Open access

Natural Compounds, Antioxidant and Antiandrogens in the Prevention of Prostate Cancer: In vivo Evidences from Murine Models and Human Clinical Studies

Written By

Rossano Lattanzio, Alessia Lamolinara, Mauro Piantelli and Manuela Iezzi

Submitted: 03 May 2012 Published: 16 January 2013

DOI: 10.5772/52292

From the Edited Volume

Advances in Prostate Cancer

Edited by Gerhard Hamilton

Chapter metrics overview

1,997 Chapter Downloads

View Full Metrics

1. Introduction

Prostate cancer (PCa)is the most frequent malignant neoplasia in men. The number of cases has continuously increased over the past decades, partly due to the higher life expectancy. Additional factors are the high caloric diet and lack of physical exercise, typically seen in the Western countries. Notably, up to 40% of cancer incidents are preventable by consuming a healthy diet, regular physical activity, and maintenance of optimum body weight, and more than 20% by consuming vegetables and fruits. PCa represents an ideal candidate disease for chemoprevention. It is typically diagnosed in elderly men and even a modest delay in the neoplastic development could result in substantial reduction in the incidence of the clinically detectable disease.In this chapter we will review the history, the development, and the applications of some of the most common animal models of PCa,and we will discuss of the role of animal models in translational research.

Advertisement

2. Body

Prostate cancer (PCa) is the most common non-cutaneous malignant neoplasm in men in Western countries, responsible for the deaths of approximately 30,000 and 85,000 men per year in the United States and Europe, respectively [1,2].The number of cases is increasing rapidly in step with the growing number of men >50 worldwide, strategies for the prevention of PCa and its progression are urgently required. Since studies of chemopreventive agents in humans are hampered by the long latency period and challenging epidemiological problems, reliable preclinical models can be useful to overcome these problems. Early prostate tumorigenesis is apparently characterised by dysplasia that starts with proliferative inflammatory atrophy as the prelude to low-grade Prostatic Intraepithelial Neoplasia (PIN), high-grade PIN, primary cancer, metastatic cancer, and hormone-refractory cancer. During this progression, genetic damage accumulates within cancer cells [3,4]. Animal modelling has made a significant contribution to the study of prostate development and disease. Identification of the molecular features of PCa pathogenesis and progression could be greatly facilitated by laboratory and clinical models. However, a prerequisite for the elaboration of useful models is a better understanding of the molecular characteristics of human PCa. This puzzle, in addition to the well-known inter- and intra-individual heterogeneity of the disease itself and its multi-faceted nature, has necessitated the development of several complementary model systems. The most effective animal models will be those that most closely mimic the phenotypic and genetic changes accompanying the progression of the human disease. Systems shown to be promising include the dog, the rat, the human xenograft, and the genetically manipulated mouse. They have been widely employed to test preventive regimens, combinations of chemopreventive agents and/or drugs, cancer vaccines, and targeted treatments [5-12].This paper reviews the history, development, and applications of some of the most common animal models, and discusses their pros and cons in translational research.

Advertisement

3. Canine models

The dog is the animal known to commonly develop high-grade PIN and PCa spontaneously in a human-like manner [13]. The many similarities between the canine and the human form include the morphologic and phenotypic heterogeneity of the tumoral lesions, the age-dependency of tumor occurrence, and the propensity to metastasize to bones in an osteoblastic manner [14,15]. Androgen-dependency, on the other hand, is ruled out by a similar incidence in castrated animals [15], while a relatively long latency, the low incidence of spontaneous disease, the impracticability of genetic manipulation, and the high expense of maintaining dog colonies [16,17] are other limitations of canine systems.

Advertisement

4. Rat models

Spontaneous PCa is sometimes observed in some strains of rats [18]. The Dunning model [19] is the most popular. The original R-3327 tumor arose spontaneously in an inbred Copenhagen rat, and was translated into a syngenic Copenhagen x Fisher F1 rat. It is a slow growing, well differentiated and non-metastatic form. Several sublines with different characteristics mimicking some aspects of the human disease have since been developed [20-23]. Copenhagen and Wistar rats also develop a wide range of PCa phenotypes [24,25]. This variability, however, coupled with the rarity and long latency of these tumors, and their lack of metastases, bar the realistic employment of such models [12], though the recent elaboration of knockout methods [26-28] indicates that greater use could be made of genetically engineered rats in the future [29].

Advertisement

5. Xenograft models

In immunodeficient nude mice tumors grow after injection of cancer cells or xenograft implantation with no evidence of a graft-versus-host response. In function of the number of cells injected, or the size of the xenograft, the tumor will develop over 1–8 weeks, 1–4 months, or longer, and its response to treatment can be studied [30]. By comparison with in vitro studies, this approach offers several advantages, especially a 3D structure complete with tumor-induced angiogenesis, hormonal, paracrine/autocrine factors, and metastasis [12]. Xenografting of human PCa began in the 1970s [31]. Thereafter several cell lines that displayed different PCa phenotypes when injected into athymic nude mice have been developed [32,33]. This model has been used to show the ability of tumor xenografts to metastasize to the lymph node and bone, the two most common human sites [34].

Mice with an autosomal recessive Severe Combined Immuno Deficiency mutation (SCID mice) were identified in 1983 [35]. This mutation results in a lack of T- and B-lymphocyte function. However, normal natural killer (NK) cells and myeloid function are present, and in some SCID mice, some B and T cells are still present [36]. In this model subcutaneous injection of HER2/neu overexpressing human CLNCaP cells has shown that HER2/neu induces androgen-independent tumor growth through modulation of the androgen receptor signalling pathway[37].

In 1995, the features of this model were improved by crossing SCID mice with nonobese diabetic (NOD) mice, which lack in NK cells, antigen-presenting cells, and circulating complement [38]. NOD-SCID mice accepted foreign tissue more successfully and were more immunodeficient than SCID mice. This strain has been used to elaborate a model for orthotopic implantation of PC-3 and DU145 cells with a tumor take efficacy of >80% for both lines [39]. Some xenograft models result in metastasis to bone after intracardiac injection of bone cells that probably survive in a niche whose microenvironment is optimal for their seeding and growth. However intracardiac injection is not an ideal procedure and attention has thus been focused on xenografts to orthotopic sites such as the prostate. The success rates depend on the host strain and the use of hormones or Matrigel to provide adequate growth factors and a scaffold for cell growth [40-42].

The immunodeficiency mouse model has been further improved by crossing NOD-SCID mice with interleukin-2 receptor gamma null mice (NOG/NSG mice). These long-living mice (median 90 weeks) totally lack B, T, and NK cell activities, and cytokine signaling, together with no age-related “leakiness”. They have a higher xenograft success rate and are more effective than other models, particularly in long-term studies involving prostate and non prostate cancer cells [43-45].

For preclinical prostate studies, most laboratories employ human PCa cell lines xenografted in mice. Many excellent reviews of the characteristics of these lines have been published [46-50]. The most widely used, each with thousands of studies published according to PubMed, are the classic three lines PC-3, LNCaP, and DU145, while each of the other lines has less than 200 citations [8]. These cell lines do not represent the steps of PCa progression. For example, almost all cell lines, including the most popular, were obtained from metastatic deposits: PC-3 from bone, LNCaP from lymph node, and DU145 from dural metastasis. In addition, PC-3 and DU145 are androgen receptor (AR) negative and LNCaP expresses a mutated AR. Again, cell lines, and their sublines in particular, are not fully genetically, functionally and phenotypically characterized, nor is there a method for standardization [8,46-48].

