Next Article in Journal
Pediatric Cancers: Insights and Novel Therapeutic Approaches
Next Article in Special Issue
Management of Acute Promyelocytic Leukemia at Extremes of Age
Previous Article in Journal
Distal Humeral Replacement in Patients with Primary Bone Sarcoma: The Functional Outcome and Return to Sports
Previous Article in Special Issue
The Coagulopathy of Acute Promyelocytic Leukemia: An Updated Review of Pathophysiology, Risk Stratification, and Clinical Management
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Promise of Retinoids in the Treatment of Cancer: Neither Burnt Out Nor Fading Away

by
Yuya Nagai
1,* and
Alexander J. Ambinder
2,*
1
Department of Hematology, Kobe City Medical Center General Hospital, Kobe 650-0047, Hyogo, Japan
2
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
*
Authors to whom correspondence should be addressed.
Cancers 2023, 15(14), 3535; https://doi.org/10.3390/cancers15143535
Submission received: 27 May 2023 / Revised: 29 June 2023 / Accepted: 3 July 2023 / Published: 8 July 2023
(This article belongs to the Special Issue Acute Promyelocytic Leukemia (APML))

Abstract

:

Simple Summary

In the realm of cancer treatment and prevention, the application of retinoids has led to unparalleled successes and humbling disappointments; all-trans retinoic acid (ATRA) has transformed APL (acute promyelocytic leukemia) into an eminently curable disease but has demonstrated little efficacy in acute myeloid leukemia (AML) more broadly. As a cancer prevention strategy, vitamin A supplementation was ineffective and appeared to increase lung cancer incidence, underscoring the complex relationship between retinoic acid signaling and carcinogenesis. This review aims to provide a survey of our current understanding of retinoic acid homeostasis and signaling, to reflect on prior successes and failures to deploy retinoids in the treatment and prevention of cancer, and to explore the unfulfilled potential of retinoids in cancer. New biological insights relating to the regulation of retinoid homeostasis and the discovery of biomarkers of retinoid sensitivity in subsets of non-APL AML have revived enthusiasm for the therapeutic potential of synthetic retinoids, and thus warrant a reacquaintance with retinoid biology.

Abstract

Since the introduction of all-trans retinoic acid (ATRA), acute promyelocytic leukemia (APL) has become a highly curable malignancy, especially in combination with arsenic trioxide (ATO). ATRA’s success has deepened our understanding of the role of the RARα pathway in normal hematopoiesis and leukemogenesis, and it has influenced a generation of cancer drug development. Retinoids have also demonstrated some efficacy in a handful of other disease entities, including as a maintenance therapy for neuroblastoma and in the treatment of cutaneous T-cell lymphomas; nevertheless, the promise of retinoids as a differentiating therapy in acute myeloid leukemia (AML) more broadly, and as a cancer preventative, have largely gone unfulfilled. Recent research into the mechanisms of ATRA resistance and the biomarkers of RARα pathway dysregulation in AML have reinvigorated efforts to successfully deploy retinoid therapy in a broader subset of myeloid malignancies. Recent studies have demonstrated that the bone marrow environment is highly protected from exogenous ATRA via local homeostasis controlled by stromal cells expressing CYP26, a key enzyme responsible for ATRA inactivation. Synthetic CYP26-resistant retinoids such as tamibarotene bypass this stromal protection and have shown superior anti-leukemic effects. Furthermore, recent super-enhancer (SE) analysis has identified a novel AML subgroup characterized by high expression of RARα through strong SE levels in the gene locus and increased sensitivity to tamibarotene. Combined with a hypomethylating agent, synthetic retinoids have shown synergistic anti-leukemic effects in non-APL AML preclinical models and are now being studied in phase II and III clinical trials.

1. Introduction

The remarkable success of all-trans retinoic acid (ATRA) therapy in APL has come to epitomize the importance of retinoic acid signaling in carcinogenesis and the anti-cancer potential of retinoids. Nonetheless, efforts to replicate the success of ATRA in APL with other non-APL leukemias and malignancies have led to disappointing results [1,2,3]. Recent improvements in the understanding of retinoic acid biology, the development of synthetic retinoids, and an improved capability to identify subsets of non-APL AML with increased sensitivity to retinoids have revived hopes for the successful use of retinoids beyond APL [4,5,6,7,8]. In this article, we review the history of the introduction of ATRA into clinical use, summarize recent developments in the understanding of the role of retinoic acid signaling in non-APL AML, and discuss promising strategies to successfully target retinoic acid pathways in the treatment of non-APL AML.

2. The History of Retinol, Health, and Cancer

Vitamin A is a fat-soluble vitamin essential in orchestrating embryogenesis, cellular differentiation, and proliferation [9]. Much of what is known about the biological role of retinol has been learned from the effects of its absence. One of the most visible adverse effects of vitamin A deficiency (VAD) is xerophthalmia or night blindness. Records from ancient Greece, Egypt, and China have all documented animal liver as an effective remedy for night blindness, likely owing to its high concentration of vitamin A stores [10]. Nutritional studies in the early 1900s first identified a fat-soluble dietary factor, Factor A, present in butter and eggs and necessary for rats to continue growing. Today, VAD remains a significant cause of morbidity and mortality worldwide. In population studies, a deficiency of vitamin A has been associated with xerophthalmia, stunted growth, anemia, dermatologic manifestations including poor wound healing, impaired immune function, and increased maternal and childhood mortality. In populations with a high prevalence of VAD, vitamin A supplementation has been demonstrated to reduce early childhood mortality. VAD has therefore been targeted for elimination in an extensive public health campaign by the World Health Organization and the United Nations International Children’s Emergency Fund (UNICEF).
The potential role of vitamin A in oncogenesis was first suggested by animal studies in the mid-1920s, though the claim was met with skepticism, and data from different groups were conflicting [11]. Animal studies continued, with one notable study in 1959 showing a higher incidence of epithelial tumor production in mice treated with a carcinogenic-treated cheek pouch and a diet deficient in vitamin A, compared to control mice who received the carcinogenic treatment to the cheek pouch but otherwise had a vitamin-A-sufficient diet [12]. Then, in the 1970s, there were a series of both retrospective and prospective epidemiologic studies that found an inverse relationship between serum vitamin A concentration and cancer incidence, particularly with respect to lung cancer [13]. These studies sparked tremendous enthusiasm to understand the biology of retinoids and to harness them for the prevention and treatment of cancer. In the case of acute promyelocytic leukemia, these efforts have led to remarkable success and the development of retinoids as a model for targeted cancer therapy; however, the outcomes of studies using retinoids as cancer prophylaxis and in non-APL acute myeloid leukemias (AML) have been sobering, underscoring the complex role of retinoid metabolism in cancer biology.

3. Retinoids

Vitamin A metabolism is complicated by the diversity of forms that it takes in the body. Animals cannot synthesize vitamin A and thus must ingest retinol, the alcohol form of vitamin A, or provitamin A carotenoids such as beta-carotene. Enzymes in the liver can esterify them for storage, or they may be bound by retinoid-binding proteins for transport to target organs, where they are converted into retinal (retinaldehyde) by retinyl ester hydrolase and then into retinoic acid, the most active form of vitamin A, by retinaldehyde dehydrogenase [14]. ATRA is the most active form of endogenous vitamin A; however, numerous other isoforms, including 13-cis retinoic acid (Isotretinoin) and 9-cis retinoic acid, have varying degrees of affinity for the various retinoic acid receptors (RARs) [10]. In addition, some forms can bind RXR, in which case they are referred to as rexinoids. CYP26 then catabolizes retinoids into inactive metabolites, namely, 4-hydroxy retinoic acid and 4-oxo retinoic acid.

4. Retinoid Signaling

ATRA controls gene expression via the transcriptional activation of nuclear retinoic acid receptors (RARs) [15]. RARs form a ligand-regulated nuclear receptor superfamily responsible for regulating transcription. RARs consist of three isotypes, including RARa, RARb, and RARg, which are expressed by varying degrees across tissues and developmental stages. RARa is highly expressed in the developing and adult nervous system, lung, spleen, and hematopoietic cells in the bone marrow [1]. RARb is expressed in the lung, esophagus, and skin, whereas RARg is predominantly expressed in skin, skeletal muscle, and adipose tissue. RARs act as a nuclear transcription factor with the retinoid X receptor (RXR) cofactor. RARs form heterodimers with RXR and bind to specific motifs called retinoic acid responsive elements (RAREs) within the promoter of target genes. In the absence of a ligand, RAR/RXR interacts with the corepressor proteins, the silencing mediator of retinoid and thyroid hormone receptor (SMRT) and the nuclear receptor corepressor (NCoR). These corepressors, in turn, recruit histone deacetylase (HDAC), which triggers the chromatin to assume a transcriptionally inactive conformation. ATRA binds to the RAR and causes a conformational change of the ligand-binding-domain (LBD), leading to the receptor’s dissociation from corepressors and the promotion of the interaction with co-activators, including histone acetyltransferase (HAT). Thus, the binding of ATRA promotes the transcriptional activation of target genes and differentiation [16]. In the case of RARa, many of its target genes, including C/EBPepsilon, PU.1, and HOX proteins, are involved in myelopoiesis. RARa signaling is therefore involved in orchestrating hematopoietic stem cell quiescence, self-renewal, and differentiation into myeloid hematopoietic cells [1,17,18,19].

5. The Unparalleled Success of Retinoids in the Treatment of Acute Promyelocytic Leukemia

APL is a distinct subtype of AML characterized by the proliferation of abnormal promyelocytes and typically a severe coagulopathy accompanied by disseminated intravascular coagulation (DIC) or hyperfibrinolysis. Historically, chemotherapy alone exacerbates bleeding tendency, leading to a high incidence of early hemorrhagic death, and achieved cure in only 35% to 45% of patients with APL [20]. The discovery in the 1980s that retinoic acid induces the terminal differentiation of APL cells in vitro [21] and a clinical response with the differentiation of APL cells and improved coagulopathy in patients [22,23,24] promoted clinical trials of ATRA as a targeted therapy for the treatment of APL. The first documented use of retinoids in the treatment of APL came in 1983 and 1984 when two case reports described the successful single agent use of 13-cis-retinoic acid, an ATRA prodrug that is itself a less-potent RAR agonist [23,25,26]. In 1985, a subsequent case report was published describing the case of a five-year-old girl with APL at Shanghai Children’s Hospital successfully treated with ATRA. The patient went into complete remission within three weeks of administration, leading to further clinical drug development [26]. Later case studies demonstrated that ATRA was effective in salvaging patients previously treated with 13-cis-retinoin, leading to its preferential use in larger subsequent studies [27]. In 1988, a Shanghai group published a study in which single-agent ATRA induced CR in a remarkable 23 of 24 patients [28]. The efficacy was confirmed by another clinical study with a larger cohort, in which CR was achieved in 72% of patients [29].
Nevertheless, remissions with single-agent ATRA are typically transient [30]. To achieve more-durable remissions, subsequent trials combined ATRA and chemotherapy in the induction phase and followed with ATRA maintenance, resulting in an improved 5-year disease-free survival of 74% [31] (Table 1). More recently, dual-targeted therapy consisting of ATRA and ATO achieved superior event-free survival (EFS) and overall survival (OS) compared to ATRA plus chemotherapy for patients with low-to-intermediate risk of APL in the phase 3 trial (APL0406) [32,33] (Table 1). The updated analysis showed excellent results, with participants having an EFS at 72 months of 96.6% in the ATRA-ATO group compared to 77.4% in the ATRA-chemotherapy group [34]. However, the optimal regimen for patients with high-risk cases remains a debated issue. Idarubicin and gemtuzumab ozogamicin are most commonly used with ATRA-ATO [35,36,37], whereas the APL15 trial showed that ATRA-ATO with hydroxyurea used to control leukocytosis has the same efficacy as ATRA-ATO plus chemotherapy, even in high-risk patients [38]. An advantage of this chemotherapy-free regimen is that it is relatively well-tolerated by patients unfit for chemotherapy due to cardiac dysfunction or severe comorbidities. Oral formulations of ATO have also been developed and appear to have similar tolerability and efficacy, promising a more accessible and convenient therapeutic regimen in the future [39]. Hence, the remarkable evolution of APL therapy over the decades has drastically changed APL from one of the most fatal to one of the most curable subtypes of AML.

