Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Oct 30.
Published in final edited form as: Adv Drug Deliv Rev. 2010 Aug 3;62(13):1285–1298. doi: 10.1016/j.addr.2010.07.003

Retinoid Pathway and Cancer Therapeutics

Nathan Bushue 1, Yu-Jui Yvonne Wan 1,*
PMCID: PMC2991380  NIHMSID: NIHMS229611  PMID: 20654663

Abstract

The retinoids are a class of compounds that are structurally related to vitamin A. Retinoic acid, which is the active metabolite of retinol, regulates a wide range of biological processes including development, differentiation, proliferation, and apoptosis. Retinoids exert their effects through a variety of binding proteins including cellular retinol binding protein (CRBP), retinol-binding proteins (RBP), cellular retinoic acid-binding protein (CRABP), and nuclear receptors i.e. retinoic acid receptor (RAR) and retinoid × receptor (RXR). Because of the pleiotropic effects of retinoids, understanding the function of these binding proteins and nuclear receptors assists us in developing compounds that have specific effects. This review summarizes our current understanding of how retinoids are processed and act with the emphasis on the application of retinoids in cancer treatment and prevention.

Keywords: Retinoids, Nuclear receptor, RAR, RXR, all-trans retinoic acid, 9-cis retinoic acid, 13-cis retinoic acid

1. History

Vitamin A and its derivatives (retinoids) exert a wide range of effects on embryonic development, cell growth, differentiation, and apoptosis. Vitamin A has been used as a treatment for thousands of years. The Egyptian papyruses Kahun 1 (ca. 1825 B.C.) and Ebers (ca. 1500 B.C.) described how the liver was used to cure eye diseases such as night blindness. Greek scholar Hippocrates (460-327 B.C.) described in the second book of “Prognostics” a method for curing night blindness: “raw beef liver, as large as possible, soaked in honey, to be taken once or twice by mouth.” Chinese medicine used pigs’ liver as a remedy for night blindness, as described by Sun-szu-mo (7th century A.D.) in his “1000 Golden Remedies”. Given that the liver is where the body stores excess vitamin A, the liver represents the best source of vitamin A available for treatment in the pre-pharmaceutical world.

One of the first experiments involving vitamin A was performed by F. Magendie (1817) [1], in which he fed dogs a diet of only sugar and water. Three weeks later, the dogs got sick and developed ulcers of the cornea. At the time, Magendie attributed these effects to nitrogen deficiency in their diet, but we now understand the sickness could have been due to vitamin A deficiency. The effect of vitamin A on growth was first described in a mouse experiment done by G. Lunin (1881) [2], in which one group of mice was fed pure casein, fat, sucrose, minerals, and water, and another group was fed whole dried milk. The milk-fed group was healthy and grew normally, while the other group was sick and ultimately died. Thus, something in milk was essential for survival. Elmer McCollum at University of Wisconsin-Madison as well as Lafayette Mendel and Thomas Burr Osborne at Yale University independently discovered vitamin A. McCollum began his study in 1907 by feeding cows hay with wheat, oats, or yellow maize.

Wheat-fed cows did not thrive, became blind and gave birth to dead calves prematurely. Oat-fed cows fared somewhat better, but the yellow maize-fed cows were in excellent condition, produced vigorous calves, and had no miscarriages. McCollum postulated that performing the same nutritional study using small animals, such as rodents, which require less food, provide faster reproduction and experimental outcome. Using rats, he found a diet of pure protein, pure milk sugar, minerals, and lard (or olive oil) inhibited growth, while addition of butterfat or an ether extract of egg yolk to the diet restored health. Thinking that he had found a fat-soluble factor that promoted growth in rats, he saponified butterfat, extracted the unsaponifiable mixture into ether, and added the extract to oliveoil and that extract could support growth. This essential component to support growth and development was named “fat-soluble factor A,” and later renamed vitamin A [1].

2. Retinoids

There are over 4,000 natural and synthetic molecules structurally and/or functionally related to vitamin A. Vitamin A cannot be synthesized by any animal species and is only obtained through diet in the form of retinol, retinyl ester, or β-carotene (Figure 1). Ingested vitamin A is stored as retinyl esters in hepatic stellate cells. Retinol is reversibly oxidized by retinol dehydrogenases to yield retinal. Subsequently, retinal may be irreversibly oxidized to all-trans retinoic acid (all-trans RA) by retinal dehydrogenases and further oxidized by cytochrome P450 enzymes (mainly CYP26) in hepatic tissue. Retinol has six biologically active isoforms that include all-trans, 11-cis, 13-cis, 9, 13-di-cis, 9-cis, and 11, 13-di-cis, with all-trans being the predominant physiological form. Endogenous retinoids with biological activity include all-trans RA, 9-cis RA, 11-cis retinaldehyde, 3,4-didehydro RA, and perhaps 14-hydroxy-4, 14-retro retinol, 4-oxo RA, and 4-oxo retinol [35]. All-trans RA isomerizes under experimental and physiological conditions. Different isomers activate different receptors and thus lead to different biological effects. RAs designed to be receptor specific can improve efficacy and avoid unwanted side effects. Retinoids that specifically bind to RXR are called rexinoids and have been effective in cancer treatment. Retinoids are comprised of three units: a bulky hydrophobic region, a linker unit, and a polar terminus, which is usually a carboxylic acid. Modification of each unit has generated many more compounds. Please refer to recent reviews [68].

Figure 1. Retinoid Pathway.

Figure 1

Retinoids absorbed from food are converted to retinol and bound to CRBP in the intestine. Then, retinol is converted to retinyl esters and enters into blood circulation. The liver up takes retinyl esters, which are converted to retinol-RBP complex in the hepatocyte. In the serum, the retinol-RBP complex is bound to transthyretin (TTR) in a 1:1 ratio to prevent elimination by the kidney and to ensure retinol is delivered to the target cell. The uptake of retinol by the target cell is mediated by a trans-membrane protein named “stimulated by retinoic acid 6” (STRA6), which is a RBP receptor. In the target cell, retinol either binds to CRBP or is oxidized to retinaldehyde by retinol dehydrogenase (RDH) in a reversible reaction. Then, retinaldehyde can be oxidized by retinaldehyde dehydrogenase (RALDH) to RA. In the target cell, RA either binds to CRABP or enters the nucleus and binds to nuclear receptors to regulate gene transcription. Alternatively, RA can mediate via nongenomic mechanism and regulate cellular function. Hepatocytes not only process retinoids, but also are the target cells. In addition, hepatocytes located next to the storage site (stellate cell). Thus, retinoid-mediated signaling must have a profound effect in regulating hepatocyte function and phenotype [36, 190, 191]

3. Retinoid Binding Proteins

There are various types of retinoid-binding proteins, which locate in intracellular and extracellular compartments and associate with isomeric forms of retinoids. Hence, retinoids are either associated with cellular membranes or bound to a specific retinoid binding protein. These binding proteins along with nuclear receptors mediate the action of retinoids. Their interactions are summarized in figure 1. Retinoid-binding proteins solubilize and stabilize retinoids in aqueous spaces. In addition to this general role, specific retinoid-binding proteins have distinct functions in regulating transport and metabolism of specific retinoids. For example, the parent vitamin A molecule, all-trans retinol, circulates in blood bound to serum retinol binding protein (RBP). Inside the cells, all-trans retinol and its oxidation product, all-trans retinal, are associated with different isoforms of cellular retinol-binding proteins (CRBP), while all-trans RA intracellularly binds to cellular retinoic acid-binding protein isoforms (CRABP).

3.1. RBP

Retinol is secreted from its storage pools and circulates in blood by binding to RBP. The main storage site for vitamin A and the main site of synthesis of RBP is the liver, although other tissues (including adipose tissue, kidney, lung, heart, skeletal muscle, spleen, eye and testis) also express this protein. Secretion of RBP from the liver is regulated by the availability of retinol [9]. Vitamin A deficiency inhibits RBP secretion, leading to protein accumulation in the endoplasmic reticulum of hepatic parenchymal cells. In the presence of retinol, RBP associates with retinol, moves to the Golgi apparatus and is secreted into blood. The mechanism by which retinol initiates RBP secretion from cells is not known. In blood, RBP is bound to the small protein transthyretin, which in addition to associating with RBP functions as a carrier protein for thyroid hormones. Binding of RBP to transthyretin prevents the loss of this smaller protein by filtration in the renal glomeruli. The transthyretin-RBP-retinol complex transports retinol in the circulation and delivers it to target tissues [10].

Important insights into the biological role of RBP have been obtained by studies of mice and humans in which the RBP gene is disrupted. RBP-deficient mice display both reduced blood retinol levels and impaired visual function during the first months of life. When maintained on a vitamin A-sufficient diet, they acquire normal vision by 5 months of age, even though their blood retinol level remains low. A striking phenotype of the RBP-null mice is that they possess larger than normal hepatic vitamin A storage, but are dependent on a continuous dietary intake of vitamin A [11], further proving the importance of RBP as a transporting protein. A study of two human siblings that harbored point mutations in their RBP gene and exhibited undetectable plasma RBP levels revealed that these sisters suffered from night blindness and mild retinal dystrophy but did not exhibit other clinical symptoms of vitamin A deficiency [12]. Taken together, RBP is critical for the mobilization of retinol from hepatic storage pools; however, RBP is not essential for the delivery of retinol to target tissues. Supply of vitamin A to target tissues in the absence of RBP is likely to be accomplished via newly absorbed retinyl esters or β-carotene present in circulating chylomicrons. Increased RBP has been shown to contribute to insulin resistance and type 2 diabetes [11]. All-trans RA has recently been shown to increase insulin sensitivity in diabetic mice while lowering RBP [13]. The effect on binding proteins must be considered when retinoids are used for disease treatment.

3.2. STRA6

The stimulated by retinoic acid gene 6 (STRA6) encodes the cell surface RBP receptor, which binds specifically to RBP and mediates retinol uptake from holo-RBP [14]. STRA6 is a widely expressed transmembrane protein. In mouse mammary epithelial cells, STRA6 expression can be up regulated by Wnt1 and retinoids. In addition, STRA6 mRNA levels are up regulated in mouse mammary gland tumors and human colorectal tumors [15]. Importantly, while the RBP-null mice and humans give rise to relative mild phenotypes, STRA6-null mice develop anophthalmia, congenital heart defects, diaphragmatic hernias, alveolar capillary dysplasia, lung hypoplasia, and mental retardation. These findings suggest that STRA6 may have additional functions that are not related to RBP transport [16].

3.3. CRBP

CRBPs belong to the family of fatty acid binding proteins in which expression of CRBP family members are tissue specific. For example, CRBP-II is expressed only in the enterocytes of the intestine, while CRBP-I and -III are expressed throughout embryonic and adult tissues [17]. Knockout studies for CRBP isoforms have identified differences in function due to altered tissue localization. CRBP-I knockout mice are healthy. However, they have low levels of hepatic retinyl esters [18], and their hepatic lipid droplets appear to be smaller and less abundant than in wild type littermates. CRBP-II-null mice have impaired retinol uptake, but they develop and reproduce normally under vitamin A-enriched diet, albeit with reduced retinol storage [19]. Reduction of vitamin A in the maternal diet of CRBP-II-null mice during gestation results in neonatal mortality immediately following birth [19]. CRBP-III null mice have impaired vitamin A incorporation into milk, but they are otherwise healthy [20]. CRBP-I and CRBP-III compensate for each other to maintain normal retinoid homeostasis, but the compensation is incomplete during lactation [20]. The binding affinity of CRBP-I towards retinol is about 100-fold higher than that of CRBP-II. They display a similar binding affinity towards retinal and CRBP-II associates with retinol and retinal with similar affinities. CRBPs, and especially CRBP-I with its high affinity for retinol, may sequester retinol from its ability to disrupt cell membranes. Epigenetic silencing of CRBP is a common event in human cancers [21]. Silencing CRBP reduces the availability of retinyl esters in the bloodstream and decreases the body’s ability to metabolize retinol [22].

3.4. CRABP

CRABP-I and -II have been identified with a high affinity for all-trans RA. In humans, these isoforms display 74 percent sequence identity and are highly conserved among species; however, these CRABP isoforms display different patterns of expression across cells and developmental stages. In adults, CRABP-I is expressed ubiquitously, while CRABP-II is only expressed in the skin, uterus, ovary, and the choroid plexus. Both CRABPs are widely expressed in the embryo, although they do not usually co-exist in the same cells. The biological functions of CRABPs are not completely understood. In mouse knockout stu dies, disruption of either CRABP-I or -II only display mild defects in limb development [23], which suggests CRABPs may be involved in generation of appropriate RA concentration gradients in the developing limb bud. Both CRABP isoforms are present in cytosol and nucleus and thus may deliver the ligand directly to the nuclear receptor. The differential role of these two binding proteins remains to be studied (reviewed in [24] and [25]). Increased CRABP-I expression may also contribute to RA resistance of cancer cells [26]. The effect of CRABP on cancer therapy deserves more attention.

4. Retinoic Acid Receptors

The major breakthrough in understanding RA’s function occurred upon identifying and cloning the receptors for RA [27, 28]. RA regulates gene expression by binding to its nuclear receptors, which in turn activates transcription of their downstream target genes. Thus, retinoids exert their biological functions primarily by regulating gene expression. This was predicted by Sporn and Roberts in 1983, when they wrote: “Ultimately, it would appear that the problem of the molecular mechanism of action of retinoids in control of differentiation and carcinogenesis is converging on one of the central problems of all biology, the control of gene expression.” [29]

4.1. RAR and RXR

Two distinct classes of receptors for retinoids have been identified: retinoic acid receptors (RAR) and retinoid × receptors (RXR). Each class of receptor contains three subtypes - α, β, and γ. RARs can be activated by both all-trans and 9-cis-RA, while, RXRs are exclusively activated by 9-cis RA. However, due to the conversion of all-trans to 9-cis RA, high concentrations (10−5 M) of all-trans RA can also activate gene transcription in cells transfected with RXRs [30].

