1
|
Lin F, Yin S, Zhang Z, Yu Y, Fang H, Liang Z, Zhu R, Zhou H, Li J, Cao K, Guo W, Qin S, Zhang Y, Lu C, Li H, Liu S, Zhang H, Ye B, Lin J, Li Y, Kang X, Xi JJ, Chen PR. Multimodal targeting chimeras enable integrated immunotherapy leveraging tumor-immune microenvironment. Cell 2024:S0092-8674(24)01198-X. [PMID: 39504957 DOI: 10.1016/j.cell.2024.10.016] [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: 07/13/2022] [Revised: 08/02/2024] [Accepted: 10/11/2024] [Indexed: 11/08/2024]
Abstract
Although immunotherapy has revolutionized cancer treatment, its efficacy is affected by multiple factors, particularly those derived from the complexity and heterogeneity of the tumor-immune microenvironment (TIME). Strategies that simultaneously and synergistically engage multiple immune cells in TIME remain highly desirable but challenging. Herein, we report a multimodal and programmable platform that enables the integration of multiple therapeutic modules into single agents for tumor-targeted co-engagement of multiple immune cells within TIME. We developed the triple orthogonal linker (T-Linker) technology to integrate various therapeutic small molecules and biomolecules as multimodal targeting chimeras (Multi-TACs). The EGFR-CD3-PDL1 Multi-TAC facilitated T-dendritic cell co-engagement to target solid tumors with excellent efficacy, as demonstrated in vitro, in several humanized mouse models and in patient-derived tumor models. Furthermore, Multi-TACs were constructed to coordinate T cells with other immune cell types. The highly modular and programmable feature of our Multi-TACs may find broad applications in immunotherapy and beyond.
Collapse
Affiliation(s)
- Feng Lin
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China; Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Shenyi Yin
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Zijian Zhang
- National Resource Center for Mutant Mice, MOE Key Laboratory of Model Animals for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, Medical School of Nanjing University, Nanjing 210061, China
| | - Ying Yu
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Haoming Fang
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Zhen Liang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Rujie Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Haitao Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Jianjie Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Medical Oncology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Kunxia Cao
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Weiming Guo
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Shan Qin
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Yuxuan Zhang
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Chenghao Lu
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Han Li
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Shibo Liu
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Heng Zhang
- Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Buqing Ye
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Jian Lin
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China; Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China.
| | - Yan Li
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210061, China; National Resource Center for Mutant Mice, MOE Key Laboratory of Model Animals for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, Medical School of Nanjing University, Nanjing 210061, China.
| | - Xiaozheng Kang
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, China.
| | - Jianzhong Jeff Xi
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China.
| | - Peng R Chen
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China; Shenzhen Bay Laboratory, Shenzhen 518055, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China.
| |
Collapse
|
2
|
Omar MH, Emam SH, Mikhail DS, Elmeligie S. Combretastatin A-4 based compounds as potential anticancer agents: A review. Bioorg Chem 2024; 153:107930. [PMID: 39504638 DOI: 10.1016/j.bioorg.2024.107930] [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: 06/11/2024] [Revised: 10/13/2024] [Accepted: 10/27/2024] [Indexed: 11/08/2024]
Abstract
The current review discusses the importance of combretastatin A-4 (CA-4) as a lead compound of microtubule targeting agents. CA-4 holds a unique place among naturally occurring compounds having cytotoxic activity. In this review an overall picture of design strategies, structure-activity relationship, synthesis, cytotoxic activity, and binding interactions of promising CA-4 analogues, are discussed and arranged chronologically from 2016 to early 2023. Also, this review emphasizes their biological activity as anticancer agents, within an overview of clinical application limitation and suggested strategies to overcome. Dual targeting tubulin inhibitors showed highpotentialto surpass medication resistance and provide synergistic efficacy. Linking platinum (IV), amino acids, and HDAC targeting moieties to active tubulin inhibitorsproduced potent active compounds. Analogues of CA-4 bridged with azetidin-2-one, pyrazole, sulfide, or carrying selenium atom exhibited cytotoxic action against a variety of malignant cell lines through different pathways.
Collapse
Affiliation(s)
- Mai H Omar
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| | - Soha H Emam
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Demiana S Mikhail
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Salwa Elmeligie
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| |
Collapse
|
3
|
Berlanga-Acosta J, Garcia-Ojalvo A, Fernández-Montequin J, Falcon-Cama V, Acosta-Rivero N, Guillen-Nieto G, Pujol-Ferrer M, Limonta-Fernandez M, Ayala-Avila M, Eriksson E. Epidermal Growth Factor Intralesional Delivery in Chronic Wounds: The Pioneer and Standalone Technique for Reversing Wound Chronicity and Promoting Sustainable Healing. Int J Mol Sci 2024; 25:10883. [PMID: 39456666 PMCID: PMC11507032 DOI: 10.3390/ijms252010883] [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: 06/11/2024] [Revised: 09/16/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
The early expectations about growth factors' (GFs') discovery as an undisputed therapeutic solution for chronic wounds progressively eclipsed when they failed to accelerate acute wound closure and restore the healing trajectory of stagnant ulcers. Critical knowledge about chronic wound biology and GF pharmacology was a conundrum at that time. Diabetes undermines keratinocytes' and fibroblasts' physiology, impairing skin healing abilities. Diabetic ulcers, as other chronic wounds, are characterized by hyperinflammation, unbalanced proteolytic activity, catabolism, and free radical cytotoxicity. This hostile scenario for the chemical stability, integrity, and functionality of GFs led to the conclusion that topical administration may jeopardize GFs' clinical effectiveness. Epidermal growth factor (EGF) has a proximal position in tissues homeostasis by activating survival and mitogenic pathways from embryonic life to adulthood. Seminal experiments disclosed unprecedented pharmacological bounties of parenterally administered EGF. Accordingly, the experience accumulated for more than 20 years of EGF intralesional infiltration of diabetic wound bottoms and edges has translated into sustained healing responses, such as low recurrences and amputation rates. This delivery route, in addition to being safe and tolerated, has shown to restore a variety of circulating biochemical markers ordinarily disturbed in diabetic conditions. EGF infiltration triggers a cascade of local fibroblast reactions, supporting its molecular integrity, prolonged mean residence time, and ultimately eliciting its receptor trafficking and nuclear translocation. The intralesional delivery route seems to warrant that EGF reaches wound fibroblasts' epigenetic core, mitigating the consequences of metabolic memory imprinting.
Collapse
Affiliation(s)
- Jorge Berlanga-Acosta
- Wound Healing, Tissue Repair and Cytoprotection Research Project, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology, Playa, P.O. Box 6162, Havana 11600, Cuba; (A.G.-O.); (V.F.-C.); (N.A.-R.); (G.G.-N.); (M.P.-F.); (M.L.-F.); (M.A.-A.)
| | - Ariana Garcia-Ojalvo
- Wound Healing, Tissue Repair and Cytoprotection Research Project, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology, Playa, P.O. Box 6162, Havana 11600, Cuba; (A.G.-O.); (V.F.-C.); (N.A.-R.); (G.G.-N.); (M.P.-F.); (M.L.-F.); (M.A.-A.)
| | - Jose Fernández-Montequin
- National Institute of Angiology and Vascular Surgery—Diabetic Angiopathy Service, Calzada del Cerro 1551 esq, Domínguez, Cerro, Havana 12000, Cuba;
| | - Viviana Falcon-Cama
- Wound Healing, Tissue Repair and Cytoprotection Research Project, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology, Playa, P.O. Box 6162, Havana 11600, Cuba; (A.G.-O.); (V.F.-C.); (N.A.-R.); (G.G.-N.); (M.P.-F.); (M.L.-F.); (M.A.-A.)
| | - Nelson Acosta-Rivero
- Wound Healing, Tissue Repair and Cytoprotection Research Project, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology, Playa, P.O. Box 6162, Havana 11600, Cuba; (A.G.-O.); (V.F.-C.); (N.A.-R.); (G.G.-N.); (M.P.-F.); (M.L.-F.); (M.A.-A.)
| | - Gerardo Guillen-Nieto
- Wound Healing, Tissue Repair and Cytoprotection Research Project, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology, Playa, P.O. Box 6162, Havana 11600, Cuba; (A.G.-O.); (V.F.-C.); (N.A.-R.); (G.G.-N.); (M.P.-F.); (M.L.-F.); (M.A.-A.)
| | - Merardo Pujol-Ferrer
- Wound Healing, Tissue Repair and Cytoprotection Research Project, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology, Playa, P.O. Box 6162, Havana 11600, Cuba; (A.G.-O.); (V.F.-C.); (N.A.-R.); (G.G.-N.); (M.P.-F.); (M.L.-F.); (M.A.-A.)
| | - Miladys Limonta-Fernandez
- Wound Healing, Tissue Repair and Cytoprotection Research Project, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology, Playa, P.O. Box 6162, Havana 11600, Cuba; (A.G.-O.); (V.F.-C.); (N.A.-R.); (G.G.-N.); (M.P.-F.); (M.L.-F.); (M.A.-A.)
| | - Marta Ayala-Avila
- Wound Healing, Tissue Repair and Cytoprotection Research Project, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology, Playa, P.O. Box 6162, Havana 11600, Cuba; (A.G.-O.); (V.F.-C.); (N.A.-R.); (G.G.-N.); (M.P.-F.); (M.L.-F.); (M.A.-A.)
| | - Elof Eriksson
- Joseph E. Murray Professor of Plastic and Reconstructive Surgery, Brigham and Women’s Hospital, Harvard Medical School, Main Pike, ASB-2, 75 Francis St, Boston, MA 02115, USA;
| |
Collapse
|
4
|
Cuan X, Yang X, Wang J, Sheng J, Wang X, Huang Y. Discovery of flavonoid-containing compound Lupalbigenin as anti-NSCLC cancer agents via suppression of EGFR and ERK1/2 pathway. Bioorg Chem 2024; 153:107808. [PMID: 39288634 DOI: 10.1016/j.bioorg.2024.107808] [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: 06/18/2024] [Revised: 08/22/2024] [Accepted: 09/03/2024] [Indexed: 09/19/2024]
Abstract
Epidermal growth factor receptor exon 20 insertions (EGFR Ex20ins) driver mutations in non-small cell lung cancer (NSCLC) is insensitive to EGFR tyrosine kinase inhibitors (TKIs). Therefore, it is necessary to develop more novel strategy to address the limitations of existing therapies targeting EGFR-mutated NSCLC. Lupalbigenin (LB), a flavonoid compound extracted from Derris scandens, has shown preclinical activity in lung cancer. However, the activity of LB in Ex20ins-driven tumors has not yet been elucidated. In this study, a series of stable BaF/3 cell-line that contains a high proportion (>90 %) of EGFR-eGFP Ex20ins were generated using an IL3-deprivation method. Ba/F3 cell models harboring dissimilar Ex20ins were used to characterize the antineoplastic mechanism of LB. Molecular docking confirmed that the LB could effectively bind to key target EGFR. The in vitro anticancer activity of LB was investigated in engineered Ba/F3 cells bearing diverse uncommon EGFR mutations. LB was shown to be more potent in inhibiting the viability of various uncommon EGFR-mutated cell lines. Mechanistic studies disclosed that LB repressed EGFR phosphorylation and downstream survival pathways in Ba/F3 cells expressing EGFR Ex20ins, resulting in caspase activation by activating the intrinsic apoptotic pathway. Further analyses showed that LB significantly induced G0/G1 cell cycle arrest and apoptosis in cells. LB also reduced the protein expression levels of CDK4, CDK6, CDK8, cyclin D1, cyclin A2, and Bcl2 and promoted the expression of cytochrome C, p27, and p53. In summary, we explored the possible potential targets of LB through network pharmacology and verified the target using in vitro experiments. Furthermore, our results demonstrated that LB showed potential anti-Ex20ins cancer activity through suppression of the EGFR and ERK1/2 signaling pathway in Ba/F3 cells bearing two to three amino acid insertion mutations. These findings suggested that LB might be valuable for further investigation as a potential candidate in the treatment of associated diseases.
Collapse
Affiliation(s)
- Xiangdan Cuan
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China; Sanmenxia Polytechnic, Sanmenxia, China
| | - Xingying Yang
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China; College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Jinxian Wang
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China; College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Jun Sheng
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China; State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Kunming, China.
| | - Xuanjun Wang
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China.
| | - Yanping Huang
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China; College of Science, Yunnan Agricultural University, Kunming, China.
| |
Collapse
|
5
|
Li S, Luo X, Sun M, Wang Y, Zhang Z, Jiang J, Hu D, Zhang J, Wu Z, Wang Y, Huang W, Xia L. Context-dependent T-BOX transcription factor family: from biology to targeted therapy. Cell Commun Signal 2024; 22:350. [PMID: 38965548 PMCID: PMC11225425 DOI: 10.1186/s12964-024-01719-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024] Open
Abstract
T-BOX factors belong to an evolutionarily conserved family of transcription factors. T-BOX factors not only play key roles in growth and development but are also involved in immunity, cancer initiation, and progression. Moreover, the same T-BOX molecule exhibits different or even opposite effects in various developmental processes and tumor microenvironments. Understanding the multiple roles of context-dependent T-BOX factors in malignancies is vital for uncovering the potential of T-BOX-targeted cancer therapy. We summarize the physiological roles of T-BOX factors in different developmental processes and their pathological roles observed when their expression is dysregulated. We also discuss their regulatory roles in tumor immune microenvironment (TIME) and the newly arising questions that remain unresolved. This review will help in systematically and comprehensively understanding the vital role of the T-BOX transcription factor family in tumor physiology, pathology, and immunity. The intention is to provide valuable information to support the development of T-BOX-targeted therapy.
Collapse
Affiliation(s)
- Siwen Li
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Xiangyuan Luo
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Mengyu Sun
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Yijun Wang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Zerui Zhang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Junqing Jiang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Dian Hu
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Jiaqian Zhang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Zhangfan Wu
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Yufei Wang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Wenjie Huang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Huazhong University of Science and Technology, Ministry of Education and Ministry of Public Health, Wuhan, Hubei, 430030, China.
| | - Limin Xia
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
6
|
Sabatelle RC, Colson YL, Sachdeva U, Grinstaff MW. Drug Delivery Opportunities in Esophageal Cancer: Current Treatments and Future Prospects. Mol Pharm 2024; 21:3103-3120. [PMID: 38888089 PMCID: PMC11331583 DOI: 10.1021/acs.molpharmaceut.4c00246] [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] [Indexed: 06/20/2024]
Abstract
With one of the highest mortality rates of all malignancies, the 5-year survival rate for esophageal cancer is under 20%. Depending on the stage and extent of the disease, the current standard of care treatment paradigm includes chemotherapy or chemoradiotherapy followed by surgical esophagogastrectomy, with consideration for adjuvant immunotherapy for residual disease. This regimen has high morbidity, due to anatomic changes inherent in surgery, the acuity of surgical complications, and off-target effects of systemic chemotherapy and immunotherapy. We begin with a review of current treatments, then discuss new and emerging targets for therapies and advanced drug delivery systems. Recent and ongoing preclinical and early clinical studies are evaluating traditional tumor targets (e.g., human epidermal growth factor receptor 2), as well as promising new targets such as Yes-associated protein 1 or mammalian target of rapamycin to develop new treatments for this disease. Due the function and location of the esophagus, opportunities also exist to pair these treatments with a drug delivery strategy to increase tumor targeting, bioavailability, and intratumor concentrations, with the two most common delivery platforms being stents and nanoparticles. Finally, early results with antibody drug conjugates and chimeric antigenic receptor T cells show promise as upcoming therapies. This review discusses these innovations in therapeutics and drug delivery in the context of their successes and failures, with the goal of identifying those solutions that demonstrate the most promise to shift the paradigm in treating this deadly disease.
Collapse
Affiliation(s)
- Robert C. Sabatelle
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston, MA, 02215, USA
| | - Yolonda L. Colson
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Uma Sachdeva
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Mark W. Grinstaff
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston, MA, 02215, USA
| |
Collapse
|
7
|
Lu W, Liu L, Kang X, Ren K, Huang Y, Cheng M, Li X, Xu F, Xu X. Combined treatment with cetuximab and STA9090 has synergistic anticancer effects on human non-small cell lung cancer. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1022-1033. [PMID: 38818581 PMCID: PMC11322868 DOI: 10.3724/abbs.2024069] [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: 12/08/2023] [Accepted: 02/02/2024] [Indexed: 06/01/2024] Open
Abstract
Cetuximab (CET), a human murine chimeric IgG monoclonal antibody and an inhibitor of epidermal growth factor receptor (EGFR), has been shown to be effective in treating various types of cancer. However, its use is hindered by limitations such as resistance development, variability in patient response, side effects, and challenges in biomarker identification. Therefore, CET is often combined with other targeted therapies or chemotherapies to enhance its effectiveness. In this study, we investigate the anticancer effects and underlying mechanisms of the combination of CET, an EGFR inhibitor, and STA9090, an inhibitor of heat shock protein 90 (Hsp90), in both in vitro and in vivo models of non-small cell lung cancer (NSCLC). The results demonstrate significantly stronger effects on NSCLC cells in response to combination therapy than to treatment with either agent alone, indicating that the combination of CET and STA9090 has potential synergistic effects. Additionally, the combination therapy inhibits tumor growth in a xenograft nude mouse model more effectively than treatment with either agent alone, suggesting improved efficacy when used together. Furthermore, the synergistic effects of the combination therapy are likely due to inactivation of the receptor tyrosine kinase (RTK) pathway, which is overly activated in cancer and contributes to tumor growth, angiogenesis, and metastasis. Consequently, our findings suggest that STA9090 has potent direct antitumor activity and synergizes with CET against NSCLC tumors. It is highly likely that these synergistic effects are mediated through RTK pathway inactivation caused by the combination. Therefore, our findings strongly and consistently support the potential synergistic effect of STA9090, an RTK inhibitor, in combination with EGFR-targeting agents.
Collapse
Affiliation(s)
- Wanjun Lu
- Jiangxi Clinical Research Center for Respiratory DiseasesJiangxi Institute of Respiratory DiseaseDepartment of Respiratory and Critical Care Medicinethe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
- The First Clinical Medical CollegeNanchang UniversityNanchang30006China
| | - Lixia Liu
- Department of Occupational Health and Occupational MedicineGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhou510515China
| | - Xiang Kang
- Jiangxi Clinical Research Center for Respiratory DiseasesJiangxi Institute of Respiratory DiseaseDepartment of Respiratory and Critical Care Medicinethe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
- The First Clinical Medical CollegeNanchang UniversityNanchang30006China
| | - Kangkang Ren
- Jiangxi Clinical Research Center for Respiratory DiseasesJiangxi Institute of Respiratory DiseaseDepartment of Respiratory and Critical Care Medicinethe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
- Jiangxi Hospital of China-Japan Friendship HospitalNanchang330052China
| | - Ye Huang
- Jiangxi Clinical Research Center for Respiratory DiseasesJiangxi Institute of Respiratory DiseaseDepartment of Respiratory and Critical Care Medicinethe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
- Jiangxi Hospital of China-Japan Friendship HospitalNanchang330052China
| | - Minzhang Cheng
- Jiangxi Clinical Research Center for Respiratory DiseasesJiangxi Institute of Respiratory DiseaseDepartment of Respiratory and Critical Care Medicinethe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
- Jiangxi Hospital of China-Japan Friendship HospitalNanchang330052China
| | - Xiaolei Li
- Jiangxi Clinical Research Center for Respiratory DiseasesJiangxi Institute of Respiratory DiseaseDepartment of Respiratory and Critical Care Medicinethe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
- Jiangxi Hospital of China-Japan Friendship HospitalNanchang330052China
| | - Fei Xu
- Jiangxi Clinical Research Center for Respiratory DiseasesJiangxi Institute of Respiratory DiseaseDepartment of Respiratory and Critical Care Medicinethe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
- Jiangxi Hospital of China-Japan Friendship HospitalNanchang330052China
| | - Xinping Xu
- Jiangxi Clinical Research Center for Respiratory DiseasesJiangxi Institute of Respiratory DiseaseDepartment of Respiratory and Critical Care Medicinethe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
- Jiangxi Hospital of China-Japan Friendship HospitalNanchang330052China
| |
Collapse
|
8
|
Rinaldi L, Chiuso F, Senatore E, Borzacchiello D, Lignitto L, Iannucci R, Donne RD, Fuggi M, Reale C, Russo F, Russo NA, Giurato G, Rizzo F, Sellitto A, Santangelo M, De Biase D, Paciello O, D'Ambrosio C, Amente S, Garbi C, Dalla E, Scaloni A, Weisz A, Ambrosino C, Insabato L, Feliciello A. Downregulation of praja2 restrains endocytosis and boosts tyrosine kinase receptors in kidney cancer. Commun Biol 2024; 7:208. [PMID: 38379085 PMCID: PMC10879500 DOI: 10.1038/s42003-024-05823-4] [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: 02/15/2023] [Accepted: 01/16/2024] [Indexed: 02/22/2024] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common kidney cancer in the adult population. Late diagnosis, resistance to therapeutics and recurrence of metastatic lesions account for the highest mortality rate among kidney cancer patients. Identifying novel biomarkers for early cancer detection and elucidating the mechanisms underlying ccRCC will provide clues to treat this aggressive malignant tumor. Here, we report that the ubiquitin ligase praja2 forms a complex with-and ubiquitylates the AP2 adapter complex, contributing to receptor endocytosis and clearance. In human RCC tissues and cells, downregulation of praja2 by oncogenic miRNAs (oncomiRs) and the proteasome markedly impairs endocytosis and clearance of the epidermal growth factor receptor (EGFR), and amplifies downstream mitogenic and proliferative signaling. Restoring praja2 levels in RCC cells downregulates EGFR, rewires cancer cell metabolism and ultimately inhibits tumor cell growth and metastasis. Accordingly, genetic ablation of praja2 in mice upregulates RTKs (i.e. EGFR and VEGFR) and induces epithelial and vascular alterations in the kidney tissue.In summary, our findings identify a regulatory loop between oncomiRs and the ubiquitin proteasome system that finely controls RTKs endocytosis and clearance, positively impacting mitogenic signaling and kidney cancer growth.
Collapse
Affiliation(s)
- Laura Rinaldi
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Francesco Chiuso
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Emanuela Senatore
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Domenica Borzacchiello
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Luca Lignitto
- Cancer Research Center of Marseille (CRCM), CNRS, Aix Marseille Univ, INSERM, Institut Paoli-Calmettes, Marseille, France
| | - Rosa Iannucci
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Rossella Delle Donne
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Mariano Fuggi
- Department of Advanced Biomedical Sciences, University Hospital Federico II, Naples, Italy
| | - Carla Reale
- Biogem, Biology and Molecular Genetics Institute, Ariano Irpino, Italy
| | - Filomena Russo
- Biogem, Biology and Molecular Genetics Institute, Ariano Irpino, Italy
| | | | - Giorgio Giurato
- Genome Research Center for Health, Baronissi (SA), Italy
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry SMS, Baronissi (SA), Italy
| | - Francesca Rizzo
- Genome Research Center for Health, Baronissi (SA), Italy
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry SMS, Baronissi (SA), Italy
| | - Assunta Sellitto
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry SMS, Baronissi (SA), Italy
| | - Michele Santangelo
- Department of Advanced Biomedical Sciences, University Hospital Federico II, Naples, Italy
| | - Davide De Biase
- Department of Pharmacy, University of Salerno, Salerno, Italy
| | - Orlando Paciello
- Department of Veterinary Medicine and Animal Production, Pathology Unit, University Federico II, Naples, Italy
| | - Chiara D'Ambrosio
- Proteomics, Metabolomics and Mass Spectrometry Laboratory, ISPAAM, National Research Council, Portici (Naples), Italy
| | - Stefano Amente
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Corrado Garbi
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Emiliano Dalla
- Department of Medicine, University of Udine, Udine, Italy
| | - Andrea Scaloni
- Proteomics, Metabolomics and Mass Spectrometry Laboratory, ISPAAM, National Research Council, Portici (Naples), Italy
| | - Alessandro Weisz
- Genome Research Center for Health, Baronissi (SA), Italy
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry SMS, Baronissi (SA), Italy
| | - Concetta Ambrosino
- Biogem, Biology and Molecular Genetics Institute, Ariano Irpino, Italy
- Department of Science and Technology University of Sannio, Sannio, Italy
| | - Luigi Insabato
- Department of Advanced Biomedical Sciences, University Hospital Federico II, Naples, Italy
| | - Antonio Feliciello
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy.
| |
Collapse
|
9
|
Scheffges C, Devy J, Giustiniani J, Francois S, Cartier L, Merrouche Y, Foussat A, Potteaux S, Bensussan A, Marie-Cardine A. Identification of CD160-TM as a tumor target on triple negative breast cancers: possible therapeutic applications. Breast Cancer Res 2024; 26:28. [PMID: 38360636 PMCID: PMC10870674 DOI: 10.1186/s13058-024-01785-x] [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] [Accepted: 02/12/2024] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND Despite major therapeutic advances, triple-negative breast cancer (TNBC) still presents a worth prognosis than hormone receptors-positive breast cancers. One major issue relies in the molecular and mutational heterogeneity of TNBC subtypes that is reinforced by the absence of reliable tumor-antigen that could serve as a specific target to further promote efficient tumor cell recognition and depletion. CD160 is a receptor mainly expressed by NK lymphocytes and presenting two isoforms, namely the GPI-anchored form (CD160-GPI) and the transmembrane isoform (CD160-TM). While CD160-GPI is constitutively expressed on resting cells and involved in the generation of NK cells' cytotoxic activity, CD160-TM is neo-synthesized upon activation and promotes the amplification of NK cells' killing ability. METHODS CD160 expression was assessed by immunohistochemistry (IHC) and flow cytometry on TNBC patient biopsies or cell lines, respectively. Antibody (Ab)-mediated tumor depletion was tested in vitro by performing antibody-dependent cell cytotoxicity (ADCC) and phagocytosis (ADCP) assays, and in vivo on a TNBC mouse model. RESULTS Preliminary data obtained by IHC on TNBC patients' tumor biopsies revealed an unconventional expression of CD160 by TNBC tumor cells. By using a specific but conformation-dependent anti-CD160-TM Ab, we established that CD160-TM, but not CD160-GPI, was expressed by TNBC tumor cells. A conformation-independent anti-CD160-TM mAb (22B12; muIgG2a isotype) was generated and selected according to pre-defined specificity and functional criterions. In vitro functional assays demonstrated that ADCC and ADCP could be induced in the presence of 22B12, resulting in TNBC cell line apoptosis. The ability of 22B12 to exert an in vivo anti-tumor activity was also demonstrated on a TNBC murine model. CONCLUSIONS Our data identify CD160-TM as a tumor marker for TNBC and provide a rational for the use of anti-CD160-TM antibodies as therapeutic tools in this tumor context.
Collapse
Affiliation(s)
- Claire Scheffges
- INSERM U976, HIPI, Team 1, 75010, Paris, France
- Université Paris Cité, IRSL, 75010, Paris, France
- Alderaan Biotechnology, 75005, Paris, France
| | - Jérôme Devy
- UMR CNRS/URCA 7369, MEDyC, Université de Reims-Champagne-Ardennes, 51100, Reims, France
| | | | | | - Lucille Cartier
- Département de Recherche, Institut Godinot, 51100, Reims, France
- UR7509, IRMAIC, Université de Reims-Champagne-Ardennes, 51097, Reims, France
| | - Yacine Merrouche
- Département de Recherche, Institut Godinot, 51100, Reims, France
- UR7509, IRMAIC, Université de Reims-Champagne-Ardennes, 51097, Reims, France
| | | | - Stéphane Potteaux
- UR7509, IRMAIC, Université de Reims-Champagne-Ardennes, 51097, Reims, France
| | - Armand Bensussan
- INSERM U976, HIPI, Team 1, 75010, Paris, France
- Université Paris Cité, IRSL, 75010, Paris, France
| | - Anne Marie-Cardine
- INSERM U976, HIPI, Team 1, 75010, Paris, France.
- Université Paris Cité, IRSL, 75010, Paris, France.
| |
Collapse
|
10
|
Bozic D, Živanović J, Živančević K, Baralić K, Đukić-Ćosić D. Trends in Anti-Tumor Effects of Pseudomonas aeruginosa Mannose-Sensitive-Hemagglutinin (PA-MSHA): An Overview of Positive and Negative Effects. Cancers (Basel) 2024; 16:524. [PMID: 38339275 PMCID: PMC10854591 DOI: 10.3390/cancers16030524] [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: 12/15/2023] [Revised: 01/13/2024] [Accepted: 01/16/2024] [Indexed: 02/12/2024] Open
Abstract
Cancer is a leading cause of death worldwide, for which finding the optimal therapy remains an ongoing challenge. Drug resistance, toxic side effects, and a lack of specificity pose significant difficulties in traditional cancer treatments, leading to suboptimal clinical outcomes and high mortality rates among cancer patients. The need for alternative therapies is crucial, especially for those resistant to conventional methods like chemotherapy and radiotherapy or for patients where surgery is not possible. Over the past decade, a novel approach known as bacteria-mediated cancer therapy has emerged, offering potential solutions to the limitations of conventional treatments. An increasing number of in vitro and in vivo studies suggest that the subtype of highly virulent Pseudomonas aeruginosa bacterium called Pseudomonas aeruginosa mannose-sensitive-hemagglutinin (PA-MSHA) can successfully inhibit the progression of various cancer types, such as breast, lung, and bladder cancer, as well as hepatocellular carcinoma. PA-MSHA inhibits the growth and proliferation of tumor cells and induces their apoptosis. Proposed mechanisms of action include cell-cycle arrest and activation of pro-apoptotic pathways regulated by caspase-9 and caspase-3. Moreover, clinical studies have shown that PA-MSHA improved the effectiveness of chemotherapy and promoted the activation of the immune response in cancer patients without causing severe side effects. Reported adverse reactions were fever, skin irritation, and pain, attributed to the overactivation of the immune response. This review aims to summarize the current knowledge obtained from in vitro, in vivo, and clinical studies available at PubMed, Google Scholar, and ClinicalTrials.gov regarding the use of PA-MSHA in cancer treatment in order to further elucidate its pharmacological and toxicological properties.
Collapse
Affiliation(s)
- Dragica Bozic
- Department of Toxicology “Akademik Danilo Soldatović”, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (J.Ž.); (K.Ž.); (K.B.); (D.Đ.-Ć.)
| | - Jovana Živanović
- Department of Toxicology “Akademik Danilo Soldatović”, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (J.Ž.); (K.Ž.); (K.B.); (D.Đ.-Ć.)
| | - Katarina Živančević
- Department of Toxicology “Akademik Danilo Soldatović”, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (J.Ž.); (K.Ž.); (K.B.); (D.Đ.-Ć.)
- Center for Laser Microscopy, Faculty of Biology, Institute of Physiology and Biochemistry “Ivan Djaja”, University of Belgrade, Studentski trg 16, 11158 Belgrade, Serbia
| | - Katarina Baralić
- Department of Toxicology “Akademik Danilo Soldatović”, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (J.Ž.); (K.Ž.); (K.B.); (D.Đ.-Ć.)
| | - Danijela Đukić-Ćosić
- Department of Toxicology “Akademik Danilo Soldatović”, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (J.Ž.); (K.Ž.); (K.B.); (D.Đ.-Ć.)
- Center for Toxicological Risk Assessment, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia
| |
Collapse
|
11
|
Hao L, Zhong X, Yu R, Chen J, Li W, Chen Y, Lu W, Wu J, Wang P. Integrating Network Pharmacology and Experimental Validation to Decipher the Anti-Inflammatory Effects of Magnolol on LPS-induced RAW264.7 Cells. Comb Chem High Throughput Screen 2024; 27:462-478. [PMID: 37818577 DOI: 10.2174/0113862073255964230927105959] [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: 04/14/2023] [Revised: 07/11/2023] [Accepted: 08/11/2023] [Indexed: 10/12/2023]
Abstract
INTRODUCTION Magnolol is beneficial against inflammation-mediated damage. However, the underlying mechanisms by which magnolol exerts anti-inflammatory effects on macrophages remain unclear. OBJECTIVE In this study, network pharmacology and experimental validation were used to assess the effect of magnolol on inflammation caused by lipopolysaccharide (LPS) in RAW264.7 cells. MATERIALS AND METHODS Genes related to magnolol were identified in the PubChem and Swiss Target Prediction databases, and gene information about macrophage polarization was retrieved from the GeneCards, OMIM, and PharmGKB databases. Analysis of protein-protein interactions was performed with STRING, and Cytoscape was used to construct a component-target-disease network. GO and KEGG enrichment analyses were performed to ascertain significant molecular biological processes and signaling pathways. LPS was used to construct the inflammatory cell model. ELISA and qRT.PCR were used to examine the expression levels of inflammationassociated factors, immunofluorescence was used to examine macrophage markers (CD86 and CD206), and western blotting was used to examine protein expression levels. RESULTS The hub target genes of magnolol that act on macrophage polarization were MDM2, MMP9, IL-6, TNF, EGFR, AKT1, and ERBB2. The experimental validation results showed that magnolol treatment decreased the levels of proinflammatory factors (TNF-α, IL-1β, and IL-6). Moreover, the levels of anti-inflammatory factors (IL-10 and IL-4) were increased. In addition, magnolol upregulated the expression of M2 markers (Agr-1, Fizzl, and CD206) and downregulated M1 markers (CD86). The cell experiment results supported the network pharmacological results and demonstrated that magnolol alleviated inflammation by modulating the PI3k-Akt and P62/keap1/Nrf2 signaling pathways. CONCLUSION According to network pharmacology and experimental validation, magnolol attenuated inflammation in LPS-induced RAW264.7 cells mainly by inhibiting M1 polarization and enhancing M2 polarization by activating the PI3K/Akt and P62/keap1/Nrf2 signaling pathways.
Collapse
Affiliation(s)
- Lei Hao
- Department of Surgery Two, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoying Zhong
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Runjia Yu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiahui Chen
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Li
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuzhong Chen
- Department of Surgery Two, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weiqi Lu
- Department of Surgery Two, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianyu Wu
- Department of Surgery Two, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peizong Wang
- State Key Laboratory of Oncology in South China, Department of Anesthesiology, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P.R. China
| |
Collapse
|
12
|
Wu H, Ding X, Chen Y, Cai Y, Yang Z, Jin J. EGFR-targeted humanized single chain antibody fragment functionalized silica nanoparticles for precision therapy of cancer. Int J Biol Macromol 2023; 253:127538. [PMID: 37866562 DOI: 10.1016/j.ijbiomac.2023.127538] [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: 05/29/2023] [Revised: 10/06/2023] [Accepted: 10/16/2023] [Indexed: 10/24/2023]
Abstract
The combination of highly specific targeting ability and potent killing effect has made antibody-drug conjugates (ADCs) a popular area of focus in the development of anti-cancer drugs. However, the large molecular weight of IgG antibodies (∼ 150 kDa) often faces challenges in penetrating capillaries and stroma in tumor tissue. Moreover, when the drug-antibody ratio (DAR) is too low (DAR < 2) or too high (DAR > 6) it decreases the effectiveness of the ADC and further increases the potential for aggregation, overall clearance of the early system payload, and release rate. In this study, an EGFR-based single-chain antibody fragment (husA)-human serum albumin (HSA)-coupled FITC-labeled mesoporous silica nanoparticle (FMSN-DOX-H-husA) was developed. Chinese hamster ovarian cells express the husA, which is a single chain antibody fragment of the EGFR that has been humanized. The small molecular weight of the single chain antibody allows for shorter penetration into solid tumors and the absence of adverse effects of the Fc fragment. The modification of HSA improves the safety of the antibody nanoparticle couples by both improving the biocompatibility of the nanoparticles, prolonging the circulation time of the nanoparticles, and avoiding early release of the payload. Also, the humanization substantially reduces the immunogenicity. More importantly, the ratio of drug antibodies on nanoparticles was experimentally and computationally derived to be 11.8, providing a more accurate guide for clinical trials. The results of both in vivo and in vitro experiments indicated promising antitumor activity and safety of FMSN-DOX-H-husA. Thus, this antibody-drug conjugate provided a hopeful option for cancer treatment.
Collapse
Affiliation(s)
- Hao Wu
- School of Life Sciences and Health Engineering, Jiangnan University, 1800 Lihu Avenue, Wuxi 214000, People's Republic of China; School of Chemical & Material Engineering, Jiangnan University, 1800 Lihu Avenue, Wuxi 214000, People's Republic of China
| | - Xuefeng Ding
- School of Biotechnology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214000, People's Republic of China
| | - Yun Chen
- School of Life Sciences and Health Engineering, Jiangnan University, 1800 Lihu Avenue, Wuxi 214000, People's Republic of China
| | - Yanfei Cai
- School of Life Sciences and Health Engineering, Jiangnan University, 1800 Lihu Avenue, Wuxi 214000, People's Republic of China
| | - Zhaoqi Yang
- School of Life Sciences and Health Engineering, Jiangnan University, 1800 Lihu Avenue, Wuxi 214000, People's Republic of China.
| | - Jian Jin
- School of Life Sciences and Health Engineering, Jiangnan University, 1800 Lihu Avenue, Wuxi 214000, People's Republic of China.
| |
Collapse
|
13
|
Li HQ, Wang Q, Zhang LY, Li JY, Wang YJ, Wei L, Yao LG. Hepatic arterial infusion chemotherapy and trastuzumab in gastric cancer with liver metastases: a case report. Front Oncol 2023; 13:1283274. [PMID: 38188301 PMCID: PMC10768022 DOI: 10.3389/fonc.2023.1283274] [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: 08/25/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024] Open
Abstract
Background Gastric cancer is a common cancer worldwide and is responsible for over one million new cases in 2020 and an estimated 769,000 deaths, ranking fifth for incidence and fourth for mortality globally. Incidence rates are highest in Eastern Asia and Eastern Europe. Gastric cancer is highly heterogeneous and progresses rapidly. The prognosis of gastric cancer with liver metastases is poor, and clinical treatment remains challenging. Human epidermal growth factor receptor 2 (HER2) positivity is correlated to a bad prognosis for gastric cancer. Trastuzumab combined with systemic chemotherapy is the preferred treatment for HER2-positive advanced gastric cancer. However, intravenous chemotherapy has severe systemic toxicity, which reduces the local drug concentration and tumor uptake rate, and the effect is unsatisfactory. Case summary We reported a 66-year-old patient with HER2-positive advanced gastric cancer with jaundice due to multiple liver metastases, after 6 cycles of trastuzumab combined with hepatic arterial infusion chemotherapy (HAIC), the tumor retracted significantly, the jaundice subsided, and the patient recovered well. The patient achieved disease control with an intensive regimen followed by less toxic maintenance therapy. Trastuzumab combined with capecitabine maintenance therapy followed up for more than 16 months. Conclusion HAIC plus trastuzumab may be a tolerable treatment option for patients with severe liver metastases from HER2-positive gastric cancer to achieve local control and prolong survival.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Li-ge Yao
- Department of Oncology, The Third People’s Hospital of Zhengzhou, Zhengzhou, China
| |
Collapse
|
14
|
Cao W, Tang Q, Zeng J, Jin X, Zu L, Xu S. A Review of Biomarkers and Their Clinical Impact in Resected Early-Stage Non-Small-Cell Lung Cancer. Cancers (Basel) 2023; 15:4561. [PMID: 37760531 PMCID: PMC10526902 DOI: 10.3390/cancers15184561] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/28/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
The postoperative survival of early-stage non-small-cell lung cancer (NSCLC) patients remains unsatisfactory. In this review, we examined the relevant literature to ascertain the prognostic effect of related indicators on early-stage NSCLC. The prognostic effects of the epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), mesenchymal-epithelial transition (MET), C-ros oncogene 1 (ROS1), or tumour protein p53 (TP53) alterations in resected NSCLC remains debatable. Kirsten rat sarcoma viral oncogene homologue (KRAS) alterations indicate unfavourable outcomes in early-stage NSCLC. Meanwhile, adjuvant or neoadjuvant EGFR-targeted agents can substantially improve prognosis in early-stage NSCLC with EGFR alterations. Based on the summary of current studies, resected NSCLC patients with overexpression of programmed death-ligand 1 (PD-L1) had worsening survival. Conversely, PD-L1 or PD-1 inhibitors can substantially improve patient survival. Considering blood biomarkers, perioperative peripheral venous circulating tumour cells (CTCs) and pulmonary venous CTCs predicted unfavourable prognoses and led to distant metastases. Similarly, patients with detectable perioperative circulating tumour DNA (ctDNA) also had reduced survival. Moreover, patients with perioperatively elevated carcinoembryonic antigen (CEA) in the circulation predicted significantly worse survival outcomes. In the future, we will incorporate mutated genes, immune checkpoints, and blood-based biomarkers by applying artificial intelligence (AI) to construct prognostic models that predict patient survival accurately and guide individualised treatment.
Collapse
Affiliation(s)
- Weibo Cao
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China; (W.C.); (Q.T.); (J.Z.); (X.J.); (L.Z.)
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Quanying Tang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China; (W.C.); (Q.T.); (J.Z.); (X.J.); (L.Z.)
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jingtong Zeng
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China; (W.C.); (Q.T.); (J.Z.); (X.J.); (L.Z.)
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xin Jin
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China; (W.C.); (Q.T.); (J.Z.); (X.J.); (L.Z.)
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Lingling Zu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China; (W.C.); (Q.T.); (J.Z.); (X.J.); (L.Z.)
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Song Xu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China; (W.C.); (Q.T.); (J.Z.); (X.J.); (L.Z.)
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
15
|
Al-Wahaibi LH, Abou-Zied HA, Hisham M, Beshr EAM, Youssif BGM, Bräse S, Hayallah AM, Abdel-Aziz M. Design, Synthesis, and Biological Evaluation of Novel 3-Cyanopyridone/Pyrazoline Hybrids as Potential Apoptotic Antiproliferative Agents Targeting EGFR/BRAF V600E Inhibitory Pathways. Molecules 2023; 28:6586. [PMID: 37764362 PMCID: PMC10537368 DOI: 10.3390/molecules28186586] [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: 08/15/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
A series of novel 3-cyanopyridone/pyrazoline hybrids (21-30) exhibiting dual inhibition against EGFR and BRAFV600E has been developed. The synthesized target compounds were tested in vitro against four cancer cell lines. Compounds 28 and 30 demonstrated remarkable antiproliferative activity, boasting GI50 values of 27 nM and 25 nM, respectively. These hybrids exhibited dual inhibitory effects on both EGFR and BRAFV600E pathways. Compounds 28 and 30, akin to Erlotinib, displayed promising anticancer potential. Compound 30 emerged as the most potent inhibitor against cancer cell proliferation and BRAFV600E. Notably, both compounds 28 and 30 induced apoptosis by elevating levels of caspase-3 and -8 and Bax, while downregulating the antiapoptotic Bcl2 protein. Molecular docking studies confirmed the potential of compounds 28 and 30 to act as dual EGFR/BRAFV600E inhibitors. Furthermore, in silico ADMET prediction indicated that most synthesized 3-cyanopyridone/pyrazoline hybrids exhibit low toxicity and minimal adverse effects.
Collapse
Affiliation(s)
- Lamya H. Al-Wahaibi
- Department of Chemistry, College of Sciences, Princess Nourah bint Abdulrahman University, Riyadh 11564, Saudi Arabia;
| | - Hesham A. Abou-Zied
- Medicinal Chemistry Department, Faculty of Pharmacy, Deraya University, Minia 61111, Egypt; (H.A.A.-Z.); (M.H.)
| | - Mohamed Hisham
- Medicinal Chemistry Department, Faculty of Pharmacy, Deraya University, Minia 61111, Egypt; (H.A.A.-Z.); (M.H.)
| | - Eman A. M. Beshr
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia 61519, Egypt; (E.A.M.B.); (M.A.-A.)
| | - Bahaa G. M. Youssif
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Stefan Bräse
- Institute of Biological and Chemical Systems, IBCS-FMS, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany
| | - Alaa M. Hayallah
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Sphinx University, Assiut 71515, Egypt
| | - Mohamed Abdel-Aziz
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia 61519, Egypt; (E.A.M.B.); (M.A.-A.)
| |
Collapse
|
16
|
Anisuzzman M, Komalla V, Tarkistani MAM, Kayser V. Anti-Tumor Activity of Novel Nimotuzumab-Functionalized Gold Nanoparticles as a Potential Immunotherapeutic Agent against Skin and Lung Cancers. J Funct Biomater 2023; 14:407. [PMID: 37623652 PMCID: PMC10456021 DOI: 10.3390/jfb14080407] [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: 07/04/2023] [Revised: 07/21/2023] [Accepted: 07/30/2023] [Indexed: 08/26/2023] Open
Abstract
The epidermal growth factor receptor (EGFR) is vital for many different types of cancer. Nimotuzumab (NmAb), an anti-EGFR monoclonal antibody (mAb), is used against some of EGFR-overexpressed cancers in various countries. It targets malignant cells and is internalized via receptor-mediated endocytosis. We hypothesized that mAb-nanoparticle conjugation would provide an enhanced therapeutic efficacy, and hence we conjugated NmAb with 27 nm spherical gold nanoparticles (AuNPs) to form AuNP-NmAb nanoconjugates. Using biophysical and spectroscopic methods, including ultraviolet-visible spectroscopy (UV-Vis), transmission electron microscopy (TEM), dynamic light scattering (DLS), nanoparticle tracking analysis (NTA), sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), and Fourier-transform infrared spectroscopy (FTIR), the AuNP-NmAb complex was characterized. Furthermore, in vitro studies were performed using a medium-level EGFR-expressing skin cancer cell (A431, EGFRmedium) and low-level EGFR-expressing lung cancer cell (A549, EGFRlow) to evaluate anti-tumor and cellular uptake efficiency via MTT assay and single-particle inductively coupled plasma mass spectrometry (spICP-MS), respectively. In comparison to NmAb monotherapy, the AuNP-NmAb treatment drastically reduced cancer cell survivability: for A431 cells, the IC50 value of AuNP-NmAb conjugate was 142.7 µg/mL, while the IC50 value of free NmAb was 561.3 µg/mL. For A549 cells, the IC50 value of the AuNP-NmAb conjugate was 163.6 µg/mL, while the IC50 value of free NmAb was 1,082.0 µg/mL. Therefore, this study highlights the unique therapeutic potential of AuNP-NmAb in EGFR+ cancers and shows the potential to develop other mAb nanoparticle complexes for a superior therapeutic efficacy.
Collapse
Affiliation(s)
| | | | | | - Veysel Kayser
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| |
Collapse
|
17
|
Hong L, Li W, Li Y, Yin S. Nanoparticle-based drug delivery systems targeting cancer cell surfaces. RSC Adv 2023; 13:21365-21382. [PMID: 37465582 PMCID: PMC10350659 DOI: 10.1039/d3ra02969g] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/11/2023] [Indexed: 07/20/2023] Open
Abstract
Traditional cancer chemotherapy easily produces serious toxic and side effects due to the lack of specific selection of tumor cells, which restricts its curative effect. Targeted delivery can increase the concentration of drugs in the target site and reduce their toxic and side effects on normal tissues and cells. Biocompatible and surface-modifiable nanocarriers are novel drug delivery systems, which are used to specifically target tumor sites in a controllable way. One of the effective ways to design effective targeting nanocarriers is to decorate with functional ligands, which can bind to specific receptors overexpressed on the surfaces of cancer cells. Various functional ligands, including transferrin, folic acid, polypeptide and hyaluronic acid, have been widely explored to develop tumor-selective drug delivery systems. This review focuses on the research progress of various receptors overexpressed on the surfaces of cancer cells and different nano-delivery systems of anticancer drugs targeted on the surfaces of cancer cells. We believe that through continuous research and development, actively targeted cancer nano-drugs will make a breakthrough and become an indispensable platform for accurate cancer treatment.
Collapse
Affiliation(s)
- Liquan Hong
- Deqing Hospital of Hangzhou Normal University, The Third People's Hospital of Deqing Deqing 313200 China
| | - Wen Li
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology Zhejiang Province Hangzhou 311121 China
| | - Yang Li
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology Zhejiang Province Hangzhou 311121 China
| | - Shouchun Yin
- Deqing Hospital of Hangzhou Normal University, The Third People's Hospital of Deqing Deqing 313200 China
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology Zhejiang Province Hangzhou 311121 China
| |
Collapse
|
18
|
Jain A, Bhattacharya S. Recent advances in nanomedicine preparative methods and their therapeutic potential for colorectal cancer: a critical review. Front Oncol 2023; 13:1211603. [PMID: 37427139 PMCID: PMC10325729 DOI: 10.3389/fonc.2023.1211603] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/05/2023] [Indexed: 07/11/2023] Open
Abstract
Colorectal cancer (CRC) is a prevalent malignancy that affects a large percentage of the global population. The conventional treatments for CRC have a number of limitations. Nanoparticles have emerged as a promising cancer treatment method due to their ability to directly target cancer cells and regulate drug release, thereby enhancing therapeutic efficacy and minimizing side effects. This compilation examines the use of nanoparticles as drug delivery systems for CRC treatment. Different nanomaterials can be used to administer anticancer drugs, including polymeric nanoparticles, gold nanoparticles, liposomes, and solid lipid nanoparticles. In addition, we discuss recent developments in nanoparticle preparation techniques, such as solvent evaporation, salting-out, ion gelation, and nanoprecipitation. These methods have demonstrated high efficacy in penetrating epithelial cells, a prerequisite for effective drug delivery. This article focuses on the various targeting mechanisms utilized by CRC-targeted nanoparticles and their recent advancements in this field. In addition, the review offers descriptive information regarding numerous nano-preparative procedures for colorectal cancer treatments. We also discuss the outlook for innovative therapeutic techniques in the management of CRC, including the potential application of nanoparticles for targeted drug delivery. The review concludes with a discussion of current nanotechnology patents and clinical studies used to target and diagnose CRC. The results of this investigation suggest that nanoparticles have great potential as a method of drug delivery for the treatment of colorectal cancer.
Collapse
|
19
|
Wei B, Lantz C, Liu W, Viner R, Loo RRO, Campuzano IDG, Loo JA. Added Value of Internal Fragments for Top-Down Mass Spectrometry of Intact Monoclonal Antibodies and Antibody-Drug Conjugates. Anal Chem 2023; 95:9347-9356. [PMID: 37278738 PMCID: PMC10954349 DOI: 10.1021/acs.analchem.3c01426] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Monoclonal antibodies (mAbs) and antibody-drug conjugates (ADCs) are two of the most important therapeutic drug classes that require extensive characterization, whereas their large size and structural complexity make them challenging to characterize and demand the use of advanced analytical methods. Top-down mass spectrometry (TD-MS) is an emerging technique that minimizes sample preparation and preserves endogenous post-translational modifications (PTMs); however, TD-MS of large proteins suffers from low fragmentation efficiency, limiting the sequence and structure information that can be obtained. Here, we show that including the assignment of internal fragments in native TD-MS of an intact mAb and an ADC can improve their molecular characterization. For the NIST mAb, internal fragments can access the sequence region constrained by disulfide bonds to increase the TD-MS sequence coverage to over 75%. Important PTM information, including intrachain disulfide connectivity and N-glycosylation sites, can be revealed after including internal fragments. For a heterogeneous lysine-linked ADC, we show that assigning internal fragments improves the identification of drug conjugation sites to achieve a coverage of 58% of all putative conjugation sites. This proof-of-principle study demonstrates the potential value of including internal fragments in native TD-MS of intact mAbs and ADCs, and this analytical strategy can be extended to bottom-up and middle-down MS approaches to achieve even more comprehensive characterization of important therapeutic molecules.
Collapse
Affiliation(s)
- Benqian Wei
- Department of Chemistry and Biochemistry, University of California Los Angeles-Los Angeles, CA, 90095 USA
| | - Carter Lantz
- Department of Chemistry and Biochemistry, University of California Los Angeles-Los Angeles, CA, 90095 USA
| | - Weijing Liu
- Thermo Fisher Scientific, San Jose, CA, 95134 USA
| | - Rosa Viner
- Thermo Fisher Scientific, San Jose, CA, 95134 USA
| | - Rachel R. Ogorzalek Loo
- Department of Chemistry and Biochemistry, University of California Los Angeles-Los Angeles, CA, 90095 USA
- UCLA-DOE Institute, University of California-Los Angeles, Los Angeles, CA, 90095 USA
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA, 90095 USA
| | - Iain D. G. Campuzano
- Amgen Research, Center for Research Acceleration and Digital Innovation, Molecular Analytics, Thousand Oaks, CA, 91320 USA
| | - Joseph A. Loo
- Department of Chemistry and Biochemistry, University of California Los Angeles-Los Angeles, CA, 90095 USA
- Department of Biological Chemistry, University of California-Los Angeles, Los Angeles, CA, 90095 USA
- UCLA-DOE Institute, University of California-Los Angeles, Los Angeles, CA, 90095 USA
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA, 90095 USA
| |
Collapse
|
20
|
Liu J, Chao T, Liu Y, Gong C, Zhang Y, Xiong H. Heterocyclic Molecular Targeted Drugs and Nanomedicines for Cancer: Recent Advances and Challenges. Pharmaceutics 2023; 15:1706. [PMID: 37376154 DOI: 10.3390/pharmaceutics15061706] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/28/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Cancer is a top global public health concern. At present, molecular targeted therapy has emerged as one of the main therapies for cancer, with high efficacy and safety. The medical world continues to struggle with the development of efficient, extremely selective, and low-toxicity anticancer medications. Heterocyclic scaffolds based on the molecular structure of tumor therapeutic targets are widely used in anticancer drug design. In addition, a revolution in medicine has been brought on by the quick advancement of nanotechnology. Many nanomedicines have taken targeted cancer therapy to a new level. In this review, we highlight heterocyclic molecular-targeted drugs as well as heterocyclic-associated nanomedicines in cancer.
Collapse
Affiliation(s)
- Junxia Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Tengfei Chao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Yingying Liu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200000, China
| | - Chen Gong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Yinan Zhang
- School of Chemical Science and Engineering, Tongji University, Shanghai 200000, China
| | - Huihua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| |
Collapse
|
21
|
Li X, Zhou J, Wang X, Li C, Ma Z, Wan Q, Peng F. New advances in the research of clinical treatment and novel anticancer agents in tumor angiogenesis. Biomed Pharmacother 2023; 163:114806. [PMID: 37163782 DOI: 10.1016/j.biopha.2023.114806] [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: 02/10/2023] [Revised: 04/24/2023] [Accepted: 04/30/2023] [Indexed: 05/12/2023] Open
Abstract
In 1971, Folkman proposed that tumors could be limited to very small sizes by blocking angiogenesis. Angiogenesis is the generation of new blood vessels from pre-existing vessels, considered to be one of the important processes in tumor growth and metastasis. Angiogenesis is a complex process regulated by various factors and involves many secreted factors and signaling pathways. Angiogenesis is important in the transport of oxygen and nutrients to the tumor during tumor development. Therefore, inhibition of angiogenesis has become an important strategy in the clinical management of many solid tumors. Combination therapies of angiogenesis inhibitors with radiotherapy and chemotherapy are often used in clinical practice. In this article, we will review common targets against angiogenesis, the most common and up-to-date anti-angiogenic drugs and clinical treatments in recent years, including active ingredients from chemical and herbal medicines.
Collapse
Affiliation(s)
- Xin Li
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jianbo Zhou
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xue Wang
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Chunxi Li
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zifan Ma
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Qiaoling Wan
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Fu Peng
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
22
|
Yu M, Liang Y, Li L, Zhao L, Kong F. Research progress of antibody-drug conjugates therapy for HER2-low expressing gastric cancer. Transl Oncol 2023; 29:101624. [PMID: 36652760 PMCID: PMC9860488 DOI: 10.1016/j.tranon.2023.101624] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 01/01/2023] [Accepted: 01/09/2023] [Indexed: 01/17/2023] Open
Abstract
Gastric cancer (GC) is a highly fatal malignant tumor in the world. Most of the patients are in an unresectable state when they have symptoms. Systemic treatment is the primary treatment for advanced patients. Among them, the Human epidermal growth factor receptor 2 (HER2) is an important therapeutic target. With the continuous optimization of chemotherapeutic drugs and chemotherapy regimens, the prognosis of some HER2-positive GC patients has been greatly improved. However, the needs of GC patients with a low level of HER2 expression still need to be met. Several targeted drugs against human epidermal growth factor receptor 2 emerged in recent years, including Antibody-drug Conjugates (ADC), novel humanized anti-HER2 monoclonal antibodies, and Tyrosine kinase inhibitors (TKI). As an important breakthrough in treating HER2-positive GC, ADC became one of the fastest-growing anti-tumor drugs. Some drugs also showed an anti-tumor effect on GC with low expression of HER2. It may also be the key to the treatment of low expression of HER2 GC in the future. This article mainly reviews several promising ADC drugs for the treatment of HER2 low-expression GC and related trials.
Collapse
Affiliation(s)
- Minghui Yu
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Anshanxi Road, Nankai District, Tianjin 300193, China,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, China
| | - Yangyueying Liang
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Anshanxi Road, Nankai District, Tianjin 300193, China,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, China
| | - Longhui Li
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Anshanxi Road, Nankai District, Tianjin 300193, China,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, China
| | - Lu Zhao
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Anshanxi Road, Nankai District, Tianjin 300193, China,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, China
| | - Fanming Kong
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Anshanxi Road, Nankai District, Tianjin 300193, China,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, China,Corresponding author at: Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Anshanxi Road, Nankai District, Tianjin 300193, China.
| |
Collapse
|
23
|
Design of Nanoparticles in Cancer Therapy Based on Tumor Microenvironment Properties. Pharmaceutics 2022; 14:pharmaceutics14122708. [PMID: 36559202 PMCID: PMC9785496 DOI: 10.3390/pharmaceutics14122708] [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: 10/19/2022] [Revised: 11/23/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer is one of the leading causes of death worldwide, and battling cancer has always been a challenging subject in medical sciences. All over the world, scientists from different fields of study try to gain a deeper knowledge about the biology and roots of cancer and, consequently, provide better strategies to fight against it. During the past few decades, nanoparticles (NPs) have attracted much attention for the delivery of therapeutic and diagnostic agents with high efficiency and reduced side effects in cancer treatment. Targeted and stimuli-sensitive nanoparticles have been widely studied for cancer therapy in recent years, and many more studies are ongoing. This review aims to provide a broad view of different nanoparticle systems with characteristics that allow them to target diverse properties of the tumor microenvironment (TME) from nanoparticles that can be activated and release their cargo due to the specific characteristics of the TME (such as low pH, redox, and hypoxia) to nanoparticles that can target different cellular and molecular targets of the present cell and molecules in the TME.
Collapse
|
24
|
Mouse Syngeneic Melanoma Model with Human Epidermal Growth Factor Receptor Expression. Pharmaceutics 2022; 14:pharmaceutics14112448. [PMID: 36432639 PMCID: PMC9697344 DOI: 10.3390/pharmaceutics14112448] [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: 09/28/2022] [Revised: 11/07/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
The development of epidermal growth factor receptor (EGFR)-targeting agents for the treatment of malignant melanoma requires cheap and easy animal tumor models for high-throughput in vivo screening. Thus, the aim of this study was to develop mouse syngeneic melanoma model that expresses human EGFR. Cloudman S91 clone M3 mouse melanoma cells were transduced with lentiviral particles carrying the human EGFR gene followed by a multistep selection process. The resulting M3-EGFR has been tested for EGFR expression and functionality in vitro and in vivo. Radioligand assay confirmed the presence of 13,900 ± 1500 EGF binding sites per cell at a dissociation constant of 5.3 ± 1.4 nM. M3-EGFR demonstrated the ability to bind and internalize specifically and provide the anticipated intracellular nuclear import of three different EGFR-targeted modular nanotransporters designed for specific anti-cancer drug delivery. Introduction of the human EGFR gene did not alter the tumorigenicity of the offspring M3-EGFR cells in host immunocompetent DBA/2J mice. Preservation of the expression of EGFR in vivo was confirmed by immunohistochemistry. To sum up, we successfully developed the first mouse syngeneic melanoma model with preserved in vivo expression of human EGFR.
Collapse
|
25
|
Yuan Y, Chen J, Fang M, Guo Y, Sun X, Yu D, Guo Y, Xin Y. Nimotuzumab combined with chemoradiotherapy for the treatment of cervical cancer: A meta-analysis of randomized controlled trials. Front Oncol 2022; 12:994726. [PMID: 36263226 PMCID: PMC9573994 DOI: 10.3389/fonc.2022.994726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/20/2022] [Indexed: 12/03/2022] Open
Abstract
Objectives To assess the clinical efficacy and toxicity of nimotuzumab in combination with chemoradiotherapy or chemoradiotherapy alone in the treatment of cervical cancer. Methods The PubMed, Embase, Cochrane Library, Web of Science, China National Knowledge Infrastructure, China Biomedical Medicine, Wanfang, and VIP databases were systematically searched for relevant literature. Ultimately, six randomised controlled trials (n=393) were included in our meta-analysis. Results A total of 393 patients were included, of which 197 were in the nimotuzumab combined with chemoradiotherapy group and 196 were in the chemoradiotherapy group. The results of our meta-analysis showed that the complete remission rate (risk ratio [RR] = 1.34, 95% confidence interval [CI]: 1.08-1.65, P = 0.007), objective response rate (RR = 1.30, 95% CI: 1.16-1.44, P < 0.05), and three-year survival rate (RR = 1.27, 95% CI: 1.06-1.51, P = 0.008) in the nimotuzumab combined with chemoradiotherapy group were significantly improved compared with the chemoradiotherapy group. This difference was not statistically significant when comparing the incidence of adverse reactions (such as leukocytopenia, gastrointestinal reaction, radiocystitis, and radioproctitis) between the two groups. Conclusions Nimotuzumab in combination with chemoradiotherapy has some advantages over chemoradiotherapy alone in the treatment of cervical cancer and does not increase toxicity. Therefore, nimotuzumab has the potential to be an effective treatment for cervical cancer; however, further evidence from large-scale randomised controlled trials is needed.
Collapse
Affiliation(s)
- Yan Yuan
- Department of Radiation, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Jiuzhou Chen
- Department of Radiation, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Miao Fang
- Department of Radiation, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Yaru Guo
- Department of Radiation, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Xueqing Sun
- Department of Radiation, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Dehong Yu
- Department of Radiation, the Affiliated Pizhou Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yilong Guo
- Department of Radiation, the Affiliated Pizhou Hospital of Xuzhou Medical University, Xuzhou, China
- *Correspondence: Yilong Guo, ; Yong Xin,
| | - Yong Xin
- Department of Radiation, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Cancer Institute, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Yilong Guo, ; Yong Xin,
| |
Collapse
|
26
|
Zhou Y, Jiang D, Chu X, Yan M, Qi H, Wu X, Tang Y, Dai Y. High expression of CD73 contributes to poor prognosis of clear-cell renal cell carcinoma by promoting cell proliferation and migration. Transl Cancer Res 2022; 11:3634-3644. [PMID: 36388013 PMCID: PMC9641103 DOI: 10.21037/tcr-22-544] [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: 03/02/2022] [Accepted: 08/01/2022] [Indexed: 10/12/2024]
Abstract
BACKGROUND Accumulating data have shown that high expression of CD73 is associated with poor prognosis in various cancers, however the role and significance of CD73 in clear-cell renal cell carcinoma (ccRCC) still remain unclear. The present study aims to evaluate the prognostic significance of CD73 in ccRCC and explore the potential function in vitro and in vivo. METHODS Firstly, the expression of CD73 in ccRCC was detected using clinical tissues and verified using TCGA and GEO data. Immunohistochemistry and Kaplan-Meier test were performed for survival analysis. Furthermore, knockdown or overexpression of CD73 was conducted by lentivirus transfection in ccRCC cells. MTT assay, colony formation assay, wound healing assay, transwell assay and xenograft assay were performed in vitro or in vivo. RESULTS Our results showed that CD73 was highly expressed in ccRCC, and high expression of CD73 was negatively correlated with prognosis. In addition, CD73 promoted cell proliferation and migration in vitro and in vivo. Our data also showed that CD73 played both enzymatic and non-enzymatic functions in the regulation of cell proliferation and migration in ccRCC. CONCLUSIONS These findings suggested that CD73 might promote the growth of ccRCC and contribute to poor prognosis. Taken together, CD73 may be a potential therapeutic target in ccRCC.
Collapse
Affiliation(s)
- Yihong Zhou
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Dong Jiang
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Xi Chu
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Minbo Yan
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Hao Qi
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Xiang Wu
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Yuxin Tang
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Yingbo Dai
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| |
Collapse
|
27
|
Xiao Y, Xu G, Cloyd JM, Du S, Mao Y, Pawlik TM. Predicting Novel Drug Candidates for Pancreatic Neuroendocrine Tumors via Gene Signature Comparison and Connectivity Mapping. J Gastrointest Surg 2022; 26:1670-1678. [PMID: 35508682 DOI: 10.1007/s11605-022-05337-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 04/19/2022] [Indexed: 02/08/2023]
Abstract
INTRODUCTION There is a paucity of effective treatment options for advanced pancreatic neuroendocrine tumors (pNETs). Genome-wide analyses may allow for potential drugs to be identified based on differentially expressed genes (DEGs). METHODS Oligo microarray data of RNA expression profiling of pNETs and normal pancreas tissues were downloaded from the Gene Expression Omnibus. Functional and pathway enrichment information of the DEGs was obtained using the Gene Ontology and Kyoto Encyclopedia of Genes and Genomes databases. Corresponding homologous proteins were analyzed and potential therapeutic drugs for pNETs were identified using the Connectivity Map and Drug-Gene Interaction Database. RESULTS Assessment of raw data from 12,610 pNET genes demonstrated that 1082 and 380 genes were upregulated and downregulated, respectively, compared with normal pancreas tissue. Upregulated pathways were associated with nitrogen metabolism (i.e., GABAergic synapse, and graft-versus-host disease), whereas downregulated pathways included C-type leptin receptor signaling pathway, pertussis and AMPK signaling pathway. In particular, the protein-protein interaction analysis revealed 10 upregulated hub genes (DYNLL1, GNB5, GNB2, GNG4, GNAI2, GNAI1, HIST2H2BE, NUP107, NUP133, and SNAP25) and 10 downregulated hub genes (CXCL8, F2, CXCL2, GCG, SST, INS, GALR3, CCL20, ADRA2B, and CXCL6). Using the Drug-Gene Interaction Database, candidate drugs for pNETs treatment included 3 EGFR inhibitors (canertinib, erlotinib, WZ-4-145), as well as other cell-signaling pathway inhibitors such as AG-592, acarbose, lonidamine, azacytidine, rottlerin, and HU-211. CONCLUSION Using available genetic atlas data, potential drug candidates for treatment of pNETs were identified based on differentially expressed genes. These results may help focus efforts on identifying targeted agents with therapeutic efficacy to treat patients with pNETs.
Collapse
Affiliation(s)
- Yao Xiao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Gang Xu
- Department of Liver Surgery and Liver Transplant Center, West China Hospital of Sichuan University, Chengdu, China
| | - Jordan M Cloyd
- Department of Surgery, The Ohio State University Wexner Medical Center and James Cancer Hospital and Solove Research Institute, 395 W. 12th Ave., Suite 670, Columbus, OH, USA
| | - Shunda Du
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Timothy M Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center and James Cancer Hospital and Solove Research Institute, 395 W. 12th Ave., Suite 670, Columbus, OH, USA.
| |
Collapse
|
28
|
Lassman AB, Pugh SL, Wang TJC, Aldape K, Gan HK, Preusser M, Vogelbaum MA, Sulman EP, Won M, Zhang P, Moazami G, Macsai MS, Gilbert MR, Bain EE, Blot V, Ansell PJ, Samanta S, Kundu MG, Armstrong TS, Wefel JS, Seidel C, de Vos FY, Hsu S, Cardona AF, Lombardi G, Bentsion D, Peterson RA, Gedye C, Bourg V, Wick A, Curran WJ, Mehta MP. Depatuxizumab mafodotin in EGFR-amplified newly diagnosed glioblastoma: A phase III randomized clinical trial. Neuro Oncol 2022; 25:339-350. [PMID: 35849035 PMCID: PMC9925712 DOI: 10.1093/neuonc/noac173] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Approximately 50% of newly diagnosed glioblastomas (GBMs) harbor epidermal growth factor receptor gene amplification (EGFR-amp). Preclinical and early-phase clinical data suggested efficacy of depatuxizumab mafodotin (depatux-m), an antibody-drug conjugate comprised of a monoclonal antibody that binds activated EGFR (overexpressed wild-type and EGFRvIII-mutant) linked to a microtubule-inhibitor toxin in EGFR-amp GBMs. METHODS In this phase III trial, adults with centrally confirmed, EGFR-amp newly diagnosed GBM were randomized 1:1 to radiotherapy, temozolomide, and depatux-m/placebo. Corneal epitheliopathy was treated with a combination of protocol-specified prophylactic and supportive measures. There was 85% power to detect a hazard ratio (HR) ≤0.75 for overall survival (OS) at a 2.5% 1-sided significance level (ie traditional two-sided p ≤ 0.05) by log-rank testing. RESULTS There were 639 randomized patients (median age 60, range 22-84; 62% men). Prespecified interim analysis found no improvement in OS for depatux-m over placebo (median 18.9 vs. 18.7 months, HR 1.02, 95% CI 0.82-1.26, 1-sided p = 0.63). Progression-free survival was longer for depatux-m than placebo (median 8.0 vs. 6.3 months; HR 0.84, 95% confidence interval [CI] 0.70-1.01, p = 0.029), particularly among those with EGFRvIII-mutant (median 8.3 vs. 5.9 months, HR 0.72, 95% CI 0.56-0.93, 1-sided p = 0.002) or MGMT unmethylated (HR 0.77, 95% CI 0.61-0.97; 1-sided p = 0.012) tumors but without an OS improvement. Corneal epitheliopathy occurred in 94% of depatux-m-treated patients (61% grade 3-4), causing 12% to discontinue. CONCLUSIONS Interim analysis demonstrated no OS benefit for depatux-m in treating EGFR-amp newly diagnosed GBM. No new important safety risks were identified.
Collapse
Affiliation(s)
- Andrew B Lassman
- Corresponding Author: Andrew B. Lassman, MD, Division of Neuro-Oncology, Department of Neurology, Vagelos College of Physicians and Surgeons, Herbert Irving Comprehensive Cancer Center, Columbia University, and New York-Presbyterian Hospital, 710 West 168th Street, New York, NY, USA. ()
| | - Stephanie L Pugh
- RTOG Foundation Statistics and Data Management Center, American College of Radiology, Philadelphia, Pennsylvania
| | - Tony J C Wang
- Department of Radiation Oncology (in Neurological Surgery), Columbia University Vagelos College of Physicians and Surgeons and New York-Presbyterian Hospital, New York, New York, USA,Herbert Irving Comprehensive Cancer Center, New York, New York, USA
| | - Kenneth Aldape
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Hui K Gan
- Cancer Therapies and Biology Group, Centre of Research Excellence in Brain Tumours, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, Melbourne, Australia,La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia,Department of Medicine, University of Melbourne, Heidelberg, Victoria, Australia
| | - Matthias Preusser
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | | | - Erik P Sulman
- Department of Radiation Oncology, New York University, Grossman School of Medicine, New York, New York, USA,Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - Minhee Won
- RTOG Foundation Statistics and Data Management Center, American College of Radiology, Philadelphia, Pennsylvania
| | | | - Golnaz Moazami
- Department of Ophthalmology, Columbia University Vagelos College of Physicians and Surgeons and New York-Presbyterian Hospital, New York, New York, USA
| | - Marian S Macsai
- NorthShore University HealthSystem, Department of Ophthalmology, University of Chicago Pritzker School of Medicine, Evanston, Illinois, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | | | | | | | | | | | | | - Jeffrey S Wefel
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Filip Y de Vos
- University Medical Center Utrecht, Cancer Center, Utrecht, The Netherlands
| | - Sigmund Hsu
- Department of Neurosurgery, University of Texas Health Sciences Center, McGovern School of Medicine, Houston, Texas, USA
| | - Andrés F Cardona
- Foundation for Clinical and Applied Cancer Research-FICMAC/Clinical and Translational Oncology Group, Brain Tumor Section, Bogotá, Colombia
| | - Giuseppe Lombardi
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | | | | | - Craig Gedye
- Calvary Mater Newcastle, Waratah, New South Wales, Australia
| | - Véronique Bourg
- Department of Neurology, Côte d’Azur University, Nice, France
| | - Antje Wick
- Heidelberg University Medical Center, Heidelberg, Germany
| | | | - Minesh P Mehta
- Miami Cancer Institute, Baptist Hospital, Miami, Florida, USA
| |
Collapse
|
29
|
Outstanding prognostic value of novel ferroptosis-related genes in chemoresistance osteosarcoma patients. Sci Rep 2022; 12:5029. [PMID: 35322804 PMCID: PMC8943205 DOI: 10.1038/s41598-022-09080-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/16/2022] [Indexed: 12/11/2022] Open
Abstract
Osteosarcoma (OS) is the most common bone-derived tumor, and chemoresistance is a pivotal factor in the poor prognosis of patients with OS. Ferroptosis, as an emerging modality of regulated cell death, has demonstrated potential value in tumor chemoresistance studies. Through the gene expression omnibus database in conjunction with the FerrDb database, we identified novel ferroptosis-related differentially expressed genes (DEGs) involving chemoresistance in OS patients. Subsequently, enrichment analysis, protein-protein interaction network analysis and survival analysis were performed sequentially to recognize the hub genes and ultimately to construct a predictive model. The model constructed from the TARGET database was exhibited in a nomogram and assessed by calibration curves. The prognostic value of the model and hub genes was validated separately by an independent cohort. Twenty-two ferroptosis-related DEGs were identified, including 16 up-regulated and 6 down-regulated. Among them, expressions of CBS, COCS1, EGFR, as hub genes, were significantly associated with the prognosis of OS patients and were evidenced as independent prognostic factors. An efficient prognostic model covering hub gene expressions and clinical variables was developed and validated. Combining the results of hub genes in differential analysis, the actions of hub genes in ferroptosis, and the prognostic relevance of hub genes in patients, we revealed that CBS, SOCS1 and EGFR might play essential roles in OS and its chemoresistance with potential research and clinical value.
Collapse
|
30
|
FITC-conjugated single chain Nimotuzumab can specifically recognize and enter EGFR-overexpressing cells. Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-022-10389-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
31
|
Chen Y, Du F, Tang L, Xu J, Zhao Y, Wu X, Li M, Shen J, Wen Q, Cho CH, Xiao Z. Carboranes as unique pharmacophores in antitumor medicinal chemistry. Mol Ther Oncolytics 2022; 24:400-416. [PMID: 35141397 PMCID: PMC8807988 DOI: 10.1016/j.omto.2022.01.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Carborane is a carbon-boron molecular cluster that can be viewed as a 3D analog of benzene. It features special physical and chemical properties, and thus has the potential to serve as a new type of pharmacophore for drug design and discovery. Based on the relative positions of two cage carbons, icosahedral closo-carboranes can be classified into three isomers, ortho-carborane (o-carborane, 1,2-C2B10H12), meta-carborane (m-carborane, 1,7-C2B10H12), and para-carborane (p-carborane, 1,12-C2B10H12), and all of them can be deboronated to generate their nido- forms. Cage compound carborane and its derivatives have been demonstrated as useful chemical entities in antitumor medicinal chemistry. The applications of carboranes and their derivatives in the field of antitumor research mainly include boron neutron capture therapy (BNCT), as BNCT/photodynamic therapy dual sensitizers, and as anticancer ligands. This review summarizes the research progress on carboranes achieved up to October 2021, with particular emphasis on signaling transduction pathways, chemical structures, and mechanistic considerations of using carboranes.
Collapse
Affiliation(s)
- Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Liyao Tang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Jinrun Xu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Qinglian Wen
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Luzhou Key Laboratory of Cell Therapy & Cell Drugs, Southwest Medical University, Luzhou 646000, China
| | - Chi Hin Cho
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Luzhou Key Laboratory of Cell Therapy & Cell Drugs, Southwest Medical University, Luzhou 646000, China
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Zhangang Xiao
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Luzhou Key Laboratory of Cell Therapy & Cell Drugs, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
32
|
Dunlap T, Cao Y. Physiological Considerations for Modeling in vivo Antibody-Target Interactions. Front Pharmacol 2022; 13:856961. [PMID: 35281913 PMCID: PMC8912916 DOI: 10.3389/fphar.2022.856961] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 02/10/2022] [Indexed: 11/25/2022] Open
Abstract
The number of therapeutic antibodies in development pipelines is increasing rapidly. Despite superior success rates relative to small molecules, therapeutic antibodies still face many unique development challenges. There is often a translational gap from their high target affinity and specificity to the therapeutic effects. Tissue microenvironment and physiology critically influence antibody-target interactions contributing to apparent affinity alterations and dynamic target engagement. The full potential of therapeutic antibodies will be further realized by contextualizing antibody-target interactions under physiological conditions. Here we review how local physiology such as physical stress, biological fluid, and membrane characteristics could influence antibody-target association, dissociation, and apparent affinity. These physiological factors in the early development of therapeutic antibodies are valuable toward rational antibody engineering, preclinical candidate selection, and lead optimization.
Collapse
Affiliation(s)
- Tyler Dunlap
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
33
|
Liu Q, Huang Q, Yu Z, Wu H. Clinical characteristics of non-small cell lung cancer patients with EGFR mutations and ALK&ROS1 fusions. THE CLINICAL RESPIRATORY JOURNAL 2022; 16:216-225. [PMID: 35081265 PMCID: PMC9060101 DOI: 10.1111/crj.13472] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 11/02/2021] [Accepted: 12/25/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To study the relationship between clinical characteristics and anaplastic lymphoma kinase (ALK) fusions, c-ros oncogene 1, receptor tyrosine kinase (ROS1) gene fusions, and epidermic growth factor receptor (EGFR) mutations in non-small cell lung cancer (NSCLC) patients to distinguish these different types. METHODS Both ALK, ROS1 gene rearrangements and EGFR mutations testing were performed. The clinical characteristics and associated pulmonary abnormalities were investigated. RESULTS Four hundred fifty-three NSCLC patients were included for analysis. One hundred seventy (37.5%), 32 (7.1%), and 9 cases (2.0%) with EGFR mutations, ALK gene fusions, and ROS1 gene fusions were identified, respectively. The EGFR-positive and ALK&ROS1-positive were more common in female (χ2 = 61.934, P < 0.001 and χ2 = 28.152, P < 0.001), non-smoking (χ2 = 59.315, P < 0.001 and χ2 = 11.080, P = 0.001), and adenocarcinoma (χ2 = 44.864, P < 0.001 and χ2 = 12.318, P = 0.002) patients; proportion of patients with emphysema was lower (χ2 = 35.494, P < 0.001 and χ2 = 15.770, P < 0.001) than the wild-type patients. The results of logistic regression analysis indicated that female (adjusted odds ratio [OR] 1.834, 95% confidence interval [CI] 1.069-3.144, P = 0.028), non-smoking (adjusted OR 2.504, 95% CI 1.456-4.306, P = 0.001), lung adenocarcinoma (adjusted OR 4.512, 95% CI 2.465-8.260, P < 0.001), stage III-IV (adjusted OR 2.232, 95% CI 1.066-4.676, P = 0.033), and no symptoms of emphysema (adjusted OR 2.139, 95% CI 1.221-3.747, P = 0.008) were independent variables associated with EGFR mutations. Young (adjusted OR 3.947, 95% CI 1.873-8.314, P < 0.001) and lung adenocarcinoma (adjusted OR 2.950, 95% CI 0.998-8.719, P = 0.050) were associated with ALK/ROS1 fusions. CONCLUSIONS EGFR mutations were more likely to occur in non-smoking, stage III-IV, and female patients with lung adenocarcinoma, whereas ALK&ROS1 gene fusions were more likely to occur in young patients with lung adenocarcinoma. Emphysema was less common in patients with EGFR mutations.
Collapse
Affiliation(s)
- Qinghua Liu
- Center for Pathological Diagnostics, Meizhou People's Hospital (Huangtang Hospital)Meizhou Academy of Medical SciencesMeizhouP. R. China
- Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou People's Hospital (Huangtang Hospital)Meizhou Academy of Medical SciencesMeizhouP. R. China
| | - Qingyan Huang
- Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou People's Hospital (Huangtang Hospital)Meizhou Academy of Medical SciencesMeizhouP. R. China
- Center for Precision Medicine, Meizhou People's Hospital (Huangtang Hospital)Meizhou Academy of Medical SciencesMeizhouP. R. China
| | - Zhikang Yu
- Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou People's Hospital (Huangtang Hospital)Meizhou Academy of Medical SciencesMeizhouP. R. China
- Center for Precision Medicine, Meizhou People's Hospital (Huangtang Hospital)Meizhou Academy of Medical SciencesMeizhouP. R. China
| | - Heming Wu
- Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou People's Hospital (Huangtang Hospital)Meizhou Academy of Medical SciencesMeizhouP. R. China
- Center for Precision Medicine, Meizhou People's Hospital (Huangtang Hospital)Meizhou Academy of Medical SciencesMeizhouP. R. China
| |
Collapse
|
34
|
Yu J, Fang T, Yun C, Liu X, Cai X. Antibody-Drug Conjugates Targeting the Human Epidermal Growth Factor Receptor Family in Cancers. Front Mol Biosci 2022; 9:847835. [PMID: 35295841 PMCID: PMC8919033 DOI: 10.3389/fmolb.2022.847835] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/08/2022] [Indexed: 12/14/2022] Open
Abstract
Members of the human epidermal growth factor receptor (HER) family, which includes HER1 (also known as EGFR), HER2, HER3 and HER4, have played a central role in regulating cell proliferation, survival, differentiation and migration. The overexpression of the HER family has been recognized as one of the most common cellular dysregulation associated with a wide variety of tumor types. Antibody-drug conjugates (ADCs) represent a new and promising class of anticancer therapeutics that combine the cancer specificity of antibodies with cytotoxicity of chemotherapeutic drugs. Two HER2-directed ADCs, trastuzumane-emtansine (T-DM1) and trastuzumab-deruxtecan (DS-8201a), have been approved for HER2-positive metastatic breast cancer by the U.S. Food and Drug Administration (FDA) in 2013 and 2019, respectively. A third HER2-directed ADC, disitamab vedotin (RC48), has been approved for locally advanced or metastatic gastric or gastroesophageal junction cancer by the NMPA (National Medical Products Administration) of China in 2021. A total of 11 ADCs that target HER family receptors (EGFR, HER2 or HER3) are currently under clinical trials. In this review article, we summarize the three approved ADCs (T-DM1, DS-8201a and RC48), together with the investigational EGFR-directed ADCs (ABT-414, MRG003 and M1231), HER2-directed ADCs (SYD985, ARX-788, A166, MRG002, ALT-P7, GQ1001 and SBT6050) and HER3-directed ADC (U3-1402). Lastly, we discuss the major challenges associated with the development of ADCs, and highlight the possible future directions to tackle these challenges.
Collapse
Affiliation(s)
| | | | | | | | - Xiaoqing Cai
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
35
|
T-box transcription factor 19 promotes hepatocellular carcinoma metastasis through upregulating EGFR and RAC1. Oncogene 2022; 41:2225-2238. [PMID: 35217793 DOI: 10.1038/s41388-022-02249-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/29/2022] [Accepted: 02/14/2022] [Indexed: 12/21/2022]
Abstract
The effect of targeted therapy for metastatic hepatocellular carcinoma (HCC) is still unsatisfactory. Exploring the underlying mechanism of HCC metastasis is favorable to provide new therapeutic strategies. T-box (TBX) transcription factor family genes, which are crucial regulators in embryo and organ development, are vital for regulating tumor initiation, growth and metastasis. Here we explored the role of TBX19 in HCC metastasis, which is one of the most upregulated TBX family genes in human HCC tissues. TBX19 expression was markedly upregulated in HCC tissues and elevated TBX19 expression predicted poor prognosis. Overexpression of TBX19 enhanced HCC metastasis through upregulating epidermal growth factor receptor (EGFR) and Rac family small GTPase 1 (RAC1) expression. Downregulation of EGFR and RAC1 inhibited TBX19-mediated HCC metastasis, while upregulation of EGFR and RAC1 restored inhibition of HCC metastasis mediated by TBX19 knockdown. Furthermore, epidermal growth factor (EGF)/EGFR signaling upregulated TBX19 expression via the extracellular signal-regulated kinase (ERK)/nuclear factor (NF)-kB axis. Besides, the combined application of EGFR inhibitor Erlotinib and RAC1 inhibitor NSC23766 markedly inhibited TBX19-mediated HCC metastasis. In HCC cohorts, TBX19 expression was positively associated with EGFR and RAC1 expression. Patients with positive coexpression of TBX19/EGFR or TBX19/RAC1 displayed the poorest prognosis. In conclusion, EGF/EGFR signaling upregulated TBX19 expression via ERK/NF-kB pathway and TBX19 fostered HCC metastasis by enhancing EGFR and RAC1 expression, which formed an EGF-TBX19-EGFR positive feedback loop. Targeting this signaling pathway may offer a potential therapeutic strategy to efficiently restrain TBX19-mediated HCC metastasis.
Collapse
|
36
|
Xue K, Luo B, Li X, Deng W, Zeng C, Zuo C. Consistency evaluation of lung adenocarcinoma tissue and circulating tumor cells related gene mutation detection based on multi-site immunomagnetic beads. J Biomater Appl 2022; 36:1700-1711. [PMID: 35029523 DOI: 10.1177/08853282211065861] [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: 11/17/2022]
Abstract
This study was designed to investigate the feasibility of genetic testing using circulating tumor cells (CTCs) instead of tumor tissues in lung adenocarcinoma to break through its limitation. Separation system for epithelial cell adhesion molecule (EpCAM), epidermal growth factor receptor (EGFR), and Vimentin expressing CTCs was constructed and used to capture CTCs in the blood samples of 57 patients with lung adenocarcinoma. Genetic mutations of genes involved in targeted therapies were detected by next-generation sequencing (NGS) in tissues from these patients. Blood CTC samples with the gene mutations identified by tissue-NGS were selected and corresponding gene mutations were detected by Sanger sequencing. The specificity of the CTC separation system was 95.48% and the sensitivity was 90.85%. The average number of CTCs in the blood of patients with lung adenocarcinoma was 12.47/7.5 mL. Comparison of the tissue-NGS with the CTC-Sanger sequencing showed that the consistencies of genetic mutations of EGFR (n = 24), KRAS (n = 9), TP53 (n = 19), BRAF (n = 1), ERBB2 (n = 2), and PIK3CA (n = 3) were 92.31%, 75.00%, 86.36%, 50.00%, 66.67%, and 75.00%, with an overall consistency of 84.06%. The CTC separation system established in this study shows high specificity and sensitivity. CTCs can be used as a suitable alternative to tumor tissues that are difficult to obtain in clinical practice and overcome the difficulties in tumor tissue collection, which is of significance in guiding clinical medication and individualized treatment with significant clinical application value in terms of genetic testing for targeted therapies in tumor treatment.
Collapse
Affiliation(s)
- Keying Xue
- 519885Xiamen Medical College Affiliated Second Hospital, Xiamen, China
| | - Bingqing Luo
- 519885Xiamen Medical College Affiliated Second Hospital, Xiamen, China
| | - Xiaoqing Li
- 519885Xiamen Medical College Affiliated Second Hospital, Xiamen, China
| | - Weixian Deng
- 519885Xiamen Medical College Affiliated Second Hospital, Xiamen, China
| | - Chunyan Zeng
- 519885Xiamen Medical College Affiliated Second Hospital, Xiamen, China
| | - Cuiyun Zuo
- 519885Xiamen Medical College Affiliated Second Hospital, Xiamen, China
| |
Collapse
|
37
|
Valle‑Mendiola A, Bustos‑Rodríguez R, Domínguez‑Melendez V, Zerecero‑Carreón O, Gutiérrez‑Hoya A, Weiss‑Steider B, Soto‑cruz I. Mutations in the helix αC of the catalytic domain from the EGFR affect its activity in cervical cancer cell lines. Oncol Lett 2022; 23:71. [PMID: 35069880 PMCID: PMC8756430 DOI: 10.3892/ol.2022.13191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/26/2021] [Indexed: 11/06/2022] Open
Abstract
The EGFR is a protein that belongs to the ErbB family of tyrosine kinase receptors. The EGFR is often overexpressed in human carcinomas. Amplification of the EGFR gene and mutations in the EGFR tyrosine kinase domain occur in patients with cancer. In cervical cancer, the expression level of the EGFR protein appears to directly associate with human papillomavirus infection. Our previous research demonstrated that in the cervical cancer cell lines, CALO and INBL, the EGFR is non-phosphorylated. The aim of the current study was to analyze the catalytic activity of the isolated EGFR and the presence of mutations in the control region αC. Catalytic activity was assessed by a universal in vitro kinase assay using polyGluTyr as a substrate, and the proteins were visualized by western blotting. For mutation analysis, DNA from CALO and INBL cell lines was isolated, and PCR was used to amplify the exons corresponding to the helix αC in the EGFR. The PCR products were visualized by agarose gel electrophoresis. The bands were isolated using a Zymoclean Gel DNA Recovery kit and directly sequenced. The EGFR, which was isolated and analyzed using the in vitro kinase assay, had catalytic activity. The receptor contained some mutations in the helix αC of the catalytic domain in both cell lines. The observed changes in the amino acid sequence may induce a different spatial arrangement and, therefore, a different conformation, which may confer different activities to this receptor. Thus, it was concluded that non-phosphorylated EGFR has catalytic activity, and it bears some amino acid changes in the helix αC of the catalytic domain in the CALO and INBL cells. These results suggest that the EGFR may function as an activator of other ErbB family receptors in these cervical cancer cells.
Collapse
Affiliation(s)
- Arturo Valle‑Mendiola
- Molecular Oncology Laboratory, Cell Differentiation and Cancer Research Unit, UMIEZ Campus II, FES Zaragoza, National University of Mexico, Iztapalapa, Mexico City 09230, Mexico
| | - Ricardo Bustos‑Rodríguez
- Molecular Oncology Laboratory, Cell Differentiation and Cancer Research Unit, UMIEZ Campus II, FES Zaragoza, National University of Mexico, Iztapalapa, Mexico City 09230, Mexico
| | | | - Octavio Zerecero‑Carreón
- Molecular Oncology Laboratory, Cell Differentiation and Cancer Research Unit, UMIEZ Campus II, FES Zaragoza, National University of Mexico, Iztapalapa, Mexico City 09230, Mexico
| | - Adriana Gutiérrez‑Hoya
- Molecular Oncology Laboratory, Cell Differentiation and Cancer Research Unit, UMIEZ Campus II, FES Zaragoza, National University of Mexico, Iztapalapa, Mexico City 09230, Mexico
| | - Benny Weiss‑Steider
- Molecular Oncology Laboratory, Cell Differentiation and Cancer Research Unit, UMIEZ Campus II, FES Zaragoza, National University of Mexico, Iztapalapa, Mexico City 09230, Mexico
| | - Isabel Soto‑cruz
- Molecular Oncology Laboratory, Cell Differentiation and Cancer Research Unit, UMIEZ Campus II, FES Zaragoza, National University of Mexico, Iztapalapa, Mexico City 09230, Mexico
| |
Collapse
|
38
|
Purba ER, Saita EI, Akhouri RR, Öfverstedt LG, Wilken G, Skoglund U, Maruyama IN. Allosteric activation of preformed EGF receptor dimers by a single ligand binding event. Front Endocrinol (Lausanne) 2022; 13:1042787. [PMID: 36531494 PMCID: PMC9748436 DOI: 10.3389/fendo.2022.1042787] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/14/2022] [Indexed: 12/02/2022] Open
Abstract
Aberrant activation of the epidermal growth factor receptor (EGFR) by mutations has been implicated in a variety of human cancers. Elucidation of the structure of the full-length receptor is essential to understand the molecular mechanisms underlying its activation. Unlike previously anticipated, here, we report that purified full-length EGFR adopts a homodimeric form in vitro before and after ligand binding. Cryo-electron tomography analysis of the purified receptor also showed that the extracellular domains of the receptor dimer, which are conformationally flexible before activation, are stabilized by ligand binding. This conformational flexibility stabilization most likely accompanies rotation of the entire extracellular domain and the transmembrane domain, resulting in dissociation of the intracellular kinase dimer and, thus, rearranging it into an active form. Consistently, mutations of amino acid residues at the interface of the symmetric inactive kinase dimer spontaneously activate the receptor in vivo. Optical observation also indicated that binding of only one ligand activates the receptor dimer on the cell surface. Our results suggest how oncogenic mutations spontaneously activate the receptor and shed light on the development of novel cancer therapies.
Collapse
Affiliation(s)
- Endang R. Purba
- Information Processing Biology Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Ei-ichiro Saita
- Information Processing Biology Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Reetesh R. Akhouri
- Cellular Structural Biology Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Lars-Goran Öfverstedt
- Cellular Structural Biology Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Gunnar Wilken
- Cellular Structural Biology Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Ulf Skoglund
- Cellular Structural Biology Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Ichiro N. Maruyama
- Information Processing Biology Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
- *Correspondence: Ichiro N. Maruyama,
| |
Collapse
|
39
|
Botticelli A, Pomati G, Marchetti P. Target therapy in cancer treatment. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00152-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
40
|
Polymorphisms in EGFR Gene Predict Clinical Outcome in Unresectable Non-Small Cell Lung Cancer Treated with Radiotherapy and Platinum-Based Chemoradiotherapy. Int J Mol Sci 2021; 22:ijms22115605. [PMID: 34070597 PMCID: PMC8197839 DOI: 10.3390/ijms22115605] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 01/29/2023] Open
Abstract
For non-small cell lung cancer (NSCLC), radiotherapy (RT) and platinum-based chemotherapy (CHT) are among the main treatment options. On the other hand, radioresistance and cytotoxic drug resistance are common causes of failure. The epidermal growth factor receptor (EGFR) plays an important role in radioresponse and therapy resistance. We hypothesized that single nucleotide polymorphisms (SNPs) in the EGFR gene might affect individual sensitivity to these treatments, and thus, therapy outcome and prognosis. The association between functional EGFR SNPs and overall (OS), locoregional recurrence-free (LFRS), and metastasis-free (MFS) survival was examined in 436 patients with unresectable NSCLC receiving RT and platinum-based CHTRT. In a multivariate analysis, the rs712830 CC homozygotes showed reduced OS in the whole group (p = 0.039) and in the curative treatment subset (p = 0.047). The rs712829 TT genotype was strongly associated with decreased LRFS (p = 0.006), and the T-C haplotype was a risk factor for locoregional recurrence in our patients (p = 0.003). The rs2227983 GG alone and in combination with rs712829 T was an indicator of unfavorable LRFS (p = 0.028 and 0.002, respectively). Moreover, significant independent effects of these SNPs on OS, LRFS, and MFS were observed. Our results demonstrate that inherited EGFR gene variants may predict clinical outcomes in NSCLC treated with DNA damage-inducing therapy.
Collapse
|
41
|
Li P, Zhu K, Mo Y, Deng X, Jiang X, Shi L, Guo C, Zhang W, Zeng Z, Li G, Xiong W, Zhang S, Gong Z. Research Progress of circRNAs in Head and Neck Cancers. Front Oncol 2021; 11:616202. [PMID: 33996542 PMCID: PMC8117014 DOI: 10.3389/fonc.2021.616202] [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: 10/11/2020] [Accepted: 04/07/2021] [Indexed: 12/13/2022] Open
Abstract
Circular RNAs (circRNAs) are a novel type of non-coding RNAs. Because of their characteristics of a closed loop structure, disease- and tissue-specificity, and high conservation and stability, circRNAs have the potential to be biomarkers for disease diagnosis. Head and neck cancers are one of the most common malignant tumors with high incidence rates globally. Affected patients are often diagnosed at the advanced stage with poor prognosis, owing to the concealment of anatomic sites. The characteristics, functions, and specific mechanisms of circRNAs in head and neck cancers are increasingly being discovered, and they have important clinical significance for the early diagnosis, treatment, and prognosis evaluation of patients with cancer. In this study, the generation, characteristics, and functions of circRNAs, along with their regulatory mechanisms in head and neck cancers have been summarized. We report that circRNAs interact with molecules such as transcription and growth factors to influence specific pathways involved in tumorigenesis. We conclude that circRNAs have an important role to play in the proliferation, invasion, metastasis, energy and substance metabolism, and treatment resistance in cancers.
Collapse
Affiliation(s)
- Panchun Li
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Kunjie Zhu
- Department of Head and Neck Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yongzhen Mo
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China
| | - Xiangying Deng
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China
| | - Xianjie Jiang
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China
| | - Lei Shi
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Can Guo
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China
| | - Wenling Zhang
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China
| | - Shanshan Zhang
- Department of Stomatology, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
| | - Zhaojian Gong
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
42
|
Rosenkranz AA, Slastnikova TA. Epidermal Growth Factor Receptor: Key to Selective Intracellular Delivery. BIOCHEMISTRY (MOSCOW) 2021; 85:967-1092. [PMID: 33050847 DOI: 10.1134/s0006297920090011] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Epidermal growth factor receptor (EGFR) is an integral surface protein mediating cellular response to a number of growth factors. Its overexpression and increased activation due to mutations is one of the most common traits of many types of cancer. Development and clinical use of the agents, which block EGFR activation, became a prime example of the personalized targeted medicine. However, despite the obvious success in this area, cancer cure remains unattainable in most cases. Because of that, as well as the result of the search for possible ways to overcome the difficulties of treatment, a huge number of new treatment methods relying on the use of EGFR overexpression and its changes to destroy cancer cells. Modern data on the structure, functioning, and intracellular transport of EGFR, its natural ligands, as well as signaling cascades triggered by the EGFR activation, peculiarities of the EGFR expression and activation in oncological disorders, as well as applied therapeutic approaches aimed at blocking EGFR signaling pathway are summarized and analyzed in this review. Approaches to the targeted delivery of various chemotherapeutic agents, radionuclides, immunotoxins, photosensitizers, as well as the prospects for gene therapy aimed at cancer cells with EGFR overexpression are reviewed in detail. It should be noted that increasing attention is being paid nowadays to the development of multifunctional systems, either carrying several different active agents, or possessing several environment-dependent transport functions. Potentials of the systems based on receptor-mediated endocytosis of EGFR and their possible advantages and limitations are discussed.
Collapse
Affiliation(s)
- A A Rosenkranz
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia. .,Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - T A Slastnikova
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| |
Collapse
|
43
|
Shah A, Rauth S, Aithal A, Kaur S, Ganguly K, Orzechowski C, Varshney GC, Jain M, Batra SK. The Current Landscape of Antibody-based Therapies in Solid Malignancies. Am J Cancer Res 2021; 11:1493-1512. [PMID: 33391547 PMCID: PMC7738893 DOI: 10.7150/thno.52614] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023] Open
Abstract
Over the past three decades, monoclonal antibodies (mAbs) have revolutionized the landscape of cancer therapy. Still, this benefit remains restricted to a small proportion of patients due to moderate response rates and resistance emergence. The field has started to embrace better mAb-based formats with advancements in molecular and protein engineering technologies. The development of a therapeutic mAb with long-lasting clinical impact demands a prodigious understanding of target antigen, effective mechanism of action, gene engineering technologies, complex interplay between tumor and host immune system, and biomarkers for prediction of clinical response. This review discusses the various approaches used by mAbs for tumor targeting and mechanisms of therapeutic resistance that is not only caused by the heterogeneity of tumor antigen, but also the resistance imposed by tumor microenvironment (TME), including inefficient delivery to the tumor, alteration of effector functions in the TME, and Fc-gamma receptor expression diversity and polymorphism. Further, this article provides a perspective on potential strategies to overcome these barriers and how diagnostic and prognostic biomarkers are being used in predicting response to mAb-based therapies. Overall, understanding these interdependent parameters can improve the current mAb-based formulations and develop novel mAb-based therapeutics for achieving durable clinical outcomes in a large subset of patients.
Collapse
|
44
|
Bhatia P, Sharma V, Alam O, Manaithiya A, Alam P, Kahksha, Alam MT, Imran M. Novel quinazoline-based EGFR kinase inhibitors: A review focussing on SAR and molecular docking studies (2015-2019). Eur J Med Chem 2020; 204:112640. [PMID: 32739648 DOI: 10.1016/j.ejmech.2020.112640] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/29/2020] [Accepted: 07/01/2020] [Indexed: 12/11/2022]
Abstract
The over expression of EGFR has been recognized as the driver mechanism in the occurrence and progression of carcinomas such as lung cancer, breast cancer, pancreatic cancer, etcetera. EGFR receptor was thus established as an important target for the management of solid tumors. The occurrence of resistance caused as a result of mutations in EGFR has presented a formidable challenge in the discovery of novel inhibitors of EGFR. This has resulted in the development of three generations of EGFR TKIs. Newer mutations like C797S cause failure of Osimertinib and other EGFR TKIs belonging to the third-generation caused by the development of resistance. In this review, we have summarized the work done in the last five years to overcome the limitations of currently marketed drugs, giving structural activity relationships of quinazoline-based lead compounds synthesized and tested recently. We have also highlighted the shortcomings of the currently used approaches and have provided guidance for circumventing these limitations. Our review would help medicinal chemists streamline and guide their efforts towards developing novel quinazoline-based EGFR inhibitors.
Collapse
Affiliation(s)
- Parth Bhatia
- Medicinal Chemistry and Molecular Modelling Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Vrinda Sharma
- Medicinal Chemistry and Molecular Modelling Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Ozair Alam
- Medicinal Chemistry and Molecular Modelling Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| | - Ajay Manaithiya
- Medicinal Chemistry and Molecular Modelling Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Perwaiz Alam
- Medicinal Chemistry and Molecular Modelling Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Kahksha
- Medicinal Chemistry and Molecular Modelling Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Md Tauquir Alam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha, Pin Code 91911, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha, Pin Code 91911, Saudi Arabia
| |
Collapse
|
45
|
Lodge JM, Schauer KL, Brademan DR, Riley NM, Shishkova E, Westphall MS, Coon JJ. Top-Down Characterization of an Intact Monoclonal Antibody Using Activated Ion Electron Transfer Dissociation. Anal Chem 2020; 92:10246-10251. [PMID: 32608969 DOI: 10.1021/acs.analchem.0c00705] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Monoclonal antibodies (mAbs) are important therapeutic glycoproteins, but their large size and structural complexity make them difficult to rapidly characterize. Top-down mass spectrometry (MS) has the potential to overcome challenges of other common approaches by minimizing sample preparation and preserving endogenous modifications. However, comprehensive mAb characterization requires generation of many, well-resolved fragments and remains challenging. While ETD retains modifications and cleaves disulfide bonds-making it attractive for mAb characterization-it can be less effective for precursors having high m/z values. Activated ion electron transfer dissociation (AI-ETD) uses concurrent infrared photoactivation to promote product ion generation and has proven effective in increasing sequence coverage of intact proteins. Here, we present the first application of AI-ETD to mAb sequencing. For the standard NIST mAb, we observe a high degree of complementarity between fragments generated using standard ETD with a short reaction time and AI-ETD with a long reaction time. Most importantly, AI-ETD reveals disulfide-bound regions that have been intractable, thus far, for sequencing with top-down MS. We conclude AI-ETD has the potential to rapidly and comprehensively analyze intact mAbs.
Collapse
|
46
|
Li WQ, Cui JW. Non-small cell lung cancer patients with ex19del or exon 21 L858R mutation: distinct mechanisms, different efficacies to treatments. J Cancer Res Clin Oncol 2020; 146:2329-2338. [PMID: 32596787 DOI: 10.1007/s00432-020-03296-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/20/2020] [Indexed: 12/11/2022]
Abstract
With the development of antitumor therapies, different treatment methods including monotherapy and combined therapy have achieved clinical efficacy in advanced epidermal growth factor receptor (EGFR) mutant non-small cell lung cancer (NSCLC) patients. Exon 19 deletion (ex19del) and exon 21 L858R mutation are common sensitive subtypes of EGFR mutation. However, potential distinct mechanisms are found from several dimensions including molecular structures, biological behaviors, concomitant mutations, resistance mechanisms and tumor mutation burdens. More evidence indicates the prognostic difference of EGFR subgroups. This review focused on the progress of potential distinct mechanisms and outcomes in clinical trials of advanced NSCLC patients with ex19del or exon 21 L858R mutation.
Collapse
Affiliation(s)
- W-Q Li
- Department of Cancer Center, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - J-W Cui
- Department of Cancer Center, The First Hospital of Jilin University, Changchun, 130021, Jilin, China.
| |
Collapse
|