1
|
Lian Q, Liu H, Li J, Luo C, Liu C, Zhao H, Dai P, Wang B, Zhou H, Jiang X, Wang Z, Qiao S. Enhancing radiosensitivity of osteosarcoma by ITGB3 knockdown: a mechanism linked to enhanced osteogenic differentiation status through JNK/c-JUN/RUNX2 pathway activation. J Exp Clin Cancer Res 2025; 44:159. [PMID: 40410897 PMCID: PMC12102912 DOI: 10.1186/s13046-025-03417-4] [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: 12/21/2024] [Accepted: 05/11/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND The prognosis of osteosarcoma has improved little over the past few decades, with radioresistance being a contributing factor. Effective radiosensitizing targets and novel mechanisms for treating osteosarcoma are urgently needed. Research on the impact of regulating differentiation levels on the radiosensitivity of malignant tumors is limited. This study aimed to explore the efficacy of ITGB3 as a novel radiosensitizing target in osteosarcoma and to explore whether the modulation of osteogenic differentiation plays a role in mediating the radiosensitizing effect. METHODS RNA sequencing was utilized to screen for potential targets that affect the radiosensitivity of osteosarcoma. In vitro assays examining cell viability, apoptosis, proliferation, migration, and invasion were conducted to verify the radiosensitizing effect of ITGB3-knockdown (KD). Furthermore, in vivo validation was performed by constructing mouse models with subcutaneous and orthotopic tibial tumors. Rescue experiments involving siRNAs and molecular inhibitors were performed to explore and validate the mechanisms through which ITGB3-KD exerts a radiosensitizing effect in vitro and in vivo. Additionally, osteogenic differentiation cultures of osteosarcoma cells were conducted as auxiliary validation for the radiosensitizing mechanism. RESULTS ITGB3-KD had a radiosensitizing effect on osteosarcoma in vitro by inhibiting cell viability, proliferation, migration, and invasion and promoting apoptosis. ITGB3-KD radiosensitized osteosarcoma in vivo in subcutaneous and orthotopic tibial tumor models. ITGB3-KD upregulated the JNK/c-JUN pathway, and rescue experiments with a JNK inhibitor revealed that the activation of this pathway was crucial for the upregulation of osteogenic markers such as RUNX2, OCN, and OPN, as well as for promoting apoptotic pathways. siRNA-based rescue experiments indicated that the upregulation of RUNX2 mediated the proapoptotic radiosensitizing effects of ITGB3-KD. Culture in osteogenic differentiation medium promoted osteosarcoma radiosensitization by enhancing the osteogenic differentiation status, working synergistically with ITGB3-KD. CONCLUSIONS Our findings indicate that ITGB3-KD enhances radiosensitivity in osteosarcoma by promoting osteogenic differentiation and apoptosis through activation of the JNK/c-JUN/RUNX2 pathway, identifying ITGB3 as a candidate therapeutic target and implicating JNK/c-JUN/RUNX2 signaling as a modulatory axis for improving the response to radiation of osteosarcoma.
Collapse
Affiliation(s)
- Qiujian Lian
- Department of Orthopedics, Fuzhou Second General Hospital, Fuzhou, Fujian, 350007, China
- Department of Orthopedics, The Third Affiliated Hospital of Naval Medical University, Shanghai, 201805, China
| | - Hu Liu
- Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Jingyan Li
- Department of Geriatric Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Cheng Luo
- Department of Orthopedics, The Third Affiliated Hospital of Naval Medical University, Shanghai, 201805, China
| | - Chang Liu
- Department of Orthopedics, The 900th Hospital of Joint Logistic Support Force, Fuzhou, Fujian, 350025, China
| | - Haonan Zhao
- Department of Orthopedics, The Third Affiliated Hospital of Naval Medical University, Shanghai, 201805, China
| | - Peijun Dai
- Department of Orthopedics, The Third Affiliated Hospital of Naval Medical University, Shanghai, 201805, China
| | - Bingxuan Wang
- Department of Orthopedics, The Third Affiliated Hospital of Naval Medical University, Shanghai, 201805, China
| | - Huipeng Zhou
- Department of Orthopedics, The Third Affiliated Hospital of Naval Medical University, Shanghai, 201805, China
| | - Xin Jiang
- Department of Anesthesiology, The Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China.