RXRs can form homo- and heterodimers with other receptors. In fact, RXRs are promiscuous receptors forming heterodimers with many different kinds of receptors, which include receptors for fatty acids [peroxisomal proliferator activated receptors (PPAR)], bile acids [farnesoid × receptor (FXR)], oxysterols [liver × receptor (LXR)], xenobiotics [pregnane × receptor (PXR) and constitutive androstane receptor (CAR)], vitamin D [vitamin D receptor (VDR)], and RA (RAR). RXRs can also form homodimers. Hypervitaminosis A leads to bone fracture suggesting that vitamin A and D compete for the same receptor [31]. Within these heterodimers, RXRs can exist as both active and silent partners. When it serves as an active partner, 9-cis RA and the ligand for the heterodimeric partner can activate the heterodimer, and addition both ligands give synergistic induction in gene transcription. For example, RXR is an active partner for PPAR. Similarly, heterodimeric complexes of RXR with LXR or FXR also retain 9-cis RA responsiveness. Thus, RAs can regulate PPAR- and FXR-mediated pathways [32]. Recently, we demonstrated that RAs could also activate PXR-, VDR, and CAR-mediated signaling and thus regulated xenobiotic metabolism and potentially its own oxidation [3335]. When RXR serves as a silent partner, the heterodimer of RXR and its partner does not respond to RA. Regardless of their active or silent role, RXRs must be present in order to exert biological actions of various nuclear receptors. Using hepatocyte RXRα-deficient mice [36, 37], we have demonstrated that RXRα does play vital roles in xenobiotic (alcohol, acetaminophen) and endobiotic (fatty acid, cholesterol, amino acid, and carbohydrate) metabolism [3340]. Thus, RXR functions as an auxiliary factor and determines the effects of other hormones, making RXR a master regulator. The structure of nuclear receptors is summarized in recent review articles [7, 38].

Existing data suggest that the binding protein and receptor work together to exert the specific effect of RAs. For example, RAs can bind to both PPARβ, the receptor for fatty acids, and RAR. Fatty acid-binding protein 5 (FABP5) and CRABP-II are specific binding proteins that channel RAs from the cytosol into the nucleus for binding to either PPARβ or RAR, respectively [39]. The ratio of FABP5/CRABP-II concentrations determines which receptor is activated. By activating PPARβ, RAs induce expression of genes affecting lipid and glucose homeostasis, such as the insulin-signaling gene pyruvate dehydrogenase kinase 1 (PDK1), which enhances insulin action. Hence, RAs stimulate lipolysis and reduce triglyceride content. RA implantation into obese mice causes up regulation of PPARβ as well as an increased expression of PPARβ target genes, including PDK1, which led to weight loss [40].

4.2. Receptor Isoforms

Multiple receptor isoforms for both RAR and RXR have been identified. RAR isoforms are either transcribed from two different promoters or produced by alternative splicing. There are two major isoforms for RARα (α1 and α2) and RARγ (γ1 and γ2) and five major isoforms for RARβ (β1–β4 and β1′). RAR isoforms can be classified as those which are transcribed from either P1 promoter (class I: RARα1, β1, and β3, γ1) or P2 promoter (class II: RARα2, β2 and β4, γ2). All class II isoform promoters have an RA response element and are RA inducible. Two major isoforms for RXRα (α1 and α2), RXRβ (β1 and β2), and RXRγ (γ1 and γ2) have been identified. These RXR isoforms have different amino-terminal regions [41]. RARs and RXRs are expressed in a tissue-specific manner during development reflecting their pleiotropic roles. The tissue distribution pattern of these receptors is summarized in a review article entitled “Developmental Expression of Retinoic Acid Receptors (RARs)” [42].

Knockout studies have been generated in order to determine the function of RAR and RXR in vivo. RARα-null mutant males are sterile due to degeneration of the seminiferous epithelium, inhibiting spermatogenesis[43]. RARβ-null mice display abnormalities in the vitreous body of the eye and impaired locomotion and motor coordination [44], while ablation of RARγ causes both skeletal and epithelial defects [45]. The loss of RXRα was found to be lethal during fetal life due to hypoplasia of the myocardium [46]. Moreover, fetuses lacking RXRα have ocular malformation [47]. In addition, it has been shown that RXRα is involved in mediating the teratogenic effect of retinoids [48]. The ablation of RXRβ led to approximately 50% lethality of mice in utero. Similar to RARα-null mice, RXRβ-null mice are also sterile: however, this is due to testicular defects and abnormal spermatid maturation [49]. Moreover, RXRβ mutation causes abnormal lipid metabolism in Sertoli cells, suggesting functional interactions of RXRβ with other nuclear receptors that control lipid metabolism [50]. In contrast, RXRγ-null mice are fertile but have elevated serum levels of both L-thyroxine and thyroid-stimulating hormone (TSH), as well as increased metabolic rates when compared to wild type animals [51]. The function of RARs during embryonic development is summarized in a recent review article [52].

4.3. Non-genomic Actions

Retinoids also exert their effects via transcription independent pathway, which can occur in the presence or absence of nuclear receptor. Retinoids mediated via RARs inhibit AP-1-regulated cell proliferation [53]. RA also inhibits NFκB activity in mice [54]. Mediated through RARβ, RA changes the intracellular Ca++ level and thus controls PI3K activation in neural cell [5557]. RARα can interact with mRNA in cytoplasm and control translation [58, 59]. Via nuclear receptor independent pathway, RA rapidly activates cAMP response element (CREB) binding protein in human bronchial epithelial cells [60]. These mechanisms add a new dimension to the action of retinoids.

5. Retinoids and Cancer

Retinoids are widely used to treat visual and dermatological diseases. Their effect on cancer prevention and treatment has received a lot of attention. This review focuses on the action of retinoids on cancer. Retinoids have been used as potential chemotherapeutic or chemopreventive agents because of their differentiation, anti-proliferative, pro-apoptotic, and anti-oxidant effects. Epidemiological studies show that lower vitamin A intake results in a higher risk of developing cancer, which aligns with observations of vitamin A-deficient animals [61]. Altered expression of RA receptors is also associated with malignant transformation of animal tissues or cultured cells. Furthermore, retinoids suppress carcinogenesis in tumorigenic animal models for skin, oral, lung, breast, bladder, ovarian, and prostate [6268]. In humans, retinoids reverse premalignant human epithelial lesions, induce the differentiation of myeloid cells, and prevent lung, liver, and breast cancer [6973].

The following is a summary of how major retinoids may work in cancer treatment or prevention.

5.1. All-trans RA (tretinoin)

All-trans RA is the most abundant natural retinoid and has been widely studied for many years. It is currently in clinical trials for the treatment of lymphoma, leukemia, melanoma, lung cancer, cervical cancer, kidney cancer, neuroblastoma, and glioblastoma. The most effective clinical usage of all-trans RA in human disease was demonstrated in treatment of a rare leukemia, acute promyelocytic leukemia (APL). APL is characterized by selected expansion of immature myeloid precursors or malignant myeloid cells blocked at the promyelocytic stage of hemopoietic development. APL cells invariably express aberrant fusion proteins involving the DNA and ligand binding domain of RARα [74, 75]. Other fusion partners include the promyelocytic leukemia zinc finger gene, the nucleophosmin gene, the nuclear mitotic apparatus gene, and the Stat5b gene, while the most common fusion partner is promyelocytic leukemia protein (PML). The PML-RARα chimeric receptor is created by a balanced reciprocal chromosomal translocation, t(15;17)(q22:q11). The expressed PML-RARα chimeric receptor alters normal function of RARs. PML-RARα can form a homodimer through the coiled-coil motif of PML, inhibiting RARα’s ability to bind to RA responsive elements, thereby preventing activation of downstream target genes [76, 77]. In addition, RXR is an essential component of the oncogenic PML/RARα complex suggesting RXR can be a drug target for APL [78, 79]. In 1995, the FDA approved all-trans RA for treating APL. The all-trans RA-induced differentiation of APL cells is due to both its ability to promote the degradation of the mutant PML-RARα and the dissociation of its co-repressors [80]. All-trans RA also causes cell cycle arrest at G1 phase and inhibits cell proliferation [81]. In addition, high concentration of all-trans RA induces post-maturation apoptosis of APL-blasts through the induction of the tumor-selective death ligand tumor necrosis factor-related apoptosis-inducing ligand TRAIL [82].

RA syndrome is a life-threatening complication seen in APL patients treated with all-trans RA. This syndrome is characterized by dyspnea, fever, weight gain, hypotension, and pulmonary infiltrates. It can be effectively treated by giving dexamethasone and holding off all-trans RA treatment in severe cases. An elevated white count is sometimes associated with this syndrome, but is not a prerequisite. The etiology of RA syndrome is not clear; several causes have been speculated including a capillary leak syndrome from cytokine release from the differentiating myeloid cells. Alternatively, all-trans RA may cause the maturing myeloid cells to acquire the ability to infiltrate organs such as the lung [83].

5.2. 9-cis RA (alitretinoin)

9-cis RA differentiates itself from all-trans RA in its ability to activate both RAR and RXR. In addition, 9-cis RA activates PPAR, FXR, PXR, VDR, and CAR via RXR. In preclinical studies, 9-cis RA is effective in the prevention of mammary and prostate cancer [84, 85] and it has also been FDA-approved for the topical treatment of cutaneous lesions of Kaposi’s sarcoma [86]. In addition, 9-cis RA and all-trans RA can individually induce apoptosis of human liver cancer cells [87]. 9-cis RA not only regulates nuclear genes, but also mitochondria gene transcription [88].

5.3. 13-cis RA (isotretinoin)

13-cis RA is unique that it exhibits immunomodulatory and anti-inflammatory responses. It inhibits ornithine decarboxylase, thereby decreasing polyamine synthesis and keratinization [89]. 13-cis RA noticeably reduces the production of sebum and shrinks the sebaceous glands [90]. It stabilizes keratinization and prevents comedones formation [91, 92]. The exact mechanism of action is unknown. This combination of regulating proliferation, differentiation, and inflammation could make 13-cis RA a more effective drug in comparison to other retinoids, which may cause inflammation and irritation [93].

13-cis RA is in clinical trial for different types of cancers, and thyroid cancer received a lot of attention. In follicular thyroid cancer cells, 13-cis RA induces radioiodine avidity of cells formerly unable to accumulate radioiodine [94]. In human thyroid carcinoma cell lines, retinoids induce the expression of type I iodothyronine-5′-deiodinase and sodium/iodide-symporter, which are the thyroid differentiation markers [95]. However, approximately 30% of thyroid tumors dedifferentiate after treatment and thus develop into highly malignant anaplastic thyroid carcinomas [96]. 13-cis RA is also used to treat non-operable thyroid follicular tumors, which fail to uptake radioiodine. 13-cis RA increases the radioiodide uptake in some patients. The beneficial outcome of this treatment was interpreted as partial re-differentiation of thyroid cancer cells. This effect of 13-cis RA requires the existence of functional RXR [96]. The effect of 13-cis RA on thyroid cancer has been reviewed extensively [97]. Besides thyroid cancer, utilizing 13-cis RA for maintenance therapy has significantly improved the outcome of patients with a high-risk form of neuroblastoma [98]. Along the same line of work, Krüppel zinc-finger protein ZNF423 is critical for RA signaling and is likely a prognostic marker for neuroblastoma [99]. 13-cis RA is also effective in preventing head and neck cancer, which is discussed below.

5.4. Synthetic Retinoids

N-(4-hydroxyphenyl) retinamide (Fenretinide or 4HPR) was first synthesized in the late 1960s by R. W. Pharmaceuticals. Since then, the biological properties of fenretinide have been of great interest. Currently, fenretinide is one of the most promising clinically tested retinoids. The modification of the carboxyl end of all-trans RA with an N-4-hydroxyphenyl group resulted in increased efficacy as a chemoprevention agent as well as reduced toxicity when compared with other retinoids [100]. Animal models have demonstrated that treatment with fenretinide prevents chemically induced cancers of the breast, prostate, bladder, and skin [101104]. Furthermore, the combination of tamoxifen with fenretinide produces efficacy greater than either chemical alone [105].

Natural retinoids like all-trans RA induce differentiation and/or cytostasis in target cells [106108], while fenretinide has distinct biologic effects including the induction of apoptosis by generating reactive oxygen species (ROS) and lipid second messengers [104]. The apoptotic effect of fenretinide has been documented in a variety of cancer cells including transformed T cells, B cells and breast epithelial cells, as well as bladder, breast, cervical, colon, embryonal, esophageal, head and neck, lung, ovarian, pancreatic, prostate, and skin carcinomas [100]. Furthermore, fenretinide does not induce point mutations or chromosomal aberrations, and is therefore not genotoxic [109]. These qualities suggest that fenretinide could be used for a long-term chemopreventive modality. In animal models, fenretinide has demonstrated chemopreventive efficacy against carcinogenesis of the breast [110], prostate, pancreas, and skin [104, 111, 112]. Moreover, in a clinical setting, fenretinide slowed the progression of prostate cancer in men diagnosed with an early stage of the disease [113]. Fenretinide protected against the development of ovarian cancer and a second breast malignancy in premenopausal women who had been treated to prevent the progression of early-stage breast cancer [114]. It also prevented relapse and the formation of secondary primary lesions in patients following the surgical removal of oral leukoplakia [115]. Recent studies also illustrated the anti-angiogenic [116] and anti-fibrotic [117] effect of fenretinide. Furthermore, long-term fenretinide treatment prevents high-fat diet-induced obesity, insulin resistance, and hepatic steatosis [118].

The mechanisms associated with fenretinide-induced apoptosis have been explored, but are not well-understood [100]. The components that lead to ROS generation and cause cell death are largely unknown. Depending on cell types and models used, the effect of fenretinide has been shown to be RARβ-dependent or -independent [119]. Our data showed that fenretinide-induced apoptosis of human liver cancer cells was RARβ-dependent [120]. Furthermore, induction and cytoplasmic localization of Nur77 dictates the sensitivity of liver cancer cell to fenretinide-induced apoptosis [121]. It seems that fenretinide enriches the cytoplasmic Nur77 to target mitochondria and induce cell death. The relationship between RARβ and Nur77 in mediating fenretinide-induced apoptosis remains to be determined.

A retinoid-related molecule 6-[3-(1-adamantyl)-4-hydroxyphenyl]-2-naphthalenecar-boxylic acid (AHPN) (also called CD437) and it’s analog (E)-4-[3-(1-adamantyl)-4-hydroxyphenyl]-3-chlorocinnamic acid (3-Cl-AHPC) also have Nur77-dependent apoptotic effects [122124]. AHPN is structurally distinct from fenretinide. AHPN-induced apoptosis activates JNK [125127], which is required for maximal apoptosis induction and precedes mitochondrial depolarization. Induction of apoptosis of breast and prostate cancer cells by AHPN is also associated with its inhibition of Akt activity [128]. Thus, induction of JNK and inhibition of Akt phosphorylation of Nur77 contribute to Nur77 nuclear export mediated by AHPN [129].

While many synthesized RAs are promising for cancer treatment, only a few are FDA-approved or currently undergoing clinical trials for cancer therapy. A number of retinoids, which have been FDA-approved for dermatological purposes, have potential for cancer treatment. Bexarotene (Targretin) is a synthetic retinoid approved by the FDA to treat skin problems caused by cutaneous T-cell lymphoma that are unresponsive to other treatments [130]. Other synthetic retinoids, such as TAC-101 (Taiho Pharmaceutical, Tokyo, Japan) has shown efficacy in inhibiting tumor growth in the liver and markedly increases survival in both the primary HCC and metastatic colon cancer models [131]. TAC-101 is currently in phase II trial for hepatocellular carcinoma and has shown good preliminary success [132]. Another, Tazarotene (AVAGE) (Allergan, Irvine, CA) is in phase I trials for the treatment of lymphoma [133]. Please see table 1 for a brief characterization of some of retinoids that are in use or in clinical trials.

Table 1.

Retinoids: action, targets, and usage

Drug Target Effect Current Clinical Trials Reference
All-trans RA (Tretinoin)
graphic file with name nihms229611t1.jpg
RAR
  • Natural ligand of RAR

  • Through RAR, all-trans RA exerts a variety of affects from transcriptional control of growth and differentiation genes to AP-1 inhibition

  • Launched: Acne, APL, Warts, and Psoriasis

  • Phase I: Asthma

  • Phase II: Rosacea, Amyotrophic Lateral Sclerosis, Neuroblastoma, Head and Neck Cancer, Photodamage, Lymphoma, Melanoma, Lung Cancer, Kidney Cancer, Emphysema, and Glioblastoma

  • Phase III: Skin Aging and Lung Cancer

[8, 192]
9-cis RA (Alitretinoin)
graphic file with name nihms229611t2.jpg
RAR and RXR
  • Activate RXR and thus activate the heterodimer of RXR and PPAR, VDR, LXR, FXR and PXR

  • This expands its effects to lipid metabolism, drug interactions, etc.

  • Launched: Acne, APL, Psoriasis, and Kaposi’s Sarcoma

  • Phase I: Breast Cancer, Low HDL Cholesterol, and Asthma

  • Phase III: Eczema and Hand Dermatitoses

[8, 192]
13-cis RA (Isotretinoin)
graphic file with name nihms229611t3.jpg
RAR
  • Reduces the production of sebum and shrinks the sebaceous glands

  • Stabilizes keratinization and prevents comedones formation.

  • Decrease cyclin B1 and Bcl-2 expression

  • Launched: Acne

  • Phase II: Photodamage, Cervical Cancer, Lung Cancer, Prostate Cancer, Lymphoma, Glioblastoma, Neuroblastoma, Skin Cancer, Penile Cancer, and Emphysema

  • Phase III: Brain and Central Nervous System Tumors, Kidney Cancer, Lung Cancer, Rosacea, and Head and Neck Cancer

[193]
Ethyl 6-[2-(4,4-dimethyl-2,3-dihydrothiochromen-6-yl)ethynyl]pyridine-3-carboxylate (Tazarotene)
graphic file with name nihms229611t4.jpg
RARβ and RARγ
  • Chemoprevention of basal cell carcinoma in the Ptch−/−mouse model

  • Clinical study revealed efficacy in human basal cell carcinoma treatment

  • Down regulates Gli1 and up regulates CRABP2

  • Launched: Acne, Psoriasis, and Photodamage

  • Phase II: Lymphoma

[133]
MDI 301
graphic file with name nihms229611t5.jpg
Not Identified
  • Anti-neoplastic in breast, prostate cancer cell lines, neuroblastoma, and leukaemia cell lines

  • Has topical efficacy and lack of skin irritation

  • Preclinical: Acne, Cancer, Photodamage and Psoriasis

[194]
R667
graphic file with name nihms229611t6.jpg
RARγ
  • Regenerates lung tissues and restores lung function in animal models

  • Phase II: Emphysema

[195]
4-[1-(3,5,5,8,8-pentamethyl-6,7-dihydronaphthalen-2-yl)ethenyl]benzoic acid (Bexarotene)
graphic file with name nihms229611t7.jpg
RXR
  • G1 and/or G2/M arrest and down regulation of cyclin D

  • Induces apoptosis and differentiation with activation of caspase-3 and cleavage of PARP, and inhibition of surviving

  • Prevents and overcomes multidrug resistance by modulating MDR1 expression

  • Inhibits angiogenesis and metastasis by reduction in MMPs, VEGF, and EGF as well as increase in TIMPs secretion

  • Launched: Cutaneous T-cell lymphoma

  • Phase 0: Thyroid Cancer

  • Phase II: Lung Cancer, Leukemia, Psoriasis, Cushing’s Disease, and Breast Cancer

  • Phase III: Schizophrenia

[130]
8-(3′,4′-dihydro-1′ (2′H)-naphthalen-1′-ylidene)-3,7-dimethyl-2,4,6-octatrienoic acid (9-cis UAB30)
graphic file with name nihms229611t8.jpg
RXR
  • Down regulates DNA methyltransferases

  • Anti-telomerase activity

  • Preclinical: Cancer

[196, 197]
N-(4-Hydroxyphenyl)retinamide (Fenretinide/4HPR)
graphic file with name nihms229611t9.jpg
RAR
  • ROS generation

  • RARβ activation

  • Nur77 nuclear export

  • Ceramide and Ca++ mobility

  • Caspase cleavage

  • Phase I: Neoblastoma,

  • Phase II: Brain and Central Nervous System Tumors, Lymphoma, Prostate Cancer, Head and Neck Cancer, Melanoma, Ovarian Cancer, Kidney Cancer, Lung Cancer, Geographic Atrophy, and Insulin Resistance

  • Phase III: Breast Cancer, Bladder Cancer, Cervical Cancer, and Ewing’s Sarcoma

[100, 120, 121]
6-[3-(1-adamantyl)-4-methoxyphenyl]naphthalene-2-carboxylic acid (Adapalene)
graphic file with name nihms229611t10.jpg
RAR
  • More chemically stable and lipophilic than all-trans RA

  • Modulates cellular keratinization

  • Inhibits lipooxygenase activity and oxidative metabolism of arachidonic acid to exert anti-inflammatory effect

  • Launched: Acne

  • Phase III: Skin Aging

  • No development reported: Multiple Sclerosis

[198]
Anhydroretinol
graphic file with name nihms229611t11.jpg
Not Identified
  • Metabolite of vitamin A

  • Induces human B lymphoblastoid 5/2 cell death by increasing intracellular oxidative stress in a time- and dose-dependent manner

No known trial currently underway [199]
6-[3-(1-adamantyl)-4-hydroxyphenyl]-2-naphthalenecarboxylic acid (AHPN/CD437)
graphic file with name nihms229611t12.jpg
RARγ
  • Induces apoptosis in various cancer cell lines

  • Causes ER stress

  • Nur77 nuclear export

  • SHP mitochondrial association.

  • Shows apoptogenic activity in retinoid resistant cells and in RARγ-negative cells

No known trial currently underway [200, 201]
3-[4-[3-(2-adamantyl)-4-hydroxyphenyl]phenyl]prop-2-enoic acid (AHPC/ ST1926)
graphic file with name nihms229611t13.jpg
Not Identified
  • Similar to AHPN

  • Causes genomic instability leading to apoptosis

  • Phase I: Ovarian cancer

[202]
3-[4-[3-(1-adamantyl)-4-hydroxyphenyl]-3-chlorophenyl]prop-2-enoic acid (3-Cl-AHPC)
graphic file with name nihms229611t14.jpg
Small Heterodimer Partner (SHP)
  • Enhanced interaction between SHP and a multiprotein repressor complex containing the repressor mSin3A, the NR corepressor NCoR, histone deacetylase 4, and heat shock protein 90

  • Attenuation of proto-oncogene c-Myc mRNA and protein expression

  • NFκβ activation and increase c- Fos/c-Jun expression

No known trial currently underway [203]
MX781 (CD 2674)
graphic file with name nihms229611t15.jpg
RAR and RXR antagonist
  • Induction of apoptosis by caspase 2

  • Inhibits Iκκβ

No known trial currently underway [204]
3,7,11,15-tetramethylhexadeca-2,4,6,10,14-pentaenoic acid (Acyclic retinoid/NIK333/polyprenoic acid)
graphic file with name nihms229611t16.jpg
RXR and RAR
  • Induces growth inhibition by suppressing the VEGF-MAPK and Ras-MAPK pathways.

  • Phase II: Hepatocellular Carcinoma

[205, 206]
(4-nitrophenyl)amino][2,2,4,4-tetramethylthiochroman-6-yl)amino]methanethione (SHetA2)
graphic file with name nihms229611t17.jpg
Not Identified
  • Sensitizes ovarian cancer cells to TNFα induction of the extrinsic apoptosis pathway

  • Exhibits receptor independent mitochondria mediated apoptosis

No known trial currently underway [207]
4-[3,5-bis(trimethylsilyl) benzamido] benzoic acid (TAC 101)
graphic file with name nihms229611t18.jpg
RARα
  • AP-1 inhibition

  • Inhibits tumor growth in the liver, exhibits low toxicity, and markedly increases survival in both the primary HCC and metastatic colon cancer models

  • Phase II: Hepatocellular Carcinoma

[132]

Note: This table represents the most studied retinoids over the past few years. Retinoids that have not been published on in the last five years were not included.

Retinoids are frequently tested in head/neck, skin, lung, and breast cancer. The followings summarize those findings.

5.5. Retinoids in Head/Neck and Skin Cancers

While RAs are considered effective for treating cancer, they are also used in cancer prevention of head/neck and skin cancers. Leukoplakia, actinic keratosis, and cervical dysplasia are the three precancerous lesions that can be effectively treated by the classical retinoids, such as 13-cis RA [134, 135]. In cancer prevention, classical retinoids delay the development of skin cancer in individuals with xeroderma pigmentosum, an inherited predisposition to UV-induced cancer [6]. RARβ plays a critical role in mediating the growth-inhibitory effect of retinoids in various types of cancer cells, which include breast cancer [136, 137], lung cancer [138], ovarian cancer [139], prostate cancer [140], neuroblastoma [141], renal cell carcinoma [142], pancreatic cancer [143], liver cancer [144], and head and neck cancer [145]. In oral cancer, after 3 months of high-dose 13-cis RA treatment, a significant induction of RARβ was noted and its expression level correlated with the intracellular RA levels [146]. Among the RARβ isoforms, RARβ2 is the most abundant and inducible form and has tumor suppressor activity. In contrast to β2, RARβ4 has oncogenic activity [147]. Thus, β2 and β4 have antagonistic effect in carcinogenesis. Benzo(a)pyrene diolepoxide, a carcinogen presented in tobacco, suppresses RARβ2 expression. Moreover, up regulation of RARβ is associated with a positive clinical response to retinoids in patients with premalignant oral lesions [148, 149]. The expression of RARβ can be transcriptionally regulated by RARβ itself and epigenetically regulated by DNA methylation and histone acetylation [150, 151].

In skin cancer, RARα and γ mRNA levels are decreased in benign skin tumors in mice [152] and the mRNA level is essentially absent in human undifferentiated squamous cell carcinomas [153]. Priming human skin with RA prevents AP-1-induced matrix-degrading metalloproteinase genes caused by UV irradiation [154, 155]. Irradiation human skin using UV rapidly reduces RARα and γ and loses RA-responsive gene expression in human skin [156]. Essentially, UV irradiation causes a functional vitamin A deficiency in skin, which begins at UV doses that do not cause detectable sunburn. In animal models, RA dramatically delays and reduces chemical-induced squamous carcinoma [157, 158]. The anti-tumor effect could be due to the anti-AP1 activity of the RA [159, 160]. In humans, retinoids reverse photodamage to skin [161].

5.6. Retinoids and Lung Cancer

Beta-carotene and retinoids were the most promising agents against common cancers when the National Cancer Institute mounted a substantial program of population-based trials in the early 1980s [162]. However, major lung cancer chemoprevention trials demonstrated these agents had no beneficial effects in preventing cancer. Furthermore, these trials showed significant increases in lung cancer incidence, cardiovascular disease, and total mortality. Several intervention trials have been completed; most of them included supplementation with β-carotene with or without other nutrients. The α-Tocopherol, β-Carotene Cancer Prevention Study (ATBC) was a randomized double-blind placebo-controlled primary-prevention trial to test whether daily supplementation with α-tocopherol (50 mg/day), β-carotene (20 mg/day), or both, would reduce the incidence of lung cancer in 29,133 male smokers [163]. The β-Carotene and Retinol Efficacy Trial (CARET) enrolled 18,314 men and women at high risk for lung cancer. This trial evaluated combined supplementation with β-carotene (30 mg/day) and retinyl palmitate (25,000 IU/day) for an average of 4 years [164]. The Physicians’ Health Study (PHS) was a randomized double blind placebo-controlled trial of β-carotene (50 mg on alternate days) supplementation for 12 years among 22,071 male physicians who were 40–84 years of age [165]. These studies concluded that retinoids reduce tumor occurrence and mortality in non-smokers, were beneficial for former smokers, but increased the risk of lung cancer in smokers. A later EUROSCAN study also showed a higher incidence of lung cancer among smoking participants who received β-carotene [166]. The highest serum concentrations of β-carotene during supplementation were observed in ATBC and CARET (medium 3.0 and 2.1 mg/l, respectively), whereas notably lower concentrations were found in the PHS (mean 1.2 mg/l). Thus, high serum concentrations of β-carotene might be detrimental to lung cancer patients who are current smokers. The mechanism that explains the higher incidence of lung cancers observed in smokers who received β-carotene is not clear. Our recent data indicate that retinoids via PXR, CAR, and VDR regulate the expression of cytochrome P450 gene expression and thus xenobiotic metabolism [3335]. Thus, it is possible that retinoids may facilitate the bio-activation of the carcinogens in cigarettes and enhance lung carcinogenesis.

Deletion of chromosome 3p14-p25 is frequently found in non-small cell lung carcinoma (NSCLC) [167]. The RARβ2 gene is mapped at 3p24, and its expression is frequently suppressed in lung cancer cell lines [168, 169]. Reduced expression of RARβ2 mRNA is also found in bronchial biopsy specimens from heavy smokers [170], head and neck [171], and breast [172] as well as liver cancer cell lines [144]. Epigenetic modification of the RARβ gene causes reduced RARβ2 expression, which may lead to carcinogenesis. Although the aberrant expression of RARβ is associated with lung cancer, the majority of human and experimental NSCLC are resistant to all-trans RA, bexarotene, and CD437 [173, 174]. Reduced expression of RARβ1′ may account for the resistance of lung cancer cells to RA [175]. In addition, the expression of RARβ is positively associated with the expression of COUP-TF and the growth inhibition by RA in lung cancer cell lines [176]. COUP-TF is required for RA to induce RARβ expression, growth inhibition, and apoptosis in cancer cells. In addition, COUP-TF induces transcriptional activity of the RARβ promoter in a RA- and RARα-dependent manner through its binding to a DR-8 element in the RARβ promoter. Thus, COUP-TF is an accessory factor for RARα to recruit its coactivator and induces RARβ promoter activity [177].

