Open access peer-reviewed chapter

Retinoids in Treatment of Colorectal Cancer

Written By

Caroline O.B. Facey and Bruce M. Boman

Submitted: 17 June 2020 Reviewed: 24 August 2020 Published: 24 September 2020

DOI: 10.5772/intechopen.93699

From the Edited Volume

Colorectal Cancer

Edited by Alberto Vannelli

Chapter metrics overview

461 Chapter Downloads

View Full Metrics

Abstract

Retinoids are vitamin A metabolites best known for their role in embryonic development. Indeed, retinoid acid (RA) signaling plays a key role in regulating the development of the embryo body-plan by controlling embryonic stem cells (SCs). Retinoids function through their ability to induce cellular differentiation. Mutations in RA signaling pathway genes occur in most human cancers. The classic example is the chromosomal translocation involving RA receptor alpha in acute promyelocytic leukemia (APL). Because all-trans retinoic acid (ATRA) is a highly effective and often curative treatment for APL patients, determining if retinoids are efficacious for other cancer types is imperative. We review the current research on retinoids in colorectal cancer (CRC) and provide bioinformatics analyses of RA signaling. Our results show that most RA pathway genes are overexpressed and often mutated in CRC. Moreover, aberrant expression of many RA signaling proteins predicts decreased CRC patient survival. We also review aldehyde dehydrogenase (ALDH) expression in CRC because ALDH is a key enzyme in RA signaling, which regulates colonic SCs. Further investigation of RA signaling mechanisms that regulate colon SCs and how dysregulation contributes to the SC overpopulation that drives CRC growth should provide insight into strategies for designing new SC-targeted therapies for CRC.

Keywords

  • retinoic acid
  • stem cells
  • colon cancer
  • adenomatous polyposis coli
  • aldehyde dehydrogenase

1. Introduction

Our goal herein is to review current research findings on retinoids in colorectal cancer (CRC), and to provide an update from our bioinformatics analysis of RA signaling components in CRC. Retinoic acid (RA) is currently being used in the treatment of specific types of human cancers [1]. The classic example is use of ATRA as first line treatment for acute promyelocytic leukemia (APL). RA therapy has also been shown to improve survival in patients with neuroblastoma [2, 3, 4]. Additionally, RA-based agents have been evaluated for clinical anti-cancer activity in breast cancer and in lung cancer [5]. In this review, we discuss the anti-cancer activity of retinoids using in vitro and in vivo models of CRC, and the use of ATRA as a differentiation agent in SC research [4, 6, 7, 8].

A strong rationale to investigate RA signaling in oncology research is that ATRA is an effective drug used to treat APL patients. Indeed, ATRA effectively induces APL cells to terminally differentiate into neutrophils [9, 10, 11]. Current treatment regimens for APL also include arsenic in combination with ATRA because the combination provides a synergic drug response that cures the majority of APL patients, who would otherwise be facing a highly fatal illness. The precise mechanism involved in triggering APL cells have been extensively studied with the hope of understanding how it can be applied to trigger differentiation in other cancer types. What appears to be the basis for clinical success in treating APL is that the RA/arsenic combination not only induces terminal differentiation, but it also abrogates self-renewal of APL SCs [12]. Thus, future retinoid-based treatments for other cancers will likely necessitate drug combinations that incorporate a RA signaling differentiation therapy and a SC-targeting therapy that inhibits cancer SC self-renewal.

Advertisement

2. Key components of the retinoic acid signaling pathway

To understand how the RA signaling pathway is altered in cancer and to provide a basis for designing retinoid-based treatment approaches to cancer, we provide a brief description of the key components in the RA signaling pathway. The reader is referred to Das et al. [13] for more detailed information. Listed below are the main proteins essential to proper functioning of the RA signaling pathway. A simplified schematic of the RA signaling pathway is illustrated in Figure 1.

Figure 1.

This figure illustrates a simplified schematic of the RA signaling pathway, which plays a key role in embryogenesis and adult tissue homeostasis. The cell surface protein STRA6 accepts all-trans retinol from the extracellular milieu to transfer it across the cell membrane into the cytoplasm. STRA6 does not transport retinoic acid. After transfer or diffusion into cytoplasm, the internalized free retinol is bound to CRBP or is oxidized to retinal by retinol dehydrogenases (RDH) or alcohol dehydrogenases (ADH) and eventually to form all-trans retinoic acid (ATRA) by aldehyde dehydrogenases (ALDHs). ATRA then binds to cellular retinoic acid-binding proteins (CRABPs), which transfers ATRA to the nucleus. Once localized in the nucleus, ATRA serves as a ligand for binding to retinoid X receptors (RXRs) and retinoic acid receptors (RARs). Once ATRA travels to the nucleus, it binds RARs to induce the transcription of retinoid-responsive genes. Specifically, bound ATRA (or other ligands such as 9-cis) induces formation of a heterodimer (RA:RAR:RXR) in a complex at retinoic acid receptor elements on DNA, which then is able to induce transcription of RA-response genes. Thus, the RAR:RXR heterodimer acts as the main transcription factor in the classical RA signaling pathway. Nonetheless, the rate of formation of the RA:RAR:RXR complex, is still affected by other intracellular RA binding proteins such as CRABPs, which can sequester RA in the cytosol and limit the amount of RA available for binding to RARs. CRABPs can also facilitate RA degradation by directing RA to CYP26A1 RA-degrading enzymes. STRA6 = stimulated by retinoic acid 6, RDHs = retinol dehydrogenases, ADHs = alcohol dehydrogenases, RBPs = retinol binding proteins, ALDHs = aldehyde dehydrogenases, CRABPs = cellular retinoic acid binding proteins, CYP26A1 = cytochrome p450 family 26 subfamily a member 1, RARE = retinoic acid response element; RXRs = retinoid X receptors, RARs = retinoic acid receptors.

2.1 STRA6 (stimulated by retinoic acid 6)

STRA6 is a cell surface protein that functions as a receptor to accept all-trans retinol from the extracellular retinol-binding protein RBP4 and to transport retinol across the cell membrane. STRA6 removes the retinol from RBP4 and transfers it to RBP1 in the cytoplasm. STRA6 does not transport RA.

