Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Mar 12.
Published in final edited form as: Pharmacol Res. 2019 Jun 26;147:104331. doi: 10.1016/j.phrs.2019.104331

The potential of retinoids for combination therapy of lung cancer: Updates and future directions

Surya Kant Tripathi a, Kamal Pandey a, Munmun Panda a, Michael J Spinella b, Kannan RR Rengasamy c, Bijesh K Biswal a,*
PMCID: PMC7066582  NIHMSID: NIHMS1564922  PMID: 31254665

Abstract

Lung cancer is the most common cancer-related death worldwide. Natural compounds have shown high biological and pharmaceutical relevance as anticancer agents. Retinoids are natural derivatives of vitamin A having many regulatory functions in the human body, including vision, cellular proliferation and differentiation, and activation of tumour suppressor genes. Retinoic acid (RA) is a highly active retinoid isoform with promising anti-lung cancer activity. The abnormal expression of retinoid receptors is associated with loss of anticancer activities and acquired resistance to RA in lung cancer. The preclinical promise has not translated to the general clinical utility of retinoids for lung cancer patients, especially those with a history of smoking. Newer retinoid nano-formulations and the combinatorial use of retinoids has been associated with lower toxicity and more favorably efficacy in both the preclinical and clinical settings. Here, we highlight epidemiological and clinical therapeutic studies involving retinoids and lung cancer. We also discuss the biological actions of retinoids in lung cancer, which include effects on cancer stem cell differentiation, angiogenesis, metastasis, and proliferative. We suggest that the use of retinoids in combination with conventional and targeted anticancer agents will broaden the utility of these potent anticancer compounds in the lung cancer clinic.

Keywords: Retinoids, Lung cancer, Retinoic acid receptor, Chemoresistance, Combination therapy

1. Introduction

Lung cancer represents the second most diagnosed cancer with a high incidence of mortality in both men and women worldwide [1]. Based on histology, there are two main types of lung cancer: small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC). Both types of lung cancer are epithelial in origin and have an inferior prognosis. A small subpopulation of cells may be responsible for the aggressive behavior and resistance to chemotherapy of these tumours [2]. This subpopulation of cells is known as tumour-inducing cells (TICs) or cancer stem cells (CSCs). Both TICs and CSCs have self-renewing, tumour-initiating, and multipotent differentiation properties. Compounds that target TICs and CSCs and promote their differentiation would be predicted to have considerable therapeutic potential in lung cancer patients in both the early and advanced settings [3].

Vitamins are essential for healthy growth, differentiation, and development of the human body. Like most vitamins, animals do not synthesize vitamin A, which must be supplemented in the diet mainly from plants sources. Retinoids are vitamers of vitamin A, derived from retinyl esters. Retinoids have several isoforms including β-carotene, retinol, retinal, all-trans-retinoic acid, and 13-cis-retinal, among others [4]. In mammals, retinoids regulate vision, cellular proliferation, differentiation, the immune system, growth of skin and bone tissue, and perhaps most importantly for cancer, the activation of tumour suppressor genes [5,6]. Retinoids inhibit the growth and progression of various malignancies, including cancers of the lung, mouth, prostate, and skin [710]. Retinoids also can modulate various signaling pathways and inhibit cell proliferation, growth factor expression, and cell cycle progression in many human cancers, including lung cancer [6]. Retinoids regulate cyclins, CDKs and cell cycle inhibitors to mainly arrest cells in the G1 phase [11]. Retinoids exert anticancer activities via binding to nuclear receptors, the retinoic acid receptors (RARs), RARα, RARβ, and RARγ and their isoforms (Table 1). The main inducible and aberrantly expressed retinoid receptor isoform in tumorigenesis is RARβ2 [12]. Silencing of the RARβ2 gene has been associated with retinoid resistance, and restoration of RARβ2 expression can restore the anticancer activity of retinoids [13]. Epigenetic silencing of the RARβ2 gene has also been documented in lung cancer [14,15]. Combinatorial use of retinoids with tyrosine kinase inhibitors (TKIs) can restore TKI sensitivity in TKI resistant lung cancer patients [16]. The retinoid derivative all-trans retinoic acid, also called retinoic acid (RA) is approved by the Food and Drug Administration (FDA) for the treatment of lymphoma and leukemia. RA inhibits many biological functions, such as tumour growth, angiogenesis, and metastasis [17,18]. RA treatment of human cancer cells and tumour xenografts suppresses cancer cell growth and promotes apoptosis and differentiation of CSCs [3].

Table 1.

Alterations in retinoid receptors associated with retinoid resistance.

SI. No Model system Retinoic acid receptors Overcoming strategy Outcome References
1. Calu-1 cell line Does not express RARβ and thus RA resistant Transfection of pMARKCD7D5 vectors containing RARα, RARβ, RXRα, and RARγ Sensitized cancer cells towards RA after transfection with RARα, RARβ or RXRα but not RARγ [19]
2. In-vitro and in-vivo model Loss of RARβ2 and RARβ4 expression due to hypermethylation of promoter region 5-aza-deoxycytidine treatment Restored RARβ expression and inhibited metastatic lung cancer [20,21]
3. H1792 cell line Expresses endogenous RXRα but does not activate DR1 RXE after treatment with 9-cis-RA Transfection of exogenous RXRα Sensitized cells towards 9-cis-RA to activate DR1 RXE [19]
4. A549, HCC827, and NCI-H460 cell lines RARβ2 downregulated thus shows less sensitivity towards ATRA Combination of panobinostat + ATRA Showed enhanced inhibitory effects by synergistic H3 acetylation [22]
5. Clinical trial RARβ loss in smokers which is a pre-neoplasia symptom 9-cis-RA treatment Restored RARβ expression in bronchial epithelium in former smokers and reduction of metaplasia [23]
6. In-vitro model RARα, RARβ, and RXRα, RXRβ expressions unchanged after treatment Treatment with ATRA or 9-cis-RA along with HDAC inhibitors SL142 or SL325 Induced enhanced antitumor activity [24]
7. A/J mouse model Upregulation of RARβ expression after treatment 9-cis-RA treatment Lowered tumour multiplicity and tumour incidence [25]
8. In-vitro study RARβ and RXRβ upregulated post treatment Combination treatment of ATRA and 9-cis-RA Showed synergistic inhibition of lung cancer cell line [26]
9. Calu-6, H460, H292, SK-MES-1, and H661 cell lines RARβ expression induced upon treatment with ATRA only in Calu-6, and H460 cell line ATRA treatment Induced apoptosis in Calu-6, and H460 cell line while little efficacy in other cell lines [27]
10. In-vitro study Treatment in H1703 cells upregulated RARβ expression which had no basal expression of RARβ Treatment with ATRA or 13-cis-RA followed by 4-HPR Synergistic growth inhibitory effect [28]

Here, we summarize recent studies focused on molecular and cellular mechanisms regulated by retinoids in lung cancer. This review will also provide details on the clinical potential of retinoids in combination therapies for treatment naive and chemotherapy resistant lung cancer patients. We hope this review on retinoids and lung cancer provides useful insight and further understanding for researchers working in the field of lung cancer prevention and therapy.

2. Retinoids

Retinoids, a class of chemical compounds derivative from vitamin A, are implicated in stimulating cellular differentiation, growth, and development but also in inducing cell death [29,30]. Retinoids are composed of a cyclic hydrophobic moiety connected to a polar moiety via a linker unit. Mammals cannot synthesize vitamin A which must be obtained in the diet mainly from plant pigments such as carotenes (β-carotene, β-cryptoxanthin, and α-carotene) or animal sources (cod liver oil, eggs, butter) [6]. After consumption, retinol can be stored in the form of retinyl esters in hepatic stellate cells. An irreversible oxidation reaction converts retinal into RA and other isoforms broadly categorized as retinoids [31]. Isoforms of retinoids are found in the all-trans; 13-cis; 9, 13-di-cis; 11-cis; 11, 13-di-cis; and 9-cis configurations. Biologically active endogenous retinoids are all-trans retinoic acid (ATRA); 11-cis-retinal; 3,4-didehydro-RA; 14-hydroxy-4, 14-retro-retinol; 4-oxo-RA; 4-oxo-retinol; and 9-cis-RA [6]. Based on molecular structure, retinoids are classified into first-, second-, and third-generations. First-generation comprises naturally occurring, nonaromatic retinoids obtain from retinyl palmitate, retinol, retinal, ATRA, 9-cis-RA, and 13-cis-RA. Second-generation consists of synthetic mono-aromatic retinoids such as etretinate and acitretin while the third-generation includes synthetic polyaromatic retinoids such as bexarotene and fenretinide. Chemical structures of common natural and synthetic retinoids are provided in Fig. 1.

Fig. 1. Chemical structures of natural (1–6) and synthetic (7–8) retinoids.

Fig. 1.

Retinol (1), Retina (2), Retinoic acid (3), 13-cis Retinoic acid (4), 9-cis Retinoic acid (5), 14-Hydroxy-retro-retinol (6), Bexarotene (7), Fenretinide (8).

3. Retinoids in lung cancer

Initially, retinoids were used to treat ophthalmological and dermatological related conditions in humans. However, in the last few decades, retinoids have received considerable attention as anti-carcinogens alone or in combination with existing therapies due to their CSCs differentiating properties. Epidemiological studies established an inverse relationship between vitamin A intake and cancer risk. Aberrant expression of retinoid receptors is also associated with malignancy in animal tissues and cancer cells. Retinoids have documented tumour suppressor activity in many animal cancer models, including tumours of the lung, breast, oral cavity and prostate. Furthermore, clinical studies supported the potential of retinoids to reverse pre-malignancy in human epithelial tissues and to promote the differentiation of CSCs, leading to growth inhibition of several cancers, including lung cancer. This review focuses on molecular and cellular mechanisms of action of retinoids in lung cancer treatment.

