Published OnlineFirst October 17, 2018; DOI: 10.1158/0008-5472.CAN-18-0345

Cancer Review Research

Glioma Cell Secretion: A Driver of Tumor Progression and a Potential Therapeutic Target Damian A. Almiron Bonnin1,2, Matthew C. Havrda1,2, and Mark A. Israel1,2,3

Abstract

Cellular secretion is an important mediator of cancer progres- ple oncogenic pathologies. In this review, we describe tumor cell sion. Secreted molecules in glioma are key components of secretion in high-grade glioma and highlight potential novel complex autocrine and paracrine pathways that mediate multi- therapeutic opportunities. Cancer Res; 78(21); 6031–9. 2018 AACR.

Introduction Glioma-Secreted Molecules Impact Disease Glial cells in the central nervous system (CNS) provide trophic Progression support for neurons (1). In glial tumors, this trophic support is Glioma cells modify their microenvironment by introducing dysregulated creating a pro-oncogenic microenvironment medi- diverse molecules into the extracellular space (Table 1). To exem- ated by a heterogeneous array of molecules secreted into the plify the pro-oncogenic role that secreted molecules can have on – extracellular space (2 15). The glioma secretome includes pro- glioma pathology, we review the functional impact of specific teins, nucleic acids, and metabolites that are often overexpressed cytokines, metabolites, and nucleic acids on glioma biology. By in malignant tissue and contribute to virtually every aspect of describing some of the potent antitumorigenic effects observed in – cancer pathology (Table 1; Fig. 1; refs. 2 15), providing a strong preclinical therapeutic studies targeting tumor cell secretion, we – rationale to target the cancer cell secretory mechanisms. also highlight how blocking secreted molecules might be of fi Although the speci c mechanisms regulating secretion in therapeutic impact in gliomas, as well as other tumors. malignant cells remain to be fully characterized, there is signif- icant evidence that the secretory mechanisms themselves are Cytokines altered during oncogenesis (8, 16–36). Well-known mediators Cytokines are essential mediators of cellular signaling (2, 13, of secretion, like the ADP-ribosylation factors (ARF) and the small 15). In glioma, secreted cytokines, including IL1b, IL6, and IL8, GTPase (RAB) have been reported to be dysregulated create a state of chronic inflammation that promotes the malig- in glioma and several other tumors (17, 20, 22, 28, 31, 32, 37–41). nant phenotype (15). These cytokines are associated with poor These proteins facilitate secretion of pro-oncogenic molecules prognosis for patients with high-grade gliomas (HGG; refs. 2, 13, (28, 42) and their inhibition diminishes multiple aspects of 15). Both in vitro and in vivo studies demonstrate that targeting cancer pathology including cellular proliferation, survival, and these mediators of inflammation inhibits important aspects of invasion (20, 22, 28, 32, 37–39, 41, 42), while showing no signs glioma pathology including angiogenesis, proliferation, and inva- of obvious toxicities in animal models (18, 19, 21, 26, 36, 43–46). sion (2, 13, 15). It has also been shown that cytokines, like IL6, This reliance of cancer cells on secretory pathways is exemplified IL8, EGF, and TGFb, promote resistance to antineoplastic therapy by the unfolded response (UPR; ref. 18). UPR activation is in glioma (15, 24, 47, 48), breast (49), and prostate cancer (50). In thought to be crucial for oncogenic progression (18), and agents melanoma, secretion has been identified as an important mech- inhibiting the UPR have shown potent antitumorigenic effects in anism facilitating the emergence of drug resistance via the acti- models of glioma, multiple myeloma, and pancreatic cancer (18). vation of the AKT pathway (51). Importantly, these cytokines also Tumor cell secretory "addiction" describes the dependence of facilitate the maintenance of cancer stem cells (Table 1; ref. 15), tumor cells on secretory pathways like the UPR (18), and suggest which are largely refractory to therapy, and play an important role a potential therapeutic window to target these pathways. The in cancer progression (52). functional impact of secreted molecules and secretory pathways Platelet-derived growth factor (PDGF), one of the best charac- on glioma biology underscores the potential therapeutic implica- terized cytokines in HGGs and other cancers (2, 13, 53–55), is the tions of targeting the tumor cell secretion (Table 1). ideal example to illustrate the functional impact of cytokines on cancer biology. Autocrine PDGF signaling was determined to play 1Department of Molecular and Systems Biology, Geisel School of Medicine at an important role in malignant transformation (2), and mouse Dartmouth, Hanover, New Hampshire. 2Norris Cotton Cancer Center, Geisel models of HGG demonstrate that PDGF signaling is sufficient for School of Medicine at Dartmouth, Lebanon, New Hampshire. 3Departments of tumor initiation and progression (13, 54). Dysregulated PDGF Medicine and Pediatrics, Geisel School of Medicine at Dartmouth, Hanover, New signaling activates MAPK-ERK and PI3K-AKT, two nodal points Hampshire. critical for cell proliferation, resistance to , and invasion Corresponding Author: Mark A. Israel, M.D., Norris Cotton Cancer Center at (10, 12, 56). PDGF-mediated PI3K/AKT activation has also been Dartmouth, One Medical Center Drive, Lebanon, NH 03756. Phone: 603-653- shown to regulate glucose metabolism facilitating the Warburg 3611; Fax: 603-653-9003; E-mail: [email protected] effect in HGGs (12). In vitro and in vivo studies confirm PDGF's doi: 10.1158/0008-5472.CAN-18-0345 recognized function enhancing tumor angiogenesis by stimulat- 2018 American Association for Cancer Research. ing endothelial cell migration and promoting endothelial cell

www.aacrjournals.org 6031

Downloaded from cancerres.aacrjournals.org on October 6, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst October 17, 2018; DOI: 10.1158/0008-5472.CAN-18-0345

Almiron Bonnin et al.

Table 1. Glioma-secreted molecules and the hallmarks of cancer they affect Secreted molecules functional in glioma Impacted hallmark of cancer Platelet-derived growth factor (2, 116), hepatocyte growth factor (56, 117), insulin-like growth factor (56, 118), Sustaining proliferative transforming growth factor a (119), adrenomedullin (120), epidermal growth factor receptor variant III (121), signaling sphingosine-1-phosphate (122) microRNA-17 (14, 123), microRNA-19 (14, 123), microRNA-21 (14, 123), microRNA-24 (123, 124), microRNA-26a (14, 123), Evading growth suppression microRNA-221/222 (123, 124) Transforming growth factor b2 (125), interleukin-10 (126), kynurenine (11), lactate dehydrogenase (14, 127), osteopontin (128) Avoiding immune destruction Vascular endothelial growth factor (43), basic fibroblast growth factor (5, 14), interleukin-6 (15, 129), interleukin-8 (15, 130), Enabling replicative C-X-C motif chemokine ligand 12 (131, 132), gremlin 1 (133), sema 3C (134), periostin (135, 136), sphingosine-1-phosphate immortality/ stemness (122), telomerase reverse transcriptase transcript (14), transferrin (137) Tumor necrosis factor a (138), interleukin-1b (138), interleukin-6 (15, 129), interleukin-8 (15, 139), glutamate (140) Tumor-promoting inflammation Transforming growth factor a (6, 119), hepatocyte growth factor (117), EGF (6), periostin (135, 136), osteopontin (128), C-X-C Activating invasion and motif chemokine ligand 12 (131), glial cell–derived neurotrophic factor (141), urokinase-type plasminogen activator metastasis (142), protease nexin 1 (143), metalloproteinase 2 (144), metalloproteinase 9 (144), autotaxin (145), kynurenine (11), glutamate (146), versican (147), laminins (148), metastasis-associated lung adenocarcinoma noncoding RNA (14, 149), sphingosine-1-phosphate (6, 122), microRNA-20a (14, 123), microRNA-21 (14, 123) Vascular endothelial growth factor (14), TGFb2 (125, 150), fibroblast growth factor (14, 150), hepatocyte growth factor Inducing angiogenesis (117, 150), epidermal growth factor (6, 150), interleukin-6 (15, 129), interleukin-8 (15, 138), C-X-C motif chemokine ligand 12 (151), angiogenin (14, 152), platelet-derived growth factor (116, 150) Kynurenine (4, 11), human endogenous retrovirus retrotransponson (3, 153), long interspersed nuclear element 1 Genome instability retrotransponson (3, 154), arthrobacter luteus (Alu) retrotransponson (3, 155) Vascular endothelial growth factor (14, 56), fibroblast growth factor (5, 14, 56), epidermal growth factor (6, 56), interleukin-6 Resisting cell death (15, 129), Sema 3C (134), microRNA-21 (14, 123), microRNA-92 (14, 123) Platelet derived growth factor (12, 116, 156), vascular endothelial growth factor (14, 156), fibroblast growth factor (14, 156), Deregulating cellular energetics hepatocyte growth factor (117, 156), epidermal growth factor (6, 156)

proliferation (2). Consistent with these findings, disrupting PDGF of disease progression (62). In glioma and other tumors, signaling markedly reduces angiogenesis, tumor growth, and nucleic acids can be secreted in extracellular vesicles (EV) and invasion in multiple mouse models of glioma (2, 10, 12, 13). deliveredtonearbycells(3,7,9, 14). EVs can carry mutated or amplified oncogene sequences, mRNA, transposable elements, Metabolites ormiRNAs(7,9,14).EVsfromHGGcontainanarrayofpro- Malignant cells reprogram their metabolism to meet the bio- oncogenic miRNAs such as miR-19b, miR-20, and miR-21 that energetic and biosynthetic demands of tumor growth (57). A promote HGG progression (14). Importantly, genetic material consequence of this altered metabolism is the aberrant produc- contained in these EVs, such as mRNA and miRNAs, can be tion of metabolites that can have profound effects on tumor delivered to nearby cells where they remain functional (7, 9, biology (57). Studies suggest that several metabolites, including 14). Glioma-derived cells incubated in medium supplemented well-known molecules like glutamate and 2-hydroxyglutarate, are with EVs from patient-derived glioblastoma cells exhibit involved in glioma progression (58, 59). increased proliferation when compared with glioma cells incu- A recently described, secreted pro-oncogenic metabolite in bated in normal, untreated growth medium (14). Although glioma is kynurenine (4, 11). Kynurenine is an intermediate in further experimentation is required to demonstrate specific L-tryptophan catabolism and an endogenous aryl hydrocarbon effects of each nucleic acid carried within EVs, it is highly likely receptor (AhR) agonist (4, 11). At levels secreted by glioma cells, that they are, at least in part, responsible for these EV-mediated but not at levels secreted by normal cells, kynurenine promotes effects in glioma. genomic instability of HGG-derived cells (4). AhR activation by Studies have demonstrated that EVs derived from glioma cells secreted kynurenine upregulates the trans-lesion synthesis poly- can transform fibroblasts and epithelial cells (8). Consistent with merase, hpol k (4). The dysregulated overexpression of this these findings, targeting proteins required for EV biogenesis polymerase, which normally mediates the DNA damage tolerance effectively blocks EV-induced oncogenic transformation (8). pathway, increases genomic instability and promotes tumorigen- These findings suggest that DNA transfer in EVs could transform esis (4, 60). Such an autocrine pathway mediating increased nearby cells contributing to the high degree of heterogeneity and genomic instability may contribute to the exceptionally high polyclonality observed in human HGGs (61, 63). levels of genomic heterogeneity characterizing HGG (61). Kynurenine also has been shown to have immunosuppressive Molecular Mechanisms of Secretion effects, to enhance cellular motility and to promote survival of HGG cells (11). Inhibition of kynurenine secretion in glioma Classical secretory pathway increases the lysis of glioma cells by alloreactive blood mononu- Malignant cells utilize the classical secretory pathway. In this clear cells, decreases cellular migration in vitro, and markedly pathway, proteins with a signal peptide are recognized by the decreases clonogenic survival of glioma cells (11). Consistent signal recognition particle and then inserted into the endoplas- with these observations, kynurenine-secreting tumors grow sig- mic reticulum (ER; ref. 64). In the ER, translation is completed nificantly more rapidly and have a higher proliferation index than and the newly synthesized proteins are transported through the nonsecreting tumors in immunocompetent mice (11). Golgi apparatus to the plasma membrane in a process mediated by numerous proteins including RABs and ARFs (64). Nucleic acids Accumulating evidence indicates that RABs and ARFs are of Secreted nucleic acids are commonly found in the blood of significant importance for cancer progression (17, 20, 28, 37, 65, patients with cancer and they have been shown to be indicative 66). Aberrant expression of RAB proteins, a family of more than

6032 Cancer Res; 78(21) November 1, 2018 Cancer Research

Downloaded from cancerres.aacrjournals.org on October 6, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst October 17, 2018; DOI: 10.1158/0008-5472.CAN-18-0345

Glioma Secretion Drives Tumor Progression

Sustaining proliferative Evading growth signaling suppressors

Deregulating Avoiding immune cellular energetics destruction Membrane Classical transport proteins secretion

Type II Type IV or or ABC Golgi transporter Golgi Bypass based Enabling Resisting secretion replicative cell death Nucleus ER immortality

Type III or auto- Exosomes Multivesicular body phagosome based secretion

Type I or pore Microvesicles based secretion Genome instability Tumor-promoting & mutation inflammation

Inducing Activating invasion angiogenesis & metastasis

© 2018 American Association for Cancer Research

Figure 1. Schematic representation of the impact of glioma secretion on the hallmarks of cancer. Pro-oncogenic molecules can be transported through the plasma membrane by classical secretory mechanisms, nonclassical secretory mechanisms (type I–IV; refs. 5, 15, 16, 19, 21, 25, 26, 29, 33, 72–81, 83), microvesicles, and exosomes (3, 8, 9, 14, 28, 32, 36). Molecules secreted by these mechanisms have been reported to contribute to each of the hallmarks of cancer (red arrows; refs. 2–15), as defined by Hanahan and Weinberg (114).

70 members, is observed in glioma and other cancers (22, 28, 31, in vitro, and this oncogenic RAB35 was sufficient to drive vesicular 32, 40, 67, 68). In HGG RAB3A, RAB34, and RAB27A expression transport in the absence of stimuli, suggesting that dysregulated levels correlate with survival and pathologic grade (22, 31, 40). vesicular trafficking can contribute to oncogenesis (70). RAB3A, which regulates vesicular transport in neurons (69), is Like the RAB family of proteins, ARFs and their associated associated with increased proliferation and chemo- and radio- guanine nucleotide exchange factors (GEF) and GTPase-activating resistance of cultured glioma cells and tumor formation and proteins (GAP) are emerging as novel mediators of disease pro- glioma growth in nude mice (22). A recent study has identified gression in gliomas, as well as other tumors (17, 20, 36–39, RAB35 as a novel activator of PI3K in multiple cells from kidney, 65, 66). Dysregulated ARF6 has been reported to be a key driver colon, prostate, and cervical cancer. Somatic RAB35 mutations of tumor cell invasion in several cancer types including glioma found in human tumors were capable of transforming cells (17, 20, 65, 66). A glioma study has shown that ARF6 knockdown

www.aacrjournals.org Cancer Res; 78(21) November 1, 2018 6033

Downloaded from cancerres.aacrjournals.org on October 6, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst October 17, 2018; DOI: 10.1158/0008-5472.CAN-18-0345

Almiron Bonnin et al.

by siRNA markedly decreases invasion of intracranial brain xeno- useful in the clinic due to the side effects they cause (19). Multiple grafts in nude mice (20). ARF6 has also been shown to orchestrate strategies to prevent such drug toxicities are currently under the activity of the oncogenic guanine nucleotide–binding protein investigation (26). G(q) subunit alpha (GNAQ) in uveal melanoma by mediating the The type III pathway of nonclassical secretion relies on autop- vesicular transport of GNAQ and b-catenin (71). Consistent with hagosomes (72), double-membraned vesicles that enclose cellu- this finding, targeting ARF6 with the small-molecule inhibitor, lar material destined to be degraded or secreted (25). Secretory NAV-2729, reduces melanoma proliferation and tumorigenesis autophagosomes carry cytokines including IL1b, and IL18 (25, in vivo (71). 72), which are well-characterized mediators of glioma progres- sion (15, 75, 76). The process of autophagosome formation is Nonclassical secretory pathway tightly controlled by highly conserved autophagy-related A substantial proportion of cancer secretion is mediated by (ATG; refs. 25, 72). These genes, as well as the process of autop- the nonclassical secretory pathway (29). This pathway is hagy, can have pro-oncogenic or antioncogenic effects in HGG induced by cellular stress (72), a characteristic of transformed (30, 79, 80). For example, targeting ATG7 or ATG13 has been cells (18, 73). There are four types of nonclassical pathway shown to result in markedly decreased KRAS-driven HGG growth secretion, which may play a role in cancer: type I or pore- in vivo (80), while inhibiting ATG5 enhances the resistance of mediated secretion, type II or ABC transporter-mediated secre- cultured HGG cells to temozolomide, a drug routinely used in the tion, type III or endosome/autophagosome-mediated secretion, treatment of glioblastoma (30). The mechanisms behind this dual and type IV or Golgi-bypass secretion (72). role of ATGs in HGG remain to be elucidated (79). Proteins secreted by the type I pathway rely on the formation of The type IV nonclassical pathway mediates the secretion of pores in the plasma membrane. The formation of these pores can proteins during ER stress that bypass the Golgi enroute to the be mediated by either the cytoplasmic protein to be secreted or by plasma membrane following synthesis (72). The IRE1a protein of inflammation (72). Secretion of FGF2, a widely studied pro- the UPR activates this pathway in response to ER stress (18, 81), oncogenic protein of importance in glioma (5), is a well-known which is provoked by many conditions common to malignant example of the former mechanism (72). FGF2 binds to phospha- tissues including hypoxia, nutrient deprivation, increased meta- tidylinositol 4, 5-bisphosphate (PI(4,5)P2) on the plasma mem- bolic activity, and high levels of proliferation (18, 73, 82). Human brane and oligomerizes promoting the formation of a lipidic pore glioma cells expressing a nonfunctional mutant of IRE1a dem- through which secretion occurs (72). Alternatively, pore forma- onstrate markedly decreased intracranial tumor growth in nude tion can be mediated by inflammation (72, 74), which charac- mice (82). Golgi bypass in the type IV pathway is mediated by an terizes the glioma microenvironment (75). During inflammation, ER-sorting machinery that includes heat shock proteins (HSP; ref. gasdermin D, gasdermin A, and gasdermin A3 are cleaved by 72). The role of HSPs in HGGs has been well described (33, 83). caspase-1 releasing the N-terminal half of these proteins: gasder- For example, HSP70, which is upregulated in HGGs (33, 83), min-N, which binds PIP2 on the plasma membrane and forms 16- increases tumor formation, survival, and chemoresistance in fold-symmetry in the pore (72, 74). These pores then mediate the glioblastoma (33, 83). secretion of IL1b (74), a cytokine known to promote further inflammation and drive glioma growth (75, 76). While a role Extracellular vesicle formation and secretion for the gasdermin protein family in glioma has not been pro- Many different terms are used in the literature to describe EVs posed, members of this family are aberrantly expressed in several including microvesicles, exosomes, oncosomes, microparticles, tumor types (16) and promote brain metastasis of tumors from and ectosomes (84, 85). Even although there is considerable outside the CNS (77). debate in the field as to the proper usage of these terms (84, 85), The type II nonclassical pathway relies on the ATP-binding EVs can be broadly classified into two main groups based on cassette (ABC) transporters (72), which are well-described med- their biogenesis: microvesicles and exosomes (36). Microvesi- iators of chemoresistance in glioma as well as other tumor types cles are derived from the plasma membrane, while exosomes (27, 78). These transporters facilitate chemoresistance by medi- originate from multivesicular endosomal compartments (36). ating the efflux of chemotherapeutic agents from malignant cells However, experimental procedures designed to isolate each of (27, 78). Overexpression of the ABC transporter subfamily B these vesicle types, including centrifugation protocols, com- member 1, which is also known as the multidrug resistance mercial kits, and filters,arebasedonthesizeandweightof protein 1 (MDR1), mediates resistance to chemotherapeutic these vesicles, and not on their intracellular origin (86, 87). agents in aggressive gliomas (27, 78). Inhibiting MDR1, in vitro Consequently, material isolated by these experimental techni- or in vivo, increases the sensitivity of HGG cells to vincristine and ques contain a mixture of different types of EVs (86, 87), and temozolomide, two antineoplastic agents commonly used in the thus it is important to consider investigational findings in the management of patients with glioblastoma (78). Furthermore, light of these experimental limitations. MDR1 inhibitors have been shown to lead to increased concen- In cancer, EVs transport proteins, lipids, and nucleic acid (36) trations of chemotherapy agents in the brain when used as and can promote multiple aspects of cancer progression (3, 9, 14, adjuvants (19, 21). MDR1 is also expressed in endothelial cells 32, 36, 66), and even induce malignant transformation (8, 88). of the blood–brain barrier (19), where it pumps metabolic waste The small Ras homolog family member A (RhoA) GTPase products out of the CNS as well as enhancing the efflux of regulates EV biogenesis in glioma cells (8, 36). Studies demon- chemotherapy agents and decreasing their availability in brain strate that RhoA facilitates the activation of Rho kinase (ROCK) tumors (19). Thus, clinical therapies that effectively inhibited and subsequently the (LIMK; refs. 8, 36). LIMK these transporters could potentially have the added advantage of phosphorylates cofilin causing a reorganization of actin fibers increasing the CNS availability of chemotherapy agents active in and microvesicle shedding (8, 36). Inhibition of these proteins glioma (21), although to date, MDR1 inhibitors have not been has been shown to markedly reduce microvesicle release in

6034 Cancer Res; 78(21) November 1, 2018 Cancer Research

Downloaded from cancerres.aacrjournals.org on October 6, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst October 17, 2018; DOI: 10.1158/0008-5472.CAN-18-0345

Glioma Secretion Drives Tumor Progression

glioma-derived cells (8). Inhibition of the RhoA–ROCK–LIMK EI, which inhibits Sec61-mediated protein translocation in pathway reduces cellular proliferation and migration in glioma the classical secretory pathway (44), has potent antioncogenic (8, 35). Pharmacologic inhibition of this pathway by the ROCK activity (46, 89). Preclinical studies demonstrated that EI treat- inhibitor, Y-27632, suppresses migration and proliferation of ment causes a significant reduction of invasion and cellular glioma-derived cells. Similarly, silencing of LIMK reduced growth proliferation of a non–smallcelllungcarcinoma–derived cell of a breast cancer xenograft in nude mice (8). Consistent with line, while increasing cell death by apoptosis (46, 89). In vivo,EI these observations, RhoA, which mediates the activation of ROCK treatment resulted in reduced xenograft growth in nude mice and LIMK as discussed previously (8, 36), is highly expressed in with no signs of toxicity (46). Other Sec61 inhibitors, like HGGs and its expression is correlated with pathologic grade (34). apratoxin (104) and cotransins (105), have also been shown RAB and ARF family members, which were discussed in previous to have significant antitumorigenic activity in preclinical stud- sections, have also been implicated in EV formation in gliomas as ies (106–108). However, these agents, as well as other Sec61 well as other types of cancer (32, 36, 66). inhibitors, like decatransin (109) and mycolactone (110), remain to be studied in the context of HGG, and they have Therapeutic Opportunities yet to be studied in clinical trials for any tumor type. These pharmacologic inhibitors of secretion represent a relatively Targeting secretion in glioma unexplored therapeutic opportunity for tumors, like HGG, that The concept of tumor cell–secretory addiction describes the rely on secretory pathways, as discussed above. reliance of cancer cells on secreted molecules and their secretory Another potential target to inhibit cancer-associated secretion machinery (18). Studies described above demonstrate the signif- is the UPR (18). Treatment of HGG cells with epigallocatechin icance of this concept in HGG (Table 1). By describing some of the gallate, which inhibits GRP78, significantly increases the cytotoxic essential functions of glioma-secreted molecules and the mechan- effect of temozolomide on HGG (90). This drug is currently being isms facilitating their secretion in glioma pathology (Fig. 1), we evaluated as an adjuvant for treatment of colorectal cancer in a have sought to underscore the potential therapeutic benefits of phase I clinical trial (111). Similarly, inhibition of the glucose- targeting secretion in HGGs. Targeting proteins of the secretory regulated protein 78 (GRP78), a key regulator of the UPR path- mechanism in glioma has been shown to inhibit key aspects of way, by the novel fusion protein EGF-SubA results in substantial glioma biology including tumor cell proliferation and survival growth reduction of HGG xenografts in nude mice (45). (Table 1; Fig. 1; refs. 8, 17–23, 26–28, 32, 34–36, 38, 39, 44– Botulinum neurotoxin–based pharmacologic agents represent 46, 89, 90). Furthermore, targeting secretion seems to selectively another promising strategy to inhibit secretion (112). These affect transformed cells, while sparing normal cells, as evidenced drugs, which are known as targeted secretion inhibitors (TSI), by the lack of toxicities reported in multiple animal experiments can target specific cells and cleave SNARE proteins (112). TSIs testing agents that inhibit secretion (18, 19, 21, 26, 36, 43–46). contain a targeting domain, a transport domain, and a blocking This suggests that targeting proteins of the secretory machinery to domain. The targeting domain can be engineered to bind classic disturb the secretion of molecules that enhance tumor pathology glioblastoma markers, such as EGFR (113). The transport domain may be a promising treatment strategy. facilitates the entry of this agent into the cytosol, where the Brefeldin A, a known secretion inhibitor (23), and EHT-1864, enzymatic blocking domain cleaves SNARE proteins to inhibit which inhibits Rac1 (91), a known mediator of secretion (92, 93), protein secretion (112). Even although TSIs have yet to be dramatically reduce VEGF secretion, decrease GSC self-renewal, explored in the context of cancer, their characteristics discussed and inhibit tumor growth in a mouse model of HGG (54). The above make them well suited for cancer therapeutics. antitumorigenic effects of these agents also have been observed in several other cancer types (38, 39, 41). Multiple high-throughput drug screens have identified novel small-molecule inhibitors that Conclusion effectively target mediators of cellular secretion (94). Two small- Cancer cell secretion is a vital process contributing to glioma molecule inhibitors of secretion that have been studied in cancer progression. Numerous studies have characterized the autocrine are AMF-26 (37) and Eeyarestatin I (EI; ref. 44). Like Brefeldin A, and paracrine pathways that facilitate many key aspects of tumor AMF-26 targets the ARF1-guanine nucleotide exchange factor progression described as hallmarks of cancer by Hanahan and (GEF) interaction, which prevents the assembly of the vesicle Weinberg (Table 1; Fig. 1; ref. 114). The capacity of cancer cells to coating protein complex I (COPI) assembly and consequently secrete a wide range of different soluble factors with redundant inhibits protein secretion (37). AMF-26 has potent antitumor functions (Table 1) could also explain cancer resistance to anti- activity (37). Oral administration of AMF-26 resulted in complete neoplastic therapies. In HGG, studies have shown that when regression of human breast cancer xenografts in nude mice, and signaling from a specific secreted factor is blocked, an alternative was nontoxic to animals (37) providing further evidence of the secreted factor can maintain the oncogenic functions of the potential of such drugs to have an acceptable therapeutic index. secreted factor that was blocked (54, 115). These secreted factors Early preclinical studies also suggest that other pharmacologic have also been observed to promote resistance to radiation and agents that result in the inhibition of ARF1-mediated COPI chemotherapy (15, 24, 47–51). Therefore, targeting the secretory assembly, like Exo2 (8), LM11 (95), and Golgicide A (96), have mechanisms of cancer cells could potentially reduce simulta- significant antitumorigenic activity in breast and prostate cancer neously the levels of multiple pro-oncogenic secreted factors and models (97–99). However, these agents, as well as other agents consequently diminish cancer drug resistance and increase patient that block ER-to-Golgi vesicular transport, including Exo1 (100), survival. However, despite accumulating evidence demonstrating LG186 (100), CI-976 (101), dispergo (102), and FLI-06 (103), the importance of these secretory mechanisms in glioma biology have not been tested in glioblastoma and their clinical value for (8, 16–36), there are currently no clinically available therapies cancer therapeutics remains largely unexplored. targeting these mechanisms.

www.aacrjournals.org Cancer Res; 78(21) November 1, 2018 6035

Downloaded from cancerres.aacrjournals.org on October 6, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst October 17, 2018; DOI: 10.1158/0008-5472.CAN-18-0345

Almiron Bonnin et al.

As pharmacologic agents targeting secretory mechanisms Disclosure of Potential Conflicts of Interest emerge, it will become important to cautiously consider their No potential conflicts of interest were disclosed. safety. Although several drugs targeting secretory mechanisms have been tested in animal models with no observable toxicities Acknowledgments (18, 19, 21, 26, 36, 43–46), inhibiting such an essential cellular Support was generously provided by the Theodora B. Betz Foundation function as secretion poses obvious risks. Many cell types, and (toM.A.Israel),theJordanandKyra Memorial Foundation (to M.A. Israel), and the Andrea Clark Nelson Medical Research Endowment especially neurons, glial cells, and immune cells, require secretion (to M.A. Israel). to function properly. Much like current chemotherapeutic drugs, dose levels and dosing schedules are likely to require careful evaluation to target secretion from malignant cells, while sparing Received February 2, 2018; revised May 30, 2018; accepted August 14, 2018; secretion from normal cells. published first October 17, 2018.

References 1. Du Y, Dreyfus CF. Oligodendrocytes as providers of growth factors. 19. Gottesman MM, Fojo T, Bates SE. Multidrug resistance in cancer: role of J Neurosci Res 2002;68:647–54. ATP–dependent transporters. Nat Rev Cancer 2002;2:48–58. 2. Andrae J, Gallini R, Betsholtz C. Role of platelet-derived growth factors in 20. Hu B, Shi B, Jarzynka MJ, Yiin JJ, D'Souza-Schorey C, Cheng SY. ADP- physiology and medicine. Genes Dev 2008;22:1276–312. ribosylation factor 6 regulates glioma cell invasion through the IQ- 3. Balaj L, Lessard R, Dai L, Cho Y-J, Pomeroy SL, Breakefield XO, et al. domain GTPase-activating protein 1-Rac1-mediated pathway. Cancer Res Tumour microvesicles contain retrotransposon elements and amplified 2009;69:794–801. oncogene sequences. Nat Commun 2011;2:180. 21. Hubensack M, Muller C, Hocherl P, Fellner S, Spruss T, Bernhardt G, 4. Bostian AC, Maddukuri L, Reed MR, Savenka T, Hartman JH, Davis L, et al. et al. Effect of the ABCB1 modulators elacridar and tariquidar on the Kynurenine signaling increases DNA polymerase kappa expression and distribution of paclitaxel in nude mice. J Cancer Res Clin Oncol promotes genomic instability in glioblastoma cells. Chem Res Toxicol 2007;134:597–607. 2016;29:101–8. 22. Kim JK, Lee SY, Park C, Park SH, Yin J, Kim J, et al. Rab3a promotes brain 5. Haley EM, Kim Y. The role of basic fibroblast growth factor in glioblas- tumor initiation and progression. Mol Biol Rep 2014;41:5903–11. toma multiforme and glioblastoma stem cells and in their in vitro culture. 23. Misumi Y, Misumi Y, Miki K, Takatsuki A, Tamura G, Ikehara Y. Novel Cancer Lett 2014;346:1–5. blockade by brefeldin A of intracellular transport of secretory proteins in 6. Hoelzinger DB, Demuth T, Berens ME. Autocrine factors that sustain cultured rat hepatocytes. J Biol Chem 1986;261:11398–403. glioma invasion and paracrine biology in the brain microenvironment. J 24. Munoz JL, Rodriguez-Cruz V, Greco SJ, Nagula V, Scotto KW, Rameshwar Natl Cancer Inst 2007;99:1583–93. P. Temozolomide induces the production of epidermal growth factor to 7. Katakowski M, Buller B, Wang X, Rogers T, Chopp M. Functional micro- regulate MDR1 expression in glioblastoma cells. Mol Cancer Ther 2014; RNA is transferred between glioma cells. Cancer Res 2010;70:8259–63. 13:2399–411. 8. Li B, Antonyak MA, Zhang J, Cerione RA. RhoA triggers a specific signaling 25. Ponpuak M, Mandell M, Kimura T, Chauhan S, Cleyrat C, Deretic V. pathway that generates transforming microvesicles in cancer cells. Secretory autophagy. Curr Opin Cell Biol 2015;35:106–16. Oncogene 2012;31:4740–9. 26. Singh MS, Tammam SN, Shetab Boushehri MA, Lamprecht A. MDR in 9. Li CCY, Eaton SA, Young PE, Lee M, Shuttleworth R, Humphreys DT, et al. cancer: addressing the underlying cellular alterations with the use of Glioma microvesicles carry selectively packaged coding and non-coding nanocarriers. Pharmacol Res 2017;126:2–30. RNAs which alter in recipient cells. RNA Biol 2013;10: 27. Tivnan A, Zakaria Z, O'Leary C, Kogel€ D, Pokorny JL, Sarkaria JN, et al. 1333–44. Inhibition of multidrug resistance protein 1 (MRP1) improves chemo- 10. Lokker N, Sullivan C, Hollenbach S, Israel M, Giese N. Platelet-derived therapy drug response in primary and recurrent glioblastoma multiforme. growth factor (PDGF) autocrine signaling regulates survival and mito- Front Neurosci 2015;9:218. genic pathways in glioblastoma cells: evidence that the novel PDGF-C and 28. Tzeng H-T, Wang Y-C. Rab-mediated vesicle trafficking in cancer. J Biomed PDGF-D ligands may play a role in the development of brain tumors. Sci 2016;23:70. Cancer Res 2002;62:3729–35. 29. Villarreal L, Mendez O, Salvans C, Gregori J, Baselga J, Villanueva J. 11. Opitz CA, Litzenburger UM, Sahm F, Ott M, Tritschler I, Trump S, et al. An Unconventional secretion is a major contributor of cancer cell line endogenous tumour-promoting ligand of the human aryl hydrocarbon secretomes. Mol Cell Proteomics 2013;12:1046–60. receptor. Nature 2011;478:197–203. 30. Voss V, Senft C, Lang V, Ronellenfitsch MW, Steinbach JP, Seifert V, et al. 12. C, Liu H, Hitoshi Y, Israel M. Proliferation-independent control of The Pan-Bcl-2 inhibitor ()-Gossypol triggers autophagic cell death in tumor glycolysis by PDGFR-mediated AKT activation. Cancer Res malignant glioma. Mol Cancer Res 2010;8:1002–16. 2013;73:1831–43. 31. WangHJ,GaoY,ChenL,LiYL,JiangCL.RAB34wasaprogression-and 13. Shih AH, Holland EC. Platelet-derived growth factor (PDGF) and glial prognosis-associated biomarker in gliomas. Tumour Biol 2014;36: tumorigenesis. Cancer Lett 2006;232:139–47. 1573–8. 14. Skog J, Wurdinger T, van Rijn S, Meijer D, Gainche L, Sena-Esteves M, et al. 32. Wang T, Gilkes DM, Takano N, Xiang L, Luo W, Bishop CJ, et al. Hypoxia- Glioblastoma microvesicles transport RNA and protein that promote inducible factors and RAB22A mediate formation of microvesicles that tumor growth and provide diagnostic biomarkers. Nat Cell Biol 2008; stimulate breast cancer invasion and metastasis. Proc Natl Acad Sci U S A 10:1470–6. 2014;111:E3234–42. 15. Yeung YT, McDonald KL, Grewal T, Munoz L. Interleukins in glioblastoma 33. Wu J, Liu T, Rios Z, Mei Q, Lin X, Cao S. Heat shock proteins and cancer. pathophysiology: implications for therapy. Br J Pharmacol 2013;168: Trends Pharmacol Sci 2017;38:226–56. 591–606. 34. Yan B, Chour HH, Peh BK, Lim C, Salto-Tellez M. RhoA protein expression 16. Carl-McGrath S, Schneider-Stock R, Ebert M, Rocken€ C. Differential correlates positively with degree of malignancy in astrocytomas. Neurosci expression and localisation of gasdermin-like (GSDML), a novel member Lett 2006;407:124–6. of the cancer-associated GSDMDC protein family, in neoplastic and non- 35. Zohrabian VM, Forzani B, Chau Z, Murali RAJ, Jhanwar-Uniyal M. Rho/ neoplastic gastric, hepatic, and colon tissues. Pathology 2008;40:13–24. ROCK and MAPK signaling pathways are involved in glioblastoma cell 17. Casalou C, Faustino A, Barral DC. Arf proteins in cancer cell migration. migration and proliferation. Anticancer Res 2009;29:119–23. Small 2016;7:270–82. 36. van Niel G, D'Angelo G, Raposo G. Shedding light on the cell biology of 18. Dejeans N, Manie S, Hetz C, Bard F, Hupp T, Agostinis P, et al. Addicted to extracellular vesicles. Nat Rev Mol Cell Biol 2018;19:213–28. secrete - novel concepts and targets in cancer therapy. Trends Mol Med 37. Ohashi Y, Iijima H, Yamaotsu N, Yamazaki K, Sato S, Okamura M, et al. 2013;20:242–50. AMF-26, a novel inhibitor of the Golgi system, targeting ADP-ribosylation

6036 Cancer Res; 78(21) November 1, 2018 Cancer Research

Downloaded from cancerres.aacrjournals.org on October 6, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst October 17, 2018; DOI: 10.1158/0008-5472.CAN-18-0345

Glioma Secretion Drives Tumor Progression

factor 1 (Arf1) with potential for cancer therapy. J Biol Chem 2011; 62. Schwarzenbach H, Hoon DSB, Pantel K. Cell-free nucleic acids as bio- 287:3885–97. markers in cancer patients. Nat Rev Cancer 2011;11:426–37. 38. Tseng CN, Hong YR, Chang HW, Yu TJ, Hung TW, Hou MF, et al. Brefeldin 63. Meyer M, Reimand J, Lan X, Head R, Zhu X, Kushida M, et al. Single cell- A reduces anchorage-independent survival, cancer stem cell potential and derived clonal analysis of human glioblastoma links functional and migration of MDA-MB-231 human breast cancer cells. Molecules 2014; genomic heterogeneity. Proc Natl Acad Sci U S A 2015;112:851–6. 19:17464–77. 64. Viotti C. ER to Golgi-dependent protein secretion: the conventional 39. Tseng CN, Huang CF, Cho CL, Chang HW, Chiu CC, Chang YF. Brefeldin a pathway. Unconventional protein secretion: methods and protocols; effectively inhibits cancer stem cell-like properties and MMP-9 activity in 2016. Springer. p. 3–29. human colorectal cancer Colo 205 cells. Molecules 2013;18:10242–53. 65. Marchesin V, Castro-Castro A, Lodillinsky C, Castagnino A, Cyrta J, 40. Wang H, Zhao Y, Zhang C, Li M, Jiang C, Li Y. Rab27a was identified as a Bonsang-Kitzis H, et al. ARF6–JIP3/4 regulate endosomal tubules prognostic biomaker by mRNA profiling, correlated with malignant for MT1-MMP exocytosis in cancer invasion. J Cell Biol 2015;211: progression and subtype preference in gliomas. PLoS One 2014;9:e89782. 339–58. 41. Hampsch RA, Shee K, Bates D, Lewis LD, Desire L, Leblond B, et al. 66. Muralidharan-Chari V, Clancy J, Plou C, Romao M, Chavrier P, Raposo G, Therapeutic sensitivity to Rac GTPase inhibition requires consequential et al. ARF6-regulated shedding of tumor cell-derived plasma membrane suppression of mTORC1, AKT, and MEK signaling in breast cancer. microvesicles. Curr Biol 2009;19:1875–85. Oncotarget 2017;8:21806–17. 67. Qin X, Wang J, Wang X, Liu F, Jiang B, Zhang Y. Targeting Rabs as a 42. Nie Z, Randazzo PA. Arf GAPs and membrane traffic. J Cell Sci 2006; novel therapeutic strategy for cancertherapy.DrugDiscovToday2017; 119:1203–11. 22:1139–47. 43. Almiron Bonnin DA, Havrda MC, Lee MC, Liu H, Zhang Z, Nguyen LN, 68. Cheng KW, Lahad JP, Gray JW, Mills GB. Emerging Role of RAB GTPases in et al. Secretion-mediated STAT3 activation promotes self-renewal of cancer and human disease. Cancer Res 2005;65:2516–9. glioma stem-like cells during hypoxia. Oncogene 2017;37:1107–18. 69. Geppert M, Bolshakov VY, Siegelbaum SA, Takei K, De Camilli P, Hammer 44. Cross BCS, McKibbin C, Callan AC, Roboti P, Piacenti M, Rabu C, et al. RE, et al. The role of Rab3A in neurotransmitter release. Nature Eeyarestatin I inhibits Sec61-mediated protein translocation at the endo- 1994;369:493–7. plasmic reticulum. J Cell Sci 2009;122:4393–400. 70. Wheeler DB, Zoncu R, Root DE, Sabatini DM, Sawyers CL. Identification 45. Prabhu A, Sarcar B, Kahali S, Shan Y, Chinnaiyan P. Targeting the unfolded of an oncogenic RAB protein. Science 2015;350:211–7. protein response in glioblastoma cells with the fusion protein EGF-SubA. 71. Yoo JH, Shi DS, Grossmann AH, Sorensen LK, Tong Z, Mleynek TM, et al. PLoS One 2012;7:e52265. ARF6 is an actionable node that orchestrates oncogenic GNAQ signaling 46. Valle CW, Min T, Bodas M, Mazur S, Begum S, Tang D, et al. Critical Role of in uveal melanoma. Cancer Cell 2016;29:889–904. VCP/p97 in the pathogenesis and progression of non-small cell lung 72. Rabouille C. Pathways of unconventional protein secretion. Trends Cell carcinoma. PLoS One 2011;6:e29073. Biol 2016;27:230–40. 47. Dubost JJ, Rolhion C, Tchirkov A, Bertrand S, Chassagne J, Dosgilbert A, 73. Senft D, Ronai ZeA. Adaptive stress responses during tumor metastasis et al. Interleukin-6-producing cells in a human glioblastoma cell line are and dormancy. Trends Cancer 2016;2:429–42. not affected by ionizing radiation. J Neurooncol 2002;56:29–34. 74. DingJ,WangK,LiuW,SheY,SunQ,ShiJ,etal.Pore-formingactivity 48. Hardee ME, Marciscano AE, Medina-Ramirez CM, Zagzag D, Narayana A, and structural autoinhibition of the gasdermin family. Nature 2016; Lonning SM, et al. Resistance of glioblastoma-initiating cells to radiation 535:111–6. mediated by the tumor microenvironment can be abolished by inhibiting 75. Ha ET, Antonios JP, Soto H, Prins RM, Yang I, Kasahara N, et al. Chronic transforming growth factor-beta. Cancer Res 2012;72:4119–29. inflammation drives glioma growth: cellular and molecular factors 49. Conze D, Weiss L, Regen PS, Bhushan A, Weaver D, Johnson P, et al. responsible for an immunosuppressive microenvironment. Neuroimmu- Autocrine production of interleukin 6 causes multidrug resistance in nology Neuroinflammation 2014;1:66–76. breast cancer cells. Cancer Res 2001;61:8851–8. 76. Tarassishin L, Casper D, Lee SC. Aberrant expression of interleukin-1b and 50. Liu C, Zhu Y, Lou W, Cui Y, Evans CP, Gao AC. Inhibition of constitutively inflammasome activation in human malignant gliomas. PLoS One active Stat3 reverses enzalutamide resistance in LNCaP derivative prostate 2014;9:e103432. cancer cells. Prostate 2014;74:201–9. 77. Hergueta-Redondo M, Sarrio D, Molina-Crespo A, Megias D, Mota A, 51. Obenauf AC, Zou Y, Ji AL, Vanharanta S, Shu W, Shi H, et al. Therapy- Rojo-Sebastian A, et al. Gasdermin-B promotes invasion and metastasis in induced tumour secretomes promote resistance and tumour progression. breast cancer cells. PLoS One 2014;9:e90099. Nature 2015;520:368–72. 78. Munoz JL, Rodriguez-Cruz V, Ramkissoon SH, Ligon KL, Greco SJ, 52. Batlle E, Clevers H. Cancer stem cells revisited. Nat Med 2017;23:1124–34. Rameshwar P. Temozolomide resistance in glioblastoma occurs by 53. Verhaak RGW, Hoadley KA, Purdom E, Wang V, Qi Y, Wilkerson MD, et al. miRNA-9-targeted PTCH1, independent of sonic hedgehog level. Onco- An integrated genomic analysis identifies clinically relevant subtypes of target 2015;6:1190–201. glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR 79. Jawhari S, Ratinaud MH, Verdier M. Glioblastoma, hypoxia and autop- and NF1. Cancer Cell 2010;17:98–110. hagy: a survival-prone 'menage-a-trois'. Cell Death Dis 2016;7:e2434. 54. Almiron Bonnin DA, Ran C, Havrda MC, Liu H, Hitoshi Y, Zhang Z, et al. 80. Gammoh N, Fraser J, Puente C, Syred HM, Kang H, Ozawa T, et al. Insulin-mediated signaling facilitates resistance to PDGFR inhibition in Suppression of autophagy impedes glioblastoma development and proneural hPDGFB-driven gliomas. Mol Cancer Ther 2017;16:705–16. induces senescence. Autophagy 2016;12:1431–9. 55. Kohler N, Lipton A. Platelets as a source of fibroblast growth-promoting 81. Gee Heon Y, Noh Shin H, Tang Bor L, Kim Kyung H, Lee Min G. Rescue of activity. Exp Cell Res 1974;87:297–301. DF508-CFTR trafficking via a GRASP-dependent unconventional secre- 56. Lemmon MA, Schlessinger J. Cell signaling by receptor tyrosine kinases. tion pathway. Cell 2011;146:746–60. Cell 2010;141:1117–34. 82. Drogat B, Auguste P, Nguyen DT, Bouchecareilh M, Pineau R, Nalban- 57. DeBerardinis RJ, Chandel NS. Fundamentals of cancer metabolism. toglu J, et al. IRE1 signaling is essential for ischemia-induced vascular Science Advances 2016;2:e1600200. endothelial growth factor-A expression and contributes to angiogenesis 58. Huang J, Weinstein SJ, Kitahara CM, Karoly ED, Sampson JN, Albanes D. A and tumor growth in vivo. Cancer Res 2007;67:6700–7. prospective study of serum metabolites and glioma risk. Oncotarget 83. Rajesh Y, Biswas A, Mandal M. Glioma progression through the prism of 2017;8:70366–77. heat shock protein mediated extracellular matrix remodeling and epithe- 59. Maus A, Peters GJ. Glutamate and a-ketoglutarate: key players in glioma lial to mesenchymal transition. Exp Cell Res 2017;359:299–311. metabolism. Amino Acids 2017;49:21–32. 84. Gould SJ, Raposo G. As we wait: coping with an imperfect nomenclature 60. Bavoux C, Leopoldino AM, Bergoglio V, O-Wang J, Ogi T, Bieth A, et al. Up- for extracellular vesicles. J Extracell Vesicles 2013;2:20389. regulation of the error-prone DNA polymerase {kappa} promotes pleio- 85. Meehan B, Rak J, Di Vizio D. Oncosomes – large and small: what are they, tropic genetic alterations and tumorigenesis. Cancer Res 2005;65:325–30. where they came from? J Extracell Vesicles 2016;5:33109. 61. Soeda A, Hara A, Kunisada T, Yoshimura S-i, Iwama T, Park DM. The 86. Lotvall€ J, Hill AF, Hochberg F, Buzas EI, Di Vizio D, Gardiner C, et al. evidence of glioblastoma heterogeneity. Sci Rep 2015;5:7979. Minimal experimental requirements for definition of extracellular vesicles

www.aacrjournals.org Cancer Res; 78(21) November 1, 2018 6037

Downloaded from cancerres.aacrjournals.org on October 6, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst October 17, 2018; DOI: 10.1158/0008-5472.CAN-18-0345

Almiron Bonnin et al.

and their functions: a position statement from the International Society 108. Serrill JD, Wan X, Hau AM, Jang HS, Coleman DJ, Indra AK, et al. for Extracellular Vesicles. J Extracell Vesicles 2014;3:26913. Coibamide A, a natural lariat depsipeptide, inhibits VEGFA/VEGFR2 87. Witwer KW, Soekmadji C, Hill AF, Wauben MH, Buzas EI, Di Vizio D, et al. expression and suppresses tumor growth in glioblastoma xenografts. Updating the MISEV minimal requirements for extracellular vesicle Invest New Drugs 2016;34:24–40. studies: building bridges to reproducibility. J Extracell Vesicles 2017;6: 109. Junne T, Wong J, Studer C, Aust T, Bauer BW, Beibel M, et al. Decatransin, a 1396823. new natural product inhibiting protein translocation at the Sec61/SecYEG 88. Antonyak MA, Li B, Boroughs LK, Johnson JL, Druso JE, Bryant KL, et al. translocon. J Cell Biol 2015;128:1217–29. Cancer cell-derived microvesicles induce transformation by transferring 110. McKenna M, Simmonds RE, High S. Mechanistic insights into the inhi- tissue transglutaminase and fibronectin to recipient cells. Proc Natl Acad bition of Sec61-dependent co- and post-translational translocation by Sci U S A 2011;108:4852–7. mycolactone. J Cell Biol 2016;129:1404–15. 89. Brem GJ, Mylonas I, Bruning A. Eeyarestatin causes cervical cancer cell 111. ClinicalTrials.gov. Chemopreventive effects of epigallocatechin gallate sensitization to bortezomib treatment by augmenting ER stress and (EGCG) in colorectal cancer (CRC) patients. Available from: https:// CHOP expression. Gynecol Oncol 2013;128:383–90. clinicaltrials.gov/ct2/show/NCT02891538. 90. PyrkoP,SchonthalAH,HofmanFM,ChenTC,LeeAS.Theunfolded 112. Keith F, John C. Targeted secretion inhibitors—innovative protein protein response regulator GRP78/BiP as a novel target for increasing therapeutics. Toxins 2010;2:2795–815. chemosensitivity in malignant gliomas. Cancer Res 2007;67: 113. Foster KA. Harnessing toxins. Manuf Chem 2006:23–6. 9809–16. 114. Hanahan D, Weinberg Robert A. Hallmarks of cancer: the next generation. 91. Shutes A, Onesto C, Picard V, Leblond B, Schweighoffer F, Der CJ. Cell 2011;144:646–74. Specificity and Mechanism of Action of EHT 1864, a novel small 115. Akhavan D, Pourzia AL, Nourian AA, Williams KJ, Nathanson D, Babic I, molecule inhibitor of family small GTPases. J Biol Chem 2007; et al. De-repression of PDGFRb transcription promotes acquired resis- 282:35666–78. tance to EGFR tyrosine kinase inhibitors in glioblastoma patients. Cancer 92. Akbar H, Kim J, Funk K, Cancelas JA, Shang X, Chen L, et al. Genetic and Discov 2013;3:534–47. pharmacologic evidence that Rac1 GTPase is involved in regulation of 116. Nister M, Heldin CH, Wasteson Å, Westermark B. A platelet-derived platelet secretion and aggregation. J Thromb Haemost 2007;5:1747–55. growth factor analog produced by a human clonal glioma cell line. Ann 93. Li Q, Ho CS, Marinescu V, Bhatti H, Bokoch GM, Ernst SA, et al. N Y Acad Sci 1982;397:25–33. Facilitation of Ca(2þ)-dependent exocytosis by Rac1-GTPase in bovine 117. Chattopadhyay N, Tfelt-Hansen J, Brown EM. PKC, p42/44 MAPK and chromaffin cells. J Physiol 2003;550:431–45. p38 MAPK regulate hepatocyte growth factor secretion from human 94. Ivanov AI.Pharmacological inhibitors of exocytosis and endocytosis: astrocytoma cells. Brain Res Mol Brain Res 2002;102:73–82. novel bullets for old targets. In:Ivanov AI, editor. Exocytosis and endo- 118. Ambrose D, Resnicoff M, Coppola D, Sell C, Miura M, Jameson S, cytosis. New York, NY: Springer; 2014. p. 3–18. et al. Growth regulation of human glioblastoma t98g cells by 95. Viaud J, Zeghouf M, Barelli H, Zeeh JC, Padilla A, Guibert B, et al. insulinlike growth factor1 and its receptor. J Cell Physiol 1994; Structure-based discovery of an inhibitor of Arf activation by Sec7 159:92–100. domains through targeting of protein-protein complexes. Proc Natl Acad 119. Tang P, Steck PA, Yung WKA. The autocrine loop of TGF-a/EGFR and Sci U S A 2007;104:10370–5. brain tumors. J Neurooncol 1997;35:303–14. 96. Saenz JB, Sun WJ, Chang JW, Li J, Bursulaya B, Gray NS, et al. Golgicide A 120. Ouafik L, Sauze S, Boudouresque F, Chinot O, Delfino C, Fina F, et al. reveals essential roles for GBF1 in Golgi assembly and function. Nat Chem Neutralization of adrenomedullin inhibits the growth of human glio- Biol 2009;5:157–65. blastoma cell lines in vitro and suppresses tumor xenograft growth in vivo 97. Lang L, Shay C, Zhao X, Teng Y. Combined targeting of Arf1 and Ras 2002;160:1279–92. potentiates anticancer activity for prostate cancer therapeutics. J Exp Clin 121. Al-Nedawi K, Meehan B, Micallef J, Lhotak V, May L, Guha A, et al. Cancer Res 2017;36:112. Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles 98. Xie X, Tang SC, Cai Y, Pi W, Deng L, Wu G, et al. Suppression of breast derived from tumour cells2008;10:619–24. cancer metastasis through the inactivation of ADP-ribosylation factor 1. 122. Marfia G, Campanella R, Navone SE, Di Vito C, Riccitelli E, Hadi LA, et al. Oncotarget 2016;7:58111–20. Autocrine/paracrine sphingosine-1-phosphate fuels proliferative and 99. Luchsinger C, Aguilar M, Burgos PV, Ehrenfeld P, Mardones GA. Functional stemness qualities of glioblastoma stem cells. Glia 2014;62:1968–81. disruption of the Golgi apparatus protein ARF1 sensitizes MDA-MB-231 123. Shea A, Harish V, Afzal Z, Chijioke J, Kedir H, Dusmatova S, et al. breast cancer cells to the antitumor drugs Actinomycin D and Vinblastine MicroRNAs in glioblastoma multiforme pathogenesis and therapeutics. through ERK and AKT signaling. PLoS One 2018;13:e0195401. Cancer Med 2016;5:1917–46. 100. Feng Y, Yu S, Lasell TKR, Jadhav AP, Macia E, Chardin P, et al. Exo1: a new 124. Wei Z, Batagov AO, Schinelli S, Wang J, Wang Y, El Fatimy R, et al. Coding chemical inhibitor of the exocytic pathway. Proc Natl Acad Sci U S A and noncoding landscape of extracellular RNA released by human glioma 2003;100:6469–74. stem cells. Nat Commun 2017;8:1145. 101. Chambers K, Judson B, Brown WJ. A unique lysophospholipid acyltrans- 125. WrannM,BodmerS,SieplC,Hofer-WarbinekR,FreiK,HoferE,etal. T ferase (LPAT) antagonist, CI-976, affects secretory and endocytic mem- cell suppressor factor from human glioblastoma cells is a 12.5-kd brane trafficking pathways. J Cell Biol 2005;118:3061–71. protein closely related to transforming growth factor-beta. EMBO J 102. Lu L, Hannoush RN, Goess BC, Varadarajan S, Shair MD, Kirchhausen T, 1987;6:1633–6. et al. The small molecule dispergo tubulates the endoplasmic reticulum 126. Hishii M, Nitta T, Ishida H, Ebato M, Kurosu A, Yagita H, et al. Human and inhibits export. Mol Biol Cell 2013;24:1020–9. glioma-derived interleukin-10 inhibits antitumor immune responses in 103. Yonemura Y, Li X, Muller€ K, Kr€amer A, Atigbire P, Mentrup T, et al. vitro. Neurosurgery 1995;37:1160–7. Inhibition of cargo export at ER exit sites and the trans-Golgi network by 127. Crane CA, Austgen K, Haberthur K, Hofmann C, Moyes KW, Avanesyan the secretion inhibitor FLI-06. J Cell Sci 2016;129:3868–77. L, et al. Immune evasion mediated by tumor-derived lactate dehydro- 104. Liu Y, Law BK, Luesch H. Apratoxin a reversibly inhibits the secretory genase induction of NKG2D ligands on myeloid cells in glioblastoma pathway by preventing cotranslational translocation. Mol Pharmacol patients. Proc Natl Acad Sci U S A 2014;111:12823–8. 2009;76:91–104. 128. Ellert-Miklaszewska A, Wisniewski P, Kijewska M, Gajdanowicz P, 105. Garrison JL, Kunkel EJ, Hegde RS, Taunton J. A substrate-specific inhibitor Pszczolkowska D, Przanowski P, et al. Tumour-processed osteopontin of protein translocation into the endoplasmic reticulum. Nature and lactadherin drive the protumorigenic reprogramming of microglia 2005;436:285–9. and glioma progression. Oncogene 2016;35:6366–77. 106. Cai W, Chen QY, Dang LH, Luesch H. Apratoxin S10, a dual inhibitor of 129. Van Meir E, Sawamura Y, Diserens AC, Hamou MF, de Tribolet N. Human angiogenesis and cancer cell growth to treat highly vascularized tumors. glioblastoma cells release interleukin 6 in vivo and in vitro. Cancer Res ACS Med Chem Lett 2017;8:1007–12. 1990;50:6683–8. 107. Huang K-C, Chen Z, Jiang Y, Akare S, Kolber-Simonds D, Condon K, et al. 130. Infanger DW, Cho Y, Lopez BS, Mohanan S, Liu SC, Gursel D, et al. Apratoxin a shows novel pancreas-targeting activity through the binding Glioblastoma stem cells are regulated by interleukin-8 signaling in a of sec 61. Mol Cancer Ther 2016;15:1208–16. tumoral perivascular niche. Cancer Res 2013;73:7079–89.

6038 Cancer Res; 78(21) November 1, 2018 Cancer Research

Downloaded from cancerres.aacrjournals.org on October 6, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst October 17, 2018; DOI: 10.1158/0008-5472.CAN-18-0345

Glioma Secretion Drives Tumor Progression

131. Wang S-C, Hong J-H, Hsueh C, Chiang C-S. Tumor-secreted SDF-1 144. Rooprai HK, Rucklidge GJ, Panou C, Pilkington GJ. The effects of promotes glioma invasiveness and TAM tropism toward hypoxia in a exogenous growth factors on matrix metalloproteinase secretion by murine astrocytoma model. Lab Invest 2011;92:151–62. human brain tumour cells. Br J Cancer 1999;82:52–5. 132. Ho SY, Ling TY, Lin HY, Liou JT, Liu FC, Chen IC, et al. SDF-1/CXCR4 145. Hoelzinger DB, Nakada M, Demuth T, Rosenstee T, Reavie LB, Berens ME. signaling maintains stemness signature in mouse neural stem/progenitor Autotaxin: a secreted autocrine/paracrine factor that promotes glioma cells. Stem Cells Int 2017;2017:2493752. invasion. J Neurooncol 2008;86:297–309. 133. Yan K, Wu Q, Yan DH, Lee CH, Rahim N, Tritschler I, et al. Glioma cancer 146. Lyons SA, Chung WJ, Weaver AK, Ogunrinu T, Sontheimer H. Autocrine stem cells secrete Gremlin1 to promote their maintenance within the glutamate signaling promotes glioma cell invasion. Cancer Res 2007; tumor hierarchy. Genes Dev 2014;28:1085–100. 67:9463–71. 134. Man J, Shoemake J, Zhou W, Fang X, Wu Q, Rizzo A, et al. Sema3C 147. Hu F, a Dzaye OD, Hahn A, Yu Y, Scavetta RJ, Dittmar G, et al. Glioma- Promotes the survival and tumorigenicity of glioma stem cells through derived versican promotes tumor expansion via glioma-associated micro- activation. Cell Rep 2014;9:1812–26. glial/macrophages Toll-like receptor 2 signaling. Neuro-oncol 2015;17: 135. Zhou W, Ke SQ, Huang Z, Flavahan W, Fang X, Paul J, et al. Periostin 200–10. secreted by glioblastoma stem cells recruits M2 tumour-associated 148. Kawataki T, Yamane T, Naganuma H, Rousselle P, Anduren I, Tryggvason macrophages and promotes malignant growth. Nat Cell Biol 2015; K, et al. Laminin isoforms and their integrin receptors in glioma cell 17:170–82. migration and invasiveness: Evidence for a role of alpha5-laminin(s) and 136. Liu AY, Zheng H, Ouyang G. Periostin, a multifunctional matricellular alpha3beta1 integrin. Exp Cell Res 2007;313:3819–31. protein in inflammatory and tumor microenvironments. Matrix Biol 149. Gutschner T, Diederichs S. The hallmarks of cancer: a long non-coding 2014;37:150–6. RNA point of view. RNA Biol 2012;9:703–19. 137. Schonberg DL, Miller TE, Wu Q, Flavahan WA, Das NK, Hale JS, et al. 150. Dunn IF, Heese O, Black PM. Growth factors in glioma angiogenesis: Preferential iron trafficking characterizes glioblastoma stem-like cells. FGFs, PDGF, EGF, and TGFs. J Neurosurg 2000;50:121–37. Cancer Cell 2015;28:441–55. 151. Aghi M, Cohen KS, Klein RJ, Scadden DT, Chiocca EA. Tumor Stromal- 138. Hwang J-S, Jung E-H, Kwon M-Y, Han I-O. Glioma-secreted soluble Derived factor-1 recruits vascular progenitors to mitotic neovasculature, factors stimulate microglial activation: The role of interleukin-1b and where microenvironment influences their differentiated phenotypes. tumor necrosis factor-a. J Neuroimmunol 2016;298:165–71. Cancer Res 2006;66:9054–64. 139. Dwyer J, Hebda JK, Le Guelte A, Galan-Moya E-M, Smith SS, Azzi S, et al. 152. Miyake M, Goodison S, Lawton A, Gomes-Giacoia E, Rosser CJ. Angio- Glioblastoma cell-secreted interleukin-8 induces brain endothelial cell genin promotes tumoral growth and angiogenesis by regulating matrix permeability via CXCR2. PLoS One 2012;7:e45562. metallopeptidase-2 expression via the ERK1/2 pathway. Oncogene 2015; 140. Ye ZC, Sontheimer H. Glioma cells release excitotoxic concentrations of 34:890–901. glutamate. Cancer Res 1999;59:4383–91. 153. Campbell IM, Gambin T, Dittwald P, Beck CR, Shuvarikov A, Hixson P, 141. Song H, Moon A. Glial cell-derived neurotrophic factor (GDNF) pro- et al. Human endogenous retroviral elements promote genome insta- motes low-grade Hs683 glioma cell migration through JNK, ERK-1/2 and bility via non-allelic homologous recombination. BMC Biol 2014; p38 MAPK signaling pathways. Neurosci Res 2006;56:29–38. 12:74. 142. Amos S, Redpath GT, diPierro CG, Carpenter JE, Hussaini IM. Epi- 154. Kines KJ, Sokolowski M, deHaro DL, Christian CM, Belancio VP. Potential dermal growth factor receptor-mediated regulation of urokinase for genomic instability associated with retrotranspositionally-incompe- plasminogen activator expression and glioblastoma invasion via C- tent L1 loci. Nucleic Acids Res 2014;42:10488–502. SRC/MAPK/AP-1 signaling pathways. J Neuropathol Exp Neurol 155. Fazza AC, Sabino FC, de Setta N, Bordin NA, da Silva EHT, Carareto CMA. 2010;69:582–92. Estimating genomic instability mediated by Alu retroelements in breast 143. Lagriffoul A, Charpentier N, Carrette J, Tougard C, Bockaert J, Homburger cancer. Genet Mol Biol 2009;32:25–31. V. Secretion of protease nexin-1 by C6 glioma cells is under the control of a 156. Soga T.Cancer metabolism: key players in metabolic reprogramming. heterotrimeric , Go1. J Biol Chem 1996;271:31508–16. Cancer Sci 2013;104:275–81.

www.aacrjournals.org Cancer Res; 78(21) November 1, 2018 6039

Downloaded from cancerres.aacrjournals.org on October 6, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst October 17, 2018; DOI: 10.1158/0008-5472.CAN-18-0345

Glioma Cell Secretion: A Driver of Tumor Progression and a Potential Therapeutic Target

Damian A. Almiron Bonnin, Matthew C. Havrda and Mark A. Israel

Cancer Res 2018;78:6031-6039. Published OnlineFirst October 17, 2018.

Updated version Access the most recent version of this article at: doi:10.1158/0008-5472.CAN-18-0345

Cited articles This article cites 150 articles, 40 of which you can access for free at: http://cancerres.aacrjournals.org/content/78/21/6031.full#ref-list-1

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at Subscriptions [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerres.aacrjournals.org/content/78/21/6031. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on October 6, 2021. © 2018 American Association for Cancer Research.