Could Inhibiting the DNA Damage Repair Checkpoint Rescue Immune-Checkpoint-Inhibitor-Resistant Endometrial Cancer?
Abstract
:1. Introduction
2. Immunotherapy in Endometrial Cancer: Progress and Problems
2.1. Clinical Efficacy of Immunotherapy in Endometrial Cancer
2.2. Limitations of Current Immunotherapy in Endometrial Cancer
2.3. Prognostic Biomarker in Endometrial Cancer
2.4. New Approaches to Improve the Efficacy of Immunotherapy and Enhance Antitumor Immunity
3. DNA Damage Response Checkpoint in Endometrial Cancer
3.1. DNA Damage Response Checkpoint Inhibitors
3.1.1. ATM/CHK2/P53 Pathway Inhibitors
3.1.2. ATR/CHK1/WEE1 Pathway Inhibitors
3.2. Prognostic Biomarker in Endometrial Cancer
4. Crosstalk between the DNA Damage Response and Innate and Adaptive Immunity
5. Rationale for “Double-Checkpoint Inhibition” in Endometrial Cancer
6. The Combination Strategies of DNA Damage Response Pathway Inhibitors and Immune Therapy
7. Summary and Future Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
- Miller, K.D.; Nogueira, L.; Devasia, T.; Mariotto, A.B.; Yabroff, K.R.; Jemal, A.; Kramer, J.; Siegel, R.L. Cancer treatment and survivorship statistics, 2022. CA Cancer J. Clin. 2022, 72, 409–436. [Google Scholar] [CrossRef] [PubMed]
- Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2022. CA Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef] [PubMed]
- Crosbie, E.J.; Kitson, S.J.; McAlpine, J.N.; Mukhopadhyay, A.; Powell, M.E.; Singh, N. Endometrial cancer. Lancet 2022, 399, 1412–1428. [Google Scholar] [CrossRef]
- Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef] [PubMed]
- Matei, D.; Filiaci, V.; Randall, M.E.; Mutch, D.; Steinhoff, M.M.; DiSilvestro, P.A.; Moxley, K.M.; Kim, Y.M.; Powell, M.A.; O’Malley, D.M.; et al. Adjuvant Chemotherapy plus Radiation for Locally Advanced Endometrial Cancer. N. Engl. J. Med. 2019, 380, 2317–2326. [Google Scholar] [CrossRef] [PubMed]
- Legge, F.; Restaino, S.; Leone, L.; Carone, V.; Ronsini, C.; Di Fiore, G.L.M.; Pasciuto, T.; Pelligra, S.; Ciccarone, F.; Scambia, G.; et al. Clinical outcome of recurrent endometrial cancer: Analysis of post-relapse survival by pattern of recurrence and secondary treatment. Int. J. Gynecol. Cancer. 2020, 30, 193–200. [Google Scholar] [CrossRef]
- Bokhman, J.V. Two pathogenetic types of endometrial carcinoma. Gynecol. Oncol. 1983, 15, 10–17. [Google Scholar] [CrossRef]
- Kandoth, C.; Schultz, N.; Cherniack, A.D.; Akbani, R.; Liu, Y.; Shen, H.; Robertson, A.G.; Pashtan, I.; Shen, R.; Benz, C.C.; et al. Integrated genomic characterization of endometrial carcinoma. Nature 2013, 497, 67–73. [Google Scholar] [CrossRef]
- Eggink, F.A.; Van Gool, I.C.; Leary, A.; Pollock, P.M.; Crosbie, E.J.; Mileshkin, L.; Jordanova, E.S.; Adam, J.; Freeman-Mills, L.; Church, D.N.; et al. Immunological profiling of molecularly classified high-risk endometrial cancers identifies POLE-mutant and microsatellite unstable carcinomas as candidates for checkpoint inhibition. Oncoimmunology. 2017, 6, e1264565. [Google Scholar] [CrossRef]
- Van Gool, I.C.; Eggink, F.A.; Freeman-Mills, L.; Stelloo, E.; Marchi, E.; de Bruyn, M.; Palles, C.; Nout, R.A.; de Kroon, C.D.; Osse, E.M.; et al. POLE Proofreading Mutations Elicit an Antitumor Immune Response in Endometrial Cancer. Clin. Cancer Res. 2015, 21, 3347–3355. [Google Scholar] [CrossRef] [PubMed]
- Marcus, L.; Lemery, S.J.; Keegan, P.; Pazdur, R. FDA Approval Summary: Pembrolizumab for the Treatment of Microsatellite Instability-High Solid Tumors. Clin. Cancer Res. 2019, 25, 3753–3758. [Google Scholar] [CrossRef]
- Marabelle, A.; Le, D.T.; Ascierto, P.A.; Di Giacomo, A.M.; De Jesus-Acosta, A.; Delord, J.P.; Geva, R.; Gottfried, M.; Penel, N.; Hansen, A.R.; et al. Efficacy of Pembrolizumab in Patients With Noncolorectal High Microsatellite Instability/Mismatch Repair-Deficient Cancer: Results From the Phase II KEYNOTE-158 Study. J. Clin. Oncol. 2020, 38, 1–10. [Google Scholar] [CrossRef] [PubMed]
- Oaknin, A.; Tinker, A.V.; Gilbert, L.; Samouëlian, V.; Mathews, C.; Brown, J.; Barretina-Ginesta, M.P.; Moreno, V.; Gravina, A.; Abdeddaim, C.; et al. Clinical Activity and Safety of the Anti-Programmed Death 1 Monoclonal Antibody Dostarlimab for Patients With Recurrent or Advanced Mismatch Repair-Deficient Endometrial Cancer: A Nonrandomized Phase 1 Clinical Trial. JAMA Oncol. 2020, 6, 1766–1772. [Google Scholar] [CrossRef] [PubMed]
- Ascierto, P.A.; Del Vecchio, M.; Mandalá, M.; Gogas, H.; Arance, A.M.; Dalle, S.; Cowey, C.L.; Schenker, M.; Grob, J.J.; Chiarion-Sileni, V.; et al. Adjuvant nivolumab versus ipilimumab in resected stage IIIB-C and stage IV melanoma (CheckMate 238): 4-year results from a multicentre, double-blind, randomised, controlled, phase 3 trial. Lancet Oncol. 2020, 21, 1465–1477. [Google Scholar] [CrossRef]
- Borghaei, H.; Gettinger, S.; Vokes, E.E.; Chow, L.Q.M.; Burgio, M.A.; de Castro Carpeno, J.; Pluzanski, A.; Arrieta, O.; Frontera, O.A.; Chiari, R.; et al. Five-Year Outcomes From the Randomized, Phase III Trials CheckMate 017 and 057: Nivolumab Versus Docetaxel in Previously Treated Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2021, 39, 723–733. [Google Scholar] [CrossRef]
- Azad, N.S.; Gray, R.J.; Overman, M.J.; Schoenfeld, J.D.; Mitchell, E.P.; Zwiebel, J.A.; Sharon, E.; Streicher, H.; Li, S.; McShane, L.M.; et al. Nivolumab Is Effective in Mismatch Repair-Deficient Noncolorectal Cancers: Results From Arm Z1D-A Subprotocol of the NCI-MATCH (EAY131) Study. J. Clin. Oncol. 2020, 38, 214–222. [Google Scholar] [CrossRef]
- Hollebecque, A.; Chung, H.C.; de Miguel, M.J.; Italiano, A.; Machiels, J.P.; Lin, C.C.; Dhani, N.C.; Peeters, M.; Moreno, V.; Su, W.C.; et al. Safety and Antitumor Activity of α-PD-L1 Antibody as Monotherapy or in Combination with α-TIM-3 Antibody in Patients with Microsatellite Instability-High/Mismatch Repair-Deficient Tumors. Clin. Cancer Res. 2021, 27, 6393–6404. [Google Scholar] [CrossRef]
- Soumerai, T.E.; Donoghue, M.T.A.; Bandlamudi, C.; Srinivasan, P.; Chang, M.T.; Zamarin, D.; Cadoo, K.A.; Grisham, R.N.; O’Cearbhaill, R.E.; Tew, W.P.; et al. Clinical Utility of Prospective Molecular Characterization in Advanced Endometrial Cancer. Clin. Cancer Res. 2018, 24, 5939–5947. [Google Scholar] [CrossRef]
- Pakish, J.B.; Zhang, Q.; Chen, Z.; Liang, H.; Chisholm, G.B.; Yuan, Y.; Mok, S.C.; Broaddus, R.R.; Lu, K.H.; Yates, M.S. Immune Microenvironment in Microsatellite-Instable Endometrial Cancers: Hereditary or Sporadic Origin Matters. Clin. Cancer Res. 2017, 23, 4473–4481. [Google Scholar] [CrossRef]
- Fiamchander, N.C.; Ryan, N.A.J.; Walker, T.D.J.; Harries, L.; Bolton, J.; Bosse, T.; Evans, D.G.; Crosbie, E.J. Distinct Immunological Landscapes Characterize Inherited and Sporadic Mismatch Repair Deficient Endometrial Cancer. Front. Immunol. 2020, 10, 3023. [Google Scholar] [CrossRef] [PubMed]
- Zhao, S.; Chen, L.; Zang, Y.; Liu, W.; Liu, S.; Teng, F.; Xue, F.; Wang, Y. Endometrial cancer in Lynch syndrome. Int. J. Cancer. 2022, 150, 7–17. [Google Scholar] [CrossRef] [PubMed]
- Makker, V.; Taylor, M.H.; Aghajanian, C.; Oaknin, A.; Mier, J.; Cohn, A.L.; Romeo, M.; Bratos, R.; Brose, M.S.; DiSimone, C.; et al. Lenvatinib Plus Pembrolizumab in Patients With Advanced Endometrial Cancer. J. Clin. Oncol. 2020, 38, 2981–2992. [Google Scholar] [CrossRef] [PubMed]
- Arora, S.; Balasubramaniam, S.; Zhang, W.; Zhang, L.; Sridhara, R.; Spillman, D.; Mathai, J.P.; Scott, B.; Golding, S.J.; Coory, M.; et al. FDA Approval Summary: Pembrolizumab plus Lenvatinib for Endometrial Carcinoma, a Collaborative International Review under Project Orbis. Clin. Cancer Res. 2020, 26, 5062–5067. [Google Scholar] [CrossRef]
- Oaknin, A.; Gilbert, L.; Tinker, A.V.; Brown, J.; Mathews, C.; Press, J.; Sabatier, R.; O’Malley, D.M.; Samouelian, V.; Boni, V.; et al. Safety and antitumor activity of dostarlimab in patients with advanced or recurrent DNA mismatch repair deficient/microsatellite instability-high (dMMR/MSI-H) or proficient/stable (MMRp/MSS) endometrial cancer: Interim results from GARNET-a phase I, single-arm study. J. Immunother. Cancer 2022, 10, e003777. [Google Scholar] [CrossRef]
- Antill, Y.; Kok, P.S.; Robledo, K.; Yip, S.; Cummins, M.; Smith, D.; Spurdle, A.; Barnes, E.; Lee, Y.C.; Friedlander, M.; et al. Clinical activity of durvalumab for patients with advanced mismatch repair-deficient and repair-proficient endometrial cancer. A nonrandomized phase 2 clinical trial. J. Immunother. Cancer 2021, 9, e002255. [Google Scholar] [CrossRef]
- Konstantinopoulos, P.A.; Gockley, A.A.; Xiong, N.; Krasner, C.; Horowitz, N.; Campos, S.; Wright, A.A.; Liu, J.F.; Shea, M.; Yeku, O.; et al. Evaluation of Treatment With Talazoparib and Avelumab in Patients With Recurrent Mismatch Repair Proficient Endometrial Cancer. JAMA Oncol. 2022, 8, 1317–1322. [Google Scholar] [CrossRef]
- Ott, P.A.; Bang, Y.J.; Berton-Rigaud, D.; Elez, E.; Pishvaian, M.J.; Rugo, H.S.; Puzanov, I.; Mehnert, J.M.; Aung, K.L.; Lopez, J.; et al. Safety and Antitumor Activity of Pembrolizumab in Advanced Programmed Death Ligand 1-Positive Endometrial Cancer: Results From the KEYNOTE-028 Study. J. Clin. Oncol. 2017, 35, 2535–2541. [Google Scholar] [CrossRef]
- O’Malley, D.M.; Bariani, G.M.; Cassier, P.A.; Marabelle, A.; Hansen, A.R.; De Jesus Acosta, A.; Miller, W.H., Jr.; Safra, T.; Italiano, A.; Mileshkin, L.; et al. Pembrolizumab in Patients With Microsatellite Instability-High Advanced Endometrial Cancer: Results From the KEYNOTE-158 Study. J. Clin. Oncol. 2022, 40, 752–761. [Google Scholar] [CrossRef]
- Bellone, S.; Roque, D.M.; Siegel, E.R.; Buza, N.; Hui, P.; Bonazzoli, E.; Guglielmi, A.; Zammataro, L.; Nagarkatti, N.; Zaidi, S.; et al. A phase 2 evaluation of pembrolizumab for recurrent Lynch-like versus sporadic endometrial cancers with microsatellite instability. Cancer 2022, 128, 1206–1218. [Google Scholar] [CrossRef]
- Konstantinopoulos, P.A.; Luo, W.; Liu, J.F.; Gulhan, D.C.; Krasner, C.; Ishizuka, J.J.; Gockley, A.A.; Buss, M.; Growdon, W.B.; Crowe, H.; et al. Phase II Study of Avelumab in Patients with Mismatch Repair Deficient and Mismatch Repair Proficient Recurrent/Persistent Endometrial Cancer. J. Clin. Oncol. 2019, 37, 2786–2794. [Google Scholar] [CrossRef] [PubMed]
- Le, D.T.; Uram, J.N.; Wang, H.; Bartlett, B.R.; Kemberling, H.; Eyring, A.D.; Skora, A.D.; Luber, B.S.; Azad, N.S.; Laheru, D.; et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N. Engl. J. Med. 2015, 372, 2509–2520. [Google Scholar] [CrossRef] [PubMed]
- Patel, S.P.; Kurzrock, R. PD-L1 Expression as a Predictive Biomarker in Cancer Immunotherapy. Mol. Cancer Ther. 2015, 14, 847–856. [Google Scholar] [CrossRef]
- Grosso, J.; Horak, C.E.; Inzunza, D.; Cardona, D.M.; Simon, J.S.; Gupta, A.K.; Sankar, V.; Park, J.-S.; Kollia, G.; Taube, J.M.; et al. Association of tumor PD-L1 expression and immune biomarkers with clinical activity in patients (pts) with advanced solid tumors treated with nivolumab (anti-PD-1; BMS-936558; ONO-4538). J. Clin. Oncol. 2013, 31, 3016. [Google Scholar] [CrossRef]
- Gettinger, S.N.; Shepherd, F.A.; Antonia, S.J.; Brahmer, J.R.; Chow, L.Q.M.; Juergens, R.A.; Borghaei, H.; Shen, Y.; Harbison, C.; Alaparthy, S.; et al. First-line nivolumab (anti-PD-1; BMS-936558, ONO-4538) monotherapy in advanced NSCLC: Safety, efficacy, and correlation of outcomes with PD-L1 status. J. Clin. Oncol. 2014, 32, 8024. [Google Scholar] [CrossRef]
- Muro, K.; Chung, H.C.; Shankaran, V.; Geva, R.; Catenacci, D.; Gupta, S.; Eder, J.P.; Golan, T.; Le, D.T.; Burtness, B.; et al. Pembrolizumab for patients with PD-L1-positive advanced gastric cancer (KEYNOTE-012): A multicentre, open-label, phase 1b trial. Lancet Oncol. 2016, 17, 717–726. [Google Scholar] [CrossRef] [PubMed]
- Bellmunt, J.; de Wit, R.; Vaughn, D.J.; Fradet, Y.; Lee, J.L.; Fong, L.; Vogelzang, N.J.; Climent, M.A.; Petrylak, D.P.; Choueiri, T.K.; et al. Pembrolizumab as Second-Line Therapy for Advanced Urothelial Carcinoma. N. Engl. J. Med. 2017, 376, 1015–1026. [Google Scholar] [CrossRef] [PubMed]
- Herbst, R.S.; Baas, P.; Kim, D.W.; Felip, E.; Pérez-Gracia, J.L.; Han, J.Y.; Molina, J.; Kim, J.H.; Arvis, C.D.; Ahn, M.J.; et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): A randomised controlled trial. Lancet 2016, 387, 1540–1550. [Google Scholar] [CrossRef]
- Goyal, G.; Lau, D.; Nagle, A.M.; Vassallo, R.; Rech, K.L.; Ryu, J.H.; Davidge-Pitts, C.J.; Tobin, W.O.; Koster, M.J.; Bennani, N.N.; et al. Tumor mutational burden and other predictive immunotherapy markers in histiocytic neoplasms. Blood. 2019, 133, 1607–1610. [Google Scholar] [CrossRef]
- Yarchoan, M.; Hopkins, A.; Jaffee, E.M. Tumor Mutational Burden and Response Rate to PD-1 Inhibition. N. Engl. J. Med. 2017, 377, 2500–2501. [Google Scholar] [CrossRef]
- Snyder, A.; Makarov, V.; Merghoub, T.; Yuan, J.; Zaretsky, J.M.; Desrichard, A.; Walsh, L.A.; Postow, M.A.; Wong, P.; Ho, T.S.; et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 2014, 371, 2189–2199. [Google Scholar] [CrossRef]
- Chalmers, Z.R.; Connelly, C.F.; Fabrizio, D.; Gay, L.; Ali, S.M.; Ennis, R.; Schrock, A.; Campbell, B.; Shlien, A.; Chmielecki, J.; et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017, 9, 34. [Google Scholar] [CrossRef] [PubMed]
- Sudo, T.; Nishida, R.; Kawahara, A.; Saisho, K.; Mimori, K.; Yamada, A.; Mizoguchi, A.; Kadoya, K.; Matono, S.; Mori, N.; et al. Clinical Impact of Tumor-Infiltrating Lymphocytes in Esophageal Squamous Cell Carcinoma. Ann. Surg. Oncol. 2017, 24, 3763–3770. [Google Scholar] [CrossRef] [PubMed]
- Stanton, S.E.; Disis, M.L. Clinical significance of tumor-infiltrating lymphocytes in breast cancer. J. Immunother. Cancer. 2016, 4, 59. [Google Scholar] [CrossRef] [PubMed]
- De Jong, R.A.; Leffers, N.; Boezen, H.M.; ten Hoor, K.A.; van der Zee, A.G.; Hollema, H.; Nijman, H.W. Presence of tumor-infiltrating lymphocytes is an independent prognostic factor in type I and II endometrial cancer. Gynecol. Oncol. 2009, 114, 105–110. [Google Scholar] [CrossRef]
- Wei, W.; Ban, X.; Yang, F.; Li, J.; Cheng, X.; Zhang, R.; Huang, X.; Huang, Y.; Li, Q.; Qiu, Y.; et al. Phase II trial of efficacy, safety and biomarker analysis of sintilimab plus anlotinib for patients with recurrent or advanced endometrial cancer. J. Immunother. Cancer 2022, 10, e004338. [Google Scholar] [CrossRef]
- Chabanon, R.M.; Rouanne, M.; Lord, C.J.; Soria, J.C.; Pasero, P.; Postel-Vinay, S. Targeting the DNA damage response in immuno-oncology: Developments and opportunities. Nat. Rev. Cancer. 2021, 21, 701–717. [Google Scholar] [CrossRef]
- Staniszewska, A.D.; Armenia, J.; King, M.; Michaloglou, C.; Reddy, A.; Singh, M.; San Martin, M.; Prickett, L.; Wilson, Z.; Proia, T.; et al. PARP inhibition is a modulator of anti-tumor immune response in BRCA-deficient tumors. Oncoimmunology 2022, 11, 2083755. [Google Scholar] [CrossRef]
- Li, S.; Wang, L.; Wang, Y.; Zhang, C.; Hong, Z.; Han, Z. The synthetic lethality of targeting cell cycle checkpoints and PARPs in cancer treatment. J. Hematol. Oncol. 2022, 15, 147. [Google Scholar] [CrossRef]
- Lord, C.J.; Ashworth, A. PARP inhibitors: Synthetic lethality in the clinic. Science. 2017, 355, 1152–1158. [Google Scholar] [CrossRef]
- Pilié, P.G.; Tang, C.; Mills, G.B.; Yap, T.A. State-of-the-art strategies for targeting the DNA damage response in cancer. Nat. Rev. Clin. Oncol. 2019, 16, 81–104. [Google Scholar] [CrossRef]
- Post, C.C.B.; Westermann, A.M.; Boere, I.A.; Witteveen, P.O.; Ottevanger, P.B.; Sonke, G.S.; Lalisang, R.I.; Putter, H.; Meershoek-Klein Kranenbarg, E.; Braak, J.; et al. Efficacy and safety of durvalumab with olaparib in metastatic or recurrent endometrial cancer (phase II DOMEC trial). Gynecol. Oncol. 2022, 165, 223–229. [Google Scholar] [CrossRef]
- Pitter, K.L.; Casey, D.L.; Lu, Y.C.; Hannum, M.; Zhang, Z.; Song, X.; Pecorari, I.; McMillan, B.; Ma, J.; Samstein, R.M.; et al. Pathogenic ATM Mutations in Cancer and a Genetic Basis for Radiotherapeutic Efficacy. J. Natl. Cancer Inst. 2021, 113, 266–273. [Google Scholar] [CrossRef]
- Durant, S.T.; Zheng, L.; Wang, Y.; Chen, K.; Zhang, L.; Zhang, T.; Yang, Z.; Riches, L.; Trinidad, A.G.; Fok, J.H.L.; et al. The brain-penetrant clinical ATM inhibitor AZD1390 radiosensitizes and improves survival of preclinical brain tumor models. Sci. Adv. 2018, 4, eaat1719. [Google Scholar] [CrossRef]
- Zimmermann, A.; Zenke, F.T.; Chiu, L.Y.; Dahmen, H.; Pehl, U.; Fuchss, T.; Grombacher, T.; Blume, B.; Vassilev, L.T.; Blaukat, A. A New Class of Selective ATM Inhibitors as Combination Partners of DNA Double-Strand Break Inducing Cancer Therapies. Mol. Cancer Ther. 2022, 21, 859–870. [Google Scholar] [CrossRef]
- Waqar, S.N.; Robinson, C.; Olszanski, A.J.; Spira, A.; Hackmaster, M.; Lucas, L.; Sponton, L.; Jin, H.; Hering, U.; Cronier, D.; et al. Phase I trial of ATM inhibitor M3541 in combination with palliative radiotherapy in patients with solid tumors. Investig. New Drugs 2022, 40, 596–605. [Google Scholar] [CrossRef]
- Scheper, J.; Hildebrand, L.S.; Faulhaber, E.M.; Deloch, L.; Gaipl, U.S.; Symank, J.; Fietkau, R.; Distel, L.V.; Hecht, M.; Jost, T. Tumor-specific radiosensitizing effect of the ATM inhibitor AZD0156 in melanoma cells with low toxicity to healthy fibroblasts. Strahlenther. Onkol. 2022. [Google Scholar] [CrossRef] [PubMed]
- Davis, S.L.; Hartman, S.J.; Bagby, S.M.; Schlaepfer, M.; Yacob, B.W.; Tse, T.; Simmons, D.M.; Diamond, J.R.; Lieu, C.H.; Leal, A.D.; et al. ATM kinase inhibitor AZD0156 in combination with irinotecan and 5-fluorouracil in preclinical models of colorectal cancer. BMC Cancer 2022, 22, 1107. [Google Scholar] [CrossRef] [PubMed]
- Koneru, B.; Farooqi, A.; Nguyen, T.H.; Chen, W.H.; Hindle, A.; Eslinger, C.; Makena, M.R.; Burrow, T.A.; Wilson, J.; Smith, A.; et al. ALT neuroblastoma chemoresistance due to telomere dysfunction-induced ATM activation is reversible with ATM inhibitor AZD0156. Sci. Transl. Med. 2021, 13, eabd5750. [Google Scholar] [CrossRef]
- Takeuchi, M.; Tanikawa, M.; Nagasaka, K.; Oda, K.; Kawata, Y.; Oki, S.; Agapiti, C.; Sone, K.; Miyagawa, Y.; Hiraike, H.; et al. Anti-Tumor Effect of Inhibition of DNA Damage Response Proteins, ATM and ATR, in Endometrial Cancer Cells. Cancers 2019, 11, 1913. [Google Scholar] [CrossRef] [PubMed]
- Teng, P.N.; Bateman, N.W.; Darcy, K.M.; Hamilton, C.A.; Maxwell, G.L.; Bakkenist, C.J.; Conrads, T.P. Pharmacologic inhibition of ATR and ATM offers clinically important distinctions to enhancing platinum or radiation response in ovarian, endometrial, and cervical cancer cells. Gynecol. Oncol. 2015, 136, 554–561. [Google Scholar] [CrossRef]
- Jobson, A.G.; Lountos, G.T.; Lorenzi, P.L.; Llamas, J.; Connelly, J.; Cerna, D.; Tropea, J.E.; Onda, A.; Zoppoli, G.; Kondapaka, S.; et al. Cellular inhibition of checkpoint kinase 2 (Chk2) and potentiation of camptothecins and radiation by the novel Chk2 inhibitor PV1019 [7-nitro-1H-indole-2-carboxylic acid {4-[1-(guanidinohydrazone)-ethyl]-phenyl}-amide]. J. Pharmacol. Exp. Ther. 2009, 331, 816–826. [Google Scholar] [CrossRef]
- Wang, L.; Wang, Y.; Chen, A.; Jalali, A.; Liu, S.; Guo, Y.; Na, S.; Nakshatri, H.; Li, B.Y.; Yokota, H. Effects of a checkpoint kinase inhibitor, AZD7762, on tumor suppression and bone remodeling. Int. J. Oncol. 2018, 53, 1001–1012. [Google Scholar] [CrossRef] [PubMed]
- Seto, T.; Esaki, T.; Hirai, F.; Arita, S.; Nosaki, K.; Makiyama, A.; Kometani, T.; Fujimoto, C.; Hamatake, M.; Takeoka, H.; et al. Phase I, dose-escalation study of AZD7762 alone and in combination with gemcitabine in Japanese patients with advanced solid tumours. Cancer Chemother. Pharmacol. 2013, 72, 619–627. [Google Scholar] [CrossRef] [PubMed]
- Tian, Y.; Wang, Y.; Xu, S.; Guan, C.; Zhang, Q.; Li, W. The Expression and Therapeutic Potential of Checkpoint Kinase 2 in Laryngeal Squamous Cell Carcinoma. Drug Des. Devel. Ther. 2020, 14, 2613–2622. [Google Scholar] [CrossRef] [PubMed]
- Hsieh, C.C.; Hsu, S.H.; Lin, C.Y.; Liaw, H.J.; Li, T.W.; Jiang, K.Y.; Chiang, N.J.; Chen, S.H.; Lin, B.W.; Chen, P.C.; et al. CHK2 activation contributes to the development of oxaliplatin resistance in colorectal cancer. Br. J. Cancer. 2022, 127, 1615–1628. [Google Scholar] [CrossRef] [PubMed]
- Galal, S.A.; Khairat, S.H.M.; Ali, H.I.; Shouman, S.A.; Attia, Y.M.; Ali, M.M.; Mahmoud, A.E.; Abdel-Halim, A.H.; Fyiad, A.A.; Tabll, A.; et al. Part II: New candidates of pyrazole-benzimidazole conjugates as checkpoint kinase 2 (Chk2) inhibitors. Eur. J. Med. Chem. 2018, 144, 859–873. [Google Scholar] [CrossRef]
- Galal, S.A.; Khattab, M.; Shouman, S.A.; Ramadan, R.; Kandil, O.M.; Kandil, O.M.; Tabll, A.; El Abd, Y.S.; El-Shenawy, R.; Attia, Y.M.; et al. Part III: Novel checkpoint kinase 2 (Chk2) inhibitors; design, synthesis and biological evaluation of pyrimidine-benzimidazole conjugates. Eur. J. Med. Chem. 2018, 146, 687–708. [Google Scholar] [CrossRef]
- Park, S.J.; Chang, S.J.; Suh, D.H.; Kong, T.W.; Song, H.; Kim, T.H.; Kim, J.W.; Kim, H.S.; Lee, S.J. A phase IA dose-escalation study of PHI-101, a new checkpoint kinase 2 inhibitor, for platinum-resistant recurrent ovarian cancer. BMC Cancer 2022, 22, 28. [Google Scholar] [CrossRef]
- Shapiro, G.I.; Wesolowski, R.; Devoe, C.; Lord, S.; Pollard, J.; Hendriks, B.S.; Falk, M.; Diaz-Padilla, I.; Plummer, R.; Yap, T.A. Phase 1 study of the ATR inhibitor berzosertib in combination with cisplatin in patients with advanced solid tumours. Br. J. Cancer 2021, 125, 520–527. [Google Scholar] [CrossRef]
- Middleton, M.R.; Dean, E.; Evans, T.R.J.; Shapiro, G.I.; Pollard, J.; Hendriks, B.S.; Falk, M.; Diaz-Padilla, I.; Plummer, R. Phase 1 study of the ATR inhibitor berzosertib (formerly M6620, VX-970) combined with gemcitabine ± cisplatin in patients with advanced solid tumours. Br. J. Cancer 2021, 125, 510–519. [Google Scholar] [CrossRef] [PubMed]
- Telli, M.L.; Tolaney, S.M.; Shapiro, G.I.; Middleton, M.; Lord, S.R.; Arkenau, H.T.; Tutt, A.; Abramson, V.; Dean, E.; Haddad, T.C.; et al. Phase 1b study of berzosertib and cisplatin in patients with advanced triple-negative breast cancer. NPJ Breast Cancer 2022, 8, 45. [Google Scholar] [CrossRef] [PubMed]
- Plummer, R.; Dean, E.; Arkenau, H.T.; Redfern, C.; Spira, A.I.; Melear, J.M.; Chung, K.Y.; Ferrer-Playan, J.; Goddemeier, T.; Locatelli, G.; et al. A phase 1b study evaluating the safety and preliminary efficacy of berzosertib in combination with gemcitabine in patients with advanced non-small cell lung cancer. Lung Cancer 2022, 163, 19–26. [Google Scholar] [CrossRef] [PubMed]
- Pal, S.K.; Frankel, P.H.; Mortazavi, A.; Milowsky, M.; Vaishampayan, U.; Parikh, M.; Lyou, Y.; Weng, P.; Parikh, R.; Teply, B.; et al. Effect of Cisplatin and Gemcitabine With or Without Berzosertib in Patients With Advanced Urothelial Carcinoma: A Phase 2 Randomized Clinical Trial. JAMA Oncol. 2021, 7, 1536–1543. [Google Scholar] [CrossRef]
- Shi, Q.; Shen, L.Y.; Dong, B.; Fu, H.; Kang, X.Z.; Yang, Y.B.; Dai, L.; Yan, W.P.; Xiong, H.C.; Liang, Z.; et al. The identification of the ATR inhibitor VE-822 as a therapeutic strategy for enhancing cisplatin chemosensitivity in esophageal squamous cell carcinoma. Cancer Lett. 2018, 432, 56–68. [Google Scholar] [CrossRef]
- Combès, E.; Andrade, A.F.; Tosi, D.; Michaud, H.A.; Coquel, F.; Garambois, V.; Desigaud, D.; Jarlier, M.; Coquelle, A.; Pasero, P.; et al. Inhibition of Ataxia-Telangiectasia Mutated and RAD3-Related (ATR) Overcomes Oxaliplatin Resistance and Promotes Antitumor Immunity in Colorectal Cancer. Cancer Res. 2019, 79, 2933–2946. [Google Scholar] [CrossRef]
