Abstract
Ceramides are a family of wax-like lipids that fall under the broader category of sphingolipids. A ceramide is composed of a sphingosine side chain coupled to a fatty acid via an amide linkage. Distinct from complex sphingolipids, the head of ceramide is a simple alcohol rather than a phosphate, phosphocholine, sugar, or more. The fatty acid chains of ceramide can also vary in chain length and degree of saturation. The degree of saturation may determine the biological activity of the ceramide species. Ceramides are highly abundant within the cell membrane of eukaryotic cells and are appreciated for their structural roles in these cells. Moreover, ceramides are well-known for their biological activity including as regulators of apoptosis, senescence, the cell cycle, and differentiation. This review discusses pathways of ceramide, roles of ceramide in various diseases, targeting ceramide metabolism in the treatment of cancer, as well as ceramide-delivering nanotechnologies.
Keywords: Ceramide, Sphingolipid Metabolism, Cancer, Leukemia, Therapeutics, Nanoliposomes, Combinatorial Therapies
Ceramide Metabolism
Ceramide metabolism consists of a complex network of interconnected pathways with many origins and many downstream biological implications (Figure 1) [1]. Broadly, ceramide metabolism can be classified into de novo synthesis and catabolism, including recycling, salvage, and degradation.
Figure 1. Basic pathway of ceramide metabolism.
De novo ceramide synthesis initially generates dihydrosphingolipid species prior to desaturation. Salvage ceramide synthesis can include turnover by the activities of acidic/lysosomal-localized catabolic enzymes, which generates sphingosine that leaves the lysosome and is re-acylated to ceramide. Glucosylceramide synthase (GCS); cerebrosidase (GBA); sphingomyelin synthase (SGMS); ceramide kinase (CERK), ceramide-1-phosphate phosphatase (C1PP); ceramidases (CDase); ceramide synthase (CERS); sphingosine kinase (SPHK); S1P phosphatase (S1PP).
De novo synthesis
De novo synthesis of ceramide occurs in the endoplasmic reticulum [1]. It starts with the condensation of the amino acid serine and palmitoyl-CoA by serine palmitoyltransferase, which produces 3-keto-dihydrosphingosine. This intermediate is reduced to dihydrosphingosine (also known as sphinganine) by 3-ketosphinganine reductase. The ceramide synthase (CERS) family catalyzes the N-acylation of dihydrosphingosine to dihydroceramide. Dihydroceramide desaturase next introduces a 4,5-trans-double bond into the backbone of dihydroceramide, producing ceramide.
Six ceramide synthases (CERS1–6) have been identified in humans (Table 1) [1]. Each CERS enzyme prefers different chain lengths of the fatty acyl-CoA substrate involved in the N-acylation of dihydrosphingosine. The resulting dihydroceramide may contain a fatty acid of varying lengths depending on the CERS enzyme utilized. The chain lengths of the fatty acyl-CoA used tend to vary from 14 to 36 carbons. Ceramides with different chain compositions impact cell physiology differently [2], as has been observed with contrast death/survival roles reported for C16 and C18 ceramides [3,4]. In addition, the various CERS enzymes can have different tissue distribution patterns. For example, CERS1 is highly expressed in the brain and skeletal muscle, which correlates with an abundance of C18 ceramide in those tissues. CERS2 is highly expressed in the liver and kidneys and favors very-long chain fatty acids (C22-C24) [5]. CERS3 is highly expressed in the skin and testis and is selective toward very-long (C22-C24) and ultra-long chains (≥ C26). CERS4 is highly expressed in the skin and leukocytes and catalyzes formation of ceramide with a selectivity towards long and very-long chains (C18-C22) [6]. CERS5 is more commonly found in the muscle and brain. It mainly produces ceramides incorporating C16 fatty acyl-CoA [5]. CERS6 also uses C16 fatty acyl-CoA as a substrate and is commonly found in the brain [7].
Table 1.
Classification and distribution of ceramide synthases
Ceramide synthase | Chain length of the fatty acyl-CoA substrate | Tissue distribution |
---|---|---|
CERS1 | C18 | Brain and skeletal muscle |
CERS2 | C22-C24 | Liver and kidneys |
CERS3 | C22-C24, ≥ C26 | Skin and testis |
CERS4 | C18-22 | Skin, leukocytes |
CERS5 | C16 | Muscle and brain |
CERS6 | C16 | Brain |
Salvage, recycling, and degradation
In addition to de novo synthesis, ceramide can be “salvaged” from complex sphingolipid reservoirs, recycled to/from other sphingolipids, or degraded into simple molecules [1]. These pathways, alongside de novo synthesis, regulate the cellular levels of ceramide. The salvage of ceramide from complex sphingolipids provides for more than half of ceramide biosynthesis. One such example is the catabolism of glycosphingolipids [1]. During the breakdown of large glycosphingolipids, sugar groups are cleaved off which leads to the formation of either glucosylceramide or galactosylceramide. In turn, specific β-glucosidases and galactosidases hydrolyze these lipids to form ceramide [8]. In addition to the formation of ceramide from larger sphingolipids such as glycosphingolipids, ceramide can be formed via the hydrolysis of sphingomyelin by various sphingomyelinases (SMase) [5], which are defined by their pH optima. Ceramide can also act as a substrate to build more complex sphingolipids. For instance, ceramide can be converted to sphingomyelin, a large component of the fatty myelin sheath, via the action of sphingomyelin synthase (SGMS). Similarly, ceramide can be utilized by glucosylceramide synthase (GCS) to generate glucosylceramide. Glucosylceramide, as well as galactosylceramide, can go on to form more complex sphingolipids in the Golgi apparatus, which include gangliosides, globosides, and sulfatides. These can then be moved to the plasma membrane, the major reservoir of these lipids [9,10]. The degradation of ceramide is also a necessary part of maintaining lipid homeostasis. Ceramides present in endothelial cells can be de-acylated to yield sphingosine by ceramidases (CDase), which are also defined by their pH optima. Sphingosine can then be used to generate sphingosine-1-phosphate (S1P) or be re-acylated to again form ceramide [11]. This re-acylation process is further important to complex sphingolipid catabolism in the lysosome as the ceramide that is initially generated is catabolized by acid CDase to sphingosine, which can subsequently be re-acylated by CERS to re-form ceramide. In a similar mechanism, short-chain ceramide analogs can be catabolized by acid CDase to sphingosine and then re-acylated to yield ceramide with physiological chain lengths [12].
Ceramide and S1P
In addition to ceramide, S1P is an important sphingolipid to highlight. Ceramide can be converted into S1P via two enzymatic reactions [1]. Typically, acid CDase (or another CDase) hydrolyzes ceramide to sphingosine and then a sphingosine kinase (SPHK), typically SPHK1, phosphorylates sphingosine to generate S1P. Although S1P and ceramide share structural similarities and are easily interconverted, the two exhibit opposite effects. Whereas ceramide is well appreciated for its role in the stimulation of apoptosis, S1P is appreciated as a pro-survival signal, leading to increased cell growth and viability [13]. S1P’s pro-survival effects are beneficial to tumors and thus an excess of S1P in tissues is associated with tumor neogenesis. S1P predominantly signals through G protein-coupled receptors (GPCRs) known as S1P receptors (S1PRs). Activation of S1PRs often leads to subsequent activation of the Raf-Mek-Erk pathway, a classic mitogenic pathway. Thus, activation of these GPCRs leads to increased tumor cell viability. The control of the titer of ceramide and S1P in tumors is thought to be a potential therapeutic target [14].
Basic role of ceramide in skin disease, metabolic syndrome, infection, and cancer
Structurally diverse ceramides can provide platforms for signaling events and protein/lipid trafficking along cellular membranes [15]. Thus, changes in ceramide levels can alter the biophysical properties of the membrane. In this way, ceramide and its metabolism can function as a sensor, affect the distribution of transmembrane proteins, and impact a diversity of other cellular processes [16]. Consequently, abnormalities in ceramide homeostasis can contribute to various pathologies. This is important when considering inflammatory links between conditions including metabolic disease or infection and cancer, or even the persistence of cancer within the skin.
Skin diseases
Skin ceramide composition, which plays crucial roles in maintaining the barrier function of the stratum corneum, is altered in patients with atopic dermatitis [17]. It is accompanied by a change of ceramide synthase isoform expression from CERS3 to CERS4, leading to a barrier repair response [18]. Atopic dermatitis, sometimes referred to as eczema, is an inflammatory condition linked with a compromised epidermal barrier [19,20]. Topical treatment of atopic dermatitis with pseudo-ceramide lotion, which contains 3% Cetyl-PG hydroxyethyl palmitamide, has been found to reduce symptoms such as pruritis. Additionally, a decrease in trans-epidermal water loss and increased water content has been shown with pseudo-ceramide treatment. Those changes are associated with a distinct switch of the ceramide profile from an atopic dermatitis to a healthy skin phenotype without any increased level of endogenous ceramides [21]. Furthermore, increased ceramide content has been shown to result in acceleration of epidermal barrier restoration after acute damage to the skin [22]. Collectively, this is noteworthy as pseudo-ceramide has become a major and well-advertised component of various skin creams.
Metabolic syndrome
Ceramide contributes to lipotoxicity that underlies metabolic syndrome and related diseases such as type 2 diabetes, as well as various types of cardiovascular disease including coronary artery disease, hypertension, and stroke [23]. The species of ceramide typically associated with cardiovascular disease progression are C16 ceramide (d18:1/16:0), C18 ceramide (d18:1/18:0), and C24:1 ceramide (d18:1/24:1). These ceramides have been shown to lead to abnormalities in glucose metabolism by blocking insulin signaling. Additionally, ceramides dysregulate lipid metabolism by promoting adipose inflammation. In contrast, C24 ceramide (d18:1/24:0) has been associated with good cardiovascular health [24]. Interestingly, skeletal muscle fatty acid oxidation was promoted alongside a decrease in whole-body fat accumulation following oral treatment with P053, which targets CERS1 [25].
Infection
The structural domains formed by different ceramides can cause changes in membrane curvature. These changes in membrane morphology may be exploited by pathogens, leading to host cell infection [26]. Bile acids and ceramides can assist the entry of viruses and to facilitate their subsequent replication in jejunal enteroids. Some viral pathogens can invade the epithelial barrier and replicate in the human small intestine. For instance, in the case of norovirus strains, ceramide can assist their release into the cytoplasm by acting as a binding factor [27]. Conversely, it has been shown that ceramide can mediate neutrophil extracellular trap formation to boost their bactericidal capacity [28]. Interestingly, studies have shown that the glucosylceramide-ceramide cycle can regulate cellular entry of certain viruses including the influenza virus [29,30]. Glucosylceramide can be generated from ceramide as well as broken down to ceramide. In one study, manipulation of glucosylceramide synthesis revealed that glucosylceramide can facilitate influenza viral binding and entry into host cells [29]. A follow-up study instead manipulated the breakdown of glucosylceramide to ceramide to demonstrated that this process was key to mediating endocytosis of influenza and other enveloped viruses [30].
Cancer
Ceramides of various chain lengths have been found to be dysregulated in different cancer types [13]. Ceramides have been shown to either mediate apoptosis via mitochondrial membrane perturbation or to exert their effects by influencing cell death signaling. Conversely, some reports have suggested that certain ceramide species may exert anti-apoptotic, protective roles [13]. The specific effect of a given species of ceramide towards cancer may depend on its molecular structure or the cellular context [31]. For example, C16 ceramide generation has been shown to be decreased in human ovarian carcinoma cells [32]. Whereas the overexpression of CERS2, which increases the generation of C24 ceramide, was shown to protect Hela cells from ionizing radiation (IR)-induced apoptosis [33]. Therefore, these reports may suggest that cancer cell survival may be due to a lack of C16 ceramide or a prevalence of C24 ceramide, or alternatively due entirely to the cancer cell type independent of the ceramide species. This later perspective may be further supported by a study that showed elevations of C14 ceramide, C16 ceramide, and C18 ceramide in breast cancer tissue as compared to normal tissue [34]. Suffice to say, while most roles attributed to ceramides are pro-stress or pro-death, there are instances where this traditional dogma does not apply for reasons that deserve further investigation. Notably though, various anticancer therapeutics have been shown to increase cellular levels of ceramides or alter the expression or activity of different CERS, potentially contributing to their anti-neoplastic activity (Table 2). Importantly, an increase or decrease in the expression or activity of a given CERS will not always manifest in an increase or decrease in the associated ceramide species alone. This is because those ceramide species may be converted to any number of ceramide metabolites, such as sphingomyelins or glycosphingolipids. Therefore, changes in CERS can potentially alter ceramide species or any of their metabolic derivatives.
Table 2.
Anticancer therapeutics that affect ceramide generation
Name of anticancer therapeutic | Mechanism of action | Effect on ceramide metabolism | Cancer type | Citation |
---|---|---|---|---|
Chemotherapy agents | ||||
Camptothecin | Inhibiting DNA synthesis | Increase de novo ceramide synthesis | Follicular thyroid carcinoma | Rath et al., 2009 [35] |
Carboplatin | Interfering with DNA replication | Increase CERS1 expression | Embryonic kidney cells | Min et al., 2007 [36] |
Cisplatin | Interfering with DNA replication | Increase CERS1, CERS6 expression | Embryonic kidney cells, Oral squamous carcinoma | Li et al., 2018 [37]; Min et al. 2007 [36] |
Crenolanib | Tyrosine kinase inhibitor against FLT3, PDGFRα and PDGFRβ | Increase CERS1 expression | Acute myeloid leukemia | Dany et al. 2016 [38] |
Dasatinib | Tyrosine kinase inhibitor against Bcr-Abl | Increase CERS1, CERS5, CERS6 expression | Chronic myeloid leukemia | Gencer et al., 2011 [39] |
Daunorubicin | DNA intercalation | Increase CERS expression | Leukemia | Bose et al., 1995 [40] |
Doxorubicin | DNA intercalation | Increase CERS1, CERS2, CERS5 expression | Embryonic kidney cells, Bladder cancer, Breast cancer | Fan et al., 2013 [41]; Huang et al., 2018 [42]; Min et al., 2007 [36] |
Etoposide | Topoisomerase II inhibitor | Increase CERS2, CERS3, CERS4 expression | Embryonic fibroblasts | Siddique et al., 2012 [43] |
Fludarabine | Purine analog; interferes with DNA synthesis | Increase CERS2 expression | Chronic lymphocytic leukemia | Biswal et al., 2000 [44] |
Fluorouracil (5-FU) | Thymidylate synthase inhibitor, inhibiting DNA replication | Decrease CERS5 expression | Colon cancer | Brachtendorf et al., 2018 [45] |
Gemcitabine | Inhibiting DNA synthesis | Increase CERS2 expression | Pancreatic cancer | Modrak et al., 2009 [46] |
Imatinib | Tyrosine kinase inhibitor against Bcr-Abl | Increase CERS1 expression | Chronic myeloid leukemia | Baran et al., 2007 [47] |
Irinotecan | Inhibiting DNA synthesis | Block formation of glucosyl-ceramide | Colon cancer | Litvak et al. 2003 [48] |
Methotrexate | Inhibiting the synthesis of DNA, RNA, via inhibition of thymidylate | Increase CERS6 expression | Liver cancer, Lung cancer | Fekry et al., 2016 [49] |
Oxaliplatin | Inhibiting DNA synthesis | Increase CERS5 expression | Colon cancer | Brachtendorf et al., 2018 [45] |
Quizartinib | Tyrosine kinase inhibitor against FLT3/STK1, CSF1R/FMS, SCFR/KIT, and PDGFRs | Increase CERS1 expression | Acute myeloid leukemia | Dany et al. 2016 [38] |
Sorafenib | Kinase inhibitor against VEGFR, PDGFR and Raf kinases. | Increase CERS1 expression | Acute myeloid leukemia | Dany et al. 2016 [38] |
Vincristine | Inhibitor of mitosis at metaphase | Increase CERS1, CERS5 expression | Embryonic kidney cells | Min et al., 2007 [36] |
Radiation | ||||
Ionizing radiation | Damaging DNA | Increase CERS2, CERS5, CERS6 expression | Cervical cancer, multiple cancer types | Mesicek et al., 2010 [33] |
UV-B radiation | Damaging DNA | Increase CERS1 expression | Lung cancer | Sridevi et al., 2009 [50] |
Alternative medicine | ||||
Staurosporine | Inhibitor of protein kinases against PKC | Increase CERS2, CERS6 expression | Glioma | Jensen et al., 2014 [51] |
Hormone inhibitors | ||||
Anastrozole | Aromatase inhibitor, inhibits the production of estrogens | Decrease CERS4, CERS5 expression | Endometrial cancer | Mojakgomo et al., 2015 [52] |
Tamoxifen | Selective estrogen receptor modulator | Inhibit ceramide glycosylation | Breast cancer | Wang et al., 2003 [53] |
Adjuvant drugs | ||||
R(+)-meth anandamide | Synthetic long-lasting anandamide analog | Increase CERS6 expression | Mantle cell lymphoma | Gustafsson et al., 2009 [54] |
Tetrahydro cannabinol (THC) | Partial agonist activity at cannabinoid receptors | Increase CERS5, CERS6 expression | Glioma | Hernández-Tiedra et al., 2016 [55] |
Anti-inflammatory drugs | ||||
Celecoxib | COX-2 inhibitor | Increase CERS6 expression | Colon cancer | Schiffmann et al., 2010 [56] |
Clinical trial drugs | ||||
Cladososide C2 | Inducing apoptosis | Increase CERS6 expression | Leukemia | Yun et al., 2015 [57] |
Fenretinide | Inducing apoptosis | Activate serine palmitoyl transferase | Prostate cancer, relapsed and refractory T-cell lymphoma, ovarian cancer, neuroblastoma, head and neck squamous cell carcinoma, recurrent small cell lung cancer, kidney cancer, breast cancer | Wang et al., 2003 [53]; Clinicaltrials.gov identifiers: NCT04234048, NCT00026091, NCT00646230, NCT00006471, NCT00009971, NCT00011973, NCT00003099 |
IL-24 | Inducing apoptosis and angiogenesis | Increase CERS6 expression | Glioblastoma | Yacoub et al., 2010 [58] |
Resveratrol | Not yet clear | Inhibition of dihydroceramide desaturase | Gastric cancer | Signorelli et al., 2009 [59] |
Stichoposide D | Inducing apoptosis | Increase CERS6 expression | Leukemia | Yun et al., 2015 [57] |
TRAIL(Apo-2L) | Inducing TNF-related apoptosis | Increase CERS6 expression | Colon cancer | White-Gilbertson et al., 2009 [60] |
Targeting ceramide metabolism for cancer therapy
Considering the known effects of ceramide and its dysregulation in many cancers, developing ceramide-based therapies for cancer is very attractive. Various therapies have aimed to target ceramide metabolism as a method to promote apoptosis in cancer cells (Table 3). These therapies upregulate endogenous production of ceramide or downregulate ceramide neutralization pathways. Enzymes that neutralize ceramide convert ceramide to non-ceramide sphingolipids. Examples include GCS, SGMS, CDases, and SPHKs (Figure 1). The goal of these therapies is to raise the intracellular concentration of ceramide to direct cancer cells towards apoptosis. In addition to augmenting endogenous ceramide synthesis via pharmacological interventions, increasing ceramide levels through delivery of exogenous analogs has been explored in various cancer models. Various analogs or inhibitors of ceramide signaling have also shown efficacy in clinical trials. Targeting ceramide is also not the only sphingolipid-based strategy that is being explored for their potential to treat cancer. For example, fingolimod (FTY720 or Gilyena™), a structural analog of sphingosine (or S1P once phosphorylated) and regulator of S1PR signaling, has been approved for clinical use for the treatment of multiple sclerosis and has also shown potential anticancer efficacy in various preclinical models [61]. Mechanistically, fingolimod suppresses tumor growth via S1PR-dependent or receptor-independent mechanisms in colon and lung cancer, as well as acute myeloid leukemia, chronic lymphocytic and large granular lymphocytic leukemia [62–67]. Most of these effects are thought to be dependent on its activity towards S1PR1, but antagonistic effects towards other S1PRs such as S1PR5 have been reported in preclinical studies of its use to treat large granular lymphocytic leukemia [64]. Fingolimod has also shown potential to treat imatinib-refractory chronic myeloid leukemia by inhibiting stem cell proliferation and expansion in vitro [68].
Table 3.
Synthetic drugs that target sphingolipid metabolism or signaling for cancer
Synthetic drug | Mechanism of action | Cancer type effected | Citation |
---|---|---|---|
AB1 | S1PR2 antagonist | Neuroblastoma | Li et al., 2015 [69] |
ABC294640 | Inhibitor of sphingosine kinase 2 | Pancreatic cancer, prostate cancer, multiple myeloma, lymphoma | Britten et al., 2017 [70] ; Lewis et al., 2016 [71] ; Qin et al., 2014 [72] ; Venant et al., 2015 [73]; Venkata et al., 2014 [74] |
Analog 315 and 403 | Ceramide analog | Lymphoma, breast cancer | Chen et al., 2020 [75] |
Analog 406 | C8-Ceramide analog | Breast cancer, ovarian cancer | Ponnapakam et al., 2014 [76] |
C16-serinol | C16-Ceramide analog | Neuroblastoma | Bieberich et al., 2000 [77] |
Ceranib-2 | Inhibitor of acid ceramidase | Breast cancer | Draper et al., 2011 [78] |
D-MAPP, B13 | Inhibitors of acid ceramidase | Colon cancer | Selzner et al., 2001 [79] |
Fingolimod (FTY720 or Gilenya™) | S1PR antagonist (predominantly S1PR1) | Colon cancer, lung cancer, chronic myeloid leukemia, chronic lymphocytic leukemia, large granular lymphocytic leukemia | Chen et al., 2014 [62]; Liang et al., 2013 [63]; Liao et al., 2011 [64]; Liu et al., 2008 [65]; Neviani et al., 2013 [68]; Saddoughi et al., 2013 [66]; Young et al., 2019 [67] |
LCL29 | C6-Ceramide analog | Head and neck squamous cell carcinoma | Separovic et al., 2011 [80] |
LCL30 | C16-Ceramide analog | Head and neck squamous cell carcinoma | Separovic et al., 2011 [80] |
LCL85 | C16-Ceramide analog | Colon cancer, breast cancer | Paschall et al., 2014 [81] |
LCL124 | C6-Ceramide analog | Pancreatic cancer | Beckham et al., 2013 [82] |
LCL204 | Inhibitor of acid ceramidase | Head and neck squamous cell carcinoma, acute myeloid leukemia | Elojeimy et al., 2007 [83] ; Tan et al., 2016 [84] |
LCL385 | Inhibitor of acid ceramidase | Prostate tumors | Mahdy et al., 2009 [85] |
LCL521 | Inhibitor of acid ceramidase | Head and neck squamous cell carcinoma | Korbelik et al., 2016 [86] |
MP-A08 | Inhibitor of sphingosine kinase 1 | Acute myeloid leukemia | Powell et al., 2017 [87] |
PF-543 | Inhibitor of S1P | Head and neck squamous cell carcinoma, colon cancer | Ju et al., 2016 [88]; Schnute et al., 2016 [89] |
SACLAC | Inhibitor of acid ceramidase | Acute myeloid leukemia | Pearson et al., 2020 [90] |
SK1-I | Inhibitor of sphingosine kinase 1 | Glioblastoma | Kapitonov et al., 2009 [91] |
SKI-178 | Inhibitor of sphingosine kinase 1 | Acute myeloid leukemia | Hengst et al., 2017 [92] |
Sonepcizumab | S1P monoclonal antibody | Renal cell carcinoma | Pal et al., 2017 [93] |
VPC03090 | S1PR1/3 antagonist | Breast cancer | Kennedy et al., 2011 [94] |
Nanotechnology for ceramide delivery
One of the issues with ceramide-based therapies is the lipid’s delivery in vivo. Since ceramide is highly hydrophobic, a suitable carrier is needed for its successful delivery without restricting its pharmacological effects [95]. Recent advances in nanotechnology have helped to develop novel drug delivery platforms that overcome many typical pharmacological limitations. Nanoparticles, measuring up to 400 nm in size, have demonstrated efficacy for carrying and delivering therapeutic molecules with diverse physiological properties. Several delivery materials have been explored to increase ceramide-based drug performance (Table 4). Importantly, the use of nanoparticles may allow for selective targeting of tumors due to the enhanced permeation and retention (EPR) effect [95,96]. The EPR effect is due to the propensity of tumor vasculature to be leaky. This is largely due to the disorganized manner that tumor-mediated neovascularization occurs. Gaps between vascular endothelium provide potential for passive targeting of anticancer nanotherapies to tumors. Nanoparticles are small enough to leave the vasculature at these points of disorganization and leakiness. In theory, this facet of tumors allows for passive targeting of cancerous tissues by nanomedicines [95,96]. A particularly optimistic aspect for the use of nanotechnology in ceramide delivery is the potential for non-toxic therapy. One of the largest issues confronting cancer therapeutics is the toxic nature of many of these drugs. Unlike many standard of care therapeutics, the C6-ceramide nanoliposome has shown promising results in terms of toxicity. In dog and murine models, the C6-ceramide nanoliposome was minimally toxic [95]. More recently, the ceramide nanoliposome completed a phase I clinical trial with solid tumor patients [97]. Importantly, despite evaluating a dose range from 36 mg/m2 to 323 mg/m2, there were no dose-limiting toxicities observed and a maximum tolerated dose was not reached [97]. For this trial, the starting dose of 36 mg/m2 was chosen based on prior canine studies yet the human trial rapidly increased beyond this dose when no dose-limiting toxicities were observed [97]. Beyond nanoliposomes, other nanotechnologies may likewise be able to effective deliver ceramide to cancer cells. Unfortunately, most nanoparticles are composed of highly toxic materials [92]. One exception, beyond nanoliposomes, is the calcium phosphosilicate nanoparticle. This platform offers many of the same advantages as nanoliposomes in that it can delivery both hydrophobic and lipophilic agents including ceramide [96, 98]. Intriguingly, calcium phosphosilicate nanoparticles can deliver ceramide of varying chain lengths, is composed of non-toxic materials, and offers controlled release inside target cells [96, 98]. Overall, the use of nanotechnology in drug delivery may lead to a new generation of cancer therapeutics, one in which toxicity is not the norm for standard of care therapies.
Table 4.
Nanotechnology for ceramide delivery
Delivery material | Advantages | Citation |
---|---|---|
C6-ceramide nanoliposome | Overcomes the lack of solubility for lipophilic compounds (drugs) in physiological solutions, high ceramide encapsulation efficiency, stealth characteristics, can be actively targeted, ability to co-encapsulate/deliver hydrophobic and lipophilic drugs, increased systemic circulation, reduced toxicity Clinicaltrials.gov identifier: NCT02834611 |
Ciner et al., 2024 [97]; Kester et al., 2015 [95]; Stover and Kester, 2003 [99] |
Ceramide-loaded calcium phosphosilicate nanoparticle | Encapsulates a wide range of compounds (hydrophobic and lipophilic), fully encapsulates active agents, can co-deliver multiple agents including imaging agents, stable at physiological pH for protected delivery, controlled release inside cells at acidic pH, stealth characteristics, can be actively targeted, increased systemic circulation, and reduced toxicity | Adair et al., 2010 [96]; Kester et al., 2008 [98] |
Chitosan-ceramide graft copolymer | Sustained release, higher cellular uptake, improved stability and reduced toxicity under physiological conditions | Battogtokh and Ko, 2014a [100]; Battogtokh and Ko, 2014b [101] |
Poly (D,L-lactide-co-glycolide) PLGA | Favorable anticancer drug delivery performance but insufficient tumor targeting | Graf et al., 2012 [102] |
Solid lipid nanoparticles | Large surface area and high loading content for hydrophobic drugs, improved stability | Balakrishnan et al., 2016 [103] |
Combined usage of ceramide and other drugs in the treatment of cancer
With the assistance of nanotechnology, exogenous ceramide delivery is being explored as a therapeutic strategy for various cancers. C6-ceramide has been found to prevent metastasis and recurrence of anaplastic thyroid carcinoma cell lines [104]. Moreover, the C6-ceramide nanoliposome slows the growth of liver tumors in mice by boosting T-cell activity [105]. In addition to being administered alone, exogenous ceramide can also be incorporated with other drugs as innovative cancer treatments [106]. The nanoliposome has potential as a delivery mechanism for combinatorial therapies because ceramide can be easily distributed into the hydrophobic membrane of nanoliposomes, alongside other lipophilic drugs, while hydrophilic therapies can be loaded into the nanoliposomal aqueous interior [95]. As studies demonstrate the ability of nano-formulated ceramide to improve the efficacy of other drugs, ceramide will likely gain more clinical interest as a combinatorial therapeutic than as a monotherapy (Table 5) [95].
Table 5.
Combination of ceramide and other drugs/agents in the treatment of cancer
Combination usage | Cancer type | Citation |
---|---|---|
7,8-Benzoflavone nanoliposome + C6-ceramide nanoliposome | Acute myeloid leukemia | Barth et al., 2014a [107] |
Curcumin + C6-ceramide | Melanoma | Yu et al., 2010 [108] |
Curcumin + C6-ceramide | Osteosarcoma | Dhule et al., 2014 [109] |
Doxorubicin + C6-ceramide | Breast cancer, melanoma | Fonseca et al., 2014 [110] |
Gemcitabine + C6-ceramide nanoliposome | Pancreatic cancer | Jiang et al., 2011 [111] |
Ibrutinib + C6-ceramide nanoliposome | Chronic lymphocytic leukemia | Doshi et al., 2017 [112] |
Myrisplatin + C6-ceramide | Ovarian cancer | Ganta et al., 2014 [113] |
Paclitaxel + C6-ceramide | Melanoma | Carvalho et al., 2017 [114] |
PDMP nanoliposome + C6-ceramide nanoliposome | Pancreatic cancer | Jiang et al., 2011 [111] |
Safingol nanoliposome + C2-ceramide nanoliposome | Acute myeloid leukemia | Tan et al., 2014 [115] |
Safingol nanoliposome + C6-ceramide nanoliposome | Acute myeloid leukemia | Brown et al., 2013 [116] |
Sorafenib + C6-ceramide nanoliposome | Breast cancer, melanoma | Tran et al., 2008 [117] |
Tamoxifen nanoliposome + C6-ceramide nanoliposome | Acute myeloid leukemia | Barth et al., 2014b [118]; Morad et al., 2016 [119] |
Venetoclax + cytarabine + C6-ceramide nanoliposome | Acute myeloid leukemia | Khokhiatchev et al., 2022 [120] |
Vinblastine nanoliposome + C6-ceramide nanoliposome | Hepatocellular carcinoma, colon cancer | Adiseshaiah et al., 2013 [121] |
Vinblastine (free drug or nanoliposome) + C6-ceramide nanoliposome | Acute myeloid leukemia | Barth et al., 2019 [12] |
Conclusions
Sphingolipids, as with other lipids, have been classically appreciated as being structural elements of cell membranes. While this is a truism, it is not the whole picture. Biologically active lipids play vital roles in the development and prevention of diseases. This is highlighted through an examination of ceramide, its metabolic pathways, as well as its demonstrative physiological effects. Ceramide plays large roles in many disease processes, including cancer. Many cancers experience a downregulation of ceramide and the enzymes responsible for its biosynthesis. This is because the presence of large quantities of endogenous ceramide is often associated with the induction of apoptosis. However, there have been contrasting findings that have challenged the dogma that ceramide exclusively promotes stress and apoptosis. For instance, by ascribing anti-apoptotic roles to specific ceramide species [4]. Moreover, the field’s perspective on the deleterious effects ascribed to certain ceramide species in cancer has evolved as elevations of various ceramide species have been observed in malignant cells and tissues [12,34]. Notably, in many lipidomic studies C16:0, C18:0, and C24:1 ceramide are the predominant ceramide species detected and so variations in these are frequently characterized. However, in many cases the change in these ceramide species can be identical and as such can sometimes be associated with similar biological effects. It is important though to recall that similar increases in multiple ceramide species may occur due to engagement of sphingomyelinases rather than an upregulation of de novo ceramide biosynthesis as the sphingomyelinases do not seem to be chain length specific. This highlights that the types of changes to endogenous ceramide levels may be driven by situationally unique molecular and cellular pathways. Furthermore, rather than changes to unique ceramide species there is emerging evidence that alterations in ratios of long-chain to very long-chain ceramide species may be more relevant to disease pathology [122]. This further underscores the cellular and molecular complexity associated with alterations to ceramide metabolism and its pathological or therapeutic relevance.
Interestingly a related sphingolipid, S1P, is highly bioactive but acts to achieve the opposite physiological effect of ceramide, promoting cell division and survival. More specifically, ceramide has antitumor therapeutic potential through its role in the stimulation of both mitochondrial mediated and death-receptor mediated apoptosis, as well as other mechanisms. The question of how to exploit the effect of endogenous ceramides is yet to be definitively answered. Some drugs, as mentioned, have aimed to control the metabolism of ceramide. The result of these therapies is an increased abundance of intracellular ceramide. They do this primarily by downregulating ceramide neutralization enzymes, which are responsible for the conversion of ceramide to non-ceramide sphingolipids. Within this category of therapeutics, therapies that aim to downregulate the activity of S1P, such as the numerous acid CDase and SPHK inhibitors or the anti-S1P monoclonal antibody Sonepcizumab, can be considered as they act to manipulate sphingolipid metabolism in an anticancer fashion. As S1P’s activity yields an opposite physiological effect of ceramide, the antagonism of S1PRs may also present therapeutic potential. However, the generation of S1P is not the sole metabolic outlet that malignant cells can use to neutralize ceramide. Moreover, the diversity of ceramide metabolic outlets may present a unique challenge to ceramide-based anticancer therapy due to a range of adaptive resistance that could develop [12,123].
Aside from pharmacological manipulations of sphingolipid metabolism, other approaches instead focus on introducing exogenous ceramide to cancer tissues. Although the goal of ceramide elevation is shared, these are distinct as delivery systems. In many ways, this is a more direct strategy to raise cellular levels of ceramide. However, there are issues that need to be solved before these ceramide-delivering therapies can be brought to the bedside. For instance, some therapies have had difficulty effectively targeting and delivering exogenous ceramide to cancer cells. The development of delivery systems such as the ceramide nanoliposome and the ceramide-loaded calcium phosphosilicate nanoparticle provide optimism for the next generation of cancer therapeutics as this platform is passively targeted by EPR, could be actively targeted, but importantly can encapsulate and protect ceramide during systemic delivery to exert limited off-target toxicity and maximal anticancer efficacy [95–98]. Most notably, the ceramide nanoliposome has completed a phase I clinical trial with solid tumor patients and no dose-limiting toxicities were observed despite evaluating an incredibly high dose of 323 mg/m2 [97]. Although a promising field, ceramide-based therapies are continuing to improve and may eventually be best suited as combinatorial therapies alongside standard of care therapies. Improvements to these delivery systems will further reduce off-target toxicity, improve targeting, and co-load therapeutics, as to improve overall therapeutic efficacy. It is hoped that in time these novel ceramide-based therapies will efficiently treat a variety of malignancies.
Acknowledgements
This work was funded in part by grants from the National Cancer Institute of the National Institutes of Health to BMB (K22-CA190674, R03-CA252825, and P01-CA171983 via subaward from the University of Virginia) and to TJB (T32-CA009679, L30-CA274783),, a pilot project grant to BMB as part of University of New Hampshire COBRE award P20-GM113131 from the National Institute of General Medical Sciences of the National Institutes of Health, as well as support from the University of New Hampshire Hamel Center for Undergraduate Research. BMB is the owner and founder of Tahosa Bio, LLC (Rapid City, SD). Portions of this work appear in the Ph.D. dissertation of WW (https://scholars.unh.edu/dissertation/2600/) at the University of New Hampshire.
References
- 1.Arsenault EJ, McGill CM, Barth BM. Sphingolipids as Regulators of Neuro-Inflammation and NADPH Oxidase 2. Neuromolecular Med. 2021. Mar;23(1):25–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Pinto SN, Laviad EL, Stiban J, Kelly SL, Merrill AH Jr, Prieto M, Futerman AH, Silva LC. Changes in membrane biophysical properties induced by sphingomyelinase depend on the sphingolipid N-acyl chain. J Lipid Res. 2014. Jan;55(1):53–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Koybasi S, Senkal CE, Sundararaj K, Spassieva S, Bielawski J, Osta W, Day TA, Jiang JC, Jazwinski SM, Hannun YA, Obeid LM, Ogretmen B. Defects in cell growth regulation by C18:0-ceramide and longevity assurance gene 1 in human head and neck squamous cell carcinomas. J Biol Chem. 2004. Oct 22;279(43):44311–9. [DOI] [PubMed] [Google Scholar]
- 4.Senkal CE, Ponnusamy S, Bielawski J, Hannun YA, Ogretmen B. Antiapoptotic roles of ceramide-synthase-6-generated C16-ceramide via selective regulation of the ATF6/CHOP arm of ER-stress-response pathways. FASEB J. 2010. Jan;24(1):296–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Wattenberg BW. The long and the short of ceramides. J Biol Chem. 2018. Jun 22;293(25):9922–9923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Kihara A Very long-chain fatty acids: elongation, physiology and related disorders. J Biochem. 2012. Nov;152(5):387–95. [DOI] [PubMed] [Google Scholar]
- 7.Kitatani K, Idkowiak-Baldys J, Hannun YA. The sphingolipid salvage pathway in ceramide metabolism and signaling. Cell Signal. 2008. Jun;20(6):1010–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Kolter T, Sandhoff K. Principles of lysosomal membrane digestion: stimulation of sphingolipid degradation by sphingolipid activator proteins and anionic lysosomal lipids. Annu Rev Cell Dev Biol. 2005;21:81–103. [DOI] [PubMed] [Google Scholar]
- 9.Hanada K, Kumagai K, Yasuda S, Miura Y, Kawano M, Fukasawa M, Nishijima M. Molecular machinery for non-vesicular trafficking of ceramide. Nature. 2003. Dec 18;426(6968):803–9. [DOI] [PubMed] [Google Scholar]
- 10.Maceyka M, Spiegel S. Sphingolipid metabolites in inflammatory disease. Nature. 2014. Jun 5;510(7503):58–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Hussain MM, Jin W, Jiang XC. Mechanisms involved in cellular ceramide homeostasis. Nutr Metab (Lond). 2012. Jul 31;9(1):71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Barth BM, Wang W, Toran PT, Fox TE, Annageldiyev C, Ondrasik RM, Keasey NR, Brown TJ, Devine VG, Sullivan EC, Cote AL, Papakotsi V, Tan SF, Shanmugavelandy SS, Deering TG, Needle DB, Stern ST, Zhu J, Liao J, Viny AD, Feith DJ, Levine RL, Wang HG, Loughran TP Jr, Sharma A, Kester M, Claxton DF. Sphingolipid metabolism determines the therapeutic efficacy of nanoliposomal ceramide in acute myeloid leukemia. Blood Adv. 2019. Sep 10;3(17):2598–2603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Ogretmen B Sphingolipid metabolism in cancer signalling and therapy. Nat Rev Cancer. 2018. Jan;18(1):33–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Wang W, Toran PT, Sabol R, Brown TJ, Barth BM. Epigenetics and Sphingolipid Metabolism in Health and Disease. Int J Biopharm Sci. 2018. Oct;1(2):105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Bieberich E Sphingolipids and lipid rafts: Novel concepts and methods of analysis. Chem Phys Lipids. 2018. Nov;216:114–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Ventura AE, Mestre B, Silva LC. Ceramide Domains in Health and Disease: A Biophysical Perspective. Adv Exp Med Biol. 2019;1159:79–108. [DOI] [PubMed] [Google Scholar]
- 17.Shen CP, Zhao MT, Jia ZX, Zhang JL, Jiao L, Ma L. Skin Ceramide Profile in Children With Atopic Dermatitis. Dermatitis. 2018. Jul/Aug;29(4):219–222. [DOI] [PubMed] [Google Scholar]
- 18.Peters F, Tellkamp F, Brodesser S, Wachsmuth E, Tosetti B, Karow U, Bloch W, Utermöhlen O, Krönke M, Niessen CM. Murine Epidermal Ceramide Synthase 4 Is a Key Regulator of Skin Barrier Homeostasis. J Invest Dermatol. 2020. Oct;140(10):1927–1937.e5. [DOI] [PubMed] [Google Scholar]
- 19.Pareek A, Kumari L, Pareek A, Chaudhary S, Ratan Y, Janmeda P, Chuturgoon S, Chuturgoon A. Unraveling Atopic Dermatitis: Insights into Pathophysiology, Therapeutic Advances, and Future Perspectives. Cells. 2024. Feb 28;13(5):425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Pavel P, Blunder S, Moosbrugger-Martinz V, Elias PM, Dubrac S. Atopic Dermatitis: The Fate of the Fat. Int J Mol Sci. 2022. Feb 14;23(4):2121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Ishida K, Takahashi A, Bito K, Draelos Z, Imokawa G. Treatment with Synthetic Pseudoceramide Improves Atopic Skin, Switching the Ceramide Profile to a Healthy Skin Phenotype. J Invest Dermatol. 2020. Sep;140(9):1762–1770.e8. [DOI] [PubMed] [Google Scholar]
- 22.Shin KO, Lim CJ, Park HY, Kim S, Kim B, Lee Y, Chung H, Jeong SK, Park K, Park K. Activation of SIRT1 Enhances Epidermal Permeability Barrier Formation through Ceramide Synthase 2- and 3-Dependent Mechanisms. J Invest Dermatol. 2020. Jul;140(7):1435–1438.e5. [DOI] [PubMed] [Google Scholar]
- 23.Fang Z, Pyne S, Pyne NJ. Ceramide and sphingosine 1-phosphate in adipose dysfunction. Prog Lipid Res. 2019. Apr;74:145–159. [DOI] [PubMed] [Google Scholar]
- 24.Poss AM, Holland WL, Summers SA. Risky lipids: refining the ceramide score that measures cardiovascular health. Eur Heart J. 2020. Jan 14;41(3):381–382. [DOI] [PubMed] [Google Scholar]
- 25.Turner N, Lim XY, Toop HD, Osborne B, Brandon AE, Taylor EN, Fiveash CE, Govindaraju H, Teo JD, McEwen HP, Couttas TA, Butler SM, Das A, Kowalski GM, Bruce CR, Hoehn KL, Fath T, Schmitz-Peiffer C, Cooney GJ, Montgomery MK, Morris JC, Don AS. A selective inhibitor of ceramide synthase 1 reveals a novel role in fat metabolism. Nat Commun. 2018. Aug 21;9(1):3165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Prévost C, Zhao H, Manzi J, Lemichez E, Lappalainen P, Callan-Jones A, Bassereau P. IRSp53 senses negative membrane curvature and phase separates along membrane tubules. Nat Commun. 2015. Oct 15;6:8529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Murakami K, Tenge VR, Karandikar UC, Lin SC, Ramani S, Ettayebi K, Crawford SE, Zeng XL, Neill FH, Ayyar BV, Katayama K, Graham DY, Bieberich E, Atmar RL, Estes MK. Bile acids and ceramide overcome the entry restriction for GII.3 human norovirus replication in human intestinal enteroids. Proc Natl Acad Sci U S A. 2020. Jan 21;117(3):1700–1710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Corriden R, Hollands A, Olson J, Derieux J, Lopez J, Chang JT, Gonzalez DJ, Nizet V. Tamoxifen augments the innate immune function of neutrophils through modulation of intracellular ceramide. Nat Commun. 2015. Oct 13;6:8369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Drews K, Calgi MP, Harrison WC, Drews CM, Costa-Pinheiro P, Shaw JJP, Jobe KA, Han JD, Fox TE, White JM, Kester M. Glucosylceramide synthase maintains influenza virus entry and infection. PLoS One. 2020. Feb 7;15(2):e0228735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Drews K, Calgi MP, Harrison WC, Drews CM, Costa-Pinheiro P, Shaw JJP, Jobe KA, Nelson EA, Han JD, Fox T, White JM, Kester M. Glucosylceramidase Maintains Influenza Virus Infection by Regulating Endocytosis. J Virol. 2019. May 29;93(12):e00017–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Jeffries KA, Krupenko NI. Ceramide Signaling and p53 Pathways. Adv Cancer Res. 2018;140:191–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Prinetti A, Millimaggi D, D’Ascenzo S, Clarkson M, Bettiga A, Chigorno V, Sonnino S, Pavan A, Dolo V. Lack of ceramide generation and altered sphingolipid composition are associated with drug resistance in human ovarian carcinoma cells. Biochem J. 2006. Apr 15;395(2):311–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Mesicek J, Lee H, Feldman T, Jiang X, Skobeleva A, Berdyshev EV, Haimovitz-Friedman A, Fuks Z, Kolesnick R. Ceramide synthases 2, 5, and 6 confer distinct roles in radiation-induced apoptosis in HeLa cells. Cell Signal. 2010. Sep;22(9):1300–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Moro K, Kawaguchi T, Tsuchida J, Gabriel E, Qi Q, Yan L, Wakai T, Takabe K, Nagahashi M. Ceramide species are elevated in human breast cancer and are associated with less aggressiveness. Oncotarget. 2018. Apr 13;9(28):19874–19890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Rath G, Schneider C, Langlois B, Sartelet H, Morjani H, Btaouri HE, Dedieu S, Martiny L. De novo ceramide synthesis is responsible for the anti-tumor properties of camptothecin and doxorubicin in follicular thyroid carcinoma. Int J Biochem Cell Biol. 2009. May;41(5):1165–72. [DOI] [PubMed] [Google Scholar]
- 36.Min J, Mesika A, Sivaguru M, Van Veldhoven PP, Alexander H, Futerman AH, Alexander S. (Dihydro)ceramide synthase 1 regulated sensitivity to cisplatin is associated with the activation of p38 mitogen-activated protein kinase and is abrogated by sphingosine kinase 1. Mol Cancer Res. 2007. Aug;5(8):801–12. [DOI] [PubMed] [Google Scholar]
- 37.Li S, Wu Y, Ding Y, Yu M, Ai Z. CerS6 regulates cisplatin resistance in oral squamous cell carcinoma by altering mitochondrial fission and autophagy. J Cell Physiol. 2018. Dec;233(12):9416–9425. [DOI] [PubMed] [Google Scholar]
- 38.Dany M, Gencer S, Nganga R, Thomas RJ, Oleinik N, Baron KD, Szulc ZM, Ruvolo P, Kornblau S, Andreeff M, Ogretmen B. Targeting FLT3-ITD signaling mediates ceramide-dependent mitophagy and attenuates drug resistance in AML. Blood. 2016. Oct 13;128(15):1944–1958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Gencer EB, Ural AU, Avcu F, Baran Y. A novel mechanism of dasatinib-induced apoptosis in chronic myeloid leukemia; ceramide synthase and ceramide clearance genes. Ann Hematol. 2011. Nov;90(11):1265–75. [DOI] [PubMed] [Google Scholar]
- 40.Bose R, Verheij M, Haimovitz-Friedman A, Scotto K, Fuks Z, Kolesnick R. Ceramide synthase mediates daunorubicin-induced apoptosis: an alternative mechanism for generating death signals. Cell. 1995. Aug 11;82(3):405–14. [DOI] [PubMed] [Google Scholar]
- 41.Fan S, Niu Y, Tan N, Wu Z, Wang Y, You H, Ke R, Song J, Shen Q, Wang W, Yao G, Shu H, Lin H, Yao M, Zhang Z, Gu J, Qin W. LASS2 enhances chemosensitivity of breast cancer by counteracting acidic tumor microenvironment through inhibiting activity of V-ATPase proton pump. Oncogene. 2013. Mar 28;32(13):1682–90. [DOI] [PubMed] [Google Scholar]
- 42.Huang L, Luan T, Chen Y, Bao X, Huang Y, Fu S, Wang H, Wang J. LASS2 regulates invasion and chemoresistance via ERK/Drp1 modulated mitochondrial dynamics in bladder cancer cells. J Cancer. 2018. Feb 28;9(6):1017–1024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Siddique MM, Bikman BT, Wang L, Ying L, Reinhardt E, Shui G, Wenk MR, Summers SA. Ablation of dihydroceramide desaturase confers resistance to etoposide-induced apoptosis in vitro. PLoS One. 2012;7(9):e44042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Biswal SS, Datta K, Acquaah-Mensah GK, Kehrer JP. Changes in ceramide and sphingomyelin following fludarabine treatment of human chronic B-cell leukemia cells. Toxicology. 2000. Nov 23;154(1–3):45–53. [DOI] [PubMed] [Google Scholar]
- 45.Brachtendorf S, Wanger RA, Birod K, Thomas D, Trautmann S, Wegner MS, Fuhrmann DC, Brüne B, Geisslinger G, Grösch S. Chemosensitivity of human colon cancer cells is influenced by a p53-dependent enhancement of ceramide synthase 5 and induction of autophagy. Biochim Biophys Acta Mol Cell Biol Lipids. 2018. Oct;1863(10):1214–1227. [DOI] [PubMed] [Google Scholar]
- 46.Modrak DE, Leon E, Goldenberg DM, Gold DV. Ceramide regulates gemcitabine-induced senescence and apoptosis in human pancreatic cancer cell lines. Mol Cancer Res. 2009. Jun;7(6):890–6. [DOI] [PubMed] [Google Scholar]
- 47.Baran Y, Salas A, Senkal CE, Gunduz U, Bielawski J, Obeid LM, Ogretmen B. Alterations of ceramide/sphingosine 1-phosphate rheostat involved in the regulation of resistance to imatinib-induced apoptosis in K562 human chronic myeloid leukemia cells. J Biol Chem. 2007. Apr 13;282(15):10922–34. [DOI] [PubMed] [Google Scholar]
- 48.Litvak DA, Bilchik AJ, Cabot MC. Modulators of ceramide metabolism sensitize colorectal cancer cells to chemotherapy: a novel treatment strategy. J Gastrointest Surg. 2003. Jan;7(1):140–148. [DOI] [PubMed] [Google Scholar]
- 49.Fekry B, Esmaeilniakooshkghazi A, Krupenko SA, Krupenko NI. Ceramide Synthase 6 Is a Novel Target of Methotrexate Mediating Its Antiproliferative Effect in a p53-Dependent Manner. PLoS One. 2016. Jan 19;11(1):e0146618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Sridevi P, Alexander H, Laviad EL, Pewzner-Jung Y, Hannink M, Futerman AH, Alexander S. Ceramide synthase 1 is regulated by proteasomal mediated turnover. Biochim Biophys Acta. 2009. Jul;1793(7):1218–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Jensen SA, Calvert AE, Volpert G, Kouri FM, Hurley LA, Luciano JP, Wu Y, Chalastanis A, Futerman AH, Stegh AH. Bcl2L13 is a ceramide synthase inhibitor in glioblastoma. Proc Natl Acad Sci U S A. 2014. Apr 15;111(15):5682–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Mojakgomo R, Mbita Z, Dlamini Z. Linking the ceramide synthases (CerSs) 4 and 5 with apoptosis, endometrial and colon cancers. Exp Mol Pathol. 2015. Jun;98(3):585–92. [DOI] [PubMed] [Google Scholar]
- 53.Wang H, Charles AG, Frankel AJ, Cabot MC. Increasing intracellular ceramide: an approach that enhances the cytotoxic response in prostate cancer cells. Urology. 2003. May;61(5):1047–52. [DOI] [PubMed] [Google Scholar]
- 54.Gustafsson K, Sander B, Bielawski J, Hannun YA, Flygare J. Potentiation of cannabinoid-induced cytotoxicity in mantle cell lymphoma through modulation of ceramide metabolism. Mol Cancer Res. 2009. Jul;7(7):1086–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Hernández-Tiedra S, Fabriàs G, Dávila D, Salanueva ÍJ, Casas J, Montes LR, Antón Z, García-Taboada E, Salazar-Roa M, Lorente M, Nylandsted J, Armstrong J, López-Valero I, McKee CS, Serrano-Puebla A, García-López R, González-Martínez J, Abad JL, Hanada K, Boya P, Goñi F, Guzmán M, Lovat P, Jäättelä M, Alonso A, Velasco G. Dihydroceramide accumulation mediates cytotoxic autophagy of cancer cells via autolysosome destabilization. Autophagy. 2016. Nov;12(11):2213–2229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Schiffmann S, Ziebell S, Sandner J, Birod K, Deckmann K, Hartmann D, Rode S, Schmidt H, Angioni C, Geisslinger G, Grösch S. Activation of ceramide synthase 6 by celecoxib leads to a selective induction of C16:0-ceramide. Biochem Pharmacol. 2010. Dec 1;80(11):1632–40. [DOI] [PubMed] [Google Scholar]
- 57.Yun SH, Park ES, Shin SW, Ju MH, Han JY, Jeong JS, Kim SH, Stonik VA, Kwak JY, Park JI. By activating Fas/ceramide synthase 6/p38 kinase in lipid rafts, stichoposide D inhibits growth of leukemia xenografts. Oncotarget. 2015. Sep 29;6(29):27596–612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Yacoub A, Hamed HA, Allegood J, Mitchell C, Spiegel S, Lesniak MS, Ogretmen B, Dash R, Sarkar D, Broaddus WC, Grant S, Curiel DT, Fisher PB, Dent P. PERK-dependent regulation of ceramide synthase 6 and thioredoxin play a key role in mda-7/IL-24-induced killing of primary human glioblastoma multiforme cells. Cancer Res. 2010. Feb 1;70(3):1120–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Signorelli P, Munoz-Olaya JM, Gagliostro V, Casas J, Ghidoni R, Fabriàs G. Dihydroceramide intracellular increase in response to resveratrol treatment mediates autophagy in gastric cancer cells. Cancer Lett. 2009. Sep 18;282(2):238–43. [DOI] [PubMed] [Google Scholar]
- 60.White-Gilbertson S, Mullen T, Senkal C, Lu P, Ogretmen B, Obeid L, Voelkel-Johnson C. Ceramide synthase 6 modulates TRAIL sensitivity and nuclear translocation of active caspase-3 in colon cancer cells. Oncogene. 2009. Feb 26;28(8):1132–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Chun J, Hartung HP. Mechanism of action of oral fingolimod (FTY720) in multiple sclerosis. Clin Neuropharmacol. 2010. Mar-Apr;33(2):91–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Chen L, Luo LF, Lu J, Li L, Liu YF, Wang J, Liu H, Song H, Jiang H, Chen SJ, Luo C, Li KK. FTY720 induces apoptosis of M2 subtype acute myeloid leukemia cells by targeting sphingolipid metabolism and increasing endogenous ceramide levels. PLoS One. 2014. Jul 22;9(7):e103033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Liang J, Nagahashi M, Kim EY, Harikumar KB, Yamada A, Huang WC, Hait NC, Allegood JC, Price MM, Avni D, Takabe K, Kordula T, Milstien S, Spiegel S. Sphingosine-1-phosphate links persistent STAT3 activation, chronic intestinal inflammation, and development of colitis-associated cancer. Cancer Cell. 2013. Jan 14;23(1):107–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Liao A, Broeg K, Fox T, Tan SF, Watters R, Shah MV, Zhang LQ, Li Y, Ryland L, Yang J, Aliaga C, Dewey A, Rogers A, Loughran K, Hirsch L, Jarbadan NR, Baab KT, Liao J, Wang HG, Kester M, Desai D, Amin S, Loughran TP Jr, Liu X. Therapeutic efficacy of FTY720 in a rat model of NK-cell leukemia. Blood. 2011. Sep 8;118(10):2793–800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Liu Q, Zhao X, Frissora F, Ma Y, Santhanam R, Jarjoura D, Lehman A, Perrotti D, Chen CS, Dalton JT, Muthusamy N, Byrd JC. FTY720 demonstrates promising preclinical activity for chronic lymphocytic leukemia and lymphoblastic leukemia/lymphoma. Blood. 2008. Jan 1;111(1):275–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Saddoughi SA, Gencer S, Peterson YK, Ward KE, Mukhopadhyay A, Oaks J, Bielawski J, Szulc ZM, Thomas RJ, Selvam SP, Senkal CE, Garrett-Mayer E, De Palma RM, Fedarovich D, Liu A, Habib AA, Stahelin RV, Perrotti D, Ogretmen B. Sphingosine analogue drug FTY720 targets I2PP2A/SET and mediates lung tumour suppression via activation of PP2A-RIPK1-dependent necroptosis. EMBO Mol Med. 2013. Jan;5(1):105–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Young MM, Bui V, Chen C, Wang HG. FTY720 induces non-canonical phosphatidylserine externalization and cell death in acute myeloid leukemia. Cell Death Dis. 2019. Nov 7;10(11):847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Neviani P, Harb JG, Oaks JJ, Santhanam R, Walker CJ, Ellis JJ, Ferenchak G, Dorrance AM, Paisie CA, Eiring AM, Ma Y, Mao HC, Zhang B, Wunderlich M, May PC, Sun C, Saddoughi SA, Bielawski J, Blum W, Klisovic RB, Solt JA, Byrd JC, Volinia S, Cortes J, Huettner CS, Koschmieder S, Holyoake TL, Devine S, Caligiuri MA, Croce CM, Garzon R, Ogretmen B, Arlinghaus RB, Chen CS, Bittman R, Hokland P, Roy DC, Milojkovic D, Apperley J, Goldman JM, Reid A, Mulloy JC, Bhatia R, Marcucci G, Perrotti D. PP2A-activating drugs selectively eradicate TKI-resistant chronic myeloid leukemic stem cells. J Clin Invest. 2013. Oct;123(10):4144–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Li MH, Swenson R, Harel M, Jana S, Stolarzewicz E, Hla T, Shapiro LH, Ferrer F. Antitumor Activity of a Novel Sphingosine-1-Phosphate 2 Antagonist, AB1, in Neuroblastoma. J Pharmacol Exp Ther. 2015. Sep;354(3):261–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Britten CD, Garrett-Mayer E, Chin SH, Shirai K, Ogretmen B, Bentz TA, Brisendine A, Anderton K, Cusack SL, Maines LW, Zhuang Y, Smith CD, Thomas MB. A Phase I Study of ABC294640, a First-in-Class Sphingosine Kinase-2 Inhibitor, in Patients with Advanced Solid Tumors. Clin Cancer Res. 2017. Aug 15;23(16):4642–4650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Lewis CS, Voelkel-Johnson C, Smith CD. Suppression of c-Myc and RRM2 expression in pancreatic cancer cells by the sphingosine kinase-2 inhibitor ABC294640. Oncotarget. 2016. Sep 13;7(37):60181–60192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Qin Z, Dai L, Trillo-Tinoco J, Senkal C, Wang W, Reske T, Bonstaff K, Del Valle L, Rodriguez P, Flemington E, Voelkel-Johnson C, Smith CD, Ogretmen B, Parsons C. Targeting sphingosine kinase induces apoptosis and tumor regression for KSHV-associated primary effusion lymphoma. Mol Cancer Ther. 2014. Jan;13(1):154–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Venant H, Rahmaniyan M, Jones EE, Lu P, Lilly MB, Garrett-Mayer E, Drake RR, Kraveka JM, Smith CD, Voelkel-Johnson C. The Sphingosine Kinase 2 Inhibitor ABC294640 Reduces the Growth of Prostate Cancer Cells and Results in Accumulation of Dihydroceramides In Vitro and In Vivo. Mol Cancer Ther. 2015. Dec;14(12):2744–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Venkata JK, An N, Stuart R, Costa LJ, Cai H, Coker W, Song JH, Gibbs K, Matson T, Garrett-Mayer E, Wan Z, Ogretmen B, Smith C, Kang Y. Inhibition of sphingosine kinase 2 downregulates the expression of c-Myc and Mcl-1 and induces apoptosis in multiple myeloma. Blood. 2014. Sep 18;124(12):1915–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Chen J, Goyal N, Dai L, Lin Z, Del Valle L, Zabaleta J, Liu J, Post SR, Foroozesh M, Qin Z. Developing new ceramide analogs and identifying novel sphingolipid-controlled genes against a virus-associated lymphoma. Blood. 2020. Nov 5;136(19):2175–2187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Ponnapakam AP, Liu J, Bhinge KN, Drew BA, Wang TL, Antoon JW, Nguyen TT, Dupart PS, Wang Y, Zhao M, Liu YY, Foroozesh M, Beckman BS. 3-Ketone-4,6-diene ceramide analogs exclusively induce apoptosis in chemo-resistant cancer cells. Bioorg Med Chem. 2014. Feb 15;22(4):1412–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Bieberich E, Kawaguchi T, Yu RK. N-acylated serinol is a novel ceramide mimic inducing apoptosis in neuroblastoma cells. J Biol Chem. 2000. Jan 7;275(1):177–81. [DOI] [PubMed] [Google Scholar]
- 78.Draper JM, Xia Z, Smith RA, Zhuang Y, Wang W, Smith CD. Discovery and evaluation of inhibitors of human ceramidase. Mol Cancer Ther. 2011. Nov;10(11):2052–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Selzner M, Bielawska A, Morse MA, Rüdiger HA, Sindram D, Hannun YA, Clavien PA. Induction of apoptotic cell death and prevention of tumor growth by ceramide analogues in metastatic human colon cancer. Cancer Res. 2001. Feb 1;61(3):1233–40. [PubMed] [Google Scholar]
- 80.Separovic D, Saad ZH, Edwin EA, Bielawski J, Pierce JS, Buren EV, Bielawska A. C16-Ceramide Analog Combined with Pc 4 Photodynamic Therapy Evokes Enhanced Total Ceramide Accumulation, Promotion of DEVDase Activation in the Absence of Apoptosis, and Augmented Overall Cell Killing. J Lipids. 2011;2011:713867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Paschall AV, Zimmerman MA, Torres CM, Yang D, Chen MR, Li X, Bieberich E, Bai A, Bielawski J, Bielawska A, Liu K. Ceramide targets xIAP and cIAP1 to sensitize metastatic colon and breast cancer cells to apoptosis induction to suppress tumor progression. BMC Cancer. 2014. Jan 15;14:24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Beckham TH, Lu P, Jones EE, Marrison T, Lewis CS, Cheng JC, Ramshesh VK, Beeson G, Beeson CC, Drake RR, Bielawska A, Bielawski J, Szulc ZM, Ogretmen B, Norris JS, Liu X. LCL124, a cationic analog of ceramide, selectively induces pancreatic cancer cell death by accumulating in mitochondria. J Pharmacol Exp Ther. 2013. Jan;344(1):167–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Elojeimy S, Liu X, McKillop JC, El-Zawahry AM, Holman DH, Cheng JY, Meacham WD, Mahdy AE, Saad AF, Turner LS, Cheng J, A Day T, Dong JY, Bielawska A, Hannun YA, Norris JS. Role of acid ceramidase in resistance to FasL: therapeutic approaches based on acid ceramidase inhibitors and FasL gene therapy. Mol Ther. 2007. Jul;15(7):1259–63. [DOI] [PubMed] [Google Scholar]
- 84.Tan SF, Liu X, Fox TE, Barth BM, Sharma A, Turner SD, Awwad A, Dewey A, Doi K, Spitzer B, Shah MV, Morad SA, Desai D, Amin S, Zhu J, Liao J, Yun J, Kester M, Claxton DF, Wang HG, Cabot MC, Schuchman EH, Levine RL, Feith DJ, Loughran TP Jr. Acid ceramidase is upregulated in AML and represents a novel therapeutic target. Oncotarget. 2016. Dec 13;7(50):83208–83222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Mahdy AE, Cheng JC, Li J, Elojeimy S, Meacham WD, Turner LS, Bai A, Gault CR, McPherson AS, Garcia N, Beckham TH, Saad A, Bielawska A, Bielawski J, Hannun YA, Keane TE, Taha MI, Hammouda HM, Norris JS, Liu X. Acid ceramidase upregulation in prostate cancer cells confers resistance to radiation: AC inhibition, a potential radiosensitizer. Mol Ther. 2009. Mar;17(3):430–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Korbelik M, Banáth J, Zhang W, Saw KM, Szulc ZM, Bielawska A, Separovic D. Interaction of acid ceramidase inhibitor LCL521 with tumor response to photodynamic therapy and photodynamic therapy-generated vaccine. Int J Cancer. 2016. Sep 15;139(6):1372–8. [DOI] [PubMed] [Google Scholar]
- 87.Powell JA, Lewis AC, Zhu W, Toubia J, Pitman MR, Wallington-Beddoe CT, Moretti PA, Iarossi D, Samaraweera SE, Cummings N, Ramshaw HS, Thomas D, Wei AH, Lopez AF, D’Andrea RJ, Lewis ID, Pitson SM. Targeting sphingosine kinase 1 induces MCL1-dependent cell death in acute myeloid leukemia. Blood. 2017. Feb 9;129(6):771–782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Ju T, Gao D, Fang ZY. Targeting colorectal cancer cells by a novel sphingosine kinase 1 inhibitor PF-543. Biochem Biophys Res Commun. 2016. Feb 12;470(3):728–734. [DOI] [PubMed] [Google Scholar]
- 89.Schnute ME, McReynolds MD, Kasten T, Yates M, Jerome G, Rains JW, Hall T, Chrencik J, Kraus M, Cronin CN, Saabye M, Highkin MK, Broadus R, Ogawa S, Cukyne K, Zawadzke LE, Peterkin V, Iyanar K, Scholten JA, Wendling J, Fujiwara H, Nemirovskiy O, Wittwer AJ, Nagiec MM. Modulation of cellular S1P levels with a novel, potent and specific inhibitor of sphingosine kinase-1. Biochem J. 2012. May 15;444(1):79–88. [DOI] [PubMed] [Google Scholar]
- 90.Pearson JM, Tan SF, Sharma A, Annageldiyev C, Fox TE, Abad JL, Fabrias G, Desai D, Amin S, Wang HG, Cabot MC, Claxton DF, Kester M, Feith DJ, Loughran TP. Ceramide Analogue SACLAC Modulates Sphingolipid Levels and MCL-1 Splicing to Induce Apoptosis in Acute Myeloid Leukemia. Mol Cancer Res. 2020. Mar;18(3):352–363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Kapitonov D, Allegood JC, Mitchell C, Hait NC, Almenara JA, Adams JK, Zipkin RE, Dent P, Kordula T, Milstien S, Spiegel S. Targeting sphingosine kinase 1 inhibits Akt signaling, induces apoptosis, and suppresses growth of human glioblastoma cells and xenografts. Cancer Res. 2009. Sep 1;69(17):6915–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Hengst JA, Dick TE, Sharma A, Doi K, Hegde S, Tan SF, Geffert LM, Fox TE, Sharma AK, Desai D, Amin S, Kester M, Loughran TP, Paulson RF, Claxton DF, Wang HG, Yun JK. SKI-178: A Multitargeted Inhibitor of Sphingosine Kinase and Microtubule Dynamics Demonstrating Therapeutic Efficacy in Acute Myeloid Leukemia Models. Cancer Transl Med. 2017. Jul-Aug;3(4):109–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Pal SK, Drabkin HA, Reeves JA, Hainsworth JD, Hazel SE, Paggiarino DA, Wojciak J, Woodnutt G, Bhatt RS. A phase 2 study of the sphingosine-1-phosphate antibody sonepcizumab in patients with metastatic renal cell carcinoma. Cancer. 2017. Feb 15;123(4):576–582. [DOI] [PubMed] [Google Scholar]
- 94.Kennedy PC, Zhu R, Huang T, Tomsig JL, Mathews TP, David M, Peyruchaud O, Macdonald TL, Lynch KR. Characterization of a sphingosine 1-phosphate receptor antagonist prodrug. J Pharmacol Exp Ther. 2011. Sep;338(3):879–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Kester M, Bassler J, Fox TE, Carter CJ, Davidson JA, Parette MR. Preclinical development of a C6-ceramide NanoLiposome, a novel sphingolipid therapeutic. Biol Chem. 2015. Jun;396(6–7):737–47. [DOI] [PubMed] [Google Scholar]
- 96.Adair JH, Parette MP, Altinoğlu EI, Kester M. Nanoparticulate alternatives for drug delivery. ACS Nano. 2010. Sep 28;4(9):4967–70. [DOI] [PubMed] [Google Scholar]
- 97.Ciner A, Gourdin T, Davidson J, Parette M, Walker SJ, Fox TE, Jiang Y. A phase I study of the ceramide nanoliposome in patients with advanced solid tumors. Cancer Chemother Pharmacol. 2024. Jan;93(1):23–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Kester M, Heakal Y, Fox T, Sharma A, Robertson GP, Morgan TT, Altinoğlu EI, Tabaković A, Parette MR, Rouse SM, Ruiz-Velasco V, Adair JH. Calcium phosphate nanocomposite particles for in vitro imaging and encapsulated chemotherapeutic drug delivery to cancer cells. Nano Lett. 2008. Dec;8(12):4116–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Stover T, Kester M. Liposomal delivery enhances short-chain ceramide-induced apoptosis of breast cancer cells. J Pharmacol Exp Ther. 2003. Nov;307(2):468–75. [DOI] [PubMed] [Google Scholar]
- 100.Battogtokh G, Ko YT. Self-assembled chitosan-ceramide nanoparticle for enhanced oral delivery of paclitaxel. Pharm Res. 2014. Nov;31(11):3019–30. [DOI] [PubMed] [Google Scholar]
- 101.Battogtokh G, Ko YT. Self-assembled polymeric nanoparticle of PEGylated chitosan-ceramide conjugate for systemic delivery of paclitaxel. J Drug Target. 2014. Nov;22(9):813–21. [DOI] [PubMed] [Google Scholar]
- 102.Graf N, Bielenberg DR, Kolishetti N, Muus C, Banyard J, Farokhzad OC, Lippard SJ. α(V)β(3) integrin-targeted PLGA-PEG nanoparticles for enhanced anti-tumor efficacy of a Pt(IV) prodrug. ACS Nano. 2012. May 22;6(5):4530–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Balakrishnan P, Song CK, Jahn A, Cho HJ. Ceramide and N,N,N-Trimethylphytosphingosine-Iodide (TMP-I)-Based Lipid Nanoparticles for Cancer Therapy. Pharm Res. 2016. Jan;33(1):206–16. [DOI] [PubMed] [Google Scholar]
- 104.Fujiwara K, Yazama H, Donishi R, Koyama S, Fukuhara T, Kitatani K, Kataoka H, Takeuchi H. C6-ceramide Inhibits the Motility of Anaplastic Thyroid Carcinoma Cells. Yonago Acta Med. 2020. Mar 23;63(2):95–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Bai AP, Guo Y. Ceramide is a Potential Activator of Immune Responses Against Tumors. Gastroenterology. 2018. Aug;155(2):579–580. [DOI] [PubMed] [Google Scholar]
- 106.Moro K, Nagahashi M, Gabriel E, Takabe K, Wakai T. Clinical application of ceramide in cancer treatment. Breast Cancer. 2019. Jul;26(4):407–415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Barth BM, Brown TJ, Adams MT, Garcia AM, Fisher LN, Fritz JL, Beck AJ, McGill CM, Kester M, Tran MA, Claxton DF. Combinatorial Efficacy of Nanoliposomal Ceramide and the Antioxidant 7,8-Benzoflavone for Acute Myeloid Leukemia. J Leuk (Los Angel). 2014. Sep;2(4):152. [Google Scholar]
- 108.Yu T, Li J, Sun H. C6 ceramide potentiates curcumin-induced cell death and apoptosis in melanoma cell lines in vitro. Cancer Chemother Pharmacol. 2010. Oct;66(5):999–1003. [DOI] [PubMed] [Google Scholar]
- 109.Dhule SS, Penfornis P, He J, Harris MR, Terry T, John V, Pochampally R. The combined effect of encapsulating curcumin and C6 ceramide in liposomal nanoparticles against osteosarcoma. Mol Pharm. 2014. Feb 3;11(2):417–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Fonseca NA, Gomes-da-Silva LC, Moura V, Simões S, Moreira JN. Simultaneous active intracellular delivery of doxorubicin and C6-ceramide shifts the additive/antagonistic drug interaction of non-encapsulated combination. J Control Release. 2014. Dec 28;196:122–31. [DOI] [PubMed] [Google Scholar]
- 111.Jiang Y, DiVittore NA, Kaiser JM, Shanmugavelandy SS, Fritz JL, Heakal Y, Tagaram HR, Cheng H, Cabot MC, Staveley-O’Carroll KF, Tran MA, Fox TE, Barth BM, Kester M. Combinatorial therapies improve the therapeutic efficacy of nanoliposomal ceramide for pancreatic cancer. Cancer Biol Ther. 2011. Oct 1;12(7):574–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Doshi UA, Shaw J, Fox TE, Claxton DF, Loughran TP, Kester M. STAT3 mediates C6-ceramide-induced cell death in chronic lymphocytic leukemia. Signal Transduct Target Ther. 2017. Oct 27;2:17051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Ganta S, Singh A, Patel NR, Cacaccio J, Rawal YH, Davis BJ, Amiji MM, Coleman TP. Development of EGFR-targeted nanoemulsion for imaging and novel platinum therapy of ovarian cancer. Pharm Res. 2014. Sep;31(9):2490–502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Carvalho VFM, Migotto A, Giacone DV, de Lemos DP, Zanoni TB, Maria-Engler SS, Costa-Lotufo LV, Lopes LB. Co-encapsulation of paclitaxel and C6 ceramide in tributyrin-containing nanocarriers improve co-localization in the skin and potentiate cytotoxic effects in 2D and 3D models. Eur J Pharm Sci. 2017. Nov 15;109:131–143. [DOI] [PubMed] [Google Scholar]
- 115.Tan KB, Ling LU, Bunte RM, Chng WJ, Chiu GN. Liposomal codelivery of a synergistic combination of bioactive lipids in the treatment of acute myeloid leukemia. Nanomedicine (Lond). 2014. Aug;9(11):1665–79. [DOI] [PubMed] [Google Scholar]
- 116.Brown TJ, Garcia AM, Kissinger LN, Shanmugavelandy SS, Wang X, Cabot MC, Kester M, Claxton DF, Barth BM. Therapeutic Combination of Nanoliposomal Safingol and Nanoliposomal Ceramide for Acute Myeloid Leukemia. J Leuk (Los Angel). 2013. Apr;1(2):110. [Google Scholar]
- 117.Tran MA, Smith CD, Kester M, Robertson GP. Combining nanoliposomal ceramide with sorafenib synergistically inhibits melanoma and breast cancer cell survival to decrease tumor development. Clin Cancer Res. 2008. Jun 1;14(11):3571–81. [DOI] [PubMed] [Google Scholar]
- 118.Barth BM, Keasey NR, Wang X, Shanmugavelandy SS, Rampal R, Hricik T, Cabot MC, Kester M, Wang HG, Shultz LD, Tallman MS, Levine RL, Loughran TP Jr, Claxton DF. Engraftment of Human Primary Acute Myeloid Leukemia Defined by Integrated Genetic Profiling in NOD/SCID/IL2rγnull Mice for Preclinical Ceramide-Based Therapeutic Evaluation. J Leuk (Los Angel). 2014. Sep;2(3):146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Morad SA, Ryan TE, Neufer PD, Zeczycki TN, Davis TS, MacDougall MR, Fox TE, Tan SF, Feith DJ, Loughran TP Jr, Kester M, Claxton DF, Barth BM, Deering TG, Cabot MC. Ceramide-tamoxifen regimen targets bioenergetic elements in acute myelogenous leukemia. J Lipid Res. 2016. Jul;57(7):1231–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Khokhlatchev AV, Sharma A, Deering TG, Shaw JJP, Costa-Pinheiro P, Golla U, Annageldiyev C, Cabot MC, Conaway MR, Tan SF, Ung J, Feith DJ, Loughran TP Jr, Claxton DF, Fox TE, Kester M. Ceramide nanoliposomes augment the efficacy of venetoclax and cytarabine in models of acute myeloid leukemia. FASEB J. 2022. Oct;36(10):e22514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Adiseshaiah PP, Clogston JD, McLeland CB, Rodriguez J, Potter TM, Neun BW, Skoczen SL, Shanmugavelandy SS, Kester M, Stern ST, McNeil SE. Synergistic combination therapy with nanoliposomal C6-ceramide and vinblastine is associated with autophagy dysfunction in hepatocarcinoma and colorectal cancer models. Cancer Lett. 2013. Sep 1;337(2):254–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Tippetts TS, Holland WL, Summers SA. The ceramide ratio: a predictor of cardiometabolic risk. J Lipid Res. 2018. Sep;59(9):1549–1550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Barth BM. Ceramide: improving Bcl-2 inhibitor therapy. Blood. 2022. Jun 30;139(26):3676–3678. [DOI] [PMC free article] [PubMed] [Google Scholar]