1
|
Wang T, Shi X, Xu X, Zhang J, Ma Z, Meng C, Jiao D, Wang Y, Chen Y, He Z, Zhu Y, Liu HN, Zhang T, Jiang Q. Emerging prodrug and nano-drug delivery strategies for the detection and elimination of senescent tumor cells. Biomaterials 2025; 318:123129. [PMID: 39922127 DOI: 10.1016/j.biomaterials.2025.123129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/15/2025] [Accepted: 01/23/2025] [Indexed: 02/10/2025]
Abstract
Tumor cellular senescence, characterized by reversible cell cycle arrest following anti-cancer therapies, presents a complex paradigm in oncology. Given that senescent tumor cells may promote angiogenesis, tumorigenesis, and metastasis, selective killing senescent cells (SCs)-a strategy termed senotherapy-has emerged as a promising approach to improve cancer treatment. However, the clinical implementation of senotherapy faces significant hurdles, including lack of precise methods for SCs identification and the potential for adverse effects associated with highly cytotoxic senolytic agents. In this account, we elucidate recent advancement in developing novel approaches for the detection and selective elimination of SCs, encompassing prodrugs, nanoparticles, and other cutting-edge drug delivery systems such as PROTAC technology and CAR T cell therapy. Furthermore, we explore the paradoxical nature of SCs, which can induce growth arrest in adjacent neoplastic cells and recruit immunomodulatory cells that contribute to tumor suppression. Therefore, we utilize SCs membrane as vehicles to elicit antitumor immunity and potentially augment existing anti-cancer therapies. Finally, the opportunities and challenges are put forward to facilitate the development and clinical transformation of SCs detection, elimination or utilization.
Collapse
Affiliation(s)
- Tao Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Xiaolan Xu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Jiaming Zhang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Zhengdi Ma
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Chen Meng
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Dian Jiao
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yubo Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yanfei Chen
- School of Hainan Provincial Drug Safety Evaluation Research Center, Hainan Medical University, Haikou, China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Ying Zhu
- Department of Neurology, The First Hospital of China Medical University, Shenyang, 110002, China.
| | - He-Nan Liu
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Tianhong Zhang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Qikun Jiang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China; Engineering Research Center of Tropical Medicine Innovation and Transformation of Ministry of Education, Hainan Medical University, Haikou, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China.
| |
Collapse
|
2
|
Wang Z, Chen Y, Fang H, Xiao K, Wu Z, Xie X, Liu J, Chen F, He Y, Wang L, Yang C, Pei R, Shao D. Reprogramming cellular senescence in the tumor microenvironment augments cancer immunotherapy through multifunctional nanocrystals. SCIENCE ADVANCES 2024; 10:eadp7022. [PMID: 39485841 PMCID: PMC11529718 DOI: 10.1126/sciadv.adp7022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 09/25/2024] [Indexed: 11/03/2024]
Abstract
Harnessing the immunogenic potential of senescent tumor cells provides an opportunity to remodel tumor microenvironment (TME) and boost antitumor immunity. However, this potential needs to be sophisticatedly wielded to avoid additional immunosuppressive capacity of senescent cells. Our study shows that blocking the JAK2/STAT3 pathway enhances immunogenic efficacy of Aurora kinase inhibitor alisertib (Ali)-induced senescence by reducing immunosuppressive senescence-associated secretory phenotype (SASP) while preserving immunogenic SASP. Hypothesizing that SASP reprogramming with Ali and JAK2 inhibitor ruxolitinib (Rux) will benefit cancer immunotherapy, we create nanoparticulate crystals (Ali-Rux) composed of Ali and Rux with a fully active pharmaceutical ingredient. Immunization with Ali-Rux-orchestrated senescent cells promotes stronger activation of antigen-presenting cells, enhancing antitumor immune surveillance. This approach remodels the TME by increasing CD8+ T cell and NK recruitment and activation while decreasing MDSCs. Combined with PD-L1 blockade, Ali-Rux elicits a durable antitumor immune response, suggesting the TME reshaping approach as a potential cancer immunotherapy.
Collapse
Affiliation(s)
- Zheng Wang
- Suzhou Institute of Nano-Tech and NanoBionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Yinglu Chen
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Hui Fang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Kai Xiao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Ziping Wu
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Xiaochun Xie
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Jie Liu
- Department of Orthopedics, Academy of Orthopedics-Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Fangman Chen
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Yi He
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - Liang Wang
- Department of Orthopedics, Academy of Orthopedics-Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Chao Yang
- Department of Orthopedics, Academy of Orthopedics-Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Renjun Pei
- Suzhou Institute of Nano-Tech and NanoBionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Dan Shao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, China
| |
Collapse
|
3
|
Tufail M, Huang YQ, Hu JJ, Liang J, He CY, Wan WD, Jiang CH, Wu H, Li N. Cellular Aging and Senescence in Cancer: A Holistic Review of Cellular Fate Determinants. Aging Dis 2024:AD.2024.0421. [PMID: 38913050 DOI: 10.14336/ad.2024.0421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 05/21/2024] [Indexed: 06/25/2024] Open
Abstract
This comprehensive review navigates the complex relationship between cellular aging, senescence, and cancer, unraveling the determinants of cellular fate. Beginning with an overview of cellular aging's significance in cancer, the review explores processes, changes, and molecular pathways influencing senescence. The review explores senescence as a dual mechanism in cancer, acting as a suppressor and contributor, focusing on its impact on therapy response. This review highlights opportunities for cancer therapies that target cellular senescence. The review further examines the senescence-associated secretory phenotype and strategies to modulate cellular aging to influence tumor behavior. Additionally, the review highlights the mechanisms of senescence escape in aging and cancer cells, emphasizing their impact on cancer prognosis and resistance to therapy. The article addresses current advances, unexplored aspects, and future perspectives in understanding cellular aging and senescence in cancer.
Collapse
Affiliation(s)
- Muhammad Tufail
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Yu-Qi Huang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Jia-Ju Hu
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Liang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Cai-Yun He
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Wen-Dong Wan
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Can-Hua Jiang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hong Wu
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, 410083, China
| | - Ning Li
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
4
|
Zhang Z, Chen L, Yang Q, Tang X, Li J, Zhang G, Wang Y, Huang H. INHBA regulates Hippo signaling to confer 5-FU chemoresistance mediated by cellular senescence in colon cancer cells. Int J Biochem Cell Biol 2024; 171:106570. [PMID: 38588888 DOI: 10.1016/j.biocel.2024.106570] [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: 09/18/2023] [Revised: 02/27/2024] [Accepted: 04/05/2024] [Indexed: 04/10/2024]
Abstract
Colon cancer has become a global public health challenge, and 5-Fluorouracil (5-FU) chemoresistance is a major obstacle in its treatment. Chemoresistance can be mediated by therapy-induced cellular senescence. This study intended to investigate mechanisms of INHBA (inhibin A) in 5-FU resistance mediated by cellular senescence in colon cancer. Bioinformatics analysis of INHBA expression in colon cancer tissues, survival analysis, and correlation analysis of cellular senescence markers were performed. The effects of INHBA on the biological characteristics and 5-FU resistance of colon cancer cells were examined through loss/gain-of-function and molecular assays. Finally, a xenograft mouse model was built to validate the mechanism of INHBA in vivo. INHBA was upregulated in colon cancer and was significantly positively correlated with cellular senescence markers uncoupling protein 2 (UCP-2), matrix metalloproteinase-1 (MMP-1), dense and erect panicle 1 (DEP1), and p21. Cellular senescence in colon cancer mediated 5-FU resistance. Downregulation of INHBA expression enhanced 5-FU sensitivity in colon cancer cells, inhibited cell proliferation, promoted apoptosis, increased the proportion of cells in G0/G1 phase, and it resulted in a lower proportion of senescent cells and lower levels of the cellular senescence markers interleukin 6 (IL-6) and interleukin 8 (IL-8). Analysis of whether to use the pathway inhibitor Verteporfin proved that INHBA facilitated colon cancer cell senescence and enhanced 5-FU chemoresistance via inactivation of Hippo signaling pathway, and consistent results were obtained in vivo. Collectively, INHBA conferred 5-FU chemoresistance mediated by cellular senescence in colon cancer cells through negative regulation of Hippo signaling.
Collapse
Affiliation(s)
- Zhan Zhang
- Department of Hematology and Oncology, Taizhou First People's Hospital, Taizhou 318020, China; Huangyan Hospital of Wenzhou Medical University, Taizhou 318020, China
| | - Lili Chen
- Department of Hematology and Oncology, Taizhou First People's Hospital, Taizhou 318020, China; Huangyan Hospital of Wenzhou Medical University, Taizhou 318020, China; Wenzhou Medical University, Wenzhou 325035, China.
| | - Qiao Yang
- Department of Hematology and Oncology, Taizhou First People's Hospital, Taizhou 318020, China
| | - Xiaowan Tang
- Department of Hematology and Oncology, Taizhou First People's Hospital, Taizhou 318020, China
| | - Jianhua Li
- Department of Hematology and Oncology, Taizhou First People's Hospital, Taizhou 318020, China
| | - Guangwen Zhang
- Department of Hematology and Oncology, Taizhou First People's Hospital, Taizhou 318020, China
| | - Youqun Wang
- Department of Hematology and Oncology, Taizhou First People's Hospital, Taizhou 318020, China
| | - Hui Huang
- Department of Hematology and Oncology, Taizhou First People's Hospital, Taizhou 318020, China
| |
Collapse
|
5
|
Tóth F, Moftakhar Z, Sotgia F, Lisanti MP. In Vitro Investigation of Therapy-Induced Senescence and Senescence Escape in Breast Cancer Cells Using Novel Flow Cytometry-Based Methods. Cells 2024; 13:841. [PMID: 38786063 PMCID: PMC11120107 DOI: 10.3390/cells13100841] [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: 04/12/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Although cellular senescence was originally defined as an irreversible form of cell cycle arrest, in therapy-induced senescence models, the emergence of proliferative senescence-escaped cancer cells has been reported by several groups, challenging the definition of senescence. Indeed, senescence-escaped cancer cells may contribute to resistance to cancer treatment. Here, to study senescence escape and isolate senescence-escaped cells, we developed novel flow cytometry-based methods using the proliferation marker Ki-67 and CellTrace CFSE live-staining. We investigated the role of a novel senescence marker (DPP4/CD26) and a senolytic drug (azithromycin) on the senescence-escaping ability of MCF-7 and MDA-MB-231 breast cancer cells. Our results show that the expression of DPP4/CD26 is significantly increased in both senescent MCF-7 and MDA-MB-231 cells. While not essential for senescence induction, DPP4/CD26 contributed to promoting senescence escape in MCF-7 cells but not in MDA-MB-231 cells. Our results also confirmed the potential senolytic effect of azithromycin in senescent cancer cells. Importantly, the combination of azithromycin and a DPP4 inhibitor (sitagliptin) demonstrated a synergistic effect in senescent MCF-7 cells and reduced the number of senescence-escaped cells. Although further research is needed, our results and novel methods could contribute to the investigation of the mechanisms of senescence escape and the identification of potential therapeutic targets. Indeed, DPP4/CD26 could be a promising marker and a novel target to potentially decrease senescence escape in cancer.
Collapse
Affiliation(s)
- Fanni Tóth
- Translational Medicine, University of Salford, Salford M5 4WT, UK; (F.T.)
- The CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, 1090 Wien, Vienna, Austria
| | - Zahra Moftakhar
- Translational Medicine, University of Salford, Salford M5 4WT, UK; (F.T.)
| | - Federica Sotgia
- Translational Medicine, University of Salford, Salford M5 4WT, UK; (F.T.)
| | - Michael P. Lisanti
- Translational Medicine, University of Salford, Salford M5 4WT, UK; (F.T.)
| |
Collapse
|
6
|
Pichol-Thievend C, Anezo O, Pettiwala AM, Bourmeau G, Montagne R, Lyne AM, Guichet PO, Deshors P, Ballestín A, Blanchard B, Reveilles J, Ravi VM, Joseph K, Heiland DH, Julien B, Leboucher S, Besse L, Legoix P, Dingli F, Liva S, Loew D, Giani E, Ribecco V, Furumaya C, Marcos-Kovandzic L, Masliantsev K, Daubon T, Wang L, Diaz AA, Schnell O, Beck J, Servant N, Karayan-Tapon L, Cavalli FMG, Seano G. VC-resist glioblastoma cell state: vessel co-option as a key driver of chemoradiation resistance. Nat Commun 2024; 15:3602. [PMID: 38684700 PMCID: PMC11058782 DOI: 10.1038/s41467-024-47985-z] [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: 11/10/2023] [Accepted: 04/17/2024] [Indexed: 05/02/2024] Open
Abstract
Glioblastoma (GBM) is a highly lethal type of cancer. GBM recurrence following chemoradiation is typically attributed to the regrowth of invasive and resistant cells. Therefore, there is a pressing need to gain a deeper understanding of the mechanisms underlying GBM resistance to chemoradiation and its ability to infiltrate. Using a combination of transcriptomic, proteomic, and phosphoproteomic analyses, longitudinal imaging, organotypic cultures, functional assays, animal studies, and clinical data analyses, we demonstrate that chemoradiation and brain vasculature induce cell transition to a functional state named VC-Resist (vessel co-opting and resistant cell state). This cell state is midway along the transcriptomic axis between proneural and mesenchymal GBM cells and is closer to the AC/MES1-like state. VC-Resist GBM cells are highly vessel co-opting, allowing significant infiltration into the surrounding brain tissue and homing to the perivascular niche, which in turn induces even more VC-Resist transition. The molecular and functional characteristics of this FGFR1-YAP1-dependent GBM cell state, including resistance to DNA damage, enrichment in the G2M phase, and induction of senescence/stemness pathways, contribute to its enhanced resistance to chemoradiation. These findings demonstrate how vessel co-option, perivascular niche, and GBM cell plasticity jointly drive resistance to therapy during GBM recurrence.
Collapse
Affiliation(s)
- Cathy Pichol-Thievend
- Institut Curie, INSERM U1021, CNRS UMR3347, Tumor Microenvironment Lab, Paris-Saclay University, 91400, Orsay, France
| | - Oceane Anezo
- Institut Curie, INSERM U1021, CNRS UMR3347, Tumor Microenvironment Lab, Paris-Saclay University, 91400, Orsay, France
| | - Aafrin M Pettiwala
- Institut Curie, INSERM U1021, CNRS UMR3347, Tumor Microenvironment Lab, Paris-Saclay University, 91400, Orsay, France
- Institut Curie, PSL University, 75005, Paris, France
| | - Guillaume Bourmeau
- Institut Curie, INSERM U1021, CNRS UMR3347, Tumor Microenvironment Lab, Paris-Saclay University, 91400, Orsay, France
| | - Remi Montagne
- Institut Curie, PSL University, 75005, Paris, France
- INSERM U900, 75005, Paris, France
- MINES ParisTeach, CBIO-Centre for Computational Biology, PSL Research University, 75006, Paris, France
| | - Anne-Marie Lyne
- Institut Curie, PSL University, 75005, Paris, France
- INSERM U900, 75005, Paris, France
- MINES ParisTeach, CBIO-Centre for Computational Biology, PSL Research University, 75006, Paris, France
| | - Pierre-Olivier Guichet
- Université de Poitiers, CHU Poitiers, ProDiCeT, F-86000, Poitiers, France
- CHU Poitiers, Laboratoire de Cancérologie Biologique, F-86000, Poitiers, France
| | - Pauline Deshors
- Institut Curie, INSERM U1021, CNRS UMR3347, Tumor Microenvironment Lab, Paris-Saclay University, 91400, Orsay, France
| | - Alberto Ballestín
- Institut Curie, INSERM U1021, CNRS UMR3347, Tumor Microenvironment Lab, Paris-Saclay University, 91400, Orsay, France
| | - Benjamin Blanchard
- Institut Curie, INSERM U1021, CNRS UMR3347, Tumor Microenvironment Lab, Paris-Saclay University, 91400, Orsay, France
| | - Juliette Reveilles
- Institut Curie, INSERM U1021, CNRS UMR3347, Tumor Microenvironment Lab, Paris-Saclay University, 91400, Orsay, France
| | - Vidhya M Ravi
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
| | - Kevin Joseph
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
| | - Dieter H Heiland
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
| | - Boris Julien
- Institut Curie, INSERM U1021, CNRS UMR3347, Tumor Microenvironment Lab, Paris-Saclay University, 91400, Orsay, France
| | | | - Laetitia Besse
- Institut Curie, PSL University, Université Paris-Saclay, CNRS UMS2016, INSERM US43, Multimodal Imaging Center, 91400, Orsay, France
| | - Patricia Legoix
- Institut Curie, PSL University, ICGex Next-Generation Sequencing Platform, 75005, Paris, France
| | - Florent Dingli
- Institut Curie, PSL University, CurieCoreTech Spectrométrie de Masse Protéomique, 75005, Paris, France
| | - Stephane Liva
- Institut Curie, PSL University, 75005, Paris, France
- INSERM U900, 75005, Paris, France
- MINES ParisTeach, CBIO-Centre for Computational Biology, PSL Research University, 75006, Paris, France
| | - Damarys Loew
- Institut Curie, PSL University, CurieCoreTech Spectrométrie de Masse Protéomique, 75005, Paris, France
| | - Elisa Giani
- Department of Biomedical Sciences, Humanitas University, 20072, Pieve Emanuele, Italy
| | - Valentino Ribecco
- Institut Curie, INSERM U1021, CNRS UMR3347, Tumor Microenvironment Lab, Paris-Saclay University, 91400, Orsay, France
| | - Charita Furumaya
- Institut Curie, INSERM U1021, CNRS UMR3347, Tumor Microenvironment Lab, Paris-Saclay University, 91400, Orsay, France
| | - Laura Marcos-Kovandzic
- Institut Curie, INSERM U1021, CNRS UMR3347, Tumor Microenvironment Lab, Paris-Saclay University, 91400, Orsay, France
| | - Konstantin Masliantsev
- Université de Poitiers, CHU Poitiers, ProDiCeT, F-86000, Poitiers, France
- CHU Poitiers, Laboratoire de Cancérologie Biologique, F-86000, Poitiers, France
| | - Thomas Daubon
- Université Bordeaux, CNRS, IBGC, UMR5095, Bordeaux, France
| | - Lin Wang
- Department of Computational and Quantitative Medicine, Hematologic Malignancies Research Institute and Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Aaron A Diaz
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Oliver Schnell
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
| | - Jürgen Beck
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
| | - Nicolas Servant
- Institut Curie, PSL University, 75005, Paris, France
- INSERM U900, 75005, Paris, France
- MINES ParisTeach, CBIO-Centre for Computational Biology, PSL Research University, 75006, Paris, France
| | - Lucie Karayan-Tapon
- Université de Poitiers, CHU Poitiers, ProDiCeT, F-86000, Poitiers, France
- CHU Poitiers, Laboratoire de Cancérologie Biologique, F-86000, Poitiers, France
| | - Florence M G Cavalli
- Institut Curie, PSL University, 75005, Paris, France
- INSERM U900, 75005, Paris, France
- MINES ParisTeach, CBIO-Centre for Computational Biology, PSL Research University, 75006, Paris, France
| | - Giorgio Seano
- Institut Curie, INSERM U1021, CNRS UMR3347, Tumor Microenvironment Lab, Paris-Saclay University, 91400, Orsay, France.
| |
Collapse
|
7
|
Lee DH, Imran M, Choi JH, Park YJ, Kim YH, Min S, Park TJ, Choi YW. CDK4/6 inhibitors induce breast cancer senescence with enhanced anti-tumor immunogenic properties compared with DNA-damaging agents. Mol Oncol 2024; 18:216-232. [PMID: 37854019 PMCID: PMC10766199 DOI: 10.1002/1878-0261.13541] [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: 05/22/2023] [Revised: 10/05/2023] [Accepted: 10/17/2023] [Indexed: 10/20/2023] Open
Abstract
Since therapy-induced senescence (TIS) can either support or inhibit cancer progression, identifying which types of chemotherapeutic agents can produce the strongest anti-tumor TIS is an important issue. Here, cyclin-dependent kinase4/6 inhibitors (CDK4/6i)-induced senescence was compared to the TIS induced by conventional DNA-damaging agents. Despite both types of agents eliciting a similar degree of senescence, we observed increased expression of the senescence-associated secretory phenotype (SASP) and ligands related to pro-tumor immunity (IL6, CXCL8, TGFβ, CD274, and CEACAM1) and angiogenesis (VEGFA) mainly in TIS induced by DNA-damaging agents rather than by CDK4/6i. Additionally, although all agents increased the expression of anti-tumor immunomodulatory proteins related to antigen presentation (MHC-I, B2M) and T cell chemokines (CXCL9, 10, 11), CDK4/6i-induced senescent cells still maintained this expression at a similar or even higher intensity than cells treated with DNA-damaging agents, despite the absence of nuclear factor-kappa-B (NF-κB) and p53 activation. These data suggest that in contrast with DNA-damaging agents, which augment the pro-tumorigenic microenvironment via pro-inflammatory SASP, CDK4/6i can generate TIS only with antitumor immunomodulatory proteins.
Collapse
Affiliation(s)
- Dong Hyun Lee
- Department of Biochemistry and Molecular BiologyAjou University School of MedicineSuwonKorea
- Department of Biomedical SciencesAjou University Graduate School of MedicineSuwonKorea
- Inflamm‐Aging Translational Research CenterAjou University Medical CenterSuwonKorea
| | - Muhammad Imran
- Department of Biochemistry and Molecular BiologyAjou University School of MedicineSuwonKorea
- Inflamm‐Aging Translational Research CenterAjou University Medical CenterSuwonKorea
| | - Jae Ho Choi
- Inflamm‐Aging Translational Research CenterAjou University Medical CenterSuwonKorea
- Department of Hematology‐OncologyAjou University School of MedicineSuwonKorea
| | - Yoo Jung Park
- Department of Hematology‐OncologyAjou University School of MedicineSuwonKorea
| | - Young Hwa Kim
- Inflamm‐Aging Translational Research CenterAjou University Medical CenterSuwonKorea
| | - Sunwoo Min
- Department of Biological SciencesKorea Advanced Institute of Science and Technology (KAIST)DaejeonKorea
| | - Tae Jun Park
- Department of Biochemistry and Molecular BiologyAjou University School of MedicineSuwonKorea
- Department of Biomedical SciencesAjou University Graduate School of MedicineSuwonKorea
- Inflamm‐Aging Translational Research CenterAjou University Medical CenterSuwonKorea
| | - Yong Won Choi
- Inflamm‐Aging Translational Research CenterAjou University Medical CenterSuwonKorea
- Department of Hematology‐OncologyAjou University School of MedicineSuwonKorea
| |
Collapse
|
8
|
Wilczyński J, Paradowska E, Wilczyńska J, Wilczyński M. Prediction of Chemoresistance-How Preclinical Data Could Help to Modify Therapeutic Strategy in High-Grade Serous Ovarian Cancer. Curr Oncol 2023; 31:229-249. [PMID: 38248100 PMCID: PMC10814576 DOI: 10.3390/curroncol31010015] [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: 11/13/2023] [Revised: 12/12/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024] Open
Abstract
High-grade serous ovarian cancer (HGSOC) is one of the most lethal tumors generally and the most fatal cancer of the female genital tract. The approved standard therapy consists of surgical cytoreduction and platinum/taxane-based chemotherapy, and of targeted therapy in selected patients. The main therapeutic problem is chemoresistance of recurrent and metastatic HGSOC tumors which results in low survival in the group of FIGO III/IV. Therefore, the prediction and monitoring of chemoresistance seems to be of utmost importance for the improvement of HGSOC management. This type of cancer has genetic heterogeneity with several subtypes being characterized by diverse gene signatures and disturbed peculiar epigenetic regulation. HGSOC develops and metastasizes preferentially in the specific intraperitoneal environment composed mainly of fibroblasts, adipocytes, and immune cells. Different HGSOC subtypes could be sensitive to distinct sets of drugs. Moreover, primary, metastatic, and recurrent tumors are characterized by an individual biology, and thus diverse drug responsibility. Without a precise identification of the tumor and its microenvironment, effective treatment seems to be elusive. This paper reviews tumor-derived genomic, mutational, cellular, and epigenetic biomarkers of HGSOC drug resistance, as well as tumor microenvironment-derived biomarkers of chemoresistance, and discusses their possible use in the novel complex approach to ovarian cancer therapy and monitoring.
Collapse
Affiliation(s)
- Jacek Wilczyński
- Department of Gynecological Surgery and Gynecological Oncology, Medical University of Lodz, 4 Kosciuszki Str., 90-419 Lodz, Poland
| | - Edyta Paradowska
- Laboratory of Virology, Institute of Medical Biology of the Polish Academy of Sciences, 106 Lodowa Str., 93-232 Lodz, Poland;
| | - Justyna Wilczyńska
- Department of Tele-Radiotherapy, Mikolaj Kopernik Provincial Multi-Specialized Oncology and Traumatology Center, 62 Pabianicka Str., 93-513 Lodz, Poland;
| | - Miłosz Wilczyński
- Department of Gynecological, Endoscopic and Oncological Surgery, Polish Mother’s Health Center—Research Institute, 281/289 Rzgowska Str., 93-338 Lodz, Poland;
- Department of Surgical and Endoscopic Gynecology, Medical University of Lodz, 4 Kosciuszki Str., 90-419 Lodz, Poland
| |
Collapse
|
9
|
Mejia Peña C, Skipper TA, Hsu J, Schechter I, Ghosh D, Dawson MR. Metronomic and single high-dose paclitaxel treatments produce distinct heterogenous chemoresistant cancer cell populations. Sci Rep 2023; 13:19232. [PMID: 37932310 PMCID: PMC10628134 DOI: 10.1038/s41598-023-46055-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 10/27/2023] [Indexed: 11/08/2023] Open
Abstract
More than 75% of epithelial ovarian cancer (EOC) patients experience disease recurrence after initial treatment, highlighting our incomplete understanding of how chemoresistant populations evolve over the course of EOC progression post chemotherapy treatment. Here, we show how two paclitaxel (PTX) treatment methods- a single high dose and a weekly metronomic dose for four weeks, generate unique chemoresistant populations. Using mechanically relevant alginate microspheres and a combination of transcript profiling and heterogeneity analyses, we found that these PTX-treatment regimens produce distinct and resilient subpopulations that differ in metabolic reprogramming signatures, acquisition of resistance to PTX and anoikis, and the enrichment for cancer stem cells (CSCs) and polyploid giant cancer cells (PGCCs) with the ability to replenish bulk populations. We investigated the longevity of these metabolic reprogramming events using untargeted metabolomics and found that metabolites associated with stemness and therapy-induced senescence were uniquely abundant in populations enriched for CSCs and PGCCs. Predictive network analysis revealed that antioxidative mechanisms were likely to be differentially active dependent on both time and exposure to PTX. Our results illustrate how current standard chemotherapies contribute to the development of chemoresistant EOC subpopulations by either selecting for intrinsically resistant subpopulations or promoting the evolution of resistance mechanisms. Additionally, our work describes the unique phenotypic signatures in each of these distinct resistant subpopulations and thus highlights potential vulnerabilities that can be exploited for more effective treatment.
Collapse
Affiliation(s)
- Carolina Mejia Peña
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, 02912, USA
| | - Thomas A Skipper
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, 02912, USA
| | - Jeffrey Hsu
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, 02912, USA
| | - Ilexa Schechter
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, 02912, USA
| | - Deepraj Ghosh
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, 02912, USA
| | - Michelle R Dawson
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, 02912, USA.
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, 02912, USA.
| |
Collapse
|
10
|
Xiao S, Qin D, Hou X, Tian L, Yu Y, Zhang R, Lyu H, Guo D, Chen XZ, Zhou C, Tang J. Cellular senescence: a double-edged sword in cancer therapy. Front Oncol 2023; 13:1189015. [PMID: 37771436 PMCID: PMC10522834 DOI: 10.3389/fonc.2023.1189015] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 08/15/2023] [Indexed: 09/30/2023] Open
Abstract
Over the past few decades, cellular senescence has been identified in cancer patients undergoing chemotherapy and radiotherapy. Senescent cells are generally characterized by permanent cell cycle arrest as a response to endogenous and exogenous stresses. In addition to exiting the cell cycle process, cellular senescence also triggers profound phenotypic changes such as senescence-associated secretory phenotype (SASP), autophagy modulation, or metabolic reprograming. Consequently, cellular senescence is often considered as a tumor-suppressive mechanism that permanently arrests cells at risk of malignant transformation. However, accumulating evidence shows that therapy-induced senescence can promote epithelial-mesenchymal transition and tumorigenesis in neighboring cells, as well as re-entry into the cell cycle and activation of cancer stem cells, thereby promoting cancer cell survival. Therefore, it is particularly important to rapidly eliminate therapy-induced senescent cells in patients with cancer. Here we review the hallmarks of cellular senescence and the relationship between cellular senescence and cancer. We also discuss several pathways to induce senescence in tumor therapy, as well as strategies to eliminate senescent cells after cancer treatment. We believe that exploiting the intersection between cellular senescence and tumor cells is an important means to defeat tumors.
Collapse
Affiliation(s)
- Shuai Xiao
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Dongmin Qin
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Xueyang Hou
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Lingli Tian
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Yeping Yu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Rui Zhang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Hao Lyu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Dong Guo
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Cefan Zhou
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Jingfeng Tang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| |
Collapse
|
11
|
Billimoria R, Bhatt P. Senescence in cancer: Advances in detection and treatment modalities. Biochem Pharmacol 2023; 215:115739. [PMID: 37562510 DOI: 10.1016/j.bcp.2023.115739] [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: 06/22/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 08/12/2023]
Abstract
Senescence is a form of irreversible cell cycle arrest. Senescence plays a dual role in cancer, as both a tumor suppressor by preventing the growth of damaged cells and a cancer promoter by creating an inflammatory milieu. Stress-induced premature senescence (SIPS) and replicative senescence are the two major sub-types of senescence. Senescence plays a dual role in cancer, depending on the context and kind of senescence involved. SIPS can cause cancer by nurturing an inflammatory environment, whereas replicative senescence may prevent cancer. Major pathways that are involved in senescence are the p53-p21, p16INK4A-Rb pathway along with mTOR, MAPK, and PI3K pathways. The lack of universal senescence markers makes it difficult to identify senescent cells in vivo. A combination of reliable detection methods of senescent cells in vivo is of utmost importance and will help in early detection and open new avenues for future treatment. New strategies that are being developed in order to tackle these shortcomings are in the field of fluorescent probes, nanoparticles, positron emission tomography probes, biosensors, and the detection of cell-free DNA from liquid biopsies. Along with detection, eradication of these senescent cells is also important to prevent cancer reoccurrence. Recently, the field of nano-senolytic and immunotherapy has also been emerging. This review provides up-to-date information on the various types of advancements made in the field of detection and treatment modalities for senescent cells that hold promise for the future treatment and prognosis of cancer, as well as their limitations and potential solutions.
Collapse
Affiliation(s)
- Rezina Billimoria
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's NMIMS (Deemed-to-be University), Vile Parle (West), Mumbai, India
| | - Purvi Bhatt
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's NMIMS (Deemed-to-be University), Vile Parle (West), Mumbai, India.
| |
Collapse
|
12
|
Mandl GA, Vettier F, Tessitore G, Maurizio SL, Bietar K, Stochaj U, Capobianco JA. Combining Pr 3+-Doped Nanoradiosensitizers and Endogenous Protoporphyrin IX for X-ray-Mediated Photodynamic Therapy of Glioblastoma Cells. ACS APPLIED BIO MATERIALS 2023. [PMID: 37267436 DOI: 10.1021/acsabm.3c00201] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Glioblastoma multiforme is an aggressive type of brain cancer with high recurrence rates due to the presence of radioresistant cells remaining after tumor resection. Here, we report the development of an X-ray-mediated photodynamic therapy (X-PDT) system using NaLuF4:25% Pr3+ radioluminescent nanoparticles in conjunction with protoporphyrin IX (PPIX), an endogenous photosensitizer that accumulates selectively in cancer cells. Conveniently, 5-aminolevulinic acid (5-ALA), the prodrug that is administered for PDT, is the only drug approved for fluorescence-guided resection of glioblastoma, enabling dual detection and treatment of malignant cells. NaLuF4:Pr3+ nanoparticles were synthesized and spectroscopically evaluated at a range of Pr3+ concentrations. This generated radioluminescent nanoparticles with strong emissions from the 1S0 excited state of Pr3+, which overlaps with the Soret band of PPIX to perform photodynamic therapy. The spectral overlap between the nanoparticles and PPIX improved treatment outcomes for U251 cells, which were used as a model for the thin tumor margin. In addition to sensitizing PPIX to induce X-PDT, our nanoparticles exhibit strong radiosensitizing properties through a radiation dose-enhancement effect. We evaluate the effects of the nanoparticles alone and in combination with PPIX on viability, death, stress, senescence, and proliferation. Collectively, our results demonstrate this as a strong proof of concept for nanomedicine.
Collapse
Affiliation(s)
- Gabrielle A Mandl
- Department of Chemistry and Biochemistry & Centre for Nanoscience Research, Concordia University, 7141 Sherbrooke St. W., Montreal, Quebec H4B 1R6, Canada
| | - Freesia Vettier
- Department of Chemistry and Biochemistry & Centre for Nanoscience Research, Concordia University, 7141 Sherbrooke St. W., Montreal, Quebec H4B 1R6, Canada
| | - Gabriella Tessitore
- Department of Chemistry and Biochemistry & Centre for Nanoscience Research, Concordia University, 7141 Sherbrooke St. W., Montreal, Quebec H4B 1R6, Canada
| | - Steven L Maurizio
- Department of Chemistry and Biochemistry & Centre for Nanoscience Research, Concordia University, 7141 Sherbrooke St. W., Montreal, Quebec H4B 1R6, Canada
| | - Kais Bietar
- Department of Physiology, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Ursula Stochaj
- Department of Physiology, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - John A Capobianco
- Department of Chemistry and Biochemistry & Centre for Nanoscience Research, Concordia University, 7141 Sherbrooke St. W., Montreal, Quebec H4B 1R6, Canada
| |
Collapse
|
13
|
Shen Q, Liu Y, Deng X, Hu CD. PRMT5 promotes chemotherapy-induced neuroendocrine differentiation in NSCLC. Thorac Cancer 2023. [PMID: 37140020 DOI: 10.1111/1759-7714.14921] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/05/2023] Open
Abstract
BACKGROUND In response to therapeutic treatments, cancer cells can exhibit a variety of resistance phenotypes including neuroendocrine differentiation (NED). NED is a process by which cancer cells can transdifferentiate into neuroendocrine-like cells in response to treatments, and is now widely accepted as a key mechanism of acquired therapy resistance. Recent clinical evidence has suggested that non-small cell lung cancer (NSCLC) can also transform into small cell lung cancer (SCLC) in patients treated with EGFR inhibitors. However, whether chemotherapy induces NED to confer therapy resistance in NSCLC remains unknown. METHODS We evaluated whether NSCLC cells can undergo NED in response to chemotherapeutic agents etoposide and cisplatin. By Knock-down of PRMT5 or pharmacological inhibition of PRMT5 to identify its role in the NED process. RESULTS We observed that both etoposide and cisplatin can induce NED in multiple NSCLC cell lines. Mechanistically, we identified protein arginine methyltransferase 5 (PRMT5) as a critical mediator of chemotherapy-induced NED. Significantly, the knock-down of PRMT5 or pharmacological inhibition of PRMT5 suppressed the induction of NED and increased the sensitivity to chemotherapy. CONCLUSION Taken together, our results suggest that targeting PRMT5 may be explored as a chemosensitization approach by inhibiting chemotherapy-induced NED.
Collapse
Affiliation(s)
- Qi Shen
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University College of Pharmacy, West Lafayette, Indiana, USA
- Department of Gastroenterology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Liu
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University College of Pharmacy, West Lafayette, Indiana, USA
- College of Chemical Engineering, Sichuan University of Science & Engineering, Zigong, China
| | - Xuehong Deng
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University College of Pharmacy, West Lafayette, Indiana, USA
| | - Chang-Deng Hu
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University College of Pharmacy, West Lafayette, Indiana, USA
| |
Collapse
|
14
|
Ozcan G. The hypoxia-inducible factor-1α in stemness and resistance to chemotherapy in gastric cancer: Future directions for therapeutic targeting. Front Cell Dev Biol 2023; 11:1082057. [PMID: 36846589 PMCID: PMC9945545 DOI: 10.3389/fcell.2023.1082057] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Hypoxia-inducible factor-1α (HIF-1α) is a crucial mediator of intra-tumoral heterogeneity, tumor progression, and unresponsiveness to therapy in tumors with hypoxia. Gastric tumors, one of the most aggressive tumors in the clinic, are highly enriched in hypoxic niches, and the degree of hypoxia is strongly correlated with poor survival in gastric cancer patients. Stemness and chemoresistance in gastric cancer are the two root causes of poor patient outcomes. Based on the pivotal role of HIF-1α in stemness and chemoresistance in gastric cancer, the interest in identifying critical molecular targets and strategies for surpassing the action of HIF-1α is expanding. Despite that, the understanding of HIF-1α induced signaling in gastric cancer is far from complete, and the development of efficacious HIF-1α inhibitors bears various challenges. Hence, here we review the molecular mechanisms by which HIF-1α signaling stimulates stemness and chemoresistance in gastric cancer, with the clinical efforts and challenges to translate anti-HIF-1α strategies into the clinic.
Collapse
Affiliation(s)
- Gulnihal Ozcan
- Department of Medical Pharmacology, School of Medicine, Koç University, Istanbul, Turkiye
- Koç University Research Center for Translational Medicine, Istanbul, Turkiye
| |
Collapse
|
15
|
Romaniello D, Gelfo V, Pagano F, Sgarzi M, Morselli A, Girone C, Filippini DM, D’Uva G, Lauriola M. IL-1 and senescence: Friends and foe of EGFR neutralization and immunotherapy. Front Cell Dev Biol 2023; 10:1083743. [PMID: 36712972 PMCID: PMC9877625 DOI: 10.3389/fcell.2022.1083743] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 12/19/2022] [Indexed: 01/13/2023] Open
Abstract
Historically, senescence has been considered a safe program in response to multiple stresses in which cells undergo irreversible growth arrest. This process is characterized by morphological and metabolic changes, heterochromatin formation, and secretion of inflammatory components, known as senescence-associated secretory phenotype (SASP). However, recent reports demonstrated that anti-cancer therapy itself can stimulate a senescence response in tumor cells, the so-called therapy-induced senescence (TIS), which may represent a temporary bypass pathway that promotes drug resistance. In this context, several studies have shown that EGFR blockage, by TKIs or moAbs, promotes TIS by increasing IL-1 cytokine production, thus pushing cells into a "pseudo-senescent" state. Today, senotherapeutic agents are emerging as a potential strategy in cancer treatment thanks to their dual role in annihilating senescent cells and simultaneously preventing their awakening into a resistant and aggressive form. Here, we summarize classic and recent findings about the cellular processes driving senescence and SASP, and we provide a state-of-the-art of the anti-cancer strategies available so far that exploits the activation and/or blockade of senescence-based mechanisms.
Collapse
Affiliation(s)
- Donatella Romaniello
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy,Centre for Applied Biomedical Research (CRBA), Bologna University Hospital Authority St. Orsola -Malpighi Polyclinic, Bologna, Italy
| | - Valerio Gelfo
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy,Centre for Applied Biomedical Research (CRBA), Bologna University Hospital Authority St. Orsola -Malpighi Polyclinic, Bologna, Italy
| | - Federica Pagano
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Michela Sgarzi
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Alessandra Morselli
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Cinzia Girone
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Daria Maria Filippini
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy,Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Gabriele D’Uva
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy,Centre for Applied Biomedical Research (CRBA), Bologna University Hospital Authority St. Orsola -Malpighi Polyclinic, Bologna, Italy,National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
| | - Mattia Lauriola
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy,Centre for Applied Biomedical Research (CRBA), Bologna University Hospital Authority St. Orsola -Malpighi Polyclinic, Bologna, Italy,*Correspondence: Mattia Lauriola,
| |
Collapse
|
16
|
Estepa-Fernández A, García-Fernández A, Lérida-Viso A, Blandez JF, Galiana I, Sancenon-Galarza F, Orzáez M, Martínez-Máñez R. Combination of palbociclib with navitoclax based-therapies enhances in vivo antitumoral activity in triple-negative breast cancer. Pharmacol Res 2023; 187:106628. [PMID: 36566002 DOI: 10.1016/j.phrs.2022.106628] [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] [Received: 05/17/2022] [Revised: 11/29/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
Triple-negative breast cancer (TNBC) is a very aggressive subtype of breast cancer with a poor prognosis and limited effective therapeutic options. Induction of senescence, arrest of cell proliferation, has been explored as an effective method to limit tumor progression in metastatic breast cancer. However, relapses occur in some patients, possibly as a result of the accumulation of senescent tumor cells in the body after treatment, which promote metastasis. In this study, we explored the combination of senescence induction and the subsequent removal of senescent cells (senolysis) as an alternative approach to improve outcomes in TNBC patients. We demonstrate that a combination treatment, using the senescence-inducer palbociclib and the senolytic agent navitoclax, delays tumor growth and reduces metastases in a mouse xenograft model of aggressive human TNBC (hTNBC). Furthermore, considering the off-target effects and toxicity derived from the use of navitoclax, we propose a strategy aimed at minimizing the associated side effects. We use a galacto-conjugated navitoclax (nav-Gal) as a senolytic prodrug that can preferentially be activated by β-galactosidase overexpressed in senescent cells. Concomitant treatment with palbociclib and nav-Gal in vivo results in the eradication of senescent hTNBC cells with consequent reduction of tumor growth, while reducing the cytotoxicity of navitoclax. Taken together, our results support the efficacy of combination therapy of senescence-induction with senolysis for hTNBC, as well as the development of a targeted approach as an effective and safer therapeutic opportunity.
Collapse
Affiliation(s)
- Alejandra Estepa-Fernández
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) Universitat Politècnica de València, Universitat de València. Camino de Vera, s/n, 46022 Valencia, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe. C/ Eduardo Primo Yúfera 3, 46012 Valencia, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Spain
| | - Alba García-Fernández
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) Universitat Politècnica de València, Universitat de València. Camino de Vera, s/n, 46022 Valencia, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe. C/ Eduardo Primo Yúfera 3, 46012 Valencia, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Spain.
| | - Araceli Lérida-Viso
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) Universitat Politècnica de València, Universitat de València. Camino de Vera, s/n, 46022 Valencia, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe. C/ Eduardo Primo Yúfera 3, 46012 Valencia, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Spain; Unidad Mixta de Investigación en Nanomedicina y Sensores. Universitat Politècnica de València, IIS La Fe. Av. Fernando Abril Martorell, 106 Torre A 7ª planta, 46026 Valencia, Spain
| | - Juan F Blandez
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) Universitat Politècnica de València, Universitat de València. Camino de Vera, s/n, 46022 Valencia, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Spain; Unidad Mixta de Investigación en Nanomedicina y Sensores. Universitat Politècnica de València, IIS La Fe. Av. Fernando Abril Martorell, 106 Torre A 7ª planta, 46026 Valencia, Spain
| | - Irene Galiana
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) Universitat Politècnica de València, Universitat de València. Camino de Vera, s/n, 46022 Valencia, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe. C/ Eduardo Primo Yúfera 3, 46012 Valencia, Spain
| | - Félix Sancenon-Galarza
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) Universitat Politècnica de València, Universitat de València. Camino de Vera, s/n, 46022 Valencia, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe. C/ Eduardo Primo Yúfera 3, 46012 Valencia, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Spain; Unidad Mixta de Investigación en Nanomedicina y Sensores. Universitat Politècnica de València, IIS La Fe. Av. Fernando Abril Martorell, 106 Torre A 7ª planta, 46026 Valencia, Spain
| | - Mar Orzáez
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe. C/ Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, 46012 Valencia, Spain.
| | - Ramón Martínez-Máñez
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) Universitat Politècnica de València, Universitat de València. Camino de Vera, s/n, 46022 Valencia, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe. C/ Eduardo Primo Yúfera 3, 46012 Valencia, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Spain; Unidad Mixta de Investigación en Nanomedicina y Sensores. Universitat Politècnica de València, IIS La Fe. Av. Fernando Abril Martorell, 106 Torre A 7ª planta, 46026 Valencia, Spain.
| |
Collapse
|
17
|
Zhai J, Han J, Li C, Lv D, Ma F, Xu B. Tumor senescence leads to poor survival and therapeutic resistance in human breast cancer. Front Oncol 2023; 13:1097513. [PMID: 36937388 PMCID: PMC10019818 DOI: 10.3389/fonc.2023.1097513] [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: 11/14/2022] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Background Breast cancer (BRCA) is the most common malignant tumor that seriously threatens the health of women worldwide. Senescence has been suggested as a pivotal player in the onset and progression of tumors as well as the process of treatment resistance. However, the role of senescence in BRCA remains unelucidated. Methods The clinical and transcriptomic data of 2994 patients with BRCA were obtained from The Cancer Genome Atlas and the METABRIC databases. Consensus clustering revealed senescence-associated subtypes of BRCA patients. Functional enrichment analysis explored biological effect of senescence. We then applied weighted gene co-expression network analysis (WGCNA) and LASSO regression to construct a senescence scoring model, Sindex. Survival analysis validated the effectiveness of Sindex to predict the overall survival (OS) of patients with BRCA. A nomogram was constructed by multivariate Cox regression. We used Oncopredict algorithm and real-world data from clinical trials to explore the value of Sindex in predicting response to cancer therapy. Results We identified two distinct senescence-associated subtypes, noted low senescence CC1 and high senescence CC2. Survival analysis revealed worse OS associated with high senescence, which was also validated with patient samples from the National Cancer Center in China. Further analysis revealed extensively cell division and suppression of extracellular matrix process, along with lower stromal and immune scores in the high senescence CC2. We then constructed a 37 signature gene scoring model, Sindex, with robust predictive capability in patients with BRCA, especially for long time OS beyond 10 years. We demonstrated that the Sene-high subtype was resistant to CDK inhibitors but sensitive to proteosome inhibitors, and there was no significant difference in paclitaxel chemotherapy and immunotherapy between patients with different senescence statuses. Conclusions We reported senescence as a previously uncharacterized hallmark of BRCA that impacts patient outcomes and therapeutic response. Our analysis demonstrated that the Sindex can be used to identify not only patients at different risk levels for the OS but also patients who would benefit from some cancer therapeutic drugs.
Collapse
Affiliation(s)
- Jingtong Zhai
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiashu Han
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- 4 + 4 Medical Doctor Program, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Cong Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dan Lv
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Binghe Xu, ; Fei Ma,
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Binghe Xu, ; Fei Ma,
| |
Collapse
|
18
|
Pardella E, Pranzini E, Nesi I, Parri M, Spatafora P, Torre E, Muccilli A, Castiglione F, Fambrini M, Sorbi F, Cirri P, Caselli A, Puhr M, Klocker H, Serni S, Raugei G, Magherini F, Taddei ML. Therapy-Induced Stromal Senescence Promoting Aggressiveness of Prostate and Ovarian Cancer. Cells 2022; 11:cells11244026. [PMID: 36552790 PMCID: PMC9776582 DOI: 10.3390/cells11244026] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/09/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022] Open
Abstract
Cancer progression is supported by the cross-talk between tumor cells and the surrounding stroma. In this context, senescent cells in the tumor microenvironment contribute to the development of a pro-inflammatory milieu and the acquisition of aggressive traits by cancer cells. Anticancer treatments induce cellular senescence (therapy-induced senescence, TIS) in both tumor and non-cancerous cells, contributing to many detrimental side effects of therapies. Thus, we focused on the effects of chemotherapy on the stromal compartment of prostate and ovarian cancer. We demonstrated that anticancer chemotherapeutics, regardless of their specific mechanism of action, promote a senescent phenotype in stromal fibroblasts, resulting in metabolic alterations and secretion of paracrine factors, sustaining the invasive and clonogenic potential of both prostate and ovarian cancer cells. The clearance of senescent stromal cells, through senolytic drug treatment, reverts the malignant phenotype of tumor cells. The clinical relevance of TIS was validated in ovarian and prostate cancer patients, highlighting increased accumulation of lipofuscin aggregates, a marker of the senescent phenotype, in the stromal compartment of tissues from chemotherapy-treated patients. These data provide new insights into the potential efficacy of combining traditional anticancer strategies with innovative senotherapy to potentiate anticancer treatments and overcome the adverse effects of chemotherapy.
Collapse
Affiliation(s)
- Elisa Pardella
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Viale Morgagni 50, 50134 Florence, Italy
| | - Erica Pranzini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Viale Morgagni 50, 50134 Florence, Italy
| | - Ilaria Nesi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Viale Morgagni 50, 50134 Florence, Italy
| | - Matteo Parri
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Viale Morgagni 50, 50134 Florence, Italy
| | - Pietro Spatafora
- Department of Minimally Invasive and Robotic Urologic Surgery and Kidney Transplantation, University of Florence, 50134 Florence, Italy
| | - Eugenio Torre
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Viale Morgagni 50, 50134 Florence, Italy
| | - Angela Muccilli
- Department of Health Sciences, Section of Pathology, University of Florence, 50134 Florence, Italy
| | - Francesca Castiglione
- Histopathology and Molecular Diagnostics, Careggi Teaching Hospital, 50134 Florence, Italy
| | - Massimiliano Fambrini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Viale Morgagni 50, 50134 Florence, Italy
| | - Flavia Sorbi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Viale Morgagni 50, 50134 Florence, Italy
| | - Paolo Cirri
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Viale Morgagni 50, 50134 Florence, Italy
| | - Anna Caselli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Viale Morgagni 50, 50134 Florence, Italy
| | - Martin Puhr
- Department of Urology, Division of Experimental Urology, Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | - Helmut Klocker
- Department of Urology, Division of Experimental Urology, Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | - Sergio Serni
- Department of Minimally Invasive and Robotic Urologic Surgery and Kidney Transplantation, University of Florence, 50134 Florence, Italy
| | - Giovanni Raugei
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Viale Morgagni 50, 50134 Florence, Italy
| | - Francesca Magherini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Viale Morgagni 50, 50134 Florence, Italy
| | - Maria Letizia Taddei
- Department of Experimental and Clinical Medicine, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
- Correspondence:
| |
Collapse
|
19
|
Hu X, Guo L, Liu G, Dai Z, Wang L, Zhang J, Wang J. Novel cellular senescence-related risk model identified as the prognostic biomarkers for lung squamous cell carcinoma. Front Oncol 2022; 12:997702. [DOI: 10.3389/fonc.2022.997702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/20/2022] [Indexed: 11/18/2022] Open
Abstract
BackgroundLung cancer is one of the top causes of cancer-related death worldwide. Cellular senescence is a characteristic of cell cycle arrest that plays a role in carcinogenesis and immune microenvironment modulation. Despite this, the clinical and immune cell infiltration features of senescence in lung squamous cell carcinoma (LUSC) are unknown.MethodsThe Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) were used to get RNA-seq data and clinical information for LUSC. The least absolute shrinkage and selection operator (LASSO)-Cox regression, receiver operating characteristic (ROC), and Kaplan-Meier analysis were used to evaluate a risk model for predicting overall survival based on six differentially expressed genes. The tumor microenvironment (TME) and immunotherapy response were also studied.ResultsTo discriminate LUSC into high- and low-risk subgroups, a risk model comprised of six cellular senescence-related genes (CDKN1A, CEBPB, MDH1, SIX1, SNAI1, and SOX5) was developed. The model could stratify patients into high-risk and low-risk groups, according to ROC and Kaplan-Meier analysis. In the TCGA-LUSC and GSE73403 cohorts, the high-risk group had a worse prognosis (P<0.05), and was associated with immune cell inactivation and being insensitive to immunotherapy in IMvigor210.ConclusionsWe discovered a new LUSC classification based on six cellular senescence-related genes, which will aid in identifying patients who will benefit from anti-PD-1 treatment. Targeting senescence-related genes appears to be another option for improving clinical therapy for LUSC.
Collapse
|
20
|
Liu H, Zhao H, Sun Y. Tumor microenvironment and cellular senescence: Understanding therapeutic resistance and harnessing strategies. Semin Cancer Biol 2022; 86:769-781. [PMID: 34799201 DOI: 10.1016/j.semcancer.2021.11.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/24/2021] [Accepted: 11/08/2021] [Indexed: 01/27/2023]
Abstract
The tumor microenvironment (TME) is a major contributor to cancer malignancy including development of therapeutic resistance, a process mediated in part through intercellular crosstalk. Besides diverse soluble factors responsible for pro-survival pathway activation, immune evasion and extracellular matrix (ECM) remodeling further promote cancer resistance. Importantly, therapy-induced senescence (TIS) of cells in the TME is frequently observed in anticancer regimens, an off-target effect that can generate profound impacts on disease progression. By conferring the resistance and fueling the repopulation of remaining cancerous cells, TIS is responsible for tumor relapse and distant metastasis in posttreatment stage. This pathological trajectory can be substantially driven by the pro-inflammatory feature of senescent cells, termed as the senescence-associated secretory phenotype (SASP). Targeting strategies to selectively and efficiently remove senescent cells before they exert non-autonomous but largely deleterious effects, are emerging as an effective solution to prevent drug resistance acquired from a treatment-remodeled TME. In this review, we summarize the TME composition and key activities that affect tissue homeostasis and support treatment resistance. Promising opportunities that allow TME-manipulation and senescent cell-targeting (senotherapy) are discussed, with translational pipelines to overcome therapeutic barriers in clinical oncology projected.
Collapse
Affiliation(s)
- Hanxin Liu
- Department of Pharmacology, Institute of Aging Medicine, Binzhou Medical University, Yantai, Shandong, 264003, China
| | - Huifang Zhao
- Department of Pharmacology, Institute of Aging Medicine, Binzhou Medical University, Yantai, Shandong, 264003, China
| | - Yu Sun
- Department of Pharmacology, Institute of Aging Medicine, Binzhou Medical University, Yantai, Shandong, 264003, China; CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China; Department of Medicine and VAPSHCS, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
21
|
Piskorz WM, Cechowska-Pasko M. Senescence of Tumor Cells in Anticancer Therapy—Beneficial and Detrimental Effects. Int J Mol Sci 2022; 23:ijms231911082. [PMID: 36232388 PMCID: PMC9570404 DOI: 10.3390/ijms231911082] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/16/2022] [Accepted: 09/18/2022] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence process results in stable cell cycle arrest, which prevents cell proliferation. It can be induced by a variety of stimuli including metabolic stress, DNA damage, telomeres shortening, and oncogenes activation. Senescence is generally considered as a process of tumor suppression, both by preventing cancer cells proliferation and inhibiting cancer progression. It can also be a key effector mechanism for many types of anticancer therapies such as chemotherapy and radiotherapy, both directly and through bioactive molecules released by senescent cells that can stimulate an immune response. Senescence is characterized by a senescence-associated secretory phenotype (SASP) that can have both beneficial and detrimental impact on cancer progression. Despite the negatives, attempts are still being made to use senescence to fight cancer, especially when it comes to senolytics. There is a possibility that a combination of prosenescence therapy—which targets tumor cells and causes their senescence—with senotherapy—which targets senescent cells, can be promising in cancer treatment. This review provides information on cellular senescence, its connection with carcinogenesis and therapeutic possibilities linked to this process.
Collapse
|
22
|
Llop-Hernández À, Verdura S, Cuyàs E, Menendez JA. Nutritional Niches of Cancer Therapy-Induced Senescent Cells. Nutrients 2022; 14:nu14173636. [PMID: 36079891 PMCID: PMC9460569 DOI: 10.3390/nu14173636] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/30/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
Therapy-induced senescence (TIS) is a state of stable proliferative arrest of both normal and neoplastic cells that is triggered by exposure to anticancer treatments. TIS cells acquire a senescence-associated secretory phenotype (SASP), which is pro-inflammatory and actively promotes tumor relapse and adverse side-effects in patients. Here, we hypothesized that TIS cells adapt their scavenging and catabolic ability to overcome the nutritional constraints in their microenvironmental niches. We used a panel of mechanistically-diverse TIS triggers (i.e., bleomycin, doxorubicin, alisertib, and palbociclib) and Biolog Phenotype MicroArrays to identify (among 190 different carbon and nitrogen sources) candidate metabolites that support the survival of TIS cells in limiting nutrient conditions. We provide evidence of distinguishable TIS-associated nutrient consumption profiles involving a core set of shared (e.g., glutamine) and unique (e.g., glucose-1-phosphate, inosine, and uridine) nutritional sources after diverse senescence-inducing interventions. We also observed a trend for an inverse correlation between the intensity of the pro-inflammatory SASP provoked by different TIS agents and diversity of compensatory nutritional niches utilizable by senescent cells. These findings support the detailed exploration of the nutritional niche as a new metabolic dimension to understand and target TIS in cancer.
Collapse
Affiliation(s)
| | - Sara Verdura
- Metabolism and Cancer Group, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, 17005 Girona, Spain
| | - Elisabet Cuyàs
- Girona Biomedical Research Institute, 17190 Girona, Spain
- Metabolism and Cancer Group, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, 17005 Girona, Spain
- Correspondence: (E.C.); or (J.A.M.)
| | - Javier A. Menendez
- Girona Biomedical Research Institute, 17190 Girona, Spain
- Metabolism and Cancer Group, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, 17005 Girona, Spain
- Correspondence: (E.C.); or (J.A.M.)
| |
Collapse
|
23
|
Bientinesi E, Lulli M, Becatti M, Ristori S, Margheri F, Monti D. Doxorubicin-induced senescence in normal fibroblasts promotes in vitro tumour cell growth and invasiveness: the role of Quercetin in modulating these processes. Mech Ageing Dev 2022; 206:111689. [PMID: 35728630 DOI: 10.1016/j.mad.2022.111689] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/05/2022] [Accepted: 06/16/2022] [Indexed: 01/10/2023]
Abstract
Ageing is a complex biological phenomenon representing the major risk factor for developing age-related diseases, such as cardiovascular pathologies, neurodegenerative diseases, and cancer. Geroscience, the new vision of gerontology, identifies cellular senescence as an interconnected biological process that characterises ageing and age-related diseases. Therefore, many strategies have been employed in the last years to reduce the harmful effects of senescence, and among these, the most intriguing ones use nutraceutical compounds. Here we show that a pre-treatment with Quercetin, a bioactive flavonoid present in many fruits and vegetables, increasing cellular antioxidant defence, can alleviate Doxorubicin (Doxo)-induced cellular senescence in human normal WI-38 fibroblasts. Furthermore, our work demonstrates that Quercetin pre-treatment, reducing the number of senescent cells and the production of the senescence-associated secretory phenotype (SASP) factors, can decrease the pro-tumour effects of conditioned medium from Doxo-induced senescent fibroblasts on osteosarcoma cells. Overall, our findings are consistent with the hypothesis that targeting senescent cells can be an emerging strategy for cancer treatment, especially in elderly patients, in which senescent cells are already abundant in several tissues and organs.
Collapse
Affiliation(s)
- Elisa Bientinesi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy 50134
| | - Matteo Lulli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy 50134.
| | - Matteo Becatti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy 50134.
| | - Sara Ristori
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy 50134.
| | - Francesca Margheri
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy 50134.
| | - Daniela Monti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy 50134.
| |
Collapse
|
24
|
Lee AH, Mejia Peña C, Dawson MR. Comparing the Secretomes of Chemorefractory and Chemoresistant Ovarian Cancer Cell Populations. Cancers (Basel) 2022; 14:1418. [PMID: 35326569 PMCID: PMC8946241 DOI: 10.3390/cancers14061418] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/04/2022] [Accepted: 03/08/2022] [Indexed: 12/13/2022] Open
Abstract
High-grade serous ovarian cancer (HGSOC) constitutes the majority of all ovarian cancer cases and has staggering rates of both refractory and recurrent disease. While most patients respond to the initial treatment with paclitaxel and platinum-based drugs, up to 25% do not, and of the remaining that do, 75% experience disease recurrence within the subsequent two years. Intrinsic resistance in refractory cases is driven by environmental stressors like tumor hypoxia which alter the tumor microenvironment to promote cancer progression and resistance to anticancer drugs. Recurrent disease describes the acquisition of chemoresistance whereby cancer cells survive the initial exposure to chemotherapy and develop adaptations to enhance their chances of surviving subsequent treatments. Of the environmental stressors cancer cells endure, exposure to hypoxia has been identified as a potent trigger and priming agent for the development of chemoresistance. Both in the presence of the stress of hypoxia or the therapeutic stress of chemotherapy, cancer cells manage to cope and develop adaptations which prime populations to survive in future stress. One adaptation is the modification in the secretome. Chemoresistance is associated with translational reprogramming for increased protein synthesis, ribosome biogenesis, and vesicle trafficking. This leads to increased production of soluble proteins and extracellular vesicles (EVs) involved in autocrine and paracrine signaling processes. Numerous studies have demonstrated that these factors are largely altered between the secretomes of chemosensitive and chemoresistant patients. Such factors include cytokines, growth factors, EVs, and EV-encapsulated microRNAs (miRNAs), which serve to induce invasive molecular, biophysical, and chemoresistant phenotypes in neighboring normal and cancer cells. This review examines the modifications in the secretome of distinct chemoresistant ovarian cancer cell populations and specific secreted factors, which may serve as candidate biomarkers for aggressive and chemoresistant cancers.
Collapse
Affiliation(s)
- Amy H. Lee
- Center for Biomedical Engineering, Brown University, Providence, RI 02912, USA;
| | - Carolina Mejia Peña
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA;
| | - Michelle R. Dawson
- Center for Biomedical Engineering, Brown University, Providence, RI 02912, USA;
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA;
| |
Collapse
|
25
|
Fakhri S, Zachariah Moradi S, DeLiberto LK, Bishayee A. Cellular senescence signaling in cancer: A novel therapeutic target to combat human malignancies. Biochem Pharmacol 2022; 199:114989. [DOI: 10.1016/j.bcp.2022.114989] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/05/2022] [Accepted: 03/07/2022] [Indexed: 12/26/2022]
|
26
|
Liu Y, Yang H, Liu Q, Pan M, Wang D, Pan S, Zhang W, Wei J, Zhao X, Ji J. Selenocystine-Derived Label-Free Fluorescent Schiff Base Nanocomplex for siRNA Delivery Synergistically Kills Cancer Cells. Molecules 2022; 27:1302. [PMID: 35209090 PMCID: PMC8878402 DOI: 10.3390/molecules27041302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/24/2022] [Accepted: 02/08/2022] [Indexed: 11/16/2022] Open
Abstract
Chemo and siRNA synergic treatments for tumors is a promising new therapeutic trend. Selenocystine, a selenium analog of cysteine, has been considered a potential antitumor agent due to its redox perturbing role. In this study, we developed a nanocarrier for siRNA based on a selenocystine analog engineered polyetherimide and achieved traceable siRNA delivery and the synergic killing of tumor cells. Notably, we applied the label-free Schiff base fluorescence mechanism, which enabled us to trace the siRNA delivery and to monitor the selenocystine analogs' local performance. A novel selenocystine-derived fluorescent Schiff base linker was used to crosslink the polyetherimide, thereby generating a traceable siRNA delivery vehicle with green fluorescence. Moreover, we found that this compound induced tumor cells to undergo senescence. Together with the delivery of a siRNA targeting the anti-apoptotic BCL-xl/w genes in senescent cells, it achieved a synergistic inhibition function by inducing both senescence and apoptosis of tumor cells. Therefore, this study provides insights into the development of label-free probes, prodrugs, and materials towards the synergic strategies for cancer therapy.
Collapse
Affiliation(s)
- Yang Liu
- Center of Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou 310058, China;
| | - Haoying Yang
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng 475004, China; (H.Y.); (Q.L.); (M.P.); (S.P.); (W.Z.)
| | - Qian Liu
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng 475004, China; (H.Y.); (Q.L.); (M.P.); (S.P.); (W.Z.)
| | - Mingming Pan
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng 475004, China; (H.Y.); (Q.L.); (M.P.); (S.P.); (W.Z.)
| | - Danli Wang
- Zhoushan Hospital of Zhejiang Province, Zhoushan 316004, China;
| | - Shiyuan Pan
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng 475004, China; (H.Y.); (Q.L.); (M.P.); (S.P.); (W.Z.)
| | - Weiran Zhang
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng 475004, China; (H.Y.); (Q.L.); (M.P.); (S.P.); (W.Z.)
| | - Jinfeng Wei
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng 475004, China; (H.Y.); (Q.L.); (M.P.); (S.P.); (W.Z.)
| | - Xiaowei Zhao
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng 475004, China; (H.Y.); (Q.L.); (M.P.); (S.P.); (W.Z.)
| | - Junfeng Ji
- Center of Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou 310058, China;
| |
Collapse
|
27
|
Different Cell Responses to Hinokitiol Treatment Result in Senescence or Apoptosis in Human Osteosarcoma Cell Lines. Int J Mol Sci 2022; 23:ijms23031632. [PMID: 35163553 PMCID: PMC8835861 DOI: 10.3390/ijms23031632] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 02/01/2023] Open
Abstract
Hinokitiol is a tropolone-related compound isolated from the heartwood of cupressaceous plants. It is known to exhibit various biological functions including antibacterial, antifungal, and antioxidant activities. In the study, we investigated the antitumor activities of hinokitiol against human osteosarcoma cells. The results revealed that hinokitiol treatment inhibited cell viability of human osteosarcoma U-2 OS and MG-63 cells in the MTT assay. Further study revealed that hinokitiol exposure caused cell cycle arrest at the S phase and a DNA damage response with the induction of γ-H2AX foci in both osteosarcoma cell lines. In U-2 OS cells with wild-type tumor suppressor p53, we found that hinokitiol exposure induced p53 expression and cellular senescence, and knockdown of p53 suppressed the senescence. However, in MG-63 cells with mutated p53, a high percentage of cells underwent apoptosis with cleaved-PARP expression and Annexin V staining after hinokitiol treatment. In addition, up-regulated autophagy was observed both in hinokitiol-exposed U-2 OS and MG-63 cells. As the autophagy was suppressed through the autophagy inhibitor chloroquine, hinokitiol-induced senescence in U-2 OS cells was significantly enhanced accompanying more abundant p53 expression. In MG-63 cells, co-treatment of chloroquine increased hinokitiol-induced apoptosis and decreased cell viability of the treated cells. Our data revealed that hinokitiol treatment could result in different cell responses, senescence or apoptosis in osteosarcoma cell lines, and suppression of autophagy could promote these effects. We hypothesize that the analysis of p53 status and co-administration of autophagy inhibitors might provide more precise and efficacious therapies in hinokitiol-related trials for treating osteosarcoma.
Collapse
|
28
|
Zhang JW, Zhang D, Yu BP. Senescent cells in cancer therapy: why and how to remove them. Cancer Lett 2021; 520:68-79. [PMID: 34237406 DOI: 10.1016/j.canlet.2021.07.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/22/2021] [Accepted: 07/01/2021] [Indexed: 01/10/2023]
Abstract
Cellular senescence is a stress response that imposes a growth arrest on cancer and nonmalignant cells during cancer therapy. By secreting a plethora of proinflammatory factors collectively termed the senescence-associated secretory phenotype (SASP), therapy-induced senescent cells can promote tumorigenesis. Moreover, the SASP from senescent cells is also able to drive therapy resistance and mediate many adverse effects of cancer therapy. Because senescent cell production often occurs during cancer therapy, it is important to carefully consider these potential detrimental effects. Senotherapy, which refers to selective removal of senescent cells, has been proposed as a promising adjuvant approach to eliminate the adverse effects of senescent cells. Thus, in this review we summarize in detail the mechanisms by which senescent cells contribute to tumorigenesis and therapeutic resistance. Also, we thoroughly discuss the potential strategies regarding how to effectively circumvent the undesirable effects of therapy-induced senescent cells.
Collapse
Affiliation(s)
- Jian-Wei Zhang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China; Key Laboratory of Hubei Province for Digestive System Diseases, Wuhan, Hubei Province, People's Republic of China
| | - Dan Zhang
- Chongqing University Cancer Hospital, Chongqing, People's Republic of China
| | - Bao-Ping Yu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China; Key Laboratory of Hubei Province for Digestive System Diseases, Wuhan, Hubei Province, People's Republic of China.
| |
Collapse
|
29
|
Chakrabarty A, Chakraborty S, Bhattacharya R, Chowdhury G. Senescence-Induced Chemoresistance in Triple Negative Breast Cancer and Evolution-Based Treatment Strategies. Front Oncol 2021; 11:674354. [PMID: 34249714 PMCID: PMC8264500 DOI: 10.3389/fonc.2021.674354] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 06/01/2021] [Indexed: 01/10/2023] Open
Abstract
Triple negative breast cancer (TNBC) is classically treated with combination chemotherapies. Although, initially responsive to chemotherapies, TNBC patients frequently develop drug-resistant, metastatic disease. Chemotherapy resistance can develop through many mechanisms, including induction of a transient growth-arrested state, known as the therapy-induced senescence (TIS). In this paper, we will focus on chemoresistance in TNBC due to TIS. One of the key characteristics of senescent cells is a complex secretory phenotype, known as the senescence-associated secretory proteome (SASP), which by prompting immune-mediated clearance of senescent cells maintains tissue homeostasis and suppresses tumorigenesis. However, in cancer, particularly with TIS, senescent cells themselves as well as SASP promote cellular reprograming into a stem-like state responsible for the emergence of drug-resistant, aggressive clones. In addition to chemotherapies, outcomes of recently approved immune and DNA damage-response (DDR)-directed therapies are also affected by TIS, implying that this a common strategy used by cancer cells for evading treatment. Although there has been an explosion of scientific research for manipulating TIS for prevention of drug resistance, much of it is still at the pre-clinical stage. From an evolutionary perspective, cancer is driven by natural selection, wherein the fittest tumor cells survive and proliferate while the tumor microenvironment influences tumor cell fitness. As TIS seems to be preferred for increasing the fitness of drug-challenged cancer cells, we will propose a few tactics to control it by using the principles of evolutionary biology. We hope that with appropriate therapeutic intervention, this detrimental cellular fate could be diverted in favor of TNBC patients.
Collapse
|
30
|
Ye X, Liu X, Gao M, Gong L, Tian F, Shen Y, Hu H, Sun G, Zou Y, Gong Y. CUL4B Promotes Temozolomide Resistance in Gliomas by Epigenetically Repressing CDNK1A Transcription. Front Oncol 2021; 11:638802. [PMID: 33869025 PMCID: PMC8050354 DOI: 10.3389/fonc.2021.638802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/19/2021] [Indexed: 01/10/2023] Open
Abstract
Resistance to temozolomide (TMZ), the first-line chemotherapeutic drug for glioblastoma (GBM) and anaplastic gliomas, is one of the most significant obstacles in clinical treatment. TMZ resistance is regulated by complex genetic and epigenetic networks. Understanding the mechanisms of TMZ resistance can help to identify novel drug targets and more effective therapies. CUL4B has been shown to be upregulated and promotes progression and chemoresistance in several cancer types. However, its regulatory effect and mechanisms on TMZ resistance have not been elucidated. The aim of this study was to decipher the role and mechanism of CUL4B in TMZ resistance. Western blot and public datasets analysis showed that CUL4B was upregulated in glioma specimens. CUL4B elevation positively correlated with advanced pathological stage, tumor recurrence, malignant molecular subtype and poor survival in glioma patients receiving TMZ treatment. CUL4B expression was correlated with TMZ resistance in GBM cell lines. Knocking down CUL4B restored TMZ sensitivity, while upregulation of CUL4B promoted TMZ resistance in GBM cells. By employing senescence β-galactosidase staining, quantitative reverse transcription PCR and Chromatin immunoprecipitation experiments, we found that CUL4B coordinated histone deacetylase (HDAC) to co-occupy the CDKN1A promoter and epigenetically silenced CDKN1A transcription, leading to attenuation of TMZ-induced senescence and rendering the GBM cells TMZ resistance. Collectively, our findings identify a novel mechanism by which GBM cells develop resistance to TMZ and suggest that CUL4B inhibition may be beneficial for overcoming resistance.
Collapse
Affiliation(s)
- Xiang Ye
- Key Laboratory of Experimental Teratology of Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaochen Liu
- Key Laboratory of Experimental Teratology of Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Min Gao
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Li Gong
- Key Laboratory of Experimental Teratology of Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fei Tian
- Key Laboratory of Experimental Teratology of Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yangli Shen
- Key Laboratory of Experimental Teratology of Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Huili Hu
- Key Laboratory of Experimental Teratology of Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Gongping Sun
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yongxin Zou
- Key Laboratory of Experimental Teratology of Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yaoqin Gong
- Key Laboratory of Experimental Teratology of Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
31
|
Berben L, Floris G, Wildiers H, Hatse S. Cancer and Aging: Two Tightly Interconnected Biological Processes. Cancers (Basel) 2021; 13:1400. [PMID: 33808654 PMCID: PMC8003441 DOI: 10.3390/cancers13061400] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 02/07/2023] Open
Abstract
Age is one of the main risk factors of cancer; several biological changes linked with the aging process can explain this. As our population is progressively aging, the proportion of older patients with cancer is increasing significantly. Due to the heterogeneity of general health and functional status amongst older persons, treatment of cancer is a major challenge in this vulnerable population. Older patients often experience more side effects of anticancer treatments. Over-treatment should be avoided to ensure an optimal quality of life. On the other hand, under-treatment due to fear of toxicity is a frequent problem and can lead to an increased risk of relapse and worse survival. There is a delicate balance between benefits of therapy and risk of toxicity. Robust biomarkers that reflect the body's biological age may aid in outlining optimal individual treatment regimens for older patients with cancer. In particular, the impact of age on systemic immunity and the tumor immune infiltrate should be considered, given the expanding role of immunotherapy in cancer treatment. In this review, we summarize current knowledge concerning the mechanistic connections between aging and cancer, as well as aging biomarkers that could be helpful in the field of geriatric oncology.
Collapse
Affiliation(s)
- Lieze Berben
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, 3000 Leuven, Belgium;
| | - Giuseppe Floris
- Department of Pathology, University Hospitals Leuven, 3000 Leuven, Belgium;
- Laboratory of Translational Cell and Tissue Research, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Hans Wildiers
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, 3000 Leuven, Belgium;
- Department of General Medical Oncology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Sigrid Hatse
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, 3000 Leuven, Belgium;
| |
Collapse
|