5.7. Retinoids and Breast Cancer

Almost every major retinoid is currently in clinical trials by itself or in combination with interferons and estrogen antagonists to treat or prevent the progression of breast cancer [70, 178, 179]. Retinoids are able to inhibit mammary gland cancer in animal models and human breast cancer. They are effective inhibitors of breast cancer cells at the early stages of tumor progression, but their effectiveness diminishes as the tumors become more aggressive. RARs are differentially expressed in lactating and post-lactating glands. Rat post-lactating mammary glands express all RAR and RXR subtypes, whereas non-lactating mammary tissues only express RARα and RXRα subtypes [180]. Retinoid receptors are also differentially expressed in normal and malignant epithelial cells and are implicated in carcinogenesis [179]. However, hormone-dependent and -independent breast cancer cells have the similar expression pattern of RARα, RARγ, RXRα, and RXRβ [181]. Growth inhibition of breast cancer cells by RA has been associated with induction of the expression of RARβ. Conversely, RARβ gene expression is up regulated in normal mammary epithelial cells upon RA treatment. Deficiency of the RARβ expression and deficient responsiveness of retinoids via RARβ may account for decreased treatment efficacy in patients with advanced breast tumors [182].

9-cis RA and LGD1069 (Targretin, Bexarotene), a synthetic RXR-selective agonist, are superior to all-trans RA as a chemopreventive compound in the 1-methyl-1-nitrosourea (MNU)-induced rat mammary gland carcinoma model [183, 184]. Targretin is well tolerated during chronic therapy with no classic signs of retinoid-associated toxicities. In animal mammary gland carcinoma models, other promising retinoids are retinyl acetate or fenretinide. The potency of retinoids to inhibit proliferation of breast cancer cells was demonstrated when retinoids were administered either alone or in combination with antiestrogens [70]. Tamoxifen and fenretinide combination therapy is an active treatment regimen in metastatic breast cancer patients, but not in estrogen receptor negative metastatic breast cancer or in patients whose disease had progressed after tamoxifen treatment [185]. Retinoids do not require estrogen receptors for their actions; they may affect neoplastic transformation in estrogen-negative cells, in contrast to tamoxifen, whose primary mechanism of action is through estrogen receptors [186, 187]. In spite of this, retinoids and rexinoids seem to be more active in estrogen-positive than in estrogen-negative precancerous tissue [188]. Furthermore, retinoids can promote apoptosis of breast cancer cells and that effect is RARβ dependent. Recent study showed that the estrogen-responsive B box protein (EBBP) restores retinoid sensitivity in retinoid-resistant cancer cells via effects on histone acetylation [189]. Despite a number of important findings achieved recently by many laboratories, the precise mechanism(s) by which natural or synthetic retinoids inhibit the growth of breast cancer cell has not been completely elucidated. Great interest has been recently focused on rexinoids in combination with selective estrogen receptor modulators, which dramatically inhibit breast cancer cell growth and induce apoptosis even with intermittent administration [179].

6. Conclusions

Retinoids have a wide spectrum of functions. They are essential for normal development, but can cause congenital abnormality. They are critical for growth, but also have apoptotic effect. They have anti-proliferative and differentiation effects thus are used for cancer prevention and treatment, but they potentially can also increase the risk of cancer. Our recent publication indicating hepatocyte RXRα dictates hepatocyte proliferation during liver regeneration clearly indicates that activation of RXRα may have beneficial (tissue repair and regeneration) as well as detrimental (hepatomegaly) effect [190]. Because of wide usage of retinoids, their effects in cell proliferation cannot be ignored [191]. Since retinoids can activate multiple nuclear receptor mediated pathways, it is crucial to identify the specificity of retinoid in activation of each nuclear receptor-mediated pathways. Recent progress in high-throughput chromatin immunoprecipitation sequencing (ChIP-seq) can help us to identify ligand/nuclear receptor-dependent signaling in vivo, which in turn assist us to design or utilize nuclear receptor specific ligand in order to avoid unwanted effects. In addition to gene transcription, the non-genomic signaling of retinoids deserves more attention [192]. By using ChIP-seq data with microarray or protemomic data, we should be able to identify genomic vs. non-genomic signaling. Knowledge earned using this type of approach can pave the way for the synthesis of new pharmacological agents with extended therapeutic repertoire, reduced toxicity, and improved therapeutic index.

