mm i.d., 5 μm, Luna C18 100 Å (Phenomenex, Torrance, CA). The analysis was performed using a 2.5% acetic acid in water (solvent A) and 2.5% acetic acid in methanol (solvent B), starting from 100% A for 5 min, 0−60% B for 15 min, and 60−100% B for the next 15 min. The flow rate was 0.3 mL/min with the pressure set at 900−1500 mmHg. Figure 1. Structures of ellagitannins and urolithins. Synthesis of Urolithins Chemicals Resorcinol, ReagentPlus (99%), 2-bromobenzoic acid (97%), 2-bromo-4,5- dimethoxybenzoic acid (98%), and chlorobenzene were purchased from Sigma Aldrich (St. Louis, MO). 2-Bromo-5-methoxybenzoic acid (98%) was purchased from Alfa Aesar (Ward Hill, MA). Pyrogallol (ACS grade) was purchased from Acros Organics. CuSO4, NaOH, and AlCl3 were purchased from Fisher Scientific (Pittsburgh, PA). Purification of Compounds The high-performance liquid chromatography (HPLC) system consisted of a Waters Delta 600, a Waters 600 controller, a Waters 996 Photodiode Array Detector, and a 3.0 mm × 150 mm column (Phenomenex, ODS 5 μm C18 100 Å). Analyses were performed in the gradient system A, 2.5% aqueous acetic acid, and B, 2.5% acetic acid in methanol, starting from 100% A for 5 min, 0−60% B for 15 min, and 60−100% B for 15 min. The flow rate was 1 mL/min, and the pressure was 600−800 mmHg. The elution of metabolites was monitored at 254 nm. Urolithins (urolithin B, 8-O-methylurolithin A, urolithin A, 8,9-di-O-methylurolithin C, urolithin C, 8,9-di-O-methylurolithin D, and urolithin D) were synthesized by the condensation of resorcinol or pyrogallol with an appropriately substituted benzoic acid by the modified protocols described by Ito et al. (15). The structures of urolithins were confirmed by their molecular mass and comparison of observed and reported 1H NMR data with reported data (Figure 1). Recombinant CYP1 Ethoxyresorufin-O-deethylase (EROD) Assay and Inhibition Kinetics
  • 29. To study the effects of pomegranate chemical constituents and their microbial metabolites on recombinant CYP1A1 and CYP1B1, a 96-well plate EROD assay was used (16). Concentrations of the test compounds ranged from 0.5 to 30 μM. Inhibition kinetics of CYP1B1-mediated EROD activity was determined similarly. Concentrations of 0.5 and 1 μM were used for urolithins A and B in triplicate. 22Rv1 Prostate Cell EROD Assay To evaluate the effects of pomegranate constituents and microbial metabolites in a cell- based CYP1 activity, an EROD assay was conducted using 22Rv1 cells in a 48-well plate format (32). The test compounds were studied for their effects on cell-based 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced CYP1-mediated EROD activity. The cells were treated with TCDD for 24 h to induce CYP1 expression. The cells were also cotreated with compounds, for either 30 min or 24 h, at concentrations ranging from 6.75 to 50 μM. The cells were treated with DMSO, ellagitannins, and urolithins alone to evaluate their ability to induce CYP1 expression in the absence of TCDD. Microsome Preparation Cells were seeded in 150 cm2 culture plates and exposed to various treatments for 24 h. The cells were harvested, washed, and spun down (250g/5 min/22 °C). An appropriate amount of the lysis buffer [10 mM Tris, pH 7.5, 10 mM KCl, and 0.5 mM ethylenediaminetetraacetic acid (EDTA)] was added, and the cells were transferred to the glass tube of a Teflon homogenizer and kept on ice for 10 min. This was followed by the addition of an appropriate amount of homogenization buffer [0.25 M KH2PO4, 0.15 M KCl, 10 mM EDTA, and 0.25 mM phenylmethanesulfonyl fluoride (PMSF)]. The cells were then broken by 12 manual strokes on a tight-fitting Teflon homogenizer. After centrifugation at 15000g/20 min/4 °C, the supernatant was again centrifuged at 105000g/90 min/4 °C. Microsomal pellets were resuspended in microsomal dilution buffer [0.1 M KH2PO4, 20% glycerol, 