Next Article in Journal
Effects of the COVID-19 Pandemic on the Interest of Google Queries in Cancer Screening and Cancers: A Retrospective Study
Previous Article in Journal
Clinical Significance of the Lymph Node Ratio of the Second Operation to Predict Re-Recurrence in Thyroid Carcinoma
Previous Article in Special Issue
Honeycomb-like Structured Film, a Novel Therapeutic Device, Suppresses Tumor Growth in an In Vivo Ovarian Cancer Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

c-MYC-Driven Polyamine Metabolism in Ovarian Cancer: From Pathogenesis to Early Detection and Therapy

by
Yihui Chen
,
Ricardo A. León-Letelier
,
Ali Hussein Abdel Sater
,
Jody Vykoukal
,
Jennifer B. Dennison
,
Samir Hanash
and
Johannes F. Fahrmann
*
Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(3), 623; https://doi.org/10.3390/cancers15030623
Submission received: 4 January 2023 / Accepted: 14 January 2023 / Published: 19 January 2023
(This article belongs to the Special Issue New Insights of Ovarian Cancer Treatment)

Abstract

:

Simple Summary

This review informs on how the MYC signaling is dysregulated and participates in ovarian cancer progression, and the unmet challenge to directly target MYC for ovarian cancer treatment. Therefore, we proposed to alternatively target essential downstream polyamine metabolism pathway of MYC. In this review we include the metabolism of polyamine, the regulation of polyamine metabolism by MYC signaling, the utility of polyamine as therapeutic targets and cancer biomarkers.

Abstract

c-MYC and its paralogues MYCN and MYCL are among the most frequently amplified and/or overexpressed oncoproteins in ovarian cancer. c-MYC plays a key role in promoting ovarian cancer initiation and progression. The polyamine pathway is a bona fide target of c-MYC signaling, and polyamine metabolism is strongly intertwined with ovarian malignancy. Targeting of the polyamine pathway via small molecule inhibitors has garnered considerable attention as a therapeutic strategy for ovarian cancer. Herein, we discuss the involvement of c-MYC signaling and that of its paralogues in promoting ovarian cancer tumorigenesis. We highlight the potential of targeting c-MYC-driven polyamine metabolism for the treatment of ovarian cancers and the utility of polyamine signatures in biofluids for early detection applications.

1. Introduction

Ovarian cancer remains among the leading causes of cancer-related mortality among women. The 5-year overall survival rate for ovarian cancer is approximately 50% and decreased to nearly 30% for patients with distant metastasis [1]. It is estimated that there will be 20,000 new ovarian cancer cases and 13,000 deaths due to ovarian cancer in the United States in 2022 [1].
The MYC family of transcription factors c-MYC, MYCN, and MYCL are closely associated with ovarian and other cancers through amplification or overexpression [2,3,4,5,6,7]. MYC family members contain several conserved regions, including the MYC homology boxes (MBs), transactivation domain (TAD), basic region (BR), and helix–loop–helix–leucine zipper (HLH-LZ) domain [8]. Several c-MYC interacting proteins have been identified, including MYC-associated factor X (MAX) [9], WD repeat-containing protein 5 (WDR5) [10], TATA box-binding protein (TBP) [11], and bridging integrator 1 (BIN1) [12], whereas Aurora kinase A (AURKA) has been shown to interact with MYCN through MBI and flanking regions on MYCN [13]. These interactors enable MYC family members to function as transcription factors to promote oncogenesis. The c-MYC heterodimer can also repress gene expression via binding to the transcription factors MYC-interaction zinc-finger protein 1 (Miz1) and specificity protein 1 (SP1) [14,15].
c-MYC expression and function are tightly regulated at both the transcriptional level by a variety of transcriptional regulatory motifs within its proximal promotor region [16], as well as at the protein level through post-translational modifications, namely phosphorylation at various serine and threonine residues that promotes proteasomal degradation through the canonical SCFFBXW7 ubiquitin ligase pathway [17].
The MYC family oncoproteins, particularly c-MYC, are essential master regulators of metabolic reprogramming in a variety of cancer types, including ovarian cancer [18,19]. Among the c-MYC-regulated metabolic pathways is the polyamine biosynthetic pathway. Polyamines are ubiquitously distributed highly charged small basic molecules that are strongly associated with hyperproliferative diseases and play essential roles in c-MYC-driven malignancies by promoting cell growth and other pro-tumoral activities [20,21]. Cancer cells upregulate polyamine pools either by increasing de novo biosynthesis or enhancing salvaging from the surrounding milieu. Catabolism of polyamines is mediated by spermidine/spermine N1-acetyltranferase (SSAT), which is responsive to intracellular polyamine pools, resulting in the formation of acetylated polyamine derivatives, such as diacetylspermine (DAS), that can be secreted into the surrounding microenvironment and can be detected in peripheral blood, providing potential utility for cancer detection [22]. Targeting dysregulated polyamine metabolism is also being increasingly explored as a therapeutic strategy for various malignancies, including ovarian cancer.
In this review, we discuss the significance of c-MYC and its paralogues in ovarian cancer. We additionally highlight opportunities for exploiting c-MYC-driven polyamine metabolism for early detection and therapeutic applications in ovarian cancer.

2. MYC Family Members and Ovarian Cancer

2.1. Genomic Aberrations and Overexpression of c-MYC in Ovarian Cancer

MYC is located on chromosome 8q24 [23,24], a commonly amplified region in ovarian carcinomas [25]. Genomic alterations in MYC family members are prevalent in ovarian cancer [2,4]. c-MYC copy-number amplifications have been reported in up to 50% of ovarian carcinomas, whereas MYCL copy-number amplifications and gene overexpression have been reported in upwards of 21% of ovarian carcinomas [6,26,27,28,29,30,31,32,33,34,35,36,37]. MYCN amplification has also been frequently found in ovarian cancers [38].
c-MYC transcript and protein levels are higher in ovarian tumors compared to normal tissues, and c-MYC overexpression is associated with more aggressive disease [39,40]. c-MYC gene expression tends to correlate with c-MYC copy-number amplification [41,42,43], although these are not mutual. MYCL and MYCN gene transcripts also are overexpressed in some ovarian cancers [38,39,44].
c-MYC transcriptional activity is, in part, modulated post-translationally through phosphorylation and proteasomal degradation [45,46]. Phosphorylation at serine 62 (Ser62) by mitogenic signaling increases c-MYC stability [47]. Studies have demonstrated that mutations at the threonine 58 residue result in constitutive Ser62 phosphorylation [48], leading to c-MYC protein overexpression and activation. Cancer cells also maintain Ser62 phosphorylation of c-MYC by downregulating protein phosphatase 2A (PP2A), a p-Ser62 targeting phosphatase [49,50,51,52]. In ovarian cancer, proto-oncogene serine/threonine-protein kinase Pim-1 interacts with c-MYC and mediates its Ser62 phosphorylation, promoting cancer cell proliferation [53]. F-Box protein 31 (FBOX31), which is suppressed by c-MYC, facilitates the ubiquitination and degradation of c-MYC independent of its phosphorylation status [54].

2.2. Prognostic Value of c-MYC in Ovarian Cancers

Several studies have assessed the prognostic value of c-MYC copy-number amplification as well as mRNA expression and protein levels in ovarian cancer. Wang found that ovarian cancer patients with c-MYC copy-number amplifications had poorer overall survival compared to those with wild-type c-MYC [29]. Similarly, Katsaros et al. reported that c-MYC copy-number amplification with high p185/p21 in ovarian tumors was associated with shorter patient survival [28]. Elevated tumor c-MYC mRNA levels were prognostic for poor disease-free survival and worse overall survival in The Cancer Genome Atlas (TCGA) ovarian cancer dataset [55]. MYC protein levels and localization account for its transcriptional activity. Two independent studies revealed positive correlations between the c-MYC protein levels and sub-cellular localization with tumor size and tumor classification, respectively [56,57]. Ning et al. found that enrichment of c-MYC in the nucleus in early-stage ovarian cancer correlated with shorter overall survival [43]. Yamamoto et al. showed that phosphorylation of c-MYC at Ser62 was prognostic for poor overall survival [42]. The above-mentioned studies suggest a prognostic role of c-MYC. However, it should be noted that several studies have reported non-significant associations between c-MYC copy numbers as well as mRNA and protein levels in ovarian tumors with survival outcomes [30,34,39,42,58,59,60]
Collectively, these findings emphasize the need to integrate the multidimensional data incorporating the complexity of c-MYC signaling at the protein level, localization, as well as PTM status, to define the relationship between c-MYC and ovarian cancer.

2.3. Therapeutic Targeting of MYC in Ovarian Cancers

Oncogenic c-MYC and its paralogues orchestrate tumor-promoting signaling through reprograming of cell growth, survival, metabolism, and shaping the tumor microenvironment (TME) and tumor immunity [18,61,62]. Many cancers, including ovarian cancer, are addicted to c-MYC signaling [62,63,64,65], which has served as the justification for targeting c-MYC [66,67,68,69]. Early investigations demonstrated anti-ovarian cancer efficacy by targeting c-MYC using triplex-forming and liposomal phosphorothioate oligonucleotides in vitro [70,71]. A small molecule 10058-F4 blocks c-MYC/MAX heterodimerization in ovarian cancer cells and thus induces cell cycle arrest and apoptosis and attenuates glutamine uptake, an essential nutrient for cancer cells [72,73]. Another molecule, JQ1, targeting c-MYC and BRD4, suppresses ovarian cancer cell proliferation and induces apoptosis [74,75,76]. Concomitant upregulation of FAK and c-MYC in HGSOC implies a co-targeting strategy. Dual targeting of FAK and MYC by VS-6063 and JQ1 resulted in cell cycle arrest and cell death of ovarian cancer cells in vitro [77]. Moreover, c-MYC amplified chemotherapy-resistant ovarian cancer cells were demonstrated to be highly sensitive to combinational JQ1 and GS-626510, a bromodomain and extra-terminal motif inhibitor [75]. Indirect inhibition of c-MYC by the CDK7 and CDK12/13 inhibitor THZ1 repressed tumor growth in patient-derived xenograft models of ovarian cancer [65]. Dual inhibition of PARP and CDK4/6 suppressed ovarian cancer cell growth in vitro and tumor growth in vivo in a c-MYC-dependent manner [78]. The insulin-like growth factor II mRNA-binding protein 1 (IGF2BP1/IMP1) inhibitor BTYNB destabilizes c-MYC mRNA via disrupting IMP1 binding to c-MYC mRNA, thereby repressing ovarian cancer cell proliferation [79]. In addition, targeting c-MYC also sensitizes ovarian cancer cells to chemotherapy [55,75,76].
Despite the above-mentioned studies, targeting c-MYC and its paralogues has remained challenging due to protein and peptides inhibitors undergoing rapid proteolytic degradation, as well as issues related to the low membrane and cell permeability, low oral bioavailability, high clearance, and poor tissue distribution [80]. Currently, there are no drugs clinically available for targeting c-MYC in human cancers. An alternative strategy is to target one or several downstream signaling pathways of c-MYC.

3. MYC and Polyamine Regulation

As an oncogenic transcription factor, c-MYC and its paralogues transcriptionally regulate hundreds of genes that impact tumor metabolism and enable tumor progression [3,19,62,81]. c-MYC is involved in nutrient sensing and regulates glucose, glutamine, and lipids metabolism by upregulating glucose transporter 1, pyruvate kinase isoenzymes M1 and M2, and lactate dehydrogenase A [19]. Among other c-MYC-regulated metabolic pathways is the polyamine metabolism pathway, which plays critical tumor-promoting roles [20,82,83,84].
Polyamines are small polycationic molecules present in nearly all living organisms. The three major mammalian polyamines (putrescine, spermidine, spermine) play essential roles in cell proliferation, and functionality and polyamines are commonly linked to hyperproliferative disorders, including cancer. Polyamine content is usually higher in rapidly growing cells and tissues and is upregulated by regenerative and growth-promoting hormonal stimuli [85,86,87]. Inhibition of polyamine biosynthesis induces cytostasis, which is reversible by the supplementation of exogenous putrescine or spermidine [88]. The best-known molecular function of spermidine is serving as the aminobutyl group donor for the hypusination of eIF5A, which is essential for the activation of this translation factor that contributes to transcription, mRNA turnover, nucleocytoplasmic transport, and apoptosis [89].

3.1. Biosynthesis of Polyamines

Putrescine, spermidine, and spermine are the only three polyamines that can be synthesized de novo in mammalian cells [21]. Putrescine is the central intermediate of polyamine biosynthesis. Ornithine decarboxylase 1 (ODC1), in the presence of cofactor PLP, produces putrescine through the decarboxylation of ornithine (Orn), a metabolic product of arginase [90,91]. ODC1 is the rate-limiting step in putrescine production and is a bona fide target of oncogenic c-MYC (Figure 1).
The aminopropyl moiety is required to form higher polyamine molecules, such as spermidine and spermine, from putrescine [20,21]. S-adenosylmethionine decarboxylase (AdoMetDC, encoded by AMD1) converts S-adenosylmethionine (AdoMet) into decarboxylated AdoMet (dcAdoMet), the aminopropyl group donor [92]. Putrescine stimulates the processing of mammalian AdoMetDC and enhances its activity (Figure 1) [92,93]. Therefore, putrescine facilitates its conversion resulting in higher levels of spermidine and spermine than putrescine in mammalian cells and tissues.
Spermidine and spermine are the most pronounced non-acetylated polyamines in mammals. Aminopropyltransferases, namely spermidine synthase (SRM) and spermine synthase (SMS) are responsible for the formation of these higher polyamines. SRM transfers the aminopropyl moiety to putrescine to generate spermidine, and SMS adds another aminopropyl group to spermidine from spermine (Figure 1) [94]. Despite their similar aminopropyl transferring activity, SRM and SMS are distinct enzymes with strict substrate specificity [95,96]. This difference prevents the cross-reaction between these enzymes and their substrates.