2.2 LRAT (lecithin retinol acyltransferase)

LRAT is an enzyme that converts retinol to all-trans retinyl esters, which is a storage form of vitamin A. LRAT also functions to enhance cellular uptake of retinol by STRA6, which contributes to the activation of the RA signaling cascade.

2.3 RDHs (retinol dehydrogenases)

RDHs are a family of dehydrogenase enzymes involved in the conversion of retinol to retinaldehyde by catalyzing the oxidation of cis-isomers of retinol, including 11-cis-, 9-cis-, and 13-cis-retinol in an NAD-dependent manner. This family of short-chain dehydrogenases/reductases functions to catalyze the final step in the biosynthesis of 11-cis retinaldehyde.

2.4 DHRS3 (retinaldehyde reductase-3)

DHRS3 is an oxidoreductase that catalyzes the oxidation/reduction of all-trans-retinal to all-trans-retinol in the presence of NADPH. DHRS3 is essential for preventing the formation of excess RA during embryonic development.

2.5 ADHs (alcohol dehydrogenases)

ADHs are a family of alcohol dehydrogenases involved in retinoid metabolism via conversion of retinol to retinaldehyde by catalyzing the NAD-dependent oxidation of all-trans-retinol and its derivatives such as all-trans-4-hydroxyretinol. These enzymes metabolize a wide variety of substrates, including ethanol, retinol, aliphatic alcohols, hydroxysteroids, and products of lipid peroxidation. ADHs consist of several homo- and heterodimers of alpha, beta, and gamma subunits, which plays major roles in ethanol catabolism. For example, three genes encoding alpha, beta, and gamma subunits of ADH1 are tandemly organized in a genomic segment as a gene cluster.

2.6 RBPs (retinol-binding proteins)

RBP1 (Retinol Binding Protein 1) and RBP2 (Retinol Binding Protein 2) are cytoplasmic retinol-binding proteins, which contribute to retinol uptake, storage, and retinoid homeostasis. Specifically, RBP1 is the carrier protein for transport of retinol from the liver storage site to peripheral tissue. RBP2 also plays an important role in the uptake and intracellular transport of retinol, which is necessary for intracellular metabolism of vitamin A.

2.7 ALDHs (aldehyde dehydrogenases)

ALDHs are cytoplasmic enzymes that convert/oxidize retinaldehyde to RA. ALDHs are the enzymes that function after the alcohol dehydrogenase step in the RA signaling pathway. Nineteen ALDH isoforms encoded by 19 different genes exist in humans with as many orthologs in the mouse plus some alternatively spliced transcriptional variants. Through its role in retinol metabolism, ALDHs play a major role in the regulation of responses to RA.

2.8 CRABPs (cellular retinoic acid-binding proteins)

CRABP1 (Cellular Retinoic Acid Binding Protein 1) and CRABP2 (Cellular Retinoic Acid Binding Protein 2) are paralogous genes that encode cellular RA binding proteins. These proteins transport RA to the nucleus and function to regulate the access of RA to the nuclear RA receptors. Specifically, CRABPs are cytosol-to-nuclear shuttling proteins, which facilitate RA binding to its cognate receptor complex and nuclear transfer. These activities in the retinoid signaling pathway play an important role in RA-mediated differentiation and proliferation processes. CRABPs are structurally similar to the cellular retinol-binding proteins, but CRABPs only bind RA, which contributes to RA-directed differentiation in epithelial tissue. Diseases associated with CRABPs include embryonal carcinomas.

2.9 CYP26A1 (cytochrome P450 family 26 subfamily A member 1)

CYP26A1 is a cytochrome P450 monooxygenase that plays a key role in the metabolism of ATRA. The cytochrome P450 superfamily contains 57 members that are monooxygenase enzymes which catalyze many processes including drug metabolism and synthesis of cholesterol, steroids, and various lipids. CYP26A1 acts on ATRA by catalyzing the hydroxylation of carbon hydrogen bonds of ATRA. This includes both 4-hydroxylation and 18-hydroxylation activities. It has little activity toward 9-cis and 13-cis RA ligands. By regulating intracellular concentrations of RA, CYP26A1 can control RA signaling mediated gene expression in both embryonic and adult tissues. There are two alternatively spliced transcript variants of CYP26A1 that encode the different isoforms. This enzyme regulates the cellular level of RA which in turn regulates gene expression in both embryonic and adult tissues. Diseases associated with CYP26A1 include embryonal carcinoma and APL.

2.10 Retinoid X receptors (RXRs) and retinoic acid receptors (RARs)

The proteins encoded by RARs (RARA, RARB, RARG) and RXR (RXRA, RXRB, RXRG) genes are classified as members of the steroid and thyroid hormone receptor superfamily of transcriptional regulators. Various receptor isoforms can result from differential splicing of RA receptor genes and alternate promoter usage. RXRs and RARs are nuclear receptors that are central to retinoid acid (RA) signaling through their role in RA-mediated gene activation in response to their ligands ATRA or 9-cis retinoic acid. The 9-cis RA ligand has a high affinity for RXRs. These receptors are localized to cytoplasm and sub-nuclear compartments where they can bind RA to activate cellular signaling by forming homodimers or heterodimers. These dimers primarily act as transcription factors via binding to the retinoic acid response elements (RARE) made of tandem 5’-AGGTCA-3′ sites known as DR1-DR5. When the ligand is absent, RXRA/RARB forms a multiprotein complex containing transcription co-repressors that can induce histone deacetylation, chromatin condensation and transcriptional suppression. When the ligand is present, it induces the co-repressors to dissociate from the receptors and co-activators are recruited which leads to transcriptional activation. Moreover, depending on the RARE DNA element condition, the heterodimer can act as a transcriptional repressor or transcriptional activator. For example, the heterodimer can act as a repressor on the DR1 element and as an activator on the DR5 element. RA receptors can also dimerize with thyroid hormone, and vitamin D receptors, which increases their DNA binding and transcriptional effects on their respective response elements. RA signaling regulates gene expression in various biological processes such as embryonic morphogenesis, granulocytopoiesis, and skeletal growth. It also plays an essential role in mediating the antiproliferative effects of RA by inducing cellular differentiation and apoptosis. In oncology, translocations between RARA and other loci are associated with the development of APL.

Now that we have briefly covered the key components in the RA signaling pathway that are critical to its proper function, we will discuss alterations of this pathway that occur in CRC.

