1
|
Chen A, Yin K, Liu Y, Hu L, Cui Q, Wan X, Yang W. WEE Family Kinase Inhibitors Combined with Sorafenib Can Selectively Inhibit HCC Cell Proliferation. Curr Cancer Drug Targets 2025; 25:370-385. [PMID: 38860904 DOI: 10.2174/0115680096298370240520093003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/10/2024] [Accepted: 04/24/2024] [Indexed: 06/12/2024]
Abstract
BACKGROUND Sorafenib is currently the first choice for the treatment of patients with advanced hepatocellular carcinoma, but its therapeutic effect is still limited. OBJECTIVES This study aims to examine whether WEE family kinase inhibitors can enhance the anticancer effect of sorafenib. METHODS We analyzed the expression levels of PKMYT1 kinase and WEE1 kinase in HCC, studied the inhibitory effect of PKMYT1 kinase inhibitor RP-6306, WEE1 kinase inhibitor adavosertib combined with sorafenib on the proliferation of HCC cells, and detected the effect of drug combination on CDK1 phosphorylation. RESULTS We found that PKMYT1 and WEE1 were upregulated in HCC and were detrimental to patient survival. Cell experiments showed that both RP-6306 and adavosertib (1-100 μM) inhibited the proliferation of HCC cell lines in a dose-dependent manner alone, and the combination of the two drugs had a synergistic effect. In HCC cell lines, sorafenib combined with RP-6306 or adavosertib showed a synergistic antiproliferation effect and less toxicity to normal cells. Sorafenib combined with RP-6306 and adavosertib further inhibited the proliferation of HCC cells and caused complete dephosphorylation of CDK1. CONCLUSION Taken together, our findings provide experimental evidence for the future use of sorafenib in combination with RP-6306 or adavosertib for the treatment of HCC.
Collapse
Affiliation(s)
- Anling Chen
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Science Island Branch, Graduate School of University of Science and Technology of China, Hefei, 230031, China
| | - Ke Yin
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Yu Liu
- School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China
| | - Lei Hu
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Qianwen Cui
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Science Island Branch, Graduate School of University of Science and Technology of China, Hefei, 230031, China
| | - Xiaofeng Wan
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Wulin Yang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Science Island Branch, Graduate School of University of Science and Technology of China, Hefei, 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| |
Collapse
|
2
|
Dong S, Zhao M, Zhu J, Li T, Yan M, Xing K, Liu P, Yu S, Ma J, He H. Natural killer cells: a future star for immunotherapy of head and neck squamous cell carcinoma. Front Immunol 2024; 15:1442673. [PMID: 39234249 PMCID: PMC11371580 DOI: 10.3389/fimmu.2024.1442673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 07/15/2024] [Indexed: 09/06/2024] Open
Abstract
The interplay between immune components and the epithelium plays a crucial role in the development and progression of head and neck squamous cell carcinoma (HNSCC). Natural killer (NK) cells, one of the main tumor-killing immune cell populations, have received increasing attention in HNSCC immunotherapy. In this review, we explore the mechanism underlying the interplay between NK cells and HNSCC. A series of immune evasion strategies utilized by cancer cells restrict HNSCC infiltration of NK cells. Overcoming these limitations can fully exploit the antineoplastic potential of NK cells. We also investigated the tumor-killing efficacy of NK cell-based immunotherapies, immunotherapeutic strategies, and new results from clinical trials. Notably, cetuximab, the most essential component of NK cell-based immunotherapy, inhibits the epidermal growth factor receptor (EGFR) signaling pathway and activates the immune system in conjunction with NK cells, inducing innate effector functions and improving patient prognosis. In addition, we compiled information on other areas for the improvement of patient prognosis using anti-EGFR receptor-based monoclonal antibody drugs and the underlying mechanisms and prognoses of new immunotherapeutic strategies for the treatment of HNSCC.
Collapse
Affiliation(s)
- Shuyan Dong
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ming Zhao
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Jin Zhu
- Department of Pathology, Xi’an Daxing Hospital, Xi’an, China
| | - Ting Li
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Mingze Yan
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Kaixun Xing
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Peng Liu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Shan Yu
- Department of Pathology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jian Ma
- Department of General Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Immunology, Harbin Medical University, Harbin, China
| | - Hongjiang He
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
3
|
Yin Y, Wang J, Yi J, Zhang K, Yin Z, Jin S, Zheng B. AZD1775 and anti-PD-1 antibody synergistically sensitize hepatoma to radiotherapy. Chin Med J (Engl) 2024; 137:222-231. [PMID: 38167245 PMCID: PMC10798739 DOI: 10.1097/cm9.0000000000002988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Radiation (IR)-induced DNA damage triggers cell cycle arrest and has a suppressive effect on the tumor microenvironment (TME). Wee1, a cell cycle regulator, can eliminate G2/M arrest by phosphorylating cyclin-dependent kinase 1 (CDK1). Meanwhile, programed death-1/programed death ligand-1 (PD-1/PDL-1) blockade is closely related to TME. This study aims to investigate the effects and mechanisms of Wee1 inhibitor AZD1775 and anti-PD-1 antibody (anti-PD-1 Ab) on radiosensitization of hepatoma. METHODS The anti-tumor activity of AZD1775 and IR was determined by 3-(4,5-dimethylthiazol-2-y1)-2,5-diphenyltetrazolium bromide (MTT) assay on human and mouse hepatoma cells HepG2, Hepa1-6, and H22. The anti-hepatoma mechanism of AZD1775 and IR revealed by flow cytometry and Western blot in vitro . A hepatoma subcutaneous xenograft mice model was constructed on Balb/c mice, which were divided into control group, IR group, AZD1775 group, IR + AZD1775 group, IR + anti-PD-1 Ab group, and the IR + AZD1775 + anti-PD-1 Ab group. Cytotoxic CD8 + T cells in TME were analyzed by flow cytometry. RESULTS Combining IR with AZD1775 synergistically reduced the viability of hepatoma cells in vitro . AZD1775 exhibited antitumor effects by decreasing CDK1 phosphorylation to reverse the IR-induced G2/M arrest and increasing IR-induced DNA damage. AZD1775 treatment also reduced the proportion of PD-1 + /CD8 + T cells in the spleen of hepatoma subcutaneous xenograft mice. Further studies revealed that AZD1775 and anti-PD-1 Ab could enhance the radiosensitivity of hepatoma by enhancing the levels of interferon γ (IFNγ) + or Ki67 + CD8 T cells and decreasing the levels of CD8 + Tregs cells in the tumor and spleen of the hepatoma mice model, indicating that the improvement of TME was manifested by increasing the cytotoxic factor IFNγ expression, enhancing CD8 + T cells proliferation, and weakening CD8 + T cells depletion. CONCLUSIONS This work suggests that AZD1775 and anti-PD-1 Ab synergistically sensitize hepatoma to radiotherapy by enhancing IR-induced DNA damage and improving cytotoxic CD8 + T cells in TME.
Collapse
Affiliation(s)
- Yichun Yin
- Public Health of College, Jilin University, Jilin, Changchun 130021, China
| | - Jian Wang
- Institute of Virology and AIDS Research, The First Hospital of Jilin University, Jilin, Changchun 130021, China
| | - Junxuan Yi
- Public Health of College, Jilin University, Jilin, Changchun 130021, China
| | - Kaiyue Zhang
- Public Health of College, Jilin University, Jilin, Changchun 130021, China
| | - Zimeng Yin
- Public Health of College, Jilin University, Jilin, Changchun 130021, China
| | - Shunzi Jin
- Public Health of College, Jilin University, Jilin, Changchun 130021, China
- NHC Key Laboratory of Radiobiology (Jilin University), Jilin, Changchun 130021, China
| | - Baisong Zheng
- Institute of Virology and AIDS Research, The First Hospital of Jilin University, Jilin, Changchun 130021, China
| |
Collapse
|
4
|
Kostecki KL, Iida M, Crossman BE, Salgia R, Harari PM, Bruce JY, Wheeler DL. Immune Escape Strategies in Head and Neck Cancer: Evade, Resist, Inhibit, Recruit. Cancers (Basel) 2024; 16:312. [PMID: 38254801 PMCID: PMC10814769 DOI: 10.3390/cancers16020312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Head and neck cancers (HNCs) arise from the mucosal lining of the aerodigestive tract and are often associated with alcohol use, tobacco use, and/or human papillomavirus (HPV) infection. Over 600,000 new cases of HNC are diagnosed each year, making it the sixth most common cancer worldwide. Historically, treatments have included surgery, radiation, and chemotherapy, and while these treatments are still the backbone of current therapy, several immunotherapies have recently been approved by the Food and Drug Administration (FDA) for use in HNC. The role of the immune system in tumorigenesis and cancer progression has been explored since the early 20th century, eventually coalescing into the current three-phase model of cancer immunoediting. During each of the three phases-elimination, equilibrium, and escape-cancer cells develop and utilize multiple strategies to either reach or remain in the final phase, escape, at which point the tumor is able to grow and metastasize with little to no detrimental interference from the immune system. In this review, we summarize the many strategies used by HNC to escape the immune system, which include ways to evade immune detection, resist immune cell attacks, inhibit immune cell functions, and recruit pro-tumor immune cells.
Collapse
Affiliation(s)
- Kourtney L. Kostecki
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA; (K.L.K.); (M.I.); (B.E.C.)
| | - Mari Iida
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA; (K.L.K.); (M.I.); (B.E.C.)
| | - Bridget E. Crossman
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA; (K.L.K.); (M.I.); (B.E.C.)
| | - Ravi Salgia
- Department of Medical Oncology and Experimental Therapeutics, Comprehensive Cancer Center, City of Hope, Duarte, CA 91010, USA;
| | - Paul M. Harari
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA; (K.L.K.); (M.I.); (B.E.C.)
- University of Wisconsin Carbone Cancer Center, Madison, WI 53705, USA;
| | - Justine Y. Bruce
- University of Wisconsin Carbone Cancer Center, Madison, WI 53705, USA;
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Deric L. Wheeler
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA; (K.L.K.); (M.I.); (B.E.C.)
- University of Wisconsin Carbone Cancer Center, Madison, WI 53705, USA;
| |
Collapse
|
5
|
Zhang C, Peng K, Liu Q, Huang Q, Liu T. Adavosertib and beyond: Biomarkers, drug combination and toxicity of WEE1 inhibitors. Crit Rev Oncol Hematol 2024; 193:104233. [PMID: 38103761 DOI: 10.1016/j.critrevonc.2023.104233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 12/10/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023] Open
Abstract
WEE1 kinase is renowned as an S-G2 checkpoint inhibitor activated by ATR-CHK1 in response to replication stress. WEE1 inhibition enhances replication stress and effectively circumvents checkpoints into mitosis, which triggers significant genetic impairs and culminates in cell death. This approach has been validated clinically for its promising anti-tumor efficacy across various cancer types, notably in cases of ovarian cancers. Nonetheless, the initial stage of clinical trials has shown that the first-in-human WEE1 inhibitor adavosertib is limited by dose-limiting adverse events. As a result, recent efforts have been made to explore predictive biomarkers and smart combination schedules to alleviate adverse effects. In this review, we focused on the exploration of therapeutic biomarkers, as well as schedules of combination utilizing WEE1 inhibitors and canonical anticancer drugs, according to the latest preclinical and clinical studies, indicating that the optimal application of WEE1 inhibitors will likely be as part of dose-reducing combination and be tailored to specific patient populations.
