1
|
Saadh MJ, Allela OQB, Kareem RA, Baldaniya L, Ballal S, Vashishth R, Parmar M, Sameer HN, Hamad AK, Athab ZH, Adil M. Prognostic gene expression profile of colorectal cancer. Gene 2025; 955:149433. [PMID: 40122415 DOI: 10.1016/j.gene.2025.149433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/26/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Colorectal cancer is a major global health burden, with significant heterogeneity in clinical outcomes among patients. Identifying robust prognostic gene expression signatures can help stratify patients, guide treatment decisions, and improve clinical management. This review provides an overview of current prognostic gene expression profiles in colorectal cancer research. We have synthesized evidence from numerous published studies investigating the association between tumor gene expression patterns and patient survival outcomes. The reviewed literature reveals several promising gene signatures that have demonstrated the ability to predict disease-free survival and overall survival in CRC patients, independent of standard clinicopathological risk factors. These genes are crucial in fundamental biological processes, including cell cycle control, epithelial-mesenchymal transition, and immune regulation. The implementation of prognostic gene expression tests in clinical practice holds great potential for enabling more personalized management strategies for colorectal cancer.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan.
| | | | | | - Lalji Baldaniya
- Marwadi University Research Center, Department of Pharmacy, Faculty of Health Sciences, Marwadi University, Rajkot 360003 Gujarat, India.
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India.
| | - Raghav Vashishth
- Department of Surgery, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India.
| | - Manisha Parmar
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India.
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar 64001, Iraq.
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq.
| | | |
Collapse
|
2
|
Sun L, Chen X, Li F, Liu S. Construction and significance of a breast cancer prognostic model based on cuproptosis-related genotyping and lncRNAs. J Formos Med Assoc 2025; 124:361-374. [PMID: 38772805 DOI: 10.1016/j.jfma.2024.05.007] [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: 06/28/2023] [Revised: 03/18/2024] [Accepted: 05/08/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND/PURPOSE Cuproptosis may play a significant role in breast cancer (BC). We aimed to investigate the prognostic impact of cuproptosis-related lncRNAs in BC. METHODS Consensus clustering analysis categorized TCGA-BRCA samples into 3 clusters, followed by survival and immune analyses of the 3 clusters. LASSO-COX analysis was performed on cuproptosis-related lncRNAs differentially expressed in BC to construct a BC prognostic model. Gene Ontology/Kyoto Encyclopedia of Genes and Genomes (GO/KEGG) enrichment, immune, and drug prediction analyses were performed on the high-risk and low-risk groups. Cell experiments were conducted to analyze the results of drug prediction and two cuproptosis-related lncRNAs (AC104211.1 and LINC01863). RESULTS Significant differences were observed in survival outcomes and immune infiltration levels among the three clusters (p < 0.05). The validation of the model showed significant differences in survival outcomes between the high-risk and low-risk groups in both the training and validation sets (p < 0.05). Differential mRNAs between the two groups were significantly enriched in the Neuroactive ligand-receptor interaction and cAMP signaling pathway. Additionally, significant differences were found in immune infiltration levels, human leukocyte antigen (HLA) expression, Immunophenoscore (IPS) scores, and Tumor Immune Dysfunction and Exclusion (TIDE) scores between the two groups (p < 0.05). Drug prediction and corresponding cell experimental results showed that Trametinib, 5-fluorouracil, and AICAR significantly inhibited the viability of MCF-7 cells (p < 0.05). AC104211.1 and LINC01863 were found to impact the proliferation of BC cells. CONCLUSION The risk-scoring model obtained in this study may serve as a robust prognostic biomarker, potentially aiding in clinical decision-making for BC patients.
Collapse
Affiliation(s)
- Lu Sun
- Department of Breast Surgery, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong, China
| | - Xinxu Chen
- Department of the Breast and Thyroid Surgery, Guiqian International General Hospital, 550018, Guiyang, China
| | - Fei Li
- Department of Public Health and Medical Technology, Xiamen Medical College, Xiamen 361023, Fujian, China
| | - Shengchun Liu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, 400042, Chongqing, China.
| |
Collapse
|
3
|
Tripathi S, Sharma Y, Kumar D. Unveiling the link between chronic inflammation and cancer. Metabol Open 2025; 25:100347. [PMID: 39876904 PMCID: PMC11772974 DOI: 10.1016/j.metop.2025.100347] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/05/2025] [Accepted: 01/06/2025] [Indexed: 01/31/2025] Open
Abstract
The highly nuanced transition from an inflammatory process to tumorigenesis is of great scientific interest. While it is well known that environmental stimuli can cause inflammation, less is known about the oncogenic modifications that chronic inflammation in the tissue microenvironment can bring about, as well as how these modifications can set off pro-tumorigenic processes. It is clear that no matter where the environmental factors come from, maintaining an inflammatory microenvironment encourages carcinogenesis. In addition to encouraging angiogenesis and metastatic processes, sustaining the survival and proliferation of malignant transformed cells, and possibly altering the efficacy of therapeutic agents, inflammation can negatively regulate the antitumoral adaptive and innate immune responses. Because chronic inflammation has multiple pathways involved in tumorigenesis and metastasis, it has gained recognition as a marker of cancer and a desirable target for cancer therapy. Recent advances in our knowledge of the molecular mechanisms that drive cancer's progression demonstrate that inflammation promotes tumorigenesis and metastasis while suppressing anti-tumor immunity. In many solid tumor types, including breast, lung, and liver cancer, inflammation stimulates the activation of oncogenes and impairs the body's defenses against the tumor. Additionally, it alters the microenvironment of the tumor. As a tactical approach to cancer treatment, these findings have underscored the importance of targeting inflammatory pathways. This review highlights the role of inflammation in cancer development and metastasis, focusing on its impact on tumor progression, immune suppression, and therapy resistance. It examines current anti-inflammatory strategies, including NSAIDs, cytokine modulators, and STAT3 inhibitors, while addressing their potential and limitations. The review emphasizes the need for further research to unravel the complex mechanisms linking inflammation to cancer progression and identify molecular targets for specific cancer subtypes.