Advertisement

3. Studies on alterations of retinoic acid signaling in CRC

Many studies have been done to identify mechanisms that explain how RA resistance occurs in solid tumors. Indeed, CRCs have been shown to lose the ability to produce ATRA and fail to growth inhibit or differentiate in response to treatment with ATRA [14, 15, 16]. Retinoic acid resistance appears to arise spontaneously in human cancers. To assess how alterations in RA signaling components effect response to RA ligands, we performed a literature search. Most of the published studies discussed below used in vitro experiments on CRC cell lines and analysis of human CRC tissues.

In a study by Jette et al. [16], seven CRC cell lines were evaluated for retinol dehydrogenase (RDH) enzymatic activity. They found CRC cells have decreased conversion of retinol into RA compared to normal cells. This inhibition of RDH expression appeared to be due to loss of adenomatous polyposis coli (APC) function. Interestingly, reintroduction of wild-type APC into an APC-mutant CRC cell line (HT29) increased expression of DHRS9 (RDHL) but not RDH5. Transfection of wild-type APC also increased production of RA. This study indicates intracellular crosstalk occurs between WNT signaling and RA signaling pathways.

Another study by Park et al. [14] examined the ability of retinol to inhibit the growth of CRC cell lines. They observed that some CRC cells are ATRA-sensitive (HCT-15) and other cells are ATRA-resistant (HCT-116, SW620, and WiDR). They also found that retinol inhibited the growth of both ATRA-sensitive and ATRA-resistant CRC cells through a RA receptor-independent mechanism.

Other studies by Shelton et al. [17] evaluated for over-expression of CYP26A1 enzymes that could lead to increased ATRA degradation. Indeed, CYP26A1 was upregulated in APC–deficient CRC tissues which provides a mechanism that might explain how increased WNT-signaling might be tied to impaired RA-signaling function in ATRA-resistant cells.

Lecithin retinol acyltransferase (LRAT), which esterifies retinol to retinyl esters, has also been evaluated by Cheng et al. [18]. Indeed, the LRAT gene promoter was hypermethylated in CRC cell lines and neoplasms compared to normal tissue [18]. A decrease in LRAT expression due to hypermethylation could lower availability of retinoids and reduce intracellular storage of retinol.

Additionally, several studies have investigated whether RA receptors are intact in CRC cells [19]. We discuss below a few studies that reported loss of RAR in CRC cells. In one study by Moison et al. [20], epigenetic changes appeared to lead to loss of RARB expression in HCT116 cells from DNA hypermethylation [20]. Interestingly, a DNA methylation inhibitor is able to restore RARB expression [21]. In a second study by Nicke et al. [22], the RA-resistant LoVo CRC line was induced to over-express RARB, which produced responsiveness to ATRA resulting in growth inhibition. A third study by Lee et al. [23] had similar results. They observed that ATRA treatment of RA-sensitive and RA-resistant CRC lines induced RARA expression in all cell lines, but ATRA only increased RARB expression in lines that were sensitive to RA. The DLD-1 RA resistant cells acquired sensitivity to ATRA when RARB was over-expressed. Additional studies that examine RA resistance due to alterations in RARs have also been reported [23, 24].

Finally, a recent study by Kropotova et al. [15] used RT-PCR to measure expression patterns of genes involved in ATRA biosynthesis. They evaluated normal human colorectal tissues, primary carcinomas, and cancer cell lines. Expression of most genes involved in ATRA synthesis was altered in CRC tumors and colorectal cell lines. Moreover, the expression of several genes, particularly ADH isoforms ADH2 and ADH3, showed decreased gene expression in adenomas when compared to more advanced carcinomas.

Overall, the studies on CRC discussed above show that RA signaling components become altered at many levels along the pathway. This includes: (i) loss of RAR expression that impairs RA response and gene transcription; (ii) decreased ability to enzymatically synthesize ATRA; (iii) LRAT alterations that impair retinoid storage; (iv) enhanced degradation of ATRA via CYP26A1. Many of these alterations appear to be a consequence of the mutations, such as APC, that drive CRC development [1, 25]. Thus, as CRC progresses, tumor cells develop resistance to ATRA by losing their ability to produce and respond to it, as well as, by causing its degradation.

Advertisement

4. Animal model studies

In addition to the studies on RA signaling in cell lines and CRC tissues discussed above, other important investigations have been done using animal models. Many of these studies were done using azoxymethane (AOM) or 1,2-dimethylhydrazine (DMH) to induce colonic neoplasms in rats to investigate the anti-tumor effects of retinoids [26]. An early study by Stopera and Bird [27] found that ATRA treatment reduced the number of AOM-induced aberrant crypt foci (ACF), a precursor to CRCs. Two studies [28, 29] using the DMH-induced colon carcinogenesis model indicated that vitamin A dietary supplementation may diminish ACF formation. Other studies by Wargovich et al. [30, 31] reported that 13-cis-retinoic acid (13-cRA), 9-cis-retinoic acid, and the synthetic Vitamin A derivative 4-hydroxy-phenretinamide (4-HPR) diminished AOM-induced ACF in rats. An interesting study by Zheng et al. [32] screened thirteen retinoids for prevention of ACF. They found that two retinoids, 9-cis-retinoic acid and 4-HPR, reduced both colonic ACF and tumor formation. In another study by Zheng et al. [33], 2-(carboxyphenyl)retinamide (2-CPR) was evaluated because it prevents ACF. However, they found that this synthetic retinoid analogue increased the number of colon tumors. Thus, these studies on rats show that ATRA, retinol, 9-cis-retinoic acid, 4-HPR, 13-cRA, and 2-CPR can inhibit the formation of carcinogen-induced ACF. However, only 9-cis-retinoic acid and 4-HPR were shown to reduce colonic tumor formation, and 2-CPR actually increased the number of colon tumors in this rat model.

Several other animal studies to evaluate the effect of retinoids have employed the ApcMin/+ mouse model. Experiments using this model are important because these mice develop intestinal tumors due to Apc mutations and APC is a driver mutation for CRC growth in humans.

A study of ApcMin/+ mice by Volate et al. [34] showed that retinoic receptors including Rara, Rarb, Rxrb, Rxrg were all expressed in ApcMin/+ adenomas. However, in AOM-treated ApcMin/+ mice, Rxra was selectively downregulated in intestinal tumors. Therefore, these findings indicate that Rxra downregulation occurs early in CRC carcinogenesis and is not dependent on Apc mutations and beta-catenin.

Another study by Mollersen et al. [35] administered ATRA to ApcMin/+ mice and discovered that ATRA treatment failed to prevent tumor formation. Three studies were then performed that gave results which provide mechanisms that helps explain this unexpected ATRA resistance.

One line of investigation focused on C-Terminal Binding Protein 1 (CTBP1), which has been reported to inactivate retinoid dehydrogenase RDH [36]. Examination of adenomas from ApcMin/+ mice and familial adenomatous polyposis coli (FAP) patients showed an increased expression of CTBP1. Because CTBP1 decreases RDH levels, upregulated CTBP1 will lead to lower ATRA levels in tumors [37].

In another study on ApcMin/+ mice, Shelton et al. [17] analyzed expression levels of CYP26A1, the major RA catabolic enzyme. They found that CYP26A1 expression was increased in tumors from ApcMin/+ mice, and in tumors from FAP patients. They also determined that CYP26A1 is a TCF4 target gene which explains why CYP26A1 expression is increased due to upregulated WNT signaling in APC mutant tissues. An increase in CYP26A1 would lead to increased ATRA degradation, which provides a mechanism that helps explain why ATRA treatment failed to prevent tumor development in ApcMin/+ mice.

A recent innovative study by Penny et al. [38] involved treating ApcMin/+ mice with the CYP26a inhibitor Liarozole. Administration of Liarozole to ApcMin/+ mice increased endogenous RA signaling (presumably by blocking ATRA metabolism) and effectively reduced intestinal adenoma numbers in these Apc mutant mice. We also found that treatment of human CRC cells with Liarozole decreased proliferation, sphere formation and size of the ALDH+ stem cell population [39]. This suggests that Liarozole might decrease tumor stem cell numbers in APC mutant tissues.

Thus, the above discussed animal model studies have provided valuable information on how the retinoid pathway might be targeted in designing treatment approaches for human CRC patients. The studies using chemical carcinogen models show that different retinoid drugs have different activities against colon tumors. The studies using the ApcMin/+ model reveal it might be an effective screen for other retinoid drugs that have anti-tumor activity against APC mutant tissues. Perhaps a reasonable place to start would be to screen other agents for their ability to inhibit specific cellular processes upregulated in tumors that lower endogenous ATRA levels and decrease RA signaling.

Advertisement

5. Clinical studies

There have been an increasing number of clinical trials done on solid tumors using retinoids. However, our search of trials listed www.clinicaltrials.gov does not show any trials on CRCs using retinoids, Tretinoin or Liarozole. There were several trials listed for breast, lung, prostate, pancreatic, renal, cervical, brain, skin, and several hematologic malignancies. Given the pre-clinical data discussed above, it seems like it would be reasonable to develop a retinoid-based trial for CRC.

Advertisement

6. Prospect for retinoid-based, stem cell-targeted therapies for CRC

We have been interested in the role of RA signaling in regulation of colonic SCs and how dysregulation of RA signaling may contribute to CRC development for several reasons: (i) RA regulates embryonic SCs during development [40] and WNT signaling, another key developmental pathway, has an opposing effect on embryonic SCs [41]. The idea that the mechanisms that regulate embryonic SCs are the same mechanisms that become dysregulated in the SC etiology of cancer [42] is intriguing because some scientists view cancer as aberrant organogenesis [43] and metastases as aberrant morphogenesis [44]. (ii) APC mutations occur in most CRCs (nearly 90%) during CRC development and APC mutation leads to constitutively activated WNT signaling. (iii) APC mutations that drive CRC development appear to do so by causing SC overpopulation [45]. (iv) ALDH, a key component in RA signaling, marks colonic SCs and tracks SC overpopulation during CRC development.

Indeed, our research team [39, 46, 47, 48, 49] and others [50, 51, 52] have been using ALDH as a marker to identify and isolate SCs from patient tissues for several years. ALDH not only marks colonic SCs, but ALDH+ cells also have SC properties of self-renewal, drug resistance, and cell differentiation potential [53]. For example, ALDH+ cells possess self-renewing ability as shown by sphere-forming ability in vitro and tumor-initiating ability in mice [46, 51, 52, 54]. The drug resistance property of ALDH+ SCs comes from aldehyde dehydrogenase’s enzymatic function, which is the cell’s natural detoxification mechanism [50, 55, 56]. The ability of ALDH+ cells to differentiate comes from ALDH’s functional role in the RA signaling pathway [5, 13, 57, 58, 59]. Moreover, we examined ALDH+ cells from colon tissues and observed that retinoid receptors RXR and RAR are selectively expressed in ALDH+ cells [39], which indicates that RA signaling mainly occurs via ALDH+ SCs. That RA signaling primarily occurs in ALDH+ stem cells provides a mechanism for selective treatment of SCs using RA analogues.

ATRA is commonly used as a differentiating agent in SC research. For example, we found that treatment of ALDH+ cancer SCs (CSCs) with ATRA inhibits cell proliferation, reduces SC proliferation, sphere formation, and SC population size, as well as enhances SC differentiation [39, 47]. Others have shown that retinoids decrease proliferation of ALDH+ SCs and, conversely, that inhibitors of ALDH increase proliferation of ALDH+ SCs [4, 6, 7, 8]. Because ALDH is key to retinoid acid (RA) signaling and retinoids are well known to promote differentiation of SCs [4], it follows that having ALDH in a SC provides the capacity for it to differentiate in response to retinoids.

Since APC mutations are known to increase WNT signaling in CRC, this raises the question: does increased WNT signaling lead to decreased retinoid signaling? Indeed, previous studies have implicated a role for APC in regulating RA biosynthesis and that APC mutations may lead to aberrant RA signaling [16, 36]. Notably, studies show that appropriately regulated WNT signaling is necessary for RA to induce neuronal differentiation of embryonic SCs [60]. Furthermore, not only does WNT suppress retinoid signaling, but conversely, increased RA signaling diminishes the ability of WNT signaling to block retinoid induction of the neural differentiation of SCs [61, 62]. That WNT signaling must be downregulated for neural differentiation to be inducible by RA treatment helps explain how APC mutation and increased WNT signaling might prevent maturation of ALDH+ colonic SCs in CRC development. Thus, it appears that APC mutations may alter the ability of ALDH+ SCs to differentiate in response to retinoids, which would lead to expansion of the ALDH+ SC population size in CRC [39, 46].