3.1. The anti-cancer effect of retinoids in lung cancer angiogenesis and metastasis

Metastasis refers to the spread of cancer cells from a primary site to a new location, thereby colonizing distant organs [32]. Cancer which has metastasized is responsible for most cancer-related mortality. Angiogenesis plays a crucial role in metastatic spread. Angiogenesis is the development of new blood vessels from pre-existing vasculature [33]. These blood vessels not only fulfil the metabolic requirement of a growing tumour but also provide a passage for the migration of cancer cells to distant sites. Malignant cells secrete multiple proangiogenic factors that trigger new blood vessel formation in nearby healthy tissues from extant cells. These factors include vascular endothelial growth factor (VEGF), epidermal growth factor (EGF), basic fibroblast growth factor (bFGF), hypoxia-inducible factors, platelet-derived growth factors (PDGF), matrix metalloproteinases (MMPs), and angiopoietin [34]. A high serum level of VEGF is an indicator of poor prognosis in lung cancer patients. Thus, VEGF has long been recognized as a potential target to inhibit tumour progression and prolong survival. Sunitinib and sorafenib are small drug molecules which act by inhibiting multiple receptor tyrosine kinase (RTK) pathways involved in proangiogenic signaling [17]. Different classes of natural and synthetic retinoids have been found to display promising in-vitro and pre-clinical activities as anti-angiogenesis and angiopreventive agents.

RA appears to repress human colon adenocarcinoma cell movement by decreasing myosin light chain kinase (MLCK) expression and cancer cell metastasis [18]. Further investigation indicated that MLCK expression is upregulated in different types of tumours including lung, prostate, brain, liver, ovarian and melanoma [3537]. Thus, preventing MLCK overexpression is a potential mechanism to oppose the metastasis of cancer cells. Syndecan-1, a proteoglycan of lung epithelium is associated with tumour cell growth and invasiveness. RA positively regulates the syndecan-1 expression and inhibits invasion and metastasis in a mouse lung cancer model [38]. RA shows differential outcomes as an anti-angiogenic agent in various cancers. RA demonstrated anti-angiogenesis and anti-metastatic property in brain cancer and esophageal squamous cell carcinoma (ESCC). In glioma cells, RA shows anti-angiogenic activity by downregulating the expression of VEGF and HIF-1α [39]. Similarly, RA suppressed the expression of angiopoietin 1 (AP1), angiopoietin 2 (AP2), and receptor Tie-2 leading to decreased angiogenesis in ESCC cells [40,41]. In contrast, RA supports survival and migration of lung tumour cells by ERK pathway activation [42] and inhibition of the ERK signaling pathway reverses this RA activity. Therefore, combinatorial treatment of RA with ERK inhibitors may serve to inhibit lung cancer migration and survival. RA showed minimal efficacy as a single agent in a phase II trial in metastatic NSCLC, while the combination with other chemotherapeutic agents has proved promising results [43]. Grace et al. showed that liposomal RA surpassed the anti-metastatic potential of free RA in a lung cancer mouse model [44]. Liposomal RA resulted in higher RA concentrations in lung tissue and restricted tumour nodule formation, thus providing a possible benefit in inhibiting lung metastasis.

Bexarotene, a synthetic rexinoid, has demonstrated anti-angiogenic and anti-metastatic effects in lung cancer in a dose-dependent manner by activation of its dimerization partner PPARγ [45]. In-vitro treatment of bexarotene decreased expression of VEGF, EGF and MMPs, and upregulated tissue inhibitors of matrix metalloproteinases (TIMPs) secretion in lung cancer cells. Furthermore, bexarotene inhibits angiogenesis by directly inhibiting human umbilical vein endothelial cell growth and indirectly inhibiting tumour cell-mediated migration of human umbilical vein endothelial cells. Fu et al. also demonstrated the inhibitory effect of bexarotene on tumour-induced angiogenesis [46]. Bexarotene negatively regulated the expression of VEGF and suppressed JNK and ERK activation. Despite these results and promising findings in a phase II trial, the addition of bexarotene to first-line chemotherapeutic drugs in a phase III trial did not increase the survival of patients with advanced NSCLC [47]. Studies also suggested that subcutaneous administration of low dose interleukin-2 with oral 13-cis-RA inhibited VEGF expression and enhanced progression-free and overall survival of advanced stage NSCLC patients [48]. The above studies suggest a potential therapeutic role for retinoids as a potent anti-metastatic and anti-angiogenic agent in suppressing lung cancer progression (Fig. 2).

Fig. 2.

Fig. 2.

Model showing the anti-angiogenic and anti-metastatic effect of retinoic acid (RA) as a combinatorial drug in lung cancer progression. RA promotes angiogenesis and metastasis in lung cancer via activation of the extracellular-signal-regulated kinase (ERK) pathway. Activation of the ERK pathway upregulates hypoxia-inducible factor-alpha (HIF-α), activator protein-1 (AP-1), and activator protein-2 (AP-2) gene expression which favors the upregulation of many factors involved in angiogenesis and metastasis such as vascular endothelial growth factor (VEGF), epidermal growth factor (EGF), and matrix metalloproteinases (MMPs). However, activation of the ERK pathway alleviates the expression of MMPs inhibitors such as TIMP metallopeptidase inhibitor (TIMP). Treatment with a combination of RA and ERK inhibitors drastically overcomes this effect of RA resulting in inhibition of angiogenesis and metastasis in lung tumours. Bexarotene also inhibits angiogenesis and metastasis of lung tumours via ERK pathway inhibition. The high concentration of liposome encapsulated RA inhibits angiogenesis and metastasis in lung tumours via ERK pathway inhibition.

3.2. Retinoids associated aberrant signaling in lung cancer

Aberrant signaling is a unique feature of cancer malignancy. In most cancers, at least one cell signaling pathway is deregulated. Deregulated signaling pathways in cancer include many oncogenic pathways that can modulate cellular activities, including cell proliferation, differentiation, angiogenesis, apoptosis, and cell cycle progression. Due to their pleiotropic nature, retinoids can alter many signaling pathways involved in cancer initiation and progression. There are several distinct binding proteins through which retinoids exert their effects, such as retinol binding protein (RBP), stimulated by retinoic acid gene 6 (STRA6), cellular retinol binding protein (CRBP), cellular retinoic acid-binding protein (CRABP), and nuclear receptors such as retinoic acid receptors (RARs) and retinoid X receptors (RXRs). These receptors are present in the extracellular and intracellular compartments and transfer retinol from ingested food to target cells. First retinol binds with RBP and translocates into the cell by STRA6 mediated endocytosis or diffusion [49]. In lung cancer, STRA6 is found to be frequently upregulated, but the mechanism is poorly understood.

Upon entry into the cell, retinol binds CRBP, and the retinol dehydrogenase (RDH) reversibly oxidizes retinol to retinal. CRBP is a key component of the retinoid pathway and is dysregulated in lung adenocarcinoma. High expression of CRBP is associated with upregulation of the EGFR/Akt/Erk pathway whereas treatment with RA inhibits EGFR/Akt/Erk leading to inhibition of cell growth [11,42,50,51]. CRABP mediates subsequent RA transportation from the cytosol to the nucleus. The two isoforms of CRABP, CRABP-I, and CRAB-II have a high affinity for RA. Favorskaya et al. reported that CRABP-II is suppressed in NSCLC [52]. It has been documented that CRABP-I mediates non-canonical RAR leads to membrane signal-free initiation of ERK1/2 by RA [53]. In some cancers including lung cancer, CRABP-I acts as a tumour suppressor while others demonstrate an association between CRABP-I and cancerous growth and reduced survival [52,54,55]. Therefore, targeting retinoid receptors and their downstream signaling pathways could inhibit lung cancer growth and promote apoptosis of lung cancer cells.

RARβ induces cell cycle arrest and apoptosis in a RARα-dependent manner. RA-bound RARα binds to the cis-acting retinoic acid response element (RARE) in the RARβ promoter and recruits various activator proteins that upregulate RARβ2 gene expression [6]. RARβ mediated transactivation and expression of target genes is responsible for cancer cell apoptosis and differentiation. RARβ2 is found to be downregulated in bronchial biopsy specimens of heavy smokers, and cell lines of the liver, breast, and head and neck cancer [5659]. RARs are also substrates for protein kinase C (PKC), protein kinase A (PKA), cyclin-dependent kinase 7 (CDK7), and p38 [6063]. Srinivas et al. demonstrated that Akt phosphorylates the DNA-binding domain of RARα at Ser96, leading to inhibition of RARα transactivation [64]. Mutation of Ser96 suppressed RARα phosphorylation and transactivation, leading to retinoid resistance in NSCLC [64]. Karamouzis and collogues developed a ‘switch on/off model to represent the crosstalk between retinoid receptors and other signaling molecules during early stage bronchial carcinogenesis [65]. It has been shown that RXR selective agents inhibit the activity of AP-1 and promote anti-proliferative activity in respiratory epithelium. Further, RARβ and RXRα can crosstalk with other nuclear hormone receptors such as PPARγ leading to apoptosis [65].

RA inhibited EGFR expression at the transcriptional level by targeting the EGFR promoter leading to inhibition of lung cancer cell growth [66]. RA arrests TKI resistant NSCLC cells in the G0/G1 phase of the cell cycle by altering the expression of GATA6 and inhibits the activation of two important pathways involved in lung cancer progression namely EGFR and Wnt signaling to overcome TKI resistance [11]. GATA6 induced the downregulation of EGFR and CTNNB1 by binding to their promoter regions and also activated the Wnt inhibitor (FZD2) leading to inhibition of the Wnt signaling pathway. G-protein coupled receptor family C group 5 member A (GPRC5A), an RA-inducible gene is prominently expressed in normal lung tissue. In addition, the physical interaction of GPRC5A with EGFR is essential for EGFR inhibition [67,68]. Suppression of GPRC5A causes EGFR dysregulation, which is associated with lung tumorigenesis [69]. Overall, retinoids have a potent suppressive effect on signaling pathways that are aberrant in lung cancer, as summarized in Fig. 3.

Fig. 3. Retinoid signaling pathways leading to lung cancer stem cell (CSC) differentiation and cell cycle arrest.

Fig. 3.