6. Disruption of Retinoid Signaling in APL

The success of ATRA in the treatment of APL may be due in part to the singular role of dysregulated retinoic acid signaling in its pathogenesis. An epidemiological study implying a single rate-limiting event in APL based on the constant incidence over life [49] and leukemia development in PML-RARa transgenic mice [50,51] suggests that the PML-RARa translocation is necessary and sufficient for triggering leukemogenesis. APL cells have remarkably stable genomes with co-occurring mutations in only a handful of genes involved in cell signaling and proliferation, including FLT3, WT1, NRAS, and KRAS, and noncoding regions within the intron of WT1 [52,53,54].
A large number of reviews have described the molecular pathogenesis of APL. In brief, the PML-RARa oncoprotein forms homodimers via the PML coiled-coil domain, interacts with RXR [55], and competes with RARα for binding to RARE within the target genes. Unlike wild-type RAR/RXR, the PML-RARa fusion receptor has a much higher affinity for corepressors and a lower sensitivity to physiological concentrations of ATRA, resulting in the constitutional repression of target genes. Consequently, PML-RARa/RXR acts in a dominant negative fashion, resulting in maturation arrest and the proliferation of myeloid progenitors and promyelocytes. PML-RARa also recruits the DNA-methylating enzymes DNMT1 and DNMT3a, leading to transcriptional repression through the hypermethylation of the promoter regions of target genes [56]. Furthermore, a genome-wide analysis of the PML-RARa/RXR binding site using Chip-seq in an APL cell line (NB4) and primary APL cells revealed that the PML-RARa /RXR dimer interacts with and represses a broader range of promoter regions than its RARa /RXR counterpart [57], likely contributing to the oncogenic action of the fusion protein [55]. The same group and others have also recently demonstrated that PML/RARa exerts transactivating functions through directly binding to target genes such as growth factor independent 1 transcriptional repressor (GFI-1), which is mediated by the recruitment of P300 and HDAC1 and the formation of super-enhancers [58,59]. Additionally, PML-RARa interferes with the formation of PML nuclear bodies, which blunts p53 signaling and leads to the enhanced self-renewal of leukemic cells [60]. The complex transcriptional dysregulation of extended target genes and disorganized PML nuclear bodies contributes to the differentiation block, aberrant self-renewal, and impaired apoptosis in APL cells.

7. Mechanisms of Action of ATRA

Mechanistically, ATRA has dual effects on the PML-RARa fusion protein: first, ATRA induces the transactivation of target genes. PML-RARa is not responsive to the physiological level (10−10–10−9 M) of ATRA, but pharmacological doses (10−6–10−7 M) of ATRA induce a conformational change in PML-RARa (followed by dissociation with corepressors including HDACs [61] and DNMTs [62]), and the recruitment of co-activators such as HAT, which allows for the reactivation of silenced target genes, thereby resuming differentiation. Transcriptomic analyses have revealed that ATRA treatment rapidly up-regulates many transcription factors related to hematopoiesis, including the C/EBP family in NB4 cells [63]. In addition, genome-wide analyses of epigenetic change in NB4 treated with ATRA also clarified the increase in H3 acetylation at the PML-RARα target genes [55]. These reports support the fact that that ATRA converts PML-RARa from a constitutive repressor into a transcriptional activator and reactivates the target genes. The other mechanism is the ATRA-induced degradation of PML-RARa through the ubiquitin/proteasome system [64]. The ATRA-induced degradation of its receptor is considered a general feature of nuclear receptor signaling, which provides a negative feedback loop of RA on its receptors [65]. Mutations in the ligand-binding domain of RARa are known to confer resistance to ATRA by reducing the binding affinity [66,67,68]. These mutations are found in a third of patients who relapsed after ATRA treatment and are associated with a reduced response to post-relapse salvage therapy [69]. Furthermore, the presence of an internal tandem duplication (ITD) in the FLT3 gene, an adverse risk factor for worse outcomes in patients treated without arsenic trioxide (ATO), blunts ATRA-induced PML-RARa degradation [70]. Although the two modes of action are generally coupled, some experimental data show that the degradation of PML-RARa (rather than the differentiation of leukemic cells) is primarily responsible for clinical response to ATRA treatment. In a PML-RARa transgenic mice model, low-dose retinoic acid triggers differentiation but does not fully degrade PML-RARa, and thus fails to eradicate leukemia-initiating cells [71]. Similarly, the rare promyelocytic leukemia zinc finger (PLZF)-RARa-driven APL cells terminally differentiate but are never cleared on RA therapy [71]. Interestingly, etretinate, a synthetic retinoid, triggers target gene activation and efficient APL cell differentiation at levels comparable to ATRA but does not efficiently degrade PML-RARa. Etretinate fails to abolish the capacity of PML-RARa-transformed cells to initiate leukemia, indicating that sustained PML-RARa protein is necessary for the self-renewal of leukemic cells [72]. These results suggest the induction of differentiation is insufficient to eradicate APL cells and that the degradation of PML-RARa is also necessary. ATO also triggers PML-RARa degradation [73], providing a rationale for the dramatic synergistic effect observed with the combination of ATRA and ATO [74].

8. Exploring the Activity of Retinoids in Non-APL AML

The remarkable success of ATRA therapy in APL, together with the preclinical data showing that ATRA induces transcriptional activation in most AML cell lines in tissue culture, encouraged the undertaking of numerous clinical trials investigating the benefit of adding ATRA to standard chemotherapy in non-APL AML. Contrary to expectations, the results from those trials were disparate from one another, and in sum, did not demonstrate clear benefit. In an initial phase 3 trial that included 242 elderly patients with AML, adding ATRA to induction and consolidation chemotherapy was associated with a statistically significant improvement in remission rate and OS [75]. However, a sub-analysis for predictive biomarkers revealed that the benefit of ATRA was restricted to the cases with an NPM1 mutation without FLT-ITD [76]. In contrast, the MRC AML 12 trial randomized 1075 younger patients with non-APL AML and high-risk MDS to induction therapy with or without ATRA and found that the addition of ATRA did not affect remission rate or OS. An absence of benefit was observed for the entire cohort and specific subgroups, including AML with mutated NPM1, FLT3-ITD, or CEBPα [2,3] (Table 1).

9. Retinoids and Rexinoids for the Treatment of Non-Leukemic Malignancies

The use of retinoids as anti-cancer therapy was also explored in other, non-leukemic malignancies. Neuroblastoma, a solid tumor of the peripheral nervous system, is the most common cancer diagnosed in infancy. High-risk neuroblastoma is a particularly aggressive form of the disease and is associated with poor prognosis. In vitro, ATRA induces differentiation in neuroblastoma cells, leading to the development of retinoids as a targeted, differentiating agent in the treatment of neuroblastoma [77]. Moreover, 13-cis-retinoic acid (isotretinoin) is an endogenous retinoic acid isoform that is less abundant and less potent than ATRA, though it is converted to ATRA in vivo. Nonetheless, 13-cis-retinoic acid has a favorable pharmacologic safety profile that led to its development as a targeted maintenance therapy in children with neuroblastoma [78]. Clinical studies of retinoids in neuroblastoma culminated in the pivotal Children’s Cancer Group study (CCG-3891), which found that isotretinoin significantly improved the event-free survival rate of children with high-risk neuroblastoma [42] (Table 1). Isotretinoin is now part of the standard-of-care maintenance therapy for high-risk neuroblastoma. There remains interest in building upon this success with alternative retinoids or combining them with other differentiating agents such as herbimycin A [77,79].
Bexarotene is a synthetic rexinoid that is approved by the Food and Drug Administration (FDA) for the treatment of cutaneous T-cell lymphoma, a subtype of peripheral T-cell lymphoma, in the relapsed/refractory setting [41] (Table 1). In the context of CTCL, bexarotene inhibits proliferation, induces apoptosis, and may even trigger the differentiation of malignant T cells [80], though its mechanism is less clearly defined than that of other retinoid-responsive malignancies.

10. Retinoids for the Prevention of Non-Hematologic Cancer

Several epidemiologic studies have suggested an inverse association between dietary vitamin A and cancer incidence. A study in 2000 found that a high nutritional intake of vitamin A and carotenoids was associated with a reduced risk of lung cancer in non-smoking women [81]. Another study published in 1999 using data from the Health Study found that women with a higher intake of vitamin A and several other vitamins had a lower incidence of post-menopausal breast cancer [82]. These observational studies inspired prospective, interventional trials that aimed to use retinoids as chemoprevention to reduce the incidence of a variety of cancers. Topical retinoids have been used with some success to prevent actinic keratosis—a precursor to non-melanoma skin cancer—but their systemic use as chemoprevention is not well studied. Studies of retinoids as a chemoprophylaxis against the development of head and neck cancer, breast cancer, and hepatocellular carcinoma have yielded mixed results [83]. The largest study, known as the CARET (Carotene and Retinol Efficacy Trial) study, found that a combination of retinyl palmitate (a retinoid) and beta-carotene actually increased the risk of lung cancer in smokers and asbestos-exposed workers [84]. Overall, these disappointing results have reduced enthusiasm for the use of retinoids as a chemoprophylaxis, though there are still some ongoing efforts to translate these theoretical benefits (NCT00201279).