Advertisement

6. Transgenic mouse models

The last ten years have witnessed a remarkable shift in animal-based cancer research from xenograftedtumor to transgenic models since it is believed that they will recapitulate the complete course of carcinogenesis more accurately[48].This assumption stems from the recognition of several advantages that transgenic models offer when compared to xenograft systems. Among these are that the process of carcinogenesis begins with normal cells, progresses through distinct genetic and histological stages, occurs in an immuno-competent host and in its own cellular microenvironment, and that metastasis can occur along routes and to sites relevant to the clinical disease. A perhaps unrecognized attribute lies in the fact that, because the disease is not initiated by human action but by a genetic program that passes through the germline, the disease process is ‘‘reset’’ each generation. Statistically, the progression of a transgenic model of cancer should therefore be precisely recapitulated across time and between colonies. Given appropriate record keeping and data analysis, this feature should allow epidemiological- style investigations of great statistical power, free from both the mathematical noise of genetic and environmental variation, and from many of the economic and ethical constraints of human medicine.

Genetically engineered mouse (GEM) models have been utilized to identify pathways involved in carcinogenesis and investigate the role of particular gene mutations/deletions, and validate key genes as therapeutic targets. These models have been widely employed to test preventive regimens, combinations of chemopreventive agents and/or drugs, cancer vaccines, and targeted PCa treatments [5-12]. To mimic the human disease, GEMs could be generated through several mechanisms, such as overexpression or activation of oncogenes, elimination of target suppressor genes (Knock-outs), or generating dominant negative proteins that disrupt the function of regulatory genes.

The methods initially reported for genetic mouse modification involved the introduction of DNA constructs designed to induce the expression of proteins under the control of strong tissue-specific promoters, such as probasin and PSA. Simian virus 40 (SV40) large T antigens (Tag) were widely used because of their transforming ability. They interact with and suppress the tumor suppressor protein p53 and retinoblastoma [51,52]. In addition, the small t antigen interacts with the serine/threonine-specific protein phosphatase 2α to induce transformation [53].

The first model involving the expression of SV40 tumor antigens to develop PCa in the mouse was the C3(1)-Tag model[54].Targeting the Tag expression to the prostate was achieved by using a region of the C3 (1) gene, the rat prostatic steroid binding protein gene. Most C3(1)-Tag mice developed PIN after about eight weeks of age. Invasive adenocarcinomas followed after 28 weeks in about 40%. These tumors rarely metastasized (<4%), and always to the lungs. However, SV 40 expression was also detected in the mammary and salivary gland, while all females develop mammary intraepithelial neoplasia that may progress to mammary carcinomas[55].More effective prostate targeting was obtained in later models. Relatively few studies have used the C3 (1)/Tag model.

The transgenic adenocarcinoma of the mouse prostate (TRAMP) mouse [56,57] is the best known and most widely used PCa model because it closely mimics the human disease.In this model, expression of both large and small SV40 early genes (T and t antigen, Tag) are driven by the prostate-specific promoter probasin that leads to cell transformation within the prostate. In this model, Tag are under the control of the minimal rat probasin –426/+C28 fragment. All male TRAMP mice develop PCa spontaneously: as in humans, they develop PIN, and well- or moderately- differentiated adenocarcinomas (between 10 and 20 weeks of age) and undifferentiated carcinomas (expressing or not AR) as well as phyllode tumors in the seminal vesicles [58,59]. Most adenocarcinomas arose in the dorsolateral lobe, which is considered most analogous to the peripheral zone where the human disease originates [10]. TRAMP was the first mouse model to display distant organ metastases, albeit rarely to the skeleton. Metastatic progression can be observed after 28 weeks of age, when almost all mice display lymphatic and >60% lung metastases from AR-, poorly differentiated (PD) tumors that constitute the main “lethal phenotype” in the TRAMP mouse on account of their fast growth and consequent acute renal damage due to compression, and also because they are the source of distant metastases and systemic cachexia [60]. These phenomena can also occur in the absence of other physiologic sequelae of metastatic disease [61]. An issue with the TRAMP model is that its most frequent lethal and metastatic malignancy (i.e. the PD tumor), has been reported to be of neuroendocrine nature and origin, while the simultaneous loss of p53 and Rb could increase susceptibility to neuroendocrine cancer [62-64].

The TRAMP mouse has become a popular preclinical model for studying chemoprevention/treatment of PCa, and elucidation of the antitumorigenic effects of many classes of chemopreventive/therapeutic regimens, including anti-androgen, anti-estrogen, anti-angiogenic, ornithine decarboxylase inhibitors, green tea polyphenols, COX-2 inhibitors, phytoestrogens, retinoic acid, grape seed extract, flavonolignans, etc (Table 1).This model enables comparison of the efficacy of treatments. A significant decrease of incidence and a delay of tumor progression was observed following anti angiogenic treatment (endostatin and angiostatin gene therapy), and lycopene and tomato supplementation. Other promising anti-oxidant agents include green tea, soy, resveratrol, crucifers, curcumin, tocotrienols, triterpenoids and methyl-selenium.

Regimen Compound Reference Year
Anti-androgen Flutamide 108 2000
Ornithine decarboxylase inhibition alpha-difluoromethylornithine 109 2000
Green tea Polyphenolic extract 110 2001
Soy Genistein 111 2001
Anti-estrogen Toremifene 112 2002
Anti-inflammatory Celecoxib 113 2004
Anti-inflammatory Celecoxib, exisulind 114 2004
Soy Genistein 115 2004
Differentiative, antiangiogenic Retinoic acid 116 2004
Green tea Polyphenolic extract 117 2004
Green tea Epigallocatechin-3-gallate (EGCG) 118 2004
Green tea Polyphenolic extract 119 2004
Green tea Polyphenolic extract 120 2005
Anti-inflammatory Etodolac 121 2005
Block of the α1-adrenergic receptors Doxazosin 122 2005
Rye Bran 123 2005
Soy Genistein 124 2005
Anti-inflammatory Celecoxib 125 2006
Anti-oxidative Spinach extract, EGCG, acetylcysteine 126 2006
DNA methyltransferase inhibition 5-aza-2’-deoxycytidine 127 2006
Estrogen metabolite 2-Methoxyestradiol 128 2006
Grape seeds Polyphenolicextracy 129 2007
Anti-β-Catenin Apigenin 130 2007
Soy Genistein 131 2007
Anti-angiogenic Endostatin and angiostatin gene therapy 132 2007
Green tea Epigallocatechin-3-gallate (EGCG) 133 2007
Milk thistle(Silybummarianum) seeds Silibin 134 2007
Combined immunoprophylaxis Allogeneic cells and recombinant IL-12 135 2007
Saw palmetto Liposterolic extract 136 2007
Grape Resveratrol 137 2007
Plant flavonoid Apigenin 138 2007
Milk thistle(Silybummarianum) seeds Silibin 139 2008
Milk thistle(Silybummarianum) seeds Silibin 140 2008
Cruciferous vegetables Sulphoraphane 141 2009
Green tea Polyphenolic extract 142 2009
Milk thistle(Silybummarianum) seeds Silibin 143 2009
Anti-oxidative γ-Tocopherol 144 2009
Systemic buffers 145 2012
Anti-oxidative γ-Tocopherol 146 2012
Anti-inflammatory Ursolic acid 147 2012
High-fat diet Whole walnuts 148 2012
Pomegranate Fruit exctract 149 2012
Plant flavonoid Apigenin 150 2012
Cancer therapy Docetaxel, Dexametasone, Octeotride 151 2012
Bitter melon Fruit exctract 152 2011
Diet Folate deficiency 153 2011
Anti-inflammatory Ursolic acid 154 2011
Anti-inflammatory + anti-hormonal Celecoxib, Hormone ablation 155 2011
Garlic Diallyltrisulfide 156 2011
Anti-oxidative Indolole-3-carbinole 157 2011
Anti-oxidative Whole tomatoes 158 2010
Anti-oxidative Lycopene beadlet, tomato paste 159 2010
Diet Western diet 160 2010
Anti-oxidative Seleniun 161 2011
Triterpenoids Synthetic CDDO 162 2011
Mitocondrial Hsp90 inhibition 163 2011
Arginine metabolism Modulators 164 2011
Anti-oxidative Methyl-seleniun 165 2009
Hormonal Methoxyestradiol 166 2009
Interferon-alpha 167 2009
3,3’-Diindolylmethane 168 2010
Anti-oxidative Mixed tocotrienols 169 2010
Diet Zinc 170 2010
Cancer therapy Treatment targeting HIF-a and Stat3 171 2011
Crucifers Indole-3-carbinol 172 2011

Table 1.

Preventive/Therapeutic Regimens Tested in the TRAMP Model of Prostate Cancer

To increase the transgene expression beyond that obtained with the minima probasin promoter, as in the TRAMP mouse, an 11.5 kb 5’ flanking fragment of the prostate-specific probasin promoter (large probasin) has since been isolated [65], and used to direct large T-antigen expression to the dorsolateral and ventral prostate (Lady mouse model). The second key difference in this model is that the large probasin promoter was linked to a deletion mutant of the SV40 T-antigen that expressed only the large T-antigen [66,67]. The Lady model is advantageous because expression is high, but the PCa progression is less aggressive, beginning with low to high-grade PIN and proceeding to carcinoma with neuroendocrine features. However, metastatic progression was not seen [5,67]. Several other trangenic mouse models have been developed with or without the involvement of SV40 antigens and with different strategies (reviewed in ref. [12]). In summary, while T antigen expression generally induces castration-resistant, aggressive and metastatic PCas, often with a neuroendocrine phenotype, the specific expression of other oncogenes in the prostate results in a mild phenotype that rarely progresses to adenocarcinoma.

Advertisement

7. Knockout mice

7.1. Whole body models

The roles of genes significant in prostate carcinogenesis can also be studied in, whole-body knockout models. Here, however, the gene involved is knocked out ubiquitously, and its specific role in a given organ cannot be readily determined. Estrogen receptor b knockout mice display hyperplastic foci in the prostate or even no pathological changes [68]. Deletion of retinoic acid receptor γ determines squamous metaplasia of prostate and seminal vesicles, but not carcinomas [69]. p27knockout mouse display prostatic hyperplasia histologically similar to that observed in human BPH, but not PIN, and a pathogenetic role of p27 loss in BPH development in both mice and humans has been suggested [70]. Inactivation of T (phosphatase and tensin homolog deleted on chromosome 10) prevents activation of AKT and apoptosis resulting in embryonic lethality. However, haploinsufficiency leads to early stages (PIN) of prostatic carcinogenesis [71]. Double-knockout models in which loss of PTEN is associated with loss of other tumor suppressors (p27, Nkx3.1, and p53), are characterized by more aggressive tumor phenotype.The highest stage of tumor progression was adenocarcinoma (PTEN x p27 mouse) [72], lymph node metastases (PTEN x Nkx3.1 mouse) [73], and high grade PIN (PTEN x p53 mouse) [74]. In addition, several mouse models with up to 5 genetic hits demonstrated, as expected, the complexity of the events required for a complete progression of prostatic tumors from low-grade PIN to metastatic disease (see review [75]).

7.2. Conditional models

The “old” (1979) [76] Cre-loxP system was used to produce mice with prostate-specific alterations. Cre is a recombinase that promotes specific genetic recombination in trans at loxP sites. The Cre-loxP system was developed and used for genetic recombination first in yeast and later in mice [77,78]. Many genes knocked out with the whole body strategy were also knocked out by using a conditional approach that results in higher prostate tumor severity. As an example, tissue-specific deletion indicated that homozygous loss of prostatic PTEN led to most stages of prostate tumor progression (metastatic disease) when compared to whole-body haploinsufficiecy, where only PIN was present [79]. At present, the Cre-lox system is diffusely employed to generate mouse models characterized by cell-type-specific and tissue-specific genetic modification (see recent review in ref. [12]). The probasin and the prostate specific antigen (PSA) promoters were extensively utilized to induce targeted Cre expression in the prostate. PB-Cre and PSA-Cre mice have been employed to delete the intraprostatic expression of PTEN, Rb, p53, APC, IGF1 and PTEN, Nkx3, respectively.

E-Resources for mouse models of human cancer, including PCa, are also available online (http://emice.nci.nih.gov/,http://cancermodels.nci.nih.gov/,andhttp://cancerimages.nci.nih.gov/).

Advertisement

8. Clinical trials

Mouse models have significantly contributed to our understanding of PCa biology through their identification of new cancer genes and biomarkers, and their illustration of the molecular and cellular mechanisms underlying tumor initiation and progression. They have also been employed in a preclinical setting to test novel preventive and/or therapeutic strategies [5,6,8-12,80]. Mice, in fact, offer several advantages. They are small, relatively inexpensive, and reproduce rapidly with large litters. More importantly, technical advances have facilitated the generation of defined genetic modifications that can also be spatially controlled, to mimic human prostate carcinogenesis. In general, and perhaps not surprisingly, a variety of phenotypes are obtained depending on the specific genetically engineered mouse model, but none exactly mimics the human disease. Although preclinical studies and the epidemiological evidence suggest that specific dietary components or nutritional supplements influence overall mortality and/or reduce the risk of PCa, randomized, controlled clinical trials provide high-quality evidence of benefit, no effect, or even harm. Examples of ongoing clinical trials are reported in Table 2. In the last ten years, several primary prevention trials have been reported (reviewed in ref. [11,81]). Preventive strategies in a clinical setting have focused on two approaches: antioxidant regimens to reduce DNA damage and suppression of androgenic stimulation [82]. Since a wealth of preclinical and epidemiologic data indicated that selenium and vitamin E reduce PCa, these compounds were evaluated in humans. The Nutritional Prevention of Cancer (NPC) trial found a 63% reduction of PCa incidence (secondary endpoint) following the administration of selenized yeast [83]. The Alpha-Tocopherol Beta-Carotene Cancer prevention study (ATBC), one of the first large studies (14,569 subjects enrolled), investigated the prevention of lung cancer among male smokers. The results indicated that beta carotene supplements increased the risk of lung cancer, rather than preventing it, and that vitamin E had no effect [84-86]. However, a significantly lower risk of PCa was observed for participants receiving vitamin E alone. The NPC and ATBC findings underpinned the NCI-sponsored selenium and vitamin E cancer prevention trial (SELECT). This randomized 35,533 men into four groups: (1) selenium/placebo, (2) vitamin E/placebo, (3) both agents, and (4) placebo alone [87]. At a mean of 5.5 years neither agent reduced risk of PCa. However, at a mean of 7 years and with an additional person-year of follow-up, men receiving vitamin E alone had a significantly increased the risk of PCa (Hazard Ratio 1.17, 99% CI 1.004– 1.36, P = 0.008) [88]. Does vitamin E prevent or promote cancer? More research on the biological activities of the forms and mixtures of tocopherols (alpha, gamma, and delta), and their baseline serum levels should be considered (analyses and discussion in ref. [81,89,90]).

The most promising agents for preventing PCa are probably the 5-alpha reductase inhibitors (5-ARIs). Five-alpha reductase catalyzes the conversion of testosterone to the more active dihydrotestosterone. The Prostate Cancer Prevention Trial (PCPT) and the Reduction by Dutasteride of Prostate Cancer Events (REDUCE) Trial evaluated the activities of two 5-ARIs, finasteride and dutasteride, respectively (reviewed in ref. [81,91]). 5-ARI use for 4-7 years reduced the overall risk of biopsy-detectable PCa by 23-25%. All the prevented cases are either low-grade (PCPT) or GS ≤3 + 4 = 7 prostatic carcinoma (REDUCE). It is unclear whether the slightly increased risk of high-grade cancers in both trials is real or an artifact. In addition to the risk of androgen-independent tumors, the side effects of 5-ARI such as neurodegeneration, osteoporosis, cardiovascular diseases, genitourinary dysfunctions, and hormonal disarrangement limit their use as primary chemopreventive drugs [92-94].

Clinical translation has thus proved to be a general failure when viewed against the optimism aroused by preventive treatments (antioxidant, anti-hormonal, anti-inflammatory, anti-angiogenic etc agents) in the preclinical setting. It has been proposed that species-specific differences, and differences in time of treatment intervention age, trial design enrolment criteria, genetic variation, and the choice, dose, and bioavailability of preventive/therapeutic agents are lie behind for the discrepancy [11]. The most substantial challenge posed by mouse models of PCa, as for other tumors, is their species-specific differences. The lifespan of a mouse is 25-50 times shorter than that of humans, and mice are 3000 times smaller, with consequent differences in pharmacokinetics [95,96]. Anatomically, the human prostate is a single alobular organ with a central, a transitional, and a peripheral zone, whereas the murine prostate comprises four paired lobes located around the urethra, namely the anterior (or coagulating gland), dorsal, lateral, and ventral prostate. The dorsal and lateral lobes are treated as one (the dorsolateral lobe) as they share a ductal system. This lobe has been described as the most similar to the human peripheral zone where most carcinomas arise [97,98]. According to the Bar Harbor Pathology Panel consensus opinion, however, there is no direct relationship between any mouse lobe and any of the human zones [58]. Histologically, the mouse and the human prostate display similar cell types (secretory, basal and neuroendocrine), but their ratio varies from one species to another [99,100]. Mice have fewer basal cells and a discontinuous layer on the basal membrane, whereas in humans, this layer is continuous between secretory cells and the basal membrane. Neuroendocrine cells, rare in humans, are even more rare in mice. The human prostate is characterized by an abundant fibromuscular stoma, whereas the murine gland has a small stromal component. Mice are susceptible to malignancies. By comparison with humans, however, they tend to have more sarcomas and lymphomas and very few epithelial tumors, probably due to differences in relative telomere activity [101-103]. Telomerase, mostly inactive in cells from adult humans, is present in mouse cells, which can thus be transformed/immortalized more easily than their human counterparts, and fewer genetic hits are required to bring about neoplastic transformation in mice than in men. Inactivation of telomerase in the mouse model may be necessary to more accurately recapitulate human cancer phenotypes [80,104].

Most primary PCa prevention studies used mice with an average age of 4-8 weeks, by which time they are considered to have attained sexual maturity and are unlikely to have sustained hormone-induced oxidative stress. In the mouse, a delay in the start of treatment results in a reduced or even no effect. Most human PCa prevention trials were conducted on men aged 50 or more. In addition, the agent dose in animals is 50-80% of the maximally tolerated dose, whereas in humans lower doses may be required for bioethical reasons. The excellent review of Pienta et al. (Prostate Cancer Model Working Group) offers a list of limitations of preclinical models that have hampered the translation of their findings to human clinical trials [8].

Agent* Trial No. Type Institution Phase Status
Green tea NCT00685516 Therapy Jonsson Comprehensive Cancer Center II Recruiting
NCT00253643 Prevention Oregon Health and Science University Recruiting
NCT00003367 Therapy Memorial Sloan-Kettering Cancer Center III Active
NCT00676780 Basic science Louisiana State University Active II Active
NCT00744549 Therapy University Health Network, Toronto II Recruiting
Genistein NCT00546039 Basic science University Hospital, Aker Active II
NCT00005827 Therapy North Carolina University LinebergerCenter I Completed
NCT00058266 Therapy Robert H. Lurie Cancer Center II Active
NCT00584532 Therapy University of California, Davis II/III Completed
NCT00376948 Therapy Barbara Ann Karmanos Cancer Institute II Suspended
NCT00499408 Therapy Wake Forest University II Recruiting
Pomegranate NCT00413530 Therapy M. D. Anderson Cancer Center Recruiting
NCT00719030 Prevention University of California, Los Angeles Recruiting
NCT00732043 Prevention Radiant Research II Recruiting
NCT00731848 Therapy Radiant Research II Recruiting
NCT00336934 Therapy Roll International Corporation III Recruiting
NCT00060086 Therapy Jonsson Comprehensive Cancer Center II Active
NCT00433797 Therapy University of Oslo I/II Recruiting
Lycopene NCT00042731 Therapy H. Lee Moffitt Cancer Center Completed
NCT00416325 Prevention University of Illinois I Completed
NCT00178113 PIN Prevention University of Pittsburgh I Completed
NCT00093561 Prevention University of Illinois Completed I Completed
NCT00450749 Therapy M. D. Anderson Cancer Center II Recruiting
NCT00006078 Prevention University of Illinois I Completed
NCT00322114 Prevention University of Illinois II Recruiting
NCT00402285 Therapy University of California San Francisco Active
NCT00450957 Prevention University of Illinois I Active
NCT00068731 Therapy North Central Cancer Treatment Group II Active
NCT00744549 Therapy University Health Network, Toronto II Recruiting
NCT00669656 Therapy Norris Comprehensive Cancer Center II Recruiting
n-3 poly NCT00458549 Therapy Dana-Farber Cancer Institute Recruiting
unsaturated
fatty acids
NCT00402285 Therapy California San Francisco Helen Diller Center Active

Table 2.

Clinical Trials of Preventive/Therapeutic Regimens for Prostate Cancer

* Data from ref. [105]


Advertisement

9. Conclusions

Genetically engineered mouse models of PCa have paved the way to many important discoveries and helped to define the molecular events of prostate tumorigenesis. However, no single model precisely recapitulates all the molecular or cellular features of the progression of PCa from the normal gland to metastatic, hormone-refractory carcinoma, especially since its early stages are not those of single-cell-type disease, but must be viewed as a complex system of epithelial cells that display dysregulated growth within both a microenvironment composed of many cells which support such growth, and the host macroenvironment with its unique genotype and immune system. Further research is needed to better define these interactions, many of which are potential therapeutic targets. Several in vivo models can be utilized to study specific components of tumor initiation and progression. Meaningful interpretation of their results, however, demands a full understanding of the properties and limits of each model, along with employment of the model most consonant with the subject to be studied. Preclinical models have been poorly predictive of results in human studies because of both their inadequacy and their inappropriate use leading to the designing of clinical trials that do not mirror the preclinical model testing [106]. However, the chemoprevention field is particularly challenging since discrepancies have also been found between initial findings in several trials, secondary analyses and epidemiologic data, and subsequent randomized studies in humans [107]. These inconsistencies may reasonably be supposed to stem from the fact that dietary agents may act long before the scheduled commencement of a chemoprevention trial. Since such trials need to find outcomes (cancers), they invariably start with populations at higher risk of developing clinically detectable cancer, namely middle-aged and older subjects. However, dietary elements may either have a lifelong effect in their changes to the baseline risk for cancer or act at key points by priming the pump for its future development. In either case, dietary chemoprevention might be possible, but its indisputable demonstration in a trial would be highly unlikely. Do these discrepancies mean that all the preclinical and epidemiologic studies are wrong? It must primarily be considered that the timing of such interventions is unclear. Their employment in very high risk subjects, indeed, may actually be too late to significantly prevent cancer formation. Future studies will require both the use of other models founded on our increased understanding of human cancer proteomic genetics and epigenetics to define the very first steps in the progression of the disease and the ability of agents to impair or retard it, and a better “translational approach” achieved through preclinical studies that utilize the appropriate agent doses, and pharmacokinetic and pharmacodynamic parameters to take into account the differences in metabolism between mice and humans, together with clinical trials whose design takes account of how the preclinical testing was accomplished.

Advertisement

Acknowledgments

We wish to thank Prof. John Iliffe for reading the manuscript and critical suggestions. The work of the authors is supported by the Italian Ministry for the Universities and Research.

References

  1. 1. Ferlay J, Autier P, Boniol M, Heanue M, Colombet M, Boyle P. Estimates of the cancer incidence and mortality in Europe in 2006. Ann Oncol 2007;18(3):581-592.
  2. 2. Jemal A, Murray T, Ward E, Samuels A, Tiwari RC, Ghafoor A, Feuer EJ, Thun MJ. Cancer statistics, 2005. CA Cancer J Clin 2005;55(1):10-30.
  3. 3. Dasgupta S, Srinidhi S, Vishwanatha JK. Oncogenic activation in prostate cancer progression and metastasis: Molecular insights and future challenges. J Carcinog;11:4.
  4. 4. Nelson WG, De MarzoAM, Isaacs WB. Prostate cancer. N Engl J Med 2003;349(4):366-381.
  5. 5. Lamb DJ, Zhang L. Challenges in prostate cancer research: animal models for nutritional studies of chemoprevention and disease progression. J Nutr 2005;135(12 Suppl):3009S-3015S.
  6. 6. Nguewa PA, Calvo A. Use of transgenic mice as models for prostate cancer chemoprevention. CurrMol Med;10(8):705-718.
  7. 7. Ozten-Kandas N, Bosland MC. Chemoprevention of prostate cancer: Natural compounds, antiandrogens, and antioxidants - In vivo evidence. J Carcinog;10:27.
  8. 8. Pienta KJ, Abate-Shen C, Agus DB, Attar RM, Chung LW, Greenberg NM, Hahn WC, Isaacs JT, Navone NM, Peehl DM, Simons JW, Solit DB, Soule HR, VanDyke TA, Weber MJ, Wu L, Vessella RL. The current state of preclinical prostate cancer animal models. Prostate 2008;68(6):629-639.
  9. 9. Roy-Burman P, Wu H, Powell WC, Hagenkord J, Cohen MB. Genetically defined mouse models that mimic natural aspects of human prostate cancer development. EndocrRelat Cancer 2004;11(2):225-254.
  10. 10. Sharma P, Schreiber-Agus N. Mouse models of prostate cancer. Oncogene 1999;18(38):5349-5355.
  11. 11. Thapa D, Ghosh R. Antioxidants for prostate cancer chemoprevention: challenges and opportunities. Biochem Pharmacol;83(10):1319-1330.
  12. 12. Valkenburg KC, Williams BO. Mouse models of prostate cancer. Prostate Cancer;2011:895238.
  13. 13. Waters DJ, Sakr WA, Hayden DW, Lang CM, McKinney L, Murphy GP, Radinsky R, Ramoner R, Richardson RC, Tindall DJ. Workgroup 4: spontaneous prostate carcinoma in dogs and nonhuman primates. Prostate 1998;36(1):64-67.
  14. 14. Leroy BE, Northrup N. Prostate cancer in dogs: comparative and clinical aspects. Vet J 2009;180(2):149-162.
  15. 15. Winter SF, Cooper AB, Greenberg NM. Models of metastatic prostate cancer: a transgenic perspective. Prostate Cancer Prostatic Dis 2003;6(3):204-211.
  16. 16. Ghoniem GM, VandenBerg TL. Dollars and sense: considerations for experimental model design. NeurourolUrodyn 1994;13(2):91-96.
  17. 17. Maini A, Archer C, Wang CY, Haas GP. Comparative pathology of benign prostatic hyperplasia and prostate cancer. In Vivo 1997;11(4):293-299.
  18. 18. Rosol TJ, Tannehill-Gregg SH, LeRoy BE, Mandl S, Contag CH. Animal models of bone metastasis. Cancer 2003;97(3 Suppl):748-757.
  19. 19. Dunning WF. Prostate Cancer in the Rat. Natl Cancer InstMonogr 1963;12:351-369.
  20. 20. Isaacs JT, Isaacs WB, Feitz WF, Scheres J. Establishment and characterization of seven Dunning rat prostatic cancer cell lines and their use in developing methods for predicting metastatic abilities of prostatic cancers. Prostate 1986;9(3):261-281.
  21. 21. Isaacs JT, Weissman RM, Coffey DS, Scott WW. Concepts in prostatic cancer biology: Dunning R-3327 H, HI, and AT tumors. ProgClinBiol Res 1980;37:311-323.
  22. 22. Lubaroff DM, Canfield L, Feldbush TL, Bonney WW. R3327 adenocarcinoma of the Copenhagen rat as a model for the study of the immunologic aspects of prostate cancer. J Natl Cancer Inst 1977;58(6):1677-1689.
  23. 23. Lubaroff DM, Canfield L, Rasmussen GT, Reynolds CW. An animal model for the study of prostate carcinoma. Natl Cancer InstMonogr 1978(49):275-281.
  24. 24. Jeet V, Russell PJ, Khatri A. Modeling prostate cancer: a perspective on transgenic mouse models. Cancer Metastasis Rev;29(1):123-142.
  25. 25. Pollard M. Animal models for prostate cancer. Prostate 1980;1(2):207-213.
  26. 26. Geurts AM, Cost GJ, Freyvert Y, Zeitler B, Miller JC, Choi VM, Jenkins SS, Wood A, Cui X, Meng X, Vincent A, Lam S, Michalkiewicz M, Schilling R, Foeckler J, Kalloway S, Weiler H, Menoret S, Anegon I, Davis GD, Zhang L, Rebar EJ, Gregory PD, Urnov FD, Jacob HJ, Buelow R. Knockout rats via embryo microinjection of zinc-finger nucleases. Science 2009;325(5939):433.
  27. 27. Izsvak Z, Frohlich J, Grabundzija I, Shirley JR, Powell HM, Chapman KM, Ivics Z, Hamra FK. Generating knockout rats by transposon mutagenesis in spermatogonial stem cells. Nat Methods;7(6):443-445.
  28. 28. Tong C, Li P, Wu NL, Yan Y, Ying QL. Production of p53 gene knockout rats by homologous recombination in embryonic stem cells. Nature;467(7312):211-213.
  29. 29. Hamra FK. Gene targeting: Enter the rat. Nature;467(7312):161-163.
  30. 30. Morton CL, Houghton PJ. Establishment of human tumorxenografts in immunodeficient mice. Nat Protoc 2007;2(2):247-250.
  31. 31. Schroder FH, Jellinghaus W. Prostatic adenoma and carcinoma in cell culture and heterotransplantation. ProgClinBiol Res 1976;6:301-312.
  32. 32. Hoehn W, Schroeder FH, Reimann JF, Joebsis AC, Hermanek P. Human prostatic adenocarcinoma: some characteristics of a serially transplantable line in nude mice (PC 82). Prostate 1980;1(1):95-104.
  33. 33. van Weerden WM, Romijn JC. Use of nude mouse xenograft models in prostate cancer research. Prostate 2000;43(4):263-271.
  34. 34. Thalmann GN, Anezinis PE, Chang SM, Zhau HE, Kim EE, Hopwood VL, Pathak S, von Eschenbach AC, Chung LW. Androgen-independent cancer progression and bone metastasis in the LNCaP model of human prostate cancer. Cancer Res 1994;54(10):2577-2581.
  35. 35. Bosma GC, Custer RP, Bosma MJ. A severe combined immunodeficiency mutation in the mouse. Nature 1983;301(5900):527-530.
  36. 36. Bosma MJ, Carroll AM. The SCID mouse mutant: definition, characterization, and potential uses. Annu Rev Immunol 1991;9:323-350.
  37. 37. Craft N, Shostak Y, Carey M, Sawyers CL. A mechanism for hormone-independent prostate cancer through modulation of androgen receptor signaling by the HER-2/neu tyrosine kinase. Nat Med 1999;5(3):280-285.
  38. 38. Shultz LD, Schweitzer PA, Christianson SW, Gott B, Schweitzer IB, Tennent B, McKenna S, Mobraaten L, Rajan TV, Greiner DL, et al. Multiple defects in innate and adaptive immunologic function in NOD/LtSz-scid mice. J Immunol 1995;154(1):180-191.
  39. 39. Bastide C, Bagnis C, Mannoni P, Hassoun J, Bladou F. A Nod Scid mouse model to study human prostate cancer. Prostate Cancer Prostatic Dis 2002;5(4):311-315.
  40. 40. Nemeth JA, Harb JF, Barroso U, Jr., He Z, Grignon DJ, Cher ML. Severe combined immunodeficient-hu model of human prostate cancer metastasis to human bone. Cancer Res 1999;59(8):1987-1993.
  41. 41. Wainstein MA, He F, Robinson D, Kung HJ, Schwartz S, Giaconia JM, Edgehouse NL, Pretlow TP, Bodner DR, Kursh ED, et al. CWR22: androgen-dependent xenograft model derived from a primary human prostatic carcinoma. Cancer Res 1994;54(23):6049-6052.
  42. 42. Yonou H, Yokose T, Kamijo T, Kanomata N, Hasebe T, Nagai K, Hatano T, Ogawa Y, Ochiai A. Establishment of a novel species- and tissue-specific metastasis model of human prostate cancer in humanized non-obese diabetic/severe combined immunodeficient mice engrafted with human adult lung and bone. Cancer Res 2001;61(5):2177-2182.
  43. 43. D'Antonio JM, Vander Griend DJ, Antony L, Ndikuyeze G, Dalrymple SL, Koochekpour S, Isaacs JT. Loss of androgen receptor-dependent growth suppression by prostate cancer cells can occur independently from acquiring oncogenic addiction to androgen receptor signaling. PLoS One;5(7):e11475.
  44. 44. Ito M, Hiramatsu H, Kobayashi K, Suzue K, Kawahata M, Hioki K, Ueyama Y, Koyanagi Y, Sugamura K, Tsuji K, Heike T, Nakahata T. NOD/SCID/gamma(c)(null) mouse: an excellent recipient mouse model for engraftment of human cells. Blood 2002;100(9):3175-3182.
  45. 45. Quintana E, Shackleton M, Sabel MS, Fullen DR, Johnson TM, Morrison SJ. Efficient tumour formation by single human melanoma cells. Nature 2008;456(7222):593-598.
  46. 46. Sobel RE, Sadar MD. Cell lines used in prostate cancer research: a compendium of old and new lines--part 1. J Urol 2005;173(2):342-359.
  47. 47. Sobel RE, Sadar MD. Cell lines used in prostate cancer research: a compendium of old and new lines--part 2. J Urol 2005;173(2):360-372.
  48. 48. Sobel RE, Wang Y, Sadar MD. Molecular analysis and characterization of PrEC, commercially available prostate epithelial cells. In Vitro Cell DevBiolAnim 2006;42(1-2):33-39.
  49. 49. van Bokhoven A, Caires A, Maria MD, Schulte AP, Lucia MS, Nordeen SK, Miller GJ, Varella-Garcia M. Spectral karyotype (SKY) analysis of human prostate carcinoma cell lines. Prostate 2003;57(3):226-244.
  50. 50. vanBokhoven A, Varella-Garcia M, Korch C, Johannes WU, Smith EE, Miller HL, Nordeen SK, Miller GJ, Lucia MS. Molecular characterization of human prostate carcinoma cell lines. Prostate 2003;57(3):205-225.
  51. 51. DeCaprio JA, Ludlow JW, Figge J, Shew JY, Huang CM, Lee WH, Marsilio E, Paucha E, Livingston DM. SV40 large tumor antigen forms a specific complex with the product of the retinoblastoma susceptibility gene. Cell 1988;54(2):275-283.
  52. 52. Lane DP, Crawford LV. T antigen is bound to a host protein in SV40-transformed cells. Nature 1979;278(5701):261-263.
  53. 53. Pallas DC, Shahrik LK, Martin BL, Jaspers S, Miller TB, Brautigan DL, Roberts TM. Polyoma small and middle T antigens and SV40 small t antigen form stable complexes with protein phosphatase 2A. Cell 1990;60(1):167-176.
  54. 54. Maroulakou IG, Anver M, Garrett L, Green JE. Prostate and mammary adenocarcinoma in transgenic mice carrying a rat C3(1) simian virus 40 large tumor antigen fusion gene. ProcNatlAcadSci U S A 1994;91(23):11236-11240.
  55. 55. Yoshidome K, Shibata MA, Maroulakou IG, Liu ML, Jorcyk CL, Gold LG, Welch VN, Green JE. Genetic alterations in the development of mammary and prostate cancer in the C3(1)/Tag transgenic mouse model. Int J Oncol 1998;12(2):449-453.
  56. 56. Gingrich JR, Barrios RJ, Kattan MW, Nahm HS, Finegold MJ, Greenberg NM. Androgen-independent prostate cancer progression in the TRAMP model. Cancer Res 1997;57(21):4687-4691.
  57. 57. Greenberg NM, DeMayo F, Finegold MJ, Medina D, Tilley WD, Aspinall JO, Cunha GR, Donjacour AA, Matusik RJ, Rosen JM. Prostate cancer in a transgenic mouse. ProcNatlAcadSci U S A 1995;92(8):3439-3443.
  58. 58. Shappell SB, Thomas GV, Roberts RL, Herbert R, Ittmann MM, Rubin MA, Humphrey PA, Sundberg JP, Rozengurt N, Barrios R, Ward JM, Cardiff RD. Prostate pathology of genetically engineered mice: definitions and classification. The consensus report from the Bar Harbor meeting of the Mouse Models of Human Cancer Consortium Prostate Pathology Committee. Cancer Res 2004;64(6):2270-2305.
  59. 59. Tani Y, Suttie A, Flake GP, Nyska A, Maronpot RR. Epithelial-stromal tumor of the seminal vesicles in the transgenic adenocarcinoma mouse prostate model. Vet Pathol 2005;42(3):306-314.
  60. 60. Gingrich JR, Barrios RJ, Foster BA, Greenberg NM. Pathologic progression of autochthonous prostate cancer in the TRAMP model. Prostate Cancer Prostatic Dis 1999;2(2):70-75.
  61. 61. Wood RW, Baggs RB, Schwarz EM, Messing EM. Initial observations of reduced uroflow in transgenic adenocarcinoma of murine prostate. Urology 2006;67(6):1324-1328.
  62. 62. Bono AV, Montironi R, Pannellini T, Sasso F, Mirone V, Musiani P, Iezzi M. Effects of castration on the development of prostate adenocarcinoma from its precursor HGPIN and on the occurrence of androgen-independent, poorly differentiated carcinoma in TRAMP mice. Prostate Cancer Prostatic Dis 2008;11(4):377-383.
  63. 63. Chiaverotti T, Couto SS, Donjacour A, Mao JH, Nagase H, Cardiff RD, Cunha GR, Balmain A. Dissociation of epithelial and neuroendocrine carcinoma lineages in the transgenic adenocarcinoma of mouse prostate model of prostate cancer. Am J Pathol 2008;172(1):236-246.
  64. 64. Huss WJ, Gray DR, Tavakoli K, Marmillion ME, Durham LE, Johnson MA, Greenberg NM, Smith GJ. Origin of androgen-insensitive poorly differentiated tumors in the transgenic adenocarcinoma of mouse prostate model. Neoplasia 2007;9(11):938-950.
  65. 65. Yan Y, Sheppard PC, Kasper S, Lin L, Hoare S, Kapoor A, Dodd JG, Duckworth ML, Matusik RJ. Large fragment of the probasin promoter targets high levels of transgene expression to the prostate of transgenic mice. Prostate 1997;32(2):129-139.
  66. 66. Kasper S, Sheppard PC, Yan Y, Pettigrew N, Borowsky AD, Prins GS, Dodd JG, Duckworth ML, Matusik RJ. Development, progression, and androgen-dependence of prostate tumors in probasin-large T antigen transgenic mice: a model for prostate cancer. Lab Invest 1998;78(6):i-xv.
  67. 67. Kasper S, Tu W, Roberts RL, Shappell SB. Transgenic mouse models for prostate cancer. Identification of an androgen-dependent promoter and creation and characterization of the long probasin promoter-Large T antigen (LPB-Tag) model. Methods Mol Med 2003;81:113-147.
  68. 68. Weihua Z, Makela S, Andersson LC, Salmi S, Saji S, Webster JI, Jensen EV, Nilsson S, Warner M, Gustafsson JA. A role for estrogen receptor beta in the regulation of growth of the ventral prostate. ProcNatlAcadSci U S A 2001;98(11):6330-6335.
  69. 69. Lohnes D, Kastner P, Dierich A, Mark M, LeMeur M, Chambon P. Function of retinoic acid receptor gamma in the mouse. Cell 1993;73(4):643-658.
  70. 70. Cordon-Cardo C, Koff A, Drobnjak M, Capodieci P, Osman I, Millard SS, Gaudin PB, Fazzari M, Zhang ZF, Massague J, Scher HI. Distinct altered patterns of p27KIP1 gene expression in benign prostatic hyperplasia and prostatic carcinoma. J Natl Cancer Inst 1998;90(17):1284-1291.
  71. 71. Podsypanina K, Ellenson LH, Nemes A, Gu J, Tamura M, Yamada KM, Cordon-Cardo C, Catoretti G, Fisher PE, Parsons R. Mutation of Pten/Mmac1 in mice causes neoplasia in multiple organ systems. ProcNatlAcadSci U S A 1999;96(4):1563-1568.
  72. 72. Di Cristofano A, De Acetis M, Koff A, Cordon-Cardo C, Pandolfi PP. Pten and p27KIP1 cooperate in prostate cancer tumor suppression in the mouse. Nat Genet 2001;27(2):222-224.
  73. 73. Abate-Shen C, Banach-Petrosky WA, Sun X, Economides KD, Desai N, Gregg JP, Borowsky AD, Cardiff RD, Shen MM. Nkx3.1; Pten mutant mice develop invasive prostate adenocarcinoma and lymph node metastases. Cancer Res 2003;63(14):3886-3890.
  74. 74. Couto SS, Cao M, Duarte PC, Banach-Petrosky W, Wang S, Romanienko P, Wu H, Cardiff RD, Abate-Shen C, Cunha GR. Simultaneous haploinsufficiency of Pten and Trp53 tumor suppressor genes accelerates tumorigenesis in a mouse model of prostate cancer. Differentiation 2009;77(1):103-111.
  75. 75. Kasper S. Survey of genetically engineered mouse models for prostate cancer: analyzing the molecular basis of prostate cancer development, progression, and metastasis. J Cell Biochem 2005;94(2):279-297.
  76. 76. Sternberg N. Demonstration and analysis of P1 site-specific recombination using lambda-P1 hybrid phages constructed in vitro. Cold Spring HarbSymp Quant Biol 1979;43Pt 2:1143-1146.
  77. 77. Metzger D, Chambon P. Site- and time-specific gene targeting in the mouse. Methods 2001;24(1):71-80.
  78. 78. Sauer B, Henderson N. Cre-stimulated recombination at loxP-containing DNA sequences placed into the mammalian genome. Nucleic Acids Res 1989;17(1):147-161.
  79. 79. Wang S, Gao J, Lei Q, Rozengurt N, Pritchard C, Jiao J, Thomas GV, Li G, Roy-Burman P, Nelson PS, Liu X, Wu H. Prostate-specific deletion of the murine Ptentumor suppressor gene leads to metastatic prostate cancer. Cancer Cell 2003;4(3):209-221.
  80. 80. Cheon DJ, Orsulic S. Mouse models of cancer. Annu Rev Pathol;6:95-119.
  81. 81. Klein EA, Thompson IM. Chemoprevention of prostate cancer: an updated view. World J Urol;30(2):189-194.
  82. 82. Walsh PC. Chemoprevention of prostate cancer. N Engl J Med;362(13):1237-1238.
  83. 83. Clark LC, Dalkin B, Krongrad A, Combs GF, Jr., Turnbull BW, Slate EH, Witherington R, Herlong JH, Janosko E, Carpenter D, Borosso C, Falk S, Rounder J. Decreased incidence of prostate cancer with selenium supplementation: results of a double-blind cancer prevention trial. Br J Urol 1998;81(5):730-734.
  84. 84. Albanes D, Heinonen OP, Taylor PR, Virtamo J, Edwards BK, Rautalahti M, Hartman AM, Palmgren J, Freedman LS, Haapakoski J, Barrett MJ, Pietinen P, Malila N, Tala E, Liippo K, Salomaa ER, Tangrea JA, Teppo L, Askin FB, Taskinen E, Erozan Y, Greenwald P, Huttunen JK. Alpha-Tocopherol and beta-carotene supplements and lung cancer incidence in the alpha-tocopherol, beta-carotene cancer prevention study: effects of base-line characteristics and study compliance. J Natl Cancer Inst 1996;88(21):1560-1570.
  85. 85. Virtamo J, Pietinen P, Huttunen JK, Korhonen P, Malila N, Virtanen MJ, Albanes D, Taylor PR, Albert P. Incidence of cancer and mortality following alpha-tocopherol and beta-carotene supplementation: a postintervention follow-up. Jama 2003;290(4):476-485.
  86. 86. Weinstein SJ, Wright ME, Lawson KA, Snyder K, Mannisto S, Taylor PR, Virtamo J, Albanes D. Serum and dietary vitamin E in relation to prostate cancer risk. Cancer Epidemiol Biomarkers Prev 2007;16(6):1253-1259.
  87. 87. Lippman SM, Klein EA, Goodman PJ, Lucia MS, Thompson IM, Ford LG, Parnes HL, Minasian LM, Gaziano JM, Hartline JA, Parsons JK, Bearden JD, 3rd, Crawford ED, Goodman GE, Claudio J, Winquist E, Cook ED, Karp DD, Walther P, Lieber MM, Kristal AR, Darke AK, Arnold KB, Ganz PA, Santella RM, Albanes D, Taylor PR, Probstfield JL, Jagpal TJ, Crowley JJ, Meyskens FL, Jr., Baker LH, Coltman CA, Jr. Effect of selenium and vitamin E on risk of prostate cancer and other cancers: the Selenium and Vitamin E Cancer Prevention Trial (SELECT). Jama 2009;301(1):39-51.
  88. 88. Klein EA, Thompson IM, Jr., Tangen CM, Crowley JJ, Lucia MS, Goodman PJ, Minasian LM, Ford LG, Parnes HL, Gaziano JM, Karp DD, Lieber MM, Walther PJ, Klotz L, Parsons JK, Chin JL, Darke AK, Lippman SM, Goodman GE, Meyskens FL, Jr., Baker LH. Vitamin E and the risk of prostate cancer: the Selenium and Vitamin E Cancer Prevention Trial (SELECT). Jama;306(14):1549-1556.
  89. 89. McNeil C. Vitamin E and prostate cancer: research focus turns to biologic mechanisms. J Natl Cancer Inst;103(23):1731-1734.
  90. 90. Yang CS, Suh N, Kong AN. Does vitamin E prevent or promote cancer? Cancer Prev Res (Phila);5(5):701-705.
  91. 91. Azzouni F, Mohler J. Role of 5alpha-reductase inhibitors in prostate cancer prevention and treatment. Urology;79(6):1197-1205.
  92. 92. Feldman BJ, Feldman D. The development of androgen-independent prostate cancer. Nat Rev Cancer 2001;1(1):34-45.
  93. 93. Jones TH. Cardiovascular risk during androgen deprivation therapy for prostate cancer. Bmj;342:d3105.
  94. 94. Taylor LG, Canfield SE, Du XL. Review of major adverse effects of androgen-deprivation therapy in men with prostate cancer. Cancer 2009;115(11):2388-2399.
  95. 95. de Jong M, Maina T. Of mice and humans: are they the same?--Implications in cancer translational research. J Nucl Med;51(4):501-504.
  96. 96. Rangarajan A, Weinberg RA. Opinion: Comparative biology of mouse versus human cells: modelling human cancer in mice. Nat Rev Cancer 2003;3(12):952-959.
  97. 97. Powell WC, Cardiff RD, Cohen MB, Miller GJ, Roy-Burman P. Mouse strains for prostate tumorigenesis based on genes altered in human prostate cancer. Curr Drug Targets 2003;4(3):263-279.
  98. 98. Xue L, Yang K, Newmark H, Lipkin M. Induced hyperproliferation in epithelial cells of mouse prostate by a Western-style diet. Carcinogenesis 1997;18(5):995-999.
  99. 99. Garabedian EM, Humphrey PA, Gordon JI. A transgenic mouse model of metastatic prostate cancer originating from neuroendocrine cells. ProcNatlAcadSci U S A 1998;95(26):15382-15387.
  100. 100. Marker PC, Donjacour AA, Dahiya R, Cunha GR. Hormonal, cellular, and molecular control of prostatic development. DevBiol 2003;253(2):165-174.
  101. 101. Artandi SE, DePinho RA. Telomeres and telomerase in cancer. Carcinogenesis;31(1):9-18.
  102. 102. Harvey M, McArthur MJ, Montgomery CA, Jr., Butel JS, Bradley A, Donehower LA. Spontaneous and carcinogen-induced tumorigenesis in p53-deficient mice. Nat Genet 1993;5(3):225-229.
  103. 103. Jacks T, Remington L, Williams BO, Schmitt EM, Halachmi S, Bronson RT, Weinberg RA. Tumor spectrum analysis in p53-mutant mice. CurrBiol 1994;4(1):1-7.
  104. 104. Maser RS, Choudhury B, Campbell PJ, Feng B, Wong KK, Protopopov A, O'Neil J, Gutierrez A, Ivanova E, Perna I, Lin E, Mani V, Jiang S, McNamara K, Zaghlul S, Edkins S, Stevens C, Brennan C, Martin ES, Wiedemeyer R, Kabbarah O, Nogueira C, Histen G, Aster J, Mansour M, Duke V, Foroni L, Fielding AK, Goldstone AH, Rowe JM, Wang YA, Look AT, Stratton MR, Chin L, Futreal PA, DePinho RA. Chromosomally unstable mouse tumours have genomic alterations similar to diverse human cancers. Nature 2007;447(7147):966-971.
  105. 105. Amin AR, Kucuk O, Khuri FR, Shin DM. Perspectives for cancer prevention with natural compounds. J ClinOncol 2009;27(16):2712-2725.
  106. 106. Gupta S. Prostate cancer chemoprevention: current status and future prospects. ToxicolApplPharmacol 2007;224(3):369-376.
  107. 107. Strope SA, Andriole GL. Update on chemoprevention for prostate cancer. CurrOpin Urol;20(3):194-197.
  108. 108. Raghow S, et al. Cancer Res 2000;60:4093–7;
  109. 109. Gupta S, et al. Cancer Res 2000;60:5125-33;
  110. 110. Gupta S, et al. Proc NatlAcadSci USA 2001;98:10350-5;
  111. 111. Mentor-Marcel R, et al. Cancer Res 2001;61:6777-82;
  112. 112. Raghow S, et al. Cancer Res 2002;62:1370-6;
  113. 113. Gupta S, et al. Cancer Res 2004;64:3334-43;
  114. 114. Narayanan BA, et al. Clin Cancer Res 2004;10:7727-37;
  115. 115. Wang J, et al. Mol CellEndocrinol 2004;219:171-80;
  116. 116. Huss WJ, et al. Prostate 2004;61:142-52;
  117. 117. Adhami VM, et al. Cancer Res 2004;64:8715-22;
  118. 118. Sartor L, et al. Int J Cancer 2004; 112, 823-9;
  119. 119. Caporali A, et al. Carcinogenesis 2004;25:2217-24;
  120. 120. Saleem M, et al. Clin Cancer Res 2005;11:147-53;
  121. 121. Kolluri SK, et al. ProcNatlAcadSci USA 2005;102:2525-30;
  122. 122. Chiang CF, et al. Prostate 2005;64:408-18;
  123. 123. Wikstrom P, et al. Nutr Cancer 2005;53:111-6;
  124. 124. Mentor Marcel R, et al. J Nutrition 2005;135:989-95;
  125. 125. Narayanan BA, et al. Prostate 2006;66:257-65;
  126. 126. Tam NN, et al. Prostate 2006;66:57-69;
  127. 127. McCabe MT, et al. Cancer Res 2006;66:385-92;
  128. 128. Garcia GE, et al. Clin Cancer Res 2006;12:980-7;
  129. 129. Raina K, et al. Cancer Res 2007;67:5976-82;
  130. 130. Shukla S, et al. Cancer Res 2007;67:6925-35;
  131. 131. Wang J, et al. J Carcinog. 2007;6:3;
  132. 132. Isayeva T, et al. Cancer Res 2007;67:5789-97;
  133. 133. Harper CE, et al. Prostate 2007;67:1576-89;
  134. 134. Raina K, et al. Cancer Res 2007;67:11083-91;
  135. 135. De Giovanni C Croci S, et al. Int J Cancer 2007;121:88-94;
  136. 136. Harper CE, et al. Prostate 2007;67:1576-89;
  137. 137. Shukla S, et al. Cancer Res 2007;67:6925-35;
  138. 138. Wadsworth TL, et al. Prostate 2007;67:661-73;
  139. 139. Raina K, et al. Cancer Res 2008;68:6822-30;
  140. 140. Singh RP, et al. Clin CanceRes 2008;14:7773-80;
  141. 141. Singh SV, et al. Cancer Res 2009;69:2117-25;
  142. 142. Adhami VM, et al. Clin Cancer Res 2009;15:1947-53;
  143. 143. Raina K, et al. Cancer Res 2009;69:3731-5;
  144. 144. Barve A, et al. Int J Cancer 2009;124:1693-9;
  145. 145. Ibrahim-Hashim A, et al. J Urol 2012;Jun 14 [Epubahead of print];
  146. 146. Huang Y, et al. J Nutr 2012;142:818-23;
  147. 147. Shanmugam MK, et al. PLoS One 2012;7:e32476 [Epub ahead of print];
  148. 148. Davis PA, et al. Br J Nutr 2012;16:1-9 [Epub ahead of print];
  149. 149. Adhami VM, et al. Carcinogenesis 2012;33:644-51;
  150. 150. Shukla S, et al. Pharm Res 2012;29:1506-17;
  151. 151. Dalezis P, et al. In Vivo 2012;26:75-86;
  152. 152. Ru P, et al. Cancer Prev Res (Phila) 2011;4:2122-30;
  153. 153. Bistulfi G, et al.Cancer Prev Res (Phila) 2011;4:1825-34;
  154. 154. Shanmugam MK, et al. Int J Cancer 2011;129:1552-63;
  155. 155. Abedinpour P, et al. Prostate 2011;71:813-23;
  156. 156. Kim SH, et al. Cancer Prev Res (Phila) 2011;4:897-06;
  157. 157. Wu TY, et al. Mol Carcinog 2011;
  158. 158. Pannellini et al. Cancer PrevRes 2010;3:1284-91;
  159. 159. Konijeti R, et al. Prostate 2010;70:1547-54;
  160. 160. Llaverias G, et al. Am J Pathol 2010;177:3180-91;
  161. 161. Wang L, et al. Prostate 2011;71:1429-40;
  162. 162. Deeb D, et al. Carcinogenesis 2011;32:757-64;
  163. 163. Kang BH, et al. Br J Cancer 2011;104:629-34;
  164. 164. Rigamonti N, et al. Clin Cancer Res 2011;17:1012-23;
  165. 165. Wang L, et al. Cancer Prev Res (Phila) 2009;2:484-95;
  166. 166. Ganapathy M, et al. Clin Cancer Res 2009;15:1601-11;
  167. 167. Persano L, et al. Carcinogenesis 2009;30:851-60;
  168. 168. Cho HJ, et al. MolCarcinog 2011;50:100-12;
  169. 169. Barve A, et al. Nutr Cancer 2010;62:789-94;
  170. 170. Prasad AS, et al. J Med Food 2010;13:70-6;
  171. 171. Reddy KR, et al. Prostate 2011;71:1796-09;
  172. 172. Wu TY, et al. Mol Carcinogenesis Aug 2011.

Written By

Rossano Lattanzio, Alessia Lamolinara, Mauro Piantelli and Manuela Iezzi

Submitted: 03 May 2012 Published: 16 January 2013