1,361
Views
2
CrossRef citations to date
0
Altmetric
Review

Strategic development of AZD1775, a Wee1 kinase inhibitor, for cancer therapy

, , &
Pages 741-751 | Received 20 May 2018, Accepted 10 Aug 2018, Published online: 21 Aug 2018

References

  • Otto T, Sicinski P. Cell cycle proteins as promising targets in cancer therapy. Nat Rev Cancer. 2017;17:93–115.
  • Ceccaldi R, Liu JC, Amunugama R, et al. Homologous-recombination-deficient tumours are dependent on Poltheta-mediated repair. Nature. 2015;518:258–262.
  • Patch AM, Christie EL, Etemadmoghadam D, et al. Whole-genome characterization of chemoresistant ovarian cancer. Nature. 2015;521:489–494.
  • Forment JV, O’Connor MJ. Targeting the replication stress response in cancer. Pharmacol Ther. 2018;188:155–167.
  • Russell P, Nurse P. Negative regulation of mitosis by wee1+, a gene encoding a protein kinase homolog. Cell. 1987;49:559–567. .
  • Aarts M, Sharpe R, Garcia-Murillas I, et al. Forced mitotic entry of S-phase cells as a therapeutic strategy induced by inhibition of WEE1. Cancer Discov. 2012;2:524–539.
  • Kogiso T, Nagahara H, Hashimoto E, et al. Efficient induction of apoptosis by wee1 kinase inhibition in hepatocellular carcinoma cells. PLoS One. 2014;9:e100495.
  • Mir SE, De Witt Hamer PC, Krawczyk PM, et al. In silico analysis of kinase expression identifies WEE1 as a gatekeeper against mitotic catastrophe in glioblastoma. Cancer Cell. 2010;18:244–257.
  • Harris PS, Venkataraman S, Alimova I, et al. Integrated genomic analysis identifies the mitotic checkpoint kinase WEE1 as a novel therapeutic target in medulloblastoma. Mol Cancer. 2014;13:72.
  • Leary A, Auguste A, Mesnage S. DNA damage response as a therapeutic target in gynecological cancers. Curr Opin Oncol. 2016;28:404–411.
  • Abroudi A, Samarasinghe S, Kulasiri D. A comprehensive complex systems approach to the study and analysis of mammalian cell cycle control system in the presence of DNA damage stress. J Theor Biol. 2017;429:204–228.
  • Bassermann F, Frescas D, Guardavaccaro D, et al. The Cdc14B-Cdh1-Plk1 axis controls the G2 DNA-damage-response checkpoint. Cell. 2008;134:256–267.
  • Kato S, Schwaederle M, Daniels GA, et al. Cyclin-dependent kinase pathway aberrations in diverse malignancies: clinical and molecular characteristics. Cell Cycle. 2015;14:1252–1259.
  • Visconti R, Della Monica R, Grieco D. Cell cycle checkpoint in cancer: a therapeutically targetable double-edged sword. J Exp Clin Cancer Res. 2016;35:153.
  • Kanska J, Zakhour M, Taylor-Harding B, et al. Cyclin E as a potential therapeutic target in high grade serous ovarian cancer. Gynecol Oncol. 2016;143:152–158.
  • Hegarat N, Rata S, Hochegger H. Bistability of mitotic entry and exit switches during open mitosis in mammalian cells. Bioessays. 2016;38:627–643.
  • Boutros R, Dozier C, Ducommun B. The when and wheres of CDC25 phosphatases. Curr Opin Cell Biol. 2006;18:185–191.
  • Vassilopoulos A, Tominaga Y, Kim HS, et al. WEE1 murine deficiency induces hyper-activation of APC/C and results in genomic instability and carcinogenesis. Oncogene. 2015;34:3023–3035.
  • Liu JC, Granieri L, Shrestha M, et al. Identification of CDC25 as a Common Therapeutic Target for Triple-Negative Breast Cancer. Cell Rep. 2018;23:112–126.
  • Schmidt M, Rohe A, Platzer C, et al. Regulation of G2/M Transition by Inhibition of WEE1 and PKMYT1 Kinases. Molecules. 2017;22:2045. doi:10.3390/molecules22122045.
  • Moseley JB. Wee1 and Cdc25: tools, pathways, mechanisms, questions. Cell Cycle. 2017;16:599–600.
  • Lucena R, Alcaide-Gavilan M, Anastasia SD, et al. Wee1 and Cdc25 are controlled by conserved PP2A-dependent mechanisms in fission yeast. Cell Cycle. 2017;16:428–435.
  • Iwai A, Bourboulia D, Mollapour M, et al. Combined inhibition of Wee1 and Hsp90 activates intrinsic apoptosis in cancer cells. Cell Cycle. 2012;11:3649–3655.
  • O’Connell MJ, Raleigh JM, Verkade HM, et al. Chk1 is a wee1 kinase in the G2 DNA damage checkpoint inhibiting cdc2 by Y15 phosphorylation. EMBO J. 1997;16:545–554.
  • Lee J, Kumagai A, Dunphy WG. Positive regulation of Wee1 by Chk1 and 14–3–3 proteins. Mol Biol Cell. 2001;12:551–563.
  • Katayama K, Fujita N, Tsuruo T. Akt/protein kinase B-dependent phosphorylation and inactivation of WEE1Hu promote cell cycle progression at G2/M transition. Mol Cell Biol. 2005;25:5725–5737.
  • Beck H, Nahse V, Larsen MS, et al. Regulators of cyclin-dependent kinases are crucial for maintaining genome integrity in S phase. J Cell Biol. 2010;188:629–638.
  • Lianga N, Williams EC, Kennedy EK, et al. A Wee1 checkpoint inhibits anaphase onset. J Cell Biol. 2013;201:843–862.
  • Mahajan K, Mahajan NP. WEE1 tyrosine kinase, a novel epigenetic modifier. Trends Genet. 2013;29:394–402.
  • Mahajan K, Fang B, Koomen JM, et al. H2B Tyr37 phosphorylation suppresses expression of replication-dependent core histone genes. Nat Struct Mol Biol. 2012;19:930–937.
  • Bartucci M, Svensson S, Romania P, et al. Therapeutic targeting of Chk1 in NSCLC stem cells during chemotherapy. Cell Death Differ. 2012;19:768–778.
  • Music D, Dahlrot RH, Hermansen SK, et al. Expression and prognostic value of the WEE1 kinase in gliomas. J Neurooncol. 2016;127:381–389.
  • Wang H, Huang M, Zhang DY, et al. Global profiling of signaling networks: study of breast cancer stem cells and potential regulation. Oncologist. 2011;16:966–979.
  • Magnussen GI, Holm R, Emilsen E, et al. High expression of Wee1 is associated with poor disease-free survival in malignant melanoma: potential for targeted therapy. PLoS One. 2012;7:e38254.
  • Slipicevic A, Holth A, Hellesylt E, et al. Wee1 is a novel independent prognostic marker of poor survival in post-chemotherapy ovarian carcinoma effusions. Gynecol Oncol. 2014;135:118–124.
  • Kreahling JM, Gemmer JY, Reed D, et al. MK1775, a selective Wee1 inhibitor, shows single-agent antitumor activity against sarcoma cells. Mol Cancer Ther. 2012;11:174–182.
  • PosthumaDeBoer J, Wurdinger T, Graat HC, et al. WEE1 inhibition sensitizes osteosarcoma to radiotherapy. BMC Cancer. 2011;11:156.
  • Magnussen GI, Hellesylt E, Nesland JM, et al. High expression of wee1 is associated with malignancy in vulvar squamous cell carcinoma patients. BMC Cancer. 2013;13:288.
  • De Witt Hamer PC, Mir SE, Noske D, et al. WEE1 kinase targeting combined with DNA-damaging cancer therapy catalyzes mitotic catastrophe. Clin Cancer Res. 2011;17:4200–4207.
  • Backert S, Gelos M, Kobalz U, et al. Differential gene expression in colon carcinoma cells and tissues detected with a cDNA array. Int J Cancer. 1999;82:868–874.
  • Kiviharju-Af Hallstrom TM, Jaamaa S, Monkkonen M, et al. Human prostate epithelium lacks Wee1A-mediated DNA damage-induced checkpoint enforcement. Proc Natl Acad Sci USA. 2007;104:7211–7216.
  • Yoshida T, Tanaka S, Mogi A, et al. The clinical significance of Cyclin B1 and Wee1 expression in non-small-cell lung cancer. Ann Oncol. 2004;15:252–256.
  • Wang Z, Fu S. An overview of tyrosine kinase inhibitors for the treatment of epithelial ovarian cancer. Expert Opin Investig Drugs. 2016;25:15–30.
  • Leach SD, Scatena CD, Keefer CJ, et al. Negative regulation of Wee1 expression and Cdc2 phosphorylation during p53-mediated growth arrest and apoptosis. Cancer Res. 1998;58:3231–3236.
  • Tong Y, Torrent M, Florjancic AS, et al. Pyrimidine-based tricyclic molecules as potent and orally efficacious inhibitors of wee1 kinase. ACS Med Chem Lett. 2015;6:58–62.
  • Wright G, Golubeva V, Remsing Rix LL, et al. Dual targeting of WEE1 and PLK1 by AZD1775 elicits single agent cellular anticancer activity. ACS Chem Biol. 2017;12:1883–1892.
  • Zhu JY, Cuellar RA, Berndt N, et al. Structural basis of Wee kinases functionality and inactivation by diverse small molecule inhibitors. J Med Chem. 2017;60:7863–7875.
  • Metz JT, Johnson EF, Soni NB, et al. Navigating the kinome. Nat Chem Biol. 2011;7:200–202.
  • Geenen JJJ, Schellens JHM. Molecular pathways: targeting the protein kinase Wee1 in cancer. Clin Cancer Res. 2017;23:4540–4544.
  • Lescarbeau RS, Lei L, Bakken KK, et al. Quantitative phosphoproteomics reveals Wee1 kinase as a therapeutic target in a model of proneural glioblastoma. Mol Cancer Ther. 2016;15:1332–1343.
  • Carrassa L, Damia G. DNA damage response inhibitors: mechanisms and potential applications in cancer therapy. Cancer Treat Rev. 2017;60:139–151.
  • Aarts M, Bajrami I, Herrera-Abreu MT, et al. Functional genetic screen identifies increased sensitivity to WEE1 inhibition in cells with defects in fanconi anemia and HR pathways. Mol Cancer Ther. 2015;14:865–876.
  • Sen T, Tong P, Diao L, et al. Targeting AXL and mTOR pathway overcomes primary and acquired resistance to WEE1 inhibition in small-cell lung cancer. Clin Cancer Res. 2017;23:6239–6253.
  • Richer AL, Cala JM, O’Brien K, et al. WEE1 kinase inhibitor AZD1775 has preclinical efficacy in LKB1-deficient non-small cell lung cancer. Cancer Res. 2017;77:4663–4672.
  • Brandsma I, Fleuren EDG, Williamson CT, et al. Directing the use of DDR kinase inhibitors in cancer treatment. Expert Opin Investig Drugs. 2017;26:1341–1355.
  • Ku BM, Bae YH, Koh J, et al. Mutational status of TP53 defines the efficacy of Wee1 inhibitor AZD1775 in KRAS-mutant non-small cell lung cancer. Oncotarget. 2017;8:67526–67537.
  • Rajeshkumar NV, De Oliveira E, Ottenhof N, et al. MK-1775, a potent Wee1 inhibitor, synergizes with gemcitabine to achieve tumor regressions, selectively in p53-deficient pancreatic cancer xenografts. Clin Cancer Res. 2011;17:2799–2806.
  • Hirai H, Iwasawa Y, Okada M, et al. Small-molecule inhibition of Wee1 kinase by MK-1775 selectively sensitizes p53-deficient tumor cells to DNA-damaging agents. Mol Cancer Ther. 2009;8:2992–3000.
  • Bridges KA, Hirai H, Buser CA, et al. MK-1775, a novel Wee1 kinase inhibitor, radiosensitizes p53-defective human tumor cells. Clin Cancer Res. 2011;17:5638–5648.
  • Zhang M, Dominguez D, Chen S, et al. WEE1 inhibition by MK1775 as a single-agent therapy inhibits ovarian cancer viability. Oncol Lett. 2017;14:3580–3586.
  • Guertin AD, Li J, Liu Y, et al. Preclinical evaluation of the WEE1 inhibitor MK-1775 as single-agent anticancer therapy. Mol Cancer Ther. 2013;12:1442–1452.
  • Mueller S, Hashizume R, Yang X, et al. Targeting Wee1 for the treatment of pediatric high-grade gliomas. Neuro Oncol. 2014;16:352–360.
  • Van Linden AA, Baturin D, Ford JB, et al. Inhibition of Wee1 sensitizes cancer cells to antimetabolite chemotherapeutics in vitro and in vivo, independent of p53 functionality. Mol Cancer Ther. 2013;12:2675–2684.
  • Webster PJ, Littlejohns AT, Gaunt HJ, et al. AZD1775 induces toxicity through double-stranded DNA breaks independently of chemotherapeutic agents in p53-mutated colorectal cancer cells. Cell Cycle. 2017;16:2176–2182.
  • Cuneo KC, Morgan MA, Davis MA, et al. Wee1 kinase inhibitor AZD1775 radiosensitizes hepatocellular carcinoma regardless of TP53 mutational status through induction of replication stress. Int J Radiat Oncol Biol Phys. 2016;95:782–790.
  • Yang Y, Gao Y, Mutter-Rottmayer L, et al. DNA repair factor RAD18 and DNA polymerase polkappa confer tolerance of oncogenic DNA replication stress. J Cell Biol. 2017;216:3097–3115.
  • Toledo CM, Ding Y, Hoellerbauer P, et al. Genome-wide CRISPR-Cas9 screens reveal loss of redundancy between PKMYT1 and WEE1 in glioblastoma stem-like cells. Cell Rep. 2015;13:2425–2439.
  • Pfister SX, Markkanen E, Jiang Y, et al. Inhibiting WEE1 selectively kills histone H3K36me3-deficient cancers by dNTP starvation. Cancer Cell. 2015;28:557–568.
  • Mahajan NP, Malla P, Bhagwat S, et al. WEE1 epigenetically modulates 5-hmC levels by pY37-H2B dependent regulation of IDH2 gene expression. Oncotarget. 2017;8:106352–106368.
  • Lindemann A, Takahashi H, Patel AA, et al. Targeting the DNA damage response in OSCC with TP53 mutations. J Dent Res. 2018;97:635–644.
  • Osman AA, Monroe MM, Ortega Alves MV, et al. Wee-1 kinase inhibition overcomes cisplatin resistance associated with high-risk TP53 mutations in head and neck cancer through mitotic arrest followed by senescence. Mol Cancer Ther. 2015;14:608–619.
  • Tanaka N, Patel AA, Wang J, et al. Wee-1 kinase inhibition sensitizes high-risk HPV+ HNSCC to apoptosis accompanied by downregulation of MCl-1 and XIAP antiapoptotic proteins. Clin Cancer Res. 2015;21:4831–4844.
  • Stewart E, Federico SM, Chen X, et al. Orthotopic patient-derived xenografts of paediatric solid tumours. Nature. 2017;549:96–100.
  • Jhuraney A, Woods NT, Wright G, et al. PAXIP1 potentiates the combination of WEE1 Inhibitor AZD1775 and platinum agents in lung cancer. Mol Cancer Ther. 2016;15:1669–1681.
  • Zhao W, Liu S, Dou Q, et al. The role and mechanism of WEE1 on the cisplatin resistance reversal of the HepG2/DDP human hepatic cancer cell line. Oncol Lett. 2015;10:3081–3086.
  • Indovina P, Marcelli E, Di Marzo D, et al. Abrogating G(2)/M checkpoint through WEE1 inhibition in combination with chemotherapy as a promising therapeutic approach for mesothelioma. Cancer Biol Ther. 2014;15:380–388.
  • Ford JB, Baturin D, Burleson TM, et al. AZD1775 sensitizes T cell acute lymphoblastic leukemia cells to cytarabine by promoting apoptosis over DNA repair. Oncotarget. 2015;6:28001–28010.
  • Kausar T, Schreiber JS, Karnak D, et al. Sensitization of pancreatic cancers to gemcitabine chemoradiation by WEE1 kinase inhibition depends on homologous recombination repair. Neoplasia. 2015;17:757–766.
  • Kreahling JM, Foroutan P, Reed D, et al. Wee1 inhibition by MK-1775 leads to tumor inhibition and enhances efficacy of gemcitabine in human sarcomas. PLoS One. 2013;8:e57523.
  • Kim HY, Cho Y, Kang H, et al. Targeting the WEE1 kinase as a molecular targeted therapy for gastric cancer. Oncotarget. 2016;7:49902–49916.
  • Hirai H, Arai T, Okada M, et al. MK-1775, a small molecule Wee1 inhibitor, enhances anti-tumor efficacy of various DNA-damaging agents, including 5-fluorouracil. Cancer Biol Ther. 2010;9:514–522.
  • Lewis CW, Jin Z, Macdonald D, et al. Prolonged mitotic arrest induced by Wee1 inhibition sensitizes breast cancer cells to paclitaxel. Oncotarget. 2017;8:73705–73722.
  • Pokorny JL, Calligaris D, Gupta SK, et al. The efficacy of the Wee1 inhibitor MK-1775 combined with temozolomide is limited by heterogeneous distribution across the blood-brain barrier in glioblastoma. Clin Cancer Res. 2015;21:1916–1924.
  • Ma H, Takahashi A, Sejimo Y, et al. Targeting of carbon ion-induced G2 checkpoint activation in lung cancer cells using Wee-1 inhibitor MK-1775. Radiat Res. 2015;184:660–669.
  • Sarcar B, Kahali S, Prabhu AH, et al. Targeting radiation-induced G(2) checkpoint activation with the Wee-1 inhibitor MK-1775 in glioblastoma cell lines. Mol Cancer Ther. 2011;10:2405–2414.
  • Sakurikar N, Thompson R, Montano R, et al. A subset of cancer cell lines is acutely sensitive to the Chk1 inhibitor MK-8776 as monotherapy due to CDK2 activation in S phase. Oncotarget. 2016;7:1380–1394.
  • Tanaka N, Patel AA, Tang L, et al. Replication stress leading to apoptosis within the S-phase contributes to synergism between vorinostat and AZD1775 in HNSCC harboring high-risk TP53 mutation. Clin Cancer Res. 2017;23:6541–6554.
  • Zheng H, Shao F, Martin S, et al. WEE1 inhibition targets cell cycle checkpoints for triple negative breast cancers to overcome cisplatin resistance. Sci Rep. 2017;7:43517.
  • Clausse V, Goloudina AR, Uyanik B, et al. Wee1 inhibition potentiates Wip1-dependent p53-negative tumor cell death during chemotherapy. Cell Death Dis. 2016;7:e2195.
  • Chila R, Basana A, Lupi M, et al. Combined inhibition of Chk1 and Wee1 as a new therapeutic strategy for mantle cell lymphoma. Oncotarget. 2015;6:3394–3408.
  • Li Y, Saini P, Sriraman A, et al. Mdm2 inhibition confers protection of p53-proficient cells from the cytotoxic effects of Wee1 inhibitors. Oncotarget. 2015;6:32339–32352.
  • Francis AM, Alexander A, Liu Y, et al. CDK4/6 inhibitors sensitize rb-positive sarcoma cells to Wee1 kinase inhibition through reversible cell-cycle arrest. Mol Cancer Ther. 2017;16:1751–1764.
  • Drean A, Williamson CT, Brough R, et al. Modeling therapy resistance in BRCA1/2-mutant cancers. Mol Cancer Ther. 2017;16:2022–2034.
  • Karnak D, Engelke CG, Parsels LA, et al. Combined inhibition of Wee1 and PARP1/2 for radiosensitization in pancreatic cancer. Clin Cancer Res. 2014;20:5085–5096.
  • Ruiz S, Mayor-Ruiz C, Lafarga V, et al. A genome-wide CRISPR screen identifies CDC25A as a determinant of sensitivity to ATR inhibitors. Mol Cell. 2016;62:307–313.
  • Busch CJ, Kroger MS, Jensen J, et al. G2-checkpoint targeting and radiosensitization of HPV/p16-positive HNSCC cells through the inhibition of Chk1 and Wee1. Radiother Oncol. 2017;122:260–266.
  • Restelli V, Vagni M, Arribas AJ, et al. Inhibition of CHK1 and WEE1 as a new therapeutic approach in diffuse large B cell lymphomas with MYC deregulation. Br J Haematol. 2018;181:129–133.
  • Hauge S, Naucke C, Hasvold G, et al. Combined inhibition of Wee1 and Chk1 gives synergistic DNA damage in S-phase due to distinct regulation of CDK activity and CDC45 loading. Oncotarget. 2017;8:10966–10979.
  • Carrassa L, Chila R, Lupi M, et al. Combined inhibition of Chk1 and Wee1: in vitro synergistic effect translates to tumor growth inhibition in vivo. Cell Cycle. 2012;11:2507–2517.
  • Wang G, Niu X, Zhang W, et al. Synergistic antitumor interactions between MK-1775 and panobinostat in preclinical models of pancreatic cancer. Cancer Lett. 2015;356:656–668.
  • Qi W, Zhang W, Edwards H, et al. Synergistic anti-leukemic interactions between panobinostat and MK-1775 in acute myeloid leukemia ex vivo. Cancer Biol Ther. 2015;16:1784–1793.
  • Zhou L, Zhang Y, Chen S, et al. A regimen combining the Wee1 inhibitor AZD1775 with HDAC inhibitors targets human acute myeloid leukemia cells harboring various genetic mutations. Leukemia. 2015;29:807–818.
  • Chen G, Zhang B, Xu H, et al. Suppression of sirt1 sensitizes lung cancer cells to WEE1 inhibitor MK-1775-induced DNA damage and apoptosis. Oncogene. 2017;36:6863–6872.
  • Wu S, Wang S, Gao F, et al. Activation of WEE1 confers resistance to PI3K inhibition in glioblastoma. Neuro Oncol. 2018;20:78–91.
  • Kuzu OF, Gowda R, Sharma A, et al. Identification of WEE1 as a target to make AKT inhibition more effective in melanoma. Cancer Biol Ther. 2018;19:53–62.
  • Hai J, Liu S, Bufe L, et al. Synergy of WEE1 and mTOR inhibition in mutant KRAS-driven lung cancers. Clin Cancer Res. 2017;23:6993–7005.
  • Dinavahi SS, Noory MA, Gowda R, et al. Moving synergistically acting drug combinations to the clinic by comparing sequential versus simultaneous drug administrations. Mol Pharmacol. 2018;93:190–196.
  • Johnson TS, Terrell CE, Millen SH, et al. Etoposide selectively ablates activated T cells to control the immunoregulatory disorder hemophagocytic lymphohistiocytosis. J Immunol. 2014;192:84–91.
  • McNally JP, Millen SH, Chaturvedi V, et al. Manipulating DNA damage-response signaling for the treatment of immune-mediated diseases. Proc Natl Acad Sci USA. 2017;114:E4782–E4791.
  • Watanabe M, Moon KD, Vacchio MS, et al. Downmodulation of tumor suppressor p53 by T cell receptor signaling is critical for antigen-specific CD4(+) T cell responses. Immunity. 2014;40:681–691.
  • Ghiasi N, Habibagahi M, Rosli R, et al. Tumor suppressive effects of WEE1 gene silencing could not enhance immunopotentiation effects of CD80 and 4–1BBL co-stimulation in human T cells. J Cancer Res Ther. 2015;11:708–716.
  • Fernando RI, Litzinger M, Trono P, et al. The T-box transcription factor brachyury promotes epithelial-mesenchymal transition in human tumor cells. J Clin Invest. 2010;120:533–544.
  • Hamilton DH, Huang B, Fernando RI, et al. WEE1 inhibition alleviates resistance to immune attack of tumor cells undergoing epithelial-mesenchymal transition. Cancer Res. 2014;74:2510–2519.
  • Li J, Wu J, Bao X, et al. Quantitative and mechanistic understanding of AZD1775 penetration across human blood-brain barrier in glioblastoma patients using an IVIVE-PBPK modeling approach. Clin Cancer Res. 2017;23:7454–7466.
  • Do K, Wilsker D, Ji J, et al. Phase I study of single-agent AZD1775 (MK-1775), a Wee1 kinase inhibitor, in patients with refractory solid tumors. J Clin Oncol. 2015;33:3409–3415.
  • Leijen S, van Geel RM, Pavlick AC, et al. Phase I study evaluating WEE1 inhibitor AZD1775 as monotherapy and in combination with gemcitabine, cisplatin, or carboplatin in patients with advanced solid tumors. J Clin Oncol. 2016;34:4371–4380.
  • Leijen S, Van Geel RM, Sonke GS, et al. Phase II study of wee1 inhibitor AZD1775 plus carboplatin in patients with TP53-mutated ovarian cancer refractory or resistant to first-line therapy within 3 months. J Clin Oncol. 2016;34:4354–4361.
  • Mendez E, Rodriguez CP, Kao M, et al. A phase I clinical trial of AZD1775 in combination with neoadjuvant weekly docetaxel and cisplatin before definitive therapy in head and neck squamous cell carcinoma. Clin Cancer Res. 2018;24:2740–2748.
  • Oza AM, Weberpals JI, Provencher DM, et al. An international, biomarker-directed, randomized, phase II trial of AZD1775 plus paclitaxel and carboplatin (P/C) for the treatment of women with platinum-sensitive, TP53-mutant ovarian cancer. J Clin Oncol. 2015;33(suppl; abstr 5506).
  • Lheureux S, Weberpals JI, Hendrickson AEW, et al. A randomized, placebo-controlled phase II trial comparing gemcitabine monotherapy to gemcitabine in combination with AZD 1775 (MK 1775) in women with recurrent, platinum-resistant epithelial ovarian, primary peritoneal, or fallopian tube cancers: trial of princess margaret, Mayo, Chicago, and California consortia. J Clin Oncol. 2015;33(suppl; abstr TPS5613).
  • Glorieux M, Dok R, Nuyts S. Novel DNA targeted therapies for head and neck cancers: clinical potential and biomarkers. Oncotarget. 2017;8:81662–81678.
  • Pappano WN, Zhang Q, Tucker LA, et al. Genetic inhibition of the atypical kinase Wee1 selectively drives apoptosis of p53 inactive tumor cells. BMC Cancer. 2014;14:430.
  • Friedman J, Morisada M, Sun L, et al. Inhibition of WEE1 kinase and cell cycle checkpoint activation sensitizes head and neck cancers to natural killer cell therapies. J Immunother Cancer. 2018;6:59.
  • Hamilton DH, McCampbell KK, Palena C. Loss of the cyclin-dependent kinase inhibitor 1 in the context of brachyury-mediated phenotypic plasticity drives tumor resistance to immune attack. Front Oncol. 2018;8:143.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.