Sphingosine metabolism as a therapeutic target in cutaneous melanoma

Sphingosine metabolism as a therapeutic target in cutaneous melanoma

Accepted Manuscript Sphingosine metabolism as a therapeutic target in cutaneous melanoma Mohammed Dany PII: S1931-5244(17)30099-3 DOI: 10.1016/j.tr...

2MB Sizes 0 Downloads 125 Views

Accepted Manuscript Sphingosine metabolism as a therapeutic target in cutaneous melanoma Mohammed Dany PII:

S1931-5244(17)30099-3

DOI:

10.1016/j.trsl.2017.04.005

Reference:

TRSL 1148

To appear in:

Translational Research

Received Date: 15 February 2017 Revised Date:

26 March 2017

Accepted Date: 25 April 2017

Please cite this article as: Dany M, Sphingosine metabolism as a therapeutic target in cutaneous melanoma, Translational Research (2017), doi: 10.1016/j.trsl.2017.04.005. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT

Sphingosine metabolism as a therapeutic target in cutaneous melanoma Mohammed Dany College of Medicine, Medical University of South Carolina, Charleston, SC

Abbreviated Title: Sphingolipids in melanoma

SC

Featured New Investigator Submission

RI PT

* Corresponding author at: Medical University of South Carolina, Charleston, SC 29425, USA. Tel: +1 843-489-1599; E-mail address: [email protected].

Keywords: Ceramide; Sphingosine-1-phosphate; melanoma; lipid signaling; sphingolipids

AC C

EP

TE D

M AN U

Conflict of interest: none

ACCEPTED MANUSCRIPT

Abstract Melanoma is by far the most aggressive type of skin cancer with a poor prognosis in its advanced stages. Understanding the mechanisms involved in melanoma pathogenesis,

RI PT

response and resistance to treatment has gained a lot of attention worldwide. Recently, the role of sphingolipid metabolism has been studied in cutaneous melanoma. Sphingolipids are bioactive lipid effector molecules involved in the regulation of various

SC

cellular signaling pathways such as inflammation, cancer cell proliferation, death, senescence, and metastasis. Recent studies suggest that sphingolipid metabolism

M AN U

impacts melanoma pathogenesis and is a potential therapeutic target. This review focuses on defining the role of sphingolipid metabolism in melanoma carcinogenesis, discussing sphingolipid-based therapeutic approaches, and highlighting the areas that

AC C

EP

TE D

require more extensive research.

ACCEPTED MANUSCRIPT

1. Introduction According to the National Cancer Institute, the incidence of invasive melanoma in the United States was estimated to be about 73,870 cases in 2015, and one American dies

RI PT

of melanoma every hour (1). Melanoma treatment depends on the stage of the cancer. Early lesions are often cured by surgical excision alone, while more advanced disease requires systemic therapy (2). The 10-year overall survival rate for advanced melanoma

SC

is improving, but is still only 10-15% (3,4). One challenge in the field of melanoma is understanding the impact of the different metabolic pathways on melanoma

M AN U

pathogenesis. In particular, it is important to understand the role of the metabolism of cancer related bioactive molecules in melanoma pathogenesis. This will help in developing novel therapeutic strategies that target the metabolism of such bioactive molecules.

TE D

Advances in sphingolipid research are continuously proving the impact of sphingolipid metabolism on cancer pathogenesis (5). Sphingolipids are membrane lipids that were discovered for their important functions in regulating membrane fluidity and subdomain

EP

structures. Indeed, sphingolipids have long been recognized as important structural components of the epidermis, securing the epidermal permeability barrier (6). Recently,

AC C

it was revealed that sphingolipids have signaling roles apart from their structural roles. In particular, several sphingolipid species were shown to act as bioactive signaling lipids in cancer cells. Ceramide is one of the most bioactive sphingolipids that plays a key role in many of the cellular functions including cell proliferation, death, migration, and senescence (7). Ceramides account for 30–40% of stratum corneum lipids (8). Ceramide metabolism is intimately involved in cancer pathogenesis. Ceramide and its

ACCEPTED MANUSCRIPT

metabolites are not only involved in cancer initiation and progression, but also in the response of cancer cells to chemotherapeutic agents and radiation induced cell death. Unlike ceramide, Sphingosine 1 Phosphate (S1P) is a tumor promoting bioactive

RI PT

sphingolipid. The intricate balance between the levels of ceramide and S1P dictates whether a cancer cell undergoes proliferation or cell death. This is known as the ceramide-S1P rheostat and can be targeted by several approaches to sway the balance

SC

towards ceramide generation or inhibition of S1P synthesis, to result in cancer cell death. In this review article, we discuss the role of sphingolipid metabolism in the tumor

target sphingolipid metabolism.

M AN U

pathogenesis of melanoma and the different potential therapeutic approaches that

2. Metabolism of biologically active sphingolipids: ceramide and S1P Ceramide is composed of a sphingosine backbone esterified to a fatty acyl chain via an

TE D

amide linkage (5,7). The length of the fatty acyl chain conjugated to the sphingosine backbone characterizes the different species of ceramide. For instance, C18-ceramide

ceramides (5).

EP

contains a fatty acyl chain with n=18 carbons. Ceramide species range from C14- to C26-

AC C

Ceramide lies at the center of sphingolipid metabolism. It can serve as a precursor for more complex sphingolipids, or as a product of the breakdown of other sphingolipids (Figure 1) (5). For example, ceramide acts as a precursor for the generation of glucosylceramides and as a product of the breakdown of sphingomyelin (9). De novo generation of ceramide involves the action of ceramide synthases 1-6 (CerS16). CerS1-6 catalyze the reaction of esterifying a fatty acyl CoA to dihydrosphingosine to

ACCEPTED MANUSCRIPT

generate dihydroceramide (10). Ceramide is then generated by the action of desaturase enzyme which inserts a double bond between carbons 4 and 5 (11).

RI PT

The catabolism of ceramide starts with the action of the enzyme ceramidase yielding sphingosine. Sphingosine then gets phosphorylated by sphingosine kinases 1 or 2 (SK1 SK2)

to

generate

dephosphorylated

sphingosine

by

S1P

1-phosphate

phosphatases

or

ethanolaminephosphate by S1P lyase (12) (Figure 1).

(S1P). lysed

S1P

to

then

is

hexadecanal

either and

SC

or

M AN U

The subcellular compartmentalization of sphingolipid metabolism is important for the biological function of sphingolipid molecules. The compartmentalization is based on the localization of sphingolipid metabolic enzymes (5,13). For example, de novo ceramide synthesis occurs in the endoplasmic reticulum (ER) because CerS1-6 enzymes are localized to the ER. For the synthesis of SM, ceramide is transported from the ER to the

TE D

Golgi apparatus by ceramide transporter protein (CERT) without the help of vesicles (14,15). Similarly, for the synthesis of glucosylceramide (GlcCer), ceramide is

EP

transported to the Golgi by Fabb2 transporter (15). Ceramide is also detected in mitochondria due to either de novo synthesis or sphingomyelin degradation (5). In

AC C

addition, Sphingosine kinase enzymes 1 and 2 differ in their localization: SK1 is mainly cytosolic whilst SK2 is mainly nuclear. Thus, it is believed that S1P plays different roles depending on whether it is generated by SK1 or SK2 (16).

ACCEPTED MANUSCRIPT

3. Biological roles of ceramide in cancer and melanoma pathogenesis

a. Biological functions of Ceramide Synthases 1-6

RI PT

Ceramide Synthases 1-6 have different subcellular localizations and exhibit specificity for the generation of endogenous ceramides with different fatty acid chain lengths (17,18). For example, CerS1 mainly generates C18-ceramide, CerS4 generates both

SC

C18-ceramide and C20-ceramide, and CerS5 and CerS6 generate mainly C16-ceramide, and to a lesser extent, C12- and C14- ceramides. CerS2 and CerS3 are known to

M AN U

generate very long chain C22-24- and C26- ceramides, respectively (5,7,9,19-21). In addition, ceramide synthases are expressed differently in human tissues. For example, CerS1 is mainly expressed in brain and skeletal muscle tissues (22). On the other hand, CerS3 is only expressed in skin and testicular tissues (23,24). CerS4 is also

TE D

expressed in the skin as well as in leukocytes, cardiomyocytes, and hepatocytes (25). Different species of ceramide with distinct fatty acyl chain lengths have diverse biological functions (Table 1). For example, CerS1 and CerS6, generating C18-ceramide

EP

and C16-ceramide respectively, have opposing roles in cell death and proliferation. CerS1/C18-ceramide axis leads to cancer cell death and decreases tumor growth (26-

AC C

29). Increased levels of serum C18-ceramide act as a potential biomarker to monitor patients’ response to chemotherapy (30). On the other hand, C16-ceramide can promote cancer cell proliferation and its increased serum levels associate with a positive lymph node status (31,32). On the other hand, there are studies showing that C16-ceramide is pro-apoptotic, whereas C24-ceramide protects from cell death (33,34).

ACCEPTED MANUSCRIPT

In vivo, Ceramide Synthases play different roles and knocking down their expression result in different phenotypes. Mice expressing a catalytically inactive mutant of CerS1 (toppler mice) exhibit neurological disorders secondary to Purkinjean cells dysfunction.

RI PT

Mice with a knockout in CerS2 suffer from liver cirrhosis. Mice with CerS4 knockout have severe alopecia secondary to ceramide content alterations in the sebum. Finally, mice with CerS6 knockout exhibit behavioral alterations (35-37).

SC

The role of CerSs in melanoma is not extensively studied yet. One study investigated

M AN U

the role of CerS6 on the malignant behavior of melanoma (38). The expression of CerS6 was decreased in several melanoma cell lines and the lower expression was correlated with malignant potential. Silencing the expression of CerS6 in three melanoma cell lines resulted in more proliferation and invasion. Silencing of CerS6 induced the expression of GLUT1 (Glucose transporter 1), which in turn downregulated

TE D

the expression of WNT5A, a growth factor that enhances the invasion and proliferation of melanoma cells (38). Thus, in melanoma, CerS6 acts as a tumor suppressor protein

EP

whose overexpression can provide a therapeutic strategy for melanoma treatment.

AC C

b. Ceramide as a tumor suppressor lipid

It is well established that ceramide is a pro-cell death molecule in several cancer models (22). Ceramide can trigger several programmed cell death mechanisms such as apoptosis, necroptosis, lethal autophagy, and lethal mitophagy (22,39-44). In fact, ceramide levels increase during stress like hypoxia, growth factor withdrawal, or DNA

ACCEPTED MANUSCRIPT

damage (45). Ceramide can then associate with death-associated proteins to trigger programmed cell death.

RI PT

There are several mechanisms by which ceramide acts as a tumor suppressor lipid. Ceramide binds and inactivates the inhibitor of protein phosphatase 2A (46). PP2A is itself a tumor suppressor protein (47,48). Ceramide binds to the biological inhibitor of PP2A, I2PP2A or SET oncoprotein, leading to PP2A reactivation. PP2A can then

SC

0inactivate anti-apoptotic proteins such as Bcl-2 (49,50).

M AN U

Another mechanism is that ceramide regulates human telomerase. Ceramide accumulation in cancer cells inhibits inactivates c-myc transcription factor and thus decreases telomerase expression. This results in less elongation of chromosomal telomeric ends after each replication cycle, leading eventually to senescence and cell

TE D

death (51,52).

Recently, a novel mechanism was discovered explaining ceramide’s role in mediating cell death by attaching the mitochondria (53). In response to tyrosine kinase inhibitor

EP

treatment, ceramide localized to mitochondria of cancer cells and served as a receptor to anchor autophagy related protein LC3B-II and to thus recruit autophagosomes.

AC C

Ceramide localization to mitochondria was mediated by dynamin related protein 1 (Drp1) activation resulting in mitochondrial fission. Inhibition of Drp1 prevented ceramide-dependent lethal mitophagy and reconstitution of WT-Drp1 restored mitochondrial fission and mitophagy (39,54). In addition to tumor suppression, ceramide plays a role in suppressing metastasis by regulating integrin cell surface expression. In particular, treating cancer cells with the

ACCEPTED MANUSCRIPT

short chain C6-ceramide resulted in decreased expression of cell surface β1 and β4 integrin subunits and αVβ6 integrin (55). Lower levels of integrin resulted in diminished cellular adhesion, reduced cellular migration, and suppression of tumor

RI PT

metastasis (55).

In the context of cutaneous melanoma, few studies have confirmed that upregulation of ceramide generation is required for melanoma induced cell death and better response

SC

to treatment. Treatment with exogenous ceramide, such as liposomal C6 ceramide, was

M AN U

cytotoxic and anti-proliferative against a panel of five human melanoma cell lines. Liposomal C6 ceramide induced caspase-dependent apoptotic cell death by activating protein phosphatase 1 (PP1) (56). In addition, treatment of melanoma cancer cells with nanoliposomal C6-ceramide for only 30 minutes suppressed cell migration and decreased tumor extravasation under shear conditions. This was secondary to reducing

TE D

integrin affinity and activating PI3K and PKCζ tumor-suppressive activities (57). Moreover, ceramide plays an important role in the metastasis of melanoma. Metastatic

EP

dissemination of melanoma cancer cells is one of the hallmarks of melanoma malignancy and accounts for most of melanoma cancer deaths. Melanoma cells in the

AC C

blood vasculature may aggregate with platelets to form clots and then adhere to endothelial cells in order to finally extravasate to form metastatic niches. Interestingly, the interaction between melanoma cells and platelets results in the activation of acid sphingomyelinase and the release of ceramide. Acid sphingomyelinase activation was shown to be required for trapping of melanoma cells in the lungs of mice. Indeed, preincubation of tumor cells with recombinant acid sphingomyelinase resulted in melanoma cell colony formation in the lung of mice (58).

RI PT

ACCEPTED MANUSCRIPT

4. Acid ceramidase is implicated in melanoma pathogenesis

Acid ceramidase is an enzyme localized to lysosomes and catalyzes the degradation of

SC

ceramide into sphingosine, thus lowering ceramide levels. One study found that melanoma cells generate lower amounts of ceramides compared with normal

M AN U

melanocytes due to suppression of ceramide de novo synthesis pathways (59). In addition, the study found that ceramide hydrolysis is increased in melanoma cancer cells. The expression of acid ceramidase, an enzyme that lowers the levels of ceramides, is significantly elevated in melanoma cell lines compared with non-

TE D

cancerous skin cells. Acid ceramidase expression is also elevated in biopsies from human subjects with Stage II melanoma. Interestingly, treatment with acid ceramidase inhibitor resulted in increased ceramide levels and induction of cell death (59). The

EP

results of this study suggest that the carcinogenesis of melanoma might involve decreasing intracellular ceramide levels via decreasing its production and increasing its

AC C

degradation by acid ceramidase. Another study found that Dacarbazine, a chemotherapy agent used for the treatment of melanoma, induces cathepsin B-mediated degradation of acid ceramidase in human melanoma cells. Indeed, Dacarbazine elicited a time and dose dependent decrease of acid ceramidase activity and an increase of intracellular ceramide levels (60). The downregulation of acid ceramidase was required for the melanoma cells to respond to

ACCEPTED MANUSCRIPT

Dacabazine as inducible overexpression of acid ceramidase reduced ceramide levels and conferred resistance to Dacarbazine. The study suggests that increasing ceramide levels, secondary to degradation of acid ceramidase, is required to induce cell death in

RI PT

melanoma cells treated with Dacarbazine (60).

Moreover, a recent study discussed the expression and localization of acid ceramidase in melanoma (59). The findings of this study supported that acid ceramidase is

SC

overexpressed in melanoma resulting in both: lower levels of ceramide and increasing

M AN U

levels of S1P. Interestingly, the localization of acid ceramidase is different in melanocytes vs melanoma cells. In melanocytes, acid ceramidase is localized to both the cytosol and the nucleus, while in melanoma cells it is primarily found in the cytosol (59). This study adds evidence to the role of acid ceramidase in controlling sphingolipid

TE D

metabolism in melanoma and thus melanoma proliferation.

5. Roles of SK in cancer and melanoma pathogenesis

EP

a. Sphingosine Kinases and S1P in cancer biology and drug resistance

AC C

The ceramide/S1P rheostat impacts the fate of a cancer cell (Figure 2). Stressful stimuli such as radiation or chemotherapy treatment shift the balance towards ceramide to halt cell proliferation and promote cell death (5). However, in some cases, there is increased activity in Sphingosine Kinases, enzymes that will convert ceramide into S1P. The shift in the balance to S1P results in pro-survival, anti-apoptosis, and resistance to radiation or chemotherapy (20).

ACCEPTED MANUSCRIPT

Sphingosine kinases (SK1 and SK2) are lipid kinases, encoded by SPHK1 and SPHK2 genes in humans (61). SK1 and SK2 differ structurally by the presence of a transmembrane domain in SK2 that is not found in SK1. In addition, SK1 and SK2

RI PT

expression is tissue specific whereby SK1 is more abundant in lung and spleen tissues, whilst SK2 is more expressed in liver and heart tissues. Intracellularly, SK1 and SK2 seem to localize to different organelles, whereby SK1 is found mainly in the cytosol

SC

whereas SK2 can be detected in both cytoplasm and nucleus (61,62).

M AN U

The importance of SK enzymes to human diseases is revealed in the knockout phenotypes; even though SphK1 and SphK2 null mice survive, SphK double knockout is embryonically lethal (63).

When it comes to the context of cancer, SK1 is well established as an onco-protein. Cells overexpressing SK1 demonstrated a transformation potential (64) and formed

TE D

tumors in nude mice (65). SK1 expression is also required for ras mediated transformation. Clinically, the mRNA and protein expression of SK1 is elevated in

EP

several cancerous tissues such as ovarian, breast, lung, and gastric tumors (66,67). In addition, microarray datasets reveal increased expression of SK1 in melanoma,

AC C

squamous cell carcinomas, cervical cancer, head and neck tumors, and hematological malignancies (68-70).

SK1 acts as an onco-protein by generating S1P onco-lipid. Intracellularly, S1P generated from SK1 can then target several proteins. For example, S1P can bind to TRAF2, an E3 ubiquitin ligase, to modulate TNF-α induced activation of NF-κB mitogenic signaling (71). In addition, extracellularly, serum S1P generated by SK1, can promote lung tumor metastasis by affecting breast cancer metastasis suppressor 1

ACCEPTED MANUSCRIPT

(Brms1) expression via S1PR2 signaling. Thus, silencing SK1 expression or treatment with anti-S1P monoclonal antibody (Sphingomab) decreased lung metastasis by

RI PT

decreasing serum S1P association with S1PR2 and increasing Brms1 expression (72). SK2 is localized mainly to the nucleus and has a nuclear export signals. Some reports also detected SK2 in cytosol, ER, and mitochondria (73). Inside the nucleus, SK2 can regulate cell cycle and cell proliferation (74,75). S1P generated by SK2 in the nucleus

SC

can bind to and inhibit HDAC1/2 enzymatic activity thereby preventing deacetylation of

M AN U

histone H3. This promoted the expression of p21 and c-Fos genes. Inside the mitochondria, S1P generated by SK2 can bind to prohibitin 2 but not to prohibitin 1, thereby regulating cytochrome c oxidase complex IV (76).

S1P can be secreted to the extracellular matrix and bind to S1P Receptors (S1PR1-5) in an autocrine or paracrine fashion. The major downstream responses are related to

TE D

increased inflammation, cell migration, and angiogenesis (77). In particular, S1P-S1PR1 interaction helps in activating receptor tyrosine kinases such as EGF and PDGF. A

EP

recent study that identified the X-ray structure of ligand bound S1PR1, suggested that S1PR activation does not necessarily require the export of S1P to the extracellular

AC C

surroundings. However, S1P can interact with its receptor within the plasma membrane via lateral movements of S1P in the membrane (78). It is well established in several studies that S1P protects cancer cells from cell death in response to various cytotoxic agents. For instance, SK1 upregulation resulted in camptothecin resistance in prostate cancer cells, which was reversed upon inhibiting SK1 pharmacologically (76). In another study, upregulation of SK1 resulted in resistance of prostate cancer cells to radiation therapy, but were re-sensitized if pre-

ACCEPTED MANUSCRIPT

treated with SK inhibitor N,N-dimethyl-sphingosine (79). Other examples include SK1 mediated resistance in FAS mediated cell death in melanoma and tamoxifen induced

RI PT

cell death in breast cancer cells (80). In the context of AML-M6, known as erythro-leukemia, SK1 overexpression in proerythroblasts transformed them into tumor cells resistant to cell death. In addition,

which was required for leukemic progression (81).

SC

microarray datasets revealed upregulation of SK1 in tumorigenic pro-erythroblasts

M AN U

In Chronic Myeloid Leukemia (CML), ceramide/S1P rheostat plays an important role in inducing drug resistance to bcr-abl targeted therapy. CML cells resistant to imatinib treatment had higher levels of SK1. Silencing SK1 expression sensitized the cells to imatinib induced cell death (81). S1P generated by SK1 binds and inhibits PP2A to prevent

bcr-abl

dephosphorylation

and

subsequent

degradation.

Inhibition

of

TE D

SK1/S1P/S1PR2 signaling either by pharmacological or molecular approaches restored PP2A mediated dephosphorylation of Bcr-Abl1 and enhanced imatinib mediated

EP

apoptosis (82).

Most of the studies highlight the role of S1P generated by SK1 in drug resistance.

AC C

However, recently S1P generated by SK2 also had a role in conferring resistance in several types of cancer. For example, in estrogen receptor positive breast cancer cells, SK2 increased transcription of ER-regulated genes like SDF1 and progesterone receptor. Pharmacological inhibition of SK2 using ABC294640 decreased the expression of these genes and sensitized the cells to treatment (83). Another described mechanism is the ability of ABC294640 (SK2 inhibitor) to decrease NF-κB pro survival

ACCEPTED MANUSCRIPT

signaling by decreasing activation p65 (84). In vivo, oral administration of ABC294640 inhibited the growth of xenograft-derived tumors mice (83).

RI PT

In the context of lung cancer, hypoxia can increase SK2 generation of S1P, which can then be secreted to activate S1PR1/S1PR3 signaling and activate MAP kinase signaling. This resulted in resistance to etoposide chemotherapeutic agent (85). In colon

outside the nucleus to confer the resistance (86).

SC

cancer cells resistant to sodium butyrate, SK2 was phosphorylated and exported

M AN U

In some cases, S1P can paradoxically induce apoptosis in cancer cells. This paradoxical effect was observed when S1P was generated by SK2. Indeed, some studies demonstrated that overexpression of SK2, but not SK1, suppresses growth and enhances apoptosis (87). These studies imply that the cellular location of S1P generation determines the impact of S1P on cell death. SK1 is mostly cytosolic and thus

TE D

S1P generated from SK1 impacts cytosolic signaling pathways and signals via S1PRs, while SK2 is mostly nuclear and thus S1P generated from SK2 impacts nuclear

EP

signaling pathways (41).

Similarly, sphingosine can also induce cell death in response to DNA damage. One

AC C

recent study showed that sphingosine generated from alkaline ceramidase 2 (ACER2) induces programmed cell death (88). Upon treating cancer cells with DNA damaging agents such as doxorubicin, ACER2 expression was upregulated significantly and sphingosine levels increased resulting in apoptosis and necroptosis (88).

b. S1P in the skin

ACCEPTED MANUSCRIPT

As mentioned earlier, ceramide inhibits keratinocyte growth and induces apoptosis as in most other systems (89). Unexpectedly, S1P, normally a ceramide antagonist, also inhibits keratinocyte proliferation. However, S1P does not drive keratinocytes into

RI PT

apoptosis, but rather induces their differentiation (90,91). Treating keratinocytes with S1P results in cell cycle arrest, inhibition of cell division, and an increase in intracellular calcium (92). Calcium is an important signaling molecule in keratinocytes for their

SC

differentiation to into corneocytes (93). S1P induces these effects in keratinocytes by

M AN U

binding to S1PR2 receptor (92).

In addition, S1P modulates some of the functions of dendritic cells such as uptake of antigens and interaction with T cells. S1P decreases the endocytic ability of dendritic cells, thus limiting their antigen uptake capacity (94). In fact, S1P topical application in a mouse model reduced foreign antigen uptake by dendritic cells down to 40%. This is

TE D

again secondary to S1P mediated inhibition of Akt (94). In addition, S1P modulates the migration of dendritic cells. Studies show that dendritic cells migrate towards lymph nodes, to areas rich with S1P, following the S1P gradient. Disrupting the gradient by

EP

topical application of S1P prevented the migration of dendritic cells from the epidermis

AC C

to the lymph nodes (95). Finally, S1P exerts anti-inflammatory roles in the skin by decreasing the levels of IL-12 release from activated dendritic cells and by decreasing the activation of T helper cells (96). c. Sphingosine kinases and S1P in melanoma pathogenesis and drug resistance Even though S1P inhibits the proliferation of keratinocytes, this is not the case in human melanoma cells. Treatment of melanoma cells with S1P inhibited drug induced cell death. This was prevented by the pre-treatment with short-chain C6-ceramide (97). In

ACCEPTED MANUSCRIPT

addition, elevated S1P levels secondary to increased activity of SK1 occur in advanced melanomas. Targeting SK1 using siRNA decreased growth and induced apoptosis in melanoma cells. Pharmacological inhibition of SK1 in melanoma using the inhibitor SKI-

RI PT

I decreased S1P content, elevated ceramide levels, caused a G2-M block, and induced apoptotic cell death (98). This suggests that targeting SK1 using siRNAs or SKI-I inhibitor has therapeutic potential for melanoma treatment.

SC

Indeed, several studies demonstrate that human melanoma cell death is induced upon

M AN U

the inhibition of sphingosine kinases or the blockade of S1P receptors. In one study, the sphingosine analogue FTY720, which functionally antagonizes S1P receptors, induced cell death in human melanoma cells through a mechanism involving the vacuolar ATPase activity. Interestingly, cell death was characterized by features of necrosis and autophagy (99). In another study, FTY720 significantly decreased the viability and

TE D

expression of apoptosis markers in cisplatin resistant human melanoma cell lines (100). These finding suggest that FTY720 or other sphingosine receptor inhibitors are promising candidates for further development as new therapeutic agents in the

EP

treatment of melanoma.

AC C

Sphingosine kinases play several roles in melanoma pathogenesis. For instance, downregulation of sphingosine kinase-1 induces protective tumor immunity in melanoma. The infiltration of melanoma tumors by M2 macrophages is often correlated with poor prognosis. Downregulation of SK1 in melanoma cells reduced the percentage of M2 macrophages in favor of increased M1 macrophages into the tumor. Infiltration by M1 macrophages recruited T lymphocytes and hence tumor rejection. This suggest that

ACCEPTED MANUSCRIPT

SK1 plays a key role in the recruitment and phenotypic shift of the tumor macrophages that promote melanoma growth (101).

RI PT

Moreover, S1P plays an important role in the metastasis of melanoma. SK1 facilitates the communication between melanoma cells and fibroblasts in metastasis. SK1 in myofibroblasts promotes S1P-dependent dissemination of melanoma cells via a S1P receptor 3 (102). These findings clarify the role of S1P in melanoma’s

SC

microenvironment.

M AN U

One paradox; however, is that one study show that S1P can inhibit cell migration in B16F10 mouse melanoma cells (103). Inhibition of migration was dependent on the endogenous expression of S1P receptor 2 (S1PR2). Overexpression of S1PR2 in melanoma cells potentiated S1P’s effect in inhibiting tumor cell migration. These observations led to the suggestion that sphingosine agonists specific for S1PR2 might

TE D

be therapeutically beneficial in the treatment of melanoma. In addition, some nonphosphorylated sphingosine analogs, N-methyl sphingosine and N-acetyl sphingosine,

EP

have been shown to protect against induction of skin carcinomas in a mouse skin carcinogenesis model (104). It seems that S1P’s effect on melanoma depends on the

AC C

receptor it is binding to. S1P signaling through S1PR2 can inhibit melanoma migration while S1P signaling through other S1PRs can result in metastasis and viability of melanoma cells. Clearly, the role of S1P and the different S1PRs need to be studies more extensively in melanoma cells, fibroblasts, myofibroblasts, and platelet. In addition, the role of SK2, which is usually located in the nucleus, needs to be studied in melanoma.

ACCEPTED MANUSCRIPT

7. Sphingolipid based anti-cancer therapeutics

a. Anti-cancerous ceramide analogues and mimetics

RI PT

Exogenous ceramide analogues or mimetics have been used in several studies as a strategy to induce programmed cell death in cancer cells. Several modifications in the structure and delivery methods of ceramide were adopted in order to maximize the

SC

efficiency of exogenous ceramide (Table 1). Most notably, modifying ceramide by adding a pyridinium ring in the sphingosine backbone is an ingenious method to

M AN U

increase water and cell membrane solubility and allows targeting and accumulation of ceramide analogues into the negatively charged mitochondria, and to a lesser extent, to the nucleus of cancer cells (39). In addition, this offers a selective delivery of the ceramide analogues to cancer cells versus normal cells since it is suggested that

pyridinium-ceramide

TE D

cancer cells acquire more negative charge in their mitochondria (105). Indeed, the analogues

L-threo-C6-Pyr-Cer

and

D-erythro-C16-Pyr-Cer

accumulate in mitochondria and to a lesser extent in nuclei fractions in vitro (106-110).

EP

In addition, ceramide analogues accumulated in vivo in the tumor site more so than in other intact organs (39,109). Therefore, Pyr-Cer can preferentially target cancer cells

AC C

with minimum toxicity to normal cells. Other structural analogs of ceramide include C16-serinol and 4,6-diene-ceramide, 5ROH-3E-C8-ceramide, adamantyl-ceramide, and benzene-C4-ceramide. These analogues were also able to either induce apoptosis in various human cancer cells (111,112) or inhibit the growth of drug-resistant human cancer cell lines (113). It should also be noted that treatment of cells with exogenous ceramides may result in the generation of

ACCEPTED MANUSCRIPT

endogenous long chain ceramides via the sphingosine-recycling pathway and this might add to the anti-cancerous effect.

RI PT

The delivery method of ceramide was also modified so that ceramide can be packaged in PEGylated liposomes, which are known to be more effective for crossing the cell membrane. This formulation increased the accumulation of ceramide and thus the procell death effect in cancer cells (114). Mechanistically, ceramide delivered in liposomes

SC

prevented the phosphorylation of Akt and stimulated the activity of caspase-3/7 more

M AN U

effectively than non-liposomal ceramide (114). The liposomal formulation was also effective in vivo, resulting in slower tumor growth in murine models of breast cancer (115).

Another approach is to encapsulated vincristine in sphingomyelin-liposomes, also called

TE D

sphingosomes. This method had a higher effect compared to the conventional vincristine injections in animal models of adult acute lymphocytic leukemia (ALL). Sphingosomal vincristine is now in phase II clinical trials for recurrent and refractory ALL

EP

(116). Another method is polymeric nano-particle delivery system to improve the delivery and efficacy of ceramides to overcome resistance (117).

AC C

b. SK/S1P anticancer therapeutics Given the importance of SK1-SK2/S1P signaling in promoting tumor progression and developing resistance, several approaches were developed to target SK enzymes and S1P for therapeutic purposes (Table 2). Non-selective sphingosine kinase inhibitors: N, N-dimethyl sphingosine (DMS) exhibited anti-tumor growth properties in cancer cell lines and nude mice. Side effects of DMS

ACCEPTED MANUSCRIPT

include liver toxicity and hemolysis (118). L-threo dihyrosphingosine (Safingol) is currently in phase I clinical trials (French, Schwartz). Another SK inhibitor is Phenoxodiol with anti-cancerous and anti-angiogenic effects, and is currently in clinical

RI PT

trials for ovarian and prostate cancers (119,120). Another inhibitor is SKI-II, which has excellent oral bioavailability and anti-tumor effects in vivo (121).

SK1 selective inhibitors: SK1-I inhibitor is specific for SK1 with established in vivo

SC

orthotopic anti-tumor effects, sub-cutaneous glioblastoma multiforme, and AML (122). In

M AN U

breast cancer mouse models, SK1-I decreased serum S1P levels and decreased tumor burden (121).

SK2 selective inhibitors: ABC294640 is the SK2 specific inhibitor. It has a very high oral bioavailability and a low toxicology profile. ABC294640 has anti-proliferative effects in a panel of cancer cell lines and in vivo mouse models (83,84,123). ABC294640 is

TE D

currently in phase II clinical trials for diffuse large B cell lymphoma. Antibody based therapeutics: A monoclonal antibody against S1P (Sphingomab) was

EP

developed and was shown to be effective in neutralizing S1P and exert anti-cancerous effects in breast, ovarian, lung, and melanoma cancer models (72). In addition,

AC C

Sphingomab (LT1009) can neutralize VEGF-induced angiogenesis (visentin). LT1009 is currently in Phase I/ II clinical trials for neovascular Age Related Macular Degeneration and some types of cancer (124).

8. Conclusion

ACCEPTED MANUSCRIPT

The last decade has witnessed a dramatic increase in researching the mechanisms involved in melanoma pathogenesis, response to treatment, and resistance. Several studies

show

convincingly

that

sphingolipid

signaling

affects

melanoma’s

RI PT

carcinogenesis, regulate the fate of treated melanoma cells, and impact migration and metastasis. Specifically, the accumulation of intracellular ceramide lipid seems to drive melanoma cell death and response to treatment, while SK generated S1P seem to

SC

oppose cell death and promote resistance. This suggests that ceramide analogues, SK inhibitors, and S1P antibodies, can potentially be used in the treatment of melanoma. In

M AN U

addition, acid ceramidase is emerging as another target in melanoma cells. By overexpressing acid ceramidase, melanoma cells hydrolyze ceramide ending up with lower levels of ceramide. There are several acid ceramidase inhibitors that can be tested in vivo or in phase 1 clinical trials.

TE D

The role of S1P in the skin should be studied more extensively. It is interesting to understand the reason behind why keratinocytes and melanoma cells respond differently to S1P: S1P promotes keratinocyte cell death but melanoma cell survival.

EP

Other questions to answer are whether the ceramide levels are the same in these two

AC C

cell types and whether the S1P downstream signaling molecules, such as Akt, are altered in melanoma.

Finally, the role of S1P receptors S1PR1-5 in melanoma should be further characterized. Even though S1P binding to S1PR3 has been reported to promote metastasis, the underlying mechanism is not fully understood. S1PR inhibitors, such as FTY720, have been shown to be very effective in inducing cell death and decreasing

ACCEPTED MANUSCRIPT

melanoma tumor burden in vivo. More studies need to be performed to characterize the

M AN U

SC

RI PT

role of each S1PR in melanoma and their role as therapeutic targets.

Acknowledgements •

There is no conflict of interest to be declared.



All authors have read the journal's policy on disclosure of potential conflicts of

TE D

interest. •

No editorial sources have been used for preparation of the manuscript.



All authors have read the journal's authorship agreement and that the manuscript

AC C

EP

has been reviewed by and approved by all named authors.

ACCEPTED MANUSCRIPT

References (1) Vennepureddy A, Thumallapally N, Nehru VM, Atallah J, Terjanian T. Novel Drugs and Combination Therapies for the Treatment of Metastatic Melanoma. Journal of clinical medicine research 2016;8(2):63.

RI PT

(2) Miller AJ, Mihm Jr MC. Melanoma. N Engl J Med 2006;355(1):51-65.

(3) Macdonald JB, Dueck AC, Gray RJ, Wasif N, Swanson DL, Sekulic A, et al. Malignant melanoma in the elderly: different regional disease and poorer prognosis. J Cancer 2011;2:538-543.

SC

(4) Dany M, Nganga R, Chidiac A, Hanna E, Matar S, Elston D. Advances in immunotherapy for melanoma management. Human Vaccines & Immunotherapeutics 2016:1-11.

M AN U

(5) Ogretmen B, Hannun YA. Biologically active sphingolipids in cancer pathogenesis and treatment. Nat Rev Cancer 2004 Aug;4(8):604-616. (6) Hamanaka S, Suzuki A, Hara M, Nishio H, Otsuka F, Uchida Y. Human epidermal glucosylceramides are major precursors of stratum corneum ceramides. J Invest Dermatol 2002;119(2):416-423.

TE D

(7) Saddoughi SA, Ogretmen B. Diverse functions of ceramide in cancer cell death and proliferation. Adv Cancer Res 2013;117:37-58. (8) Behne M, Uchida Y, Seki T, de Montellano PO, Elias PM, Holleran WM. Omegahydroxyceramides are required for corneocyte lipid envelope (CLE) formation and normal epidermal permeability barrier function. J Invest Dermatol 2000;114(1):185-192.

EP

(9) Saddoughi SA, Song P, Ogretmen B. Roles of bioactive sphingolipids in cancer biology and therapeutics. Subcell Biochem 2008;49:413-440.

AC C

(10) Pewzner-Jung Y, Ben-Dor S, Futerman AH. When do Lasses (longevity assurance genes) become CerS (ceramide synthases)?: Insights into the regulation of ceramide synthesis. J Biol Chem 2006 Sep 1;281(35):25001-25005. (11) Kraveka JM, Li L, Szulc ZM, Bielawski J, Ogretmen B, Hannun YA, et al. Involvement of dihydroceramide desaturase in cell cycle progression in human neuroblastoma cells. J Biol Chem 2007 Jun 8;282(23):16718-16728. (12) Selvam SP, Ogretmen B. Sphingosine kinase/sphingosine 1-phosphate signaling in cancer therapeutics and drug resistance. Handb Exp Pharmacol 2013;(216):3-27. doi(216):3-27.

ACCEPTED MANUSCRIPT

(13) Regulation and signaling mechanism of cancer cell migration by TGF-Beta receptors and ceramide metabolism. FEBS JOURNAL: WILEY-BLACKWELL 111 RIVER ST, HOBOKEN 07030-5774, NJ USA; 2015.

RI PT

(14) Hanada K, Kumagai K, Yasuda S, Miura Y, Kawano M, Fukasawa M, et al. Molecular machinery for non-vesicular trafficking of ceramide. Nature 2003 Dec 18;426(6968):803-809. (15) Kumagai K, Yasuda S, Okemoto K, Nishijima M, Kobayashi S, Hanada K. CERT mediates intermembrane transfer of various molecular species of ceramides. J Biol Chem 2005 Feb 25;280(8):6488-6495.

SC

(16) Selvam SP, Ogretmen B. Sphingosine kinase/sphingosine 1-phosphate signaling in cancer therapeutics and drug resistance. Handb Exp Pharmacol 2013;(216):3-27. doi(216):3-27.

M AN U

(17) Schulz A, Mousallem T, Venkataramani M, Persaud-Sawin DA, Zucker A, Luberto C, et al. The CLN9 protein, a regulator of dihydroceramide synthase. J Biol Chem 2006 Feb 3;281(5):2784-2794. (18) Imgrund S, Hartmann D, Farwanah H, Eckhardt M, Sandhoff R, Degen J, et al. Adult ceramide synthase 2 (CERS2)-deficient mice exhibit myelin sheath defects, cerebellar degeneration, and hepatocarcinomas. J Biol Chem 2009 Nov 27;284(48):33549-33560.

TE D

(19) Jiang W, Ogretmen B. Autophagy paradox and ceramide. Biochim Biophys Acta 2014 May;1841(5):783-792.

EP

(20) Ponnusamy S, Meyers-Needham M, Senkal CE, Saddoughi SA, Sentelle D, Selvam SP, et al. Sphingolipids and cancer: ceramide and sphingosine-1-phosphate in the regulation of cell death and drug resistance. Future Oncol 2010 Oct;6(10):16031624.

AC C

(21) Mizutani Y, Kihara A, Igarashi Y. Mammalian Lass6 and its related family members regulate synthesis of specific ceramides. Biochem J 2005 Aug 15;390(Pt 1):263-271. (22) Dany M, Ogretmen B. Ceramide induced mitophagy and tumor suppression. Biochim Biophys Acta 2015 Jan 26. (23) Jennemann R, Rabionet M, Gorgas K, Epstein S, Dalpke A, Rothermel U, et al. Loss of ceramide synthase 3 causes lethal skin barrier disruption. Hum Mol Genet 2012 Feb 1;21(3):586-608. (24) Rabionet M, Bayerle A, Jennemann R, Heid H, Fuchser J, Marsching C, et al. Male meiotic cytokinesis requires ceramide synthase 3-dependent sphingolipids with unique membrane anchors. Hum Mol Genet 2015 Sep 1;24(17):4792-4808.

ACCEPTED MANUSCRIPT

(25) Levy M, Futerman 2010;62(5):347-356.

AH.

Mammalian

ceramide

synthases.

IUBMB

Life

RI PT

(26) Senkal CE, Ponnusamy S, Rossi MJ, Bialewski J, Sinha D, Jiang JC, et al. Role of human longevity assurance gene 1 and C18-ceramide in chemotherapy-induced cell death in human head and neck squamous cell carcinomas. Mol Cancer Ther 2007 Feb;6(2):712-722.

SC

(27) Karahatay S, Thomas K, Koybasi S, Senkal CE, Elojeimy S, Liu X, et al. Clinical relevance of ceramide metabolism in the pathogenesis of human head and neck squamous cell carcinoma (HNSCC): attenuation of C(18)-ceramide in HNSCC tumors correlates with lymphovascular invasion and nodal metastasis. Cancer Lett 2007 Oct 18;256(1):101-111.

M AN U

(28) Koybasi S, Senkal CE, Sundararaj K, Spassieva S, Bielawski J, Osta W, et al. 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):44311-44319. (29) Gencer S, Oleinik N, Dany M, Ogretmen B. Ceramide is A Key Factor That Regulates The Crosstalk Between TGF-β and Sonic Hedgehog Signaling at The Basal Cilia To Control Cell Migration and Tumor Metastasis. The FASEB Journal 2016;30(1 Supplement):1108.2-1108.2.

TE D

(30) Saddoughi SA, Garrett-Mayer E, Chaudhary U, O'Brien PE, Afrin LB, Day TA, et al. Results of a phase II trial of gemcitabine plus doxorubicin in patients with recurrent head and neck cancers: serum C(1)(8)-ceramide as a novel biomarker for monitoring response. Clin Cancer Res 2011 Sep 15;17(18):6097-6105.

EP

(31) 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.

AC C

(32) Senkal CE, Ponnusamy S, Manevich Y, Meyers-Needham M, Saddoughi SA, Mukhopadyay A, et al. Alteration of ceramide synthase 6/C16-ceramide induces activating transcription factor 6-mediated endoplasmic reticulum (ER) stress and apoptosis via perturbation of cellular Ca2+ and ER/Golgi membrane network. J Biol Chem 2011 Dec 9;286(49):42446-42458. (33) Holliday MW,Jr, Cox SB, Kang MH, Maurer BJ. C22:0- and C24:0dihydroceramides confer mixed cytotoxicity in T-cell acute lymphoblastic leukemia cell lines. PLoS One 2013 Sep 9;8(9):e74768. (34) Sassa T, Suto S, Okayasu Y, Kihara A. A shift in sphingolipid composition from C24 to C16 increases susceptibility to apoptosis in HeLa cells. Biochim Biophys Acta 2012 Jul;1821(7):1031-1037.

ACCEPTED MANUSCRIPT

(35) Zhao L, Spassieva SD, Jucius TJ, Shultz LD, Shick HE, Macklin WB, et al. A deficiency of ceramide biosynthesis causes cerebellar purkinje cell neurodegeneration and lipofuscin accumulation. PLoS Genet 2011 May;7(5):e1002063.

RI PT

(36) Ebel P, Imgrund S, Vom Dorp K, Hofmann K, Maier H, Drake H, et al. Ceramide synthase 4 deficiency in mice causes lipid alterations in sebum and results in alopecia. Biochem J 2014 Jul 1;461(1):147-158. (37) Pewzner-Jung Y, Brenner O, Braun S, Laviad EL, Ben-Dor S, Feldmesser E, et al. A critical role for ceramide synthase 2 in liver homeostasis: II. insights into molecular changes leading to hepatopathy. J Biol Chem 2010 Apr 2;285(14):10911-10923.

SC

(38) Tang Y, Cao K, Wang Q, Chen J, Liu R, Wang S, et al. Silencing of CerS6 increases the invasion and glycolysis of melanoma WM35, WM451 and SK28 cell lines via increased GLUT1-induced downregulation of WNT5A. Oncol Rep 2016.

M AN U

(39) Dany M, Gencer S, Nganga R, Thomas RJ, Oleinik N, Baron KD, et al. Targeting FLT3-ITD signaling mediates ceramide-dependent mitophagy and attenuates drug resistance in AML. Blood 2016 Aug 18. (40) Saddoughi SA, Gencer S, Peterson YK, Ward KE, Mukhopadhyay A, Oaks J, et al. 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):105-121.

TE D

(41) Panneer Selvam S, De Palma RM, Oaks JJ, Oleinik N, Peterson YK, Stahelin RV, et al. Binding of the sphingolipid S1P to hTERT stabilizes telomerase at the nuclear periphery by allosterically mimicking protein phosphorylation. Sci Signal 2015 Jun 16;8(381):ra58.

EP

(42) Beckham TH, Lu P, Jones EE, Marrison T, Lewis CS, Cheng JC, et al. LCL124, a cationic analog of ceramide, selectively induces pancreatic cancer cell death by accumulating in mitochondria. J Pharmacol Exp Ther 2013 Jan;344(1):167-178.

AC C

(43) Baran Y, Bielawski J, Gunduz U, Ogretmen B. Targeting glucosylceramide synthase sensitizes imatinib-resistant chronic myeloid leukemia cells via endogenous ceramide accumulation. J Cancer Res Clin Oncol 2011 Oct;137(10):1535-1544. (44) Madi N, Dany M, Abdoun S, Usta J. Moringa oleifera's Nutritious Aqueous Leaf Extract Has Anticancerous Effects by Compromising Mitochondrial Viability in an ROSDependent Manner. J Am Coll Nutr 2016;35(7):604-613. (45) Pettus BJ, Chalfant CE, Hannun YA. Ceramide in apoptosis: an overview and current perspectives. Biochim Biophys Acta 2002 Dec 30;1585(2-3):114-125.

ACCEPTED MANUSCRIPT

(46) Chalfant CE, Kishikawa K, Mumby MC, Kamibayashi C, Bielawska A, Hannun YA. Long chain ceramides activate protein phosphatase-1 and protein phosphatase-2A. Activation is stereospecific and regulated by phosphatidic acid. J Biol Chem 1999 Jul 16;274(29):20313-20317.

RI PT

(47) Yeh E, Cunningham M, Arnold H, Chasse D, Monteith T, Ivaldi G, et al. A signalling pathway controlling c-Myc degradation that impacts oncogenic transformation of human cells. Nat Cell Biol 2004 Apr;6(4):308-318.

SC

(48) Salas A, Ponnusamy S, Senkal CE, Meyers-Needham M, Selvam SP, Saddoughi SA, et al. Sphingosine kinase-1 and sphingosine 1-phosphate receptor 2 mediate BcrAbl1 stability and drug resistance by modulation of protein phosphatase 2A. Blood 2011 Jun 2;117(22):5941-5952.

M AN U

(49) Mukhopadhyay A, Saddoughi SA, Song P, Sultan I, Ponnusamy S, Senkal CE, et al. Direct interaction between the inhibitor 2 and ceramide via sphingolipid-protein binding is involved in the regulation of protein phosphatase 2A activity and signaling. FASEB J 2009 Mar;23(3):751-763. (50) Saddoughi SA, Gencer S, Peterson YK, Ward KE, Mukhopadhyay A, Oaks J, et al. 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):105-121.

TE D

(51) Ogretmen B, Schady D, Usta J, Wood R, Kraveka JM, Luberto C, et al. Role of ceramide in mediating the inhibition of telomerase activity in A549 human lung adenocarcinoma cells. J Biol Chem 2001 Jul 6;276(27):24901-24910.

EP

(52) Kraveka JM, Li L, Bielawski J, Obeid LM, Ogretmen B. Involvement of endogenous ceramide in the inhibition of telomerase activity and induction of morphologic differentiation in response to all-trans-retinoic acid in human neuroblastoma cells. Arch Biochem Biophys 2003 Nov 15;419(2):110-119.

AC C

(53) Dany M, Ogretmen B. 522 Ceramide accumulation in mitochondria: A novel anticancerous approach via inducing mitophagy. J Invest Dermatol 2016;136(5):S92. (54) Dany M, Ogretmen B. Ceramide Mediated Lethal Mitophagy: A Novel Cell Death Mechanism in FLT3 Targeted Therapy for Acute Myeloid Leukemia. The FASEB Journal 2015;29(1 Supplement):147.1. (55) Morad SA, Bridges LC, Larrea ADA, Mayen AL, MacDougall MR, Davis TS, et al. Short-chain ceramides depress integrin cell surface expression and function in colorectal cancer cells. Cancer Lett 2016;376(2):199-204.

ACCEPTED MANUSCRIPT

(56) Jiang F, Jin K, Huang S, Bao Q, Shao Z, Hu X, et al. Liposomal C6 Ceramide Activates Protein Phosphatase 1 to Inhibit Melanoma Cells. PloS one 2016;11(9):e0159849.

RI PT

(57) Zhang P, Fu C, Hu Y, Dong C, Song Y, Song E. C6-ceramide nanoliposome suppresses tumor metastasis by eliciting PI3K and PKCζ tumor-suppressive activities and regulating integrin affinity modulation. Scientific reports 2015;5. (58) Becker KA, Beckmann N, Adams C, Hessler G, Kramer M, Gulbins E, et al. Melanoma cell metastasis via P-selectin-mediated activation of acid sphingomyelinase in platelets. Clin Exp Metastasis 2016:1-11.

SC

(59) Realini N, Palese F, Pizzirani D, Pontis S, Basit A, Bach A, et al. Acid Ceramidase in Melanoma: EXPRESSION, LOCALIZATION, AND EFFECTS OF PHARMACOLOGICAL INHIBITION. J Biol Chem 2016 Jan 29;291(5):2422-2434.

M AN U

(60) Bedia C, Casas J, Andrieu-Abadie N, Fabrias G, Levade T. Acid ceramidase expression modulates the sensitivity of A375 melanoma cells to dacarbazine. J Biol Chem 2011 Aug 12;286(32):28200-28209. (61) Pitson SM. Regulation of sphingosine kinase and sphingolipid signaling. Trends Biochem Sci 2011 Feb;36(2):97-107.

TE D

(62) Liu H, Chakravarty D, Maceyka M, Milstien S, Spiegel S. Sphingosine kinases: a novel family of lipid kinases. Prog Nucleic Acid Res Mol Biol 2002;71:493-511. (63) Spiegel S, Milstien S. Sphingosine-1-phosphate: an enigmatic signalling lipid. Nat Rev Mol Cell Biol 2003 May;4(5):397-407.

EP

(64) Vadas M, Xia P, McCaughan G, Gamble J. The role of sphingosine kinase 1 in cancer: oncogene or non-oncogene addiction? Biochim Biophys Acta 2008 Sep;1781(9):442-447.

AC C

(65) Xia P, Gamble JR, Wang L, Pitson SM, Moretti PA, Wattenberg BW, et al. An oncogenic role of sphingosine kinase. Curr Biol 2000 Nov 30;10(23):1527-1530. (66) Johnson KR, Johnson KY, Crellin HG, Ogretmen B, Boylan AM, Harley RA, et al. Immunohistochemical distribution of sphingosine kinase 1 in normal and tumor lung tissue. J Histochem Cytochem 2005 Sep;53(9):1159-1166. (67) French KJ, Schrecengost RS, Lee BD, Zhuang Y, Smith SN, Eberly JL, et al. Discovery and evaluation of inhibitors of human sphingosine kinase. Cancer Res 2003 Sep 15;63(18):5962-5969.

ACCEPTED MANUSCRIPT

(68) Nindl I, Dang C, Forschner T, Kuban RJ, Meyer T, Sterry W, et al. Identification of differentially expressed genes in cutaneous squamous cell carcinoma by microarray expression profiling. Mol Cancer 2006 Aug 8;5:30.

RI PT

(69) Talantov D, Mazumder A, Yu JX, Briggs T, Jiang Y, Backus J, et al. Novel genes associated with malignant melanoma but not benign melanocytic lesions. Clin Cancer Res 2005 Oct 15;11(20):7234-7242. (70) Chan MM, Lu X, Merchant FM, Iglehart JD, Miron PL. Gene expression profiling of NMU-induced rat mammary tumors: cross species comparison with human breast cancer. Carcinogenesis 2005 Aug;26(8):1343-1353.

SC

(71) Alvarez SE, Harikumar KB, Hait NC, Allegood J, Strub GM, Kim EY, et al. Sphingosine-1-phosphate is a missing cofactor for the E3 ubiquitin ligase TRAF2. Nature 2010 Jun 24;465(7301):1084-1088.

M AN U

(72) Ponnusamy S, Selvam SP, Mehrotra S, Kawamori T, Snider AJ, Obeid LM, et al. Communication between host organism and cancer cells is transduced by systemic sphingosine kinase 1/sphingosine 1-phosphate signalling to regulate tumour metastasis. EMBO Mol Med 2012 Aug;4(8):761-775. (73) Maceyka M, Sankala H, Hait NC, Le Stunff H, Liu H, Toman R, et al. SphK1 and SphK2, sphingosine kinase isoenzymes with opposing functions in sphingolipid metabolism. J Biol Chem 2005 Nov 4;280(44):37118-37129.

TE D

(74) Liu H, Toman RE, Goparaju SK, Maceyka M, Nava VE, Sankala H, et al. Sphingosine kinase type 2 is a putative BH3-only protein that induces apoptosis. J Biol Chem 2003 Oct 10;278(41):40330-40336.

EP

(75) Hait NC, Bellamy A, Milstien S, Kordula T, Spiegel S. Sphingosine kinase type 2 activation by ERK-mediated phosphorylation. J Biol Chem 2007 Apr 20;282(16):1205812065.

AC C

(76) Strub GM, Paillard M, Liang J, Gomez L, Allegood JC, Hait NC, et al. Sphingosine1-phosphate produced by sphingosine kinase 2 in mitochondria interacts with prohibitin 2 to regulate complex IV assembly and respiration. FASEB J 2011 Feb;25(2):600-612. (77) Strub GM, Maceyka M, Hait NC, Milstien S, Spiegel S. Extracellular and intracellular actions of sphingosine-1-phosphate. Adv Exp Med Biol 2010;688:141-155. (78) Hanson MA, Roth CB, Jo E, Griffith MT, Scott FL, Reinhart G, et al. Crystal structure of a lipid G protein-coupled receptor. Science 2012 Feb 17;335(6070):851855.

ACCEPTED MANUSCRIPT

(79) Nava VE, Cuvillier O, Edsall LC, Kimura K, Milstien S, Gelmann EP, et al. Sphingosine enhances apoptosis of radiation-resistant prostate cancer cells. Cancer Res 2000 Aug 15;60(16):4468-4474.

RI PT

(80) Sukocheva O, Wang L, Verrier E, Vadas MA, Xia P. Restoring endocrine response in breast cancer cells by inhibition of the sphingosine kinase-1 signaling pathway. Endocrinology 2009 Oct;150(10):4484-4492. (81) Le Scolan E, Pchejetski D, Banno Y, Denis N, Mayeux P, Vainchenker W, et al. Overexpression of sphingosine kinase 1 is an oncogenic event in erythroleukemic progression. Blood 2005 Sep 1;106(5):1808-1816.

M AN U

SC

(82) Salas A, Ponnusamy S, Senkal CE, Meyers-Needham M, Selvam SP, Saddoughi SA, et al. Sphingosine kinase-1 and sphingosine 1-phosphate receptor 2 mediate BcrAbl1 stability and drug resistance by modulation of protein phosphatase 2A. Blood 2011 Jun 2;117(22):5941-5952. (83) Antoon JW, White MD, Slaughter EM, Driver JL, Khalili HS, Elliott S, et al. Targeting NFkB mediated breast cancer chemoresistance through selective inhibition of sphingosine kinase-2. Cancer Biol Ther 2011 Apr 1;11(7):678-689. (84) Antoon JW, White MD, Meacham WD, Slaughter EM, Muir SE, Elliott S, et al. Antiestrogenic effects of the novel sphingosine kinase-2 inhibitor ABC294640. Endocrinology 2010 Nov;151(11):5124-5135.

TE D

(85) Schnitzer SE, Weigert A, Zhou J, Brune B. Hypoxia enhances sphingosine kinase 2 activity and provokes sphingosine-1-phosphate-mediated chemoresistance in A549 lung cancer cells. Mol Cancer Res 2009 Mar;7(3):393-401.

EP

(86) Xiao M, Liu Y, Zou F. Sensitization of human colon cancer cells to sodium butyrateinduced apoptosis by modulation of sphingosine kinase 2 and protein kinase D. Exp Cell Res 2012 Jan 1;318(1):43-52.

AC C

(87) Hait NC, Oskeritzian CA, Paugh SW, Milstien S, Spiegel S. Sphingosine kinases, sphingosine 1-phosphate, apoptosis and diseases. Biochimica et Biophysica Acta (BBA)-Biomembranes 2006;1758(12):2016-2026. (88) Xu R, Wang K, Mileva I, Hannun YA, Obeid LM, Mao C. Alkaline ceramidase 2 and its bioactive product sphingosine are novel regulators of the DNA damage response. Oncotarget 2016 Apr 5;7(14):18440-18457. (89) Bektas M, Orfanos CE, Geilen CC. Different vitamin D analogues induce sphingomyelin hydrolysis and apoptosis in the human keratinocyte cell line HaCaT. Cell Mol Biol (Noisy-le-grand) 2000 Feb;46(1):111-119.

ACCEPTED MANUSCRIPT

(90) Vogler R, Sauer B, Kim D, Schäfer-Korting M, Kleuser B. Sphingosine-1-phosphate and its potentially paradoxical effects on critical parameters of cutaneous wound healing. J Invest Dermatol 2003;120(4):693-700.

RI PT

(91) Japtok L, Bäumer W, Kleuser B. Sphingosine-1-phosphate as signaling molecule in the skin. Allergo journal international 2014;23(2):54-59. (92) Schüppel M, Kürschner U, Kleuser U, Schäfer-Korting M, Kleuser B. Sphingosine 1-phosphate restrains insulin-mediated keratinocyte proliferation via inhibition of Akt through the S1P2 receptor subtype. J Invest Dermatol 2008;128(7):1747-1756.

SC

(93) Lichte K, Rossi R, Danneberg K, ter Braak M, Kürschner U, Jakobs KH, et al. Lysophospholipid receptor-mediated calcium signaling in human keratinocytes. J Invest Dermatol 2008;128(6):1487-1498.

M AN U

(94) Japtok L, Schaper K, Bäumer W, Radeke HH, Jeong SK, Kleuser B. Sphingosine 1-phosphate modulates antigen capture by murine Langerhans cells via the S1P 2 receptor subtype. PLoS One 2012;7(11):e49427. (95) Radeke HH, von Wenckstern H, Stoidtner K, Sauer B, Hammer S, Kleuser B. Overlapping signaling pathways of sphingosine 1-phosphate and TGF-beta in the murine Langerhans cell line XS52. J Immunol 2005 Mar 1;174(5):2778-2786.

TE D

(96) Schröder M, Richter C, San Juan MH, Maltusch K, Giegold O, Quintini G, et al. The sphingosine kinase 1 and S1P1 axis specifically counteracts LPS-induced IL-12p70 production in immune cells of the spleen. Mol Immunol 2011;48(9):1139-1148. (97) Zou Y, Fan G, Wang X. Pre-clinical assessment of A-674563 as an anti-melanoma agent. Biochem Biophys Res Commun 2016;477(1):1-8.

EP

(98) Madhunapantula SV, Hengst J, Gowda R, Fox TE, Yun JK, Robertson GP. Targeting sphingosine kinase-1 to inhibit melanoma. Pigment Cell Melanoma Res 2012 Mar;25(2):259-274.

AC C

(99) Tay KH, Liu X, Chi M, Jin L, Jiang CC, Guo ST, et al. Involvement of vacuolar H ‐ ATPase in killing of human melanoma cells by the sphingosine kinase analogue FTY720. Pigment cell & melanoma research 2015;28(2):171-183. (100) Ishitsuka A, Fujine E, Mizutani Y, Tawada C, Kanoh H, Banno Y, et al. FTY720 and cisplatin synergistically induce the death of cisplatin-resistant melanoma cells through the downregulation of the PI3K pathway and the decrease in epidermal growth factor receptor expression. Int J Mol Med 2014 Oct;34(4):1169-1174. (101) Mrad M, Imbert C, Garcia V, Rambow F, Therville N, Carpentier S, et al. Downregulation of sphingosine kinase-1 induces protective tumor immunity by

ACCEPTED MANUSCRIPT

promoting M1 macrophage response in melanoma. Oncotarget 2016;7(44):7187371886.

RI PT

(102) Pyne NJ, Pyne S. Sphingosine kinase 1 enables communication between melanoma cells and fibroblasts that provides a new link to metastasis. Oncogene 2014;33(26):3361-3363. (103) Arikawa K, Takuwa N, Yamaguchi H, Sugimoto N, Kitayama J, Nagawa H, et al. Ligand-dependent inhibition of B16 melanoma cell migration and invasion via endogenous S1P2 G protein-coupled receptor. Requirement of inhibition of cellular RAC activity. J Biol Chem 2003 Aug 29;278(35):32841-32851.

SC

(104) Birt D, Merrill Jr A, Barnett T, Enkvetchakul B, Pour P, Liotta D, et al. Inhibition of skin carcinomas but not papillomas by sphingosine, N‐methylsphingosine, and N‐ acetylsphingosine. 1998.

M AN U

(105) Modica-Napolitano JS, Aprille JR. Delocalized lipophilic cations selectively target the mitochondria of carcinoma cells. Adv Drug Deliv Rev 2001 Jul 2;49(1-2):63-70. (106) Novgorodov SA, Szulc ZM, Luberto C, Jones JA, Bielawski J, Bielawska A, et al. Positively charged ceramide is a potent inducer of mitochondrial permeabilization. J Biol Chem 2005 Apr 22;280(16):16096-16105.

TE D

(107) Rossi MJ, Sundararaj K, Koybasi S, Phillips MS, Szulc ZM, Bielawska A, et al. Inhibition of growth and telomerase activity by novel cationic ceramide analogs with high solubility in human head and neck squamous cell carcinoma cells. Otolaryngol Head Neck Surg 2005 Jan;132(1):55-62.

EP

(108) Dindo D, Dahm F, Szulc Z, Bielawska A, Obeid LM, Hannun YA, et al. Cationic long-chain ceramide LCL-30 induces cell death by mitochondrial targeting in SW403 cells. Mol Cancer Ther 2006 Jun;5(6):1520-1529.

AC C

(109) Senkal CE, Ponnusamy S, Rossi MJ, Sundararaj K, Szulc Z, Bielawski J, et al. Potent antitumor activity of a novel cationic pyridinium-ceramide alone or in combination with gemcitabine against human head and neck squamous cell carcinomas in vitro and in vivo. J Pharmacol Exp Ther 2006 Jun;317(3):1188-1199. (110) Dahm F, Bielawska A, Nocito A, Georgiev P, Szulc ZM, Bielawski J, et al. Mitochondrially targeted ceramide LCL-30 inhibits colorectal cancer in mice. Br J Cancer 2008 Jan 15;98(1):98-105. (111) 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):177-181.

ACCEPTED MANUSCRIPT

(112) Struckhoff AP, Bittman R, Burow ME, Clejan S, Elliott S, Hammond T, et al. Novel ceramide analogs as potential chemotherapeutic agents in breast cancer. J Pharmacol Exp Ther 2004 May;309(2):523-532.

RI PT

(113) Crawford KW, Bittman R, Chun J, Byun HS, Bowen WD. Novel ceramide analogues display selective cytotoxicity in drug-resistant breast tumor cell lines compared to normal breast epithelial cells. Cell Mol Biol (Noisy-le-grand) 2003 Nov;49(7):1017-1023. (114) Stover T, Kester M. Liposomal delivery enhances short-chain ceramide-induced apoptosis of breast cancer cells. J Pharmacol Exp Ther 2003 Nov;307(2):468-475.

SC

(115) Stover TC, Sharma A, Robertson GP, Kester M. Systemic delivery of liposomal short-chain ceramide limits solid tumor growth in murine models of breast adenocarcinoma. Clin Cancer Res 2005 May 1;11(9):3465-3474.

M AN U

(116) Thomas DA, Sarris AH, Cortes J, Faderl S, O'Brien S, Giles FJ, et al. Phase II study of sphingosomal vincristine in patients with recurrent or refractory adult acute lymphocytic leukemia. Cancer 2006 Jan 1;106(1):120-127. (117) van Vlerken LE, Duan Z, Seiden MV, Amiji MM. Modulation of intracellular ceramide using polymeric nanoparticles to overcome multidrug resistance in cancer. Cancer Res 2007 May 15;67(10):4843-4850.

TE D

(118) Kedderis LB, Bozigian HP, Kleeman JM, Hall RL, Palmer TE, Harrison SD,Jr, et al. Toxicity of the protein kinase C inhibitor safingol administered alone and in combination with chemotherapeutic agents. Fundam Appl Toxicol 1995 May;25(2):201217.

EP

(119) De Luca T, Morre DM, Zhao H, Morre DJ. NAD+/NADH and/or CoQ/CoQH2 ratios from plasma membrane electron transport may determine ceramide and sphingosine-1phosphate levels accompanying G1 arrest and apoptosis. Biofactors 2005;25(1-4):4360.

AC C

(120) Gamble JR, Xia P, Hahn CN, Drew JJ, Drogemuller CJ, Brown D, et al. Phenoxodiol, an experimental anticancer drug, shows potent antiangiogenic properties in addition to its antitumour effects. Int J Cancer 2006 May 15;118(10):2412-2420. (121) Nagahashi M, Ramachandran S, Kim EY, Allegood JC, Rashid OM, Yamada A, et al. Sphingosine-1-phosphate produced by sphingosine kinase 1 promotes breast cancer progression by stimulating angiogenesis and lymphangiogenesis. Cancer Res 2012 Feb 1;72(3):726-735. (122) Paugh SW, Paugh BS, Rahmani M, Kapitonov D, Almenara JA, Kordula T, et al. A selective sphingosine kinase 1 inhibitor integrates multiple molecular therapeutic targets in human leukemia. Blood 2008 Aug 15;112(4):1382-1391.

ACCEPTED MANUSCRIPT

(123) Beljanski V, Lewis CS, Smith CD. Antitumor activity of sphingosine kinase 2 inhibitor ABC294640 and sorafenib in hepatocellular carcinoma xenografts. Cancer Biol Ther 2011 Mar 1;11(5):524-534.

RI PT

(124) Sabbadini RA. Sphingosine-1-phosphate antibodies as potential agents in the treatment of cancer and age-related macular degeneration. Br J Pharmacol 2011 Mar;162(6):1225-1238.

SC

Figure Legends

M AN U

Figure 1. Metabolism of ceramide and S1P. Ceramide lies at the center of sphingolipid metabolism. de novo generation of ceramide requires the condensation of Serine and Palmitoyl CoA by the enzyme Serine Palmitoyl CoA Transferase (SPT), to generate 3-ketosphinganine, which is then converted to dihydrosphingosine. Ceramide

TE D

Synthases (CerS1-6) generate dihydroceramide which gets desaturated to ceramide by Desaturase enzyme (DES). Ceramide can be converted to S1P via ceramidase followed by the action of SK1 or SK2. Ceramide can be generated back from S1P via

EP

sphingosine-1-phosphate-phosphatase (S1PP). Ceramide can be generated from sphingomyelin via sphingomyelinase (SMase) and from Glycosphingolipids via

AC C

hexosylceramidase. Ceramide also serves as the precursor of sphingomyelin via Sphingomyelin Synthase (SMS) and gangliosides using several enzymes such as hexosylceramide synthase and GM synthase.

Figure 2. Ceramide/S1P rheostat. The balance between ceramide and S1P signaling can determine whether a cancer cell would survive or proceed to cell death.

ACCEPTED MANUSCRIPT

Accumulation of ceramide promotes susceptibility to anti-cancer drugs and cell death;

AC C

EP

TE D

M AN U

SC

RI PT

however, accumulation of S1P is implicated with drug resistance and survival.

ACCEPTED MANUSCRIPT

Table 1. Ceramide based anticancer therapeutics Mechanism of action

Cancer Type

Pyridinium-Ceramides

Mitochondrial accumulation

Lung, breast, melanoma

C16-serinol

Ceramide analogue

Breast

4,6-diene-ceramide

Ceramide analogue

Neuroblastoma

Ceramide nanoparticles

Liposomes to improve delivery

D-MAPP

Ceramidase inhibitor

B13

Acid Ceramidase inhibitor

RI PT

Compound

Breast, leukemia

Squamous Cell Carcinomas

AC C

EP

TE D

M AN U

SC

Prostate, Colon, Head and Neck

ACCEPTED MANUSCRIPT

Table 2. Sphingosine kinase/S1P/S1PR based anti-cancer therapeutics Target

Status

Cancer Type

Dimethyl sphingosine

SK1,SK2

in vitro, in vivo

Leukemia, colon, breast, melanoma

Safingol

SK1,SK2

Phase I

Solid tumors

SKI-II

SK1,SK2

in vitro, in vivo

Breast

Phenoxodiol

SK1,SK2

in vitro, in vivo

Ovarian and prostate

SKI-I

SK1

in vitro, in vivo

Glioblastoma, breast, AML, melanoma

ABC294640

SK2

Phase II

Breast, prostate and renal

FTY-720

S1PR

in vitro, in vivo

Bladder, prostate, breast, lymphoma, melanoma

Anti-S1P-mAb

S1P

Phase I/II

Lung, ovarian, breast and melanoma

SC

M AN U

TE D EP AC C

RI PT

Compound

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT