1
|
Luo Y, Tan L, Meng C, Gao J, Chen H, Fang R, Wu X. Integrating bulk RNA-seq and scRNA-seq data to explore diverse cell death patterns and develop a programmed cell death-related relapse prediction model in pediatric B-ALL. Sci Rep 2025; 15:5620. [PMID: 39955305 PMCID: PMC11829959 DOI: 10.1038/s41598-025-86148-y] [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: 10/08/2024] [Accepted: 01/08/2025] [Indexed: 02/17/2025] Open
Abstract
Acute B-lymphoblastic leukemia (B-ALL) is a hematologic malignancy with diverse mechanisms of PCD influencing its progression. This study aimed to identify PCD-related biomarkers and develop a predictive model for relapse in pediatric B-ALL patients. Initially, we examined the activity of 16 PCD patterns in B-ALL patients using scRNA-seq. Following this, we employed both univariate and multivariate Cox regression analyses to identify relapse-related PCD patterns and constructed a relapse prediction model comprising seven key PCD-related genes: Bcl-2-interacting killer (BIK), translocator protein (TSPO), BCL2L2, PIP4K2C, mixed-lineage kinase-like (MLKL), STAT2, and WW domain-containing oxidoreductase (WWOX). Based on the optimal cut-off value derived from the cell death index(CDI) model, patients were categorized into high-CDI and low-CDI groups. Additionally, we evaluated the association between CDI scores and immune cell infiltration, tumor microenvironment (TME) characteristics, and drug sensitivity. Nine PCD patterns, encompassing ferroptosis, autophagy, necroptosis, entotic cell death, alkaliptosis, apoptosis, netotic cell death, oxeiptosis, and NETosis, exhibited strong associations with relapse in B-cell acute lymphoblastic leukemia (B-ALL). The CDI model, validated across multiple cohorts, demonstrated substantial predictive power for relapse-free survival (RFS) and was identified as an independent risk factor. This study offers a comprehensive analysis of PCD patterns in pediatric B-ALL, yielding valuable insights into potential novel therapeutic strategies and opportunities for personalized treatment approaches.
Collapse
Affiliation(s)
- Yaxin Luo
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lin Tan
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chuikai Meng
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jingyu Gao
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongxin Chen
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ruihan Fang
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuedong Wu
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Cui H, Liu Z, Peng L, Liu L, Xie X, Zhang Y, Gao Z, Zhang C, Yu X, Hu Y, Liu J, Shang L, Li L. A novel 5'tRNA-derived fragment tRF-Tyr inhibits tumor progression by targeting hnRNPD in gastric cancer. Cell Commun Signal 2025; 23:88. [PMID: 39953522 PMCID: PMC11829405 DOI: 10.1186/s12964-025-02086-2] [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: 10/02/2024] [Accepted: 02/05/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Transfer RNA-derived small RNAs (tsRNAs), including tRNA-derived fragments (tRFs) and tRNA halves (tiRNAs), constitute a novel class of small noncoding RNAs (sncRNAs). tsRNAs have been linked to tumorigenesis and the progression of carcinogenesis; however, the precise molecular mechanism through which tRFs act in gastric cancer (GC) remains unknown. METHODS tRF-Tyr is a potential GC tumor suppressor that was identified through high-throughput sequencing technology. The expression and subcellular localization of tRF-Tyr in GC were detected by via qRT‒PCR and FISH. RNA pull-down, mass spectrometry, RNA immunoprecipitation (RIP), dual-luciferase reporter and rescue assays were performed to explore the regulatory mechanisms through which tRF-Tyr acts in GC. RESULTS tRF-Tyr was significantly downregulated and the downregulation of its mainly concentrated in the nuclei of GC cells. Functionally, tRF-Tyr inhibited the proliferation, invasiveness and migration of GC cells and promoted GC cells apoptosis in vitro; meanwhile, tRF-Tyr inhibited tumor growth in vivo. Mechanistically, tRF-Tyr bound directly to the hnRNPD protein and competitively inhibited the binding of hnRNPD to the c-Myc 3'UTR, thereby, regulating the c-Myc/Bcl2/Bax pathway and ultimately inhibiting the progression of GC. CONCLUSIONS This study focused on a novel GC suppressor, tRF-Tyr, and revealed a previously undiscovered mechanism that tRF-Tyr inhibits tumor progression by targeting hnRNPD. These findings provide new insight into the involvement of tRFs in GC and suggest a novel target for GC treatment.
Collapse
Affiliation(s)
- Huaiping Cui
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwu Weiqi Road, Jinan, Shandong, 250021, China.
- Department of Gastrointestinal Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, Shandong, 250021, China.
| | - Zhaodong Liu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwu Weiqi Road, Jinan, Shandong, 250021, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, Shandong, 250021, China
| | - Lipan Peng
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwu Weiqi Road, Jinan, Shandong, 250021, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, Shandong, 250021, China
| | - Lijun Liu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwu Weiqi Road, Jinan, Shandong, 250021, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, Shandong, 250021, China
| | - Xiaozhou Xie
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwu Weiqi Road, Jinan, Shandong, 250021, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, Shandong, 250021, China
| | - Yudi Zhang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwu Weiqi Road, Jinan, Shandong, 250021, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, Shandong, 250021, China
| | - Zi Gao
- Department of Gastrointestinal Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, Shandong, 250021, China
| | - Chi Zhang
- Department of Gastrointestinal Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, Shandong, 250021, China
| | - Xinshuai Yu
- Department of Gastrointestinal Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, Shandong, 250021, China
| | - Yonghao Hu
- Department of Gastrointestinal Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, Shandong, 250021, China
| | - Jin Liu
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China.
| | - Liang Shang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwu Weiqi Road, Jinan, Shandong, 250021, China.
- Department of Gastrointestinal Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, Shandong, 250021, China.
| | - Leping Li
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwu Weiqi Road, Jinan, Shandong, 250021, China.
- Department of Gastrointestinal Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, Shandong, 250021, China.
| |
Collapse
|
3
|
Zheng Y, Li X, Huang G, Li Q, Yao X, Zhang W, Zhang Y, Meng X, Li Q, Huang G. Shuang Bailian mixture enhanced the anti-cancer effect of cisplatin by regulating PI3K-Akt-Bcl 2 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 343:119462. [PMID: 39923956 DOI: 10.1016/j.jep.2025.119462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/01/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Esophageal cancer (EC) is a prevalent malignant tumor with the characteristics of poor prognosis and high mortality in the clinic. Cisplatin (CP), a first-line chemotherapeutic agent, faces the challenge of drug resistance, which hampers its clinical efficacy. In recent decades, traditional Chinese medicine has earned increasing attention for its potential as an adjunct to chemotherapy. Shuang Bailian mixture (SBLM) could significantly enhances the anti-cancer effect of CP in our clinical practices. However, the underlying mechanism of SBLM still needs further investigation. AIM OF THIS STUDY To investigate the underlying mechanisms that SBLM enhanced the anti-cancer effect of CP. MATERIALS AND METHODS This study used network pharmacology and molecular docking to obtain the potential active targets of SBLM. Moreover, CCK-8 assay, wound healing, colony formation assays, flow cytometry and Western blotting were used to evaluate the properties of SBLM enhancing the anti-cancer effect of CP. Furthermore, tumor xenograft in nude mice model was used to further determine the potential mechanism that SBLM enhanced the anti-cancer effect of CP. RESULTS Network pharmacology analysis showed that the PI3K-Akt-Bcl 2 signaling pathway served as the primary target of SBLM. Moreover, SBLM could significantly improve the anti-cancer effect of CP, including inhibited cell proliferation, suppressed colony formation and invasion, and induced cell cycle arrest and apoptosis. Furthermore, SLBM + CP could significantly reduce the tumor sizes on tumor-bearing mice when compared to SBLM and CP treatment. Mechanistically, SLBM could enhance CP effect by inducing cancer cells apoptosis via PI3K-Akt-Bcl 2 signaling pathway. CONCLUSION Our study revealed the underlying mechanisms that SBLM enhanced the anti-cancer effect of CP through inhibiting the PI3K/Akt/Bcl-2 signaling pathway to induce the cells apoptosis. These results suggest that SBLM could serve as a promising adjuvant in chemotherapeutic regimens for EC.
Collapse
Affiliation(s)
- Yilin Zheng
- Clinical Research Center, Shantou Key Laboratory of Basic and Translational Research of Malignant Tumor, Shantou Central Hospital, Shantou, China.
| | - Xiaoyan Li
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Guohai Huang
- Blood Purification Center, Shantou Central Hospital, Shantou, Guangdong, China.
| | - Qingrui Li
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Xiaohua Yao
- Tropical Agriculture and Forestry College, Guangdong AIB Polytechnic College, Guangzhou, China.
| | - Weijin Zhang
- Department of Rheumatology and Immunology, Shantou Central Hospital, Shantou, China.
| | - Yuanfeng Zhang
- Clinical Research Center, Shantou Key Laboratory of Basic and Translational Research of Malignant Tumor, Shantou Central Hospital, Shantou, China.
| | - Xiongyu Meng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Qingnan Li
- Clinical Research Center, Shantou Key Laboratory of Basic and Translational Research of Malignant Tumor, Shantou Central Hospital, Shantou, China; Department of Pharmacy, Shantou Central Hospital, Shantou, China.
| | - Guoxin Huang
- Clinical Research Center, Shantou Key Laboratory of Basic and Translational Research of Malignant Tumor, Shantou Central Hospital, Shantou, China.
| |
Collapse
|
4
|
Tian F, Sun S, Ge Z, Ge Y, Ge X, Shi Z, Qian X. Understanding the Anticancer Effects of Phytochemicals: From Molecular Docking to Anticarcinogenic Signaling. J Nutr 2025; 155:431-444. [PMID: 39581266 DOI: 10.1016/j.tjnut.2024.11.015] [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: 11/13/2024] [Accepted: 11/20/2024] [Indexed: 11/26/2024] Open
Abstract
As nontraditional nutrients, the biological activity of phytochemicals have been extensively studied for their antioxidant, anti-inflammatory, and apoptosis-promoting effects in various diseases. The general anticancer benefits of phytochemicals have been demonstrated in both basic researches and clinical trials. However, researchers understanding of how phytochemicals target cancer-related signaling pathways is still in its infancy. Molecular docking simulation analyses have yielded a large amount of cellular target molecules of phytochemicals. Herein, we review the potential signaling pathways that may be involved in the phytochemical-driven cancer benefits. We expect these findings to help in the design of potential cancer treatments designed by manipulating the binding modes and sites of these plant chemicals.
Collapse
Affiliation(s)
- Fuwei Tian
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shuhong Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zehe Ge
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuqian Ge
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xin Ge
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhumei Shi
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China; Department of Neurosurgery of the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xu Qian
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China; Department of Neurosurgery of the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
5
|
Martí-Quijal FJ, Franco-Campos F, Barba FJ, Ruiz MJ. Citrinin-Induced Cellular Damage: Insights from SH-SY5Y Cell Line Studies. Foods 2025; 14:356. [PMID: 39941949 PMCID: PMC11817462 DOI: 10.3390/foods14030356] [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: 12/26/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 02/16/2025] Open
Abstract
Citrinin (CIT), a mycotoxin commonly found in cereals, is produced by fungi from the Aspergillus, Penicillium, and Monascus genera. While its nephrotoxic effects are well studied, its impact on neurons is less understood. This study investigates CIT-induced toxicity in human neuroblastoma cells (SH-SY5Y). The IC50 values for cells treated with CIT were 77.1 μM at 24 h and 74.7 μM at 48 h using MTT assay, and 101.0 μM at 24 h and 54.7 μM at 48 h using neutral red assay. CIT exposure caused G2/M phase arrest, with cells in this phase increasing from 11.83% (control) to 33.10% at 50 μM CIT. At 50 μM, the percentage of cells in the S phase also increased, which may suggest that cellular stress pathways were activated. Moreover, an increase in late apoptosis process was noted in cells exposed to CIT for 24 h, particularly at the highest concentrations (38.75 and 50 µM). Western blot analysis confirmed a rapid change in the anti-apoptotic protein Bcl-2, but no significant changes in Bax. In conclusion, CIT induces apoptosis and cell cycle arrest in SH-SY5Y cells. However, further transcriptomic studies in specific proteins involved in different pathways described in this work are needed to gain a comprehensive understanding of the specific mechanisms underlying CIT's toxicity in SH-SY5Y cells.
Collapse
Affiliation(s)
- Francisco J. Martí-Quijal
- Research Group in Innovative Technologies for Sustainable Food (ALISOST), Nutrition, Food Science and Toxicology Department, Faculty of Pharmacy, Universitat de València, Avda. Vicent Andrés Estellés, s/n, 46100 Burjassot, València, Spain; (F.J.M.-Q.); (F.J.B.)
- Research Group in Alternative Methods for Determining Toxics Effects and Risk Assessment of Contaminants and Mixtures (RiskTox), Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy and Food Science, University of Valencia, Av. Vicent Andrés Estellés, s/n, 46100 Burjassot, València, Spain;
| | - Felipe Franco-Campos
- Research Group in Alternative Methods for Determining Toxics Effects and Risk Assessment of Contaminants and Mixtures (RiskTox), Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy and Food Science, University of Valencia, Av. Vicent Andrés Estellés, s/n, 46100 Burjassot, València, Spain;
| | - Francisco J. Barba
- Research Group in Innovative Technologies for Sustainable Food (ALISOST), Nutrition, Food Science and Toxicology Department, Faculty of Pharmacy, Universitat de València, Avda. Vicent Andrés Estellés, s/n, 46100 Burjassot, València, Spain; (F.J.M.-Q.); (F.J.B.)
| | - María-José Ruiz
- Research Group in Alternative Methods for Determining Toxics Effects and Risk Assessment of Contaminants and Mixtures (RiskTox), Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy and Food Science, University of Valencia, Av. Vicent Andrés Estellés, s/n, 46100 Burjassot, València, Spain;
| |
Collapse
|
6
|
Lv S, Zhang X, Lang F, Wu Y, Zhang C, Qi Q, Jiang J. Long non-coding RNA LINC01224 plays an oncogenic role in endometrial cancer via miR-4673/TPX2 axis and activating Wnt/β-catenin signaling pathway. Biofactors 2025; 51:e2153. [PMID: 39780463 DOI: 10.1002/biof.2153] [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: 09/19/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025]
Abstract
Endometrial cancer (EC) is a prevalent gynecological malignancy with a rising incidence and poor prognosis in advanced cases. Long non-coding RNAs (lncRNAs) have been implicated in various cancers, including EC. This study explores the role of lncRNA Linc01224 in EC. Analyzing TCGA data, we found Linc01224 expression significantly elevated in EC tissues, correlating with poor prognosis. Clinical samples validated these findings, showing higher Linc01224 levels in tumor tissues. Knockdown of Linc01224 in EC cell lines (Hec-1-B and Ishikawa) inhibited proliferation, migration, and promoted apoptosis, alongside increased Bax and decreased BCL2 expression. Furthermore, Linc01224 knockdown notably reduced Wnt2/β-catenin pathway activation. We identified TPX2 as a target of miR-4673, which is regulated by Linc01224 through a competing endogenous RNA (ceRNA) mechanism. Dual-luciferase reporter assays confirmed miR-4673 binding to Linc01224 and TPX2. Rescue experiments revealed that TPX2 knockdown reversed Linc01224-induced proliferation and migration, highlighting TPX2's pivotal role in Linc01224's oncogenic function. In vivo, Linc01224 knockdown significantly impeded tumor growth and metastasis in a xenograft model, with decreased expression of c-Myc, Cyclin D1, and β-catenin. These findings reveal a novel ceRNA regulatory axis involving Linc01224, miR-4673, and TPX2, elucidating Linc01224's role in EC progression through the Wnt2/β-catenin pathway. Linc01224 emerges as a potential biomarker and therapeutic target for EC prognosis and treatment.
Collapse
Affiliation(s)
- Shuqing Lv
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, China
| | - Xinlu Zhang
- Department of Reproductive Medicine, Central Hospital Affiliation to Shandong First Medical University, Jinan, China
| | - Fangfang Lang
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, China
| | - Yanmei Wu
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, China
| | - Cancan Zhang
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, China
| | - Qi Qi
- Department of Pathology, The First Affiliated Hospital of Shangdong First Medical University, Jinan, China
| | - Jie Jiang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
7
|
Lou W, Zhang L, Wang J. Current status of nucleic acid therapy and its new progress in cancer treatment. Int Immunopharmacol 2024; 142:113157. [PMID: 39288629 DOI: 10.1016/j.intimp.2024.113157] [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: 06/11/2024] [Revised: 07/05/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024]
Abstract
Nucleic acid is an essential biopolymer in all living cells, performing the functions of storing and transmitting genetic information and synthesizing protein. In recent decades, with the progress of science and biotechnology and the continuous exploration of the functions performed by nucleic acid, more and more studies have confirmed that nucleic acid therapy for living organisms has great medical therapeutic potential. Nucleic acid drugs began to become independent therapeutic agents. As a new therapeutic method, nucleic acid therapy plays an important role in the treatment of genetic diseases, viral infections and cancers. There are currently 19 nucleic acid drugs approved by the Food and Drug Administration (FDA). In the following review, we start from principles and advantages of nucleic acid therapy, and briefly describe development history of nucleic acid drugs. And then we give examples of various RNA therapeutic drugs, including antisense oligonucleotides (ASO), mRNA vaccines, small interfering RNA (siRNA) and microRNA (miRNA), aptamers, and small activating RNA (saRNA). In addition, we also focused on the current status of nucleic acid drugs used in cancer therapy and the breakthrough in recent years. Clinical trials of nucleic acid drugs for cancer treatment are under way, conventional radiotherapy and chemotherapy combined with the immunotherapies such as checkpoint inhibitors and nucleic acid drugs may be the main prospects for successful cancer treatment.
Collapse
Affiliation(s)
- Wenting Lou
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Leqi Zhang
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Jianwei Wang
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China; Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, 2nd Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou 310009, China.
| |
Collapse
|
8
|
Sogbein O, Paul P, Umar M, Chaari A, Batuman V, Upadhyay R. Bortezomib in cancer therapy: Mechanisms, side effects, and future proteasome inhibitors. Life Sci 2024; 358:123125. [PMID: 39413903 DOI: 10.1016/j.lfs.2024.123125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/07/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024]
Abstract
The ubiquitin-proteasome pathway (UPP) regulates protein stability and normal cellular functions with the help of autocatalytic proteasome complex. Studies have linked aberrant proteasome activity to malignant cells and found that proteasome inhibitors play a significant role as therapeutic drugs for various types of cancer, specifically multiple myeloma and mantle cell lymphoma. Bortezomib, the first FDA-approved proteasome inhibitor for treating different stages of multiple myeloma, acts on cancer cells by inhibiting the 26S proteasome, modulating NF-κB, phosphorylating Bcl-2, upregulating of NOXA, blocking p53 degradation, activating caspase, generating reactive oxygen species (ROS), and inhibiting angiogenesis. However, its efficacy is limited due to side effects such as peripheral neuropathy (PN), thrombotic microangiopathy (TMA), and acute interstitial nephritis (AIN). Therefore, a better understanding of its precise mechanism of action may help mitigate these side effects. In this review, we have discussed the proposed mechanisms of action and off target effects of Bortezomib, along with the prospects of next generation potential proteasome inhibitor drugs in the treatment of cancer.
Collapse
Affiliation(s)
- Olusola Sogbein
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Pradipta Paul
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Qatar
| | - Meenakshi Umar
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Ali Chaari
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Qatar
| | - Vecihi Batuman
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| | - Rohit Upadhyay
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| |
Collapse
|
9
|
Sun KY, Bai XY, Zhang L, Zhang X, Hu QQ, Song YX, Qiang RR, Zhang N, Zou JL, Yang YL, Xiang Y. A new strategy for the treatment of intracerebral hemorrhage: Ferroptosis. Exp Neurol 2024; 382:114961. [PMID: 39288829 DOI: 10.1016/j.expneurol.2024.114961] [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: 06/07/2024] [Revised: 08/28/2024] [Accepted: 09/13/2024] [Indexed: 09/19/2024]
Abstract
Intracerebral hemorrhage, is a cerebrovascular disease with high morbidity, mortality, and disability. Due to the lack of effective clinical treatments, the development of new drugs to treat intracerebral hemorrhage is necessary. In recent years, ferroptosis has been found to play an important role in the pathophysiological process of intracerebral hemorrhage, which can be treated by inhibiting ferroptosis and thus intracerebral hemorrhage. This article aims to explain the mechanism of ferroptosis and its relationship to intracerebral hemorrhage. In the meantime, it briefly discusses the molecules identified to alleviate intracerebral hemorrhage by inhibiting ferroptosis, along with other clinical agents that are expected to treat intracerebral hemorrhage through this mechanism. In addition, a brief overview of the morphological alterations of different forms of cell death and their role in ICH is provided. Finally, the challenges that may arise in translating ferroptosis inhibitors from basic research to clinical use are presented. This article serves as a reference and provides insights to aid in the treatment of intracerebral hemorrhage in the clinic.
Collapse
Affiliation(s)
- Ke Yao Sun
- School of Medicine, Yan'an University, Yan'an, China
| | - Xin Yue Bai
- School of Medicine, Yan'an University, Yan'an, China
| | - Lei Zhang
- School of Medicine, Yan'an University, Yan'an, China
| | - Xin Zhang
- School of Medicine, Yan'an University, Yan'an, China
| | - Qian Qian Hu
- School of Medicine, Yan'an University, Yan'an, China
| | - Yu Xuan Song
- School of Medicine, Yan'an University, Yan'an, China
| | | | - Ning Zhang
- School of Medicine, Yan'an University, Yan'an, China
| | - Jia Lun Zou
- School of Medicine, Yan'an University, Yan'an, China
| | - Yan Ling Yang
- School of Medicine, Yan'an University, Yan'an, China
| | - Yang Xiang
- School of Medicine, Yan'an University, Yan'an, China; College of Physical Education, Yan'an University, Yan'an, China.
| |
Collapse
|
10
|
Ezzat RS, Abdel-Moneim A, Zoheir KMA, Mohamed EE, Abou-Seif HS, Hefnawy M, Ahmed OM. Anti-carcinogenic effects and mechanisms of actions of Citrus limon fruit peel hydroethanolic extract and limonene in diethylnitrosmine/2-acetylaminofluorene-induced hepatocellular carcinoma in Wistar rats. Am J Cancer Res 2024; 14:5193-5215. [PMID: 39659918 PMCID: PMC11626260 DOI: 10.62347/foyi6658] [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: 09/23/2024] [Accepted: 11/01/2024] [Indexed: 12/12/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the third most common cause of cancer death and disability in the world. Citrus species and their constituents have many biological activities including antioxidant, anti-inflammatory and anti-carcinogenic properties. This study aimed to assess the anti-carcinogenic effects and postulate the possible mechanisms of action for Citrus limon fruit peel hydroethanolic extract (CLFPHE) and limonene in diethylnitrosamine (DEN)/2-acetylaminofluorene (2AAF)-induced HCC in male Wistar rats. For analysis and characterization of CLFPHE, gas chromatography-mass spectrum (GC-MS) and high performance liquid chromatography (HPLC) methods were applied. A HCC was elaborated by DEN intraperitoneal injection (150 mg/kg/week) for two weeks followed by oral delivery of 2AAF (20 mg/kg) four times a week for three weeks. The DEN/2AAF-administered rats were treated with CLFPHE (50 mg/kg) and limonene (20 mg/kg) by oral gavage every other day for 24 weeks. CLFPHE and limonene significantly attenuated the harmful effects of DEN on liver function. Histopathological analysis confirmed that both treatments inhibited DEN/2AAF-induced tumorigenesis in association with the suppression of serum tumor markers including AFP, CEA, and CA19.9 and liver proliferator indicator (Ki-67). Moreover, CLFPHE and limonene prevented the oxidative stress and enhanced the antioxidant defenses in DEN/2AAF-administered rats. These ameliorations were manifested by decreases in liver lipid peroxidation, increases in GSH, SOD and GPx levels and upregulation of Nrf2. The treatments also abated inflammation by suppressing TNF-α and IL-1β levels and IL-8 and NF-κB expression. CLFPHE and limonene substantially decreased hepatic BCL-2, IQGAP1, IQGAP3, HRAS, KRAS and Ki-67 while they elevated BAX, P53, PDCD5 and IQGAP2 expressions. Our findings suggest that CLFPHE and limonene may abate HCC development via enhancement of apoptotic, antioxidant, cell anti-proliferatory and anti-inflammatory effects.
Collapse
Affiliation(s)
- Rania S Ezzat
- Physiology Division, Faculty of Science, Beni-Suef UniversityP.O. Box 62521, Beni-Suef, Egypt
| | - Adel Abdel-Moneim
- Physiology Division, Faculty of Science, Beni-Suef UniversityP.O. Box 62521, Beni-Suef, Egypt
| | - Khairy MA Zoheir
- Department of Cell Biology, Biotechnology Research Institute, National Research CentreCairo 12622, Egypt
| | - Eman E Mohamed
- Physiology Division, Faculty of Science, Beni-Suef UniversityP.O. Box 62521, Beni-Suef, Egypt
| | - Howida S Abou-Seif
- Department of Medical Physiology, Medical Research and Clinical Studies Institute, National Research CentreCairo 12622, Egypt
| | - Mohamed Hefnawy
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud UniversityRiyadh 11451, Saudi Arabia
| | - Osama M Ahmed
- Physiology Division, Faculty of Science, Beni-Suef UniversityP.O. Box 62521, Beni-Suef, Egypt
| |
Collapse
|
11
|
López-Gil CI, Téllez-Jurado A, Velasco-Velázquez MA, Anducho-Reyes MA. Identification and Analysis of Anticancer Therapeutic Targets from the Polysaccharide Krestin (PSK) and Polysaccharopeptide (PSP) Using Inverse Docking. Molecules 2024; 29:5390. [PMID: 39598781 PMCID: PMC11596896 DOI: 10.3390/molecules29225390] [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: 09/23/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024] Open
Abstract
The natural compounds PSK and PSP have antitumor and immunostimulant properties. These pharmacological benefits have been documented in vitro and in vivo, although there is no information in silico which describes the action mechanisms at the molecular level. In this study, the inverse docking method was used to identify the interactions of PSK and PSP with two local databases: BPAT with 66 antitumor proteins, and BPSIC with 138 surfaces and intracellular proteins. This led to the identification interactions and similarities of PSK and the AB680 inhibitor in the active site of CD73. It was also found that PSK binds to CD59, interacting with the amino acids APS22 and PHE23, which coincide with the rlLYd4 internalization inhibitor. With the isoform of the K-RAS protein, PSK bonded to the TYR32 amino acid at switch 1, while with BAK it bonded to the region of the α1 helix, while PSP bonded to the activation site and the C-terminal and N-terminal ends of that helix. In Bcl-2, PSK interacted at the binding site of the Venetoclax inhibitor, showing similarities with the amino acids ASP111, VAL133, LEU137, MET115, PHE112, and TYR108, while PSP had similarities with THR132, VAL133, LEU137, GLN118, MET115, APS111, PHE112, and PHE104.
Collapse
Affiliation(s)
- Carlos Iván López-Gil
- Department of Biotechnology, Universidad Politécnica de Pachuca, Zempoala 43830, Mexico; (C.I.L.-G.); (A.T.-J.)
| | - Alejandro Téllez-Jurado
- Department of Biotechnology, Universidad Politécnica de Pachuca, Zempoala 43830, Mexico; (C.I.L.-G.); (A.T.-J.)
| | | | - Miguel Angel Anducho-Reyes
- Department of Biotechnology, Universidad Politécnica de Pachuca, Zempoala 43830, Mexico; (C.I.L.-G.); (A.T.-J.)
| |
Collapse
|
12
|
Zhang L, Song Z, He L, Zhong S, Ju X, Sha H, Xu J, Qin Q, Peng J, Liang H. Unveiling the toxicological effects and risks of prometryn on red swamp crayfish (Procambarus clarkii): Health assessments, ecological, and molecular insights. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 951:175512. [PMID: 39151629 DOI: 10.1016/j.scitotenv.2024.175512] [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: 05/31/2024] [Revised: 07/21/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024]
Abstract
Prometryn is commonly used in agricultural and non-agricultural settings. However, possible harm to aquatic organisms remains a persistent concern. Prometryn was also the only one of the 26 triazine herbicides detected in this study. Numerous studies have assessed the harmful effects of prometryn in teleost fish and shrimp. There is a lack of information regarding the ecological and human health risks, as well as the toxic mechanisms affecting crayfish. In this study, human health risk assessment (THQ) and ecological risk assessment (RQ) were conducted on P. clarkii in the rice-crayfish co-culture (IRCC) farming model. The 96 h of exposure to 0.286 mg/L and 1.43 mg/L prometryn was conducted to investigate the potential effects and molecular mechanisms of hepatopancreatic resistance to prometryn in P. clarkii. The original sample analysis revealed that the THQ calculated from the prometryn levels in the muscle and hepatopancreas was below 0.1, suggesting no threat to human health. However, the calculated RQ values were >0.1, indicating a risk to P. clarkii. Histological analysis and biochemical index detection of the experimental samples revealed that the hepatopancreatic injury and oxidative damage in P. clarkii were caused by prometryn. Moreover, transcriptome analysis identified 2512 differentially expressed genes (DEGs) after 96 h of prometryn exposure. Prometryn exposure caused significant changes in metabolic pathways, including oxoacid metabolic processes and cytochrome P450-associated drug metabolism. Further hub gene analysis via PPI indicated that exposure to prometryn may inhibit lipid synthesis, storage, and amino acid transport and affect glucose metabolic pathways and hormone synthesis. Additionally, we hypothesized that prometryn-triggered cell death could be linked to the PI3K-Akt signaling cascade. This study's findings have significant meaning for the efficient and logical application of herbicides in IRCC, ultimately aiding in advancing a highly productive agricultural system.
Collapse
Affiliation(s)
- Lang Zhang
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China.
| | - Ziwei Song
- Department of Genetics, Wuhan University, Wuhan, Hubei 430071, China; School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Li He
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China
| | - Shan Zhong
- Department of Genetics, Wuhan University, Wuhan, Hubei 430071, China; Hubei Province Key Laboratory of Allergy and Immunology, Wuhan, Hubei 430071, China
| | - Xiaoqian Ju
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China
| | - Hang Sha
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China
| | - Jing Xu
- College of Pharmacy, South Central University for Nationalities, Wuhan 430074, China
| | - Qiuying Qin
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China; College of Pharmacy, South Central University for Nationalities, Wuhan 430074, China
| | - Jie Peng
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China.
| | - Hongwei Liang
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China.
| |
Collapse
|
13
|
Khuu A, Verreault M, Colin P, Tran H, Idbaih A. Clinical Applications of Antisense Oligonucleotides in Cancer: A Focus on Glioblastoma. Cells 2024; 13:1869. [PMID: 39594617 PMCID: PMC11592788 DOI: 10.3390/cells13221869] [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: 09/09/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Antisense oligonucleotides (ASOs) are promising drugs capable of modulating the protein expression of virtually any target with high specificity and high affinity through complementary base pairing. However, this requires a deep understanding of the target sequence and significant effort in designing the correct complementary drug. In addition, ASOs have been demonstrated to be well tolerated during their clinical use. Indeed, they are already used in many diseases due to pathogenic RNAs of known sequences and in several neurodegenerative diseases and metabolic diseases, for which they were given marketing authorizations (MAs) in Europe and the United States. Their use in oncology is gaining momentum with several identified targets, promising preclinical and clinical results, and recent market authorizations in the US. However, many challenges remain for their clinical use in cancer. It seems necessary to take a step back and review our knowledge of ASOs and their therapeutic uses in oncology. The objectives of this review are (i) to summarize the current state of the art of ASOs; (ii) to discuss the therapeutic use of ASOs in cancer; and (iii) to focus on ASO usage in glioblastoma, the challenges, and the perspective ahead.
Collapse
Affiliation(s)
- Alexandre Khuu
- AP-HP, Institut du Cerveau, Paris Brain Institute, ICM, Inserm U 1127, CNRS UMR 7225, Hôpitaux Universitaires La Pitié Salpêtrière, Charles Foix, DMU Neurosciences, Service de Neuro-Oncologie-Institut de Neurologie, Sorbonne Université, 75013 Paris, France; (A.K.); (M.V.)
- Institut de Recherche Servier, Rue Francis Perrin, 91190 Gif-sur-Yvette, France;
| | - Maïté Verreault
- AP-HP, Institut du Cerveau, Paris Brain Institute, ICM, Inserm U 1127, CNRS UMR 7225, Hôpitaux Universitaires La Pitié Salpêtrière, Charles Foix, DMU Neurosciences, Service de Neuro-Oncologie-Institut de Neurologie, Sorbonne Université, 75013 Paris, France; (A.K.); (M.V.)
| | - Philippe Colin
- Institut de Recherche Servier, Rue Francis Perrin, 91190 Gif-sur-Yvette, France;
| | - Helene Tran
- Institut de Recherche Servier, Rue Francis Perrin, 91190 Gif-sur-Yvette, France;
| | - Ahmed Idbaih
- AP-HP, Institut du Cerveau, Paris Brain Institute, ICM, Inserm U 1127, CNRS UMR 7225, Hôpitaux Universitaires La Pitié Salpêtrière, Charles Foix, DMU Neurosciences, Service de Neuro-Oncologie-Institut de Neurologie, Sorbonne Université, 75013 Paris, France; (A.K.); (M.V.)
| |
Collapse
|
14
|
Jiang RY, Zhu JY, Zhang HP, Yu Y, Dong ZX, Zhou HH, Wang X. STAT3: Key targets of growth-promoting receptor positive breast cancer. Cancer Cell Int 2024; 24:356. [PMID: 39468521 PMCID: PMC11520424 DOI: 10.1186/s12935-024-03541-9] [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: 07/26/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024] Open
Abstract
Breast cancer has become the malignant tumor with the first incidence and the second mortality among female cancers. Most female breast cancers belong to luminal-type breast cancer and HER2-positive breast cancer. These breast cancer cells all have different driving genes, which constantly promote the proliferation and metastasis of breast cancer cells. Signal transducer and activator of transcription 3 (STAT3) is an important breast cancer-related gene, which can promote the progress of breast cancer. It has been proved in clinical and basic research that over-expressed and constitutively activated STAT3 is involved in the progress, proliferation, metastasis and chemotherapy resistance of breast cancer. STAT3 is an important key target in luminal-type breast cancer and HER2-positive cancer, which has an important impact on the curative effect of related treatments. In breast cancer, the activation of STAT3 will change the spatial position of STAT3 protein and cause different phenotypic changes of breast cancer cells. In the current basic research and clinical research, small molecule inhibitors activated by targeting STAT3 can effectively treat breast cancer, and enhance the efficacy level of related treatment methods for luminal-type and HER2-positive breast cancers.
Collapse
Affiliation(s)
- Rui-Yuan Jiang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, NO.548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Jia-Yu Zhu
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, NO.548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Huan-Ping Zhang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Department of Graduate Student, Wenzhou Medical University, No.270, Xueyuan West Road, Lucheng District, Wenzhou, 325027, Zhejiang, China
| | - Yuan Yu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Zhi-Xin Dong
- Department of Oncology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, No.89-9, Dongge Road, Qingxiu District, Nanning, 530000, Guangxi, China
| | - Huan-Huan Zhou
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, NO.548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China.
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| | - Xiaojia Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
15
|
Sudaarsan ASK, Ghosh AR. Appraisal of postbiotics in cancer therapy. Front Pharmacol 2024; 15:1436021. [PMID: 39372197 PMCID: PMC11449718 DOI: 10.3389/fphar.2024.1436021] [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: 05/21/2024] [Accepted: 09/05/2024] [Indexed: 10/08/2024] Open
Abstract
Cancer remains a multifactorial disease with an increased mortality rate around the world for the past several decades. Despite advancements in treatment strategies, lower survival rates, drug-associated side effects, and drug resistance create a need for novel anticancer agents. Ample evidence shows that imbalances in the gut microbiota are associated with the formation of cancer and its progression. Altering the gut microbiota via probiotics and their metabolites has gained attention among the research community as an alternative therapy to treat cancer. Probiotics exhibit health benefits as well as modulate the immunological and cellular responses in the host. Apart from probiotics, their secreted products like bacteriocins, exopolysaccharides, short-chain fatty acids, conjugated linoleic acid, peptidoglycan, and other metabolites are found to possess anticancer activity. The beneficiary role of these postbiotic compounds is widely studied for characterizing their mechanism and mode of action that reduces cancer growth. The present review mainly focuses on the postbiotic components that are employed against cancer with their reported mechanism of action. It also describes recent research works carried out so far with specific strain and anticancer activity of derived compounds both in vitro and in vivo, validating that the probiotic approach would pave an alternative way to reduce the burden of cancer.
Collapse
|
16
|
El-Naggar NEA, Shweqa NS, Abdelmigid HM, Alyamani AA, Elshafey N, Soliman HM, Heikal YM. Myco-Biosynthesis of Silver Nanoparticles, Optimization, Characterization, and In Silico Anticancer Activities by Molecular Docking Approach against Hepatic and Breast Cancer. Biomolecules 2024; 14:1170. [PMID: 39334936 PMCID: PMC11429812 DOI: 10.3390/biom14091170] [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: 07/31/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
This study explored the green synthesis of silver nanoparticles (AgNPs) using the extracellular filtrate of Fusarium oxysporum as a reducing agent and evaluated their antitumor potential through in vitro and in silico approaches. The biosynthesis of AgNPs was monitored by visual observation of the color change and confirmed by UV-Vis spectroscopy, revealing a characteristic peak at 418 nm. Scanning electron microscopy (SEM) and transmission electron microscopy (TEM) analyses showed spherical nanoparticles ranging from 6.53 to 21.84 nm in size, with stable colloidal behavior and a negative zeta potential of -15.5 mV. Selected area electron diffraction (SAED) confirmed the crystalline nature of the AgNPs, whereas energy-dispersive X-ray (EDX) indicated the presence of elemental silver at 34.35%. A face-centered central composite design (FCCD) was employed to optimize the biosynthesis process, yielding a maximum AgNPs yield of 96.77 µg/mL under the optimized conditions. The antitumor efficacy of AgNPs against MCF-7 and HepG2 cancer cell lines was assessed, with IC50 values of 35.4 µg/mL and 7.6 µg/mL, respectively. Molecular docking revealed interactions between Ag metal and key amino acids of BCL-2 (B-cell lymphoma-2) and FGF19 (fibroblast growth factor 19), consistent with in vitro data. These findings highlight the potential of biologically derived AgNPs as promising therapeutic agents for cancer treatment and demonstrate the utility of these methods for understanding the reaction mechanisms and optimizing nanomaterial synthesis.
Collapse
Affiliation(s)
- Noura El-Ahmady El-Naggar
- Department of Bioprocess Development, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg El Arab City 21934, Egypt
| | - Nada S Shweqa
- Botany Department, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Hala M Abdelmigid
- Department of Biotechnology, College of Science, Taif University, Taif 21944, Saudi Arabia
| | - Amal A Alyamani
- Department of Biotechnology, College of Science, Taif University, Taif 21944, Saudi Arabia
| | - Naglaa Elshafey
- Botany and Microbiology Department, Faculty of Science, Arish University, Al-Arish 45511, Egypt
| | - Hoda M Soliman
- Botany Department, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Yasmin M Heikal
- Botany Department, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
17
|
An G, Hui J, Zhang W, Fan A, Zhou Y, Zhao X, Lu Y, Wang X. A novel lncRNA associated with the prognosis of patients with colorectal cancer resists apoptosis through the LYN/BCL-2 pathway. Biochem Biophys Res Commun 2024; 723:150177. [PMID: 38810320 DOI: 10.1016/j.bbrc.2024.150177] [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: 03/22/2024] [Revised: 05/14/2024] [Accepted: 05/24/2024] [Indexed: 05/31/2024]
Abstract
PURPOSE We found a novel lncRNA named lncAC138150.2 related to the overall survival and staging of patients with colorectal cancer (CRC) by bioinformatic analysis using data from the Cancer Genome Atlas (TCGA), and the study aimed to elucidate the function of lncAC138150.2 and underlying mechanisms. METHODS Target molecules were knocked down by transfection with antisense oligonucleotides (ASOs), siRNAs, or lentiviruses and overexpressed by transfection with plasmids. The function of lncAC138150.2 was determined using histological, cytological, and molecular biology methods. The underlying mechanism of lncAC138150.2 function was investigated using RNA-seq, bioinformatics analysis, and molecular biology methods. RESULTS The expression of lncAC138150.2 was increased in colorectal tissues compared with paired normal tissues. The lncAC138150.2 knockdown increased apoptosis but did not change the cell proliferation, cell cycle distribution, or cell migration ability of CRC cells, while lncAC138150.2 overexpression decreased CRC apoptosis. lncAC138150.2 was mainly located in the cell nucleus, and each lncAC138150.2 transcript knockdown increased CRC apoptosis. BCL-2 pathway was significantly altered in apoptosis induced by lncAC138150.2 knockdown, which was alleviated by BAX knockdown. The expression of LYN was significantly decreased with lncAC138150.2 knockdown, LYN knockdown increased CRC apoptosis, and its overexpression completely alleviated CRC apoptosis induced by lncAC138150.2 knockdown. CONCLUSION lncAC138150.2 significantly inhibited CRC apoptosis and affected the prognosis of patients with CRC, through the LYN/BCL-2 pathway.
Collapse
Affiliation(s)
- Guangzhou An
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China; Department of Radiation Protection Medicine, Ministry of Education Key Laboratory of Hazard Assessment and Control in Special Operational Environment, Faculty of Preventive Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Juan Hui
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Wenyao Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, China
| | - Ahui Fan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, China
| | - Yun Zhou
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Xiaodi Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, China
| | - Yuanyuan Lu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, China.
| | - Xin Wang
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China.
| |
Collapse
|
18
|
Rigillo G, Cappellucci G, Baini G, Vaccaro F, Miraldi E, Pani L, Tascedda F, Bruni R, Biagi M. Comprehensive Analysis of Berberis aristata DC. Bark Extracts: In Vitro and In Silico Evaluation of Bioaccessibility and Safety. Nutrients 2024; 16:2953. [PMID: 39275269 PMCID: PMC11397700 DOI: 10.3390/nu16172953] [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: 07/30/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
Berberine (BER) is an alkaloid found, together with other protoberberinoids (PROTBERs), in several species used in medicines and food supplements. While some herbal preparations containing BER and PROTBERs, such as Berberis aristata DC. bark extracts, have shown promising potential for human health, their safety has not been fully assessed. Recently, the EFSA issued a call for data to deepen the pharmacokinetic and pharmacodynamic understanding of products containing BER and PROTBERs and to comprehensively assess their safety, especially when used in food supplements. In this context, new data were collected in this work by assessing: (i) the phytochemical profile of 16 different commercial B. aristata dry extracts, which are among the most widely used preparations containing BER and PROTBERs in Europe; (ii) the In Vitro and In Silico investigation of the pharmacokinetic properties of BER and PROTBERs; (iii) the In Vitro cytotoxicity of selected extracts in different human cell lines, including tests on hepatic cells in the presence of CYP450 substrates; (iv) the effects of the extracts on cancer cell migration; and (v) the In Vitro molecular effects of extracts in non-cancer human cells. Results showed that commercial B. aristata extracts contain BER as the main constituent, with jatrorrhizine as main secondary PROTBER. BER and jatrorrhizine were found to have a good bioaccessibility rate, but they interact with P-gp. B. aristata extracts showed limited cytotoxicity and minimal interaction with CYP450 substrates. Furthermore, tested extracts demonstrated inhibition of cancer cell migration and were devoid of any pro-tumoral effects in normal cells. Overall, our work provides a valuable overview to better elucidate important concerns regarding botanicals containing BER and PROTBERs.
Collapse
Affiliation(s)
- Giovanna Rigillo
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Laboratory of Italian Society of Phytoterapy-SIFITLab, 53100 Siena, Italy
| | - Giorgio Cappellucci
- Laboratory of Italian Society of Phytoterapy-SIFITLab, 53100 Siena, Italy
- Department of Physical Sciences, Earth and Environment, University of Siena, 53100 Siena, Italy
| | - Giulia Baini
- Laboratory of Italian Society of Phytoterapy-SIFITLab, 53100 Siena, Italy
- Department of Physical Sciences, Earth and Environment, University of Siena, 53100 Siena, Italy
| | - Federica Vaccaro
- Laboratory of Italian Society of Phytoterapy-SIFITLab, 53100 Siena, Italy
- Department of Physical Sciences, Earth and Environment, University of Siena, 53100 Siena, Italy
| | - Elisabetta Miraldi
- Laboratory of Italian Society of Phytoterapy-SIFITLab, 53100 Siena, Italy
- Department of Physical Sciences, Earth and Environment, University of Siena, 53100 Siena, Italy
| | - Luca Pani
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Department of Psychiatry and Behavioral Sciences, University of Miami, Miami, FL 33136, USA
| | - Fabio Tascedda
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Consorzio Interuniversitario Biotecnologie (CIB), 34148 Trieste, Italy
| | - Renato Bruni
- Department of Food and Drug, University of Parma, 43124 Parma, Italy
| | - Marco Biagi
- Laboratory of Italian Society of Phytoterapy-SIFITLab, 53100 Siena, Italy
- Department of Food and Drug, University of Parma, 43124 Parma, Italy
| |
Collapse
|
19
|
Beilankouhi EAV, Maghsoodi MS, Sani MZ, Khosroshahi NS, Zarezadeh R, Nargesi MM, Safaralizadeh R, Valilo M. miRNAs that regulate apoptosis in breast cancer and cervical cancer. Cell Biochem Biophys 2024; 82:1993-2006. [PMID: 38969951 DOI: 10.1007/s12013-024-01405-7] [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] [Accepted: 07/03/2024] [Indexed: 07/07/2024]
Abstract
In today's world, one of the main problems is cancer, which still has a long way to go to cure it, and it brings a lot of financial and emotional costs to the people of society and governments. Breast cancer (BC) and cervical cancer (CC), two of the most common cancers, are caused by several genetic and environmental factors in women. These two cancers' involvement rate is higher than other cancers in women. microRNAs (miRNAs) are non-coding RNA molecules with a length of 18 to 24 nucleotides, which play an important role in post-translational changes. miRNAs themselves are divided into two categories, oncomiRs and tumor suppressors. OncomiRs have a part in tumor expansion and tumor suppressors prevent tumor development and progress. miRNAs can control cellular processes by regulating various pathways including autophagy, apoptosis, and signaling. Apoptosis is a type of programmed cell death that includes intrinsic and extrinsic pathways and is different from other cell death pathways such as necrosis and ferroptosis. Apoptosis controls the growth, differentiation, and death of cells by regulating the death of damaged and old cells, and since miRNAs are one of the factors that regulate apoptosis, and divided into two categories: pro-apoptotic and anti-apoptotic. We decided in this study to investigate the relationship between miRNAs and apoptosis in the most common women's cancers, BC and CC.
Collapse
Affiliation(s)
| | - Maral Salek Maghsoodi
- Department of Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Maryam Zamani Sani
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Negin Sadi Khosroshahi
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Reza Zarezadeh
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mirsaed Miri Nargesi
- Molecular Virology and Covid Unit, LabPlus, Department of Pathology and Laboratory Medicine, Auckland City Hospital, Te Whatu Ora Health New Zealand, Auckland, New Zealand
| | - Reza Safaralizadeh
- Department of Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Mohammad Valilo
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
20
|
Zhao Q, Han B, Peng C, Zhang N, Huang W, He G, Li JL. A promising future of metal-N-heterocyclic carbene complexes in medicinal chemistry: The emerging bioorganometallic antitumor agents. Med Res Rev 2024; 44:2194-2235. [PMID: 38591229 DOI: 10.1002/med.22039] [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: 01/19/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/10/2024]
Abstract
Metal complexes based on N-heterocyclic carbene (NHC) ligands have emerged as promising broad-spectrum antitumor agents in bioorganometallic medicinal chemistry. In recent decades, studies on cytotoxic metal-NHC complexes have yielded numerous compounds exhibiting superior cytotoxicity compared to cisplatin. Although the molecular mechanisms of these anticancer complexes are not fully understood, some potential targets and modes of action have been identified. However, a comprehensive review of their biological mechanisms is currently absent. In general, apoptosis caused by metal-NHCs is common in tumor cells. They can cause a series of changes after entering cells, such as mitochondrial membrane potential (MMP) variation, reactive oxygen species (ROS) generation, cytochrome c (cyt c) release, endoplasmic reticulum (ER) stress, lysosome damage, and caspase activation, ultimately leading to apoptosis. Therefore, a detailed understanding of the influence of metal-NHCs on cancer cell apoptosis is crucial. In this review, we provide a comprehensive summary of recent advances in metal-NHC complexes that trigger apoptotic cell death via different apoptosis-related targets or signaling pathways, including B-cell lymphoma 2 (Bcl-2 family), p53, cyt c, ER stress, lysosome damage, thioredoxin reductase (TrxR) inhibition, and so forth. We also discuss the challenges, limitations, and future directions of metal-NHC complexes to elucidate their emerging application in medicinal chemistry.
Collapse
Affiliation(s)
- Qian Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Dermatology & Venerolog, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Gu He
- Department of Dermatology & Venerolog, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jun-Long Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Anti-Infective Agent Creation Engineering Research Centre of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, China
| |
Collapse
|
21
|
Jang H, Seo S, Park S, Kim BJ, Choi GW, Choi J, Park C. De novo drug design through gradient-based regularized search in information-theoretically controlled latent space. J Comput Aided Mol Des 2024; 38:32. [PMID: 39190191 PMCID: PMC11349835 DOI: 10.1007/s10822-024-00571-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/31/2024] [Indexed: 08/28/2024]
Abstract
Over the last decade, automatic chemical design frameworks for discovering molecules with drug-like properties have significantly progressed. Among them, the variational autoencoder (VAE) is a cutting-edge approach that models the tractable latent space of the molecular space. In particular, the usage of a VAE along with a property estimator has attracted considerable interest because it enables gradient-based optimization of a given molecule. However, although successful results have been achieved experimentally, the theoretical background and prerequisites for the correct operation of this method have not yet been clarified. In view of the above, we theoretically analyze and rigorously reconstruct the entire framework. From the perspective of parameterized distribution and the information theory, we first describe how the previous model overcomes the limitations of the beta VAE in discovering molecules with the desired properties. Furthermore, we describe the prerequisites for training the above model. Next, from the log-likelihood perspective of each term, we reformulate the objectives for exploring latent space to generate drug-like molecules. The distributional constraints are defined in this study, which will break away from the invalid molecular search. We demonstrated that our model could discover a novel chemical compound for targeting BCL-2 family proteins in de novo approach. Through the theoretical analysis and practical implementation, the importance of the aforementioned prerequisites and constraints to operate the model was verified.
Collapse
Affiliation(s)
- Hyosoon Jang
- Graduate School of AI, POSTECH, 77 Cheongam-Ro, Pohang, 37673, Gyeongbuk, Republic of Korea
| | - Sangmin Seo
- Department of Computer Science, Yonsei University, Yonsei-ro 50, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Sanghyun Park
- Department of Computer Science, Yonsei University, Yonsei-ro 50, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Byung Ju Kim
- UBLBio Corporation, Yeongtong-ro 237, Suwon, 16679, Gyeonggi-do, Republic of Korea
| | - Geon-Woo Choi
- Department of Medical Bigdata Convergence, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, 24341, Gangwon-do, Republic of Korea
| | - Jonghwan Choi
- College of Information Science, Hallym University, 1 Hallymdaehak-gil, Chuncheon, 24252, Gangwon-do, Republic of Korea.
| | - Chihyun Park
- Department of Medical Bigdata Convergence, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, 24341, Gangwon-do, Republic of Korea.
- Department of Compupter Science and Engineering, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, 24341, Gangwon-do, Republic of Korea.
| |
Collapse
|
22
|
de Melo Silva AJ, de Melo Gama JE, de Oliveira SA. The Role of Bcl-2 Family Proteins and Sorafenib Resistance in Hepatocellular Carcinoma. Int J Cell Biol 2024; 2024:4972523. [PMID: 39188653 PMCID: PMC11347034 DOI: 10.1155/2024/4972523] [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: 04/13/2024] [Revised: 07/10/2024] [Accepted: 08/02/2024] [Indexed: 08/28/2024] Open
Abstract
Liver cancer has been reported to be one of the most malignant diseases in the world. It is late diagnosis consequently leads to a difficult treatment, as the cancer reached an advanced stage. Hepatocellular carcinoma (HCC) is the primary type of cancer diagnosed in the liver, with deadly characteristics and a poor prognosis. The first-in-line treatment for advanced HCC is sorafenib. Sorafenib acts by inhibiting cell proliferation and by inducing apoptosis as well as blocks receptors associated with these mechanisms. Due to its constant use, sorafenib resistance has been described, especially to proteins of the Bcl-2 family, and their overexpression of Bcl-XL and Mcl-1. This review focuses on the role of the Bcl-2 proteins in relation to sorafenib resistance as a consequence of first-in-line treatment in HCC.
Collapse
|
23
|
Ruocco N, Nuzzo G, Federico S, Esposito R, Gallo C, Ziaco M, Manzo E, Fontana A, Bertolino M, Zagami G, Zupo V, Sansone C, Costantini M. Potential of Polar Lipids Isolated from the Marine Sponge Haliclona ( Halichoclona) vansoesti against Melanoma. Int J Mol Sci 2024; 25:7418. [PMID: 39000524 PMCID: PMC11242152 DOI: 10.3390/ijms25137418] [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: 05/24/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
Marine sponges represent a good source of natural metabolites for biotechnological applications in the pharmacological, cosmeceutical, and nutraceutical fields. In the present work, we analyzed the biotechnological potential of the alien species Haliclona (Halichoclona) vansoesti de Weerdt, de Kluijver & Gomez, 1999, previously collected in the Mediterranean Sea (Faro Lake, Sicily). The bioactivity and chemical content of this species has never been investigated, and information in the literature on its Caribbean counterpart is scarce. We show that an enriched extract of H. vansoesti induced cell death in human melanoma cells with an IC50 value of 36.36 µg mL-1, by (i) triggering a pro-inflammatory response, (ii) activating extrinsic apoptosis mediated by tumor necrosis factor receptors triggering the mitochondrial apoptosis via the involvement of Bcl-2 proteins and caspase 9, and (iii) inducing a significant reduction in several proteins promoting human angiogenesis. Through orthogonal SPE fractionations, we identified two active sphingoid-based lipid classes, also characterized by nuclear magnetic resonance and mass spectrometry, as the main components of two active fractions. Overall, our findings provide the first evaluation of the anti-cancer potential of polar lipids isolated from the marine sponge H. (Halichoclona) vansoesti, which may lead to new lead compounds with biotechnological applications in the pharmaceutical field.
Collapse
Affiliation(s)
- Nadia Ruocco
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Calabria Marine Centre, C. da Torre Spaccata, 87071 Amendolara, Italy
| | - Genoveffa Nuzzo
- Bio-Organic Chemistry Unit, Institute of Biomolecular Chemistry CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Serena Federico
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Via Ammiraglio Ferdinando Acton, 55, 80133 Napoli, Italy
- Department of Earth, Environmental and Life Sciences, University of Genoa, Corso Europa 26, 16132 Genova, Italy
| | - Roberta Esposito
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Via Ammiraglio Ferdinando Acton, 55, 80133 Napoli, Italy
| | - Carmela Gallo
- Bio-Organic Chemistry Unit, Institute of Biomolecular Chemistry CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Marcello Ziaco
- Bio-Organic Chemistry Unit, Institute of Biomolecular Chemistry CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Emiliano Manzo
- Bio-Organic Chemistry Unit, Institute of Biomolecular Chemistry CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Angelo Fontana
- Bio-Organic Chemistry Unit, Institute of Biomolecular Chemistry CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Italy
- Laboratory of Bio-Organic Chemistry and Chemical Biology, Dipartimento di Biologia, Università di Napoli "Federico II", Via Cupa Nuova Cinthia 21, 80126 Napoli, Italy
| | - Marco Bertolino
- Department of Earth, Environmental and Life Sciences, University of Genoa, Corso Europa 26, 16132 Genova, Italy
| | - Giacomo Zagami
- Department of Biological, Chemical, Pharmaceutical and Environmental Sciences, University of Messina, 98100 Messina, Italy
| | - Valerio Zupo
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Ischia Marine Center, 80077 Ischia, Italy
| | - Clementina Sansone
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Via Ammiraglio Ferdinando Acton, 55, 80133 Napoli, Italy
| | - Maria Costantini
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Via Ammiraglio Ferdinando Acton, 55, 80133 Napoli, Italy
| |
Collapse
|
24
|
High P, Guernsey C, Subramanian S, Jacob J, Carmon KS. The Evolving Paradigm of Antibody-Drug Conjugates Targeting the ErbB/HER Family of Receptor Tyrosine Kinases. Pharmaceutics 2024; 16:890. [PMID: 39065587 PMCID: PMC11279420 DOI: 10.3390/pharmaceutics16070890] [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: 06/14/2024] [Revised: 06/28/2024] [Accepted: 06/30/2024] [Indexed: 07/28/2024] Open
Abstract
Current therapies targeting the human epidermal growth factor receptor (HER) family, including monoclonal antibodies (mAbs) and tyrosine kinase inhibitors (TKIs), are limited by drug resistance and systemic toxicities. Antibody-drug conjugates (ADCs) are one of the most rapidly expanding classes of anti-cancer therapeutics with 13 presently approved by the FDA. Importantly, ADCs represent a promising therapeutic option with the potential to overcome traditional HER-targeted therapy resistance by delivering highly potent cytotoxins specifically to HER-overexpressing cancer cells and exerting both mAb- and payload-mediated antitumor efficacy. The clinical utility of HER-targeted ADCs is exemplified by the immense success of HER2-targeted ADCs including trastuzumab emtansine and trastuzumab deruxtecan. Still, strategies to improve upon existing HER2-targeted ADCs as well as the development of ADCs against other HER family members, particularly EGFR and HER3, are of great interest. To date, no HER4-targeting ADCs have been reported. In this review, we extensively detail clinical-stage EGFR-, HER2-, and HER3-targeting monospecific ADCs as well as novel clinical and pre-clinical bispecific ADCs (bsADCs) directed against this receptor family. We close by discussing nascent trends in the development of HER-targeting ADCs, including novel ADC payloads and HER ligand-targeted ADCs.
Collapse
Affiliation(s)
- Peyton High
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (P.H.); (C.G.); (S.S.); (J.J.)
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center and UTHealth Houston, Houston, TX 77030, USA
| | - Cara Guernsey
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (P.H.); (C.G.); (S.S.); (J.J.)
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center and UTHealth Houston, Houston, TX 77030, USA
| | - Shraddha Subramanian
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (P.H.); (C.G.); (S.S.); (J.J.)
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center and UTHealth Houston, Houston, TX 77030, USA
| | - Joan Jacob
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (P.H.); (C.G.); (S.S.); (J.J.)
| | - Kendra S. Carmon
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (P.H.); (C.G.); (S.S.); (J.J.)
| |
Collapse
|
25
|
Atmaca H, Ilhan S, Çamli Pulat Ç, Dundar BA, Zora M. Evaluation of Novel Spiro-pyrrolopyridazine Derivatives as Anticancer Compounds: In Vitro Selective Cytotoxicity, Induction of Apoptosis, EGFR Inhibitory Activity, and Molecular Docking Analysis. ACS OMEGA 2024; 9:23713-23723. [PMID: 38854531 PMCID: PMC11154717 DOI: 10.1021/acsomega.4c00794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/26/2024] [Accepted: 05/13/2024] [Indexed: 06/11/2024]
Abstract
Cancer, characterized by uncontrolled cell proliferation, remains a global health challenge. Despite advancements in cancer treatment, drug resistance and adverse effects on normal cells remain challenging. The epidermal growth factor receptor (EGFR), a transmembrane tyrosine kinase protein, is crucial in controlling cell proliferation and is implicated in various cancers. Here, the cytotoxic and apoptotic potential of 21 newly synthesized spiro-pyrrolopyridazine (SPP) derivatives was investigated on breast (MCF-7), lung (H69AR), and prostate (PC-3) cancer cells. XTT assay was used for cytotoxicity assessment. Flow cytometry and western blot (WB) analyses were conducted for apoptosis detection. Additionally, the EGFR inhibitory potential of these derivatives was evaluated via a homogeneous time-resolved fluorescence (HTRF) assay, and WB and molecular docking studies were conducted to analyze the binding affinities of SPP10 with EGFR. SPPs, especially SPP10, exhibit significant cytotoxicity across MCF-7, H69AR, and PC-3 cancer cells with IC50 values of 2.31 ± 0.3, 3.16 ± 0.8, and 4.2 ± 0.2 μM, respectively. Notably, SPP10 demonstrates selective cytotoxicity against cancer cells with a low impact on nontumorigenic cells (IC50 value: 26.8 ± 0.4 μM). Flow cytometric analysis demonstrated the potent induction of apoptotic cell death by SPP10 in all of the tested cancer cells. Western blot analysis revealed the involvement of key apoptotic proteins, with SPP10 notably inhibiting antiapoptotic Bcl-2 while inducing pro-apoptotic Bax and cytochrome c. SPP10 exhibited significant EGFR kinase inhibitory activity, surpassing the efficacy of the reference drug erlotinib. Molecular docking studies support these findings, revealing strong binding affinities of SPP10 with both wild-type and mutated EGFR. The study underscores the significance of heterocyclic compounds, particularly spiro-class heterocyclic molecules, in advancing cancer research. Overall, SPP10 emerges as a promising candidate for further investigations in cancer treatment, combining potent cytotoxicity, apoptotic induction, and targeted EGFR inhibition.
Collapse
Affiliation(s)
- Harika Atmaca
- Department
of Biology, Faculty of Engineering and Natural Sciences, Manisa Celal Bayar University, Manisa 45140, Turkey
| | - Suleyman Ilhan
- Department
of Biology, Faculty of Engineering and Natural Sciences, Manisa Celal Bayar University, Manisa 45140, Turkey
| | - Çisil Çamli Pulat
- Applied
Science Research Center, Manisa Celal Bayar
University, Manisa 45140, Turkey
| | - Buse Aysen Dundar
- Department
of Chemistry, Middle East Technical University, Ankara 06800, Turkey
| | - Metin Zora
- Department
of Chemistry, Middle East Technical University, Ankara 06800, Turkey
| |
Collapse
|
26
|
Choudhury SD, Kumar P, Choudhury D. Bioactive nutraceuticals as G4 stabilizers: potential cancer prevention and therapy-a critical review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3585-3616. [PMID: 38019298 DOI: 10.1007/s00210-023-02857-z] [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: 08/14/2023] [Accepted: 11/13/2023] [Indexed: 11/30/2023]
Abstract
G-quadruplexes (G4) are non-canonical, four-stranded, nucleic acid secondary structures formed in the guanine-rich sequences, where guanine nucleotides associate with each other via Hoogsteen hydrogen bonding. These structures are widely found near the functional regions of the mammalian genome, such as telomeres, oncogenic promoters, and replication origins, and play crucial regulatory roles in replication and transcription. Destabilization of G4 by various carcinogenic agents allows oncogene overexpression and extension of telomeric ends resulting in dysregulation of cellular growth-promoting oncogenesis. Therefore, targeting and stabilizing these G4 structures with potential ligands could aid cancer prevention and therapy. The field of G-quadruplex targeting is relatively nascent, although many articles have demonstrated the effect of G4 stabilization on oncogenic expressions; however, no previous study has provided a comprehensive analysis about the potency of a wide variety of nutraceuticals and some of their derivatives in targeting G4 and the lattice of oncogenic cell signaling cascade affected by them. In this review, we have discussed bioactive G4-stabilizing nutraceuticals, their sources, mode of action, and their influence on cellular signaling, and we believe our insight would bring new light to the current status of the field and motivate researchers to explore this relatively poorly studied arena.
Collapse
Affiliation(s)
- Satabdi Datta Choudhury
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India
| | - Prateek Kumar
- School of Basic Sciences, Indian Institute of Technology (IIT), Mandi, Himachal Pradesh, 175005, India
| | - Diptiman Choudhury
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India.
- Centre for Excellence in Emerging Materials, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India.
| |
Collapse
|
27
|
Patil D, Raut S, Joshi M, Bhatt P, Bhatt LK. PAQR4 oncogene: a novel target for cancer therapy. Med Oncol 2024; 41:161. [PMID: 38767705 DOI: 10.1007/s12032-024-02382-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: 01/18/2024] [Accepted: 04/06/2024] [Indexed: 05/22/2024]
Abstract
Despite decades of basic and clinical research and trials of promising new therapies, cancer remains a major cause of morbidity and mortality due to the emergence of drug resistance to anticancer drugs. These resistance events have a very well-understood underlying mechanism, and their therapeutic relevance has long been recognized. Thus, drug resistance continues to be a major obstacle to providing cancer patients with the intended "cure". PAQR4 (Progestin and AdipoQ Receptor Family Member 4) gene is a recently identified novel protein-coding gene associated with various human cancers and acts through different signaling pathways. PAQR4 has a significant influence on multiple proteins that may regulate various gene expressions and may develop chemoresistance. This review discusses the roles of PAQR4 in tumor immunity, carcinogenesis, and chemoresistance. This paper is the first review, discussing PAQR4 in the pathogenesis of cancer. The review further explores the PAQR4 as a potential target in various malignancies.
Collapse
Affiliation(s)
- Dipti Patil
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India
| | - Swapnil Raut
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India
| | - Mitesh Joshi
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's NMIMS (Deemed-to-be University), Vile Parle (West), Mumbai, India
| | - Purvi Bhatt
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's NMIMS (Deemed-to-be University), Vile Parle (West), Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India.
| |
Collapse
|
28
|
Shui S, Scheller L, Correia BE. Protein-based bandpass filters for controlling cellular signaling with chemical inputs. Nat Chem Biol 2024; 20:586-593. [PMID: 37957273 PMCID: PMC11062894 DOI: 10.1038/s41589-023-01463-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 09/29/2023] [Indexed: 11/15/2023]
Abstract
Biological signal processing is vital for cellular function. Similar to electronic circuits, cells process signals via integrated mechanisms. In electronics, bandpass filters transmit frequencies with defined ranges, but protein-based counterparts for controlled responses are lacking in engineered biological systems. Here, we rationally design protein-based, chemically responsive bandpass filters (CBPs) showing OFF-ON-OFF patterns that respond to chemical concentrations within a specific range and reject concentrations outside that range. Employing structure-based strategies, we designed a heterodimeric construct that dimerizes in response to low concentrations of a small molecule (ON), and dissociates at high concentrations of the same molecule (OFF). The CBPs have a multidomain architecture in which we used known drug receptors, a computationally designed protein binder and small-molecule inhibitors. This modular system allows fine-tuning for optimal performance in terms of bandwidth, response, cutoff and fold changes. The CBPs were used to regulate cell surface receptor signaling pathways to control cellular activities in engineered cells.
Collapse
Affiliation(s)
- Sailan Shui
- Laboratory of Protein Design and Immunoengineering (LPDI)-STI-EPFL, Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Leo Scheller
- Laboratory of Protein Design and Immunoengineering (LPDI)-STI-EPFL, Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Bruno E Correia
- Laboratory of Protein Design and Immunoengineering (LPDI)-STI-EPFL, Lausanne, Switzerland.
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland.
| |
Collapse
|
29
|
Rajiv Gandhi G, Sharanya CS, Jayanandan A, Haridas M, Edwin Hillary V, Rajiv Gandhi S, Sridharan G, Sivasubramanian R, Silva Vasconcelos AB, Montalvão MM, Antony Ceasar S, Sousa NFD, Scotti L, Scotti MT, Gurgel RQ, Quintans-Júnior LJ. Multitargeted molecular docking and dynamics simulation studies of flavonoids and volatile components from the peel of Citrus sinensis L. (Osbeck) against specific tumor protein markers. J Biomol Struct Dyn 2024; 42:3051-3080. [PMID: 37203996 DOI: 10.1080/07391102.2023.2212062] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 05/01/2023] [Indexed: 05/20/2023]
Abstract
Citrus sinensis (L.) Osbeck (Rutaceae), commonly known as the sweet orange, is a popular and widely consumed fruit with several medicinal properties. The present study aimed to perform the in silico screening of 18 flavonoids and eight volatile components from the peel of C. sinensis against apoptotic and inflammatory proteins, metalloprotease, and tumor suppressor markers. Flavonoids obtained higher probabilities than volatile components against selected anti-cancer drug targets. Hence, the data from the binding energies against the essential apoptotic and cell proliferation proteins substantiate that they may be promising compounds in developing effective candidates to block cell growth, proliferation, and induced cell death by activating the apoptotic pathway. Further, the binding stability of the selected targets and the corresponding molecules were analyzed by 100 ns molecular dynamics (MD) simulations. Chlorogenic acid has the most binding affinity against the important anti-cancer targets iNOS, MMP-9, and p53. The congruent binding mode to different drug targets focused on cancer shown by chlorogenic acid suggests that it may be a compound with significant therapeutic potential. Moreover, the binding energy predictions indicated that the compound had stable electrostatic and van der Waal energies. Thus, our data reinforce the medicinal importance of flavonoids from C. sinensis and expand the need for more studies, seeking to optimize results and amplify the impacts of further in vitro and in vivo studies. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Gopalsamy Rajiv Gandhi
- Division of Phytochemistry and Drug Design, Department of Biosciences, Rajagiri College of Social Sciences, Kalamassery, Kochi, India
| | - Chelankara Suresh Sharanya
- Division of Phytochemistry and Drug Design, Department of Biosciences, Rajagiri College of Social Sciences, Kalamassery, Kochi, India
| | - Abhithaj Jayanandan
- Department of Biotechnology and Microbiology, Dr. Janaki Ammal Campus, Kannur University, Thalassery, Kannur, India
| | - Madathilkovilakath Haridas
- Department of Biotechnology and Microbiology, Dr. Janaki Ammal Campus, Kannur University, Thalassery, Kannur, India
| | - Varghese Edwin Hillary
- Division of Plant Molecular Biology and Biotechnology, Department of Biosciences, Rajagiri College of Social Sciences, Kalamassery, Kochi, India
| | - Sathiyabama Rajiv Gandhi
- Laboratory of Neuroscience and Pharmacological Assays (LANEF), Department of Physiology (DFS), Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
- Postgraduate Program of Health Sciences (PPGCS), University Hospital, Federal University of Sergipe (HU-UFS), Aracaju, Sergipe, Brazil
| | - Gurunagarajan Sridharan
- Department of Biochemistry, Srimad Andavan Arts and Science College (Autonomous), Affiliated to Bharathidasan University, Tiruchirapalli, India
| | - Rengaraju Sivasubramanian
- Department of Biochemistry, Srimad Andavan Arts and Science College (Autonomous), Affiliated to Bharathidasan University, Tiruchirapalli, India
| | - Alan Bruno Silva Vasconcelos
- Postgraduate Program of Physiological Sciences (PROCFIS), Federal University of Sergipe (UFS), São Cristóvão, Sergipe, Brazil
| | - Monalisa Martins Montalvão
- Postgraduate Program of Health Sciences (PPGCS), University Hospital, Federal University of Sergipe (HU-UFS), Aracaju, Sergipe, Brazil
| | - Stanislaus Antony Ceasar
- Division of Plant Molecular Biology and Biotechnology, Department of Biosciences, Rajagiri College of Social Sciences, Kalamassery, Kochi, India
| | - Natália Ferreira de Sousa
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, Paraíba, Brazil
| | - Luciana Scotti
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, Paraíba, Brazil
| | - Marcus Tullius Scotti
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, Paraíba, Brazil
| | - Ricardo Queiroz Gurgel
- Postgraduate Program of Health Sciences (PPGCS), University Hospital, Federal University of Sergipe (HU-UFS), Aracaju, Sergipe, Brazil
| | - Lucindo José Quintans-Júnior
- Laboratory of Neuroscience and Pharmacological Assays (LANEF), Department of Physiology (DFS), Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
- Postgraduate Program of Health Sciences (PPGCS), University Hospital, Federal University of Sergipe (HU-UFS), Aracaju, Sergipe, Brazil
| |
Collapse
|
30
|
Chatzikalil E, Roka K, Diamantopoulos PT, Rigatou E, Avgerinou G, Kattamis A, Solomou EE. Venetoclax Combination Treatment of Acute Myeloid Leukemia in Adolescents and Young Adult Patients. J Clin Med 2024; 13:2046. [PMID: 38610812 PMCID: PMC11012941 DOI: 10.3390/jcm13072046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 03/25/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
Over the past two decades, the prognosis in adolescents and young adults (AYAs) diagnosed with acute myeloid leukemia (AML) has significantly improved. The standard intensive cytotoxic treatment approach for AYAs with AML, consisting of induction chemotherapy with anthracycline/cytarabine combination followed by consolidation chemotherapy or stem cell transplantation, has lately been shifting toward novel targeted therapies, mostly in the fields of clinical trials. One of the most recent advances in treating AML is the combination of the B-cell lymphoma 2 (Bcl-2) inhibitor venetoclax with hypomethylating agents, which has been studied in elderly populations and was approved by the Food and Drug Administration (FDA) for patients over 75 years of age or patients excluded from intensive chemotherapy induction schemas due to comorbidities. Regarding the AYA population, venetoclax combination therapy could be a therapeutic option for patients with refractory/relapsed (R/R) AML, although data from real-world studies are currently limited. Venetoclax is frequently used by AYAs diagnosed with advanced hematologic malignancies, mainly acute lymphoblastic leukemia and myelodysplastic syndromes, as a salvage therapeutic option with considerable efficacy and safety. Herein, we aim to summarize the evidence obtained from clinical trials and observational studies on venetoclax use in AYAs with AML. Based on the available evidence, venetoclax is a safe and effective therapeutic option for R/R AML AYA patients. However, further research in larger cohorts is needed to confirm these data, establishing the benefits of a venetoclax-based regimen for this special population.
Collapse
Affiliation(s)
- Elena Chatzikalil
- Division of Pediatric Hematology-Oncology, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece; (E.C.); (K.R.); (E.R.); (G.A.); (A.K.)
- “Aghia Sofia” Children’s Hospital ERN-PeadCan Center, 11527 Athens, Greece
| | - Kleoniki Roka
- Division of Pediatric Hematology-Oncology, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece; (E.C.); (K.R.); (E.R.); (G.A.); (A.K.)
- “Aghia Sofia” Children’s Hospital ERN-PeadCan Center, 11527 Athens, Greece
| | - Panagiotis T. Diamantopoulos
- First Department of Internal Medicine, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece;
| | - Efthymia Rigatou
- Division of Pediatric Hematology-Oncology, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece; (E.C.); (K.R.); (E.R.); (G.A.); (A.K.)
- “Aghia Sofia” Children’s Hospital ERN-PeadCan Center, 11527 Athens, Greece
| | - Georgia Avgerinou
- Division of Pediatric Hematology-Oncology, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece; (E.C.); (K.R.); (E.R.); (G.A.); (A.K.)
- “Aghia Sofia” Children’s Hospital ERN-PeadCan Center, 11527 Athens, Greece
| | - Antonis Kattamis
- Division of Pediatric Hematology-Oncology, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece; (E.C.); (K.R.); (E.R.); (G.A.); (A.K.)
- “Aghia Sofia” Children’s Hospital ERN-PeadCan Center, 11527 Athens, Greece
| | - Elena E. Solomou
- Department of Internal Medicine, University of Patras Medical School, 26500 Rion, Greece
| |
Collapse
|
31
|
Nayak D, Lv D, Yuan Y, Zhang P, Hu W, Nayak A, Ruben EA, Lv Z, Sung P, Hromas R, Zheng G, Zhou D, Olsen SK. Development and crystal structures of a potent second-generation dual degrader of BCL-2 and BCL-xL. Nat Commun 2024; 15:2743. [PMID: 38548768 PMCID: PMC10979003 DOI: 10.1038/s41467-024-46922-4] [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: 02/24/2023] [Accepted: 03/14/2024] [Indexed: 04/01/2024] Open
Abstract
Overexpression of BCL-xL and BCL-2 play key roles in tumorigenesis and cancer drug resistance. Advances in PROTAC technology facilitated recent development of the first BCL-xL/BCL-2 dual degrader, 753b, a VHL-based degrader with improved potency and reduced toxicity compared to previous small molecule inhibitors. Here, we determine crystal structures of VHL/753b/BCL-xL and VHL/753b/BCL-2 ternary complexes. The two ternary complexes exhibit markedly different architectures that are accompanied by distinct networks of interactions at the VHL/753b-linker/target interfaces. The importance of these interfacial contacts is validated via functional analysis and informed subsequent rational and structure-guided design focused on the 753b linker and BCL-2/BCL-xL warhead. This results in the design of a degrader, WH244, with enhanced potency to degrade BCL-xL/BCL-2 in cells. Using biophysical assays followed by in cell activities, we are able to explain the enhanced target degradation of BCL-xL/BCL-2 in cells. Most PROTACs are empirically designed and lack structural studies, making it challenging to understand their modes of action and specificity. Our work presents a streamlined approach that combines rational design and structure-based insights backed with cell-based studies to develop effective PROTAC-based cancer therapeutics.
Collapse
Affiliation(s)
- Digant Nayak
- Department of Biochemistry & Structural Biology and Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Dongwen Lv
- Department of Biochemistry & Structural Biology and Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Yaxia Yuan
- Department of Biochemistry & Structural Biology and Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Peiyi Zhang
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Wanyi Hu
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Anindita Nayak
- Department of Biochemistry & Structural Biology and Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Eliza A Ruben
- Department of Biochemistry & Structural Biology and Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Zongyang Lv
- Department of Biochemistry & Structural Biology and Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Patrick Sung
- Department of Biochemistry & Structural Biology and Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Robert Hromas
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Guangrong Zheng
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA.
| | - Daohong Zhou
- Department of Biochemistry & Structural Biology and Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
| | - Shaun K Olsen
- Department of Biochemistry & Structural Biology and Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
32
|
Saladino GM, Brodin B, Kakadiya R, Toprak MS, Hertz HM. Iterative nanoparticle bioengineering enabled by x-ray fluorescence imaging. SCIENCE ADVANCES 2024; 10:eadl2267. [PMID: 38517973 DOI: 10.1126/sciadv.adl2267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 02/16/2024] [Indexed: 03/24/2024]
Abstract
Nanoparticles (NPs) are currently developed for drug delivery and molecular imaging. However, they often get intercepted before reaching their target, leading to low targeting efficacy and signal-to-noise ratio. They tend to accumulate in organs like lungs, liver, kidneys, and spleen. The remedy is to iteratively engineer NP surface properties and administration strategies, presently a time-consuming process that includes organ dissection at different time points. To improve this, we propose a rapid iterative approach using whole-animal x-ray fluorescence (XRF) imaging to systematically evaluate NP distribution in vivo. We applied this method to molybdenum-based NPs and clodronate liposomes for tumor targeting with transient macrophage depletion, leading to reduced accumulations in lungs and liver and eventual tumor detection. XRF computed tomography (XFCT) provided 3D insight into NP distribution within the tumor. We validated the results using a multiscale imaging approach with dye-doped NPs and gene expression analysis for nanotoxicological profiling. XRF imaging holds potential for advancing therapeutics and diagnostics in preclinical pharmacokinetic studies.
Collapse
Affiliation(s)
- Giovanni M Saladino
- Department of Applied Physics, Biomedical and X-Ray Physics, KTH Royal Institute of Technology, SE 10691, Stockholm, Sweden
| | - Bertha Brodin
- Department of Applied Physics, Biomedical and X-Ray Physics, KTH Royal Institute of Technology, SE 10691, Stockholm, Sweden
| | - Ronak Kakadiya
- Department of Applied Physics, Biomedical and X-Ray Physics, KTH Royal Institute of Technology, SE 10691, Stockholm, Sweden
| | - Muhammet S Toprak
- Department of Applied Physics, Biomedical and X-Ray Physics, KTH Royal Institute of Technology, SE 10691, Stockholm, Sweden
| | - Hans M Hertz
- Department of Applied Physics, Biomedical and X-Ray Physics, KTH Royal Institute of Technology, SE 10691, Stockholm, Sweden
| |
Collapse
|
33
|
Egbulefu C, Black K, Su X, Karmakar P, Habimana-Griffin L, Sudlow G, Prior J, Chukwu E, Zheleznyak A, Xu B, Xu Y, Esser A, Mixdorf M, Moss E, Manion B, Reed N, Gubin M, Lin CY, Schreiber R, Weilbaecher K, Achilefu S. Induction of complementary immunogenic necroptosis and apoptosis cell death pathways inhibits cancer metastasis and relapse. RESEARCH SQUARE 2024:rs.3.rs-3992212. [PMID: 38558990 PMCID: PMC10980095 DOI: 10.21203/rs.3.rs-3992212/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Interactions of light-sensitive drugs and materials with Cerenkov radiation-emitting radiopharmaceuticals generate cytotoxic reactive oxygen species (ROS) to inhibit localized and disseminated cancer progression, but the cell death mechanisms underlying this radionuclide stimulated dynamic therapy (RaST) remain elusive. Using ROS-regenerative nanophotosensitizers coated with a tumor-targeting transferrin-titanocene complex (TiO2-TC-Tf) and radiolabeled 2-fluorodeoxyglucose (18FDG), we found that adherent dying cells maintained metabolic activity with increased membrane permeabilization. Mechanistic assessment of these cells revealed that RaST activated the expression of RIPK-1 and RIPK-3, which mediate necroptosis cell death. Subsequent recruitment of the nuclear factors kappa B and the executioner mixed lineage kinase domain-like pseudo kinase (MLKL) triggered plasma membrane permeabilization and pore formation, respectively, followed by the release of cytokines and immunogenic damage-associated molecular patterns (DAMPs). In immune-deficient breast cancer models with adequate stroma and growth factors that recapitulate the human tumor microenvironment, RaST failed to inhibit tumor progression and the ensuing lung metastasis. A similar aggressive tumor model in immunocompetent mice responded to RaST, achieving a remarkable partial response (PR) and complete response (CR) with no evidence of lung metastasis, suggesting active immune system engagement. RaST recruited antitumor CD11b+, CD11c+, and CD8b+ effector immune cells after initiating dual immunogenic apoptosis and necroptosis cell death pathways in responding tumors in vivo. Over time, cancer cells upregulated the expression of negative immune regulating cytokine (TGF-β) and soluble immune checkpoints (sICP) to challenge RaST effect in the CR mice. Using a signal-amplifying cancer-imaging agent, LS301, we identified latent minimal residual disseminated tumors in the lymph nodes (LNs) of the CR group. Despite increased protumor immunogens in the CR mice, RaST prevented cancer relapse and metastasis through dynamic redistribution of ROS-regenerative TiO2 from bones at the early treatment stage to the spleen and LNs, maintaining active immunity against cancer progression and migration. This study reveals the immune-mechanistic underpinnings of RaST-mediated antitumor immune response and highlights immunogenic reprogramming of tumors in response to RaST. Overcoming apoptosis resistance through complementary necroptosis activation paves the way for strategic drug combinations to improve cancer treatment.
Collapse
Affiliation(s)
- Christopher Egbulefu
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
- Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75235-9397, USA
| | - Kvar Black
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
| | - Xinming Su
- Department of Medicine, Washington University in St. Louis, MO 63110, USA
| | - Partha Karmakar
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
| | | | - Gail Sudlow
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
| | - Julie Prior
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
| | - Ezeugo Chukwu
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
| | - Alex Zheleznyak
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
| | - Baogang Xu
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
| | - Yalin Xu
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
| | - Alison Esser
- Department of Medicine, Washington University in St. Louis, MO 63110, USA
| | - Matthew Mixdorf
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
| | - Evan Moss
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
| | - Brad Manion
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
| | - Nathan Reed
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
| | - Matthew Gubin
- Department of Pathology and Immunology, Washington University in St. Louis, MO 63110, USA
| | - Chieh-Yu Lin
- Department of Pathology and Immunology, Washington University in St. Louis, MO 63110, USA
| | - Robert Schreiber
- Department of Pathology and Immunology, Washington University in St. Louis, MO 63110, USA
| | | | - Samuel Achilefu
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
- Department of Medicine, Washington University in St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, MO 63110, USA
- Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75235-9397, USA
| |
Collapse
|
34
|
Chi H, Su L, Yan Y, Gu X, Su K, Li H, Yu L, Liu J, Wang J, Wu Q, Yang G. Illuminating the immunological landscape: mitochondrial gene defects in pancreatic cancer through a multiomics lens. Front Immunol 2024; 15:1375143. [PMID: 38510247 PMCID: PMC10953916 DOI: 10.3389/fimmu.2024.1375143] [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: 01/23/2024] [Accepted: 02/16/2024] [Indexed: 03/22/2024] Open
Abstract
This comprehensive review delves into the complex interplay between mitochondrial gene defects and pancreatic cancer pathogenesis through a multiomics approach. By amalgamating data from genomic, transcriptomic, proteomic, and metabolomic studies, we dissected the mechanisms by which mitochondrial genetic variations dictate cancer progression. Emphasis has been placed on the roles of these genes in altering cellular metabolic processes, signal transduction pathways, and immune system interactions. We further explored how these findings could refine therapeutic interventions, with a particular focus on precision medicine applications. This analysis not only fills pivotal knowledge gaps about mitochondrial anomalies in pancreatic cancer but also paves the way for future investigations into personalized therapy options. This finding underscores the crucial nexus between mitochondrial genetics and oncological immunology, opening new avenues for targeted cancer treatment strategies.
Collapse
Affiliation(s)
- Hao Chi
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Lanqian Su
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Yalan Yan
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Xiang Gu
- Biology Department, Southern Methodist University, Dallas, TX, United States
| | - Ke Su
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Han Li
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Lili Yu
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
| | - Jie Liu
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China
| | - Jue Wang
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
| | - Qibiao Wu
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
| | - Guanhu Yang
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
- Department of Specialty Medicine, Ohio University, Athens, OH, United States
| |
Collapse
|
35
|
Lee D, V AADLR, Kim Y. Optimal strategies of oncolytic virus-bortezomib therapy via the apoptotic, necroptotic, and oncolysis signaling network. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2024; 21:3876-3909. [PMID: 38549312 DOI: 10.3934/mbe.2024173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Bortezomib and oncolytic virotherapy are two emerging targeted cancer therapies. Bortezomib, a proteasome inhibitor, disrupts protein degradation in cells, leading to the accumulation of unfolded proteins that induce apoptosis. On the other hand, virotherapy uses genetically modified oncolytic viruses (OVs) to infect cancer cells, trigger cell lysis, and activate anti-tumor response. Despite progress in cancer treatment, identifying administration protocols for therapeutic agents remains a significant concern, aiming to strike a balance between efficacy, minimizing toxicity, and administrative costs. In this work, optimal control theory was employed to design a cost-effective and efficient co-administration protocols for bortezomib and OVs that could significantly diminish the population of cancer cells via the cell death program with the NF$ \kappa $B-BAX-RIP1 signaling network. Both linear and quadratic control strategies were explored to obtain practical treatment approaches by adapting necroptosis protocols to efficient cell death programs. Our findings demonstrated that a combination therapy commencing with the administration of OVs followed by bortezomib infusions yields an effective tumor-killing outcome. These results could provide valuable guidance for the development of clinical administration protocols in cancer treatment.
Collapse
Affiliation(s)
- Donggu Lee
- Department of Mathematics, Konkuk University, Seoul, Republic of Korea
| | - Aurelio A de Los Reyes V
- Institute of Mathematics, University of the Philippines Diliman, Quezon City 1101, Philippines
- Biomedical Mathematics Group, Pioneer Research Center for Mathematical and Computational Sciences, Institute for Basic Science, Daejeon 34126, Republic of Korea
| | - Yangjin Kim
- Department of Mathematics, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
36
|
Ahmadi M, Ritter CA, von Woedtke T, Bekeschus S, Wende K. Package delivered: folate receptor-mediated transporters in cancer therapy and diagnosis. Chem Sci 2024; 15:1966-2006. [PMID: 38332833 PMCID: PMC10848714 DOI: 10.1039/d3sc05539f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/31/2023] [Indexed: 02/10/2024] Open
Abstract
Neoplasias pose a significant threat to aging society, underscoring the urgent need to overcome the limitations of traditional chemotherapy through pioneering strategies. Targeted drug delivery is an evolving frontier in cancer therapy, aiming to enhance treatment efficacy while mitigating undesirable side effects. One promising avenue utilizes cell membrane receptors like the folate receptor to guide drug transporters precisely to malignant cells. Based on the cellular folate receptor as a cancer cell hallmark, targeted nanocarriers and small molecule-drug conjugates have been developed that comprise different (bio) chemistries and/or mechanical properties with individual advantages and challenges. Such modern folic acid-conjugated stimuli-responsive drug transporters provide systemic drug delivery and controlled release, enabling reduced dosages, circumvention of drug resistance, and diminished adverse effects. Since the drug transporters' structure-based de novo design is increasingly relevant for precision cancer remediation and diagnosis, this review seeks to collect and debate the recent approaches to deliver therapeutics or diagnostics based on folic acid conjugated Trojan Horses and to facilitate the understanding of the relevant chemistry and biochemical pathways. Focusing exemplarily on brain and breast cancer, recent advances spanning 2017 to 2023 in conjugated nanocarriers and small molecule drug conjugates were considered, evaluating the chemical and biological aspects in order to improve accessibility to the field and to bridge chemical and biomedical points of view ultimately guiding future research in FR-targeted cancer therapy and diagnosis.
Collapse
Affiliation(s)
- Mohsen Ahmadi
- Leibniz Institute for Plasma Science and Technology (INP), Center for Innovation Competence (ZIK) Plasmatis Felix Hausdorff-Str. 2 17489 Greifswald Germany
| | - Christoph A Ritter
- Institute of Pharmacy, Section Clinical Pharmacy, University of Greifswald Greifswald Germany
| | - Thomas von Woedtke
- Leibniz Institute for Plasma Science and Technology (INP), Center for Innovation Competence (ZIK) Plasmatis Felix Hausdorff-Str. 2 17489 Greifswald Germany
- Institute for Hygiene and Environmental Medicine, Greifswald University Medical Center Ferdinand-Sauerbruch-Straße 17475 Greifswald Germany
| | - Sander Bekeschus
- Leibniz Institute for Plasma Science and Technology (INP), Center for Innovation Competence (ZIK) Plasmatis Felix Hausdorff-Str. 2 17489 Greifswald Germany
- Clinic and Policlinic for Dermatology and Venereology, Rostock University Medical Center Strempelstr. 13 18057 Rostock Germany
| | - Kristian Wende
- Leibniz Institute for Plasma Science and Technology (INP), Center for Innovation Competence (ZIK) Plasmatis Felix Hausdorff-Str. 2 17489 Greifswald Germany
| |
Collapse
|
37
|
Ibrahim IAA, Alzahrani AR, Alanazi IM, Shahzad N, Shahid I, Falemban AH, Nur Azlina MF, Arulselvan P. Synthesis and Characterization of Graphene Oxide/Polyethylene Glycol/Folic Acid/Brucine Nanocomposites and Their Anticancer Activity on HepG2 Cells. Int J Nanomedicine 2024; 19:1109-1124. [PMID: 38344441 PMCID: PMC10854405 DOI: 10.2147/ijn.s445206] [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: 10/17/2023] [Accepted: 01/19/2024] [Indexed: 02/15/2024] Open
Abstract
Background Liver cancer is the sixth most prevalent form of cancer and the second major cause of cancer-associated mortalities worldwide. Cancer nanotechnology has the ability to fundamentally alter cancer treatment, diagnosis, and detection. Objective In this study, we explained the development of graphene oxide/polyethylene glycol/folic acid/brucine nanocomposites (GO/PEG/Bru-FA NCs) and evaluated their antimicrobial and anticancer effect on the liver cancer HepG2 cells. Methodology The GO/PEG/Bru-FA NCs were prepared using the co-precipitation technique and characterized using various techniques. The cytotoxicity of the GO/PEG/Bru-FA NCs was tested against both liver cancer HepG2 and non-malignant Vero cells using an MTT assay. The antimicrobial activity of the GO/PEG/Bru-FA NCs was tested against several pathogens using the well diffusion technique. The effects of GO/PEG/Bru-FA NCs on endogenous ROS accumulation, apoptosis, and MMP levels were examined using corresponding fluorescent staining assays, respectively. The apoptotic protein expressions, such as Bax, Bcl-2, and caspases, were studied using the corresponding kits. Results The findings of various characterization assays revealed the development of GO/PEG/Bru-FA NCs with face-centered spherical morphology and an agglomerated appearance with an average size of 197.40 nm. The GO/PEG/Bru-FA NCs treatment remarkably inhibited the growth of the tested pathogens. The findings of the MTT assay evidenced that the GO/PEG/Bru-FA NCs effectively reduced the HepG2 cell growth while not showing toxicity to the Vero cells. The findings of the fluorescent assay proved that the GO/PEG/Bru-FA NCs increased ROS generation, reduced MMP levels, and promoted apoptosis in the HepG2 cells. The levels of Bax, caspase-9, and -3 were increased, and Bcl-2 was reduced in the GO/PEG/Bru-FA NCs-treated HepG2 cells. Conclusion The results of this work demonstrate that GO/PEG/Bru-FA NCs suppress viability and induce apoptosis in HepG2 cells, indicating their potential as an anticancer candidate.
Collapse
Affiliation(s)
- Ibrahim Abdel Aziz Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura university, Makkah, Saudi Arabia
| | - Abdullah R Alzahrani
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura university, Makkah, Saudi Arabia
| | - Ibrahim M Alanazi
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura university, Makkah, Saudi Arabia
| | - Naiyer Shahzad
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura university, Makkah, Saudi Arabia
| | - Imran Shahid
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura university, Makkah, Saudi Arabia
| | - Alaa Hisham Falemban
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura university, Makkah, Saudi Arabia
| | - Mohd Fahami Nur Azlina
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan, Bangi, Selangor, Malaysia
| | - Palanisamy Arulselvan
- Department of Chemistry, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu, 602 105, India
| |
Collapse
|
38
|
Yu Y, Baral S, Sun Q, Ding J, Zhang Q, Zhao F, Gao S, Yao Q, Yu H, Liu B, Wang D. PLCD3 inhibits apoptosis and promotes proliferation, invasion and migration in gastric cancer. Discov Oncol 2024; 15:26. [PMID: 38305998 PMCID: PMC10837395 DOI: 10.1007/s12672-024-00881-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 01/31/2024] [Indexed: 02/03/2024] Open
Abstract
Gastric cancer (GC) is a heterogeneous disease whose development is accompanied by alterations in a variety of pathogenic genes. The phospholipase C Delta 3 enzyme is a member of the phospholipase C family, which controls substance transport between cells in the body. However, its role in gastric cancer has not been discovered. The purpose of this study was to investigate the expression and mechanism of action of PLCD3 in connection to gastric cancer. By Western blot analysis and immunohistochemistry, PLCD3 mRNA and protein expression levels were measured, with high PLCD3 expression suggesting poor prognosis. In N87 and HGC-27 cells, the silencing of PLCD3 using small interfering RNA effectively induced apoptosis and inhibited tumor cell proliferation, invasion, and migration. Conversely, overexpression of PLCD3 using overexpressed plasmids inhibited apoptosis in AGS and BGC-823 cells and promoted proliferation, migration, and invasion. In order to investigate the underlying mechanisms, we conducted further analysis of PLCD3, which indicates that this protein is closely related to the cell cycle and EMT. Additionally, we found that overexpression of PLCD3 inhibits apoptosis and promotes the development of GC cells through JAK2/STAT3 signaling. In conclusion, PLCD3 inhibits apoptosis and promotes proliferation, invasion, and migration, which indicated that PLCD3 might serve as a therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Yantao Yu
- Dalian Medical University, Dalian, 116044, Liaoning, China
- Yangzhou Clinical Medical College, Dalian Medical University, Yangzhou, 225001, Jiangsu, China
| | - Shantanu Baral
- Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, Jiangsu, China
- Yangzhou Key Laboratory of Basic and Clinical Translation of Gastroenterology/Metabolic Diseases, Yangzhou, 225001, Jiangsu, China
| | - Qiannan Sun
- Northern Jiangsu People's Hospital, Yangzhou, 225001, Jiangsu, China
- Medical Research Center of Northern Jiangsu People's Hospital, Yangzhou, 225001, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, Jiangsu, China
- Yangzhou Key Laboratory of Basic and Clinical Translation of Gastroenterology/Metabolic Diseases, Yangzhou, 225001, Jiangsu, China
| | - Jianyue Ding
- Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Qi Zhang
- Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu, China
- Northern Jiangsu People's Hospital, Yangzhou, 225001, Jiangsu, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, Jiangsu, China
- Yangzhou Key Laboratory of Basic and Clinical Translation of Gastroenterology/Metabolic Diseases, Yangzhou, 225001, Jiangsu, China
| | - Fanyu Zhao
- Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Shuyang Gao
- Dalian Medical University, Dalian, 116044, Liaoning, China
- Yangzhou Clinical Medical College, Dalian Medical University, Yangzhou, 225001, Jiangsu, China
| | - Qing Yao
- Dalian Medical University, Dalian, 116044, Liaoning, China
- Yangzhou Clinical Medical College, Dalian Medical University, Yangzhou, 225001, Jiangsu, China
| | - Haoyue Yu
- Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Bin Liu
- Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu, China.
- Northern Jiangsu People's Hospital, Yangzhou, 225001, Jiangsu, China.
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, Jiangsu, China.
- Yangzhou Key Laboratory of Basic and Clinical Translation of Gastroenterology/Metabolic Diseases, Yangzhou, 225001, Jiangsu, China.
| | - Daorong Wang
- Yangzhou Clinical Medical College, Dalian Medical University, Yangzhou, 225001, Jiangsu, China.
- Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu, China.
- Northern Jiangsu People's Hospital, Yangzhou, 225001, Jiangsu, China.
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, Jiangsu, China.
- Yangzhou Key Laboratory of Basic and Clinical Translation of Gastroenterology/Metabolic Diseases, Yangzhou, 225001, Jiangsu, China.
| |
Collapse
|
39
|
Du S, Cao K, Yan Y, Wang Y, Wang Z, Lin D. Developments and current status of cell-free DNA in the early detection and management of hepatocellular carcinoma. J Gastroenterol Hepatol 2024; 39:231-244. [PMID: 37990622 DOI: 10.1111/jgh.16416] [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: 07/25/2023] [Revised: 10/23/2023] [Accepted: 10/30/2023] [Indexed: 11/23/2023]
Abstract
Nowadays, hepatocellular carcinoma (HCC) is still a major threat to human health globally, with a disappointing prognosis. Regular monitoring of patients at high risk, utilizing abdominal ultrasonography combined with alpha-fetoprotein (AFP) serum analysis, enables the early detection of potentially treatable tumors. However, the approach has limitations due to its lack of sensitivity. Meanwhile, the current standard procedure for obtaining a tumor biopsy in cases of HCC is invasive and lacks the ability to assess the dynamic progression of cancer or account for tumor heterogeneity. Hence, there is a pressing need to develop non-invasive, highly sensitive biomarkers for HCC which can improve the accuracy of early diagnosis, assess treatment response and accurately predict the prognosis. In contrast to the conventional method of tissue biopsy, liquid biopsy offers a non-invasive approach that can be readily repeated. As a liquid biopsy approach, the analysis of cell-free DNA (cfDNA) offers real-time insights that can accurately portray the tumor burden and provide a comprehensive depiction of the genetic profile associated with HCC. In this review, we present a comprehensive summary of the recent research findings pertaining to the significance and potential practicality of cfDNA analysis in the early detection and effective management of HCC.
Collapse
Affiliation(s)
- Sihao Du
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ke Cao
- Department of General Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yadong Yan
- Department of General Surgery, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yupeng Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhenshun Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Dongdong Lin
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
40
|
Tripathi N, Fortuna GG, Gebrael G, Dal E, Mathew Thomas V, Gupta S, Swami U. Predictors of response to neoadjuvant therapy in urothelial cancer. Crit Rev Oncol Hematol 2024; 194:104236. [PMID: 38128631 DOI: 10.1016/j.critrevonc.2023.104236] [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: 05/22/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
Neoadjuvant cisplatin-based chemotherapy (NACC) followed by radical cystectomy is the standard treatment for localized muscle-invasive bladder cancer (MIBC). Patients who achieve a complete pathological response following NACC have better overall survival than those with residual disease. However, a subset of patients does not derive benefit from NACC while experiencing chemotherapy-related side effects that may delay cystectomy, which can be detrimental. There is a need for predictive and prognostic biomarkers to better stratify patients who will derive benefits from NACC. This review summarizes the currently available literature on various predictors of response to neoadjuvant chemotherapy. Covered predictors include clinical factors, treatment regimens (including chemotherapy and immunotherapy), histological predictors, and molecular predictors such as DNA repair genes, p53, FGFR3, ERBB2, Bcl-2, EMMPRIN, survivin, choline-phosphate cytidylyltransferase-α, epigenetic markers, immunological markers, other molecular predictors and gene expression profiling. Further, we elaborate on the potential role of neoadjuvant immunotherapy and the correlative biomarkers of response.
Collapse
Affiliation(s)
- Nishita Tripathi
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Gliceida Galarza Fortuna
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Georges Gebrael
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Emre Dal
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Vinay Mathew Thomas
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Sumati Gupta
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Umang Swami
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
41
|
Lakhani NJ, Rasco D, Wang H, Men L, Liang E, Fu T, Collins MC, Min P, Yin Y, Davids MS, Yang D, Zhai Y. First-in-Human Study with Preclinical Data of BCL-2/BCL-xL Inhibitor Pelcitoclax in Locally Advanced or Metastatic Solid Tumors. Clin Cancer Res 2024; 30:506-521. [PMID: 37971712 DOI: 10.1158/1078-0432.ccr-23-1525] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 09/15/2023] [Accepted: 11/14/2023] [Indexed: 11/19/2023]
Abstract
PURPOSE B-cell lymphoma-extra-large (BCL-xL) regulates apoptosis and is an attractive anticancer therapeutic target. However, BCL-xL inhibition also kills mature platelets, hampering clinical development. Using an innovative prodrug strategy, we have developed pelcitoclax (APG-1252), a potent, dual BCL-2 and BCL-xL inhibitor. Aims of this study were to characterize the antitumor activity and safety of pelcitoclax and explore its underlying mechanisms of action (MOA). PATIENTS AND METHODS Cell line-derived xenograft and patient-derived xenograft (PDX) models were tested to evaluate antitumor activity and elucidate MOA. Subjects (N = 50) with metastatic small-cell lung cancer and other solid tumors received intravenous pelcitoclax once or twice weekly. Primary outcome measures were safety and tolerability; preliminary efficacy (responses every 2 cycles per RECIST version 1.1) represented a secondary endpoint. RESULTS Pelcitoclax exhibited strong BAX/BAK‒dependent and caspase-mediated antiproliferative and apoptogenic activity in various cancer cell lines. Consistent with cell-based apoptogenic activity, pelcitoclax disrupted BCL-xL:BIM and BCL-xL:PUMA complexes in lung and gastric cancer PDX models. Levels of BCL-xL complexes correlated with tumor growth inhibition by pelcitoclax. Combined with taxanes, pelcitoclax enhanced antitumor activity by downregulating antiapoptotic protein myeloid cell leukemia-1 (MCL-1). Importantly, pelcitoclax was well tolerated and demonstrated preliminary therapeutic efficacy, with overall response and disease control rates of 6.5% and 30.4%, respectively. Most common treatment-related adverse events included transaminase elevations and reduced platelets that were less frequent with a once-weekly schedule. CONCLUSIONS Our data demonstrate that pelcitoclax has antitumor activity and is well tolerated, supporting its further clinical development for human solid tumors, particularly combined with agents that downregulate MCL-1.
Collapse
Affiliation(s)
| | | | - Hengbang Wang
- Ascentage Pharma (Suzhou) Co., Ltd., Suzhou, Jiangsu, China
| | - Lichuang Men
- Ascentage Pharma (Suzhou) Co., Ltd., Suzhou, Jiangsu, China
| | - Eric Liang
- Ascentage Pharma Group Inc., Rockville, Maryland
| | - Tommy Fu
- Ascentage Pharma Group Inc., Rockville, Maryland
| | - Mary C Collins
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ping Min
- Ascentage Pharma (Suzhou) Co., Ltd., Suzhou, Jiangsu, China
| | - Yan Yin
- Ascentage Pharma (Suzhou) Co., Ltd., Suzhou, Jiangsu, China
| | - Matthew S Davids
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Dajun Yang
- Ascentage Pharma (Suzhou) Co., Ltd., Suzhou, Jiangsu, China
- Ascentage Pharma Group Inc., Rockville, Maryland
- Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yifan Zhai
- Ascentage Pharma (Suzhou) Co., Ltd., Suzhou, Jiangsu, China
- Ascentage Pharma Group Inc., Rockville, Maryland
| |
Collapse
|
42
|
Kumar R, Afrin H, Bhatt HN, Beaven E, Gangavarap A, Esquivel SV, Zahid MI, Nurunnabi M. Mucoadhesive Carrier-Mediated Oral Co-delivery of Bcl2 Inhibitors Improves Gastric Cancer Treatment. ACS APPLIED MATERIALS & INTERFACES 2024; 16:305-317. [PMID: 38157479 DOI: 10.1021/acsami.3c15226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Gastric cancer treatment is challenging due to the lack of early-stage diagnostic technology and targeted delivery systems. Currently, the available treatments for gastric cancer are surgery, chemotherapy, immunotherapy, and radiation. These strategies are either invasive or require systemic delivery, exerting toxicities within healthy tissues. By creation of a targeted delivery system to the stomach, gastric cancer can be treated in the early stages. Such an approach reduces the negative effects on the rest of the body by minimizing systemic absorbance and random localization. With this in mind, we developed a mucoadhesive vehicle composed of β-Glucan And Docosahexaenoic Acid (GADA) for controlled drug/gene delivery. In the current study, we investigated the therapeutic effect of codelivery Bcl2 inhibitors navitoclax (NAVI) and siRNA (Bcl2) via oral using GADA. The therapeutic efficacy of the GADA-mediated oral NAVI/siRNA was investigated in a gastric cancer mouse model. Higher Bcl2 inhibition efficacy was observed in Western blotting and TUNEL assay in mice treated with GADA/NAVI/siRNA compared to free NAVI, siRNA, and NAVI/siRNA. Histology (H&E) and immunohistochemistry (Ki67, TUNEL, and BCl2) analyses confirmed a significant reduction of the tumor region. Interaction between GADA and mucus resulted in retention for over 6 h and thereby sustained local payload release. The developed oral carrier GADA is an emerging vehicle that has promising potential in oral delivery of both small and large molecules, and their mucoadhesive property results in improved therapeutic efficacy with minimal side effects compared to conventional treatment. This study opens a new window for the effective delivery of oral medicine for the treatment of gastric cancer and other gastrointestinal diseases.
Collapse
Affiliation(s)
- Raj Kumar
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
- Department of Biomedical Engineering, University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Humayra Afrin
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
- Environmental Science & Engineering, University of Texas at El Paso, El Paso, Texas 79965,United States
| | - Himanshu N Bhatt
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
- Department of Biomedical Engineering, University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Elfa Beaven
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
- Department of Biomedical Engineering, University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Anushareddy Gangavarap
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
| | - Stephanie V Esquivel
- Department of Aerospace & Mechanical Engineering, University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Md Ikhtiar Zahid
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
- Department of Biomedical Engineering, University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
- Department of Biomedical Engineering, University of Texas at El Paso, El Paso, Texas 79968, United States
- Environmental Science & Engineering, University of Texas at El Paso, El Paso, Texas 79965,United States
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas 79968, United States
| |
Collapse
|
43
|
Singh T, Bhattacharya M, Mavi AK, Gulati A, Rakesh, Sharma NK, Gaur S, Kumar U. Immunogenicity of cancer cells: An overview. Cell Signal 2024; 113:110952. [PMID: 38084844 DOI: 10.1016/j.cellsig.2023.110952] [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: 09/28/2023] [Revised: 10/25/2023] [Accepted: 10/25/2023] [Indexed: 12/18/2023]
Abstract
The immune system assumes a pivotal role in the organism's capacity to discern and obliterate malignant cells. The immunogenicity of a cancer cell pertains to its proficiency in inciting an immunological response. The prowess of immunogenicity stands as a pivotal determinant in the triumph of formulating immunotherapeutic methodologies. Immunotherapeutic strategies include immune checkpoint inhibitors, chimeric antigen receptor (CAR) T-cell therapy, and on vaccines. Immunogenic cell death (ICD) epitomizes a form of cellular demise that incites an immune response against dying cells. ICD is characterized by the liberation of distinct specific molecules that activate the immune system, thereby leading to the identification and elimination of dying cells by immunocytes. One of the salient characteristics inherent to the ICD phenomenon resides in the vigorous liberation of adenosine triphosphate (ATP) by cellular entities dedicated to embarking upon the process of programmed cell death, yet refraining from complete apoptotic demise. ICD is initiated by a sequence of molecular events that occur during cell death. These occurrences encompass the unveiling or discharge of molecules such as calreticulin, high-mobility group box 1 (HMGB1), and adenosine triphosphate (ATP) from dying cells. These molecules act as "eat me" signals, which are recognized by immune cells, thereby prompting the engulfment and deterioration of expiring cells by phagocytes including various pathways such as Necroptosis, Apoptosis, and pyroptosis. Here, we review our current understanding of the pathophysiological importance of the immune responses against dying cells and the mechanisms underlying their activation. Overall, the ICD represents an important mechanism by which the immune system recognizes and eliminates dying cells, including cancer cells. Understanding the molecular events that underlie ICD bears the potential to engender innovative cancer therapeutics that harness the power of the immune system to combat cancer.
Collapse
Affiliation(s)
- Tanya Singh
- Department of Microbiology, Ram Lal Anand College, University of Delhi, Delhi 110021, India
| | - Madhuri Bhattacharya
- Department of Microbiology, Ram Lal Anand College, University of Delhi, Delhi 110021, India
| | - Anil Kumar Mavi
- Department of Botany, Sri Aurobindo College, University of Delhi, Delhi 110017, India.
| | - Anita Gulati
- Department of Zoology, Deen Dayal Upadhyaya College, University of Delhi, Delhi 110078, India
| | - Rakesh
- Janki Devi Memorial College, University of Delhi, Delhi 110060, India
| | - Naresh Kumar Sharma
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sonal Gaur
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Umesh Kumar
- School of Biosciences, Institute of Management Studies Ghaziabad (University Courses Campus), NH9, Adhyatmik Nagar, Ghaziabad, Uttar Pradesh 201015, India.
| |
Collapse
|
44
|
Darvish M. LncRNA FTH1P3: A New Biomarker for Cancer-Related Therapeutic Development. Curr Mol Med 2024; 24:576-584. [PMID: 37491858 DOI: 10.2174/1566524023666230724141353] [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: 04/15/2023] [Revised: 06/17/2023] [Accepted: 06/22/2023] [Indexed: 07/27/2023]
Abstract
Cancer is a persistent and urgent health problem that affects the entire world. Not long ago, regulatory biomolecules referred to as long noncoding RNAs (lncRNAs) might have value for their innate abundance and stability. These single-stranded RNAs potentially interfere with several physiological and biochemical cellular processes involved in many human pathological situations, particularly cancer diseases. Ferritin heavy chain1 pseudogene 3 (FTH1P3), a lncRNA that is ubiquitously transcribed and belongs to the ferritin heavy chain (FHC) family, represents a novel class of lncRNAs primarily found in oral squamous cell carcinoma. Further research has shown that FTH1P3 is involved in other malignancies such as uveal melanoma, glioma, esophageal squamous cell carcinoma, non-small cell lung cancer, breast cancer, laryngeal squamous cell carcinoma, and cervical cancer. Accordingly, FTH1P3 significantly enhances cancer symptoms, including cell proliferation, invasion, metastasis, chemoresistance, and inhibition of apoptosis through many specific mechanisms. Notably, the clinical data significantly demonstrated the association of FTH1P3 overexpression with poor prognosis and poor overall survival within the examined samples. Here, we summarize all the research published to date (13 articles) on FTH1P3, focusing on the biological function underlying the regulatory mechanism and its possible clinical relevance.
Collapse
Affiliation(s)
- Maryam Darvish
- Department of Medical Biotechnology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| |
Collapse
|
45
|
Burkett WC, Zhao Z, Newton MA, Sun W, Deng B, Secord AA, Zhou C, Bae-Jump V. Ipatasertib, an oral AKT inhibitor, in combination with carboplatin exhibits anti-proliferative effects in uterine serous carcinoma. Ann Med 2023; 55:603-614. [PMID: 36773034 PMCID: PMC9930841 DOI: 10.1080/07853890.2023.2177883] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/18/2022] [Accepted: 02/03/2023] [Indexed: 02/12/2023] Open
Abstract
PURPOSE Uterine serous carcinoma (USC) exhibits worse survival rates compared to the endometrioid subtype, and there is currently no effective treatment options for recurrence of this disease after platinum-based chemotherapy. Activation of PIK3CA/AKT/mTOR signaling pathway is a common biological feature in USC. MATERIALS AND METHODS Ipatasertib (IPAT) is an investigational, orally administered, ATP-competitive, highly selective inhibitor of pan AKT that has demonstrated anti-proliferative activity in a variety of tumor cells and tumor models. In this study, we used IPAT, carboplatin and their combination to investigate the anti-tumor activity in SPEC-2 and ARK-1 cells. RESULTS Our results indicate that IPAT combined with carboplatin at low doses was more effective at reducing proliferation, inducing apoptosis and causing cellular stress than IPAT or carboplatin alone. In particular, inhibition of the PIK3CA/AKT/mTOR pathway and induction of DNA damage were involved in the synergistic inhibition by combination treatment of cell viability in USC cells treated with the combination. Furthermore, IPAT in combination with carboplatin significantly reduced cell adhesion and inhibited cell invasion. CONCLUSIONS These findings suggest that the combination of IPAT and carboplatin has potential clinical implications for developing new USC treatment strategies.
Collapse
Affiliation(s)
- Wesley C. Burkett
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, NC
| | - Ziyi Zhao
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, NC
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health care Hospital, Beijing, P. R. China
| | - Meredith A. Newton
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, NC
| | - Wenchuan Sun
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, NC
| | - Boer Deng
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, NC
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health care Hospital, Beijing, P. R. China
| | - Angeles Alvarez Secord
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Duke University, Durham, NC
| | - Chunxiao Zhou
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, NC
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, NC
| | - Victoria Bae-Jump
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, NC
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, NC
| |
Collapse
|
46
|
Abd Elhameed AA, Ali AR, Ghabbour HA, Bayomi SM, El-Gohary NS. Design, synthesis, and antitumor screening of new thiazole, thiazolopyrimidine, and thiazolotriazine derivatives as potent inhibitors of VEGFR-2. Drug Dev Res 2023; 84:1664-1698. [PMID: 37661648 DOI: 10.1002/ddr.22109] [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: 05/01/2023] [Revised: 07/09/2023] [Accepted: 08/05/2023] [Indexed: 09/05/2023]
Abstract
New thiazole, thiazolopyrimidine, and thiazolotriazine derivatives 3-12 and 14a-f were synthesized. The newly synthesized analogs were tested for in vitro antitumor activity against HepG2, HCT-116, MCF-7, HeP-2, and Hela cancer cells. Results indicated that compound 5 displayed the highest potency toward the tested cancer cells. Compound 11b possessed enhanced effectiveness over MCF-7, HepG2, HCT-116, and Hela cancer cells. In addition, compounds 4 and 6 showed promising activity toward HCT-116, MCF-7, and Hela cancer cells and eminent activity against HepG2 and HeP-2 cells. Moreover, compounds 3-6 and 11b were tested for their capability to inhibit vascular endothelial growth factor receptor-2 (VEGFR-2) activity. The obtained results showed that compound 5 displayed significant inhibitory activity against VEGFR-2 (half-maximal inhibitory concentration [IC50 ] = 0.044 μM) comparable to sunitinib (IC50 = 0.100 μM). Also, the synthesized compounds 3-6 and 11b were subjected to in vitro cytotoxicity tests over WI38 and WISH normal cells. It was found that the five tested compounds displayed significantly lower cytotoxicity than doxorubicin toward normal cell lines. Cell cycle analysis proved that compound 5 induces cell cycle arrest in the S phase for HCT-116 and Hela cancer cell lines and in the G2/M phase for the MCF-7 cancer cell line. Moreover, compound 5 induced cancer cell death through apoptosis accompanied by a high ratio of BAX/BCL-2 in the screened cancer cells. Furthermore, docking results revealed that compound 5 showed the essential interaction bonds with VEGFR-2, which agreed with in vitro enzyme assay results. In silico studies showed that most of the analyzed compounds complied with the requirements of good oral bioavailability with minimal toxicity threats in humans.
Collapse
Affiliation(s)
- Alaa A Abd Elhameed
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Ahmed R Ali
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Hazem A Ghabbour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Said M Bayomi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Nadia S El-Gohary
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
47
|
Pemmaraju N, Garcia JS, Perkins A, Harb JG, Souers AJ, Werner ME, Brown CM, Passamonti F. New era for myelofibrosis treatment with novel agents beyond Janus kinase-inhibitor monotherapy: Focus on clinical development of BCL-X L /BCL-2 inhibition with navitoclax. Cancer 2023; 129:3535-3545. [PMID: 37584267 DOI: 10.1002/cncr.34986] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/20/2023] [Accepted: 06/30/2023] [Indexed: 08/17/2023]
Abstract
Myelofibrosis is a heterogeneous myeloproliferative neoplasm characterized by chronic inflammation, progressive bone marrow failure, and hepatosplenic extramedullary hematopoiesis. Treatments like Janus kinase inhibitor monotherapy (e.g., ruxolitinib) provide significant spleen and symptom relief but demonstrate limited ability to lead to a durable disease modification. There is an urgent unmet medical need for treatments with a novel mechanism of action that can modify the underlying pathophysiology and affect the disease course of myelofibrosis. This review highlights the role of B-cell lymphoma (BCL) protein BCL-extra large (BCL-XL ) in disease pathogenesis and the potential role that navitoclax, a BCL-extra large/BCL-2 inhibitor, may have in myelofibrosis treatment.
Collapse
Affiliation(s)
- Naveen Pemmaraju
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Andrew Perkins
- Australian Centre for Blood Diseases, Monash University, and the Alfred Hospital, Melbourne, Victoria, Australia
| | | | | | | | | | - Francesco Passamonti
- Department of Oncology and Onco-Hematology, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
48
|
Wang J, Zhang M, Huang X, Yue H. Multiproperty Polyethylenimine-Caged Platinum Nanoclusters Promote Apoptosis of Osteosarcoma Cells via Regulating the BAX-Bcl-2/Caspase-3/PARP Axis. Mol Pharm 2023; 20:5607-5615. [PMID: 37831437 DOI: 10.1021/acs.molpharmaceut.3c00501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Osteosarcoma, a prevalent primary bone cancer in children, exhibits a poor prognosis due to the high prevalence of drug resistance. The objective of this study was to investigate the potential of fluorescent ultrafine polyethylenimine-coated caged platinum nanoclusters (PEI-Pt NCs) as an antitumor agent in osteosarcoma. The primary focus of this study involved the utilization of osteosarcoma cells (U2-OS and MG-63) and normal control cells (hBMSC) as the primary subjects of investigation. The capacity of PEI-Pt NCs to enter osteosarcoma cells was observed through the implementation of confocal microscopy. The impact of PEI-Pt NCs on migration and proliferation was assessed through the utilization of various methodologies, including the CCK8 assay, Ki-67 immunofluorescence, clone formation assay, transwell assay, and wound healing assay. Furthermore, the influence of PEI-Pt NCs on apoptosis and its underlying mechanism was explored through the implementation of flow cytometry and Western blotting techniques. The PEI-Pt NCs demonstrated the capability to enter osteosarcoma cells, including the nucleus, while also exhibiting fluorescent labeling properties. Furthermore, the PEI-Pt NCs effectively impeded the migration and proliferation of osteosarcoma cells. Additionally, the PEI-Pt NCs facilitated apoptosis by modulating the BAX-Bcl-2/Caspase 3/PARP axis. The novel nanomaterial PEI-Pt NCs possess diverse advantageous capabilities, including the ability to impede cell proliferation and migration, as well as the capacity to modulate the BAX-Bcl-2/Caspase 3/PARP axis, thereby promoting cell apoptosis. Consequently, this nanomaterial exhibits promising potential in addressing the issue of inadequate platinum-based treatment for osteosarcoma.
Collapse
Affiliation(s)
- Jialin Wang
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, 100000, China
| | - Mengjun Zhang
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, 7 Rehabilitation Front Street, Zhengzhou 450052, China
| | - Xin Huang
- Department of Light Chemical Engineering, School of Textiles, Zhongyuan University of Technology, No. 41 Zhongyuan Road (M), Zhengzhou 450007, China
| | - Haodi Yue
- Department of Center for Clinical Single Cell Biomedicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu Street, Zhengzhou 450003, China
| |
Collapse
|
49
|
Bhattacharya D, Mukhopadhyay M, Shivam K, Tripathy S, Patra R, Pramanik A. Recent developments in photodynamic therapy and its application against multidrug resistant cancers. Biomed Mater 2023; 18:062005. [PMID: 37827172 DOI: 10.1088/1748-605x/ad02d4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 10/12/2023] [Indexed: 10/14/2023]
Abstract
Recently, photodynamic therapy (PDT) has received a lot of attention for its potential use in cancer treatment. It enables the therapy of a multifocal disease with the least amount of tissue damage. The most widely used prodrug is 5-aminolevulinic acid, which undergoes heme pathway conversion to protoporphyrin IX, which acts as a photosensitizer (PS). Additionally, hematoporphyrin, bacteriochlorin, and phthalocyanine are also studied for their therapeutic potential in cancer. Unfortunately, not every patient who receives PDT experiences a full recovery. Resistance to different anticancer treatments is commonly observed. A few of the resistance mechanisms by which cancer cells escape therapeutics are genetic factors, drug-drug interactions, impaired DNA repair pathways, mutations related to inhibition of apoptosis, epigenetic pathways, etc. Recently, much research has been conducted to develop a new generation of PS based on nanomaterials that could be used to overcome cancer cells' multidrug resistance (MDR). Various metal-based, polymeric, lipidic nanoparticles (NPs), dendrimers, etc, have been utilized in the PDT application against cancer. This article discusses the detailed mechanism by which cancer cells evolve towards MDR as well as recent advances in PDT-based NPs for use against multidrug-resistant cancers.
Collapse
Affiliation(s)
- Debalina Bhattacharya
- Department of Microbiology, Maulana Azad College, Kolkata, West Bengal 700013, India
| | - Mainak Mukhopadhyay
- Department of Biotechnology, JIS University, Kolkata, West Bengal 700109, India
| | - Kumar Shivam
- Amity Institute of Click Chemistry Research & Studies, Amity University, Noida 201301, India
| | - Satyajit Tripathy
- Department of Pharmacology, University of Free State, Bloemfontein, Free State, 9301, South Africa
- Amity Institute of Allied Health Science, Amity University, Noida 201301, India
| | - Ranjan Patra
- Amity Institute of Click Chemistry Research & Studies, Amity University, Noida 201301, India
- Department of Chemistry, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Arindam Pramanik
- School of Medicine, University of Leeds, Leeds, LS9 7TF, United Kingdom
- Amity Institute of Biotechnology, Amity University, Noida 201301, India
| |
Collapse
|
50
|
Mohamed EE, Ahmed OM, Zoheir KMA, El-Shahawy AAG, Tamur S, Shams A, Burcher JT, Bishayee A, Abdel-Moneim A. Naringin-Dextrin Nanocomposite Abates Diethylnitrosamine/Acetylaminofluorene-Induced Lung Carcinogenesis by Modulating Oxidative Stress, Inflammation, Apoptosis, and Cell Proliferation. Cancers (Basel) 2023; 15:5102. [PMID: 37894468 PMCID: PMC10605195 DOI: 10.3390/cancers15205102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/29/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Nanotechnology has proven advantageous in numerous scientific applications, one being to enhance the delivery of chemotherapeutic agents. This present study aims to evaluate the mechanisms underlying the chemopreventive action of naringin-dextrin nanocomposites (Nar-Dx-NCs) against diethylnitrosamine (DEN)/2-acetylaminofluorene (2AAF)-induced lung carcinogenesis in male Wistar rats. DEN was administered intraperitoneally (i.p.) (150 mg/kg/week) for two weeks, followed by the oral administration of 2AAF (20 mg/kg) four times a week for three weeks. Rats receiving DEN/2AAF were concurrently treated with naringin or Nar-Dx-NCs orally at a dose of 10 mg/kg every other day for 24 weeks. Naringin and Nar-Dx-NCs treatments prevented the formation of tumorigenic cells within the alveoli of rats exposed to DEN/2AAF. These findings were associated with a significant decrease in lipid peroxidation, upregulation of antioxidant enzyme (glutathione peroxidase and superoxide dismutase) activity, and enhanced glutathione and nuclear factor erythroid 2-related factor 2 expression in the lungs. Naringin and Nar-Dx-NCs exerted anti-inflammatory actions manifested by a decrease in lung protein expression of tumor necrosis factor-α and interleukin-1β and mRNA expression of interleukin-6, interferon-γ, nuclear factor-κB, and inducible nitric oxide synthase, with a concurrent increase in interleukin-10 expression. The anti-inflammatory effect of Nar-Dx-NCs was more potent than naringin. Regarding the effect on apoptosis, both naringin and Nar-Dx-NCs significantly reduced Bcl-2 and increased Bax and P53 expressions. Moreover, naringin or Nar-Dx-NCs induced a significant decrease in the expression of the proliferator marker, Ki-67, and the effect of Nar-Dx-NCs was more marked. In conclusion, Nar-Dx-NCs improved naringin's preventive action against DEN/2AAF-induced lung cancer and exerted anticarcinogenic effects by suppressing oxidative stress and inflammation and improving apoptotic signal induction and propagation.
Collapse
Affiliation(s)
- Eman E. Mohamed
- Physiology Division, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt; (E.E.M.)
| | - Osama M. Ahmed
- Physiology Division, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt; (E.E.M.)
| | - Khairy M. A. Zoheir
- Cell Biology Department, Biotechnology Research Institute, National Research Centre, Cairo 12622, Egypt;
| | - Ahmed A. G. El-Shahawy
- Materials Science and Nanotechnology Department, Faculty of Postgraduate Studies for Advanced Sciences, Beni-Suef University, Beni-Suef 62521, Egypt;
| | - Shadi Tamur
- Departement of Pediatrics, College of Medicine, Taif University, Taif 21944, Saudi Arabia;
| | - Anwar Shams
- Departement of Pharmacology, College of Medicine, Taif University, Taif 21944, Saudi Arabia
- Centre of Biomedical Sciences Research, Deanship of Scientific Research, Taif University, Taif 21974, Saudi Arabia
- High Altitude Research Center, Taif University, Taif 21944, Saudi Arabia
| | - Jack T. Burcher
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA; (J.T.B.); or (A.B.)
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA; (J.T.B.); or (A.B.)
| | - Adel Abdel-Moneim
- Physiology Division, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt; (E.E.M.)
| |
Collapse
|