Retinol binds with retinol binding protein (RBP) and translocates into lung cancer cells by retinoic acid gene 6 (STRA6) receptor mediated endocytosis or diffusion. Inside the cell, it interacts with cellular retinol binding protein (CRBP) and oxidizes into RA by enzyme retinol dehydrogenase (RDH). RA inhibits the EGFR/Wnt pathways via activation of transcription factor GATA-binding factor 6 (GATA6). GATA6 upregulates the expression of (SEPTB), mucin4 (MUC4), frizzled class receptor 2 (FZD2) and downregulates the expression of epidermal growth factor receptor (EGFR), catenin beta (CTNNB), and cyclin D1. Importantly, EGFR and CTNNB downregulation inhibit the activation of the Wnt signaling pathway; however, downregulation of cyclin D1 promotes lung CSC differentiation and cell cycle arrest. Furthermore, RA alone may promote cancer cell survival, proliferation, migration, and invasion via activation of PI3-Akt/ERK pathways. Combinatorial treatment of RA with PI3K/ERK inhibitors represses these pro-cancer effects of RA in lung tumours.

3.3. Retinoids as an epigenetic modulator in lung cancer

Aberrant methylation of CpG islands in gene promoters is a primary epigenetic mechanism for transcriptional silencing of tumour suppressor genes in cancer [7072]. Promoter hypermethylation of specific genes has the potential to be used to diagnosis and predict the risk for many types of cancers, including lung cancer [73]. Previous studies suggested that frequent aberrant methylation of the RARβ promoter silences RARβ expression during tumour progression, which indicated that RARβ is a tumour suppressor protein [7476]. It has been shown that inhibitors of DNA methyltransferases (DNMTs) and histone deacetylases (HDACs) can re-express epigenetically silenced RARβ [22]. RA or the pan-HDAC inhibitor (panobinostat) showed moderate efficacy in NSCLC cells. However, combined treatment had synergistic effects on global histone H3 acetylation and differentiation. Also, the combined treatment of RA and panobinostat showed enhanced efficacy in upregulating p53 and p21CIP1/WAF1 proteins and downregulating phospho-ERK and phospho-AKT. Han et al. showed that combination therapy of RA or 9-cis-RA and HDAC inhibitors (SL142 and SL325) showed a significant reduction in single cell colony forming ability with increased apoptosis in NSCLC cells [24]. Moreover, combination treatment enhanced Bax expression and caspase-3 activity. These results indicate that RA combined with HDAC inhibitors is a promising treatment for lung cancer [24].

Virmani and colleagues reported a role for methylation of the RARβ P2 promoter in gene silencing of RARβ2 and RARβ4 in lung cancer, especially in SCLC [21]. In contrast, Dutkowska and colleagues did not find a correlation between RARβ gene expression and methylation, but RARβ gene expression was significantly downregulated. Adenocarcinoma and large cell carcinoma patients showed lower expression of RARβ as compared to squamous cell carcinoma patients, supporting the use of the differential expression of the RARβ gene as a prognostic biomarker to differentiate lung cancer subtypes [77]. A meta-analysis of 18 relevant studies that included 1871 participants suggested an increase in the frequency of RARβ hyper-methylation in NSCLC patients compared to non-malignant lung tissues. The RARβ gene of NSCLC patients who smoke was hypermethylated (2.46 fold) compared to nonsmokers [78].

Furthermore, another meta-analysis of 16 studies including 1780 lung cancer patients also observed RARβ hyper-methylation [79]. Interestingly, RARβ gene hypermethylation was again significantly higher in lung adenocarcinoma patients as compared to squamous cell carcinoma patients. Overall, both meta-analyses support a correlation between RARβ hyper-methylation and high risk of NSCLC [79]. It has been shown that treatment with glucocorticoids plus RA (GC/RA) in combination with epigenetic drugs (azacytidine and SAHA; A/S) enhanced growth inhibitory effect of A/S in MYC amplified lung cancer cells which supports a pro-differentiation action of GC/RA [80]. These findings suggest an epigenetic regulatory effect of retinoids as a combination drug during lung cancer treatment. Overall findings indicate that deregulated histone H3 acetylation and RARβ methylation have roles in lung cancer development and could be targeted to restore retinoic acid sensitivity to lung cancer patients.

3.4. Chemoresistance associated with retinoids in lung cancer

Cancer recurrence after treatment implies the dormancy of some cancer cells during the treatment period that reemerges later. This rare subpopulation of chemoresistant cancer cells is defined as cancer stem cells (CSCs) or tumour-initiating cells (TICs) [2]. CSCs possess similar properties to normal stem cells (self-renewal and multi-lineage differentiation) [81]. CSCs can sustain a tumour, undergo extensive proliferation, and mediate resistance against chemo- and radiation therapy in many cancers [2]. CSCs were first identified in acute myeloid leukemia, but further studies revealed their existence in other cancer types including cancer of the lung [82]. Vitamin A derived RA controls the expression and activity of many genes involved in cellular development and differentiation. In the past two decades, vitamin A and its natural and synthetic derivatives (retinoids) have been shown to regulate many cellular activities including proliferation, differentiation, and apoptosis [29,83,84]. Tumour-initiating cells (TICs) have great therapeutic importance due to their tumour-inducing potential and chemotherapy resistance.

Moro et al. described TICs in cisplatin-resistant NSCLC that express CD133 and CXCR4 [3]. The authors reported heterogeneity in TICs through in-vitro cell fate tracing systems. TICs were of two types: (1) a highly quiescent pool and (2) a slowly dividing subpopulation. The highly quiescent pool was enriched for CD133+/CXCR4- cells while the slowly dividing subpopulation was enriched for CD133+/CXCR4+ cells. Further, pre-treatment with RA neutralized cisplatin resistance; mainly in the slowly dividing CD133+/CXCR4+ subpopulation [3]. Similar results were observed in patient-derived xenografts. Treatment with RA in combination with cisplatin reduced the fraction of TICs and tumour metastasis [3]. Cells exhibiting aberrant retinol signaling show a high level of resistance to chemo- and radiation therapy [85].

Several clinical studies support that resistance to retinoids in lung cancer is mediated by repression of RARβ [13,21,58,59,8690]. It was shown that RARβ2 overexpression inhibits lung cancer cell proliferation as restores retinoid sensitivity to retinoid-resistant human lung cancer cells [13]. However, RARβ suppression has a modulatory effect on the expression of other retinoid receptors involved in retinoid resistance [91]. Petty and colleagues also observed hypermethylation of the RARβ P2 promoter in RA resistant lung cancer cells leading to RARβ silencing [92]. RARβ1′ (RARβ isoform) is expressed in normal and RA-sensitive lung cells but not in RA resistant lung cancer cells or clinical lung cancer specimens. Transfection of RARβ1′ into RA resistant lung cancer cells with suppressed RARβ2 restored RA sensitivity and mediated growth inhibition [92]. These results confirmed that loss of RARβ and RARβ1′ play a role in retinoid resistance in lung carcinogenesis. Interestingly, overexpression of exogenous nuclear retinoid receptors could also restore growth inhibitory effects of RA in lung cancer cells resistant to RA [19].

García-Regalado et al. revealed a new molecular mechanism responsible for RA resistance in lung cancer. They suggested that Akt pathway activation induces resistance to RA in lung cancer by promoting a RARα-Akt interaction leading to activation of PI3k-Akt through transcription-independent mechanisms (Rac-GTPase activation) [51]. Overexpression of Akt or combination treatment of RA and a PI3k inhibitor suppressed the survival and invasion of lung cancer cells through overexpression of caspase-3 and RARβ2 [51]. Similarly, inhibitors of the ERK pathway restored the effects of RA including inhibition of proliferation and migration in lung cancer cells. This suggests that targeting PI3k-Akt or ERK in combination with RA treatment is a promising therapeutic strategy [42]. Zito et al. found that RA treatment inhibits the growth of tyrosine kinase inhibitor (TKI) resistant NSCLC cells [11].

SCLC also demonstrate resistant to RA due to loss of RARβ expression and retroviral transduction of the RARβ gene re-sensitized SCLC cells to RA [16]. G1 phase arrest with downregulation of L-myc and cyclin-dependent kinase 2 (CDK2) and enhanced expression of CDK inhibitor p27Kip1 has been observed after RA treatment in RARβ overexpressed SCLC cells. In conclusion, repression of RARα and RARβ has critical roles in retinoid intrinsic and acquired resistance in human lung cancer cells, which limits the use of retinoids as a therapeutic against lung cancer. Alternative clinical strategies are needed to overcome retinoid resistance such as the use of non-classical retinoids that have retinoid receptor-independent activity. The pathways involved in retinoids resistance in lung cancer are summarized in Fig. 4. The use of retinoids as a re-sensitizing agent for TKIs resistant lung cancer is summarized in Fig. 5.

Fig. 4. Retinoid resistant pathways in lung cancer.

Fig. 4.

Lung tumour inducing cells (TICs) or cancer stem cells (CSCs) positive for CD133+/CXCR4+ are sensitive to retinoids via inhibition of the PI3K-Akt pathway. However, CD133+/CXCR4 lung TICs/CSCs exhibit resistance to retinoid treatment. How CD133+/CXCR4- lung TICs/CSCs modulate the PI3K-Akt pathway during retinoid treatment and the potential to target this pathway is an active area of research.

Fig. 5. Re-sensitizing effect of retinoids as a combinatorial drug in TKIs resistant lung cancer.

Fig. 5.

Combinatorial treatment of retinoids with TKIs drugs in TKIs resistance lung cancer cells promotes the activation of a transcription factor named GATA-binding factor 6 (GATA6). Activation of GATA6 inhibits the activation of EGFR/Wnt signaling pathways and favors the association of retinoid X receptors (RXR), retinoic acid receptorβ (RARβ), and cellular retinoic acid-binding protein-2 (CRABP2). This complex inhibits the proliferation and promotes the differentiation of lung tumour cells via inhibiting activating protein-2 (AP-2), which result in re-sensitization of TKIs resistant lung cancer cells.