3.2. Exogenous Sources of Polyamines

Although mammalian cells have an intact pathway for de novo biosynthesis of polyamines, exogenous sources are additional means to maintain polyamine homeostasis (Figure 1) [97,98,99,100]. Polyamines are found in all types of food and provide hundreds of micromoles of polyamines to the gut lumen daily [101,102,103]. For instance, agmatine, which is not synthesized by mammalian cells but is still widely present in mammalian tissues, is a polyamine produced by plants and microorganisms and mainly obtained from the diet and gut microbiota [21,104]. Sawada et al. suggested that ingested food is the major source of polyamines in the lumen of the upper small bowel in humans [105], while the gut microbiota, to some extent, is responsible for polyamines level mainly in the lower part of the intestine [106,107]. A polyamine-rich diet significantly increases levels of circulating polyamines in mouse and human [108,109].
Luminal polyamines are rapidly absorbed in the upper intestine [110]. In addition to passive diffusion [111,112], endocytosis and solute carrier-dependent mechanism also participate in polyamine uptake [113]. A recent study demonstrated that ingestion of exogenous putrescine is mediated by a caveolin-1 and NOS2-dependent process, while the solute carrier transporter SLC3A2 imports putrescine from the polyamine-rich lumen [114]. Colonic mucosa is also involved in transporting polyamines from the colonic lumen into the bloodstream [115]. It is becoming increasingly evident that exogenous polyamines affect whole-body polyamine levels and have an impact on a variety of activities in health and diseases [97,98,101,103,116,117]. Understanding the importance of exogenous polyamines requires further investigations.

3.3. Mechanisms of Extracellular Polyamines Uptake

Importing polyamines from the extracellular compartment has been shown to be of equal importance as de novo polyamine biosynthesis. Several studies have demonstrated the diverse activities of extracellular polyamines [118,119,120,121]. However, the mechanisms driving polyamine uptake at the molecular level remain incomplete. A study suggested that chemicals modulating the membrane potential affect the polyamine import system, and protein kinase C inhibits polyamine uptake [122]. Heparan sulfate is also implicated in polyamine import, which can be blocked by the single-chain fragment of an anti-heparan sulfate antibody [123]. Glypican-1 (GPC-1), along with other glypicans, is a family of heparan sulfate proteoglycans [124]. The Heparan sulfate side chains of recycling GPC-1 sequester spermine for its uptake [125]. By using a fluorescent polyamine probe, Soulet et al. demonstrated the internalization and localization of polyamines in discrete vesicles [126]. Another study provided a supportive observation of caveolin-1-dependent endocytosis-mediated polyamine uptake [127]. A previous study suggested that the solute carrier transporter SLC3A2 is an exporter of polyamines [128], while an independent study demonstrated that SLC3A2 also mediates polyamine import [129]. To this end, Uemura et al. demonstrated that SLC3A2 is a bidirectional polyamines transporter that can either import or export polyamines depending on the concentration gradient [114]. In addition, the ATP13A2 and ATP13A3 ATPases are also implicated in polyamine uptake, although the biochemical mechanism is not fully understood [130,131,132]. Multiple mechanisms may cooperate to regulate polyamines import, and their involvement could be tissue-specific and context-dependent.
The export of polyamines is important to maintain polyamine homeostasis. This process is well-studied in bacteria, yeast and protozoa [133,134,135,136,137,138,139,140,141]. Although the excretion of these compounds is also reported in mammalian cells [142,143,144,145,146], the underlying molecular mechanism remains elusive. To date, SLC3A2 is the only well-studied exporter of polyamines [114,128]. The solute carrier 18B1 (SLC18B1), a member of the vesicular amine transporter family, mediates the exocytosis of polyamines from mast cells [147], and another study reported the requirement of SLC18B1 to maintain polyamine pool in the brain [148]. There are many other transporters that have been implicated in polyamine effluxion [149], but extensive research is required to characterize these polyamines’ secretory pathways.

3.4. Catabolism of Polyamines

Polyamines are interconvertible, and several enzymes involved in these processes have been identified (Figure 1) [150,151,152,153]. Spermine oxidase (SMOX) selectively catalyzes spermine into spermidine and 3-aminopropanaldehyde [151]. Several SMOX isoforms resulting from alternative splicing have also been identified, although the functional consequence of these distinct isoforms is largely unknown. These isoforms share similar catalytic activities but differ in their localization [154,155,156,157,158]. There is also an alternative pathway through which spermine is converted into spermidine and spermidine into putrescine [153]. In this pathway, a less specific acetyltransferase SSAT adds acetyl groups to either spermine or spermidine [159,160]. The N1-acetylspermine (AcSpm) or N1-acetylspermidine (AcSpmd) is then catabolized into spermidine or putrescine, respectively, in the presence of the acetylpolyamine oxidase (PAOX) [156,161,162,163]. PAOX prefers to oxidize acetylated polyamines and exhibits negligible affinity towards spermidine and spermine [157]. The SMOX and SSAT/PAOX pathways work against the polyamine biosynthesis reactions to degrade the higher polyamines into putrescine, controlling the polyamines dynamics. Putrescine can be oxidized by the diamine oxidase [164,165] or excreted in poorly characterized manners [166]. In addition, the higher polyamines, as well as their acetylated derivatives, can also be secreted from cells [166,167,168,169].

3.5. Regulation of Polyamines by c-MYC

Polyamine metabolism is exquisitely regulated by c-MYC signaling [83]. The first piece of evidence for the interplay between polyamine and the c-MYC oncogene was revealed with the observation that ODC1 is a transcriptional target of c-MYC [170]. c-MYC-induced growth factor-independent transcription results in increased ODC1 protein expression and enhanced enzymatic activity, resulting in cell cycle progression and transformation [171,172,173,174,175]. A single nucleotide A/G polymorphism at position +317 in the human ODC1 gene relative to transcription initiation is located between the two E-boxes and may affect c-MYC binding and, therefore, ODC1 expression [176,177]. Moreover, ODC1 is a critical determinant of c-MYC oncogenesis and a potential therapeutic target in MYC-driven tumors [178].
In addition to ODC1, other genes in the polyamine metabolism pathway regulated by c-MYC include SRM, SMS, AMD1, and OAZ2, an ornithine decarboxylase antizyme [178,179,180]. c-MYC also indirectly modulates AdoMetDC/AMD1 expression by promoting mTOR activation [181,182,183], which stabilizes AdoMetDC [184]. Regarding polyamine catabolism, c-MYC has been demonstrated to repress the expression of SSAT and SMOX [178,179], thus maintaining high cellular polyamine levels.
There is no direct evidence for c-MYC-regulated polyamine transport, but some indirect evidence suggests that c-MYC promotes polyamine uptake. However, the polyamine transporter SLC3A2 is a direct transcriptional target of c-MYC [185,186].

4. Polyamines as Therapeutic Targets in MYC-Driven Ovarian Cancer

Polyamines act as critical regulators of tumor initiation, and progression and dysregulation of polyamine homeostasis are prevalent in various human cancers [20,187,188]. The potential application of targeting polyamine metabolism in cancer therapy remains an active area of intensive investigation in a variety of cancers, including ovarian cancer [20,187]. An overview of small molecular inhibitors targeting various aspects of polyamine metabolism is provided in Figure 1.

4.1. Targeting Polyamine Metabolism and Transport for Ovarian Cancer Treatment

The ODC1 inhibitor eflornithine (also known as α-difluoromethylornithine or DFMO) was developed in 1978 [189]. Early studies have investigated the cytotoxicity of DFMO in ovarian cancer cells [190], and a recent study demonstrated that DFMO induces apoptosis in ovarian cancer cells by regulating AP-1 signaling [188]. In addition to a stand-alone treatment modality, researchers have also explored the effect of DFMO in combination with other drugs. Together with the DNA methyl transferase inhibitor 5-azacytidine, DFMO increased the accumulation of anti-tumor M1 macrophages and reversed the immunosuppressive TME, promoting tumoricidal immune responses and prolonging the survival of mice with ovarian cancer [191]. In another study, DFMO synergistically enhanced the cytotoxicity of PARP inhibitors toward ovarian cancer cells in vitro [192].
AdoMetDC is essential to produce the aminopropyl moiety donor for the biosynthesis of polyamines. Targeting AdoMetDC can therefore interrupt polyamine homeostasis in cancer cells resulting in anti-cancer effects. Several AdoMetDC inhibitors have been developed, including methylglyoxal bis(guanylhydrazone) (MGBG)[193], 4-amidinoindan-1-one 2′amidinohydrazone (SAM486A) [194,195,196], as well as 5′(((z)-4-amino-20butenyl)methylamino)-5′deoxyadenosine (AbeAdo) and its 8-methyl derivative (Genz-644131) [187,193]. Evaluation of the anti-cancer efficacy of these small molecule inhibitors for the treatment of ovarian cancer has been limited, although earlier studies demonstrated that DFMO, in combination with AbeAdo, promoted G1 arrest in OVCAR-3 ovarian cancer cells [197].
The anti-cancer effects of small molecule inhibitors targeting de novo polyamine biosynthesis may be attenuated due to increased scavenging of extracellular polyamines by cancer cells. AMXT 1501 (D-Lys(C16acyl)-Spm) exhibited extraordinary cytotoxicity towards a variety of cancer types, including ovarian cancer, by interfering with the extracellular polyamine transport apparatus [187]. Moreover, the combination of DFMO and AMXT 1501 treatment also yielded exceptional tumoricidal efficacy [187]. A phase I clinical study has recently been initiated to assess the safety and dose level of AMXT 1501 alone and in combination with DFMO for the treatment of solid cancers, including ovarian cancer (NCT05500508). F14512, another polyamine transport blocker, demonstrated potent anti-proliferative and pro-apoptotic activities in vitro and in a mouse ovarian tumor model. Notably, the anti-cancer effects of F14512 were most profound in ovarian cancer cells that exhibited highly active polyamine transport systems [198]. A phase I clinical study using F14512 was conducted in patients with platinum-refractory or resistant ovarian cancer. However, this study was discontinued due to the occurrence of grade 4 neutropenia [199].

4.2. Exploiting Synthetic Polyamine Analogues to Deplete Polyamine Pools in Ovarian Cancer Cells for Anti-Cancer Treatment

Initial attempts have led to the generation of several analogs that are structurally similar to natural polyamines, and several additional analogs have gradually been introduced with an advanced understanding of polyamine metabolism and synthesis methods (Figure 1) [200,201,202]. The representative polyamine analogs that have been intensively studied in cancers include N1,N11-bis(ethyl)norspermine (BENSpm, also known as DENSpm), N1,N12-bis(ethyl)spermine (BESpm), N1,N12-bis(ethyl)-cis-6,7-dehydrospermine (PG-11047), and diethyl dihydroxyhomospermine (SBP-101). Mechanistic studies reveal that BENSpm and BESpm not only interfere with polyamine biosynthesis and transport but also drastically induce the polyamine catabolic enzymes SSAT and SMOX, resulting in the depletion of cellular polyamines and cancer cell death [203,204,205]. Intraperitoneal administration of BENSpm into A121 ovarian carcinoma tumor-bearing mice resulted in pronounced anti-cancer effects, with 40% of tumor-bearing mice being tumor-free following the intervention [206]. Mechanistically, BESpm treatment depleted ovarian cancer cell polyamine pools, partially through induction of SSAT, resulting in cancer cell death [207]. Furthermore, studies using ovarian cancer cell lines demonstrated that the combination of BENSpm and BESpm with chemotherapeutic drugs results in improved anti-cancer effects compared to chemotherapy alone [208,209,210]. SBP-101, unlike other polyamine analogs, had modest induction of polyamine catabolism but robustly repressed the activity of ODC1 in ovarian cancer cells [211]. More recently, a preclinical study reported that, through modulating polyamine metabolism and immunosuppressive microenvironment, SBP-101 marked prolonged the median survival of mice in a VDID8+ murine ovarian cancer model [212].
Early studies revealed that platinum-based drugs and polyamine analogs induced SSAT expression in the cisplatin-sensitive, but not the cisplatin-resistant, human ovarian cancer cells [213,214]. Other studies further demonstrated that treatment of ovarian cancer cells with cisplatin or oxaliplatin in combination with DENSpm results in enhanced anti-proliferative effects via enhancing SSAT expression [208,209,215]. These findings are consistent with a previous study that also reported increased sensitivity of ovarian cancer cells to cisplatin when SSAT1 was transiently overexpressed [216]. Notably, in these studies, other polyamines catabolism enzymes SSAT2, SMOX and PAOX were also induced, while ODC1 was downregulated [209,215].
The folate cycle inhibitor-induced SSAT expression results in the depletion of cellular polyamines with subsequent production of reactive oxygen species and cell death in ovarian cancer cell lines [210,217]. Moreover, folate cycle inhibitors, when combined with small molecular inhibitors of polyamine metabolism, were demonstrated to elicit synergistic cytotoxicity toward ovarian cancer cells [210]. This combinational treatment also increased chemosensitivity in drug-resistant ovarian cancer cells [210,217].

5. Utility of Polyamines as Biomarkers for Early Detection of Ovarian Cancer

Polyamines exert potent tumor-promoting activities, and their homeostasis is often dysregulated in cancers with increased biosynthesis and excretion, thus providing opportunities for early detection applications. Indeed, polyamines and their metabolites are reported as biomarkers for various cancers, including ovarian cancer (Table 1) [22,218,219,220].

5.1. Urinary Polyamines and Their Acetylated Derivates as Biomarkers for Early Detection of Ovarian Cancer

Increased urinary excretion of putrescine, spermidine, and spermine has been observed in ovarian cancer cases [221,222,226,227]. Putrescine, spermidine, and spermine are part of a urine-based biomarker test developed by Waalkes et al. for the detection of ovarian cancer [223]. In their study, the frequency of the three polyamines was markedly elevated in the urine of cases with advanced ovarian cancer compared to disease-free controls (42% vs. 13% for putrescine, 88% vs. 0% for spermidine, and 59% vs. 7% for spermine) [223]. Moreover, the urinary spermidine/creatinine ratio was drastically increased in advanced ovarian cancer patients [223]. Suh et al. evaluated free and acetylated urinary polyamines and found consistent increases in putrescine, spermidine, and spermine levels in the urine of ovarian cancer patients compared with controls [224]. Moreover, N-acetylputrescine (NAcPut) and AcSpmd were also concomitantly elevated in cases compared to healthy controls [224]. In an independent study, 14 urinary-free, mono- and di-acetylpolyamines were measured in patients with ovarian cancer and individuals presenting with benign disease. These studies revealed DAS, among other polyamines, to be able to distinguish malignant from benign masses [225]. Notably, DAS had better sensitivity (86.5%) but lower specificity (65.2%) than CA-125 (75.7% sensitivity, 69.6% specificity) [225].

