Article Data

  • Views 1000
  • Dowloads 184

Original Research

Open Access

Identifying and overcoming a mechanism of resistance to WEE1 kinase inhibitor AZD1775 in high grade serous ovarian cancer cells

  • Miriam K. Gomez1,§
  • John P. Thomson1
  • Graeme R. Grimes2
  • Anderson T. Wang3,§
  • Michael Churchman1
  • Mark J. O’Connor3
  • Charlie Gourley1
  • David W. Melton1,*,

1Nicola Murray Centre for Ovarian Cancer Research, University of Edinburgh, EH4 2XU Edinburgh, UK

2MRC Human Genetics Unit, University of Edinburgh, EH4 2XU Edinburgh, UK

3Bioscience, Oncology R&D, AstraZeneca, CB2 0AA Cambridge, UK

DOI: 10.31083/j.ejgo4302024 Vol.43,Issue 2,April 2022 pp.183-195

Submitted: 10 December 2021 Accepted: 15 February 2022

Published: 15 April 2022

*Corresponding Author(s): David W. Melton E-mail: David.Melton@ed.ac.uk

§ The author’s own special request.

Abstract

Objective: As a result of TP53 gene mutation high grade serous ovarian cancer (HGSOC) is dependent on the G2 checkpoint for the repair of DNA damage and survival. The key role of WEE1 kinase at this checkpoint makes inhibition of WEE1 kinase in combination with DNA damaging agents an attractive therapeutic strategy for HGSOC. Our aim was to characterise resistance mechanisms to WEE1 inhibitor AZD1775 and identify ways to overcome resistance ready for use in the clinic. Methods: AZD1775-resistant HGSOC cell clones were isolated and western blotting, cell cycle analysis, growth assays, RNA-Seq and gene expression analysis were used to characterise resistance mechanisms and investigate a way to overcome resistance. Results: A resistance mechanism previously reported in small cell lung cancer did not operate in HGSOC. Instead, resistance resulted from different cell cycle control pathway changes that slow AZD1775-induced cell cycle progression and reduce accumulation of replication associated DNA damage. One major change was reduced levels of CDK1, the substrate for WEE1 kinase inhibition; another was increased levels of PKMYT1, which can also inhibit CDK1. Increased expression of TGFβ signalling to slow cell cycle progression occurred in resistant clones. A TGFβR1 inhibitor overcame resistance in a clone with the highest TGFβR1 receptor expression. Conclusions: Although overexpression of the membrane glycoprotein MDR1 is a common mechanism of drug resistance, it was not involved in our HGSOC cells. Instead AZD1775 resistance resulted from cell cycle control pathway changes that combine to slow AZD1775-induced cell cycle progression and so reduce accumulation of replication-associated DNA damage.


Keywords

ovarian cancer; cell cycle control; WEE1 kinase inhibition; resistance mechanism; DNA repair

Cite and Share

Miriam K. Gomez,John P. Thomson,Graeme R. Grimes,Anderson T. Wang,Michael Churchman,Mark J. O’Connor,Charlie Gourley,David W. Melton. Identifying and overcoming a mechanism of resistance to WEE1 kinase inhibitor AZD1775 in high grade serous ovarian cancer cells. European Journal of Gynaecological Oncology. 2022. 43(2);183-195.

References

[1] Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. A Cancer Journal for Clinicians. 2018; 68: 394–424.

[2] Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature. 2011; 474: 609–615.

[3] Lawrie TA, Winter-Roach BA, Heus P, Kitchener HC. Adjuvant (post-surgery) chemotherapy for early stage epithelial ovarian cancer. The Cochrane Database of Systematic Reviews. 2015; 2015: CD004706.

[4] Moschetta M, Boussios S, Rassy E, Samartzis EP, Funingana G, Uccello M. Neoadjuvant treatment for newly diagnosed advanced ovarian cancer: where do we stand and where are we going? Annals of Translational Medicine. 2020; 8: 1710.

[5] Moore K, Colombo N, Scambia G, Kim B, Oaknin A, Friedlander M, et al. Maintenance Olaparib in Patients with Newly Diagnosed Advanced Ovarian Cancer. The New England Journal of Medicine. 2018; 379: 2495–2505.

[6] D’Andrea AD. Mechanisms of PARP inhibitor sensitivity and resistance. DNA Repair. 2018; 71: 172–176.

[7] McGowan CH, Russell P. Cell cycle regulation of human WEE1. The EMBO Journal. 1995; 14: 2166–2175.