3.5. Clinical significance of retinoids in lung cancer

Lung cancer is a significant cause of mortality and morbidity worldwide, with 1.38 million deaths annually [1]. The majority of patients are diagnosed at clinical stages III b and IV resulting in a survival rate of less than 5%. Natural and synthetic retinoids are well-known chemotherapeutic and chemopreventive agents and play a pivotal role in cancer cell differentiation, proliferation, and apoptosis. Hong and Itri in 1994 reviewed epidemiological evidence that found an inverse relationship between β-carotene and serum vitamin A levels and cancer incidence [93]. Wolbach and Howe published the first preclinical study on vitamin A in cancer in 1925 [94]. Their study suggested that rodents with vitamin A deficiency undergo squamous metaplasia of lung epithelia that was overcome after repletion with vitamin A [94]. Similar types of metaplastic changes are observed in smokers. Experimental chemopreventive studies on retinoids in animal models revealed a mutation suppressive role for retinoids [95]. According to National Cancer Institute population-based trials, β-carotene and retinoids were promising anticancer agents in the early 1980s [96]. Studies also suggested that deficiency of vitamin A promoted preneoplastic changes in the bronchial epithelium which was associated with a higher risk of lung cancer development.

Initial clinical trials supported retinoids in the treatment of aerodigestive tract cancers. High dose vitamin A was given as adjuvant therapy to 307 patients with stage I NSCLC. Upon curative surgery, patients were randomly assigned to retinol palmitate (300,000 IU orally daily for 12 months) or the control group. Forty-six months of observation revealed a 37% rate of new primary tumour recurrence in the treated group compared to 48% in the control group. This study concluded that vitamin A could reduce the number of new primary tumours in tobacco-related stage I lung cancer patients [97]. A program on vitamin A to prevent cancer in asbestos-exposed workers (Wittenoom Gorge between 1943 and 1966) was initiated by Musk et al. [98]. Selected workers were supplemented with vitamin A (either synthetic β-carotene or retinol) from June 1990 to May 1995 with no smoking whereas the control groups were not supplemented with vitamin A. A total of 12 cases of lung cancer was observed in the vitamin A-supplemented group compared to 30 cases in the control group, suggesting vitamin A supplementation can protect against lung cancer malignancy in asbestos-exposed workers [98].

Lippman et al. conducted a National Cancer Institute Intergroup randomized, double-blind, phase III trial of placebo or isotretinoin (13-cis-RA; 30 mg/day) for three years in 1166 patients with stage I NSCLC [99]. The trial suggested that isotretinoin treatment did not improve the mortality of NSCLC patients. Isotretinoin was found to benefit only never smokers and harmed current smokers [99]. A double-blind, randomized clinical trial, the α-Tocopherol, β-Carotene Cancer Prevention Study (ATBC), was performed in which doses of β-carotene (20 mg/day), α-tocopherol (50 mg/day) or both were daily supplemented to 29,133 male smokers [100]. The β-Carotene and Retinol Efficacy Trial (CARET) enlisted 18,314 men and women at high risk of lung cancer for supplementation with placebo or combined doses of β-carotene (30 mg/day) and retinyl palmitate (25,000 IU/day) for approximately four years [101]. In addition, the double-blind, randomized trial of β-carotene (50 mg on alternate days) supplementation among 22,071 male physicians (40–84 years of age) for 12 years was conducted by the Physicians’ Health Study (PHS) [102]. These clinical trials revealed that retinoids were effective in reducing tumour induction and mortality in non-smokers and former smokers. However, retinoids increased the incidence of lung cancer in smokers, which was further supported in the EUROSCAN study [103].

Bushue et al. suggested that retinoids could cause bio-activation of cigarette carcinogens leading to an enhanced risk of lung cancer in smokers. A combination study on the effects of vitamin and mineral supplementation on lung cancer mortality among 29,584 healthy adults from Linxian, China suggested that combinations of vitamins and minerals failed to reduce lung cancer-related mortality in the selected population [104]. Another study based on cyclophosphamide and biotherapeutic compounds such as retinoids were conducted on 28 patients with advanced NSCLC and low-performance status with no previous record of surgery, chemotherapy or radiation [105]. Patients received a daily dose of (bromocriptine, cyclophosphamide, melatonin, retinoids, somatostatin, and vitamin D) every day for an indefinite period until unacceptable toxicity. This combined treatment was associated with improved survival and quality of life in advanced NSCLC patients with low-performance status [105].

RA and 13-cis-RA are the most active metabolites of vitamin A and both have effects on cell growth, differentiation, and apoptosis. Use of RA alone for treatment of twenty-eight metastatic patients exhibited minimal anti-lung cancer activity with an acceptable toxicity profile [43]. During a randomized phase II multi-center trial, combined treatment of interferon (IFN)-α and RA improved the survival rate of lung cancer patients who were treated with conventional cancer therapeutics [106]. However, the combined treatment of INF-α and RA in twenty-nine un-resurrected NSCLC patients showed only modest anticancer activity with moderate/severe hypertriglyceridemia and low toxicity in patients (Athanasiadis et al.1995). Combined treatment of RA and cisplatin or paclitaxel enhanced the median progression-free survival of advanced NSCLC patients [107]. RA shows less bioavailability after prolonged periods of administration. Thiruvengadam and colleagues treated 20 stage IIIB and IV NSCLC patients with an interrupted schedule of RA alone or in combination with cisplatin-VP 16 [108]. Ten patients out of 19 had a partial response whereas four patients had a minor response with neutropenia (most common acute toxicity). The average median survival rate was 25.5 weeks [108].

Further, the nano-encapsulation of RA using poly (lactic-co-glycolic) acid (PLGA) polymer has been successful in overcoming bioavailability related issues during treatment [109]. Zhang et al. formulated RA loaded PLGA nanoparticles with CD133 aptamers that have targeting efficiency for CD133+ lung cancer initiating cells. This PLGA nano-formulation showed higher inhibitory effects toward CD133+ lung cancer TICs as compared to non-targeted PLGA nanoparticles and RA [109]. Therefore, an interrupting schedule or nano-formulation of RA in combination with traditional chemotherapeutics may be an option to overcome bioavailability related issues in lung cancer therapy.

A phase II randomized clinical trial of 13-cis-RA with or without α-tocopherol was conducted by Kelly et al. on subjects with a high risk of lung cancer [110]. Bronchial biopsy histology showed only minor effects of 13-cis-RA and α-tocopherol did not alleviate 13-cis-RA toxicity [110]. Pillai and collogues conducted a phase II trial to evaluate the effects of 13-cis-RA, interferon-α, and paclitaxel combination therapy on 37 recurrent SCLC patients [111]. Three patients showed a partial response with 13-cis-RA, whereas five patients had stable disease. The median progression-free survival and median overall survival was 2 and 6.2 months, respectively.

Bexarotene (targretin) is approved for the treatment of cutaneous T-cell lymphoma and is an agonist of the retinoid-X-receptor and inducer of CYP3A4-mediated metabolism. Bexarotene induces cell differentiation and apoptosis leading to the prevention of drug resistance in cancer cells including solid tumours. Interestingly, bexarotene showed good tolerability and disease stabilization in NSCLC patients in phase I trials [112,113]. Bexarotene also demonstrated excellent tolerability in lung cancer patients when used in combination with cisplatin or vinorelbine with an improved patient survival rate. Some common side effects observed during treatment were anemia, asthenia, hyperlipidemia, leukopenia, nausea, and vomiting [114]. Another, phase II trial on combined bexarotene and gemcitabine or carboplatin found that stage IIIB or stage IV NSCLC patients benefited from the combination with higher tolerance to treatment and improved progression time and overall survival [115]. A pharmacokinetic study of bexarotene with established chemotherapeutic drugs (such as paclitaxel, carboplatin, cisplatin, and vinorelbine) suggested drug-drug interactions during bexarotene co-administration. Co-administration of atorvastatin or fenofibrate with bexarotene suggested that co-administration of atorvastatin controls bexarotene-mediated hypertriglyceridemia more effectively than fenofibrate in NSCLC patients [111,112]. The SPIRIT I and SPIRIT II (randomized phase III trials of bexarotene) failed to improve the survival rate of chemotherapy-naive advanced or metastatic NSCLC patients [47,116]. However, a subpopulation of bexarotene treated NSCLC patients showed high-grade hypertriglyceridemia and an improved survival rate. Both SPRIT I and SPRIT II suggests that triglyceride induction could be a biomarker for bexarotene treatment of lung cancer patients. Overall, these findings suggest that bexarotene could be safely added to platinum-based chemotherapeutics for lung cancer treatment and warrant further clinical evaluations. Clinical use of retinoids as combinatorial drug therapeutics is summarized in Table 2.

Table 2.

Clinical use of retinoids as combinatorial drug therapeutics.

Retinoids Combination Consequence Stage of study Reference
All-trans retinoic acid Somatostatin, melatonin, vitamin D, bromocriptine, and cyclophosphamide Overall survival 12.9 months Stage IIIB or stage IV [105]
C-phycocyanin Synergistic In-vitro and in-vivo [117]
Arsenic trioxide Synergistic In-vitro [118]
Paclitaxel and cisplatin Progression-free survival 8.9 months Stage IIIB, stage IV [107]
c-myc antisense DNA Synergistic In-vitro [119]
IFN-α Overall survival eight months Phase II trial [120]
Bexarotene, capecitabine, and docetaxel Survival benefit Stage IIIB and IV [121]
PLGA nanoparticles with CD133 aptamers Enhanced inhibitory effect In-vitro [109]
Bexarotene Gemcitabine, Carboplatin Increased overall survival Stage IIIB, stage IV [115]
Carboplatin, Paclitaxel May benefit a segment of first-line NSCLC patients Phase III trial [116]
Paclitaxel, vinorelbine Enhanced inhibitory effect In-vitro and in-vivo [122]
Cisplatin, vinorelbine Overall survival 8.7 months Phase I/II trial [114]
Cisplatin, vinorelbine No survival benefits in advanced NSCLC Phase III trial [47]
13-cis-retinoic acid IFN-α, paclitaxel Overall survival 6.2 months Phase II study [111]
αTocopherol Reduced bronchial epithelial cell proliferation Previous smokers [123]
Interleukin-2 Improves progression-free and overall survival Phase II study [48]
Fenretinide Curcumin Synergistic effect In-vitro, in-vivo [124]
Etoposide, paclitaxel, cisplatin Enhanced inhibitory effect In-vitro [125]
Retinyl Palmitate 5 FU, Cisplatin, IFN-β, Overall survival 9.1 months Phase II study [126]

4. Conclusion and future perspective

Lung cancer is one of the most aggressive and life-threatening forms of cancer worldwide. Improvement in the treatment of lung cancer is an urgent human health need. Retinoids have various anticancer properties including anti-proliferative, anti-invasive, anti-migratory, anti-angiogenic, antimetastatic, and pro-apoptotic effects through modulating aberrant oncogenic signaling pathways. Retinoids also regulate the survival and differentiation of tumour-initiating cells or cancer stem cells. Studies highlight the potential of retinoids in the epigenetic regulation of oncogenes and tumour suppressor genes in lung cancer. However, despite this preclinical promise, retinoids have failed to show significant clinical utility as single agents for the treatment of naive and drug-resistant lung cancer. One possible reason for single agent failure could be poor solubility and low bioavailability of retinoids, which may be overcome through retinoid nano-formulation. Retinoids can be used as a combination therapeutics for the resenstitization of TKIs resistant lung cancer. The retinoid resistance in lung cancer can be overcomed by cotreatment of drugs which target the cancer stem cells. The best way to harness these powerful anticancer agents to treat lung cancer is likely combination strategies with conventional and targeted therapies to improve efficacy and limit toxicity.

Acknowledgements

The authors thank (a) MHRD and National Institute of Technology, Rourkela, Odisha, India for providing laboratory and fellowship to Surya Kant Tripathi; (b) Department of Science and Technology, Science and Engineering Research Board (DST, SERB) (Grant Number: ECR/2016/000792) for providing fellowship to Munmun Panda; (c) Department of Science and Technology, Odisha, India (Grant number: 1201) for providing fellowship to Kamal Pandey and (d) National Institutes of Health grants R01-CA211875 and R03CA223709 and a Reach Grant from the Alex’s Lemonade Stand Foundation to MJS, and (e) Konkuk University, Seoul, South Korea for providing support through KU Brain Pool Program to KRRR.

Footnotes

Declaration of Competing Interest

The authors declare no conflict of interest regarding the publication of this review article.

References

  • [1].Miller KD, Siegel RL, Lin CC, Mariotto AB, Kramer JL, Rowland JH, Stein KD, Alteri R, Jemal A, Cancer treatment and survivorship statistics, 2016, CA Cancer J. Clin 66 (4) (2016) 271–289. [DOI] [PubMed] [Google Scholar]
  • [2].Reya T, Morrison SJ, Clarke MF, Weissman IL, Stem cells, cancer, and cancer stem cells, Nature 414 (6859) (2001) 105. [DOI] [PubMed] [Google Scholar]
  • [3].Moro M, Bertolini G, Pastorino U, Roz L, Sozzi G, Combination treatment with all-trans retinoic acid prevents cisplatin-induced enrichment of CD133+ tumor-initiating cells and reveals heterogeneity of cancer stem cell compartment in lung cancer, J. Thorac. Oncol 10 (7) (2015) 1027–1036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].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 129 (4) (2007) 723–733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].McKenna NJ, EMBO Retinoids 2011: mechanisms, biology and pathology of signaling by retinoic acid and retinoic acid receptors, Nucl. Recept. Signal 10 (1) (2012) nrs. 10003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Bushue N, Wan Y-JY, Retinoid pathway and cancer therapeutics, Adv. Drug Deliv. Rev 62 (13) (2010) 1285–1298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].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 46 (11) (2007) 1160–1165. [DOI] [PubMed] [Google Scholar]
  • [8].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 (2009) 1940–6207 CAPR-09-0221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].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 111 (7) (2001) 1203–1208. [DOI] [PubMed] [Google Scholar]
  • [10].Liu B, Lee K-W, Li H, Ma L, Lin GL, Chandraratna RA, Cohen P, Combination therapy of insulin-like growth factor binding protein-3 and retinoid X receptor ligands synergize on prostate cancer cell apoptosis in vitro and in vivo, Clin. Cancer Res 11 (13) (2005) 4851–4856. [DOI] [PubMed] [Google Scholar]
  • [11].Zito G, Naselli F, Saieva L, Raimondo S, Calabrese G, Guzzardo C, Forte S, Rolfo C, Parenti R, Alessandro R, Retinoic Acid affects Lung Adenocarcinoma growth by inducing differentiation via GATA6 activation and EGFR and Wnt inhibition, Sci. Rep 7 (1) (2017) 4770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Muñiz-Hernández S, Hernández-Pedro N, Macedo-Pérez OE, Arrieta O, Alterations in retinoic acid receptors in non-small cell lung cancer and their clinical implications, J. Cancer Ther 6 (08) (2015) 648. [Google Scholar]
  • [13].Houle B, Rochette-Egly C, Bradley W, Tumor-suppressive effect of the retinoic acid receptor beta in human epidermoid lung cancer cells, Proc. Natl. Acad. Sci 90 (3) (1993) 985–989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Kim Y, Jin D, Lee BB, Cho EY, Han J, Shim YM, Kim D-H, RARβ2 hypermethylation is associated with poor recurrence-free survival in never-smokers with adenocarcinoma of the lung, Clin. Epigenetics 7 (1) (2015) 32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Kim JS, Lee H, Kim H, Shim YM, Han J, Park J, Kim D-H, Promoter methylation of retinoic acid receptor Beta 2 and the development of second primary lung cancers in non-Small-Cell lung Cancer, J. Clin. Oncol 22 (17) (2004) 3443–3450. [DOI] [PubMed] [Google Scholar]
  • [16].Geradts J, Chen J-Y, Russell EK, Yankaskas JR, Nieves L, Minna JD, Human lung cancer cell lines exhibit resistance to retinoic acid treatment, Metabolism 1 (1993) 7. [PubMed] [Google Scholar]
  • [17].Barrascout E, Medioni J, Scotte F, Ayllon J, Mejean A, Cuenod C, Tartour E, Elaidi R, Oudard S, Angiogenesis inhibition: review of the activity of sorafenib, sunitinib and bevacizumab, Bull. Cancer 97 (2010) 29–43. [DOI] [PubMed] [Google Scholar]
  • [18].Zuo L, Yang X, Lu M, Hu R, Zhu H, Zhang S, Zhou Q, Chen F, Gui S, Wang Y, All-Trans Retinoic Acid Inhibits Human Colorectal Cancer Cells RKO Migration via Downregulating Myosin Light Chain Kinase Expression through MAPK Signaling Pathway, Nutr. Cancer 68 (7) (2016) 1225–1233. [DOI] [PubMed] [Google Scholar]
  • [19].Wan H, Hong WK, Lotan R, Increased retinoic acid responsiveness in lung carcinoma cells that are nonresponsive despite the presence of endogenous retinoic acid receptor (RAR) β by expression of exogenous retinoid receptors retinoid X receptor α, RARα, and RARγ, Cancer Res. 61 (2) (2001) 556–564. [PubMed] [Google Scholar]
  • [20].Momparler RL, Bouffard DY, Momparler LF, Dionne J, Bélanger K, Ayoub J, Pilot phase I-II study on 5-aza-2′-deoxycytidine (Decitabine) in patients with metastatic lung cancer, Anticancer Drugs 8 (4) (1997) 358–368. [DOI] [PubMed] [Google Scholar]
  • [21].Virmani AK, Rathi A, Zöchbauer-Müller S, Sacchi N, Fukuyama Y, Bryant D, Maitra A, Heda S, Fong KM, Thunnissen F, Promoter methylation and silencing of the retinoic acid receptor-β gene in lung carcinomas, J. Natl. Cancer Inst 92 (16) (2000) 1303–1307. [DOI] [PubMed] [Google Scholar]
  • [22].Greve G, Schiffmann I, Lübbert M, Epigenetic priming of non-small cell lung cancer cell lines to the antiproliferative and differentiating effects of all-trans retinoic acid, J. Cancer Res. Clin. Oncol 141 (12) (2015) 2171–2180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Kurie JM, Lotan R, Lee JJ, Lee JS, Morice RC, Liu DD, Xu X-C, Khuri FR, Ro JY, Hittelman WN, Treatment of former smokers with 9-cis-retinoic acid reverses loss of retinoic acid receptor-β expression in the bronchial epithelium: results from a randomized placebo-controlled trial, J. Natl. Cancer Inst 95 (3) (2003) 206–214. [DOI] [PubMed] [Google Scholar]
  • [24].Han S, Fukazawa T, Yamatsuji T, Matsuoka J, Miyachi H, Maeda Y, Durbin M, Naomoto Y, Anti-tumor effect in human lung cancer by a combination treatment of novel histone deacetylase inhibitors: SL142 or SL325 and retinoic acids, PLoS One 5 (11) (2010) e13834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Mernitz H, Smith DE, Zhu AX, Wang X-D, 9-cis-Retinoic acid inhibition of lung carcinogenesis in the A/J mouse model is accompanied by increased expression of RAR-β but no change in cyclooxygenase-2, Cancer Lett. 244 (1) (2006) 101–108. [DOI] [PubMed] [Google Scholar]
  • [26].Bama ES, Grace VB, Sundaram V, Jesubatham PD, Synergistic effect of cotreatment with all-trans retinoic acid and 9-cis retinoic acid on human lung cancer cell line at molecular level, 3 Biotech 9 (4) (2019) 159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Li Y, Dawson MI, Agadir A, Lee MO, Jong L, Hobbs PD, Zhang Xk., Regulation of RARβ expression by RAR-and RXR-selective retinoids in human lung cancer cell lines: effect on growth inhibition and apoptosis induction, Int. J. Cancer 75 (1) (1998) 88–95. [DOI] [PubMed] [Google Scholar]
  • [28].Choi EJ, Whang YM, Kim SJ, Kim HJ, Kim YH, Combinational treatment with retinoic acid derivatives in non-small cell lung carcinoma in vitro, J. Korean Med. Sci 22 (Suppl) (2007) S52–S60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Mark M, Ghyselinck NB, Chambon P, Function of retinoic acid receptors during embryonic development, Nucl. Recept. Signal 7 (1) (2009) nrs. 07002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Sakoe Y, Sakoe K, Kirito K, Ozawa K, Komatsu N, FOX03A as a key molecule for all-trans retinoic acid-induced granulocytic differentiation and apoptosis in acute promyelocytic leukemia, Blood 115 (18) (2010) 3787–3795. [DOI] [PubMed] [Google Scholar]
