1
|
Sun S, Qian S, Wang R, Zhao M, Li R, Gu W, Zhao M, Qian C, Liu L, Tang X, Li Y, Shi H, Pan Y, Xiao H, Yang K, Hu C, Huang Y, Wei L, Zhang Y, Ji J, Chen Y, Liu H. Targeting GOLPH3L improves glioblastoma radiotherapy by regulating STING-NLRP3-mediated tumor immune microenvironment reprogramming. Sci Transl Med 2025; 17:eado0020. [PMID: 40043140 DOI: 10.1126/scitranslmed.ado0020] [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: 01/11/2024] [Revised: 08/04/2024] [Accepted: 02/12/2025] [Indexed: 04/18/2025]
Abstract
Radiotherapy (RT) has been the standard-of-care treatment for patients with glioblastoma (GBM); however, the clinical effectiveness is hindered by therapeutic resistance. Here, we demonstrated that the tumor immune microenvironment (TIME) exhibited immunosuppressive properties and high expression of Golgi phosphoprotein 3 like (GOLPH3L) in RT-resistant GBM. Our study showed that GOLPH3L interacted with stimulator of interferon genes (STING) at the aspartic acid residue 184 in Golgi after RT, leading to coat protein complex II-mediated retrograde transport of STING from Golgi to endoplasmic reticulum. This suppressed the STING-NOD-like receptor thermal protein domain associated protein 3 (NLRP3)-mediated pyroptosis, resulting in suppressive TIME, driving GBM resistance to RT. Genetic GOLPH3L ablation in RT-resistant GBM cells augmented antitumor immunity and overcame tumor resistance to RT. Moreover, we have identified a small molecular inhibitor of GOLPH3L, vitamin B5 calcium (VB5), which improved the therapeutic efficacy of RT and immune checkpoint blockade by inducing a robust antitumor immune response in mouse models. Clinically, patients with GBM treated with VB5 exhibited improved responses to RT. Thus, reprogramming the TIME by targeting GOLPH3L may offer a potential opportunity to improve RT in GBM.
Collapse
Affiliation(s)
- Shuo Sun
- Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Shiyu Qian
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Ran Wang
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Mengya Zhao
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
- Research Center of Surgery, Nanjing BenQ Medical Center, Affiliated BenQ Hospital of Nanjing Medical University, Department of Immunology, Nanjing Medical University, Nanjing 211166, China
| | - Ran Li
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Wei Gu
- Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Mengjie Zhao
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chunfa Qian
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Liang Liu
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xianglong Tang
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yangyang Li
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Hui Shi
- Department of Neurosurgery, First Hospital of Lianyungang, Lianyungang 222000, China
| | - Yunsong Pan
- Department of Neurosurgery, First Hospital of Lianyungang, Lianyungang 222000, China
| | - Hong Xiao
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Kun Yang
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chupeng Hu
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Yedi Huang
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Liangnian Wei
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Yuhan Zhang
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Jing Ji
- Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Yun Chen
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Hongyi Liu
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
2
|
Grazini U, Markovets A, Ireland L, O'Neill D, Phillips B, Xu M, Pfeifer M, Vaclova T, Martin MJ, Bigot L, Friboulet L, Hartmaier R, Cuomo ME, Barry ST, Smith PD, Floc'h N. Overcoming Osimertinib Resistance with AKT Inhibition in EGFRm-Driven Non-Small Cell Lung Cancer with PIK3CA/PTEN Alterations. Clin Cancer Res 2024; 30:4143-4154. [PMID: 38630555 DOI: 10.1158/1078-0432.ccr-23-2540] [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: 08/23/2023] [Revised: 12/31/2023] [Accepted: 04/05/2024] [Indexed: 04/19/2024]
Abstract
PURPOSE Osimertinib is an EGFR tyrosine kinase inhibitor indicated for the treatment of EGFR-mutated (EGFRm)-driven lung adenocarcinomas. Osimertinib significantly improves progression-free survival in first-line-treated patients with EGFRm advanced non-small cell lung cancer (NSCLC). Despite the durable disease control, the majority of patients receiving osimertinib eventually develop disease progression. EXPERIMENTAL DESIGN ctDNA profiling analysis of on-progression plasma samples from patients treated with osimertinib in both first- (phase III, FLAURA trial) and second-line trials (phase III, AURA3 trial) revealed a high prevalence of PIK3CA/AKT/PTEN alterations. In vitro and in vivo evidence using CRISPR-engineered NSCLC cell lines and patient-derived xenograft (PDX) models supports a functional role for PIK3CA and PTEN mutations in the development of osimertinib resistance. RESULTS These alterations are functionally relevant as EGFRm NSCLC cells with engineered PIK3CA/AKT/PTEN alterations develop resistance to osimertinib and can be resensitized by treatment with the combination of osimertinib and the AKT inhibitor capivasertib. Moreover, xenograft and PDX in vivo models with PIK3CA/AKT/PTEN alterations display limited sensitivity to osimertinib relative to models without alterations, and in these double-mutant models, capivasertib and osimertinib combination elicits an improved antitumor effect versus osimertinib alone. CONCLUSIONS Together, this approach offers a potential treatment strategy for patients with EGFRm-driven NSCLC who have a suboptimal response or develop resistance to osimertinib through PIK3CA/AKT/PTEN alterations. See related commentary by Vokes et al., p. 3968.
Collapse
Affiliation(s)
- Ursula Grazini
- Bioscience, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | | | - Lucy Ireland
- Bioscience, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Daniel O'Neill
- Bioscience, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Benjamin Phillips
- Data Sciences and Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, United Kingdom
| | - Man Xu
- Bioscience, Oncology R&D, AstraZeneca, Boston, Massachusetts
| | - Matthias Pfeifer
- Leibniz-Institute of Virology, Universität Sklinikum Hamburg-Eppendorf (UKE) Hamburg, Germany
| | - Tereza Vaclova
- Translational Medicine, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Matthew J Martin
- Bioscience, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Ludovic Bigot
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France
| | - Luc Friboulet
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France
| | - Ryan Hartmaier
- Translational Medicine, Oncology R&D, AstraZeneca, Boston, Massachusetts
| | - Maria E Cuomo
- Bioscience, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Simon T Barry
- Bioscience, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Paul D Smith
- Bioscience, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Nicolas Floc'h
- Bioscience, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| |
Collapse
|
3
|
Wang L, Dou X, Chen S, Yu X, Huang X, Zhang L, Chen Y, Wang J, Yang K, Bugno J, Pitroda S, Ding X, Piffko A, Si W, Chen C, Jiang H, Zhou B, Chmura SJ, Luo C, Liang HL, He C, Weichselbaum RR. YTHDF2 inhibition potentiates radiotherapy antitumor efficacy. Cancer Cell 2023; 41:1294-1308.e8. [PMID: 37236197 PMCID: PMC10524856 DOI: 10.1016/j.ccell.2023.04.019] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/23/2022] [Accepted: 04/28/2023] [Indexed: 05/28/2023]
Abstract
RNA N6-methyladenosine (m6A) modification is implicated in cancer progression. However, the impact of m6A on the antitumor effects of radiotherapy and the related mechanisms are unknown. Here we show that ionizing radiation (IR) induces immunosuppressive myeloid-derived suppressor cell (MDSC) expansion and YTHDF2 expression in both murine models and humans. Following IR, loss of Ythdf2 in myeloid cells augments antitumor immunity and overcomes tumor radioresistance by altering MDSC differentiation and inhibiting MDSC infiltration and suppressive function. The remodeling of the landscape of MDSC populations by local IR is reversed by Ythdf2 deficiency. IR-induced YTHDF2 expression relies on NF-κB signaling; YTHDF2 in turn leads to NF-κB activation by directly binding and degrading transcripts encoding negative regulators of NF-κB signaling, resulting in an IR-YTHDF2-NF-κB circuit. Pharmacological inhibition of YTHDF2 overcomes MDSC-induced immunosuppression and improves combined IR and/or anti-PD-L1 treatment. Thus, YTHDF2 is a promising target to improve radiotherapy (RT) and RT/immunotherapy combinations.
Collapse
Affiliation(s)
- Liangliang Wang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA
| | - Xiaoyang Dou
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, University of Chicago, Chicago, IL 60637, USA
| | - Shijie Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xianbin Yu
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, University of Chicago, Chicago, IL 60637, USA
| | - Xiaona Huang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA
| | - Linda Zhang
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, University of Chicago, Chicago, IL 60637, USA
| | - Yantao Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jiaai Wang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA
| | - Kaiting Yang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA
| | - Jason Bugno
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA; The Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, IL 600637, USA
| | - Sean Pitroda
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA
| | - Xingchen Ding
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Andras Piffko
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA; Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Wei Si
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Chao Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hualiang Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Bing Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Steven J Chmura
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA
| | - Cheng Luo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528437, China.
| | - Hua Laura Liang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA.
| | - Chuan He
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, University of Chicago, Chicago, IL 60637, USA; Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA.
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
4
|
Han D, Zhang N, Zhao S, Liu H, Wang X, Yang M, Wang S, Li Y, Liu Z, Teng L. AKIP1 promotes glioblastoma viability, mobility and chemoradiation resistance via regulating CXCL1 and CXCL8 mediated NF-κB and AKT pathways. Am J Cancer Res 2021; 11:1185-1205. [PMID: 33948353 PMCID: PMC8085855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 01/19/2021] [Indexed: 06/12/2023] Open
Abstract
This study aimed to investigate the interaction of A-kinase-interacting protein 1 (AKIP1) with C-X-C motif chemokine ligand (CXCL)1, CXCL2, CXCL8, and their effects on regulating glioblastoma multiforme (GBM) malignant behaviors. AKIP1 expression was modified by pcDNA and pGPH1 vectors in U-87 MG and U-251 MG cells. Subsequently, multiple compensative experiments were conducted via adding CXCL1, CXCL2 and CXCL8 in the pGPH1-AKIP1 (AKIP1 knockdown) transfected U-87 MG and U-251 MG cells, respectively. Furthermore, AKIP1, CXCL1/2/8 expressions in 10 GBM and 10 low-grade glioma (LGG) tumor samples were detected. AKIP1 was elevated in various GBM cell lines compared to normal human astrocytes. AKIP1 overexpression promoted U-87 MG and U-251 MG cell proliferation and invasion while inhibited apoptosis; and it enhanced chemoresistance to temozolomide (but not cisplatin) and radiation resistance; then AKIP1 knockdown showed the opposite effects. Meanwhile, AKIP1 positively regulated CXCL1/2/8, NF-κB pathway, AKT pathway and PD-L1 expression. Further multiple compensative experiments uncovered that CXCL1 and CXCL8 promoted proliferation, invasion, chemoradiation resistance, NF-κB pathway, AKT pathway and PD-L1 expression in U-87 MG and U-251 MG cells, also in pGPH1-AKIP1 (AKIP1 knockdown) transfected U-87 MG and U-251 MG cells; although CXCL2 exhibited similar treads, but its effect was much weaker. Besides, NF-κB pathway inhibitor and AKT pathway inhibitor attenuated the effect of CXCL1&CXCL8 on promoting GBM cell malignant behaviors. Clinically AKIP1 and CXCL1/8 were elevated in GBM compared to LGG tumor samples, and they were inter-correlated. AKIP1 promotes GBM viability, mobility and chemoradiation resistance via regulating CXCL1 and CXCL8 mediated NF-κB and AKT pathways.
Collapse
Affiliation(s)
- Dayong Han
- Department of Neurosurgery, The First Clinical College of Harbin Medical UniversityNangang District, Harbin 150001, China
| | - Na Zhang
- Department of Laboratory Diagnostics, The First Clinical College of Harbin Medical UniversityNangang District, Harbin 150001, China
| | - Shu Zhao
- Department of Emergency Internal Medicine, The First Clinical College of Harbin Medical UniversityNangang District, Harbin 150001, China
| | - Huailei Liu
- Department of Neurosurgery, The First Clinical College of Harbin Medical UniversityNangang District, Harbin 150001, China
| | - Xiaoxiong Wang
- Department of Neurosurgery, The First Clinical College of Harbin Medical UniversityNangang District, Harbin 150001, China
| | - Mingchun Yang
- Department of Neurosurgery, The First Clinical College of Harbin Medical UniversityNangang District, Harbin 150001, China
| | - Shengtao Wang
- Department of Neurosurgery, The First Clinical College of Harbin Medical UniversityNangang District, Harbin 150001, China
| | - Yue Li
- Department of Neurosurgery, The First Clinical College of Harbin Medical UniversityNangang District, Harbin 150001, China
| | - Zhanwen Liu
- Department of Neurosurgery, The First Clinical College of Harbin Medical UniversityNangang District, Harbin 150001, China
| | - Lei Teng
- Department of Neurosurgery, The First Clinical College of Harbin Medical UniversityNangang District, Harbin 150001, China
| |
Collapse
|
5
|
Mroweh M, Roth G, Decaens T, Marche PN, Lerat H, Macek Jílková Z. Targeting Akt in Hepatocellular Carcinoma and Its Tumor Microenvironment. Int J Mol Sci 2021; 22:1794. [PMID: 33670268 PMCID: PMC7917860 DOI: 10.3390/ijms22041794] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/07/2021] [Accepted: 02/09/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common causes of cancer-related deaths worldwide, and its incidence is rising. HCC develops almost exclusively on the background of chronic liver inflammation, which can be caused by chronic alcohol consumption, viral hepatitis, or an unhealthy diet. The key role of chronic inflammation in the process of hepatocarcinogenesis, including in the deregulation of innate and adaptive immune responses, has been demonstrated. The inhibition of Akt (also known as Protein Kinase B) directly affects cancer cells, but this therapeutic strategy also exhibits indirect anti-tumor activity mediated by the modulation of the tumor microenvironment, as demonstrated by using Akt inhibitors AZD5363, MK-2206, or ARQ 092. Moreover, the isoforms of Akt converge and diverge in their designated roles, but the currently available Akt inhibitors fail to display an isoform specificity. Thus, selective Akt inhibition needs to be better explored in the context of HCC and its possible combination with immunotherapy. This review presents a compact overview of the current knowledge concerning the role of Akt in HCC and the effect of Akt inhibition on the HCC and liver tumor microenvironment.
Collapse
Affiliation(s)
- Mariam Mroweh
- Institute for Advanced Biosciences, Research Center Inserm U 1209/CNRS 5309, 38700 La Tronche, France; (M.M.); (G.R.); (T.D.); (P.N.M.); (H.L.)
- Université Grenoble-Alpes, 38000 Grenoble, France
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath Beirut 6573-14, Lebanon
| | - Gaël Roth
- Institute for Advanced Biosciences, Research Center Inserm U 1209/CNRS 5309, 38700 La Tronche, France; (M.M.); (G.R.); (T.D.); (P.N.M.); (H.L.)
- Université Grenoble-Alpes, 38000 Grenoble, France
- Service D’hépato-Gastroentérologie, Pôle Digidune, CHU Grenoble Alpes, 38700 La Tronche, France
| | - Thomas Decaens
- Institute for Advanced Biosciences, Research Center Inserm U 1209/CNRS 5309, 38700 La Tronche, France; (M.M.); (G.R.); (T.D.); (P.N.M.); (H.L.)
- Université Grenoble-Alpes, 38000 Grenoble, France
- Service D’hépato-Gastroentérologie, Pôle Digidune, CHU Grenoble Alpes, 38700 La Tronche, France
| | - Patrice N. Marche
- Institute for Advanced Biosciences, Research Center Inserm U 1209/CNRS 5309, 38700 La Tronche, France; (M.M.); (G.R.); (T.D.); (P.N.M.); (H.L.)
- Université Grenoble-Alpes, 38000 Grenoble, France
| | - Hervé Lerat
- Institute for Advanced Biosciences, Research Center Inserm U 1209/CNRS 5309, 38700 La Tronche, France; (M.M.); (G.R.); (T.D.); (P.N.M.); (H.L.)
- Université Grenoble-Alpes, 38000 Grenoble, France
| | - Zuzana Macek Jílková
- Institute for Advanced Biosciences, Research Center Inserm U 1209/CNRS 5309, 38700 La Tronche, France; (M.M.); (G.R.); (T.D.); (P.N.M.); (H.L.)
- Université Grenoble-Alpes, 38000 Grenoble, France
- Service D’hépato-Gastroentérologie, Pôle Digidune, CHU Grenoble Alpes, 38700 La Tronche, France
| |
Collapse
|
6
|
Guo ZF, Kong FL. Akt regulates RSK2 to alter phosphorylation level of H2A.X in breast cancer. Oncol Lett 2021; 21:187. [PMID: 33574926 PMCID: PMC7816342 DOI: 10.3892/ol.2021.12448] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 08/13/2020] [Indexed: 02/06/2023] Open
Abstract
Histone H2AX (H2A.X) is a variant of the histone H2A family. Phosphorylation of H2A.X is a marker of DNA strand breaks and the presence or absence of H2A.X is closely related to tumor susceptibility and drug resistance. The present study found that the activity of the serine/threonine kinase Akt was negatively associated with H2A.X phosphorylated at the Ser16 site (H2A.X S16ph), but the mechanism of the inverse relationship remains elusive. The aim of the present study was to elucidate the mechanism of action between Akt and H2A.X S16ph and the exact role of this mechanism. Western blot analysis was performed to detect the regulatory association between p-Akt and H2A.X S16ph/p-RSK2, and immunoprecipitation and chromatin immunoprecipitation were performed to prove that Akt, RSK2 and H2A.X combine and interact in human breast cancer cells. The changes of cellular proliferation and migration induced by the interaction of Akt, RSK2 and H2A.X was determined by MTT, soft agar colony formation and cell migration experiments. The effect of interaction of Akt, RSK2 and H2A.X on cancer-promoting genes, such as PSAT-1 was determined via reverse transcription-quantitative PCR analysis. The current study indicated that the serine/threonine kinase ribosomal S6 kinase 2 (RSK2) as a kinase of H2A.X could be phosphorylated by Akt at Ser19 site. Moreover, Akt positively regulated the phosphorylation of RSK2 to inhibit phosphorylation of H2A.X, thereby affecting the affinity between RSK2 and substrate histone, promoting the survival and migration of breast cancer cells. In conclusion, Akt-mediated phosphorylation of RSK2 regulated the phosphorylation of H2A.X, thereby promoting oncogenic activity. This finding provides new insights to understand the pathogenesis and treatment mechanisms of breast cancer.
Collapse
Affiliation(s)
- Zhi-Feng Guo
- Department of Oncology, Section II, Chifeng Municipal Hospital, Chifeng, Inner Mongolia Autonomous Region 024000, P.R. China
| | - Fan-Long Kong
- Department of Oncology, Section II, Chifeng Municipal Hospital, Chifeng, Inner Mongolia Autonomous Region 024000, P.R. China
| |
Collapse
|
7
|
Qin S, Jiang J, Lu Y, Nice EC, Huang C, Zhang J, He W. Emerging role of tumor cell plasticity in modifying therapeutic response. Signal Transduct Target Ther 2020; 5:228. [PMID: 33028808 PMCID: PMC7541492 DOI: 10.1038/s41392-020-00313-5] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/25/2020] [Accepted: 08/30/2020] [Indexed: 02/07/2023] Open
Abstract
Resistance to cancer therapy is a major barrier to cancer management. Conventional views have proposed that acquisition of resistance may result from genetic mutations. However, accumulating evidence implicates a key role of non-mutational resistance mechanisms underlying drug tolerance, the latter of which is the focus that will be discussed here. Such non-mutational processes are largely driven by tumor cell plasticity, which renders tumor cells insusceptible to the drug-targeted pathway, thereby facilitating the tumor cell survival and growth. The concept of tumor cell plasticity highlights the significance of re-activation of developmental programs that are closely correlated with epithelial-mesenchymal transition, acquisition properties of cancer stem cells, and trans-differentiation potential during drug exposure. From observations in various cancers, this concept provides an opportunity for investigating the nature of anticancer drug resistance. Over the years, our understanding of the emerging role of phenotype switching in modifying therapeutic response has considerably increased. This expanded knowledge of tumor cell plasticity contributes to developing novel therapeutic strategies or combination therapy regimens using available anticancer drugs, which are likely to improve patient outcomes in clinical practice.
Collapse
Affiliation(s)
- Siyuan Qin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, People's Republic of China
| | - Jingwen Jiang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, People's Republic of China
| | - Yi Lu
- School of Medicine, Southern University of Science and Technology Shenzhen, Shenzhen, Guangdong, 518055, People's Republic of China
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen, Guangdong, People's Republic of China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, People's Republic of China.
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Road, 611137, Chengdu, People's Republic of China.
| | - Jian Zhang
- School of Medicine, Southern University of Science and Technology Shenzhen, Shenzhen, Guangdong, 518055, People's Republic of China.
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen, Guangdong, People's Republic of China.
| | - Weifeng He
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China.
- Chongqing Key Laboratory for Disease Proteomics, Chongqing, People's Republic of China.
| |
Collapse
|
8
|
Circumventing AKT-Associated Radioresistance in Oral Cancer by Novel Nanoparticle-Encapsulated Capivasertib. Cells 2020; 9:cells9030533. [PMID: 32106632 PMCID: PMC7140405 DOI: 10.3390/cells9030533] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 02/11/2020] [Accepted: 02/24/2020] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Development of radioresistance in oral squamous cell carcinoma (OSCC) remains a significant problem in cancer treatment, contributing to the lack of improvement in survival trends in recent decades. Effective strategies to overcome radioresistance are necessary to improve the therapeutic outcomes of radiotherapy in OSCC patients. METHODS Cells and xenograft tumors were irradiated using the Small Animal Radiation Research Platform. AKT inhibitor capivasertib (AZD5363) was encapsulated into cathepsin B-responsible nanoparticles (NPs) for tumor-specific delivery. Cell viability was measured by alamarBlue, cell growth was determined by colony formation and 3D culture, and apoptosis was assessed by flow cytometry with the staining of Fluorescein isothiocyanate (FITC) Annexin V and PI. An orthotopic tongue tumor model was used to evaluate the in vivo therapeutic effects. The molecular changes induced by the treatments were assessed by Western blotting and immunohistochemistry. RESULTS We show that upregulation of AKT signaling is the critical mechanism for radioresistance in OSCC cells, and AKT inactivation by a selective and potent AKT inhibitor capivasertib results in radiosensitivity. Moreover, relative to irradiation (IR) alone, IR combined with the delivery of capivasertib in association with tumor-seeking NPs greatly enhanced tumor cell repression in 3D cell cultures and OSCC tumor shrinkage in an orthotopic mouse model. CONCLUSIONS These data indicate that capivasertib is a potent agent that sensitizes radioresistant OSCC cells to IR and is a promising strategy to overcome failure of radiotherapy in OSCC patients.
Collapse
|
9
|
Wang P, Yang Z, Ye T, Shao F, Li J, Sun N, He J. lncTUG1/miR-144-3p affect the radiosensitivity of esophageal squamous cell carcinoma by competitively regulating c-MET. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:7. [PMID: 31918742 PMCID: PMC6953235 DOI: 10.1186/s13046-019-1519-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 12/30/2019] [Indexed: 02/07/2023]
Abstract
Background Long noncoding RNAs (lncRNAs) are involved in the progression of various cancers and affect the response to radiotherapy. This study focused on clarifying the underlying mechanism by which lncTUG1 affects the radiosensitivity of esophageal squamous cell carcinoma (ESCC). Methods lncTUG1, miR-144-3p and MET expression levels were detected in ESCC tissues and cells by qRT-PCR. Western blotting was used to examine the protein levels of MET, p-AKT and EGFR. The dual-luciferase reporter system and RNA immunoprecipitation (RIP) assays were used to confirm the interaction between lncTUG1 and miR-144-3p or miR-144-3p and MET. MTT, colony formation and flow cytometry assays were applied to examine the behavioral changes in EC9706 and KYSE30 cells. Results lncTUG1 was upregulated in ESCC cells and tissues, and lncTUG1 expression was associated with an advanced pathological stage. The bioinformatics analysis revealed that lncTUG1 could specifically bind to miR-144-3p, which was downregulated in ESCC. There was a negative correlation between lncTUG1 and miR-144-3p. LncTUG1 inhibition retarded proliferation and colony formation and induced apoptosis in ESCC cells. Moreover, lncTUG1 knockdown dramatically improved the effect of radiotherapy on ESCC development both in vivo and in vitro. Furthermore, MET was revealed as a downstream target of miR-144-3p and is downregulated by it. LncTUG1 promoted the progression of ESCC and elevated radiotherapy resistance in ESCC cells, accompanied by a high level of MET expression. Moreover, we found that knockdown of lncTUG1 enhanced the radiosensitivity of ESCC cells via the p-AKT signaling pathway. Conclusion Our results indicate that lncTUG1 enhances the radiotherapy resistance of ESCC by lowering the miR-144-3p level and modulating the MET/EGFR/AKT axis.
Collapse
Affiliation(s)
- Pan Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17 Panjiayuannanli, Beijing, 100021, China
| | - Zhuanbo Yang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ting Ye
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, 266071, Shandong, China.,Qingdao Cancer Institute, Qingdao, 266071, Shandong, China
| | - Fei Shao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17 Panjiayuannanli, Beijing, 100021, China.,Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, 266071, Shandong, China.,Qingdao Cancer Institute, Qingdao, 266071, Shandong, China
| | - Jiagen Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17 Panjiayuannanli, Beijing, 100021, China
| | - Nan Sun
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17 Panjiayuannanli, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17 Panjiayuannanli, Beijing, 100021, China.
| |
Collapse
|
10
|
Lang L, Shay C, Zhao X, Xiong Y, Wang X, Teng Y. Simultaneously inactivating Src and AKT by saracatinib/capivasertib co-delivery nanoparticles to improve the efficacy of anti-Src therapy in head and neck squamous cell carcinoma. J Hematol Oncol 2019; 12:132. [PMID: 31805962 PMCID: PMC6896687 DOI: 10.1186/s13045-019-0827-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 11/13/2019] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Src, an oncoprotein that drives progression of head and neck squamous cell carcinoma (HNSCC), is commonly hyperactivated in this disease. Unfortunately, the clinical benefit of targeting Src is significantly dampened in HNSCC patients, because the cytotoxic effects of anti-Src therapy and tumor resistance to it are less predictable. Thus, understanding the mechanism of tumor resistance to Src inhibition and seeking a way to overcome it are warranted. METHODS Dual drug-loaded nanoparticles (NPs) were developed to co-deliver Src inhibitor saracatinib (AZD0530) and AKT inhibitor capivasertib (AZD5363) into the same population of tumor cells. An orthotopic tongue tumor model was generated to evaluate the in vivo therapeutic effects. Cell growth was determined by CellTiter-Glo® Luminescent Cell Viability Kit, colony formation, and 3D culture, and tumor growth was determined by bioluminescence and tumor size. The molecular changes induced by the treatments were assessed by Western blotting and immunohistochemistry. RESULTS Capivasertib inactivated the AKT-S6 signaling and re-sensitized saracatinib-resistant HNSCC cells to saracatinib. Combination of capivasertib with saracatinib suppressed HNSCC growth more efficiently than either drug alone. Cathepsin B-sensitive NPs for co-delivering saracatinib and capivasertib significantly improved the efficacy of tumor repression without increasing side effects, which were due to highly specific tumor-targeting drug delivery system and synergistic anticancer effects by co-inactivation of AKT and Src in HNSCC cells. CONCLUSIONS Addition of AKT blockade improves anti-HNSCC efficacy of anti-Src therapy, and co-delivery of capivasertib and saracatinib by tumor-targeting NPs has the potential to achieve better treatment outcomes than the free drug combination.
Collapse
Affiliation(s)
- Liwei Lang
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Chloe Shay
- Department of Pediatrics, Emory Children's Center, Emory University, Atlanta, GA, USA
| | - Xiangdong Zhao
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Yuanping Xiong
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Xuli Wang
- Department of Radiology and Imaging Sciences, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Yong Teng
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA. .,Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA. .,Department of Medical Laboratory, Imaging and Radiologic Sciences, College of Allied Health, Augusta University, Augusta, GA, USA.
| |
Collapse
|
11
|
Au JLS, Lu Z, Abbiati RA, Wientjes MG. Systemic Bioequivalence Is Unlikely to Equal Target Site Bioequivalence for Nanotechnology Oncologic Products. AAPS J 2019; 21:24. [PMID: 30710324 PMCID: PMC6432930 DOI: 10.1208/s12248-019-0296-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 01/09/2019] [Indexed: 11/30/2022] Open
Abstract
Approval of generic drugs by the US Food and Drug Administration (FDA) requires the product to be pharmaceutically equivalent to the reference listed drug (RLD) and demonstrate bioequivalence (BE) in effectiveness when administered to patients under the conditions in the RLD product labeling. Effectiveness is determined by drug exposure at the target sites. However, since such measurement is usually unavailable, systemic exposure is assumed to equal target site exposure and systemic BE to equal target site BE. This assumption, while it often applies to small molecule drug products that are readily dissolved in biological fluids and systemically absorbed, is unlikely to apply to nanotechnology products (NP) that exist as heterogeneous systems and are subjected to dimension- and material-dependent changes. This commentary provides an overview of the intersecting and spatial-dependent processes and variables governing the delivery and residence of oncologic NP in solid tumors. In order to provide a quantitative perspective of the collective effects of these processes, we used quantitative systems pharmacology (QSP) multi-scale modeling to capture the physicochemical and biological events on several scales (whole-body, organ/suborgan, cell/subcellular, spatial locations, time). QSP is an emerging field that entails using modeling and computation to facilitate drug development; an analogous approach (i.e., model-informed drug development) is advocated by to FDA. The QSP model-based simulations illustrated that small changes in NP attributes (e.g., size variations during manufacturing, interactions with proteins in biological milieu) could lead to disproportionately large differences in target site exposure, rending systemic BE unlikely to equal target site BE.
Collapse
Affiliation(s)
- Jessie L-S Au
- Institute of Quantitative Systems Pharmacology, 1815 Aston Avenue, suite 107, Carlsbad, California, 92008, USA.
- Optimum Therapeutics LLC, Carlsbad, California, 92008, USA.
- Department of Pharmaceutical Sciences, University of Oklahoma, Oklahoma City, Oklahoma, 73117, USA.
- College of Pharmacy, Taipei Medical University, Taipei, Taiwan, Republic of China.
| | - Ze Lu
- Institute of Quantitative Systems Pharmacology, 1815 Aston Avenue, suite 107, Carlsbad, California, 92008, USA
- Optimum Therapeutics LLC, Carlsbad, California, 92008, USA
| | - Roberto A Abbiati
- Institute of Quantitative Systems Pharmacology, 1815 Aston Avenue, suite 107, Carlsbad, California, 92008, USA
- Department of Pharmaceutical Sciences, University of Oklahoma, Oklahoma City, Oklahoma, 73117, USA
| | - M Guillaume Wientjes
- Institute of Quantitative Systems Pharmacology, 1815 Aston Avenue, suite 107, Carlsbad, California, 92008, USA
- Optimum Therapeutics LLC, Carlsbad, California, 92008, USA
| |
Collapse
|
12
|
Ion L, Petre BA. Immuno-Affinity Mass Spectrometry: A Novel Approaches with Biomedical Relevance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1140:377-388. [DOI: 10.1007/978-3-030-15950-4_21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
13
|
Yang X, Geng KY, Zhang YS, Zhang JF, Yang K, Shao JX, Xia WL. Sirt3 deficiency impairs neurovascular recovery in ischemic stroke. CNS Neurosci Ther 2018; 24:775-783. [PMID: 29777578 DOI: 10.1111/cns.12853] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 03/06/2018] [Accepted: 03/10/2018] [Indexed: 12/19/2022] Open
Abstract
AIMS Sirt3 is one member of the NAD+ -dependent protein deacetylase family and plays crucial roles in diverse aspects of mammalian biological function. Then the role of Sirt3 on ischemia stroke is unknown. METHODS To examine the effect of Sirt3 on ischemic stroke, we performed transient middle cerebral artery occlusion (tMCAO) in adult male Sirt3 knockout (KO) and wild-type (WT) mice. RESULTS The level of Sirt3 in infarct region is decreased after ischemic stroke. In addition, we found that Sirt3 KO mice showed worse neurobehavioral outcome compared with WT mice, accompanied by decreased neurogenesis and angiogenesis as shown by the reduction in number of DCX+ /BrdU+ cells, NeuN+ /BrdU+ cells, and CD31+ /BrdU+ cells in the perifocal region during recovery phase after ischemic stroke. Furthermore, Sirt3 deficiency reduced the activation of vascular endothelial growth factor (VEGF), AKT, and extracellular signal-regulated kinases (ERK) signaling pathways. CONCLUSION Our results indicated that Sirt3 is beneficial to neurovascular and functional recovery following chronic ischemic stroke.
Collapse
Affiliation(s)
- Xiao Yang
- State Key Laboratory of Oncogenes and Related Genes, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ke-Yi Geng
- State Key Laboratory of Oncogenes and Related Genes, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yan-Shuang Zhang
- State Key Laboratory of Oncogenes and Related Genes, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jin-Fan Zhang
- State Key Laboratory of Oncogenes and Related Genes, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ke Yang
- State Key Laboratory of Oncogenes and Related Genes, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jia-Xiang Shao
- State Key Laboratory of Oncogenes and Related Genes, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Wei-Liang Xia
- State Key Laboratory of Oncogenes and Related Genes, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
14
|
Searle EJ, Telfer BA, Mukherjee D, Forster DM, Davies BR, Williams KJ, Stratford IJ, Illidge TM. Akt inhibition improves long-term tumour control following radiotherapy by altering the microenvironment. EMBO Mol Med 2017; 9:1646-1659. [PMID: 29084756 PMCID: PMC5709765 DOI: 10.15252/emmm.201707767] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 09/27/2017] [Accepted: 09/28/2017] [Indexed: 01/01/2023] Open
Abstract
Radiotherapy is an important anti-cancer treatment, but tumour recurrence remains a significant clinical problem. In an effort to improve outcomes further, targeted anti-cancer drugs are being tested in combination with radiotherapy. Here, we have studied the effects of Akt inhibition with AZD5363. AZD5363 administered as an adjuvant after radiotherapy to FaDu and PE/CA PJ34 tumours leads to long-term tumour control, which appears to be secondary to effects on the irradiated tumour microenvironment. AZD5363 reduces the downstream effectors VEGF and HIF-1α, but has no effect on tumour vascularity or oxygenation, or on tumour control, when administered prior to radiotherapy. In contrast, AZD5363 given after radiotherapy is associated with marked reductions in tumour vascular density, a decrease in the influx of CD11b+ myeloid cells and a failure of tumour regrowth. In addition, AZD5363 is shown to inhibit the proportion of proliferating tumour vascular endothelial cells in vivo, which may contribute to improved tumour control with adjuvant treatment. These new insights provide promise to improve outcomes with the addition of AZD5363 as an adjuvant therapy following radiotherapy.
Collapse
Affiliation(s)
- Emma J Searle
- Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Manchester, UK
- Division of Cancer Sciences, School of Medical Sciences, University of Manchester, Manchester, UK
- Christie Hospital Manchester Academic Health Sciences Centre University of Manchester, Manchester, UK
| | - Brian A Telfer
- Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Manchester, UK
| | - Debayan Mukherjee
- Division of Cancer Sciences, School of Medical Sciences, University of Manchester, Manchester, UK
- Christie Hospital Manchester Academic Health Sciences Centre University of Manchester, Manchester, UK
| | - Duncan M Forster
- Division of Informatics, Imaging & Data Sciences, School of Health Sciences, University of Manchester, Manchester, UK
| | | | - Kaye J Williams
- Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Manchester, UK
- CRUK-EPSRC Cancer Imaging Centre in Cambridge and Manchester, Cambridge, UK
- CRUK-EPSRC Cancer Imaging Centre in Cambridge and Manchester, Manchester, UK
| | - Ian J Stratford
- Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Manchester, UK
| | - Tim M Illidge
- Division of Cancer Sciences, School of Medical Sciences, University of Manchester, Manchester, UK
- Christie Hospital Manchester Academic Health Sciences Centre University of Manchester, Manchester, UK
| |
Collapse
|