[8] Zhou BB, Bartek J. Targetting the checkpoint kinases: chemosensitization versus chemoprotection. Nature Reviews. Cancer. 2004; 4: 216–225.

[9] Kawabe T. G2/M checkpoint abrogators as anticancer drugs. Molecular Cancer Therapeutics. 2004; 3: 513–519.

[10] Buche, N, Britten CD. G2 checkpoint abrogation and checkpoint kinase-1 targeting in the treatment of cancer. British Journal of Cancer. 2008; 98: 523–528.

[11] De Witt Hamer PC, Mir SE, Noske D, Van Noorden CJF, Würdinger T. WEE1 kinase targeting combined with DNA-damaging cancer therapy catalyzes mitotic catastrophe. Clinical Cancer Research. 2011; 17: 4200–4207.

[12] Leijen S, van Geel RM, Pavlick AC, Tibes R, Rosen L, Razak AR, 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. Journal of Clinical Oncology. 2016; 34: 4371–4380.

[13] Mittra A, Coyne GO, Kummar S, Do K, Bruns A, Juwara L, et al. Abstract CT099: DNA damage response and therapeutic activity following once-daily administration of the Wee 1 inhibitor AZD1775 (adavosertib). Cancer Research. 2019.

[14] Leijen S, van Geel RM, Sonke GS, de Jong D, Rosenberg EH, Marchetti S, 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. Journal of Clinical Oncology. 2016; 34: 4354–4361.

[15] Oza AM, Estevez-Diz M, Grischke EM, Hall M, Marmé F, Provencher D, et al. A Biomarker-enriched, Randomized Phase II Trial of Adavosertib (AZD1775) Plus Paclitaxel and Carboplatin for Women with Platinum-sensitive TP53-mutant Ovarian Cancer. Clinical Cancer Research. 2020; 26: 4767–4776.

[16] Lheureux S, Cristea MC, Bruce JP, Garg S, Cabanero M, Mantia-Smaldone G, et al. Adavosertib plus gemcitabine for platinum-resistant or platinum-refractory recurrent ovarian cancer: a double-blind, randomised, placebo-controlled, phase 2 trial. The Lancet. 2021; 397: 281–292.

Keenan TE, Li T, Vallius T, Guerriero JL, Tayob N, Kochupu-rakkal B, et al. Clinical Efficacy and Molecular Response Correlates of the WEE1 Inhibitor Adavosertib Combined with Cisplatin in Patients with Metastatic Triple-Negative Breast Cancer. Clinical Cancer Research. 2021; 27: 983–991.

Liu JF, Xiong N, Campos SM, Wright AA, Krasner C, Schumer S, et al. Phase II Study of the WEE1 Inhibitor Adavosertib in Recurrent Uterine Serous Carcinoma. Journal of Clinical Oncology. 2021; 39: 1531–1539.

[19] Moore KN, Chambers SK, Hamilton EP, Chen L, Oza AM, Ghamande SA, et al. Adavosertib with Chemotherapy in Patients with Primary Platinum-Resistant Ovarian, Fallopian Tube, or Peritoneal Cancer: an Open-Label, Four-Arm, Phase II Study. Clinical Cancer Research. 2022; 28: 36–44.

[20] Mueller PR, Coleman TR, Kumagai A, Dunphy WG. Myt1: a Membrane-Associated Inhibitory Kinase that Phosphorylates Cdc2 on both Threonine-14 and Tyrosine-15. Science. 1995; 270: 86–90.

[21] Sen T, Tong P, Diao L, Li L, Fan Y, Hoff J, et al. Targeting AXL and mTOR Pathway Overcomes Primary and Acquired Resistance to WEE1 Inhibition in Small-Cell Lung Cancer. Clinical Cancer Research. 2017; 23: 6239–6253.

[22] Skehan P, Storeng R, Scudiero D, Monks A, McMahon J, Vistica D, et al. New Colorimetric Cytotoxicity Assay for Anticancer-Drug Screening. Journal of the National Cancer Institute. 1990; 82: 1107–1112.

[23] Harrison C, Ketchen AM, Redhead NJ, O’Sullivan MJ, Melton DW. Replication failure, genome instability, and increased cancer susceptibility in mice with a point mutation in the DNA ligase I gene. Cancer Research. 2002; 62: 4065–4074.

[24] Song L, Ritchie A, McNeil EM, Li W, Melton DW. Identification of DNA repair gene Ercc1 as a novel target in melanoma. Pigment Cell & Melanoma Research. 2011; 24: 966–971.