Advertisement

7. Bioinformatics analysis of retinoid signaling components in CRC

We extended our study of RA signaling in CRC herein by using bioinformatics to analyze expression and mutation of RA signaling genes in CRCs and identify RA pathway genes that predict CRC patient survival. We found that most genes in the RA pathway are overexpressed and many are mutated in CRC (Figure 2). This is consonant with our previous result showing that RAR, RXR and other RA signaling proteins are overexpressed in CRC, which parallels overpopulation of ALDH-positive SCs that occurs during CRC tumorigenesis [39, 46]. Moreover, we found that aberrant expression of many RA signaling proteins (10 of 27) predicted (p < 0.05) decreased survival of CRC patients (Figure 3). We refer the reader to the meta-analysis by Chen et al. [63] which reveals that increased ALDH also indicates a poor prognosis in CRC patients. These updated findings provide insight into the complexity of RA signaling mechanisms and how RA signaling, when dysregulated, contributes to the development of CRC.

Figure 2.

Bioinformatics analyses of RA signaling genes in CRC, including overexpression (a) and mutations (b). Bioinformatics data derived from cosmic catalog of somatic mutations in cancer (https://cancer.sanger.ac.uk/cosmic).

Figure 3.

Kaplan-Meier survival analysis of RA signaling genes that predict (p < 0.05) CRC patient survival. (A) shows survival curves for RARs (retinoic acid receptors), RXRs (retinoid X receptors), CRABPs (cellular retinoic acid binding proteins), and RBPs (retinol binding proteins). (B) shows survival curves for LRAT (lecithin retinol acyltransferase), RDHs (retinol dehydrogenases), and DHRS3 (retinaldehyde reductase-3). (B) shows survival curves for CYP26A1 (cytochrome P450 family 26 subfamily A member 1), STRA6 (stimulated by retinoic acid 6), and ADHs (alcohol dehydrogenases). Curves reflect low (blue) and high (red) gene expression. Y axis = survival probability (0.0–1.0). X axis = time (years 0–12). Bioinformatics data derived from the human protein atlas (https://www.proteinatlas.org).

Advertisement

8. Conclusion and future perspectives

Our results indicate that RA signaling, when dysregulated, plays a major role in the SC origin of CRC. Overall, our review provides a strong rationale for future exploration of retinoid therapies for CRC in precision oncology. A few clues gleaned from our review are as follows: (i) drug screens using CRC cell lines (Table 1) and knockout of RA-signaling genes in human CRC cells might identify which retinoid drugs are active against cells with specific mutations; (ii) ApcMin/+ mice may be useful to identify additional retinoid agents that are active against Apc mutant tissues; (iii) strategies for designing retinoid-based CRC therapies will likely need to incorporate retinoids into drug combination regimens; (4) CRCs will likely need to be genotyped to determine the status of RA signaling genes when administering RA-based treatments to CRC patients. Finally, continued discovery of the mechanisms that explain how RA signaling regulate normal colon SCs and how dysregulation of RA signaling in cancer SCs drive CRC growth should provide insight into how new SC-targeted therapies might be designed for CRC.

CRC Cell lineAPC mutationMicrosatellite instabilityBeta-catenin mutationRARA mutationRARB mutationRARG mutationRXRA mutationRXRB mutationRXRG mutation
HT-29YesStableNoNoNoNoNoNoNo
SW480YesStabeNoYesNoNoNoNoYes
HCT116YesHighYesYesNoNoNoNoNo
LoVoYesLowYesNoNoNoYesNoNo
DiFiYesStabeYesNoNoNoNoNoNo
SW48YesHighYesYesNoNoNoNoNo
SW1116YesStabeNoNoNoNoYesNoNo
COLO320YesStabeNoNoNoNoNoNoNo
LIM1863YesStabeYes??????
DLD-1YesStabeNo??????
RKONoHighNoNoNoNoYesNoYes

Table 1.

Bioinformatics results on RXR and RAR mutations in CRC cell lines.

Analysis done using the COSMIC database on human somatic mutations in cancer (https://cancer.sanger.ac.uk/cosmic).

8.1 Materials and methods

The bioinformatics analysis on overexpression and mutation of RA signaling component genes in CRCs was done through the COSMIC website (cancer.sanger.ac.uk/cosmic). Bioinformatics analysis to identify RA signaling genes that predict CRC patient survival was done through The Human Protein Atlas (https://www.proteinatlas.org).

Advertisement

Conflict of interest

The authors do not have any conflicts of interest.

Advertisement

Funding

This study was supported in part by the Lisa Dean Moseley Foundation (BB), Cancer B*Ware Foundation (BB), and Cawley Center for Translational Cancer Research Fund (BB, CF).