11. ATRA Metabolism in Vivo

The reasons for retinoids’ failure to improve outcomes outside of APL and neuroblastoma are complex but are illuminated by a deepened understanding of retinoid metabolism and homeostasis. Continuous oral ATRA monotherapy induces only temporary remissions, with durations often being less than six months [30]. Researchers have speculated that these remissions are short-lived because of the rapid decline in steady-state ATRA concentrations that occur within days of the initiation of the drug [85,86]. A liposomal formulation of all-trans retinoic acid (L-ATRA) with better pharmacokinetic properties and higher maintained ATRA concentrations [87,88] led to sustained molecular complete remissions (CRs) with a median duration of 6.4 years in 10 out of 26 responders with low-risk APL [89]. Consistent with this result, experimental data demonstrated that only high concentrations of ATRA can fully degrade PML-RARa protein and eradicate leukemia-initiating cells in a PML-RARα transgenic mice model [71]. Collectively, this clinical and experimental evidence underscores that the levels of ATRA reaching leukemic cells are critical for complete eradication.
The balanced synthesis of ATRA by ALDH1A enzymes and metabolism by CYP26 enzymes, as well as other regulating enzymes, maintain ATRA homeostasis [90,91,92]. ATRA induces its catabolism by stimulating the up-regulation (up to a thousand-fold) of CYP26A1 mRNA expression in human hepatocytes, suggesting that its auto-induced catabolism is responsible for the accelerated clearance of ATRA and progressive decline of plasma ATRA concentrations in patients on continuous oral ATRA treatment [93,94]. Intermittent ATRA dosing schedules may be advantageous over continuous dosing schedules because they minimize the induction of auto catabolism [95]. Notably, systemic RA concentrations do not reflect RA homeostasis in specific tissues due to an intra-organ expression of CYP26 enzymes, which regulate RA levels locally [96]. The local production of the CYP26 enzyme within the bone marrow microenvironment may provide a protective niche for leukemic cells and contribute to leukemic cells acquiring resistance to ATRA.
Recent reports have implicated that the feedback mechanisms that control RA signaling are essential in regulating hematopoiesis within the bone marrow microenvironment. Although hematopoietic stem cells (HSC) rapidly differentiate in liquid culture, the co-culture system has shown that the human bone marrow-derived stromal cells are capable of metabolizing retinoids through CYP26A1 and CYP26B1 expression, thereby preserving the primitive phenotype and self-renewal capacity of HSCs through the maintenance of a low retinoid in an environment [97]. Similarly, pharmacological doses of ATRA exhibit activity against non-APL AML cells in liquid culture; however, stromal cells, which also up-regulate their CYP26 expression upon exposure to ATRA, blocked the pro-differentiation effect of ATRA on APL and AML cell lines as well as CD34+CD38ALDHint leukemic cells from primary samples, which were rescued by CYP26 inhibition [98]. These experimental data suggest that HSCs and leukemic cells are protected from RA signaling by CYP26 activity in the bone marrow niche, which provides a plausible explanation as to why clinical studies failed to demonstrate any benefit from the addition of ATRA to treatment regimens in non-APL AML [2,99]. Tamibarotene and IRX195183 are CYP26-resistant synthetic retinoids that primarily function as RARα agonists. Unlike ATRA, these synthetic retinoids maintain their pro-differentiating activity on APL cells in co-culture with bone marrow stromal cells [6]. Thus, CYP26-resistant synthetic retinoids have the potential to bypass the stromal-mediated protection and fully exert an anti-leukemic effect via the RARα-mediated differentiation of leukemic cells.

12. Synthetic Retinoids

Building on the success of ATRA, many synthetic retinoids have been developed and evaluated for enhanced anti-tumor therapeutic potential [100,101] (Figure 1). Synthesized at the University of Tokyo in 1984, tamibarotene (formerly known as Am80) is a selective retinoid with a high affinity for RARa, resulting in greater potency as a differentiating agent [100] but a minimal affinity for RARγ [4]. It bears two methyl groups at C-4 that render it resistant to CYP26-mediated catabolism [6,102]. Thus, tamibarotene has a favorable pharmacokinetic profile with more sustained plasma drug levels than ATRA [46,103]. Furthermore, this agent is associated with a lower incidence and severity of cutaneous reactions, presumably related to its lower affinity for RARγ, mainly expressed in the dermal epithelium [104]. In Japan, tamibarotene has been studied in several clinical studies as a potentially superior alternative to ATRA. A prospective multicenter phase 2 study evaluated tamibarotene as a single agent in 24 patients who had relapsed after ATRA-containing treatment. Although those patients generally have difficulty in achieving a second CR with a second course of ATRA alone [105], tamibarotene achieved CR in 14 patients (68%) in this study, and four of them, after subsequent consolidation chemotherapy, have remained in CR for more than four years without stem cell transplantation [46,106] (Table 1). These results imply that tamibarotene overcomes the mechanisms that confer resistance to ATRA in vivo, including CYP26-mediated protection in bone marrow niche. Based on these results, tamibarotene was approved in Japan for the treatment of relapsed APL. A subsequent phase 3 study compared tamibarotene with ATRA as a maintenance therapy for newly diagnosed APL patients [47] (Table 1). An updated long-term analysis showed a statistically significant difference in relapse-free survival (RFS) of seven years between the ATRA group (84%) and the tamibarotene group (93%). When the analysis was restricted to high-risk patients, RFS at seven years was 62% in the ATRA group and 89% in the tamibarotene group, suggesting the potential role of tamibarotene as a maintenance therapy, especially in high-risk cases. Although the utility of tamibarotene is uncertain in the ATO era, the optimal management of patients relapsing after ATRA and ATO has been an unresolved issue. In this context, a multicenter phase 2 clinical study was conducted to evaluate the efficacy of tamibarotene in the relapsed/refractory setting. Tamibarotene as a single agent achieved CR/CRi in 64% of the cases, including molecular CR in 21%, although subsequent relapse occurred within six months in most cases. This result suggests that tamibarotene has significant activity in substantial patients with relapsed APL after ATRA and ATO treatment [107], and a more significant role in the cases that acquire PML mutations associated with the resistance to ATO [108,109].
IRX195183 (formerly known as NRX195183) is another promising CYP26-resistant synthetic retinoid. This agent is an exclusive RARα agonist, whereas ATRA is a pan-RAR agonist with relatively similar binding affinities for all three RAR isotypes [6]. The interplay between RARs and their ligands in normal hematopoiesis is complex and depends upon the differential stage of hematopoietic cells and physiological levels of retinoids [19]. The RARγ pathway appears to promote self-renewal of hematopoietic stem cells, while the RARα pathway promotes the myeloid differentiation of committed myeloid progenitors [110]. Consistent with this, a study using selective RAR agonists revealed the differentiation of AML cells appears to be mediated by mainly RARα signaling and only minimally by RARβ and RARγ [6]. Intriguingly, a study revealed that IRX195183 efficiently induced myeloid differentiation and the loss of the self-renewal of AML1/ETO-transformed progenitors, whereas ATRA potentiated the clonogenicity of them, probably through RARγ activation [5]. Transactivation studies revealed that a 0.24 nM level of ATRA was needed to transactivate RARγ, whereas 19.3 nM was needed for RARα, indicating that a substantially lower level of ATRA is sufficient for the activation of RARγ [111]. As described above, exogenous ATRA up-regulates CYP26 activity in bone marrow stromal cells, rendering the bone marrow niche a retinoid-deficient environment [6]. As a result, ATRA levels might fall below the threshold necessary for the transactivation of RARα but sufficient for RARγ transactivation, leading to the enhanced self-renewal of leukemic cells [5]. In this context, RARa-selective and CYP26-resistant retinoids may be preferable differentiation agents for AML cells. A phase 1 study of IRX195183 in patients with MDS and AML was recently conducted and showed that IRX195183 is safe and potentially effective in a subset of cases [112]. The clinical benefit of IRX195183 as a single agent or in combination with other drugs needs to be investigated in further studies. These synthetic retinoids could expand the application of retinoid-based therapy in both APL and non-APL AML.

13. Renewed Promise for Retinoids in Non-APL AML

More recent studies have tried to identify specific genetic subgroups, which may have greater retinoid sensitivity through functional RAR signaling [110]. AML with an overexpression of ecotropic viral integration site 1 (EVI-1) occurs in approximately 10% of patients with AML and is associated with poor prognosis. A study using AML cell lines showed that EVI-1 enhanced the transcriptional response to ATRA [113], and another study using primary samples showed that ATRA induces differentiation and reduces the clonogenic capacity for most of primary EVI-1-positive AML cells in vitro and in vivo, whereas other subtype AML cells lack such a response [114]. IDH mutations are discovered in approximately 15% of AML cases [115] and result in the production of the oncometabolite (R)-2-hydroxyglutarate (2-HG). Moreover, 2-HG is a competitive inhibitor of multiple α-ketoglutarate-dependent dioxygenases such as the TET family of DNA demethylases and the Jumonji family of histone demethylases, including lysine-specific demethylase 1 (LSD1) [116]. Furthermore, 2-HG dysregulates differentiation programs in leukemic cells through DNA and histone hypermethylation for several genes involved in RA signaling pathways [117,118]. A laboratory experiment revealed that IDH1-R132H mutation leads to the specific gene signatures enriched in myeloid differentiation and retinoid responsiveness. IDH1-mutated cell lines and IDH1-mutated primary cells are more sensitive to ATRA-induced differentiation and apoptosis than IDH1 wild-type cells in vitro and in vivo [119]. These experimental data suggest that EVI-1 overexpression and IDH1 mutation are possible biomarkers for ATRA sensitivity, although this assumption has not been tested in clinical trials.
The crosstalk between LSD1 and the ATRA-driven differentiation pathway has been well investigated. LSD1 is a mono- and di-methyl lysine demethylase targeting lysine 4 of histone H3 [120] and regulates hematopoietic myeloid differentiation as a transcriptional corepressor, partly through the interaction with GFI-1, which is a pro-differentiation transcription factor [121]. LSD1 is commonly overexpressed in AML [122] and is required to maintain AML cells [123]. Intriguingly, the inhibition of LSD1 potentiated an ATRA-driven differentiation of AML cell lines and primary cells together with the transcriptional activation of RAR target genes and also diminished the engraftment of primary AML samples in xenotransplantation models [7,124], suggesting that LSD1 is a promising target in combination with ATRA. Initially, the synergistic activity of this combination was thought to depend on the inhibition of LSD1 enzymatic activity; however, recent mechanistic studies have demonstrated that dissociation with LSD1 and GFI-1 is rather critical to sensitize AML cells to ATRA-driven differentiation [125]. Based on these laboratory results, phase 1 clinical trials of the LSD1 inhibitor tranylcypromine combined with ATRA have been conducted for patients with refractory/relapsed AML and MDS [43,44] (Table 1). The results showed an overall response rate of approximately 20% and a clinical benefit rate of roughly 40%. In one trial, gene signatures enriched in responders suggested more quiescent and less proliferative leukemic cells. In contrast, those increased in non-responders suggested a more proliferative phenotype characterized by mTOR signaling and MYC-regulated genes [43]. This evidence supports the further investigation of this combination strategy with more suitable patient selection and/or novel LSD1 inhibitors.
Super-enhancer (SE) is a large and highly active enhancer region that regulates essential genes to define cell identity and cell state [126,127]. More recently, SE analysis of AML patients has identified a novel subgroup with a strong SE in the RARα locus, resulting in a high level of RARα mRNA in leukemic cells, which accounted for approximately 25% of the samples analyzed and was not found in normal CD34-positive cells [8]. Intriguingly, this feature predisposes leukemic cells to an extremely high sensitivity to tamibarotene. Preclinical data showed that tamibarotene selectively induced RARα target genes such as ITGAX, ITGAM, and CD38, and inhibits the proliferation in in vitro and patient-derived xenograft (PDX) models of leukemic cells with high RARα, but not in RARα-low AML cells. Additionally, gene expression and enhancer responses to tamibarotene in RARα-high AML cells are like those of APL cells to retinoids. These results suggest a model where SE-induced over-expression of RARα results in an unbalanced excess of unliganded RARα, favoring the more repressive control of the myeloid differentiation program, as the PML-RARA fusion protein does in APL. Tamibarotene efficiently converts RARα from a repressive form to a transcriptional activator and reactivates differentiation pathways in RARα-high AML cells. Thus, a SE at the RARα locus could be a promising biomarker to select patients for tamibarotene treatment. In this study, the RARα-high PDX model was treated with tamibarotene or ATRA with doses corresponding to human exposure observed in the clinic. Intriguingly, tamibarotene showed better tumor reduction and more prolonged survival than ATRA, indicating that tamibarotene has higher potency against RARα-high AML cells, consistent with previous clinical studies showing the superiority of tamibarotene over ATRA in APL [47]. Based on these preclinical results, a biomarker-directed phase 2 trial of tamibarotene was conducted in AML and MDS patients with high RARα expression [128]. High mRNA expression was identified in approximately 30% of screened patients, and tamibarotene as a single agent achieved biological and clinical activity in 43% of the enrolled patients. Furthermore, synergistic activity with tamibarotene and hypomethylating agents (HMAs) such as azacitidine and decitabine was evaluated in RARα-high AML in a preclinical model [129]. This combination showed synergistic anti-proliferative effects in vitro and more profound and durable responses in PDX models than either agent alone, providing a rationale for designing a clinical trial of this combination strategy. In this context, a phase 2 study of tamibarotene and azacitidine combination was conducted in 51 newly diagnosed unfit AMLs including both RARα-high (N = 22) and RARα-low patients (N = 29), which were prospectively identified through real-time quantitative PCR assay (Table 1) [48]. Consistent with previous preclinical results, complete remission was much higher in RARα-high cases than in low cases (61% vs. 32%). The median time to initial complete response was rapid, at 1.2 months, and the median duration of composite complete remission was 10.8 months in RARα-high cases. Furthermore, exploratory RNA-seq analysis revealed that RARα-high patients show higher monocytic gene expression signatures than RARα-low patients, characterized by higher MCL-1 and lower BCL-2 expression. This finding is supported by TCGA non-APL AML datasets, demonstrating that approximately 70% of RARα-high patients show high monocytic gene expression signatures [48]. Notably, the BEAT AML dataset revealed that RARα-high samples show much lower ex vivo sensitivity to venetoclax than RARα-low samples [130], which is consistent with the accumulating reports of the association of monocytic AML with resistance to venetoclax [131,132]. Collectively, these results indicate that RARα-high AML cases are enriched for monocytic features associated with resistance to venetoclax-based therapies, and instead they are highly sensitive to tamibarotene and azacitidine combination. Currently, the phase 3 study of tamibarotene and azacitidine combination therapy for RARα-high high-risk MDS patients (NCT04797780), and a phase 2 study of tamibarotene plus venetoclax/azacitidine for newly diagnosed RARα-high AML patients (NCT04905407), are ongoing (Table 1).