[25] Domcke S, Sinha R, Levine DA, Sander C, Schultz N. Evaluating cell lines as tumour models by comparison of genomic profiles. Nature Communications. 2013; 4: 2126.

[26] Stordal B, Timms K, Farrelly A, Gallagher D, Busschots S, Renaud M, et al. BRCA1/2 mutation analysis in 41 ovarian cell lines reveals only one functionally deleterious BRCA1 mutation. Molecular Oncology. 2013; 7: 567–579.

[27] Tumiati M, Hietanen S, Hynninen J, Pietilä E, Färkkilä A, Kaipio K, et al. A Functional Homologous Recombination Assay Predicts Primary Chemotherapy Response and Long-Term Survival in Ovarian Cancer Patients. Clinical Cancer Research. 2018; 24: 4482–4493.

[28] Gomez MK, Illuzzi G, Colomer C, Churchman M, Hollis RL, O’Connor MJ, et al. Identifying and Overcoming Mechanisms of PARP Inhibitor Resistance in Homologous Recombination Repair-Deficient and Repair-Proficient High Grade Serous Ovarian Cancer Cells. Cancers. 2020; 12: 1503.

[29] The Cancer Cell Line Encyclopaedia (CCLE). Available at: ht tps://portals.broadinstitute.org/ccle (Accessed: 16 April 2020).

[30] Vaidyanathan A, Sawers L, Gannon A, Chakravarty P, Scott AL, Bray SE, et al. ABCB1 (MDR1) induction defines a common resistance mechanism in paclitaxel- and olaparib-resistant ovarian cancer cells. British Journal of Cancer. 2016; 115: 431–441.

[31] Nakumura T, Sakaeda T, Ohmoto N, Moriya Y, Komoto C, Shirakawa T, et al. Gene expression profiles of ABC transporters and cytochrome P450 3a in Caco-2 and human colorectal cancer cell lines. Pharmaceutical Research. 2003; 20: 324–327.

[32] Parker L, Piwnica-Worms H. Inactivation of the p34cdc2-cyclin B complex by the human WEE1 tyrosine kinase. Science. 1992; 257: 1955–1957.

[33] Beck H, Nähse V, Larsen MSY, Groth P, Clancy T, Lees M, et al. Regulators of cyclin-dependent kinases are crucial for maintaining genome integrity in S phase. Journal of Cell Biology. 2010; 188: 629–638.

[34] Beck H, Nähse-Kumpf V, Larsen MSY, O’Hanlon KA, Patzke S, Holmberg C, et al. Cyclin-dependent kinase suppression by WEE1 kinase protects the genome through control of replication initiation and nucleotide consumption. Molecular and Cellular Biology. 2012; 32: 4226–4236.

[35] Kinner A, Wu W, Staudt C, Iliakis G. Gamma-H2AX in recognition and signaling of DNA double-strand breaks in the context of chromatin. Nucleic Acids Research. 2008; 36: 5678–5694.

[36] Jette N, Lees-Miller SP. The DNA-dependent protein kinase: a multifunctional protein kinase with roles in DNA double strand break repair and mitosis. Progress in Biophysics and Molecular Biology. 2015; 117: 194–205.

[37] Vassin VM, Anantha RW, Sokolova E, Kanner S, Borowiec JA. Human RPA phosphorylation by ATR stimulates DNA synthesis and prevents ssDNA accumulation during DNA-replication stress. Journal of Cell Science. 2009; 122: 4070–4080.

[38] Hendzel MJ, Wei Y, Mancini MA, Van Hooser A, Ranalli T, Brinkley BR, et al. Mitosis-specific phosphorylation of histone H3 initiates primarily within pericentromeric heterochromatin during G2 and spreads in an ordered fashion coincident with mitotic chromosome condensation. Chromosoma. 1997; 106: 348–360.

[39] Gomis RR, Alarcón C, Nadal C, Van Poznak C, Massagué J. C/EBPbeta at the core of the TGFbeta cytostatic response and its evasion in metastatic breast cancer cells. Cancer Cell. 2006; 10: 203–214.

[40] Gellibert F, Woolven J, Fouchet M, Mathews N, Goodland H, Lovegrove V, et al. Identification of 1,5-naphthyridine derivatives as a novel series of potent and selective TGF-beta type i receptor inhibitors. Journal of Medicinal Chemistry. 2004; 47: 4494–4506.

[41] Boussios S, Karihtala P, Moschetta M, Karathanasi A, Sadauskaite A, Rassy E, et al. Combined Strategies with Poly (ADP-Ribose) Polymerase (PARP) Inhibitors for the Treatment of Ovarian Cancer: A Literature Review. Diagnostics (Basel). 2019; 9: 87.

[42] Do K, Wilsker D, Ji J, Zlott J, Freshwater T, Kinders RJ, et al. Phase i Study of Single-Agent AZD1775 (MK-1775), a Wee1 Kinase Inhibitor, in Patients with Refractory Solid Tumors. Journal of Clinical Oncology. 2015; 33: 3409–3415.

[43] Garcia TB, Fosmire SP, Porter CC. Increased activity of both CDK1 and CDK2 is necessary for the combinatorial activity of WEE1 inhibition and cytarabine. Leukemia Research. 2018; 64: 30–33.

[44] Boutros R, Lobjois V, Ducommun B. CDC25 phosphatases in cancer cells: key players? Good targets? Nature Reviews. Cancer. 2007; 7: 495–507.

[45] Izumi T, Maller JL. Elimination of cdc2 phosphorylation sites in the cdc25 phosphatase blocks initiation of M-phase. Molecular Biology of the Cell. 1993; 4: 1337–1350.

[46] Ovejero S, Ayala P, Bueno A, Sacristán MP. Human Cdc14a regulates Wee1 stability by counteracting CDK-mediated phosphorylation. Molecular Biology of the Cell. 2012; 23: 4515–4525.

[47] Vázquez-Novelle MD, Mailand N, Ovejero S, Bueno A, Sacristán MP. Human Cdc14a phosphatase modulates the G2/M transition through Cdc25a and Cdc25B. The Journal of Biological Chemistry. 2010; 285: 40544–40553.

[48] Sacristán MP, Ovejero S, Bueno A. Human Cdc14a becomes a cell cycle gene in controlling Cdk1 activity at the G₂/M transition. Cell Cycle. 2011; 10: 387–391.

[49] Lewis CW, Bukhari AB, Xiao EJ, Choi W, Smith JD, Homola E, et al. Upregulation of Myt1 Promotes Acquired Resistance of Cancer Cells to Wee1 Inhibition. Cancer Research. 2019; 79: 5971–5985.

[50] Guertin AD, Li J, Liu Y, Hurd MS, Schuller AG, Long B, et al. Preclinical evaluation of the WEE1 inhibitor MK-1775 as single-agent anticancer therapy. Molecular Cancer Therapeutics. 2013; 12: 1442–1452.

[51] Garcia TB, Uluisik RC, van Linden AA, Jones KL, Venkataraman S, Vibhakar R, et al. Increased HDAC Activity and c-MYC Expression Mediate Acquired Resistance to WEE1 Inhibition in Acute Leukemia. Frontiers in Oncology. 2020; 10: 296.

[52] Hauge S, Macurek L, Syljuåsen RG. P21 limits S phase DNA damage caused by the Wee1 inhibitor MK1775. Cell Cycle. 2019; 18: 834–847.

Abstracted / indexed in

Science Citation Index Expanded (SciSearch) Created as SCI in 1964, Science Citation Index Expanded now indexes over 9,500 of the world’s most impactful journals across 178 scientific disciplines. More than 53 million records and 1.18 billion cited references date back from 1900 to present.

Biological Abstracts Easily discover critical journal coverage of the life sciences with Biological Abstracts, produced by the Web of Science Group, with topics ranging from botany to microbiology to pharmacology. Including BIOSIS indexing and MeSH terms, specialized indexing in Biological Abstracts helps you to discover more accurate, context-sensitive results.

Google Scholar Google Scholar is a freely accessible web search engine that indexes the full text or metadata of scholarly literature across an array of publishing formats and disciplines.

JournalSeek Genamics JournalSeek is the largest completely categorized database of freely available journal information available on the internet. The database presently contains 39226 titles. Journal information includes the description (aims and scope), journal abbreviation, journal homepage link, subject category and ISSN.

Current Contents - Clinical Medicine Current Contents - Clinical Medicine provides easy access to complete tables of contents, abstracts, bibliographic information and all other significant items in recently published issues from over 1,000 leading journals in clinical medicine.

BIOSIS Previews BIOSIS Previews is an English-language, bibliographic database service, with abstracts and citation indexing. It is part of Clarivate Analytics Web of Science suite. BIOSIS Previews indexes data from 1926 to the present.

Journal Citation Reports/Science Edition Journal Citation Reports/Science Edition aims to evaluate a journal’s value from multiple perspectives including the journal impact factor, descriptive data about a journal’s open access content as well as contributing authors, and provide readers a transparent and publisher-neutral data & statistics information about the journal.

Submission Turnaround Time

Conferences

Top