Collapse
Affiliation(s)
- Siddhant Tripathi
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
| | - Yashika Sharma
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
| | - Dileep Kumar
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| |
Collapse
|
4
|
Laila UE, Zhao ZL, Liu H, Xu ZX. Aspirin in Cancer Therapy: Pharmacology and Nanotechnology Advances. Int J Nanomedicine 2025; 20:2327-2365. [PMID: 40017626 PMCID: PMC11866938 DOI: 10.2147/ijn.s505636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 02/04/2025] [Indexed: 03/01/2025] Open
Abstract
Aspirin, a non-steroidal anti-inflammatory drug (NSAID), has garnered significant attention for its anti-cancer potential. This review explores the pharmacological properties, chemical dynamics, and evolving therapeutic applications of aspirin, with an emphasis on its integration into advanced cancer therapies. Aspirin demonstrates broad-spectrum efficacy across diverse cancer types by modulating signaling pathways such as COX-dependent and COX-independent mechanisms, including Wnt, NF-κB, β-catenin/TCF, and IL-6/STAT3. Recent advancements highlight the role of nanotechnology in enhancing aspirin's targeted delivery, therapeutic effectiveness, and patient outcomes. Nanoparticle-based formulations, including liposomes, solid lipid nanoparticles, and mesoporous silica nanoparticles, offer improved solubility, stability, and bioavailability, enabling controlled drug release and tumor-specific targeting. These innovations reduce systemic toxicity and enhance therapeutic effects, paving the way for aspirin's integration into personalized cancer treatments. Ongoing clinical studies reinforce its safety profile, underscoring aspirin's role in cancer pharmacotherapy. This review calls for continued research into aspirin's repurposing in combination therapies and novel delivery systems to maximize its therapeutic potential.
Collapse
Affiliation(s)
- Umm E Laila
- School of Life Sciences, Henan University, Kaifeng, Henan Province, 475001, People’s Republic of China
| | - Zi Lon Zhao
- School of Life Sciences, Henan University, Kaifeng, Henan Province, 475001, People’s Republic of China
| | - Huai Liu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, 475001, People’s Republic of China
| | - Zhi-Xiang Xu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, 475001, People’s Republic of China
| |
Collapse
|
5
|
Sun M, Yu J, Wan J, Dou X, Chen X, Ye F. Role of aspirin in cancer prevention. Cancer Treat Res Commun 2025; 43:100884. [PMID: 39923320 DOI: 10.1016/j.ctarc.2025.100884] [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: 09/27/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
Aspirin [1], also known as acetylsalicylic acid, is a common non-steroidal anti-inflammatory drug (NSAID). The available evidence shows that aspirin can effectively prevent and treat diseases such as rectal, gastric, ovarian, prostate, and lung cancer. Aspirin has been extensively studied for the prevention and treatment of colorectal cancer. In recent years, studies have found that the potential mechanism of aspirin in the prevention and treatment of cancer may lie in immune regulation, the role of DNA repair pathways, the regulation of cell metabolism, anti-inflammatory effects and antiplatelet effects.
Collapse
Affiliation(s)
- Meng Sun
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210003, China.
| | - Jun Yu
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210003, China
| | - Jie Wan
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210003, China
| | - Xiaoyun Dou
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210003, China
| | - Xiaoying Chen
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210003, China
| | - Fang Ye
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210003, China; Zhou Zhongying's studio, Nanjing, Jiangsu 210003, China
| |
Collapse
|
6
|
Ezenkwa US, Omenai SA, Iyapo O, Ezekekwu CA, Adetona AE, Akunwata CU, Ale AO, Ebili HO. Low prostaglandin-endoperoxide synthase-2 gene expression in colorectal carcinomas may predict poorer survival. Ecancermedicalscience 2024; 18:1814. [PMID: 40171466 PMCID: PMC11959140 DOI: 10.3332/ecancer.2024.1814] [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: 03/13/2024] [Indexed: 04/03/2025] Open
Abstract
Introduction Prostaglandin-endoperoxide synthase-2 (ptgs2), otherwise called Cyclooxygenase 2, is overexpressed in colorectal carcinoma (CRC) compared to normal tissues. However, the impact of differential expression among ptgs2-positive tumours on CRC prognosis has not been well investigated. By sub-stratifying positive tumour expression, this study determined its potential influence on patients' outcomes. Methods The Cancer Genome Atlas database was explored to determine CRC cases with RNA-Sequence (RNA-Seq) transcript data and matched clinicopathological data alongside gene copy number variation and methylation status. Descriptive, chi-square, Fisher exact, Linear-by-Linear associations, logistic and Kaplan-Meier statistics were used to determine proportions, associations, predictors and survival between ptgs2 and tumour parameters using Statistical Package for Social Sciences version 20. Two-tailed p-value <0.05 was accepted as statistically significant. Results There were 534 CRC classified predominantly as adenocarcinoma not otherwise specified (86.3%) and mucinous carcinoma (12.4) histologically included in this study. Marker (ptgs2) expression ranged from 0.02 FPKM-131.89 FPKM, (Median 1.4 FPKM). The majority of the cases (53.4%) were diagnosed at an early stage and showed high ptgs2 RNA-Sequence (RNA-seq) expression in 51.5% (275/534). Significant associations were seen between ptgs2 expression and histological subtype (p < 0.001), lymphovascular invasion (p = 0.013), pN2 stage (> 6 positive lymph nodes) (p = 0.011) and American Joint Committee on Cancer Staging stage (p = 0.028), and these all had lower ptgs2 expression. On regression analysis, histological differentiation emerged as a predictor of ptgs2 expression (Odds ratio 2.749, 95% confidence interval 1.479-5.108, p < 0.001). Also, gene methylation was associated with reduced ptgs2 expression. Overall survival was significantly inferior among individuals with low ptgs2 tumours (p = 0.018) while that for disease-free survival was non-significant (p = 0.327). Conclusion CRCs with low ptgs2 transcripts are associated with poorer survival. This finding suggests a need for closer follow up and tailored adjuvant therapy for these patients.
