1
|
Liu X, Cui Y, Gong J, Yu X, Cui Y, Xuan Y. SETD5 facilitates stemness and represses ferroptosis via m6A-mediating PKM2 stabilization in non-small cell lung cancer. Oncogene 2025:10.1038/s41388-025-03426-9. [PMID: 40307507 DOI: 10.1038/s41388-025-03426-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 04/12/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025]
Abstract
SETD5, an atypical member of the histone lysine methyltransferase family known for its association with cancer stemness, is a significant predictor of unfavorable survival outcomes in non-small cell lung cancer (NSCLC). However, the function of SETD5 in NSCLC stemness remains unclear, and whether it is an active H3K36me3 is controversial. Consequently, further investigation is required to clarify the pivotal role of SETD5 in NSCLC stemness and its related mechanism. Thus, this study employed the NSCLC tissue microarray and bioinformatics tools to analyze SETD5 expression and determine its effect on stemness and investigated the role of SETD5 in the metastasis of NSCLC using in vitro and in vivo analyses. The findings indicated high SETD5 expression in embryonic and NSCLC tissues, which was related to the pathological tumor stage, lymph node metastasis, and clinical stage, indicating that SETD5 could be used as a biomarker and prognostic factor in NSCLC. In addition, we found that SETD5 can promote glycolysis, thereby inhibiting ferroptosis and promoting the stemness of NSCLC, causing tumor metastasis and adverse prognosis in patients. In terms of mechanism, SETD5 as H3K36me3 facilitates the m6A modification of METTL14 and the recruitment of YTHDF1 and mediates PKM2 nuclear translocation and phosphorylation of p-PKM2 Tyr105, regulating GPX4 mediated ferroptosis resistance and SOX9 mediated stemness in NSCLC. The findings emphasize that SETD5 may serve as a promising indicator of stemness in NSCLC, which can help develop therapeutic targets for NSCLC and prognostic evaluation. This study provides evidence that SETD5 as H3K36me3 facilitates the m6A modification of METTL14 and the recruitment of YTHDF1 and mediates the nuclear translocation of PKM2, regulating GPX4 mediated ferroptosis resistance and SOX9 mediated stemness, causing tumor metastasis and adverse prognosis in patients.
Collapse
Affiliation(s)
- Xingzhe Liu
- Department of Pathology, Yanbian University College of Medicine, Yanji, China
| | - Yuzhen Cui
- Department of Oncology, Affiliated Hospital of Yanbian University, Yanji, China
| | - Jie Gong
- Department of Pathology, Yanbian University College of Medicine, Yanji, China
| | - Xinhui Yu
- Department of Oncology, Affiliated Hospital of Yanbian University, Yanji, China
| | - Yan Cui
- Department of Oncology, Affiliated Hospital of Yanbian University, Yanji, China
| | - Yanhua Xuan
- Department of Pathology, Yanbian University College of Medicine, Yanji, China.
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, China.
- Institute of Regenerative Medicine, Yanbian University College of Medicine, Yanji, China.
| |
Collapse
|
2
|
Lalmuansangi C, Lalfakawmi, Nghakliana F, Sailo H, Tochhawng L, Trivedi AK, Kharat KR, Vellingiri B, Kumar NS, Siama Z. Anticancer activity of Stemona tuberosa (wild asparagus) against type-II human lung adenocarcinoma (A549) cells and identification of SRC inhibitor using integrated network pharmacology and molecular dynamic simulation. Discov Oncol 2025; 16:429. [PMID: 40159570 PMCID: PMC11955439 DOI: 10.1007/s12672-025-02138-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/12/2025] [Indexed: 04/02/2025] Open
Abstract
Stemona tuberosa is widely recognized for its traditional applications as an anti-cancer agent. This study aimed to assess the anti-cancer properties of S. tuberosa in human lung adenocarcinoma A549 cells. Among the various solvent extracts of S. tuberosa, the methanolic extract showed the highest toxicity against A549 cells. The S. tuberosa extract elicited cytotoxic effects and suppressed colony formation in A549 cells in a dose-dependent manner. S. tuberosa activity was further supported by AO/EtBr staining, increased caspase 3/6 activity, upregulation of pro-apoptotic genes, DNA damage, and elevated lipid peroxidation, with decreasing antioxidant levels. LC-MS analysis identified 80 predominant secondary metabolites in the methanolic extracts of S. tuberosa. A network pharmacology study identified SRC as the primary target of compounds identified from S. tuberosa. SRC protein is crucial for advancing lung cancer because of its function in cell proliferation, survival, and metastasis. Among the various compounds identified from S. tuberosa extract, 4-Azatricyclo [4.3.1.13,8] undecan-5-one (ADE) (- 10.88 kcal/mol) and Dihydro-normorphine, 3-desoxy- (DNY) (- 10.83 kcal/mol) exhibited notable binding affinities for SRC. Further analysis using molecular dynamics simulations (100 ns) validated the stability of SRC-ligand complexes, with RMSD of 1.8 and 2.2 Å for ADE and DNY, respectively, alongside the establishment of essential hydrogen bonds with pivotal residues, including ASP408, ALA403, and THR438. Finally, gmx._MMPBSA showed favourable ΔGbind values for ADE (- 15.06 ± 0.11 kcal/mol) and DNY (- 15.66 ± 0.25 kcal/mol), which highlights the significant potential of ADE and DNY as effective SRC inhibitors, suggesting S. tuberosa as a novel candidate for cancer therapy.
Collapse
Affiliation(s)
- C Lalmuansangi
- Department of Zoology, Mizoram University (a Central University), Aizawl, 796004, India
| | - Lalfakawmi
- Department of Zoology, Mizoram University (a Central University), Aizawl, 796004, India
| | - Fanai Nghakliana
- Department of Zoology, Mizoram University (a Central University), Aizawl, 796004, India
| | - Hmingremhlua Sailo
- Department of Botany, Mizoram University (a Central University), Aizawl, 796004, India
| | - Lalchhandami Tochhawng
- Mizoram Science, Technology and Innovation Council (MISTIC), Aizawl, 796001, Mizoram, India
| | - Amit Kumar Trivedi
- Department of Zoology, Mizoram University (a Central University), Aizawl, 796004, India
| | - Kiran R Kharat
- Department of Zoology, Mizoram University (a Central University), Aizawl, 796004, India
| | - Balachandar Vellingiri
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, India
| | | | - Zothan Siama
- Department of Zoology, Mizoram University (a Central University), Aizawl, 796004, India.
| |
Collapse
|
3
|
Cheng Y, Han R, Wang M, Wang S, Zhou J, Wang J, Xu H. M 6A -mediated lncRNA SCIRT stability promotes NSCLC progression through binding to SFPQ and activating the PI3K/Akt pathway. Cell Mol Life Sci 2025; 82:63. [PMID: 39869159 PMCID: PMC11772919 DOI: 10.1007/s00018-025-05594-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 01/28/2025]
Abstract
Non-small cell lung cancer (NSCLC) has emerged as one of the most prevalent malignancies worldwide. N6-methyladenosine (m6A) methylation, a pervasive epigenetic modification in long noncoding RNAs (lncRNAs), plays a crucial role in NSCLC progression. Here, we report that m6A modification and the expression of the lncRNA stem cell inhibitory RNA transcript (SCIRT) was significantly upregulated in NSCLC tissues and cells. Functional analysis revealed that SCIRT enhanced NSCLC cell proliferation, migration, invasion, and epithelial‒mesenchymal transition. The m6A modification of SCIRT can be installed by METTL3, which enhanced the stability of this lncRNA. Notably, SCIRT overexpression in response to DNA double-strand breaks (DSBs) sensitized cells to camptothecin (CPT) and impairs DNA homologous recombination repair. SCIRT directly interacted with SFPQ in vitro and was primarily localized in the nucleus. Furthermore, ectopic SCIRT expression upregulated SFPQ and activated the PI3K/Akt pathway following CPT treatment, suggesting an unexpected role of SCIRT in facilitating SFPQ-mediated DSB repair. In brief, our findings highlight the oncogenic role of SCIRT in NSCLC by binding SFPQ and activating PI3K/Akt signaling, presenting a promising therapeutic target for personalized NSCLC treatment.
Collapse
Affiliation(s)
- Yongming Cheng
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150081, China
| | - Rong Han
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150081, China
| | - Meiqi Wang
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150081, China
| | - Shuqing Wang
- Department of Clinical Laboratory, Harbin Fourth Hospital, 119 Jingyu Road, Harbin, 150001, China
| | - Junliang Zhou
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150081, China
| | - Jianyi Wang
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150081, China
| | - Hui Xu
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150081, China.
| |
Collapse
|
4
|
Wang J, Fang S, Jiang Y, Hua Q. Unraveling the Mechanism of Action of Ubiquitin-Specific Protease 5 and Its Inhibitors in Tumors. Clin Med Insights Oncol 2024; 18:11795549241281932. [PMID: 39391229 PMCID: PMC11465303 DOI: 10.1177/11795549241281932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 08/21/2024] [Indexed: 10/12/2024] Open
Abstract
Ubiquitin-specific protease 5 (USP5), a member of the ubiquitin-specific proteases (USPs) family, functions by specifically removing ubiquitin chains from target proteins for stabilization and degrading unbound polyubiquitin chains to maintain a steady-state monoubiquitin pool. Ubiquitin-specific protease 5 regulates various cellular activities, including DNA double-strand break repair, transmission of neuropathic and inflammatory pain signals, immune response, and tumor cell proliferation. Furthermore, USP5 is involved in the development of multiple tumors such as liver, lung, pancreatic, and breast cancers as well as melanoma. Downstream regulatory mechanisms associated with USP5 are complex and diverse. Ubiquitin-specific protease 5 has been revealed as an emerging target for tumor treatment. This study has introduced some molecules upstream to control the expression of USP5 at the levels of transcription, translation, and post-translation. Furthermore, the study incorporated inhibitors known to be associated with USP5, including partially selective deubiquitinase (DUB) inhibitors such as WP1130, EOAI3402143, vialinin A, and chalcone derivatives. It also included the ubiquitin-activating enzyme E1 inhibitor, PYR-41. These small molecule inhibitors impact the occurrence and development of various tumors. Therefore, this article comprehensively reviews the pivotal role of USP5 in different signaling pathways during tumor progression and resumes the progress made in developing USP5 inhibitors, providing a theoretical foundation for their clinical translation.
Collapse
Affiliation(s)
| | | | - Yang Jiang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qingquan Hua
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
5
|
Blobner J, Dengler L, Eberle C, Herold JJ, Xu T, Beck A, Mühlbauer A, Müller KJ, Teske N, Karschnia P, van den Heuvel D, Schallerer F, Ishikawa-Ankerhold H, Thon N, Tonn JC, Subklewe M, Kobold S, Harter PN, Buchholz VR, von Baumgarten L. PD-1 blockade does not improve efficacy of EpCAM-directed CAR T-cell in lung cancer brain metastasis. Cancer Immunol Immunother 2024; 73:255. [PMID: 39358663 PMCID: PMC11447167 DOI: 10.1007/s00262-024-03837-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/15/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Lung cancer brain metastasis has a devastating prognosis, necessitating innovative treatment strategies. While chimeric antigen receptor (CAR) T-cell show promise in hematologic malignancies, their efficacy in solid tumors, including brain metastasis, is limited by the immunosuppressive tumor environment. The PD-L1/PD-1 pathway inhibits CAR T-cell activity in the tumor microenvironment, presenting a potential target to enhance therapeutic efficacy. This study aims to evaluate the impact of anti-PD-1 antibodies on CAR T-cell in treating lung cancer brain metastasis. METHODS We utilized a murine immunocompetent, syngeneic orthotopic cerebral metastasis model for repetitive intracerebral two-photon laser scanning microscopy, enabling in vivo characterization of red fluorescent tumor cells and CAR T-cell at a single-cell level over time. Red fluorescent EpCAM-transduced Lewis lung carcinoma cells (EpCAM/tdtLL/2 cells) were implanted intracranially. Following the formation of brain metastasis, EpCAM-directed CAR T-cell were injected into adjacent brain tissue, and animals received either anti-PD-1 or an isotype control. RESULTS Compared to controls receiving T-cell lacking a CAR, mice receiving EpCAM-directed CAR T-cell showed higher intratumoral CAR T-cell densities in the beginning after intraparenchymal injection. This finding was accompanied with reduced tumor growth and translated into a survival benefit. Additional anti-PD-1 treatment, however, did not affect intratumoral CAR T-cell persistence nor tumor growth and thereby did not provide an additional therapeutic effect. CONCLUSION CAR T-cell therapy for brain malignancies appears promising. However, additional anti-PD-1 treatment did not enhance intratumoral CAR T-cell persistence or effector function, highlighting the need for novel strategies to improve CAR T-cell therapy in solid tumors.
Collapse
Affiliation(s)
- Jens Blobner
- Department of Neurosurgery, LMU University Hospital, Ludwig Maximilians University (LMU), 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
- Division of Neuro-Oncology, Department of Neurosurgery, Ludwig Maximilians University School of Medicine, Marchioninistrasse 15, 81377, Munich, Germany
| | - Laura Dengler
- German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | - Constantin Eberle
- German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | - Julika J Herold
- German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | - Tao Xu
- German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
- Department of Neurology, LMU University Hospital, Ludwig Maximilians University (LMU), 81377, Munich, Germany
| | - Alexander Beck
- German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
- Center for Neuropathology and Prion Research, Faculty of Medicine LMU Munich, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Anton Mühlbauer
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, 81675, Munich, Germany
| | - Katharina J Müller
- German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
- Department of Neurology, LMU University Hospital, Ludwig Maximilians University (LMU), 81377, Munich, Germany
| | - Nico Teske
- Department of Neurosurgery, LMU University Hospital, Ludwig Maximilians University (LMU), 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | - Philipp Karschnia
- Department of Neurosurgery, LMU University Hospital, Ludwig Maximilians University (LMU), 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | - Dominic van den Heuvel
- Department of Medicine I, Ludwig-Maximilians-University School of Medicine, Munich, Germany
| | - Ferdinand Schallerer
- German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | | | - Niklas Thon
- Department of Neurosurgery, LMU University Hospital, Ludwig Maximilians University (LMU), 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
- Division of Neuro-Oncology, Department of Neurosurgery, Ludwig Maximilians University School of Medicine, Marchioninistrasse 15, 81377, Munich, Germany
| | - Joerg-Christian Tonn
- Department of Neurosurgery, LMU University Hospital, Ludwig Maximilians University (LMU), 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | - Marion Subklewe
- German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
- Department of Medicine III, Ludwig-Maximilians-University School of Medicine, Munich, Germany
- Bavarian Cancer Research Center (BZKF), 91054, Erlangen, Germany
| | - Sebastian Kobold
- German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
- Department of Medicine IV, Division of Clinical Pharmacology, LMU University Hospital Munich, Munich, Germany
| | - Patrick N Harter
- German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
- Center for Neuropathology and Prion Research, Faculty of Medicine LMU Munich, Ludwig-Maximilians-University (LMU), Munich, Germany
- Bavarian Cancer Research Center (BZKF), 91054, Erlangen, Germany
| | - Veit R Buchholz
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, 81675, Munich, Germany
| | - Louisa von Baumgarten
- Department of Neurosurgery, LMU University Hospital, Ludwig Maximilians University (LMU), 81377, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany.
- Department of Neurology, LMU University Hospital, Ludwig Maximilians University (LMU), 81377, Munich, Germany.
- Bavarian Cancer Research Center (BZKF), 91054, Erlangen, Germany.
- Division of Neuro-Oncology, Department of Neurosurgery, Ludwig Maximilians University School of Medicine, Marchioninistrasse 15, 81377, Munich, Germany.
| |
Collapse
|
6
|
Guo J, Zhao W, Xiao X, Liu S, Liu L, Zhang L, Li L, Li Z, Li Z, Xu M, Peng Q, Wang J, Wei Y, Jiang N. Reprogramming exosomes for immunity-remodeled photodynamic therapy against non-small cell lung cancer. Bioact Mater 2024; 39:206-223. [PMID: 38827172 PMCID: PMC11141154 DOI: 10.1016/j.bioactmat.2024.05.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 04/11/2024] [Accepted: 05/16/2024] [Indexed: 06/04/2024] Open
Abstract
Traditional treatments against advanced non-small cell lung cancer (NSCLC) with high morbidity and mortality continue to be dissatisfactory. Given this situation, there is an urgent requirement for alternative modalities that provide lower invasiveness, superior clinical effectiveness, and minimal adverse effects. The combination of photodynamic therapy (PDT) and immunotherapy gradually become a promising approach for high-grade malignant NSCLC. Nevertheless, owing to the absence of precise drug delivery techniques as well as the hypoxic and immunosuppressive characteristics of the tumor microenvironment (TME), the efficacy of this combination therapy approach is less than ideal. In this study, we construct a novel nanoplatform that indocyanine green (ICG), a photosensitizer, loads into hollow manganese dioxide (MnO2) nanospheres (NPs) (ICG@MnO2), and then encapsulated in PD-L1 monoclonal antibodies (anti-PD-L1) reprogrammed exosomes (named ICG@MnO2@Exo-anti-PD-L1), to effectively modulate the TME to oppose NSCLC by the synergy of PDT and immunotherapy modalities. The ICG@MnO2@Exo-anti-PD-L1 NPs are precisely delivered to the tumor sites by targeting specially PD-L1 highly expressed cancer cells to controllably release anti-PD-L1 in the acidic TME, thereby activating T cell response. Subsequently, upon endocytic uptake by cancer cells, MnO2 catalyzes the conversion of H2O2 to O2, thereby alleviating tumor hypoxia. Meanwhile, ICG further utilizes O2 to produce singlet oxygen (1O2) to kill tumor cells under 808 nm near-infrared (NIR) irradiation. Furthermore, a high level of intratumoral H2O2 reduces MnO2 to Mn2+, which remodels the immune microenvironment by polarizing macrophages from M2 to M1, further driving T cells. Taken together, the current study suggests that the ICG@MnO2@Exo-anti-PD-L1 NPs could act as a novel drug delivery platform for achieving multimodal therapy in treating NSCLC.
Collapse
Affiliation(s)
- Jiao Guo
- School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, China
| | - Wei Zhao
- School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, China
| | - Xinyu Xiao
- School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, China
| | - Shanshan Liu
- Department of Plastic and Maxillofacial Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Liang Liu
- School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, China
| | - La Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lu Li
- School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, China
| | - Zhenghang Li
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhi Li
- Traditional Chinese Medicine Hospital of Bijie City, Guizhou province, 551700, China
| | - Mengxia Xu
- Traditional Chinese Medicine Hospital of Bijie City, Guizhou province, 551700, China
| | - Qiling Peng
- School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, China
- Bijie Municipal Health Bureau, Guizhou province, 551700, China
- Health Management Center, the Affiliated Hospital of Guizhou Medical University
| | - Jianwei Wang
- School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, China
| | - Yuxian Wei
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ning Jiang
- Department of Pathology, School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, 400016, China
- Department of Pathology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
7
|
Wu W, Guo J, He L, Deng Q, Huang X. Case report: Long-term intracranial effect of zimberelimab monotherapy following surgical resection of high PD-L1-expressing brain metastases from NSCLC. Front Oncol 2024; 14:1390343. [PMID: 38800395 PMCID: PMC11116670 DOI: 10.3389/fonc.2024.1390343] [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: 02/23/2024] [Accepted: 04/24/2024] [Indexed: 05/29/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) accounted for the majority of lung cancer cases worldwide. Brain metastases (BM) frequently complicate NSCLC and portend a dismal prognosis. To control neurological symptoms, surgical resection is commonly followed by brain radiotherapy (RT). However, RT is often complicated by neurotoxicity. For patients with tumors that harbor positive driver genes, tyrosine kinase inhibitors are considered the standard of care. Nevertheless, treatment options for those without driver gene mutations are still debated. Programmed death receptor 1 (PD-1)/ligand 1 (PD-L1) inhibition has emerged as a novel therapeutic strategy for NSCLC patients with PD-L1-positive tumors, as well as for those with asymptomatic BM. However, the effect of anti-PD-1 antibodies on active BM within such specific populations is undetermined. Herein we present a case of a 65-year-old patient with NSCLC and high PD-L1-expressing BM. The patient underwent surgical resection of BM followed by first-line monotherapy with 31 cycles of zimberelimab, a novel anti-PD-1 antibody, and has already achieved 24 months of progression-free survival and intracranial recurrence-free survival. To our knowledge, this is the first report regarding the intracranial effect of zimberelimab on BM from primary lung cancer. This case report might facilitate an understanding of the intracranial effects of different anti-PD-1 antibodies for such populations.
Collapse
Affiliation(s)
- Weijia Wu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jinyou Guo
- Department of Oncology, Yuhuan Second People’s Hospital, Yuhuan, China
| | - Lianxiang He
- Department of Medical Affairs, Guangzhou Gloria Bioscience Co., Ltd., Beijing, China
| | - Qi Deng
- Department of Medical Affairs, Guangzhou Gloria Bioscience Co., Ltd., Beijing, China
| | - Xianping Huang
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
8
|
Karati D, Mukherjee S, Roy S. Deciphering the molecular mechanistic paths describing the chemotherapeutic potential and epigenetic regulation of curcumin in lung cancer: a mini review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2715-2725. [PMID: 37982888 DOI: 10.1007/s00210-023-02838-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 11/03/2023] [Indexed: 11/21/2023]
Abstract
In an uncontrolled inflammatory environment, the complex process of lung carcinogenesis occurs. Lung cancer remains the leading cause of cancer-related mortality worldwide. The average 5-year survival rate is still low despite significant advancements in our knowledge of lung carcinogenesis and the development of innovative therapies in recent decades. Research on adjuvant treatment, lung carcinogenesis pathways, and possible prognostic indicators has to be refocused using an innovative approach. The majority of lung cancers are discovered at an advanced stage when there is little chance of recovery. It has grown in popularity in recent years to supplement already available chemotherapeutic therapies with adjuvant herbal medications, which may lessen toxicity and adverse effects without sacrificing therapeutic efficiency. One such prospective contender is curcumin. In-depth research has been done on curcumin as a multi-target anti-tumor and anti-inflammatory molecule. A pharmacologically active polyphenol produced from turmeric is called curcumin. Over the past few decades, curcumin's therapeutic potential has been thoroughly studied, and data indicate that curcumin may play a part in a variety of biological processes, most notably its potent anticancer activity. Being a pleiotropic chemical, curcumin regulates a variety of molecules that are key players in many cell signaling pathways. It has been shown to stifle transformation, restrain proliferation, and trigger apoptosis. Curcumin can reduce the development of non-small cell LC by downregulating Circular RNA hsa_circ_0007580, which in turn controls the expression of integrin subunit beta 1 by adsorbing miR-384. Nevertheless, despite all these advantages, curcumin's effectiveness is still restricted because of its weak bioavailability, poor absorption within the systemic circulation, and quick removal from the body. In an effort to overcome these constraints, scientists from all around the world are working to develop a synthetic and improved curcuminoid by appropriately altering the parent skeleton structurally. These curcuminoids will simultaneously improve the physicochemical properties and efficacy. This review presents evidence from the most recent clinical trials coupled with the molecular mechanisms of curcumin in LC.
Collapse
Affiliation(s)
- Dipanjan Karati
- Department of Pharmaceutical Technology, School of Pharmacy, Techno India University, Kolkata, 700091, West Bengal, India
| | - Swarupananda Mukherjee
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata, 124 B.L. Saha Road, Kolkata, West Bengal, 700053, India
| | - Souvik Roy
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata, 124 B.L. Saha Road, Kolkata, West Bengal, 700053, India.
| |
Collapse
|
9
|
Parera Roig M, Colomé DC, Colomer GB, Sardo EG, Tournour MA, Fernández SG, Ominetti AI, Juvanteny EP, Polo JLM, Jobal DB, Espejo-Herrera N. Evolution of Diagnoses, Survival, and Costs of Oncological Medical Treatment for Non-Small-Cell Lung Cancer over 20 Years in Osona, Catalonia. Curr Oncol 2024; 31:2145-2157. [PMID: 38668062 PMCID: PMC11049066 DOI: 10.3390/curroncol31040159] [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: 03/11/2024] [Revised: 04/04/2024] [Accepted: 04/06/2024] [Indexed: 04/28/2024] Open
Abstract
Non-small-cell lung cancer (NSCLC) has experienced several diagnostic and therapeutic changes over the past two decades. However, there are few studies conducted with real-world data regarding the evolution of the cost of these new drugs and the corresponding changes in the survival of these patients. We collected data on patients diagnosed with NSCLC from the tumor registry of the University Hospital of Vic from 2002 to 2021. We analyzed the epidemiological and pathological characteristics of these patients, the diverse oncological treatments administered, and the survival outcomes extending at least 18 months post-diagnosis. We also collected data on pharmacological costs, aligning them with the treatments received by each patient to determine the cost associated with individualized treatments. Our study included 905 patients diagnosed with NSCLC. We observed a dynamic shift in histopathological subtypes from squamous carcinoma in the initial years to adenocarcinoma. Regarding the treatment approach, the use of chemotherapy declined over time, replaced by immunotherapy, while molecular therapy showed relative stability. An increase in survival at 18 months after diagnosis was observed in patients with advanced stages over the most recent years of this study, along with the advent of immunotherapy. Mean treatment costs per patient ranged from EUR 1413.16 to EUR 22,029.87 and reached a peak of EUR 48,283.80 in 2017 after the advent of immunotherapy. This retrospective study, based on real-world data, documents the evolution of pathological characteristics, survival rates, and medical treatment costs for NSCLC over the last two decades. After the introduction of immunotherapy, patients in advanced stages showed an improvement in survival at 18 months, coupled with an increase in treatment costs.
Collapse
Affiliation(s)
- Marta Parera Roig
- Oncohematology Unit, Consorci Hospitalari de Vic (University Hospital of Vic), 08500 Vic, Spain; (E.G.S.); (M.A.T.); (S.G.F.); (A.I.O.)
- School of Medicine, University of Vic-Central University of Catalonia (UVIC-UCC), 08500 Vic, Spain; (D.B.J.); (N.E.-H.)
- Doctoral College, Medicine and Biomedical Sciences, University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Spain
- Mechanisms of Disease Laboratory Research Group (MoD Lab), IRIS-CC, University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Spain
| | - David Compte Colomé
- Planning and Information Systems Department, Consorci Hospitalari de Vic (University Hospital of Vic), 08500 Vic, Spain;
| | - Gemma Basagaña Colomer
- Pharmacy Department, Consorci Hospitalari de Vic (University Hospital of Vic), 08500 Vic, Spain;
| | - Emilia Gabriela Sardo
- Oncohematology Unit, Consorci Hospitalari de Vic (University Hospital of Vic), 08500 Vic, Spain; (E.G.S.); (M.A.T.); (S.G.F.); (A.I.O.)
- School of Medicine, University of Vic-Central University of Catalonia (UVIC-UCC), 08500 Vic, Spain; (D.B.J.); (N.E.-H.)
| | - Mauricio Alejandro Tournour
- Oncohematology Unit, Consorci Hospitalari de Vic (University Hospital of Vic), 08500 Vic, Spain; (E.G.S.); (M.A.T.); (S.G.F.); (A.I.O.)
| | - Silvia Griñó Fernández
- Oncohematology Unit, Consorci Hospitalari de Vic (University Hospital of Vic), 08500 Vic, Spain; (E.G.S.); (M.A.T.); (S.G.F.); (A.I.O.)
| | - Arturo Ivan Ominetti
- Oncohematology Unit, Consorci Hospitalari de Vic (University Hospital of Vic), 08500 Vic, Spain; (E.G.S.); (M.A.T.); (S.G.F.); (A.I.O.)
| | - Emma Puigoriol Juvanteny
- Epidemiology Department, Consorci Hospitalari de Vic (University Hospital of Vic), 08500 Vic, Spain;
- Multidisciplinary Inflammation Research Group (MIRG), IRIS-CC, 08500 Vic, Spain
| | - José Luis Molinero Polo
- Pathology Department, Consorci Hospitalari de Vic (University Hospital of Vic), 08500 Vic, Spain;
| | - Daniel Badia Jobal
- School of Medicine, University of Vic-Central University of Catalonia (UVIC-UCC), 08500 Vic, Spain; (D.B.J.); (N.E.-H.)
- Pathology Department, Consorci Hospitalari de Vic (University Hospital of Vic), 08500 Vic, Spain;
| | - Nadia Espejo-Herrera
- School of Medicine, University of Vic-Central University of Catalonia (UVIC-UCC), 08500 Vic, Spain; (D.B.J.); (N.E.-H.)
- Multidisciplinary Inflammation Research Group (MIRG), IRIS-CC, 08500 Vic, Spain
- Pathology Department, Consorci Hospitalari de Vic (University Hospital of Vic), 08500 Vic, Spain;
| |
Collapse
|
10
|
Zywiciel JF, Verm RA, Raad W, Baker M, Freeman R, Abdelsattar ZM. En bloc chest wall resection in locally advanced cT3N2 (stage IIIB) lung cancer involving the chest wall: Revisiting guidelines. JTCVS OPEN 2024; 18:221-231. [PMID: 38690419 PMCID: PMC11056476 DOI: 10.1016/j.xjon.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 11/11/2023] [Accepted: 12/10/2023] [Indexed: 05/02/2024]
Abstract
Objectives Current National Comprehensive Cancer Network guidelines recommend definitive chemoradiation rather than surgery for patients with locally advanced clinical stage T3 and N2 (stage IIIB) lung cancer involving the chest wall. The data supporting this recommendation are controversial. We studied whether surgery confers a survival advantage over definitive chemoradiation in the National Cancer Database. Methods We identified all patients with clinical stage T3 and N2 lung cancer in the National Cancer Database from 2004 to 2017 who underwent a lobectomy with en bloc chest wall resection and compared them with patients with clinical stage T3 and N2 lung cancer who had definitive chemoradiation. We used propensity score matching to minimize confounding by indication while excluding patients with tumors in the upper lobes to exclude Pancoast tumors. We used 1:1 propensity score matching and Kaplan-Meir survival analyses to estimate associations. Results Of 4467 patients meeting all inclusion/exclusion criteria, 210 (4.49%) had an en bloc chest wall resection. Patients undergoing surgical resection were younger (mean age = 60.3 ± 10.3 years vs 67.5 ± 10.4 years; P < .001) and had more adenocarcinoma (59.0% vs 44.5%; P < .001) but were otherwise similar in terms of sex (37.1% female vs 42.0%; P = .167) and race (Whites 84.3% vs 84.0%; P = .276) compared with the definitive chemoradiation group. After resection, there was an unadjusted 30- and 90-day mortality rate of 3.3% and 9.5%, respectively. A substantial survival benefit with surgical resection persisted after propensity score matching (log-rank P < .001). Conclusions In this large observational study, we found that in select patients, en bloc chest wall resection for locally advanced clinical stage T3 and N2 lung cancer was associated with improved survival compared with definitive chemoradiation. National Comprehensive Cancer Network guidelines should be revisited.
Collapse
Affiliation(s)
| | - Raymond A. Verm
- Department of Thoracic & Cardiovascular Surgery, Loyola University Medical Center, Maywood, Ill
| | - Wissam Raad
- Stritch School of Medicine, Loyola University Chicago, Chicago, Ill
- Department of Thoracic & Cardiovascular Surgery, Loyola University Medical Center, Maywood, Ill
| | - Marshall Baker
- Stritch School of Medicine, Loyola University Chicago, Chicago, Ill
- Department of Surgery, Loyola University Medical Center, Maywood, Ill
- Edward Hines, Jr VA Hospital, US Department of Veterans Affairs, Hines, Ill
| | - Richard Freeman
- Stritch School of Medicine, Loyola University Chicago, Chicago, Ill
- Department of Thoracic & Cardiovascular Surgery, Loyola University Medical Center, Maywood, Ill
| | - Zaid M. Abdelsattar
- Stritch School of Medicine, Loyola University Chicago, Chicago, Ill
- Department of Thoracic & Cardiovascular Surgery, Loyola University Medical Center, Maywood, Ill
- Edward Hines, Jr VA Hospital, US Department of Veterans Affairs, Hines, Ill
| |
Collapse
|
11
|
Cheng Z, Xue K, Xiong C, Zheng Z, Li J, Qiao X. MRPS16 promotes lung adenocarcinoma growth via the PI3K/AKT/Frataxin signalling axis. J Cell Mol Med 2024; 28:e18166. [PMID: 38506080 PMCID: PMC10951875 DOI: 10.1111/jcmm.18166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 12/26/2023] [Accepted: 01/24/2024] [Indexed: 03/21/2024] Open
Abstract
Although MRPS16 is involved in cancer development, its mechanisms in developing LAUD remain unclear. Herein, qRT-PCR, WB and IHC were utilized for evaluating MRPS16 expression levels, while functional assays besides animal experiments were performed to measure MRPS16 effect on LAUD progression. Using WB, the MRPS16 effect on PI3K/AKT/Frataxin signalling pathway was tested. According to our study, MRPS16 was upregulated in LAUD and was correlated to the advanced TNM stage as well as poor clinical outcomes, which represent an independent prognostic factor. Based on functional assays, MRPS16 is involved in promoting LAUD growth, migration and invasion, which was validated further in subsequent analyses through PI3K/AKT/Frataxin pathway activation. Moreover, MRPS16-knockdown-mediated Frataxin overexpression was shown to restore the reduction in tumour cells proliferation, migration and invasion. Our results revealed that MRPS16 caused an aggressive phenotype to LAUD and was a poor prognosticator; thus, targeting MRPS16 may be effectual in LAUD treatment.
Collapse
Affiliation(s)
- Zaixing Cheng
- Department of Thoracic SurgeryUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Kaming Xue
- Department of Traditional Chinese MedicineUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Cui Xiong
- Department of EndocrinologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Zhikun Zheng
- Department of Thoracic SurgeryUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Jinsong Li
- Department of Thoracic SurgeryUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Xinwei Qiao
- Department of Thoracic SurgeryUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| |
Collapse
|
12
|
Wang X, Qin ZL, Li N, Jia MQ, Liu QG, Bai YR, Song J, Yuan S, Zhang SY. Annual review of PROTAC degraders as anticancer agents in 2022. Eur J Med Chem 2024; 267:116166. [PMID: 38281455 DOI: 10.1016/j.ejmech.2024.116166] [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: 10/31/2023] [Revised: 01/10/2024] [Accepted: 01/18/2024] [Indexed: 01/30/2024]
Abstract
Following nearly two decades of development, significant advancements have been achieved in PROTAC technology. As of the end of 2022, more than 20 drugs have entered clinical trials, with ARV-471 targeting estrogen receptor (ER) showing remarkable progress by entering phase III clinical studies. In 2022, significant progress has been made on multiple targets. The first reversible covalent degrader designed to target the KRASG12C mutant protein, based on cyclopropionamide, has been reported. Additionally, the activity HDCA1 degrader surpassed submicromolar levels during the same year. A novel FEM1B covalent ligand called EN106 was also discovered, expanding the range of available ligands. Furthermore, the first PROTAC drug targeting SOS1 was reported. Additionally, the first-in-class degraders that specifically target BRD4 isoforms (BRD4 L and BRD4 S) have recently been reported, providing a valuable tool for further investigating the biological functions of these isoforms. Lastly, a breakthrough was also achieved with the first degrader targeting both CDK9 and Cyclin T1. In this review, we aimed to update the PROTAC degraders as potential anticancer agents covering articles published in 2022. The design strategies, degradation effects, and anticancer activities were highlighted, which might provide an updated sight to develop novel PROTAC degraders with great potential as anticancer agents as well as favorable drug-like properties.
Collapse
Affiliation(s)
- Xiao Wang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Zhao-Long Qin
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Na Li
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Mei-Qi Jia
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Qiu-Ge Liu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yi-Ru Bai
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Jian Song
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| | - Shuo Yuan
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China.
| | - Sai-Yang Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China; State Key Laboratory of Esophageal Cancer Prevention &Treatment, Zhengzhou 450001, China.
| |
Collapse
|
13
|
Korucu Aktas P, Baysal I, Yabanoglu-Ciftci S, Lamprecht A, Arica B. Recent progress in drug delivery systems for tyrosine kinase inhibitors in the treatment of lung cancer. Int J Pharm 2024; 650:123703. [PMID: 38092263 DOI: 10.1016/j.ijpharm.2023.123703] [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: 08/16/2023] [Revised: 12/01/2023] [Accepted: 12/10/2023] [Indexed: 12/22/2023]
Abstract
Lung cancer ranks as the second most commonly diagnosed cancer in both men and women worldwide. Despite the availability of diverse diagnostic and treatment strategies, it remains the leading cause of cancer-related deaths globally. The current treatment approaches for lung cancer involve the utilization of first generation (e.g., erlotinib, gefitinib) and second generation (e.g., afatinib) tyrosine kinase inhibitors (TKIs). These TKIs exert their effects by inhibiting a crucial enzyme called tyrosine kinase, which is responsible for cell survival signaling. However, their clinical effectiveness is hindered by limited solubility and oral bioavailability. Nanotechnology has emerged as a significant application in modern cancer therapy. Nanoparticle-based drug delivery systems, including lipid, polymeric, hybrid, inorganic, dendrimer, and micellar nanoparticles, have been designed to enhance the bioavailability, stability, and retention of these drugs within the targeted lung area. Furthermore, these nanoparticle-based delivery systems offer several advantages, such as increased therapeutic efficacy and reduced side effects and toxicity. This review focuses on the recent advancements in drug delivery systems for some of the most important TKIs, shedding light on their potential in improving lung cancer treatment.
Collapse
Affiliation(s)
- Pelinsu Korucu Aktas
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Ipek Baysal
- Vocational School of Health Services, Hacettepe University, Ankara,Turkey
| | | | - Alf Lamprecht
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Germany
| | - Betul Arica
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
14
|
Sayin AZ, Abali Z, Senyuz S, Cankara F, Gursoy A, Keskin O. Conformational diversity and protein-protein interfaces in drug repurposing in Ras signaling pathway. Sci Rep 2024; 14:1239. [PMID: 38216592 PMCID: PMC10786864 DOI: 10.1038/s41598-023-50913-8] [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: 08/14/2023] [Accepted: 12/27/2023] [Indexed: 01/14/2024] Open
Abstract
We focus on drug repurposing in the Ras signaling pathway, considering structural similarities of protein-protein interfaces. The interfaces formed by physically interacting proteins are found from PDB if available and via PRISM (PRotein Interaction by Structural Matching) otherwise. The structural coverage of these interactions has been increased from 21 to 92% using PRISM. Multiple conformations of each protein are used to include protein dynamics and diversity. Next, we find FDA-approved drugs bound to structurally similar protein-protein interfaces. The results suggest that HIV protease inhibitors tipranavir, indinavir, and saquinavir may bind to EGFR and ERBB3/HER3 interface. Tipranavir and indinavir may also bind to EGFR and ERBB2/HER2 interface. Additionally, a drug used in Alzheimer's disease can bind to RAF1 and BRAF interface. Hence, we propose a methodology to find drugs to be potentially used for cancer using a dataset of structurally similar protein-protein interface clusters rather than pockets in a systematic way.
Collapse
Affiliation(s)
- Ahenk Zeynep Sayin
- Department of Chemical and Biological Engineering, College of Engineering, Koc University, Rumeli Feneri Yolu Sariyer, 34450, Istanbul, Turkey
| | - Zeynep Abali
- Graduate School of Science and Engineering, Computational Sciences and Engineering, Koc University, 34450, Istanbul, Turkey
| | - Simge Senyuz
- Graduate School of Science and Engineering, Computational Sciences and Engineering, Koc University, 34450, Istanbul, Turkey
| | - Fatma Cankara
- Graduate School of Science and Engineering, Computational Sciences and Engineering, Koc University, 34450, Istanbul, Turkey
| | - Attila Gursoy
- Department of Computer Engineering, Koc University, 34450, Istanbul, Turkey
| | - Ozlem Keskin
- Department of Chemical and Biological Engineering, College of Engineering, Koc University, Rumeli Feneri Yolu Sariyer, 34450, Istanbul, Turkey.
| |
Collapse
|
15
|
Quirk J, Mac Donnchadha C, Vaantaja J, Mitchell C, Marchi N, AlSaleh J, Dalton B. Future implications of artificial intelligence in lung cancer screening: a systematic review. BJR Open 2024; 6:tzae035. [PMID: 39444460 PMCID: PMC11498893 DOI: 10.1093/bjro/tzae035] [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: 02/12/2024] [Revised: 08/04/2024] [Accepted: 10/05/2024] [Indexed: 10/25/2024] Open
Abstract
Objectives The aim of this study was to systematically review the literature to assess the application of AI-based interventions in lung cancer screening, and its future implications. Methods Relevant published literature was screened using PRISMA guidelines across three databases: PubMed, Scopus, and Web of Science. Search terms for article selection included "artificial intelligence," "radiology," "lung cancer," "screening," and "diagnostic." Included studies evaluated the use of AI in lung cancer screening and diagnosis. Results Twelve studies met the inclusion criteria. All studies concerned the role of AI in lung cancer screening and diagnosis. The AIs demonstrated promising ability across four domains: (1) detection, (2) characterization and differentiation, (3) augmentation of the work of human radiologists, (4) AI implementation of the LUNG-RADS framework and its ability to augment this framework. All studies reported positive results, demonstrating in some cases AI's ability to perform these tasks to a level close to that of human radiologists. Conclusions The AI systems included in this review were found to be effective screening tools for lung cancer. These findings hold important implications for the future use of AI in lung cancer screening programmes as they may see use as an adjunctive tool for lung cancer screening that would aid in making early and accurate diagnosis. Advances in knowledge AI-based systems appear to be powerful tools that can assist radiologists with lung cancer screening and diagnosis.
Collapse
Affiliation(s)
- Joseph Quirk
- Trinity College Dublin School of Medicine, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, D02R590, Ireland
- St James’s Hospital, James’s Street, Dublin 8, Dublin, D08 NHY1, Ireland
| | - Conor Mac Donnchadha
- Trinity College Dublin School of Medicine, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, D02R590, Ireland
- Tallaght University Hospital, Tallaght, Dublin 24, Dublin, D24 NR0A, Ireland
| | - Jonathan Vaantaja
- Trinity College Dublin School of Medicine, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, D02R590, Ireland
- Aintree University Hospital (Liverpool University Hospitals Foundation Trust NHS), Lower Ln, Fazakerley, Liverpool, L9 7AL, United Kingdom
| | - Cameron Mitchell
- Trinity College Dublin School of Medicine, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, D02R590, Ireland
- Vassar Brothers Medical Center, Nuvance Health, 45 Reade Pl, Poughkeepsie, NY 12601, United States
| | - Nicolas Marchi
- Trinity College Dublin School of Medicine, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, D02R590, Ireland
- University Hospitals Plymouth NHS Trust, Derriford Road, Plymouth, Devon, PL6 8DH, United Kingdom
| | - Jasmine AlSaleh
- Trinity College Dublin School of Medicine, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, D02R590, Ireland
- Mubarak Al Kabeer Hospital, Street 103, Jabriya, Kuwait
| | - Bryan Dalton
- Radiology Department, St James’s Hospital, James’s Street, Dublin 8, Dublin, D08 NHY1, Ireland
| |
Collapse
|
16
|
Zhen Y, Xu YB, Deng RY, Li M, Ma MT, Zhou ZG, Meng QJ, Gong YN, Zhao LY, Liu YB. Correlation Analysis between T790M Status and Clinical Characteristics of Patients with EGFR-sensitive Mutation Advanced NSCLC who Progressed after the First Generation and First-line EGFR-TKIs Administration: A Real-world Exploratory Study. Comb Chem High Throughput Screen 2024; 27:845-853. [PMID: 37282652 DOI: 10.2174/1386207326666230606100729] [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: 10/21/2022] [Revised: 04/18/2023] [Accepted: 05/04/2023] [Indexed: 06/08/2023]
Abstract
AIM The present study is to investigate the association between T790M status and clinical characteristics of patients with EGFR-sensitive advanced non-small cell lung cancer (NSCLC) who progressed the initial epidermal growth factor receptor tyrosine kinase inhibitors (EGFRTKIs) administration. METHODS A total of 167 patients with EGFR-sensitive mutations advanced NSCLC who had successful genetic tests and progressed the initial EGFR-TKI treatment were included in this study retrospectively. The clinical and demographic characteristics of these patients were collected, which were manifested as pathological type, metastasis location, initial biopsy method, initial genetic test specimens, and baseline gene mutations status. Correlation analysis between T790M status and these characteristics was performed and prognostic analysis regarding the different subgroups was carried out accordingly. RESULTS The prevalence of secondary T790M after resistance to initial EGFR-TKIs among the 167 patients was 52.7%. Correlation analysis indicated that the median progression-free Survival (PFS) to initial EGFR-TKIs >12 months were more likely to develop secondary T790M in univariate analysis. However, the conclusion failed to show statistically significant in multivariate analysis. Additionally, patients with intracranial progression of initial EGFR-TKIs therapy were associated with secondary EGFR-T790M. However, it should be noted that those whose best overall response was partial response (PR) during the EGFR-TKI therapy were relevant to secondary T790M. Furthermore, The median PFS of the initial EGFR-TKIs administration was longer among patients with T790M positive mutation and patients with PR reaction than those without T790M mutation and patients with stable disease (SD), respectively (median PFS: 13.6 vs. 10.9 months, P=0.023) and (median PFS: 14.0 vs. 10.1 months, P=0.001). CONCLUSION This retrospective study highlighted the real-world evidence that the best efficacy and intracranial progression with initial EGFR-TKIs therapy among patients with advanced NSCLC might be the promising indicators to predict the occurrence of EGFR-T790M. Patients with PR reaction and T790M positive mutation conferred longer PFS of the initial EGFR-TKIs administration. Also, the conclusion should be confirmed in more patients with advanced NSCLC subsequently.
Collapse
Affiliation(s)
- Ye Zhen
- Department of Medical Oncology, The First Affiliated Hospital of Xingtai Medical College, Hebei Province, China
| | - Ying-Bo Xu
- Department of Medical Oncology, The First Affiliated Hospital of Xingtai Medical College, Hebei Province, China
| | - Ruo-Ying Deng
- Department of Medical Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Meng Li
- Department of Medical Oncology, Quyang Cancer Hospital/ Hengzhou Hospital, Baoding, Hebei Province, China
| | - Min-Ting Ma
- Department of Medical Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Zhi-Guo Zhou
- Department of Medical Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Qing-Ju Meng
- Department of Orthopedics, The First Affiliated Hospital of Xingtai Medical College, Hebei Province, China
| | - Ya-Ning Gong
- Department of Medical Oncology, the First Hospital of Xingtai, Hebei Province, China
| | - Li-Yan Zhao
- Department of Medical Oncology, The First Affiliated Hospital of Xingtai Medical College, Hebei Province, China
| | - Yi-Bing Liu
- Department of Medical Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| |
Collapse
|
17
|
Mohammad-Jafari K, Naghib SM, Mozafari MR. Cisplatin-based Liposomal Nanocarriers for Drug Delivery in Lung Cancer Therapy: Recent Progress and Future Outlooks. Curr Pharm Des 2024; 30:2850-2881. [PMID: 39051580 DOI: 10.2174/0113816128304923240704113319] [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: 02/13/2024] [Accepted: 05/23/2024] [Indexed: 07/27/2024]
Abstract
In order to improve the treatment of lung cancer, this paper looks at the development of cisplatinbased liposomal nanocarriers. It focuses on addressing the drawbacks of conventional cisplatin therapy, including systemic toxicity, inadequate tumor targeting, and drug resistance. Liposomes, or spherical lipid vesicles, offer a potentially effective way to encapsulate cisplatin, enhancing its transport and minimizing harmful effects on healthy tissues. The article discusses many liposomal cisplatin formulations, including pH-sensitive liposomes, sterically stabilized liposomes, and liposomes coupled with specific ligands like EGFR antibodies. These novel formulations show promise in reducing cisplatin resistance, optimizing pharmacokinetics, and boosting therapeutic results in the two in vitro and in vivo models. They also take advantage of the Enhanced Permeability and Retention (EPR) effect in the direction of improved tumor accumulation. The study highlights the need for more investigation to move these liposomal formulations from experimental to clinical settings, highlighting their potential to offer less harmful and more effective cancer therapy alternatives.
Collapse
Affiliation(s)
- Kave Mohammad-Jafari
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - M R Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| |
Collapse
|
18
|
Zhang G, Tang Z, Fan S, Li C, Li Y, Liu W, Long X, Zhang W, Zhang Y, Li Z, Wang Z, Chen D, Ouyang G. Synthesis and biological assessment of indole derivatives containing penta-heterocycles scaffold as novel anticancer agents towards A549 and K562 cells. J Enzyme Inhib Med Chem 2023; 38:2163393. [PMID: 36629428 PMCID: PMC9848270 DOI: 10.1080/14756366.2022.2163393] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Herein, a new series of 2-chloro-N-(5-(2-oxoindolin-3-yl)-4H-pyrazol-3-yl) acetamide derivatives containing 1,3,4-thiadiazole (10a-i) and 4H-1,2,4-triazol-4-amine (11a-r) moiety was designed, synthesised as novel anticancer agents. The antiproliferative activity values indicated that compound 10 b stood as the most potent derivative with IC50 values of 12.0 nM and 10 nM against A549 and K562 cells, respectively. Mechanism investigation and docking studies of 10 b indicated that it possessed good apoptosis characteristic and dose-dependent growth arrest of A549 and K562 cells, blocked cell cycle into G2/M phase. Interestingly, 10 b suppressed the growth of A549 and K562 cells via modulation of EGFR and p53-MDM2 mediated pathway.
Collapse
Affiliation(s)
- Guanglong Zhang
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals of Guizhou University, Guiyang, China
| | - Zhenhua Tang
- College of Pharmacy, Guizhou University, Guiyang, China
| | - Sili Fan
- College of Pharmacy, Guizhou University, Guiyang, China
| | - Chengpeng Li
- College of Pharmacy, Guizhou University, Guiyang, China
| | - Yan Li
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals of Guizhou University, Guiyang, China
| | - Weiqin Liu
- College of Pharmacy, Guizhou University, Guiyang, China
| | - Xuesha Long
- College of Pharmacy, Guizhou University, Guiyang, China
| | - Wenjing Zhang
- College of Pharmacy, Guizhou University, Guiyang, China
| | - Yi Zhang
- College of Pharmacy, Guizhou University, Guiyang, China
| | - Zhurui Li
- College of Pharmacy, Guizhou University, Guiyang, China
| | - Zhenchao Wang
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals of Guizhou University, Guiyang, China,College of Pharmacy, Guizhou University, Guiyang, China,Guizhou Engineering Laboratory for Synthetic Drugs, Guizhou University, Guiyang, China,CONTACT Zhenchao Wang
| | - Danping Chen
- College of Pharmacy, Guizhou University, Guiyang, China,Danping Chen
| | - Guiping Ouyang
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals of Guizhou University, Guiyang, China,College of Pharmacy, Guizhou University, Guiyang, China,Guizhou Engineering Laboratory for Synthetic Drugs, Guizhou University, Guiyang, China,Guiping Ouyang
| |
Collapse
|
19
|
Zhang L, Liang J, Qin H, Lv Y, Liu X, Li Z, Chao Z, Jia C, Qin X, Zhang H. Lnc AC016727.1/BACH1/HIF-1 α signal loop promotes the progression of non-small cell lung cancer. J Exp Clin Cancer Res 2023; 42:296. [PMID: 37946265 PMCID: PMC10636976 DOI: 10.1186/s13046-023-02875-y] [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: 07/16/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) have been reported to play vital roles in the development and progression of cancer. However, their biological significance and functional mechanisms in non-small cell lung cancer (NSCLC) are mostly unclear. METHODS We performed RNA-sequencing to predict the differential expression of lncRNAs in clinical NSCLC and paired paracancerous lung tissues. To identify lncRNA expression, quantitative polymerase chain reaction (qPCR) was used. Using both cell and mouse models, We studied lncRNA AC016727.1's function in NSCLC growth and metastasis. Western blot assays, dual luciferase reporter assays, and chromatin immunoprecipitation were used to analyze the functional mechanism of lncRNA AC016727.1. RESULTS Our larger NSCLC cohorts validated that the lncRNA AC016727.1 was upregulated in 94 paired NSCLC tissues and correlated with poor survival. Functionally, lncRNA AC016727.1 downregulation inhibited NSCLC cell proliferation, aerobic glycolysis, EMT, and migration, inducing apoptosis. Conversely, upregulated lncRNA AC016727.1 expression exhibited the opposite effect, promoting NSCLC cell survival. Importantly, lncRNA AC016727.1 knockdown inhibited lung cancer growth and slowed the progression of lung metastasis in nude mouse models. Mechanistically, lncRNA AC016727.1 upregulated BACH1 target gene expression by acting as a sponge for miR-98-5p, thereby functioning as a competing endogenous RNA. The function of lncRNA AC016727.1 is mediated by the miR-98-5p/BACH1 axis in NSCLC cells. Meanwhile, the transcription factor HIF-1α can bind to the promoter and activate lncRNA AC016727.1 transcription. lncRNA AC016727.1 regulates HIF-1α expression via BACH1 in NSCLC and forms the lncRNA AC016727.1/BACH1/HIF-1α signaling loop under hypoxic conditions. CONCLUSION Our study reveals a novel lncRNA AC016727.1/BACH1/HIF-1α signaling loop in the progression of NSCLC under hypoxic conditions, suggesting that lncRNA AC016727.1 could act as a useful biomarker for NSCLC and a new therapeutic target.
Collapse
Affiliation(s)
- Li Zhang
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, 221006, Jiangsu Province, China
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, China
| | - Jingtian Liang
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, 221006, Jiangsu Province, China
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, China
| | - Hao Qin
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, 221006, Jiangsu Province, China
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, China
| | - Yin Lv
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, 221006, Jiangsu Province, China
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, China
| | - Xiucheng Liu
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, 221006, Jiangsu Province, China
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, China
| | - Zhuoqun Li
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, 221006, Jiangsu Province, China
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, China
| | - Zhixiang Chao
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, 221006, Jiangsu Province, China
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, China
| | - Caili Jia
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, 221006, Jiangsu Province, China
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, China
| | - Xichun Qin
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, 221006, Jiangsu Province, China
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, China
| | - Hao Zhang
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, 221006, Jiangsu Province, China.
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, China.
| |
Collapse
|
20
|
Yennurajalingam S, Mott F, Lu Z, Urbauer D, Stanton P, Torres H, Rantanen PA, Davis S, Dev R, Hui D, Bruera E. Perception of subjective lived experiences of individuals with anorexia-cachexia in patients with advanced lung cancer. Asia Pac J Oncol Nurs 2023; 10:100314. [PMID: 38197040 PMCID: PMC10772162 DOI: 10.1016/j.apjon.2023.100314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 10/04/2023] [Indexed: 01/11/2024] Open
Abstract
Objective Cancer cachexia (CC) is a frequent and debilitating syndrome in patients with cancer. It has serious implications for patients, extending beyond physical problems into psychological, and social domains. The objective of our study was to qualitatively understand the experiences related to CC in patients with advanced lung cancer. Methods Patients with advanced lung cancer with anorexia (≤ 37 points on Functional Assessment of Anorexia/Cachexia Treatment-ACS) and weight loss were eligible. Patients participated in semi-structured interviews prior to study treatment (n = 19). Qualitative analysis was conducted using interpretative phenomenological approach. Results Two super-ordinate themes emerged (anorexia and weight loss). Patients reported experiencing distress related to anorexia, weight loss, lack of social eating, worsening function, body image, and eating habits. The encouragement to eat by the family was often distressing to the patient. The treatment recommendations by their oncologist for anorexia and weight loss was felt inadequate. Patients felt that the treatment for CC should improve appetite and weight gain as well as their mood and be independent. Conclusions The findings of the study suggests that anorexia and weight loss results in high levels of distress due to their effects on physical and psychosocial domains. Further studies are needed to better understand the experience of anorexia and weight loss to develop strategies to effectively treat CC. Trial registration NCT03637816.
Collapse
Affiliation(s)
- Sriram Yennurajalingam
- Department of Palliative, Rehabilitation, and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Frank Mott
- Department of Thoracic, Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Zhanni Lu
- Department of Palliative, Rehabilitation, and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Diana Urbauer
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Penny Stanton
- Department of Palliative, Rehabilitation, and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Hilda Torres
- Department of Palliative, Rehabilitation, and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Petra Ann Rantanen
- Department of Palliative, Rehabilitation, and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Sara Davis
- Department of Palliative, Rehabilitation, and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Rony Dev
- Department of Palliative, Rehabilitation, and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - David Hui
- Department of Palliative, Rehabilitation, and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Eduardo Bruera
- Department of Palliative, Rehabilitation, and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| |
Collapse
|
21
|
Wang B, Pei J, Xu S, Liu J, Yu J. System analysis based on glutamine catabolic-related enzymes identifies GPT2 as a novel immunotherapy target for lung adenocarcinoma. Comput Biol Med 2023; 165:107415. [PMID: 37657356 DOI: 10.1016/j.compbiomed.2023.107415] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/31/2023] [Accepted: 08/28/2023] [Indexed: 09/03/2023]
Abstract
BACKGROUND In recent years, targeting glutamine metabolism has gained attention as a promising therapeutic approach. Glutamine catabolic-related enzymes play a crucial role in modulating glutamine metabolism and influencing immune responses in the tumor immune microenvironment (TME). However, current literature on the function of glutamine catabolic enzymes in lung adenocarcinoma (LUAD) is limited. METHODS We validated the glutamine dependency of LUAD cells in vitro, followed by transcriptome data to identify differentially expressed genes (DEGs), with transcriptome and single-cell data analysis utilized to explore the role of such genes within the tumor immune microenvironment. We performed employed subcutaneous injection of lewis lung carcinoma cells in C57BL/6 mice to confirm the role of candidate genes in tumor growth and anti-tumor immunity. RESULTS Our study revealed that glutamine is essential for the growth of LUAD cells. Subsequently, we identified four DEGs - glutamate pyruvate transaminase 1 (GPT1), glutamate pyruvate transaminase 2 (GPT2), glutamic-oxaloacetic transaminase 1 (GOT1), and glutamic-oxaloacetic transaminase 2 (GOT2) - in LUAD patients, which were highly expressed in tumor tissue and associated with an immunosuppressive TME. Single-cell sequencing analysis detected high expression levels of GOT1 and GOT2 in immune and stromal cell subpopulations, while GPT1 and GPT2 showed relatively lower expression. Based on the lower immune score and lower expression in immune and stromal cells, we validated the role of GPT2 in vivo for modulating the TME and tumor growth. Inhibition of GPT2 resulted in suppressed tumor growth and increased the expression of CD4 and CD8. Additionally, GPT2 inhibitors induced a stronger antitumor immunity when used in combination with anti-programmed cell death ligand 1. CONCLUSION This study is the first to show the critical role of glutamine catabolic-related enzymes in the TME, and identified GPT2 as a promising therapeutic target for inhibiting tumor growth and improving anti-tumour immune responses for LUAD. Additional studies will be required to define the roles glutamine catabolic-related enzymes play in LUAD.
Collapse
Affiliation(s)
- Bolin Wang
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences Jinan, Shandong, China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China
| | - Jinli Pei
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences Jinan, Shandong, China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China
| | - Shengnan Xu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences Jinan, Shandong, China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China
| | - Jie Liu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences Jinan, Shandong, China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China.
| | - Jinming Yu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences Jinan, Shandong, China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
22
|
Elasbali AM, Al-Soud WA, Mousa Elayyan AE, Alhassan HH, Danciu C, Elfaki EM, Alharethi SH, Alharbi B, Alanazi HH, Mohtadi ME, Patel M, Adnan M. Antioxidative and ROS-dependent apoptotic effects of Cuscuta reflexa Roxb. stem against human lung cancer: network pharmacology and in vitro experimental validation. J Biomol Struct Dyn 2023; 42:11651-11676. [PMID: 37776015 DOI: 10.1080/07391102.2023.2263889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/21/2023] [Indexed: 10/01/2023]
Abstract
Lung cancer remains a formidable global health challenge, necessitating the exploration of novel therapeutic approaches. This study investigates the potential of Cuscuta reflexa Roxb. stem extract as an anticancer agent against human lung cancer, focusing on its antioxidative and ROS-dependent apoptotic effects. Utilizing a combination of network pharmacology and in-vitro experimental validation, we delineate the multifaceted molecular mechanisms underlying the observed effects. The antioxidant potential of C. reflexa stem extract was evaluated by the 2,2-diphenyl-1-picrylhydrazyl (DPPH•), 2,2-azinobis (3-ethyl-benzothiazoline-6-sulfonic acid) (ABTS•+) and ferric reducing/antioxidant power (FRAP), hydroxyl free radical scavenging, reactive nitrogen oxide scavenging and super oxide anion radical scavenging assays. Furthermore, the antiproliferative and proapoptotic effect of C. reflexa stem extract was evaluated against A549 lung adenocarcinoma cell line using the consecrated sulforhodamine B (SBR) and Annexin V-PI assays. Additionally, the mitochondrial membrane potential (MMP) and the total reactive oxygen species (ROS) estimation assays were performed. As a result, network pharmacology analysis revealed a complex interaction network between the bioactive constituents of C. reflexa and key proteins implicated in lung cancer progression. The C. reflexa stem extract showed dose-dependent antioxidant activity against DPPH• (IC50 - 87.38 µg/mL), reactive nitrogen oxide (IC50 - 318.34 µg/mL), FRAP (IC50 - 359.96 µg/mL), hydroxy free radicals (IC50 - 526.12 µg/mL) than ABTS●+ (IC50 - 698.45 µg/mL) and super oxide anion (IC50 - 892.71 µg/mL) as well as cytotoxic activity against A549 cells (IC50 - 436.80 µg/mL). Observations of morphological features in treated cells have revealed hallmark of apoptosis properties. Furthermore, as a result of treatment with C. reflexa stem extract, ROS generation and mitochondrial depolarization were increased in A549 cells, suggesting that this treatment has significant apoptotic properties. . These findings highlight the potential utility of this natural extract as an innovative therapeutic strategy for lung cancer treatment. The integration of network pharmacology and experimental validation enhances our understanding of the underlying mechanisms and provide the way for further translational research.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Abdelbaset Mohamed Elasbali
- Department of Clinical Laboratory Science, College of Applied Sciences-Qurayyat, Jouf University, Qurayyat, Saudi Arabia
| | - Waleed Abu Al-Soud
- Department of Clinical Laboratory Science, College of Applied Sciences-Sakaka, Jouf University, Sakaka, Saudi Arabia
| | - Afnan Elayyan Mousa Elayyan
- Department of Clinical Laboratory Science, College of Applied Sciences-Qurayyat, Jouf University, Qurayyat, Saudi Arabia
| | - Hassan H Alhassan
- Department of Clinical Laboratory Science, College of Applied Sciences-Sakaka, Jouf University, Sakaka, Saudi Arabia
| | - Corina Danciu
- Department of Pharmacognosy, Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Elyasa Mustafa Elfaki
- Department of Clinical Laboratory Science, College of Applied Sciences-Qurayyat, Jouf University, Qurayyat, Saudi Arabia
| | - Salem Hussain Alharethi
- Department of Biological Science, College of Arts and Science, Najran University, Najran, Saudi Arabia
| | - Bandar Alharbi
- Department of Clinical Laboratory, College of Applied Medical Sciences, University of Hail, Hail, Saudi Arabia
| | - Hamad H Alanazi
- Department of Clinical Laboratory Science, College of Applied Sciences-Qurayyat, Jouf University, Qurayyat, Saudi Arabia
| | | | - Mitesh Patel
- Research and Development Cell, Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Vadodara, India
| | - Mohd Adnan
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| |
Collapse
|
23
|
Tabassum M, Lone BA, Bhat MN, Bhushan A, Banjare N, Manrique E, Gupta P, Mondhe DM, Gupta PN. Apoptotic Potential and Antitumor Efficacy of Trilliumoside A: A New Steroidal Saponin Isolated from Rhizomes of Trillium govanianum. ACS OMEGA 2023; 8:31914-31927. [PMID: 37692233 PMCID: PMC10483520 DOI: 10.1021/acsomega.3c03649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/08/2023] [Indexed: 09/12/2023]
Abstract
Natural product-derived molecules exhibit potential as anticancer agents. Trilliumoside A, a new steroidal saponin, was obtained from rhizomes of Trillium govanianum, and its anticancer activity was investigated in the presented study. Trilliumoside A was investigated in a panel of cell lines, and it exhibited promising cytotoxic activity on the A549 cells (human lung cancer cells) with an IC50 of 1.83 μM. The mechanism of cell death induced by Trilliumoside A in A549 cells and its anticancer potential in murine tumor models (EAC and EAT) were presented in the current research. Trilliumoside A was found to induce apoptosis in A549 cells by increasing the expression of various apoptotic proteins, such as Bax, Puma, cytochrome C, cleaved PARP, and cleaved caspase 3. Additionally, Trilliumoside A regulates the expression of p53, CDK2, and Cyclin A by decreasing the mitochondrial membrane potential, elevating reactive oxygen species, and stopping the growth of A549 cells in the synthesis phase (S) of the cell cycle. Trilliumoside A showed a considerable reduction in the tumor volume, the amount of ascitic fluid, and the total cell number without affecting the body weight of animals. Our results demonstrate that Trilliumoside A inhibits the proliferation of human lung cancer cells by inducing DNA damage, arresting the cell cycle, and activating the mitochondrial signaling pathway. The study demonstrated the potential of Trilliumoside A as a potential anticancer agent.
Collapse
Affiliation(s)
- Misbah Tabassum
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Bashir Ahmad Lone
- Natural
Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Mudasir Nazir Bhat
- Plant
Science and Agrotechnology Division, CSIR-Indian
Institute of Integrative Medicine, Canal Road, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Anil Bhushan
- Natural
Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Nagma Banjare
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Esteban Manrique
- Real
Jardin Botanico-CSIC, Claudio Moyano 1, 28760 Madrid, Spain
| | - Prasoon Gupta
- Natural
Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Dilip M. Mondhe
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Prem N. Gupta
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
24
|
Korucu Aktas P, Baysal I, Yabanoglu-Ciftci S, Arica B. Development and In Vitro Evaluation of Crizotinib-Loaded Lipid-Polymer Hybrid Nanoparticles Using Box-Behnken Design in Non-small Cell Lung Cancer. AAPS PharmSciTech 2023; 24:178. [PMID: 37658977 DOI: 10.1208/s12249-023-02634-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/07/2023] [Indexed: 09/05/2023] Open
Abstract
The goal of the study was to produce, optimize, characterize, and compare crizotinib-loaded lipid-polymer hybrid nanoparticles (CL-LPHNPs), representing a novel contribution to the existing literature, and to determine their anticancer activity in non-small cell lung cancer cells (NSCLC). Box-Behnken design was used to investigate the effect of three independent variables: polymer amount (X1), soy phosphatidylcholine (X2), and DSPE-PEG (X3), on three responses: particle size (Y1), polydispersity index (Y2), and zeta potential (Y3). Different parameters were evaluated on the optimized LPHNP formulations such as encapsulation efficiency, drug release study, transmission electron microscopy (TEM) image analysis, and in vitro cell evaluations. The mean particle size of the optimized formulation is between 120 and 220 nm with a PDI< 0.2 and a zeta potential of -10 to -15 mV. The encapsulation efficiency values of crizotinib-loaded PLGA-LPHNPs (CL-PLGA-LPHNPs) and crizotinib-loaded PCL-LPHNPs (CL-PCL-LPHNPs) were 79.25±0.07% and 70.93±1.81%, respectively. Drug release study of CL-PLGA-LPHNPs and CL-PCL-LPHNPs showed a controlled and sustained release pattern as a result of core-shell type. Additionally, after 48 h, CL-PLGA-LPHNPs and CL-PCL-LPHNPs significantly reduced the viability of NCI-H2228 cells compared to free crizotinib. Moreover, CL-PLGA-LPHNPs and CL-PCL-LPHNPs exhibited a significant decrease in RAS, RAF, MEK, and ERK gene/protein expression levels after 48-h incubation. In conclusion, this pioneering study introduces lipid-polymer hybrid nanoparticles containing crizotinib as a novel treatment approach, uniting the advantages of a polymeric core and a lipid shell. The successful formulation optimization using Box-Behnken design yielded nanoparticles with adjustable size, remarkable stability, high drug loading, and a customizable drug release profile. Extensive investigations of key parameters, including particle size, PDI, ZP, TEM analysis, drug release, EE%, and in vitro evaluations, validate the potential of these nanoparticles. Moreover, the examination of two different polymers, PLGA and PCL, highlights their distinct impacts on nanoparticle performance. This research opens up new prospects for advanced therapeutic interventions with lipid-polymer hybrid nanoparticles.
Collapse
Affiliation(s)
- Pelinsu Korucu Aktas
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, 06100, Ankara, Turkey
| | - Ipek Baysal
- Vocational School of Health Services, Hacettepe University, Ankara, Turkey
| | | | - Betul Arica
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, 06100, Ankara, Turkey.
| |
Collapse
|
25
|
Wei C, Lan X, Qiu M, Cui R, Fu Q, Shinge SAU, Muluh TA, Jiang O. Expanding the role of combined immunochemotherapy and immunoradiotherapy in the management of head and neck cancer (Review). Oncol Lett 2023; 26:372. [PMID: 37965160 PMCID: PMC10641411 DOI: 10.3892/ol.2023.13958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 06/13/2023] [Indexed: 11/16/2023] Open
Abstract
Immunotherapy has become one of the most promising approaches in tumor therapy, and there are numerous associated clinical trials in China. As an immunosuppressive tumor, head and neck squamous cell carcinoma (HNSCC) carries a high mutation burden, making immune checkpoint inhibitors promising candidates in this field due to their unique mechanism of action. The present review outlines a comprehensive multidisciplinary cancer treatment approach and elaborates on how combining immunochemotherapy and immunoradiotherapy guidelines could enhance clinical efficacy in patients with HNSCC. Furthermore, the present review explores the immunology of HNSCC, current immunotherapeutic strategies to enhance antitumor activity, ongoing clinical trials and the future direction of the current immune landscape in HNSCC. Advanced-stage HNSCC presents with a poor prognosis, low survival rates and minimal improvement in patient survival trends over time. Understanding the potential of immunotherapy and ways to combine it with surgery, chemotherapy and radiotherapy confers good prospects for the management of human papillomavirus (HPV)-positive HNSCC, as well as other HPV-positive malignancies. Understanding the immune system and its effect on HNSCC progression and metastasis will help to uncover novel biomarkers for the selection of patients and to enhance the efficacy of treatments. Further research on why current immune checkpoint inhibitors and targeted drugs are only effective for some patients in the clinic is needed; therefore, further research is required to improve the overall survival of affected patients.
Collapse
Affiliation(s)
- Chun Wei
- Department of Oncology, The Second People's Hospital of Neijiang City, Neijiang, Sichuan 641000, P.R. China
| | - Xiaojun Lan
- Department of Oncology, The Second People's Hospital of Neijiang City, Neijiang, Sichuan 641000, P.R. China
| | - Maona Qiu
- Department of Oncology, The Second People's Hospital of Neijiang City, Neijiang, Sichuan 641000, P.R. China
| | - Ran Cui
- Department of Oncology, The First People's Hospital of Neijiang City, Neijiang, Sichuan 641000, P.R. China
| | - Qiuxia Fu
- Department of General Medicine, The People's Hospital of Luzhou City, Luzhou, Sichuan 646000, P.R. China
| | - Shafiu A. Umar Shinge
- Department of Cardiothoracic Surgery, Sun Yat Sen Memorial Hospital, Sun Yat Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Tobias Achu Muluh
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Ou Jiang
- Department of Oncology, The Second People's Hospital of Neijiang City, Neijiang, Sichuan 641000, P.R. China
| |
Collapse
|
26
|
Zhang Y, Gao Z, Pan Z, Fu H, Jiang F, Yan H, Yang B, He Q, Luo P, Xu Z, Yang X. Crizotinib induces pulmonary toxicity by blocking autophagy flux in alveolar epithelial cells. Biochem Pharmacol 2023; 215:115636. [PMID: 37290598 DOI: 10.1016/j.bcp.2023.115636] [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: 02/03/2023] [Revised: 05/29/2023] [Accepted: 05/31/2023] [Indexed: 06/10/2023]
Abstract
Crizotinib is the first-line drug for advanced non-small cell lung cancer with the abnormal expression of anaplastic lymphoma kinase gene. Severe, life-threatening, or fatal interstitial lung disease/pneumonia has been reported in patients treated with crizotinib. The clinical benefit of crizotinib is limited by its pulmonary toxicity, but the underlying mechanisms have not been adequately studied, and protective strategies are relatively scarce. Here, we established an in vivo mouse model in which crizotinib was continuously administered to C57BL/6 at 100 mg/kg/day for 6 weeks and verified that crizotinib induced interstitial lung disease in vivo, which was consistent with the clinical observations. We further treated BEAS-2B and TC-1 cells, the alveolar epithelial cell lines, with crizotinib and found the increased apoptosis rate. We proved that crizotinib-blocked autophagic flux caused apoptosis of the alveolar epithelial cells and then promoted the recruitment of immune cells, suggesting that limited autophagy activity was the key reason for pulmonary injury and inflammation caused by crizotinib. Subsequently, we found that metformin could reduce the macrophage recruitment and pulmonary fibrosis by recovering the autophagy flux, thereby ameliorating impaired lung function caused by crizotinib. In conclusion, our study revealed the mechanism of crizotinib-induced apoptosis of alveolar epithelial cells and activation of inflammation during the onset of pulmonary toxicity and provided a promising therapeutic strategy for the treatment of crizotinib-induced pulmonary toxicity.
Collapse
Affiliation(s)
- Yuanteng Zhang
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China; Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Zizheng Gao
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Zezheng Pan
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Huangxi Fu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Feng Jiang
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Hao Yan
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Bo Yang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, Zhejiang, China; Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China; Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhifei Xu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China.
| | - Xiaochun Yang
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
27
|
Varlı M, Ngo MT, Kim SM, Taş İ, Zhou R, Gamage CD, Pulat S, Park SY, Sesal NC, Hur JS, Kang KB, Kim H. A fatty acid-rich fraction of an endolichenic fungus Phoma sp. suppresses immune checkpoint markers via AhR/ARNT and ESR1. Heliyon 2023; 9:e19185. [PMID: 37662726 PMCID: PMC10474435 DOI: 10.1016/j.heliyon.2023.e19185] [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: 03/27/2023] [Revised: 07/29/2023] [Accepted: 08/15/2023] [Indexed: 09/05/2023] Open
Abstract
Lung cancer has the highest mortality rates worldwide. The disease is caused by environmental pollutants, smoking, and many other factors. Recent treatments include immunotherapeutics, which have shown some success; however, the search for new therapeutics is ongoing. Endolichenic fungi produce a whale of a lot of secondary metabolites, the therapeutic effects of which are being evaluated. Here, we used a crude extract and subfractions of the endolichenic fungus, Phoma sp. (EL006848), isolated from the Pseudevernia furfuracea. It was identified the fatty acid components, palmitic acid, stearic acid, and oleic acid, exist in subfractions E1 and E2. In addition, EL006848 and its fatty acids fractions suppressed benzo[a]pyrene (an AhR ligand)- induced expression of PD-L1 to inhibit the activity of multiple immune checkpoints. E2 subfraction, which had a higher fatty acid content than E1, downregulated expression of AhR/ARNT and several human transcription factors related to ESR1. Moreover, E2 showed a strong inhibitory effect on STAT3 expression and mild effect on NF-kB activity. These results suggest that fatty acids extracted from an endolichenic fungus can exert strong immunotherapeutic effects.
Collapse
Affiliation(s)
- Mücahit Varlı
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Men Thi Ngo
- College of Pharmacy, Sookmyung Women's University, 100 Cheongpa-ro 47 gil, Seoul 04310, Republic of Korea
| | - Seong-Min Kim
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - İsa Taş
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Rui Zhou
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Chathurika D.B. Gamage
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Sultan Pulat
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - So-Yeon Park
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Nüzhet Cenk Sesal
- Faculty of Arts and Sciences, Department of Biology, Marmara University, Istanbul, Turkey
| | - Jae-Seoun Hur
- Korean Lichen Research Institute, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Kyo Bin Kang
- College of Pharmacy, Sookmyung Women's University, 100 Cheongpa-ro 47 gil, Seoul 04310, Republic of Korea
| | - Hangun Kim
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| |
Collapse
|
28
|
Zhao S, Song P, Zhou G, Zhang D, Hu Y. METTL3 promotes the malignancy of non-small cell lung cancer by N6-methyladenosine modifying SFRP2. Cancer Gene Ther 2023; 30:1094-1104. [PMID: 37106069 DOI: 10.1038/s41417-023-00614-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 01/08/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023]
Abstract
This study aimed to investigate the roles of METTL3, a regulator of m6A, in NSCLC. RT-qPCR was applied to determine mRNA of m6A-associated genes and SFRP2, and western blot were used for ZEB1 and MMP9 protein expression. Total m6A level was measured using methylated RNA immunoprecipitation (MeRIP) assay, and RIP was used to access m6A level of SFRP2. Cellular behaviors were detected using CCK-8 and tranwell assays. Xenograft assays were conducted to further verify the roles of METTL3 and SFRP2 in NSCLC. The expression level of METTL3 was higher in NSCLC than normal controls. However, downregulation of METTL3 restrained the proliferation, migration and invasion of NSCLC cells. Enhanced expression of METTL3 caused the inverse consequences. Moreover, SFRP2 was found to be negatively regulated by METTL3. Intriguingly, the anti-tumor functions of METTL3 knockdown in the phenotype of NSCLC cells and xenograft mice were overturned by inhibition of SFRP2. Silencing METTL3 resulted in the enhanced stability of SFRP2. Finally, downregulation of SFRP2 induced by METTL3 activated the Wnt/β-catenin signaling pathway in NSCLC. METTL3 acted as an oncogene in the pathogenesis of NSCLC via suppressing SFRP2 to activate Wnt/β-catenin signaling pathway, indicating that METTL3 might be a promising predictor in NSCLC.
Collapse
Affiliation(s)
- Shu Zhao
- Medical School of Chinese PLA, Chinese PLA General Hospital, 100853, Beijing, China
- Department of Medical Oncology, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, 100853, Beijing, China
| | - Peng Song
- Department of Medical Oncology, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, 100853, Beijing, China
| | - Gang Zhou
- Department of Medical Oncology, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, 100853, Beijing, China
| | - Dong Zhang
- Department of Medical Oncology, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, 100853, Beijing, China.
| | - Yi Hu
- Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, 100071, Beijing, China.
| |
Collapse
|
29
|
Hernandez-Con P, Shults J, Willis AW, Yang YX. Dopamine agonists and risk of lung cancer in patients with restless legs syndrome. Pharmacoepidemiol Drug Saf 2023; 32:726-734. [PMID: 36760024 PMCID: PMC10766437 DOI: 10.1002/pds.5596] [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: 08/23/2022] [Revised: 01/20/2023] [Accepted: 01/28/2023] [Indexed: 02/11/2023]
Abstract
PURPOSE To examine the association between long-term use of dopamine agonists (DAs) and the risk of lung cancer in patients with restless legs syndrome (RLS). METHODS We conducted a retrospective cohort study using Optum Clinformatics® database. We included adults ≥40 years diagnosed with RLS during the study period (1/2006-12/2016). Follow-up started with the first RLS diagnosis and ended on the earliest of: incident diagnosis of lung cancer, end of enrollment in the database or end of the study period. The exposure of interest was cumulative duration of DAs use, measured in a time-varying manner. We constructed a multivariable Cox regression model to estimate HRs and 95% CIs for the association between lung cancer and cumulative durations of DA use, adjusting for potential confounding variables. RESULTS We identified 295 042 patients with a diagnosis of RLS. The mean age of the cohort was 62.9; 66.6% were women and 82.3% were white. The prevalence of any DA exposure was 40.3%. Compared to the reference group (no use and ≤1 year), the crude HRs for lung cancer were 1.16 (95% CI 0.99-1.36) and 1.14 (95% CI 0.86-1.51) for 1-3 years and >3 years of cumulative DA use, respectively. The adjusted HR for lung cancer was 1.05 (95% CI 0.88-1.25) for 1-3 years and 1.02 (95% CI 0.76-1.37) for >3 years of cumulative DA use, respectively. CONCLUSIONS At typical doses for the clinical management of RLS, long-term DA use was not associated with risk of lung cancer.
Collapse
Affiliation(s)
- Pilar Hernandez-Con
- Department of Pharmaceutical Outcomes and Policy, College of Pharmacy, University of Florida, Gainesville, Florida, USA
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Justine Shults
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Roberts Center for Pediatric Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Allison W Willis
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Yu-Xiao Yang
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
30
|
Karaağaç M, Geredeli Ç, Yıldırım MS, Altınok T, Dede İ, İnal A, Zamani AG, Kaya B, Demirkazık A, Artaç M. The XRCC1 and TP53 gene polymorphisms are associated with advanced-stage disease and early distant metastasis at diagnosis in non-small cell lung cancer. J Cancer Res Ther 2023; 19:1248-1254. [PMID: 37787291 DOI: 10.4103/jcrt.jcrt_1657_21] [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] [Indexed: 10/04/2023]
Abstract
Background Studies on single nucleotide polymorphisms (SNPs) in non-small cell lung cancer (NSCLC) suggest that DNA repair capacity may have prognostic implications for disease recurrence and survival. However, there is no study investigating the relationship between SNPs and the risk of metastasis at the time of initial diagnosis in patients with NSCLC. Objective This study aimed to investigate the potential predictive value of SNPs in detecting the risk of metastasis at the time of initial diagnosis and poor prognosis in patients with NSCLC. Material and Methods In this prospective cohort study, we evaluated 275 patients with NSCLC. Analysis of SNPs from peripheral blood cells was performed by a polymerase chain reaction. Excision repair cross-complementing group 1 (ERCC1)- Asn118Asn, excision repair cross-complementing group 2 (ERCC2)-Lys751Gln, X-ray repair cross-complementing group 1 (XRCC1)-Arg399Gln, and tumor protein 53 (TP53)-Arg72Pro polymorphisms were evaluated in conjunction with the development of metastasis. Results The ERCC1 normal genotype, ERCC2 heterozygote genotype, XRCC1 normal genotype, and TP53 normal genotype were associated with a higher stage and more advanced-stage disease at the time of initial diagnosis (P = 0.027, 0.005, <0.001, and 0.006, respectively). Also, XRCC1 normal genotype and TP53 normal genotype were associated with the risk of metastasis at the time of initial diagnosis (P = <0.001 and 0.002, respectively). Moreover, the XRCC1 normal genotype was associated with the risk of brain metastasis at the time of initial diagnosis (P = 0.031). Conclusions We showed that SNPs are related to a higher stage and more advanced-stage disease at the time of initial diagnosis in patients with NSCLC, and XRCC1 and TP53 gene polymorphisms are associated with the risk of metastasis. These results may contribute to the identification of high-risk groups and may help to earlier diagnosis and treatment in patients with NSCLC.
Collapse
Affiliation(s)
- Mustafa Karaağaç
- Department of Medical Oncology, Meram Medical Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Çağlayan Geredeli
- Department of Medical Oncology, Meram Medical Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Mahmut Selman Yıldırım
- Department of Medical Oncology, Meram Medical Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Tamer Altınok
- Department of Thoracic Surgery, Meram Medical Faculty, Necmettin Erbakan University, Konya, Turkey
| | - İsa Dede
- Department of Medical Oncology, Medical Faculty, Ankara University, Ankara, Turkey
| | - Ali İnal
- Department of Medical Oncology, Medical Faculty, Dicle University, Diyarbakir, Turkey
| | - Ayşe Gül Zamani
- Department of Medical Oncology, Meram Medical Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Buğra Kaya
- Department of Nuclear Medicine, Meram Medical Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Ahmet Demirkazık
- Department of Medical Oncology, Medical Faculty, Ankara University, Ankara, Turkey
| | - Mehmet Artaç
- Department of Medical Oncology, Meram Medical Faculty, Necmettin Erbakan University, Konya, Turkey
| |
Collapse
|
31
|
Yang X, Ling L, Li C, Hu T, Zhou C, Chen J, Wang Y, Hu L. STAMBPL1 promotes the progression of lung adenocarcinoma by inhibiting DHRS2 expression. Transl Oncol 2023; 35:101728. [PMID: 37393834 DOI: 10.1016/j.tranon.2023.101728] [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: 04/17/2023] [Revised: 06/04/2023] [Accepted: 06/19/2023] [Indexed: 07/04/2023] Open
Abstract
BACKGROUND Lung cancer is responsible for the majority of cancer deaths in the world. We found a significant increase of STAMBPL1 expression in lung adenocarcinoma (LUAD) tissues and cells. However, its mechanism has not been clarified. METHODS LUAD tissues and adjacent normal tissues were collected from 62 patients treated in the First Affiliated Hospital of Wenzhou Medical University from August 2018 to August 2021. In vivo, the clinical data and STAMBPL1 expression of 62 patients with LUAD were analyzed by qPCR. In vitro, cell experiments were carried out after STAMBPL1 knockdown in A549 and H1299 cells to determine cell growth, migration rate, evasiveness, colony-forming ability, and apoptosis. Gene sequencing was used to explore the expression of various genes in A549 and H1299 cells to verify that DHRS2 was up-regulated after STAMBPL1 knockdown; cell experiments further detected the role of the DHRS2 gene after DHRS2 overexpression in A549 and H1299 cells. A rescue experiment was conducted to certify that STAMBPL1 promotes NSCLC progression by regulating DHRS2 expression. RESULTS After STAMBPL1 knockdown by siRNA. Migration, invasion, colony formation, and proliferation of siRNA groups were suppressed than those of NC groups in A549 and H1299 cells, while the cell apoptosis rate of siRNA groups increased significantly. By using gene-sequence analysis, we found that the expression level of the DHRS2 gene was up-regulated in STAMBPL1 siRNA groups, compared with STAMBPL1 NC (negative control) groups in A549 and H1299, which was verified by qPCR and WB. Further experiments showed that the DHRS2 OE group was suppressed in cell proliferation, migration, and invasion in the A549 and H1299 cell lines compared to the DHRS2 NC group, while DHRS2 OE group was significantly enhanced in the cell apoptosis in the A549 and H1299 cell lines. According to the rescue experiment, cell proliferation, migration, and invasion of the STAMBPL1 SI+DHRS2 SI group were enhanced compared with the STAMBPL1 SI+DHRS2 NC group in A549 and H1299 cells, while the STAMBPL1 SI+DHRS2 OE group were further decreased. CONCLUSIONS The expression of STAMBPL1 mRNA is significantly up-regulated in LUAD, promoting the progression of LUAD by down-regulating the expression of DHRS2 and acting as a potential biomarker of LUAD.
Collapse
Affiliation(s)
- Xiang Yang
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, PR China
| | - Liqun Ling
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, PR China
| | - Changhong Li
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, PR China
| | - Tianqi Hu
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, PR China
| | - Chenkang Zhou
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, PR China
| | - Jian Chen
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, PR China
| | - Yumin Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, PR China.
| | - Lijuan Hu
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, PR China.
| |
Collapse
|
32
|
Zhou H, Sun D, Tao J, Xu M, Zhang X, Hou H. Role of YES1 signaling in tumor therapy resistance. CANCER INNOVATION 2023; 2:210-218. [PMID: 38089407 PMCID: PMC10686156 DOI: 10.1002/cai2.51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/06/2022] [Accepted: 12/28/2022] [Indexed: 10/15/2024]
Abstract
YES proto-oncogene 1 (YES1) is an SRC family kinase (SFK) that plays a key role in cancer cell proliferation, adhesion, invasion, survival, and angiogenesis during tumorigenesis and tumor development. Reports suggest that YES1 amplification is associated with resistance to chemotherapeutic drugs and tyrosine kinase inhibitors (TKIs) in human malignancies. However, the mechanisms of drug resistance have not been fully elucidated. In this article, we review the literature on YES1 and discuss the implications of YES1 signaling for targeted therapy and chemotherapy resistance in malignancies. Moreover, recent advances in targeted therapy for YES1-amplified malignancies are summarized. Finally, we conclude that targeting YES1 may reverse drug resistance and serve as a valuable tumor treatment strategy.
Collapse
Affiliation(s)
- Hai Zhou
- Precision Medicine Center of OncologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| | - Dantong Sun
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Junyan Tao
- Qingdao Sixth People's HospitalQingdaoShandongChina
| | - Mingjin Xu
- Department of Radiation OncologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| | - Xiaochun Zhang
- Precision Medicine Center of OncologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| | - Helei Hou
- Precision Medicine Center of OncologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| |
Collapse
|
33
|
Hazer S, Gülhan SŞE, Solak N, Yenibertiz D, Akıllı MS, Sayilir Guven E, Bıçakçıoğlu P. The Effect of Prognostic Nutritional Index in Postoperative Infection Following Lobectomy in Non-Small Cell Lung Cancer Patients. Cureus 2023; 15:e37611. [PMID: 37197130 PMCID: PMC10184591 DOI: 10.7759/cureus.37611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2023] [Indexed: 05/19/2023] Open
Abstract
Background The prognostic nutritional index (PNI) is a valuable marker for evaluating the nutritional status associated with postoperative complications and the prognosis of patients with cancer. However, the role and clinical value of PNI in infection after lung cancer surgery remains unclear. This study examined the association between PNI and infection after lobectomy for lung cancer, focusing on the predictive value of PNI. Methods We conducted this retrospective cohort study on 139 patients with non-small cell lung cancer (NSCLC) who underwent surgery between September 2013 and December 2018. Two groups were composed according to their PNI values (≥ 50 or <50 ), and the relationship was assessed with infection after lobectomy. Results Only PNI values, atelectasis, and prolonged air leaks were significantly associated with the development of infection. The median preoperative PNI was 52.97±5.69. Postoperative infection was seen in patients (15.5%) with PNI≥ 50 and 38.1% in patients with PNI <50. The mean PNI in patients with postoperative infection, empyema, and prolonged air leakage was lower than in patients without these conditions. Conclusions Malnutrition is commonly seen in patients with malignancy. The overall malnutrition rate is 45% in lung cancer patients. Patients with metastatic diseases are malnourished in a 73% ratio compared to 5% for localized diseases. Furthermore, malnutrition increases the tendency of postoperative infection and reduces wound healing. We aim to determine whether PNI can be a predictive index marker for postoperative infection in patients with NSCLC who underwent lobectomy. Postoperative infection was seen in 15.5% of patients with PNI>50 and 38.1% in patients with PNI <50.
Collapse
Affiliation(s)
- Seray Hazer
- Department of Thoracic Surgery, Ankara Atatürk Sanatoryum Training and Research Hospital, Ankara, TUR
| | - Selim Şakir Erkmen Gülhan
- Department of Thoracic Surgery, Ankara Atatürk Sanatoryum Training and Research Hospital, Ankara, TUR
| | - Necati Solak
- Department of Thoracic Surgery, Dr. Nafiz Korez Sincan State Hospital, Ankara, TUR
| | - Derya Yenibertiz
- Department of Pulmonary Medicine, Ankara Atatürk Sanatoryum Training and Research Hospital, Ankara, TUR
| | - Mahmut Sami Akıllı
- Emergency Department, Ankara Atatürk Sanatoryum Training and Research Hospital, Ankara, TUR
| | - Ebru Sayilir Guven
- Department of Thoracic Surgery, Ankara Atatürk Sanatoryum Training and Research Hospital, Ankara, TUR
| | - Pınar Bıçakçıoğlu
- Department of Thoracic Surgery, Ankara Atatürk Sanatoryum Training and Research Hospital, Ankara, TUR
| |
Collapse
|
34
|
Sun Z, Han J, Wang J. Circular RNA PIP5K1A promotes glycolysis and malignancy of non-small cell lung cancer via miR-656-3p/GBE1 axis under hypoxia. Mol Cell Toxicol 2023. [DOI: 10.1007/s13273-023-00343-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
|
35
|
Tang H, Huang H, Guo Z, Huang H, Niu Z, Ji Y, Zhang Y, Bian H, Hu W. Heavy Ion-Responsive lncRNA EBLN3P Functions in the Radiosensitization of Non-Small Cell Lung Cancer Cells Mediated by TNPO1. Cancers (Basel) 2023; 15:cancers15020511. [PMID: 36672460 PMCID: PMC9856274 DOI: 10.3390/cancers15020511] [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: 11/30/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
In recent decades, the rapid development of radiotherapy has dramatically increased the cure rate of malignant tumors. Heavy-ion radiotherapy, which is characterized by the "Bragg Peak" because of its excellent physical properties, induces extensive unrepairable DNA damage in tumor tissues, while normal tissues in the path of ion beams suffer less damage. However, there are few prognostic molecular biomarkers that can be used to assess the efficacy of heavy ion radiotherapy. In this study, we focus on non-small cell lung cancer (NSCLC) radiotherapy and use RNA sequencing and bioinformatic analysis to investigate the gene expression profiles of A549 cells exposed to X-ray or carbon ion irradiation to screen the key genes involved in the stronger tumor-killing effect induced by carbon ions. The potential ceRNA network was predicted and verified by polymerase chain amplification, western blotting analysis, colony formation assay, and apoptosis assay. The results of the experiments indicated that lncRNA EBLN3P plays a critical role in inhibiting carbon ion-induced cell proliferation and inducing apoptosis of NSCLC cells. These functions were achieved by the EBLN3P/miR-144-3p/TNPO1 (transportin-1) ceRNA network. In summary, the lncRNA EBLN3P functions as a ceRNA to mediate lung cancer inhibition induced by carbon ion irradiation by sponging miR-144-3p to regulate TNPO1 expression, indicating that EBLN3P may be a promising target for increasing the treatment efficacy of conventional radiotherapy for NSCLC.
Collapse
Affiliation(s)
- Haoyi Tang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Hao Huang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Zi Guo
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Haitong Huang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Zihe Niu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yi Ji
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yuyang Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Huahui Bian
- Nuclear and Radiation Incident Medical Emergency Office, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
- Correspondence: (H.B.); (W.H.)
| | - Wentao Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
- Correspondence: (H.B.); (W.H.)
| |
Collapse
|
36
|
Zhong M, Chalbatani GM, Deng M, Li Q, Gharagouzloo E, Hamblin MR, Suarez ER, Hu L, Wang D. Functional characterization and development of novel human kinase insert domain receptor chimeric antigen receptor T-cells for immunotherapy of non-small cell lung cancer. Eur J Pharm Sci 2023; 180:106331. [PMID: 36384203 DOI: 10.1016/j.ejps.2022.106331] [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: 04/07/2022] [Revised: 11/10/2022] [Accepted: 11/12/2022] [Indexed: 11/15/2022]
Abstract
CAR-T cell therapy, in which T cells are transfected or transduced with a chimeric antigen receptor (CAR), is a transformative type of cancer immunotherapy. Despite outstanding success in hematological malignancies, their efficacy against solid tumors has been limited. Here, we aimed to explore whether T cells modified by a CAR targeting the vascular endothelial growth factor 2 receptor/ kinase insert domain receptor (KDR) could destroy tumors and their vasculature. A second-generation KDR-CAR was constructed and transfected into T cells using lentivirus. The 3D structure of the CAR construct and target antigen was predicted. Moreover, in silico analysis, including molecular docking and molecular dynamics (MD) simulation, were used to evaluate the minimum energy of interaction and stability of the complex. The anti-cancer effect of KDR-specific CAR-T cells was tested with KDR-expressing and KDR overexpressing A549 cell line. The in-silico study suggested that this CAR construct could be effective for lung cancer therapy. We evaluated this using both in vitro and in vivo experiments. The KDR-CAR-T cells targeted and killed KDR-A549 with high efficiency by expressing IFN-γ and releasing granzyme B. The in vivo study showed that KDR-CAR-T cells dramatically inhibited the growth of lung cancer KDR-A549 xenografts in BALB/c-nu mice at day 10. The characterization of T cells modified by KDR-CAR by computational biology and wet-lab experiments suggested its applicability as a new treatment strategy for lung cancer and, potentially, for other vascularized solid tumors.
Collapse
Affiliation(s)
- Ming Zhong
- Department of Biopharmaceuticals, School of Life Science and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, China; School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | | | - Meifang Deng
- Department of Biopharmaceuticals, School of Life Science and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, China; School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Qiuyi Li
- Department of Biopharmaceuticals, School of Life Science and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, China
| | - Elahe Gharagouzloo
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Science, Tehran, Iran
| | - Michael R Hamblin
- Laser Research Centre, University of Johannesburg, Doornfontein 2028, South Africa
| | - Eloah Rabello Suarez
- Center for Natural and Human Sciences, Federal University of ABC, Santo Andre, SP 09210-580, Brazil
| | - Lili Hu
- Department of Biopharmaceuticals, School of Life Science and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangdong Pharmaceutical University, Guangzhou, China.
| | - Dingding Wang
- Department of Biopharmaceuticals, School of Life Science and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
37
|
García-Cuellar CM, Hernández-Delgadillo R, Torres-Betancourt JA, Solis-Soto JM, Meester I, Sánchez-Pérez Y, Pineda-Aguilar N, Nakagoshi-Cepeda SE, Sánchez-Nájera RI, Nakagoshi-Cepeda MAA, Chellam S, Cabral-Romero C. Cumulative antitumor effect of bismuth lipophilic nanoparticles and cetylpyridinium chloride in inhibiting the growth of lung cancer. J Appl Biomater Funct Mater 2023; 21:22808000231161177. [PMID: 36942951 DOI: 10.1177/22808000231161177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023] Open
Abstract
OBJECTIVE To determine the combined antitumor effect of bismuth lipophilic nanoparticles (BisBAL NP) and cetylpyridinium chloride (CPC) on human lung tumor cells. MATERIAL AND METHODS The human lung tumor cells A549 were exposed to 1-100 µM BisBAL NP or CPC, either separately or in a 1:1 combination. Cell viability was measured with the PrestoBlue assay, the LIVE/DEAD assay, and fluorescence microscopy. The integrity and morphology of cellular microtubules were analyzed by immunofluorescence. RESULTS A 24-h exposure to 1 µM solutions reduced A549 growth with 21.5% for BisBAL NP, 70.5% for CPC, and 92.4% for the combination (p < 0.0001), while a 50 µM BisBAL NP/CPC mixture inhibited cell growth with 99% (p < 0.0001). BisBAL NP-curcumin conjugates were internalized within 30 min of exposure and could be traced within the nucleus of tumor cells within 2 h. BisBAL NP, but not CPC, interfered with microtubule organization, thus interrupting cell replication, similar to the action mechanism of docetaxel. CONCLUSION The growth inhibition of A549 human tumor cells by BisBAL NP and CPC was cumulative as of 1 µM. The BisBAL NP/CPC combination may constitute an innovative and cost-effective alternative for treating human lung cancer.
Collapse
Affiliation(s)
| | - Rene Hernández-Delgadillo
- Laboratorio de Biología Molecular, Facultad de Odontología, Universidad Autónoma de Nuevo León, UANL, Monterrey, Nuevo León, México
| | | | - Juan Manuel Solis-Soto
- Laboratorio de Biología Molecular, Facultad de Odontología, Universidad Autónoma de Nuevo León, UANL, Monterrey, Nuevo León, México
| | - Irene Meester
- Departamento de Ciencias Básicas, Universidad de Monterrey, San Pedro Garza García, México
| | - Yesennia Sánchez-Pérez
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Ciudad de México, México
| | - Nayely Pineda-Aguilar
- Centro de Investigación en Materiales Avanzados, S.C. (CIMAV), Unidad Monterrey, Nuevo León, México
| | - Sergio Eduardo Nakagoshi-Cepeda
- Laboratorio de Biología Molecular, Facultad de Odontología, Universidad Autónoma de Nuevo León, UANL, Monterrey, Nuevo León, México
| | - Rosa Isela Sánchez-Nájera
- Laboratorio de Biología Molecular, Facultad de Odontología, Universidad Autónoma de Nuevo León, UANL, Monterrey, Nuevo León, México
| | | | | | - Claudio Cabral-Romero
- Laboratorio de Biología Molecular, Facultad de Odontología, Universidad Autónoma de Nuevo León, UANL, Monterrey, Nuevo León, México
| |
Collapse
|
38
|
Li J, Du Q, Sun J, Xiang L, Wang S. Identification and validation of a novel phagocytosis regulators-related signature with potential prognostic and immunotherapeutic value in patients with lung adenocarcinoma. Front Oncol 2022; 12:988332. [PMID: 36408131 PMCID: PMC9666737 DOI: 10.3389/fonc.2022.988332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/21/2022] [Indexed: 02/22/2024] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is a malignant tumor that seriously affects the prognosis of patients. Tumor-associated macrophages (TAMs) play a vital role in the tumor microenvironment and can be used as a potential target for tumor therapy, and phagocytosis regulators (PRs) are particularly important in this process. However, the PRs-related signature that can predict the potential prognostic and immunotherapeutic value in patients with LUAD has not been discovered. METHODS In this study, we mainly analyzed the effect of phagocytosis regulators on the prognosis of LUAD, and based on multiple screening analyses including differential analysis, univariate Cox analysis, and Lasso analysis, we constructed a prognostic risk model consisting of five genes. To verify the stability of the model, survival analysis and ROC curve verification were carried out through multiple data sets. In addition, we also combined many factors, such as immune infiltrating cells, clinical grouping characteristics, immune examination sites, pro-inflammatory factors, and other factors as well as in vitro cell experiments and clinical tissue samples for further validation analysis. RESULTS After identifying 29 differentially expressed PRs in LUAD samples, we further constructed a prognostic model consisting of five prognostic signatures (FURIN, KIF23, SASH3, GNPNAT1, and ITGAL). Further survival analysis tests, ROC verification, as well as univariate and multivariate Cox regression analysis showed that the risk score of the model could well predict the prognosis of LUAD patients and could be used as an independent prognostic factor. In addition, we further found that these phagocytic regulators-related signatures were closely related to the immune microenvironment and immunotherapy in LUAD patients, and could well predict the efficacy of immunotherapy in patients. In vitro cell experiments and Immunohistochemistry of clinical tissues showed that the expressions of FURIN, KIF23, SASH3, GNPNAT1 and ITGAL in normal lung cells/tissues and LUAD cells/tissues were consistent with bioinformatics results, and 3 of them had significant differences. CONCLUSION Our study identified a novel PRs-related signature that has potential application value in predicting the prognosis of LUAD patients and predicting the efficacy of immunotherapy. This provides a new basis for the prognosis assessment of LUAD patients and provides a novel target for immunotherapy of LUAD patients.
Collapse
Affiliation(s)
- Jingyang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qinyun Du
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiayi Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Xiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shaohui Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
39
|
S. M. S, Naveen NR, Rao GSNK, Gopan G, Chopra H, Park MN, Alshahrani MM, Jose J, Emran TB, Kim B. A spotlight on alkaloid nanoformulations for the treatment of lung cancer. Front Oncol 2022; 12:994155. [PMID: 36330493 PMCID: PMC9623325 DOI: 10.3389/fonc.2022.994155] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/12/2022] [Indexed: 07/30/2023] Open
Abstract
Numerous naturally available phytochemicals have potential anti-cancer activities due to their vast structural diversity. Alkaloids have been extensively used in cancer treatment, especially lung cancers, among the plant-based compounds. However, their utilization is limited by their poor solubility, low bioavailability, and inadequacies such as lack of specificity to cancer cells and indiscriminate distribution in the tissues. Incorporating the alkaloids into nanoformulations can overcome the said limitations paving the way for effective delivery of the alkaloids to the site of action in sufficient concentrations, which is crucial in tumor targeting. Our review attempts to assess whether alkaloid nanoformulation can be an effective tool in lung cancer therapy. The mechanism of action of each alkaloid having potential is explored in great detail in the review. In general, Alkaloids suppress oncogenesis by modulating several signaling pathways involved in multiplication, cell cycle, and metastasis, making them significant component of many clinical anti-cancerous agents. The review also explores the future prospects of alkaloid nanoformulation in lung cancer. So, in conclusion, alkaloid based nanoformulation will emerge as a potential gamechanger in treating lung cancer in the near future.
Collapse
Affiliation(s)
- Sindhoor S. M.
- Department of Pharmaceutics, P.A. College of Pharmacy, Mangalore, Karnataka, India
| | - N. Raghavendra Naveen
- Department of Pharmaceutics, Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University, B. G. Nagar, Karnataka, India
| | - GSN Koteswara Rao
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Gopika Gopan
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Sciences, Nitte (Deemed to be University), Mangalore, Karnataka, India
| | - Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Moon Nyeo Park
- Department of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Mohammed Merae Alshahrani
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Jobin Jose
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Sciences, Nitte (Deemed to be University), Mangalore, Karnataka, India
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Bonglee Kim
- Department of Korean Medicine, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
40
|
Barretta P, Ponte F, Scoditti S, Vigna V, Mazzone G, Sicilia E. Computational Analysis of the Behavior of BODIPY Decorated Monofunctional Platinum(II) Complexes in the Dark and under Light Irradiation. J Phys Chem A 2022; 126:7159-7167. [PMID: 36194386 PMCID: PMC9574924 DOI: 10.1021/acs.jpca.2c04544] [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] [Indexed: 11/28/2022]
Abstract
Dual-action drugs are occupying an important place in the scientific landscape of cancer research owing to the possibility to combine different therapeutic strategies into a single molecule. In the present work, the behavior of two BODIPY-appended monofunctional Pt(II) complexes, one mononuclear and one binuclear, recently synthesized and tested for their cytotoxicity have been explored both in the dark and under light irradiation. Quantum mechanical DFT calculations have been used to carry out the exploration of the key steps, aquation and guanine attack, of the mechanism of action of Pt(II) complexes in the dark. Due to the presence of the BODIPY chromophore and the potential capability of the two investigated complexes to work as photosensitizers in PDT, time dependent DFT has been employed to calculate their photophysical properties and to inspect how the sensitizing properties of BODIPY are affected by the presence of the platinum "heavy atom". Furthermore, also the eventual influence on of the photophysical properties due to the displacement of chlorido ligands by water and of water by guanine has been taken into consideration.
Collapse
Affiliation(s)
- Pierraffaele Barretta
- Department of Chemistry and Chemical Technologies, University of Calabria, Ponte P. Bucci, 87036 Arcavacata di Rende (CS), Italy
| | - Fortuna Ponte
- Department of Chemistry and Chemical Technologies, University of Calabria, Ponte P. Bucci, 87036 Arcavacata di Rende (CS), Italy
| | - Stefano Scoditti
- Department of Chemistry and Chemical Technologies, University of Calabria, Ponte P. Bucci, 87036 Arcavacata di Rende (CS), Italy
| | - Vincenzo Vigna
- Department of Chemistry and Chemical Technologies, University of Calabria, Ponte P. Bucci, 87036 Arcavacata di Rende (CS), Italy
| | - Gloria Mazzone
- Department of Chemistry and Chemical Technologies, University of Calabria, Ponte P. Bucci, 87036 Arcavacata di Rende (CS), Italy
| | - Emilia Sicilia
- Department of Chemistry and Chemical Technologies, University of Calabria, Ponte P. Bucci, 87036 Arcavacata di Rende (CS), Italy
| |
Collapse
|
41
|
Zhang H, Wang R, Deng Q. miR-29b Regulates Lung Cancer Progression by Downregulating FEM1B and Inhibiting the FOX01/AKT Pathway. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:3110330. [PMID: 36003920 PMCID: PMC9393195 DOI: 10.1155/2022/3110330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/30/2022] [Indexed: 12/24/2022]
Abstract
Purpose Lung cancer is a relatively common type of cancer, and the incidence rate has been on the rise in recent years. MicroRNAs are a class of endogenous small RNA molecules, which are essential for the posttranscriptional regulation of genes. miR-29b is closely related to the occurrence and development of tumors, including prostate cancer, colon cancer, and breast cancer. However, few studies have been performed to explore the expression and pathway of miR-29b in non-small-cell lung cancer (NSCLC). Methods Using bioinformatics analysis, we found that patients with low relative expression of the miR-29b gene have a low long-term survival rate. The results of in vitro research showed that when miR-29b expression was upregulated, the invasion, migration, and proliferation of A549 and NCI-H-1792 cells was inhibited, and the apoptosis was accelerated. Results The results showed that FEM1B is a miR-29b target gene, and the expressions of FEM1B and miR-29b were negatively correlated. Like the upregulation of miR-29b expression, silencing the FEM1B expression could also impair the invasion, migration, and proliferation abilities of A549 and NCI-H-1792 cells. When FEM1B expression was restored, the inhibitory effect of miR-29b could be reversed. Reverse transcription-polymerase chain reaction (RT-PCR) and western blot (WB) analysis showed that overexpression of miR-29b could inhibit the expression of FEM1B, AKT, vascular endothelial growth factor (VEGF), and Sirt3 in A549 and NCI-H-1792 cells and upregulate the expression of FOXO1 protein. Conclusion The results of this study indicate that miR-29b inhibits the proliferation and deterioration of NSCLC cells by targeting FEM1B and inhibiting the activation of the FOXO1/AKT pathway. miR-29b may become a new target for the clinical diagnosis and treatment of lung cancer, and it is expected to become a new inhibitor of NSCLC.
Collapse
Affiliation(s)
- Huanrong Zhang
- Department of Thoracic Surgery, Meizhou People's Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou, China
| | - Rong Wang
- Department of Thoracic Surgery, Meizhou People's Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou, China
| | - Qiuhua Deng
- The Translational Medicine Laboratory, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
42
|
Circ_001042 Inhibits TGF-β1/P38 MAPK Signaling Axis-Mediated Epithelial-Mesenchymal Transition and Metastasis in Lung Adenocarcinoma. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7890490. [PMID: 35966723 PMCID: PMC9371840 DOI: 10.1155/2022/7890490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/29/2022] [Accepted: 07/09/2022] [Indexed: 12/24/2022]
Abstract
Objective To explore the role and molecular mechanism of circ_001042 in lung adenocarcinoma (LUAD). Methods The expression level of circ_001042 and linear RNA MRPS35 in cells and clinical tissues was detected by real-time PCR (qRT-PCR). The expression of circ_001042 and transforming growth factor β1 (TGF-β1) in LUAD cells was elevated by the respective transfection of overexpression vectors OE-circ_001042 and TGF-β1; MTT and transwell assays were applied to test the proliferation, migration, and invasion abilities of cells, respectively. The E-cadherin expression level in the cells was assessed by immunofluorescence staining, and western blot was utilized to determine the expression level of epithelial-mesenchymal transition (EMT) and TGF-β1/P38 MAPK signaling axis-related proteins in the cells. Results Circ_001042 was significantly downregulated in LUAD tissues and cells, and high circ_001042 expression could inhibit the proliferation, invasion, and migration of LUAD cells. In addition, circ_001042 also inhibited the EMT process (the E-cadherin level was upregulated; and the levels of N-cadherin, vimentin, and Snail were downregulated) and TGF-β1/P38 MAPK signaling axis activity in LUAD cells. Moreover, circ_001042 could suppress the promotion of TGF-β1 on the proliferation, invasion, migration, and EMT process of LUAD cells and the activation of TGF-β1/P38 MAPK signaling axis. Conclusion By inhibiting TGF-β1, circ_001042 not only suppresses the proliferation, migration, invasion, and EMT of LUAD but also inhibits the activation of TGF-β1/P38 MAPK signaling axis. Therefore, circ_001042 can act as a potential target for early diagnosis and targeted therapy of LUAD.
Collapse
|
43
|
Zhao HY, Xi XX, Xin M, Zhang SQ. Overcoming C797S Mutation: The Challenges and Prospects of the Fourth-Generation EGFR-TKIs. Bioorg Chem 2022; 128:106057. [DOI: 10.1016/j.bioorg.2022.106057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/15/2022] [Accepted: 07/20/2022] [Indexed: 01/07/2023]
|
44
|
Luo DD, Zhao F. KLF4 suppresses the proliferation and metastasis of NSCLC cells via inhibition of MSI2 and regulation of the JAK/STAT3 signaling pathway. Transl Oncol 2022; 22:101396. [PMID: 35580385 PMCID: PMC9117691 DOI: 10.1016/j.tranon.2022.101396] [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: 10/20/2021] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 11/18/2022] Open
Abstract
KLF4 is downregulated, while MSI2 is upregulated in NSCLC tissues. Overexpression of KLF4 suppresses NSCLC cell migration and invasion. Knockdown of MSI2 suppresses the migration and invasion of NSCLC cells. KLF4 inhibits the transcription of MSI2 in NSCLC cells. KLF4 suppressed the invasion and migration of NSCLC cells via inhibition of JAK2/STAT3 signalling pathway.
Background Non-small cell lung cancer (NSCLC) remains an aggresive tumor with poor survival rates. Krüppel-like factor 4 (KLF4) is known to be involved in progression of NSCLC; however, the detailed mechanism by which KLF4 regulates the progression of NSCLC remains unclear. Methods In order to investigate the function of KLF4 in NSCLC, cell proliferation was measured by MTT and colony formation assays. The migration and invasion of NSCLC cells were detected via wound healing and Transwell assays, respectively. Then, the interaction between KLF4 and MSI2 was confirmed using a dual-luciferase reporter assay, and the mechanism by which KLF4 regulates the tumorigenesis of NSCLC was assessed by RT-qPCR and Western blotting. Results The results showed that KLF4 was downregulated, while MSI2 was upregulated in NSCLC. Additionally, KLF4 could inhibit transcription of MSI2, and overexpression of KLF4 or knockdown of MSI2 could inhibit the proliferation, migration and invasion of NSCLC cells. Moreover, KLF4 could inhibit JAK2/STAT3 signalling pathway. Conclusions In conclusion, KLF4 significantly inhibited the proliferation, invasion and migration of NSCLC cells via inactivation of MSI2/JAK2/STAT3 signalling pathway. Thereby, our finding might shed new lights on exploring the new strategies against NSCLC.
Collapse
|
45
|
Evaluating the utility of an immune checkpoint-related lncRNA signature for identifying the prognosis and immunotherapy response of lung adenocarcinoma. Sci Rep 2022; 12:12785. [PMID: 35896612 PMCID: PMC9329438 DOI: 10.1038/s41598-022-16715-0] [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: 07/30/2021] [Accepted: 07/14/2022] [Indexed: 12/03/2022] Open
Abstract
Lung adenocarcinoma (LUAD) is the most frequent subtype of lung cancer globally. However, the survival rate of lung adenocarcinoma patients remains low. Immune checkpoints and long noncoding RNAs are emerging as vital tools for predicting the immunotherapeutic response and outcomes of patients with lung adenocarcinoma. It is critical to identify lncRNAs associated with immune checkpoints in lung adenocarcinoma patients. In this study, immune checkpoint-related lncRNAs (IClncRNAs) were analysed and identified by coexpression. Based on the immune checkpoint-related lncRNAs, we divided patients with lung adenocarcinoma into two clusters and constructed a risk model. Kaplan–Meier analysis, Gene Set Enrichment Analysis, and nomogram analysis of the 2 clusters and the risk model were performed. Finally, the potential immunotherapeutic prediction value of this model was discussed. The risk model consisting of 6 immune checkpoint-related lncRNAs was an independent predictor of survival. Through regrouping the patients with this model, we can distinguish between them more effectively in terms of their immunotherapeutic response, tumour microenvironment, and chemotherapy response. This risk model based on immune checkpoint-based lncRNAs may have an excellent clinical value for predicting the immunotherapeutic response and outcomes of patients with LUAD.
Collapse
|
46
|
Targeting ferroptosis as a vulnerability in pulmonary diseases. Cell Death Dis 2022; 13:649. [PMID: 35882850 PMCID: PMC9315842 DOI: 10.1038/s41419-022-05070-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/29/2022] [Accepted: 07/04/2022] [Indexed: 01/21/2023]
Abstract
Ferroptosis is an iron-dependent regulated cell death marked by excessive oxidative phospholipids (PLs). The polyunsaturated fatty acids-containing phospholipids (PUFA-PLs) are highly susceptible to lipid peroxidation under oxidative stress. Numerous pulmonary diseases occurrences and degenerative pathologies are driven by ferroptosis. This review discusses the role of ferroptosis in the pathogenesis of pulmonary diseases including asthma, lung injury, lung cancer, fibrotic lung diseases, and pulmonary infection. Additionally, it is proposed that targeting ferroptosis is a potential treatment for pulmonary diseases, particularly drug-resistant lung cancer or antibiotic-resistant pulmonary infection, and reduces treatment-related adverse events.
Collapse
|
47
|
Ubenimex Combined with Pemetrexed Upregulates SOCS1 to Inhibit Lung Adenocarcinoma Progression via the JAK2-STAT3 Signaling Pathway. DISEASE MARKERS 2022; 2022:5614939. [PMID: 35789603 PMCID: PMC9250433 DOI: 10.1155/2022/5614939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/23/2022] [Accepted: 06/09/2022] [Indexed: 11/18/2022]
Abstract
To study the effects of ubenimex (UBE) combined with pemetrexed (PEM) on lung adenocarcinoma cell behavior and its molecular mechanism, the tissue samples from lung adenocarcinoma patients who received PEM chemotherapy, those with PEM combined with UBE chemotherapy, and healthy volunteers were retrieved and analyzed. The expression levels of the suppressor of cytokine signaling 1 (SOCS1) in the human lung adenocarcinoma cancer cell lines A549 and PC-9 and tissues were detected by qRT-PCR. MTT assay was performed for cell proliferation. Cell apoptosis was detected by flow cytometry. Cell invasion ability was assessed using the Transwell assay. The expression levels of the JAK2/STAT3 signaling pathway proteins and apoptosis-related proteins were detected by Western blot. The antitumor effect of PEM combined with UBE was tested in nude mice using the tumor formation assay. Our results showed that UBE treatment, alone or combined with PEM, inhibited lung adenocarcinoma cell migration, invasion, and proliferation; promoted apoptosis; significantly increased the G0/G1-phase cell ratio; reduced the S-phase cell ratio; and inhibited the in vivo growth of tumor cells. UBE alone or in combination with PEM also inhibited the JAK2/STAT3 signaling pathway in lung adenocarcinoma cells. In addition, UBE combined with PEM therapy was associated with increased SOCS1 expression in patients' serum and knocking down SOCS1 reversed the antitumor effects of UBE and PEM. Overall, combination therapy with UBE and PEM could inhibit the JAK2-STAT3 signaling pathway by upregulating SOCS1 expression to hinder the progression of lung adenocarcinoma cells.
Collapse
|
48
|
Li H, Harrison EB, Li H, Hirabayashi K, Chen J, Li QX, Gunn J, Weiss J, Savoldo B, Parker JS, Pecot CV, Dotti G, Du H. Targeting brain lesions of non-small cell lung cancer by enhancing CCL2-mediated CAR-T cell migration. Nat Commun 2022; 13:2154. [PMID: 35443752 PMCID: PMC9021299 DOI: 10.1038/s41467-022-29647-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/28/2022] [Indexed: 12/15/2022] Open
Abstract
Metastatic non-small cell lung cancer (NSCLC) remains largely incurable and the prognosis is extremely poor once it spreads to the brain. In particular, in patients with brain metastases, the blood brain barrier (BBB) remains a significant obstacle for the biodistribution of antitumor drugs and immune cells. Here we report that chimeric antigen receptor (CAR) T cells targeting B7-H3 (B7-H3.CAR) exhibit antitumor activity in vitro against tumor cell lines and lung cancer organoids, and in vivo in xenotransplant models of orthotopic and metastatic NSCLC. The co-expression of the CCL2 receptor CCR2b in B7-H3.CAR-T cells, significantly improves their capability of passing the BBB, providing enhanced antitumor activity against brain tumor lesions. These findings indicate that leveraging T-cell chemotaxis through CCR2b co-expression represents a strategy to improve the efficacy of adoptive T-cell therapies in patients with solid tumors presenting with brain metastases.
Collapse
Affiliation(s)
- Hongxia Li
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Emily B Harrison
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Huizhong Li
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Cancer Immunotherapy Center, Cancer Research Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Koichi Hirabayashi
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jing Chen
- Crown Bioscience Inc, San Diego, CA, USA
| | | | - Jared Gunn
- Crown Bioscience Inc, San Diego, CA, USA
| | - Jared Weiss
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Division of Hematology/Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Barbara Savoldo
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Joel S Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Chad V Pecot
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Division of Hematology/Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA. .,Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Hongwei Du
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA. .,Cancer Immunotherapy Center, Cancer Research Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, China.
| |
Collapse
|
49
|
PLOD3 regulates the expression of YAP1 to affect the progression of non-small cell lung cancer via the PKCδ/CDK1/LIMD1 signaling pathway. J Transl Med 2022; 102:440-451. [PMID: 35039611 DOI: 10.1038/s41374-021-00674-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 08/19/2021] [Accepted: 08/30/2021] [Indexed: 12/25/2022] Open
Abstract
Procollagen-lysine, 2-oxoglutarate 5-dioxygenase (PLOD3) is a crucial oncogene in human lung cancer, whereas protein kinase C δ (PKCδ) acts as a tumor suppressor. In this study, we aimed to explore the regulation by PLOD3 on the expression of YAP1 to affect the progression of non-small cell lung cancer (NSCLC) via the PKCδ/CDK1/LIMD1 signaling pathway. We found that PLOD3, CDK1, and YAP1 were highly expressed, while LIMD1 was poorly expressed in NSCLC tissues. Mechanistic investigation demonstrated that silencing PLOD3 promoted the cleavage of PKCδ in a caspase-dependent manner to generate a catalytically active fragment cleaved PKCδ, enhanced phosphorylation levels of CDK1, and LIMD1 but suppressed nuclear translocation of YAP1. Furthermore, functional experimental results suggested that loss of PLOD3 led to increased phosphorylation levels of CDK1 and LIMD1 and downregulated YAP1, thereby suppressing the proliferation, colony formation, cell cycle entry, and resistance to apoptosis of NSCLC cells in vitro and inhibiting tumor growth in vivo. Taken together, these results show that PLOD3 silencing activates the PKCδ/CDK1/LIMD1 signaling pathway to prevent the progression of NSCLC, thus providing novel insight into molecular targets for treating NSCLC.
Collapse
|
50
|
Gencer A, Baysal I, Nemutlu E, Yabanoglu-Ciftci S, Arica B. Efficacy of Sirna-Loaded Nanoparticles in the Treatment of k-ras Mutant Lung Cancer in vitro. J Microencapsul 2022; 39:261-275. [PMID: 35356841 DOI: 10.1080/02652048.2022.2061058] [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: 10/18/2022]
Abstract
AIM To design and develop K-RAS silencing small interfering RNA (siRNA)-loaded poly (D, L-lactic-co-glycolic acid) nanoparticles and evaluate their efficacy in the treatment of K-RAS mutant lung cancer. METHODS The nanoparticles prepared by the double emulsion solvent evaporation method were characterized by TEM, FTIR and XPS analyzes and evaluated in vitro by XTT, PCR, ELISA, and Western-Blot. Metabolomic analyzes were performed to evaluate the changes in metabolic profiles of the cells after nanoparticles treatment. RESULTS The nanoparticles were obtained with a particle size less than 250 nm, a polydispersity index around 0.1, a surface charge of (-12) - (+14) mV, and 80% of the siRNA encapsulation. The nanoparticles didn't affect cell viability of the cells after 72 hours. In cancer cells, KRAS expression was decreased by up to 50%, protein levels were decreased by more than 90%. CONCLUSION The formulated siRNA delivery nanoparticles can be promising treatment in lung cancer.
Collapse
Affiliation(s)
- Ayse Gencer
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Ipek Baysal
- Vocational School of Health Services, Hacettepe University, Ankara, Turkey
| | - Emirhan Nemutlu
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | | | - Betul Arica
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|