Collapse
Affiliation(s)
- Chi Zhang
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ke Peng
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qing Liu
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qihong Huang
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Tianshu Liu
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
6
|
Tan J, Sun X, Zhao H, Guan H, Gao S, Zhou P. Double-strand DNA break repair: molecular mechanisms and therapeutic targets. MedComm (Beijing) 2023; 4:e388. [PMID: 37808268 PMCID: PMC10556206 DOI: 10.1002/mco2.388] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/29/2023] [Accepted: 09/08/2023] [Indexed: 10/10/2023] Open
Abstract
Double-strand break (DSB), a significant DNA damage brought on by ionizing radiation, acts as an initiating signal in tumor radiotherapy, causing cancer cells death. The two primary pathways for DNA DSB repair in mammalian cells are nonhomologous end joining (NHEJ) and homologous recombination (HR), which cooperate and compete with one another to achieve effective repair. The DSB repair mechanism depends on numerous regulatory variables. DSB recognition and the recruitment of DNA repair components, for instance, depend on the MRE11-RAD50-NBS1 (MRN) complex and the Ku70/80 heterodimer/DNA-PKcs (DNA-PK) complex, whose control is crucial in determining the DSB repair pathway choice and efficiency of HR and NHEJ. In-depth elucidation on the DSB repair pathway's molecular mechanisms has greatly facilitated for creation of repair proteins or pathways-specific inhibitors to advance precise cancer therapy and boost the effectiveness of cancer radiotherapy. The architectures, roles, molecular processes, and inhibitors of significant target proteins in the DSB repair pathways are reviewed in this article. The strategy and application in cancer therapy are also discussed based on the advancement of inhibitors targeted DSB damage response and repair proteins.
Collapse
Affiliation(s)
- Jinpeng Tan
- Hengyang Medical CollegeUniversity of South ChinaHengyangHunan ProvinceChina
- Department of Radiation BiologyBeijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| | - Xingyao Sun
- Hengyang Medical CollegeUniversity of South ChinaHengyangHunan ProvinceChina
- Department of Radiation BiologyBeijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| | - Hongling Zhao
- Department of Radiation BiologyBeijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| | - Hua Guan
- Department of Radiation BiologyBeijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| | - Shanshan Gao
- Department of Radiation BiologyBeijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| | - Ping‐Kun Zhou
- Hengyang Medical CollegeUniversity of South ChinaHengyangHunan ProvinceChina
- Department of Radiation BiologyBeijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| |
Collapse
|
7
|
Shang J, Nie X, Qi Y, Zhou J, Qi Y. Short-term smoking cessation leads to a universal decrease in whole blood genomic DNA methylation in patients with a smoking history. World J Surg Oncol 2023; 21:227. [PMID: 37496025 PMCID: PMC10369823 DOI: 10.1186/s12957-023-03099-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/12/2023] [Indexed: 07/28/2023] Open
Abstract
BACKGROUND Epigenetics is involved in various human diseases. Smoking is one of the most common environmental factors causing epigenetic changes. The DNA methylation changes and mechanisms after quitting smoking have yet to be defined. The present study examined the changes in DNA methylation levels before and after short-term smoking cessation and explored the potential mechanism. METHODS Whole blood and clinical data were collected from 8 patients before and after short-term smoking cessation, DNA methylation was assessed, and differentially methylated sites were analyzed, followed by a comprehensive analysis of the differentially methylated sites with clinical data. GO/KEGG enrichment and protein-protein interaction (PPI) network analyses identified the hub genes. The differentially methylated sites between former and current smokers in GSE50660 from the GEO database were detected by GEO2R. Then, a Venn analysis was carried out using the differentially methylated sites. GO/KEGG enrichment analysis was performed on the genes corresponding to the common DNA methylation sites, the PPI network was constructed, and hub genes were predicted. The enriched genes associated with the cell cycle were selected, and the pan-cancer gene expression and clinical significance in lung cancer were analyzed based on the TCGA database. RESULTS Most genes showed decreased DNA methylation levels after short-term smoking cessation; 694 upregulated methylation CpG sites and 3184 downregulated methylation CpG sites were identified. The DNA methylation levels were altered according to the clinical data (body weight, expiratory, and tobacco dependence score). Enrichment analysis, construction of the PPI network, and pan-cancer analysis suggested that smoking cessation may affect various biological processes. CONCLUSIONS Smoking cessation leads to epigenetic changes, mainly decreased in the decline of most DNA methylation levels. Bioinformatics further identified the biologically relevant changes after short-term smoking cessation.
Collapse
Affiliation(s)
- Junyi Shang
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University; People's Hospital of Henan University, No. 7 Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan, China
| | - Xinran Nie
- Academy of Medical Science, Zhengzhou University, Zhengzhou, China
- Department of Respiratory and Critical Care Medicine, People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Yanan Qi
- Department of Respiratory and Critical Care Medicine, Central China Fuwai Hospital; Central China Fuwai Hospital of Zhengzhou University; People's Hospital of Zhengzhou University; Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China
| | - Jing Zhou
- Department of Health Management, Henan Provincial People's Hospital, Henan University People's Hospital, Zhengzhou, 450003, China
| | - Yong Qi
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University; People's Hospital of Henan University, No. 7 Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan, China.
| |
Collapse
|
8
|
Xie D, Jiang B, Wang S, Wang Q, Wu G. The mechanism and clinical application of DNA damage repair inhibitors combined with immune checkpoint inhibitors in the treatment of urologic cancer. Front Cell Dev Biol 2023; 11:1200466. [PMID: 37305685 PMCID: PMC10248030 DOI: 10.3389/fcell.2023.1200466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/18/2023] [Indexed: 06/13/2023] Open
Abstract
Urologic cancers such as kidney, bladder, prostate, and uroepithelial cancers have recently become a considerable global health burden, and the response to immunotherapy is limited due to immune escape and immune resistance. Therefore, it is crucial to find appropriate and effective combination therapies to improve the sensitivity of patients to immunotherapy. DNA damage repair inhibitors can enhance the immunogenicity of tumor cells by increasing tumor mutational burden and neoantigen expression, activating immune-related signaling pathways, regulating PD-L1 expression, and reversing the immunosuppressive tumor microenvironment to activate the immune system and enhance the efficacy of immunotherapy. Based on promising experimental results from preclinical studies, many clinical trials combining DNA damage repair inhibitors (e.g., PARP inhibitors and ATR inhibitors) with immune checkpoint inhibitors (e.g., PD-1/PD-L1 inhibitors) are underway in patients with urologic cancers. Results from several clinical trials have shown that the combination of DNA damage repair inhibitors with immune checkpoint inhibitors can improve objective rates, progression-free survival, and overall survival (OS) in patients with urologic tumors, especially in patients with defective DNA damage repair genes or a high mutational load. In this review, we present the results of preclinical and clinical trials of different DNA damage repair inhibitors in combination with immune checkpoint inhibitors in urologic cancers and summarize the potential mechanism of action of the combination therapy. Finally, we also discuss the challenges of dose toxicity, biomarker selection, drug tolerance, drug interactions in the treatment of urologic tumors with this combination therapy and look into the future direction of this combination therapy.
Collapse
Affiliation(s)
| | | | | | - Qifei Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
9
|
Su YL, Xiao LY, Huang SY, Wu CC, Chang LC, Chen YH, Luo HL, Huang CC, Liu TT, Peng JM. Inhibiting WEE1 Augments the Antitumor Efficacy of Cisplatin in Urothelial Carcinoma by Enhancing the DNA Damage Process. Cells 2023; 12:1471. [PMID: 37296592 PMCID: PMC10252844 DOI: 10.3390/cells12111471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Urothelial carcinoma (UC) is characterized by a high incidence of TP53 mutation, and overcoming resistance to cisplatin-based chemotherapy in UC is a major concern. Wee1 is a G2/M phase regulator that controls the DNA damage response to chemotherapy in TP53-mutant cancers. The combination of Wee1 blockade with cisplatin has shown synergistic efficacy in several types of cancers, but little is known regarding UC. The antitumor efficacy of the Wee1 inhibitor (AZD-1775) alone or in combination with cisplatin was evaluated in UC cell lines and a xenograft mouse model. AZD-1775 enhanced the anticancer activity of cisplatin by increasing cellular apoptosis. AZD-1775 inhibited the G2/M checkpoint, improving the sensitivity of mutant TP53 UC cells to cisplatin by enhancing the DNA damage process. We confirmed that AZD-1775 combined with cisplatin reduced tumor volume and proliferation activity and increased the markers of cell apoptosis and DNA damage in the mouse xenograft model. In summary, the Wee1 inhibitor AZD-1775 combined with cisplatin elicited a promising anticancer efficacy in UC, and constitutes an innovative and promising therapeutic strategy.
Collapse
Affiliation(s)
- Yu-Li Su
- Division of Hematology Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung 83301, Taiwan
- Genomic & Proteomic Core Laboratory, Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Ling-Yi Xiao
- Division of Hematology Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung 83301, Taiwan
| | - Shih-Yu Huang
- Division of Hematology Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung 83301, Taiwan
| | - Chia-Che Wu
- Division of Hematology Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung 83301, Taiwan
| | - Li-Chung Chang
- Division of Hematology Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung 83301, Taiwan
| | - Yi-Hua Chen
- Division of Hematology Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung 83301, Taiwan
| | - Hao-Lun Luo
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung 83301, Taiwan
| | - Chun-Chieh Huang
- Department of Radiation Oncology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung 83301, Taiwan
| | - Ting-Ting Liu
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung 83301, Taiwan
| | - Jei-Ming Peng
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| |
Collapse
|
10
|
Rodriguez CP, Kang H, Geiger JL, Burtness B, Chung CH, Pickering CR, Fakhry C, Le QT, Yom SS, Galloway TJ, Golemis E, Li A, Shoop J, Wong S, Mehra R, Skinner H, Saba NF, Flores ER, Myers JN, Ford JM, Karchin R, Ferris RL, Kunos C, Lynn JM, Malik S. Clinical Trial Development in TP53-Mutated Locally Advanced and Recurrent and/or Metastatic Head and Neck Squamous Cell Carcinoma. J Natl Cancer Inst 2022; 114:1619-1627. [PMID: 36053203 PMCID: PMC9745425 DOI: 10.1093/jnci/djac163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/03/2022] [Accepted: 06/15/2022] [Indexed: 01/11/2023] Open
Abstract
TP53 mutation is the most frequent genetic event in head and neck squamous cell carcinoma (HNSCC), found in more than 80% of patients with human papillomavirus-negative disease. As mutations in the TP53 gene are associated with worse outcomes in HNSCC, novel therapeutic approaches are needed for patients with TP53-mutated tumors. The National Cancer Institute sponsored a Clinical Trials Planning Meeting to address the issues of identifying and developing clinical trials for patients with TP53 mutations. Subcommittees, or breakout groups, were tasked with developing clinical studies in both the locally advanced and recurrent and/or metastatic (R/M) disease settings as well as considering signal-seeking trial designs. A fourth breakout group was focused on identifying and standardizing biomarker integration into trial design; this information was provided to the other breakout groups prior to the meeting to aid in study development. A total of 4 concepts were prioritized to move forward for further development and implementation. This article summarizes the proceedings of the Clinical Trials Planning Meeting with the goal of developing clinical trials for patients with TP53-mutant HNSCC that can be conducted within the National Clinical Trials Network.
Collapse
Affiliation(s)
| | - Hyunseok Kang
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Jessica L Geiger
- Department of Hematology and Medical Oncology, Cleveland Clinic Foundation, Cleveland, OH, USA
| | | | - Christine H Chung
- Department of Head and Neck-Endocrine Oncology, Moffit Cancer Center, Tampa, FL, USA
| | - Curtis R Pickering
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carole Fakhry
- Division of Head and Neck Surgery, Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, Baltimore, MD, USA
| | - Quynh Thu Le
- Department of Radiation Oncology-Radiation Therapy, Stanford University, Palo Alto, CA, USA
| | - Sue S Yom
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Thomas J Galloway
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Erica Golemis
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Alice Li
- Kaiser Permanente Oakland, Oakland, CA, USA
| | | | - Stuart Wong
- Division of Neoplastic Diseases, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ranee Mehra
- Division of Hematology/Oncology, Department of Medicine, University of Maryland, Baltimore, MD, USA
| | - Heath Skinner
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nabil F Saba
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Elsa R Flores
- Department of Molecular Oncology, Moffit Cancer Center, Tampa, FL, USA
| | - Jeffrey N Myers
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James M Ford
- Department of Medicine, Stanford University, Palo Alto, CA, USA
| | - Rachel Karchin
- Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - Robert L Ferris
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Jean M Lynn
- National Institutes of Health, Bethesda, MD, USA
| | - Shakun Malik
- National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
11
|
Carlsen L, El-Deiry WS. Anti-cancer immune responses to DNA damage response inhibitors: Molecular mechanisms and progress toward clinical translation. Front Oncol 2022; 12:998388. [PMID: 36276148 PMCID: PMC9583871 DOI: 10.3389/fonc.2022.998388] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/09/2022] [Indexed: 11/13/2022] Open
Abstract
DNA damage response inhibitors are widely used anti-cancer agents that have potent activity against tumor cells with deficiencies in various DNA damage response proteins such as BRCA1/2. Inhibition of other proteins in this pathway including PARP, DNA-PK, WEE1, CHK1/2, ATR, or ATM can sensitize cancer cells to radiotherapy and chemotherapy, and such combinations are currently being tested in clinical trials for treatment of many malignancies including breast, ovarian, rectal, and lung cancer. Unrepaired DNA damage induced by DNA damage response inhibitors alone or in combination with radio- or chemotherapy has a direct cytotoxic effect on cancer cells and can also engage anti-cancer innate and adaptive immune responses. DNA damage-induced immune stimulation occurs by a variety of mechanisms including by the cGAS/STING pathway, STAT1 and downstream TRAIL pathway activation, and direct immune cell activation. Whether or not the relative contribution of these mechanisms varies after treatment with different DNA damage response inhibitors or across cancers with different genetic aberrations in DNA damage response enzymes is not well-characterized, limiting the design of optimal combinations with radio- and chemotherapy. Here, we review how the inhibition of key DNA damage response enzymes including PARP, DNA-PK, WEE1, CHK1/2, ATR, and ATM induces innate and adaptive immune responses alone or in combination with radiotherapy, chemotherapy, and/or immunotherapy. We also discuss current progress in the clinical translation of immunostimulatory DNA-damaging treatment regimens and necessary future directions to optimize the immune-sensitizing potential of DNA damage response inhibitors.
Collapse
Affiliation(s)
- Lindsey Carlsen
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI, United States
- The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI, United States
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI, United States
- Pathobiology Graduate Program, The Warren Alpert Medical School, Brown University, Providence, RI, United States
- Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Wafik S. El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI, United States
- The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI, United States
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI, United States
- Pathobiology Graduate Program, The Warren Alpert Medical School, Brown University, Providence, RI, United States
- Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI, United States
- Department of Medicine, Hematology-Oncology Division, Rhode Island Hospital, Brown University, Providence, RI, United States
- *Correspondence: Wafik S. El-Deiry,
| |
Collapse
|
12
|
Porter VL, Marra MA. The Drivers, Mechanisms, and Consequences of Genome Instability in HPV-Driven Cancers. Cancers (Basel) 2022; 14:4623. [PMID: 36230545 PMCID: PMC9564061 DOI: 10.3390/cancers14194623] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/17/2022] [Accepted: 09/19/2022] [Indexed: 11/28/2022] Open
Abstract
Human papillomavirus (HPV) is the causative driver of cervical cancer and a contributing risk factor of head and neck cancer and several anogenital cancers. HPV's ability to induce genome instability contributes to its oncogenicity. HPV genes can induce genome instability in several ways, including modulating the cell cycle to favour proliferation, interacting with DNA damage repair pathways to bring high-fidelity repair pathways to viral episomes and away from the host genome, inducing DNA-damaging oxidative stress, and altering the length of telomeres. In addition, the presence of a chronic viral infection can lead to immune responses that also cause genome instability of the infected tissue. The HPV genome can become integrated into the host genome during HPV-induced tumorigenesis. Viral integration requires double-stranded breaks on the DNA; therefore, regions around the integration event are prone to structural alterations and themselves are targets of genome instability. In this review, we present the mechanisms by which HPV-dependent and -independent genome instability is initiated and maintained in HPV-driven cancers, both across the genome and at regions of HPV integration.
Collapse
Affiliation(s)
- Vanessa L. Porter
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Marco A. Marra
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
13
|
Kono M, Saito S, Egloff AM, Allen CT, Uppaluri R. The mouse oral carcinoma (MOC) model: A 10-year retrospective on model development and head and neck cancer investigations. Oral Oncol 2022; 132:106012. [PMID: 35820346 PMCID: PMC9364442 DOI: 10.1016/j.oraloncology.2022.106012] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 07/02/2022] [Indexed: 11/21/2022]
Abstract
Preclinical models of cancer have long been paramount to understanding tumor development and advancing the treatment of cancer. Creating preclinical models that mimic the complexity and heterogeneity of human tumors is a key challenge in the advancement of cancer therapy. About ten years ago, we created the mouse oral carcinoma (MOC) cell line models that were derived from 7, 12-dimethylbenz(a) anthracene (DMBA)-induced mouse oral squamous cell cancers. This model has been used in numerous investigations, including studies on tumor biology and therapeutics. We have seen remarkable progress in cancer immunology in recent years, and these cell lines, which are syngeneic to C57BL/6 background, have also been used to study the anti-tumor immune response. Herein, we aim to review the MOC model from its development and characterization to its use in non-immunological and immunological preclinical head and neck squamous cell carcinoma (HNSCC) studies. Integrating and refining these MOC model studies and extending findings to other systems will provide crucial insights for translational approaches aimed at improving head and neck cancer treatment.
Collapse
Affiliation(s)
- Michihisa Kono
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States; Department of Otolaryngology - Head and Neck Surgery, Asahikawa Medical University, Asahikawa, Japan.
| | - Shin Saito
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States; Department of Otolaryngology - Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan.
| | - Ann Marie Egloff
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States; Department of Surgery/Otolaryngology, Brigham and Women's Hospital, United States.
| | - Clint T Allen
- Section on Translational Tumor Immunology, National Institutes on Deafness and Communication Disorders, NIH, Bethesda, MD, United States.
| | - Ravindra Uppaluri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States; Department of Surgery/Otolaryngology, Brigham and Women's Hospital, United States.
| |
Collapse
|
14
|
Borgovan T, Yanamandra N, Schmidt H. INNATE IMMUNITY AS A TARGET FOR NOVEL THERAPEUTICS IN TRIPLE NEGATIVE BREAST CANCER. Expert Opin Investig Drugs 2022; 31:781-794. [DOI: 10.1080/13543784.2022.2096005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Theo Borgovan
- Oncology Research and DevelopmentGlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA 19426,
| | - Niranjan Yanamandra
- Immuno-Oncology & Combinations Research Unit.GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA 19426,
| | - Hank Schmidt
- Oncology Research and DevelopmentGlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA 19426,
| |
Collapse
|
15
|
Bickett TE, Knitz MW, Piper M, Oweida AJ, Gadwa J, Darragh LB, Nguyen D, Bhatia S, Bhuvane S, Phan AV, Van Court B, Corbo S, Pham T, Dent AL, Lenz L, Karam SD. Dichotomous effects of cellular expression of STAT3 on tumor growth of HNSCC. Mol Ther 2022; 30:1149-1162. [PMID: 34793974 PMCID: PMC8899526 DOI: 10.1016/j.ymthe.2021.11.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 10/17/2021] [Accepted: 11/11/2021] [Indexed: 01/12/2023] Open
Abstract
STAT3 signaling has been shown to regulate cellular function and cytokine production in the tumor microenvironment (TME). Within the head and neck squamous cell carcinoma (HNSCC) TME, we previously showed that therapeutic targeting of STAT3 in combination with radiation resulted in improved tumor growth delay. However, given the independent regulatory effects STAT3 has on anti-tumor immunity, we aimed to decipher the effects of individually targeting STAT3 in the cancer cell, regulatory T cells (Tregs), and natural killer (NK) cell compartments in driving tumor growth and resistance to therapy in HNSCCs. We utilized a CRISPR knockout system for genetic deletion of STAT3 within the cancer cell as well as two genetic knockout mouse models, FoxP3-Cre/STAT3 fl and NKp46-Cre/STAT3 fl, for Tregs and NK cell targeting, respectively. Our data revealed differences in development of resistance to treatment with STAT3 CRISPR knockout in the cancer cell, driven by differential recruitment of immune cells. Knockout of STAT3 in Tregs overcomes this resistance and results in Treg reprogramming and recruitment and activation of antigen-presenting cells. In contrast, knockout of STAT3 in the NK cell compartment results in NK cell inactivation and acceleration of tumor growth. These data underscore the complex interplay between the cancer cell and the immune TME and carry significant implications for drug targeting and design of combination approaches in HNSCCs.
Collapse
Affiliation(s)
- Thomas E Bickett
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Michael W Knitz
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Miles Piper
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ayman J Oweida
- Département de Médecine Nucléaire et Radiobiologie, Université de Sherbrooke, Sherbrooke, Quebec J1K 2R1, Canada
| | - Jacob Gadwa
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Laurel B Darragh
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Diemmy Nguyen
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Shilpa Bhatia
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Shiv Bhuvane
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Andy V Phan
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Benjamin Van Court
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sophia Corbo
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Tiffany Pham
- Department of Otolaryngology, Head and Neck Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Alexander L Dent
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Laurel Lenz
- Department of Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sana D Karam
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
16
|
Targeting Wee1 kinase to suppress proliferation and survival of cisplatin-resistant head and neck squamous cell carcinoma. Cancer Chemother Pharmacol 2022; 89:469-478. [PMID: 35212780 DOI: 10.1007/s00280-022-04410-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 02/11/2022] [Indexed: 01/10/2023]
Abstract
PURPOSE We investigated the role of Wee1 kinase in cisplatin-resistant head and neck squamous cell carcinoma (HNSCC) in multiple cisplatin-resistant HNSCC cell lines and determined the efficacy of either Wee1 inhibitor, AZD1775 alone, or in combination with cisplatin, on cisplatin-resistant HNSCC inhibition. METHODS Phosphorylation and total protein levels of cells were assessed by Western blot analysis. Cell viability and apoptosis were examined by MTS assay and flow cytometry, respectively. RESULTS Wee1 kinase protein expression levels in five cisplatin-resistant HNSCC cell types were higher than those in their parental cisplatin-sensitive partners. Importantly, Wee1 knockdown inhibited cell proliferation and re-sensitized cells to cisplatin treatment. Interestingly, previous studies have also shown that Wee1 inhibitor AZD1775 synergizes with cisplatin to suppress cell proliferation of cisplatin-sensitive HNSCC. We found that AZD1775 inhibited both cisplatin-sensitive and resistant HNSCC with similar IC50 values, which suggested that AZD1775 could overcome cisplatin resistance in cisplatin-resistant HNSCC. Mechanistically, AZD1775 and cisplatin cooperatively induced DNA damage and apoptosis. CONCLUSION Wee1 inhibitor, AZD1775, and cisplatin coordinately suppressed proliferation and survival of HNSCC.
Collapse
|
17
|
Deng B, Chen X, Xu L, Zheng L, Zhu X, Shi J, Yang L, Wang D, Jiang D. Chordin-like 1 is a novel prognostic biomarker and correlative with immune cell infiltration in lung adenocarcinoma. Aging (Albany NY) 2022; 14:389-409. [PMID: 35021154 PMCID: PMC8791215 DOI: 10.18632/aging.203814] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 12/29/2021] [Indexed: 11/25/2022]
Abstract
Chordin-like 1 (CHRDL1), an inhibitor of bone morphogenetic proteins(BMPs), has been recently reported to participate in the progression of numerous tumors, however, its role in lung adenocarcinoma (LUAD) remains unclear. Our study aimed to demonstrate relationship between CHRDL1 and LUAD based on data from The Cancer Genome Atlas (TCGA). Among them, CHRDL1 expression revealed promising power for distinguishing LUAD tissues form normal sample. Low CHRDL1 was correlated with poor clinicopathologic features, including high T stage (OR=0.45, P<0.001), high N stage (OR=0.57, P<0.003), bad treatment effect (OR=0.64, P=0.047), positive tumor status (OR=0.63, P=0.018), and TP53 mutation (OR=0.49, P<0.001). The survival curve illustrated that low CHRDL1 was significantly correlative with a poor overall survival (HR=0.60, P<0.001). At multivariate Cox regression analysis, CHRDL1 remained independently correlative with overall survival. GSEA identified that the CHRDL1 expression was related to cell cycle and immunoregulation. Immune infiltration analysis suggested that CHRDL1 was significantly correlative with 7 kinds of immune cells. Immunohistochemical validation showed that CHRDL1 was abnormally elevated and negatively correlated with Th2 cells in LUAD tissues. In conclusion, CHRDL1 might become a novel prognostic biomarker and therapy target in LUAD. Moreover, CHRDL1 may improve the effectiveness of immunotherapy by regulating immune infiltration.
Collapse
Affiliation(s)
- Bing Deng
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaorui Chen
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lingfang Xu
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Zheng
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoqian Zhu
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Junwei Shi
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lei Yang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dian Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Depeng Jiang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
18
|
Lutfi N, Galindo-Campos MA, Yélamos J. Impact of DNA Damage Response-Targeted Therapies on the Immune Response to Tumours. Cancers (Basel) 2021; 13:6008. [PMID: 34885119 PMCID: PMC8656491 DOI: 10.3390/cancers13236008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/22/2021] [Accepted: 11/26/2021] [Indexed: 01/02/2023] Open
Abstract
The DNA damage response (DDR) maintains the stability of a genome faced with genotoxic insults (exogenous or endogenous), and aberrations of the DDR are a hallmark of cancer cells. These cancer-specific DDR defects present new therapeutic opportunities, and different compounds that inhibit key components of DDR have been approved for clinical use or are in various stages of clinical trials. Although the therapeutic rationale of these DDR-targeted agents initially focused on their action against tumour cells themselves, these agents might also impact the crosstalk between tumour cells and the immune system, which can facilitate or impede tumour progression. In this review, we summarise recent data on how DDR-targeted agents can affect the interactions between tumour cells and the components of the immune system, both by acting directly on the immune cells themselves and by altering the expression of different molecules and pathways in tumour cells that are critical for their relationship with the immune system. Obtaining an in-depth understanding of the mechanisms behind how DDR-targeted therapies affect the immune system, and their crosstalk with tumour cells, may provide invaluable clues for the rational development of new therapeutic strategies in cancer.
Collapse
Affiliation(s)
- Nura Lutfi
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain; (N.L.); (M.A.G.-C.)
| | | | - José Yélamos
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain; (N.L.); (M.A.G.-C.)
- Immunology Unit, Department of Pathology, Hospital del Mar, 08003 Barcelona, Spain
| |
Collapse
|
19
|
Meng X, Gao JZ, Gomendoza SMT, Li JW, Yang S. Recent Advances of WEE1 Inhibitors and Statins in Cancers With p53 Mutations. Front Med (Lausanne) 2021; 8:737951. [PMID: 34671620 PMCID: PMC8520942 DOI: 10.3389/fmed.2021.737951] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/01/2021] [Indexed: 01/12/2023] Open
Abstract
p53 is among the most frequently mutated tumor suppressor genes given its prevalence in >50% of all human cancers. One critical tumor suppression function of p53 is to regulate transcription of downstream genes and maintain genomic stability by inducing the G1/S checkpoint in response to DNA damage. Tumor cells lacking functional p53 are defective in the G1/S checkpoint and become highly dependent on the G2/M checkpoint to maintain genomic stability and are consequently vulnerable to Wee1 inhibitors, which override the cell cycle G2/M checkpoint and induce cell death through mitotic catastrophe. In addition to the lost tumor suppression function, many mutated p53 (Mutp53) proteins acquire gain-of-function (GOF) activities as oncogenes to promote cancer progression, which manifest through aberrant expression of p53. In cancer cells with GOF Mutp53, statins can induce CHIP-mediated degradation of Mutp53 within the mevalonate pathway by blocking the interaction between mutp53 and DNAJA1. Therefore, targeting critical downstream pathways of Mutp53 provides an alternative strategy for treating cancers expressing Mutp53. In this review, we summarize recent advances with Wee1 inhibitors, statins, and mevalonate pathway inhibitors in cancers with p53 mutations.
Collapse
Affiliation(s)
- Xiangbing Meng
- Department of Pathology, The University of Iowa, Iowa City, IA, United States.,Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Jason Z Gao
- Department of Pathology, The University of Iowa, Iowa City, IA, United States
| | | | - John W Li
- Department of Pathology, The University of Iowa, Iowa City, IA, United States.,Department of Human and Evolutionary Biology, University of Southern California, Los Angeles, CA, United States
| | - Shujie Yang
- Department of Pathology, The University of Iowa, Iowa City, IA, United States.,Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
20
|
WEE1 inhibition reverses trastuzumab resistance in HER2-positive cancers. Gastric Cancer 2021; 24:1003-1020. [PMID: 33723720 DOI: 10.1007/s10120-021-01176-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 02/19/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND To date, many efforts have been made to understand the resistance mechanism of trastuzumab in human epidermal growth factor receptor 2 (HER2)-positive breast and gastric cancer. However, there is still a huge unmet medical need for patients with trastuzumab resistance. METHODS In our study, we generated four trastuzumab-resistant (HR) cancer cell lines from ERBB2-amplified gastric and biliary tract cancer cell lines (SNU-216, NCI-N87, SNU-2670, and SNU-2773). RESULTS Here, we found higher PD-L1 expression in trastuzumab-resistant (HR) HER2-positive cancer cells than in parental cells, and blocking PD-L1 reversed the resistance to trastuzumab in HR cells. Trastuzumab upregulated PD-L1 expression via NF-κB activation in both parental and HR cells, however, led to DNA damage only in parental cells. The WEE1 inhibitor adavosertib, which downregulates PD-L1 expression, enhanced trastuzumab efficacy by blocking BRCA1-CMTM6-PD-L1 signals and the HER2-CDCP-1-SRC axis. Additionally, the levels of galectin-9, CD163, FoxP3, and CTLA-4 were diminished by blocking WEE1 in the presence of human PBMCs in vitro. CONCLUSION Taken together, the strategy of co-targeting HER2 and WEE1 could overcome resistance to trastuzumab in HER2-positive cancers, supporting further clinical development in HER2-positive cancer patients.
Collapse
|
21
|
Solocinski K, Padget MR, Fabian KP, Wolfson B, Cecchi F, Hembrough T, Benz SC, Rabizadeh S, Soon-Shiong P, Schlom J, Hodge JW. Overcoming hypoxia-induced functional suppression of NK cells. J Immunother Cancer 2021; 8:jitc-2019-000246. [PMID: 32345623 PMCID: PMC7213912 DOI: 10.1136/jitc-2019-000246] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2020] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Natural killer (NK) cells are immune cells capable of killing virally infected cells and tumor cells without the need for antigen stimulation. Tumors, however, can create a suppressive microenvironment that decreases NK function. A feature of many tumors is hypoxia (low oxygen perfusion), which has been previously shown to decrease NK function. A high affinity NK (haNK) cell has been engineered to express a high affinity CD16 receptor as well as internal interleukin (IL)-2 for increased antibody-dependent cellular cytotoxicity (ADCC) and activation, respectively. We sought to investigate the tolerance of NK cells versus haNK cells to hypoxia. METHODS We exposed healthy donor (HD) NK and X-irradiated haNK cells to normoxia (20% oxygen) as well as hypoxia (0% oxygen) and investigated their ability to kill prostate, breast and lung tumor cell lines after 5 hours. We also used monoclonal antibodies cetuximab (anti-EGFR) or avelumab (antiprogrammed death-ligand 1) to investigate the effects of hypoxia on NK ADCC. Genomic and proteomic analyzes were done to determine the effect of hypoxia on the expression of factors important to NK cell function. RESULTS While HD NK cell cytolytic abilities were markedly and significantly impaired under hypoxic conditions, haNK cells maintained killing capacity under hypoxic conditions. NK killing, serial killing and ADCC were maintained under hypoxia in haNK cells. IL-2 has been previously implicated in serial killing and perforin regeneration and thus the endogenous IL-2 produced by haNK cells is likely a driver of the maintained killing capacity of haNK cells under hypoxic conditions. Activation of signal transducer and activator of transcription 3 (STAT3) is not seen in haNKs under hypoxia but is significant in HD NK cells. Pharmaceutical activation of STAT3 in haNKs led to reduced killing, implicating active STAT3 in reduced NK cell function. CONCLUSIONS In contrast to HD NK cells, haNK cells are resistant to acute hypoxia. The potent cytolytic function of haNK cells was maintained in an environment comparable to what would be encountered in a tumor. The data presented here provide an additional mechanism of action for haNK cells that are currently being evaluated in clinical trials for several tumor types.
Collapse
Affiliation(s)
- Kristen Solocinski
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Michelle R Padget
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Kellsye P Fabian
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Benjamin Wolfson
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | | | | | | | - Shahrooz Rabizadeh
- NantOmics, Culver City, California, USA.,ImmunityBio, Culver City, California, USA
| | - Patrick Soon-Shiong
- NantOmics, Culver City, California, USA.,ImmunityBio, Culver City, California, USA
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - James W Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
22
|
Takebe N, Naqash AR, O'Sullivan Coyne G, Kummar S, Do K, Bruns A, Juwara L, Zlott J, Rubinstein L, Piekarz R, Sharon E, Streicher H, Mittra A, Miller SB, Ji J, Wilsker D, Kinders RJ, Parchment RE, Chen L, Chang TC, Das B, Mugundu G, Doroshow JH, Chen AP. 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. [PMID: 33863809 PMCID: PMC8282703 DOI: 10.1158/1078-0432.ccr-21-0329] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/24/2021] [Accepted: 04/13/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE The Wee1 kinase inhibitor adavosertib abrogates cell-cycle arrest, leading to cell death. Prior testing of twice-daily adavosertib in patients with advanced solid tumors determined the recommended phase II dose (RPh2D). Here, we report results for once-daily adavosertib. PATIENTS AND METHODS A 3 + 3 dose-escalation design was used, with adavosertib given once daily on days 1 to 5 and 8 to 12 in 21-day cycles. Molecular biomarkers of Wee1 activity, including tyrosine 15-phosphorylated Cdk1/2 (pY15-Cdk), were assessed in paired tumor biopsies. Whole-exome sequencing and RNA sequencing of remaining tumor tissue identified potential predictive biomarkers. RESULTS Among the 42 patients enrolled, the most common toxicities were gastrointestinal and hematologic; dose-limiting toxicities were grade 4 hematologic toxicity and grade 3 fatigue. The once-daily RPh2D was 300 mg. Six patients (14%) had confirmed partial responses: four ovarian, two endometrial. Adavosertib plasma exposures were similar to those from twice-daily dosing. On cycle 1 day 8 (pre-dose), tumor pY15-Cdk levels were higher than baseline in four of eight patients, suggesting target rebound during the day 5 to 8 dosing break. One patient who progressed rapidly had a tumor WEE1 mutation and potentially compensatory PKMYT1 overexpression. Baseline CCNE1 overexpression occurred in both of two responding patients, only one of whom had CCNE1 amplification, and in zero of three nonresponding patients. CONCLUSIONS We determined the once-daily adavosertib RPh2D and observed activity in patients with ovarian or endometrial carcinoma, including two with baseline CCNE1 mRNA overexpression. Future studies will determine whether CCNE1 overexpression is a predictive biomarker for adavosertib.
Collapse
Affiliation(s)
- Naoko Takebe
- Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland
- Center for Cancer Research, NCI, Bethesda, Maryland
| | | | - Geraldine O'Sullivan Coyne
- Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland
- Center for Cancer Research, NCI, Bethesda, Maryland
| | - Shivaani Kummar
- Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland
- Center for Cancer Research, NCI, Bethesda, Maryland
| | - Khanh Do
- Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland
- Center for Cancer Research, NCI, Bethesda, Maryland
| | - Ashley Bruns
- Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland
| | - Lamin Juwara
- Clinical Monitoring Research Program, Clinical Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Jennifer Zlott
- Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland
| | - Larry Rubinstein
- Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland
| | - Richard Piekarz
- Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland
| | - Elad Sharon
- Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland
| | - Howard Streicher
- Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland
| | - Arjun Mittra
- Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland
| | - Sarah B Miller
- Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland
| | - Jiuping Ji
- Clinical Pharmacodynamic Biomarkers Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Deborah Wilsker
- Clinical Pharmacodynamic Biomarkers Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Robert J Kinders
- Clinical Pharmacodynamic Biomarkers Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Ralph E Parchment
- Clinical Pharmacodynamic Biomarkers Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Li Chen
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Ting-Chia Chang
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Biswajit Das
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Ganesh Mugundu
- AstraZeneca, Clinical Pharmacology, Waltham, Massachusetts
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland
- Center for Cancer Research, NCI, Bethesda, Maryland
| | - Alice P Chen
- Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland.
- Center for Cancer Research, NCI, Bethesda, Maryland
| |
Collapse
|
23
|
Marin-Acevedo JA, Kimbrough EO, Manochakian R, Zhao Y, Lou Y. Immunotherapies targeting stimulatory pathways and beyond. J Hematol Oncol 2021; 14:78. [PMID: 33980266 PMCID: PMC8117548 DOI: 10.1186/s13045-021-01085-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 04/27/2021] [Indexed: 12/20/2022] Open
Abstract
Co-stimulatory and co-inhibitory molecules play a critical role in T cell function. Tumor cells escape immune surveillance by promoting immunosuppression. Immunotherapy targeting inhibitory molecules like anti-CTLA-4 and anti-PD-1/PD-L1 were developed to overcome these immunosuppressive effects. These agents have demonstrated remarkable, durable responses in a small subset of patients. The other mechanisms for enhancing anti-tumor activities are to target the stimulatory pathways that are expressed on T cells or other immune cells. In this review, we summarize current phase I/II clinical trials evaluating novel immunotherapies targeting stimulatory pathways and outline their advantages, limitations, and future directions.
Collapse
Affiliation(s)
- Julian A Marin-Acevedo
- Department of Hematology and Oncology, H. Lee Moffitt Cancer Center, 12902 USF Magnolia Drive, Tampa, 33612, FL, USA
| | - ErinMarie O Kimbrough
- Division of Hematology and Oncology, Mayo Clinic, 4500 San Pablo Road S., Jacksonville, FL, 32224, USA
| | - Rami Manochakian
- Division of Hematology and Oncology, Mayo Clinic, 4500 San Pablo Road S., Jacksonville, FL, 32224, USA
| | - Yujie Zhao
- Division of Hematology and Oncology, Mayo Clinic, 4500 San Pablo Road S., Jacksonville, FL, 32224, USA
| | - Yanyan Lou
- Division of Hematology and Oncology, Mayo Clinic, 4500 San Pablo Road S., Jacksonville, FL, 32224, USA.
| |
Collapse
|
24
|
Yap TA, Parkes EE, Peng W, Moyers JT, Curran MA, Tawbi HA. Development of Immunotherapy Combination Strategies in Cancer. Cancer Discov 2021; 11:1368-1397. [PMID: 33811048 DOI: 10.1158/2159-8290.cd-20-1209] [Citation(s) in RCA: 208] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 01/03/2021] [Accepted: 02/01/2021] [Indexed: 12/11/2022]
Abstract
Harnessing the immune system to treat cancer through inhibitors of CTLA4 and PD-L1 has revolutionized the landscape of cancer. Rational combination strategies aim to enhance the antitumor effects of immunotherapies, but require a deep understanding of the mechanistic underpinnings of the immune system and robust preclinical and clinical drug development strategies. We review the current approved immunotherapy combinations, before discussing promising combinatorial approaches in clinical trials and detailing innovative preclinical model systems being used to develop rational combinations. We also discuss the promise of high-order immunotherapy combinations, as well as novel biomarker and combinatorial trial strategies. SIGNIFICANCE: Although immune-checkpoint inhibitors are approved as dual checkpoint strategies, and in combination with cytotoxic chemotherapy and angiogenesis inhibitors for multiple cancers, patient benefit remains limited. Innovative approaches are required to guide the development of novel immunotherapy combinations, ranging from improvements in preclinical tumor model systems to biomarker-driven trial strategies.
Collapse
Affiliation(s)
- Timothy A Yap
- Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Khalifa Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eileen E Parkes
- Oxford Institute of Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Weiyi Peng
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Justin T Moyers
- Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael A Curran
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hussein A Tawbi
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
25
|
Chen J, Jia X, Li Z, Song W, Jin C, Zhou M, Xie H, Zheng S, Song P. Targeting WEE1 by adavosertib inhibits the malignant phenotypes of hepatocellular carcinoma. Biochem Pharmacol 2021; 188:114494. [PMID: 33684390 DOI: 10.1016/j.bcp.2021.114494] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/20/2021] [Accepted: 02/24/2021] [Indexed: 10/22/2022]
Abstract
Targeting the cell cycle checkpoints and DNA damage response are promising therapeutic strategies for cancer. Adavosertib is a potent inhibitor of WEE1 kinase, which plays a critical role in regulating cell cycle checkpoints. However, the effect of adavosertib on hepatocellular carcinoma (HCC) treatment, including sorafenib-resistant HCC, has not been thoroughly studied. In this study, we comprehensively investigated the efficacy and pharmacology of adavosertib in HCC therapy. Adavosertib effectively inhibited the proliferation of HCC cells in vitro and suppressed tumor growth in HCC xenografts and patient-derived xenograft (PDX) models in vivo. Additionally, adavosertib treatment effectively inhibited the motility of HCC cells by impairing pseudopodia formation. Further, we revealed that adavosertib induced DNA damage and premature mitosis entrance by disturbing the cell cycle. Thus, HCC cells accumulating DNA damage underwent mitosis without G2/M checkpoint arrest, thereby leading to mitotic catastrophe and apoptosis under adavosertib administration. Given that sorafenib resistance is common in HCC in clinical practice, we also explored the efficacy of adavosertib in sorafenib-resistant HCC. Notably, adavosertib still showed a desirable inhibitory effect on the growth of sorafenib-resistant HCC cells. Adavosertib markedly induced G2/M checkpoint arrest and cell apoptosis in a dose-dependent manner, confirming the similar efficacy of adavosertib in sorafenib-resistant HCC. Collectively, our results highlight the treatment efficacy of adavosertib in HCC regardless of sorafenib resistance, providing insights into exploring novel strategies for HCC therapy.
Collapse
Affiliation(s)
- Jian Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Xing Jia
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Zequn Li
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Wenfeng Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Cheng Jin
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Mengqiao Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Haiyang Xie
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China.
| | - Penghong Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China.
| |
Collapse
|
26
|
Charap AJ, Enokida T, Brody R, Sfakianos J, Miles B, Bhardwaj N, Horowitz A. Landscape of natural killer cell activity in head and neck squamous cell carcinoma. J Immunother Cancer 2020; 8:jitc-2020-001523. [PMID: 33428584 PMCID: PMC7754625 DOI: 10.1136/jitc-2020-001523] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2020] [Indexed: 12/13/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) encompasses a set of cancers arising from the epithelia of the upper aerodigestive tract, accounting for a significant burden of disease worldwide due to the disease’s mortality, morbidity, and predilection for recurrence. Prognosis of HNSCC in the recurrent and/or metastatic (R/M-HNSCC) setting is especially poor and effective treatment options increasingly rely on modulating T-cell antitumor responses. Still, immunotherapy response rates are generally low, prompting the exploration of novel strategies that incorporate other effector cells within the tumor microenvironment. Within the last decade, important advances have been made leveraging the powerful innate antitumor function of natural killer (NK) cells to treat solid tumors, including head and neck squamous cell carcinoma. NK cells are hybrid innate-adaptive effector cells capable of directly eliminating tumor cells in addition to initiating adaptive antitumor immune responses. In the setting of HNSCC, NK cells are important for tumor surveillance and control, and NK cell infiltration has repeatedly been associated with a favorable prognosis. Yet, HNSCC-infiltrating NK cells are susceptible to an array of immune evasion strategies employed by tumors that must be overcome to fully realize the antitumor potential of NK cells. We believe that a conceptual framework informed by the basic biological understanding of the mechanisms underlying NK cell activation can improve treatment of HNSCC, in part by selecting for patients most likely to respond to NK cell-based immunotherapy. Herein, we review the activity of NK cells in HNSCC, paying special attention to the role of environmental and genetic determinants of NK cell antitumor function. Moreover, we explore the evidence that NK cells are a crucial determinant of the efficacy of both established and emerging treatments for HNSCC.
Collapse
Affiliation(s)
- Andrew J Charap
- Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Tomohiro Enokida
- Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rachel Brody
- Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - John Sfakianos
- Urology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Brett Miles
- Otolaryngology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Nina Bhardwaj
- Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Amir Horowitz
- Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
27
|
MacDonald KM, Benguerfi S, Harding SM. Alerting the immune system to DNA damage: micronuclei as mediators. Essays Biochem 2020; 64:753-764. [PMID: 32844183 PMCID: PMC7588664 DOI: 10.1042/ebc20200016] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/01/2020] [Accepted: 08/13/2020] [Indexed: 12/18/2022]
Abstract
Healthy cells experience thousands of DNA lesions per day during normal cellular metabolism, and ionizing radiation and chemotherapeutic drugs rely on DNA damage to kill cancer cells. In response to such lesions, the DNA damage response (DDR) activates cell-cycle checkpoints, initiates DNA repair mechanisms, or promotes the clearance of irreparable cells. Work over the past decade has revealed broader influences of the DDR, involving inflammatory gene expression following unresolved DNA damage, and immune surveillance of damaged or mutated cells. Subcellular structures called micronuclei, containing broken fragments of DNA or whole chromosomes that have been isolated away from the rest of the genome, are now recognized as one mediator of DDR-associated immune recognition. Micronuclei can initiate pro-inflammatory signaling cascades, or massively degrade to invoke distinct forms of genomic instability. In this mini-review, we aim to provide an overview of the current evidence linking the DDR to activation of the immune response through micronuclei formation, identifying key areas of interest, open questions, and emerging implications.
Collapse
Affiliation(s)
- Kate M MacDonald
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Soraya Benguerfi
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
| | - Shane M Harding
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
- Department of Radiation Oncology and Immunology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
28
|
Abstract
Head and neck squamous cell carcinoma (HNSCC) associated with high-risk human papilloma virus (HPV) infection is a growing clinical problem. The WEE1 kinase inhibitor AZD1775 (WEE1i) overrides cell cycle checkpoints and is being studied in HNSCC regimens. We show that the HPV16 E6/E7 oncoproteins sensitize HNSCC cells to single-agent WEE1i treatment through activation of a FOXM1-CDK1 circuit that drives mitotic gene expression and DNA damage. An isogenic cell system indicated that E6 largely accounts for these phenotypes in ways that extend beyond p53 inactivation. A targeted genomic analysis implicated FOXM1 signaling downstream of E6/E7 expression and analyses of primary tumors and The Cancer Genome Atlas (TCGA) data revealed an activated FOXM1-directed promitotic transcriptional signature in HPV+ versus HPV- HNSCCs. Finally, we demonstrate the causality of FOXM1 in driving WEE1i sensitivity. These data suggest that elevated basal FOXM1 activity predisposes HPV+ HNSCC to WEE1i-induced toxicity and provide mechanistic insights into WEE1i and HPV+ HNSCC therapies.
Collapse
|
29
|
Du X, Li J, Luo X, Li R, Li F, Zhang Y, Shi J, He J. Structure-activity relationships of Wee1 inhibitors: A review. Eur J Med Chem 2020; 203:112524. [PMID: 32688199 DOI: 10.1016/j.ejmech.2020.112524] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/28/2020] [Accepted: 06/01/2020] [Indexed: 02/05/2023]
Abstract
Wee1 kinase plays an important role in regulating G2/M checkpoint and S phase, and the inhibition of it will lead to mitotic catastrophe in cancer cells with p53 mutation or deletion. Therefore, the mechanism of Wee1 kinase in cancer treatment and the development of its inhibitors have become a research hotspot. However, although a variety of Wee1 inhibitors with different scaffolds and considerable activity have been successfully identified, so far no one has systematically summarized the structure-activity relationships (SARs) of Wee1 inhibitors. Previous reviews mainly focused on its mechanism and clinical application. To facilitate the rational design and development of Wee1 inhibitors in the future, this paper systematically summarizes its structural types, SARs and binding modes according to the Wee1 inhibitors reported in scientific journals, and also summarizes the regulatory effect of Wee1 kinase on cell cycle and the progress of its inhibitors in clinical application.
Collapse
Affiliation(s)
- Xingkai Du
- State Key Laboratory of Biotherapy & Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Jian Li
- Department of Pharmacy, West China Hospital Sichuan University, Chengdu, 610041, China
| | - Xiaojiao Luo
- State Key Laboratory of Biotherapy & Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Rong Li
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Feng Li
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yiwen Zhang
- State Key Laboratory of Biotherapy & Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China.
| | - Jianyou Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of pharmacy, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China.
| | - Jun He
- State Key Laboratory of Biotherapy & Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
30
|
Wang B, Sun L, Yuan Z, Tao Z. Wee1 kinase inhibitor AZD1775 potentiates CD8+ T cell-dependent antitumour activity via dendritic cell activation following a single high dose of irradiation. Med Oncol 2020; 37:66. [PMID: 32696094 DOI: 10.1007/s12032-020-01390-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/05/2020] [Indexed: 12/12/2022]
Abstract
As standard treatments for cancer, DNA-damaging chemotherapeutic agents and irradiation therapy improve survival in patients with various cancers. Wee1, a kinase associated with the cell cycle, causes G2/M cell cycle arrest to allow repair of injured DNA in cancer cells, and a Wee1 inhibitor has been confirmed to lead to apoptosis in cancer cells. Recently, there has been renewed interest in exploring the immune environment which plays a significant role in tumour suppression. A Wee1 inhibitor combined with radiotherapy has been tested in lung, pancreatic, and prostate cancer and melanoma in vivo or in vitro. There is still no research evaluating the immunoregulatory effects of AZD1775 plus high-dose irradiation (IR) in vivo. T cell killing and CD8+ T cell depletion assays demonstrated that the combination of AZD1775 and IR delayed tumour growth in breast cancer mouse models. Additionally, combination treatment also suppressed the expression of PD-L1, a co-inhibitor, through the STAT3-IRF1 axis. The importance and originality of this study are that it explores the internal and external mechanisms of AZD1775 combined with a single high dose of IR and provides a rationale for applying the combination therapy described above in a clinical trial.
Collapse
Affiliation(s)
- Bin Wang
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China
- Department of Radiation Oncology, Xijing Hospital, The Fourth Military Medical University, No. 127, Chang Le West Road, Xi'an, 710032, China
| | - Lin Sun
- Department of Pathology, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zhiyong Yuan
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China.
- Department of Radiation Oncology and Cyberknife Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.
| | - Zhen Tao
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China.
- Department of Radiation Oncology and Cyberknife Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.
| |
Collapse
|
31
|
Robbins Y, Greene S, Friedman J, Clavijo PE, Van Waes C, Fabian KP, Padget MR, Abdul Sater H, Lee JH, Soon-Shiong P, Gulley J, Schlom J, Hodge JW, Allen CT. Tumor control via targeting PD-L1 with chimeric antigen receptor modified NK cells. eLife 2020; 9:e54854. [PMID: 32633234 PMCID: PMC7340502 DOI: 10.7554/elife.54854] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 06/24/2020] [Indexed: 12/13/2022] Open
Abstract
Failed T cell-based immunotherapies in the presence of genomic alterations in antigen presentations pathways may be overcome by NK cell-based immunotherapy. This approach may still be limited by the presence of immunosuppressive myeloid populations. Here, we demonstrate that NK cells (haNKs) engineered to express a PD-L1 chimeric antigen receptor (CAR) haNKs killed a panel of human and murine head and neck cancer cells at low effector-to-target ratios in a PD-L1-dependent fashion. Treatment of syngeneic tumors resulted in CD8 and PD-L1-dependent tumor rejection or growth inhibition and a reduction in myeloid cells endogenously expressing high levels of PD-L1. Treatment of xenograft tumors resulted in PD-L1-dependent tumor growth inhibition. PD-L1 CAR haNKs reduced levels of macrophages and other myeloid cells endogenously expressing high PD-L1 in peripheral blood from patients with head and neck cancer. The clinical study of PD-L1 CAR haNKs is warranted.
Collapse
Affiliation(s)
- Yvette Robbins
- Translational Tumor Immunology Program, National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Sarah Greene
- Translational Tumor Immunology Program, National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Jay Friedman
- Translational Tumor Immunology Program, National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Paul E Clavijo
- Translational Tumor Immunology Program, National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Carter Van Waes
- Tumor Biology Section, National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Kellsye P Fabian
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, National Institutes of HealthBethesdaUnited States
| | - Michelle R Padget
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, National Institutes of HealthBethesdaUnited States
| | - Houssein Abdul Sater
- Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of HealthBethesdaUnited States
| | | | | | - James Gulley
- Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of HealthBethesdaUnited States
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, National Institutes of HealthBethesdaUnited States
| | - James W Hodge
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, National Institutes of HealthBethesdaUnited States
| | - Clint T Allen
- Translational Tumor Immunology Program, National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of MedicineBaltimoreUnited States
| |
Collapse
|
32
|
Chew HY, Dolcetti R, Simpson F. Scientifically based combination therapies with immuno-oncology checkpoint inhibitors. Br J Clin Pharmacol 2020; 86:1711-1725. [PMID: 32372470 DOI: 10.1111/bcp.14338] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 04/14/2020] [Accepted: 04/27/2020] [Indexed: 12/20/2022] Open
Abstract
The discovery of immune checkpoints and their role in modulating immune response have revolutionised cancer treatment in recent years. The immune checkpoints, cytotoxic T-lymphocyte-associated protein 4, programmed cell death protein 1 and its ligand, programmed cell death-ligand 1, have been extensively studied. Currently 7 monoclonal antibodies targeting these immune checkpoints are approved for treatment of various cancers. Inhibiting immune checkpoints has shown some success in clinic, however, a proportion of patients do not benefit from this treatment. Several other inhibitory molecules, in addition to lymphocyte-associated protein 4 and programmed cell death protein 1, are known to be involved in regulating immune response. To further improve patient outcomes, studies have examined targeting these inhibitory molecules through combination therapies. This review discusses the current landscape of combination therapies of checkpoint inhibitors.
Collapse
Affiliation(s)
- Hui Yi Chew
- The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| | - Riccardo Dolcetti
- The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| | - Fiona Simpson
- The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| |
Collapse
|
33
|
Zeng L, Nikolaev A, Xing C, Della Manna DL, Yang ES. CHK1/2 Inhibitor Prexasertib Suppresses NOTCH Signaling and Enhances Cytotoxicity of Cisplatin and Radiation in Head and Neck Squamous Cell Carcinoma. Mol Cancer Ther 2020; 19:1279-1288. [PMID: 32371584 DOI: 10.1158/1535-7163.mct-19-0946] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 01/08/2020] [Accepted: 04/01/2020] [Indexed: 11/16/2022]
Abstract
Platinum-based chemoradiotherapy is a mainstay of organ-preserving therapy for patients with head and neck squamous cell carcinoma cancer (HNSCC). However, the disease eventually becomes resistant to treatment necessitating new therapies. Checkpoint kinase 1 and 2 (CHK1/2) are serine/threonine kinases that activate cell-cycle checkpoints and serve a critical role in the DNA-damage response (DDR). As resistance to cisplatin and radiation may involve a heightened DDR, we hypothesized that prexasertib, an inhibitor of CHK1/2, may enhance the cytotoxicity induced by cisplatin and irradiation in HNSCC. In this study, we found that combining prexasertib with cisplatin and radiation significantly decreased the in vitro survival fraction in HNSCC cell lines both with and without radiotherapy. Reduced survival was accompanied by inhibition of DNA repair checkpoint activation, which resulted in persistent DNA damage and increased apoptosis. In addition, NanoString analysis with the PanCancer Pathways Panel revealed that prexasertib downregulated NOTCH signaling target genes (NOTCH1, NOTCH2, and NOTCH3) and their associated ligands (JAG1, JAG2, SKP2, MAML2, and DLL1). Prexasertib also reduced NOTCH1, NOTCH3 and HES1 protein expression. Importantly, a significant tumor growth delay was observed in vivo in both human papillomavirus (HPV)-positive UM-SCC47 and HPV-negative UM-SCC1 cell line xenografts treated with prexasertib, cisplatin, and radiotherapy without increased toxicity as measured by mouse body weight. Taken together, prexasertib reduced NOTCH signaling and enhanced the in vitro and in vivo response of HNSCCs to cisplatin and radiation, suggesting combination therapy may increase clinical benefit. A clinical trial has recently completed accrual (NCT02555644).
Collapse
Affiliation(s)
- Ling Zeng
- Department of Radiation Oncology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - Anatoly Nikolaev
- Department of Radiation Oncology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - Chuan Xing
- Department of Radiation Oncology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - Deborah L Della Manna
- Department of Radiation Oncology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - Eddy S Yang
- Department of Radiation Oncology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama. .,Department of Pharmacology and Toxiology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama.,Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama.,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| |
Collapse
|
34
|
Huang RX, Zhou PK. DNA damage response signaling pathways and targets for radiotherapy sensitization in cancer. Signal Transduct Target Ther 2020; 5:60. [PMID: 32355263 PMCID: PMC7192953 DOI: 10.1038/s41392-020-0150-x] [Citation(s) in RCA: 643] [Impact Index Per Article: 128.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/20/2020] [Accepted: 03/16/2020] [Indexed: 12/19/2022] Open
Abstract
Radiotherapy is one of the most common countermeasures for treating a wide range of tumors. However, the radioresistance of cancer cells is still a major limitation for radiotherapy applications. Efforts are continuously ongoing to explore sensitizing targets and develop radiosensitizers for improving the outcomes of radiotherapy. DNA double-strand breaks are the most lethal lesions induced by ionizing radiation and can trigger a series of cellular DNA damage responses (DDRs), including those helping cells recover from radiation injuries, such as the activation of DNA damage sensing and early transduction pathways, cell cycle arrest, and DNA repair. Obviously, these protective DDRs confer tumor radioresistance. Targeting DDR signaling pathways has become an attractive strategy for overcoming tumor radioresistance, and some important advances and breakthroughs have already been achieved in recent years. On the basis of comprehensively reviewing the DDR signal pathways, we provide an update on the novel and promising druggable targets emerging from DDR pathways that can be exploited for radiosensitization. We further discuss recent advances identified from preclinical studies, current clinical trials, and clinical application of chemical inhibitors targeting key DDR proteins, including DNA-PKcs (DNA-dependent protein kinase, catalytic subunit), ATM/ATR (ataxia-telangiectasia mutated and Rad3-related), the MRN (MRE11-RAD50-NBS1) complex, the PARP (poly[ADP-ribose] polymerase) family, MDC1, Wee1, LIG4 (ligase IV), CDK1, BRCA1 (BRCA1 C terminal), CHK1, and HIF-1 (hypoxia-inducible factor-1). Challenges for ionizing radiation-induced signal transduction and targeted therapy are also discussed based on recent achievements in the biological field of radiotherapy.
Collapse
Affiliation(s)
- Rui-Xue Huang
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, 410078, Changsha, People's Republic of China
| | - Ping-Kun Zhou
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, AMMS, 100850, Beijing, People's Republic of China.
- Institute for Chemical Carcinogenesis, State Key Laboratory of Respiratory, Guangzhou Medical University, 511436, Guangzhou, People's Republic of China.
| |
Collapse
|
35
|
Lee MY, Allen CT. Mechanisms of resistance to T cell-based immunotherapy in head and neck cancer. Head Neck 2020; 42:2722-2733. [PMID: 32275098 DOI: 10.1002/hed.26158] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/09/2020] [Accepted: 03/24/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Most current approved or investigational immunotherapeutic approaches for head and neck squamous cell carcinoma are aimed at activating T cells. The majority of patients receiving such immunotherapy do not demonstrate durable tumor remission. METHODS Original articles covering tumor heterogeneity, immunoediting, immune escape, and local tumor immunosuppression were reviewed. RESULTS In the face of immune pressure, subclones susceptible to T cell killing are eliminated, leaving behind resistant tumor clones in a process known as immunoediting. Such subclones of tumor cells that are resistant to T cell killing may remain sensitive to natural killer (NK) cell detection and elimination, suggesting that patients harboring such tumors may benefit from combination of T and NK cell-based immunotherapy. Even in the setting of optimal immunotherapy, the immunosuppressive tumor microenvironment may arrogate effector immune responses through a number of distinct mechanisms. CONCLUSIONS Highly effective immunotherapy will likely require multimodality approaches targeting independent mechanisms of immune activation.
Collapse
Affiliation(s)
- Maxwell Y Lee
- Translational Tumor Immunology Program, National Institute on Deafness and Other Commination Disorders, National Institutes of Health, Bethesda, Maryland, USA
| | - Clint T Allen
- Translational Tumor Immunology Program, National Institute on Deafness and Other Commination Disorders, National Institutes of Health, Bethesda, Maryland, USA.,Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
36
|
McLaughlin M, Patin EC, Pedersen M, Wilkins A, Dillon MT, Melcher AA, Harrington KJ. Inflammatory microenvironment remodelling by tumour cells after radiotherapy. Nat Rev Cancer 2020; 20:203-217. [PMID: 32161398 DOI: 10.1038/s41568-020-0246-1] [Citation(s) in RCA: 534] [Impact Index Per Article: 106.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/12/2020] [Indexed: 12/17/2022]
Abstract
The development of immune checkpoint inhibitors (ICIs) is revolutionizing the way we think about cancer treatment. Even so, for most types of cancer, only a minority of patients currently benefit from ICI therapies. Intrinsic and acquired resistance to ICIs has focused research towards new combination therapy approaches that seek to increase response rates, the depth of remission and the durability of benefit. In this Review, we describe how radiotherapy, through its immunomodulating effects, represents a promising combination partner with ICIs. We describe how recent research on DNA damage response (DDR) inhibitors in combination with radiotherapy may be used to augment this approach. Radiotherapy can kill cancer cells while simultaneously triggering the release of pro-inflammatory mediators and increasing tumour-infiltrating immune cells - phenomena often described colloquially as turning immunologically 'cold' tumours 'hot'. Here, we focus on new developments illustrating the key role of tumour cell-autonomous signalling after radiotherapy. Radiotherapy-induced tumour cell micronuclei activate cytosolic nucleic acid sensor pathways, such as cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING), and propagation of the resulting inflammatory signals remodels the immune contexture of the tumour microenvironment. In parallel, radiation can impact immunosurveillance by modulating neoantigen expression. Finally, we highlight how tumour cell-autonomous mechanisms might be exploited by combining DDR inhibitors, ICIs and radiotherapy.
Collapse
Affiliation(s)
- Martin McLaughlin
- Targeted Therapy Team, The Institute of Cancer Research, London, UK.
| | - Emmanuel C Patin
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | - Malin Pedersen
- Translational Immunotherapy Team, The Institute of Cancer Research, London, UK
| | | | - Magnus T Dillon
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Alan A Melcher
- Translational Immunotherapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Kevin J Harrington
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| |
Collapse
|
37
|
Ben-Shmuel A, Biber G, Barda-Saad M. Unleashing Natural Killer Cells in the Tumor Microenvironment-The Next Generation of Immunotherapy? Front Immunol 2020; 11:275. [PMID: 32153582 PMCID: PMC7046808 DOI: 10.3389/fimmu.2020.00275] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 02/03/2020] [Indexed: 12/12/2022] Open
Abstract
The emergence of immunotherapy for cancer treatment bears considerable clinical promise. Nevertheless, many patients remain unresponsive, acquire resistance, or suffer dose-limiting toxicities. Immune-editing of tumors assists their escape from the immune system, and the tumor microenvironment (TME) induces immune suppression through multiple mechanisms. Immunotherapy aims to bolster the activity of immune cells against cancer by targeting these suppressive immunomodulatory processes. Natural Killer (NK) cells are a heterogeneous subset of immune cells, which express a diverse array of activating and inhibitory germline-encoded receptors, and are thus capable of directly targeting and killing cancer cells without the need for MHC specificity. Furthermore, they play a critical role in triggering the adaptive immune response. Enhancing the function of NK cells in the context of cancer is therefore a promising avenue for immunotherapy. Different NK-based therapies have been evaluated in clinical trials, and some have demonstrated clinical benefits, especially in the context of hematological malignancies. Solid tumors remain much more difficult to treat, and the time point and means of intervention of current NK-based treatments still require optimization to achieve long term effects. Here, we review recently described mechanisms of cancer evasion from NK cell immune surveillance, and the therapeutic approaches that aim to potentiate NK function. Specific focus is placed on the use of specialized monoclonal antibodies against moieties on the cancer cell, or on both the tumor and the NK cell. In addition, we highlight newly identified mechanisms that inhibit NK cell activity in the TME, and describe how biochemical modifications of the TME can synergize with current treatments and increase susceptibility to NK cell activity.
Collapse
Affiliation(s)
- Aviad Ben-Shmuel
- Laboratory of Molecular and Applied Immunology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Guy Biber
- Laboratory of Molecular and Applied Immunology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Mira Barda-Saad
- Laboratory of Molecular and Applied Immunology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
38
|
Flow cytometry-based assessment of direct-targeting anti-cancer antibody immune effector functions. Methods Enzymol 2020; 632:431-456. [PMID: 32000909 PMCID: PMC7000137 DOI: 10.1016/bs.mie.2019.07.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Monoclonal antibody-based therapies are increasingly being used to treat cancer. Some mediate their therapeutic effects through modifying the function of immune cells globally, while others bind directly to tumor cells and can recruit immune effector cells through their Fc regions. As new direct-binding agents are developed, having the ability to test their Fc-mediated functions in a high-throughput manner is important for selecting antibodies with immune effector properties. Here, using monoclonal anti-CD20 antibody (rituximab) as an example and the CD20+ Raji cell line as tumor target, we describe flow cytometry-based assays for determining an antibody's capacity for mediating antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP) and complement-dependent cytotoxicity (CDC). These assays are sensitive, reliable, affordable and avoid the use of radioactivity.
Collapse
|
39
|
Greene S, Robbins Y, Mydlarz WK, Huynh AP, Schmitt NC, Friedman J, Horn LA, Palena C, Schlom J, Maeda DY, Zebala JA, Clavijo PE, Allen C. Inhibition of MDSC Trafficking with SX-682, a CXCR1/2 Inhibitor, Enhances NK-Cell Immunotherapy in Head and Neck Cancer Models. Clin Cancer Res 2019; 26:1420-1431. [PMID: 31848188 DOI: 10.1158/1078-0432.ccr-19-2625] [Citation(s) in RCA: 198] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 11/06/2019] [Accepted: 12/13/2019] [Indexed: 12/31/2022]
Abstract
PURPOSE Natural killer (NK)-cell-based immunotherapy may overcome obstacles to effective T-cell-based immunotherapy such as the presence of genomic alterations in IFN response genes and antigen presentation machinery. All immunotherapy approaches may be abrogated by the presence of an immunosuppressive tumor microenvironment present in many solid tumor types, including head and neck squamous cell carcinoma (HNSCC). Here, we studied the role of myeloid-derived suppressor cells (MDSC) in suppressing NK-cell function in HNSCC. EXPERIMENTAL DESIGN The ability of peripheral and tumor-infiltrating MDSC from mice bearing murine oral cancer 2 (MOC2) non-T-cell-inflamed tumors and from patients with HNSCC to suppress NK-cell function was studied with real-time impedance and ELISpot assays. The therapeutic efficacy of SX-682, a small-molecule inhibitor of CXCR1 and CXCR2, was assessed in combination with adoptively transferred NK cells. RESULTS Mice bearing MOC2 tumors pathologically accumulate peripheral CXCR2+ neutrophilic-MDSC (PMN-MDSC) that traffic into tumors and suppress NK-cell function through TGFβ and production of H2O2. Inhibition of MDSC trafficking with orally bioavailable SX-682 significantly abrogated tumor MDSC accumulation and enhanced the tumor infiltration, activation, and therapeutic efficacy of adoptively transferred murine NK cells. Patients with HNSCC harbor significant levels of circulating and tumor-infiltrating CXCR1/2+ CD15+ PMN-MDSC and CD14+ monocytic-MDSC. Tumor MDSC exhibited greater immunosuppression than those in circulation. HNSCC tumor MDSC immunosuppression was mediated by multiple, independent, cell-specific mechanisms including TGFβ and nitric oxide. CONCLUSIONS The clinical study of CXCR1/2 inhibitors in combination with adoptively transferred NK cells is warranted.
Collapse
Affiliation(s)
- Sarah Greene
- Translational Tumor Immunology Program, National Institute on Deafness and Other Communication Disorders, Bethesda, Maryland
- NIH Medical Research Scholars Program, Bethesda, Maryland
| | - Yvette Robbins
- Translational Tumor Immunology Program, National Institute on Deafness and Other Communication Disorders, Bethesda, Maryland
| | - Wojciech K Mydlarz
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Angel P Huynh
- Translational Tumor Immunology Program, National Institute on Deafness and Other Communication Disorders, Bethesda, Maryland
| | - Nicole C Schmitt
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Jay Friedman
- Translational Tumor Immunology Program, National Institute on Deafness and Other Communication Disorders, Bethesda, Maryland
| | - Lucas A Horn
- Laboratory of Tumor Immunology and Biology, NCI, Bethesda, Maryland
| | - Claudia Palena
- Laboratory of Tumor Immunology and Biology, NCI, Bethesda, Maryland
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, NCI, Bethesda, Maryland
| | | | | | - Paul E Clavijo
- Translational Tumor Immunology Program, National Institute on Deafness and Other Communication Disorders, Bethesda, Maryland
| | - Clint Allen
- Translational Tumor Immunology Program, National Institute on Deafness and Other Communication Disorders, Bethesda, Maryland.
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
40
|
Greene S, Patel P, Allen CT. How patients with an intact immune system develop head and neck cancer. Oral Oncol 2019; 92:26-32. [PMID: 31010619 DOI: 10.1016/j.oraloncology.2019.03.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/28/2019] [Accepted: 03/13/2019] [Indexed: 12/18/2022]
Abstract
Although the adaptive immune system can detect and eliminate malignant cells, patients with intact and fully functional immune systems develop head and neck cancer. How is this paradox explained? Manuscripts published in the English language from 1975 to 2018 were reviewed using search inputs related to tumor cell antigenicity and immunogenicity, immunodominance, cancer immunoediting and genomic alterations present within carcinomas. Early in tumor development, T cell responses to immunodominant antigens may lead to the elimination of cancer cells expressing these antigens and a tumor composed to tumor cells expressing only immunorecessive antigens. Conversely, other tumor cells may acquire genomic or epigenetic alterations that result in an antigen processing or presentation defect or other inability to be detected or killed by T cells. Such T cell insensitive tumor cells may also be selected for in a progressing tumor. Tumors harboring subpopulations of cells that cannot be eliminated by T cells may require non-T cell-based treatments, such as NK cell immunotherapies. Recognition of such tumor cell populations within a heterogeneous cancer may inform the selection of treatment for HNSCC in the future.
Collapse
Affiliation(s)
- Sarah Greene
- Translational Tumor Immunology Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - Priya Patel
- Translational Tumor Immunology Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - Clint T Allen
- Translational Tumor Immunology Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
41
|
Shi CJ, Tian R, Wang M, Zhao Y, Qin RY. Effect of WEE1 inhibitor MK-1775 on self-renewal of gallbladder cancer stem cells. Shijie Huaren Xiaohua Zazhi 2018; 26:1586-1591. [DOI: 10.11569/wcjd.v26.i27.1586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM To assess the inhibitory effect of Wee1 inhibitor MK-1775 on self-renewal of cancer stem like cells in GBC-SD cells.
METHODS The GBC-SD cell line was cultured in vitro, and the suspended cancer stem cell spheres were cultured in serum free stem cell culture medium containing MK-1775. The expression of Wee1 was detected by Western blot, and the changes of the sphere size and sphere formation rate were analyzed. After the establishment of a subcutaneously transplanted tumor model in nude mice, MK-17751 was given for 2 wk, and the weight of the transplanted tumors was detected 2 wk later.
RESULTS After 8 d of culture with MK-1775, the expression of Wee1 in GBC-SD cells was downregulated and the sphere size and sphere formation rate were reduced. After MK-17751 treatment, the growth of subcutaneous xenografts in nude mice was inhibited.
CONCLUSION MK-1775 can inhibit the self-renewal of cancer stem like cell in gallbladder cancer GBC-SD cells.
Collapse
Affiliation(s)
- Cheng-Jian Shi
- Department of Pancreatobiliary Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Rui Tian
- Department of Pancreatobiliary Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Ming Wang
- Department of Pancreatobiliary Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Yan Zhao
- Department of Pancreatobiliary Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Ren-Yi Qin
- Department of Pancreatobiliary Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| |
Collapse
|
42
|
Fu S, Wang Y, Keyomarsi K, Meric-Bernstein F. Strategic development of AZD1775, a Wee1 kinase inhibitor, for cancer therapy. Expert Opin Investig Drugs 2018; 27:741-751. [DOI: 10.1080/13543784.2018.1511700] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Siqing Fu
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yudong Wang
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People’s Republic of China
| | - Khandan Keyomarsi
- Department of Experimental Radiation, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Funda Meric-Bernstein
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|