  • [31].Shirakami Y, Lee S-A, Clugston RD, Blaner WS, Hepatic metabolism of retinoids and disease associations, Biochimica et Biophysica Acta (BBA)-Mol. Cell Biol. Lipids 1821 (1) (2012) 124–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Steeg PS, Targeting metastasis, Nat. Rev. Cancer 16 (4) (2016) 201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Tahergorabi Z, Khazaei M, A review on angiogenesis and its assays, Iran. J. Basic Med. Sci 15 (6) (2012) 1110. [PMC free article] [PubMed] [Google Scholar]
  • [34].Cao Y, Liu Q, Therapeutic targets of multiple angiogenic factors for the treatment of cancer and metastasis, Adv. Cancer Res 97 (2007) 203–224. [DOI] [PubMed] [Google Scholar]
  • [35].Minamiya Y, Nakagawa T, Saito H, Matsuzaki I, Taguchi K, Ito M, Ogawa J.-i., Increased expression of myosin light chain kinase mRNA is related to metastasis in non-small cell lung cancer, Tumor Biol. 26 (3) (2005) 153–157. [DOI] [PubMed] [Google Scholar]
  • [36].Borowicz SJ, Tauler J, Rizzo A, Garcia JG, A potential role for myosin light chain kinase in lung adenocarcinoma tumorigenesis, Am. Soc. Clin. Oncol (2013). [Google Scholar]
  • [37].Cui W.-j, Liu Y, Zhou X.-l., Wang F.-z., Zhang X.-d., Ye L.-h., Myosin light chain kinase is responsible for high proliferative ability of breast cancer cells via anti-apoptosis involving p38 pathway, Acta Pharmacol. Sin 31 (6) (2010) 725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Ramya D, Siddikuzzaman VB, Grace, Effect of all-trans retinoic acid (ATRA) on syndecan-1 expression and its chemoprotective effect in benzo (α) pyrene-induced lung cancer mice model, Immunopharmacol. Immunotoxicol 34 (6) (2012) 1020–1027. [DOI] [PubMed] [Google Scholar]
  • [39].Liang C, Guo S, Yang L, Effects of all-trans retinoic acid on VEGF and HIF-1α expression in glioma cells under normoxia and hypoxia and its anti-angiogenic effect in an intracerebral glioma model, Mol. Med. Rep 10 (5) (2014) 2713–2719. [DOI] [PubMed] [Google Scholar]
  • [40].Li N, Lu Y, Li D, Zheng X, Lian J, Li S, Cui H, Zhang L, Sang L, Wang Y, All-trans retinoic acid suppresses the angiopoietin-Tie2 pathway and inhibits angiogenesis and metastasis in esophageal squamous cell carcinoma, PLoS One 12 (4) (2017) e0174555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Lu T.-y., Li W.-c., Chen R.-y., Fan Q.-x., Wang L.-x., Wang R.-l, Lu S.-x., Hui M, Inhibition effects of all trans-retinoic acid on the growth and angiogenesis of esophageal squamous cell carcinoma in nude mice, Chinese Med. J 124 (17) (2011) 2723–2727. [PubMed] [Google Scholar]
  • [42].Barceinas Q, Sara R, García-Regalado A, Aréchaga-Ocampo E, Villegas-Sepúlveda N, All-trans retinoic acid induces proliferation, survival, and migration in A549 lung cancer cells by activating the ERK signaling pathway through a transcription-independent mechanism, Biomed Res. Int 2015. (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Treat J, Friedland D, Luginbuhl W, Meehan L, Gorman G, Miller W, Bavaria J, Kaiser L, Phase II trial of all-trans retinoic acid in metastatic non-small cell lung cancer, Cancer Invest. 14 (5) (1996) 415–420. [DOI] [PubMed] [Google Scholar]
  • [44].Grace VB, Inhibition of metastatic lung cancer in C57BL/6 mice by liposome encapsulated all trans retinoic acid (ATRA), Int. Immunopharmacol 14 (4) (2012) 570–579. [DOI] [PubMed] [Google Scholar]
  • [45].Yen W, Prudente R, Corpuz M, Negro-Vilar A, Lamph W, A selective retinoid X receptor agonist bexarotene (LGD1069, targretin) inhibits angiogenesis and metastasis in solid tumours, Br. J. Cancer 94 (5) (2006) 654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Fu J, Ding Y, Huang D, Li H, Chen X, The retinoid X receptor-selective ligand, LGD1069, inhibits tumor-induced angiogenesis via suppression of VEGF in human non-small cell lung cancer, Cancer Lett. 248 (1) (2007) 153–163. [DOI] [PubMed] [Google Scholar]
  • [47].Ramlau R, Zatloukal P, Jassem J, Schwarzenberger P, Orlov SV, Gottfried M, Pereira JR, Temperley G, Negro-Vilar R, Rahal S, Randomized phase III trial comparing bexarotene (L1069–49)/cisplatin/vinorelbine with cisplatin/vinorelbine in chemotherapy-naive patients with advanced or metastatic non–small-cell lung cancer: SPIRIT I, J. Clin. Oncol 26 (11) (2008) 1886–1892. [DOI] [PubMed] [Google Scholar]
  • [48].Recchia F, Saggio G, Nuzzo A, Biondi E, Di Blasio A, Cesta A, Candeloro G, Alesse E, Rea S, Multicenter phase 2 study of interleukin-2 and 13-cis retinoic acid as maintenance therapy in advanced non-small-cell lung cancer, J. Immunother 29 (1) (2006) 87–94. [DOI] [PubMed] [Google Scholar]
  • [49].Berry DC, Levi L, Noy N, Holo-retinol-binding protein and its receptor STRA6 drive oncogenic transformation, Cancer Res. (2014) canres. 1052.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Doldo E, Costanza G, Ferlosio A, Pompeo E, Agostinelli S, Bellezza G, Mazzaglia D, Giunta A, Sidoni A, Orlandi A, High expression of cellular retinol binding protein-1 in lung adenocarcinoma is associated with poor prognosis, Genes Cancer 6 (11–12) (2015) 490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].García-Regalado A, Vargas M, García-Carrancá A, Aréchaga-Ocampo E, González-De la Rosa CH, Activation of Akt pathway by transcription-independent mechanisms of retinoic acid promotes survival and invasion in lung cancer cells, Mol. Cancer 12 (1) (2013) 44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Favorskaya I, Kainov Y, Chemeris G, Komelkov A, Zborovskaya I, Tchevkina E, Expression and clinical significance of CRABP1 and CRABP2 in non-small cell lung cancer, Tumor Biol. 35 (10) (2014) 10295–10300. [DOI] [PubMed] [Google Scholar]
  • [53].Persaud SD, Lin Y-W, Wu C-Y, Kagechika H, Wei L-N, Cellular retinoic acid binding protein I mediates rapid non-canonical activation of ERK1/2 by all-trans retinoic acid, Cell. Signal 25 (1) (2013) 19–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Hawthorn L, Stein L, Varma R, Wiseman S, Loree T, Tan D, TIMP1 and SERPIN-A overexpression and TFF3 and CRABP1 underexpression as biomarkers for papillary thyroid carcinoma, Head Neck 26 (12) (2004) 1069–1083. [DOI] [PubMed] [Google Scholar]
  • [55].Miyake T, Ueda Y, Matsuzaki S, Miyatake T, Yoshino K, Fujita M, Nomura T, Enomoto T, Kimura T, CRABPl-reduced expression is associated with poorer prognosis in serous and clear cell ovarian adenocarcinoma, J. Cancer Res. Clin. Oncol 137 (4) (2011) 715–722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Li C, Wan Y-JY, Differentiation and antiproliferation effects of retinoic acid receptor β in hepatoma cells, Cancer Lett. 124 (2) (1998) 205–211. [DOI] [PubMed] [Google Scholar]
  • [57].Widschwendter M, Berger J, Daxenbichler G, Müller-Holzner E, Widschwendter A, Mayr A, Marth C, Zeimet AG, Loss of retinoic acid receptor β expression in breast cancer and morphologically normal adjacent tissue but not in the normal breast tissue distant from the cancer, Cancer Res. 57 (19) (1997) 4158–4161. [PubMed] [Google Scholar]
  • [58].Xu X-C, 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. 54 (13) (1994) 3580–3587. [PubMed] [Google Scholar]
  • [59].Xu X-C, Sozzi G, Lee JS, Lee JJ, Pastorino U, Pilotti S, Kurie JM, Hong WK, Lotan R, Suppression of retinoic acid receptor β in non-small-cell lung cancer in vivo: implications for lung cancer development, J. Natl. Cancer Inst 89 (9) (1997) 624–629. [DOI] [PubMed] [Google Scholar]
  • [60].Delmotte M-H, Tahayato A, Formstecher P, Lefebvre P, 157 Serine, A retinoic acid receptor α residue phosphorylated by protein kinase C in vitro, is involved in RXR· RARα heterodimerization and transcriptional activity, J. Biol. Chem 274 (53) (1999) 38225–38231. [DOI] [PubMed] [Google Scholar]
  • [61].Gianni M, Bauer A, Garattini E, Chambon P, Rochette-Egly C, Phosphorylation by p38MAPK and recruitment of SUG-1 are required for RA-induced RARγ degradation and transactivation, EMBO J. 21 (14) (2002) 3760–3769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Rochette-Egly C, Oulad-Abdelghani M, Staub A, Pfister V, Scheuer I, Chambon P, Gaub M-P, Phosphorylation of the retinoic acid receptor-alpha by protein kinase A, Mol. Endocrinol 9 (7) (1995) 860–871. [DOI] [PubMed] [Google Scholar]