Acknowledgments

This work was supported by NIH grants CA53596 and AA14147. The authors thank Dr. Rebecca Marquez, Mr. David Johnson, and Ms. Julia Straud for editing this manuscript.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Lanska DJ. Chapter 29 Historical aspects of the major neurological vitamin deficiency disorders Overview and fat-soluble vitamin A. Handb Clin Neurol. 2009;95:435–444. doi: 10.1016/S0072-9752(08)02129-5. [DOI] [PubMed] [Google Scholar]
  • 2.Voss HE, Lunin Nicolai I. 1853–1937; a biographical assay. J Am Diet Assoc. 1956;32:317–320. [PubMed] [Google Scholar]
  • 3.Napoli JL. Biochemical pathways of retinoid transport, metabolism, and signal transduction. Clin Immunol Immunopathol. 1996;80:S52–62. doi: 10.1006/clin.1996.0142. [DOI] [PubMed] [Google Scholar]
  • 4.Achkar CC, Derguini F, Blumberg B, Langston A, Levin AA, Speck J, Evans RM, Bolado J, Jr, Nakanishi K, Buck J, Gudas LJ. 4-Oxoretinol, a new natural ligand and transactivator of the retinoic acid receptors. Proc Natl Acad Sci U S A. 1996;93:4879–4884. doi: 10.1073/pnas.93.10.4879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Buck J, Derguini F, Levi E, Nakanishi K, Hammerling U. Intracellular signaling by 14-hydroxy-4,14-retro-retinol. Science. 1991;254:1654–1656. doi: 10.1126/science.1749937. [DOI] [PubMed] [Google Scholar]
  • 6.Altucci L, Leibowitz MD, Ogilvie KM, de Lera AR, Gronemeyer H. RAR and RXR modulation in cancer and metabolic disease. Nat Rev Drug Discov. 2007;6:793–810. doi: 10.1038/nrd2397. [DOI] [PubMed] [Google Scholar]
  • 7.de Lera AR, Bourguet W, Altucci L, Gronemeyer H. Design of selective nuclear receptor modulators: RAR and RXR as a case study. Nat Rev Drug Discov. 2007;6:811–820. doi: 10.1038/nrd2398. [DOI] [PubMed] [Google Scholar]
  • 8.Barnard JH, Collings JC, Whiting A, Przyborski SA, Marder TB. Synthetic retinoids: structure-activity relationships. Chemistry. 2009;15:11430–11442. doi: 10.1002/chem.200901952. [DOI] [PubMed] [Google Scholar]
  • 9.Wolf G. Identification of a membrane receptor for retinol-binding protein functioning in the cellular uptake of retinol. Nutr Rev. 2007;65:385–388. doi: 10.1301/nr.2007.aug.385-388. [DOI] [PubMed] [Google Scholar]
  • 10.Zanotti G, Berni R. Plasma retinol-binding protein: structure and interactions with retinol, retinoids, and transthyretin. Vitam Horm. 2004;69:271–295. doi: 10.1016/S0083-6729(04)69010-8. [DOI] [PubMed] [Google Scholar]
  • 11.Yang Q, Graham TE, Mody N, Preitner F, Peroni OD, Zabolotny JM, Kotani K, Quadro L, Kahn BB. Serum retinol binding protein 4 contributes to insulin resistance in obesity and type 2 diabetes. Nature. 2005;436:356–362. doi: 10.1038/nature03711. [DOI] [PubMed] [Google Scholar]
  • 12.Seeliger MW, Biesalski HK, Wissinger B, Gollnick H, Gielen S, Frank J, Beck S, Zrenner E. Phenotype in retinol deficiency due to a hereditary defect in retinol binding protein synthesis. Invest Ophthalmol Vis Sci. 1999;40:3–11. [PubMed] [Google Scholar]
  • 13.Manolescu DC, Sima A, Bhat PV. All-trans retinoic acid lowers serum retinol-binding protein 4 concentrations and increases insulin sensitivity in diabetic mice. J Nutr. 2010;140:311–316. doi: 10.3945/jn.109.115147. [DOI] [PubMed] [Google Scholar]
  • 14.Kawaguchi R, Yu J, Honda J, Hu J, Whitelegge J, Ping P, Wiita P, Bok D, Sun H. A membrane receptor for retinol binding protein mediates cellular uptake of vitamin A. Science. 2007;315:820–825. doi: 10.1126/science.1136244. [DOI] [PubMed] [Google Scholar]
  • 15.Szeto W, Jiang W, Tice DA, Rubinfeld B, Hollingshead PG, Fong SE, Dugger DL, Pham T, Yansura DG, Wong TA, Grimaldi JC, Corpuz RT, Singh JS, Frantz GD, Devaux B, Crowley CW, Schwall RH, Eberhard DA, Rastelli L, Polakis P, Pennica D. Overexpression of the retinoic acid-responsive gene Stra6 in human cancers and its synergistic induction by Wnt-1 and retinoic acid. Cancer Res. 2001;61:4197–4205. [PubMed] [Google Scholar]
  • 16.Pasutto F, Sticht H, Hammersen G, Gillessen-Kaesbach G, Fitzpatrick DR, Nurnberg G, Brasch F, Schirmer-Zimmermann H, Tolmie JL, Chitayat D, Houge G, Fernandez-Martinez L, Keating S, Mortier G, Hennekam RC, von der Wense A, Slavotinek A, Meinecke P, Bitoun P, Becker C, Nurnberg P, Reis A, Rauch A. Mutations in STRA6 cause a broad spectrum of malformations including anophthalmia, congenital heart defects, diaphragmatic hernia, alveolar capillary dysplasia, lung hypoplasia, and mental retardation. Am J Hum Genet. 2007;80:550–560. doi: 10.1086/512203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Albalat R. The retinoic acid machinery in invertebrates: ancestral elements and vertebrate innovations. Mol Cell Endocrinol. 2009;313:23–35. doi: 10.1016/j.mce.2009.08.029. [DOI] [PubMed] [Google Scholar]
  • 18.Ghyselinck NB, Bavik C, Sapin V, Mark M, Bonnier D, Hindelang C, Dierich A, Nilsson CB, Hakansson H, Sauvant P, Azais-Braesco V, Frasson M, Picaud S, Chambon P. Cellular retinol-binding protein I is essential for vitamin A homeostasis. EMBO J. 1999;18:4903–4914. doi: 10.1093/emboj/18.18.4903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.XE, Zhang L, Lu J, Tso P, Blaner WS, Levin MS, Li E. Increased neonatal mortality in mice lacking cellular retinol-binding protein II. J Biol Chem. 2002;277:36617–36623. doi: 10.1074/jbc.M205519200. [DOI] [PubMed] [Google Scholar]
  • 20.Piantedosi R, Ghyselinck N, Blaner WS, Vogel S. Cellular retinol-binding protein type III is needed for retinoid incorporation into milk. J Biol Chem. 2005;280:24286–24292. doi: 10.1074/jbc.M503906200. [DOI] [PubMed] [Google Scholar]
  • 21.Esteller M, Guo M, Moreno V, Peinado MA, Capella G, Galm O, Baylin SB, Herman JG. Hypermethylation-associated Inactivation of the Cellular Retinol-Binding-Protein 1 Gene in Human Cancer. Cancer Res. 2002;62:5902–5905. [PubMed] [Google Scholar]
  • 22.Matt N, Schmidt CK, Dupe V, Dennefeld C, Nau H, Chambon P, Mark M, Ghyselinck NB. Contribution of cellular retinol-binding protein type 1 to retinol metabolism during mouse development. Dev Dyn. 2005;233:167–176. doi: 10.1002/dvdy.20313. [DOI] [PubMed] [Google Scholar]
  • 23.Fawcett D, Pasceri P, Fraser R, Colbert M, Rossant J, Giguere V. Postaxial polydactyly in forelimbs of CRABP-II mutant mice. Development. 1995;121:671–679. doi: 10.1242/dev.121.3.671. [DOI] [PubMed] [Google Scholar]
  • 24.Wolf G. Cellular retinoic acid-binding protein II: a coactivator of the transactivation by the retinoic acid receptor complex RAR.RXR. Nutr Rev. 2000;58:151–153. doi: 10.1111/j.1753-4887.2000.tb01851.x. [DOI] [PubMed] [Google Scholar]
  • 25.Noy N. Retinoid-binding proteins: mediators of retinoid action. Biochem J 348 Pt. 2000;3:481–495. [PMC free article] [PubMed] [Google Scholar]
  • 26.Blaese MA, Santo-Hoeltje L, Rodemann HP. CRABP I expression and the mediation of the sensitivity of human tumour cells to retinoic acid and irradiation. Int J Radiat Biol. 2003;79:981–991. doi: 10.1080/09553000310001632949. [DOI] [PubMed] [Google Scholar]
  • 27.Petkovich M, Brand NJ, Krust A, Chambon P. A human retinoic acid receptor which belongs to the family of nuclear receptors. Nature. 1987;330:444–450. doi: 10.1038/330444a0. [DOI] [PubMed] [Google Scholar]
  • 28.Giguere V, Ong ES, Segui P, Evans RM. Identification of a receptor for the morphogen retinoic acid. Nature. 1987;330:624–629. doi: 10.1038/330624a0. [DOI] [PubMed] [Google Scholar]
  • 29.Sporn MB, Roberts AB. Role of retinoids in differentiation and carcinogenesis. Cancer Res. 1983;43:3034–3040. [PubMed] [Google Scholar]
  • 30.Heyman RA, Mangelsdorf DJ, Dyck JA, Stein RB, Eichele G, Evans RM, Thaller C. 9-cis retinoic acid is a high affinity ligand for the retinoid X receptor. Cell. 1992;68:397–406. doi: 10.1016/0092-8674(92)90479-v. [DOI] [PubMed] [Google Scholar]
  • 31.Forsmo S, Fjeldbo SK, Langhammer A. Childhood cod liver oil consumption and bone mineral density in a population-based cohort of peri- and postmenopausal women: the Nord-Trondelag Health Study. Am J Epidemiol. 2008;167:406–411. doi: 10.1093/aje/kwm320. [DOI] [PubMed] [Google Scholar]
  • 32.Mangelsdorf DJ, Evans RM. The RXR heterodimers and orphan receptors. Cell. 1995;83:841–850. doi: 10.1016/0092-8674(95)90200-7. [DOI] [PubMed] [Google Scholar]
  • 33.Wang K, Chen S, Xie W, Wan YJ. Retinoids induce cytochrome P450 3A4 through RXR/VDR-mediated pathway. Biochem Pharmacol. 2008;75:2204–2213. doi: 10.1016/j.bcp.2008.02.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Chen S, Wang K, Wan YJ. Retinoids activate RXR/CAR-mediated pathway and induce CYP3A. Biochem Pharmacol. 2010;79:270–276. doi: 10.1016/j.bcp.2009.08.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Wang K, Mendy AJ, Dai G, Luo HR, He L, Wan YJ. Retinoids activate the RXR/SXR-mediated pathway and induce the endogenous CYP3A4 activity in Huh7 human hepatoma cells. Toxicol Sci. 2006;92:51–60. doi: 10.1093/toxsci/kfj207. [DOI] [PubMed] [Google Scholar]
  • 36.Wan YJ, An D, Cai Y, Repa JJ, Hung-Po Chen T, Flores M, Postic C, Magnuson MA, Chen J, Chien KR, French S, Mangelsdorf DJ, Sucov HM. Hepatocyte-specific mutation establishes retinoid × receptor alpha as a heterodimeric integrator of multiple physiological processes in the liver. Mol Cell Biol. 2000;20:4436–4444. doi: 10.1128/mcb.20.12.4436-4444.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Wan YJ, Cai Y, Lungo W, Fu P, Locker J, French S, Sucov HM. Peroxisome proliferator-activated receptor alpha-mediated pathways are altered in hepatocyte-specific retinoid × receptor alpha-deficient mice. J Biol Chem. 2000;275:28285–28290. doi: 10.1074/jbc.M000934200. [DOI] [PubMed] [Google Scholar]
  • 38.Gyamfi M, Wan YJ. Pathogenesis of alcoholic liver diseasse: the role of nuclear receptors. Exp Biol Med. 2010 doi: 10.1258/ebm.2009.009249. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Schug TT, Berry DC, Shaw NS, Travis SN, Noy N. Opposing effects of retinoic acid on cell growth result from alternate activation of two different nuclear receptors. Cell. 2007;129:723–733. doi: 10.1016/j.cell.2007.02.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Berry DC, Noy N. All-trans-retinoic acid represses obesity and insulin resistance by activating both peroxisome proliferation-activated receptor beta/delta and retinoic acid receptor. Mol Cell Biol. 2009;29:3286–3296. doi: 10.1128/MCB.01742-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Brocard J, Kastner P, Chambon P. Two novel RXR alpha isoforms from mouse testis. Biochem Biophys Res Commun. 1996;229:211–218. doi: 10.1006/bbrc.1996.1782. [DOI] [PubMed] [Google Scholar]
  • 42.Dolle P. Developmental expression of retinoic acid receptors (RARs) Nucl Recept Signal. 2009;7:e006. doi: 10.1621/nrs.07006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Lufkin T, Lohnes D, Mark M, Dierich A, Gorry P, Gaub MP, LeMeur M, Chambon P. High postnatal lethality and testis degeneration in retinoic acid receptor alpha mutant mice. Proc Natl Acad Sci U S A. 1993;90:7225–7229. doi: 10.1073/pnas.90.15.7225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Ghyselinck NB, Dupe V, Dierich A, Messaddeq N, Garnier JM, Rochette-Egly C, Chambon P, Mark M. Role of the retinoic acid receptor beta (RARbeta) during mouse development. Int J Dev Biol. 1997;41:425–447. [PubMed] [Google Scholar]
  • 45.Lohnes D, Kastner P, Dierich A, Mark M, LeMeur M, Chambon P. Function of retinoic acid receptor gamma in the mouse. Cell. 1993;73:643–658. doi: 10.1016/0092-8674(93)90246-m. [DOI] [PubMed] [Google Scholar]
  • 46.Sucov HM, Dyson E, Gumeringer CL, Price J, Chien KR, Evans RM. RXR alpha mutant mice establish a genetic basis for vitamin A signaling in heart morphogenesis. Genes Dev. 1994;8:1007–1018. doi: 10.1101/gad.8.9.1007. [DOI] [PubMed] [Google Scholar]
  • 47.Kastner P, Grondona JM, Mark M, Gansmuller A, LeMeur M, Decimo D, Vonesch JL, Dolle P, Chambon P. Genetic analysis of RXR alpha developmental function: convergence of RXR and RAR signaling pathways in heart and eye morphogenesis. Cell. 1994;78:987–1003. doi: 10.1016/0092-8674(94)90274-7. [DOI] [PubMed] [Google Scholar]
  • 48.Sucov HM, Izpisua-Belmonte JC, Ganan Y, Evans RM. Mouse embryos lacking RXR alpha are resistant to retinoic-acid-induced limb defects. Development. 1995;121:3997–4003. doi: 10.1242/dev.121.12.3997. [DOI] [PubMed] [Google Scholar]
  • 49.Kastner P, Mark M, Leid M, Gansmuller A, Chin W, Grondona JM, Decimo D, Krezel W, Dierich A, Chambon P. Abnormal spermatogenesis in RXR beta mutant mice. Genes Dev. 1996;10:80–92. doi: 10.1101/gad.10.1.80. [DOI] [PubMed] [Google Scholar]
  • 50.Mascrez B, Ghyselinck NB, Watanabe M, Annicotte JS, Chambon P, Auwerx J, Mark M. Ligand-dependent contribution of RXRbeta to cholesterol homeostasis in Sertoli cells. EMBO Rep. 2004;5:285–290. doi: 10.1038/sj.embor.7400094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Brown NS, Smart A, Sharma V, Brinkmeier ML, Greenlee L, Camper SA, Jensen DR, Eckel RH, Krezel W, Chambon P, Haugen BR. Thyroid hormone resistance and increased metabolic rate in the RXR-gamma-deficient mouse. J Clin Invest. 2000;106:73–79. doi: 10.1172/JCI9422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Mark M, Ghyselinck NB, Chambon P. Function of retinoic acid receptors during embryonic development. Nucl Recept Signal. 2009;7:e002. doi: 10.1621/nrs.07002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Fanjul A, Dawson MI, Hobbs PD, Jong L, Cameron JF, Harlev E, Graupner G, Lu XP, Pfahl M. A new class of retinoids with selective inhibition of AP-1 inhibits proliferation. Nature. 1994;372:107–111. doi: 10.1038/372107a0. [DOI] [PubMed] [Google Scholar]
  • 54.Austenaa LM, Carlsen H, Ertesvag A, Alexander G, Blomhoff HK, Blomhoff R. Vitamin A status significantly alters nuclear factor-kappaB activity assessed by in vivo imaging. FASEB J. 2004;18:1255–1257. doi: 10.1096/fj.03-1098fje. [DOI] [PubMed] [Google Scholar]
  • 55.Liao YP, Ho SY, Liou JC. Non-genomic regulation of transmitter release by retinoic acid at developing motoneurons in Xenopus cell culture. J Cell Sci. 2004;117:2917–2924. doi: 10.1242/jcs.01153. [DOI] [PubMed] [Google Scholar]