- Yap, T.A.; O’Carrigan, B.; Penney, M.S.; Lim, J.S.; Brown, J.S.; de Miguel Luken, M.J.; Tunariu, N.; Perez-Lopez, R.; Rodrigues, D.N.; Riisnaes, R.; et al. Phase I Trial of First-in-Class ATR Inhibitor M6620 (VX-970) as Monotherapy or in Combination With Carboplatin in Patients With Advanced Solid Tumors. J. Clin. Oncol. 2020, 38, 3195–3204. [Google Scholar] [CrossRef]
- Gibson, W.J.; Hoivik, E.A.; Halle, M.K.; Taylor-Weiner, A.; Cherniack, A.D.; Berg, A.; Holst, F.; Zack, T.I.; Werner, H.M.J.; Staby, K.M.; et al. The genomic landscape and evolution of endometrial carcinoma progression and abdominopelvic metastasis. Nat. Genet. 2016, 48, 848–855. [Google Scholar] [CrossRef]
- Guan, B.; Mao, T.-L.; Panuganti, P.K.; Kuhn, E.; Kurman, R.J.; Maeda, D.; Chen, E.; Jeng, Y.-M.; Wang, T.-L.; Shih, I.-M. Mutation and Loss of Expression of ARID1A in Uterine Low-grade Endometrioid Carcinoma. Am. J. Surg. Pathol. 2011, 35, 625–632. [Google Scholar] [CrossRef]
- Wiegand, K.C.; Lee, A.F.; Al-Agha, O.M.; Chow, C.; Kalloger, S.E.; Scott, D.W.; Steidl, C.; Wiseman, S.M.; Gascoyne, R.D.; Gilks, B.; et al. Loss of BAF250a (ARID1A) is frequent in high-grade endometrial carcinomas. J. Pathol. 2011, 224, 328–333. [Google Scholar] [CrossRef]
- Thomas, A.; Redon, C.E.; Sciuto, L.; Padiernos, E.; Ji, J.; Lee, M.J.; Yuno, A.; Lee, S.; Zhang, Y.; Tran, L.; et al. Phase I Study of ATR Inhibitor M6620 in Combination With Topotecan in Patients With Advanced Solid Tumors. J. Clin. Oncol. 2018, 36, 1594–1602. [Google Scholar] [CrossRef] [PubMed]
- Yap, T.A.; Krebs, M.G.; Postel-Vinay, S.; El-Khouiery, A.; Soria, J.C.; Lopez, J.; Berges, A.; Cheung, S.Y.A.; Irurzun-Arana, I.; Goldwin, A.; et al. Ceralasertib (AZD6738), an Oral ATR Kinase Inhibitor, in Combination with Carboplatin in Patients with Advanced Solid Tumors: A Phase I Study. Clin. Cancer Res. 2021, 27, 5213–5224. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.T.; Smith, S.A.; Mortimer, P.; Loembé, A.B.; Cho, H.; Kim, K.M.; Smith, C.; Willis, S.; Irurzun-Arana, I.; Berges, A.; et al. Phase I Study of Ceralasertib (AZD6738), a Novel DNA Damage Repair Agent, in Combination with Weekly Paclitaxel in Refractory Cancer. Clin. Cancer Res. 2021, 27, 4700–4709. [Google Scholar] [CrossRef] [PubMed]
- Wengner, A.M.; Siemeister, G.; Lücking, U.; Lefranc, J.; Wortmann, L.; Lienau, P.; Bader, B.; Bömer, U.; Moosmayer, D.; Eberspächer, U.; et al. The Novel ATR Inhibitor BAY 1895344 Is Efficacious as Monotherapy and Combined with DNA Damage-Inducing or Repair-Compromising Therapies in Preclinical Cancer Models. Mol. Cancer Ther. 2020, 19, 26–38. [Google Scholar] [CrossRef] [PubMed]
- Yap, T.A.; Tan, D.S.P.; Terbuch, A.; Caldwell, R.; Guo, C.; Goh, B.C.; Heong, V.; Haris, N.R.M.; Bashir, S.; Drew, Y.; et al. First-in-Human Trial of the Oral Ataxia Telangiectasia and RAD3-Related (ATR) Inhibitor BAY 1895344 in Patients with Advanced Solid Tumors. Cancer Discov. 2021, 11, 80–91. [Google Scholar] [CrossRef]
- Jo, U.; Senatorov, I.S.; Zimmermann, A.; Saha, L.K.; Murai, Y.; Kim, S.H.; Rajapakse, V.N.; Elloumi, F.; Takahashi, N.; Schultz, C.W.; et al. Novel and Highly Potent ATR Inhibitor M4344 Kills Cancer Cells With Replication Stress, and Enhances the Chemotherapeutic Activity of Widely Used DNA Damaging Agents. Mol. Cancer Ther. 2021, 20, 1431–1441. [Google Scholar] [CrossRef]
- Laroche-Clary, A.; Lucchesi, C.; Rey, C.; Verbeke, S.; Bourdon, A.; Chaire, V.; Algéo, M.P.; Cousin, S.; Toulmonde, M.; Vélasco, V.; et al. CHK1 inhibition in soft-tissue sarcomas: Biological and clinical implications. Ann. Oncol. 2018, 29, 1023–1029. [Google Scholar] [CrossRef]
- Mani, C.; Jonnalagadda, S.; Lingareddy, J.; Awasthi, S.; Gmeiner, W.H.; Palle, K. Prexasertib treatment induces homologous recombination deficiency and synergizes with olaparib in triple-negative breast cancer cells. Breast Cancer Res. 2019, 21, 104. [Google Scholar] [CrossRef]
- Hong, D.S.; Moore, K.; Patel, M.; Grant, S.C.; Burris, H.A., 3rd; William, W.N., Jr.; Jones, S.; Meric-Bernstam, F.; Infante, J.; Golden, L.; et al. Evaluation of Prexasertib, a Checkpoint Kinase 1 Inhibitor, in a Phase Ib Study of Patients with Squamous Cell Carcinoma. Clin. Cancer Res. 2018, 24, 3263–3272. [Google Scholar] [CrossRef]
- Yang, E.S.; Deutsch, E.; Mehmet, A.; Fayette, J.; Tao, Y.; Nabell, L.; Spencer, S.A.; Wang, X.A.; Spoljoric, E.A.; Zhang, W.; et al. A Phase 1b trial of prexasertib in combination with chemoradiation in patients with locally advanced head and neck squamous cell carcinoma. Radiother. Oncol. 2021, 157, 203–209. [Google Scholar] [CrossRef]
- Moore, K.N.; Hong, D.S.; Patel, M.R.; Pant, S.; Ulahannan, S.V.; Jones, S.; Meric-Bernstam, F.; Wang, J.S.; Aljumaily, R.; Hamilton, E.P.; et al. A Phase 1b Trial of Prexasertib in Combination with Standard-of-Care Agents in Advanced or Metastatic Cancer. Target Oncol. 2021, 16, 569–589. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.M.; Nair, J.; Zimmer, A.; Lipkowitz, S.; Annunziata, C.M.; Merino, M.J.; Swisher, E.M.; Harrell, M.I.; Trepel, J.B.; Lee, M.J.; et al. Prexasertib, a cell cycle checkpoint kinase 1 and 2 inhibitor, in BRCA wild-type recurrent high-grade serous ovarian cancer: A first-in-class proof-of-concept phase 2 study. Lancet. Oncol. 2018, 19, 207–215. [Google Scholar] [CrossRef]
- Konstantinopoulos, P.A.; Lee, J.M.; Gao, B.; Miller, R.; Lee, J.Y.; Colombo, N.; Vergote, I.; Credille, K.M.; Young, S.R.; McNeely, S.; et al. A Phase 2 study of prexasertib (LY2606368) in platinum resistant or refractory recurrent ovarian cancer. Gynecol. Oncol. 2022, 167, 213–225. [Google Scholar] [CrossRef]
- Gatti-Mays, M.E.; Karzai, F.H.; Soltani, S.N.; Zimmer, A.; Green, J.E.; Lee, M.J.; Trepel, J.B.; Yuno, A.; Lipkowitz, S.; Nair, J.; et al. A Phase II Single Arm Pilot Study of the CHK1 Inhibitor Prexasertib (LY2606368) in BRCA Wild-Type, Advanced Triple-Negative Breast Cancer. Oncologist 2020, 25, 1013-e1824. [Google Scholar] [CrossRef] [PubMed]
- Byers, L.A.; Navarro, A.; Schaefer, E.; Johnson, M.; Özgüroğlu, M.; Han, J.Y.; Bondarenko, I.; Cicin, I.; Dragnev, K.H.; Abel, A.; et al. A Phase II Trial of Prexasertib (LY2606368) in Patients With Extensive-Stage Small-Cell Lung Cancer. Clin. Lung Cancer 2021, 22, 531–540. [Google Scholar] [CrossRef] [PubMed]
- Do, K.T.; Kochupurakkal, B.; Kelland, S.; de Jonge, A.; Hedglin, J.; Powers, A.; Quinn, N.; Gannon, C.; Vuong, L.; Parmar, K.; et al. Phase 1 Combination Study of the CHK1 Inhibitor Prexasertib and the PARP Inhibitor Olaparib in High-grade Serous Ovarian Cancer and Other Solid Tumors. Clin. Cancer Res. 2021, 27, 4710–4716. [Google Scholar] [CrossRef] [PubMed]
- Hong, D.S.; Moore, K.N.; Bendell, J.C.; Karp, D.D.; Wang, J.S.; Ulahannan, S.V.; Jones, S.; Wu, W.; Donoho, G.P.; Ding, Y.; et al. Preclinical Evaluation and Phase Ib Study of Prexasertib, a CHK1 Inhibitor, and Samotolisib (LY3023414), a Dual PI3K/mTOR Inhibitor. Clin. Cancer Res. 2021, 27, 1864–1874. [Google Scholar] [CrossRef]
- Patties, I.; Kallendrusch, S.; Böhme, L.; Kendzia, E.; Oppermann, H.; Gaunitz, F.; Kortmann, R.D.; Glasow, A. The Chk1 inhibitor SAR-020106 sensitizes human glioblastoma cells to irradiation, to temozolomide, and to decitabine treatment. J. Exp. Clin. Cancer Res. 2019, 38, 420. [Google Scholar] [CrossRef]
- Chung, S.W.; Kim, G.C.; Kweon, S.; Lee, H.; Choi, J.U.; Mahmud, F.; Chang, H.W.; Kim, J.W.; Son, W.C.; Kim, S.Y.; et al. Metronomic oral doxorubicin in combination of Chk1 inhibitor MK-8776 for p53-deficient breast cancer treatment. Biomaterials 2018, 182, 35–43. [Google Scholar] [CrossRef]
- Liu, J.F.; Xiong, N.; Campos, S.M.; Wright, A.A.; Krasner, C.; Schumer, S.; Horowitz, N.; Veneris, J.; Tayob, N.; Morrissey, S.; et al. Phase II Study of the WEE1 Inhibitor Adavosertib in Recurrent Uterine Serous Carcinoma. J. Clin. Oncol. 2021, 39, 1531–1539. [Google Scholar] [CrossRef]
- Sanai, N.; Li, J.; Boerner, J.; Stark, K.; Wu, J.; Kim, S.; Derogatis, A.; Mehta, S.; Dhruv, H.D.; Heilbrun, L.K.; et al. Phase 0 Trial of AZD1775 in First-Recurrence Glioblastoma Patients. Clin. Cancer Res. 2018, 24, 3820–3828. [Google Scholar] [CrossRef]
- Seligmann, J.F.; Fisher, D.J.; Brown, L.C.; Adams, R.A.; Graham, J.; Quirke, P.; Richman, S.D.; Butler, R.; Domingo, E.; Blake, A.; et al. Inhibition of WEE1 Is Effective in TP53- and RAS-Mutant Metastatic Colorectal Cancer: A Randomized Trial (FOCUS4-C) Comparing Adavosertib (AZD1775) With Active Monitoring. J. Clin. Oncol. 2021, 39, 3705–3715. [Google Scholar] [CrossRef]
- Takebe, N.; Naqash, A.R.; O’Sullivan Coyne, G.; Kummar, S.; Do, K.; Bruns, A.; Juwara, L.; Zlott, J.; Rubinstein, L.; Piekarz, R.; et al. Safety, Antitumor Activity, and Biomarker Analysis in a Phase I Trial of the Once-daily Wee1 Inhibitor Adavosertib (AZD1775) in Patients with Advanced Solid Tumors. Clin. Cancer Res. 2021, 27, 3834–3844. [Google Scholar] [CrossRef]
- Fu, S.; Yao, S.; Yuan, Y.; Previs, R.A.; Elias, A.D.; Carvajal, R.D.; George, T.J.; Yuan, Y.; Yu, L.; Westin, S.N.; et al. Multicenter Phase II Trial of the WEE1 Inhibitor Adavosertib in Refractory Solid Tumors Harboring CCNE1 Amplification. J. Clin. Oncol. 2022, 41, 1725–1734. [Google Scholar] [CrossRef]
- Xu, H.; George, E.; Kinose, Y.; Kim, H.; Shah, J.B.; Peake, J.D.; Ferman, B.; Medvedev, S.; Murtha, T.; Barger, C.J.; et al. CCNE1 copy number is a biomarker for response to combination WEE1-ATR inhibition in ovarian and endometrial cancer models. Cell Rep. Med. 2021, 2, 100394. [Google Scholar] [CrossRef] [PubMed]
- Nakayama, K.; Rahman, M.T.; Rahman, M.; Nakamura, K.; Ishikawa, M.; Katagiri, H.; Sato, E.; Ishibashi, T.; Iida, K.; Ishikawa, N.; et al. CCNE1 amplification is associated with aggressive potential in endometrioid endometrial carcinomas. Int. J. Oncol. 2016, 48, 506–516. [Google Scholar] [CrossRef] [PubMed]
- Zhao, S.; Choi, M.; Overton, J.D.; Bellone, S.; Roque, D.M.; Cocco, E.; Guzzo, F.; English, D.P.; Varughese, J.; Gasparrini, S.; et al. Landscape of somatic single-nucleotide and copy-number mutations in uterine serous carcinoma. Proc. Natl. Acad. Sci. USA 2013, 110, 2916–2921. [Google Scholar] [CrossRef] [PubMed]
- Cherniack, A.D.; Shen, H.; Walter, V.; Stewart, C.; Murray, B.A.; Bowlby, R.; Hu, X.; Ling, S.; Soslow, R.A.; Broaddus, R.R.; et al. Integrated Molecular Characterization of Uterine Carcinosarcoma. Cancer Cell. 2017, 31, 411–423. [Google Scholar] [CrossRef]
- Keenan, T.E.; Li, T.; Vallius, T.; Guerriero, J.L.; Tayob, N.; Kochupurakkal, B.; Davis, J.; Pastorello, R.; Tahara, R.K.; Anderson, L.; et al. Clinical Efficacy and Molecular Response Correlates of the WEE1 Inhibitor Adavosertib Combined with Cisplatin in Patients with Metastatic Triple-Negative Breast Cancer. Clin. Cancer Res. 2021, 27, 983–991. [Google Scholar] [CrossRef]
- Kato, H.; de Souza, P.; Kim, S.W.; Lickliter, J.D.; Naito, Y.; Park, K.; Kumar, S.; Mugundu, G.M.; Bang, Y.J. Safety, Pharmacokinetics, and Clinical Activity of Adavosertib in Combination with Chemotherapy in Asian Patients with Advanced Solid Tumors: Phase Ib Study. Target Oncol. 2020, 15, 75–84. [Google Scholar] [CrossRef]
- Oza, A.M.; Estevez-Diz, M.; Grischke, E.M.; Hall, M.; Marmé, F.; Provencher, D.; Uyar, D.; Weberpals, J.I.; Wenham, R.M.; Laing, N.; 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. Clin. Cancer Res. 2020, 26, 4767–4776. [Google Scholar] [CrossRef] [PubMed]
- Lheureux, S.; Cristea, M.C.; Bruce, J.P.; Garg, S.; Cabanero, M.; Mantia-Smaldone, G.; Olawaiye, A.B.; Ellard, S.L.; Weberpals, J.I.; Wahner Hendrickson, A.E.; et al. Adavosertib plus gemcitabine for platinum-resistant or platinum-refractory recurrent ovarian cancer: A double-blind, randomised, placebo-controlled, phase 2 trial. Lancet 2021, 397, 281–292. [Google Scholar] [CrossRef] [PubMed]
- Chera, B.S.; Sheth, S.H.; Patel, S.A.; Goldin, D.; Douglas, K.E.; Green, R.L.; Shen, C.J.; Gupta, G.P.; Moore, D.T.; Grilley Olson, J.E.; et al. Phase 1 trial of adavosertib (AZD1775) in combination with concurrent radiation and cisplatin for intermediate-risk and high-risk head and neck squamous cell carcinoma. Cancer 2021, 127, 4447–4454. [Google Scholar] [CrossRef] [PubMed]
- Cuneo, K.C.; Morgan, M.A.; Sahai, V.; Schipper, M.J.; Parsels, L.A.; Parsels, J.D.; Devasia, T.; Al-Hawaray, M.; Cho, C.S.; Nathan, H.; et al. Dose Escalation Trial of the Wee1 Inhibitor Adavosertib (AZD1775) in Combination With Gemcitabine and Radiation for Patients With Locally Advanced Pancreatic Cancer. J. Clin. Oncol. 2019, 37, 2643–2650. [Google Scholar] [CrossRef] [PubMed]
- Italiano, A.; Infante, J.R.; Shapiro, G.I.; Moore, K.N.; LoRusso, P.M.; Hamilton, E.; Cousin, S.; Toulmonde, M.; Postel-Vinay, S.; Tolaney, S.; et al. Phase I study of the checkpoint kinase 1 inhibitor GDC-0575 in combination with gemcitabine in patients with refractory solid tumors. Ann. Oncol. 2018, 29, 1304–1311. [Google Scholar] [CrossRef]
- Stelloo, E.; Bosse, T.; Nout, R.A.; MacKay, H.J.; Church, D.N.; Nijman, H.W.; Leary, A.; Edmondson, R.J.; Powell, M.E.; Crosbie, E.J.; et al. Refining prognosis and identifying targetable pathways for high-risk endometrial cancer; a TransPORTEC initiative. Mod. Pathol. 2015, 28, 836–844. [Google Scholar] [CrossRef]
- Kommoss, S.; McConechy, M.K.; Kommoss, F.; Leung, S.; Bunz, A.; Magrill, J.; Britton, H.; Kommoss, F.; Grevenkamp, F.; Karnezis, A.; et al. Final validation of the ProMisE molecular classifier for endometrial carcinoma in a large population-based case series. Ann. Oncol. 2018, 29, 1180–1188. [Google Scholar] [CrossRef]
- Talhouk, A.; McConechy, M.K.; Leung, S.; Li-Chang, H.H.; Kwon, J.S.; Melnyk, N.; Yang, W.; Senz, J.; Boyd, N.; Karnezis, A.N.; et al. A clinically applicable molecular-based classification for endometrial cancers. Br. J. Cancer. 2015, 113, 299–310. [Google Scholar] [CrossRef]
- Bosse, T.; Nout, R.A.; McAlpine, J.N.; McConechy, M.K.; Britton, H.; Hussein, Y.R.; Gonzalez, C.; Ganesan, R.; Steele, J.C.; Harrison, B.T.; et al. Molecular Classification of Grade 3 Endometrioid Endometrial Cancers Identifies Distinct Prognostic Subgroups. Am. J. Surg. Pathol. 2018, 42, 561–568. [Google Scholar] [CrossRef]
- León-Castillo, A.; de Boer, S.M.; Powell, M.E.; Mileshkin, L.R.; Mackay, H.J.; Leary, A.; Nijman, H.W.; Singh, N.; Pollock, P.M.; Bessette, P.; et al. Molecular Classification of the PORTEC-3 Trial for High-Risk Endometrial Cancer: Impact on Prognosis and Benefit From Adjuvant Therapy. J. Clin. Oncol. 2020, 38, 3388–3397. [Google Scholar] [CrossRef]
- Randon, G.; Fucà, G.; Rossini, D.; Raimondi, A.; Pagani, F.; Perrone, F.; Tamborini, E.; Busico, A.; Peverelli, G.; Morano, F.; et al. Prognostic impact of ATM mutations in patients with metastatic colorectal cancer. Sci. Rep. 2019, 9, 2858. [Google Scholar] [CrossRef] [PubMed]
- Suh, K.J.; Ryu, H.S.; Lee, K.H.; Kim, H.; Min, A.; Kim, T.Y.; Yang, Y.; Lee, H.B.; Moon, H.G.; Han, S.W.; et al. Prognostic effects of abnormal DNA damage response protein expression in breast cancer. Breast Cancer Res. Treat. 2019, 175, 117–127. [Google Scholar] [CrossRef] [PubMed]
- Jha, J.; Singh, M.K.; Singh, L.; Pushker, N.; Bajaj, M.S.; Sen, S.; Kashyap, S. Prognostic relevance of ATM protein in uveal melanoma and its association with clinicopathological factors. Int. J. Clin. Oncol. 2019, 24, 1526–1535. [Google Scholar] [CrossRef]
- Joshi, M.; Grivas, P.; Mortazavi, A.; Monk, P.; Clinton, S.K.; Sue-Ann Woo, M.; Holder, S.L.; Drabick, J.J.; Yin, M. Alterations of DNA damage response genes correlate with response and overall survival in anti-PD-1/PD-L1-treated advanced urothelial cancer. Cancer Med. 2020, 9, 9365–9372. [Google Scholar] [CrossRef] [PubMed]
- Gachechiladze, M.; Skarda, J.; Bouchalova, K.; Soltermann, A.; Joerger, M. Predictive and Prognostic Value of DNA Damage Response Associated Kinases in Solid Tumors. Front. Oncol. 2020, 10, 581217. [Google Scholar] [CrossRef] [PubMed]
- Mhawech-Fauceglia, P.; Wang, D.; Kim, G.; Sharifian, M.; Chen, X.; Liu, Q.; Lin, Y.G.; Liu, S.; Pejovic, T. Expression of DNA repair proteins in endometrial cancer predicts disease outcome. Gynecol. Oncol. 2014, 132, 593–598. [Google Scholar] [CrossRef] [PubMed]
- Sun, L.; Wang, R.C.; Zhang, Q.; Guo, L.L. ATM mutations as an independent prognostic factor and potential biomarker for immune checkpoint therapy in endometrial cancer. Pathol. Res. Pract. 2020, 216, 153032. [Google Scholar] [CrossRef]
- McGranahan, N.; Furness, A.J.; Rosenthal, R.; Ramskov, S.; Lyngaa, R.; Saini, S.K.; Jamal-Hanjani, M.; Wilson, G.A.; Birkbak, N.J.; Hiley, C.T.; et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016, 351, 1463–1469. [Google Scholar] [CrossRef]
- Egger, T.; Bordignon, B.; Coquelle, A. A clinically relevant heterozygous ATR mutation sensitizes colorectal cancer cells to replication stress. Sci. Rep. 2022, 12, 5422. [Google Scholar] [CrossRef]
- Falchetti, M.; Saieva, C.; Lupi, R.; Masala, G.; Rizzolo, P.; Zanna, I.; Ceccarelli, K.; Sera, F.; Mariani-Costantini, R.; Nesi, G.; et al. Gastric cancer with high-level microsatellite instability: Target gene mutations, clinicopathologic features, and long-term survival. Hum. Pathol. 2008, 39, 925–932. [Google Scholar] [CrossRef]
- Vassileva, V.; Millar, A.; Briollais, L.; Chapman, W.; Bapat, B. Genes involved in DNA repair are mutational targets in endometrial cancers with microsatellite instability. Cancer Res. 2002, 62, 4095–4099. [Google Scholar] [PubMed]
- Fang, Y.; Tsao, C.C.; Goodman, B.K.; Furumai, R.; Tirado, C.A.; Abraham, R.T.; Wang, X.F. ATR functions as a gene dosage-dependent tumor suppressor on a mismatch repair-deficient background. EMBO J. 2004, 23, 3164–3174. [Google Scholar] [CrossRef] [PubMed]
- Lewis, K.A.; Mullany, S.; Thomas, B.; Chien, J.; Loewen, R.; Shridhar, V.; Cliby, W.A. Heterozygous ATR mutations in mismatch repair-deficient cancer cells have functional significance. Cancer Res. 2005, 65, 7091–7095. [Google Scholar] [CrossRef] [PubMed]
- Zighelboim, I.; Ali, S.; Lankes, H.A.; Backes, F.; Moore, K.; Mutch, D.; Robison, K.; Behbakht, K.; Waggoner, S.; Ghebre, R.G.; et al. Assessing the prognostic role of ATR mutation in endometrioid endometrial cancer: An NRG Oncology/Gynecologic Oncology Group study. Gynecol. Oncol. 2015, 138, 614–619. [Google Scholar] [CrossRef]
- Dinoi, G.; Mariani, A.; Martinelli, E.; Ciucci, A.; Zannoni, G.F.; Weaver, A.L.; Keeney, G.L.; Vasmatzis, G.; Anastasiadis, P.Z.; Fanfani, F.; et al. In search for biomarkers and potential drug targets for uterine serous endometrial cancer. J. Cancer Res. Clin. Oncol. 2021, 147, 1647–1658. [Google Scholar] [CrossRef]
- Lee, H.E.; Han, N.; Kim, M.A.; Lee, H.S.; Yang, H.K.; Lee, B.L.; Kim, W.H. DNA damage response-related proteins in gastric cancer: ATM, Chk2 and p53 expression and their prognostic value. Pathobiology 2014, 81, 25–35. [Google Scholar] [CrossRef] [PubMed]
- Eichenauer, T.; Federlein, F.; Möller, K.; Chirico, V.; Kind, S.; Lennartz, M.; Lutz, F.; Hube-Magg, C.; Höflmayer, D.; Fisch, M.; et al. High CHK2 protein expression is a strong and independent prognostic feature in ERG negative prostate cancer. Pathology 2020, 52, 421–430. [Google Scholar] [CrossRef] [PubMed]
- Magnussen, G.I.; Holm, R.; Emilsen, E.; Rosnes, A.K.; Slipicevic, A.; Flørenes, V.A. High expression of Wee1 is associated with poor disease-free survival in malignant melanoma: Potential for targeted therapy. PLoS ONE 2012, 7, e38254. [Google Scholar] [CrossRef]
- Magnussen, G.I.; Hellesylt, E.; Nesland, J.M.; Trope, C.G.; Flørenes, V.A.; Holm, R. High expression of wee1 is associated with malignancy in vulvar squamous cell carcinoma patients. BMC Cancer 2013, 13, 288. [Google Scholar] [CrossRef]
- Slipicevic, A.; Holth, A.; Hellesylt, E.; Tropé, C.G.; Davidson, B.; Flørenes, V.A. Wee1 is a novel independent prognostic marker of poor survival in post-chemotherapy ovarian carcinoma effusions. Gynecol. Oncol. 2014, 135, 118–124. [Google Scholar] [CrossRef]
- Ge, X.C.; Wu, F.; Li, W.T.; Zhu, X.J.; Liu, J.W.; Wang, B.L. Upregulation of WEE1 is a potential prognostic biomarker for patients with colorectal cancer. Oncol. Lett. 2017, 13, 4341–4348. [Google Scholar] [CrossRef] [PubMed]
- De Nonneville, A.; Finetti, P.; Birnbaum, D.; Mamessier, E.; Bertucci, F. WEE1 Dependency and Pejorative Prognostic Value in Triple-Negative Breast Cancer. Adv. Sci. 2021, 8, e2101030. [Google Scholar] [CrossRef] [PubMed]
- Music, D.; Dahlrot, R.H.; Hermansen, S.K.; Hjelmborg, J.; de Stricker, K.; Hansen, S.; Kristensen, B.W. Expression and prognostic value of the WEE1 kinase in gliomas. J. Neurooncol. 2016, 127, 381–389. [Google Scholar] [CrossRef] [PubMed]
- Qing, T.; Jun, T.; Lindblad, K.E.; Lujambio, A.; Marczyk, M.; Pusztai, L.; Huang, K.L. Diverse immune response of DNA damage repair-deficient tumors. Cell Rep. Med. 2021, 2, 100276. [Google Scholar] [CrossRef] [PubMed]
- Ballhausen, A.; Przybilla, M.J.; Jendrusch, M.; Haupt, S.; Pfaffendorf, E.; Seidler, F.; Witt, J.; Hernandez Sanchez, A.; Urban, K.; Draxlbauer, M.; et al. The shared frameshift mutation landscape of microsatellite-unstable cancers suggests immunoediting during tumor evolution. Nat. Commun. 2020, 11, 4740. [Google Scholar] [CrossRef]
- van Wilpe, S.; Simnica, D.; Slootbeek, P.; van Ee, T.; Pamidimarri Naga, S.; Gorris, M.A.J.; van der Woude, L.L.; Sultan, S.; Koornstra, R.H.T.; van Oort, I.M.; et al. Homologous recombination repair deficient prostate cancer represents an immunologically distinct subtype. Oncoimmunology 2022, 11, 2094133. [Google Scholar] [CrossRef]
- Zhou, Z.; Ding, Z.; Yuan, J.; Shen, S.; Jian, H.; Tan, Q.; Yang, Y.; Chen, Z.; Luo, Q.; Cheng, X.; et al. Homologous recombination deficiency (HRD) can predict the therapeutic outcomes of immuno-neoadjuvant therapy in NSCLC patients. J. Hematol. Oncol. 2022, 15, 62. [Google Scholar] [CrossRef]
- Chen, M.; Linstra, R.; van Vugt, M. Genomic instability, inflammatory signaling and response to cancer immunotherapy. Biochim. Biophys. Acta Rev. Cancer 2022, 1877, 188661. [Google Scholar] [CrossRef]
- Zheng, J.; Mo, J.; Zhu, T.; Zhuo, W.; Yi, Y.; Hu, S.; Yin, J.; Zhang, W.; Zhou, H.; Liu, Z. Comprehensive elaboration of the cGAS-STING signaling axis in cancer development and immunotherapy. Mol. Cancer 2020, 19, 133. [Google Scholar] [CrossRef]
- de Queiroz, N.; Xia, T.; Konno, H.; Barber, G.N. Ovarian Cancer Cells Commonly Exhibit Defective STING Signaling Which Affects Sensitivity to Viral Oncolysis. Mol. Cancer Res. 2019, 17, 974–986. [Google Scholar] [CrossRef]
- Reisländer, T.; Groelly, F.J.; Tarsounas, M. DNA Damage and Cancer Immunotherapy: A STING in the Tale. Mol. Cell. 2020, 80, 21–28. [Google Scholar] [CrossRef] [PubMed]
- Diamond, M.S.; Kinder, M.; Matsushita, H.; Mashayekhi, M.; Dunn, G.P.; Archambault, J.M.; Lee, H.; Arthur, C.D.; White, J.M.; Kalinke, U.; et al. Type I interferon is selectively required by dendritic cells for immune rejection of tumors. J. Exp. Med. 2011, 208, 1989–2003. [Google Scholar] [CrossRef] [PubMed]
- Reboulet, R.A.; Hennies, C.M.; Garcia, Z.; Nierkens, S.; Janssen, E.M. Prolonged antigen storage endows merocytic dendritic cells with enhanced capacity to prime anti-tumor responses in tumor-bearing mice. J. Immunol. 2010, 185, 3337–3347. [Google Scholar] [CrossRef]
- Lorenzi, S.; Mattei, F.; Sistigu, A.; Bracci, L.; Spadaro, F.; Sanchez, M.; Spada, M.; Belardelli, F.; Gabriele, L.; Schiavoni, G. Type I IFNs control antigen retention and survival of CD8α(+) dendritic cells after uptake of tumor apoptotic cells leading to cross-priming. J. Immunol. 2011, 186, 5142–5150. [Google Scholar] [CrossRef] [PubMed]
- MacNabb, B.W.; Tumuluru, S.; Chen, X.; Godfrey, J.; Kasal, D.N.; Yu, J.; Jongsma, M.L.M.; Spaapen, R.M.; Kline, D.E.; Kline, J. Dendritic cells can prime anti-tumor CD8(+) T cell responses through major histocompatibility complex cross-dressing. Immunity 2022, 55, 982–997.e988. [Google Scholar] [CrossRef] [PubMed]
- Padovan, E.; Spagnoli, G.C.; Ferrantini, M.; Heberer, M. IFN-alpha2a induces IP-10/CXCL10 and MIG/CXCL9 production in monocyte-derived dendritic cells and enhances their capacity to attract and stimulate CD8+ effector T cells. J. Leukoc. Biol. 2002, 71, 669–676. [Google Scholar] [CrossRef] [PubMed]
- Sato, H.; Niimi, A.; Yasuhara, T.; Permata, T.B.M.; Hagiwara, Y.; Isono, M.; Nuryadi, E.; Sekine, R.; Oike, T.; Kakoti, S.; et al. DNA double-strand break repair pathway regulates PD-L1 expression in cancer cells. Nat. Commun. 2017, 8, 1751. [Google Scholar] [CrossRef]
- Permata, T.B.M.; Hagiwara, Y.; Sato, H.; Yasuhara, T.; Oike, T.; Gondhowiardjo, S.; Held, K.D.; Nakano, T.; Shibata, A. Base excision repair regulates PD-L1 expression in cancer cells. Oncogene 2019, 38, 4452–4466. [Google Scholar] [CrossRef]
- Garcia-Diaz, A.; Shin, D.S.; Moreno, B.H.; Saco, J.; Escuin-Ordinas, H.; Rodriguez, G.A.; Zaretsky, J.M.; Sun, L.; Hugo, W.; Wang, X.; et al. Interferon Receptor Signaling Pathways Regulating PD-L1 and PD-L2 Expression. Cell Rep. 2017, 19, 1189–1201. [Google Scholar] [CrossRef]
- Zhang, Q.; Green, M.D.; Lang, X.; Lazarus, J.; Parsels, J.D.; Wei, S.; Parsels, L.A.; Shi, J.; Ramnath, N.; Wahl, D.R.; et al. Inhibition of ATM Increases Interferon Signaling and Sensitizes Pancreatic Cancer to Immune Checkpoint Blockade Therapy. Cancer Res. 2019, 79, 3940–3951. [Google Scholar] [CrossRef]
- Wang, L.; Yang, L.; Wang, C.; Zhao, W.; Ju, Z.; Zhang, W.; Shen, J.; Peng, Y.; An, C.; Luu, Y.T.; et al. Inhibition of the ATM/Chk2 axis promotes cGAS/STING signaling in ARID1A-deficient tumors. J. Clin. Invest. 2020, 130, 5951–5966. [Google Scholar] [CrossRef] [PubMed]
- Chiu, L.Y.; Sun, Q.; Zenke, F.T.; Blaukat, A.; Vassilev, L.T. Selective ATM inhibition augments radiation-induced inflammatory signaling and cancer cell death. Aging 2023, 15, 492–512. [Google Scholar] [CrossRef] [PubMed]
- Hu, M.; Zhou, M.; Bao, X.; Pan, D.; Jiao, M.; Liu, X.; Li, F.; Li, C.Y. ATM inhibition enhances cancer immunotherapy by promoting mtDNA leakage and cGAS/STING activation. J. Clin. Invest. 2021, 131, e139333. [Google Scholar] [CrossRef]
- Shen, M.; Xu, Z.; Xu, W.; Jiang, K.; Zhang, F.; Ding, Q.; Xu, Z.; Chen, Y. Inhibition of ATM reverses EMT and decreases metastatic potential of cisplatin-resistant lung cancer cells through JAK/STAT3/PD-L1 pathway. J. Exp. Clin. Cancer Res. 2019, 38, 149. [Google Scholar] [CrossRef] [PubMed]
- Lopez-Pelaez, M.; Young, L.; Vazquez-Chantada, M.; Nelson, N.; Durant, S.; Wilkinson, R.W.; Poon, E.; Gaspar, M.; Valge-Archer, V.; Smith, P.; et al. Targeting DNA damage response components induces enhanced STING-dependent type-I IFN response in ATM deficient cancer cells and drives dendritic cell activation. Oncoimmunology 2022, 11, 2117321. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Li, Y.; Zhao, Z.; Miao, N.; Liu, G.; Deng, L.; Wei, S.; Hou, J. Immunogenicity of small-cell lung cancer associates with STING pathway activation and is enhanced by ATR and TOP1 inhibition. Cancer Med. 2023, 12, 4864–4881. [Google Scholar] [CrossRef] [PubMed]
- Sun, L.L.; Yang, R.Y.; Li, C.W.; Chen, M.K.; Shao, B.; Hsu, J.M.; Chan, L.C.; Yang, Y.; Hsu, J.L.; Lai, Y.J.; et al. Inhibition of ATR downregulates PD-L1 and sensitizes tumor cells to T cell-mediated killing. Am. J. Cancer Res. 2018, 8, 1307–1316. [Google Scholar] [PubMed]
- Tang, Z.; Pilié, P.G.; Geng, C.; Manyam, G.C.; Yang, G.; Park, S.; Wang, D.; Peng, S.; Wu, C.; Peng, G.; et al. ATR Inhibition Induces CDK1-SPOP Signaling and Enhances Anti-PD-L1 Cytotoxicity in Prostate Cancer. Clin. Cancer Res. 2021, 27, 4898–4909. [Google Scholar] [CrossRef] [PubMed]
- Chao, H.H.; Karagounis, I.V.; Thomas, C.; François, N.B.; Facciabene, A.; Koumenis, C.; Maity, A. Combination of CHEK1/2 inhibition and ionizing radiation results in abscopal tumor response through increased micronuclei formation. Oncogene 2020, 39, 4344–4357. [Google Scholar] [CrossRef] [PubMed]
- Sen, T.; Della Corte, C.M.; Milutinovic, S.; Cardnell, R.J.; Diao, L.; Ramkumar, K.; Gay, C.M.; Stewart, C.A.; Fan, Y.; Shen, L.; et al. Combination Treatment of the Oral CHK1 Inhibitor, SRA737, and Low-Dose Gemcitabine Enhances the Effect of Programmed Death Ligand 1 Blockade by Modulating the Immune Microenvironment in SCLC. J. Thorac. Oncol. 2019, 14, 2152–2163. [Google Scholar] [CrossRef]
- Sen, T.; Rodriguez, B.L.; Chen, L.; Corte, C.M.D.; Morikawa, N.; Fujimoto, J.; Cristea, S.; Nguyen, T.; Diao, L.; Li, L.; et al. Targeting DNA Damage Response Promotes Antitumor Immunity through STING-Mediated T-cell Activation in Small Cell Lung Cancer. Cancer Discov. 2019, 9, 646–661. [Google Scholar] [CrossRef] [PubMed]
- Guo, E.; Xiao, R.; Wu, Y.; Lu, F.; Liu, C.; Yang, B.; Li, X.; Fu, Y.; Wang, Z.; Li, Y.; et al. WEE1 inhibition induces anti-tumor immunity by activating ERV and the dsRNA pathway. J. Exp. Med. 2022, 219, e20210789. [Google Scholar] [CrossRef] [PubMed]
- Taniguchi, H.; Caeser, R.; Chavan, S.S.; Zhan, Y.A.; Chow, A.; Manoj, P.; Uddin, F.; Kitai, H.; Qu, R.; Hayatt, O.; et al. WEE1 inhibition enhances the antitumor immune response to PD-L1 blockade by the concomitant activation of STING and STAT1 pathways in SCLC. Cell Rep. 2022, 39, 110814. [Google Scholar] [CrossRef] [PubMed]
- Wu, X.; Kang, X.; Zhang, X.; Xie, W.; Su, Y.; Liu, X.; Guo, L.; Guo, E.; Li, F.; Hu, D.; et al. WEE1 inhibitor and ataxia telangiectasia and RAD3-related inhibitor trigger stimulator of interferon gene-dependent immune response and enhance tumor treatment efficacy through programmed death-ligand 1 blockade. Cancer Sci. 2021, 112, 4444–4456. [Google Scholar] [CrossRef]
- Jin, M.H.; Nam, A.R.; Park, J.E.; Bang, J.H.; Bang, Y.J.; Oh, D.Y. Therapeutic Co-targeting of WEE1 and ATM Downregulates PD-L1 Expression in Pancreatic Cancer. Cancer Res. Treat. 2020, 52, 149–166. [Google Scholar] [CrossRef]
- Lampert, E.J.; Cimino-Mathews, A.; Lee, J.S.; Nair, J.; Lee, M.J.; Yuno, A.; An, D.; Trepel, J.B.; Ruppin, E.; Lee, J.M. Clinical outcomes of prexasertib monotherapy in recurrent BRCA wild-type high-grade serous ovarian cancer involve innate and adaptive immune responses. J. Immunother. Cancer 2020, 8, e000516. [Google Scholar] [CrossRef] [PubMed]
- Kwon, M.; Kim, G.; Kim, R.; Kim, K.T.; Kim, S.T.; Smith, S.; Mortimer, P.G.S.; Hong, J.Y.; Loembé, A.B.; Irurzun-Arana, I.; et al. Phase II study of ceralasertib (AZD6738) in combination with durvalumab in patients with advanced gastric cancer. J. Immunother. Cancer 2022, 10, e005041. [Google Scholar] [CrossRef]
- Kim, R.; Kwon, M.; An, M.; Kim, S.T.; Smith, S.A.; Loembé, A.B.; Mortimer, P.G.S.; Armenia, J.; Lukashchuk, N.; Shah, N.; et al. Phase II study of ceralasertib (AZD6738) in combination with durvalumab in patients with advanced/metastatic melanoma who have failed prior anti-PD-1 therapy. Ann. Oncol. 2022, 33, 193–203. [Google Scholar] [CrossRef]
- Do, K.T.; Manuszak, C.; Thrash, E.; Giobbie-Hurder, A.; Hu, J.; Kelland, S.; Powers, A.; de Jonge, A.; Shapiro, G.I.; Severgnini, M. Immune modulating activity of the CHK1 inhibitor prexasertib and anti-PD-L1 antibody LY3300054 in patients with high-grade serous ovarian cancer and other solid tumors. Cancer Immunol. Immunother. 2021, 70, 2991–3000. [Google Scholar] [CrossRef]
- Lv, M.; Chen, M.; Zhang, R.; Zhang, W.; Wang, C.; Zhang, Y.; Wei, X.; Guan, Y.; Liu, J.; Feng, K.; et al. Manganese is critical for antitumor immune responses via cGAS-STING and improves the efficacy of clinical immunotherapy. Cell Res. 2020, 30, 966–979. [Google Scholar] [CrossRef]
Trial | ICIs | Research Object/Sample Size | Efficacy Index | Phase | Status | References |
---|---|---|---|---|---|---|
NCT02054806 (KEYNOTE-028) | Pembrolizumab | Advanced PD-L1-positive endometrial cancer | ORR 13%, | 1b | Published | [28] |
(Keytruda) | PR 13%, | |||||
(n = 24.23 evaluable) | SD 13%, | |||||
Median PFS 1.8 months, | ||||||
PFS—6 months 19.0%, | ||||||
PFS—12 months 14.3%, | ||||||
OS—6-month rate 67.0%, | ||||||
OS—12-month rate 51.0% | ||||||
NCT02628067 | Pembrolizumab | Advanced MSI-H/dMMR tumors (including endometrial cancer) | ORR 48%, | 2 | Published | [29] |
(KEYNOTE-158) | (Keytruda) | CR 14%, | ||||
PR 34%, | ||||||
(n = 90.79 evaluable) | SD 18%, | |||||
Median PFS 13.1 months | ||||||
NCT02899793 | Pembrolizumab | Recurrent MSI-H endometrial cancer with Lynch-like vs. MLH-1 methylated characteristics | Overall ORR 58% | 2 | Published | [30] |
(Keytruda) | Lynch-like: | |||||
PFS—3-year rate 100%, | ||||||
OS-3-year rate 100% | ||||||
(n = 25) | MLH-1 methylated: | |||||
PFS—3-year rate 30%, | ||||||
OS—3-year rate 43% | ||||||
NCT02715284 | Dostarlimab | Recurrent or advanced dMMR endometrial cancer | ORR 42.3%, | 1 | Published | [14] |
(GARNET) | (TSR-042) | CR 12.7%, | ||||
(n = 104.71 evaluable) | PR 29.6%, | |||||
Median PFS 8.1 months, | ||||||
NCT02465060 | Nivolumab | dMMR non-colorectal cancers (including 13 endometrioid endometrial adenocarcinomas and 4 uterine carcinosarcomas) | ORR 36%, | 2 | Published | [17] |
(NCI-MATCH/EAY131) | (Opdivo) | SD 21%, | ||||
PFS—6 months 51.3%, | ||||||
PFS—12 months 46.2%, | ||||||
(n = 42) | PFS—18 months 31.4%, | |||||
Median OS 17.3 months | ||||||
NCT02982486 | Nivolumab | Non-resectable sarcoma and endometrial carcinoma | PR, CR, | 2 | Unknown | - |
(Opdivo), | Median PFS, PFS—12 weeks, PFS—24 weeks, OS—3 years | |||||
Ipilimumab | (n = 60, estimated enrollment) | |||||
(Yervoy, CTLA-4 inhibitor) | ||||||
NCT04106414 | Nivolumab | Endometrial adenocarcinoma and endometrial carcinosarcoma | ORR | 2 | Active, not recruiting | - |
(Opdivo), | ||||||
BMS–986205 | (n = 24, actual enrollment) | |||||
(IDO-1 inhibitor) | ||||||
NCT02791334 | LY3300054 | Advanced refractory MSI-H/dMMR solid tumors (including 14 endometrial cancers) | LY3300054 (n = 40): | 1b | Published | [18] |
(PD-L1 inhibitor) | ORR 32.5% | |||||
LY3321367 | CR 12.5, | |||||
(TIM-3 inhibitor) | PR 20%, | |||||
(n = 82) | SD 27.5%, | |||||
PFS rate 62.5%, | ||||||
Median PFS 7.4 months | ||||||
LY3300054 + LY3321367 (n = 42): | ||||||
ORR 45%, | ||||||
CR 10%, | ||||||
PR 35%, | ||||||
SD 25%, | ||||||
PFS rate 55%, | ||||||
Median PFS 7.6 months | ||||||
NCT03015129 | Durvalumab (Imfinzi) | Advanced dMMR and pMMR endometrial cancer | dMMR (n = 36): | 2 | Published | [26] |
ORR 47% | ||||||
(n = 71) | PR 30.6%, | |||||
PFS—6 months 53%, | ||||||
Median PFS 8.3 months, | ||||||
OS—12-month rate 71% | ||||||
pMMR (n = 35): | ||||||
ORR 3%, | ||||||
PR 3%, | ||||||
PFS—6 months 14%, | ||||||
Median PFS 1.8 months, | ||||||
OS-12-month rate 51% | ||||||
NCT02912572 | Avelumab | Recurrent/persistent dMMR and pMMR endometrial cancer | 31 included in analysis: | 2 | Published | [31] |
(BAVENCIO) | dMMR (n = 15): | |||||
(n = 33) | ORR 26.7%, | |||||
CR 6.7%, | ||||||
PR 20%, | ||||||
PFS—6-month rate 40%, | ||||||
Median PFS 4.4 months | ||||||
pMMR (n = 16): | ||||||
ORR 6.25%, | ||||||
CR 0%, | ||||||
PR 6.25%, | ||||||
PFS—6-month rate 6.25%, | ||||||
Median PFS 1.9 months, | ||||||
Median OS 6.6 months |
DDR Target | DDR Inhibitor | Research Object/Sample | Research Type | Phase | Efficacy Index | Status | References |
---|---|---|---|---|---|---|---|
ATM | KU60019 | Endometrial cancer cells (HEC-1-B, HEC-6) | Preclinical | - | IC50 20 µM | Published | [60] |
ATM | KU55933 | Endometrial cancer cells (HEC-1-B, KLE) | Preclinical | - | LD50 38.3 ± 7.6 µM | Published | [61] |
ATR | ETP-46464 | Endometrial cancer cells (HEC-1-B, KLE) | Preclinical | - | LD50 10.0 ± 8.7 µM | Published | [61] |
ATR | VE822 | Endometrial cancer cells (HEC-1-B, HEC-6) | Preclinical | - | IC50 1.5 µM | Published | [60] |
ATR | Berzosertib (M6620/VX-970) | Advanced solid tumors (including 1 endometrial cancer) | Clinical | 1 | PR continuing at 18 months | Published | [81] |
(n = 1) | |||||||
CHK1 | GDC-0575 | Refractory solid tumors (including 4 endometrial cancers) | Clinical | 1 | SD or PR 66% | Published | [115] |
(n = 102, 90 evaluable) | |||||||
ATR | VE822 | Endometrial cancer cells (HEC-1-B, HEC-6) | Preclinical | - | CI in HEC-1-B: 0.43 | Published | [60] |
CHK1 | AZD7762 | CI in HEC-6: 0.28 | |||||
Wee1 | Adavosertib (AZD1775) | Advanced solid tumors (including 3 endometrial cancers and 1 uterine carcinosarcoma) | Clinical | 1 | PR 14.3%, | Published | [103] |
(n = 42) | median DOR 4.9 months, | ||||||
SD 47.6% | |||||||
Wee1 | Adavosertib (AZD1775) | Advanced or refractory malignant solid neoplasm (including 3 uterine tumors) | Clinical | 2 | ORR 27%, | Published | [104] |
(n = 30) | median PFS 4.1 months, | ||||||
median OS 9.9 months | |||||||
Wee1 | Adavosertib (AZD1775) | Recurrent uterine serous carcinoma | Clinical | 2 | ORR 29.4%, | Published | [100] |
(n = 35, 34 evaluable) | PFS6 47.1%, | ||||||
median PFS 6.1 months |
Targets | Inhibitors | Research Object/Sample Size | Phase | Status | NCT Number |
---|---|---|---|---|---|
ATR, | Ceralasertib, | Advanced or metastatic non-small-cell lung cancer | 3 | Recruiting | NCT05450692 |
PD-L1 | Durvalumab | ||||
(n = 580) | |||||
ATR, | Ceralasertib, | Melanoma | 2 | Recruiting | NCT05061134 |
PD-L1 | Durvalumab | (n = 195) | |||
ATR, | Ceralasertib, | Triple-negative breast cancer | 2 | Recruiting | NCT05582538 |
PD-L1 | Durvalumab | (n = 37) | |||
ATR, | Ceralasertib, | Solid tumors (including endometrial cancer) | 2 | Recruiting | NCT03682289 |
PD-L1 | Durvalumab | ||||
(n = 89) | |||||
ATR, | Ceralasertib, | Advanced solid malignancies-HNSCC, ATM Pro/Def NSCLC, gastric, breast and ovarian cancer | 1/2 | Recruiting | NCT02264678 |
PD-L1 | Durvalumab | ||||
(n = 330) | |||||
ATR, | Ceralasertib, | Non-small-cell lung cancer | 2 | Recruiting | NCT03334617 |
PD-L1 | Durvalumab | (n = 570) | |||
ATR, | Ceralasertib, | Non-small-cell lung cancer | 2 | Recruiting | NCT03833440 |
PD-L1 | Durvalumab | (n = 120) | |||
ATR, | Ceralasertib, | Advanced solid tumors | 1 | Recruiting | NCT05514132 |
PD-L1 | Durvalumab | (n = 12) | |||
ATR, | Elimusertib, | Advanced or recurrent head and neck cancer | 1 | Recruiting | NCT04576091 |
PD-1 | Pembrolizumab | ||||
(n = 37) | |||||
ATR, | M1774, | Metastatic or locally advanced unresectable solid tumors | 1 | Recruiting | NCT05396833 |
IC | Unknown | (n = 72) |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Li, Y.; Wang, X.; Hou, X.; Ma, X. Could Inhibiting the DNA Damage Repair Checkpoint Rescue Immune-Checkpoint-Inhibitor-Resistant Endometrial Cancer? J. Clin. Med. 2023, 12, 3014. https://doi.org/10.3390/jcm12083014
Li Y, Wang X, Hou X, Ma X. Could Inhibiting the DNA Damage Repair Checkpoint Rescue Immune-Checkpoint-Inhibitor-Resistant Endometrial Cancer? Journal of Clinical Medicine. 2023; 12(8):3014. https://doi.org/10.3390/jcm12083014
Chicago/Turabian StyleLi, Yinuo, Xiangyu Wang, Xin Hou, and Xiangyi Ma. 2023. "Could Inhibiting the DNA Damage Repair Checkpoint Rescue Immune-Checkpoint-Inhibitor-Resistant Endometrial Cancer?" Journal of Clinical Medicine 12, no. 8: 3014. https://doi.org/10.3390/jcm12083014
APA StyleLi, Y., Wang, X., Hou, X., & Ma, X. (2023). Could Inhibiting the DNA Damage Repair Checkpoint Rescue Immune-Checkpoint-Inhibitor-Resistant Endometrial Cancer? Journal of Clinical Medicine, 12(8), 3014. https://doi.org/10.3390/jcm12083014