5.2. Plasma Acetylated Polyamines for Risk Prediction of Malignancy for Ovarian Cancer

Polyamines have also been reported to be elevated in the blood of individuals with ovarian cancer [228,229]. Recently, Fahrmann et al. reported a plasma polyamine signature for the early detection of ovarian cancer [22]. Specifically, a logistic regression model based on DAS + N-(3-acetamidopropyl)pyrrolidin-2-one (N3AP) + CA125 was developed for the detection of ovarian cancer. At a >99% specificity threshold, the model yielded a sensitivity of 73.7% sensitivity in an independent validation set for the detection of early-stage ovarian cancers, which was markedly better than CA-125 alone (62.2% sensitivity; McNemar exact test 2-sided P: 0.019) [22]. In an independent study, Irajizad et al. developed a deep learning model based on a panel of seven cancer-associated metabolites, which included DAS, DiAcSpmd, and N3AP, for risk prediction of malignancy among women presenting with ovarian cysts. The 7-marker metabolite panel had an AUC of 0.86 for differentiating early-stage ovarian cancers from benign pelvic masses. The authors further demonstrated that the combination of the 7-marker metabolite panel with the risk of ovarian malignancy algorithm (ROMA) yielded a significantly higher positive predictive value (PPV) compared to ROMA alone (0.68 vs. 0.52) for early-stage ovarian cancer [220].

6. Conclusions

c-MYC and its paralogues are among the most frequently amplified and/or overexpressed oncoproteins in ovarian cancer. MYC family members contribute to cancer initiation and progression and are regulated at genomic, mRNA, and protein levels. The tumor-promoting polyamine pathway is tightly linked to c-MYC signaling and intertwines with the malignancy of ovarian cancer. Multiple strategies for inhibiting polyamine biosynthesis, metabolism, and transport have been explored in the context of ovarian cancer, with several small molecule inhibitors having been developed to target various aspects of polyamine metabolism. Preclinical investigations have shown considerable promise of polyamine metabolism as a metabolic vulnerability for the anti-cancer treatment of ovarian cancer. Clinical trials targeting polyamine metabolism using small molecule inhibitors for the treatment of solid malignancies, including ovarian cancer, are ongoing. The elevation of polyamines and their acetylated derivates in urine and plasmas of ovarian cancer cases provides additional opportunities for risk prediction and early detection applications and provides a potential means to better select individuals who may best benefit from polyamine targeting therapies.

Author Contributions

Conceptualization, Y.C., J.F.F. and S.H.; Draft preparation: Y.C., R.A.L.-L., A.H.A.S. and J.F.F.; Review and editing, Y.C., J.V., J.B.D., J.F.F. and S.H.; Supervision, J.F.F. and S.H. 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 interest.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2022. CA Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef]
  2. Meyer, N.; Penn, L.Z. Reflecting on 25 years with MYC. Nat. Rev. Cancer 2008, 8, 976–990. [Google Scholar] [CrossRef] [PubMed]
  3. Dang, C.V. MYC on the path to cancer. Cell 2012, 149, 22–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Kalkat, M.; De Melo, J.; Hickman, K.A.; Lourenco, C.; Redel, C.; Resetca, D.; Tamachi, A.; Tu, W.B.; Penn, L.Z. MYC Deregulation in Primary Human Cancers. Genes 2017, 8, 151. [Google Scholar] [CrossRef] [Green Version]
  5. Schaub, F.X.; Dhankani, V.; Berger, A.C.; Trivedi, M.; Richardson, A.B.; Shaw, R.; Zhao, W.; Zhang, X.; Ventura, A.; Liu, Y.; et al. Pan-cancer Alterations of the MYC Oncogene and Its Proximal Network across the Cancer Genome Atlas. Cell Syst. 2018, 6, 282–300.e282. [Google Scholar] [CrossRef] [Green Version]
  6. Wu, R.; Lin, L.; Beer, D.G.; Ellenson, L.H.; Lamb, B.J.; Rouillard, J.M.; Kuick, R.; Hanash, S.; Schwartz, D.R.; Fearon, E.R.; et al. Amplification and overexpression of the L-MYC proto-oncogene in ovarian carcinomas. Am. J. Pathol. 2003, 162, 1603–1610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Reyes-Gonzalez, J.M.; Vivas-Mejia, P.E. c-MYC and Epithelial Ovarian Cancer. Front. Oncol. 2021, 11, 601512. [Google Scholar] [CrossRef] [PubMed]
  8. Lourenco, C.; Resetca, D.; Redel, C.; Lin, P.; MacDonald, A.S.; Ciaccio, R.; Kenney, T.M.G.; Wei, Y.; Andrews, D.W.; Sunnerhagen, M.; et al. MYC protein interactors in gene transcription and cancer. Nat. Rev. Cancer 2021, 21, 579–591. [Google Scholar] [CrossRef]
  9. Amati, B.; Brooks, M.W.; Levy, N.; Littlewood, T.D.; Evan, G.I.; Land, H. Oncogenic activity of the c-Myc protein requires dimerization with Max. Cell 1993, 72, 233–245. [Google Scholar] [CrossRef]
  10. Thomas, L.R.; Wang, Q.; Grieb, B.C.; Phan, J.; Foshage, A.M.; Sun, Q.; Olejniczak, E.T.; Clark, T.; Dey, S.; Lorey, S.; et al. Interaction with WDR5 promotes target gene recognition and tumorigenesis by MYC. Mol. Cell 2015, 58, 440–452. [Google Scholar] [CrossRef]
  11. Wei, Y.; Resetca, D.; Li, Z.; Johansson-Åkhe, I.; Ahlner, A.; Helander, S.; Wallenhammar, A.; Morad, V.; Raught, B.; Wallner, B.; et al. Multiple direct interactions of TBP with the MYC oncoprotein. Nat. Struct. Mol. Biol. 2019, 26, 1035–1043. [Google Scholar] [CrossRef] [PubMed]
  12. Pineda-Lucena, A.; Ho, C.S.; Mao, D.Y.; Sheng, Y.; Laister, R.C.; Muhandiram, R.; Lu, Y.; Seet, B.T.; Katz, S.; Szyperski, T.; et al. A structure-based model of the c-Myc/Bin1 protein interaction shows alternative splicing of Bin1 and c-Myc phosphorylation are key binding determinants. J. Mol. Biol. 2005, 351, 182–194. [Google Scholar] [CrossRef]
  13. Richards, M.W.; Burgess, S.G.; Poon, E.; Carstensen, A.; Eilers, M.; Chesler, L.; Bayliss, R. Structural basis of N-Myc binding by Aurora-A and its destabilization by kinase inhibitors. Proc. Natl. Acad. Sci. USA 2016, 113, 13726–13731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Staller, P.; Peukert, K.; Kiermaier, A.; Seoane, J.; Lukas, J.; Karsunky, H.; Möröy, T.; Bartek, J.; Massagué, J.; Hänel, F.; et al. Repression of p15INK4b expression by Myc through association with Miz-1. Nat. Cell Biol. 2001, 3, 392–399. [Google Scholar] [CrossRef] [PubMed]
  15. Molander, C.; Hackzell, A.; Ohta, M.; Izumi, H.; Funa, K. Sp1 is a key regulator of the PDGF beta-receptor transcription. Mol. Biol. Rep. 2001, 28, 223–233. [Google Scholar] [CrossRef] [PubMed]
  16. Miller, D.M.; Thomas, S.D.; Islam, A.; Muench, D.; Sedoris, K. c-Myc and cancer metabolism. Clin. Cancer Res. 2012, 18, 5546–5553. [Google Scholar] [CrossRef] [Green Version]
  17. Dingar, D.; Tu, W.B.; Resetca, D.; Lourenco, C.; Tamachi, A.; De Melo, J.; Houlahan, K.E.; Kalkat, M.; Chan, P.K.; Boutros, P.C.; et al. MYC dephosphorylation by the PP1/PNUTS phosphatase complex regulates chromatin binding and protein stability. Nat. Commun. 2018, 9, 3502. [Google Scholar] [CrossRef] [Green Version]
  18. Dong, Y.; Tu, R.; Liu, H.; Qing, G. Regulation of cancer cell metabolism: Oncogenic MYC in the driver’s seat. Signal Transduct. Target. Ther. 2020, 5, 124. [Google Scholar] [CrossRef]
  19. Stine, Z.E.; Walton, Z.E.; Altman, B.J.; Hsieh, A.L.; Dang, C.V. MYC, Metabolism, and Cancer. Cancer Discov. 2015, 5, 1024–1039. [Google Scholar] [CrossRef] [Green Version]
  20. Casero, R.A., Jr.; Murray Stewart, T.; Pegg, A.E. Polyamine metabolism and cancer: Treatments, challenges and opportunities. Nat. Rev. Cancer 2018, 18, 681–695. [Google Scholar] [CrossRef]
  21. Pegg, A.E. Mammalian polyamine metabolism and function. IUBMB Life 2009, 61, 880–894. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Fahrmann, J.F.; Irajizad, E.; Kobayashi, M.; Vykoukal, J.; Dennison, J.B.; Murage, E.; Wu, R.; Long, J.P.; Do, K.A.; Celestino, J.; et al. A MYC-Driven Plasma Polyamine Signature for Early Detection of Ovarian Cancer. Cancers 2021, 13, 913. [Google Scholar] [CrossRef] [PubMed]
  23. Xu, J.; Chen, Y.; Olopade, O.I. MYC and Breast Cancer. Genes Cancer 2010, 1, 629–640. [Google Scholar] [CrossRef] [PubMed]
  24. Nesbit, C.E.; Tersak, J.M.; Prochownik, E.V. MYC oncogenes and human neoplastic disease. Oncogene 1999, 18, 3004–3016. [Google Scholar] [CrossRef] [Green Version]
  25. Cancer Genome Atlas Research, N. Integrated genomic analyses of ovarian carcinoma. Nature 2011, 474, 609–615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Berns, E.M.; Klijn, J.G.; Henzen-Logmans, S.C.; Rodenburg, C.J.; van der Burg, M.E.; Foekens, J.A. Receptors for hormones and growth factors and (onco)-gene amplification in human ovarian cancer. Int. J. Cancer 1992, 52, 218–224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Schreiber, G.; Dubeau, L. C-myc proto-oncogene amplification detected by polymerase chain reaction in archival human ovarian carcinomas. Am. J. Pathol. 1990, 137, 653–658. [Google Scholar] [PubMed]
  28. Katsaros, D.; Theillet, C.; Zola, P.; Louason, G.; Sanfilippo, B.; Isaia, E.; Arisio, R.; Giardina, G.; Sismondi, P. Concurrent abnormal expression of erbB-2, myc and ras genes is associated with poor outcome of ovarian cancer patients. Anticancer Res. 1995, 15, 1501–1510. [Google Scholar]
  29. Wang, Z.R.; Liu, W.; Smith, S.T.; Parrish, R.S.; Young, S.R. c-myc and chromosome 8 centromere studies of ovarian cancer by interphase FISH. Exp. Mol. Pathol. 1999, 66, 140–148. [Google Scholar] [CrossRef]
  30. Diebold, J.; Suchy, B.; Baretton, G.B.; Blasenbreu, S.; Meier, W.; Schmidt, M.; Rabes, H.; Lohrs, U. DNA ploidy and MYC DNA amplification in ovarian carcinomas. Correlation with p53 and bcl-2 expression, proliferative activity and prognosis. Virchows Arch. 1996, 429, 221–227. [Google Scholar] [CrossRef]
  31. Lee, Y.J.; Kim, D.; Kim, H.S.; Na, K.; Lee, J.Y.; Nam, E.J.; Kim, S.W.; Kim, S.; Kim, Y.T. Integrating a Next Generation Sequencing Panel into Clinical Practice in Ovarian Cancer. Yonsei Med. J. 2019, 60, 914–923. [Google Scholar] [CrossRef] [PubMed]
  32. Ross, J.S.; Ali, S.M.; Wang, K.; Palmer, G.; Yelensky, R.; Lipson, D.; Miller, V.A.; Zajchowski, D.; Shawver, L.K.; Stephens, P.J. Comprehensive genomic profiling of epithelial ovarian cancer by next generation sequencing-based diagnostic assay reveals new routes to targeted therapies. Gynecol. Oncol. 2013, 130, 554–559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Dimova, I.; Raitcheva, S.; Dimitrov, R.; Doganov, N.; Toncheva, D. Correlations between c-myc gene copy-number and clinicopathological parameters of ovarian tumours. Eur. J. Cancer 2006, 42, 674–679. [Google Scholar] [CrossRef] [PubMed]
  34. Baker, V.V.; Borst, M.P.; Dixon, D.; Hatch, K.D.; Shingleton, H.M.; Miller, D. c-myc amplification in ovarian cancer. Gynecol. Oncol. 1990, 38, 340–342. [Google Scholar] [CrossRef] [PubMed]
  35. Bian, M.; Fan, Q.; Huang, S.; Ma, J.; Lang, J. Amplifications of proto-oncogenes in ovarian carcinoma. Chin. Med. J. 1995, 108, 844–848. [Google Scholar] [PubMed]
  36. Sasano, H.; Garrett, C.T.; Wilkinson, D.S.; Silverberg, S.; Comerford, J.; Hyde, J. Protooncogene amplification and tumor ploidy in human ovarian neoplasms. Hum. Pathol. 1990, 21, 382–391. [Google Scholar] [CrossRef]
  37. Zhou, D.J.; Gonzalez-Cadavid, N.; Ahuja, H.; Battifora, H.; Moore, G.E.; Cline, M.J. A unique pattern of proto-oncogene abnormalities in ovarian adenocarcinomas. Cancer 1988, 62, 1573–1576. [Google Scholar] [CrossRef]
  38. Helland, A.; Anglesio, M.S.; George, J.; Cowin, P.A.; Johnstone, C.N.; House, C.M.; Sheppard, K.E.; Etemadmoghadam, D.; Melnyk, N.; Rustgi, A.K.; et al. Deregulation of MYCN, LIN28B and LET7 in a molecular subtype of aggressive high-grade serous ovarian cancers. PLoS ONE 2011, 6, e18064. [Google Scholar] [CrossRef] [Green Version]
  39. Jung, M.; Russell, A.J.; Kennedy, C.; Gifford, A.J.; Australian Ovarian Cancer Study, G.; Mallitt, K.A.; Sivarajasingam, S.; Bowtell, D.D.; DeFazio, A.; Haber, M.; et al. Clinical Importance of Myc Family Oncogene Aberrations in Epithelial Ovarian Cancer. JNCI Cancer Spectr. 2018, 2, pky047. [Google Scholar] [CrossRef] [PubMed]
  40. Skírnisdóttir, I.A.; Sorbe, B.; Lindborg, K.; Seidal, T. Prognostic impact of p53, p27, and C-MYC on clinicopathological features and outcome in early-stage (FIGO I-II) epithelial ovarian cancer. Int. J. Gynecol. Cancer 2011, 21, 236–244. [Google Scholar] [CrossRef]
  41. Slamon, D.J.; deKernion, J.B.; Verma, I.M.; Cline, M.J. Expression of cellular oncogenes in human malignancies. Science 1984, 224, 256–262. [Google Scholar] [CrossRef] [PubMed]
  42. Yamamoto, A.; Kurata, M.; Yamamoto, K.; Nogawa, D.; Inoue, M.; Ishibashi, S.; Ikeda, M.; Miyasaka, N.; Kitagawa, M. High amplification of PVT1 and MYC predict favorable prognosis in early ovarian carcinoma. Pathol. Res. Pract. 2020, 216, 153175. [Google Scholar] [CrossRef] [PubMed]
  43. Ning, Y.X.; Luo, X.; Xu, M.; Feng, X.; Wang, J. Let-7d increases ovarian cancer cell sensitivity to a genistein analog by targeting c-Myc. Oncotarget 2017, 8, 74836–74845. [Google Scholar] [CrossRef] [Green Version]
  44. Jung, M.; Gao, J.; Cheung, L.; Bongers, A.; Somers, K.; Clifton, M.; Ramsay, E.E.; Russell, A.J.; Valli, E.; Gifford, A.J.; et al. ABCC4/MRP4 contributes to the aggressiveness of Myc-associated epithelial ovarian cancer. Int. J. Cancer 2020, 147, 2225–2238. [Google Scholar] [CrossRef]
  45. Allen-Petersen, B.L.; Sears, R.C. Mission Possible: Advances in MYC Therapeutic Targeting in Cancer. BioDrugs 2019, 33, 539–553. [Google Scholar] [CrossRef] [Green Version]
  46. Sears, R.; Nuckolls, F.; Haura, E.; Taya, Y.; Tamai, K.; Nevins, J.R. Multiple Ras-dependent phosphorylation pathways regulate Myc protein stability. Genes Dev. 2000, 14, 2501–2514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Farrell, A.S.; Joly, M.M.; Allen-Petersen, B.L.; Worth, P.J.; Lanciault, C.; Sauer, D.; Link, J.; Pelz, C.; Heiser, L.M.; Morton, J.P.; et al. MYC regulates ductal-neuroendocrine lineage plasticity in pancreatic ductal adenocarcinoma associated with poor outcome and chemoresistance. Nat. Commun. 2017, 8, 1728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Wang, X.; Cunningham, M.; Zhang, X.; Tokarz, S.; Laraway, B.; Troxell, M.; Sears, R.C. Phosphorylation regulates c-Myc’s oncogenic activity in the mammary gland. Cancer Res. 2011, 71, 925–936. [Google Scholar] [CrossRef] [Green Version]
  49. Arnold, H.K.; Sears, R.C. Protein phosphatase 2A regulatory subunit B56alpha associates with c-myc and negatively regulates c-myc accumulation. Mol. Cell. Biol. 2006, 26, 2832–2844. [Google Scholar] [CrossRef] [Green Version]
  50. Kauko, O.; O’Connor, C.M.; Kulesskiy, E.; Sangodkar, J.; Aakula, A.; Izadmehr, S.; Yetukuri, L.; Yadav, B.; Padzik, A.; Laajala, T.D.; et al. PP2A inhibition is a druggable MEK inhibitor resistance mechanism in KRAS-mutant lung cancer cells. Sci. Transl. Med. 2018, 10, eaaq1093. [Google Scholar] [CrossRef] [Green Version]
  51. Reavie, L.; Buckley, S.M.; Loizou, E.; Takeishi, S.; Aranda-Orgilles, B.; Ndiaye-Lobry, D.; Abdel-Wahab, O.; Ibrahim, S.; Nakayama, K.I.; Aifantis, I. Regulation of c-Myc ubiquitination controls chronic myelogenous leukemia initiation and progression. Cancer Cell 2013, 23, 362–375. [Google Scholar] [CrossRef]
  52. Ruvolo, P.P. The broken “Off” switch in cancer signaling: PP2A as a regulator of tumorigenesis, drug resistance, and immune surveillance. BBA Clin. 2016, 6, 87–99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Wu, Y.; Deng, Y.; Zhu, J.; Duan, Y.; Weng, W.; Wu, X. Pim1 promotes cell proliferation and regulates glycolysis via interaction with MYC in ovarian cancer. OncoTargets Ther. 2018, 11, 6647–6656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Islam, S.; Dutta, P.; Sahay, O.; Gopalakrishnan, K.; Muhury, S.R.; Parameshwar, P.; Shetty, P.; Santra, M.K. Feedback-regulated transcriptional repression of FBXO31 by c-Myc triggers ovarian cancer tumorigenesis. Int. J. Cancer 2022, 150, 1512–1524. [Google Scholar] [CrossRef]