  • 56.Chen N, Onisko B, Napoli JL. The Nuclear Transcription Factor RARα Associates with Neuronal RNA Granules and Suppresses Translation. Journal of Biological Chemistry. 2008;283:20841–20847. doi: 10.1074/jbc.M802314200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Masia S, Alvarez S, de Lera AR, Barettino D. Rapid, nongenomic actions of retinoic acid on phosphatidylinositol-3-kinase signaling pathway mediated by the retinoic acid receptor. Mol Endocrinol. 2007;21:2391–2402. doi: 10.1210/me.2007-0062. [DOI] [PubMed] [Google Scholar]
  • 58.Chen N, Napoli JL. All-trans-retinoic acid stimulates translation and induces spine formation in hippocampal neurons through a membrane-associated RARalpha. FASEB J. 2008;22:236–245. doi: 10.1096/fj.07-8739com. [DOI] [PubMed] [Google Scholar]
  • 59.Poon MM, Chen L. Retinoic acid-gated sequence-specific translational control by RARalpha. Proc Natl Acad Sci U S A. 2008;105:20303–20308. doi: 10.1073/pnas.0807740105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Aggarwal S, Kim SW, Cheon K, Tabassam FH, Yoon JH, Koo JS. Nonclassical action of retinoic acid on the activation of the cAMP response element-binding protein in normal human bronchial epithelial cells. Mol Biol Cell. 2006;17:566–575. doi: 10.1091/mbc.E05-06-0519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Sun SY, Lotan R. Retinoids and their receptors in cancer development and chemoprevention. Crit Rev Oncol Hematol. 2002;41:41–55. doi: 10.1016/s1040-8428(01)00144-5. [DOI] [PubMed] [Google Scholar]
  • 62.Bukhari MH, Qureshi SS, Niazi S, Asef M, Naheed M, Khan SA, Chaudhry NA, Tayyab M, Hasan M. Chemotherapeutic/chemopreventive role of retinoids in chemically induced skin carcinogenesis in albino mice. Int J Dermatol. 2007;46:1160–1165. doi: 10.1111/j.1365-4632.2007.03425.x. [DOI] [PubMed] [Google Scholar]
  • 63.Wang Y, Wen W, Yi Y, Zhang Z, Lubet RA, You M. Preventive effects of bexarotene and budesonide in a genetically engineered mouse model of small cell lung cancer. Cancer Prev Res (Phila Pa) 2009;2:1059–1064. doi: 10.1158/1940-6207.CAPR-09-0221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Pisano C, Vesci L, Fodera R, Ferrara FF, Rossi C, De Cesare M, Zuco V, Pratesi G, Supino R, Zunino F. Antitumor activity of the combination of synthetic retinoid ST1926 and cisplatin in ovarian carcinoma models. Ann Oncol. 2007;18:1500–1505. doi: 10.1093/annonc/mdm195. [DOI] [PubMed] [Google Scholar]
  • 65.Shah RK, Valdez TA, Wang Z, Shapshay SM. Pulsed-dye laser and retinoic acid delay progression of oral squamous cell carcinoma: a murine model. Laryngoscope. 2001;111:1203–1208. doi: 10.1097/00005537-200107000-00013. [DOI] [PubMed] [Google Scholar]
  • 66.Li Y, Zhang Y, Hill J, Kim HT, Shen Q, Bissonnette RP, Lamph WW, Brown PH. The rexinoid, bexarotene, prevents the development of premalignant lesions in MMTV-erbB2 mice. Br J Cancer. 2008;98:1380–1388. doi: 10.1038/sj.bjc.6604320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Moon RC, Kelloff GJ, Detrisac CJ, Steele VE, Thomas CF, Sigman CC. Chemoprevention of OH-BBN-induced bladder cancer in mice by oltipraz, alone and in combination with 4-HPR and DFMO. Anticancer Res. 1994;14:5–11. [PubMed] [Google Scholar]
  • 68.Liu B, Lee KW, Li H, Ma L, Lin GL, Chandraratna RA, Cohen P. Combination therapy of insulin-like growth factor binding protein-3 and retinoid × receptor ligands synergize on prostate cancer cell apoptosis in vitro and in vivo. Clin Cancer Res. 2005;11:4851–4856. doi: 10.1158/1078-0432.CCR-04-2160. [DOI] [PubMed] [Google Scholar]
  • 69.Muto Y, Moriwaki H, Shiratori Y. Prevention of second primary tumors by an acyclic retinoid, polyprenoic acid, in patients with hepatocellular carcinoma. Digestion 59 Suppl. 1998;2:89–91. doi: 10.1159/000051435. [DOI] [PubMed] [Google Scholar]
  • 70.Recchia F, Sica G, Candeloro G, Necozione S, Bisegna R, Bratta M, Rea S. Beta-interferon, retinoids and tamoxifen in metastatic breast cancer: long-term follow-up of a phase II study. Oncol Rep. 2009;21:1011–1016. doi: 10.3892/or_00000317. [DOI] [PubMed] [Google Scholar]
  • 71.Edelman MJ, Smith R, Hausner P, Doyle LA, Kalra K, Kendall J, Bedor M, Bisaccia S. Phase II trial of the novel retinoid, bexarotene, and gemcitabine plus carboplatin in advanced non-small-cell lung cancer. J Clin Oncol. 2005;23:5774–5778. doi: 10.1200/JCO.2005.14.373. [DOI] [PubMed] [Google Scholar]
  • 72.Yamane A, Tsukamoto N, Saitoh T, Uchiumi H, Handa H, Karasawa M, Nojima Y, Murakami H. Successful treatment by all-trans retinoic acid in a patient with acute promyelocytic leukemia complicated by liver cirrhosis and polycystic kidney. Intern Med. 2009;48:1691–1694. doi: 10.2169/internalmedicine.48.2358. [DOI] [PubMed] [Google Scholar]
  • 73.Colombo N, Formelli F, Cantu MG, Parma G, Gasco M, Argusti A, Santinelli A, Montironi R, Cavadini E, Baglietto L, Guerrieri-Gonzaga A, Viale G, Decensi A. A phase I–II preoperative biomarker trial of fenretinide in ascitic ovarian cancer. Cancer Epidemiol Biomarkers Prev. 2006;15:1914–1919. doi: 10.1158/1055-9965.EPI-06-0183. [DOI] [PubMed] [Google Scholar]
  • 74.Borrow J, Goddard AD, Sheer D, Solomon E. Molecular analysis of acute promyelocytic leukemia breakpoint cluster region on chromosome 17. Science. 1990;249:1577–1580. doi: 10.1126/science.2218500. [DOI] [PubMed] [Google Scholar]
  • 75.de The H, Chomienne C, Lanotte M, Degos L, Dejean A. The t(15;17) translocation of acute promyelocytic leukaemia fuses the retinoic acid receptor alpha gene to a novel transcribed locus. Nature. 1990;347:558–561. doi: 10.1038/347558a0. [DOI] [PubMed] [Google Scholar]
  • 76.Grignani F, De Matteis S, Nervi C, Tomassoni L, Gelmetti V, Cioce M, Fanelli M, Ruthardt M, Ferrara FF, Zamir I, Seiser C, Lazar MA, Minucci S, Pelicci PG. Fusion proteins of the retinoic acid receptor-alpha recruit histone deacetylase in promyelocytic leukaemia. Nature. 1998;391:815–818. doi: 10.1038/35901. [DOI] [PubMed] [Google Scholar]
  • 77.Fenrick R, Hiebert SW. Role of histone deacetylases in acute leukemia. J Cell Biochem Suppl. 1998;30–31:194–202. [PubMed] [Google Scholar]
  • 78.Zeisig BB, Kwok C, Zelent A, Shankaranarayanan P, Gronemeyer H, Dong S, So CW. Recruitment of RXR by homotetrameric RARalpha fusion proteins is essential for transformation. Cancer Cell. 2007;12:36–51. doi: 10.1016/j.ccr.2007.06.006. [DOI] [PubMed] [Google Scholar]
  • 79.Zhu J, Nasr R, Peres L, Riaucoux-Lormiere F, Honore N, Berthier C, Kamashev D, Zhou J, Vitoux D, Lavau C, de The H. RXR is an essential component of the oncogenic PML/RARA complex in vivo. Cancer Cell. 2007;12:23–35. doi: 10.1016/j.ccr.2007.06.004. [DOI] [PubMed] [Google Scholar]
  • 80.Yoshida H, Kitamura K, Tanaka K, Omura S, Miyazaki T, Hachiya T, Ohno R, Naoe T. Accelerated degradation of PML-retinoic acid receptor alpha (PML-RARA) oncoprotein by all-trans-retinoic acid in acute promyelocytic leukemia: possible role of the proteasome pathway. Cancer Res. 1996;56:2945–2948. [PubMed] [Google Scholar]
  • 81.Casini T, Pelicci PG. A function of p21 during promyelocytic leukemia cell differentiation independent of CDK inhibition and cell cycle arrest. Oncogene. 1999;18:3235–3243. doi: 10.1038/sj.onc.1202630. [DOI] [PubMed] [Google Scholar]
  • 82.Jimenez-Lara AM, Clarke N, Altucci L, Gronemeyer H. Retinoic-acid-induced apoptosis in leukemia cells. Trends Mol Med. 2004;10:508–515. doi: 10.1016/j.molmed.2004.08.006. [DOI] [PubMed] [Google Scholar]
  • 83.Patatanian E, Thompson DF. Retinoic acid syndrome: a review. J Clin Pharm Ther. 2008;33:331–338. doi: 10.1111/j.1365-2710.2008.00935.x. [DOI] [PubMed] [Google Scholar]
  • 84.Christov KT, Moon RC, Lantvit DD, Boone CW, Steele VE, Lubet RA, Kelloff GJ, Pezzuto JM. 9-cis-retinoic acid but not 4-(hydroxyphenyl)retinamide inhibits prostate intraepithelial neoplasia in Noble rats. Cancer Res. 2002;62:5178–5182. [PubMed] [Google Scholar]
  • 85.Wu K, Kim HT, Rodriquez JL, Munoz-Medellin D, Mohsin SK, Hilsenbeck SG, Lamph WW, Gottardis MM, Shirley MA, Kuhn JG, Green JE, Brown PH. 9-cis-Retinoic acid suppresses mammary tumorigenesis in C3(1)-simian virus 40 T antigen-transgenic mice. Clin Cancer Res. 2000;6:3696–3704. [PubMed] [Google Scholar]
  • 86.Baumann L, Vujevich J, Halem M, Martin LK, Kerdel F, Lazarus M, Pacheco H, Black L, Bryde J. Open-label pilot study of alitretinoin gel 0.1% in the treatment of photoaging. Cutis. 2005;76:69–73. [PubMed] [Google Scholar]
  • 87.Wan YJ, Cai Y, Cowan C, Magee TR. Fatty acyl-CoAs inhibit retinoic acid-induced apoptosis in Hep3B cells. Cancer Lett. 2000;154:19–27. doi: 10.1016/s0304-3835(00)00341-4. [DOI] [PubMed] [Google Scholar]
  • 88.Lin YW, Lien LM, Yeh TS, Wu HM, Liu YL, Hsieh RH. 9-cis retinoic acid induces retinoid × receptor localized to the mitochondria for mediation of mitochondrial transcription. Biochem Biophys Res Commun. 2008;377:351–354. doi: 10.1016/j.bbrc.2008.09.122. [DOI] [PubMed] [Google Scholar]
  • 89.Zheng ZS, Xue GZ, Prystowsky JH. Regulation of the induction of ornithine decarboxylase in keratinocytes by retinoids. Biochem J. 1995;309(Pt 1):159–165. doi: 10.1042/bj3090159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Geiger JM, Hommel L, Harms M, Saurat JH. Oral 13-cis retinoic acid is superior to 9-cis retinoic acid in sebosuppression in human beings. J Am Acad Dermatol. 1996;34:513–515. doi: 10.1016/s0190-9622(96)90462-4. [DOI] [PubMed] [Google Scholar]
  • 91.Bouclier M, Chatelus A, Ferracin J, Delain C, Shroot B, Hensby CN. Quantification of epidermal histological changes induced by topical retinoids and CD271 in the rhino mouse model using a standardized image analysis technique. Skin Pharmacol. 1991;4:65–73. doi: 10.1159/000210926. [DOI] [PubMed] [Google Scholar]
  • 92.Gilgor RS, Chiaramonti A, Goldsmith LA, Lazarus GS. Evaluation of 13-cis retinoic acid in lamellar ichthyosis, pityriasis rubra pilaris and Darier’s disease. Cutis. 1980;25:380–381. 385. [PubMed] [Google Scholar]
  • 93.Thielitz A, Gollnick H. Topical retinoids in acne vulgaris: update on efficacy and safety. Am J Clin Dermatol. 2008;9:369–381. doi: 10.2165/0128071-200809060-00003. [DOI] [PubMed] [Google Scholar]
  • 94.Van Herle AJ, Agatep ML, Padua DN, 3rd, Totanes TL, Canlapan DV, Van Herle HM, Juillard GJ. Effects of 13 cis-retinoic acid on growth and differentiation of human follicular carcinoma cells (UCLA R0 82 W-1) in vitro. J Clin Endocrinol Metab. 1990;71:755–763. doi: 10.1210/jcem-71-3-755. [DOI] [PubMed] [Google Scholar]
  • 95.Schreck R, Schnieders F, Schmutzler C, Kohrle J. Retinoids stimulate type I iodothyronine 5′-deiodinase activity in human follicular thyroid carcinoma cell lines. J Clin Endocrinol Metab. 1994;79:791–798. doi: 10.1210/jcem.79.3.8077363. [DOI] [PubMed] [Google Scholar]
  • 96.Schmutzler C, Koehrle J. Innovative strategies for the treatment of thyroid cancer. Eur J Endocrinol. 2000;143:15–24. doi: 10.1530/eje.0.1430015. [DOI] [PubMed] [Google Scholar]
  • 97.Brtko J. Retinoids, rexinoids and their cognate nuclear receptors: character and their role in chemoprevention of selected malignant diseases. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. 2007;151:187–194. doi: 10.5507/bp.2007.033. [DOI] [PubMed] [Google Scholar]
  • 98.Matthay KK, Reynolds CP, Seeger RC, Shimada H, Adkins ES, Haas-Kogan D, Gerbing RB, London WB, Villablanca JG. Long-term results for children with high-risk neuroblastoma treated on a randomized trial of myeloablative therapy followed by 13-cis-retinoic acid: a children’s oncology group study. J Clin Oncol. 2009;27:1007–1013. doi: 10.1200/JCO.2007.13.8925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Huang S, Laoukili J, Epping MT, Koster J, Holzel M, Westerman BA, Nijkamp W, Hata A, Asgharzadeh S, Seeger RC, Versteeg R, Beijersbergen RL, Bernards R. ZNF423 is critically required for retinoic acid-induced differentiation and is a marker of neuroblastoma outcome. Cancer Cell. 2009;15:328–340. doi: 10.1016/j.ccr.2009.02.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Hail N, Jr, Kim HJ, Lotan R. Mechanisms of fenretinide-induced apoptosis. Apoptosis. 2006;11:1677–1694. doi: 10.1007/s10495-006-9289-3. [DOI] [PubMed] [Google Scholar]
  • 101.Moon RC, Pritchard JF, Mehta RG, Nomides CT, Thomas CF, Dinger NM. Suppression of rat mammary cancer development by N-(4-hydroxyphenyl)retinamide (4-HPR) following surgical removal of first palpable tumor. Carcinogenesis. 1989;10:1645–1649. doi: 10.1093/carcin/10.9.1645. [DOI] [PubMed] [Google Scholar]
  • 102.Pollard M, Luckert PH, Sporn MB. Prevention of primary prostate cancer in Lobund-Wistar rats by N-(4-hydroxyphenyl)retinamide. Cancer Res. 1991;51:3610–3611. [PubMed] [Google Scholar]
  • 103.McCormick DL, Becci PJ, Moon RC. Inhibition of mammary and urinary bladder carcinogenesis by a retinoid and a maleic anhydride-divinyl ether copolymer (MVE-2) Carcinogenesis. 1982;3:1473–1476. doi: 10.1093/carcin/3.12.1473. [DOI] [PubMed] [Google Scholar]
  • 104.McCormick DL, Moon RC. Antipromotional activity of dietary N-(4-hydroxyphenyl)retinamide in two-stage skin tumorigenesis in CD-1 and SENCAR mice. Cancer Lett. 1986;31:133–138. doi: 10.1016/0304-3835(86)90003-0. [DOI] [PubMed] [Google Scholar]
  • 105.McCormick DL, Moon RC. Retinoid-tamoxifen interaction in mammary cancer chemoprevention. Carcinogenesis. 1986;7:193–196. doi: 10.1093/carcin/7.2.193. [DOI] [PubMed] [Google Scholar]
  • 106.Hong WK, Sporn MB. Recent advances in chemoprevention of cancer. Science. 1997;278:1073–1077. doi: 10.1126/science.278.5340.1073. [DOI] [PubMed] [Google Scholar]
  • 107.Formelli F, Barua AB, Olson JA. Bioactivities of N-(4-hydroxyphenyl) retinamide and retinoyl beta-glucuronide. Faseb J. 1996;10:1014–1024. doi: 10.1096/fasebj.10.9.8801162. [DOI] [PubMed] [Google Scholar]
  • 108.Lotan R. Retinoids and apoptosis: implications for cancer chemoprevention and therapy. J Natl Cancer Inst. 1995;87:1655–1657. doi: 10.1093/jnci/87.22.1655. [DOI] [PubMed] [Google Scholar]
  • 109.Paulson JD, Oldham JW, Preston RF, Newman D. Lack of genotoxicity of the cancer chemopreventive agent N-(4-hydroxyphenyl)retinamide. Fundam Appl Toxicol. 1985;5:144–150. doi: 10.1016/0272-0590(85)90058-2. [DOI] [PubMed] [Google Scholar]