14. Conclusions and Future Perspectives

ATRA was the first successful targeted cancer therapy, and it established a new paradigm for cancer treatment. Key to its success was the fact that it was first deployed in the treatment of a narrowly defined genetic subtype of AML. It is certainly serendipitous that the only genetically defined subtype of AML in the 1980s happened to be exquisitely responsive to ATRA, a retinoid that had been discovered 30 years earlier. Subsequent efforts to apply retinoids more broadly in the treatment and prevention of other cancers were disappointing, which is perhaps unsurprising in retrospect. Continued research has illuminated the reasons for these failures, including the body’s tight control of retinoid homeostasis and the heterogeneity of cancers, only some of which are genetically predisposed to being susceptible to retinoids. Breakthroughs in the understanding of retinoid metabolism as well as the revolution in cancer genetics are now leading to more thoughtful applications of synthetic retinoids with superior pharmacokinetics and to more precisely defined subsets of cancer, renewing hope that the full potential of retinoids and differentiating agents more broadly have not yet been realized.

Author Contributions

Y.N. and A.J.A. contributed equally to this work. Y.N. and A.J.A.; writing the manuscript, designing a figure and a table. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interests.

References

  1. Johnson, D.E.; Redner, R.L. An ATRActive future for differentiation therapy in AML. Blood. Rev. 2015, 29, 263–268. [Google Scholar]
  2. Burnett, A.K.; Hills, R.K.; Green, C.; Jenkinson, S.; Koo, K.; Patel, Y.; Guy, C.; Gilkes, A.; Milligan, D.W.; Goldstone, A.H.; et al. The impact on outcome of the addition of all-trans retinoic acid to intensive chemotherapy in younger patients with nonacute promyelocytic acute myeloid leukemia: Overall results and results in genotypic subgroups defined by mutations in NPM1, FLT3, and CEBPA. Blood 2010, 115, 948–956. [Google Scholar]
  3. Burnett, A.K.; Hills, R.K.; Milligan, D.W.; Goldstone, A.H.; Prentice, A.G.; McMullin, M.F.; Duncombe, A.; Gibson, B.; Wheatley, K. Attempts to optimize induction and consolidation treatment in acute myeloid leukemia: Results of the MRC AML12 trial. J. Clin. Oncol. 2010, 28, 586–595. [Google Scholar]
  4. Fukasawa, H.; Iijima, T.; Kagechika, H.; Hashimoto, Y.; Shudo, K. Expression of the ligand-binding domain-containing region of retinoic acid receptors alpha, beta and gamma in Escherichia coli and evaluation of ligand-binding selectivity. Biol. Pharm. Bull. 1993, 16, 343–348. [Google Scholar]
  5. Chee, L.C.Y.; Hendy, J.; Purton, L.E.; McArthur, G.A. ATRA and the specific RARα agonist, NRX195183, have opposing effects on the clonogenicity of pre-leukemic murine AML1-ETO bone marrow cells. Leukemia 2013, 27, 1369–1380. [Google Scholar] [PubMed] [Green Version]
  6. Hernandez, D.; Palau, L.; Norsworthy, K.; Anders, N.M.; Alonso, S.; Su, M.; Petkovich, M.; Chandraratna, R.; Rudek, M.A.; Smith, B.D.; et al. Overcoming microenvironment-mediated protections from ATRA using CYP26-resistant retinoids. Leukemia 2020, 34, 3077–3081. [Google Scholar] [PubMed]
  7. Schenk, T.; Chen, W.C.; Göllner, S.; Howell, L.; Jin, L.; Hebestreit, K.; Klein, H.U.; Popescu, A.C.; Burnett, A.; Mills, K.; et al. Inhibition of the LSD1 (KDM1A) demethylase reactivates the all-trans-retinoic acid differentiation pathway in acute myeloid leukemia. Nat. Med. 2012, 18, 605–611. [Google Scholar]
  8. McKeown, M.R.; Corces, M.R.; Eaton, M.L.; Fiore, C.; Lee, E.; Lopez, J.T.; Chen, M.W.; Smith, D.; Chan, S.M.; Koenig, J.L.; et al. Superenhancer Analysis Defines Novel Epigenomic Subtypes of Non-APL AML, Including an RARα Dependency Targetable by SY-1425, a Potent and Selective RARα Agonist. Cancer Discov. 2017, 7, 1136–1153. [Google Scholar]
  9. Berenguer, M.; Duester, G. Retinoic acid, RARs and early development. J. Mol. Endocrinol. 2022, 69, T59–T67. [Google Scholar] [PubMed]
  10. Bushue, N.; Wan, Y.J. Retinoid pathway and cancer therapeutics. Adv. Drug. Deliv. Rev. 2010, 62, 1285–1298. [Google Scholar] [PubMed] [Green Version]
  11. Sugiura, K.; Benedict, S.R. A Critical Study of Vitamin A and Carcinogenesis. Cancer Res. 1930, 14, 306–310. [Google Scholar]
  12. Rowe, N.H.; Gorlin, R.J. The effect of vitamin A deficiency upon experimental oral carcinogenesis. J. Dent. Res. 1959, 38, 72–83. [Google Scholar] [PubMed]
  13. Smith, A.H. Relationship between vitamin A and lung cancer. Natl. Cancer. Inst. Monogr. 1982, 62, 165–166. [Google Scholar] [PubMed]
  14. Blomhoff, R.; Blomhoff, H.K. Overview of retinoid metabolism and function. J. Neurobiol. 2006, 66, 606–630. [Google Scholar]
  15. Chambon, P. A decade of molecular biology of retinoic acid receptors. FASEB J. 1996, 10, 940–954. [Google Scholar]
  16. Collins, S.J. The role of retinoids and retinoic acid receptors in normal hematopoiesis. Leukemia 2002, 16, 1896–1905. [Google Scholar]
  17. Kastner, P.; Lawrence, H.J.; Waltzinger, C.; Ghyselinck, N.B.; Chambon, P.; Chan, S. Positive and negative regulation of granulopoiesis by endogenous RARalpha. Blood 2001, 97, 1314–1320. [Google Scholar]
  18. Qian, P.; Kumar, B.D.; He, X.C.; Nolte, C.; Gogol, M.; Ahn, Y.; Chen, S.; Li, Z.; Xu, H.; Perry, J.M.; et al. Retinoid-Sensitive Epigenetic Regulation of the Hoxb Cluster Maintains Normal Hematopoiesis and Inhibits Leukemogenesis. Cell. Stem. Cell. 2018, 22, 740–754. [Google Scholar]
  19. Brown, G. Retinoic acid receptor regulation of decision-making for cell differentiation. Front. Cell. Dev. Biol. 2023, 11, 1182204. [Google Scholar]
  20. Fenaux, P.; Wang, Z.Z.; Degos, L. Treatment of acute promyelocytic leukemia by retinoids. Curr. Top. Microbiol. Immunol. 2007, 313, 101–128. [Google Scholar]
  21. Breitman, T.R.; Selonick, S.E.; Collins, S.J. Induction of differentiation of the human promyelocytic leukemia cell line (HL-60) by retinoic acid. Proc. Natl. Acad. Sci. USA 1980, 77, 2936–2940. [Google Scholar] [PubMed]
  22. Breitman, T.R.; Collins, S.J.; Keene, B.R. Terminal differentiation of human promyelocytic leukemic cells in primary culture in response to retinoic acid. Blood 1981, 57, 1000–1004. [Google Scholar] [PubMed]
  23. Flynn, P.J.; Miller, W.J.; Weisdorf, D.J.; Arthur, D.C.; Brunning, R.; Branda, R.F. Retinoic acid treatment of acute promyelocytic leukemia: In vitro and in vivo observations. Blood 1983, 62, 1211–1217. [Google Scholar] [PubMed]
  24. Daenen, S.; Vellenga, E.; van Dobbenburgh, O.A.; Halie, M.R. Retinoic acid as antileukemic therapy in a patient with acute promyelocytic leukemia and Aspergillus pneumonia. Blood 1986, 67, 559–561. [Google Scholar]
  25. Nilsson, B. Probable in vivo induction of differentiation by retinoic acid of promyelocytes in acute promyelocytic leukaemia. Br. J. Haematol. 1984, 57, 365–371. [Google Scholar]
  26. Wang, Z.Y.; Chen, Z. Acute promyelocytic leukemia: From highly fatal to highly curable. Blood 2008, 111, 2505–2515. [Google Scholar]
  27. Runde, V.; Aul, C.; Südhoff, T.; Heyll, A.; Schneider, W. Retinoic acid in the treatment of acute promyelocytic leukemia: Inefficacy of the 13-cis isomer and induction of complete remission by the all-trans isomer complicated by thromboembolic events. Ann. Hematol. 1992, 64, 270–272. [Google Scholar]
  28. Huang, M.E.; Ye, Y.C.; Chen, S.R.; Chai, J.R.; Lu, J.X.; Zhoa, L.; Gu, L.J.; Wang, Z.Y. Use of all-trans retinoic acid in the treatment of acute promyelocytic leukemia. Blood 1988, 72, 567–572. [Google Scholar]
  29. Tallman, M.S.; Andersen, J.W.; Schiffer, C.A.; Appelbaum, F.R.; Feusner, J.H.; Ogden, A.; Shepherd, L.; Willman, C.; Bloomfield, C.D.; Rowe, J.M.; et al. All-trans-retinoic acid in acute promyelocytic leukemia. N. Engl. J. Med. 1997, 337, 1021–1028. [Google Scholar]
  30. Degos, L.; Dombret, H.; Chomienne, C.; Daniel, M.T.; Micléa, J.M.; Chastang, C.; Castaigne, S.; Fenaux, P. All-trans-retinoic acid as a differentiating agent in the treatment of acute promyelocytic leukemia. Blood 1995, 85, 2643–2653. [Google Scholar]