10 mM EDTA, 0.25 mM PMSF, and 0.1 mM dithiothreitol (DTT)] and stored in aliquots at −80 °C. Western Blotting Microsomal protein (5−10 μg) samples were prepared by heating at 95 °C for 5 min in the sample buffer comprising 0.5 μM Tris HCl (pH 6.8), 10% glycerol, 2% sodium dodecyl sulfate (SDS), 5% mercaptoethanol, and 0.001% bromophenol blue. The samples were resolved by precast criterion SDS-polyacrylamide gel electrophoresis (PAGE) gel (10%) at 200 V for 45−50 min (Bio-Rad Laboratories, Hercules, CA). The proteins were then transferred to a PVDF membrane (Bio-Rad Laboratories) at 100 V for 90 min. Following transfer, the membranes were blocked in blocking buffer for 1 h followed by incubation in CYP1B1 primary antibody (1:1000, antirat CYP1B1 polyclonal antibody, kindly donated by Dr. Thomas R. Sutter). After they were washed, the membranes were incubated in buffer containing the horseradish peroxidase-conjugated secondary antibody (1:30000, antigoat IgG peroxidase conjugate, Sigma Chemicals) for 2 h. The membrane was washed and developed using LumiGLO Reserve chemiluminescent substrate (KPL, Inc., Gaithersburg, MD). The signals were detected using a CCD camera (VersaDoc Imaging System, Bio-Rad Laboratories). Human CYP1B1 supersomes were used to make standard curves of known protein concentrations (0.25−2 pmol). Standard curves were used to quantitate the CYP1 protein amounts in the samples using the Quantity One quantitation software (Bio-Rad Laboratories). Statistical differences between the control and the treated samples were determined using one-way analysis of variance (ANOVA) followed by Newman−Keuls posthoc (p < 0.05) using GraphPad Prism software. Neutral Red Cytotoxicity Assay
  • 30. The assay was performed in 96-well microplates. Cells were seeded at a density of 10000 cells/well and allowed to settle for 30 min at 37 °C. The compounds, diluted appropriately in RPMI-1640 medium, were added to the cells and again incubated for 48 h. The number of viable cells was determined using the neutral red assay procedure (17). Induction of Phase II Conjugating Enzymes Assay The assays were performed according to standard procedures described by Kirlin et al. (18) Cellular Uptake of Urolithins A and B 22Rv1 cells were incubated with 20 μM urolithins A and B in the RPMI-1640 media for 0.5, 6, 12, and 24 h time periods. After incubation, the cells were washed and harvested by trypsinization. The cells were extracted with acidified MeOH. The samples were analyzed by reverse phase HPLC at 288 nm detection. The experiment was done in triplicate. A standard curve of urolithins A and B was prepared from which the amount of urolithin uptake was determined. The amount of urolithin uptake was adjusted to the amount of protein. Results and Discussion Pomegranate fruit consists of two major classes of polyphenols, flavonoids and ellagitannins. The flavonoids include quercetin, kaempferol, and myricetin (19). These flavonoids have been reported to exhibit CYP1 inhibitory activities (16). Pomegranate juice consumption has been found to decrease the expression/activity of total hepatic CYP content (9). Pomegranate juice obtained by hydrostatic pressing of whole fruit predominantly contains ellagitannins. Ellagitannins, with the exception of ellagic acid, have not been previously studied for their CYP1 inhibitory activities. Ellagic acid has been shown to inhibit CYP2A2, 3A1, 2C11, 2B1, 2B2, and 2C6 in rat liver microsomes (20) and also inhibited the CYP1A1-dependent activation of benzo[a]pyrene (BaP) (3). Inhibition of CYP1 protein expression and activity by some flavonoids, such as diosmin, diosmetin, quercetin, kaempferol, and myricetin, was believed to be through AhR antagonism or their effects on the downstream products of AhR signal transduction pathways (21). However, ellagic acid decreased CYP1A1-dependent BaP activity independent of the AhR-responsive element (3). The proposed mechanism of inhibition of CYP1A1-dependent BaP activation involved scavenging of the carcinogen by ellagic acid through chemical binding (22). Therefore, our objective was to study the effects of a selection of pomegranate ellagitannins and urolithins on the inhibition of CYP1- dependent carcinogen activation. The ability of ellagitannins and urolithins to inhibit CYP1 activity was tested in a recombinant CYP1A1- and CYP1B1-dependent EROD assay. The IC50 values for CYP1B1 inhibition ranged from 1.15 ± 0.65 μM for urolithin A to 137 ± 11.08 μM for urolithin D. CYP1A1 IC50 values ranged from 1.5 ± 0.32 μM for punicalins to 2907 ± 168 μM for urolithin D (Table 1, section A). Urolithins exhibited higher selectivity toward CYP1B1 EROD inhibition as compared to CYP1A1, although the selectivity was not significant. The Ki values of CYP1B1 and CYP1A1 depicted in Table 1, section A, indicate that 8-O-methylurolithin A exhibited a 7.5-fold selectivity toward CYP1B1 inhibition. Punicalins and punicalgins were 10- and 5-fold more selective toward CYP1A1 inhibition. Table 1. Results of Recombinant CYP1-Mediated EROD Assay and Kinetic Parametersa
  • 31. Section A CYP1B1 CYP1A1 IC50 ± SEM Ki ± SEM IC50 ± SEM Ki ± SEM Ki(CYP1A1:CYP1B1) (μM) (μM) (μM) (μM) UA 1.15 ± 0.65 0.25 ± 0.14 12.4 ± 4.7 0.51 ± 0.18 2.05 UB 1.55 ± 0.49 0.34 ± 0.17 26.8 ± 12.9 1.11 ± 0.53 3.28 UC 39.9 ± 29 8.74 ± 0.65 790 ± 75 32.7 ± 3.11 3.74 UD 137 ± 11.08 30.10 ± 25 2907 ± 168 120.3 ± 69.6 3.99 MUA 1.49 ± 0.39 0.327 ± 0.08 59.8 ± 8.7 2.47 ± 0.36 7.57 DMUC 89.6 ± 9.7 19.6 ± 2.13 657 ± 74.3 27.2 ± 3.00 1.39 PL 2.82 ± 0.33 0.618 ± 0.07 1.5 ± 0.32 0.062 ± 0.00 0.10 PG 2.6 ± 0.79 0.58 ± 0.02 2.67 ± 0.48 0.109 ± 0.20 0.19 Section B urolithin A urolithin B DMSO 0.5 μM 1 μM DMSO 0.5 μM 1 μM Vmax 339 ± 148 97.3 ± 14.6 66 ± 6.02 355 ± 103 150.9 ± 14.9 99.5 ± 8.53 Km 3.414 ± 2 2 ± 0.47 1.6 ± 0.25 4.78 ± 1.81 2.10 ± 0.33 1.9 ± 0.26 IC50 and Ki values (±SEM) and the ratio of CYP1A1 to CYP1B1 Ki for urolithins A (UA), B (UB), C (UC), D (UD), 8-O-methylurolithin A (MUA), 8,9-di-O-methylurolithin C (DMUC), punicalins (PL), and punicalagins (PG) mediated inhibition of EROD acitivity using recombinant human CYP1B1 and CYP1A1 enzymes. For section B, kinetic parameters, Vmax (pmol/mg/min), and Km (μM) ± standard errors (n = 3) for the inhibition of recombinant human CYP1B1 by urolithin A and B (0.5 and 1 μM), determined by nonlinear regression curve fit using the Michaelis−Menten equation ([S] vs V) plot using GraphPad Prism. Urolithins A and B are the major microbial metabolites of pomegranate chemical constituents detected in human systemic circulation. These metabolites exhibited lower IC50 values in recombinant CYP1 inhibition as compared to all other tested compounds. Therefore, a study was conducted to investigate the mechanism of action of CYP1B1 inhibition by urolithins A and B. The concentrations of inhibitors used were in the vicinity of their calculated IC50 values, that is, 0.5 and 1 μM. EROD activities were determined with substrate concentrations ranging from 0.1 to 2.0 μM. Kinetic parameters, Vmax and Km, were calculated using the Michaelis−Menten equation ([S] vs V curve). Double reciprocal plots were plotted using 1/[S] and 1/V (Figure 2) from which Ki values were calculated (Table 1, section B). The calculated Ki values for urolihins A and B (1.51 ± 0.91 and 1.33 ± 0.08 μM) were not statistically different from those calculated by using Cheng−Prusoff equations (16). The calculated Vmax and Km for urolithin A changed significantly with an increasing concentration of inhibitor, suggesting an uncompetitive type of inhibition. However, the Vmax and Km of urolithin B did not differ significantly upon increase of inhibitor concentration, suggesting a noncompetitive type of inhibition.
  • 32. Figure 2. Double reciprocal plots for the inhibition of in vitro EROD activity of CYP1B1 by urolithins A and B at 0.5 and 1 μM. EROD substrate concentrations used were 0.1, 0.2, 0.3, 0.4, 0.6, 1.0, and 2.0 μM. Recombinant CYP1B1 was preincubated with the inhibitor or DMSO prior to initiation of reaction. Each experiment was done three times in duplicate. In our study, urolithins A and B, structural analogues of ellagic acid, exhibited a significant inhibition of CYP1-dependent EROD activity. The results suggested that a hydroxy group at C-8 and C-3 (urolithins A and B), corresponding to the C-4 and C-4′ position of ellagic acid, were required for full CYP1-dependent EROD activity inhibition in accordance with a previous study by Barch et al. (7). However, in our study, additional hydroxy groups at C-4 and C-9 of the urolithin pharmacophore (urolithins C and D) resulted in decreased CYP1-dependent EROD inhibitory activity. Methylation of the hydroxy groups to give 8-O-methylurolithin A and 8,9-di-O-methylurolithin C decreased the activity, suggesting that the phenolic hydroxy groups are important for CYP1 inhibitory activity. To probe the importance of the lactone group for CYP1-dependent EROD activity inhibition, HHDP and gallic acid were tested for their CYP1-dependent EROD activity. The results showed that hydrolysis of the lactone functionality did not result in CYP1 inhibition (data not shown).
  • 33. The punicalins and punicalagins were potent inhibitors of CYP1-dependent EROD activity with Ki values (Table 1, section A) comparable to the dietary flavonoids. The dietary flavonoids, inhibiting CYP1-dependent metabolic activation of procarcinogens, include quercetin, kaempferol, apigenin, myricetin, and rutin. Quercetin and kaempferol, which are the predominant flavonoids in the human diet, inhibited CYP1-mediated EROD activities. The apparent Ki values of inhibition of human recombinant CYP1B1 and CYP1A1 were 14 ± 3 and 52 ± 2 nM for kaempferol, whereas they were 23 ± 2 and 77 ± 5 nM for quercetin (23). In a different study, quercetin inhibited epoxidation of 7,8- dihydro-7,8-benzo[a]pyrene-7,8-diol by CYP1A1 allelic variants with Ki values ranging from 2.0 to 9.3 μM with mixed type inhibition (24). In another study, quercetin inhibited recombinant CYP1A1 and CYP1B1 activities with Ki values of 0.25 ± 0.04 and 0.12 ± 0.02 μM with a mixed type inhibition (16). The bioavailability of the flavonoids depends upon the source of food; for example, quercetin absorption from tomato puree, apples, and onions was 0.082, 0.34, and 0.74 μM, respectively (25, 26). Kaempferol plasma concentrations ranged from 0.01, 0.05, and 0.1 μM upon consumption of onion, tea, and endive, respectively (27, 28). Bioavailable concentrations of quercetin and kaempferol from some food sources were less than their reported Ki values of CYP1 inhibition. Apigenin, a flavone present in parsley, exhibits CYP1B1 and CYP1A1 inhibition with Ki values ranging from 60 nM to 0.2 μM. These concentrations are bioavailable (127 ± 81 nM) upon consumption of 2 g of blanched parsley (14). Bioavailability of rutin from tomato puree as detected in plasma was calculated to be 0.1 μM (25), which was around 60-fold lower than the reported Ki of CYP1B1 inhibition. The bioavailability of many other flavonoids is still unclear. However, dietary flavonoids could inhibit CYP1-mediated bioactivation of environmental and dietary carcinogens into genotoxic compounds and prevent cancer initiation in alimentary canal-related cancers because they come into direct contact with the digestive epithelium of the digestive system (29). However, the prostate cancer chemopreventive effects of flavonoids (30) depending on the bioavailability are still debated. It is therefore important to choose an appropriate dietary
  • 34. supplement that can release adequate amounts of cancer chemopreventive compounds into plasma, whenever it is consumed, with an intended pharmacological activity. Pomegranate juice ellagitannins have been extensively studied for their bioavailability and their biological effects. In one study, it was established that the punicalagins hydrolyze into ellagic acid and other smaller polyphenols that are responsible for the bioactivity of ellagitannins. In a study performed on human subjects, consumption of 180 mL of pomegranate juice (equivalent to 25 mg of ellagic acid and 319 mg of punicalagins) resulted in detection of ellagic acid in plasma with a maximum concentration of 31.9 ng/mL (0.1 μM) (31). Previous studies also indicated that the bioavailability of ellagic acid from pomegranate juice, pomegranate liquid concentrate extract, and pomegranate powder extract was not statistically different (32). In our study, punicalins and punicalagins inhibited CYP1A1 with Ki values of 0.062−0.109 μM and CYP1B1 with Ki values 0.618 and 0.58 μM, respectively. Punicalins and punicalagins exhibited approximately a 10- and 5-fold selectivity for CYP1A1 over CYP1B1. Thus, consumption of pomegranate juice could be beneficial in decreasing CYP1-mediated oral, esophageal, and colon cancers. However, these ellagitannins cannot exhibit a systemic CYP1 inhibition activity because they are metabolized in the colon by microflora into smaller organic molecules called urolithins. Bioavailability studies indicate that maximum plasma concentrations of urolithins A and B reach concentrations of 4−18 μM in human subjects (8, 32). Urolithins A and B inhibited human recombinant CYP1A1 and CYP1B1 with Ki values in their bioavailable concentration range. Our particular interest was to explore the beneficial effects of urolithins in prostate cancer. Studies indicate that 15% of prostate cancer patients, who have undergone a radical prostectomy, had a biochemical recurrence of prostate- specific antigen (PSA). Among them, 34% of patients developed distant metastases within 15 years (33). It was evident that consumption of pomegranate juice delayed the doubling time of the PSA by 39 months after primary therapy (7). The effects were ascribed to the antiproliferative, apoptotic, and antioxidant effects of pomegranate constituents, observed in LNCaP, PC-3, 22Rv1, and DU 145 prostate cancer cell lines (8, 34). A study about gene polymorphisms and risk of prostate cancer showed that polymorphisms in CYP1B1 and PSA genes increased the risk and aggressiveness of prostate cancer (35). Any dietary constituent with CYP1B1 inhibitory activity could potentially lead to prostate cancer chemoprevention. There were no previous reports about the ability of pomegranate constituents/metabolites to inhibit CYP1B1-dependent carcinogenesis. Therefore, we studied the capability of pomegranate constituents to inhibit CYP1B1- induced metabolic activation in a prostate cancer cell line, 22Rv1. The cells were treated with TCDD for 24 h to induce CYP1B1 protein expression. Then, the cells were treated with punicalins, punicalagins, or urolithins for 30 min. Following 30 min of incubation, urolithins A and B significantly decreased TCDD-induced EROD activity at the highest concentration used (Figure 3A). The IC50 values calculated for cell-based CYP1 inhibition by urolithins A and B were 32 ± 8.9 and 38.2 ± 3.94 μM, respectively (Table 2). The results indicate a 28- and 26-fold increase in IC50 values for cell-based CYP1- mediated EROD activity of urolithins A and B, respectively, as compared to their in vitro recombinant CYP1-mediated EROD inhibitory IC50. Punicalins and punicalagins did not inhibit cell-based CYP1-mediated EROD activity at the highest concentration used (50 μM). To ascertain whether a decrease in IC50 occurred upon longer incubation, the cells were allowed to grow in the presence of compounds and TCDD for 24 h (Figure 3B). The EROD activity results indicated that the compounds more effectively inhibited CYP1-mediated EROD activity and had IC50 values lower than those following 30 min of
  • 35. incubation. After 24 h of cotreatment, urolithins A and B inhibited TCDD-induced EROD activity in prostate cells with IC50 values of 13.3 ± 1.32 and 17.9 ± 1.8 μM, respectively, which were in the vicinity of their bioavailability (8, 32). Punicalins and punicalagins did not exhibit EROD inhibition even upon 24 h of incubation. Urolithin C, 8-O- methylurolithin A, and 8,9-di-O-methyl urolithin C demonstrated IC50 values of 26.8 ± 2.5, 14.8 ± 2.24, and 11.5 ± 2.8 μM, respectively, which were lower than those of urolithins A and B (Table 2). The compounds alone did not induce EROD activity after 24 h of incubation. Because the prostate cells were treated with the test compounds for 24 h, it was imperative to investigate if the compounds exhibited any cytotoxicity. A neutral red dye uptake assay was used to measure the cytotoxicity of the test compounds. The data were probit transformed followed by linear regression and IC50 calculation. The cytotoxic IC50 values ranged from 20.6 ± 4.58 μM for 8,9-di-O-methylurolithin C to 108 ± 3.994 μM for urolithin B (Table 3). The results indicate that the cytotoxicities of urolithin A and B had no contribution toward decreased CYP1-mediated EROD activity. The results also indicate that urolithin C, 8,9-di-O-methylurolithin C, and 8-O-methylurolithin A were false positives in the prostate cell EROD assay. The activity was not because of CYP1 inhibition but due to cytotoxicity. This conclusion was verified based on four facts: (1) These compounds inhibited recombinant CYP1-mediated EROD activity at higher concentrations as compared to urolithins A and B; (2) these compounds did not inhibit prostate cell EROD activity upon 30 min of incubation; (3) they inhibited prostate cell EROD activity upon 24 h of treatment, with IC50 values lower than those exhibited in the recombinant CYP1-mediated EROD assay; and (4) they exhibited cytotoxicity in the vicinity of their prostate cell EROD inhibition IC50 values. Figure 3. Effects of increasing concentration (6.75, 12.5, 25, and 50 μM) of urolithin A (UA), urolithin B (UB), urolithin C (UC), 8-O-methylurolithin A (MUA), 8,9-di-O- methylurolithin C (DMUC), punicalins (PL), and punicalagins (PG) on 50 nM TCDD- induced EROD activity in intact 22Rv1 human prostate cancer cells. The cells were exposed to the compounds for (A) 30 min or (B) 24 h prior to the EROD measurement.
  • 36. Each experiment was done three times in triplicate. Global one-way (ANOVA) with a Student−Newman−Keuls posthoc test was used to determine treatment effects. In the same treatment groups, bars with different letters are statistically different. Table 2. IC50 Values for Pomegranate Chemical Constituents/Microbial Metabolite Mediated Inhibition of EROD Activity in TCDD-Induced 22Rv1 Prostate Cancer Cells IC50 ± SEM (μM) chemical constituent 30 min 24 h urolithin A 32 ± 8.9 13.3 ± 1.3 urolithin B 38 ± 3.9 17.9 ± 1.8 urolithin C NA 26.89 ± 2.5 8-O-methylurolithin A NA 14.8 ± 2.2 8,9-di-O-methylurolithin C NA 11.5 ± 2.8 punicalins NA NA punicalagins NA NA Table 3. IC50 Values for Urolithin-Mediated Cytotoxicity of 22Rv1 Prostate Cells chemical constituents IC50 ± SEM (μM) urolithin A 98.7 ± 4.2 (a) urolithin B 108 ± 3.9 (a) urolithin C 36 ± 3.5 (b) 8-O-methylurolithin A 23.3 ± 3.9 (c) 8,9-di-O-methylurolithin C 20.6 ± 4.6 (c) Following cell-based EROD assays, cellular uptake experiments were performed for urolithins A and B. These experiments were performed to determine if the decrease in IC50 values for CYP1-mediated EROD inhibition in 22Rv1 cells was related to increased uptake over a 24 h time period. The experiments indicated that there was a 4.5-fold increase in urolithin uptake upon 24 h of incubation as compared to 30 min of incubation. The results also indicated that there was a dramatic increase in urolithin uptake between 6 and 12 h, beyond which the uptake was constant. The results suggested that increased availability of urolithins (from 30 min to 24 h) could have contributed to the decrease in IC50 of CYP1 inhibition (Table 4). The results also indicated that urolithins were metabolically stable in 22Rv1 cells for up to 24 h. Table 4. Uptake of Urolithins A and B by 22Rv1 Cells over a Period of 0.5−24 h uptake in μmol/mg protein time (h) urolithin A urolithin B 0.5 5.2 ± 0.4 5.7 ± 0.8 6 9.6 ± 0.4 8.0 ± 1.1 12 20.1 ± 0.5 21.9 ± 1.1 24 23.9 ± 0.6 25.2 ± 1.6 The decrease in IC50 values could also be attributed to the decrease in the TCDD- induced CYP1B1 protein expression. To examine if the treatment affected CYP1 protein expression levels, Western blots were performed (Figure 4). Cell treatments were DMSO, TCDD (50 nM), UA (50 μM), UB (50 μM), TCDD + UA (50 μM), TCDD + UB (50 μM), TCDD + UA (25 μM), and TCDD + UB (25 μM). CYP1B1 protein levels were
  • 37. significantly increased ( 4.5-fold) by TCDD (2.8 ± 0.6 pmol/mg) as compared to DMSO (0.62 ± 0.28 pmol/mg). Cotreatment of TCDD with urolithin A (50 μM) significantly decreased CYP1B1 protein (1.26 ± 0.38 pmol/mg) production by 54%, while urolithin A (25 μM) decreased CYP1B1 protein (2.00 ± 0.5 pmol/mg) production by 28% as compared to TCDD-induced cells. Cotreatment of TCDD with urolithin B (50 μM) decreased CYP1B1 protein (0.96 ± 0.4 pmol/mg) production by 65%, and urolithin B (25 μM) treatment decreased protein (1.9 ± 0.5 pmol/mg) production by 29% as compared to TCDD-induced levels. Western blot analyses suggest that none of the urolithins induce CYP1B1 basal levels significantly as compared to DMSO. However, cotreatments with urolithins A and B 50 μM and TCDD decreased CYP1B1 protein expression levels as compared to TCDD alone (Figure 4). Figure 4. (A) Western blot showing the effect of urolithins A and B and their cotreatment with TCDD on CYP1B1 protein expression. Microsomes were prepared after treating cells for 24 h with different treatments (DMSO, 50 nM TCDD, UA 50 μM, UB 50 μM, TCDD + UA 50 μM, TCDD + UA 25 μM, TCDD + UB 50 μM, and TCDD + UB 25 μM), along with standards, were loaded on 10% SDS-PAGE. Protein levels were expressed in pmol/mg. (B) Effect of urolithin A and B on the TCDD-induced CYP1B1 protein expression in 22Rv1 prostate cancer cells. Each experiment was repeated three times. Global one- way ANOVA with a Student−Newman−Keuls posthoc test was used to determine treatment effects. Bars with different letters are statistically different. Urolithins A and B inhibited CYP1 EROD activity by inhibiting both the protein expression and the activity of CYP1B1. While the influence of urolithin C, 8,9-di-O- methylurolithin C, and 8-O-methylurolithin A on CYP1B1 activity/expression was not clear, they are believed to exert an antiproliferative activity on prostate cancer cells, in accordance with previous studies (32, 34). An ideal anticarcinogenic agent would inhibit phase I enzymes, involved in carcinogen activation while inducing the phase II enzymes, responsible for the deactivation of carcinogens by assisting their excretion via increased water solubility. The pomegranate ellagitannins and urolithins were tested for their capacity to induce glutathione S-transferase and quinone O-reductase enzymes. The basal levels of quinone O-reductase and glutathione S-transferase enzymes in 22Rv1 cells were determined to be 1.2 ± 0.32 and 0.51 ± 0.17 μmol min−1 mg protein−1, respectively. However, none of the compounds exhibited (data not shown) induction of quinone O-reductase or glutathione S-transferase as compared to normal proliferating cells.