References

  1. 1. Mongan NP, Gudas LJ. Diverse actions of retinoid receptors in cancer prevention and treatment. Differentiation. 2007;75(9):853-870. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0301468109601778
  2. 2. Niles RM. Recent advances in the use of vitamin A (retinoids) in the prevention and treatment of cancer. Nutrition. 2000;16(11-12):1084-1089. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0899900700004366
  3. 3. Reynolds CP. Differentiating agents in pediatric malignancies: Retinoids in neuroblastoma. Vol. 2, Current Oncology Reports. Springer; 2000. pp. 511-518. Available from: https://link.springer.com/article/10.1007/s11912-000-0104-y
  4. 4. Gudas LJ, Wagner JA. Retinoids regulate stem cell differentiation. Journal of Cellular Physiology. 2010;226(2):322-330
  5. 5. Ma I, Allan AL. The role of human aldehyde dehydrogenase in normal and cancer stem cells. Stem Cell Reviews and Reports. 2010;7(2):292-306. DOI: 10.1007%2Fs12015-010-9208-4
  6. 6. Ginestier C, Wicinski J, Cervera N, Monville F, Finetti P, Bertucci F, et al. Retinoid signaling regulates breast cancer stem cell differentiation. Cell Cycle. 2009;8(20):3297-3302. Available from: http://www.tandfonline.com/doi/abs/10.4161/cc.8.20.9761
  7. 7. Tonge PD, Andrews PW. Retinoic acid directs neuronal differentiation of human pluripotent stem cell lines in a noncell-autonomous manner. Differentiation. 2010;80(1):20-30. DOI: 10.1016%2Fj.diff.2010.04.001
  8. 8. Chute JP, Muramoto GG, Whitesides J, Colvin M, Safi R, Chao NJ, et al. Inhibition of aldehyde dehydrogenase and retinoid signaling induces the expansion of human hematopoietic stem cells. Proceedings of the National Academy of Sciences. 2006;103(31):11707-11712. DOI: 10.1073%2Fpnas.0603806103
  9. 9. Idres N, Benoı̂t G, Flexor MA, Lanotte M, Chabot GG. Granulocytic differentiation of human NB4 promyelocytic leukemia cells induced by all-trans retinoic acid metabolites. Cancer Research. 2001;61:2
  10. 10. Zhu J-W, Shi XG, Chu HY, Tong JH, Wang Z, Naoe T, et al. Effect of retinoic acid isomers on proliferation, differentiation and PML relocalization in the APL cell line NB4. Leukemia. 1995;9(2):302-309
  11. 11. Fang J, Chen S-J, Tong J-H, Wang Z-G, Chen G-Q. Treatment of acute promyelocytic leukemia with ATRA and As2O3: A model of molecular. Cancer Biology & Therapy. 2002;1(6):614-620. DOI: 10.4161%2Fcbt.308
  12. 12. de Thé H. Differentiation therapy revisited. Nature Reviews Cancer. 2018;18(2):117-127. Available from: http://www.nature.com/articles/nrc.2017.103
  13. 13. Das BC, Thapa P, Karki R, Das S, Mahapatra S, Liu T-C, et al. Retinoic acid signaling pathways in development and diseases. Bioorganic & Medicinal Chemistry. 2014;22(2):673-683. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0968089613009565
  14. 14. Park EY, Dillard A, Williams EA, Wilder ET, Pepper MR, Lane MA. Retinol inhibits the growth of all-trans -retinoic acid–sensitive and all-trans-retinoic acid–resistant colon cancer cells through a retinoic acid receptor–independent mechanism. Cancer Research. 2005;65(21):9923-9933. Available from: http://cancerres.aacrjournals.org/lookup/doi/10.1158/0008-5472.CAN-05-1604
  15. 15. Kropotova ES, Zinovieva OL, Zyryanova AF, Dybovaya VI, Prasolov VS, Beresten SF, et al. Altered expression of multiple genes involved in retinoic acid biosynthesis in human colorectal cancer. Pathology Oncology Research. 2014;20(3):707-717. Available from: http://link.springer.com/10.1007/s12253-014-9751-4
  16. 16. Jette C, Peterson PW, Sandoval IT, Manos EJ, Hadley E, Ireland CM, et al. The tumor suppressor adenomatous polyposis coli and caudal related homeodomain protein regulate expression of retinol dehydrogenase L. Journal of Biological Chemistry. 2004;279(33):34397-34405. Available from: http://www.jbc.org/lookup/doi/10.1074/jbc.M314021200
  17. 17. Shelton DN, Sandoval IT, Eisinger A, Chidester S, Ratnayake A, Ireland CM, et al. Up-regulation of CYP26A1 in adenomatous polyposis coli–deficient vertebrates via a WNT-dependent mechanism: Implications for intestinal cell differentiation and colon tumor development. Cancer Research. 2006;66(15):7571-7577. Available from: http://cancerres.aacrjournals.org/lookup/doi/10.1158/0008-5472.CAN-06-1067
  18. 18. Cheng Y-W, Pincas H, Huang J, Zachariah E, Zeng Z, Notterman DA, et al. High incidence of LRAT promoter hypermethylation in colorectal cancer correlates with tumor stage. Medical Oncology. 2014;31(11):254. Available from: http://link.springer.com/10.1007/s12032-014-0254-7
  19. 19. Xu X-C. Tumor-suppressive activity of retinoic acid receptor-β in cancer. Cancer Letters. 2007;253(1):14-24. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0304383506006549
  20. 20. Moison C, Senamaud-Beaufort C, Fourrière L, Champion C, Ceccaldi A, Lacomme S, et al. DNA methylation associated with polycomb repression in retinoic acid receptor β silencing. The FASEB Journal. 2013;27(4):1468-1478. Available from: https://onlinelibrary.wiley.com/doi/abs/10.1096/fj.12-210971
  21. 21. Côté S, Sinnett D, Momparler RL. Demethylation by 5-aza-2′-deoxycytidine of specific 5-methylcytosine sites in the promoter region of the retinoic acid receptor beta gene in human colon carcinoma cells. Anti-Cancer Drugs. 1998;9(9):743-750. Available from: http://journals.lww.com/00001813-199810000-00001
  22. 22. Nicke B, Riecken E-O, Rosewicz S. Induction of retinoic acid receptor beta mediates growth inhibition in retinoid resistant human colon carcinoma cells. Gut. 1999;45(1):51-57. Available from: http://gut.bmj.com/cgi/doi/10.1136/gut.45.1.51
  23. 23. Lee M-O, Han S-Y, Jiang S, Han Park J, Kim SJ. Differential effects of retinoic acid on growth and apoptosis in human colon cancer cell lines associated with the induction of retinoic acid receptor β. Biochemical Pharmacology. 2000;59(5):485-496. Available from: https://linkinghub.elsevier.com/retrieve/pii/S000629529900355X
  24. 24. Freemantle SJ, Spinella MJ, Dmitrovsky E. Retinoids in cancer therapy and chemoprevention: Promise meets resistance. Oncogene. 23;22(47):7305-7315. Available from: http://www.nature.com/articles/1206936
  25. 25. Applegate CC, Lane MA. Role of retinoids in the prevention and treatment of colorectal cancer. World Journal of Gastrointestinal Oncology. 2015;7(10):184. Available from: http://www.wjgnet.com/1948-5204/full/v7/i10/184.htm
  26. 26. Pereira MA, Barnes LH, Rassman VL, Kelloff GV, Steele VE. Use of azoxymethane-induced foci of aberrant crypts in rat colon to identify potential cancer chemopreventive agents. Carcinogenesis. 1994;15(5):1049-1054. Available from: https://academic.oup.com/carcin/article-lookup/doi/10.1093/carcin/15.5.1049
  27. 27. Stopera SA, Bird RP. Effects of all-trans retinoic acid as a potential chemopreventive agent on the formation of azoxymethane-induced aberrant crypt foci: Differential expression of c-myc and c-fos mrna and protein. International Journal of Cancer. 1993;53(5):798-803. Available from: http://doi.wiley.com/10.1002/ijc.2910530516
  28. 28. Rogers AE, Herndon BJ, Newberne PM. Induction by dimethylhydrazine of intestinal carcinoma in normal rats and rats fed high or low levels of vitamin A. Cancer Research. 1973;33:5
  29. 29. Delage B, Groubet R, Pallet V, Bairras C, Higueret P, Cassand P. Vitamin A prevents high fat diet-induced ACF development and modifies the pattern of expression of peroxisome proliferator and retinoic acid receptor m-RNA. Nutrition and Cancer. 2004;48(1):28-36. Available from: http://www.tandfonline.com/doi/abs/10.1207/s15327914nc4801_5
  30. 30. Wargovich MJ, Chen CD, Harris C, Yang E, Velasco M. Inhibition of aberrant crypt growth by nonsteroidal anti-inflammatory agents and differentiation agents in the rat colon. International Journal of Cancer. 1995;60(4):515-519. Available from: http://doi.wiley.com/10.1002/ijc.2910600415
  31. 31. Wargovich MJ, Jimenez A, McKee K, Steele VE, Velasco M, Woods J, et al. Efficacy of potential chemopreventive agents on rat colon aberrant crypt formation and progression. Carcinogenesis. 2000;21(6):1149-1155. Available from: https://academic.oup.com/carcin/article-lookup/doi/10.1093/carcin/21.6.1149
  32. 32. Zheng Y. Prevention by retinoids of azoxymethane-induced tumors and aberrant crypt foci and their modulation of cell proliferation in the colon of rats. Carcinogenesis. 1997;18(11):2119-2125. Available from: https://academic.oup.com/carcin/article-lookup/doi/10.1093/carcin/18.11.2119
  33. 33. Zheng Y. Effect of retinoids on AOM-induced colon cancer in rats: Modulation of cell proliferation, apoptosis and aberrant crypt foci. Carcinogenesis. 1999;20(2):255-260. Available from: https://academic.oup.com/carcin/article-lookup/doi/10.1093/carcin/20.2.255
  34. 34. Volate SR, Muga SJ, Issa AY, Nitcheva D, Smith T, Wargovich MJ. Epigenetic modulation of the retinoid X receptor α by green tea in the azoxymethane-Apc min/+ mouse model of intestinal cancer. Molecular Carcinogenesis. 2009;48(10):920-933. Available from: http://doi.wiley.com/10.1002/mc.20542
  35. 35. Mollersen L. Dietary retinoic acid supplementation stimulates intestinal tumour formation and growth in multiple intestinal neoplasia (min)/+ mice. Carcinogenesis. 2003;25(1):149-153. Available from: https://academic.oup.com/carcin/article-lookup/doi/10.1093/carcin/bgg176
  36. 36. Nadauld LD, Chidester S, Shelton DN, Rai K, Broadbent T, Sandoval IT, et al. Dual roles for adenomatous polyposis coli in regulating retinoic acid biosynthesis and Wnt during ocular development. Proceedings of the National Academy of Sciences. 2006;103(36):13409-13414. Available from: http://www.pnas.org/cgi/doi/10.1073/pnas.0601634103
  37. 37. Phelps RA, Chidester S, Dehghanizadeh S, Phelps J, Sandoval IT, Rai K, et al. A two-step model for colon adenoma initiation and progression caused by APC loss. Cell. 2009;137(4):623-634. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0092867409002517
  38. 38. Penny HL, Prestwood TR, Bhattacharya N, Sun F, Kenkel JA, Davidson MG, et al. Restoring retinoic acid attenuates intestinal inflammation and tumorigenesis in APCMin/mice. Cancer Immunology Research. 2016;4(11):917-926. DOI: 10.1158%2F2326-6066.cir-15-0038
  39. 39. Modarai SR, Gupta A, Opdenaker LM, Kowash R, Masters G, Viswanathan V, et al. The anti-cancer effect of retinoic acid signaling in CRC occurs via decreased growth of ALDH colon cancer stem cells and increased differentiation of stem cells. Oncotarget. 2018;9(78):34658-34669. DOI: 10.18632%2Foncotarget.26157
  40. 40. Ross SA, McCaffery PJ, Drager UC, de Luca LM. Retinoids in embryonal development. Physiological Reviews. 2000;80(3):1021-1054. Available from: https://www.physiology.org/doi/10.1152/physrev.2000.80.3.1021
  41. 41. Mallo M, Alonso CR. The regulation of Hox gene expression during animal development. Development. 2013;140(19):3951. Available from: http://dev.biologists.org/content/140/19/3951.abstract
  42. 42. Bertrand FE, Angus CW, Partis WJ, Sigounas G. Developmental pathways in colon cancer. Cell Cycle. 2012;11(23):4344-4351. Available from: http://www.tandfonline.com/doi/abs/10.4161/cc.22134
  43. 43. Pierce GB, Speers WC. Tumors as caricatures of the process of tissue renewal: Prospects for therapy by directing differentiation. Cancer Research. 1988;48:8
