<<

Published OnlineFirst February 24, 2020; DOI: 10.1158/0008-5472.CAN-19-3652

CANCER RESEARCH | REVIEW

Targeting the SAGA and ATAC Transcriptional Complexes in -Driven Lisa Maria Mustachio1,2, Jason Roszik3,4, Aimee Farria1,2,5, and Sharon Y.R. Dent1,2,5

ABSTRACT ◥ Targeting epigenetic regulators, such as -modifying disruption of MYC-driven functions. GCN5 is part of two , provides novel strategies for therapy. The GCN5 distinct multiprotein histone-modifying complexes, SAGA and acetyltransferase (KAT) functions together with MYC both ATAC. This review summarizes key findings on the roles of during normal development and in oncogenesis. As SAGA and ATAC in embryo development and in cancer to better factors, MYC family members are difficult to target with small- understand the functional relationships of these complexes with molecule inhibitors, but the acetyltransferase domain and the MYC family members, as well as their future potential as in GCN5 might provide alternative targets for therapeutic targets.

Introduction with sequence-specific transcription factors, and may be stabilized at certain regions through interactions of “reader” domains with GCN5 was defined in the late 1990s as a component of the specifichistonemodifications. Some studies indicate that SAGA Spt-Ada-Gcn5-Acetyl transferase (SAGA) complex in (i) yeast and acts at select , whereas others indicate that it functions as a (ii) related TFTC/STAGA complexes in (1, 2). These general (9–11). Recent studies may resolve multiprotein assemblies posttranslationally modify as these differences, as chromatin immunoprecipitation experiments well as other proteins (1). SAGA is composed of lysine acetyltrans- indicate that the SAGA subunit SUS1 is recruited to yeast genes ferase (KAT), deubiquitinase (DUB), SPT, and TAF modules. These regulated by the general transcription factor IID (TFIID), as well as modules maintain SAGA architecture (SPT), regulate histone acet- to known SAGA-regulated genes and ribosomal protein ylation and deubiquitination (KAT and DUB), and interact with genes (12, 13). However, under conditions of heat shock, SUS1 general transcriptional machinery (TAF), as well as specifictran- binding shifts away from SAGA-dominated and ribosomal protein scription factors (TRRAP; TF binding module; ref. 3). GCN5 genes to stress-responsive genes (12), and SAGA colocalizes with (KAT2A) was later identified as a component of the ADA Two heat shock factor 1 (Hsf1) at these genes (14). These findings A Containing (ATAC) complex, which also regulates tran- indicate that SAGA distributions across the genome can be scription and chromatin organization (4). GCN5 KAT activity dynamic. and specificity is augmented by association with the alteration/ SAGA regulates both transcription initiation and elongation deficiency in activation (ADA) proteins (1, 5), and independent through its KAT and DUB activities. SAGA also has nonhistone ADA–GCN5 complexes have been identified in yeast and metazo- substrates, such as cMYC, which is acetylated by GCN5 and its ans(6).GCN5requiresassociationwithbothADA2andADA3 ortholog PCAF (15) and TRF1, which is deubiquitylated by for robust of histone and nonhistone targets (7). GCN5 USP22 (16). As such, SAGA influences at many levels, is associated with two forms of ADA2 in metazoans, termed from transcription to protein stability (17). GCN5 functions extend ADA2A and ADA2B (8). ADA2B is exclusively found in SAGA, beyond the nucleus, independent of the SAGA and ATAC complexes, while ADA2A is found in ATAC (8). Outside of the KAT modules, including mitochondrial functions in S. cerevisiae (18). Genetic studies SAGA and ATAC are distinct in composition, likely reflecting in mice indicate GCN5 (19, 20) and USP22 (21) are required for unique functions (3, 4). normal embryo development. SAGA also impacts signaling pathways, Both SAGA and ATAC function as transcriptional coactivators. genome integrity, and metabolic control in mammalian cells (17). These complexes are targeted to genomic loci through interactions Although less studied, components of ATAC are also required for normal embryo development, modulating ribosome biogenesis, cell cycle, and DNA repair (22). This review will further explore the 1 Departments of Epigenetics and Molecular Carcinogenesis, The University of functions of SAGA and ATAC in development and in cancer, espe- 2 Texas MD Anderson Cancer Center, Houston, Texas. Center for Cancer Epige- cially in regards to MYC functions. netics, The University of Texas MD Anderson Cancer Center, Houston, Texas. 3Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. 4Genomic Medicine, The University of Texas MD Links between SAGA and MYC Family 5 Anderson Cancer Center, Houston, Texas. Graduate School of Biomedical fi fi Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas. GCN5 was the rst identi ed transcription-related KAT (23), and it acetylates lysine residues in histones H3 and H2B (17). Corresponding Author: Sharon Y.R. Dent, University of Texas MD Anderson Mammals also express PCAF (P300/CBP-associated factor, or Cancer Center, 1808 Park Road 1C, Smithville, TX 78957. Phone: 512-237-2403; Fax: 512-237-3439; E-mail: [email protected] KAT2B), which is highly homologous to GCN5 (24). GCN5 and PCAF are incorporated into SAGA and ATAC complexes in Cancer Res 2020;XX:XX–XX a mutually exclusive way. In contrast to Gcn5, no abnormal doi: 10.1158/0008-5472.CAN-19-3652 phenotypes are associated with Pcaf deletion in mice (19, 20). 2020 American Association for Cancer Research. Interestingly, combined knockout of Gcn5 and Pcaf results in more

AACRJournals.org | OF1

Downloaded from cancerres.aacrjournals.org on October 4, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst February 24, 2020; DOI: 10.1158/0008-5472.CAN-19-3652

Mustachio et al.

severe developmental phenotypes than Gcn5 deletion alone (19), SAGA in Cancer suggesting both redundant and synergistic roles for these two GCN5 likely plays a role in the oncogenic functions of MYC, KATs. although GCN5 and other components of SAGA may contribute to Conditional deletion of Gcn5 in neural progenitor cell popula- cancer formation or progression independently of MYC as well. GCN5 tions reduces brain mass in mice, with a phenotype similar to overexpression is observed in multiple cancer types (Fig. 1A), and conditional loss of cMyc or N-myc (25, 26). Interestingly, MYC GCN5 depletion inhibits cell proliferation or induces , regulates chromatin structures in neural progenitors at least, in part, consistent with a role in oncogenesis. For example, inhibition of GCN5 through upregulation of GCN5 (26). Regulatory connections activity reduces viability of lung cancer stem–like cells and reduces between GCN5 and MYC family members have been observed in non–small cell lung cancer growth (34, 35). GCN5 has also been linked numerous settings. The TRRAP protein in SAGA directly interacts to regulation of cell proliferation and invasion in glioma (36). In breast with MYC in cancer cells, recruiting GCN5 for activation of MYC cancer, GCN5 regulates the epithelial-to-mesenchymal transition by target genes (27–29). Moreover, acetylation of MYC by GCN5 or enhancing STAT3, AKT, and E2F1 signaling pathways (37). GCN5 PCAF extends its half-life in vivo, augmenting MYC functions overexpression in hepatocellular carcinoma is linked to increased (15). In embryonic stem cells, GCN5 (30) and other SAGA com- AIB1 expression, which drives progression of these cancers (38). In ponents (31) colocalize with MYC for upregulation of cell-cycle cells, AND-1, a protein required for the stability of GCN5, is genes, promoting embryonic stem cell self-renewal. Moreover, upregulated (39). A CRISPR screen for factors required for AML functions of MYC as a “Yamanaka factor” in somatic cell repro- identified GCN5 (KAT2A) as a “top-hit” (40). In lymphoma cells, gramming involve upregulation of GCN5 expression followed by GCN5 inhibition reduces cell survival through reduction of MYC recruitment of GCN5 protein to MYC target genes (30). Loss of target gene expression (41). Many of the cancer types that overexpress Gcn5 also hinders transcription of cMYC target genes associated GCN5 also harbor MYC amplifications or increased expression of with FGF signaling in embryoid bodies and early differentia- MYC family proteins. tion (32). Altogether, these studies indicate that GCN5 and MYC Although highly related to GCN5, PCAF may play different roles work together in a feed forward loop. These functions are likely to in cancer, acting as either an oncogene or tumor suppressor based involve SAGA, as the TAD domain of MYC interacts with SAGA on the tissue of origin. In hepatocellular carcinoma, PCAF pro- but not ATAC components (33), and MYC upregulates expression motes autophagy (42). PCAF acts as a suppressor of gastric cancer of several components of SAGA in addition to GCN5 (30).

AC Dependency score (YEATS2) Dependency score (ATAC2)

Fibroblast Fibroblast Eye Cervix Mesothelioma Eye Bone Mesothelioma Bile duct Urinary tract Ovary Bile duct Urinary tract Bone Prostate Breast Rhabdoid Thyroid Prostate Kidney chromophobe Kidney Normal thyroid adenocarcinoma Sarcoma endometrial carcinoma Uterine Normal esophagus carcinosarcoma Uterine Liver cancer Liver glioma grade Lower Normal cervix Breast carcinoma Adrenocortical cancer Pancreatic Normal thymus adenocarcinoma Colon Bladder cancer Kidney clear cell carcinoma clear cell Kidney melanoma Uveal Ovarian cancer Ovarian Prostate adenocarcinoma Prostate Thyroid carcinoma Thyroid Head and neck cancer and parag. Pheochromocytoma Normal skin tumor germ cell Testicular Normal bile duct Normal kidney cancer Normal lung Normal uterus carcinoma Lung squamous cell Esophageal carcinoma Normal liver lymphoma B-cell Cholangiocarcinoma myeloid Acute Normal soft tissue Lung adenocarcinoma cancer Cervical Glioblastoma Thymoma Normal bladder Normal breast Normal pancreas papillary carcinoma Kidney Normal prostate Normal adrenal tissue Normal adrenal Skin cutaneous melanoma Mesothelioma Normal head and neck Normal stomach Gastric Central nervous system Central nervous system ** *** *** *** *** *** *** **** *** *** *** *** Liver Skin Ovary Lung Lung *** *** Liver Kidney Pancreas *** Uterus 200 Rhabdomyosarcoma Colorectal Uterus Upper aerodigestive Thyroid Upper aerodigestive 100 Rhabdomyosarcoma Leukemia Skin Multiple myeloma Gastric Peripheral nervous system 50 Soft tissue Colorectal Breast Soft tissue Peripheral nervous system Esophagus Esophagus Cervix Multiple myeloma Rhabdoid 20 Lymphoma Kidney Leukemia Lymphoma

10 –1.5 –1 –0.5 0 –1.5 –1 –0.5 0

5 KAT2A mRNA levels (TPM) mRNA levels KAT2A

2 1 USP22 GCN5 Ub BDDependency score (KAT2A) Dependency score (USP22) Ac

Prostate Prostate Ac Rhabdomyosarcoma Multiple myeloma Fibroblast Lymphoma Ub MYC Cervix Breast Peripheral nervous system Liver Esophagus Lung Ovary Pancreas Ac Colorectal Leukemia Ub Leukemia Peripheral nervous system Bone Rhabdomyosarcoma Breast Colorectal Uterus Esophagus Soft tissue Gastric Lung Bile duct GCN5 Liver Mesothelioma Urinary tract Upper aerodigestive Multiple myeloma Ovary Coactivator Gastric Urinary tract (SAGA Components) Central nervous system Uterus Skin Soft tissue Upper aerodigestive Central nervous system Bile duct Cervix Pancreas Fibroblast Mesothelioma Skin Lymphoma Eye Rhabdoid Bone MYC Target genes Eye Thyroid Thyroid Rhabdoid Kidney Kidney

–1 –0.5 0 0.5 –1 –0.5 0 0.5 1 1.5

Figure 1. Targeting SAGA and ATAC in cancer. A, GCN5 mRNA levels are significantly upregulated in most human cancers (red) relative to adjacent normal tissues (green), as indicated by analysis of The Cancer Genome Atlas. Ã, P < 0.05; ÃÃ, P < 0.01; ÃÃÃ, P < 0.001. B and C, Multiple cancer types show dependencies on GCN5 and USP22 (B)as well as YEATS2 and ATAC2 (C), as shown by DepMap, where a lower CERES score indicates that a gene is essential to a cell line, where a score of 0 indicates that the gene is not critical. Cell lines containing damaging, hotspot, or other nonconserving in YEATS2 or ATAC2 are highlighted in red, orange, and blue, respectively. D, Model summarizing how SAGA components promote MYC functions, through acting as a coactivator for MYC target genes and by increasing the stability of MYC protein through acetylation (GCN5) and deubiquitination (USP22).

OF2 Cancer Res; 2020 CANCER RESEARCH

Downloaded from cancerres.aacrjournals.org on October 4, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst February 24, 2020; DOI: 10.1158/0008-5472.CAN-19-3652

Targeting SAGA and ATAC in MYC-Driven Cancers

by inhibiting and through increased immunity (43). In Further work is needed to determine the relationship between the contrast, upon loss of function and perturbation of DNA KAT and DUB modules in cancer, and whether both enzymes might be damage response, PCAF may promote Hedgehog-dependent cell simultaneously targeted for a synergistic response. Loss of GCN5 survival and proliferation (44, 45). Double knockdown of both results in the depletion of USP22 from SAGA, leading to telomere PCAF and GCN5 significantly affects cMYC levels in urothelial fusions in human and mouse cells (16). The DUB module may carcinoma cells (46). Given the differential, but overlapping, expres- stimulate the KAT activity in SAGA (62) and USP22 is acetylated at sion patterns of GCN5 and PCAF in specific tissues, and their multiple (63). Cross-talk between these enzymatic modules is different functions during development, these KATS also likely have consistent with functional relationships between other DUBs and shared and unique roles in differentcancersthatareimportantto KATs in oncogenesis (64). define for therapy development.

ADA proteins Overview of ATAC and Its Role in Functions of the ADA proteins (ADA2 A and B isoforms and Cancer fi ADA3) in cancer have not yet been de ned, but they are likely to be The YEATS2 component of ATAC is emerging as potentially important as these proteins are critical regulators of GCN5 cataly- important in cancer. YEATS2 contains a histone fold domain as fi – sis (6, 17). A recent study to de ne gene dependencies in N-MYC well as a YEATS domain, which selectively binds to (“reads”) acetyl- fi driven neuroblastomas identi ed ADA2B and several other SAGA lysine residues in histones and likely other nonhistone proteins (65). components as top hits (47). Importantly, loss of ADA2A or ADA2B In particular, YEATS2 binds to acetylated H3K27, recruiting the inactivates both GCN5- and PCAF-containing versions of ATAC and ATAC complex to chromatin (66). This recruitment allows for the fi SAGA, respectively, providing an ef cient way to eliminate redundant maintenanceofH3K9acandH3K14acfacilitatinggenetranscrip- functions of these KATs in cancer cells. tion. YEATS2 is amplified in lung, ovarian, cervical, and uterine cancers and is necessary for survival of non–small cell lung cancer USP22 cells (66). Whether ATAC plays a role in MYC functions is fi USP22 is a member of the -speci c processing currently unknown. YEATS2 physically interacts with MYC, but (USP) family, and it targets both histone and nonhistone substrates. at much lower levels than SAGA components (33). Cancers, such as USP22 is particularly known for targeting monoubiquitinated H2B to lung, liver, breast, and prostate, are dependent on YEATS2 and facilitate gene regulation (45, 48, 49). USP22 has been described as a ATAC2, as shown by DepMap analyses (Fig. 1C). The YEATS “ ” member of an 11-gene death from cancer signature (50) and like domain and the ATAC2 KAT domain might provide unique targets GCN5, genetic studies indicate that USP22 has important functions in for cancer therapies. both normal development and oncogenesis. Deletion of Usp22 in mice leads to vascular defects in the placental labyrinth linked to decreased activity of several signaling Distributions of the SAGA and ATAC pathways including those driven by TGFb,VEGF,andother Complexes receptor tyrosine kinases (21). Similarly, USP22 is also critical for Identification of specific oncogenic or tumor-suppressive path- angiogenesis of non–small cell lung cancer where its functions ways affected by SAGA or ATAC is needed to fully understand mirror those observed in placenta development (51). Early embry- functions of these complexes in cancers. Chromatin immunopre- onic lethality of USP22-deficient mice is also linked to increased cipitation studies in GM12878 and HeLa cell lines indicate that p53 expression (52). USP22 affects the stability and expression of SAGA and ATAC are targeted to different genomic loci by distinct nonhistone proteins such as TRF1, which impacts telomere struc- regulatory elements (67). SAGA and ATAC also are involved in ture and genome integrity, and FBP1, which regulates MYC expres- distinct signaling pathways (68). Unlike ADA2A (ATAC), ADA2B sion (16, 53). Other studies indicate USP22 is important for (SAGA) interacts with the tumor suppressor protein p53 and counteracting silencing in heterochromatin and serves as a positive p53-response genes (68). ATAC interacts with several MAPK cofactor for activation (53, 54). The SAGA DUB pathway components during osmotic stress induction and may module is also known to impact early stages of the DNA damage play a more prominent role than SAGA in this process (68). response (DDR) and is required for the DNA repair phase of class switch recombination (CSR; ref. 55). USP22 is overexpressed in highly aggressive tumors (56), and Targetable Domains within the SAGA knockdown of USP22 in cancer cells often leads to cell-cycle arrest and ATAC Complexes and decreased tumor growth (57). Here again, USP22 functions include both gene regulation, likely due to changes in H2B ubiqui- SAGA and ATAC subunits provide unique targets for potential tination, and regulation of nonhistone protein stability. USP22 therapy development. Both complexes contain multiple enzymatic deubiquitinates cMYC in breast cancer cells, increasing MYC sta- activities, the HAT and DUB modules in SAGA and the two KATs in bility (58). USP22 also regulates levels and ATAC. The acetyltransferase domains of the KATs, the reader coordinates with MYC signaling to drive prostate adenocarcino- domains in GCN5 (bromodomain) and YEATS2 (the YEATS fi ma (59). USP22 deubiquitinates and stabilizes the PU.1 transcrip- domain), and ubiquitin-speci c domain of USP22 all provide tion factor. Loss of USP22 in a mouse model of KRAS-driven potential targets for inhibitor development. leukemia exacerbated disease due to decreased PU.1 stability and subsequent blocks to myeloid differentiation associated with Targeting KAT activities increased expression of MYC target genes in progenitor cells (60). KAT activity can be repressed by directly inhibiting enzymatic Multiple cancer types are dependent on GCN5 and USP22, as shown activity or disrupting interactions between a KAT and recruitment by DepMap (61) analyses (Fig. 1B). proteins necessary for its functions. KAT inhibitors include natural

AACRJournals.org Cancer Res; 2020 OF3

Downloaded from cancerres.aacrjournals.org on October 4, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst February 24, 2020; DOI: 10.1158/0008-5472.CAN-19-3652

Mustachio et al.

products, carboxylic acids, protein–protein interaction inhibitors, through epigenetic interactions between and acet- bi-substrate inhibitors, and synthetic compounds (69). Oridonin, ylated histones (78). Although BET inhibitors have efficacy as a lysine acetyltransferase inhibitor that targets multiple KATs single agents, resistance to these is common and they may work including GCN5/PCAF, inhibits proliferation of breast cancer both best in combination with agents such as checkpoint in vitro and in vivo (70) and induces apoptosis in gastric cancer cell inhibitors (76). lines (71). In the same study, butyrolactone 3 (MB-3), a more BET domains are just one subfamily of the more than 60 differ- specific GCN5/PCAF KAT inhibitor, was also shown to inhibit ent bromodomain-containing proteins in mammals (79). The gastric cancer cell proliferation, but less potently than oridonin. bromodomains of CBP/P300 are essential for proliferation of Similarly, garcinol, a PCAF and P300 KAT probe, inhibits nonho- leukemia and lymphoma cell lines (80), suggesting inhibitors mologous end joining and radiosensitizes lung and cervical cancer targeting these may have therapeutic potential. Only a few small cells in vitro (72).PU139,aGCN5,PCAF,CREB,andP300KAT molecules targeting GCN5/PCAF bromodomains have been devel- inhibitor, triggers caspase-independent cell death in neuroblastoma oped. L-Moses was the first selective bromodomain inhibitor cells and blocks the growth of neuroblastoma xenografts in reported for GCN5/PCAF (81). L-Moses shows no cytotoxicity mice(73).Finally,theGCN5/PCAF KAT inhibitor CPTH6 affects in normal blood cells and is metabolically stable in the human growth and viability of lung cancer stem–like cells and affects the liver, suggesting it might be a good candidate for testing as an viability of other cancer cell lines (74). Key GCN5/PCAF KAT anticancer agent. The GSK4027 chemical probe is a more selective activity probes having therapeutic potential in cancer are summa- and potent GCN5/PCAF bromodomain inhibitor in vitro (82). rized in Table 1. Future work should identify whether ATAC2, the Additional studies are needed to determine the effects of L-Moses second KAT within the ATAC complex, might also be targeted for and the GSK probes on cancer cell lines and xenograft models. therapy. Development of PROTAC degraders of BET domains has proven Unfortunately, available KAT inhibitors have limited utility useful in avoiding resistance mechanisms in cancer (83). Such for clinical trials due to issues with selectivity, toxicity, and degraders targeting GCN5/PCAF bromodomains might also pro- potency (69). Not only is it difficult to develop inhibitors specific vide effective means of blocking functions of these KATs in to particular KATs, these inhibitors also affect other acetyl coen- cancer (84). zyme A (Ac-CoA)–specific functions (69). Because KATs are The discovery that YEATS domains, which are structurally bisubstrate enzymes, multiple factors influence potency of inhibi- distinct from bromodomains, also read acetyl-lysine moieties tors including the catalytic mechanism and concentration of raises the possibility that chemical probes against these domains, substrates (69). Finally, because KATs function in multiple cellular including the YEATS2 protein in ATAC, may have therapeutic processes beyond gene transcription, their inhibition may increase potential (85, 86). toxicity. All KATs have nonhistone substrates that may also be directly Targeting DUBs k involved in tumorigenesis, such as p53, Rb, and NF B (75). The SAGA functions can also be targeted through USP22 inhibition. interplay between HATs and HDACs, which have also been targeted DUB catalytic centers are structurally homologous making devel- fi for therapeutic bene t, must also be considered. Use of an HDAC opment of selective inhibitors difficult (87). In addition, USP27X inhibitor on a HAT-dependent cancer, for example, might have more and USP51 are not only structurally similar to USP22, they also fi detrimental than bene cial effects. compete with USP22 for activation by ATXN7L3 and ENY2 (88). In some cases, a DUB may function as an oncoprotein in one cancer Targeting bromodomains in KATs andatumorsuppressorinanother,suchasUSP18,whichplaysan Bromodomains are found in KATs, methyltransferases, and oncogenic role in lung cancer (89), but is a tumor suppressor in transcriptional coactivators, and some proteins, such as Brd4, sarcomas. In addition, the DUB USP7 plays various roles depending help organize superenhancers that drive expression of oncogenes on substrate abundance and physiologic state (90). Whether target- such as MYC (76). Bromodomain and extra-terminal motif (BET) ing USP22 will be effective in conferring antineoplastic effects is not inhibitors provide an alternate way to target MYC and have shown clear. inhibitors have been approved for therapies therapeutic potential in hematologic malignancies (77). The BET against myelomas and lymphomas (87). Even though many DUB family of proteins are characterized by two tandem bromodomains inhibitors show antitumor effects in vitro, they are not in clinical and an extra-terminal domain and regulate gene transcription trials.

Table 1. Key probes targeting GCN5/PCAF HAT activities and cancers, where they have in vitro and in vivo therapeutic potential.

Probe/inhibitor Known HAT targets Cancer cells sensitive to probe/inhibitor Citations

Oridonin GCN5, PCAF, TIP60, P300 Breast, gastric, glioblastoma, leukemia, lung, melanoma, prostate (62, 63, 68) Garcinol PCAF, TIP60, P300 Breast, cervical, colon, esophageal, hepatocellular, kidney, leukemia, lung, (64, 69, 70) lymphoma, myeloma, pancreatic, prostate PU139 GCN5, PCAF, CREB, P300 Breast, colon, glioblastoma, hepatocellular, neuroblastoma, ovarian (65) Butyrolactone 3 GCN5, PCAF Gastric (63) CPTH6 GCN5, PCAF Colon, glioblastoma, leukemia, lung, melanoma, ovarian, pancreatic, prostate (66) Anacardic acid PCAF, TIP60, P300 Leukemia, lung, lymphoma, prostate (70) Curcumin PCAF, TIP60, P300 Lymphoma, prostate (70, 71)

OF4 Cancer Res; 2020 CANCER RESEARCH

Downloaded from cancerres.aacrjournals.org on October 4, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst February 24, 2020; DOI: 10.1158/0008-5472.CAN-19-3652

Targeting SAGA and ATAC in MYC-Driven Cancers

Conclusions and Future Perspectives targeting other epigenetic regulators that interact with cMYC, such as TIP60 (91). Understanding the division of labor between transcrip- The proper balance between acetylation/deacetylation or ubiquiti- tional coactivator complexes in mediating oncogenic pathways will nation/deubiquitination is essential in normal physiology. This bal- hopefully help identify new strategies to tackle cancers. ance makes targeting enzymes controlling these states a double-edged sword. However, selective inhibition of KATs in cancers that are “addicted” to high expression of oncogenes may allow definition of Disclosure of Potential Conflicts of Interest therapeutic windows. By simultaneously repressing pathways that No potential conflicts of interest were disclosed. cancer cells depend on, there is a greater chance in reducing severity and reoccurrence. Acknowledgments The SAGA complex appears to play a key role in promoting MYC- This work was supported by the Postdoctoral Fellowship 131779-PF-18- driven cancers (Fig. 1D). Both GCN5 and USP22 affect MYC protein 034-01-DMC from the American Cancer Society (to L.M. Mustachio), the stability and SAGA serves as a coactivator of MYC target genes, which Postdoctoral Fellowship Award from the Center for Cancer Epigenetics at include genes encoding SAGA components. Inhibitors of SAGA and MD Anderson Cancer Center (to L.M. Mustachio), and a Tobacco Pilot Grant from MD Anderson Cancer Center (to S.Y.R. Dent) and in part from ATAC activities may be useful in targeting the functions of difficult to NIH grants R01 HD094400 and R35 GM131678 (to S.Y.R. Dent). We thank drug proteins, like cMYC. Identifying whether cancers driven by Drs. Michael Johnson McAndrew and Evangelia Koutelou for their helpful “undruggable” oncogenes are sensitive to GCN5, PCAF, and ADA2B consultation. inhibition is an area of active investigation. How SAGA components affect the functions of different MYC family members is also an Received November 22, 2019; revised January 28, 2020; accepted February 19, 2020; important question. Studies on GCN5 may also provide insight into published first February 24, 2020.

References 1. Grant PA, Duggan L, Cot^ e J, Roberts SM, Brownell JE, Candau R, et al. Yeast 15. Patel JH, Du Y, Ard PG, Phillips C, Carella B, Chen CJ, et al. The c-MYC Gcn5 functions in two multisubunit complexes to acetylate nucleosomal his- oncoprotein is a substrate of the acetyltransferases hGCN5/PCAF and TIP60. tones: characterization of an Ada complex and the SAGA (Spt/Ada) complex. Mol Cell Biol 2004;24:10826–34. Genes Dev 1997;11:1640–50. 16. Atanassov BS, Evrard YA, Multani AS, Zhang Z, Tora L, Devys D, et al. Gcn5 and 2. Martinez E. Multi-protein complexes in eukaryotic gene transcription. Plant Mol SAGA regulate shelterin protein turnover and telomere maintenance. Mol Cell Biol 2002;50:925–47. 2009;35:352–64. 3. Wang L, Dent SY. Functions of SAGA in development and disease. Epigenomics 17. Baker SP, Grant PA. The SAGA continues: expanding the cellular role of a 2014;6:329–39. transcriptional co- complex. Oncogene 2007;26:5329–40. 4. Riss A, Scheer E, Joint M, Trowitzsch S, Berger I, Tora L. Subunits of ADA-two- 18. Montanari A, Leo M, De Luca V, Filetici P, Francisci S. Gcn5 histone acetyl- A-containing (ATAC) or Spt-Ada-Gcn5-acetyltrasferase (SAGA) coactivator transferase is present in the mitoplasts. Biol Open 2019. DOI: 10.1242/ complexes enhance the acetyltransferase activity of GCN5. J Biol Chem 2015; bio.041244. 290:28997–9009. 19. Xu W, Edmondson DG, Evrard YA, Wakamiya M, Behringer RR, Roth SY. Loss 5. Sterner DE, Grant PA, Roberts SM, Duggan LJ, Belotserkovskaya R, Pacella LA, of Gcn5l2 leads to increased apoptosis and mesodermal defects during mouse et al. Functional organization of the yeast SAGA complex: distinct components development. Nat Genet 2000;26:229–32. involved in structural integrity, acetylation, and TATA-binding 20. Yamauchi T, Yamauchi J, Kuwata T, Tamura T, Yamashita T, Bae N, et al. protein interaction. Mol Cell Biol 1999;19:86–98. Distinct but overlapping roles of histone acetylase PCAF and of the closely 6. Soffers JHM, Li X, Saraf A, Seidel CW, Florens L, Washburn MP, et al. Char- related PCAF-B/GCN5 in mouse embryogenesis. Proc Natl Acad Sci U S A 2000; acterization of a metazoan ADA acetyltransferase complex. Nucleic Acids Res 97:11303–6. 2019;47:3383–94. 21. Koutelou E, Wang L, Schibler AC, Chao HP, Kuang X, Lin K, et al. USP22 7. Rossl A, Denoncourt A, Lin MS, Downey M. A synthetic non-histone substrate to controls multiple signaling pathways that are essential for vasculature study substrate targeting by the Gcn5 HAT and sirtuin HDACs. J Biol Chem formation in the mouse placenta. Development 2019;146. DOI: 10.1242/ 2019;294:6227–39. dev.174037. 8. Torres-Zelada EF, Stephenson RE, Alpsoy A, Anderson BD, Swanson SK, 22. Guelman S, Kozuka K, Mao Y, Pham V, Solloway MJ, Wang J, et al. The double- Florens L, et al. The Drosophila Dbf4 ortholog Chiffon forms a complex with histone-acetyltransferase complex ATAC is essential for mammalian develop- Gcn5 that is necessary for histone acetylation and viability. J Cell Sci 2019;132. ment. Mol Cell Biol 2009;29:1176–88. DOI: 10.1242/jcs.214072. 23. Brownell JE, Zhou J, Ranalli T, Kobayashi R, Edmondson DG, Roth SY, et al. 9. Huisinga KL, Pugh BF. A genome-wide housekeeping role for TFIID and a highly Tetrahymena histone acetyltransferase A: a homolog to yeast Gcn5p linking regulated stress-related role for SAGA in Saccharomyces cerevisiae. Mol Cell histone acetylation to gene activation. Cell 1996;84:843–51. 2004;13:573–85. 24. Yang XJ, Ogryzko VV, Nishikawa J, Howard BH, Nakatani Y. A p300/CBP- 10. Lee TI, Causton HC, Holstege FC, Shen WC, Hannett N, Jennings EG, et al. associated factor that competes with the adenoviral oncoprotein E1A. Nature Redundant roles for the TFIID and SAGA complexes in global transcription. 1996;382:319–24. Nature 2000;405:701–4. 25. Martinez-Cerdeno V, Lemen JM, Chan V, Wey A, Lin W, Dent SR, et al. N-Myc 11. Baptista T, Grunberg€ S, Minoungou N, Koster MJE, Timmers HTM, Hahn S, and GCN5 regulate significantly overlapping transcriptional programs in neural et al. SAGA is a general cofactor for RNA polymerase II transcription. Mol Cell stem cells. PLoS One 2012;7:e39456. 2017;68:130–43. 26. Knoepfler PS, Zhang XY, Cheng PF, Gafken PR, McMahon SB, Eisen- 12. Garcia-Molinero V, García-Martínez J, Reja R, Furio-Tarí P, Antunez O, Vinaya- man RN. Myc influences global chromatin structure. EMBO J 2006;25: chandran V, et al. The SAGA/TREX-2 subunit Sus1 binds widely to transcribed 2723–34. genesandaffectsmRNAturnoverglobally.EpigeneticsChromatin2018;11:13. 27. McMahon SB, Wood MA, Cole MD. The essential cofactor TRRAP recruits 13. Antonova SV, Boeren J, Timmers HTM, Snel B. Epigenetics and transcription the histone acetyltransferase hGCN5 to c-Myc. Mol Cell Biol 2000;20: regulation during eukaryotic diversification: the saga of TFIID. Genes Dev 2019; 556–62. 33:888–902. 28.LiuX,TesfaiJ,EvrardYA,DentSY,MartinezE.c-Myctransformation 14. Vinayachandran V, Reja R, Rossi MJ, Park B, Rieber L, Mittal C, et al. Widespread domain recruits the human STAGA complex and requires TRRAP and and precise reprogramming of yeast protein-genome interactions in response to GCN5 acetylase activity for transcription activation. J Biol Chem 2003; heat shock. Genome Res 2018;28:357–66. 278:20405–12.

AACRJournals.org Cancer Res; 2020 OF5

Downloaded from cancerres.aacrjournals.org on October 4, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst February 24, 2020; DOI: 10.1158/0008-5472.CAN-19-3652

Mustachio et al.

29. McMahon SB, Van Buskirk HA, Dugan KA, Copeland TD, Cole MD. The novel 51. Zhang K, Yang L, Wang J, Sun T, Guo Y, Nelson R, et al. Ubiquitin-specific ATM-related protein TRRAP is an essential cofactor for the c-Myc and E2F protease 22 is critical to in vivo angiogenesis, growth and of non-small oncoproteins. Cell 1998;94:363–74. cell lung cancer. Cell Commun Signal 2019;17:167. 30. Hirsch CL, Coban Akdemir Z, Wang L, Jayakumaran G, Trcka D, Weiss A, et al. 52. LinZ,YangH,KongQ,LiJ,LeeSM,GaoB,etal.USP22antagonizesp53 Myc and SAGA rewire an alternative splicing network during early somatic cell transcriptional activation by deubiquitinating Sirt1 to suppress cell apo- reprogramming. Genes Dev 2015;29:803–16. ptosis and is required for mouse embryonic development. Mol Cell 2012; 31. Seruggia D, Oti M, Tripathi P, Canver MC, LeBlanc L, Di Giammartino DC, et al. 46:484–94. TAF5L and TAF6L maintain self-renewal of embryonic stem cells via the MYC 53. Atanassov BS, Dent SY. USP22 regulates cell proliferation by deubiquitinating regulatory network. Mol Cell 2019;74:1148–63. the transcriptional regulator FBP1. EMBO Rep 2011;12:924–30. 32. Wang L, Koutelou E, Hirsch C, McCarthy R, Schibler A, Lin K, et al. GCN5 54. Zhao Y, Lang G, Ito S, Bonnet J, Metzger E, Sawatsubashi S, et al. A TFTC/ regulates FGF signaling and activates selective MYC target genes during early STAGA module mediates histone H2A and H2B deubiquitination, coactivates embryoid body differentiation. Stem Cell Reports 2018;10:287–99. nuclear receptors, and counteracts heterochromatin silencing. Mol Cell 2008;29: 33. Zhang N, Ichikawa W, Faiola F, Lo SY, Liu X, Martinez E. MYC interacts with the 92–101. human STAGA coactivator complex via multivalent contacts with the GCN5 and 55. Ramachandran S, Haddad D, Li C, Le MX, Ling AK, So CC, et al. The SAGA TRRAP subunits. Biochim Biophys Acta 2014;1839:395–405. deubiquitination module promotes DNA repair and class switch recombination 34. ChenL,WeiT,SiX,WangQ,LiY,LengY,etal.Lysineacetyltransferase through ATM and DNAPK-mediated gammaH2AX formation. Cell Rep 2016; GCN5 potentiates the growth of non-small cell lung cancer via promotion 15:1554–65. of E2F1, cyclin D1, and cyclin E1 expression. J Biol Chem 2013;288: 56. Yang X, Zang H, Luo Y, Wu J, Fang Z, Zhu W, et al. High expression of USP22 14510–21. predicts poor prognosis and advanced clinicopathological features in solid 35. Mustachio LM, Roszik J, Farria AT, Guerra K, Dent SY. Repression of GCN5 tumors: a meta-analysis. Onco Targets Ther 2018;11:3035–46. expression or activity attenuates c-MYC expression in non-small cell lung 57. Melo-Cardenas J, Zhang Y, Zhang DD, Fang D. Ubiquitin-specific peptidase 22 cancer. Am J Cancer Res 2019;9:1830–45. functions and its involvement in disease. Oncotarget 2016;7:44848–56. 36. Liu K, Zhang Q, Lan H, Wang L, Mou P, Shao W, et al. GCN5 Potentiates Glioma 58. Kim D, Hong A, Park HI, Shin WH, Yoo L, Jeon SJ, et al. Deubiquitinating Proliferation and Invasion via STAT3 and AKT Signaling Pathways. Int J Mol Sci USP22 positively regulates c-Myc stability and tumorigenic activity in 2015;16:21897–910. mammalian and breast cancer cells. J Cell Physiol 2017;232:3664–76. 37. Zhao L, Pang A, Li Y. Function of GCN5 in the TGF-beta1-induced epithelial-to- 59. Schrecengost RS, Dean JL, Goodwin JF, Schiewer MJ, Urban MW, Stanek TJ, mesenchymal transition in breast cancer. Oncol Lett 2018;16:3955–63. et al. USP22 regulates oncogenic signaling pathways to drive lethal cancer 38. Majaz S, Tong Z, Peng K, Wang W, Ren W, Li M, et al. Histone acetyl transferase progression. Cancer Res 2014;74:272–86. GCN5 promotes human hepatocellular carcinoma progression by enhancing 60. Melo-Cardenas J, Xu Y, Wei J, Tan C, Kong S, Gao B, et al. USP22 deficiency leads AIB1 expression. Cell Biosci 2016;6:47. to myeloid leukemia upon oncogenic Kras activation through a PU.1-dependent 39. Li Y, Jaramillo-Lambert AN, Yang Y, Williams R, Lee NH, Zhu W. And-1 is mechanism. Blood 2018;132:423–34. required for the stability of histone acetyltransferase Gcn5. Oncogene 2012;31: 61. Meyers RM, Bryan JG, McFarland JM, Weir BA, Sizemore AE, Xu H, et al. 643–52. Computational correction of copy number effect improves specificity 40. Tzelepis K, Koike-Yusa H, De Braekeleer E, Li Y, Metzakopian E, Dovey OM, of CRISPR-Cas9 essentiality screens in cancer cells. Nat Genet 2017;49: et al. A CRISPR dropout screen identifies genetic vulnerabilities and therapeutic 1779–84. targets in . Cell Rep 2016;17:1193–205. 62. Han Y, Luo J, Ranish J, Hahn S. Architecture of the Saccharomyces cerevisiae 41. Farria AT, Mustachio LM, Akdemir ZHC, Dent SYR. GCN5 HAT inhibition SAGA transcription coactivator complex. EMBO J 2014;33:2534–46. reduces human Burkitt lymphoma cell survival through reduction of MYC target 63. ArmourSM,BennettEJ,BraunCR,ZhangXY,McMahonSB,GygiSP,etal. gene expression and impeding BCR signaling pathways. Oncotarget 2019;10: Ahigh-confidence interaction map identifies SIRT1 as a of acet- 5847–58. ylation of USP22 and the SAGA coactivator complex. Mol Cell Biol 2013;33: 42. Jia YL, Xu M, Dou CW, Liu ZK, Xue YM, Yao BW, et al. P300/CBP-associated 1487–502. factor (PCAF) inhibits the growth of hepatocellular carcinoma by promoting cell 64. Pinto-Fernandez A, Kessler BM. DUBbing cancer: deubiquitylating enzymes autophagy. Cell Death Dis 2016;7:e2400. involved in epigenetics, DNA damage and the cell cycle as therapeutic targets. 43. Fei HJ, Zu LD, Wu J, Jiang XS, Wang JL, Chin YE, et al. PCAF acts as a gastric Front Genet 2016;7:133. cancer suppressor through a novel PCAF-p16-CDK4 axis. Am J Cancer Res 65. Zhao D, Guan H, Zhao S, Mi W, Wen H, Li Y, et al. YEATS2 is a selective histone 2016;6:2772–86. crotonylation reader. Cell Res 2016;26:629–32. 44. Infante P, Canettieri G, Gulino A, Di Marcotullio L. Yin-Yang strands of PCAF/ 66.MiW,GuanH,LyuJ,ZhaoD,XiY,JiangS,etal.YEATS2linkshistone Hedgehog axis in cancer control. Trends Mol Med 2014;20:416–8. acetylation to tumorigenesis of non-small cell lung cancer. Nat Commun 45. Malatesta M, Steinhauer C, Mohammad F, Pandey DP, Squatrito M, Helin K. 2017;8:1088. Histone acetyltransferase PCAF is required for Hedgehog-Gli-dependent tran- 67.KrebsAR,KarmodiyaK,Lindahl-AllenM,StruhlK,ToraL.SAGAand scription and cancer cell proliferation. Cancer Res 2013;73:6323–33. ATAC histone acetyl transferase complexes regulate distinct sets of genes 46. Koutsogiannouli EA, Wagner N, Hader C, Pinkerneil M, Hoffmann MJ, and ATAC defines a class of p300-independent enhancers. Mol Cell 2011; Schulz WA. Differential effects of histone acetyltransferase GCN5 or PCAF 44:410–23. knockdown on urothelial carcinoma cells. Int J Mol Sci 2017;18. DOI: 68. Spedale G, Timmers HT, Pijnappel WW. ATAC-king the complexity of SAGA 10.3390/ijms18071449. during evolution. Genes Dev 2012;26:527–41. 47. Durbin AD, Zimmerman MW, Dharia NV, Abraham BJ, Iniguez AB, 69. Wapenaar H, Dekker FJ. Histone acetyltransferases: challenges in targeting bi- Weichert-Leahey N, et al. Selective gene dependencies in MYCN- substrate enzymes. Clin Epigenetics 2016;8:59. amplified neuroblastoma include the core transcriptional regulatory circuit- 70. Xia S, Zhang X, Li C, Guan H. Oridonin inhibits breast cancer growth and ry. Nat Genet 2018;50:1240–6. metastasis through blocking the Notch signaling. Saudi Pharm J 2017;25: 48. Durand A, Bonnet J, Fournier M, Chavant V, Schultz P. Mapping the deubi- 638–43. quitination module within the SAGA complex. Structure 2014;22:1553–9. 71. Shi M, Lu XJ, Zhang J, Diao H, Li G, Xu L, et al. Oridonin, a novel lysine 49. Zhang XY, Varthi M, Sykes SM, Phillips C, Warzecha C, Zhu W, et al. The acetyltransferases inhibitor, inhibits proliferation and induces apoptosis putative cancer stem cell marker USP22 is a subunit of the human SAGA in gastric cancer cells through p53- and caspase-3-mediated mechanisms. complex required for activated transcription and cell-cycle progression. Mol Cell Oncotarget 2016;7:22623–31. 2008;29:102–11. 72. Oike T, Ogiwara H, Torikai K, Nakano T, Yokota J, Kohno T. Garcinol, a histone 50. Glinsky GV. Genomic models of metastatic cancer: functional analysis of acetyltransferase inhibitor, radiosensitizes cancer cells by inhibiting non- death-from-cancer signature genes reveals aneuploid, anoikis-resistant, homologous end joining. Int J Radiat Oncol Biol Phys 2012;84:815–21. metastasis-enabling phenotype with altered cell cycle control and activated 73. Gajer JM, Furdas SD, Grunder€ A, Gothwal M, Heinicke U, Keller K, et al. Polycomb Group (PcG) protein chromatin silencing pathway. Cell Cycle Histone acetyltransferase inhibitors block neuroblastoma cell growth in vivo. 2006;5:1208–16. Oncogenesis 2015;4:e137.

OF6 Cancer Res; 2020 CANCER RESEARCH

Downloaded from cancerres.aacrjournals.org on October 4, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst February 24, 2020; DOI: 10.1158/0008-5472.CAN-19-3652

Targeting SAGA and ATAC in MYC-Driven Cancers

74. Di Martile M, Desideri M, De Luca T, Gabellini C, Buglioni S, Eramo A, et al. 83. An S, Fu L. Small-molecule PROTACs: an emerging and promising Histone acetyltransferase inhibitor CPTH6 preferentially targets lung cancer approach for the development of targeted therapy drugs. EBioMedicine stem-like cells. Oncotarget 2016;7:11332–48. 2018;36:553–62. 75. SinghBN,ZhangG,HwaYL,LiJ,DowdySC,JiangSW.Nonhistoneprotein 84. Bassi ZI, Fillmore MC, Miah AH, Chapman TD, Maller C, Roberts EJ, et al. acetylation as cancer therapy targets. Expert Rev Anticancer Ther 2010;10: Modulating PCAF/GCN5 immune cell function through a PROTAC approach. 935–54. ACS Chem Biol 2018;13:2862–7. 76. Doroshow DB, Eder JP, LoRusso PM. BET inhibitors: a novel epigenetic 85. Moustakim M, Christott T, Monteiro OP, Bennett J, Giroud C, Ward J, et al. approach. Ann Oncol 2017;28:1776–87. Discovery of an MLLT1/3 YEATS domain chemical probe. Angew Chem Int Ed 77. Garcia-Carpizo V, Ruiz-Llorente S, Sarmentero J, Grana-Castro~ O, Pisano DG, Engl 2018;57:16302–7. Barrero MJ. CREBBP/EP300 bromodomains are critical to sustain the GATA1/ 86. Li X, Li XM, Jiang Y, Liu Z, Cui Y, Fung KY, et al. Structure-guided development MYC regulatory axis in proliferation. Epigenetics Chromatin 2018;11:30. of YEATS domain inhibitors by targeting pi-pi-pi stacking. Nat Chem Biol 2018; 78. Taniguchi Y. The bromodomain and extra-terminal domain (BET) family: 14:1140–9. functional anatomy of BET paralogous proteins. Int J Mol Sci 2016;17. DOI: 87. Huang X, Dixit VM. Drugging the undruggables: exploring the ubiquitin system 10.3390/ijms17111849. for drug development. Cell Res 2016;26:484–98. 79. Stathis A, Bertoni F. BET proteins as targets for anticancer treatment. 88. Atanassov BS, Mohan RD, Lan X, Kuang X, Lu Y, Lin K, et al. ATXN7L3 and Cancer Discov 2018;8:24–36. ENY2 coordinate activity of multiple H2B deubiquitinases important for cellular 80. Sunami Y, Araki M, Kan S, Ito A, Hironaka Y, Imai M, et al. Histone proliferation and tumor growth. Mol Cell 2016;62:558–71. acetyltransferase p300/CREB-binding protein-associated factor (PCAF) is 89. Mustachio LM, Kawakami M, Lu Y, Rodriguez-Canales J, Mino B, Behrens C, required for all-trans-retinoic acid-induced granulocytic differentiation in leu- et al. The ISG15-specific protease USP18 regulates stability of PTEN. Oncotarget kemia cells. J Biol Chem 2017;292:2815–29. 2017;8:3–14. 81. Moustakim M, Clark PG, Trulli L, Fuentes de Arriba AL, Ehebauer MT, 90.WangX,ZhangQ,WangY,ZhuangH,ChenB.Clinicalsignificance of Chaikuad A, et al. Discovery of a PCAF bromodomain chemical probe. ubiquitin specific protease 7 (USP7) in predicting prognosis of hepatocel- Angew Chem Int Ed Engl 2017;56:827–31. lular carcinoma and its functional mechanisms. Med Sci Monit 2018;24: 82. Humphreys PG, Bamborough P, Chung CW, Craggs PD, Gordon L, Grandi P, 1742–50. et al. Discovery of a potent, cell penetrant, and selective p300/CBP-associated 91. Frank SR, Parisi T, Taubert S, Fernandez P, Fuchs M, Chan HM, et al. MYC factor (PCAF)/general control nonderepressible 5 (GCN5) bromodomain chem- recruits the TIP60 histone acetyltransferase complex to chromatin. EMBO Rep ical probe. J Med Chem 2017;60:695–709. 2003;4:575–80.

AACRJournals.org Cancer Res; 2020 OF7

Downloaded from cancerres.aacrjournals.org on October 4, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst February 24, 2020; DOI: 10.1158/0008-5472.CAN-19-3652

Targeting the SAGA and ATAC Transcriptional Coactivator Complexes in MYC-Driven Cancers

Lisa Maria Mustachio, Jason Roszik, Aimee Farria, et al.

Cancer Res Published OnlineFirst February 24, 2020.

Updated version Access the most recent version of this article at: doi:10.1158/0008-5472.CAN-19-3652

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 Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerres.aacrjournals.org/content/early/2020/04/07/0008-5472.CAN-19-3652. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on October 4, 2021. © 2020 American Association for Cancer Research.