  55. Reyes-Gonzalez, J.M.; Armaiz-Pena, G.N.; Mangala, L.S.; Valiyeva, F.; Ivan, C.; Pradeep, S.; Echevarria-Vargas, I.M.; Rivera-Reyes, A.; Sood, A.K.; Vivas-Mejia, P.E. Targeting c-MYC in Platinum-Resistant Ovarian Cancer. Mol. Cancer Ther. 2015, 14, 2260–2269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Chen, C.H.; Shen, J.; Lee, W.J.; Chow, S.N. Overexpression of cyclin D1 and c-Myc gene products in human primary epithelial ovarian cancer. Int. J. Gynecol. Cancer 2005, 15, 878–883. [Google Scholar] [CrossRef]
  57. Li, X.S.; Sun, J.; He, X.L. Expression of c-myc and mutation of the KRAS gene in patients with ovarian mucinous tumors. Genet. Mol. Res. 2015, 14, 10752–10759. [Google Scholar] [CrossRef]
  58. Darcy, K.M.; Brady, W.E.; Blancato, J.K.; Dickson, R.B.; Hoskins, W.J.; McGuire, W.P.; Birrer, M.J. Prognostic relevance of c-MYC gene amplification and polysomy for chromosome 8 in suboptimally-resected, advanced stage epithelial ovarian cancers: A Gynecologic Oncology Group study. Gynecol. Oncol. 2009, 114, 472–479. [Google Scholar] [CrossRef]
  59. Tanner, B.; Hengstler, J.G.; Luch, A.; Meinert, R.; Kreutz, E.; Arand, M.; Wilkens, C.; Hofmann, M.; Oesch, F.; Knapstein, P.G.; et al. C-myc mRNA expression in epithelial ovarian carcinomas in relation to estrogen receptor status, metastatic spread, survival time, FIGO stage, and histologic grade and type. Int. J. Gynecol. Pathol. 1998, 17, 66–74. [Google Scholar] [CrossRef] [Green Version]
  60. Curling, M.; Stenning, S.; Hudson, C.N.; Watson, J.V. Multivariate analyses of DNA index, p62c-myc, and clinicopathological status of patients with ovarian cancer. J. Clin. Pathol. 1998, 51, 455–461. [Google Scholar] [CrossRef] [Green Version]
  61. Huang, H.; Weng, H.; Zhou, H.; Qu, L. Attacking c-Myc: Targeted and combined therapies for cancer. Curr. Pharm. Des. 2014, 20, 6543–6554. [Google Scholar] [CrossRef] [PubMed]
  62. Dhanasekaran, R.; Deutzmann, A.; Mahauad-Fernandez, W.D.; Hansen, A.S.; Gouw, A.M.; Felsher, D.W. The MYC oncogene—the grand orchestrator of cancer growth and immune evasion. Nat. Rev. Clin. Oncol. 2022, 19, 23–36. [Google Scholar] [CrossRef]
  63. Felsher, D.W. MYC Inactivation Elicits Oncogene Addiction through Both Tumor Cell-Intrinsic and Host-Dependent Mechanisms. Genes Cancer 2010, 1, 597–604. [Google Scholar] [CrossRef] [PubMed]
  64. Prathapam, T.; Aleshin, A.; Guan, Y.; Gray, J.W.; Martin, G.S. p27Kip1 mediates addiction of ovarian cancer cells to MYCC (c-MYC) and their dependence on MYC paralogs. J. Biol. Chem. 2010, 285, 32529–32538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Zeng, M.; Kwiatkowski, N.P.; Zhang, T.; Nabet, B.; Xu, M.; Liang, Y.; Quan, C.; Wang, J.; Hao, M.; Palakurthi, S.; et al. Targeting MYC dependency in ovarian cancer through inhibition of CDK7 and CDK12/13. Elife 2018, 7, e39030. [Google Scholar] [CrossRef]
  66. Chen, H.; Liu, H.; Qing, G. Targeting oncogenic Myc as a strategy for cancer treatment. Signal Transduct. Target. Ther. 2018, 3, 5. [Google Scholar] [CrossRef] [Green Version]
  67. Llombart, V.; Mansour, M.R. Therapeutic targeting of “undruggable” MYC. EBioMedicine 2022, 75, 103756. [Google Scholar] [CrossRef]
  68. Wang, C.; Zhang, J.; Yin, J.; Gan, Y.; Xu, S.; Gu, Y.; Huang, W. Alternative approaches to target Myc for cancer treatment. Signal Transduct. Target. Ther. 2021, 6, 117. [Google Scholar] [CrossRef]
  69. Wolf, E.; Eilers, M. Targeting MYC Proteins for Tumor Therapy. Annu. Rev. Cancer Biol. 2020, 4, 61–75. [Google Scholar] [CrossRef] [Green Version]
  70. Fei, R.; Shaoyang, L. Combination antigene therapy targeting c-myc and c-erbB(2) in the ovarian cancer COC(1) cell line. Gynecol. Oncol. 2002, 85, 40–44. [Google Scholar] [CrossRef]
  71. Helm, C.W.; Shrestha, K.; Thomas, S.; Shingleton, H.M.; Miller, D.M. A unique c-myc-targeted triplex-forming oligonucleotide inhibits the growth of ovarian and cervical carcinomas in vitro. Gynecol. Oncol. 1993, 49, 339–343. [Google Scholar] [CrossRef]
  72. Hudson, C.D.; Savadelis, A.; Nagaraj, A.B.; Joseph, P.; Avril, S.; DiFeo, A.; Avril, N. Altered glutamine metabolism in platinum resistant ovarian cancer. Oncotarget 2016, 7, 41637–41649. [Google Scholar] [CrossRef] [Green Version]
  73. Wang, J.; Ma, X.; Jones, H.M.; Chan, L.L.; Song, F.; Zhang, W.; Bae-Jump, V.L.; Zhou, C. Evaluation of the antitumor effects of c-Myc-Max heterodimerization inhibitor 100258-F4 in ovarian cancer cells. J. Transl. Med. 2014, 12, 226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Baratta, M.G.; Schinzel, A.C.; Zwang, Y.; Bandopadhayay, P.; Bowman-Colin, C.; Kutt, J.; Curtis, J.; Piao, H.; Wong, L.C.; Kung, A.L.; et al. An in-tumor genetic screen reveals that the BET bromodomain protein, BRD4, is a potential therapeutic target in ovarian carcinoma. Proc. Natl. Acad. Sci. USA 2015, 112, 232–237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Li, C.; Bonazzoli, E.; Bellone, S.; Choi, J.; Dong, W.; Menderes, G.; Altwerger, G.; Han, C.; Manzano, A.; Bianchi, A.; et al. Mutational landscape of primary, metastatic, and recurrent ovarian cancer reveals c-MYC gains as potential target for BET inhibitors. Proc. Natl. Acad. Sci. USA 2019, 116, 619–624. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Bagratuni, T.; Mavrianou, N.; Gavalas, N.G.; Tzannis, K.; Arapinis, C.; Liontos, M.; Christodoulou, M.I.; Thomakos, N.; Haidopoulos, D.; Rodolakis, A.; et al. JQ1 inhibits tumour growth in combination with cisplatin and suppresses JAK/STAT signalling pathway in ovarian cancer. Eur. J. Cancer 2020, 126, 125–135. [Google Scholar] [CrossRef]
  77. Xu, B.; Lefringhouse, J.; Liu, Z.; West, D.; Baldwin, L.A.; Ou, C.; Chen, L.; Napier, D.; Chaiswing, L.; Brewer, L.D.; et al. Inhibition of the integrin/FAK signaling axis and c-Myc synergistically disrupts ovarian cancer malignancy. Oncogenesis 2017, 6, e295. [Google Scholar] [CrossRef] [Green Version]
  78. Yi, J.; Liu, C.; Tao, Z.; Wang, M.; Jia, Y.; Sang, X.; Shen, L.; Xue, Y.; Jiang, K.; Luo, F.; et al. MYC status as a determinant of synergistic response to Olaparib and Palbociclib in ovarian cancer. EBioMedicine 2019, 43, 225–237. [Google Scholar] [CrossRef] [Green Version]
  79. Mahapatra, L.; Andruska, N.; Mao, C.; Le, J.; Shapiro, D.J. A Novel IMP1 Inhibitor, BTYNB, Targets c-Myc and Inhibits Melanoma and Ovarian Cancer Cell Proliferation. Transl. Oncol. 2017, 10, 818–827. [Google Scholar] [CrossRef]
  80. Madden, S.K.; de Araujo, A.D.; Gerhardt, M.; Fairlie, D.P.; Mason, J.M. Taking the Myc out of cancer: Toward therapeutic strategies to directly inhibit c-Myc. Mol. Cancer 2021, 20, 3. [Google Scholar] [CrossRef]
  81. Gabay, M.; Li, Y.; Felsher, D.W. MYC activation is a hallmark of cancer initiation and maintenance. Cold Spring Harb. Perspect. Med. 2014, 4, a014241. [Google Scholar] [CrossRef] [PubMed]
  82. Li, J.; Meng, Y.; Wu, X.; Sun, Y. Polyamines and related signaling pathways in cancer. Cancer Cell Int. 2020, 20, 539. [Google Scholar] [CrossRef] [PubMed]
  83. Flynn, A.T.; Hogarty, M.D. Myc, Oncogenic Protein Translation, and the Role of Polyamines. Med. Sci. 2018, 6, 41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Bachmann, A.S.; Geerts, D. Polyamine synthesis as a target of MYC oncogenes. J. Biol. Chem. 2018, 293, 18757–18769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Pegg, A.E.; Lockwood, D.H.; Williams-Ashman, H.G. Concentrations of putrescine and polyamines and their enzymic synthesis during androgen-induced prostatic growth. Biochem. J. 1970, 117, 17–31. [Google Scholar] [CrossRef] [Green Version]
  86. Raina, A.; Janne, J.; Siimes, M. Stimulation of polyamine synthesis in relation to nucleic acids in regenerating rat liver. Biochim. Biophys. Acta 1966, 123, 197–201. [Google Scholar] [CrossRef]
  87. Russell, D.; Snyder, S.H. Amine synthesis in rapidly growing tissues: Ornithine decarboxylase activity in regenerating rat liver, chick embryo, and various tumors. Proc. Natl. Acad. Sci. USA 1968, 60, 1420–1427. [Google Scholar] [CrossRef] [Green Version]
  88. Mamont, P.S.; Bohlen, P.; McCann, P.P.; Bey, P.; Schuber, F.; Tardif, C. Alpha-methyl ornithine, a potent competitive inhibitor of ornithine decarboxylase, blocks proliferation of rat hepatoma cells in culture. Proc. Natl. Acad. Sci. USA 1976, 73, 1626–1630. [Google Scholar] [CrossRef] [Green Version]
  89. Wu, G.Q.; Xu, Y.M.; Lau, A.T.Y. Recent insights into eukaryotic translation initiation factors 5A1 and 5A2 and their roles in human health and disease. Cancer Cell Int. 2020, 20, 142. [Google Scholar] [CrossRef]
  90. Pegg, A.E. Regulation of ornithine decarboxylase. J. Biol. Chem. 2006, 281, 14529–14532. [Google Scholar] [CrossRef] [Green Version]
  91. Lee, J.; Michael, A.J.; Martynowski, D.; Goldsmith, E.J.; Phillips, M.A. Phylogenetic diversity and the structural basis of substrate specificity in the beta/alpha-barrel fold basic amino acid decarboxylases. J. Biol. Chem. 2007, 282, 27115–27125. [Google Scholar] [CrossRef] [PubMed]
  92. Pegg, A.E.; Xiong, H.; Feith, D.J.; Shantz, L.M. S-adenosylmethionine decarboxylase: Structure, function and regulation by polyamines. Biochem. Soc. Trans. 1998, 26, 580–586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Bale, S.; Lopez, M.M.; Makhatadze, G.I.; Fang, Q.; Pegg, A.E.; Ealick, S.E. Structural basis for putrescine activation of human S-adenosylmethionine decarboxylase. Biochemistry 2008, 47, 13404–13417. [Google Scholar] [CrossRef] [PubMed]
  94. Ikeguchi, Y.; Bewley, M.C.; Pegg, A.E. Aminopropyltransferases: Function, structure and genetics. J. Biochem. 2006, 139, 1–9. [Google Scholar] [CrossRef]
  95. Wu, H.; Min, J.; Ikeguchi, Y.; Zeng, H.; Dong, A.; Loppnau, P.; Pegg, A.E.; Plotnikov, A.N. Structure and mechanism of spermidine synthases. Biochemistry 2007, 46, 8331–8339. [Google Scholar] [CrossRef]
  96. Wu, H.; Min, J.; Zeng, H.; McCloskey, D.E.; Ikeguchi, Y.; Loppnau, P.; Michael, A.J.; Pegg, A.E.; Plotnikov, A.N. Crystal structure of human spermine synthase: Implications of substrate binding and catalytic mechanism. J. Biol. Chem. 2008, 283, 16135–16146. [Google Scholar] [CrossRef] [Green Version]
  97. Larque, E.; Sabater-Molina, M.; Zamora, S. Biological significance of dietary polyamines. Nutrition 2007, 23, 87–95. [Google Scholar] [CrossRef]
  98. Ramos-Molina, B.; Queipo-Ortuno, M.I.; Lambertos, A.; Tinahones, F.J.; Penafiel, R. Dietary and Gut Microbiota Polyamines in Obesity- and Age-Related Diseases. Front. Nutr. 2019, 6, 24. [Google Scholar] [CrossRef] [Green Version]
  99. Tofalo, R.; Cocchi, S.; Suzzi, G. Polyamines and Gut Microbiota. Front. Nutr. 2019, 6, 16. [Google Scholar] [CrossRef] [Green Version]
  100. Munoz-Esparza, N.C.; Latorre-Moratalla, M.L.; Comas-Baste, O.; Toro-Funes, N.; Veciana-Nogues, M.T.; Vidal-Carou, M.C. Polyamines in Food. Front. Nutr. 2019, 6, 108. [Google Scholar] [CrossRef] [Green Version]
  101. Bard6cz, S.; Grant, G.; Brown, D.S.; Ralph, A.; Pusztai, A. Polyamines in food-implications for growth and health. J. Nutr. Biochem. 1993, 4, 66–71. [Google Scholar] [CrossRef]
  102. Romain, N.; Dandrifosse, G.; Jeusette, F.; Forget, P. Polyamine concentration in rat milk and food, human milk, and infant formulas. Pediatr. Res. 1992, 32, 58–63. [Google Scholar] [CrossRef] [Green Version]
  103. Bardocz, S. The role of dietary polyamines. Eur. J. Clin. Nutr. 1993, 47, 683–690. [Google Scholar]
  104. Coleman, C.S.; Hu, G.; Pegg, A.E. Putrescine biosynthesis in mammalian tissues. Biochem. J. 2004, 379, 849–855. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Sawada, Y.; Pereira, S.; Murphy, G.; Dowling, R. Origins of intestinal luminal polyamines in man. Gut 1994, 35, S20. [Google Scholar] [CrossRef] [Green Version]
  106. Sawada, Y.; Pereira, S.P.; Murphy, G.M.; Dowling, R.H. Polyamines in the intestinal lumen of patients with small bowel bacterial overgrowth. Biochem. Soc. Trans. 1994, 22, 392S. [Google Scholar] [CrossRef] [Green Version]
  107. Matsumoto, M.; Benno, Y. The relationship between microbiota and polyamine concentration in the human intestine: A pilot study. Microbiol. Immunol. 2007, 51, 25–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Soda, K.; Kano, Y.; Sakuragi, M.; Takao, K.; Lefor, A.; Konishi, F. Long-term oral polyamine intake increases blood polyamine concentrations. J. Nutr. Sci. Vitaminol. 2009, 55, 361–366. [Google Scholar] [CrossRef] [Green Version]
  109. Soda, K.; Uemura, T.; Sanayama, H.; Igarashi, K.; Fukui, T. Polyamine-Rich Diet Elevates Blood Spermine Levels and Inhibits Pro-Inflammatory Status: An Interventional Study. Med. Sci. 2021, 9, 22. [Google Scholar] [CrossRef] [PubMed]
  110. Benamouzig, R.; Mahe, S.; Luengo, C.; Rautureau, J.; Tome, D. Fasting and postprandial polyamine concentrations in the human digestive lumen. Am. J. Clin. Nutr. 1997, 65, 766–770. [Google Scholar] [CrossRef] [Green Version]
  111. Milovic, V.; Faust, D.; Turchanowa, L.; Stein, J.; Caspary, W.F. Permeability characteristics of polyamines across intestinal epithelium using the Caco-2 monolayer system: Comparison between transepithelial flux and mitogen-stimulated uptake into epithelial cells. Nutrition 2001, 17, 462–466. [Google Scholar] [CrossRef] [PubMed]
  112. Milovic, V.; Turchanowa, L.; Stein, J.; Caspary, W.F. Transepithelial transport of putrescine across monolayers of the human intestinal epithelial cell line, Caco-2. World J. Gastroenterol. 2001, 7, 193–197. [Google Scholar] [CrossRef] [PubMed]
  113. Sugiyama, Y.; Nara, M.; Sakanaka, M.; Gotoh, A.; Kitakata, A.; Okuda, S.; Kurihara, S. Comprehensive analysis of polyamine transport and biosynthesis in the dominant human gut bacteria: Potential presence of novel polyamine metabolism and transport genes. Int. J. Biochem. Cell Biol. 2017, 93, 52–61. [Google Scholar] [CrossRef] [PubMed]
  114. Uemura, T.; Stringer, D.E.; Blohm-Mangone, K.A.; Gerner, E.W. Polyamine transport is mediated by both endocytic and solute carrier transport mechanisms in the gastrointestinal tract. Am. J. Physiol.-Gastrointest. Liver Physiol. 2010, 299, G517–G522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Kibe, R.; Kurihara, S.; Sakai, Y.; Suzuki, H.; Ooga, T.; Sawaki, E.; Muramatsu, K.; Nakamura, A.; Yamashita, A.; Kitada, Y.; et al. Upregulation of colonic luminal polyamines produced by intestinal microbiota delays senescence in mice. Sci. Rep. 2014, 4, 4548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Seiler, N.; Sarhan, S.; Grauffel, C.; Jones, R.; Knodgen, B.; Moulinoux, J.P. Endogenous and exogenous polyamines in support of tumor growth. Cancer Res. 1990, 50, 5077–5083. [Google Scholar]
  117. Mantziari, A.; Mannila, E.; Collado, M.C.; Salminen, S.; Gomez-Gallego, C. Exogenous Polyamines Influence In Vitro Microbial Adhesion to Human Mucus According to the Age of Mucus Donor. Microorganisms 2021, 9, 1239. [Google Scholar] [CrossRef]
  118. Dai, F.; Yu, W.; Song, J.; Li, Q.; Wang, C.; Xie, S. Extracellular polyamines-induced proliferation and migration of cancer cells by ODC, SSAT, and Akt1-mediated pathway. Anticancer Drugs 2017, 28, 457–464. [Google Scholar] [CrossRef]
  119. Wu, R.; Chen, X.; Kang, S.; Wang, T.; Gnanaprakasam, J.R.; Yao, Y.; Liu, L.; Fan, G.; Burns, M.R.; Wang, R. De novo synthesis and salvage pathway coordinately regulate polyamine homeostasis and determine T cell proliferation and function. Sci. Adv. 2020, 6, eabc4275. [Google Scholar] [CrossRef]
  120. McCubbrey, A.L.; McManus, S.A.; McClendon, J.D.; Thomas, S.M.; Chatwin, H.B.; Reisz, J.A.; D’Alessandro, A.; Mould, K.J.; Bratton, D.L.; Henson, P.M.; et al. Polyamine import and accumulation causes immunomodulation in macrophages engulfing apoptotic cells. Cell Rep. 2022, 38, 110222. [Google Scholar] [CrossRef]
  121. Malpica-Nieves, C.J.; Rivera-Aponte, D.E.; Tejeda-Bayron, F.A.; Mayor, A.M.; Phanstiel, O.; Veh, R.W.; Eaton, M.J.; Skatchkov, S.N. The involvement of polyamine uptake and synthesis pathways in the proliferation of neonatal astrocytes. Amino Acids 2020, 52, 1169–1180. [Google Scholar] [CrossRef] [PubMed]
  122. Dot, J.; Lluch, M.; Blanco, I.; Rodriguez-Alvarez, J. Polyamine uptake in cultured astrocytes: Characterization and modulation by protein kinases. J. Neurochem. 2000, 75, 1917–1926. [Google Scholar] [CrossRef] [PubMed]
  123. Welch, J.E.; Bengtson, P.; Svensson, K.; Wittrup, A.; Jenniskens, G.J.; Ten Dam, G.B.; Van Kuppevelt, T.H.; Belting, M. Single chain fragment anti-heparan sulfate antibody targets the polyamine transport system and attenuates polyamine-dependent cell proliferation. Int. J. Oncol. 2008, 32, 749–756. [Google Scholar] [PubMed]
  124. Wang, S.; Qiu, Y.; Bai, B. The Expression, Regulation, and Biomarker Potential of Glypican-1 in Cancer. Front. Oncol. 2019, 9, 614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Belting, M.; Mani, K.; Jonsson, M.; Cheng, F.; Sandgren, S.; Jonsson, S.; Ding, K.; Delcros, J.G.; Fransson, L.A. Glypican-1 is a vehicle for polyamine uptake in mammalian cells: A pivital role for nitrosothiol-derived nitric oxide. J. Biol. Chem. 2003, 278, 47181–47189. [Google Scholar] [CrossRef] [Green Version]
  126. Soulet, D.; Gagnon, B.; Rivest, S.; Audette, M.; Poulin, R. A fluorescent probe of polyamine transport accumulates into intracellular acidic vesicles via a two-step mechanism. J. Biol. Chem. 2004, 279, 49355–49366. [Google Scholar] [CrossRef] [Green Version]
  127. Roy, U.K.; Rial, N.S.; Kachel, K.L.; Gerner, E.W. Activated K-RAS increases polyamine uptake in human colon cancer cells through modulation of caveolar endocytosis. Mol. Carcinog. 2008, 47, 538–553. [Google Scholar] [CrossRef] [Green Version]
  128. Uemura, T.; Yerushalmi, H.F.; Tsaprailis, G.; Stringer, D.E.; Pastorian, K.E.; Hawel, L., 3rd; Byus, C.V.; Gerner, E.W. Identification and characterization of a diamine exporter in colon epithelial cells. J. Biol. Chem. 2008, 283, 26428–26435. [Google Scholar] [CrossRef] [Green Version]
  129. Gamble, L.D.; Purgato, S.; Murray, J.; Xiao, L.; Yu, D.M.T.; Hanssen, K.M.; Giorgi, F.M.; Carter, D.R.; Gifford, A.J.; Valli, E.; et al. Inhibition of polyamine synthesis and uptake reduces tumor progression and prolongs survival in mouse models of neuroblastoma. Sci. Transl. Med. 2019, 11, eaau1099. [Google Scholar] [CrossRef] [Green Version]
  130. Sekhar, V.; Andl, T.; Phanstiel, O.t. ATP13A3 facilitates polyamine transport in human pancreatic cancer cells. Sci. Rep. 2022, 12, 4045. [Google Scholar] [CrossRef]
  131. Heinick, A.; Urban, K.; Roth, S.; Spies, D.; Nunes, F.; Phanstiel, O.t.; Liebau, E.; Luersen, K. Caenorhabditis elegans P5B-type ATPase CATP-5 operates in polyamine transport and is crucial for norspermidine-mediated suppression of RNA interference. FASEB J. 2010, 24, 206–217. [Google Scholar] [CrossRef] [Green Version]
  132. De La Hera, D.P.; Corradi, G.R.; Adamo, H.P.; De Tezanos Pinto, F. Parkinson’s disease-associated human P5B-ATPase ATP13A2 increases spermidine uptake. Biochem. J. 2013, 450, 47–53. [Google Scholar] [CrossRef] [PubMed]
  133. Kashiwagi, K.; Miyamoto, S.; Suzuki, F.; Kobayashi, H.; Igarashi, K. Excretion of putrescine by the putrescine-ornithine antiporter encoded by the potE gene of Escherichia coli. Proc. Natl. Acad. Sci. USA 1992, 89, 4529–4533. [Google Scholar] [CrossRef]
  134. Woolridge, D.P.; Vazquez-Laslop, N.; Markham, P.N.; Chevalier, M.S.; Gerner, E.W.; Neyfakh, A.A. Efflux of the natural polyamine spermidine facilitated by the Bacillus subtilis multidrug transporter Blt. J. Biol. Chem. 1997, 272, 8864–8866. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Soksawatmaekhin, W.; Kuraishi, A.; Sakata, K.; Kashiwagi, K.; Igarashi, K. Excretion and uptake of cadaverine by CadB and its physiological functions in Escherichia coli. Mol. Microbiol. 2004, 51, 1401–1412. [Google Scholar] [CrossRef] [PubMed]
  136. Higashi, K.; Ishigure, H.; Demizu, R.; Uemura, T.; Nishino, K.; Yamaguchi, A.; Kashiwagi, K.; Igarashi, K. Identification of a spermidine excretion protein complex (MdtJI) in Escherichia coli. J. Bacteriol. 2008, 190, 872–878. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Tomitori, H.; Kashiwagi, K.; Sakata, K.; Kakinuma, Y.; Igarashi, K. Identification of a gene for a polyamine transport protein in yeast. J. Biol. Chem. 1999, 274, 3265–3267. [Google Scholar] [CrossRef] [Green Version]
  138. Tomitori, H.; Kashiwagi, K.; Asakawa, T.; Kakinuma, Y.; Michael, A.J.; Igarashi, K. Multiple polyamine transport systems on the vacuolar membrane in yeast. Biochem. J. 2001, 353, 681–688. [Google Scholar] [CrossRef]
  139. Hasne, M.P.; Ullman, B. Identification and characterization of a polyamine permease from the protozoan parasite Leishmania major. J. Biol. Chem. 2005, 280, 15188–15194. [Google Scholar] [CrossRef] [Green Version]
  140. Uemura, T.; Tachihara, K.; Tomitori, H.; Kashiwagi, K.; Igarashi, K. Characteristics of the polyamine transporter TPO1 and regulation of its activity and cellular localization by phosphorylation. J. Biol. Chem. 2005, 280, 9646–9652. [Google Scholar] [CrossRef] [Green Version]
  141. Tachihara, K.; Uemura, T.; Kashiwagi, K.; Igarashi, K. Excretion of putrescine and spermidine by the protein encoded by YKL174c (TPO5) in Saccharomyces cerevisiae. J. Biol. Chem. 2005, 280, 12637–12642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Tjandrawinata, R.R.; Hawel, L., 3rd; Byus, C.V. Characterization of putrescine and cadaverine export in mammalian cells. A pharmacological approach. Biochem. Pharmacol. 1994, 48, 2237–2249. [Google Scholar] [CrossRef] [PubMed]
  143. Hawel, L., 3rd; Tjandrawinata, R.R.; Fukumoto, G.H.; Byus, C.V. Biosynthesis and selective export of 1,5-diaminopentane (cadaverine) in mycoplasma-free cultured mammalian cells. J. Biol. Chem. 1994, 269, 7412–7418. [Google Scholar] [CrossRef] [PubMed]
  144. Tjandrawinata, R.R.; Hawel, L., 3rd; Byus, C.V. Regulation of putrescine export in lipopolysaccharide or IFN-gamma-activated murine monocytic-leukemic RAW 264 cells. J. Immunol. 1994, 152, 3039–3052. [Google Scholar] [CrossRef]
  145. Hawel, L., 3rd; Tjandrawinata, R.R.; Byus, C.V. Selective putrescine export is regulated by insulin and ornithine in Reuber H35 hepatoma cells. Biochim. Biophys. Acta 1994, 1222, 15–26. [Google Scholar] [CrossRef]
  146. Coleman, C.S.; Wallace, H.M. Polyamine excretion from human cancer cells. Biochem. Soc. Trans. 1990, 18, 1228–1229. [Google Scholar] [CrossRef] [Green Version]
  147. Moriyama, Y.; Hatano, R.; Moriyama, S.; Uehara, S. Vesicular polyamine transporter as a novel player in amine-mediated chemical transmission. Biochim. Biophys. Acta-Biomembr. 2020, 1862, 183208. [Google Scholar] [CrossRef]
  148. Fredriksson, R.; Sreedharan, S.; Nordenankar, K.; Alsio, J.; Lindberg, F.A.; Hutchinson, A.; Eriksson, A.; Roshanbin, S.; Ciuculete, D.M.; Klockars, A.; et al. The polyamine transporter Slc18b1(VPAT) is important for both short and long time memory and for regulation of polyamine content in the brain. PLoS Genet. 2019, 15, e1008455. [Google Scholar] [CrossRef] [Green Version]
  149. Abdulhussein, A.A.; Wallace, H.M. Polyamines and membrane transporters. Amino Acids 2014, 46, 655–660. [Google Scholar] [CrossRef]
  150. Seiler, N. Catabolism of polyamines. Amino Acids 2004, 26, 217–233. [Google Scholar] [CrossRef]
  151. Wang, Y.; Casero, R.A., Jr. Mammalian polyamine catabolism: A therapeutic target, a pathological problem, or both? J. Biochem. 2006, 139, 17–25. [Google Scholar] [CrossRef] [PubMed]
  152. Yuan, F.; Zhang, L.; Cao, Y.; Gao, W.; Zhao, C.; Fang, Y.; Zahedi, K.; Soleimani, M.; Lu, X.; Fang, Z.; et al. Spermidine/spermine N1-acetyltransferase-mediated polyamine catabolism regulates beige adipocyte biogenesis. Metabolism 2018, 85, 298–304. [Google Scholar] [CrossRef] [PubMed]
  153. Pegg, A.E. Spermidine/spermine-N1-acetyltransferase: A key metabolic regulator. Am. J. Physiol.-Endocrinol. Metab. 2008, 294, E995–E1010. [Google Scholar] [CrossRef]
  154. Wang, Y.; Devereux, W.; Woster, P.M.; Stewart, T.M.; Hacker, A.; Casero, R.A., Jr. Cloning and characterization of a human polyamine oxidase that is inducible by polyamine analogue exposure. Cancer Res. 2001, 61, 5370–5373. [Google Scholar] [PubMed]
  155. Cervelli, M.; Polticelli, F.; Federico, R.; Mariottini, P. Heterologous expression and characterization of mouse spermine oxidase. J. Biol. Chem. 2003, 278, 5271–5276. [Google Scholar] [CrossRef] [Green Version]
  156. Vujcic, S.; Liang, P.; Diegelman, P.; Kramer, D.L.; Porter, C.W. Genomic identification and biochemical characterization of the mammalian polyamine oxidase involved in polyamine back-conversion. Biochem. J. 2003, 370, 19–28. [Google Scholar] [CrossRef]
  157. Wu, T.; Yankovskaya, V.; McIntire, W.S. Cloning, sequencing, and heterologous expression of the murine peroxisomal flavoprotein, N1-acetylated polyamine oxidase. J. Biol. Chem. 2003, 278, 20514–20525. [Google Scholar] [CrossRef] [Green Version]
  158. Murray-Stewart, T.; Wang, Y.; Goodwin, A.; Hacker, A.; Meeker, A.; Casero, R.A., Jr. Nuclear localization of human spermine oxidase isoforms—Possible implications in drug response and disease etiology. FEBS J. 2008, 275, 2795–2806. [Google Scholar] [CrossRef]
  159. Matsui, I.; Wiegand, L.; Pegg, A.E. Properties of spermidine N-acetyltransferase from livers of rats treated with carbon tetrachloride and its role in the conversion of spermidine into putrescine. J. Biol. Chem. 1981, 256, 2454–2459. [Google Scholar] [CrossRef]
  160. Holtta, E.; Sinervirta, R.; Janne, J. Synthesis and accumulation of polyamines in rat liver regenerating after treatment with carbon tetrachloride. Biochem. Biophys. Res. Commun. 1973, 54, 350–357. [Google Scholar] [CrossRef]
  161. Takao, K.; Shibata, S.; Ozawa, T.; Wada, M.; Sugitia, Y.; Samejima, K.; Shirahata, A. A conceptual model of the polyamine binding site of N1-acetylpolyamine oxidase developed from a study of polyamine derivatives. Amino Acids 2009, 37, 401–405. [Google Scholar] [CrossRef] [PubMed]
  162. Henderson Pozzi, M.; Gawandi, V.; Fitzpatrick, P.F. pH dependence of a mammalian polyamine oxidase: Insights into substrate specificity and the role of lysine 315. Biochemistry 2009, 48, 1508–1516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Wang, Y.; Hacker, A.; Murray-Stewart, T.; Frydman, B.; Valasinas, A.; Fraser, A.V.; Woster, P.M.; Casero, R.A., Jr. Properties of recombinant human N1-acetylpolyamine oxidase (hPAO): Potential role in determining drug sensitivity. Cancer Chemother. Pharmacol. 2005, 56, 83–90. [Google Scholar] [CrossRef] [PubMed]
  164. Fogel, W.A.; Bieganski, T.; Schayer, R.W.; Maslinski, C. Involvement of diamine oxidase in catabolism of 14C-putrescine in mice in vivo with special reference to the formation of gamma-aminobutyric acid. Agents Actions 1981, 11, 679–684. [Google Scholar] [CrossRef] [PubMed]
  165. Equi, A.M.; Brown, A.M.; Cooper, A.; Ner, S.K.; Watson, A.B.; Robins, D.J. Oxidation of Putrescine and Cadaverine Derivatives by Diamine Oxidases. Tetrahedron 1991, 47, 507–518. [Google Scholar] [CrossRef]
  166. Wallace, H.M.; Keir, H.M. Factors affecting polyamine excretion from mammalian cells in culture. Inhibitors of polyamine biosynthesis. FEBS Lett. 1986, 194, 60–63. [Google Scholar] [CrossRef] [Green Version]
  167. Hiasa, M.; Miyaji, T.; Haruna, Y.; Takeuchi, T.; Harada, Y.; Moriyama, S.; Yamamoto, A.; Omote, H.; Moriyama, Y. Identification of a mammalian vesicular polyamine transporter. Sci. Rep. 2014, 4, 6836. [Google Scholar] [CrossRef] [Green Version]
  168. Hyvonen, T. Excretion of acetylated and free polyamines by polyamine depleted Chinese hamster ovary cells. Int. J. Biochem. 1989, 21, 313–316. [Google Scholar] [CrossRef]
  169. Hyvonen, T.; Alakuijala, L.; Andersson, L.; Khomutov, A.R.; Khomutov, R.M.; Eloranta, T.O. 1-Aminooxy-3-aminopropane reversibly prevents the proliferation of cultured baby hamster kidney cells by interfering with polyamine synthesis. J. Biol. Chem. 1988, 263, 11138–11144. [Google Scholar] [CrossRef]
  170. Bello-Fernandez, C.; Packham, G.; Cleveland, J.L. The ornithine decarboxylase gene is a transcriptional target of c-Myc. Proc. Natl. Acad. Sci. USA 1993, 90, 7804–7808. [Google Scholar] [CrossRef] [Green Version]
  171. Auvinen, M.; Paasinen, A.; Andersson, L.C.; Holtta, E. Ornithine decarboxylase activity is critical for cell transformation. Nature 1992, 360, 355–358. [Google Scholar] [CrossRef] [PubMed]
  172. Bowlin, T.L.; McKown, B.J.; Sunkara, P.S. Ornithine decarboxylase induction and polyamine biosynthesis are required for the growth of interleukin-2- and interleukin-3-dependent cell lines. Cell. Immunol. 1986, 98, 341–350. [Google Scholar] [CrossRef]
  173. Brand, K. Role of ornithine decarboxylase on glycolytic enzyme induction during thymocyte proliferation. J. Biol. Chem. 1987, 262, 15232–15235. [Google Scholar] [CrossRef] [PubMed]
  174. Moshier, J.A.; Dosescu, J.; Skunca, M.; Luk, G.D. Transformation of NIH/3T3 cells by ornithine decarboxylase overexpression. Cancer Res. 1993, 53, 2618–2622. [Google Scholar]
  175. Heiskala, M.; Zhang, J.; Hayashi, S.; Holtta, E.; Andersson, L.C. Translocation of ornithine decarboxylase to the surface membrane during cell activation and transformation. EMBO J. 1999, 18, 1214–1222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Martinez, M.E.; O’Brien, T.G.; Fultz, K.E.; Babbar, N.; Yerushalmi, H.; Qu, N.; Guo, Y.; Boorman, D.; Einspahr, J.; Alberts, D.S.; et al. Pronounced reduction in adenoma recurrence associated with aspirin use and a polymorphism in the ornithine decarboxylase gene. Proc. Natl. Acad. Sci. USA 2003, 100, 7859–7864. [Google Scholar] [CrossRef] [Green Version]
  177. O’Brien, T.G.; Guo, Y.; Visvanathan, K.; Sciulli, J.; McLaine, M.; Helzlsouer, K.J.; Watkins-Bruner, D. Differences in ornithine decarboxylase and androgen receptor allele frequencies among ethnic groups. Mol. Carcinog. 2004, 41, 120–123. [Google Scholar] [CrossRef]
  178. Hogarty, M.D.; Norris, M.D.; Davis, K.; Liu, X.; Evageliou, N.F.; Hayes, C.S.; Pawel, B.; Guo, R.; Zhao, H.; Sekyere, E.; et al. ODC1 is a critical determinant of MYCN oncogenesis and a therapeutic target in neuroblastoma. Cancer Res. 2008, 68, 9735–9745. [Google Scholar] [CrossRef] [Green Version]
  179. Nilsson, J.A.; Keller, U.B.; Baudino, T.A.; Yang, C.; Norton, S.; Old, J.A.; Nilsson, L.M.; Neale, G.; Kramer, D.L.; Porter, C.W.; et al. Targeting ornithine decarboxylase in Myc-induced lymphomagenesis prevents tumor formation. Cancer Cell 2005, 7, 433–444. [Google Scholar] [CrossRef] [Green Version]
  180. Fahrmann, J.F.; Vykoukal, J.; Fleury, A.; Tripathi, S.; Dennison, J.B.; Murage, E.; Wang, P.; Yu, C.Y.; Capello, M.; Creighton, C.J.; et al. Association Between Plasma Diacetylspermine and Tumor Spermine Synthase With Outcome in Triple-Negative Breast Cancer. J. Natl. Cancer Inst. 2020, 112, 607–616. [Google Scholar] [CrossRef]
  181. Patterson, D.G.; Kania, A.K.; Price, M.J.; Rose, J.R.; Scharer, C.D.; Boss, J.M. An IRF4-MYC-mTORC1 Integrated Pathway Controls Cell Growth and the Proliferative Capacity of Activated B Cells during B Cell Differentiation In Vivo. J. Immunol. 2021, 207, 1798–1811. [Google Scholar] [CrossRef] [PubMed]
  182. Pourdehnad, M.; Truitt, M.L.; Siddiqi, I.N.; Ducker, G.S.; Shokat, K.M.; Ruggero, D. Myc and mTOR converge on a common node in protein synthesis control that confers synthetic lethality in Myc-driven cancers. Proc. Natl. Acad. Sci. USA 2013, 110, 11988–11993. [Google Scholar] [CrossRef] [Green Version]
  183. Liu, P.; Ge, M.; Hu, J.; Li, X.; Che, L.; Sun, K.; Cheng, L.; Huang, Y.; Pilo, M.G.; Cigliano, A.; et al. A functional mammalian target of rapamycin complex 1 signaling is indispensable for c-Myc-driven hepatocarcinogenesis. Hepatology 2017, 66, 167–181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Zabala-Letona, A.; Arruabarrena-Aristorena, A.; Martin-Martin, N.; Fernandez-Ruiz, S.; Sutherland, J.D.; Clasquin, M.; Tomas-Cortazar, J.; Jimenez, J.; Torres, I.; Quang, P.; et al. mTORC1-dependent AMD1 regulation sustains polyamine metabolism in prostate cancer. Nature 2017, 547, 109–113. [Google Scholar] [CrossRef]
  185. Kim, J.; Lee, J.H.; Iyer, V.R. Global identification of Myc target genes reveals its direct role in mitochondrial biogenesis and its E-box usage in vivo. PLoS ONE 2008, 3, e1798. [Google Scholar] [CrossRef] [Green Version]
  186. El Ansari, R.; Craze, M.L.; Diez-Rodriguez, M.; Nolan, C.C.; Ellis, I.O.; Rakha, E.A.; Green, A.R. The multifunctional solute carrier 3A2 (SLC3A2) confers a poor prognosis in the highly proliferative breast cancer subtypes. Br. J. Cancer 2018, 118, 1115–1122. [Google Scholar] [CrossRef] [PubMed]
  187. Murray-Stewart, T.R.; Woster, P.M.; Casero, R.A., Jr. Targeting polyamine metabolism for cancer therapy and prevention. Biochem. J. 2016, 473, 2937–2953. [Google Scholar] [CrossRef] [Green Version]
  188. Hwang, W.Y.; Park, W.H.; Suh, D.H.; Kim, K.; Kim, Y.B.; No, J.H. Difluoromethylornithine Induces Apoptosis through Regulation of AP-1 Signaling via JNK Phosphorylation in Epithelial Ovarian Cancer. Int. J. Mol. Sci. 2021, 22, 255. [Google Scholar] [CrossRef]
  189. Metcalf, B.W.; Bey, P.; Danzin, C.; Jung, M.J.; Casara, P.; Vevert, J.P. Catalytic irreversible inhibition of mammalian ornithine decarboxylase (E.C.4.1.1.17) by substrate and product analogs. J. Am. Chem. Soc. 1978, 100, 2551–2553. [Google Scholar] [CrossRef]
  190. Manetta, A.; Satyaswarcoop, P.G.; Podczaski, E.S.; Hamilton, T.; Ozols, R.F.; Mortel, R. Effect of alpha-difluoromethylornithine (DFMO) on the growth of human ovarian carcinoma. Eur. J. Gynaecol. Oncol. 1988, 9, 222–227. [Google Scholar]
  191. Travers, M.; Brown, S.M.; Dunworth, M.; Holbert, C.E.; Wiehagen, K.R.; Bachman, K.E.; Foley, J.R.; Stone, M.L.; Baylin, S.B.; Casero, R.A., Jr.; et al. DFMO and 5-Azacytidine Increase M1 Macrophages in the Tumor Microenvironment of Murine Ovarian Cancer. Cancer Res. 2019, 79, 3445–3454. [Google Scholar] [CrossRef] [PubMed]
  192. El Naggar, O.; Doyle, B.; Mariner, K.; Gilmour, S.K. Difluoromethylornithine (DFMO) Enhances the Cytotoxicity of PARP Inhibition in Ovarian Cancer Cells. Med. Sci. 2022, 10, 28. [Google Scholar] [CrossRef]
  193. Pegg, A.E. S-Adenosylmethionine decarboxylase. Essays Biochem. 2009, 46, 25–45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Millward, M.J.; Joshua, A.; Kefford, R.; Aamdal, S.; Thomson, D.; Hersey, P.; Toner, G.; Lynch, K. Multi-centre Phase II trial of the polyamine synthesis inhibitor SAM486A (CGP48664) in patients with metastatic melanoma. Investig. New Drugs 2005, 23, 253–256. [Google Scholar] [CrossRef]
  195. Pless, M.; Belhadj, K.; Menssen, H.D.; Kern, W.; Coiffier, B.; Wolf, J.; Herrmann, R.; Thiel, E.; Bootle, D.; Sklenar, I.; et al. Clinical efficacy, tolerability, and safety of SAM486A, a novel polyamine biosynthesis inhibitor, in patients with relapsed or refractory non-Hodgkin’s lymphoma: Results from a phase II multicenter study. Clin. Cancer Res. 2004, 10, 1299–1305. [Google Scholar] [CrossRef] [PubMed]
  196. Siu, L.L.; Rowinsky, E.K.; Hammond, L.A.; Weiss, G.R.; Hidalgo, M.; Clark, G.M.; Moczygemba, J.; Choi, L.; Linnartz, R.; Barbet, N.C.; et al. A phase I and pharmacokinetic study of SAM486A, a novel polyamine biosynthesis inhibitor, administered on a daily-times-five every-three-week schedule in patients with Advanced solid malignancies. Clin. Cancer Res. 2002, 8, 2157–2166. [Google Scholar]
  197. Desiderio, M.A.; Bergamaschi, D.; Mascellani, E.; De Feudis, P.; Erba, E.; D’Incalci, M. Treatment with inhibitors of polyamine biosynthesis, which selectively lower intracellular spermine, does not affect the activity of alkylating agents but antagonizes the cytotoxicity of DNA topoisomerase II inhibitors. Br. J. Cancer 1997, 75, 1028–1034. [Google Scholar] [CrossRef] [Green Version]
  198. Wang, C.; Delcros, J.G.; Biggerstaff, J.; Phanstiel, O.t. Synthesis and biological evaluation of N1-(anthracen-9-ylmethyl)triamines as molecular recognition elements for the polyamine transporter. J. Med. Chem 2003, 46, 2663–2671. [Google Scholar] [CrossRef]
  199. Leary, A.; Le Tourneau, C.; Varga, A.; Sablin, M.P.; Gomez-Roca, C.; Guilbaud, N.; Petain, A.; Pavlyuk, M.; Delord, J.P. Phase I dose-escalation study of F14512, a polyamine-vectorized topoisomerase II inhibitor, in patients with platinum-refractory or resistant ovarian cancer. Investig. New Drugs 2019, 37, 693–701. [Google Scholar] [CrossRef] [Green Version]
  200. Edwards, M.L.; Prakash, N.J.; Stemerick, D.M.; Sunkara, S.P.; Bitonti, A.J.; Davis, G.F.; Dumont, J.A.; Bey, P. Polyamine analogues with antitumor activity. J. Med. Chem. 1990, 33, 1369–1375. [Google Scholar] [CrossRef]
  201. Porter, C.W.; Bergeron, R.J. Regulation of polyamine biosynthetic activity by spermidine and spermine analogs--a novel antiproliferative strategy. Adv. Exp. Med. Biol. 1988, 250, 677–690. [Google Scholar] [CrossRef] [PubMed]
  202. Casero, R.A., Jr.; Woster, P.M. Recent advances in the development of polyamine analogues as antitumor agents. J. Med. Chem. 2009, 52, 4551–4573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Casero, R.A.; Pegg, A.E. Polyamine catabolism and disease. Biochem. J. 2009, 421, 323–338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Pledgie, A.; Huang, Y.; Hacker, A.; Zhang, Z.; Woster, P.M.; Davidson, N.E.; Casero, R.A., Jr. Spermine oxidase SMO(PAOh1), Not N1-acetylpolyamine oxidase PAO, is the primary source of cytotoxic H2O2 in polyamine analogue-treated human breast cancer cell lines. J. Biol. Chem. 2005, 280, 39843–39851. [Google Scholar] [CrossRef] [Green Version]
  205. Gabrielson, E.W.; Pegg, A.E.; Casero, R.A., Jr. The induction of spermidine/spermine N1-acetyltransferase (SSAT) is a common event in the response of human primary non-small cell lung carcinomas to exposure to the new antitumor polyamine analogue N1,N11-bis(ethyl)norspermine. Clin. Cancer Res. 1999, 5, 1638–1641. [Google Scholar]
  206. Bernacki, R.J.; Oberman, E.J.; Seweryniak, K.E.; Atwood, A.; Bergeron, R.J.; Porter, C.W. Preclinical antitumor efficacy of the polyamine analogue N1, N11-diethylnorspermine administered by multiple injection or continuous infusion. Clin. Cancer Res. 1995, 1, 847–857. [Google Scholar] [PubMed]
  207. Marverti, G.; Piccinini, G.; Ghiaroni, S.; Barbieri, D.; Quaglino, D.; Moruzzi, M.S. N1,N12-bis(ethyl)spermine effect on growth of cis-diamminedichloroplatinum(II)-sensitive and -resistant human ovarian-carcinoma cell lines. Int. J. Cancer 1998, 78, 33–40. [Google Scholar] [CrossRef]
  208. Tummala, R.; Diegelman, P.; Hector, S.; Kramer, D.L.; Clark, K.; Zagst, P.; Fetterly, G.; Porter, C.W.; Pendyala, L. Combination effects of platinum drugs and N1, N11 diethylnorspermine on spermidine/spermine N1-acetyltransferase, polyamines and growth inhibition in A2780 human ovarian carcinoma cells and their oxaliplatin and cisplatin-resistant variants. Cancer Chemother. Pharmacol. 2011, 67, 401–414. [Google Scholar] [CrossRef] [Green Version]
  209. Hector, S.; Porter, C.W.; Kramer, D.L.; Clark, K.; Prey, J.; Kisiel, N.; Diegelman, P.; Chen, Y.; Pendyala, L. Polyamine catabolism in platinum drug action: Interactions between oxaliplatin and the polyamine analogue N1,N11-diethylnorspermine at the level of spermidine/spermine N1-acetyltransferase. Mol. Cancer Ther. 2004, 3, 813–822. [Google Scholar] [CrossRef]
  210. Marverti, G.; Ligabue, A.; Guerrieri, D.; Paglietti, G.; Piras, S.; Costi, M.P.; Farina, D.; Frassineti, C.; Monti, M.G.; Moruzzi, M.S. Spermidine/spermine N1-acetyltranferase modulation by novel folate cycle inhibitors in cisplatin-sensitive and -resistant human ovarian cancer cell lines. Gynecol. Oncol. 2010, 117, 202–210. [Google Scholar] [CrossRef]
  211. Holbert, C.E.; Foley, J.R.; Murray Stewart, T.; Casero, R.A., Jr. Expanded Potential of the Polyamine Analogue SBP-101 (Diethyl Dihydroxyhomospermine) as a Modulator of Polyamine Metabolism and Cancer Therapeutic. Int. J. Mol. Sci. 2022, 23, 6798. [Google Scholar] [CrossRef] [PubMed]
  212. Holbert, C.E.; Stewart, T.M.; Simpson, J.K.; Walker, M.J.; Casero, R.A. The potential of spermine analogue SBP-101 (diethyl dihydroxyhomospermine) as a polyamine metabolism modulator in ovarian cancer. Cancer Res. 2022, 82, 5488. [Google Scholar] [CrossRef]
  213. Marverti, G.; Bettuzzi, S.; Astancolle, S.; Pinna, C.; Monti, M.G.; Moruzzi, M.S. Differential induction of spermidine/spermine N1-acetyltransferase activity in cisplatin-sensitive and -resistant ovarian cancer cells in response to N1,N12-bis(ethyl)spermine involves transcriptional and post-transcriptional regulation. Eur. J. Cancer 2001, 37, 281–289. [Google Scholar] [CrossRef] [PubMed]
  214. Marverti, G.; Monti, M.G.; Bettuzzi, S.; Caporali, A.; Astancolle, S.; Moruzzi, M.S. Cisplatin-resistance modulates the effect of protein synthesis inhibitors on spermidine/spermine N(1)-acetyltransferase expression. Int. J. Biochem. Cell Biol. 2004, 36, 123–137. [Google Scholar] [CrossRef]
  215. Tummala, R.; Diegelman, P.; Fiuza, S.M.; Batista de Carvalho, L.A.; Marques, M.P.; Kramer, D.L.; Clark, K.; Vujcic, S.; Porter, C.W.; Pendyala, L. Characterization of Pt-, Pd-spermine complexes for their effect on polyamine pathway and cisplatin resistance in A2780 ovarian carcinoma cells. Oncol. Rep. 2010, 24, 15–24. [Google Scholar] [CrossRef] [Green Version]
  216. Marverti, G.; Giuseppina Monti, M.; Pegg, A.E.; McCloskey, D.E.; Bettuzzi, S.; Ligabue, A.; Caporali, A.; D’Arca, D.; Moruzzi, M.S. Spermidine/spermine N1-acetyltransferase transient overexpression restores sensitivity of resistant human ovarian cancer cells to N1,N12-bis(ethyl)spermine and to cisplatin. Carcinogenesis 2005, 26, 1677–1686. [Google Scholar] [CrossRef] [Green Version]
  217. Marverti, G.; Ligabue, A.; Lombardi, P.; Ferrari, S.; Monti, M.G.; Frassineti, C.; Costi, M.P. Modulation of the expression of folate cycle enzymes and polyamine metabolism by berberine in cisplatin-sensitive and -resistant human ovarian cancer cells. Int. J. Oncol. 2013, 43, 1269–1280. [Google Scholar] [CrossRef] [PubMed]
  218. Bachrach, U. Polyamines and cancer: Minireview article. Amino Acids 2004, 26, 307–309. [Google Scholar] [CrossRef] [PubMed]
  219. Park, M.H.; Igarashi, K. Polyamines and their metabolites as diagnostic markers of human diseases. Biomol. Ther. 2013, 21, 1–9. [Google Scholar] [CrossRef] [Green Version]
  220. Irajizad, E.; Han, C.Y.; Celestino, J.; Wu, R.; Murage, E.; Spencer, R.; Dennison, J.B.; Vykoukal, J.; Long, J.P.; Do, K.A.; et al. A blood-based metabolite panel for distinguishing ovarian cancer from benign pelvic masses. Clin. Cancer Res. 2022, 28, 4669–4676. [Google Scholar] [CrossRef]
  221. Draga, N.V.; Berdinskikh, N.K.; Zaletok, S.P.; Vinnitskaia, V.K.; Evtushenko, G.V. Diagnostic and prognostic importance of the polyamine test of patients with ovarian tumors. Vopr. Onkol. 1987, 33, 52–57. [Google Scholar] [PubMed]
  222. Lawton, F.; Griffin, M.; Slack, J.; Blackledge, G. Urinary polyamine excretion patterns in patients with epithelial ovarian cancer. Gynecol. Obstet. Investig. 1989, 28, 212–214. [Google Scholar] [CrossRef] [PubMed]
  223. Waalkes, T.P.; Rosenshein, N.B.; Shaper, J.H.; Ettinger, D.S.; Woo, K.B.; Paone, J.F.; Gehrke, C.W. A feasibility study in the development of biological markers for ovarian cancer. J. Surg. Oncol. 1982, 21, 207–214. [Google Scholar] [CrossRef] [PubMed]
  224. Suh, J.W.; Lee, S.H.; Chung, B.C.; Park, J. Urinary polyamine evaluation for effective diagnosis of various cancers. J. Chromatogr. B Biomed. Sci. Appl. 1997, 688, 179–186. [Google Scholar] [CrossRef] [PubMed]
  225. Maenpaa, J.U.; Niemi, R.; Roine, A.; Hakkinen, M.; Kumpulainen, P.; Vepsalainen, T.K.; Lehtimaki, T.; Oksala, N. Urinary acetylated polyamines in ovarian cancer. J. Clin. Oncol. 2015, 33, 5543. [Google Scholar] [CrossRef]
  226. Russell, D.H. Increased polyamine concentrations in the urine of human cancer patients. Nat. New Biol. 1971, 233, 144–145. [Google Scholar] [CrossRef] [PubMed]
  227. Russell, D.H.; Levy, C.C.; Schimpff, S.C.; Hawk, I.A. Urinary polyamines in cancer patients. Cancer Res. 1971, 31, 1555–1558. [Google Scholar]
  228. Lundgren, D.W.; Farrell, P.M.; Cohen, L.F.; Hankins, J. Fluctuations of unbound whole blood polyamine levels during the menstrual cycle. Proc. Soc. Exp. Biol. Med. 1976, 152, 81–85. [Google Scholar] [CrossRef]
  229. Lundgren, D.W.; Oka, T. Alterations in polyamine levels in rat blood during pregnancy and lactation. Am. J. Physiol. 1978, 234, E451–E456. [Google Scholar] [CrossRef]
Figure 1. Biosynthesis, metabolism, transport, and targeting of polyamines. (1) Polyamine biosynthesis: ornithine is generated from arginine and converted into putrescine by ornithine decarboxylase (ODC1). Spermidine synthase (SRM) turns putrescine into spermidine, which is further converted into spermine by spermine synthase (SMS). Synthesis of spermidine and spermine requires aminopropyl moiety from decarboxylated S-adenosylmethionine (dcAdoMet), which is converted from S-adenosylmethionine (AdoMet) by S-adenosylmethionine decarboxylase (AdoMetDC). AdoMet is produced from methionine by methionine adenosine transferase (MAT). (2) Polyamine catabolism: spermidine and spermine are decomposed by spermidine/spermine-N1-acetyltransferase (SSAT) into N-acetylspermidine (AcSpmd) and N-acetylspermine (AcSpm), respectively. AcSpmd and AcSpm are further catabolized by acetylpolymine oxidase (PAOX) into spermidine and putrescine, respectively. In addition, spermine oxidase (SMOX) specifically degrades spermine into spermidine. (3) Polyamine transport: exogenous sources of polyamines, including foods and gut microbiota. Uptake of polyamines from the gut may be mediated by caveoline-1- and NOS-2-dependent processes. Solute carrier transporter SLC3A2 imports putrescine. At the cellular level, heparan sulfate proteoglycans glypican-1 (GPC-1) sequesters spermine for its uptake, likely through endocytosis. Caveoline-1-dependent endocytosis mediated polyamine uptake from extracellular compartments. SLC3A2 can either import or export polyamines following their concentration gradient. SLC18B1 promotes polyamine exocytosis in mast cells but is also required for polyamine uptake in the brain. ATP13A2 and ATP13A3 are involved in polyamine internalization via poorly characterized mechanism(s). (4) Targeting polyamine pathways: polyamine analogs or small molecule inhibitors targeting the polyamine metabolism pathway antagonize the tumor-promoting activities of polyamines. Small molecules AMXT 1501, F14512, and polyamine analogs DENSpm and BESpm block the uptake of polyamines by cancer cells. In addition, DENSpm and BESpm also inhibit polyamine synthesis by suppressing SRM or promote polyamine catabolism by upregulating SMOX and SSAT. DFMO and SBP-101 repress ODC, while MGBG, SAM486A, AbeAdo, and Genz-644131 target AMD to downregulate polyamine synthesis.
Figure 1. Biosynthesis, metabolism, transport, and targeting of polyamines. (1) Polyamine biosynthesis: ornithine is generated from arginine and converted into putrescine by ornithine decarboxylase (ODC1). Spermidine synthase (SRM) turns putrescine into spermidine, which is further converted into spermine by spermine synthase (SMS). Synthesis of spermidine and spermine requires aminopropyl moiety from decarboxylated S-adenosylmethionine (dcAdoMet), which is converted from S-adenosylmethionine (AdoMet) by S-adenosylmethionine decarboxylase (AdoMetDC). AdoMet is produced from methionine by methionine adenosine transferase (MAT). (2) Polyamine catabolism: spermidine and spermine are decomposed by spermidine/spermine-N1-acetyltransferase (SSAT) into N-acetylspermidine (AcSpmd) and N-acetylspermine (AcSpm), respectively. AcSpmd and AcSpm are further catabolized by acetylpolymine oxidase (PAOX) into spermidine and putrescine, respectively. In addition, spermine oxidase (SMOX) specifically degrades spermine into spermidine. (3) Polyamine transport: exogenous sources of polyamines, including foods and gut microbiota. Uptake of polyamines from the gut may be mediated by caveoline-1- and NOS-2-dependent processes. Solute carrier transporter SLC3A2 imports putrescine. At the cellular level, heparan sulfate proteoglycans glypican-1 (GPC-1) sequesters spermine for its uptake, likely through endocytosis. Caveoline-1-dependent endocytosis mediated polyamine uptake from extracellular compartments. SLC3A2 can either import or export polyamines following their concentration gradient. SLC18B1 promotes polyamine exocytosis in mast cells but is also required for polyamine uptake in the brain. ATP13A2 and ATP13A3 are involved in polyamine internalization via poorly characterized mechanism(s). (4) Targeting polyamine pathways: polyamine analogs or small molecule inhibitors targeting the polyamine metabolism pathway antagonize the tumor-promoting activities of polyamines. Small molecules AMXT 1501, F14512, and polyamine analogs DENSpm and BESpm block the uptake of polyamines by cancer cells. In addition, DENSpm and BESpm also inhibit polyamine synthesis by suppressing SRM or promote polyamine catabolism by upregulating SMOX and SSAT. DFMO and SBP-101 repress ODC, while MGBG, SAM486A, AbeAdo, and Genz-644131 target AMD to downregulate polyamine synthesis.
Cancers 15 00623 g001
Table 1. Polyamine as biomarkers for ovarian cancer.
Table 1. Polyamine as biomarkers for ovarian cancer.
PolyaminesSources of
Polyamines
ObservationsReference
PolyaminesUrineIncreased polyamines correlate with clinical status.[221]
Free and acetylated polyaminesUrineFree and acetylated polyamines were elevated in cases compared to controls.[222]
Putrescine
Spermidine
Spermine
UrinePolyamines are elevated in patients with progressive diseases;
Spermidine/creatinine ratio is increased.
[223]
Putrescine
Spermidine
Spermine
NAcPuT
AcSpmd
UrinePolyamines are drastically elevated in cancer patients[224]
DASUrineDAS has 65% specificity and 91% sensitivity (AUC 0.82), better than CA-125 (65% specificity, 68% sensitivity, AUC 0.75) and RMI (70% specificity, 68% sensitivity, AUC 0.72)[225]
DAS
N3AP
DAS
AcSpmd
PlasmaPolyamine signature consisting of DAS and N3AP in combination with CA-125 yields improvement in sensitivity at >99% specificity relative to CA-125 alone (73.7% vs. 62.2%) and can capture 30.4% more cases than CA-125 alone[22]
DAS
N3AP
DiAcSpmd
Serum7MetP yields an AUC of 0.86;
7MetP+ROMA increase AUC from 0.91 (ROMA alone) to 0.93;
7MetP+ROMA has a higher positive predictive value (0.68 vs. 0.52) with improved specificity (0.89 vs. 0.78) compared to ROMA alone.
[220]
DAS: Diacetylspermine, NAcPut: N-acetylputrescine, AcSpmd: N-acetylspermidine, N3AP: N-(3-acetamidopropyl)pyrrolidine-2-one, DiAcSpmd: Diacetylspermidine, 7MetP: 7-marker metabolite panel, ROMA: risk of ovarian malignancy algorithm.
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

Chen, Y.; León-Letelier, R.A.; Abdel Sater, A.H.; Vykoukal, J.; Dennison, J.B.; Hanash, S.; Fahrmann, J.F. c-MYC-Driven Polyamine Metabolism in Ovarian Cancer: From Pathogenesis to Early Detection and Therapy. Cancers 2023, 15, 623. https://doi.org/10.3390/cancers15030623

AMA Style

Chen Y, León-Letelier RA, Abdel Sater AH, Vykoukal J, Dennison JB, Hanash S, Fahrmann JF. c-MYC-Driven Polyamine Metabolism in Ovarian Cancer: From Pathogenesis to Early Detection and Therapy. Cancers. 2023; 15(3):623. https://doi.org/10.3390/cancers15030623

Chicago/Turabian Style

Chen, Yihui, Ricardo A. León-Letelier, Ali Hussein Abdel Sater, Jody Vykoukal, Jennifer B. Dennison, Samir Hanash, and Johannes F. Fahrmann. 2023. "c-MYC-Driven Polyamine Metabolism in Ovarian Cancer: From Pathogenesis to Early Detection and Therapy" Cancers 15, no. 3: 623. https://doi.org/10.3390/cancers15030623

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