  44. 44. Brabletz T, Jung A, Spaderna S, Hlubek F, Kirchner T. Migrating cancer stem cells — An integrated concept of malignant tumour progression. Nature Reviews Cancer. 2005;5(9):744-749. Available from: http://www.nature.com/articles/nrc1694
  45. 45. Boman BM, Huang E. Human colon cancer stem cells: A new paradigm in gastrointestinal oncology. Journal of Clinical Oncology. 2008;26(17):2828-2838. Available from: http://ascopubs.org/doi/10.1200/JCO.2008.17.6941
  46. 46. Huang EH, Hynes MJ, Zhang T, Ginestier C, Dontu G, Appelman H, et al. Aldehyde dehydrogenase 1 is a marker for normal and malignant human colonic stem cells (SC) and tracks SC overpopulation during colon tumorigenesis. Cancer Research. 2009;69(8):3382-3389. DOI: 10.1158%2F0008-5472.can-08-4418
  47. 47. Modarai SR, Opdenaker LM, Viswanathan V, Fields JZ, Boman BM. Somatostatin signaling via SSTR1 contributes to the quiescence of colon cancer stem cells. BMC Cancer. 2016;16(1):941-952. DOI: 10.1186/s12885-016-2969-7
  48. 48. Opdenaker LM, Modara SR, Boman BM. The proportion of ALDEFLUOR-positive cancer stem cells changes with cell culture density due to the expression of different ALDH isoforms. Cancer Studies and Molecular Medicine. 2015;2(2):87-95. Available from: http://openventio.org/Volume2-Issue2/The-Proportion-of-ALDEFLUOR-Positive-Cancer-Stem-Cells-Changes-with-Cell-Culture-Density-Due-to-the-Expression-of-Different-ALDH-Isoforms-CSMMOJ-2-113.pdf
  49. 49. Viswanathan V, Damle S, Zhang T, Opdenaker L, Modarai S, Accerbi M, et al. An miRNA expression signature for the human colonic stem cell niche distinguishes malignant from normal epithelia. Cancer Research. 2017;77(14):3778-3790. Available from: http://cancerres.aacrjournals.org/lookup/doi/10.1158/0008-5472.CAN-16-2388
  50. 50. Dylla SJ, Beviglia L, Park I-K, Chartier C, Raval J, Ngan L, et al. Colorectal cancer stem cells are enriched in xenogeneic tumors following chemotherapy. PLoS One. 2008;3(6):e2428. Available from: https://dx.plos.org/10.1371/journal.pone.0002428
  51. 51. Lin L, Liu A, Peng Z, Lin H-J, Li P-K, Li C, et al. STAT3 is necessary for proliferation and survival in colon cancer-initiating cells. Cancer Research. 2011;71(23):7226-7237. DOI: 10.1158%2F0008-5472.can-10-4660
  52. 52. Carpentino JE, Hynes MJ, Appelman HD, Zheng T, Steindler DA, Scott EW, et al. Aldehyde dehydrogenase-expressing colon stem cells contribute to tumorigenesis in the transition from colitis to cancer. Cancer Research. 2009;69(20):8208-8215. DOI: 10.1158%2F0008-5472.can-09-1132
  53. 53. Rodriguez-Torres M, Allan AL. Aldehyde dehydrogenase as a marker and functional mediator of metastasis in solid tumors. Clinical & Experimental Metastasis. 2016;33(1):97-113. Available from: http://link.springer.com/10.1007/s10585-015-9755-9
  54. 54. Shenoy A, Butterworth E, Huang EH. ALDH as a marker for enriching tumorigenic human colonic stem cells. In: Methods in Molecular Biology. Totowa, NJ: Humana Press; 2012. pp. 373-385. DOI: 10.1007/978-1-61779-980-8_27
  55. 55. Kozovska Z, Patsalias A, Bajzik V, Durinikova E, Demkova L, Jargasova S, et al. ALDH1A inhibition sensitizes colon cancer cells to chemotherapy. BMC Cancer. 2018;18(1):656-666. DOI: 10.1186/s12885-018-4572-6
  56. 56. Croker AK, Allan AL. Inhibition of aldehyde dehydrogenase (ALDH) activity reduces chemotherapy and radiation resistance of stem-like ALDHhiCD44+ human breast cancer cells. Breast Cancer Research and Treatment. 2012;133(1):75-87. Available from: http://link.springer.com/10.1007/s10549-011-1692-y
  57. 57. Marchitti SA, Brocker C, Stagos D, Vasiliou V. Non-P450 aldehyde oxidizing enzymes: The aldehyde dehydrogenase superfamily. Expert Opinion on Drug Metabolism & Toxicology. 2008;4(6):697-720. DOI: 10.1517%2F17425255.4.6.697
  58. 58. Bagirova M, Nehir O, Yaman S, Sefik E, Cakir R, Canim S, et al. Aldehyde dehydrogenase: Cancer and stem cells. In: Dehydrogenases. 2012. Available from: http://www.intechopen.com/books/dehydrogenases/aldehyde-dehydrogenase-cancer-and-stem-cells
  59. 59. Black WJ, Stagos D, Marchitti SA, Nebert DW, Tipton KF, Bairoch A, et al. Human aldehyde dehydrogenase genes: Alternatively spliced transcriptional variants and their suggested nomenclature. Pharmacogenetics and Genomics. 2009;19(11):893-902. Available from: http://journals.lww.com/01213011-200911000-00008
  60. 60. Engberg N, Kahn M, Petersen DR, Hansson M, Serup P. Retinoic acid synthesis promotes development of neural progenitors from mouse embryonic stem cells by suppressing endogenous, Wnt-dependent nodal signaling. Stem Cells. 2010;28(9):1498-1509. Available from: http://doi.wiley.com/10.1002/stem.479
  61. 61. Verani R, Cappuccio I, Spinsanti P, Gradini R, Caruso A, Magnotti MC, et al. Expression of the Wnt inhibitor Dickkopf-1 is required for the induction of neural markers in mouse embryonic stem cells differentiating in response to retinoic acid. Journal of Neurochemistry. 2007;100(1):242-250. Available from: http://doi.wiley.com/10.1111/j.1471-4159.2006.04207.x
  62. 62. Chuang J-H. Neural differentiation from embryonic stem cells in vitro: An overview of the signaling pathways. World Journal of Stem Cells. 2015;7(2):437. Available from: http://www.wjgnet.com/1948-0210/full/v7/i2/437.htm
  63. 63. Chen J, Xia Q , Jiang B, Chang W, Yuan W, Ma Z, et al. Prognostic value of cancer stem cell marker ALDH1 expression in colorectal cancer: A systematic review and meta-analysis. Suzuki H, editor. PLoS One. 2015;10(12):e0145164. DOI: 10.1371%2Fjournal.pone.0145164

Written By

Caroline O.B. Facey and Bruce M. Boman

Submitted: 17 June 2020 Reviewed: 24 August 2020 Published: 24 September 2020