  • 110.Abou-Issa H, Moeschberger M, el-Masry W, Tejwani S, Curley RW, Jr, Webb TE. Relative efficacy of glucarate on the initiation and promotion phases of rat mammary carcinogenesis. Anticancer Res. 1995;15:805–810. [PubMed] [Google Scholar]
  • 111.Ohshima M, Ward JM, Wenk ML. Preventive and enhancing effects of retinoids on the development of naturally occurring tumors of skin, prostate gland, and endocrine pancreas in aged male ACI/segHapBR rats. J Natl Cancer Inst. 1985;74:517–524. [PubMed] [Google Scholar]
  • 112.McCormick DL, Bagg BJ, Hultin TA. Comparative activity of dietary or topical exposure to three retinoids in the promotion of skin tumor induction in mice. Cancer Res. 1987;47:5989–5993. [PubMed] [Google Scholar]
  • 113.Chodak GW. Questioning the value of screening for prostate cancer in asymptomatic men. Urology. 1993;42:116–118. doi: 10.1016/0090-4295(93)90633-l. [DOI] [PubMed] [Google Scholar]
  • 114.Veronesi U, Paganelli G, Viale G, Galimberti V, Luini A, Zurrida S, Robertson C, Sacchini V, Veronesi P, Orvieto E, De Cicco C, Intra M, Tosi G, Scarpa D. Sentinel lymph node biopsy and axillary dissection in breast cancer: results in a large series. J Natl Cancer Inst. 1999;91:368–373. doi: 10.1093/jnci/91.4.368. [DOI] [PubMed] [Google Scholar]
  • 115.Chiesa F, Tradati N, Grigolato R, Boracchi P, Biganzoli E, Crose N, Cavadini E, Formelli F, Costa L, Giardini R, Zurrida S, Costa A, De Palo G, Veronesi U. Randomized trial of fenretinide (4-HPR) to prevent recurrences, new localizations and carcinomas in patients operated on for oral leukoplakia: long-term results. Int J Cancer. 2005;115:625–629. doi: 10.1002/ijc.20923. [DOI] [PubMed] [Google Scholar]
  • 116.Sogno I, Vene R, Sapienza C, Ferrari N, Tosetti F, Albini A. Anti-angiogenic properties of chemopreventive drugs: fenretinide as a prototype. Recent Results Cancer Res. 2009;181:71–76. doi: 10.1007/978-3-540-69297-3_8. [DOI] [PubMed] [Google Scholar]
  • 117.Qian J, Zhang JS, Wang XQ, Ji JL, Mei S. Fenretinide stimulates the apoptosis of hepatic stellate cells and ameliorates hepatic fibrosis in mice. Hepatol Res. 2009;39:1229–1247. doi: 10.1111/j.1872-034X.2009.00562.x. [DOI] [PubMed] [Google Scholar]
  • 118.Preitner F, Mody N, Graham TE, Peroni OD, Kahn BB. Long-term Fenretinide treatment prevents high-fat diet-induced obesity, insulin resistance, and hepatic steatosis. Am J Physiol Endocrinol Metab. 2009;297:E1420–1429. doi: 10.1152/ajpendo.00362.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Clifford JL, Menter DG, Wang M, Lotan R, Lippman SM. Retinoid receptor-dependent and -independent effects of N-(4-hydroxyphenyl)retinamide in F9 embryonal carcinoma cells. Cancer Res. 1999;59:14–18. [PubMed] [Google Scholar]
  • 120.Bu P, Wan YJ. Fenretinide-induced apoptosis of Huh-7 hepatocellular carcinoma is retinoic acid receptor beta dependent. BMC Cancer. 2007;7:236. doi: 10.1186/1471-2407-7-236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Yang H, Bushue N, Bu P, Yvonne Wan YJ. Induction and intracellular localization of Nur77 dictate fenretinide-induced apoptosis of human liver cancer cells. Biochem Pharmacol. 2010;79:948–954. doi: 10.1016/j.bcp.2009.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Li H, Kolluri SK, Gu J, Dawson MI, Cao X, Hobbs PD, Lin B, Chen G, Lu J, Lin F, Xie Z, Fontana JA, Reed JC, Zhang X. Cytochrome c release and apoptosis induced by mitochondrial targeting of nuclear orphan receptor TR3. Science. 2000;289:1159–1164. doi: 10.1126/science.289.5482.1159. [DOI] [PubMed] [Google Scholar]
  • 123.Holmes WF, Soprano DR, Soprano KJ. Comparison of the mechanism of induction of apoptosis in ovarian carcinoma cells by the conformationally restricted synthetic retinoids CD437 and 4-HPR. J Cell Biochem. 2003;89:262–278. doi: 10.1002/jcb.10505. [DOI] [PubMed] [Google Scholar]
  • 124.Lin B, Kolluri SK, Lin F, Liu W, Han YH, Cao X, Dawson MI, Reed JC, Zhang XK. Conversion of Bcl-2 from protector to killer by interaction with nuclear orphan receptor Nur77/TR3. Cell. 2004;116:527–540. doi: 10.1016/s0092-8674(04)00162-x. [DOI] [PubMed] [Google Scholar]
  • 125.Zhu YK, Liu X, Ertl RF, Kohyama T, Wen FQ, Wang H, Spurzem JR, Romberger DJ, Rennard SI. Retinoic acid attenuates cytokine-driven fibroblast degradation of extracellular matrix in three-dimensional culture. Am J Respir Cell Mol Biol. 2001;25:620–627. doi: 10.1165/ajrcmb.25.5.4495. [DOI] [PubMed] [Google Scholar]
  • 126.Garattini E, Parrella E, Diomede L, Gianni M, Kalac Y, Merlini L, Simoni D, Zanier R, Ferrara FF, Chiarucci I, Carminati P, Terao M, Pisano C. ST1926, a novel and orally active retinoid-related molecule inducing apoptosis in myeloid leukemia cells: modulation of intracellular calcium homeostasis. Blood. 2004;103:194–207. doi: 10.1182/blood-2003-05-1577. [DOI] [PubMed] [Google Scholar]
  • 127.Boisvieux-Ulrich E, Sourdeval M, Marano F. CD437, a synthetic retinoid, induces apoptosis in human respiratory epithelial cells via caspase-independent mitochondrial and caspase-8-dependent pathways both up-regulated by JNK signaling pathway. Exp Cell Res. 2005;307:76–90. doi: 10.1016/j.yexcr.2005.02.005. [DOI] [PubMed] [Google Scholar]
  • 128.Farhana L, Dawson MI, Huang Y, Zhang Y, Rishi AK, Reddy KB, Freeman RS, Fontana JA. Apoptosis signaling by the novel compound 3-Cl-AHPC involves increased EGFR proteolysis and accompanying decreased phosphatidylinositol 3-kinase and AKT kinase activities. Oncogene. 2004;23:1874–1884. doi: 10.1038/sj.onc.1207311. [DOI] [PubMed] [Google Scholar]
  • 129.Han YH, Cao X, Lin B, Lin F, Kolluri SK, Stebbins J, Reed JC, Dawson MI, Zhang XK. Regulation of Nur77 nuclear export by c-Jun N-terminal kinase and Akt. Oncogene. 2006;25:2974–2986. doi: 10.1038/sj.onc.1209358. [DOI] [PubMed] [Google Scholar]
  • 130.Qu L, Tang X. Bexarotene: a promising anticancer agent. Cancer Chemother Pharmacol. 2010;65:201–205. doi: 10.1007/s00280-009-1140-4. [DOI] [PubMed] [Google Scholar]
  • 131.Minagawa N, Nakayama Y, Inoue Y, Onitsuka K, Katsuki T, Tsurudome Y, Shibao K, Hirata K, Sako T, Nagata N, Ohie S, Kohno K, Itoh H. 4-[3,5-Bis(trimethylsilyl)benzamido] benzoic acid inhibits angiogenesis in colon cancer through reduced expression of vascular endothelial growth factor. Oncol Res. 2004;14:407–414. doi: 10.3727/0965040041791464. [DOI] [PubMed] [Google Scholar]
  • 132.Higginbotham KB, Lozano R, Brown T, Patt YZ, Arima T, Abbruzzese JL, Thomas MB. A phase I/II trial of TAC-101, an oral synthetic retinoid, in patients with advanced hepatocellular carcinoma. J Cancer Res Clin Oncol. 2008;134:1325–1335. doi: 10.1007/s00432-008-0406-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.So PL, Fujimoto MA, Epstein EH., Jr Pharmacologic retinoid signaling and physiologic retinoic acid receptor signaling inhibit basal cell carcinoma tumorigenesis. Mol Cancer Ther. 2008;7:1275–1284. doi: 10.1158/1535-7163.MCT-07-2043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Lippman SM, Sudbo J, Hong WK. Oral cancer prevention and the evolution of molecular-targeted drug development. J Clin Oncol. 2005;23:346–356. doi: 10.1200/JCO.2005.09.128. [DOI] [PubMed] [Google Scholar]
  • 135.Mao L, El-Naggar AK, Papadimitrakopoulou V, Shin DM, Shin HC, Fan Y, Zhou X, Clayman G, Lee JJ, Lee JS, Hittelman WN, Lippman SM, Hong WK. Phenotype and genotype of advanced premalignant head and neck lesions after chemopreventive therapy. J Natl Cancer Inst. 1998;90:1545–1551. doi: 10.1093/jnci/90.20.1545. [DOI] [PubMed] [Google Scholar]
  • 136.Li XS, Shao ZM, Sheikh MS, Eiseman JL, Sentz D, Jetten AM, Chen JC, Dawson MI, Aisner S, Rishi AK, et al. Retinoic acid nuclear receptor beta inhibits breast carcinoma anchorage independent growth. J Cell Physiol. 1995;165:449–458. doi: 10.1002/jcp.1041650302. [DOI] [PubMed] [Google Scholar]
  • 137.Liu Y, Lee MO, Wang HG, Li Y, Hashimoto Y, Klaus M, Reed JC, Zhang X. Retinoic acid receptor beta mediates the growth-inhibitory effect of retinoic acid by promoting apoptosis in human breast cancer cells. Mol Cell Biol. 1996;16:1138–1149. doi: 10.1128/mcb.16.3.1138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Li Y, Dawson MI, Agadir A, Lee MO, Jong L, Hobbs PD, Zhang XK. Regulation of RAR beta expression by RAR- and RXR-selective retinoids in human lung cancer cell lines: effect on growth inhibition and apoptosis induction. Int J Cancer. 1998;75:88–95. doi: 10.1002/(sici)1097-0215(19980105)75:1<88::aid-ijc14>3.0.co;2-9. [DOI] [PubMed] [Google Scholar]
  • 139.Pergolizzi R, Appierto V, Crosti M, Cavadini E, Cleris L, Guffanti A, Formelli F. Role of retinoic acid receptor overexpression in sensitivity to fenretinide and tumorigenicity of human ovarian carcinoma cells. Int J Cancer. 1999;81:829–834. doi: 10.1002/(sici)1097-0215(19990531)81:5<829::aid-ijc26>3.0.co;2-3. [DOI] [PubMed] [Google Scholar]
  • 140.Campbell MJ, Park S, Uskokovic MR, Dawson MI, Koeffler HP. Expression of retinoic acid receptor-beta sensitizes prostate cancer cells to growth inhibition mediated by combinations of retinoids and a 19-nor hexafluoride vitamin D3 analog. Endocrinology. 1998;139:1972–1980. doi: 10.1210/endo.139.4.5943. [DOI] [PubMed] [Google Scholar]
  • 141.Ferrari N, Pfahl M, Levi G. Retinoic acid receptor gamma1 (RARgamma1) levels control RARbeta2 expression in SK-N-BE2(c) neuroblastoma cells and regulate a differentiation-apoptosis switch. Mol Cell Biol. 1998;18:6482–6492. doi: 10.1128/mcb.18.11.6482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Hoffman AD, Engelstein D, Bogenrieder T, Papandreou CN, Steckelman E, Dave A, Motzer RJ, Dmitrovsky E, Albino AP, Nanus DM. Expression of retinoic acid receptor beta in human renal cell carcinomas correlates with sensitivity to the antiproliferative effects of 13-cis-retinoic acid. Clin Cancer Res. 1996;2:1077–1082. [PubMed] [Google Scholar]
  • 143.Kaiser A, Herbst H, Fisher G, Koenigsmann M, Berdel WE, Riecken EO, Rosewicz S. Retinoic acid receptor beta regulates growth and differentiation in human pancreatic carcinoma cells. Gastroenterology. 1997;113:920–929. doi: 10.1016/s0016-5085(97)70188-4. [DOI] [PubMed] [Google Scholar]
  • 144.Li C, Wan YJ. Differentiation and antiproliferation effects of retinoic acid receptor beta in hepatoma cells. Cancer Lett. 1998;124:205–211. doi: 10.1016/s0304-3835(97)00475-8. [DOI] [PubMed] [Google Scholar]
  • 145.Hayashi K, Yokozaki H, Naka K, Yasui W, Lotan R, Tahara E. Overexpression of retinoic acid receptor beta induces growth arrest and apoptosis in oral cancer cell lines. Jpn J Cancer Res. 2001;92:42–50. doi: 10.1111/j.1349-7006.2001.tb01046.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Xu XC, Zile MH, Lippman SM, Lee JS, Lee JJ, Hong WK, Lotan R. Anti-retinoic acid (RA) antibody binding to human premalignant oral lesions, which occurs less frequently than binding to normal tissue, increases after 13-cis-RA treatment in vivo and is related to RA receptor beta expression. Cancer Res. 1995;55:5507–5511. [PubMed] [Google Scholar]
  • 147.Berard J, Gaboury L, Landers M, De Repentigny Y, Houle B, Kothary R, Bradley WE. Hyperplasia and tumours in lung, breast and other tissues in mice carrying a RAR beta 4-like transgene. EMBO J. 1994;13:5570–5580. doi: 10.1002/j.1460-2075.1994.tb06894.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Lotan R, Xu XC, Lippman SM, Ro JY, Lee JS, Lee JJ, Hong WK. Suppression of retinoic acid receptor-beta in premalignant oral lesions and its up-regulation by isotretinoin. N Engl J Med. 1995;332:1405–1410. doi: 10.1056/NEJM199505253322103. [DOI] [PubMed] [Google Scholar]
  • 149.Shin DM, Xu XC, Lippman SM, Lee JJ, Lee JS, Batsakis JG, Ro JY, Martin JW, Hittelman WN, Lotan R, Hong WK. Accumulation of p53 protein and retinoic acid receptor beta in retinoid chemoprevention. Clin Cancer Res. 1997;3:875–880. [PubMed] [Google Scholar]
  • 150.Youssef EM, Lotan D, Issa JP, Wakasa K, Fan YH, Mao L, Hassan K, Feng L, Lee JJ, Lippman SM, Hong WK, Lotan R. Hypermethylation of the retinoic acid receptor-beta(2) gene in head and neck carcinogenesis. Clin Cancer Res. 2004;10:1733–1742. doi: 10.1158/1078-0432.ccr-0989-3. [DOI] [PubMed] [Google Scholar]
  • 151.Cras A, Darsin-Bettinger D, Balitrand N, Cassinat B, Soulie A, Toubert ME, Delva L, Chomienne C. Epigenetic patterns of the retinoic acid receptor beta2 promoter in retinoic acid-resistant thyroid cancer cells. Oncogene. 2007;26:4018–4024. doi: 10.1038/sj.onc.1210178. [DOI] [PubMed] [Google Scholar]
  • 152.Darwiche N, Celli G, Tennenbaum T, Glick AB, Yuspa SH, De Luca LM. Mouse skin tumor progression results in differential expression of retinoic acid and retinoid × receptors. Cancer Res. 1995;55:2774–2782. [PubMed] [Google Scholar]
  • 153.Lotan R. Retinoids in cancer chemoprevention. FASEB J. 1996;10:1031–1039. doi: 10.1096/fasebj.10.9.8801164. [DOI] [PubMed] [Google Scholar]
  • 154.Fisher GJ, Datta SC, Talwar HS, Wang ZQ, Varani J, Kang S, Voorhees JJ. Molecular basis of sun-induced premature skin ageing and retinoid antagonism. Nature. 1996;379:335–339. doi: 10.1038/379335a0. [DOI] [PubMed] [Google Scholar]
  • 155.Fisher GJ, Talwar HS, Lin J, Lin P, McPhillips F, Wang Z, Li X, Wan Y, Kang S, Voorhees JJ. Retinoic acid inhibits induction of c-Jun protein by ultraviolet radiation that occurs subsequent to activation of mitogen-activated protein kinase pathways in human skin in vivo. J Clin Invest. 1998;101:1432–1440. doi: 10.1172/JCI2153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Wang Z, Boudjelal M, Kang S, Voorhees JJ, Fisher GJ. Ultraviolet irradiation of human skin causes functional vitamin A deficiency, preventable by all-trans retinoic acid pre-treatment. Nat Med. 1999;5:418–422. doi: 10.1038/7417. [DOI] [PubMed] [Google Scholar]
  • 157.Greenhalgh DA, Welty DJ, Player A, Yuspa SH. Two oncogenes, v-fos and v-ras, cooperate to convert normal keratinocytes to squamous cell carcinoma. Proc Natl Acad Sci U S A. 1990;87:643–647. doi: 10.1073/pnas.87.2.643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Leder A, Kuo A, Cardiff RD, Sinn E, Leder P. v-Ha-ras transgene abrogates the initiation step in mouse skin tumorigenesis: effects of phorbol esters and retinoic acid. Proc Natl Acad Sci U S A. 1990;87:9178–9182. doi: 10.1073/pnas.87.23.9178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Huang C, Ma WY, Dawson MI, Rincon M, Flavell RA, Dong Z. Blocking activator protein-1 activity, but not activating retinoic acid response element, is required for the antitumor promotion effect of retinoic acid. Proc Natl Acad Sci U S A. 1997;94:5826–5830. doi: 10.1073/pnas.94.11.5826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Chen JY, Penco S, Ostrowski J, Balaguer P, Pons M, Starrett JE, Reczek P, Chambon P, Gronemeyer H. RAR-specific agonist/antagonists which dissociate transactivation and AP1 transrepression inhibit anchorage-independent cell proliferation. EMBO J. 1995;14:1187–1197. doi: 10.1002/j.1460-2075.1995.tb07102.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Kang S, Bergfeld W, Gottlieb AB, Hickman J, Humeniuk J, Kempers S, Lebwohl M, Lowe N, McMichael A, Milbauer J, Phillips T, Powers J, Rodriguez D, Savin R, Shavin J, Sherer D, Silvis N, Weinstein R, Weiss J, Hammerberg C, Fisher GJ, Nighland M, Grossman R, Nyirady J. Long-term efficacy and safety of tretinoin emollient cream 0.05% in the treatment of photodamaged facial skin: a two-year, randomized, placebo-controlled trial. Am J Clin Dermatol. 2005;6:245–253. doi: 10.2165/00128071-200506040-00005. [DOI] [PubMed] [Google Scholar]
  • 162.Blot WJ, Li JY, Taylor PR, Guo W, Dawsey S, Wang GQ, Yang CS, Zheng SF, Gail M, Li GY, et al. Nutrition intervention trials in Linxian, China: supplementation with specific vitamin/mineral combinations, cancer incidence, and disease-specific mortality in the general population. J Natl Cancer Inst. 1993;85:1483–1492. doi: 10.1093/jnci/85.18.1483. [DOI] [PubMed] [Google Scholar]
  • 163.The effect of vitamin E and beta carotene on the incidence of lung cancer and other cancers in male smokers. The Alpha-Tocopherol. Beta Carotene Cancer Prevention Study Group. N Engl J Med. 1994;330:1029–1035. doi: 10.1056/NEJM199404143301501. [DOI] [PubMed] [Google Scholar]
  • 164.Omenn GS, Goodman GE, Thornquist MD, Balmes J, Cullen MR, Glass A, Keogh JP, Meyskens FL, Valanis B, Williams JH, Barnhart S, Hammar S. Effects of a combination of beta carotene and vitamin A on lung cancer and cardiovascular disease. N Engl J Med. 1996;334:1150–1155. doi: 10.1056/NEJM199605023341802. [DOI] [PubMed] [Google Scholar]
  • 165.Hennekens CH, Buring JE, Manson JE, Stampfer M, Rosner B, Cook NR, Belanger C, LaMotte F, Gaziano JM, Ridker PM, Willett W, Peto R. Lack of effect of long-term supplementation with beta carotene on the incidence of malignant neoplasms and cardiovascular disease. N Engl J Med. 1996;334:1145–1149. doi: 10.1056/NEJM199605023341801. [DOI] [PubMed] [Google Scholar]
  • 166.De Vries N, Van Zandwijk N, Pastorino U. The EUROSCAN study: a progress report. Am J Otolaryngol. 1993;14:62–66. doi: 10.1016/0196-0709(93)90014-x. [DOI] [PubMed] [Google Scholar]
  • 167.Whang-Peng J, Kao-Shan CS, Lee EC, Bunn PA, Carney DN, Gazdar AF, Minna JD. Specific chromosome defect associated with human small-cell lung cancer; deletion 3p(14–23) Science. 1982;215:181–182. doi: 10.1126/science.6274023. [DOI] [PubMed] [Google Scholar]
  • 168.Houle B, Leduc F, Bradley WE. Implication of RARB in epidermoid (Squamous) lung cancer. Genes Chromosomes Cancer. 1991;3:358–366. doi: 10.1002/gcc.2870030506. [DOI] [PubMed] [Google Scholar]
  • 169.Picard E, Seguin C, Monhoven N, Rochette-Egly C, Siat J, Borrelly J, Martinet Y, Martinet N, Vignaud JM. Expression of retinoid receptor genes and proteins in non-small-cell lung cancer. J Natl Cancer Inst. 1999;91:1059–1066. doi: 10.1093/jnci/91.12.1059. [DOI] [PubMed] [Google Scholar]
  • 170.Xu XC, Sozzi G, Lee JS, Lee JJ, Pastorino U, Pilotti S, Kurie JM, Hong WK, Lotan R. Suppression of retinoic acid receptor beta in non-small-cell lung cancer in vivo: implications for lung cancer development. J Natl Cancer Inst. 1997;89:624–629. doi: 10.1093/jnci/89.9.624. [DOI] [PubMed] [Google Scholar]
  • 171.Xu XC, Ro JY, Lee JS, Shin DM, Hong WK, Lotan R. Differential expression of nuclear retinoid receptors in normal, premalignant, and malignant head and neck tissues. Cancer Res. 1994;54:3580–3587. [PubMed] [Google Scholar]
  • 172.Widschwendter M, Berger J, Daxenbichler G, Muller-Holzner E, Widschwendter A, Mayr A, Marth C, Zeimet AG. Loss of retinoic acid receptor beta expression in breast cancer and morphologically normal adjacent tissue but not in the normal breast tissue distant from the cancer. Cancer Res. 1997;57:4158–4161. [PubMed] [Google Scholar]
  • 173.Tyagi P. Bexarotene in combination with chemotherapy fails to prolong survival in patients with advanced non-small-cell lung cancer: results from the SPIRIT I and II trials. Clin Lung Cancer. 2005;7:17–19. doi: 10.1016/S1525-7304(11)70385-0. [DOI] [PubMed] [Google Scholar]
  • 174.Crowe DL. Receptor selective synthetic retinoids as potential cancer chemotherapy agents. Curr Cancer Drug Targets. 2002;2:77–86. doi: 10.2174/1568009023333935. [DOI] [PubMed] [Google Scholar]
  • 175.Petty WJ, Li N, Biddle A, Bounds R, Nitkin C, Ma Y, Dragnev KH, Freemantle SJ, Dmitrovsky E. A novel retinoic acid receptor beta isoform and retinoid resistance in lung carcinogenesis. J Natl Cancer Inst. 2005;97:1645–1651. doi: 10.1093/jnci/dji371. [DOI] [PubMed] [Google Scholar]
  • 176.Wu Q, Li Y, Liu R, Agadir A, Lee MO, Liu Y, Zhang X. Modulation of retinoic acid sensitivity in lung cancer cells through dynamic balance of orphan receptors nur77 and COUP-TF and their heterodimerization. EMBO J. 1997;16:1656–1669. doi: 10.1093/emboj/16.7.1656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Lin B, Chen GQ, Xiao D, Kolluri SK, Cao X, Su H, Zhang XK. Orphan receptor COUP-TF is required for induction of retinoic acid receptor beta, growth inhibition, and apoptosis by retinoic acid in cancer cells. Mol Cell Biol. 2000;20:957–970. doi: 10.1128/mcb.20.3.957-970.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Veronesi U, Mariani L, Decensi A, Formelli F, Camerini T, Miceli R, Di Mauro MG, Costa A, Marubini E, Sporn MB, De Palo G. Fifteen-year results of a randomized phase III trial of fenretinide to prevent second breast cancer. Ann Oncol. 2006;17:1065–1071. doi: 10.1093/annonc/mdl047. [DOI] [PubMed] [Google Scholar]
  • 179.Zanardi S, Serrano D, Argusti A, Barile M, Puntoni M, Decensi A. Clinical trials with retinoids for breast cancer chemoprevention. Endocr Relat Cancer. 2006;13:51–68. doi: 10.1677/erc.1.00938. [DOI] [PubMed] [Google Scholar]
  • 180.Macejova D, Baranova M, Liska J, Brtko J. Expression of nuclear hormone receptors, their coregulators and type I iodothyronine 5′-deiodinase gene in mammary tissue of nonlactating and postlactating rats. Life Sci. 2005;77:2584–2593. doi: 10.1016/j.lfs.2005.02.024. [DOI] [PubMed] [Google Scholar]
  • 181.Wang Y, Zhang JJ, Dai W, Pike JW. Production of granulocyte colony-stimulating factor by THP-1 cells in response to retinoic acid and phorbol ester is mediated through the autocrine production of interleukin-1. Biochem Biophys Res Commun. 1996;225:639–646. doi: 10.1006/bbrc.1996.1223. [DOI] [PubMed] [Google Scholar]
  • 182.Zhang XK, Liu Y, Lee MO. Retinoid receptors in human lung cancer and breast cancer. Mutat Res. 1996;350:267–277. doi: 10.1016/0027-5107(95)00102-6. [DOI] [PubMed] [Google Scholar]
  • 183.Gottardis MM, Bischoff ED, Shirley MA, Wagoner MA, Lamph WW, Heyman RA. Chemoprevention of mammary carcinoma by LGD1069 (Targretin): an RXR-selective ligand. Cancer Res. 1996;56:5566–5570. [PubMed] [Google Scholar]
  • 184.Anzano MA, Byers SW, Smith JM, Peer CW, Mullen LT, Brown CC, Roberts AB, Sporn MB. Prevention of breast cancer in the rat with 9-cis-retinoic acid as a single agent and in combination with tamoxifen. Cancer Res. 1994;54:4614–4617. [PubMed] [Google Scholar]
  • 185.Zujewski J, Pai L, Wakefield L, Giusti R, Dorr FA, Flanders C, Caruso R, Kaiser M, Goodman L, Merino M, Gossard M, Noone MA, Denicoff A, Venzon D, Cowan KH, O’Shaughnessy JA. Tamoxifen and fenretinide in women with metastatic breast cancer. Breast Cancer Res Treat. 1999;57:277–283. doi: 10.1023/a:1006216409688. [DOI] [PubMed] [Google Scholar]
  • 186.Liu L, Gudas LJ. Retinoic acid induces expression of the interleukin-1beta gene in cultured normal human mammary epithelial cells and in human breast carcinoma lines. J Cell Physiol. 2002;193:244–252. doi: 10.1002/jcp.10173. [DOI] [PubMed] [Google Scholar]
  • 187.Fabian CJ, Kimler BF. Chemoprevention for high-risk women: tamoxifen and beyond. Breast J. 2001;7:311–320. doi: 10.1046/j.1524-4741.2001.21570.x. [DOI] [PubMed] [Google Scholar]
  • 188.Li Y, Brown PH. Prevention of ER-negative breast cancer. Recent Results Cancer Res. 2009;181:121–134. doi: 10.1007/978-3-540-69297-3_13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Raif A, Marshall GM, Bell JL, Koach J, Tan O, D’Andreti C, Thomas W, Sekyere E, Norris M, Haber M, Kavallaris M, Cheung BB. The estrogen-responsive B box protein (EBBP) restores retinoid sensitivity in retinoid-resistant cancer cells via effects on histone acetylation. Cancer Lett. 2009;277:82–90. doi: 10.1016/j.canlet.2008.11.030. [DOI] [PubMed] [Google Scholar]
  • 190.Yang X, Guo M, Wan YJ. Deregulation of growth factor, circadian clock, and cell cycle signaling in regenerating hepatocyte RXRalpha-deficient mouse livers. Am J Pathol. 2010;176:733–743. doi: 10.2353/ajpath.2010.090524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Bushue N, Wan YJ. Retinoic Acid-mediated Nuclear Receptor Activation and Hepatocyte Proliferation. 2009;1:23–30. doi: 10.1016/S1878-3317(09)60007-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Theodosiou M, Laudet V, Schubert M. From carrot to clinic: an overview of the retinoic acid signaling pathway. Cell Mol Life Sci. 2010 doi: 10.1007/s00018-010-0268-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Vuletic A, Konjevic G, Milanovic D, Ruzdijic S, Jurisic V. Antiproliferative Effect of 13-cis-Retinoic Acid is Associated with Granulocyte Differentiation and Decrease in Cyclin B1 and Bcl-2 Protein Levels in G0/G1 Arrested HL-60 Cells. Pathol Oncol Res. 2010 doi: 10.1007/s12253-009-9241-2. [DOI] [PubMed] [Google Scholar]
  • 194.Varani J, Fay K, Perone P. MDI 301, a non-irritating retinoid, induces changes in human skin that underlie repair. Arch Dermatol Res. 2007;298:439–448. doi: 10.1007/s00403-006-0720-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Chiu YY, Roth MD, Kolis S, Rogovitz D, Davies B. Pharmacokinetics of a novel agent, R667, in patients with emphysema. Br J Clin Pharmacol. 2007;63:527–533. doi: 10.1111/j.1365-2125.2006.02808.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Hansen NJ, Wylie RC, Phipps SM, Love WK, Andrews LG, Tollefsbol TO. The low-toxicity 9-cis UAB30 novel retinoid down-regulates the DNA methyltransferases and has anti-telomerase activity in human breast cancer cells. Int J Oncol. 2007;30:641–650. [PMC free article] [PubMed] [Google Scholar]
  • 197.Jiang W, Deng W, Bailey SK, Nail CD, Frost AR, Brouillette WJ, Muccio DD, Grubbs CJ, Ruppert JM, Lobo-Ruppert SM. Prevention of KLF4-mediated tumor initiation and malignant transformation by UAB30 rexinoid. Cancer Biol Ther. 2009;8:289–298. doi: 10.4161/cbt.8.3.7486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Piskin S, Uzunali E. A review of the use of adapalene for the treatment of acne vulgaris. Ther Clin Risk Manag. 2007;3:621–624. [PMC free article] [PubMed] [Google Scholar]
  • 199.Yin JJ, Xia Q, Fu PP. UVA photoirradiation of anhydroretinol--formation of singlet oxygen and superoxide. Toxicol Ind Health. 2007;23:625–631. doi: 10.1177/0748233708090909. [DOI] [PubMed] [Google Scholar]
  • 200.Zhang XK. Targeting Nur77 translocation. Expert Opin Ther Targets. 2007;11:69–79. doi: 10.1517/14728222.11.1.69. [DOI] [PubMed] [Google Scholar]
  • 201.Zhang Y, Soto J, Park K, Viswanath G, Kuwada S, Abel ED, Wang L. Nuclear receptor SHP, a death receptor that targets mitochondria, induces apoptosis and inhibits tumor growth. Mol Cell Biol. 2010 doi: 10.1128/MCB.01076-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Zuco V, Benedetti V, De Cesare M, Zunino F. Sensitization of ovarian carcinoma cells to the atypical retinoid ST1926 by the histone deacetylase inhibitor, RC307: enhanced DNA damage response. Int J Cancer. 2010;126:1246–1255. doi: 10.1002/ijc.24819. [DOI] [PubMed] [Google Scholar]
  • 203.Farhana L, Dawson MI, Dannenberg JH, Xu L, Fontana JA. SHP and Sin3A expression are essential for adamantyl-substituted retinoid-related molecule-mediated nuclear factor-kappaB activation, c-Fos/c-Jun expression, and cellular apoptosis. Mol Cancer Ther. 2009;8:1625–1635. doi: 10.1158/1535-7163.MCT-08-0964. [DOI] [PubMed] [Google Scholar]
  • 204.Perez-Rodriguez S, Ortiz MA, Pereira R, Rodriguez-Barrios F, de Lera AR, Piedrafita FJ. Highly twisted adamantyl arotinoids: synthesis, antiproliferative effects and RXR transactivation profiles. Eur J Med Chem. 2009;44:2434–2446. doi: 10.1016/j.ejmech.2009.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Komi Y, Sogabe Y, Ishibashi N, Sato Y, Moriwaki H, Shimokado K, Kojima S. Acyclic retinoid inhibits angiogenesis by suppressing the MAPK pathway. Lab Invest. 2010;90:52–60. doi: 10.1038/labinvest.2009.110. [DOI] [PubMed] [Google Scholar]
  • 206.Shimizu M, Takai K, Moriwaki H. Strategy and mechanism for the prevention of hepatocellular carcinoma: phosphorylated retinoid × receptor alpha is a critical target for hepatocellular carcinoma chemoprevention. Cancer Sci. 2009;100:369–374. doi: 10.1111/j.1349-7006.2008.01045.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Chengedza S, Benbrook DM. NF-kappaB is involved in SHetA2 circumvention of TNF-alpha resistance, but not induction of intrinsic apoptosis. Anticancer Drugs. 2010;21:297–305. doi: 10.1097/CAD.0b013e3283350e43. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES