Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-3373

CLINICAL CANCER RESEARCH | TRANSLATIONAL CANCER MECHANISMS AND THERAPY

Wee1 Kinase Inhibitor AZD1775 Effectively Sensitizes Esophageal Cancer to Radiotherapy Linlin Yang1, Changxian Shen1, Cory J. Pettit1, Tianyun Li1, Andrew J. Hu1, Eric D. Miller1, Junran Zhang1, Steven H. Lin2, and Terence M. Williams1

ABSTRACT ◥ Purpose: Esophageal cancer is a deadly malignancy with a 5-year (100 nmol/L) as monotherapy did not alter the viability of survival rate of only 5% to 20%, which has remained unchanged for esophageal cancer cells, but significantly radiosensitized esoph- decades. Esophageal cancer possesses a high frequency of TP53 ageal cancer cells. AZD1775 significantly abrogated radiation- mutations leading to dysfunctional G1 cell-cycle checkpoint, which induced G2–M phase arrest and attenuation of p-CDK1-Y15. – likely makes esophageal cancer cells highly reliant upon G2 M Moreover, AZD1775 increased radiation-induced mitotic checkpoint for adaptation to DNA replication stress and DNA catastrophe, which was accompanied by increased gH2AX damage after radiation. We aim to explore whether targeting levels, and subsequently reduced survival after radiation. – kinase to abolish G2 M checkpoint sensitizes esophageal cancer Importantly, AZD1775 in combination with radiotherapy cells to radiotherapy. resulted in marked tumor regression of esophageal cancer tumor Experimental Design: Cell viability was assessed by cytotoxicity xenografts. – and colony-forming assays, cell-cycle distribution was analyzed by Conclusions: Abrogation of G2 M checkpoint by targeting flow cytometry, and mitotic catastrophe was assessed by immuno- Wee1 kinase with AZD1775 sensitizes esophageal cancer cells to fluorescence staining. Human esophageal cancer xenografts were radiotherapy in vitro and in mouse xenografts. Our findings generated to explore the radiosensitizing effect of AZD1775 in vivo. suggest that inhibition of Wee1 by AZD1775 is an effective Results: The IC50 concentrations of AZD1775 on esophageal strategy for radiosensitization in esophageal cancer and warrants cancer cell lines were between 300 and 600 nmol/L. AZD1775 clinical testing.

Introduction response to ever-changing intracellular and extracellular genotoxic insults, cells activate DNA damage, replication, and mitotic check- Esophageal cancer is the sixth leading cause of cancer-related death points, which function to inhibit the activity of CDKs and halt cell- and affects more than 450,000 people worldwide (1). Standard-of-care cycle progression to provide time to repair DNA damage and fix therapy for localized esophageal cancer is radiotherapy and chemo- chromatin disruption (7). The fine coupling of and DNA therapy followed by surgery, but recurrence rates remain high. More- damage checkpoints ensures genome integrity and cell survival (7). over, approximately of 50% patients diagnosed with esophageal cancer Aberrant activation of CDKs and hence uncontrolled cell-cycle pro- present with unresectable or metastatic disease (2). In the past decade, gression is a hallmark of cancer cells (8). Many human cancers have although great advances have been made for the prevention and deficits in G –S checkpoint due to mutations in the signaling axis control of many cancers such as lung cancer and breast cancer, the 1 including mutations of TP53, CDKN2A, and RB (9). Treatment of overall 5-year survival rate of patients with esophageal cancer remains these cells with radiation induces a G –M arrest, allowing time for below 20% and the incidence is increasing rapidly worldwide (1, 3, 4). 2 DNA repair, thus leading to a higher level of dependence of these Therefore, there is an urgent need to develop novel effective therapies cancer cells on G –M checkpoint for survival. In these cases, genetic for the management of esophageal cancer (2, 5). 2 abrogation of the G –M checkpoint may allow entry of cells into Proper cell proliferation and accurate genetic material duplication 2 with incompletely-repaired damaged DNA, ultimately leading depends on the tight and fine coordination of the cell-cycle surveillance to mitotic catastrophe and cell death (10). It has therefore been systems including G –G ,S,G, and M cell-cycle checkpoints (6). 0 1 2 proposed that small molecules targeting G –M checkpoint are prom- Cell-cycle progression is controlled by -dependent kinases 2 ising cancer therapy agents either as monotherapy or in combination (CDK), which are regulated by cell growth and mitogenic signals. In with radiotherapy and chemotherapy (5, 11–13). Wee1 kinase is essential for scheduled through inhib- itory phosphorylation of CDK1 and CDK2 at the conserved tyrosine15 1The Ohio State University Medical Center, Arthur G. James Comprehensive 2 residue (14). Particularly, Wee1-mediated phosphorylation and inhi- Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio. The – University of Texas MD Anderson Cancer Center, Houston, Texas. bition of CDK1 plays a critical role in G2 M checkpoint under normal cell growth and in response to DNA damage or replication stress (15). Note: Supplementary data for this article are available at Clinical Cancer DNA damage or replication stress activates ATM/CHK2 and Research Online (http://clincancerres.aacrjournals.org/). ATR/CHK1 signaling cascades to maintain genome stability and cell Corresponding Author: Terence M. Williams, Ohio State University Wexner viability (13). Activation of CHK1 by ATR in response to various types Medical Center, 460 W. 12th Avenue, Room 492, Columbus, OH 43235. of DNA lesions phosphorylates and stimulates Wee1 activation to Phone: 614-366-2621; Fax: 614-293-4044; E-mail: [email protected] suppress CDK1 activity thereby preventing entry into mitosis (15). Forced cell-cycle progression in the setting of DNA damage perpe- Clin Cancer Res 2020;XX:XX–XX tuates DNA and chromatin damage, and leads to cell death because doi: 10.1158/1078-0432.CCR-19-3373 of irreparable genetic lesions (11). Interestingly, Wee1 expression is 2020 American Association for Cancer Research. upregulated in many cancers and associated with the survival of

AACRJournals.org | OF1

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-3373

Yang et al.

xenografts. Our findings suggest that AZD1775 in combination with Translational Relevance radiotherapy may improve the therapeutic outcome of patients with Stage II/III esophageal cancers are commonly treated with esophageal cancer. radiation or chemoradiation, with or without surgery. However, esophageal cancer has very poor prognosis, and preclinical studies have shown esophageal cancer cells are resistant to radiation. The Materials and Methods majority of both esophageal adenocarcinoma and squamous cell Antibodies, chemicals, and cell culture carcinomas harbor mutations in TP53, an important tumor sup- OE33, SK4, FLO1, KYSE30, and AGS cell lines were maintained at – pressor gene that also functions to promote cell-cycle arrest in G1 S 37 Cin5%CO2 in DMEM medium supplemented with 10% FBS after DNA damage from radiation. In this preclinical study, we (Sigma), 1% penicillin/streptomycin (Life Technologies). The detailed target the G2–M cell-cycle checkpoint with AZD1775, a Wee1 cell line information is listed in Supplementary Table S1. AZD1775 was kinase inhibitor, in combination with radiation to enhance ther- obtained under a material transfer agreement from NCI-CTEP apeutic efficacy. We find that in TP53-mutated cells lacking an through AstraZeneca and was dissolved in DMSO (Sigma) and added – effective G1 S checkpoint, AZD1775 markedly radiosensitizes to medium with a final concentration of no more than 0.1% DMSO. esophageal cancer cells to radiation both in cell culture assays and Total CDK1, phospho-CDK1 (Tyr15), phospho-Wee1 (Ser642), phos- animal studies. Our results justify a clinical trial to determine the pho-H2AX (S139), phospho-histone H3 (S10), , , safety and efficacy of combining AZD1775 and radiation in , cyclin E2, and GAPDH primary antibodies were purchased patients with esophageal cancer. from Cell Signaling Technology. Anti-rabbit and anti-mouse second- ary antibodies were purchased from LI-COR Biosciences.

AlamarBlue assay and IC50 determination patients with cancer (16–18). Given the pivotal role for Wee1 in AlamarBlue assay was performed according to manufacturer's instruc- the regulation of CDK1 activity, targeting Wee1 has been proposed tions (Roche). Briefly, cells were seeded in 96-well plates in six replicates at for the sensitization of cancer cells to radiotherapy and a density of 2,000 cells per well in 100 mL medium. The next day, the cells chemotherapy (11, 19–21). Large-scale genomic studies have found were treated with AZD1775 at various concentrations. After 72 hours, that esophageal cancer has an extremely high frequency of TP53 alamarBlue reagent was added and incubated at 37C for 4 hours, and mutations, ranging from 44% to 93% (22, 23). Recently, The Cancer absorbance was measured at 490 nm. IC50 was determined using the Genome Atlas (TCGA) demonstrated that TP53 mutations were the nonlinear four-parameter regression function in GraphPad Prism. single most common significantly mutated gene in ESCA, occurring in 71% and 91% of esophageal adenocarcinoma and esophageal Immunoblotting squamous cell carcinoma, respectively (24). Therefore, esophageal Immunoblotting was performed as described previously (36). Brief- – cancer cells may depend on G2 M checkpoint for survival and may ly, cell lysates were prepared using RIPA buffer (Thermo Fisher – fi be very sensitive to G2 M checkpoint abrogation by Wee1 inhibition. Scienti c) supplemented with 1 protease inhibitors (Complete; – AZD1775 is a novel small molecule inhibitor that disrupts G2 M Roche) and phosphatase inhibitors (PhosSTOP; Roche) followed by checkpoint by directly inhibiting Wee1 kinase (25). Previous studies protein quantification with the Dc Protein Assay Kit (Bio-Rad). Equal have demonstrated that the sensitivity of AZD1775 depends on p53 amounts of protein were loaded and resolved by SDS-PAGE and functional loss in various types of cancers including non–small cell transferred onto nitrocellulose membranes. Membranes were incu- lung cancer (26–29). However, it has also been reported that Wee1 bated in 5% BSA in Tris-buffered saline with 0.1% Tween-20 (TBST) inhibition could radiosensitize carcinoma cells without TP53 muta- blocking buffer for 1 hour at room temperature. Primary antibodies – tions (30). In addition to inhibiting G2 M checkpoint, AZD1775 has with dilution of 1:200 to 1,000 were allowed to bind overnight at 4 C, been shown to induce DNA replication stress via nucleotide exhaus- or for 2 hours at room temperature. After washing in TBST, the tion (31, 32), and to reduce homologous recombination repair (33). membranes were incubated with immunofluorescent secondary anti- Although Wee1 is not the core component of the replication stress bodies at a 1:5,000 dilution for 1 hour at room temperature. Mem- response pathway, activation of Wee1 by CHK1 induces CDK1 and branes were washed with TBST and allowed to air dry prior to imaging CDK2 to halt cell-cycle progression in response to DNA damage (34). via LI-COR Odyssey CLx Imaging System (Thermo Fisher Scientific). Thus, targeting Wee1 may force cells to enter mitosis in the presence of incomplete DNA replication, which might exacerbate replication Radiation clonogenic assay stress and development of lethal DNA damage (21, 35). AZD1775 Radiation clonogenic assays were performed essentially as described has been tested preclinically in many types of cancers, and has been previously (37). In brief, single cells seeded in 60 or 100 mm tissue shown to radiosensitize and chemosensitize certain cancers, including culture plates were incubated with DMSO or AZD1775 for 3 hours and pancreatic, breast, prostate, lung, and glioblastoma cancers (20, 21). then irradiated with various doses (0–8 Gy). Radiation was performed However, the effects of AZD1775 on esophageal cancer (a disease with with 160 kV, 25 mA at a dose rate of approximately 113 cGy/min using an extraordinary high rate of TP53 mutations) as monotherapy or in a RS-2000 biological irradiator (RadSource), and cells were fixed 7 to combination with other therapeutics remains to be determined. 10 days after seeding. The number of colonies containing at least 50 In this study, we investigated the anti-neoplastic properties of cells was counted using a dissecting microscope (Leica Microsystems, AZD1775 in combination with radiation in esophageal cancer. We Inc.) and dose–enhancement ratio (DER) was calculated as reported TP53 fi – hypothesized that -de cient cells are sensitive to a G2 M check- previously (38). point inhibitor and as such, combining Wee1 inhibition and radiation should target TP53-deficient ESCA cells. We found that abrogation of Immunofluorescence for mitotic catastrophe – fi G2 M checkpoint by targeting Wee1 kinase with AZD1775 markedly Cells on coverslips were xed with 2% paraformaldehyde, permea- sensitizes esophageal cancer cells to radiotherapy in vitro and in mouse bilized with 1% Triton X-100, and blocked with 3% BSA in PBS. Briefly,

OF2 Clin Cancer Res; 2020 CLINICAL CANCER RESEARCH

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-3373

Targeting Wee1 for Radiosensitization of Esophageal Cancer

for paraffin-embedded tissue sections from tumor xenografts, sections also higher WEE1 mRNA expression in esophageal cancer tumors than were cut onto slides and deparaffinized in xylene and rehydrated that in normal tissues, although not statistically significant. Interest- through a graded alcohol series. Then, slides were washed in distilled ingly, MCM10, CCNE1, CCNE2, FBXO5, and CLSPN have been shown water. Cells on coverslips or tissue sections were stained with anti- to be biomarkers predictive for responsiveness to AZD1775 (39). tubulin antibody (Cell Signaling Technology), washed, and incubated Similarly, analysis of TCGA and GTE databases revealed that these with a fluorophore-conjugated secondary antibody (Biotium). Fol- five genes were significantly overexpressed in esophageal cancer tumor lowing nucleus counterstaining with DAPI, the slides were examined versus normal tissues (Fig. 1B). Further analysis of the Oncomine on a Zeiss fluorescence microscope. For each experiment, mitotic database (http://oncomine.org/) confirmed overexpression of these catastrophe was determined in at least 100 cells. genes in human ESCA tumors relative to normal tissues (Supplemen- tary Fig. S1). These findings of CDK1 upregulation and AZD1775 Flow cytometry responsiveness gene signature suggested targeting Wee1 with Cells were seeded into six-well plates at a density of 200,000 cells per AZD1775 may sensitize esophageal cancer to radiotherapy. well in 2 mL medium for 16 hours. The cells were treated with AZD1775 for 3 hours, followed by ionizing radiation (IR) and then Wee1 kinase inhibition with AZD1775 sensitizes esophageal cultured for 24 hours. Cells were fixed in 70% ethanol at 20C and cancer cells to radiation stained with DNA staining solution containing propidium iodide and To evaluate the potential radiation-sensitizing efficacy of Wee1 RNaseA (Sigma-Aldrich) overnight. All data were acquired on LSRII inhibition by AZD1775 in ESCA, we initially investigated the inde- cytometry (BD Biosciences) and each sample was assessed using a pendent cytotoxic effect of AZD1775 on esophageal cancer cell lines. collection of 10,000 events, followed by analysis using FlowJo software All four cell lines used in our study have TP53 mutation, including (FlowJo). esophageal adenocarcinoma cell lines (OE33, SK4, and FLO1) and an esophageal squamous cell carcinoma cell line (KYSE30). The cytotoxic In vivo studies effect of AZD1775 in esophageal cancer cell lines was assessed by Animal studies were conducted in accordance with an approved alamarBlue assay. We found that the IC50 values of AZD1775 in these protocol adhering to the IACUC policies and procedures at The Ohio esophageal cancer cell lines ranged from 252 to 624 nmol/L (Fig. 2A). State University. Six- to eight-week-old male athymic nude mice To explore the potential of AZD1775 as a radiosensitizer for esoph- (Taconic Farms Inc.) were caged in groups of five or less, and fed ageal cancer, FLO-1 and OE33 cells were treated with increasing doses with a diet of animal chow and water ad libitum. OE33 and FLO1 cells of radiation in the presence or absence of 100 nmol/L AZD1775, were injected subcutaneously into the flanks of each mouse at 2 106 followed by clonogenic (colony-forming) assay. At 100 nmol/L of and 2.5 106 cells per injection, respectively. Treatment regimens AZD1775 alone, we noted minimal cytotoxicity of the drug (Supple- were started once tumors reached 150 mm3 in size, 2 to 4 weeks mentary Fig. S2). In combination with radiation however, AZD1775 postinjection. AZD1775 powder was suspended in 0.9% sodium could effectively sensitize esophageal cancer cells to radiation treat- chloride containing 5% dextrose. AZD1775 was administered orally ment, with DER up to 3.14 in SK4 cells, 1.46 in OE33 cells, 1.34 in FLO1 using a sterile 18G gavage needle at 50 mg/kg twice a day for 5 cells, and 1.23 in KYSE cells (Fig. 2B). consecutive days. Using a custom shielding apparatus to block non- targeted areas, 4 Gy of radiation was administered directly to tumors Wee1 kinase inhibition abrogates radiation-induced G2–M once daily for 5 consecutive days. For combination treatment, mice phase cell-cycle arrest fi – were treated with radiation 2 to 3 hours after the rst daily dosing of Wee1 kinase plays a key role in promoting G2 M cell-cycle arrest AZD1775. To obtain a tumor growth curve, perpendicular diameter after DNA damage (e.g., by ionizing radiation) to allow time for cells to measurements of each tumor were measured every 2 to 5 days from the undergo DNA repair, by inactivating CDK1. We explored whether the first day of injection with digital calipers, and volumes were calculated radiosensitizing effects of AZD1775 are associated with abrogation of using the formula (L W W)/2. Two mice from each group were radiation-induced G2–M cell-cycle arrest in asynchronously growing sacrificed after three days treatment, to isolate tumor xenografts for cells by flow cytometry assay. In both FLO1 and OE33 cells, AZD1775 immunoblotting and immunofluorescence staining. treatment (100 nmol/L) decreased whereas 4 Gy radiation alone – increased cells in G2 M phase. However, pretreatment of cells with Data analysis AZD1775 at 3 hours before radiation significantly reduced the accu- – Data were analyzed similarly as described previously (37), and are mulation of G2 M phase cells after radiation (Fig. 3A). Activation of presented as the mean SEM. Wee1 kinase prevents cells from entering into mitosis through phos- phorylating and subsequent inactivation of CDK1. Immunoblotting analyses showed AZD1775 inhibited Wee1 and CDK1 phosphoryla- Results tion in a time-dependent manner, with maximal changes noted Wee1 is a potential therapeutic target in human esophageal 24 hours after treatment (Fig. 3B). Of note, radiation alone mildly cancer increased the phosphorylation of both Wee1 and CDK1 at 24 hours The sensitization of many cancers to radiotherapy and chemother- after radiation. Exposing cells to AZD1775 before radiation attenuated apy by targeting Wee1 and the high mutation rate of TP53 suggests that radiation-induced Wee1 and CDK1 phosphorylation, most notable at Wee1 inhibition is a potential therapeutic strategy for ESCA. To this 24 hours after radiation. Interestingly, AZD1775 alone induced end, we first analyzed the TCGA and GTEx databases with GEPIA gH2AX expression and enhanced radiation-mediated increase of (http://gepia.cancer-pku.cn/) for WEE1 and CDK1 mRNA expression gH2AX particularly after 24 hours of treatment. Most studies have in esophageal cancer. Significant elevation of CDK1 mRNA expression shown that the therapeutic effects AZD1775 is related to the abro- was found in esophageal cancer tumors compared with normal tissues, gation of the G2 checkpoint and/or unscheduled mitotic entry. How- suggesting increased cell proliferation or mechanisms to promote ever, emerging evidence suggest that Wee1 inhibition suppresses DNA – transition through G2 M phases (Fig. 1A). Interestingly, there was damage repair and induces replication stress (32, 33), both of which

AACRJournals.org Clin Cancer Res; 2020 OF3

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-3373

Yang et al.

A CDK1 WEE1 86 6 4 4 2 2 0 0

Esophageal cancer Esophageal cancer [num (T) = 182; num (N) = 286] [num (T) = 182; num (N) = 286] B MCM10 CCNE1 CCNE2 FBXO5 CLSPN 6 54321 86 6 6 23 23 24 24 145 145 0 0 0 0 0

Esophageal cancer Esophageal cancer Esophageal cancer Esophageal cancer Esophageal cancer [num (T) = 182; num (N) = 286] [num (T) = 182; num (N) = 286] [num (T) = 182; num (N) = 286] [num (T) = 182; num (N) = 286] [num (T) = 182; num (N) = 286]

Figure 1. CDK1 and genes associated with Wee1 inhibitor sensitivity are overexpressed in ESCA. Relative mRNA expression levels of CDK1 and WEE1 (A) or genes associated with sensitivity to Wee1 inhibitor (B) in esophageal carcinoma tumor (T) versus normal (N) tissues. Box plots were derived from Gene Expression Profiling Interactive Analysis (GEPIA) based on TCGA and GTEx databases. Red and black boxes represent the relative mRNA expression levels of the genes in the tumor and normal samples, respectively. The y-axis represents the relative mRNA expression levels of the genes in terms of log2 (TPMþ1). Tumor samples ¼ 182; normal samples ¼ 286; , P < 0.05. TPM, transcripts per million.

Figure 2.

AZD1775 effectively sensitizes esophageal cancer cells to radiation. A, IC50s of AZD1775 in esophageal cancer cells. OE33, SK4, FLO1, and KYSE cells were treated with different concentrations of AZD1775 for 72 hours. The cell viability was assessed by alamarBlue assay, and IC50 values were calculated. B, Cells were cultured in media containing 100 nmol/L AZD1775 at 3 hours prior to radiation with 0 (no IR), 2, 4, 6, and 8 Gy doses, followed by radiation clonogenic survival assay. Each dose was prepared in triplicate per experiment, and no less than two experiments were performed per cell line. DER at 2 Gy were compared between vehicle and AZD1755 (, P < 0.05; , P < 0.01).

OF4 Clin Cancer Res; 2020 CLINICAL CANCER RESEARCH

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-3373

Targeting Wee1 for Radiosensitization of Esophageal Cancer

Figure 3.

AZD1775 inhibition of Wee1 abrogates radiation-induced G2–M cell-cycle arrest. A, FLO1 and OE33 cells were cultured in media containing 100 nmol/L AZD1775 (AZD-100) at 3 hours prior to radiation with 0 (ctr) or 4 Gy doses. Twenty-four hours after 4 Gy or sham radiation, cells were prepared for flow cytometry analysis of cell-cycle distribution. Each dose was prepared in triplicate per experiment, and no less than two experiments were performed per cell line. Note AZD1775 significantly reduced G2–M phase fractions after 4 Gy radiation ( , P < 0.001). B, FLO1 cells were cultured in media containing 100 nmol/L AZD1775 (AZD-100) for 3 hours prior to radiation with 0 or 4 Gy doses. At the indicated time points following radiation (1, 4, 16, and 24 hours), the cells were lysed and subjected to immunoblotting with GAPDH as loading control.

leads to phosphorylation of H2AX. These results suggest that Wee1 inhibition attenuates DNA damage repair during – AZD1775 inhibition of Wee1 abrogates radiation induced G2 M fractionated radiation – cell-cycle checkpoint arrest by promoting CDK1 activity and increas- We found that AZD1775 attenuates radiation-induced G2 M phase ing replication stress, thereby potentiating radiation-mediated DNA arrest, enhances radiation mediated DNA damage, causes premature damage in esophageal cancer cells. entrance into mitosis, and finally leads to mitotic cell death. To further support our observations with more clinically relevant doses of Wee1 inhibitor enhances radiation-induced mitotic cell death radiation (i.e., 2 Gy per day), we extended our study to investigate Premature entrance into mitosis with unrepaired DNA lesions the capacity of AZD1775 for radiosensitization using standard frac- (particularly DSBs) leads to lethal consequences in cells. The abro- tionated radiation doses (i.e., 2 Gy per fraction) using colony formation gation of G2–M phase cell-cycle arrest and enhancement of DNA assays. In the single fraction ionizing radiation experiments, FLO1 and damage by AZD1775 in ESCA cells treated with radiation suggests that OE33 cells were pretreated with 100 nmol/L AZD1775 or vehicle AZD1775 can promote irradiated ESCA cells to prematurely enter into control for 3 hours, followed by treatment with a single radiation dose cell mitosis before completion of DNA repair. To test this hypothesis, of 4 Gy. In the fractionated ionizing radiation experiment, the cells FLO1 and OE33 cells were cultured on cover slides, and treated with were pretreated with 100 nmol/L AZD1775 or vehicle control for vehicle, AZD1775, 4 Gy radiation, or the combination of AZD1775 for 3 hours, followed by treatment with 2 Gy, which was repeated 24 hours 3 hours followed by 4 Gy. After 72 hours, the cover slides were later (Fig. 5A). Twenty-four hours postradiation, the cells were collected, and stained with DAPI and tubulin by immunofluorescence. cultured in fresh medium without AZD1775/vehicle for 10 additional Mitotic cell death was determined by the number of cells demonstrat- days before colony fixation and staining. Cell recovery rate was ing mitotic catastrophe (multinuclear cells with more than two nuclear calculated by dividing the percentage of colonies formed following lobes, or cells with several micronuclei; Fig. 4A). In comparison to fractionated radiation (2 Gy 2 fractions) by the percentage of vehicle control, AZD1775 alone did not induce mitotic catastrophe. colonies formed following a single fraction of 4 Gy. As expected, 2 Conversely, 4 Gy radiation treatment resulted in accumulation of cells Gy 2 led to a higher surviving fraction compared with a single 4 Gy experiencing mitotic catastrophe, which was significantly enhanced by dose of radiation, likely due to sublethal DNA repair occurring AZD1775 in both cell lines (Fig. 4B and C). Thus, the combination of between fractions, leading to a survival enhancement rate of 1.37 and AZD1775 with radiation resulted in a significantly higher incidence of 1.84 for FLO1 and OE33 cells, respectively. Similar to our previous data mitotic catastrophe than radiation treatment alone. with single fraction radiation in Fig. 2B, exposure of FLO1 or OE33

AACRJournals.org Clin Cancer Res; 2020 OF5

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-3373

Yang et al.

Figure 4. AZD1775 enhanced radiation-induced mitotic catastrophe in ESCA cells. A, FLO1 and OE33 cells were treated with 4 Gy radiation, 100 nmol/L AZD1775 alone, or in combination (100 nmol/L AZD1775 added 3 hours prior to 4 Gy radiation). At 72 hours postradiation, the cells were subjected to immunofluorescence staining of tubulin and DAPI to show the signs of mitotic catastrophe (micro- and multinucleated cells as shown by red arrows). Representative images of mitotic catastrophe in OE33 cells are shown. The graphs show percentage of mitotic catastrophe cells in 100 counted FLO1 (B) and OE33 (C) cells (, P < 0.001).

cells to AZD1775 before and during fractionated radiation still effec- tent with our in vitro data, the combination of AZD1775 and IR tively radiosensitized esophageal cancer cells and abrogated cell recov- significantly increased mitotic catastrophe in tumor xenografts com- ery (Fig. 5B). These results indicate a comparable enhancement of cell pare to IR treatment alone (Fig. 6D). Moreover, the majority of FLO1 death by AZD1775 in esophageal cancer cells when combining and OE33 tumors showed no evidence of tumor recurrence after AZD1775 with either fractionated or single fraction radiation. These treatment with AZD1775 in combination with radiation (survival findings have important clinical implications as patients with esoph- curves shown in Supplementary Fig. S3). In terms of toxicity, mice ageal cancer are treated with fractionated radiation to typical doses of tolerated the treatment well, and no mice died early from treatment 1.8 to 2.0 Gy/day. toxicity. Mice who received IR treatment did lose weight during the first week, but fully recovered within 2 weeks (Supplementary Fig. S4). Wee1 inhibition markedly radiosensitizes esophageal cancer We further performed immunoblotting of the tumor lysates 2 hours cells in mouse tumor xenografts after Day 3 treatment in each group to assess pharmacodynamics To determine if Wee1 inhibition could effectively radiosensitize effects of AZD1775. AZD1775 reduced the phosphorylation of ESCA cells in vivo, we further explored the combined treatment of Wee1 and CDK1 as well as the protein levels of cyclin A2, B1, AZD1775 and radiation in vivo using nude mice xenografts with FLO1 E1, and E2, while increasing levels of phospho-histone H3 (a marker and OE33 cells. When tumors reached 100 to 150 mm3, the mice were of mitotic cells; Fig. 6E). Taken together, these findings indicate – randomized to groups of treatment with vehicle, AZD1775 alone, 4 Gy AZD1775 is promoting G2 M phase cell-cycle progression. Radi- radiation alone, or the combination of AZD1775 þ 4 Gy (mice were ation increased the phosphorylation of Wee1 and CDK1, as well as treated with AZD1775 2 hours before radiation). The treatments lasted the protein levels of cyclin A2, B1, E1, and E2, which were all for 5 consecutive days during Days 1–5(Fig. 6A). AZD1775 was prevented by Wee1 inhibition. These results demonstrate that delivered by oral gavage with a dose of 50 mg/kg, twice a day as potent inhibition of Wee1 leads to promotion of cells through previously described (29). AZD1775 monotherapy and radiation alone mitosis despite DNA damage from radiation, thus leading to resulted in partial tumor growth delay. However, AZD1775 in com- marked radiosensitization by AZD1775 in vivo. bination with radiation treatment led to remarkable and sustained tumor regression of both FLO1 and OE33 xenografts (Fig. 6B and C). To investigate whether the treatment combination is indeed inducing Discussion tumor cell death through mitotic catastrophe, we performed mitotic Radiotherapy induces DNA damage, resulting in activation of catastrophe assay in tumors derived from mouse xenografts. Consis- apoptotic pathways or inducing postmitotic death due to unrepaired

OF6 Clin Cancer Res; 2020 CLINICAL CANCER RESEARCH

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-3373

Targeting Wee1 for Radiosensitization of Esophageal Cancer

Figure 5. AZD1775 effectively radiosensitizes ESCA cells during fractionated radiation. A, Schema of two radiation treatment schedules for clonogenic assay. B, AZD1775 sensitized FLO1 and OE33 cells to radiation whether cells were treated with fractionated radiation (2 Gy 2) or single fraction radiation (4 Gy). IR, ionizing radiation; w/o, without; AZD, AZD1775 (100 nmol/L; , P < 0.05).

DNA damage. Following DNA damage, cells rely on cell-cycle check- shortage, and subsequent double-strand DNA breaks (42). It has been points to provide time for DNA repair prior to cell division. Esophageal proposed that targeting Wee1 kinase is a promising strategy for the cancer cells often lack a functional G1 checkpoint due to a high radiosensitization and chemosensitization of cancer cells with a defec- TP53 frequency of mutations. On the basis of TCGA data, up to tive G1 cell-cycle checkpoint (21). Accordingly, several Wee1 kinase 91% of squamous cell carcinoma and 71% of adenocarcinoma esoph- small molecule inhibitors, including AZD1775, have been devel- ageal cancers possess a TP53 mutation making them heavily dependent oped (27). Previous studies have shown that AZD1775 is a potent on the G2–M checkpoint to survive DNA damage and replication and selective small molecule inhibitor of Wee1 kinase and has been stress. In this study, we demonstrated that a potent Wee1 kinase shown to sensitize tumor cells to both chemotherapy and radia- inhibitor AZD1775 sensitized ESCA cells to radiation therapy in tion (27, 28, 33). In this study, we found that both CDK1 and WEE1 in vitro cell cultures and mouse tumor xenografts. In addition, are overexpressed in ESCA, as well as genes associated with an AZD1775 treatment led to a comparable enhancement of cytotoxicity AZD1775 responsiveness gene signature (39). Moreover, we demon- in esophageal cancer cells treated with either fractionated radiation or strated that AZD1775 potently inhibited the phosphorylation of both single dose radiation. Mechanistically, AZD1775 attenuated radiation- Wee1 and CDK1 in the absence or presence of radiation. Consistent – – induced G2 M phase arrest, which was accompanied by enhanced with the role for Wee1 in G2 M checkpoint regulation, AZD1775 fi – radiation-induced mitotic catastrophe and DNA damage. Our ndings prevented IR-induced G2 M phase cell-cycle arrest, which was accom- suggest that Wee1 kinase specific inhibitor AZD1775 is an effective panied with enhanced mitotic catastrophe and gH2AX, indicative of radiosensitizer for esophageal cancer. enhanced cell death and DNA damage. Interestingly, we noted Wee1 is a tyrosine kinase and is activated following DNA damage. AZD1775 caused reductions in E, A, and B-type . This obser- – Wee1 kinase is a critical regulator of the G2 M checkpoint and thus vation is consistent with our knowledge of the temporal expression of genomic stability by mediating inhibitory phosphorylation of CDK1, cyclins during the cell cycle. Specifically, E-type cyclins are upregulated – resulting in cell-cycle arrest and permitting DNA repair prior to during the G1 transition and then fall down, whereas A-type proceeding with mitosis (40, 41). It has been shown that inhibiting cyclins are upregulated in the and then fall down prior to and Wee1 results in replication stress, loss of genomic integrity, nucleotide during entry of calls into mitosis. Finally, B-type cyclins are

AACRJournals.org Clin Cancer Res; 2020 OF7

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-3373

Yang et al.

A -Day 14 Day 1 Day 5 Day 52–90 D FLO1 70 60 50 Tumor cell injection 5 days treatment Study end 20 B FLO1 cells Vehicle 10 400.00% AZD1775 ALONE Vehicle + IR 0

300.00% % Cell with mitotic catastrophe AZD1775 + IR Ctr IR

AZD1775 200.00% AZD1775 + IR

100.00% E Vehicle AZD1775 4 Gy 4 Gy + AZD1775 % Volume change % Volume 0.00% 122 12121 15 10 151924295485 p-WEE (S642) −100.00% Days p-CDK1 (Y15)

C 1400.00% Cyclin A2 OE33 cells 1200.00% Vehicle Cyclin B1 1000.00% AZD1775 ALONE Vehicle + IR 800.00% Cyclin E1 AZD1775 + IR 600.00% Cyclin E2 400.00% % Volume change % Volume 200.00% p-HH3 (S10)

0.00% GAPDH 15 10 151924293338 43 47 52 Days

Figure 6. Wee1 inhibition effectively radiosensitizes esophageal carcinoma cells in xenograft tumor models. Mice were injected with 2 106 FLO1 or OE33 cells, and randomized to start treatment once tumors reached 100 to 200 mm3. AZD1775 was administered via oral gavage at 50 mg/kg twice a day for 5 days (control group received vehicle at same intervals), 2 hours prior to radiation when radiation was given. Radiation was administered at 4 Gy daily for 5 consecutive days. Tumor size was calculated by measuring length and width via calipers. A, Schema of in vivo experimental plan using tumor xenografts in mice. B, Growth curves of FLO1 xenograft tumors. C, Growth curves of OE33 xenograft tumors. D and E, FLO1 tumor xenografts were isolated from mice on Day 3 (2 hours after treatment completed) and subjected to mitotic catastrophe assay (D) or immunoblotting analysis (E) of the indicated proteins with GAPDH as loading control. HH3, histone H3; Ctr, control (vehicle; , P < 0.05).

upregulated at the G2–M transition then fall down sharply upon entry tion (45, 46). Overall, approximately 30% of patients with esophageal to mitosis. Taken together, AZD1775 potently inhibits Wee1 in cancer will demonstrate a pathologic complete response (pCR) at the esophageal cancer cells, thereby promoting entry from S and G2 time of surgery (46–48) with pCR rates closer to 43% to 49% for through M phase, and thus preventing Wee1 from protecting esoph- squamous cell cancer (46, 49) and 16% to 25% for adenocarcino- ageal cancer cells from the effects of radiotherapy. ma (46, 50, 51). A pCR is associated with an improvement in overall Esophageal cancer remains a global problem and is the eighth most survival (47, 48). Patients with a pCR, arguably, may be able to avoid common cancer worldwide (1). Globally, although esophageal squa- esophagectomy without compromising survival (52). Esophagectomy mous cell carcinoma remains the predominant histology in Asia, is associated with impaired quality of life and remains a morbid Africa, and South America, the incidence of esophageal adenocarci- operation with substantial mortality (53–57). With current pCR rates noma has surpassed that of squamous cell carcinoma in North at or below 50%, improvements in therapy, including the potential America, Australia, and Europe. Current predictions are that by addition of radiosensitizing agents, may help to make organ preser- 2030, up to 1 in 100 men will be diagnosed with esophageal adeno- vation a reality in esophageal cancer. carcinoma during their lifetime in the Netherlands and the United Currently, even with the most aggressive approach of trimodality Kingdom (43). In the United States, the incidence of cancers of the therapy, approximately 50% of patients will experience a locoregional esophagus and gastroesophageal junction has increased dramatically and/or distant recurrence within 5 years of treatment completion (45). in recent decades, largely driven by the rising incidence of adenocar- Therefore, there is an urgent need to develop novel strategies that will cinoma (44). Nearly 40% to 50% of patients present with either improve clinical outcomes of patients diagnosed with localized esoph- unresectable disease or evidence of distant metastasis (44). For patients ageal cancer. Strategies aimed at improving local control, including with resectable disease, the current standard of care includes admin- modalities such as radiation therapy, will likely have an impact on istering chemotherapy, most commonly carboplatin and paclitaxel, improving survival. Our results showed that AZD1775 in combination concurrently with radiation therapy followed by surgical resec- with radiotherapy resulted in virtually complete tumor regression of

OF8 Clin Cancer Res; 2020 CLINICAL CANCER RESEARCH

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-3373

Targeting Wee1 for Radiosensitization of Esophageal Cancer

esophageal cancer tumor xenografts. Therefore, our study suggests with multiple chemotherapy regimens with preliminary data showing that AZD1775 inhibition of Wee1 is an effective strategy for radiation that combination therapy appears to be reasonably tolerated (61–64). sensitization in esophageal cancer cells. Interestingly, we observed that Because of the high incidence of TP53 mutations and the dependence – the DER of SK4 was much higher than that of the other three cell lines on the Wee1-mediated G2 M checkpoint to survive DNA damage, (Fig. 2B). All the four esophageal cancer cell lines used in this study esophageal cancer is a logical site to consider the addition of a Wee1 have TP53 mutations. However, besides TP53 mutation, SK4 cells have kinase inhibitor to standard neoadjuvant therapy. We believe our additional KRAS and PIK3CA mutation (https://portals.broadinstitute. study warrants a phase I trial testing AZD1775 in combination with org/ccle). KRAS and PIK3CA mutations drive tumorigenesis via radiation or chemoradiation for esophageal cancer. multiple mechanisms, one of which is to induce DNA replication stress leading to genomic instability (58, 59). As mentioned, Wee1 has Disclosure of Potential Conflicts of Interest an important role in replication stress response (35). Thus, esophageal E.D. Miller reports receiving other remuneration from a CTEP-approved career cancer cells with KRAS and PIK3CA mutations might may have development LOI combining AZD1775 with radiation therapy for inoperable or become more dependent on Wee1 kinase for survival due to increased metastatic esophageal cancer. S.H. Lin reports receiving commercial research grants from Beyond Spring Pharmaceuticals, Genentech, and Hitachi Chemical Diagnostic, replication stress. It will be important to determine whether esophageal speakers bureau honoraria from AstraZeneca and Varian Medical Systems, holds KRAS PIK3CA cancer with and/or mutations are hypersensitive to ownership interest (including patents) in STCube Pharmaceuticals, and is an advisory Wee1 inhibitors in future preclinical and clinical studies. In addition, it board member/unpaid consultant for AstraZeneca and Beyond Spring Pharmaceu- will be critical to determine whether TP53 mutant status confers ticals. No potential conflicts of interest were disclosed by the other authors. increased sensitivity to AZD1775 and radiation. In our preliminary studies, we found that the combination of AZD1775 and radiation did Authors’ Contributions not radiosensitize TP53 intact AGS adenocarcinoma cells (Supple- Conception and design: L. Yang, C. Shen, J. Zhang, T.M. Williams mentary Fig. S5). Finally, further studies are needed to assess whether Development of methodology: L. Yang, C. Shen, S.H. Lin, T.M. Williams Acquisition of data (provided animals, acquired and managed patients, provided AZD1755 sensitizes esophageal cancer cells to DNA-damaging che- facilities, etc.): L. Yang, C.J. Pettit, T. Li, A.J. Hu, T.M. Williams motherapies such as platinum-based drugs or other chemotherapeu- Analysis and interpretation of data (e.g., statistical analysis, biostatistics, tics which promote replication stress. computational analysis): L. Yang, C. Shen, C.J. Pettit, T. Li, E.D. Miller, S.H. Lin, On the basis of our results, we feel that our data support a clinical T.M. Williams trial testing the combination of Wee1 kinase inhibitor and radiation for Writing, review, and/or revision of the manuscript: L. Yang, C. Shen, C.J. Pettit, esophageal cancer. It would be interesting to also explore combining E.D. Miller, S.H. Lin, T.M. Williams Administrative, technical, or material support (i.e., reporting or organizing data, Wee1 inhibitor with chemoradiation, or replacing a chemotherapy constructing databases): J. Zhang drug commonly used in esophageal cancer with a Wee1 kinase inhibitor. Standard chemotherapy regimens used in combination with Acknowledgments radiation for esophageal cancer include paclitaxel and carboplatin, or This work was supported by the following grants: The Ohio State University 5FU and oxaliplatin. Recently, a phase II trial of AZD1775 in com- Comprehensive Cancer Center (OSU-CCC), NIH (P30 CA016058 and R01 bination with gemcitabine and radiation was published for pancreatic CA198128) and National Center for Advancing Translational Sciences cancer, showing tolerable safety profile of the combination and (KL2TR001068). These data were presented in part at the American Association of significant efficacy compared with historical controls (60). This trial Cancer Research (AACR) annual meeting 2018 and Radiation Research Society annual meeting 2019. Research reported in this publication was supported by The was based on preclinical data combining Wee1 inhibitor with gemci- Ohio State University Comprehensive Cancer Center (OSU-CCC) and the National tabine and radiation. Thus, additional preclinical studies may be Institutes of Health under grant number P30 CA016058, as well as RSG-17-221-01- needed to establish the optimal combination of Wee1 inhibitor with TBG (to T.M. Williams), award number grant KL2TR001068 from the National certain chemotherapy drugs and radiation for the treatment of esoph- Center for Advancing Translational Sciences (to T.M. Williams), and NIH grant R01 ageal cancer. CA198128 (to T.M. Williams). The content is solely the responsibility of the authors fi In summary, our results demonstrated a potent inhibitory role for and does not necessarily represent the of cial views of the NIH. Wee1 kinase inhibitor AZD1775 in cell-cycle checkpoints in response The costs of publication of this article were defrayed in part by the payment of page to IR-induced DNA double-strand breaks; importantly, AZD1775 charges. This article must therefore be hereby marked advertisement in accordance – in vitro enhanced IR mediated cell death and maintained the sup- with 18 U.S.C. Section 1734 solely to indicate this fact. pression of esophageal cancer mouse xenografts by radiotherapy in vivo. There are several phase I and II studies which have been Received October 14, 2019; revised February 20, 2020; accepted March 24, 2020; performed using AZD1775 both as monotherapy and in combination published first March 27, 2020.

References 1. Lagergren J, Smyth E, Cunningham D, Lagergren P. Oesophageal cancer. Lancet 6. Otto T, Sicinski P. Cell cycle proteins as promising targets in cancer therapy. 2017;390:2383–96. Nat Rev Cancer 2017;17:93–115. 2. van Rossum PSN, Mohammad NH, Vleggaar FP, van Hillegersberg R. Treatment 7. Jeggo PA, Pearl LH, Carr AM. DNA repair, genome stability and cancer: a for unresectable or metastatic oesophageal cancer: current evidence and trends. historical perspective. Nat Rev Cancer 2016;16:35–42. Nat Rev Gastroenterol Hepatol 2018;15:235–49. 8. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011; 3. Pennathur A, Gibson MK, Jobe BA, Luketich JD. Oesophageal carcinoma. Lancet 144:646–74. 2013;381:400–12. 9. Ciccia A, Elledge SJ. The DNA damage response: making it safe to play with 4. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin 2018;68: knives. Mol Cell 2010;40:179–204. 7–30. 10. Castedo M, Perfettini JL, Roumier T, Andreau K, Medema R, Kroemer G. Cell 5. Smyth EC, Lagergren J, Fitzgerald RC, Lordick F, Shah MA, Lagergren P, et al. death by mitotic catastrophe: a molecular definition. Oncogene 2004;23: Oesophageal cancer. Nat Rev Dis Primers 2017;3:17048. 2825–37.

AACRJournals.org Clin Cancer Res; 2020 OF9

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-3373

Yang et al.

11. Rundle S, Bradbury A, Drew Y, Curtin NJ. Targeting the ATR-CHK1 axis in 35. Geenen JJJ, Schellens JHM. Molecular pathways: targeting the protein kinase cancer therapy. Cancers 2017;9. doi: 10.3390/cancers9050041. Wee1 in cancer. Clin Cancer Res 2017;23:4540–4. 12. Goldstein M, Kastan MB. The DNA damage response: implications for tumor 36. Williams TM, Flecha AR, Keller P, Ram A, Karnak D, Galban S, et al. Cotargeting responses to radiation and chemotherapy. Annu Rev Med 2015;66:129–43. MAPK and PI3K signaling with concurrent radiotherapy as a strategy for the 13. Blackford AN, Jackson SP. ATM, ATR, and DNA-PK: the trinity at the heart of treatment of pancreatic cancer. Mol Cancer Ther 2012;11:1193–202. the DNA damage response. Mol Cell 2017;66:801–17. 37. Estrada-Bernal A, Chatterjee M, Haque SJ, Yang L, Morgan MA, Kotian S, et al. 14. Parker LL, Piwnica-Worms H. Inactivation of the p34cdc2- complex by MEK inhibitor GSK1120212-mediated radiosensitization of pancreatic cancer the human WEE1 tyrosine kinase. Science 1992;257:1955–7. cells involves inhibition of DNA double-strand break repair pathways. Cell Cycle 15. Bartek J, Lukas J. Chk1 and Chk2 kinases in checkpoint control and cancer. 2015;14:3713–24. Cancer Cell 2003;3:421–9. 38. Fertil B, Dertinger H, Courdi A, Malaise EP. Mean inactivation dose: a useful 16. Masaki T, Shiratori Y, Rengifo W, Igarashi K, Yamagata M, Kurokohchi K, et al. concept for intercomparison of human cell survival curves. Radiat Res 1984;99: Cyclins and cyclin-dependent kinases: comparative study of hepatocellular 73–84. carcinoma versus cirrhosis. Hepatology 2003;37:534–43. 39. Mizuarai S, Yamanaka K, Itadani H, Arai T, Nishibata T, Hirai H, et al. Discovery 17. Iorns E, Lord CJ, Grigoriadis A, McDonald S, Fenwick K, Mackay A, et al. of gene expression-based pharmacodynamic biomarker for a p53 context- Integrated functional, gene expression and genomic analysis for the identifica- specific anti-tumor drug Wee1 inhibitor. Mol Cancer 2009;8:34. tion of cancer targets. PLoS One 2009;4:e5120. 40. Dominguez-Kelly R, Martin Y, Koundrioukoff S, Tanenbaum ME, Smits VAJ, 18. Mir SE, De Witt Hamer PC, Krawczyk PM, Balaj L, Claes A, Niers JM, et al. In Medema RH, et al. Wee1 controls genomic stability during replication by silico analysis of kinase expression identifies WEE1 as a gatekeeper against regulating the Mus81-Eme1 endonuclease. J Cell Biol 2011;194:567–79. mitotic catastrophe in glioblastoma. Cancer Cell 2010;18:244–57. 41. Watanabe N, Broome M, Hunter T. Regulation of the human Wee1hu Cdk 19. Benada J, Macurek L. Targeting the checkpoint to kill cancer cells. Biomolecules tyrosine 15-kinase during the cell-cycle. EMBO J 1995;14:1878–91. 2015;5:1912–37. 42. Beck H, Nahse-Kumpf V, Larsen MS, O'Hanlon KA, Patzke S, Holmberg C, et al. 20. Do K, Doroshow JH, Kummar S. Wee1 kinase as a target for cancer therapy. Cyclin-dependent kinase suppression by WEE1 kinase protects the genome Cell Cycle 2013;12:3159–64. through control of replication initiation and nucleotide consumption. Mol Cell 21. Matheson CJ, Backos DS, Reigan P. Targeting WEE1 kinase in cancer. Biol 2012;32:4226–36. Trends Pharmacol Sci 2016;37:872–81. 43. Arnold M, Laversanne M, Brown LM, Devesa SS, Bray F. Predicting the future 22. Song Y, Li L, Ou Y, Gao Z, Li E, Li X, et al. Identification of genomic alterations in burden of esophageal cancer by histological subtype: international trends in oesophageal squamous cell cancer. Nature 2014;509:91–5. incidence up to 2030. Am J Gastroenterol 2017;112:1247–55. 23. Dulak AM, Stojanov P, Peng S, Lawrence MS, Fox C, Stewart C, et al. Exome 44. Thrift AP. The epidemic of oesophageal carcinoma: where are we now? and whole-genome sequencing of esophageal adenocarcinoma identifies Cancer Epidemiol 2016;41:88–95. recurrent driver events and mutational complexity. Nat Genet 2013;45: 45. Shapiro J, van Lanschot JJB, Hulshof M, van Hagen P, van Berge Henegouwen 478–86. MI, Wijnhoven BPL, et al. Neoadjuvant chemoradiotherapy plus surgery versus 24. Kim J, Bowlby R, Mungall AJ, Robertson AG, Odze RD, Cherniack AD, et al. surgery alone for oesophageal or junctional cancer (CROSS): long-term results of Integrated genomic characterization of oesophageal carcinoma. Nature 2017; a randomised controlled trial. Lancet Oncol 2015;16:1090–8. 541:169. 46. van Hagen P, Hulshof MC, van Lanschot JJ, Steyerberg EW, van Berge Hene- 25. Leijen S, Beijnen JH, Schellens JH. Abrogation of the G2 checkpoint by inhibition gouwen MI, Wijnhoven BP, et al. Preoperative chemoradiotherapy for esoph- of Wee-1 kinase results in sensitization of p53-deficient tumor cells to DNA- ageal or junctional cancer. N Engl J Med 2012;366:2074–84. damaging agents. Curr Clin Pharmacol 2010;5:186–91. 47. Alnaji RM, Du W, Gabriel E, Singla S, Attwood K, Nava H, et al. Pathologic 26. Ku BM, Bae YH, Koh J, Sun JM, Lee SH, Ahn JS, et al. Mutational status of TP53 complete response is an independent predictor of improved survival following defines the efficacy of Wee1 inhibitor AZD1775 in KRAS-mutant non-small cell neoadjuvant chemoradiation for esophageal adenocarcinoma. J Gastrointest lung cancer. Oncotarget 2017;8:67526–37. Surg 2016;20:1541–6. 27. Hirai H, Iwasawa Y, Okada M, Arai T, Nishibata T, Kobayashi M, et al. Small- 48. Donahue JM, Nichols FC, Li Z, Schomas DA, Allen MS, Cassivi SD, et al. molecule inhibition of Wee1 kinase by MK-1775 selectively sensitizes p53- Complete pathologic response after neoadjuvant chemoradiotherapy for deficient tumor cells to DNA-damaging agents. Mol Cancer Ther 2009;8:2992– esophageal cancer is associated with enhanced survival. Ann Thorac Surg 3000. 2009;87:392–8. 28. Bridges KA, Hirai H, Buser CA, Brooks C, Liu H, Buchholz TA, et al. MK-1775, a 49. Yang H, Liu H, Chen Y, Zhu C, Fang W, Yu Z, et al. Neoadjuvant chemor- novel Wee1 kinase inhibitor, radiosensitizes p53-defective human tumor cells. adiotherapy followed by surgery versus surgery alone for locally advanced Clin Cancer Res 2011;17:5638–48. squamous cell carcinoma of the esophagus (NEOCRTEC5010): a phase III 29. Rajeshkumar NV, De Oliveira E, Ottenhof N, Watters J, Brooks D, Demuth T, multicenter, randomized, open-label clinical trial. J Clin Oncol 2018;36:2796–803. et al. MK-1775, a potent Wee1 inhibitor, synergizes with gemcitabine to achieve 50. Stahl M, Walz MK, Stuschke M, Lehmann N, Meyer HJ, Riera-Knorrenschild J, tumor regressions, selectively in p53-deficient pancreatic cancer xenografts. et al. Phase III comparison of preoperative chemotherapy compared with Clin Cancer Res 2011;17:2799–806. chemoradiotherapy in patients with locally advanced adenocarcinoma of the 30. Cuneo KC, Morgan MA, Davis MA, Parcels LA, Parcels J, Karnak D, et al. Wee1 esophagogastric junction. J Clin Oncol 2009;27:851–6. kinase inhibitor AZD1775 radiosensitizes hepatocellular carcinoma regardless of 51. Walsh TN, Noonan N, Hollywood D, Kelly A, Keeling N, Hennessy TP. A TP53 mutational status through induction of replication stress. Int J Radiat comparison of multimodal therapy and surgery for esophageal adenocarcinoma. Oncol Biol Phys 2016;95:782–90. N Engl J Med 1996;335:462–7. 31. Parsels LA, Karnak D, Parsels JD, Zhang Q, Velez-Padilla J, Reichert ZR, 52. Borggreve AS, Mook S, Verheij M, Mul VEM, Bergman JJ, Bartels-Rutten A, et al. et al. PARP1 trapping and DNA replication stress enhance radiosensitiza- Preoperative image-guided identification of response to neoadjuvant chemor- tion with combined WEE1 and PARP inhibitors. Mol Cancer Res 2018;16: adiotherapy in esophageal cancer (PRIDE): a multicenter observational study. 222–32. BMC Cancer 2018;18:1006. 32. Pfister SX, Markkanen E, Jiang Y, Sarkar S, Woodcock M, Orlando G, et al. 53. Busweiler LA, Wijnhoven BP, van Berge Henegouwen MI, Henneman D, van Inhibiting WEE1 selectively kills histone H3K36me3-deficient cancers by dNTP Grieken NC, Wouters MW, et al. Early outcomes from the Dutch Upper starvation. Cancer Cell 2015;28:557–68. Gastrointestinal Cancer Audit. Br J Surg 2016;103:1855–63. 33. Kausar T, Schreiber JS, Karnak D, Parsels LA, Parsels JD, Davis MA, et al. 54. Djarv T, Lagergren J, Blazeby JM, Lagergren P. Long-term health-related quality Sensitization of pancreatic cancers to gemcitabine chemoradiation by WEE1 of life following surgery for oesophageal cancer. Br J Surg 2008;95:1121–6. kinase inhibition depends on homologous recombination repair. Neoplasia 55. Kassis ES, Kosinski AS, Ross P Jr, Koppes KE, Donahue JM, Daniel VC. Predictors 2015;17:757–66. of anastomotic leak after esophagectomy: an analysis of the Society of Thoracic 34. Nam AR, Park JE, Bang JH, Jin MH, Bang YJ, Oh DY. DNA damage response Surgeons general thoracic database. Ann Thorac Surg 2013;96:1919–26. (DDR)-targeting strategy by targeting WEE1 and or ATM/ATR works in biliary 56. Mc Cormack O, Zaborowski A, King S, Healy L, Daly C, O'Farrell N, et al. New- tract cancer [abstract]. In: Proceedings of the American Association for Cancer onset atrial fibrillation post-surgery for esophageal and junctional cancer: Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): incidence, management, and impact on short- and long-term outcomes. AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 323. Ann Surg 2014;260:772–8.

OF10 Clin Cancer Res; 2020 CLINICAL CANCER RESEARCH

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-3373

Targeting Wee1 for Radiosensitization of Esophageal Cancer

57. Schandl A, Lagergren J, Johar A, Lagergren P. Health-related quality of life 62. Leijen S, van Geel RM, Pavlick AC, Tibes R, Rosen L, Razak AR, et al. Phase I 10 years after oesophageal cancer surgery. Eur J Cancer 2016;69:43–50. study evaluating WEE1 inhibitor AZD1775 as monotherapy and in combination 58. Forment JV, O'Connor MJ. Targeting the replication stress response in cancer. with gemcitabine, cisplatin, or carboplatin in patients with advanced solid Pharmacol Therapeut 2018;188:155–67. tumors. J Clin Oncol 2016;34:4371–80. 59. Macheret M, Halazonetis TD. DNA replication stress as a hallmark of cancer. 63. Leijen S, van Geel RM, Sonke GS, de Jong D, Rosenberg EH, Annu Rev Pathol 2015;10:425–48. Marchetti S, et al. Phase II study of WEE1 inhibitor AZD1775 plus 60. Cuneo KC, Morgan MA, Sahai V, Schipper MJ, Parsels LA, Parsels JD, et al. Dose carboplatin in patients with TP53-mutated ovarian cancer refractory or escalation trial of the Wee1 inhibitor adavosertib (AZD1775) in combination resistant to first-line therapy within 3 months. J Clin Oncol 2016;34: with gemcitabine and radiation for patients with locally advanced pancreatic 4354–61. cancer. J Clin Oncol 2019;37:2643–50. 64. Mendez E, Rodriguez CP, Kao MC, Raju S, Diab A, Harbison RA, et al. A phase I 61. Do K, Wilsker D, Ji J, Zlott J, Freshwater T, Kinders RJ, et al. Phase I study of clinical trial of AZD1775 in combination with neoadjuvant weekly docetaxel and single-agent AZD1775 (MK-1775), a Wee1 kinase inhibitor, in patients with cisplatin before definitive therapy in head and neck squamous cell carcinoma. refractory solid tumors. J Clin Oncol 2015;33:3409–15. Clin Cancer Res 2018;24:2740–8.

AACRJournals.org Clin Cancer Res; 2020 OF11

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-3373

Wee1 Kinase Inhibitor AZD1775 Effectively Sensitizes Esophageal Cancer to Radiotherapy

Linlin Yang, Changxian Shen, Cory J. Pettit, et al.

Clin Cancer Res Published OnlineFirst March 27, 2020.

Updated version Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-19-3373

Supplementary Access the most recent supplemental material at: Material http://clincancerres.aacrjournals.org/content/suppl/2020/03/27/1078-0432.CCR-19-3373.DC1

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://clincancerres.aacrjournals.org/content/early/2020/05/24/1078-0432.CCR-19-3373. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research.