  31. Tallman, M.S.; Andersen, J.W.; Schiffer, C.A.; Appelbaum, F.R.; Feusner, J.H.; Woods, W.G.; Ogden, A.; Weinstein, H.; Shepherd, L.; Willman, C.; et al. All-trans retinoic acid in acute promyelocytic leukemia: Long-term outcome and prognostic factor analysis from the North American Intergroup protocol. Blood 2002, 100, 4298–4302. [Google Scholar] [PubMed]
  32. Lo-CoCo, F.; Avvisati, G.; Vignetti, M.; Thiede, C.; Orlando, S.M.; Iacobelli, S.; Ferrara, F.; Fazi, P.; Cicconi, L.; Di Bona, E.; et al. Retinoic acid and arsenic trioxide for acute promyelocytic leukemia. N. Engl. J. Med. 2013, 369, 111–121. [Google Scholar]
  33. Lo-CoCo, F.; Donato, L.D.; Schlenk, R.F. Targeted Therapy Alone for Acute Promyelocytic Leukemia. N. Engl. J. Med. 2016, 374, 1197–1198. [Google Scholar] [PubMed]
  34. Cicconi, L.; Platzbecker, U.; Avvisati, G.; Paoloni, F.; Thiede, C.; Vignetti, M.; Fazi, P.; Ferrara, F.; Divona, M.; Albano, F.; et al. Long-term results of all-trans retinoic acid and arsenic trioxide in non-high-risk acute promyelocytic leukemia: Update of the APL0406 Italian-German randomized trial. Leukemia 2020, 34, 914–918. [Google Scholar] [PubMed]
  35. Iland, H.J.; Collins, M.; Bradstock, K.; Supple, S.G.; Catalano, A.; Hertzberg, M.; Browett, P.; Grigg, A.; Firkin, F.; Campbell, L.J.; et al. Use of arsenic trioxide in remission induction and consolidation therapy for acute promyelocytic leukaemia in the Australasian Leukaemia and Lymphoma Group (ALLG) APML4 study: A non-randomised phase 2 trial. Lancet Haematol. 2015, 2, e357–e366. [Google Scholar]
  36. Burnett, A.K.; Russell, N.H.; Hills, R.K.; Bowen, D.; Kell, J.; Knapper, S.; Morgan, Y.G.; Lok, J.; Grech, A.; Jones, G.; et al. Arsenic trioxide and all-trans retinoic acid treatment for acute promyelocytic leukaemia in all risk groups (AML17): Results of a randomised, controlled, phase 3 trial. Lancet Oncol. 2015, 16, 1295–1305. [Google Scholar] [PubMed]
  37. Abaza, Y.; Kantarjian, H.; Garcia-Manero, G.; Estey, E.; Borthakur, G.; Jabbour, E.; Faderl, S.; O’Brien, S.; Wierda, W.; Pierce, S.; et al. Long-term outcome of acute promyelocytic leukemia treated with all- trans-retinoic acid, arsenic trioxide, and gemtuzumab. Blood 2017, 129, 1275–1283. [Google Scholar] [PubMed] [Green Version]
  38. Wang, H.Y.; Gong, S.; Li, G.H.; Yao, Y.Z.; Zheng, Y.S.; Lu, X.H.; Wei, S.H.; Qin, W.W.; Liu, H.B.; Wang, M.C.; et al. An effective and chemotherapy-free strategy of all-trans retinoic acid and arsenic trioxide for acute promyelocytic leukemia in all risk groups (APL15 trial). Blood Cancer J. 2022, 12, 158. [Google Scholar] [PubMed]
  39. Zhu, H.H.; Wu, D.P.; Du, X.; Zhang, X.; Liu, L.; Ma, J.; Shao, Z.H.; Ren, H.Y.; Hu, J.D.; Xu, K.L.; et al. Oral arsenic plus retinoic acid versus intravenous arsenic plus retinoic acid for non-high-risk acute promyelocytic leukaemia: A non-inferiority, randomised phase 3 trial. Lancet Oncol. 2018, 19, 871–879. [Google Scholar] [PubMed]
  40. Fenaux, P.; Le Deley, M.C.; Castaigne, S.; Archimbaud, E.; Chomienne, C.; Link, H.; Guerci, A.; Duarte, M.; Daniel, M.T.; Bowen, D. Effect of all transretinoic acid in newly diagnosed acute promyelocytic leukemia. Results of a multicenter randomized trial. European APL 91 Group. Blood 1993, 82, 3241–3249. [Google Scholar]
  41. Duvic, M.; Hymes, K.; Heald, P.; Breneman, D.; Martin, A.G.; Myskowski, P.; Crowley, C.; Yocum, R.C. Bexarotene is effective and safe for treatment of refractory advanced-stage cutaneous T-cell lymphoma: Multinational phase II-III trial results. J. Clin. Oncol. 2001, 19, 2456–2471. [Google Scholar]
  42. Matthay, K.K.; Villablanca, J.G.; Seeger, R.C.; Stram, D.O.; Harris, R.E.; Ramsay, N.K.; Swift, P.; Shimada, H.; Black, C.T.; Brodeur, G.M.; et al. Treatment of high-risk neuroblastoma with intensive chemotherapy, radiotherapy, autologous bone marrow transplantation, and 13-cis-retinoic acid. Children’s Cancer Group. N. Engl. J. Med. 1999, 341, 1165–1173. [Google Scholar] [PubMed]
  43. Tayari, M.M.; Santos, H.G.D.; Kwon, D.; Bradley, T.J.; Thomassen, A.; Chen, C.; Dinh, Y.; Perez, A.; Zelent, A.; Morey, L.; et al. Clinical Responsiveness to All-trans Retinoic Acid Is Potentiated by LSD1 Inhibition and Associated with a Quiescent Transcriptome in Myeloid Malignancies. Clin. Cancer Res. 2021, 27, 1893–1903. [Google Scholar] [PubMed]
  44. Wass, M.; Göllner, S.; Besenbeck, B.; Schlenk, R.F.; Mundmann, P.; Göthert, J.R.; Noppeney, R.; Schliemann, C.; Mikesch, J.H.; Lenz, G.; et al. A proof of concept phase I/II pilot trial of LSD1 inhibition by tranylcypromine combined with ATRA in refractory/relapsed AML patients not eligible for intensive therapy. Leukemia 2021, 35, 701–711. [Google Scholar]
  45. Lübbert, M.; Grishina, O.; Schmoor, C.; Schlenk, R.F.; Jost, E.; Crysandt, M.; Heuser, M.; Thol, F.; Salih, H.R.; Schittenhelm, M.M.; et al. Valproate and Retinoic Acid in Combination With Decitabine in Elderly Nonfit Patients With Acute Myeloid Leukemia: Results of a Multicenter, Randomized, 2 × 2, Phase II Trial. J. Clin. Oncol. 2020, 38, 257–270. [Google Scholar]
  46. Tobita, T.; Takeshita, A.; Kitamura, K.; Ohnishi, K.; Yanagi, M.; Hiraoka, A.; Karasuno, T.; Takeuchi, M.; Miyawaki, S.; Ueda, R.; et al. Treatment with a new synthetic retinoid, Am80, of acute promyelocytic leukemia relapsed from complete remission induced by all-trans retinoic acid. Blood 1997, 90, 967–973. [Google Scholar]
  47. Shinagawa, K.; Yanada, M.; Sakura, T.; Ueda, Y.; Sawa, M.; Miyatake, J.; Dobashi, N.; Kojima, M.; Hatta, Y.; Emi, N.; et al. Tamibarotene as maintenance therapy for acute promyelocytic leukemia: Results from a randomized controlled trial. J. Clin. Oncol. 2014, 32, 3729–3735. [Google Scholar]
  48. de Botton, S.; Cluzeau, T.; Vigil, C.; Cook, R.J.; Rousselot, P.; Rizzieri, D.A.; Liesveld, J.L.; Fenaux, P.; Braun, T.; Banos, A.; et al. Targeting RARA overexpression with tamibarotene, a potent and selective RARα agonist, is a novel approach in AML. Blood Adv. 2023, 7, 1858–1870. [Google Scholar] [PubMed]
  49. Vickers, M.; Jackson, G.; Taylor, P. The incidence of acute promyelocytic leukemia appears constant over most of a human lifespan, implying only one rate limiting mutation. Leukemia 2000, 14, 722–726. [Google Scholar]
  50. He, L.Z.; Tribioli, C.; Rivi, R.; Peruzzi, D.; Pelicci, P.G.; Soares, V.; Cattoretti, G.; Pandolfi, P.P. Acute leukemia with promyelocytic features in PML/RARalpha transgenic mice. Proc. Natl. Acad. Sci. USA 1997, 94, 5302–5307. [Google Scholar]
  51. Westervelt, P.; Lane, A.A.; Pollock, J.L.; Oldfather, K.; Holt, M.S.; Zimonjic, D.B.; Popescu, N.C.; DiPersio, J.F.; Ley, T.J. High-penetrance mouse model of acute promyelocytic leukemia with very low levels of PML-RARalpha expression. Blood 2003, 102, 1857–1865. [Google Scholar]
  52. Fasan, A.; Haferlach, C.; Perglerovà, K.; Kern, W.; Haferlach, T. Molecular landscape of acute promyelocytic leukemia at diagnosis and relapse. Haematologica 2017, 102, e222–e224. [Google Scholar] [PubMed]
  53. Iaccarino, L.; Ottone, T.; Alfonso, V.; Cicconi, L.; Divona, M.; Lavorgna, S.; Travaglini, S.; Ferrantini, A.; Falconi, G.; Baer, C.; et al. Mutational landscape of patients with acute promyelocytic leukemia at diagnosis and relapse. Am. J. Hematol. 2019, 94, 1091–1097. [Google Scholar]
  54. Song, H.; Liu, Y.; Tan, Y.; Zhang, Y.; Jin, W.; Chen, L.; Wu, S.; Yan, J.; Li, J.; Chen, Z.; et al. Recurrent noncoding somatic and germline WT1 variants converge to disrupt MYB binding in acute promyelocytic leukemia. Blood 2022, 140, 1132–1144. [Google Scholar]
  55. Martens, J.H.; Brinkman, A.B.; Simmer, F.; Francoijs, K.J.; Nebbioso, A.; Ferrara, F.; Altucci, L.; Stunnenberg, H.G. PML-RARalpha/RXR Alters the Epigenetic Landscape in Acute Promyelocytic Leukemia. Cancer Cell. 2010, 17, 173–185. [Google Scholar] [PubMed] [Green Version]
  56. Lo-Coco, F.; Ammatuna, E. The biology of acute promyelocytic leukemia and its impact on diagnosis and treatment. Hematol. Am. Soc. Hematol. Educ. Program. 2006, 514, 156–161. [Google Scholar]
  57. Wang, K.; Wang, P.; Shi, J.; Zhu, X.; He, M.; Jia, X.; Yang, X.; Qiu, F.; Jin, W.; Qian, M.; et al. PML/RARalpha targets promoter regions containing PU.1 consensus and RARE half sites in acute promyelocytic leukemia. Cancer Cell. 2010, 17, 186–197. [Google Scholar]
  58. Tan, Y.; Wang, X.; Song, H.; Zhang, Y.; Zhang, R.; Li, S.; Jin, W.; Chen, S.; Fang, H.; Chen, Z.; et al. A PML/RARα direct target atlas redefines transcriptional deregulation in acute promyelocytic leukemia. Blood 2021, 137, 1503–1516. [Google Scholar] [PubMed]
  59. Villiers, W.; Kelly, A.; He, X.; Kaufman-Cook, J.; Elbasir, A.; Bensmail, H.; Lavender, P.; Dillon, R.; Mifsud, B.; Osborne, C.S. Multi-omics and machine learning reveal context-specific gene regulatory activities of PML::RARA in acute promyelocytic leukemia. Nat. Commun. 2023, 14, 724. [Google Scholar]
  60. Insinga, A.; Monestiroli, S.; Ronzoni, S.; Carbone, R.; Pearson, M.; Pruneri, G.; Viale, G.; Appella, E.; Pelicci, P.; Minucci, S. Impairment of p53 acetylation, stability and function by an oncogenic transcription factor. EMBO J. 2004, 23, 1144–1154. [Google Scholar]
  61. Lin, R.J.; Nagy, L.; Inoue, S.; Shao, W.; Miller, W.H., Jr.; Evans, R.M. Role of the histone deacetylase complex in acute promyelocytic leukaemia. Nature 1998, 391, 811–814. [Google Scholar]
  62. Di Croce, L.; Raker, V.A.; Corsaro, M.; Fazi, F.; Fanelli, M.; Faretta, M.; Fuks, F.; Lo Coco, F.; Kouzarides, T.; Nervi, C.; et al. Methyltransferase recruitment and DNA hypermethylation of target promoters by an oncogenic transcription factor. Science 2002, 295, 1079–1082. [Google Scholar]
  63. Zheng, P.Z.; Wang, K.K.; Zhang, Q.Y.; Huang, Q.H.; Du, Y.Z.; Zhang, Q.H.; Xiao, D.K.; Shen, S.H.; Imbeaud, S.; Eveno, E.; et al. Systems analysis of transcriptome and proteome in retinoic acid/arsenic trioxide-induced cell differentiation/apoptosis of promyelocytic leukemia. Proc. Natl. Acad. Sci. USA 2005, 102, 7653–7658. [Google Scholar]
  64. vom Baur, E.; Zechel, C.; Heery, D.; Heine, M.J.; Garnier, J.M.; Vivat, V.; Le Douarin, B.; Gronemeyer, H.; Chambon, P.; Losson, R. Differential ligand-dependent interactions between the AF-2 activating domain of nuclear receptors and the putative transcriptional intermediary factors mSUG1 and TIF1. EMBO J. 1996, 15, 110–124. [Google Scholar]
  65. Zhu, J.; Gianni, M.; Kopf, E.; Honoré, N.; Chelbi-Alix, M.; Koken, M.; Quignon, F.; Rochette-Egly, C.; de Thé, H. Retinoic acid induces proteasome-dependent degradation of retinoic acid receptor alpha (RARalpha) and oncogenic RARalpha fusion proteins. Proc. Natl. Acad. Sci. USA 1999, 96, 14807–14812. [Google Scholar] [PubMed]
  66. Schachter-Tokarz, E.; Kelaidi, C.; Cassinat, B.; Chomienne, C.; Gardin, C.; Raffoux, E.; Dombret, H.; Fenaux, P.; Gallagher, R. PML-RARalpha ligand-binding domain deletion mutations associated with reduced disease control and outcome after first relapse of APL. Leukemia 2010, 24, 473–476. [Google Scholar] [PubMed] [Green Version]
  67. Gallagher, R.E.; Moser, B.K.; Racevskis, J.; Poiré, X.; Bloomfield, C.D.; Carroll, A.J.; Ketterling, R.P.; Roulston, D.; Schachter-Tokarz, E.; Zhou, D.C.; et al. Treatment-influenced associations of PML-RARα mutations, FLT3 mutations, and additional chromosome abnormalities in relapsed acute promyelocytic leukemia. Blood 2012, 120, 2098–2108. [Google Scholar] [PubMed] [Green Version]
  68. Hattori, H.; Ishikawa, Y.; Kawashima, N.; Akashi, A.; Yamaguchi, Y.; Harada, Y.; Hirano, D.; Adachi, Y.; Miyao, K.; Ushijima, Y.; et al. Identification of the novel deletion-type PML-RARA mutation associated with the retinoic acid resistance in acute promyelocytic leukemia. PLoS ONE 2018, 13, e0204850. [Google Scholar]
  69. Ding, W.; Li, Y.P.; Nobile, L.M.; Grills, G.; Carrera, I.; Paietta, E.; Tallman, M.S.; Wiernik, P.H.; Gallagher, R.E. Leukemic cellular retinoic acid resistance and missense mutations in the PML-RARalpha fusion gene after relapse of acute promyelocytic leukemia from treatment with all-trans retinoic acid and intensive chemotherapy. Blood 1998, 92, 1172–1183. [Google Scholar]
  70. Esnault, C.; Rahmé, R.; Rice, K.L.; Berthier, C.; Gaillard, C.; Quentin, S.; Maubert, A.L.; Kogan, S.; de Thé, H. FLT3-ITD impedes retinoic acid, but not arsenic, responses in murine acute promyelocytic leukemias. Blood 2019, 133, 1495–1506. [Google Scholar]
  71. Nasr, R.; Guillemin, M.C.; Ferhi, O.; Soilihi, H.; Peres, L.; Berthier, C.; Rousselot, P.; Robledo-Sarmiento, M.; Lallemand-Breitenbach, V.; Gourmel, B.; et al. Eradication of acute promyelocytic leukemia-initiating cells through PML-RARA degradation. Nat. Med. 2008, 14, 1333–1342. [Google Scholar]
  72. Ablain, J.; Leiva, M.; Peres, L.; Fonsart, J.; Anthony, E.; de Thé, H. Uncoupling RARA transcriptional activation and degradation clarifies the bases for APL response to therapies. J. Exp. Med. 2013, 210, 647–653. [Google Scholar] [PubMed]
  73. Zhang, X.W.; Yan, X.J.; Zhou, Z.R.; Yang, F.F.; Wu, Z.Y.; Sun, H.B.; Liang, W.X.; Song, A.X.; Lallemand-Breitenbach, V.; Jeanne, M.; et al. Arsenic trioxide controls the fate of the PML-RARalpha oncoprotein by directly binding PML. Science 2010, 328, 240–243. [Google Scholar] [PubMed]
  74. Lallemand-Breitenbach, V.; Jeanne, M.; Benhenda, S.; Nasr, R.; Lei, M.; Peres, L.; Zhou, J.; Zhu, J.; Raught, B.; de Thé, H. Arsenic degrades PML or PML-RARalpha through a SUMO-triggered RNF4/ubiquitin-mediated pathway. Nat. Cell. Biol. 2008, 10, 547–555. [Google Scholar]
  75. Schlenk, R.F.; Fröhling, S.; Hartmann, F.; Fischer, J.T.; Glasmacher, A.; del Valle, F.; Grimminger, W.; Götze, K.; Waterhouse, C.; Schoch, R.; et al. Phase III study of all-trans retinoic acid in previously untreated patients 61 years or older with acute myeloid leukemia. Leukemia 2004, 18, 1798–1803. [Google Scholar]
  76. Schlenk, R.F.; Döhner, K.; Kneba, M.; Götze, K.; Hartmann, F.; Del Valle, F.; Kirchen, H.; Koller, E.; Fischer, J.T.; Bullinger, L.; et al. Gene mutations and response to treatment with all-trans retinoic acid in elderly patients with acute myeloid leukemia. Results from the AMLSG Trial AML HD98B. Haematologica 2009, 94, 54–60. [Google Scholar] [PubMed]
  77. Preis, P.N.; Saya, H.; Nádasdi, L.; Hochhaus, G.; Levin, V.; Sadée, W. Neuronal cell differentiation of human neuroblastoma cells by retinoic acid plus herbimycin A. Cancer Res. 1988, 48, 6530–6534. [Google Scholar] [PubMed]
  78. Veal, G.J.; Cole, M.; Errington, J.; Pearson, A.D.; Foot, A.B.; Whyman, G.; Boddy, A.V. Pharmacokinetics and metabolism of 13-cis-retinoic acid (isotretinoin) in children with high-risk neuroblastoma—A study of the United Kingdom Children’s Cancer Study Group. Br. J. Cancer 2007, 96, 424–431. [Google Scholar]
  79. Bayeva, N.; Coll, E.; Piskareva, O. Differentiating Neuroblastoma: A Systematic Review of the Retinoic Acid, Its Derivatives, and Synergistic Interactions. J. Pers. Med. 2021, 11, 211. [Google Scholar]
  80. Zhang, C.; Hazarika, P.; Ni, X.; Weidner, D.A.; Duvic, M. Induction of apoptosis by bexarotene in cutaneous T-cell lymphoma cells: Relevance to mechanism of therapeutic action. Clin. Cancer Res. 2002, 8, 1234–1240. [Google Scholar]
  81. Yuan, J.M.; Stram, D.O.; Arakawa, K.; Lee, H.P.; Yu, M.C. Dietary cryptoxanthin and reduced risk of lung cancer: The Singapore Chinese Health Study. Cancer Epidemiol. Biomarkers Prev. 2003, 12, 890–898. [Google Scholar] [PubMed]
  82. Zhang, S.; Hunter, D.J.; Forman, M.R.; Rosner, B.A.; Speizer, F.E.; Colditz, G.A.; Manson, J.E.; Hankinson, S.E.; Willett, W.C. Dietary carotenoids and vitamins A, C, and E and risk of breast cancer. J. Natl. Cancer Inst. 1999, 91, 547–556. [Google Scholar] [PubMed]
  83. Connolly, R.M.; Nguyen, N.K.; Sukumar, S. Molecular pathways: Current role and future directions of the retinoic acid pathway in cancer prevention and treatment. Clin. Cancer Res. 2013, 19, 1651–1659. [Google Scholar] [PubMed] [Green Version]
  84. Omenn, G.S.; Goodman, G.E.; Thornquist, M.D.; Balmes, J.; Cullen, M.R.; Glass, A.; Keogh, J.P.; Meyskens, F.L.; Valanis, B.; Williams, J.H.; et al. Effects of a combination of beta carotene and vitamin A on lung cancer and cardiovascular disease. N. Engl. J. Med. 1996, 334, 1150–1155. [Google Scholar] [PubMed] [Green Version]
  85. Muindi, J.; Frankel, S.R.; Miller, W.H., Jr.; Jakubowski, A.; Scheinberg, D.A.; Young, C.W.; Dmitrovsky, E.; Warrell, R.P., Jr. Continuous treatment with all-trans retinoic acid causes a progressive reduction in plasma drug concentrations: Implications for relapse and retinoid “resistance” in patients with acute promyelocytic leukemia. Blood 1992, 79, 299–303. [Google Scholar]
  86. Jing, J.; Nelson, C.; Paik, J.; Shirasaka, Y.; Amory, J.K.; Isoherranen, N. Physiologically Based Pharmacokinetic Model of All- trans-Retinoic Acid with Application to Cancer Populations and Drug Interactions. J. Pharmacol. Exp. Ther. 2017, 361, 246–258. [Google Scholar]
  87. Estey, E.; Thall, P.F.; Mehta, K.; Rosenblum, M.; Brewer, T., Jr.; Simmons, V.; Cabanillas, F.; Kurzrock, R.; Lopez-Berestein, G. Alterations in tretinoin pharmacokinetics following administration of liposomal all-trans retinoic acid. Blood 1996, 87, 3650–3654. [Google Scholar]
  88. Ozpolat, B.; Lopez-Berestein, G.; Adamson, P.; Fu, C.J.; Williams, A.H. Pharmacokinetics of intravenously administered liposomal all-trans-retinoic acid (ATRA) and orally administered ATRA in healthy volunteers. J. Pharm. Pharm. Sci. 2003, 6, 292–301. [Google Scholar]
  89. Tsimberidou, A.M.; Tirado-Gomez, M.; Andreeff, M.; O’Brien, S.; Kantarjian, H.; Keating, M.; Lopez-Berestein, G.; Estey, E. Single-agent liposomal all-trans retinoic acid can cure some patients with untreated acute promyelocytic leukemia: An update of The University of Texas M. D. Anderson Cancer Center Series. Leuk. Lymphoma 2006, 47, 1062–1068. [Google Scholar]
  90. White, J.A.; Beckett-Jones, B.; Guo, Y.D.; Dilworth, F.J.; Bonasoro, J.; Jones, G.; Petkovich, M. cDNA cloning of human retinoic acid-metabolizing enzyme (hP450RAI) identifies a novel family of cytochromes P450. J. Biol. Chem. 1997, 272, 18538–18541. [Google Scholar]
  91. White, J.A.; Ramshaw, H.; Taimi, M.; Stangle, W.; Zhang, A.; Everingham, S.; Creighton, S.; Tam, S.P.; Jones, G.; Petkovich, M. Identification of the human cytochrome P450, P450RAI-2, which is predominantly expressed in the adult cerebellum and is responsible for all-trans-retinoic acid metabolism. Proc. Natl. Acad. Sci. USA 2000, 97, 6403–6408. [Google Scholar] [PubMed]
  92. Snyder, J.M.; Zhong, G.; Hogarth, C.; Huang, W.; Topping, T.; LaFrance, J.; Palau, L.; Czuba, L.C.; Griswold, M.; Ghiaur, G.; et al. Knockout of Cyp26a1 and Cyp26b1 during postnatal life causes reduced lifespan, dermatitis, splenomegaly, and systemic inflammation in mice. FASEB J. 2020, 34, 15788–15804. [Google Scholar] [PubMed]
  93. Ozpolat, B.; Mehta, K.; Lopez-Berestein, G. Regulation of a highly specific retinoic acid-4-hydroxylase (CYP26A1) enzyme and all-trans-retinoic acid metabolism in human intestinal, liver, endothelial, and acute promyelocytic leukemia cells. Leuk. Lymphoma 2005, 46, 1497–1506. [Google Scholar]
  94. Topletz, A.R.; Tripathy, S.; Foti, R.S.; Shimshoni, J.A.; Nelson, W.L.; Isoherranen, N. Induction of CYP26A1 by metabolites of retinoic acid: Evidence that CYP26A1 is an important enzyme in the elimination of active retinoids. Mol. Pharmacol. 2015, 87, 430–441. [Google Scholar]
  95. Adamson, P.C.; Bailey, J.; Pluda, J.; Poplack, D.G.; Bauza, S.; Murphy, R.F.; Yarchoan, R.; Balis, F.M. Pharmacokinetics of all-trans-retinoic acid administered on an intermittent schedule. J. Clin. Oncol. 1995, 13, 1238–1241. [Google Scholar]
  96. Kurlandsky, S.B.; Gamble, M.V.; Ramakrishnan, R.; Blaner, W.S. Plasma delivery of retinoic acid to tissues in the rat. J. Biol. Chem. 1995, 270, 17850–17857. [Google Scholar]
  97. Ghiaur, G.; Yegnasubramanian, S.; Perkins, B.; Gucwa, J.L.; Gerber, J.M.; Jones, R.J. Regulation of human hematopoietic stem cell self-renewal by the microenvironment’s control of retinoic acid signaling. Proc. Natl. Acad. Sci. USA 2013, 110, 16121–16126. [Google Scholar]
  98. Su, M.; Alonso, S.; Jones, J.W.; Yu, J.; Kane, M.A.; Jones, R.J.; Ghiaur, G. All-Trans Retinoic Acid Activity in Acute Myeloid Leukemia: Role of Cytochrome P450 Enzyme Expression by the Microenvironment. PLoS ONE 2015, 10, e0127790. [Google Scholar]
  99. Milligan, D.W.; Wheatley, K.; Littlewood, T.; Craig, J.I.; Burnett, A.K. Fludarabine and cytosine are less effective than standard ADE chemotherapy in high-risk acute myeloid leukemia, and addition of G-CSF and ATRA are not beneficial: Results of the MRC AML-HR randomized trial. Blood 2006, 107, 4614–4622. [Google Scholar] [PubMed]
  100. Hashimoto, Y.; Kagechika, H.; Kawachi, E.; Fukasawa, H.; Saito, G.; Shudo, K. Correlation of differentiation-inducing activity of retinoids on human leukemia cell lines HL-60 and NB4. J. Cancer Res. Clin. Oncol. 1995, 121, 696–698. [Google Scholar] [PubMed]
  101. Liang, C.; Qiao, G.; Liu, Y.; Tian, L.; Hui, N.; Li, J.; Ma, Y.; Li, H.; Zhao, Q.; Cao, W.; et al. Overview of all-trans-retinoic acid (ATRA) and its analogues: Structures, activities, and mechanisms in acute promyelocytic leukaemia. Eur. J. Med. Chem. 2021, 220, 113451. [Google Scholar] [PubMed]
  102. Osanai, M.; Petkovich, M. Expression of the retinoic acid-metabolizing enzyme CYP26A1 limits programmed cell death. Mol. Pharmacol. 2005, 67, 1808–1817. [Google Scholar] [PubMed] [Green Version]
  103. Miwako, I.; Kagechika, H. Tamibarotene. Drugs Today 2007, 43, 563–568. [Google Scholar]
  104. Zelent, A.; Krust, A.; Petkovich, M.; Kastner, P.; Chambon, P. Cloning of murine alpha and beta retinoic acid receptors and a novel receptor gamma predominantly expressed in skin. Nature 1989, 339, 714–717. [Google Scholar]
  105. Warrell, R.P., Jr.; de Thé, H.; Wang, Z.Y.; Degos, L. Acute promyelocytic leukemia. N. Engl. J. Med. 1993, 329, 177–189. [Google Scholar] [PubMed]
  106. Shinjo, K.; Takeshita, A.; Ohnishi, K.; Sakura, T.; Miyawaki, S.; Hiraoka, A.; Takeuchi, M.; Tomoyasu, S.; Wakita, H.; Ata, K.; et al. Good prognosis of patients with acute promyelocytic leukemia who achieved second complete remission (CR) with a new retinoid, Am80, after relapse from CR induced by all-trans-retinoic acid. Int. J. Hematol. 2000, 72, 470–473. [Google Scholar]
  107. Sanford, D.; Lo-Coco, F.; Sanz, M.A.; Di Bona, E.; Coutre, S.; Altman, J.K.; Wetzler, M.; Allen, S.L.; Ravandi, F.; Kantarjian, H.; et al. Tamibarotene in patients with acute promyelocytic leukaemia relapsing after treatment with all-trans retinoic acid and arsenic trioxide. Br. J. Haematol. 2015, 171, 471–477. [Google Scholar]
  108. Zhu, H.H.; Qin, Y.Z.; Huang, X.J. Resistance to arsenic therapy in acute promyelocytic leukemia. N. Engl. J. Med. 2014, 370, 1864–1866. [Google Scholar]
  109. Goto, E.; Tomita, A.; Hayakawa, F.; Atsumi, A.; Kiyoi, H.; Naoe, T. Missense mutations in PML-RARA are critical for the lack of responsiveness to arsenic trioxide treatment. Blood 2011, 118, 1600–1609. [Google Scholar]
  110. Geoffroy, M.C.; Esnault, C.; de Thé, H. Retinoids in hematology: A timely revival? Blood 2021, 137, 2429–2437. [Google Scholar]
  111. Brown, G.; Marchwicka, A.; Cunningham, A.; Toellner, K.M.; Marcinkowska, E. Antagonizing Retinoic Acid Receptors Increases Myeloid Cell Production by Cultured Human Hematopoietic Stem Cells. Arch. Immunol. Ther. Exp. 2017, 65, 69–81. [Google Scholar]
  112. Ambinder, A.J.; Norsworthy, K.; Hernandez, D.; Palau, L.; Paun, B.; Duffield, A.; Chandraratna, R.; Sanders, M.; Varadhan, R.; Jones, R.J.; et al. A Phase 1 Study of IRX195183, a RARα-Selective CYP26 Resistant Retinoid, in Patients With Relapsed or Refractory AML. Front. Oncol. 2020, 10, 587062. [Google Scholar] [PubMed]
  113. Steinmetz, B.; Hackl, H.; Slabáková, E.; Schwarzinger, I.; Smějová, M.; Spittler, A.; Arbesu, I.; Shehata, M.; Souček, K.; Wieser, R. The oncogene EVI1 enhances transcriptional and biological responses of human myeloid cells to all-trans retinoic acid. Cell Cycle 2014, 13, 2931–2943. [Google Scholar] [PubMed] [Green Version]
  114. Verhagen, H.J.; Smit, M.A.; Rutten, A.; Denkers, F.; Poddighe, P.J.; Merle, P.A.; Ossenkoppele, G.J.; Smit, L. Primary acute myeloid leukemia cells with overexpression of EVI-1 are sensitive to all-trans retinoic acid. Blood 2016, 127, 458–463. [Google Scholar]
  115. Mardis, E.R.; Ding, L.; Dooling, D.J.; Larson, D.E.; McLellan, M.D.; Chen, K.; Koboldt, D.C.; Fulton, R.S.; Delehaunty, K.D.; McGrath, S.D.; et al. Recurring mutations found by sequencing an acute myeloid leukemia genome. N. Engl. J. Med. 2009, 361, 1058–1066. [Google Scholar]
  116. Xu, W.; Yang, H.; Liu, Y.; Yang, Y.; Wang, P.; Kim, S.H.; Ito, S.; Yang, C.; Wang, P.; Xiao, M.T.; et al. Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of α-ketoglutarate-dependent dioxygenases. Cancer Cell. 2011, 19, 17–30. [Google Scholar]