| | - Zhiwei Wang
- Department of Orthopedics, The Third Affiliated Hospital of Naval Medical University, Shanghai, 201805, China.
| | - Suchi Qiao
- Department of Orthopedics, The Third Affiliated Hospital of Naval Medical University, Shanghai, 201805, China.
| |
Collapse
|
2
|
Shen S, Wang S, Zhou D, Wu X, Gao M, Wu J, Yang Y, Pan X, Wang N. A clinician's perspective on boron neutron capture therapy: promising advances, ongoing trials, and future outlook. Int J Radiat Biol 2024; 100:1126-1142. [PMID: 38986056 DOI: 10.1080/09553002.2024.2373746] [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: 01/09/2024] [Revised: 05/15/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024]
Abstract
PURPOSE This comprehensive review aims to provide a unique clinical perspective on the latest advances and ongoing boron neutron capture therapy (BNCT) trials for various cancers. METHODS We critically analyzed clinical data from BNCT trials for head and neck cancer, glioblastoma, melanoma, meningioma, breast cancer, and liver tumors. We investigated differences in tumor responses and normal tissue toxicities among trials and discussed potential contributing factors. We also identified the limitations of early BNCT trials and proposed strategies to optimize future trial design. RESULTS BNCT has shown promising results in treating head and neck cancer, with high response rates and improved survival in patients with recurrent disease. In glioblastoma, BNCT combined with surgery and chemotherapy has demonstrated survival benefits compared to standard treatments. BNCT has also been successfully used for recurrent high-grade meningiomas and shows potential for melanomas, extramammary Paget's disease, and liver tumors. However, differences in tumor responses and toxicities were observed among trials, potentially attributable to variations in treatment protocols, patient characteristics, and evaluation methods. CONCLUSIONS BNCT is a promising targeted radiotherapy for various cancers. Further optimization and well-designed randomized controlled trials are needed to establish its efficacy and safety. Future studies should focus on standardizing treatment protocols and addressing limitations to guide clinical decision-making and research priorities.
Collapse
Affiliation(s)
- Shumin Shen
- Department of Oncology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Shanghu Wang
- Department of Radiotherapy, Anhui Chest Hospital, Hefei, China
| | - Dachen Zhou
- Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei, China
| | - Xiuwei Wu
- Department of Oncology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Mingzhu Gao
- Department of Oncology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Jinjin Wu
- Department of Oncology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Yucai Yang
- Department of Oncology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Xiaoxi Pan
- Department of Nuclear Medicine, The Second Hospital of Anhui Medical University, Hefei, China
| | - Nianfei Wang
- Department of Oncology, The Second Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
3
|
Huang L, Zhu Y, Kong Q, Guan X, Lei X, Zhang L, Yang H, Yao X, Liang S, An X, Yu J. Inhibition of Integrin α vβ 3-FAK-MAPK signaling constrains the invasion of T-ALL cells. Cell Adh Migr 2023; 17:1-14. [PMID: 36944577 PMCID: PMC10038045 DOI: 10.1080/19336918.2023.2191913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023] Open
Abstract
The role of adhesion receptor integrin αvβ3 in T-ALL was unclear. Firstly, we performed quantitative real-time PCR to assess medullary expression of integrin β3(ITGB3) in T-ALL patients and high ITGB3 expression was relevant with the central nervous system leukemia(CNSL) incidence. Decreasing of cell invasion was observed in Jurkat and Molt4 treated with integrin αvβ3 specific antibody and inhibitor as well as cells with ITGB3 interference. Further, phosphorylation of FAK, cRAF, MEK and ERK decreased in cells with integrin αvβ3 inhibition or interference. Invasion decreased in T-ALL cells treated with FAK and ERK inhibitors. In conclusion, inhibition of integrin αvβ3 signals significantly limits the cell invasion of T-ALL cells.
Collapse
Affiliation(s)
- Lan Huang
- Department of hematology and oncology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yao Zhu
- Department of hematology and oncology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Qinglin Kong
- Department of Hematology and Oncology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xianmin Guan
- Department of hematology and oncology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xiaoying Lei
- Department of hematology and oncology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Luying Zhang
- Department of hematology and oncology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Hui Yang
- Department of hematology and oncology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xinyuan Yao
- Department of hematology and oncology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Shaoyan Liang
- Department of hematology and oncology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xizhou An
- Department of hematology and oncology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Jie Yu
- Department of hematology and oncology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
4
|
Im H, Lee J, Lee HJ, Kim DY, Kim EJ, Yi JY. Cyclin D1 promotes radioresistance through regulation of RAD51 in melanoma. Exp Dermatol 2023; 32:1706-1716. [PMID: 37421206 DOI: 10.1111/exd.14877] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 06/22/2023] [Accepted: 06/23/2023] [Indexed: 07/10/2023]
Abstract
Melanoma is a notoriously radioresistant type of skin cancer. Elucidation of the specific mechanisms underlying radioresistance is necessary to improve the clinical efficacy of radiation therapy. To identify the key factors contributing to radioresistance, five melanoma cell lines were selected for study and genes that were upregulated in relatively radioresistant melanomas compared with radiosensitive melanoma cells determined via RNA sequencing technology. In particular, we focused on cyclin D1 (CCND1), a well known cell cycle regulatory molecule. In radiosensitive melanoma, overexpression of cyclin D1 reduced apoptosis. In radioresistant melanoma cell lines, suppression of cyclin D1 with a specific inhibitor or siRNA increased apoptosis and decreased cell proliferation in 2D and 3D spheroid cultures. In addition, we observed increased expression of γ-H2AX, a molecular marker of DNA damage, even at a later time after γ-irradiation, under conditions of inhibition of cyclin D1, with a response pattern similar to that of radiosensitive SK-Mel5. In the same context, expression and nuclear foci formation of RAD51, a key enzyme for homologous recombination (HR), were reduced upon inhibition of cyclin D1. Downregulation of RAD51 also reduced cell survival to irradiation. Overall, suppression of cyclin D1 expression or function led to reduced radiation-induced DNA damage response (DDR) and triggered cell death. Our collective findings indicate that the presence of increased cyclin D1 potentially contributes to the development of radioresistance through effects on RAD51 in melanoma and could therefore serve as a therapeutic target for improving the efficacy of radiation therapy.
Collapse
Affiliation(s)
- Hyuntaik Im
- Division of Basic Radiation Bioscience, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
- Department of Life Science, University of Seoul, Seoul, South Korea
| | - Jeeyong Lee
- Division of Basic Radiation Bioscience, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Hae Jin Lee
- Division of Basic Radiation Bioscience, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Da Yeon Kim
- Division of Basic Radiation Bioscience, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Eun Ju Kim
- Division of Basic Radiation Bioscience, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Jae Youn Yi
- Division of Basic Radiation Bioscience, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| |
Collapse
|
5
|
Integrin αvβ3 Is a Master Regulator of Resistance to TKI-Induced Ferroptosis in HER2-Positive Breast Cancer. Cancers (Basel) 2023; 15:cancers15041216. [PMID: 36831558 PMCID: PMC9954089 DOI: 10.3390/cancers15041216] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Human epidermal growth factor receptor-2 (HER2)-targeting therapies provide clinical benefits for patients with HER2-positive breast cancer. However, the resistance to monotherapies invariably develops and leads to disease relapse and treatment failure. Previous studies have demonstrated a link between the potency of HER2-targeting tyrosine kinase inhibitors (TKIs) and their ability to induce an iron-dependent form of cell death called ferroptosis. The aim of this study was to understand the mechanisms of resistance to TKI-induced ferroptosis and identify novel approaches to overcome treatment resistance. We used mouse and human HER2-positive models of acquired TKI resistance to demonstrate an intimate link between the resistance to TKIs and to ferroptosis and present the first evidence that the cell adhesion receptor αvβ3 integrin is a critical mediator of resistance to TKI-induced ferroptosis. Our findings indicate that αvβ3 integrin-mediated resistance is associated with the re-wiring of the iron/antioxidant metabolism and persistent activation of AKT signalling. Moreover, using gene manipulation approaches and pharmacological inhibitors, we show that this "αvβ3 integrin addiction" can be targeted to reverse TKI resistance. Collectively, these findings provide critical insights into new therapeutic strategies to improve the treatment of advanced HER2-positive breast cancer patients.
Collapse
|
6
|
Zhao L, Ma X, Li G, Zhao P, Lin H, Ma Y, Li H, Yu J. Downregulation of ITGβ3 in colon adenocarcinoma reveals poor prognosis by affecting genome stability, cell cycle, and the tumor immune microenvironment. Front Oncol 2023; 12:1047648. [PMID: 36741730 PMCID: PMC9895777 DOI: 10.3389/fonc.2022.1047648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 12/12/2022] [Indexed: 01/22/2023] Open
Abstract
Introduction Abnormal expression of integrin subunit beta 3 (ITGβ3), a gene-encoding protein, is related to the occurrence and development of cancers; however, the biological role of ITGβ3 in colon adenocarcinoma (COAD) remains unclear. Methods We used the Cancer Genome Atlas database to obtain the clinical data of patients with COAD, analyzed the mRNA gene clusters related to ITGβ3, and analyzed the interaction signal pathway and interaction protein network of the differentially expressed gene clusters. The results showed that ITGβ3 expression in COAD tumor tissues was significantly downregulated compared with that in paracancerous tissues. Low ITGβ3 expression in tumor tissues is associated with poor overall survival of patients with COAD. In multivariate analysis, stage IV and ITGβ3 low expression were independent prognostic factors. Gene Ontology analysis showed that differentially expressed genes (DEGs) were significantly enriched in leukocyte migration, cell adhesion, and extracellular matrix (ECM) organization. Kyoto Encyclopedia of Genes and Genomes analysis revealed that the DEGs were mainly enriched in ECM-receptor interactions, focal adhesion, and the PI3K-Akt signaling pathway. Protein-protein interaction network analysis revealed the hub and seed genes of the key modules related to ITGβ3. Finally, we analyzed the correlation between TGβ3 and immune-related genes and found that ITGβ3 expression was significantly correlated with tumor purity and infiltration level of dominant immune cells. Discussion These findings indicate that ITGβ3 downregulation in COAD may profoundly affect genome stability and multiple steps of the cell cycle, alter the tumor immune microenvironment, and be related to the prognosis of patients with COAD.
Collapse
Affiliation(s)
- Lei Zhao
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaoting Ma
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Guangxin Li
- Radiation Oncology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Pengfei Zhao
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Haishan Lin
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yingjie Ma
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Huihui Li
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China,*Correspondence: Jing Yu, ; Huihui Li,
| | - Jing Yu
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China,*Correspondence: Jing Yu, ; Huihui Li,
| |
Collapse
|
7
|
Lacombe J, Zenhausern F. Effect of mechanical forces on cellular response to radiation. Radiother Oncol 2022; 176:187-198. [PMID: 36228760 DOI: 10.1016/j.radonc.2022.10.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/08/2022] [Accepted: 10/05/2022] [Indexed: 12/14/2022]
Abstract
While the cellular interactions and biochemical signaling has been investigated for long and showed to play a major role in the cell's fate, it is now also evident that mechanical forces continuously applied to the cells in their microenvironment are as important for tissue homeostasis. Mechanical cues are emerging as key regulators of cellular drug response and we aimed to demonstrate in this review that such effects should also be considered vital for the cellular response to radiation. In order to explore the mechanobiology of the radiation response, we reviewed the main mechanoreceptors and transducers, including integrin-mediated adhesion, YAP/TAZ pathways, Wnt/β-catenin signaling, ion channels and G protein-coupled receptors and showed their implication in the modulation of cellular radiosensitivity. We then discussed the current studies that investigated a direct effect of mechanical stress, including extracellular matrix stiffness, shear stress and mechanical strain, on radiation response of cancer and normal cells and showed through preliminary results that such stress effectively can alter cell response after irradiation. However, we also highlighted the limitations of these studies and emphasized some of the contradictory data, demonstrating that the effect of mechanical cues could involve complex interactions and potential crosstalk with numerous cellular processes also affected by irradiation. Overall, mechanical forces alter radiation response and although additional studies are required to deeply understand the underlying mechanisms, these effects should not be neglected in radiation research as they could reveal new fundamental knowledge for predicting radiosensitivity or understanding resistance to radiotherapy.
Collapse
Affiliation(s)
- Jerome Lacombe
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, 475 North 5th Street, Phoenix, AZ 85004, USA; Department of Basic Medical Sciences, College of Medicine Phoenix, University of Arizona, 425 N 5th St, Phoenix, AZ 85004, USA.
| | - Frederic Zenhausern
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, 475 North 5th Street, Phoenix, AZ 85004, USA; Department of Basic Medical Sciences, College of Medicine Phoenix, University of Arizona, 425 N 5th St, Phoenix, AZ 85004, USA; Department of Biomedical Engineering, College of Engineering, University of Arizona, 1127 E. James E. Rogers Way, Tucson, AZ 85721, USA.
| |
Collapse
|
8
|
Krayem M, Ghanem GE, Van Gestel D. Recent advances in radiosensitivity determinants in melanoma. Curr Opin Oncol 2022; 34:131-138. [PMID: 35013044 DOI: 10.1097/cco.0000000000000818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Radiotherapy has been proven to be useful but insufficient in melanoma management due to the intrinsic radioresistance of melanoma cells. Elucidation of the molecular mechanisms and pathways related to resistance/sensitivity to radiotherapy in melanoma is of paramount importance. In this review, we will summarize and discuss the recent 'discoveries' and advances in radiosensitivity determinants in melanoma. RECENT FINDINGS The different levels of radiosensitivity among the various melanoma tumors could be attributed to the DNA damage signaling and repair proteins, tumor microenvironment, hypoxia, cell metabolism, glutathione and redox balance, protein kinase signaling pathways as well as pigmentation and melanin content. SUMMARY It is therapeutically important to elucidate the factors involved in radiation resistance/sensitivity of melanoma. More importantly, improving radiosensitivity may 'widen the clinical utility' in melanoma of this important therapeutic modality.
Collapse
Affiliation(s)
- Mohammad Krayem
- Laboratory of Clinical and Experimental Oncology (LOCE), Institut Jules Bordet
- Department of Radiation Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Ghanem E Ghanem
- Laboratory of Clinical and Experimental Oncology (LOCE), Institut Jules Bordet
| | - Dirk Van Gestel
- Department of Radiation Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
9
|
Pan X, Yi M, Liu C, Jin Y, Liu B, Hu G, Yuan X. Cilengitide, an αvβ3-integrin inhibitor, enhances the efficacy of anti-programmed cell death-1 therapy in a murine melanoma model. Bioengineered 2022; 13:4557-4572. [PMID: 35142593 PMCID: PMC8974133 DOI: 10.1080/21655979.2022.2029236] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Integrins play an important role in multiple stages of tumor progression and metastasis. Previous studies have shown synergistic effects of combined αvβ6-integrin and αvβ8-integrin inhibitors with immunotherapy. However, the role of αvβ3-integrin inhibitor in tumor immunity is still unclear. In this study, we aimed to elucidate the impact of the αvβ3-integrin inhibitor on PD-L1 expression and sensitivity to immune checkpoint blockade in melanoma. We investigated the effects of cilengitide, an αvβ3-integrin inhibitor, on cell viability and apoptosis of melanoma cell lines. And we explored how cilengitide regulated the expression of PD-L1 in melanoma cells in vitro and in vivo, using immunofluorescence, flow cytometry, Western blotting, and immunohistochemistry. A subcutaneous B16 murine melanoma model was utilized to determine whether combining cilengitide with anti-PD1 therapy inhibited tumor growth and positively regulated tumor microenvironment (TME). Our results showed that cilengitide inhibited cell viability and induced apoptosis in B16 and A375 cell lines. Furthermore, cilengitide decreased PD-L1 expression by reducing STAT3 phosphorylation in two melanoma cell lines. Cilengitide also reduced subcutaneous tumor PD-L1 expression in the B16 murine melanoma model. Accordingly, cilengitide positively regulated antitumor immune responses and provided durable therapy when combined with anti-PD1 monoclonal antibody in the murine melanoma model. This combination therapy reduced tumor growth and extended survival. Our study highlights that cilengitide enhances the efficacy of anti-PD1 therapy and produces a stronger antitumor immune response. This combination therefore represents a novel therapeutic regimen that may improve immunotherapy treratment.
Collapse
Affiliation(s)
- Xin Pan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Minxiao Yi
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Chaofan Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yu Jin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Bo Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Guangyuan Hu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
10
|
Manoukian P, Bijlsma M, van Laarhoven H. The Cellular Origins of Cancer-Associated Fibroblasts and Their Opposing Contributions to Pancreatic Cancer Growth. Front Cell Dev Biol 2021; 9:743907. [PMID: 34646829 PMCID: PMC8502878 DOI: 10.3389/fcell.2021.743907] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 08/30/2021] [Indexed: 12/17/2022] Open
Abstract
Pancreatic tumors are known to harbor an abundant and highly desmoplastic stroma. Among the various cell types that reside within tumor stroma, cancer-associated fibroblasts (CAFs) have gained a lot of attention in the cancer field due to their contributions to carcinogenesis and tumor architecture. These cells are not a homogeneous population, but have been shown to have different origins, phenotypes, and contributions. In pancreatic tumors, CAFs generally emerge through the activation and/or recruitment of various cell types, most notably resident fibroblasts, pancreatic stellate cells (PSCs), and tumor-infiltrating mesenchymal stem cells (MSCs). In recent years, single cell transcriptomic studies allowed the identification of distinct CAF populations in pancreatic tumors. Nonetheless, the exact sources and functions of those different CAF phenotypes remain to be fully understood. Considering the importance of stromal cells in pancreatic cancer, many novel approaches have aimed at targeting the stroma but current stroma-targeting therapies have yielded subpar results, which may be attributed to heterogeneity in the fibroblast population. Thus, fully understanding the roles of different subsets of CAFs within the stroma, and the cellular dynamics at play that contribute to heterogeneity in CAF subsets may be essential for the design of novel therapies and improving clinical outcomes. Fortunately, recent advances in technologies such as microfluidics and bio-printing have made it possible to establish more advanced ex vivo models that will likely prove useful. In this review, we will present the different roles of stromal cells in pancreatic cancer, focusing on CAF origin as a source of heterogeneity, and the role this may play in therapy failure. We will discuss preclinical models that could be of benefit to the field and that may contribute to further clinical development.
Collapse
Affiliation(s)
- Paul Manoukian
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Maarten Bijlsma
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Hanneke van Laarhoven
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
11
|
Smart JA, Oleksak JE, Hartsough EJ. Cell Adhesion Molecules in Plasticity and Metastasis. Mol Cancer Res 2021; 19:25-37. [PMID: 33004622 PMCID: PMC7785660 DOI: 10.1158/1541-7786.mcr-20-0595] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/08/2020] [Accepted: 09/25/2020] [Indexed: 12/12/2022]
Abstract
Prior to metastasis, modern therapeutics and surgical intervention can provide a favorable long-term survival for patients diagnosed with many types of cancers. However, prognosis is poor for patients with metastasized disease. Melanoma is the deadliest form of skin cancer, yet in situ and localized, thin melanomas can be biopsied with little to no postsurgical follow-up. However, patients with metastatic melanoma require significant clinical involvement and have a 5-year survival of only 34% to 52%, largely dependent on the site of colonization. Melanoma metastasis is a multi-step process requiring dynamic changes in cell surface proteins regulating adhesiveness to the extracellular matrix (ECM), stroma, and other cancer cells in varied tumor microenvironments. Here we will highlight recent literature to underscore how cell adhesion molecules (CAM) contribute to melanoma disease progression and metastasis.
Collapse
Affiliation(s)
- Jessica A Smart
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Julia E Oleksak
- Graduate School of Biomedical Sciences and Professional Studies, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Edward J Hartsough
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania.
| |
Collapse
|