  • [63].Rochette-Egly C, Adam S, Rossignol M, Egly J-M, Chambon P, Stimulation of RARα activation function AF-1 through binding to the general transcription factor TFIIH and phosphorylation by CDK7, Cell 90 (1) (1997) 97–107. [DOI] [PubMed] [Google Scholar]
  • [64].Srinivas H, Xia D, Moore NL, Uray IP, Kim H, Ma L, Weigel NL, Brown PH, Kurie JM, Akt phosphorylates and suppresses the transactivation of retinoic acid receptor α, Biochem. J 395 (3) (2006) 653–662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Karamouzis MV, Papavassiliou AG, Retinoid receptor cross-talk in respiratory epithelium cancer chemoprevention, Trends Mol. Med 11 (1) (2005) 10–16. [DOI] [PubMed] [Google Scholar]
  • [66].Averbuch SD, Lung cancer prevention: retinoids and the epidermal growth factor receptor—a phoenix rising? Clin. Cancer Res 8 (1) (2002) 1–3. [PubMed] [Google Scholar]
  • [67].Cheng Y, Lotan R, Molecular cloning and characterization of a novel retinoic acid-inducible gene that encodes a putative G protein-coupled receptor, J. Biol. Chem 273 (52) (1998) 35008–35015. [DOI] [PubMed] [Google Scholar]
  • [68].Tao Q, Fujimoto J, Men T, Ye X, Deng J, Lacroix L, Clifford JL, Mao L, Van Pelt CS, Lee JJ, Identification of the retinoic acid-inducible Gprc5a as a new lung tumor suppressor gene, JNCI: J. Nat. Cancer Inst 99 (22) (2007) 1668–1682. [DOI] [PubMed] [Google Scholar]
  • [69].Zhong S, Yin H, Liao Y, Yao F, Li Q, Zhang J, Jiao H, Zhao Y, Xu D, Liu S, Lung tumor suppressor GPRC5A binds EGFR and restrains its effector signaling, Cancer Res. 75 (9) (2015) 1801–1814. [DOI] [PubMed] [Google Scholar]
  • [70].Delpu Y, Cordelier P, Cho W, Torrisani J, DNA methylation and cancer diagnosis, Int. J. Mol. Sci 14 (7) (2013) 15029–15058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Ma X, Wang Y-W, Zhang MQ, Gazdar AF, DNA methylation data analysis and its application to cancer research, Epigenomics 5 (3) (2013) 301–316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Ghavifekr Fakhr M, Farshdousti Hagh M, Shanehbandi D, Baradaran B, DNA methylation pattern as important epigenetic criterion in cancer, Genet. Res. Int 2013. (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Fleischhacker M, Dietrich D, Liebenberg V, Field JK, Schmidt B, The role of DNA methylation as biomarkers in the clinical management of lung cancer, Expert Rev. Respir. Med 7 (4) (2013) 363–383. [DOI] [PubMed] [Google Scholar]
  • [74].Alvarez S, Germain P, Alvarez R, Rodríguez-Barrios F, Gronemeyer H, De Lera AR, Structure, function and modulation of retinoic acid receptor beta, a tumor suppressor, Int. J. Biochem. Cell Biol 39 (7–8) (2007) 1406–1415. [DOI] [PubMed] [Google Scholar]
  • [75].Mongan NP, Gudas LJ, Diverse actions of retinoid receptors in cancer prevention and treatment, Differentiation 75 (9) (2007) 853–870. [DOI] [PubMed] [Google Scholar]
  • [76].Swift CB, Hays JL, Petty WJ, Distinct functions of retinoic acid receptor beta isoforms: implications for targeted therapy, Endocrine, Metabolic & Immune Disorders-Drug Targets (formerly Current Drug Targets-Immune, Endocrine & Metab. Dis 8 (1) (2008) 47–50. [DOI] [PubMed] [Google Scholar]
  • [77].Dutkowska A, Antczak A, Pastuszak-Lewandoska D, Migdalska-Sęk M, Czarnecka K, Górski P, Kordiak J, Nawrot E, Brzeziańska-Lasota E, RARβ promoter methylation as an epigenetic mechanism of gene silencing in non-small cell lung Cancer, Adv. Clin. Sci (2015) 29–38 Springer. [DOI] [PubMed] [Google Scholar]
  • [78].Song X, Shi K, Zhou S-J, Yu D-P, Liu Z, Han Y, Clinicopathological significance and a potential drugtarget of RARβ in non-small-cell lung carcinoma: a meta-analysis and a systematic review, Drug design, Devel. Therapy 10 (2016) 1345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Li Y, Lu D.-g., Ma Y.-m., Liu H, Association between Retinoic acid receptor-β hypermethylation and NSCLC risk: a meta-analysis and literature review, Oncotarget 8 (4) (2017) 5814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Romero O, Verdura S, Torres-Diz M, Gomez A, Moran S, Condom E. Esteller M, Villanueva A, Sanchez-Cespedes M, Sensitization of retinoids and corticoids to epigenetic drugs in MYC-activated lung cancers by antitumor reprogramming, Oncogene 36 (9) (2017) 1287. [DOI] [PubMed] [Google Scholar]
  • [81].Clarke MF, Hass AT, Cancer stem cells, Reviews in Cell Biol. Mol. Med (2006). [Google Scholar]
  • [82].Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, Minden M, Paterson B, Caligiuri MA, Dick JE, A cell initiating human acute myeloid leukaemia after transplantation into SCID mice, Nature 367 (6464) (1994) 645. [DOI] [PubMed] [Google Scholar]
  • [83].Gudas LJ, Retinoids and vertebrate development, J. Biol. Chem 269 (22) (1994) 15399–15402. [PubMed] [Google Scholar]
  • [84].Dollé P, Developmental expression of retinoic acid receptors (RARs), Nucl. Recept. Signal 7 (1) (2009) nrs. 07006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Sharma RB, Wang Q, Khillan JS, Amplification of tumor inducing putative cancer stem cells (CSCs) by vitamin A/retinol from mammary tumors, Biochem. Biophys. Res. Commun 436 (4) (2013) 625–631. [DOI] [PubMed] [Google Scholar]
  • [86].Freemantle SJ, Spinella MJ, Dmitrovsky E, Retinoids in cancer therapy and chemoprevention: promise meets resistance, Oncogene 22 (47) (2003) 7305. [DOI] [PubMed] [Google Scholar]
  • [87].Zhang X.-k., Liu Y, Lee M-O, Pfahl M, A specific defect in the retinoic acid response associated with human lung cancer cell lines, Cancer Res. 54 (21) (1994) 5663–5669. [PubMed] [Google Scholar]
  • [88].Lotan R, Xu X-C, Lippman SM, Ro JY, Lee JS, Lee JJ, Hong WK, Suppression of retinoic acid receptor–β in premalignant oral lesions and its upregulation by isotretinoin, N. Engl. J. Med 332 (21) (1995) 1405–1411. [DOI] [PubMed] [Google Scholar]
  • [89].Vuillemenot BR, Pulling LC, Palmisano WA, Hutt JA, Belinsky SA, Carcinogen exposure differentially modulates RAR-β promoter hypermethylation, an early and frequent event in mouse lung carcinogenesis, Carcinogenesis 25 (4) (2004) 623–629. [DOI] [PubMed] [Google Scholar]
  • [90].Sun S-Y, Wan H, Yue P, Hong WK, Lotan R, Evidence that retinoic acid receptor β induction by retinoids is important for tumor cell growth inhibition, J. Biol. Chem 275 (22) (2000) 17149–17153. [DOI] [PubMed] [Google Scholar]
  • [91].Brabender J, Metzger R, Salonga D, Danenberg KD, Danenberg PV, Hölscher AH, Schneider PM, Comprehensive expression analysis of retinoic acid receptors and retinoid X receptors in non-small cell lung cancer: implications for tumor development and prognosis, Carcinogenesis 26 (3) (2005) 525–530. [DOI] [PubMed] [Google Scholar]
  • [92].Petty WJ, Li N, Biddle A, Bounds R, Nitkin C, Ma Y, Dragnev KH, Freemantle SJ, Dmitrovsky E, A novel retinoic acid receptor β isoform and retinoid resistance in lung carcinogenesis, J. Natl. Cancer Inst 97 (22) (2005) 1645–1651. [DOI] [PubMed] [Google Scholar]
  • [93].Hong W, Chemistry And medicine, Retinoids and Human Cancer, the Retinoids: Biology, 2nd ed., (1994), pp. 597–630. [Google Scholar]
  • [94].Wolbach SB, Howe PR, Tissue changes following deprivation of fat-soluble A vitamin, J. Exp. Med 42 (6) (1925) 753–777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Moon R, Metha R, Rao K, Sporn MB, Roberts AB, Goodman DS (Eds.), Retinoids and Cancer in Experimental Animals in The Retinoids: Biology, Chemistry, and Medicine, Raven Press, New York, 1994, pp. 573–595. [Google Scholar]
  • [96].Blot WJ, Li J-Y, Taylor PR, Guo W, Dawsey S, Wang G-Q, Yang CS, Zheng S-F, Gail M, Li G-Y, Nutrition intervention trials in Linxian, China: supplementation with specific vitamin/mineral combinations, cancer incidence, and disease-specific mortality in the general population, JNCI: J. Nat. Cancer Inst 85 (18) (1993) 1483–1491. [DOI] [PubMed] [Google Scholar]
  • [97].Pastorino U, Infante M, Maioli M, Chiesa G, Buyse M, Firket P, Rosmentz N, Clerici M, Soresi E, Valente M, Adjuvant treatment of stage I lung cancer with high-dose vitamin A, J. Clin. Oncol 11 (7) (1993) 1216–1222. [DOI] [PubMed] [Google Scholar]
  • [98].Musk AW, de Klerk NH, Ambrosini GL, Eccles JL, Hansen J, Olsen NJ, Watts VL, Lund HG, Pang S, Beilby J, Vitamin A and cancer prevention I: observations in workers previously exposed to asbestos at Wittenoom, Western Australia, Int. J. Cancer 75 (3) (1998) 355–361. [DOI] [PubMed] [Google Scholar]
  • [99].Lippman SM, Lee JJ, Karp DD, Vokes EE, Benner SE, Goodman GE, Khuri FR, Marks R, Winn RJ, Fry W, Randomized phase III intergroup trial of isotretinoin to prevent second primary tumors in stage I non-small-cell lung cancer, J. Natl. Cancer Inst 93 (8) (2001) 605–618. [DOI] [PubMed] [Google Scholar]
  • [100].A.-T.B.C.C.P.S. Group, The effect of vitamin E and beta carotene on the incidence of lung cancer and other cancers in male smokers, N. Engl. J. Med 330 (15) (1994) 1029–1035. [DOI] [PubMed] [Google Scholar]
  • [101].Omenn GS, Goodman GE, Thornquist MD, Balmes J, Cullen MR, Glass A, Keogh JP, Meyskens FL Jr., Valanis B, Williams JH Jr., Effects of a combination of beta carotene and vitamin A on lung cancer and cardiovascular disease, N. Engl. J. Med 334 (18) (1996) 1150–1155. [DOI] [PubMed] [Google Scholar]
  • [102].Hennekens CH, Buring JE, Manson JE, Stampfer M, Rosner B, Cook NR, Belanger C, LaMotte F, Gaziano JM, Ridker PM, Lack of effect of long-term supplementation with beta carotene on the incidence of malignant neoplasms and cardiovascular disease, N. Engl. J. Med 334 (18) (1996) 1145–1149. [DOI] [PubMed] [Google Scholar]
  • [103].De NV, Van NZ, Pastorino U, The EUROSCAN study: a progress report, Am. J. Otolaryngol 14 (1) (1993) 62–66. [DOI] [PubMed] [Google Scholar]
  • [104].Kamangar F, Qiao Y-L, Yu B, Sun X-D, Abnet CC, Fan J-H, Mark SD, Zhao P, Dawsey SM, Taylor PR, Lung cancer chemoprevention: a randomized, double-blind trial in Linxian, China, Cancer Epidemiol. Prev. Biomark 15 (8) (2006) 1562–1564. [DOI] [PubMed] [Google Scholar]
  • [105].Norsa A, Martino V, Retinoids Somatostatin, Vitamin D. Melatonin, Bromocriptine, and cyclophosphamide in advanced non-Small-Cell lung Cancer patients with low performance status, Cancer Biother. Radiopharm 21 (1) (2006) 68–73. [DOI] [PubMed] [Google Scholar]
  • [106].Ruotsalainen T, Halme M, Isokangas O-P, Pyrhönen S, Mäntylä M, Pekonen M, Sarna S, Joensuu H, Mattson K, Interferon-α and 13-cis-retinoic acid as maintenance therapy after high-dose combination chemotherapy with growth factor support for small cell lung cancer-a feasibility study, Anticancer Drugs 11 (2) (2000) 101–108. [DOI] [PubMed] [Google Scholar]
  • [107].Arrieta O, González-De la Rosa CH, Aréchaga-Ocampo E, Villanueva-Rodríguez G, Cerón-Lizárraga TL, Martínez-Barrera L, Vázquez-Manríquez ME, Ríos-Trejo MÁ, Álvarez-Avitia MÁ, Hernández-Pedro N, Randomized phase II trial of All-trans-retinoic acid with chemotherapy based on paclitaxel and cisplatin as first-line treatment in patients with advanced non-small-cell lung cancer, J. Clin. Oncol 28 (21) (2010) 3463–3471. [DOI] [PubMed] [Google Scholar]
  • [108].Thiruvengadam R, Atiba JO, Azawi SH, A phase II trial of a differentiating agent (tRA) with cisplatin-VP 16 chemotherapy in advanced non-small cell lung cancer, Invest. New Drugs 14 (4) (1996) 395–401. [DOI] [PubMed] [Google Scholar]
  • [109].Zhang Y, Zhao J, Sun J, Huang L, Li Q, Targeting lung cancer initiating cells by all-trans retinoic acid-loaded lipid-PLGA nanoparticles with CD133 aptamers, Exp. Ther. Med 16 (6) (2018) 4639–4649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Kelly K, Kittelson J, Franklin WA, Kennedy TC, Klein CE, Keith RL, Dempsey EC, Lewis M, Jackson MK, Hirsch FR, A randomized phase II chemoprevention trial of 13-CIS retinoic acid with or without α tocopherol or observation in subjects at high risk for lung cancer, Cancer Prev. Res (2009) 1940–6207 CAPR-08-0136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Pillai RN, Aisner J, Dahlberg SE, Rogers JS, DiPaola RS, Aisner S, Ramalingam SS, Schiller JH, Interferon alpha plus 13-cis-retinoic acid modulation of BCL-2 plus paclitaxel for recurrent small-cell lung cancer (SCLC): an Eastern Cooperative Oncology Group study (E6501), Cancer Chemother. Pharmacol 74 (1) (2014) 177–183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [112].Wakelee HA, Takimoto CH, Lopez-Anaya A, Chu Q, Middleton G, Dunlop D, Ramlau R, Leighl N, Rowinsky EK, Hao D, The effect of bexarotene on atorvastatin pharmacokinetics: results from a phase I trial of bexarotene plus chemotherapy in patients with advanced non-small cell lung cancer, Cancer Chemother. Pharmacol 69 (2) (2012) 563–571. [DOI] [PubMed] [Google Scholar]
  • [113].Miller VA, Benedetti FM, Rigas JR, Verret AL, Pfister DG, Straus D, Kris MG, Crisp M, Heyman R, Loewen GR, Initial clinical trial of a selective retinoid X receptor ligand, LGD1069, J. Clin. Oncol 15 (2) (1997) 790–795. [DOI] [PubMed] [Google Scholar]
  • [114].Khuri FR, Rigas JR, Figlin RA, Gralla RJ, Shin DM, Munden R, Fox N, Huyghe MR, Kean Y, Reich SD, Multi-institutional phase I/II trial of oral bexarotene in combination with cisplatin and vinorelbine in previously untreated patients with advanced non-small-cell lung cancer, J. Clin. Oncol 19 (10) (2001) 2626–2637. [DOI] [PubMed] [Google Scholar]
  • [115].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 23 (24) (2005) 5774–5778. [DOI] [PubMed] [Google Scholar]
  • [116].Blumenschein GR Jr., Khuri FR, von Pawel J, Gatzemeier U, Miller WH Jr., Jotte RM, Le Treut J, Sun S-L, Zhang JK, Dziewanowska ZE, Phase III trial comparing carboplatin, paclitaxel, and bexarotene with carboplatin and paclitaxel in chemotherapy-naive patients with advanced or metastatic non–small-cell lung cancer: SPIRIT II, J. Clin. Oncol 26 (11) (2008) 1879–1885. [DOI] [PubMed] [Google Scholar]
  • [117].Li B, Gao M-H, Chu X-M, Teng L, Lv C-Y, Yang P, Yin Q-F, The synergistic antitumor effects of all-trans retinoic acid and C-phycocyanin on the lung cancer A549 cells in vitro and in vivo, Eur. J. Pharmacol 749 (2015) 107–114. [DOI] [PubMed] [Google Scholar]
  • [118].Lin L-M, Li B-X, Xiao J-B, Lin D-H, Yang B-F, Synergistic effect of all-trans-retinoic acid and arsenic trioxide on growth inhibition and apoptosis in human hepatoma, breast cancer, and lung cancer cells in vitro, World J. Gastroenterol.: WJG 11 (36) (2005) 5633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Akie K, Dosaka-Akita H, Murakami A, Kawakami Y, A combination treatment of c-myc antisense DNA with all-trans-retinoic acid inhibits cell proliferation by downregulating c-myc expression in small cell lung cancer, Antisense Nucleic Acid Drug Dev. 10 (4) (2000) 243–249. [DOI] [PubMed] [Google Scholar]
  • [120].Athanasiadis I, Kies MS, Miller M, Ganzenko N, Joob A, Marymont M, Rademaker A, Gradishar WJ, Phase II study of all-trans-retinoic acid and alpha-interferon in patients with advanced non-small cell lung cancer, Clin. Cancer Res 1 (9) (1995) 973–979. [PubMed] [Google Scholar]
  • [121].Nandan R, Promising results achieved with a combination of chemotherapy and two retinoids in patients with advanced non-small-cell lung cancer, Lung Cancer 51 (3) (2006) 387–388. [DOI] [PubMed] [Google Scholar]
  • [122].Hermann TW, Yen W-C, Tooker P, Fan B, Roegner K, Negro-Vilar A, Lamph WW, Bissonnette RP, The retinoid X receptor agonist bexarotene (Targretin) synergistically enhances the growth inhibitory activity of cytotoxic drugs in non-small cell lung cancer cells, Lung Cancer 50 (1) (2005) 9–18. [DOI] [PubMed] [Google Scholar]
  • [123].Hittelman WN, Liu DD, Kurie JM, Lotan R, Lee JS, Khuri F, Ibarguen H, Morice RC, Walsh G, Roth JA, Proliferative changes in the bronchial epithelium of former smokers treated with retinoids, J. Natl. Cancer Inst 99 (21) (2007) 1603–1612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [124].Chen H, Chen L, Wang L, Zhou X, Chan JY-W, Li J, Cui G, Lee SM-Y, Synergistic effect of fenretinide and curcumin for treatment of non-small cell lung cancer, Cancer Biol. Ther 17 (10) (2016) 1022–1029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [125].Kalemkerian GP, Ou X, Activity of fenretinide plus chemotherapeutic agents in small-cell lung cancer cell lines, Cancer Chemother. Pharmacol 43 (2) (1999) 145–150. [DOI] [PubMed] [Google Scholar]
  • [126].Recchia F, Sica G, De SF, Rea S, Frati L, Combined chemotherapy and differentiation therapy in the treatment of advanced non-small-cell lung cancer, Anticancer Res. 17 (5B) (1997) 3761–3765. [PubMed] [Google Scholar]

RESOURCES