  117. Figueroa, M.E.; Abdel-Wahab, O.; Lu, C.; Ward, P.S.; Patel, J.; Shih, A.; Li, Y.; Bhagwat, N.; Vasanthakumar, A.; Fernandez, H.F.; et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell. 2010, 18, 553–567. [Google Scholar]
  118. Chou, A.P.; Chowdhury, R.; Li, S.; Chen, W.; Kim, A.J.; Piccioni, D.E.; Selfridge, J.M.; Mody, R.R.; Chang, S.; Lalezari, S.; et al. Identification of retinol binding protein 1 promoter hypermethylation in isocitrate dehydrogenase 1 and 2 mutant gliomas. J. Natl. Cancer Inst. 2012, 104, 1458–1469. [Google Scholar]
  119. Boutzen, H.; Saland, E.; Larrue, C.; de Toni, F.; Gales, L.; Castelli, F.A.; Cathebas, M.; Zaghdoudi, S.; Stuani, L.; Kaoma, T.; et al. Isocitrate dehydrogenase 1 mutations prime the all-trans retinoic acid myeloid differentiation pathway in acute myeloid leukemia. J. Exp. Med. 2016, 213, 483–497. [Google Scholar]
  120. Shi, Y.; Lan, F.; Matson, C.; Mulligan, P.; Whetstine, J.R.; Cole, P.A.; Casero, R.A.; Shi, Y. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell 2004, 119, 941–953. [Google Scholar]
  121. Saleque, S.; Kim, J.; Rooke, H.M.; Orkin, S.H. Epigenetic regulation of hematopoietic differentiation by Gfi-1 and Gfi-1b is mediated by the cofactors CoREST and LSD1. Moll. Cell. 2007, 27, 562–572. [Google Scholar]
  122. Berglund, L.; Björling, E.; Oksvold, P.; Fagerberg, L.; Asplund, A.; Szigyarto, C.A.; Persson, A.; Ottosson, J.; Wernérus, H.; Nilsson, P.; et al. A genecentric Human Protein Atlas for expression profiles based on antibodies. Mol. Cell. Proteom. 2008, 7, 2019–2027. [Google Scholar]
  123. Harris, W.J.; Huang, X.; Lynch, J.T.; Spencer, G.J.; Hitchin, J.R.; Li, Y.; Ciceri, F.; Blaser, J.G.; Greystoke, B.F.; Jordan, A.M.; et al. The histone demethylase KDM1A sustains the oncogenic potential of MLL-AF9 leukemia stem cells. Cancer Cell. 2012, 21, 473–487. [Google Scholar] [PubMed] [Green Version]
  124. Smitheman, K.N.; Severson, T.M.; Rajapurkar, S.R.; McCabe, M.T.; Karpinich, N.; Foley, J.; Pappalardi, M.B.; Hughes, A.; Halsey, W.; Thomas, E.; et al. Lysine specific demethylase 1 inactivation enhances differentiation and promotes cytotoxic response when combined with all- trans retinoic acid in acute myeloid leukemia across subtypes. Haematologica 2019, 104, 1156–1167. [Google Scholar] [PubMed] [Green Version]
  125. Ravasio, R.; Ceccacci, E.; Nicosia, L.; Hosseini, A.; Rossi, P.L.; Barozzi, I.; Fornasari, L.; Zuffo, R.D.; Valente, S.; Fioravanti, R.; et al. Targeting the scaffolding role of LSD1 (KDM1A) poises acute myeloid leukemia cells for retinoic acid-induced differentiation. Sci. Adv. 2020, 6, eaax2746. [Google Scholar]
  126. Lovén, J.; Hoke, H.A.; Lin, C.Y.; Lau, A.; Orlando, D.A.; Vakoc, C.R.; Bradner, J.E.; Lee, T.I.; Young, R.A. Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 2013, 153, 320–334. [Google Scholar]
  127. Hnisz, D.; Abraham, B.J.; Lee, T.I.; Lau, A.; Saint-André, V.; Sigova, A.A.; Hoke, H.A.; Young, R.A. Super-enhancers in the control of cell identity and disease. Cell 2013, 155, 934–947. [Google Scholar]
  128. Jurcic, J.G.; Raza, A.; Vlad, G.; Stein, E.M.; Roshal, M.; Bixby, D.L.; Boyer, D.F.; Vigil, C.E.; Syrbu, S.; Sekeres, M.A.; et al. Early Results from a Biomarker-Directed Phase 2 Trial of Sy-1425 in Acute Myeloid Leukemia (AML) and Myelodysplastic Syndrome (MDS) Demonstrate DHRS3 Induction and Myeloid Differentiation Following Sy-1425 Treatment. Blood 2017, 130 (Suppl. 1), 2633. [Google Scholar]
  129. McKeown, M.R.; Johannessen, L.; Lee, E.; Fiore, C.; di Tomaso, E. Antitumor synergy with SY-1425, a selective RARα agonist, and hypomethylating agents in retinoic acid receptor pathway activated models of acute myeloid leukemia. Haematologica 2019, 104, e138–e142. [Google Scholar]
  130. Tyner, J.W.; Tognon, C.E.; Bottomly, D.; Wilmot, B.; Kurtz, S.E.; Savage, S.L.; Long, N.; Schultz, A.R.; Traer, E.; Abel, M.; et al. Functional genomic landscape of acute myeloid leukaemia. Nature 2018, 562, 526–531. [Google Scholar]
  131. Pei, S.; Pollyea, D.A.; Gustafson, A.; Stevens, B.M.; Minhajuddin, M.; Fu, R.; Riemondy, K.A.; Gillen, A.E.; Sheridan, R.M.; Kim, J.; et al. Monocytic Subclones Confer Resistance to Venetoclax-Based Therapy in Patients with Acute Myeloid Leukemia. Cancer Discov. 2020, 10, 536–551. [Google Scholar]
  132. DiNardo, C.D.; Jonas, B.A.; Pullarkat, V.; Thirman, M.J.; Garcia, J.S.; Wei, A.H.; Konopleva, M.; Döhner, H.; Letai, A.; Fenaux, P.; et al. Azacitidine and Venetoclax in Previously Untreated Acute Myeloid Leukemia. N. Engl. J. Med. 2020, 383, 617–629. [Google Scholar]
Figure 1. Molecular structures of ATRA and synthetic retinoids.
Figure 1. Molecular structures of ATRA and synthetic retinoids.
Cancers 15 03535 g001
Table 1. Summary of clinical trials of retinoids in APL and other malignancies.
Table 1. Summary of clinical trials of retinoids in APL and other malignancies.
Studies of Retinoids Leading to Approved Use for the Study Population
Patient PopulationStudy DesignRetinoid RegimenResponse RatesSurvivalReference
Newly diagnosed APLRandomized controlled trialChemotherapy vs. ATRA + chemotherapyCR 81% vs. 91%, p = 0.251-year EFS 50% vs. 79% at 12 months, p = 0.001[40]
Newly diagnosed APLRandomized controlled trialATRA + chemotherapy vs. ATRA + arsenic trioxideCR 95% vs. 100%, p = 0.122-year EFS 86% vs. 97%. p = 0.02 for superiority[32]
Advanced stage (stage IIB-IVB) CTCLPhase IIb/III clinical trialBexaroteneORR 55%, CR 13%Relapse rate after response 36%, Median duration of response 299 days[41]
High Risk NeuroblastomaRandomized controlled trialIsotretinoin maintenance vs. placeboNA3-year EFS 46% vs. 29%, p = 0.027[42]
Studies of Retinoids in Diseases for Which They Are Not Specifically Approved
Patient PopulationStudy DesignRetinoid RegimenResponse RatesSurvivalReference
AML (Non-APL)Randomized controlled trialATRA + chemotherapy vs. chemotherapy aloneORR 83% vs. 84% (p = NS)8-year OS was 33% vs. 30%; HR, 0.98 [0.85–1.14])[2]
Relapsed/Refractory AML and MDSPhase IATRA + TranylcypromineORR was 30.8% (95% CI = 10.4–61.1)OS 5.0 months (95% CI = 3.2–21.6)[43]
Relapsed/Refractory AMLPhase I/IIATRA + TranylcypromineORR 20%one-year OS 22%[44]
Unfit, Newly diagnosed AML (Non-APL)Phase II Randomized controlled trialATRA + Decitabine vs. Decitabine aloneORR 21.9% vs 13.5%; OR, 1.80; 95% CI, 0.86–3.79; one-sided p = 0.06Median OS 8.2 months vs. 5.1 (HR, 0.65; 95% CI, 0.48–0.89; two-sided p = 0.006)[45]
APL relapsed after ATRAPhase IITamibarotene58% CRNot reported[46]
Newly Diagnosed APLRandomized controlled trialTamibarotene vs. ATRA MaintenanceNA4-year RFS was 87% vs. 58% (HR, 0.26; 95% CI, 0.07–0.95)[47]
Newly Diagnosed AML (RARA positive)Phase IITamibarotene + AzacitidineCR/CRi rate of 61%median OS was 8.4 months (95% CI: 5.2, 15.6)[48]
Ongoing Clinical Trials Exploring New Applications for Retinoids
Patient PopulationStudy DesignRetinoid RegimenResponse RatesSurvivalReference
Newly diagnosed or Relapsed AML (Non-APL)Phase IATRA + ATO + Realgar-Indigo naturalis formulaNANANCT05297123
Relapsed or Refractory AML (Non-APL)Phase ITAS1440 (Oral LSD1 inhibitor) + ATRANANANCT04282668
Newly Diagnosed AML (Non-APL, RARA positive)Phase IITamibarotene + Azacitidine/VenetoclaxNANANCT04905407
Newly diagnosed Multiple MyelomaPhase IIDaratumumab, Pomalidomide, Dexamethasone, and ATRANANANCT04700176
Newly Diagnosed MDS (RARA positive)Phase III Randomized Clinical TrialTamibarotene + AzacitidineNANANCT04797780
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Nagai, Y.; Ambinder, A.J. The Promise of Retinoids in the Treatment of Cancer: Neither Burnt Out Nor Fading Away. Cancers 2023, 15, 3535. https://doi.org/10.3390/cancers15143535

AMA Style

Nagai Y, Ambinder AJ. The Promise of Retinoids in the Treatment of Cancer: Neither Burnt Out Nor Fading Away. Cancers. 2023; 15(14):3535. https://doi.org/10.3390/cancers15143535

Chicago/Turabian Style

Nagai, Yuya, and Alexander J. Ambinder. 2023. "The Promise of Retinoids in the Treatment of Cancer: Neither Burnt Out Nor Fading Away" Cancers 15, no. 14: 3535. https://doi.org/10.3390/cancers15143535

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop