51
|
Fan W, Wu Z, Xu S, Liu Z, Huang Y, Wang P. Mendelian randomization analysis of plasma proteins reveals potential novel tumor markers for gastric cancer. Sci Rep 2025; 15:3537. [PMID: 39875498 PMCID: PMC11775103 DOI: 10.1038/s41598-025-88118-w] [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: 08/21/2024] [Accepted: 01/24/2025] [Indexed: 01/30/2025] Open
Abstract
This study aimed to elucidate the potential causal relationship between 4,907 plasma proteins and the risk of gastric cancer using a two-sample Mendelian randomization approach. We utilized genome-wide association study (GWAS) data to perform two-sample Mendelian randomization analyses, treating the 4,907 plasma proteins as exposure factors and gastric cancer as the outcome. Instrumental variables for plasma proteins were selected based on strongly correlated SNPs identified through data processing and screening of the GWAS data provided by the deCode database. We employed a set of statistical methods centered on inverse variance weighting (IVW) for Mendelian randomization analysis to estimate the odds ratios (ORs) for the effects of these plasma proteins on gastric cancer susceptibility. According to the IVW method, 14 plasma proteins were associated with gastric cancer (p < 0.005). Specifically, CHST15 (OR = 0.7553, 95% CI = 0.6346 - 0.8988), L1CAM (OR = 0.7230, 95% CI = 0.5876 - 0.8896), FTMT (OR = 0.8246, 95% CI = 0.7241 - 0.9391), and PMM2 (OR = 0.5767, 95% CI = 0.3943 - 0.8433) were negatively correlated with GASTRIC CANCER, whereas ABO (OR = 1.1868, 95% CI = 1.0638 - 1.3240), FAM3D (OR = 1.2109, 95% CI = 1.0850 - 1.3515), FAM3B (OR = 1.2988, 95% CI = 1.0953 - 1.5402), ADH7 (OR = 1.3568, 95% CI = 1.1044 - 1.6670), MAP1LC3A (OR = 1.3704, 95% CI = 1.1194 - 1.6778), PGLYRP1 (OR = 1.4071, 95% CI = 1.1235 - 1.7623), PDE5A (OR = 1.7446, 95% CI = 1.2693 - 2.3978), GLUL (OR = 3.1203, 95% CI = 1.5017 - 6.4839), NFE2L1 (OR = 3.1759, 95% CI = 1.6163 - 6.2402), and MAFG (OR = 3.1945, 95% CI = 1.5329 - 6.6575) were positively correlated. Convergent results from Weighted Median and MR-Egger analyses confirmed these associations. Reverse Mendelian randomization analysis indicated that gastric cancer does not significantly alter the levels of these 14 plasma proteins (p > 0.05). Sensitivity analyses, including assessments of heterogeneity and horizontal pleiotropy, confirmed the robustness and reliability of our findings without significant bias. Pathway enrichment analysis of gene expression associated with these 14 plasma proteins, using GO and KEGG pathways, revealed that CHST15, L1CAM, FTMT, and PMM2 may serve as protective factors against gastric cancer, while ABO, FAM3D, FAM3B, ADH7, MAP1LC3A, PGLYRP1, PDE5A, GLUL, NFE2L1, and MAFG may contribute to gastric cancer pathogenesis. These results highlight the complex biological interactions between plasma proteins and tumorigenesis, providing valuable insights for preventive and therapeutic strategies in gastric malignancy management.
Collapse
Affiliation(s)
- Wenhai Fan
- Department of Gastrointestinal Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Zhenjiang Wu
- Department of Gastrointestinal Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Shenghao Xu
- Department of Gastrointestinal Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Zhiheng Liu
- Department of Gastrointestinal Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Yiming Huang
- Department of Gastrointestinal Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Pan Wang
- Department of Gastrointestinal Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, China.
- Department of Gastrointestinal Surgery, Institute of Hepatobiliology and Pancreaticoenterology of the Affiliated Hospital of North Sichuan Medical College, Nanchong, China.
| |
Collapse
|
52
|
Shou S, Maolan A, Zhang D, Jiang X, Liu F, Li Y, Zhang X, Geer E, Pu Z, Hua B, Guo Q, Zhang X, Pang B. Telomeres, telomerase, and cancer: mechanisms, biomarkers, and therapeutics. Exp Hematol Oncol 2025; 14:8. [PMID: 39871386 PMCID: PMC11771031 DOI: 10.1186/s40164-025-00597-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 01/15/2025] [Indexed: 01/29/2025] Open
Abstract
Telomeres and telomerase play crucial roles in the initiation and progression of cancer. As biomarkers, they aid in distinguishing benign from malignant tissues. Despite the promising therapeutic potential of targeting telomeres and telomerase for therapy, translating this concept from the laboratory to the clinic remains challenging. Many candidate drugs remain in the experimental stage, with only a few advancing to clinical trials. This review explores the relationship between telomeres, telomerase, and cancer, synthesizing their roles as biomarkers and reviewing the outcomes of completed trials. We propose that changes in telomere length and telomerase activity can be used to stratify cancer stages. Furthermore, we suggest that differential expression of telomere and telomerase components at the subcellular level holds promise as a biomarker. From a therapeutic standpoint, combining telomerase-targeted therapies with drugs that mitigate the adverse effects of telomerase inhibition may offer a viable strategy.
Collapse
Affiliation(s)
- Songting Shou
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ayidana Maolan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Di Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaochen Jiang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fudong Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yi Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiyuan Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - En Geer
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhenqing Pu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baojin Hua
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Qiujun Guo
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Xing Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Bo Pang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
53
|
Mishra A, Patel TN. Telomerase in cancer- ongoing quest and future discoveries. Mol Biol Rep 2025; 52:161. [PMID: 39862360 DOI: 10.1007/s11033-025-10251-6] [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: 06/30/2024] [Accepted: 01/10/2025] [Indexed: 01/27/2025]
Abstract
Telomerase, constituted by the dynamic duo of telomerase reverse transcriptase (TERT), the catalytic entity, and an integral RNA component (TERC), is predominantly suppressed in differentiated human cells due to postnatal transcriptional repression of the TERT gene. Dysregulation of telomerase significantly contributes to cancer development via telomere-dependent and independent mechanisms. Telomerase activity is often elevated in advanced cancers, with TERT reactivation and upregulation of TERC observed in early tumorigenesis. Beyond their primary function of telomere maintenance, TERT and TERC exhibit multifaceted roles in regulating gene expression, signal transduction pathways, and cellular metabolism. The presence of the enzymatic component TERT in both the nucleus and mitochondria underscores its non-canonical roles. Cell death is prevented in TERT-upregulated cells regardless of the DNA damage events and safeguards mitochondrial DNA from oxidative damage. This highlights its protective role in cancer cells where it intersects with glucose metabolism and epigenetic regulation, shaping tumor phenotypes. Oncogenic viruses exploit various strategies to manipulate telomerase activity, aiding cancer progression. The perpetual cell proliferation facilitated by telomerase is a hallmark of cancer, making it an attractive therapeutic target. Inhibitors targeting the catalytic subunit of telomerase, nutraceutical-based compounds, and telomerase-based vaccines represent promising avenues for cancer therapy. Considering the pivotal roles played by the complete enzyme telomerase and TERT component in cancer initiation, substantial endeavors have been dedicated to unravel the mechanisms driving telomerase activation and TERT induction. This review also explores how computational modeling can be leveraged to uncover new insights in telomere research, and efficient targeted therapies.
Collapse
Affiliation(s)
- Apurwa Mishra
- Department of Integrative Biology, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Trupti N Patel
- Department of Integrative Biology, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
54
|
Zheng J, Gong J. SLC1A4 Promotes Malignant Transformation of Hepatocellular Carcinoma by Activating the AKT Signaling. Anal Cell Pathol (Amst) 2025; 2025:1115184. [PMID: 39949345 PMCID: PMC11824774 DOI: 10.1155/ancp/1115184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 10/04/2024] [Accepted: 11/30/2024] [Indexed: 02/16/2025] Open
Abstract
Due to the difficulty in early diagnosis and the lack of treatment for advanced disease, the mortality rate of hepatocellular carcinoma (HCC) is high, and the 5-year overall survival rate is low at present. SLC1A4 is a neutral amino acid transporter, but its regulatory role and mechanism in HCC are still unclear. Through analyzing the TCGA database and clinical tissue specimens, this study uncovered the high expression of SLC1A4 in tumor tissues of HCC. Worse more, a high level of SLC1A4 may lead to a poor prognosis of HCC. Mechanically, silencing SLC1A4 inhibited the phosphorylation activation of AKT by suppressing the ubiquitin modification of AKT at lysine 63 and amino acid influx represented by D-serine, decreasing the protein level of β-catenin in the cell nucleus and suppressing the transcriptional activity of c-Myc and EpCAM promoters. As a result, silencing SLC1A4 inhibited the proliferation, migration, and stemness of hepatic cancer cells, which was successfully reversed by the introduction of exogenous AKT. Moreover, epithelial-mesenchymal transition (EMT) in vitro and metastasis potential in vivo of hepatic cancer cells was suppressed by the downregulated SLC1A4 level. In conclusion, SLC1A4 promotes the malignant transformation of HCC through activating signal transduction mediated by AKT. The findings in this study suggested that SLC1A4 may be a diagnostic indicator for the early HCC and a therapeutic target for the advanced HCC.
Collapse
MESH Headings
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/genetics
- Humans
- Liver Neoplasms/pathology
- Liver Neoplasms/metabolism
- Liver Neoplasms/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Signal Transduction
- Epithelial-Mesenchymal Transition/genetics
- Animals
- Cell Transformation, Neoplastic/pathology
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/genetics
- Cell Line, Tumor
- Cell Proliferation/genetics
- Cell Movement/genetics
- Gene Expression Regulation, Neoplastic
- Mice, Nude
- Male
- Mice
- Mice, Inbred BALB C
- Amino Acid Transport System A/metabolism
- Amino Acid Transport System A/genetics
Collapse
Affiliation(s)
- Jiaoyun Zheng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Jian Gong
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
- Research Center of Digestive Disease, Central South University, Changsha 410011, Hunan, China
- Clinical Research Center of Digestive Diseases of Hunan Province, Changsha 410011, Hunan, China
| |
Collapse
|
55
|
Iskandar M, Xiao Barbero M, Jaber M, Chen R, Gomez-Guevara R, Cruz E, Westerheide S. A Review of Telomere Attrition in Cancer and Aging: Current Molecular Insights and Future Therapeutic Approaches. Cancers (Basel) 2025; 17:257. [PMID: 39858038 PMCID: PMC11764024 DOI: 10.3390/cancers17020257] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES As cells divide, telomeres shorten through a phenomenon known as telomere attrition, which leads to unavoidable senescence of cells. Unprotected DNA exponentially increases the odds of mutations, which can evolve into premature aging disorders and tumorigenesis. There has been growing academic and clinical interest in exploring this duality and developing optimal therapeutic strategies to combat telomere attrition in aging and cellular immortality in cancer. The purpose of this review is to provide an updated overview of telomere biology and therapeutic tactics to address aging and cancer. METHODS We used the Rayyan platform to review the PubMed database and examined the ClinicalTrial.gov registry to gain insight into clinical trials and their results. RESULTS Cancer cells activate telomerase or utilize alternative lengthening of telomeres to escape telomere shortening, leading to near immortality. Contrarily, normal cells experience telomeric erosion, contributing to premature aging disorders, such as Werner syndrome and Hutchinson-Gilford Progeria, and (2) aging-related diseases, such as neurodegenerative and cardiovascular diseases. CONCLUSIONS The literature presents several promising therapeutic approaches to potentially balance telomere maintenance in aging and shortening in cancer. This review highlights gaps in knowledge and points to the potential of these optimal interventions in preclinical and clinical studies to inform future research in cancer and aging.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sandy Westerheide
- Department of Molecular Biosciences, University of South Florida, 4202 East Fowler Avenue, ISA2015, Tampa, FL 33620, USA; (M.I.); (M.X.B.); (M.J.); (R.C.); (R.G.-G.); (E.C.)
| |
Collapse
|
56
|
Bozcuk HŞ, Sert L, Kaplan MA, Tatlı AM, Karaca M, Muğlu H, Bilici A, Kılıçtaş BŞ, Artaç M, Erel P, Yumuk PF, Bilgin B, Şendur MAN, Kılıçkap S, Taban H, Ballı S, Demirkazık A, Akdağ F, Hacıbekiroğlu İ, Güzel HG, Koçer M, Gürsoy P, Köylü B, Selçukbiricik F, Karakaya G, Alemdar MS. Enhancing Treatment Decisions for Advanced Non-Small Cell Lung Cancer with Epidermal Growth Factor Receptor Mutations: A Reinforcement Learning Approach. Cancers (Basel) 2025; 17:233. [PMID: 39858018 PMCID: PMC11763509 DOI: 10.3390/cancers17020233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 12/29/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Although higher-generation TKIs are associated with improved progression-free survival in advanced NSCLC patients with EGFR mutations, the optimal selection of TKI treatment remains uncertain. To address this gap, we developed a web application powered by a reinforcement learning (RL) algorithm to assist in guiding initial TKI treatment decisions. METHODS Clinical and mutational data from advanced NSCLC patients were retrospectively collected from 14 medical centers. Only patients with complete data and sufficient follow-up were included. Multiple supervised machine learning models were tested, with the Extra Trees Classifier (ETC) identified as the most effective for predicting progression-free survival. Feature importance scores were calculated by the ETC, and features were then integrated into a Deep Q-Network (DQN) RL algorithm. The RL model was designed to select optimal TKI generation and a treatment line for each patient and was embedded into an open-source web application for experimental clinical use. RESULTS In total, 318 cases of EGFR-mutant advanced NSCLC were analyzed, with a median patient age of 63. A total of 52.2% of patients were female, and 83.3% had ECOG scores of 0 or 1. The top three most influential features identified were neutrophil-to-lymphocyte ratio (log-transformed), age (log-transformed), and the treatment line of TKI administration, as tested by the ETC algorithm, with an area under curve (AUC) value of 0.73, whereas the DQN RL algorithm achieved a higher AUC value of 0.80, assigning distinct Q-values across four TKI treatment categories. This supports the decision-making process in the web-based 'EGFR Mutant NSCLC Treatment Advisory System', where clinicians can input patient-specific data to receive tailored recommendations. CONCLUSIONS The RL-based web application shows promise in assisting TKI treatment selection for EGFR-mutant advanced NSCLC patients, underscoring the potential for reinforcement learning to enhance decision-making in oncology care.
Collapse
Affiliation(s)
- Hakan Şat Bozcuk
- Independent Researcher, Antalya 07100, Turkey
- Private Practice, Burhanettin Onat Caddesi, 1419. Sokak, No:59, Ocean City, C-Blok, Kat:3, Daire:5, Muratpaşa, Antalya 07100, Turkey
| | - Leyla Sert
- Department of Medical Oncology, Dicle University, Diyarbakır 21280, Turkey; (L.S.); (M.A.K.)
| | - Muhammet Ali Kaplan
- Department of Medical Oncology, Dicle University, Diyarbakır 21280, Turkey; (L.S.); (M.A.K.)
| | - Ali Murat Tatlı
- Department of Medical Oncology, Akdeniz University, Antalya 07058, Turkey; (A.M.T.); (M.K.)
| | - Mustafa Karaca
- Department of Medical Oncology, Akdeniz University, Antalya 07058, Turkey; (A.M.T.); (M.K.)
| | - Harun Muğlu
- Department of Medical Oncology, Faculty of Medicine, İstanbul Medipol University, İstanbul 34810, Turkey; (H.M.); (A.B.)
| | - Ahmet Bilici
- Department of Medical Oncology, Faculty of Medicine, İstanbul Medipol University, İstanbul 34810, Turkey; (H.M.); (A.B.)
| | - Bilge Şah Kılıçtaş
- Department of Medical Oncology, Necmettin Erbakan University, Konya 42090, Turkey; (B.Ş.K.); (M.A.)
| | - Mehmet Artaç
- Department of Medical Oncology, Necmettin Erbakan University, Konya 42090, Turkey; (B.Ş.K.); (M.A.)
| | - Pınar Erel
- Department of Medical Oncology, Marmara University, İstanbul 34722, Turkey; (P.E.); (P.F.Y.)
| | - Perran Fulden Yumuk
- Department of Medical Oncology, Marmara University, İstanbul 34722, Turkey; (P.E.); (P.F.Y.)
- Division of Medical Oncology, School of Medicine, Koç University, İstanbul 34450, Turkey; (B.K.); (F.S.)
| | - Burak Bilgin
- Faculty of Medicine, Ankara Yıldırım Beyazıt University, Ankara 06010, Turkey; (B.B.); (M.A.N.Ş.)
| | - Mehmet Ali Nahit Şendur
- Faculty of Medicine, Ankara Yıldırım Beyazıt University, Ankara 06010, Turkey; (B.B.); (M.A.N.Ş.)
| | - Saadettin Kılıçkap
- Department of Medical Oncology, Faculty of Medicine, İstinye University, İstanbul 34010, Turkey; (S.K.); (M.S.A.)
| | - Hakan Taban
- Department of Medical Oncology, Medical Park Keçiören Hospital, Ankara 06120, Turkey;
| | - Sevinç Ballı
- Department of Medical Oncology, Ankara University, Ankara 06100, Turkey; (S.B.); (A.D.)
| | - Ahmet Demirkazık
- Department of Medical Oncology, Ankara University, Ankara 06100, Turkey; (S.B.); (A.D.)
| | - Fatma Akdağ
- Department of Medical Oncology, Sakarya University, Sakarya 54050, Turkey; (F.A.); (İ.H.)
| | - İlhan Hacıbekiroğlu
- Department of Medical Oncology, Sakarya University, Sakarya 54050, Turkey; (F.A.); (İ.H.)
| | - Halil Göksel Güzel
- Antalya Education and Research Hospital, Antalya 07100, Turkey; (H.G.G.); (M.K.)
| | - Murat Koçer
- Antalya Education and Research Hospital, Antalya 07100, Turkey; (H.G.G.); (M.K.)
| | - Pınar Gürsoy
- Department of Medical Oncology, Ege University, İzmir 35040, Turkey;
| | - Bahadır Köylü
- Division of Medical Oncology, School of Medicine, Koç University, İstanbul 34450, Turkey; (B.K.); (F.S.)
| | - Fatih Selçukbiricik
- Division of Medical Oncology, School of Medicine, Koç University, İstanbul 34450, Turkey; (B.K.); (F.S.)
| | - Gökhan Karakaya
- Department of Medical Oncology, ASV Yaşam Hospital, Antalya 07300, Turkey;
| | - Mustafa Serkan Alemdar
- Department of Medical Oncology, Faculty of Medicine, İstinye University, İstanbul 34010, Turkey; (S.K.); (M.S.A.)
| |
Collapse
|
57
|
Li Y, Li C, Fan J, Liu Y, Xu Y, Pang G. The Application of Traditional Chinese Medicine-Derived Formulations in Cancer Immunotherapy: A Review. Cancer Manag Res 2025; 17:23-34. [PMID: 39816489 PMCID: PMC11734505 DOI: 10.2147/cmar.s503071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 12/21/2024] [Indexed: 01/18/2025] Open
Abstract
Background Cancer immunotherapy is an advanced therapeutic approach that harnesses the body's immune system to target and eliminate tumor cells. Traditional Chinese medicine (TCM), with a history rooted in centuries of clinical practice, plays a crucial role in enhancing immune responses, alleviating cancer-related symptoms, and reducing the risks of infections and complications in cancer patients. Methodology This review systematically examines the current literature on TCM-based formulations in cancer immunotherapy. It explores the mechanisms by which TCM augments immune responses, particularly focusing on how these formulations influence both innate and adaptive immunity. Various TCM herbs and compounds, their active ingredients, and their application in cancer prevention and treatment were analyzed through an integrated review of preclinical studies, clinical trials, and molecular mechanistic investigations. Results TCM formulations contribute to cancer therapy by modulating the body's internal environment to improve immune defense. They enhance the immune system's ability to detect and destroy cancer cells, promote apoptosis in tumor cells, inhibit tumor growth and metastasis, and augment the effectiveness of conventional cancer treatments. The review highlights specific TCM herbs and formulations that have demonstrated significant anti-cancer properties, including their ability to strengthen immune responses and provide synergistic effects with existing cancer therapies. Conclusion TCM-derived formulations represent a promising addition to cancer immunotherapy. The mechanisms through which these formulations enhance anti-tumor immunity are multifaceted, involving modulation of immune cell activity, apoptosis induction, and suppression of tumor progression. As cancer immunotherapy evolves, the integration of TCM into conventional treatment regimens may offer enhanced therapeutic efficacy, reduced side effects, and improved overall outcomes for cancer patients. Further clinical research is needed to fully elucidate the therapeutic potential and safety of TCM-based immunotherapeutic strategies in cancer care.
Collapse
Affiliation(s)
- Yanyun Li
- School of Acupuncture and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Changying Li
- School of Ophthalmology and optometry, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Junzi Fan
- School of Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Yutong Liu
- School of Acupuncture and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Yincong Xu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Guowei Pang
- School of Acupuncture and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| |
Collapse
|
58
|
Liu W, Zhang J, Zhang J, Ye Y, Zhu J, Yu Q, Li T, Sun X, Chen H. EIF4A3-induced circ_0022382 promotes breast cancer cell progression through the let-7a-5p/PI3K/AKT/mTOR signaling pathway and SLC7A11 axis. Front Oncol 2025; 14:1476731. [PMID: 39868374 PMCID: PMC11758171 DOI: 10.3389/fonc.2024.1476731] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 12/16/2024] [Indexed: 01/28/2025] Open
Abstract
Introduction Breast cancer is one of the most common cancers in women and poses a serious threat to women's health. Circular RNAs (circRNAs) have been found to be specifically expressed in cancers and regulate the growth and death of tumor cells. The role of circRNAs in breast cancer remain unknown. In this study, we explored the impacts of circRNAs on the progression of breast cancer cells. Methods Using bioinformatics analysis, we screened out one up-regulated circRNA in breast cancer, and its function and regulatory mechanisms were confirmed by quantitative real-time PCR, cell counting kit-8 experiment, migration assay, dual luciferase reporter assay, Kyoto Encyclopedia of Genes and Genomes enrichment analysis, cell immunofluorescence, clone formation assay, scratch wound healing experiment, RNA immunoprecipitation and subcutaneous tumor-bearing experiments. Results Circ_0022382 was highly expressed in breast cancer cell lines MDA-MB-231, MCF-7 as well as breast cancer tissues, and promoted the proliferative and migratory capacity of breast cancer cells. In terms of regulatory mechanisms, circ_0022382 activated PI3K/AKT/mTOR signaling pathway and SLC7A11 by sponging let-7a-5p, while knockdown of circ_0022382 contributed to the occurrence of disulfidptosis. In addition, EIF4A3 promoted the expression of circ_0022382 in MDA-MB-231 and MCF-7. Consistently, knockdown of circ_0022382 inhibited the growth of breast cancer cells in vivo. Discussion Circ_0022382 and its related molecules may be effective targets for diagnosis or targeted therapy of breast cancer.
Collapse
Affiliation(s)
- Wei Liu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
- HEALTH BioMed Research & Development Center, Health BioMed Co., Ltd., Ningbo, Zhejiang, China
| | - Jun Zhang
- Department of Clinical Laboratory, The Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China
| | - Jiawen Zhang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yu Ye
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jianqin Zhu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Qiwen Yu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Tao Li
- HEALTH BioMed Research & Development Center, Health BioMed Co., Ltd., Ningbo, Zhejiang, China
- School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Xiaochun Sun
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Huabiao Chen
- HEALTH BioMed Research & Development Center, Health BioMed Co., Ltd., Ningbo, Zhejiang, China
- School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
59
|
Wang CC, Yeh SA, Chen WH, Li HJ, Yang CC, Huang TJ, Su YC. Evaluating Cetuximab Regimens in Head and Neck Cancer: Insights from a Retrospective Cohort Study. Cancers (Basel) 2025; 17:210. [PMID: 39857992 PMCID: PMC11764294 DOI: 10.3390/cancers17020210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES The aim of this study is to assess the effectiveness of cetuximab combination therapy in patients with recurrent or metastatic head and neck cancer treated at a hospital in Southern Taiwan. METHODS This study analyzed a retrospective cohort of 67 patients who were treated between January 2020 and May 2024 with two cetuximab regimens, cetuximab combined with cisplatin and 5-Fu, which were administered every four weeks during hospitalization (CPF4) and every two weeks as outpatient treatment (CPF2), respectively. The clinical outcomes, including overall survival and progression-free survival (PFS), were compared across the treatment regimens and age groups using Kaplan-Meier survival curves and Cox proportional hazard models. RESULTS The median overall survival was 11.1 months (95% confidence interval, 7.8-14.5), with CPF2 showing a potential PFS advantage in patients aged 46-60 years (p = 0.049). No significant differences in overall survival were observed between CPF2 and CPF4. CPF2, which was administered in an outpatient setting, was associated with improved convenience, reduced hospitalization, and potentially lower risks of hospital-acquired infections. CONCLUSIONS CPF2 exhibits practical advantages and comparable effectiveness, making it the preferred treatment regimen for eligible patients. Further studies with larger populations and molecular stratifications are needed to confirm these findings and develop better treatment strategies.
Collapse
Affiliation(s)
- Chih-Chun Wang
- Department of Otolaryngology, E-Da Hospital, I-Shou University, Kaohsiung 82445, Taiwan; (C.-C.W.); (C.-C.Y.); (T.-J.H.)
- School of Chinese Medicine for Post-Baccalaureate, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - Shyh-An Yeh
- Department of Radiation Oncology, E-Da Hospital, I-Shou University, Kaohsiung 82445, Taiwan;
- Department of Medical Imaging and Radiological Sciences, I-Shou University, Kaohsiung 82445, Taiwan
| | - Wen-Hui Chen
- Department of Dentistry, E-Da Hospital, I-Shou University, Kaohsiung 82445, Taiwan;
- School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - Hung-Ju Li
- Division of Hematology-Oncology, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung 82445, Taiwan;
| | - Chuan-Chien Yang
- Department of Otolaryngology, E-Da Hospital, I-Shou University, Kaohsiung 82445, Taiwan; (C.-C.W.); (C.-C.Y.); (T.-J.H.)
- School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - Tse-Jen Huang
- Department of Otolaryngology, E-Da Hospital, I-Shou University, Kaohsiung 82445, Taiwan; (C.-C.W.); (C.-C.Y.); (T.-J.H.)
- School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - Yu-Chieh Su
- Division of Hematology-Oncology, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung 82445, Taiwan;
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
- Graduate Institute of Medicine, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| |
Collapse
|
60
|
Hassan YM, Mohamed AS, Hassan YM, El-Sayed WM. Recent developments and future directions in point-of-care next-generation CRISPR-based rapid diagnosis. Clin Exp Med 2025; 25:33. [PMID: 39789283 PMCID: PMC11717804 DOI: 10.1007/s10238-024-01540-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 12/15/2024] [Indexed: 01/12/2025]
Abstract
The demand for sensitive, rapid, and affordable diagnostic techniques has surged, particularly following the COVID-19 pandemic, driving the development of CRISPR-based diagnostic tools that utilize Cas effector proteins (such as Cas9, Cas12, and Cas13) as viable alternatives to traditional nucleic acid-based detection methods. These CRISPR systems, often integrated with biosensing and amplification technologies, provide precise, rapid, and portable diagnostics, making on-site testing without the need for extensive infrastructure feasible, especially in underserved or rural areas. In contrast, traditional diagnostic methods, while still essential, are often limited by the need for costly equipment and skilled operators, restricting their accessibility. As a result, developing accessible, user-friendly solutions for at-home, field, and laboratory diagnostics has become a key focus in CRISPR diagnostic innovations. This review examines the current state of CRISPR-based diagnostics and their potential applications across a wide range of diseases, including cancers (e.g., colorectal and breast cancer), genetic disorders (e.g., sickle cell disease), and infectious diseases (e.g., tuberculosis, malaria, Zika virus, and human papillomavirus). Additionally, the integration of machine learning (ML) and artificial intelligence (AI) to enhance the accuracy, scalability, and efficiency of CRISPR diagnostics is discussed, alongside the challenges of incorporating CRISPR technologies into point-of-care settings. The review also explores the potential for these cutting-edge tools to revolutionize disease diagnosis and personalized treatment in the future, while identifying the challenges and future directions necessary to address existing gaps in CRISPR-based diagnostic research.
Collapse
Affiliation(s)
- Youssef M Hassan
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566, Egypt
| | - Ahmed S Mohamed
- Biotechnology Program, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566, Egypt
| | - Yaser M Hassan
- Biotechnology Program, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566, Egypt
| | - Wael M El-Sayed
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566, Egypt.
| |
Collapse
|
61
|
Schoenherr C, Pietzsch S, Barca C, Müller FE, Bahr FS, Kasten M, Zeug A, Erschow S, Falk CS, Ponimaskin E, Thackeray JT, Hilfiker-Kleiner D, Ricke-Hoch M. Immune-checkpoint-inhibitor therapy directed against PD-L1 is tolerated in the heart without manifestation of cardiac inflammation in a preclinical reversible melanoma mouse model. FRONTIERS IN MOLECULAR MEDICINE 2025; 4:1487526. [PMID: 39834851 PMCID: PMC11743445 DOI: 10.3389/fmmed.2024.1487526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025]
Abstract
Immune-checkpoint-inhibitors (ICI) target key regulators of the immune system expressed by cancer cells that mask those from recognition by the immune system. They have improved the outcome for patients with various cancer types, such as melanoma. ICI-based therapy is frequently accompanied by immune-related adverse side effects (IRAEs). The reversible melanoma cancer mouse model (B16F10 cells stably expressing a ganciclovir (GCV)-inducible suicide gene in C57BL/6N mice: B16F10-GCV) allows chemotherapy-free tumor elimination in advanced disease stage and demonstrates almost complete recovery of the mouse heart from cancer-induced atrophy, molecular impairment and heart failure. Thus, enabling the study of anti-cancer-therapy effects. Here, we analyzed potential cardiac side effects of antibody-mediated PD-L1 inhibition in the preclinical B16F10-GCV mouse model after tumor elimination and 2 weeks recovery (50 days after tumor inoculation). Anti-PD-L1 treatment was associated with improved survival as compared to isotype control (Ctrl) treated mice. Surviving anti-PD-L1 and Ctrl mice showed similar cardiac function, dimensions and the expression of cardiac stress and hypertrophy markers. Although anti-PD-L1 treatment was associated with increased troponin I type 3 cardiac (TNNI3) blood levels, cardiac mRNA expression of macrophage markers and elevated cardiac levels of secreted inflammatory factors compared to Ctrl treatment, both groups showed a comparable density of inflammatory cells in the heart (using CXCR4-ligand 68Ga-Pentixafor in PET-CT and immunohistochemistry). Thus, anti-PD-L1 therapy improved survival in mice with advanced melanoma cancer with no major cardiac phenotype or inflammation 50 days after tumor inoculation. Without a second hit that triggers the inflammatory response, anti-PD-L1 treatment appears to be safe for the heart in the preclinical melanoma mouse model.
Collapse
Affiliation(s)
- Caroline Schoenherr
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Stefan Pietzsch
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Cristina Barca
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Franziska E. Müller
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Frauke S. Bahr
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Martina Kasten
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Andre Zeug
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Sergej Erschow
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Christine S. Falk
- Institute of Transplant Immunology, IFB-Tx, Hannover Medical School, Hannover, Germany
| | - Evgeni Ponimaskin
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - James T. Thackeray
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Denise Hilfiker-Kleiner
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
- Department of Cardiovascular Complications of Oncologic Therapies, Medical Faculty of the Philipps University Marburg, Marburg, Germany
| | - Melanie Ricke-Hoch
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
62
|
Saha SK, Arya V, Jadhav A, Jhanana Kailash S, Panigrahy BK, Joshi A, Singh R, Dubey K. Solid dispersion of alectinib HCl: preclinical evaluation for improving bioavailability and establishing an IVIVC model. Drug Dev Ind Pharm 2025; 51:50-63. [PMID: 39757594 DOI: 10.1080/03639045.2024.2447276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 12/06/2024] [Accepted: 12/20/2024] [Indexed: 01/07/2025]
Abstract
OBJECTIVE Alectinib HCl (ALB-HCl) is a BCS class IV molecule with low solubility and low oral bioavailability. Owing to its low bioavailability, a high dose of ALB-HCl is recommended with food to meet clinical efficacy. Thus, there is a need for a delivery system to overcome the bioavailability concerns. METHODS Three solid dispersion (SD) formulations (I, II, and III) were evaluated for in-vitro dissolution and in-vivo pharmacokinetics (PK) study in Wistar rats. An in-vitro and in-vivo correlation (IVIVC) model was developed to establish a relationship between in-vitro dissolution data and in-vivo PK data. The formulations were subjected to stability studies. RESULTS All formulations showed enhanced dissolution in all the media except Formulation I in FaSSIF media. In-vivo PK studies displayed that Formulation I was inferior to API alone. Formulations II and III (amorphous SD [ASD]) exhibited two-fold higher Cmax and AUC0-last than API alone. Level A IVIVC model was established for Cmax and AUC0-last with an acceptable % prediction error (PE). When evaluated for external predictability, the model was found validated for Cmax (% PE <10%), however, it was inconclusive for AUC0-last (%PE -14.03). Stability studies showed ASD formulations were stable during storage. CONCLUSION A stable ASD formulation of ALB-HCl was successfully developed with improved bioavailability. Developing an IVIVC model can act as a surrogate to predict in-vivo performance. The selection of formulation components in ASD shall be rationalized for bioavailability and stability before clinical evaluation.
Collapse
Affiliation(s)
- Sumit Kumar Saha
- Department of Pharmacology, School of Pharmaceutical Education & Research, New Delhi, India
- Formulation Research and Development - Orals, Gurugram, India
| | - Vipin Arya
- CPP, Sun Pharmaceuticals Industries Limited, Gurugram, India
| | - Ajinkya Jadhav
- Formulation Research and Development - Orals, Vadodara, India
| | | | | | | | - Romi Singh
- Formulation Research and Development - Orals, Gurugram, India
| | - Kiran Dubey
- Department of Pharmacology, School of Pharmaceutical Education & Research, New Delhi, India
| |
Collapse
|
63
|
Nassar AH, Jayakrishnan R, Feng J, Shepherd F, Adib E, Cheung JM, Lin JJ, Liu Y, Lin SH, Parikh K, Sridhar A, Shakya P, Dilling TJ, Kaldas D, Gray JE, Lobachov A, Bar J, Luders H, Grohe C, Gupta S, Leal T, Fitzgerald B, Crowley F, Fujiwara Y, Marron TU, Wilgucki M, Reuss J, Chen L, Sankar K, Aredo JV, Neal JW, Wakelee HA, Thummalapalli R, Yu H, Whitaker R, Velazquez A, Ragavan M, Cortellini A, Kwiatkowski DJ, Naqash AR, Goldberg SB, Kim SY. Consolidation ALK Tyrosine Kinase Inhibitors Versus Durvalumab or Observation After Chemoradiation in Unresectable Stage III ALK-Positive NSCLC. J Thorac Oncol 2025; 20:109-118. [PMID: 39260522 DOI: 10.1016/j.jtho.2024.09.1379] [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: 07/01/2024] [Revised: 08/19/2024] [Accepted: 09/01/2024] [Indexed: 09/13/2024]
Abstract
INTRODUCTION Patients with advanced ALK-positive NSCLC typically have poor response to immunotherapy; the benefit of consolidation durvalumab in patients with unresectable stage III ALK-positive NSCLC remains unclear. Herein, we compare the efficacy and safety of consolidation ALK tyrosine kinase inhibitor (TKI) versus durvalumab or observation after concurrent chemoradiation. METHODS We conducted a retrospective study using a multicenter study of 17 institutions globally. Patients with unresectable stage III ALK-positive NSCLC treated between 2015 and 2022 were included. Patients received ALK TKI, durvalumab, or observation after concurrent chemoradiation. Real-world progression-free survival (rwPFS) and overall survival (OS) were estimated using Kaplan-Meier method. Treatment-related adverse events (trAEs) were classified by Common Terminology Criteria for Adverse Events version 5.0. Outcomes were assessed by multivariable Cox regression analysis. RESULTS A total of 67 patients were included, of whom 39 (58%) were female. Median age was 57 (interquartile range: 49-67) years. Furthermore, 15 received consolidation ALK TKI, 30 received durvalumab, and 22 underwent observation. Baseline characteristics were similar across the three groups other than differences in race. After adjusting for stage, age, and nodal status, median rwPFS was significantly longer for ALK TKI (rwPFS not reached, 95% confidence interval [CI]: 22.7- not reached) versus durvalumab (11.3 mo, 95% CI: 8.9-18.5, hazard ratio [HR] = 0.12, 95% CI: 0.026-0.5, p-adjusted [p-adj] = 0.006) or observation (7.2 mo, 95% CI: 3.4-10.6, HR = 0.04, 95% CI: 0.009-0.2, p-adj < 0.0001). Durvalumab significantly improved median rwPFS compared with observation (HR = 0.37, 95% CI: 0.19-0.71, p-adj = 0.002). Median OS in the ALK TKI and durvalumab cohorts was significantly improved compared with patients on observation (ALK TKI-observation: p = 0.04; durvalumab-observation: p = 0.03). TrAE of any grade occurred in eight (53%) and 11 (37%) patients treated with ALK TKI and durvalumab, respectively. Grade greater than or equal to three trAEs occurred in 27% (n = 4) of patients treated with ALK TKI and 6.7% of patients treated with durvalumab. CONCLUSIONS Patients with ALK-positive NSCLC experience significantly improved rwPFS when treated with consolidation ALK TKI therapy, surpassing outcomes found with either durvalumab or observation. Although both ALK TKI therapy and durvalumab offer an extension in OS compared with observation alone, it seems that ALK TKI therapy is the superior choice, underscoring its pivotal role in enhancing patient survival.
Collapse
Affiliation(s)
- Amin H Nassar
- Yale University School of Medicine, New Haven, Connecticut.
| | | | - Jamie Feng
- Department of Medical Oncology and Hematology, University Health Network, Princess Margaret Cancer Centre, Toronto, Canada
| | - Frances Shepherd
- Department of Medical Oncology and Hematology, University Health Network, Princess Margaret Cancer Centre, Toronto, Canada
| | - Elio Adib
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Justin M Cheung
- Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Jessica J Lin
- Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Yufei Liu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Steven H Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | | | - Thomas J Dilling
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, Florida
| | - David Kaldas
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, Florida; Department of Internal Medicine, University of South Florida, Tampa, Florida; Department of Clinical Oncology, Cairo University, Cairo, Egypt
| | - Jhanelle E Gray
- Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Anastasiya Lobachov
- Institute of Oncology, Chaim Sheba Medical Center, Ramat Gan, Israel; School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jair Bar
- Institute of Oncology, Chaim Sheba Medical Center, Ramat Gan, Israel; School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Heike Luders
- Klinik für Pneumologie-Evangelische Lungenklinik Berlin Buch, Berlin, Germany
| | - Christian Grohe
- Klinik für Pneumologie-Evangelische Lungenklinik Berlin Buch, Berlin, Germany
| | - Shruti Gupta
- Department of Hematology and Medical Oncology, Thoracic Medical Oncology Program, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Ticiana Leal
- Department of Hematology and Medical Oncology, Thoracic Medical Oncology Program, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Bailey Fitzgerald
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Fionnuala Crowley
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Brookdale Department of Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yu Fujiwara
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Thomas U Marron
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Molly Wilgucki
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Joshua Reuss
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Luxi Chen
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Kamya Sankar
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jacqueline V Aredo
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Joel W Neal
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Heather A Wakelee
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Rohit Thummalapalli
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Helena Yu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Ryan Whitaker
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Ana Velazquez
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Meera Ragavan
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California; Division of Research, Kaiser Permanente Northern California, The Permanente Medical Group, Oakland, California
| | - Alessio Cortellini
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy; Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Roma, Italy; Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - David J Kwiatkowski
- Department of Medical Oncology and Hematology, University Health Network, Princess Margaret Cancer Centre, Toronto, Canada
| | | | | | - So Yeon Kim
- Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
64
|
Cheung AH, Mui Z, Yeung WW, Chow C, Yu MF, Chen OH, Wong KY, Xie F, Lau YM, Cheng ASL, Kang W, To KF, Mok TS, Li MS. Germline Human Leukocyte Antigen Status is Associated With Immunotherapy-Induced Pneumonitis and Treatment Response in Patients With Non-Small Cell Lung Cancer With High Programmed Death-Ligand 1 Expression. JTO Clin Res Rep 2025; 6:100754. [PMID: 39897119 PMCID: PMC11786012 DOI: 10.1016/j.jtocrr.2024.100754] [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: 05/23/2024] [Revised: 10/13/2024] [Accepted: 10/20/2024] [Indexed: 02/04/2025] Open
Abstract
Introduction The germline human leukocyte antigen (HLA) status has been found to be associated with immunotherapy outcomes in patients with NSCLC, but its correlation to immunotherapy-induced pneumonitis and prognostic impact in the Asian population remains largely unknown. Methods We evaluated the HLA genotype of the germline and available tumor samples in 42 patients with programmed death-ligand 1 expression of 50% or higher undergoing pembrolizumab immunotherapy. The HLA allele expression was correlated with tumor response, disease survival, and the occurrence of pneumonitis. Results It was observed that the germline HLA-C homozygosity and HLA-DRB1∗13 expression were related to a worse progression-free survival and treatment response. Importantly, all patients (7/7 patients) who developed pneumonitis in our cohort expressed the HLA-DPB1∗02 allele, and the incidence of pneumonitis was 31.8% (7/22 patients) in patients expressing this allele compared with 0% (0/20 patients) in those without this allele (p = 0.009). Investigation of the tumor samples from 15 patients revealed some degree of HLA loss in the HLA class I loci in 40% (6/15) of patients, and no significant difference in tumor mutation burden was found among patients with different treatment responses. Conclusion Taken together, this study evaluated the impact of HLA status in both germline and tumor samples in patients with NSCLC with high programmed death-ligand 1 expression, and the high incidence of immunotherapy-induced pneumonitis in patients expressing the HLA-DPB1∗02 allele may suggest a routine HLA typing and closer monitoring in this patient subset.
Collapse
Affiliation(s)
- Alvin H.K. Cheung
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
- State Key Laboratory of Translational Oncology, the Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Zeta Mui
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Walter W. Yeung
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Chit Chow
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
- State Key Laboratory of Translational Oncology, the Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Mandy F. Yu
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Olivia H. Chen
- Department of Clinical Oncology, Prince of Wales Hospital, the Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Kit-Yee Wong
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Fuda Xie
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Yat Ming Lau
- Department of Clinical Oncology, Prince of Wales Hospital, the Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Alfred S-L. Cheng
- School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Ka-Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
- State Key Laboratory of Translational Oncology, the Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Tony S. Mok
- State Key Laboratory of Translational Oncology, the Chinese University of Hong Kong, Hong Kong, People's Republic of China
- Department of Clinical Oncology, Prince of Wales Hospital, the Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Molly S.C. Li
- State Key Laboratory of Translational Oncology, the Chinese University of Hong Kong, Hong Kong, People's Republic of China
- Department of Clinical Oncology, Prince of Wales Hospital, the Chinese University of Hong Kong, Hong Kong, People's Republic of China
| |
Collapse
|
65
|
Yang H, Zhang Y, Li Y, Peng S, An R, Du N, Cao J, Chu F, Min J. Bispecific c-Met/PD-1 CAR-T Cells Have Enhanced Therapeutic Effects on Solid Tumor. Technol Cancer Res Treat 2025; 24:15330338251336850. [PMID: 40261343 PMCID: PMC12035267 DOI: 10.1177/15330338251336850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 03/05/2024] [Accepted: 03/28/2025] [Indexed: 04/24/2025] Open
Abstract
ObjectiveTo evaluate the killing effect of c-Met CAR-T on tumor cells with different degrees of c-Met expression. It was demonstrated that CAR-T autocrine PD-1 antibody could alleviate immune checkpoint inhibition and enhance the anti-tumor effect of T cells.MethodsThe specificity and clinical significance of c-Met and PD-L1 expression in various solid tumors were verified by bioinformatics analysis. c-Met specific CAR-T and c-Met specific CAR-T secreted by PD-L1 were synthesized, and c-Met CAR-T and c-Met/PD-1 CAR-T were prepared by constructing lentivirus. Flow cytometry was used to verify the positive rate and cell population of CAR-T, western blot was used to verify the secretion of PD-1 antibody, and cck-8 was used to detect the proliferation of CAR-T in tumor cells with different c-Met expression. LDH and ELISA further evaluated the antitumor effects of c-Met CAR-T and c-Met/PD-1 CAR-T in vitro.Resultsc-Met and PD-L1 were expressed in pancreatic cancer, ovarian cancer, esophageal cancer, bladder cancer, glioma and other tumors, and were associated with a variety of immune cell infiltration. Tumor cells with high expression of c-Met can strongly stimulate the proliferation of c-Met CAR-T, and c-Met CAR-T has strong cell lysis ability on tumor cells with high expression of c-Met. Autocrine PD-1 antibody can significantly improve the activity of c-Met CAR T cells, tumor lysis ability and cytokine secretion level.ConclusionThe antitumor activity of c-Met CAR-T is positively correlated with the expression of c-Met. c-Met CAR-T secreted by PD-1 showed enhanced antitumor function in solid tumor treatment.
Collapse
Affiliation(s)
- HuanRan Yang
- Department of Stomatology, BengBu Medical University, BengBu, China
| | - YanJun Zhang
- Department of Life Science, BengBu Medical University, BengBu, China
| | - YanQiu Li
- Department of Basic Medicine, BengBu Medical University, BengBu, China
| | - Shang Peng
- Department of Basic Medicine, BengBu Medical University, BengBu, China
| | - Ran An
- Department of Public foundation, BengBu Medical University, BengBu, China
| | - NaNa Du
- Department of Basic Medicine, BengBu Medical University, BengBu, China
| | - JiaWei Cao
- Department of Clinical medicine, Bengbu Medical University, Bengbu, China
| | - Fei Chu
- The first affiliated Hospital, BengBu Medical University, BengBu, China
| | - JingTing Min
- Department of Basic Medicine, BengBu Medical University, BengBu, China
| |
Collapse
|
66
|
Yang C, Chen Y, Tao T, Xu P, Li M, Deng B, Lu S, Yang M, Wang W, Wang J, Liu SB. ERCC3-Related Genes May Aid in the Prognostic and Immunotherapeutic Analysis of Hepatocellular Carcinoma. Comb Chem High Throughput Screen 2025; 28:808-824. [PMID: 38859773 DOI: 10.2174/0113862073288597240522064027] [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: 12/12/2023] [Revised: 04/06/2024] [Accepted: 04/17/2024] [Indexed: 06/12/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) has high morbidity and mortality worldwide. Excision repair cross-complement 3 (ERCC3), a key functional gene in the nucleotide excision repair (NER) pathway, is commonly mutated or overexpressed in cancers and is thought to be a key gene contributing to the development of HCC. The characteristics of immune cell infiltration in the global tumor microenvironment (TME) mediated by ERCC3 and its related key genes in HCC are still unclear. The aim of this study was to integrate the role of ERCC3-related key genes in assessing the TME cell infiltration characteristics, immunotherapy efficacy, and prognosis of HCC patients. This study provides a theoretical basis for the study of immunological mechanisms and prognosis prediction in HCC. METHODS The HCC cohort from the TCGA database included 50 normal samples and 374 tumor samples to compare the differences in ERCC3-related gene expression and prognosis between liver tumor tissues and normal liver tissues and to analyze the extent to which different genes infiltrated TME cells by quantifying the relative abundance of 24 cells through single-sample genome enrichment analysis (ssGSEA). A risk score associated with the ERCC3 gene was constructed using the least absolute shrinkage and selection operator (LASSO) Cox regression model. RESULTS The expression of 11 ERCC3-related genes was significantly upregulated in HCC tumor tissues compared to normal liver tissues, and high expression of these genes was significantly associated with poor prognosis in HCC patients. The key genes (11 ERCC3-related genes) were closely associated with the nucleic acid reduction signaling pathway in nucleic acid metabolism and the viral oncogenic pathway, suggesting that these key genes may play a role in tumor cell proliferation, migration, and invasion, as well as in the pathogenesis of virus-associated HCC. In addition, the infiltration characteristics of TME immune cells in normal and tumor tissues were different. Immune and mesenchymal activity was significantly lower in tumor tissues than in healthy liver tissues. This study revealed that key genes were significantly positively correlated with CTLA4 and enriched in central memory CD4 T cells, effector memory CD4 T cells, activated CD4 T cells, and type 2 T helper cells. The prognostic model constructed by regression analysis could better distinguish patients into high-risk and low-risk groups, and the survival analysis showed that the survival time of patients with high-risk score subtypes was significantly lower than that of patients with low-risk scores and that the high-risk group contained higher levels of immune-suppressive cells, which may be a mediator of immune escape. Moreover, multivariate analyses showed that the risk score profile is a reliable and unbiased biomarker for assessing the prognosis of HCC patients, and its value in predicting the outcome of immunotherapy was also confirmed. CONCLUSION This study revealed a novel genetic signature that is significantly associated with TME cell infiltration and prognosis in HCC patients. It demonstrated that the combined action of multiple key genes associated with ERCC3 plays a crucial role in shaping the diversity and complexity of TME cell infiltrates. Evaluating the combined characteristics of multiple key genes associated with ERCC3 can help predict the outcome of immunotherapy in patients and provide new potential targets for immuno-individualized therapeutic studies on HCC.
Collapse
Affiliation(s)
- Chen Yang
- College of Life Science, North China University of Science and Technology, Bohai Avenue 21, Tangshan 063210, China
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, Kehua Road 28, Suzhou, China 215009
| | - Yao Chen
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, Kehua Road 28, Suzhou, China 215009
| | - Tao Tao
- Department of Respiratory and Critical Medicine, The Fifth People's Hospital of Suzhou, The Affiliated Infectious Diseases Hospital, Suzhou Medical College of Soochow University, Suzhou 215100, China
| | - Ping Xu
- Department of Clinical Laboratory, The Fifth People's Hospital of Suzhou, The Affiliated Infectious Diseases Hospital, Suzhou Medical College of Soochow University, Suzhou 215100, China
| | - Miaomiao Li
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, Kehua Road 28, Suzhou, China 215009
| | - Bicheng Deng
- College of Life Science, North China University of Science and Technology, Bohai Avenue 21, Tangshan 063210, China
| | - Sihan Lu
- College of Life Science, North China University of Science and Technology, Bohai Avenue 21, Tangshan 063210, China
| | - Minfeng Yang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Weijie Wang
- College of Life Science, North China University of Science and Technology, Bohai Avenue 21, Tangshan 063210, China
| | - Jinghan Wang
- Department of Hepatobiliary and Pancreatic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Song-Bai Liu
- College of Life Science, North China University of Science and Technology, Bohai Avenue 21, Tangshan 063210, China
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, Kehua Road 28, Suzhou, China 215009
| |
Collapse
|
67
|
Beigi A, Naghib SM, Matini A, Tajabadi M, Mozafari MR. Lipid-Based Nanocarriers for Targeted Gene Delivery in Lung Cancer Therapy: Exploring a Novel Therapeutic Paradigm. Curr Gene Ther 2025; 25:92-112. [PMID: 38778601 DOI: 10.2174/0115665232292768240503050508] [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: 11/14/2023] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 05/25/2024]
Abstract
Lung cancer is a significant cause of cancer-related death worldwide. It can be broadly categorised into small-cell lung cancer (SCLC) and Non-small cell lung cancer (NSCLC). Surgical intervention, radiation therapy, and the administration of chemotherapeutic medications are among the current treatment modalities. However, the application of chemotherapy may be limited in more advanced stages of metastasis due to the potential for adverse effects and a lack of cell selectivity. Although small-molecule anticancer treatments have demonstrated effectiveness, they still face several challenges. The challenges at hand in this context comprise insufficient solubility in water, limited bioavailability at specific sites, adverse effects, and the requirement for epidermal growth factor receptor inhibitors that are genetically tailored. Bio-macromolecular drugs, including small interfering RNA (siRNA) and messenger RNA (mRNA), are susceptible to degradation when exposed to the bodily fluids of humans, which can reduce stability and concentration. In this context, nanoscale delivery technologies are utilised. These agents offer encouraging prospects for the preservation and regulation of pharmaceutical substances, in addition to improving the solubility and stability of medications. Nanocarrier-based systems possess the notable advantage of facilitating accurate and sustained drug release, as opposed to traditional systemic methodologies. The primary focus of scientific investigation has been to augment the therapeutic efficacy of nanoparticles composed of lipids. Numerous nanoscale drug delivery techniques have been implemented to treat various respiratory ailments, such as lung cancer. These technologies have exhibited the potential to mitigate the limitations associated with conventional therapy. As an illustration, applying nanocarriers may enhance the solubility of small-molecule anticancer drugs and prevent the degradation of bio-macromolecular drugs. Furthermore, these devices can administer medications in a controlled and extended fashion, thereby augmenting the therapeutic intervention's effectiveness and reducing adverse reactions. However, despite these promising results, challenges remain that must be addressed. Multiple factors necessitate consideration when contemplating the application of nanoparticles in medical interventions. To begin with, the advancement of more efficient delivery methods is imperative. In addition, a comprehensive investigation into the potential toxicity of nanoparticles is required. Finally, additional research is needed to comprehend these treatments' enduring ramifications. Despite these challenges, the field of nanomedicine demonstrates considerable promise in enhancing the therapy of lung cancer and other respiratory diseases.
Collapse
Affiliation(s)
- Anahita Beigi
- 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
| | - Amir Matini
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - Maryam Tajabadi
- School of Metallurgy and Materials Engineering, Iran University of Science and Technology (IUST), Narmak, Tehran, 16844, Iran
| | - Mohammad Reza Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| |
Collapse
|
68
|
Nath P, Alfarsi LH, El-Ansari R, Masisi BK, Erkan B, Fakroun A, Ellis IO, Rakha EA, Green AR. The amino acid transporter SLC7A11 expression in breast cancer. Cancer Biol Ther 2024; 25:2291855. [PMID: 38073087 PMCID: PMC10761065 DOI: 10.1080/15384047.2023.2291855] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
Breast cancer (BC), characterized by its diverse molecular profiles and clinical outcomes, presents a significant challenge in the development of effective therapeutic strategies. Metabolic reprogramming, a defining characteristic of cancer, has emerged as a promising target for novel therapies. SLC7A11, an amino acid transporter that facilitates cysteine uptake in exchange for glutamate, plays a crucial role in sustaining the altered metabolism of cancer cells. This study delves into the comprehensive analysis of SLC7A11 at the genomic, transcriptomic, and protein levels in extensive BC datasets to elucidate its potential role in different BC subtypes. SLC7A11 gene copy number and mRNA expression were evaluated using the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) cohort (n = 1,980) and Breast Cancer Gene Expression Miner (n = 4,712). SLC7A11 protein was assessed using immunohistochemistry in a large BC cohort (n = 1,981). Additionally, The Cancer Genome Atlas (TCGA) dataset was used to explore SLC7A11 DNA methylation patterns using MethSurv (n = 782) and association of SLC7A11 mRNA expression with immune infiltrates using TIMER (n = 1,100). High SLC7A11 mRNA and SLC7A11 protein expression were significantly associated with high tumor grade (p ≤ .02), indicating a potential role in cancer progression. Interestingly, SLC7A11 copy number gain was observed in HER2+ tumors (p = .01), suggesting a subtype-specific association. In contrast, SLC7A11 mRNA expression was higher in the basal-like/triple-negative (TN; p < .001) and luminal B tumors (p = .02), highlighting its differential expression across BC subtypes. Notably, high SLC7A11 protein expression was predominantly observed in Estrogen Receptor (ER)-negative and Triple Negative (TN) BC, suggesting a role in these aggressive subtypes. Further analysis revealed that SLC7A11 was positively correlated with other amino acid transporters and enzymes associated with glutamine metabolism, implying a coordinated role in metabolic regulation. Additionally, SLC7A11 gene expression was positively associated with neutrophil and macrophage infiltration, suggesting a potential link between SLC7A11 and tumor immunity. Our findings suggest that SLC7A11 plays a significant role in BC metabolism, demonstrating differential expression across subtypes and associations with poor patient outcomes. Further functional studies are warranted to elucidate the precise mechanisms by which SLC7A11 contributes to BC progression and to explore its potential as a therapeutic target.
Collapse
Affiliation(s)
- Preyanka Nath
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Lutfi H. Alfarsi
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Rokaya El-Ansari
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Brendah K. Masisi
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Busra Erkan
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Ali Fakroun
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Ian O. Ellis
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
- Cellular Pathology, Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Emad A. Rakha
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
- Cellular Pathology, Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Andrew R. Green
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| |
Collapse
|
69
|
Guerini AE, Marvaso G, Tonoli S, Corrao G, Teriaca MA, Sepulcri M, Scricciolo M, Gava A, Montrone S, Giaj-Levra N, Chiorda BN, Mantello G, Fiorica F, Borghesi S, Belgioia L, Caroli A, Fiorentino A, Radioncovid Study Group, Jereczek-Fossa BA, Magrini SM, Buglione M. RadiOnCOVID: Multicentric Analysis of the Impact of COVID-19 on Patients Undergoing Radiation Therapy in Italy. Cancers (Basel) 2024; 17:86. [PMID: 39796715 PMCID: PMC11720150 DOI: 10.3390/cancers17010086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
METHODS AND MATERIALS Patients with ongoing or planned anticancer treatment at 19 Italian Radiation Oncology centers were included in the study retrospectively from 3 February 2020 to 31 December 2020 and prospectively from 1 January 2021 to 31 May 2021. Anonymized data were processed through a specific website and database. Antineoplastic treatment characteristics and timing and outcomes of COVID-19 and its impact on radiotherapy or systemic therapy were described. RESULTS The retrieved cohort included 41,039 patients that received treatment or were planned for therapy in the study period. Overall, 123 patients had a confirmed COVID-19 diagnosis during antineoplastic treatment (group A) and 99 patients before treatment start (group B). The incidence of COVID-19 across the whole cohort in the index period was 0.54% (groups A + B) and 0.30% considering only group A. A total of 60 patients developed severe COVID-19, and a total of 45 patients died as a consequence of the infection (incidence of 0.15% and 0.11%, respectively). Nonetheless, mortality among COVID-19 patients was high, with an attributable death rate after confirmed infection of 20.27%. Among the 123 patients in group A, 37.4% required temporary treatment suspension, 32.5% definitive suspension and 37 patients continued treatment while positive. As for the 99 patients in group B, 53.5% experienced temporary delay, 20.2% experienced definitive treatment suspension and 26.3% had no delay. CONCLUSIONS Most of the patients with a COVID-19 diagnosis in our cohort recovered and completed their treatment; nonetheless, the attributable death rate after confirmed infection was 20.27%, and mortality was high among cancer patients with severe COVID-19 presentation. The global incidence of death due to COVID-19 or severe COVID-19 was low and decreased over time. Radiation oncology activity could be safely continued during the COVID-19 pandemic with the adoption of adequate preventive measures.
Collapse
Affiliation(s)
- Andrea Emanuele Guerini
- Department of Radiation Oncology, Istituto del Radio O. Alberti, Spedali Civili Hospital, Piazzale Spedali Civili 1, 25121 Brescia, Italy; (S.M.M.); (M.B.)
- Department of Radiation Oncology, Università degli Studi di Brescia, 25123 Brescia, Italy
| | - Giulia Marvaso
- Division of Radiation Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (G.M.); (G.C.); (B.A.J.-F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Sandro Tonoli
- UOC Radioterapia e Medicina Nucleare, ASST di Cremona, 26100 Cremona, Italy;
| | - Giulia Corrao
- Division of Radiation Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (G.M.); (G.C.); (B.A.J.-F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Maria Ausilia Teriaca
- Radiotherapy and Radiosurgery Department, Humanitas Research Hospital, 20089 Rozzano, Italy;
| | - Matteo Sepulcri
- Radiotherapy, Veneto Institute of Oncology, IOV-IRCCS Padua, 35128 Padova, Italy;
| | - Melissa Scricciolo
- Radiation Oncology Division, Clinical Oncology Department, Ospedale dell’Angelo, 30174 Venice, Italy;
| | - Alessandro Gava
- Department of Radiation Oncology, Azienda Ospedaliera ULSS 9, 31100 Treviso, Italy;
| | - Sabrina Montrone
- U.O. Radioterapia, Azienda Ospedaliero-Universitaria Pisana, Via Roma 69, 56120 Pisa, Italy;
| | - Niccolò Giaj-Levra
- Dipartimento di Radioterapia Oncologica Avanzata, IRCCS Sacro Cuore Don Calabria Hospital, Via Don Angelo Sempreboni, 37124 Negrar di Valpolicella, Italy;
| | - Barbara Noris Chiorda
- Department of Radiation Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Giovanna Mantello
- SOD Radioterapia (Dipartimento Medicina Interna), Azienda Ospedaliero Universitaria delle Marche, 60002 Ancona, Italy;
| | - Francesco Fiorica
- Dipartimento di Oncologia Clinica, AULSS 9 Scaligera, 37122 Verona, Italy;
| | - Simona Borghesi
- Radiation Oncology Unit of Arezzo-Valdarno, Azienda USL Toscana Sud Est, 52100 Arezzo, Italy;
| | - Liliana Belgioia
- U.O. Radioterapia Oncologica, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
- Dipartimento di Scienze della Salute (DISSAL), Università Degli Studi di Genova, 16126 Genova, Italy
| | - Angela Caroli
- SOC Oncologia Radioterapica, Centro di Riferimento Oncologico, Istituto di Ricovero e Cura a Carattere Scientifico, 33081 Aviano, Italy;
| | - Alba Fiorentino
- Department of Medicine and Surgery, LUM University, 70010 Casamassima, Italy;
- Department of Radiation Oncology, Generale Regional Hospital F Miulli, 70021 Acquaviva delle Fonti, Italy
| | | | - Barbara Alicja Jereczek-Fossa
- Division of Radiation Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (G.M.); (G.C.); (B.A.J.-F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Stefano Maria Magrini
- Department of Radiation Oncology, Istituto del Radio O. Alberti, Spedali Civili Hospital, Piazzale Spedali Civili 1, 25121 Brescia, Italy; (S.M.M.); (M.B.)
- Department of Radiation Oncology, Università degli Studi di Brescia, 25123 Brescia, Italy
| | - Michela Buglione
- Department of Radiation Oncology, Istituto del Radio O. Alberti, Spedali Civili Hospital, Piazzale Spedali Civili 1, 25121 Brescia, Italy; (S.M.M.); (M.B.)
- Department of Radiation Oncology, Università degli Studi di Brescia, 25123 Brescia, Italy
| |
Collapse
|
70
|
Seishima R, Tachimori H, Fukuda K, Ikeda N, Miyata H, Fushimi K, Kitagawa Y. Impact of COVID-19 on hospital visit behaviour in cancer patients in Japan: a nationwide study. BMJ Open 2024; 14:e084630. [PMID: 39725419 PMCID: PMC11683943 DOI: 10.1136/bmjopen-2024-084630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 11/04/2024] [Indexed: 12/28/2024] Open
Abstract
OBJECTIVES Lockdowns and outing restrictions during the COVID-19 pandemic may have altered attitudes towards hospital visits. This study aimed to investigate changes in long-distance visits outside of secondary healthcare service areas (SHSA) among cancer patients in Japan. DESIGN Retrospective observational study. SETTING Inpatient data from the Japanese Diagnosis Procedure Combination database. PRIMARY AND SECONDARY OUTCOME MEASURES We retrospectively analysed inpatient data from January 2018 to December 2021, extracted from the Japanese Diagnosis Procedure Combination database. The study examined whether the patient's hospital was within their residence's SHSA and compared the admission rates before and after the pandemic. RESULTS The data of a total of 2 394 760 patients were analysed. Admission rates outside the SHSA significantly decreased after the pandemic compared with prepandemic years (26.67% and 27.58%, respectively, p<0.001). Significant reductions were observed in nearly all age groups over 30 years old, but not in younger ones. Characteristics of patients' residences, including COVID-19 infection rates, population density and the number of regional cancer hospitals, were also influential factors. The cancer site was a significant factor, with the respiratory system showing a 3.77% decrease, particularly a 5.29% decrease in those who needed surgeries that are not highly specialised (p<0.001). CONCLUSIONS Admission to distant hospitals among cancer patients decreased following the COVID-19 pandemic, indicating a restraint in their behaviour of visiting distant hospitals. The appropriate allocation of specialised hospitals will serve as a measure for the next pandemic.
Collapse
Affiliation(s)
- Ryo Seishima
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hisateru Tachimori
- Department of Health Policy and Management, Keio University School of Medicine, Tokyo, Japan
| | - Kazumasa Fukuda
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Norihiko Ikeda
- Department of Surgery, Tokyo Medical University, Tokyo, Japan
| | - Hiroaki Miyata
- Department of Health Policy and Management, Keio University School of Medicine, Tokyo, Japan
- Department of Healthcare Quality Assessment, The University of Tokyo, Graduate School of Medicine, Tokyo, Japan
| | - Kiyohide Fushimi
- Department of Health Policy and Informatics, Institute of Science Graduate School, Tokyo, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
71
|
Alsharksi AN, Sirekbasan S, Gürkök-Tan T, Mustapha A. From Tradition to Innovation: Diverse Molecular Techniques in the Fight Against Infectious Diseases. Diagnostics (Basel) 2024; 14:2876. [PMID: 39767237 PMCID: PMC11674978 DOI: 10.3390/diagnostics14242876] [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: 10/22/2024] [Revised: 11/15/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025] Open
Abstract
Infectious diseases impose a significant burden on global health systems due to high morbidity and mortality rates. According to the World Health Organization, millions die from infectious diseases annually, often due to delays in accurate diagnosis. Traditional diagnostic methods in clinical microbiology, primarily culture-based techniques, are time-consuming and may fail with hard-to-culture pathogens. Molecular biology advancements, notably the polymerase chain reaction (PCR), have revolutionized infectious disease diagnostics by allowing rapid and sensitive detection of pathogens' genetic material. PCR has become the gold standard for many infections, particularly highlighted during the COVID-19 pandemic. Following PCR, next-generation sequencing (NGS) has emerged, enabling comprehensive genomic analysis of pathogens, thus facilitating the detection of new strains and antibiotic resistance tracking. Innovative approaches like CRISPR technology are also enhancing diagnostic precision by identifying specific DNA/RNA sequences. However, the implementation of these methods faces challenges, particularly in low- and middle-income countries due to infrastructural and financial constraints. This review will explore the role of molecular diagnostic methods in infectious disease diagnosis, comparing their advantages and limitations, with a focus on PCR and NGS technologies and their future potential.
Collapse
Affiliation(s)
- Ahmed Nouri Alsharksi
- Department of Microbiology, Faculty of Medicine, Misurata University, Misrata 93FH+66F, Libya;
| | - Serhat Sirekbasan
- Department of Medical Laboratory Techniques, Şabanözü Vocational School, Çankırı Karatekin University, Çankırı 18650, Turkey
| | - Tuğba Gürkök-Tan
- Department of Field Crops, Food and Agriculture Vocational School, Çankırı Karatekin University, Çankırı 18100, Turkey;
| | - Adam Mustapha
- Department of Microbiology, Faculty of Life Sciences, University of Maiduguri, Maiduguri 600104, Nigeria;
| |
Collapse
|
72
|
Chen C, Lu F, Huang H, Pan Y. Translating CD47-targeted therapy in gastrointestinal cancers: Insights from preclinical to clinical studies. iScience 2024; 27:111478. [PMID: 39720535 PMCID: PMC11667074 DOI: 10.1016/j.isci.2024.111478] [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] [Indexed: 12/26/2024] Open
Abstract
This review presents a thorough investigation of the role of CD47 in gastrointestinal cancers. We performed a comprehensive, in-depth review of over 100 preclinical and clinical studies focused on inhibiting CD47. The research highlights the potential of targeted CD47 to enhance existing treatments by boosting the immune response to cancer cells. Considering the essential need to balance the toxicity and efficacy of CD47 inhibition, our review emphasizes the need to optimize CD47 inhibitors. We also demonstrate the necessity of combining CD47 antibodies with conventional chemotherapy, radiotherapy, or other targeted therapies to enhance treatment effectiveness. Finally, we propose the integration of CD47-targeted therapies into treatment plans as a promising approach to reshape the therapeutic landscape of gastrointestinal cancers. Continued research in this field holds great potential for improving the outcomes of gastrointestinal cancer patients and overcoming the challenges associated with this formidable spectrum of diseases.
Collapse
Affiliation(s)
- Changgan Chen
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou 350001, People's Republic of China
| | - Fengchun Lu
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou 350001, People's Republic of China
| | - Heguang Huang
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou 350001, People's Republic of China
| | - Yu Pan
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou 350001, People's Republic of China
- The Cancer Center, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| |
Collapse
|
73
|
Kou M, Deng Y. Circulating tumor DNA as a predictive biomarker for treatment response and survival in metastatic colorectal cancer. Int J Colorectal Dis 2024; 39:203. [PMID: 39681775 DOI: 10.1007/s00384-024-04785-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/09/2024] [Indexed: 12/18/2024]
Abstract
PURPOSE To explore the potential of circulating tumor DNA (ctDNA) as a prognostic biomarker to predict treatment response and survival outcomes in patients with metastatic colorectal cancer (mCRC). METHODS A retrospective analysis was conducted on 134 patients with mCRC who were treated between January 2020 and December 2021. The patients were classified into ctDNA-negative and ctDNA-positive groups based on plasma ctDNA detection. Demographic, clinical, and laboratory parameters, treatment response, survival outcomes, and adverse events were recorded and analyzed. RESULTS No significant differences were observed in baseline characteristics between the two groups. Compared to the ctDNA-positive patients, ctDNA-negative patients exhibited superior outcomes, including a higher objective response rate (65.22% vs. 46.15%), disease control rate (81.16% vs. 63.08%), progression-free survival (8.24 ± 1.02 vs. 7.86 ± 0.91 months), overall survival (24.58 ± 3.58 vs. 23.27 ± 3.46 months), and 1-year survival rate (73.91% vs. 55.38%). The ctDNA-positive group had a significantly higher incidence of adverse events. Correlation analyses revealed significant associations between ctDNA status, tumor markers, treatment response, and survival outcomes. CONCLUSIONS ctDNA is a promising noninvasive biomarker for predicting treatment response, survival, and adverse events in mCRC, potentially guiding personalized therapeutic strategies.
Collapse
Affiliation(s)
- Mengying Kou
- Department of Oncology, Maoming People's Hospital, Maoming City, Guangdong Province, China
| | - Ying Deng
- Department of Gastroenterology, Maoming People's Hospital, No.101 Weimin Road, Maonan District, Maoming City, Guangdong Province, 525000, China.
| |
Collapse
|
74
|
Lee WL, Chou WH, Chang WP, Chang TW, Kuo CN, Chang WC. Cost-effectiveness analysis of pembrolizumab with chemotherapy for metastatic nonsquamous non-small cell lung cancer in Taiwan. J Food Drug Anal 2024; 32:568-576. [PMID: 39752871 PMCID: PMC11698590 DOI: 10.38212/2224-6614.3536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/29/2024] [Indexed: 01/07/2025] Open
Abstract
This study was aimed to evaluate the cost-effectiveness of pembrolizumab with chemotherapy (pembrolizumab combination therapy) and compare it with standard-of-care platinum-based chemotherapy (chemotherapy alone) as a first-line treatment for metastatic nonsquamous NSCLC from the perspective of Taiwan's third-party-payer public health-care system. We used a partitioned survival model with an estimated time horizon of 10 years. The partitioned survival model uses Kaplan-Meier estimates of progression-free and overall survival from the KEYNOTE-189 clinical trial. The quality-adjusted life-year (QALY) values were based on utility values by progression status calculated from the KEYNOTE-189 trial. This study examined costs related to treatment regimens, disease management, second-line therapy, end-of-life care, and adverse event management. Cost and utility were discounted at 3% per year. Probabilistic and deterministic sensitivity analyses were performed to test the robustness of the results. The willingness-to-pay threshold was set at 3 × Taiwan's gross domestic product (GDP), equivalent to NT$2,788,290. In the base-case scenario, pembrolizumab combination therapy resulted in an expected gain of 0.89 QALYs and an incremental cost of NT$2,201,203 relative to chemotherapy alone. The ICER was NT$2,478,601/QALY. In the analysis of the PD-L1 tumor proportion score (TPS) ≥ 50% subgroup, the patients who received pembrolizumab combination therapy gained 1.12 QALYs more than those who received chemotherapy alone, and the incremental cost was NT$2,522,528. Therefore, the ICER for this subset of patients was NT$2,258,358/QALY. In conclusion, pembrolizumab combination therapy is a cost-effective option for first-line treatment of metastatic nonsquamous NSCLC. The relative cost-effectiveness of pembrolizumab combination therapy is greatest for patients with PD-L1 TPS ≥50%.
Collapse
Affiliation(s)
- Wei-Ling Lee
- Department of Pharmacy, Taipei Chang Gung Memorial Hospital, Taipei,
Taiwan
- Department of Clinical Pharmacy, School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei,
Taiwan
| | - Wan-Hsuan Chou
- Department of Clinical Pharmacy, School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei,
Taiwan
| | - Wei-Pin Chang
- School of Health Care Administration, College of Management, Taipei Medical University, Taipei,
Taiwan
| | - Tsung-Wei Chang
- Department of Pharmacy, Yuanlin Christian Hospital, Changhua,
Taiwan
| | - Chun-Nan Kuo
- Department of Clinical Pharmacy, School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei,
Taiwan
- Department of Pharmacy, Wan Fang Hospital, Taipei Medical University, Taipei,
Taiwan
| | - Wei-Chiao Chang
- Department of Clinical Pharmacy, School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei,
Taiwan
- Department of Pharmacy, Wan Fang Hospital, Taipei Medical University, Taipei,
Taiwan
- Master Program in Clinical Genomics and Proteomics, College of Pharmacy, Taipei Medical University, Taipei,
Taiwan
- Integrative Research Center for Critical Care, Department of Pharmacy, Wan Fang Hospital, Taipei Medical University, Taipei,
Taiwan
| |
Collapse
|
75
|
Lutimba S, Saleem B, Aleem E, Mansour MA. In Silico Analysis of Triamterene as a Potential Dual Inhibitor of VEGFR-2 and c-Met Receptors. J Xenobiot 2024; 14:1962-1987. [PMID: 39728413 DOI: 10.3390/jox14040105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024] Open
Abstract
The vascular endothelial growth factor receptor 2 (VEGFR2) and the hepatocyte growth factor receptor (C-Met) are critical receptors for signaling pathways controlling crucial cellular processes such as cell growth, angiogenesis and tissue regeneration. However, dysregulation of these proteins has been reported in different diseases, particularly cancer, where these proteins promote tumour growth, invasiveness, metastasis and resistance to conventional therapies. The identification of dual inhibitors targeting both VEGFR-2 and c-Met has emerged as a strategic therapeutic approach to overcome the limitations and resistance mechanisms associated with single-target therapies in clinical settings. Through molecular dynamics simulations and comparative docking analysis, we tested the inhibitory potential of 2,016 Food and Drug Administration (FDA)-approved drugs targeting VEGFR-2 and/or c-Met receptors. The results revealed that entacapone and telmisartan are potent and selective inhibitors for c-Met and VEGFR-2, respectively. Interestingly, triamterene was identified as a promising dual inhibitor, demonstrating specific and significant binding affinity to both proteins. Molecular dynamics simulations revealed key interactions between the identified compounds and critical residues in the catalytic domains of both VEGFR-2 (e.g., Lys868, Asp1028, Asp1046) and c-Met (e.g., Asp1204, His1202, Asp1222), providing insights into their mechanism of action. These findings underscore the therapeutic potential of triamterene in targeting multiple signaling pathways involved in cancer progression, metastasis and poor prognosis in patients. Our study provides a foundational framework for the development of novel anticancer compounds able to target multiple pathways in cancer. Further preclinical and clinical investigations are needed to validate the efficacy of these compounds in clinical settings and to test their ability to overcome resistance and improve patient outcome.
Collapse
Affiliation(s)
- Stuart Lutimba
- Cancer Biology and Therapy Laboratory, School of Applied and Health Sciences, London South Bank University, London SE1 0AA, UK
| | - Baraya Saleem
- Cancer Biology and Therapy Laboratory, School of Applied and Health Sciences, London South Bank University, London SE1 0AA, UK
| | - Eiman Aleem
- Cancer Biology and Therapy Laboratory, School of Applied and Health Sciences, London South Bank University, London SE1 0AA, UK
| | - Mohammed A Mansour
- Cancer Biology and Therapy Laboratory, School of Applied and Health Sciences, London South Bank University, London SE1 0AA, UK
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta 31527, Egypt
| |
Collapse
|
76
|
Bairqdar A, Karitskaya PE, Stepanov GA. Expanding Horizons of CRISPR/Cas Technology: Clinical Advancements, Therapeutic Applications, and Challenges in Gene Therapy. Int J Mol Sci 2024; 25:13321. [PMID: 39769084 PMCID: PMC11678091 DOI: 10.3390/ijms252413321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
CRISPR-Cas technology has transformed the field of gene editing, opening new possibilities for treatment of various genetic disorders. Recent years have seen a surge in clinical trials using CRISPR-Cas-based therapies. This review examines the current landscape of CRISPR-Cas implementation in clinical trials, with data from key registries, including the Australian New Zealand Clinical Trials Registry, the Chinese Clinical Trial Register, and ClinicalTrials.gov. Emphasis is placed on the mechanism of action of tested therapies, the delivery method, and the most recent findings of each clinical trial.
Collapse
Affiliation(s)
- Ahmad Bairqdar
- Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia;
| | - Polina E. Karitskaya
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk 630090, Russia;
| | - Grigory A. Stepanov
- Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia;
| |
Collapse
|
77
|
Lycan Jr TW, Norton DL, Ohar JA. COPD and Immune Checkpoint Inhibitors for Cancer: A Literature Review. Int J Chron Obstruct Pulmon Dis 2024; 19:2689-2703. [PMID: 39677829 PMCID: PMC11639883 DOI: 10.2147/copd.s490252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 12/02/2024] [Indexed: 12/17/2024] Open
Abstract
Purpose Immune checkpoint inhibitors are a standard treatment option for many patients with cancer and are most frequently used to treat lung cancer. Chronic obstructive pulmonary disease (COPD) is the most common comorbidity of patients with lung cancer. As the cancer-specific survival of patients with lung cancer continues to increase with modern treatments, it is critical to optimize comorbidities to improve overall survival. This literature review aimed to summarize current research on the impact of COPD upon immunotherapy outcomes. Methods A comprehensive search was conducted in the PubMed database using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Inclusion criteria focused on peer-reviewed articles published between 2010 and 2024 that addressed COPD, cancer, and immune checkpoint inhibitors. The study team screened the studies for relevance and then synthesized them narratively. Results This review identified 37 studies that met the inclusion criteria. Findings suggest that COPD is predictive of improved efficacy but slightly worse toxicity from immune checkpoint inhibitor therapy. The chronic inflammation of COPD leads to immune exhaustion including the overexpression of immune checkpoints on T-cells. Particularly within "hot" tumors that have higher concentrations of tumor-infiltrating lymphocytes, the COPD-related increase in programmed cell death protein 1 (PD-1) signaling likely creates sensitivity to immune checkpoint inhibitors. However, COPD can also lead to respiratory dysfunction, debility, and interstitial lung disease; each of which increases the severity of immune-related adverse events. Conclusion COPD is a critical comorbidity that has a significant impact on many patients with cancer who receive treatment with immune checkpoint inhibitors. Future research is needed to design interventions to optimize COPD care in this high-risk patient population.
Collapse
Affiliation(s)
- Thomas W Lycan Jr
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Dustin L Norton
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Jill A Ohar
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
78
|
Chen P, Ye Q, Liang S, Zeng L. Cephaeline promotes ferroptosis by targeting NRF2 to exert anti-lung cancer efficacy. PHARMACEUTICAL BIOLOGY 2024; 62:195-206. [PMID: 38339810 PMCID: PMC10860416 DOI: 10.1080/13880209.2024.2309891] [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: 05/15/2023] [Accepted: 01/20/2024] [Indexed: 02/12/2024]
Abstract
CONTEXT Cephaeline is a natural product isolated from ipecac (Cephaelis ipecacuanha [Brot.] A. Rich. [Rubiaceae]). It exhibits promising anti-lung cancer activity and ferroptosis induction may be a key mechanism for its anti-lung cancer effect. OBJECTIVES This study investigates the anti-lung cancer activity and mechanisms of cephaeline both in vitro and in vivo. MATERIALS AND METHODS H460 and A549 lung cancer cells were used. The cephaeline inhibition rate on lung cancer cells was detected via a Cell Counting Kit-8 assay after treatment with cephaeline for 24 h. Subsequently, the concentrations of 25, 50 and 100 nM were used for in vitro experiments. In addition, the antitumour effects of cephaeline (5, 10 mg/kg) in vivo were evaluated after 12 d of cephaeline treatment. RESULTS Cephaeline showed significant inhibitory effects on lung cancer cells, and the IC50 of cephaeline on H460 and A549 at 24, 48 and 72 h were 88, 58 and 35 nM, respectively, for H460 cells and 89, 65 and 43 nM, respectively, for A549 cells. Meanwhile, we demonstrated that ferroptosis is the key mechanism of cephaeline against lung cancer. Finally, we found that cephaeline induced ferroptosis in lung cancer cells by targeting NRF2. DISCUSSION AND CONCLUSION We demonstrated for the first time that cephaeline inhibits NRF2, leading to ferroptosis in lung cancer cells. These findings may contribute to the development of innovative therapeutics for lung cancer.
Collapse
Affiliation(s)
- Peng Chen
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, PR China
| | - Qingxuan Ye
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, PR China
| | - Shang Liang
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, PR China
| | - Linghui Zeng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, PR China
| |
Collapse
|
79
|
Wang X, Li QQ, Tang YX, Li Y, Zhang L, Xu FF, Fu XL, Ye K, Ma JQ, Guo SM, Ma FY, Liu ZY, Shi XH, Li XM, Sun HM, Wu Y, Zhang WY, Ye LH. Oncoprotein LAMTOR5-mediated CHOP silence via DNA hypermethylation and miR-182/miR-769 in promotion of liver cancer growth. Acta Pharmacol Sin 2024; 45:2625-2645. [PMID: 38942954 PMCID: PMC11579023 DOI: 10.1038/s41401-024-01310-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/08/2024] [Indexed: 06/30/2024]
Abstract
C/EBP homologous protein (CHOP) triggers the death of multiple cancers via endoplasmic reticulum (ER) stress. However, the function and regulatory mechanism of CHOP in liver cancer remain elusive. We have reported that late endosomal/lysosomal adapter, mitogen-activated protein kinase and mTOR activator 5 (LAMTOR5) suppresses apoptosis in various cancers. Here, we show that the transcriptional and posttranscriptional inactivation of CHOP mediated by LAMTOR5 accelerates liver cancer growth. Clinical bioinformatic analysis revealed that the expression of CHOP was low in liver cancer tissues and that its increased expression predicted a good prognosis. Elevated CHOP contributed to destruction of LAMTOR5-induced apoptotic suppression and proliferation. Mechanistically, LAMTOR5-recruited DNA methyltransferase 1 (DNMT1) to the CpG3 region (-559/-429) of the CHOP promoter and potentiated its hypermethylation to block its interaction with general transcription factor IIi (TFII-I), resulting in its inactivation. Moreover, LAMTOR5-enhanced miR-182/miR-769 reduced CHOP expression by targeting its 3'UTR. Notably, lenvatinib, a first-line targeted therapy for liver cancer, could target the LAMTOR5/CHOP axis to prevent liver cancer progression. Accordingly, LAMTOR5-mediated silencing of CHOP via the regulation of ER stress-related apoptosis promotes liver cancer growth, providing a theoretical basis for the use of lenvatinib for the treatment of liver cancer.
Collapse
Affiliation(s)
- Xue Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Qian-Qian Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yan-Xin Tang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Ye Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Lu Zhang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
- Chinese Academy of Medical Sciences & Peking Union Medical College Institute of Biomedical Engineering, Tianjin, 300192, China
| | - Fei-Fei Xu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
- Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, 300192, China
| | - Xue-Li Fu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Kai Ye
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jia-Qi Ma
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Shi-Man Guo
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Fang-Yuan Ma
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Zhi-Yu Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Xu-He Shi
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Xian-Meng Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Hui-Min Sun
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yue Wu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, 250014, China
| | - Wei-Ying Zhang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| | - Li-Hong Ye
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
80
|
Thapa R, Gupta S, Gupta G, Bhat AA, Smriti, Singla M, Ali H, Singh SK, Dua K, Kashyap MK. Epithelial-mesenchymal transition to mitigate age-related progression in lung cancer. Ageing Res Rev 2024; 102:102576. [PMID: 39515620 DOI: 10.1016/j.arr.2024.102576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/27/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Epithelial-Mesenchymal Transition (EMT) is a fundamental biological process involved in embryonic development, wound healing, and cancer progression. In lung cancer, EMT is a key regulator of invasion and metastasis, significantly contributing to the fatal progression of the disease. Age-related factors such as cellular senescence, chronic inflammation, and epigenetic alterations exacerbate EMT, accelerating lung cancer development in the elderly. This review describes the complex mechanism among EMT and age-related pathways, highlighting key regulators such as TGF-β, WNT/β-catenin, NOTCH, and Hedgehog signalling. We also discuss the mechanisms by which oxidative stress, mediated through pathways involving NRF2 and ROS, telomere attrition, regulated by telomerase activity and shelterin complex, and immune system dysregulation, driven by alterations in cytokine profiles and immune cell senescence, upregulate or downregulate EMT induction. Additionally, we highlighted pathways of transcription such as SNAIL, TWIST, ZEB, SIRT1, TP53, NF-κB, and miRNAs regulating these processes. Understanding these mechanisms, we highlight potential therapeutic interventions targeting these critical molecules and pathways.
Collapse
Affiliation(s)
- Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Saurabh Gupta
- Chameli Devi Institute of Pharmacy, Department of Pharmacology, Indore, Madhya Pradesh, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome-Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Smriti
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Madhav Singla
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Manoj Kumar Kashyap
- Molecular Oncology Laboratory, Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Panchgaon (Manesar), Gurugram, Haryana, India.
| |
Collapse
|
81
|
Razavi Z, Soltani M, Souri M, van Wijnen AJ. CRISPR innovations in tissue engineering and gene editing. Life Sci 2024; 358:123120. [PMID: 39426588 DOI: 10.1016/j.lfs.2024.123120] [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: 08/24/2024] [Revised: 09/22/2024] [Accepted: 10/05/2024] [Indexed: 10/21/2024]
Abstract
The CRISPR/Cas9 system is a powerful tool for genome editing, utilizing the Cas9 nuclease and programmable single guide RNA (sgRNA). However, the Cas9 nuclease activity can be disabled by mutation, resulting in catalytically deactivated Cas9 (dCas9). By combining the customizable sgRNA with dCas9, researchers can inhibit specific gene expression (CRISPR interference, CRISPRi) or activate the expression of a target gene (CRISPR activation, CRISPRa). In this review, we present the principles and recent advancements of these CRISPR technologies, as well as their delivery vectors. We also explore their applications in stem cell engineering and regenerative medicine, with a focus on in vitro stem cell fate manipulation and in vivo treatments. These include the prevention of retinal and muscular degeneration, neural regeneration, bone regeneration, cartilage tissue engineering, and the treatment of blood, skin, and liver diseases. Furthermore, we discuss the challenges of translating CRISPR technologies into regenerative medicine and provide future perspectives. Overall, this review highlights the potential of CRISPR in advancing regenerative medicine and offers insights into its application in various areas of research and therapy.
Collapse
Affiliation(s)
- ZahraSadat Razavi
- Physiology Research Center, Iran University Medical Sciences, Tehran, Iran; Biochemistry Research Center, Iran University Medical Sciences, Tehran, Iran
| | - Madjid Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran; Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Canada; Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, Canada; Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, Canada; Centre for Sustainable Business, International Business University, Toronto, Canada.
| | - Mohammad Souri
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Andre J van Wijnen
- Department of Biochemistry, University of Vermont, Burlington, VT, USA; Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
82
|
El Azzouzi M, El Ahanidi H, Hassan I, Tetou M, Ameur A, Bensaid M, Al Bouzidi A, Oukabli M, Alaoui CH, Addoum B, Chaoui I, Benbacer L, Mzibri ME, Attaleb M. Comprehensive behavioural assessment of TERT in bladder cancer. Urol Oncol 2024; 42:451.e19-451.e29. [PMID: 39147693 DOI: 10.1016/j.urolonc.2024.06.024] [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: 06/20/2024] [Accepted: 06/27/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Telomerase activity plays a crucial role in cancer development and progression. Thus, telomerase activation through the interplay of mutations and epigenetic alterations in the telomerase reverse transcriptase (TERT) promoter may provide further insight into bladder cancer induction and progression. METHODS In this study 100 bladder tumour tissues were selected, and four molecular signatures were analysed: THOR methylation status, TERT promotor mutation, telomere length, and TERT expression. RESULTS In our study, 88% of bladder cancer patients had an hypermethylation of the THOR region and 60% had mutations in the TERT promoter region. TERT promoter methylation was observed in all stages and grades of bladder cancer. While, TERT promoter mutations were detected in advanced stages and grades. In our cohort, high levels of TERT expression and long telomeres have been found in noninvasive cases of bladder cancer, with a significant association between TERT expression and Telomere length. Interestingly, patients with low TERT expression and cases with long telomeres had significantly longer Disease-free survival and overall survival. CONCLUSION The methylation and mutations occurring in the TERT promoter are implicated in bladder carcinogenesis, offering added prognostic and supplying novel insight into telomere biology in cancer.
Collapse
Affiliation(s)
- Meryem El Azzouzi
- Biology and Medical Research Unit, CNESTEN, Rabat, Morocco; Rabat Medical and Pharmacy School, Mohammed V University in Rabat, Rabat, Morocco
| | - Hajar El Ahanidi
- Biology and Medical Research Unit, CNESTEN, Rabat, Morocco; Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland; Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
| | - Ilias Hassan
- Rabat Medical and Pharmacy School, Mohammed V University in Rabat, Rabat, Morocco; Department of Urology, Military Hospital Mohammed V, Rabat, Morocco
| | - Mohammed Tetou
- Rabat Medical and Pharmacy School, Mohammed V University in Rabat, Rabat, Morocco; Department of Urology, Military Hospital Mohammed V, Rabat, Morocco
| | - Ahmed Ameur
- Rabat Medical and Pharmacy School, Mohammed V University in Rabat, Rabat, Morocco; Department of Urology, Military Hospital Mohammed V, Rabat, Morocco
| | - Mounia Bensaid
- Laboratory of Pathological Anatomy, Military Hospital Mohamed V, Rabat, Morocco; Royal School of Military Health Service, Rabat, Morocco
| | | | - Mohamed Oukabli
- Rabat Medical and Pharmacy School, Mohammed V University in Rabat, Rabat, Morocco; Laboratory of Pathological Anatomy, Military Hospital Mohamed V, Rabat, Morocco
| | - Chaimae Hafidi Alaoui
- Biology and Medical Research Unit, CNESTEN, Rabat, Morocco; Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco
| | | | - Imane Chaoui
- Biology and Medical Research Unit, CNESTEN, Rabat, Morocco
| | - Laila Benbacer
- Biology and Medical Research Unit, CNESTEN, Rabat, Morocco
| | | | | |
Collapse
|
83
|
Catalano M, Fancelli S, Caliman E, Mazzoni F, Michelet MG, Mancini S, Manneschi C, Shabani S, Napolitano B, Pillozzi S, Antonuzzo L, Roviello G. Impact of natremia on metastatic non small cell lung cancer patients receiving immune checkpoint inhibitors. Sci Rep 2024; 14:29655. [PMID: 39609621 PMCID: PMC11604652 DOI: 10.1038/s41598-024-81458-z] [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: 06/26/2024] [Accepted: 11/26/2024] [Indexed: 11/30/2024] Open
Abstract
Hyponatremia has been established as a prognostic indicator of survival in metastatic non-small cell lung cancer (mNSCLC). Conversely, the influence of normal sodium levels remains unexplored. This study aims to investigate the impact of natremia in mNSCLC patients undergoing treatment with immune checkpoint inhibitors (ICIs). Clinical and biochemical data of patients treated with ICIs for mNSCLC were obtained. Availability of baseline sodium values was a study inclusion criterion. Patients were categorized into two groups based on the cut off sodium value, determined using the receiver operating characteristic curve. Subsequently, the influence of sodium levels on response rate (RR), disease control rate (DCR), progression-free survival (PFS), and overall survival (OS) was analyzed. PFS and OS were assessed via the Kaplan-Meier method. Univariate and multivariate Cox regression analyses were conducted to evaluate prognostic factors for PFS and OS. The analysis included 88 patients, of whom 73.1% were men, with a median age of 71 years (range, 47-91). A comparison between patients with baseline natremia ≥ 140 mEq/L (n = 43) and those with < 140 mEq/L (n = 45) revealed PFS durations of 7.0 vs. 2.1 months (p < .01) and OS durations of 15.6 vs. 6.8 months, respectively (p = .02). In the univariate survival analysis, pre-ICI serum sodium ≥ 140 mEq/L (p = .01) was associated with improved PFS, while factors associated with OS included brain metastasis (p = .05) and pre-ICI serum sodium ≥ 140 mEq/L (p = .02). In the multivariate analysis, pre-ICI serum sodium ≥ 140 mEq/L maintained a statistically significant association with OS (p = .04)..This study represents the first investigation into the impact of normonatremia in mNSCLC. Our findings suggest that serum sodium levels < 140 mEq/L at baseline and initial assessment are independently associated with poorer PFS and OS in mNSCLC patients undergoing first-line treatment with ICIs.
Collapse
Affiliation(s)
- Martina Catalano
- Department of Health Science, University of Florence, Florence, 50134, Italy
| | - Sara Fancelli
- Clinical Oncology Unit, Careggi University Hospital, Florence, 50234, Italy
| | - Enrico Caliman
- Clinical Oncology Unit, Careggi University Hospital, Florence, 50234, Italy
| | - Francesca Mazzoni
- Clinical Oncology Unit, Careggi University Hospital, Florence, 50234, Italy
| | - Marta Gatta Michelet
- Department of Experimental and Clinical Medicine, University of Florence, Florence, 50134, Italy
| | - Silvia Mancini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, 50134, Italy
| | - Clara Manneschi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, 50134, Italy
| | - Sonia Shabani
- Department of Experimental and Clinical Medicine, University of Florence, Florence, 50134, Italy
| | - Brunella Napolitano
- Department of Experimental and Clinical Medicine, University of Florence, Florence, 50134, Italy
| | - Serena Pillozzi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Lorenzo Antonuzzo
- Clinical Oncology Unit, Careggi University Hospital, Florence, 50234, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence, 50134, Italy
| | | |
Collapse
|
84
|
Woods K, Rants'o TA, Chan AM, Sapre T, Mastin GE, Maguire KM, Ong SE, Golkowski M. diaPASEF-Powered Chemoproteomics Enables Deep Kinome Interaction Profiling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624841. [PMID: 39605566 PMCID: PMC11601655 DOI: 10.1101/2024.11.22.624841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Protein-protein interactions (PPIs) underlie most biological functions. Devastating human conditions like cancers, neurological disorders, and infections, hijack PPI networks to initiate disease, and to drive disease progression. Understanding precisely how diseases remodel PPI networks can, therefore, help clarify disease mechanisms and identify therapeutic targets. Protein kinases control most cellular processes through protein phosphorylation. The 518 human kinases, known as the kinome, are frequently dysregulated in disease and highly druggable with ATP-competitive inhibitors. Kinase activity, localization, and substrate recognition are regulated by dynamic PPI networks composed of scaffolding and adapter proteins, other signaling enzymes like small GTPases and E3 ligases, and phospho-substrates. Accordingly, mapping kinase PPI networks can help determine kinome activation states, and, in turn, cellular activation states; this information can be used for studying kinase-mediated cell signaling, and for prioritizing kinases for drug discovery. Previously, we have developed a high-throughput method for kinome PPI mapping based on mass spectrometry (MS)-based chemoproteomics that we named kinobead competition and correlation analysis (kiCCA). Here, we introduce 2 nd generation (gen) kiCCA which utilizes data-independent acquisition (dia) with parallel accumulation serial fragmentation (PASEF) MS and a re-designed CCA algorithm with improved selection criteria and the ability to predict multiple kinase interaction partners of the same proteins. Using neuroblastoma cell line models of the noradrenergic-mesenchymal transition (NMT), we demonstrate that 2 nd gen kiCCA (1) identified 6.1-times more kinase PPIs in native cell extracts compared to our 1 st gen approach, (2) determined kinase-mediated signaling pathways that underly the neuroblastoma NMT, and (3) accurately predicted pharmacological targets for manipulating NMT states. Our 2 nd gen kiCCA method is broadly useful for cell signaling research and kinase drug discovery.
Collapse
|
85
|
Okuyan D. Epidermal Growth Factor Downregulates Carbon Anhydrase III (CAIII) in Colon Cancer. Curr Issues Mol Biol 2024; 46:12994-13002. [PMID: 39590368 PMCID: PMC11593170 DOI: 10.3390/cimb46110774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer-related death in the world. Dysregulations in the EGF signaling pathway have been associated with colon cancer. Some members of the carbonic anhydrase family serve as biomarkers in cancer. Carbonic anhydrase III (CAIII), a member of this family, shows different activities than the other members of its family and has been associated with cancer. However, there are no studies on the effective regulation of EGF. In this study, we investigated the EGF-influenced regulation of CAIII in the HT29, SW480, and HUVEC cell lines and showed that CAIII regulation decreased with the effect of EGF. We aimed to investigate the EGF-affected mRNA and protein regulation of the CAIII gene in HT29, SW480, and HUVEC cell lines. For this purpose, we determined time-dependent CAIII mRNA and protein expression by applying EGF to HT29, SW480, and HUVEC cells. Time-dependent EGF-induced mRNA and protein level regulation of the CAIII gene decreased in the HT29, SW480, and HUVEC cell lines. EGF regulates the motility, adhesion, and metastasis of cancer cells. CAIII prevents cells from metastasizing through cell acidification. Therefore, our findings explained why the EGF-effective regulation of CAIII decreased. We suggest that the CAIII gene is promising as a targeted therapy due to the decrease in EGF-effected CAIII gene regulation in colon carcinoma.
Collapse
Affiliation(s)
- Derya Okuyan
- Department of Veterinary Medicine, Susurluk Agriculture and Forestry Vocational School, Bandırma Onyedi Eylül University, Susurluk 10600, Balıkesir, Türkiye
| |
Collapse
|
86
|
Li B, Rong D, Lin H. Atezolizumab monotherapy as first-line treatment for non-small cell lung cancer ineligible for treatment with a platinum-containing regimen (IPSOS): a cost-effectiveness analysis in the USA. BMJ Open 2024; 14:e083716. [PMID: 39532375 PMCID: PMC11574426 DOI: 10.1136/bmjopen-2023-083716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVE This study explores the cost-effectiveness of atezolizumab monotherapy compared with chemotherapy as first-line treatment for stage IIIB or IV non-small cell lung cancer (IIIB/IV-NSCLC) ineligible for platinum-based chemotherapy from a US payer perspective. DESIGN This is based on the IPSOS clinical trial. We conducted a comprehensive assessment of the cost-effectiveness of atezolizumab monotherapy versus single-agent chemotherapy over a 15-year duration. Employing a robust Markov model incorporating data from 453 patients, we calculated total costs, life-years (LYs), quality-adjusted life-years (QALYs) and the incremental cost-effectiveness ratio (ICER) at a willingness-to-pay (WTP) threshold of $150 000 per QALY. We performed one-way, two-way and probabilistic sensitivity analyses to validate our model. SETTING The US payer perspective. PARTICIPANTS A cohort with NSCLC ineligible for treatment with a platinum-containing regimen from IPSOS clinical trial. INTERVENTIONS Atezolizumab monotherapy versus chemotherapy. PRIMARY OUTCOME MEASURE Cost, QALYs, LYs and ICER. RESULT Chemotherapy resulted in an average survival of 0.930 QALYs (1.528 LYs) per patient at an average cost of $67 579. Atezolizumab treatment provided an additional 0.309 QALYs but incurred an extra cost of $66 472, leading to an ICER of $215 069 per QALY compared with chemotherapy. The cost of atezolizumab had the most significant impact on the model outcomes. Probabilistic sensitivity analysis showed that atezolizumab had a 30.2% probability of being considered cost-effective at a WTP threshold of $150 000 per QALY in the USA. These results remained consistent across various scenarios and sensitivity analyses employing both deterministic and probabilistic approaches. CONCLUSION The current price of atezolizumab renders it an unlikely cost-effective treatment option for patients with IIIB/IV-NSCLC from the payer's perspective in the USA. To achieve cost-effectiveness, substantial discounts are necessary. TRIAL REGISTRATION NUMBER The IMpower-110, an open-label, randomised, phase 3 clinical trial (NCT02409342). The IPSOS clinical trial (NCT03191786).
Collapse
Affiliation(s)
- Biao Li
- Department of Radiotherapy, Guangdong Provincial People's Hospital Affiliated to Southern Medical University, Guangzhou, Guangdong, China
| | - Dingchao Rong
- Department of Orthopedics, The First Affiliated Hospital of Shaoyang University, Shaoyang, Hunan, China
| | - Hui Lin
- Department of Radiotherapy, Guangdong Provincial People's Hospital Affiliated to Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
87
|
Chen HH, Luo CW, Chen YL, Chiang JY, Huang CR, Wang YT, Chen CH, Guo J, Yip HK. Probiotic-facilitated cytokine-induced killer cells suppress peritoneal carcinomatosis and liver metastasis in colorectal cancer cells. Int J Biol Sci 2024; 20:6162-6180. [PMID: 39664585 PMCID: PMC11628340 DOI: 10.7150/ijbs.101051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/19/2024] [Indexed: 12/13/2024] Open
Abstract
Background: This study tested the hypothesis that combined therapy with probiotics and cytokine-induced killer (CIK) cells was superior to merely one on suppressing the peritoneal carcinomatosis and liver metastasis of colorectal cancer (CRC) cells in nude mice. Methods and Results: The in vitro study revealed that in HCT 116/SW620 CRC cell lines, cell viability, proliferation, colony formation, migratory ability, wound healing, and protein expression of PD-L1 and FAK were significantly and comparably suppressed and that apoptosis was significantly and comparably increased by probiotics and CIK cells, and these effects were further significantly enhanced by combined probiotics + CIK cell therapy (all p<0.001). Nude mice were categorized into Groups 1 (SC), 2 (HCT 116), 3 (HCT 116 + probiotics), 4 (HCT 116 + CIK cells), and 5 (HCT 116 + probiotics + CIK cells). CRC cells were intraperitoneally implanted into Groups 2 to 5, and the animals were euthanized by Day 28. The results demonstrated that the abdominal dissemination of CRC cells, tumor numbers, tumor weights, liver weights, liver necrosis areas and the expression of γ-H2AX/PD-L1/FAK in harvested liver tumors were lowest in Group 1, highest in Group 2, and significantly and progressively decreased in Groups 3 to 5 (all p<0.0001). The protein expression levels of apoptotic and DNA damage biomarkers (Bax/c-caspase 3/c-PARP/γ-H2AX), a metastatic biomarker (FAK) and three tumor proliferation and survival signaling biomarkers (JAK-STAT1, PI3K/Akt/m-TOR and Ras/Raf/MEK/ERK) exhibited identical patterns to that of a tumor immune escape biomarker (PD-L1) among the groups (all p<0.0001). Conclusion: The combination of probiotics and CIK cells was superior to either therapy alone in suppressing CRC cell growth, proliferation, liver metastasis and survival, mainly through downregulating cell proliferation and survival signaling pathways.
Collapse
Affiliation(s)
- Hong-Hwa Chen
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Chi-Wen Luo
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung City 807, Taiwan
- Department of Cosmetic Science and Institute of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan
| | - Yi-Ling Chen
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - John Y. Chiang
- Department of Computer Science and Engineering, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
| | - Chi-Ruei Huang
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital Kaohsiung 833401, Taiwan
| | - Yi-Ting Wang
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Chih-Hung Chen
- Divisions of General Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Jun Guo
- Department of Cardiology, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
| | - Hon-Kan Yip
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital Kaohsiung 833401, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
88
|
Li W, Liu Y. Development and validation of a risk predictive nomogram for colon cancer-specific mortality: a competing risk model based on the SEER database. Discov Oncol 2024; 15:621. [PMID: 39503842 PMCID: PMC11541964 DOI: 10.1007/s12672-024-01498-9] [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: 09/12/2024] [Accepted: 11/01/2024] [Indexed: 11/09/2024] Open
Abstract
BACKGROUND Utilizing the SEER database, we developed a competing risk model along with a nomogram designed for the early identification of colon cancer-specific mortality (CSM) risk. METHODS Clinical and pathological information, along with other significant data, were obtained from the SEER database. Patients were randomly divided into a training set and a validation set. We investigated the independent factors affecting CSM among colon cancer patients using univariate and multivariate analyses within a competing risk framework, ultimately developing a predictive tool for CSM in colon cancer. RESULTS Involving 40,261 individuals diagnosed with colon cancer, our study included 10,397 deaths directly due to the disease and an additional 5,828 from other causes. We used a competing risk model to predict cancer-specific mortality (CSM) in these patients. For the training dataset, the model's area under the curve (AUC) for predicting 1-, 3-, and 5-year cancer-specific survival (CSS) was 0.835 (95% confidence interval [CI] 0.826 to 0.844), 0.849 (95% CI 0.843 to 0.855), and 0.843 (95% CI 0.836 to 0.850), respectively. In the validation group, the AUC values for the same time periods were 0.846 (95% CI 0.833 to 0.860), 0.853 (95% CI 0.843 to 0.862), and 0.846 (95% CI 0.835 to 0.856), respectively. In comparison, traditional survival analysis yielded higher cumulative CSM rates over time than those provided by our competing risk approach. CONCLUSION We created a competitive risk assessment model along with a predictive tool designed to estimate CSM in patients with colon cancer. This nomogram demonstrates high accuracy and reliability, aiding medical professionals in making clinical decisions and developing patient follow-up plans.
Collapse
Affiliation(s)
- Wei Li
- Department of Oncology, Shuyang Hospital, The Affiliated Shuyang Hospital of Xuzhou Medical University, No. 9 Yingbin Avenue, Shucheng Town, Shuyang County, Suqian, 223600, Jiangsu, China.
| | - Yiting Liu
- Department of Oncology, Shuyang Hospital, The Affiliated Shuyang Hospital of Xuzhou Medical University, No. 9 Yingbin Avenue, Shucheng Town, Shuyang County, Suqian, 223600, Jiangsu, China
| |
Collapse
|
89
|
Kramer A, Rubio‐Alarcón C, van den Broek D, Vessies DCL, van't Erve I, Meijer GA, Vink GR, Schuuring E, Fijneman RJA, Coupé VMH, Retèl VP. A scenario-drafting study to explore potential future implementation pathways of circulating tumor DNA testing in oncology. Mol Oncol 2024; 18:2730-2742. [PMID: 38060377 PMCID: PMC11547223 DOI: 10.1002/1878-0261.13562] [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: 07/07/2023] [Revised: 11/17/2023] [Accepted: 12/05/2023] [Indexed: 01/13/2024] Open
Abstract
Circulating tumor DNA (ctDNA) detection has multiple promising applications in oncology, but the road toward implementation in clinical practice is unclear. We aimed to support the implementation process by exploring potential future pathways of ctDNA testing. To do so, we studied four ctDNA-testing applications in two cancer types and elicited opinions from 30 ctDNA experts in the Netherlands. Our results showed that the current available evidence differed per application and cancer type. Tumor profiling and monitoring treatment response were found most likely to be implemented in non-small cell lung cancer (NSCLC) within 5 years. For colorectal cancer, applications of ctDNA testing were found to be at an early stage in the implementation process. Demonstrating clinical utility was found a key aspect for successful implementation, but there was no consensus regarding the evidence requirements. The next step toward implementation is to define how clinical utility of biomarkers should be evaluated. Finally, these data indicate that specific challenges for each clinical application and tumor type should be appropriately addressed in a deliberative process involving all stakeholders to ensure implementation of ctDNA testing and timely access for patients.
Collapse
Affiliation(s)
- Astrid Kramer
- Department of Epidemiology and Data ScienceAmsterdam UMCThe Netherlands
| | | | - Daan van den Broek
- Department of Laboratory MedicineNetherlands Cancer InstituteAmsterdamThe Netherlands
| | - Daan C. L. Vessies
- Department of Laboratory MedicineNetherlands Cancer InstituteAmsterdamThe Netherlands
| | - Iris van't Erve
- Department of PathologyNetherlands Cancer InstituteAmsterdamThe Netherlands
| | - Gerrit A. Meijer
- Department of PathologyNetherlands Cancer InstituteAmsterdamThe Netherlands
| | - Geraldine R. Vink
- Department of Medical Oncology, University Medical Center UtrechtUniversity of UtrechtThe Netherlands
- Department of Research and DevelopmentIKNLUtrechtThe Netherlands
| | - Ed Schuuring
- Department of Pathology and Medical BiologyUniversity Medical Center GroningenThe Netherlands
| | | | | | - Valesca P. Retèl
- Department of Psychosocial Research and EpidemiologyNetherlands Cancer InstituteAmsterdamThe Netherlands
- Erasmus School of Health Policy and ManagementErasmus University RotterdamThe Netherlands
| |
Collapse
|
90
|
Ma S, Wang X, Yan S, Miao L, Wan X, Ding D, Yu D, Diao X, Wang X, Zhang H. Pharmacokinetics, mass balance, and metabolism of [ 14C]envonalkib (TQ-B3139), a novel ALK tyrosine kinase inhibitor, in healthy Chinese subjects. Cancer Chemother Pharmacol 2024; 94:647-657. [PMID: 38507062 DOI: 10.1007/s00280-024-04647-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 01/30/2024] [Indexed: 03/22/2024]
Abstract
PURPOSE Envonalkib (TQ-B3139) is a novel, potent anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitor used to treat ALK-positive non-small cell lung cancer. This phase I mass balance study investigated the pharmacokinetics, metabolism, and excretion of 14C-radiolabeled envonalkib in healthy Chinese male subjects. METHODS A single oral dose of 600 mg (150 µCi) [14C]envonalkib was administered to healthy male subjects under fasted state. Samples of blood, urine and feces were collected for quantitative determination of total radioactivity and unchanged envonalkib, and the metabolites identification. RESULTS After dosing, the median Tmax of radioactivity was 4 h and the mean t1/2 was 65.2 h in plasma. The exposure of total radioactivity was much higher than that of unchanged envonalkib in plasma. The mean total recovery of the radiolabeled dose was 93.93% over 504 h post-dose, with 15.23% in urine and 78.71% in feces. Envonalkib underwent extensive metabolism and a total of 15 metabolites were identified in plasma, urine, and feces. Unchanged envonalkib and its major metabolite M315 were the main components in plasma, accounting for 20.37% and 33.33% of total plasma radioactivity. In urine, O-dealkylation metabolite M315 was the major component accounted for 7.98% of dose. In feces, 16.01% of dose was excreted as cysteine conjugate M434-1. Envonalkib was well tolerated and there were no serious adverse events observed in the study. CONCLUSION Envonalkib was extensively metabolized prior to excretion and eliminated primarily as metabolites via feces.
Collapse
Affiliation(s)
- Sheng Ma
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Xin Wang
- Department of Clinical Research Center, Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Nanjing, China
| | - Shu Yan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Liyan Miao
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Xiaojing Wan
- Department of Clinical Research Center, Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Nanjing, China
| | - Dawei Ding
- Department of Clinical Research Center, Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Nanjing, China
| | - Ding Yu
- Department of Clinical Research Center, Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Nanjing, China
| | - Xingxing Diao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xunqiang Wang
- Department of Clinical Research Center, Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Nanjing, China.
| | - Hua Zhang
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China.
- Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China.
| |
Collapse
|
91
|
Hinojosa-Gonzalez DE, Saffati G, Salgado-Garza G, Patel S, Kronstedt S, Jones JA, Taylor JM, Yen AE, Slawin JR. Novel therapeutic regimens in previously untreated metastatic urothelial carcinoma: A systematic review and bayesian network meta-analysis. Urol Oncol 2024; 42:361-369. [PMID: 39112104 DOI: 10.1016/j.urolonc.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/18/2024] [Accepted: 07/04/2024] [Indexed: 09/07/2024]
Abstract
Metastatic urothelial carcinoma (muC) has historically had few effective therapeutic options. Recently, immune checkpoint inhibitors (ICIs), were introduced as therapeutic options for cisplatin-ineligible patients, however, direct head-to-head trials comparing these treatments are lacking. To address this gap, this study employs a Bayesian framework to indirectly compare the performance of ICIs as first-line agents for muC. A systematic review was performed to identify randomized controlled trials evaluating different ICI for mUC. Data was inputted into Review Manager 5.4 for pairwise meta-analysis. Data was then used to build a network in R Studio. These networks were used to model 200,000 Markov Chains via MonteCarlo sampling. The results are expressed as hazard ratios (HR) with 95% credible intervals (CrI). Six studies with 5,449 patients were included, 3,255 received ICI monotherapy or combination. Moreover, a total of 3,006 had PD-L1 positive tumors and 2,362 were PD-L1 negative. Median overall survival (OS) ranged from 12.1 to 31.5 months across the studies, with the combination of enfortumab vedotin and pembrolizumab demonstrating the most substantial reduction in the risk of death (HR 0.47 [95% CrI: 0.38, 0.58]), followed by avelumab monotherapy (HR 0.69 [95% CrI: 0.56, 0.86]). The limitations of this network meta-analysis include variability in study follow-up duration, lack of standardized methods for assessing PD-L1 positivity, and potential bias introduced by control arms with poorer survival outcomes across included trials. The enfortumab vedotin/pembrolizumab combination significantly improved survival and response rates. Avelumab showed notable single-agent activity. These findings provide a valuable framework to guide clinical decision-making and highlight priority areas for future research, including biomarker refinement and novel combination strategies to enhance antitumor immunity in this challenging malignancy.
Collapse
Affiliation(s)
| | - Gal Saffati
- Scott Department of Urology, Baylor College of Medicine, Houston, TX
| | | | - Sagar Patel
- Scott Department of Urology, Baylor College of Medicine, Houston, TX
| | - Shane Kronstedt
- Scott Department of Urology, Baylor College of Medicine, Houston, TX
| | - Jeffrey A Jones
- Scott Department of Urology, Baylor College of Medicine, Houston, TX; Michael E. DeBakey VA Medical Center, Houston, TX
| | - Jennifer M Taylor
- Scott Department of Urology, Baylor College of Medicine, Houston, TX; Michael E. DeBakey VA Medical Center, Houston, TX
| | - Aihua E Yen
- Bladder Cancer Center, Daniel L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| | - Jeremy R Slawin
- Scott Department of Urology, Baylor College of Medicine, Houston, TX; Michael E. DeBakey VA Medical Center, Houston, TX
| |
Collapse
|
92
|
Wang K, Leyba A, Hsu R. Addressing the unmet need in NSCLC progression with advances in second-line therapeutics. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:1297-1320. [PMID: 39759220 PMCID: PMC11700623 DOI: 10.37349/etat.2024.00277] [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: 07/31/2024] [Accepted: 10/16/2024] [Indexed: 01/07/2025] Open
Abstract
Lung cancer is the leading cause of cancer mortality globally, with non-small cell lung cancer (NSCLC) accounting for 85% of cases. Despite advancements in first-line treatments such as immunotherapy and targeted therapies, resistance to these treatments is common, creating a significant unmet need for effective second-line therapies. This review evaluates current and emerging second-line therapeutic options for advanced or metastatic NSCLC, focusing on their efficacy and potential to improve patient outcomes. Anti-angiogenic drugs like ramucirumab combined with chemotherapy, particularly docetaxel, have shown moderate success. Antibody-drug conjugates (ADCs) targeting specific tumor antigens offer a promising avenue for targeted therapy, while chimeric antigen receptor (CAR)-T cell therapy and T-cell receptor therapy leverage the patient's immune system to combat cancer more effectively. mRNA vaccines, although in early stages, show potential for inducing robust immune responses against cancer-specific antigens. Building on this foundation, recent advancements in molecular testing and the exploration of the tumor microenvironment are opening new therapeutic avenues, further enhancing the potential for personalized second-line treatments in NSCLC. While ADCs and bispecific antibodies are gaining traction, more precise biomarkers are needed to optimize treatment response. Regular monitoring through techniques like liquid biopsies allows real-time tracking of mutations such as EGFR T790M, enabling timely therapeutic adjustments. Additionally, the role of neutrophils and macrophages in the tumor microenvironment is increasingly being recognized as a potential therapeutic avenue, with Smad3 emerging as a key target. Further research into drug sequencing, toxicity management, and biomarker development remains crucial to improving NSCLC treatment outcomes.
Collapse
Affiliation(s)
- Kinsley Wang
- Department of Medicine, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Alexis Leyba
- Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Robert Hsu
- Department of Medicine, Division of Medical Oncology, University of Southern California Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| |
Collapse
|
93
|
Ozbay MF, Cetinkaya AM, Balcik OY, Ilhan Y, Genc TB, Goksu SS. The ascendancy of eosinophil counts in non-small cell lung cancer: a potential marker for predicting response and survival under nivolumab treatment. Am J Cancer Res 2024; 14:5095-5104. [PMID: 39553208 PMCID: PMC11560823 DOI: 10.62347/krth2276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 10/04/2024] [Indexed: 11/19/2024] Open
Abstract
Lung cancer is the leading cause of cancer-related death globally and is often diagnosed at an advanced stage. Nivolumab represents a significant advancement for treating advanced non-small cell lung cancer (NSCLC). However, the absence of reliable biomarkers predicting treatment response hinders personalized therapy. Eosinophils play a notable role in cancer biology, particularly when treated with immune checkpoint inhibitors. Eosinophils can infiltrate tumor tissues, directly interacting with tumor cells or modifying the tumor microenvironment. This study aims to assess the potential of PD-L1 expression and peripheral blood eosinophil count in predicting treatment response and patient survival. This retrospective cohort study was conducted in three major cancer centers in Turkey, including 174 advanced NSCLC patients who had progressed after chemotherapy between July 2019 and November 2023. Demographic and clinical data, PD-L1 levels, and eosinophil counts were analyzed using SPSS 27.0. Survival analyses were performed with Kaplan-Meier and Cox regression models. Increased peripheral blood eosinophil count was positively associated with response to Nivolumab treatment and overall survival. Among treatment responders, 54.1% had eosinophil levels between 100-499 cells/mm3 before treatment, increasing to 70.8% post-treatment. In patients with high PD-L1 positivity (>50%), eosinophil levels averaged 266.0 cells/mm3, with improved survival outcomes (mean survival: 24.06 months, median: 20.0 months). Non-responders had a mean survival of 19.05 months and a median survival of 15.2 months. Peripheral eosinophil count appears to be a potential biomarker for predicting response to Nivolumab treatment and survival in NSCLC patients. Combined evaluation of eosinophil count and PD-L1 expression may enhance personalized treatment strategies. Further validation in prospective, randomized studies is necessary.
Collapse
Affiliation(s)
- Mehmet Fatih Ozbay
- Department of Medical Oncology, Kirsehir Training and Research HospitalKirsehir 40100, Turkey
| | - Aysegul Merc Cetinkaya
- Department of Medical Oncology, Faculty of Medicine, Akdeniz UniversityAntalya 07000, Turkiye
| | - Onur Yazdan Balcik
- Alanya Alaattin Keykubat University, Department of Medical OncologyAntalya, Türkiye
| | - Yusuf Ilhan
- Antalya City Hospital, Department of Medical OncologyAntalya, Türkiye
| | | | - Sema Sezgin Goksu
- Department of Medical Oncology, Faculty of Medicine, Akdeniz UniversityAntalya 07000, Turkiye
| |
Collapse
|
94
|
Huang J, Liao Z, Hu Y, Cheng Y, Zhong J. Prognostic factors of cutaneous soft tissue sarcomas in children: a SEER population-based study. Arch Dermatol Res 2024; 316:700. [PMID: 39424676 DOI: 10.1007/s00403-024-03450-x] [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: 08/07/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Abstract
This study aims to analyze the clinicopathological characteristics and survival outcomes of cutaneous soft tissue sarcomas (CSTS) in children. We selected pediatric cases of CSTS diagnosed between 2000 and 2019 from the Surveillance, Epidemiology, and End Results (SEER) database. Survival rates were calculated using Kaplan-Meier methods. We performed univariate analyses with the log-rank test and multivariate survival analyses using Cox proportional-hazards models to determine factors affecting overall survival (OS). Additionally, we constructed a predictive nomogram based on the outcomes of the Cox regression. A total of 148 pediatric patients with CSTS were reviewed. The median age at diagnosis was 13 years (range: 0-18 years). Prognostically, tumors located on the extremities showed better outcomes compared to those on the head, neck, or trunk. Among the histological types, angiosarcoma had the lowest five-year survival rate at 51.3%, which was substantially lower compared to fibrous histiocytoma and leiomyosarcoma. Cox regression analysis highlighted surgical intervention as the only significant independent prognostic factor for OS, with an increased risk of mortality observed in patients not undergoing surgery. Additionally, patients with distant-stage disease exhibited significantly lower survival rates than those with localized conditions. Pediatric CSTS represents a diverse and infrequent group of tumors, predominantly fibrous histiocytoma and leiomyosarcoma. Surgery was identified as the crucial determinant of survival, underscoring its role in effectively managing these patients.
Collapse
Affiliation(s)
- Jian Huang
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, Anhui, 230601, China
| | - Zhenqi Liao
- Department of Cardiology, Ganxian Renmin Hospital, Ganzhou, Jiangxi, China
| | - Yilan Hu
- Department of Rehabilitation, Hangzhou First People's Hospita Xiasha Campus, Hangzhou Rehabilitation Hospital, Hangzhou, Zhejiang, China
| | - Yan Cheng
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, Anhui, 230601, China.
| | - Jiawang Zhong
- Department of Stomatology, Anji County People's Hospital, Huzhou, Zhejiang, 313300, China.
| |
Collapse
|
95
|
Tirone B, Scarabosio A, Surico PL, Parodi PC, D’Esposito F, Avitabile A, Foti C, Gagliano C, Zeppieri M. Targeted Drug Delivery in Periorbital Non-Melanocytic Skin Malignancies. Bioengineering (Basel) 2024; 11:1029. [PMID: 39451404 PMCID: PMC11504966 DOI: 10.3390/bioengineering11101029] [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: 09/16/2024] [Revised: 10/05/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024] Open
Abstract
Targeted drug delivery has emerged as a transformative approach in the treatment of periorbital skin malignancies, offering the potential for enhanced efficacy and reduced side effects compared to traditional therapies. This review provides a comprehensive overview of targeted therapies in the context of periorbital malignancies, including basal cell carcinoma, squamous cell carcinoma, sebaceous gland carcinoma, and Merkel cell carcinoma. It explores the mechanisms of action for various targeted therapies, such as monoclonal antibodies, small molecule inhibitors, and immunotherapies, and their applications in treating these malignancies. Additionally, this review addresses the management of ocular and periocular side effects associated with these therapies, emphasizing the importance of a multidisciplinary approach to minimize impact and ensure patient adherence. By integrating current findings and discussing emerging trends, this review aims to highlight the advancements in targeted drug delivery and its potential to improve treatment outcomes and quality of life for patients with periorbital skin malignancies.
Collapse
Affiliation(s)
- Benedetta Tirone
- Dermatology and Venerology Section, Department of Precision and Regenerative Medicine and Ionan Area (DiMePRe-J), University of Bari Aldo Moro, 70124 Bari, Italy
| | - Anna Scarabosio
- Clinic of Plastic and Reconstructive Surgery, Ospedale Santa Maria della Misericordia, 33100 Udine, Italy
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Pier Luigi Surico
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
- Department of Ophthalmology, Campus Bio-Medico University, 00128 Rome, Italy
| | - Pier Camillo Parodi
- Clinic of Plastic and Reconstructive Surgery, Ospedale Santa Maria della Misericordia, 33100 Udine, Italy
| | - Fabiana D’Esposito
- Imperial College Ophthalmic Research Group (ICORG) Unit, Imperial College, 153-173 Marylebone Rd, London NW15QH, UK
- Department of Neurosciences, Reproductive Sciences and Dentistry, University of Naples Federico II, Via Pansini 5, 80131 Napoli, Italy
| | - Alessandro Avitabile
- Eye Clinic Catania San Marco Hospital, Viale Carlo Azeglio Ciampi, 95121 Catania, Italy
| | - Caterina Foti
- Dermatology and Venerology Section, Department of Precision and Regenerative Medicine and Ionan Area (DiMePRe-J), University of Bari Aldo Moro, 70124 Bari, Italy
| | - Caterina Gagliano
- Mediterranean Foundation “G.B. Morgagni”, 95125 Catania, Italy
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, p.le S. Maria della Misericordia 15, 33100 Udine, Italy
| |
Collapse
|
96
|
Gorzelak-Magiera A, Domagała-Haduch M, Kabut J, Gisterek-Grocholska I. The Use of Anaplastic Lymphoma Kinase Inhibitors in Non-Small-Cell Lung Cancer Treatment-Literature Review. Biomedicines 2024; 12:2308. [PMID: 39457620 PMCID: PMC11504905 DOI: 10.3390/biomedicines12102308] [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: 09/15/2024] [Revised: 10/01/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Lung cancer is the leading cause of cancer-related morbidity and mortality. The median survival time for patients with advanced non-small-cell lung cancer before the era of molecular-based personalized treatment was 7.9 months. The discovery of predictive factors and the introduction of molecular diagnostics into daily practice made a breakthrough, enabling several years of survival in patients with advanced disease. The discovery of rearrangements in the ALK gene and ALK tyrosine kinase inhibitors has resulted in a dramatic improvement in the prognosis of patients with this subtype of cancer. Currently, three generations of ALK inhibitors differing in activity, toxicity and degree of penetration into the central nervous system are available in clinical practice. The current state of knowledge on ALK inhibitors used in clinical practice is summarised in this research paper. Methods of diagnosis of abnormalities in ALK have been shown, and the review of research that contributed to the development of the next generation of ALK inhibitors has been presented.
Collapse
Affiliation(s)
- Anita Gorzelak-Magiera
- Department of Oncology and Radiotherapy, Medical University of Silesia, 40-615 Katowice, Poland; (M.D.-H.); (J.K.); (I.G.-G.)
| | | | | | | |
Collapse
|
97
|
Shen X, Zhou H, Zhou X, Liu Z, Meng X, Zhang L, Song Y, Guo R, Wang F, Li K, Li W, Yang Z, Liu Z, Li N. 68Ga-grazytracer PET for noninvasive assessment of response to immunotherapy in solid tumors and lymphomas: a phase 1/2 clinical trial. Nat Commun 2024; 15:8791. [PMID: 39389969 PMCID: PMC11467221 DOI: 10.1038/s41467-024-53197-2] [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: 07/02/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024] Open
Abstract
To tackle the clinical challenge of noninvasively assessing immunotherapy efficacy in patients, here we used positron emission tomography (PET) with 68Ga-grazytracer, which targets granzyme B, a crucial effector molecule secreted by activated CD8+ T cells. In this phase 1/2 clinical trial (NCT05000372) involving a diverse cohort of 24 patients with solid tumors and lymphomas who received immunotherapies, including immune checkpoint inhibitors (either alone or with chemotherapies) and chimeric antigen receptor-T cell therapy, we examined the in vivo behaviors of 68Ga-grazytracer. Primary endpoints were safety, biodistribution, granzyme B specificity, and the predictive utility of 68Ga-grazytracer, while secondary endpoint was the relationship between 68Ga-grazytracer uptake and tumor immune phenotype. 68Ga-grazytracer exhibited a safe profile and specifically targeted granzyme B in patients. 68Ga-grazytracer PET showed superior predictive value for short-term prognosis and progression-free survival than those of conventional assessment criteria, including RECIST 1.1 and PERCIST. Moreover, the uptake of 68Ga-grazytracer in tumors was significantly higher in those with a "non-desert" immune phenotype than those with an immune "desert" phenotype, thereby meeting the primary and secondary endpoints of this trial. Collectively, we successfully visualized CD8+ T cell effector function in humans using 68Ga-grazytracer PET, offering insights for enhancing immunotherapy assessment, patient stratification and treatment planning.
Collapse
Affiliation(s)
- Xiuling Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, 100142, Beijing, China
- Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Haoyi Zhou
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
| | - Xin Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, 100142, Beijing, China
- Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Zongchao Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Xiangxi Meng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, 100142, Beijing, China
- Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Linyu Zhang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
| | - Yufei Song
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, 100142, Beijing, China
- Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Rui Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, 100142, Beijing, China
- Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Fei Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, 100142, Beijing, China
- Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Kui Li
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
| | - Wenqing Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Zhi Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, 100142, Beijing, China.
- Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, 100142, Beijing, China.
| | - Zhaofei Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, 100142, Beijing, China.
- Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, 100142, Beijing, China.
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China.
- Department of Nuclear Medicine, Peking University Third Hospital, 100191, Beijing, China.
| | - Nan Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, 100142, Beijing, China.
- Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, 100142, Beijing, China.
| |
Collapse
|
98
|
Zagardo V, Martorana E, Harikar M, Pergolizzi S, Ferini G. Effectiveness of radiotherapy in delaying treatment changes in primary or secondary immunorefractory oligoprogressive patients: preliminary results from a single-center study. Discov Oncol 2024; 15:531. [PMID: 39377996 PMCID: PMC11461402 DOI: 10.1007/s12672-024-01360-y] [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: 04/06/2024] [Accepted: 09/17/2024] [Indexed: 10/11/2024] Open
Abstract
AIMS To investigate whether the addition of radiotherapy could be an appropriate option to delay the time-to-next systemic treatment (TTNsT) in patients with oligoprogressive solid tumors who had acquired or innate resistance to immune checkpoint inhibitors (ICIs). MATERIAL AND METHODS Patients with oligoprogressive disease treated with ICIs and radiotherapy at our Institute from January 2019 to June 2023 were retrospectively identified. Patients were stratified as primary or secondary immunorefractory according to the time of onset of ICI resistance. TTNsT and Time-To-Resistance (TTR) were the primary outcomes. Secondary outcomes included: post-radiotherapy first progression-free survival (pR-PFS), Local Control (LC), Overall Survival (OS), and treatment-related toxicities. In addition, out-of-field effects (such as the abscopal effect) of radiotherapy have been hypothesized. The survival rates were analyzed using the Kaplan-Meier method and long-rank test. RESULTS 40 out of 105 screened patients with oligoprogressive disease met the inclusion criteria. Of these, 28 had an acquired drug resistance while 12 had an innate drug resistance. Radiotherapy was offered as a local treatment approach in all patients. RT techniques were classified into three regimens: standard palliative hypofractionated radiotherapy (hypo-RT), stereotactic radiotherapy (SRS/SBRT), and lattice radiotherapy (LRT). After a median follow-up of 22.5 months, the median TTR was 4 months (range 3-4) in patients with innate resistance vs 14 months (range 7-36) in patients with acquired resistance. Median TTNsT among patients with acquired and those with innate resistance was not reached (NR) vs 24 months (range 7-72). Overall, only six patients suffered from a local failure. Although out-of-field effects of radiotherapy were hypothesized, we were unable to record them as they did not occur during the observation period. Regardless of the radiation dose, there was no observable ≥ Grade 2 acute or late treatment-related toxicity. CONCLUSION Our preliminary results seem to confirm that the integration of radiotherapy and ICIs may allow for the continuation of systemic therapy beyond progression, which can have a subsequent benefit in terms of survival outcomes even in patients with innate resistance.
Collapse
Affiliation(s)
- Valentina Zagardo
- Radiation Oncology Unit, REM Radioterapia Srl, 95029, Viagrande, Italy
| | | | - Mandara Harikar
- Clinical Trials Programme, The University of Edinburgh, Edinburgh, UK
| | - Stefano Pergolizzi
- Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, 98122, Messina, Italy
| | - Gianluca Ferini
- Radiation Oncology Unit, REM Radioterapia Srl, 95029, Viagrande, Italy.
- Department of Medicine and Surgery, Kore University of Enna, Enna, Italy.
| |
Collapse
|
99
|
Wei Y, Li R, Wang Y, Fu J, Liu J, Ma X. Nanomedicines Targeting Tumor Cells or Tumor-Associated Macrophages for Combinatorial Cancer Photodynamic Therapy and Immunotherapy: Strategies and Influencing Factors. Int J Nanomedicine 2024; 19:10129-10144. [PMID: 39381025 PMCID: PMC11460276 DOI: 10.2147/ijn.s466315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 09/17/2024] [Indexed: 10/10/2024] Open
Abstract
Immunotherapy is a promising cancer treatment because of its ability to sustainably enhance the natural immune response. However, the effects of multiple immunotherapies, including ICIs, are limited by resistance to these agents, immune-related adverse events, and a lack of reasonable therapeutic targets available at the right time and place. The tumor microenvironment (TME), which features tumor-associated macrophages (TAMs), plays a significant role in resistance owing to its hypoxic microenvironment and lack of blood vessels, resulting in cancer immune evasion. To enhance immunotherapy, photodynamic therapy (PDT) can increase innate and adaptive immune responses through immunogenic cell death (ICD) and improve the TME. Traditional photosensitizers (PSs) also include novel nanomedicines to precisely target tumor cells or TAMs. Here, we reviewed and summarized current strategies and possible influencing factors for nanomedicines for cancer photoimmunotherapy.
Collapse
Affiliation(s)
- Yuhao Wei
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Renwei Li
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Yusha Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Jiali Fu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, People’s Republic of China
| | - Jifeng Liu
- Department of Otolaryngology Head and Neck Surgery/Deep Underground Space Medical Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
- State Key Laboratory of Intelligent Construction and Healthy Operation and Maintenance of Deep Underground Engineering, Sichuan University, Chengdu, People’s Republic of China
| | - Xuelei Ma
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| |
Collapse
|
100
|
Fang Z, Zhao G, Wang Y, Li F, Ding Z. The updated relationship between the cleft‑lip and palate transmembrane protein‑1‑like rs401681 and lung cancer risk: A systematic review and meta‑analysis. Mol Clin Oncol 2024; 21:70. [PMID: 39113849 PMCID: PMC11304168 DOI: 10.3892/mco.2024.2768] [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: 01/30/2024] [Accepted: 07/04/2024] [Indexed: 08/10/2024] Open
Abstract
Currently, the role of cleft-lip and palate transmembrane protein-1-like (CLPTM1L) rs401681 in various tumor types, particularly lung cancer, has garnered significant attention. However, the findings across studies have shown discrepancies. The aim of the present meta-analysis was to provide a more nuanced understanding of the involvement of CLPTM1L rs401681 in lung cancer development. Several electronic databases were systematically searched, including PubMed, Cochrane Library, Embase, Medline, Wanfang, Google Scholar and Chinese National Knowledge Infrastructure. Odds ratios (ORs) and 95% confidence intervals (CIs) were synthesized using random-effects models. Heterogeneity of included studies was assessed using the I2 statistic and Q test. Sensitivity analysis was conducted to evaluate the stability of overall estimates. Moreover, Egger's test was utilized to detect potential publication bias. The collective ORs indicated a significant association between the CLPTM1L rs401681 polymorphism and susceptibility to lung cancer across various genetic comparisons. These encompass allele T vs. allele C (OR=0.93, 95% CI=0.88-0.99, P<0.001), TT + CT vs. CC (OR=0.91, 95% CI=0.87-0.96, P<0.001), TT vs. CC + CT (OR=0.88, 95% CI=0.80-0.96, P<0.001), TT vs. CC (OR=0.84, 95% CI=0.75-0.94, P<0.001) and CT vs. CC (OR=0.84, 95% CI=0.75-0.94, P<0.001). Examination through statistical Q test and I2 statistic revealed pronounced heterogeneity across four genetic comparisons (allele T vs. allele C, TT + CT vs. CC, TT vs. CC and CT vs. CC). Ethnical distinctions emerged as the primary, if not exclusive, sources of the significant heterogeneity. Upon stratification by ethnicity, a notable reduction in heterogeneity was discernible within the Caucasian demographic. However, heterogeneity persisted within the Asian population. Furthermore, lung cancer risks were statistically significantly decreased for individuals possessing allele T through all genetic comparisons within Caucasians; whereas among Asians, significant reduction was observed solely in the TT vs. CC comparison. The present meta-analysis uncovers a significant association between the CLPTM1L rs401681 polymorphism and altered susceptibility to lung cancer.
Collapse
Affiliation(s)
- Zemin Fang
- Department of Lung Transplant, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Gaofeng Zhao
- Department of Lung Transplant, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Yuebin Wang
- Department of Lung Transplant, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Fengke Li
- Department of Lung Transplant, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Zhidan Ding
- Department of Lung Transplant, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| |
Collapse
|