Collapse
Affiliation(s)
- Uchenna Simon Ezenkwa
- Department of Histopathology, Federal University of Health Sciences Azare, Azare 751101, Bauchi, Nigeria
- Department of Pathology, Federal Medical Centre Azare, Azare 751101, Bauchi, Nigeria
| | | | - Oluwadamilare Iyapo
- Department of Pathology, Federal Medical Centre, Ebute Metta, Lagos 101212, Nigeria
| | - Chinedu Anthony Ezekekwu
- Bristol Haematology and Oncology Centre, University Hospital, Bristol and Weston NHS Trust, Bristol BS28ED, UK
| | - Adesoji E Adetona
- Department of Morbid Anatomy & Histopathology, Olabisi Onabanjo University, Ago-Iwoye 121101, Ogun State, Nigeria
| | - Chima Uzoma Akunwata
- Department of Haematology, University College Hospital Ibadan, Ibadan 200221, Oyo, Nigeria
| | - Ayotunde Oladunmi Ale
- Department of Medicine, Olabisi Onabanjo University/University Teaching Hospital, Sagamu 121101, Ogun, Nigeria
| | - Henry Okwuchukwu Ebili
- Department of Morbid Anatomy & Histopathology, Olabisi Onabanjo University, Ago-Iwoye 121101, Ogun State, Nigeria
| |
Collapse
|
7
|
Hu H, Chen WJ, Xiong ZY, Luo LF, Sun C, Xie JP. Association of prophylactic low-dose aspirin use with all-cause and cause-specific mortality in cancer patients. Sci Rep 2024; 14:25918. [PMID: 39472644 PMCID: PMC11522375 DOI: 10.1038/s41598-024-75612-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024] Open
Abstract
The long-term use of aspirin for preventing cardiovascular disease has been recommended for decades. However, there is currently uncertainty regarding the long-term effects of aspirin use on the risk of all-cause, cardiovascular, and cancer mortality in cancer patients. The aim of this work was to analyze the connection between the prophylactic use of low-dose aspirin and the risk of all-cause death, cardiovascular death, and carcinoma death in carcinoma patients in the United States. A cohort study was conducted using National Health and Nutrition Examination Survey (NHANES) data (2011-2012, 2013-2014, 2015-2016, and 2017-2018) and associated mortality data. The 95% confidence intervals (CIs) and hazard ratios (HRs) between non-aspirin use and prophylactic low-dose aspirin use and the risk of death were measured via Cox proportional hazard regression models. A total of 1819 participants were included in the present research, of whom 945 were nonaspirin users and 874 were prophylactic aspirin users. Compared with non-aspirin users, prophylactic low-dose aspirin users had a decreased risk of all-cause death (HR = 0.647, 95% CI = 0.489-0.857). There was no statistically significant difference in the risk of cardiovascular death (HR = 0.623, 95% CI = 0.362-1.074) or cancer death (HR = 0.709, 95% CI = 0.410-1.226). Prophylactic use of low-dose aspirin may lower all-cause mortality in individuals with cancer but does not have a substantial effect on cardiovascular risk or cancer-specific mortality in this patient population.
Collapse
Affiliation(s)
- Hui Hu
- Department of Respiratory and Critical Care, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Wen-Jun Chen
- Department of Oncology, Anqing Hospital of PLA Navy, Anqing, 246004, China
| | - Zi-Yi Xiong
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Lin-Fei Luo
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Chuang Sun
- Department of Respiratory and Critical Care, Xinyu People's Hospital, Xinyu, 338099, China
| | - Jun-Ping Xie
- Department of Respiratory and Critical Care, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
8
|
Mneimneh AT, Darwiche N, Mehanna MM. Investigating the therapeutic promise of drug-repurposed-loaded nanocarriers: A pioneering strategy in advancing colorectal cancer treatment. Int J Pharm 2024; 664:124473. [PMID: 39025341 DOI: 10.1016/j.ijpharm.2024.124473] [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: 04/22/2024] [Revised: 07/06/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024]
Abstract
Globally, colorectal cancer is a major health problem that ranks in third place in terms of occurrence and second in terms of mortality worldwide. New cases increase annually, with the absence of effective therapies, especially for metastatic colorectal cancer, emphasizing the need for novel therapeutic approaches. Although conventional treatments are commonly used in oncotherapy, their success rate is low, which leads to the exploration of novel technologies. Recent efforts have focused on developing safe and efficient cancer nanocarriers. With their nanoscale properties, nanocarriers have the potential to utilize internal metabolic modifications amid cancer and healthy cells. Drug repurposing is an emerging strategy in cancer management as it is a faster, cheaper, and safer method than conventional drug development. However, most repurposed drugs are characterized by low-key pharmacokinetic characteristics, such as poor aqueous solubility, permeability, retention, and bioavailability. Nanoparticles formulations and delivery have expanded over the past few decades, creating opportunities for drug repurposing and promises as an advanced cancer modality. This review provides a concise and updated overview of colorectal cancer treatment regimens and their therapeutic limitations. Furthermore, the chemotherapeutic effect of various FDA-approved medications, including statins, non-steroidal anti-inflammatory drugs, antidiabetic and anthelmintic agents, and their significance in colorectal cancer management. Along with the role of various nanocarrier systems in achieving the desired therapeutic outcomes of employing these redefined drugs.
Collapse
Affiliation(s)
- Amina T Mneimneh
- Pharmaceutical Nanotechnology Research lab, Faculty of Pharmacy, Beirut Arab University, Beirut, Lebanon.
| | - Nadine Darwiche
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.
| | - Mohammed M Mehanna
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Department of Pharmaceutical Sciences, School of Pharmacy, Lebanese American University, Byblos, Lebanon.
| |
Collapse
|
9
|
Xu X, Gao Y, Dai J, Wang Q, Wang Z, Liang W, Zhang Q, Ma W, Liu Z, Luo H, Qiao Z, Li L, Wang Z, Chen L, Zhang Y, Xiong Z. Gastric Cancer Assembloids Derived from Patient-Derived Xenografts: A Preclinical Model for Therapeutic Drug Screening. SMALL METHODS 2024; 8:e2400204. [PMID: 38948952 DOI: 10.1002/smtd.202400204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/18/2024] [Indexed: 07/02/2024]
Abstract
The construction of reliable preclinical models is crucial for understanding the molecular mechanisms involved in gastric cancer and for advancing precision medicine. Currently, existing in vitro tumor models often do not accurately replicate the human gastric cancer environment and are unsuitable for high-throughput therapeutic drug screening. In this study, droplet microfluidic technology is employed to create novel gastric cancer assembloids by encapsulating patient-derived xenograft gastric cancer cells and patient stromal cells in Gelatin methacryloyl (GelMA)-Gelatin-Matrigel microgels. The usage of GelMA-Gelatin-Matrigel composite hydrogel effectively alleviated cell aggregation and sedimentation during the assembly process, allowing for the handling of large volumes of cell-laden hydrogel and the uniform generation of assembloids in a high-throughput manner. Notably, the patient-derived xenograft assembloids exhibited high consistency with primary tumors at both transcriptomic and histological levels, and can be efficiently scaled up for preclinical drug screening efforts. Furthermore, the drug screening results clearly demonstrated that the in vitro assembloid model closely mirrored in vivo drug responses. Thus, these findings suggest that gastric cancer assembloids, which effectively replicate the in vivo tumor microenvironment, show promise for enabling more precise high-throughput drug screening and predicting the clinical outcomes of various drugs.
Collapse
Affiliation(s)
- Xinxin Xu
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Medical School of Chinese PLA, Beijing, 100853, China
- Senior Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yunhe Gao
- Senior Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jianli Dai
- Institute of New Materials and Advanced Manufacturing, Beijing Academy of Science and Technology, Beijing, 100089, China
| | - Qianqian Wang
- Institute of New Materials and Advanced Manufacturing, Beijing Academy of Science and Technology, Beijing, 100089, China
| | - Zixuan Wang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Wenquan Liang
- Senior Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Qing Zhang
- Institute of New Materials and Advanced Manufacturing, Beijing Academy of Science and Technology, Beijing, 100089, China
| | - Wenbo Ma
- Institute of New Materials and Advanced Manufacturing, Beijing Academy of Science and Technology, Beijing, 100089, China
| | - Zibo Liu
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Hao Luo
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Zhi Qiao
- Senior Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Li Li
- Medical School of Chinese PLA, Beijing, 100853, China
- Senior Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Zijian Wang
- Medical School of Chinese PLA, Beijing, 100853, China
- Senior Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Lin Chen
- Senior Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yanmei Zhang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Institute of New Materials and Advanced Manufacturing, Beijing Academy of Science and Technology, Beijing, 100089, China
| | - Zhuo Xiong
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
10
|
Tufail M, Wan WD, Jiang C, Li N. Targeting PI3K/AKT/mTOR signaling to overcome drug resistance in cancer. Chem Biol Interact 2024; 396:111055. [PMID: 38763348 DOI: 10.1016/j.cbi.2024.111055] [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: 03/27/2024] [Revised: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 05/21/2024]
Abstract
This review comprehensively explores the challenge of drug resistance in cancer by focusing on the pivotal PI3K/AKT/mTOR pathway, elucidating its role in oncogenesis and resistance mechanisms across various cancer types. It meticulously examines the diverse mechanisms underlying resistance, including genetic mutations, feedback loops, and microenvironmental factors, while also discussing the associated resistance patterns. Evaluating current therapeutic strategies targeting this pathway, the article highlights the hurdles encountered in drug development and clinical trials. Innovative approaches to overcome resistance, such as combination therapies and precision medicine, are critically analyzed, alongside discussions on emerging therapies like immunotherapy and molecularly targeted agents. Overall, this comprehensive review not only sheds light on the complexities of resistance in cancer but also provides a roadmap for advancing cancer treatment.
Collapse
Affiliation(s)
- Muhammad Tufail
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Wen-Dong Wan
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Canhua Jiang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China; Institute of Oral Precancerous Lesions, Central South University, Changsha, China; Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ning Li
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China; Institute of Oral Precancerous Lesions, Central South University, Changsha, China; Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
11
|
Gonçalves J, Pinto S, Carmo F, Silva C, Andrade N, Martel F. Additive Cytotoxic and Colony-Formation Inhibitory Effects of Aspirin and Metformin on PI3KCA-Mutant Colorectal Cancer Cells. Int J Mol Sci 2024; 25:5381. [PMID: 38791419 PMCID: PMC11121714 DOI: 10.3390/ijms25105381] [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: 04/01/2024] [Revised: 05/08/2024] [Accepted: 05/12/2024] [Indexed: 05/26/2024] Open
Abstract
Human malignancies are one of the major health-related issues throughout the world and are anticipated to rise in the future. Despite huge investments made in anticancer drug development, limited success has been obtained and the average number of FDA approvals per year is declining. So, an increasing interest in drug repurposing exists. Metformin (MET) and aspirin (ASP) possess anticancer properties. This work aims to test the effect of these two drugs in combination on colorectal cancer (CRC) cells in vitro. The effects of MET and/or ASP on cell proliferation, viability, migratory ability, anchorage-independent growth ability (colony formation), and nutrient uptake were determined in two (HT-29 and Caco-2) human CRC cell lines. Individually, MET and ASP possessed antiproliferative, cytotoxic, and antimigratory effects and reduced colony formation in HT-29 cells (BRAF- and phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α (PI3KCA)-mutant), although MET did not affect either 3H-deoxy-D-glucose or 14C-butyrate uptake and lactate production, and ASP caused only a small decrease in 14C-butyrate uptake. Moreover, in these cells, the combination of MET and ASP resulted in a tendency to an increase in the cytotoxic effect and in a potentiation of the inhibitory effect on colony formation, although no additive antiproliferative and antimigratory effects, and no effect on nutrient uptake and lactate production were observed. In contrast, MET and ASP, both individually and in combination, were almost devoid of effects on Caco-2 cells (BRAF- and PI3KCA-wild type). We suggest that inhibition of PI3K is the common mechanism involved in the anti-CRC effect of both MET, ASP and their combination and, therefore, that the combination of MET + ASP may especially benefit PI3KCA-mutant CRC cases, which currently have a poor prognostic.
Collapse
Affiliation(s)
- Joana Gonçalves
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (J.G.); (S.P.); (F.C.); (N.A.)
| | - Sara Pinto
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (J.G.); (S.P.); (F.C.); (N.A.)
| | - Francisca Carmo
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (J.G.); (S.P.); (F.C.); (N.A.)
| | - Cláudia Silva
- LAQV/REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-453 Porto, Portugal;
| | - Nelson Andrade
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (J.G.); (S.P.); (F.C.); (N.A.)
- LAQV/REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-453 Porto, Portugal;
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal
| | - Fátima Martel
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (J.G.); (S.P.); (F.C.); (N.A.)
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal
| |
Collapse
|
12
|
Ogurchenok NE, Khalin KD, Bryukhovetskiy IS. Chemoprophylaxis of precancerous lesions in patients who are at a high risk of developing colorectal cancer (Review). MEDICINE INTERNATIONAL 2024; 4:25. [PMID: 38628384 PMCID: PMC11019464 DOI: 10.3892/mi.2024.149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 03/07/2024] [Indexed: 04/19/2024]
Abstract
The diagnostics of colorectal cancer (CRC) and precancerous lesions in the colon is one of the most urgent matters to be considered for the modern protocols of complex examination, recommended for use from the age of 45 years, and including both instrumental and laboratory methods of research: Colonoscopy, CT colonography, flexible sigmoidoscopy, fecal occult blood test, fecal immunohistochemistry test and stool DNA test Nevertheless, the removal of those precancerous lesions does not solve the issue, and, apart from the regular endoscopic monitoring of patients who are at a high risk of developing CRC, the pharmacological treatment of certain key pathogenic mechanisms leading to the development of CRC is required. The present review to discusses the function of β-catenin in the transformation of precancerous colorectal lesions into CRC, when collaborating with PI3K/AKT/mTOR signaling pathway and other mechanisms. The existing methods for the early diagnostics and prevention of discovered anomalies are described and categorized. The analysis of the approaches to chemoprophylaxis of CRC, depending on the results of endoscopic, morphological and molecular-genetic tests, is presented.
Collapse
Affiliation(s)
- Nonna E. Ogurchenok
- Far Eastern Federal University, School of Medicine and Life Sciences, FEFU Medical Center, Russky Island, 690091 Vladivostok, Russian Federation
- Primorskiy Regional Clinical Hospital N1, Medical Center, Russky Island, 690091 Vladivostok, Russian Federation
| | - Konstantin D. Khalin
- Far Eastern Federal University, School of Medicine and Life Sciences, FEFU Medical Center, Russky Island, 690091 Vladivostok, Russian Federation
- Far Eastern Federal University, Medical Center, Russky Island, 690091 Vladivostok, Russian Federation
| | - Igor S. Bryukhovetskiy
- Far Eastern Federal University, Medical Center, Russky Island, 690091 Vladivostok, Russian Federation
| |
Collapse
|
13
|
Zha X, Ji R, Li Y, Cao R, Zhou S. Network pharmacology, molecular docking, and molecular dynamics simulation analysis reveal the molecular mechanism of halociline against gastric cancer. Mol Divers 2024:10.1007/s11030-024-10822-y. [PMID: 38504075 DOI: 10.1007/s11030-024-10822-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 02/06/2024] [Indexed: 03/21/2024]
Abstract
Halociline, a derivative of alkaloids, was isolated from the marine fungus Penicillium griseofulvum by our group. This remarkable compound exhibits promising antineoplastic activity, yet the precise molecular mechanisms underlying its anticancer properties remain enigmatic. To unravel these mechanisms, we employed an integrated approach of network pharmacology analysis, molecular docking simulations, and molecular dynamics simulations to explore halociline therapeutic targets for gastric cancer. The data from network pharmacology indicate that halociline targets MAPK1, MMP-9, and PIK3CA in gastric cancer cells, potentially mediated by diverse pathways including cancer, lipid metabolism, atherosclerosis, and EGFR tyrosine kinase inhibitor resistance. Notably, molecular docking and dynamics simulations revealed a high affinity between halociline and these targets, with free binding energies (ΔEtotal) of - 20.28, - 27.94, and - 25.97 kcal/mol for MAPK1, MMP-9, and PIK3CA, respectively. This study offers valuable insights into the potential molecular mechanism of halociline's inhibition of gastric cancer cells and serves as a valuable reference for future basic research efforts.
Collapse
Affiliation(s)
- Xiangru Zha
- NHC Key Laboratory of Control of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, 3#, Xueyuan Road, Haikou, 571199, Hainan, China
| | - Rong Ji
- NHC Key Laboratory of Control of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, 3#, Xueyuan Road, Haikou, 571199, Hainan, China
| | - Yang Li
- NHC Key Laboratory of Control of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, 3#, Xueyuan Road, Haikou, 571199, Hainan, China
| | - Rong Cao
- NHC Key Laboratory of Control of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, 3#, Xueyuan Road, Haikou, 571199, Hainan, China
| | - Songlin Zhou
- NHC Key Laboratory of Control of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, 3#, Xueyuan Road, Haikou, 571199, Hainan, China.
| |
Collapse
|
14
|
Pandey M, Rajput M, Singh P, Shukla M, Zhu B, Koshiol J. Aspirin and Cancer Survival: An Analysis of Molecular Mechanisms. Cancers (Basel) 2024; 16:223. [PMID: 38201650 PMCID: PMC10778469 DOI: 10.3390/cancers16010223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/25/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
The benefit of aspirin on cancer survival is debated. Data from randomized clinical trials and cohort studies are discordant, although a meta-analysis shows a clear survival advantage when aspirin is added to the standard of care. However, the mechanism by which aspirin improves cancer survival is not clear. A PubMed search was carried out to identify articles reporting genes and pathways that are associated with aspirin and cancer survival. Gene ontology and pathway enrichment analysis was carried out using web-based tools. Gene-gene and protein-protein interactions were evaluated. Crosstalk between pathways was identified and plotted. Forty-one genes were identified and classified into primary genes (PTGS2 and PTGES2), genes regulating cellular proliferation, interleukin and cytokine genes, and DNA repair genes. The network analysis showed a rich gene-gene and protein-protein interaction between these genes and proteins. Pathway enrichment showed the interleukin and cellular transduction pathways as the main pathways involved in aspirin-related survival, in addition to DNA repair, autophagy, extracellular matrix, and apoptosis pathways. Crosstalk of PTGS2 with EGFR, JAK/AKT, TP53, interleukin/TNFα/NFκB, GSK3B/BRCA/PARP, CXCR/MUC1, and WNT/CTNNB pathways was identified. The results of the present study demonstrate that aspirin improves cancer survival by the interplay of 41 genes through a complex mechanism. PTGS2 is the primary target of aspirin and impacts cancer survival through six primary pathways: the interleukin pathway, extracellular matrix pathway, signal transduction pathway, apoptosis pathway, autophagy pathway, and DNA repair pathway.
Collapse
Affiliation(s)
- Manoj Pandey
- Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India; (M.R.)
| | - Monika Rajput
- Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India; (M.R.)
| | - Pooja Singh
- Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India; (M.R.)
| | - Mridula Shukla
- RRL, Dr. Lalpath Labs Ltd., Shivpur, Varanasi 221003, India
| | - Bin Zhu
- Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), National Institutes of Health (NIH), 9609 Medical Center Drive, RM 6-E212, Rockville, MD 20850, USA (J.K.)
| | - Jill Koshiol
- Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), National Institutes of Health (NIH), 9609 Medical Center Drive, RM 6-E212, Rockville, MD 20850, USA (J.K.)
| |
Collapse
|
15
|
Sarabi PZ, Moradi M, Bagheri M, Khalili MR, Moradifard S, Jamialahmadi T, Ghasemi F, Sahebkar A. A Contemporary Review on the Critical Role of Nonsteroidal Anti-inflammatory Agents in Colorectal Cancer Therapy. Anticancer Agents Med Chem 2024; 24:559-570. [PMID: 38275052 DOI: 10.2174/0118715206271583231206052403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/25/2023] [Accepted: 10/30/2023] [Indexed: 01/27/2024]
Abstract
Nonsteroidal Anti-Inflammatory Drugs (NSAIDs) are widely recognized as effective pain relievers and function by inhibiting the cyclooxygenase enzyme (COXs). Moreover, they have been found to participate in various cellular processes through different signaling pathways, such as WNT, MAPK, NF-κB, and PI3K/AKT/mTOR. This makes them potential candidates for chemoprevention of several malignancies, particularly colorectal cancer (CRC). However, the use of NSAIDs in cancer prevention and treatment is a complex issue due to their adverse effects and gastrointestinal toxicity. Therefore, it is crucial to explore combination therapies that can minimize side effects while maximizing synergistic effects with other agents and to evaluate the success rate of such approaches in both pre-clinical and clinical studies. In this review, we aim to provide an overview of the effects of NSAIDs in the prevention and treatment of CRC. We will focus on elucidating the possible mechanisms of action of these drugs, the signaling pathways involved in CRC, and the potential synergistic effects when combined with other therapeutic agents.
Collapse
Affiliation(s)
- Parisa Zia Sarabi
- Laboratorio de Psicobiología, Campus Santiago Ramón y Cajal, University of Sevilla, 41018, Sevilla, Spain
| | - Mohammad Moradi
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Malihe Bagheri
- Department of Biotechnology and Molecular Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Mohammad Reza Khalili
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Next to Milad Tower, Tehran, Iran
| | - Shahrzad Moradifard
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Next to Milad Tower, Tehran, Iran
| | - Tannaz Jamialahmadi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Faezeh Ghasemi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Next to Milad Tower, Tehran, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
16
|
Liu Z, Cui L, Wang J, Zhao W, Teng Y. Aspirin boosts the synergistic effect of EGFR/p53 inhibitors on lung cancer cells by regulating AKT/mTOR and p53 pathways. Cell Biochem Funct 2024; 42:e3902. [PMID: 38100146 DOI: 10.1002/cbf.3902] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/25/2023] [Accepted: 11/27/2023] [Indexed: 01/26/2024]
Abstract
The regimen of afatinib and vinorelbine has been used to treat breast or lung cancer cells with some limitations. Aspirin alone or in combination with other agents has shown unique efficacy in the treatment of cancer. We designed a preclinical study to investigate whether the triple therapy of aspirin, afatinib, and vinorelbine could synergistically inhibit the growth of p53 wild-type nonsmall cell lung cancer (NSCLC) cells. Three NSCLC cells A549, H460, and H1975 were selected to study the effect of triple therapy on cell proliferation and apoptosis. Compared to single agents, triple therapy synergistically inhibited the proliferation of lung cancer cells with combination index <1. Meanwhile, the therapeutic index of triple therapy was superior to that of single agents, indicating a balance between efficacy and safety in the combination of three agents. Mechanistic studies showed that triple therapy significantly induced apoptosis by decreasing mitochondrial membrane potential, increasing reactive oxygen species, and regulating mitochondria-related proteins. Moreover, epidermal growth factor receptor (EGFR) downstream signaling proteins including JNK, AKT, and mTOR were dramatically suppressed and p53 was substantially increased after NSCLC cells were exposed to the triple therapy. We provided evidence that the triple therapy of aspirin, afatinib and vinorelbine synergistically inhibited lung cancer cell growth through inactivation of the EGFR/AKT/mTOR pathway and accumulation of p53, providing a new treatment strategy for patients with p53 wild-type NSCLC.
Collapse
Affiliation(s)
- Zhen Liu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Li Cui
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Jinyao Wang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Wanshun Zhao
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
- National & Local United Engineering Laboratory of TCM Advanced Manufacturing Technology, Tasly Pharmaceutical Group Co. Ltd., Tianjin, China
| | - Yuou Teng
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| |
Collapse
|
17
|
Hoskin AJ, Holt AK, Legge DN, Collard TJ, Williams AC, Vincent EE. Aspirin and the metabolic hallmark of cancer: novel therapeutic opportunities for colorectal cancer. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:600-615. [PMID: 37720350 PMCID: PMC10501897 DOI: 10.37349/etat.2023.00155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 06/08/2023] [Indexed: 09/19/2023] Open
Abstract
Aspirin is a well-known nonsteroidal anti-inflammatory drug (NSAID) that has a recognized role in cancer prevention as well as evidence to support its use as an adjuvant for cancer treatment. Importantly there has been an increasing number of studies contributing to the mechanistic understanding of aspirins' anti-tumour effects and these studies continue to inform the potential clinical use of aspirin for both the prevention and treatment of cancer. This review focuses on the emerging role of aspirin as a regulator of metabolic reprogramming, an essential "hallmark of cancer" required to support the increased demand for biosynthetic intermediates needed for sustained proliferation. Cancer cells frequently undergo metabolic rewiring driven by oncogenic pathways such as hypoxia-inducible factor (HIF), wingless-related integration site (Wnt), mammalian target of rapamycin (mTOR), and nuclear factor kappa light chain enhancer of activated B cells (NF-κB), which supports the increased proliferative rate as tumours develop and progress. Reviewed here, cellular metabolic reprogramming has been identified as a key mechanism of action of aspirin and include the regulation of key metabolic drivers, the regulation of enzymes involved in glycolysis and glutaminolysis, and altered nutrient utilisation upon aspirin exposure. Importantly, as aspirin treatment exposes metabolic vulnerabilities in tumour cells, there is an opportunity for the use of aspirin in combination with specific metabolic inhibitors in particular, glutaminase (GLS) inhibitors currently in clinical trials such as telaglenastat (CB-839) and IACS-6274 for the treatment of colorectal and potentially other cancers. The increasing evidence that aspirin impacts metabolism in cancer cells suggests that aspirin could provide a simple, relatively safe, and cost-effective way to target this important hallmark of cancer. Excitingly, this review highlights a potential new role for aspirin in improving the efficacy of a new generation of metabolic inhibitors currently undergoing clinical investigation.
Collapse
Affiliation(s)
- Ashley J. Hoskin
- Department of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, BS8 1TW Bristol, UK
| | - Amy K. Holt
- Department of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, BS8 1TW Bristol, UK
| | - Danny N. Legge
- Department of Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, BS1 3NY Bristol, UK
| | - Tracey J. Collard
- Department of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, BS8 1TW Bristol, UK
| | - Ann C. Williams
- Department of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, BS8 1TW Bristol, UK
| | - Emma E. Vincent
- Department of Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, BS1 3NY Bristol, UK
- MRC Integrative Epidemiology Unit, Oakfield House, University of Bristol, BS8 2BN Bristol, UK
| |
Collapse
|
18
|
Han R, Lin C, Zhang C, Kang J, Lu C, Zhang Y, Wang Y, Hu C, He Y. The potential therapeutic regimen for overcoming resistance to osimertinib due to rare mutations in NSCLC. iScience 2023; 26:107105. [PMID: 37416479 PMCID: PMC10320197 DOI: 10.1016/j.isci.2023.107105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 05/07/2023] [Accepted: 06/08/2023] [Indexed: 07/08/2023] Open
Abstract
The mechanisms of osimertinib resistance have not been well characterized. We conducted next-generation sequencing to recognize novel resistance mechanism and used cell line-derived xenograft (CDX) and patient-derived xenograft (PDX) models to evaluate the anti-proliferative effects of aspirin in vivo and in vitro. We observed that PIK3CG mutations led to acquired resistance to osimertinib in a patient and further confirmed that both PIK3CG and PIK3CA mutations caused osimertinib resistance. Mechanistically, the expression of PI3Kγ or PI3Kα was up-regulated after PIK3CG or PIK3CA lentivirus transfection, respectively, and which can be effectively suppressed by aspirin. Lastly, our results from in vivo studies indicate that aspirin can reverse osimertinib resistance caused by PIK3CG or PIK3CA mutations in both CDX and PDX models. Herein, we first confirmed that mutations in PIK3CG can lead to resistance to osimertinib, and the combined therapy may be a strategy to reverse PIK3CG/PIK3CA mutation-induced osimertinib resistance.
Collapse
Affiliation(s)
- Rui Han
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Caiyu Lin
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Chong Zhang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun Kang
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Conghua Lu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Yiming Zhang
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Yubo Wang
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Chen Hu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Yong He
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
19
|
Zhang Y, Wang Y, Zhang B, Li P, Zhao Y. Methods and biomarkers for early detection, prediction, and diagnosis of colorectal cancer. Biomed Pharmacother 2023; 163:114786. [PMID: 37119736 DOI: 10.1016/j.biopha.2023.114786] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/17/2023] [Accepted: 04/24/2023] [Indexed: 05/01/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common digestive diseases worldwide. It has steadily ascended to the top three cancers in terms of incidence and mortality. The primary cause is the inability to diagnose it at an early stage. Therefore, early detection and diagnosis are essential for colorectal cancer prevention. Although there are now various methods for CRC early detection, in addition to recent developments in surgical and multimodal therapy, the poor prognosis and late detection of CRC still remain significant. Thus, it is important to investigate novel technologies and biomarkers to improve the sensitization and specification of CRC diagnosis. Here, we present some common methods and biomarkers for early detection and diagnosis of CRC, we hope this review will encourage the adoption of screening programs and the clinical use of these potential molecules as biomarkers for CRC early detection and prognosis.
Collapse
Affiliation(s)
- Yue Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China; Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province affiliated to Qingdao University, Shandong Province, China
| | - Bingqiang Zhang
- Key Laboratory of Cancer and Immune Cells of Qingdao, Qingdao 266021, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| | - Yi Zhao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| |
Collapse
|
20
|
Manzi J, Hoff CO, Ferreira R, Pimentel A, Datta J, Livingstone AS, Vianna R, Abreu P. Targeted Therapies in Colorectal Cancer: Recent Advances in Biomarkers, Landmark Trials, and Future Perspectives. Cancers (Basel) 2023; 15:cancers15113023. [PMID: 37296986 DOI: 10.3390/cancers15113023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/30/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
In 2022, approximately 600,000 cancer deaths were expected; more than 50,000 of those deaths would be from colorectal cancer (CRC). The CRC mortality rate in the US has decreased in recent decades, with a 51% drop between 1976 and 2014. This drop is attributed, in part, to the tremendous therapeutic improvements, especially after the 2000s, in addition to increased social awareness regarding risk factors and diagnostic improvement. Five-fluorouracil, irinotecan, capecitabine, and later oxaliplatin were the mainstays of mCRC treatment from the 1960s to 2002. Since then, more than a dozen drugs have been approved for the disease, betting on a new chapter in medicine, precision oncology, which uses patient and tumor characteristics to guide the therapeutic choice. Thus, this review will summarize the current literature on targeted therapies, highlighting the molecular biomarkers involved and their pathways.
Collapse
Affiliation(s)
- Joao Manzi
- Miami Transplant Institute, Jackson Memorial Hospital, University of Miami, Miami, FL 33136, USA
| | - Camilla O Hoff
- Miami Transplant Institute, Jackson Memorial Hospital, University of Miami, Miami, FL 33136, USA
| | - Raphaella Ferreira
- Miami Transplant Institute, Jackson Memorial Hospital, University of Miami, Miami, FL 33136, USA
| | - Agustin Pimentel
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Jashodeep Datta
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Alan S Livingstone
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Rodrigo Vianna
- Miami Transplant Institute, Jackson Memorial Hospital, University of Miami, Miami, FL 33136, USA
| | - Phillipe Abreu
- Miami Transplant Institute, Jackson Memorial Hospital, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
21
|
The Cytokine Network in Colorectal Cancer: Implications for New Treatment Strategies. Cells 2022; 12:cells12010138. [PMID: 36611932 PMCID: PMC9818504 DOI: 10.3390/cells12010138] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 01/01/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most frequent tumor entities worldwide with only limited therapeutic options. CRC is not only a genetic disease with several mutations in specific oncogenes and/or tumor suppressor genes such as APC, KRAS, PIC3CA, BRAF, SMAD4 or TP53 but also a multifactorial disease including environmental factors. Cancer cells communicate with their environment mostly via soluble factors such as cytokines, chemokines or growth factors to generate a favorable tumor microenvironment (TME). The TME, a heterogeneous population of differentiated and progenitor cells, plays a critical role in regulating tumor development, growth, invasion, metastasis and therapy resistance. In this context, cytokines from cancer cells and cells of the TME influence each other, eliciting an inflammatory milieu that can either enhance or suppress tumor growth and metastasis. Additionally, several lines of evidence exist that the composition of the microbiota regulates inflammatory processes, controlled by cytokine secretion, that play a role in carcinogenesis and tumor progression. In this review, we discuss the cytokine networks between cancer cells and the TME and microbiome in colorectal cancer and the related treatment strategies, with the goal to discuss cytokine-mediated strategies that could overcome the common therapeutic resistance of CRC tumors.
Collapse
|
22
|
Chen H, Lin Y, Zeng L, Liu S. Elucidating the mechanism of Hongjinshen decoction in the treatment of pulmonary fibrosis based on network pharmacology and molecular docking. Medicine (Baltimore) 2022; 101:e32323. [PMID: 36595795 PMCID: PMC9794259 DOI: 10.1097/md.0000000000032323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND To explore the mechanism of compound Hongginshen decoction in improving pulmonary fibrosis based on network pharmacology. METHODS The active components and targets of ginseng and Salvia miltiorrhiza were screened from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) database. The chemical components of Rhodiola, Ophiopogon japonicus, and Dendrobium were screened using the Traditional Chinese Medicine Integrated Database (TCMID), and the target compounds were predicted by the Swisstargets method. The related target genes of pulmonary fiber (PF) were screened by the Genecards database and the National Center of Biotechnology Information (NCBI) database. The protein-protein interaction network was drawn using the string database and Cytoscape software, and the network topology was analyzed. Then, using R3.6.3 software, biological processes, molecular function, cell component enrichment, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment were carried out on the common targets of drugs and diseases. The network diagram of the "traditional Chinese medicine composition disease target" of Compound Hongginshen Decoction was constructed and analyzed with the software of Cytoscape 3.6.1. RESULTS We identified 159 active components and 2820 targets in Compound Hongginshen Decoction, and 2680 targets in pulmonary fibrosis. A total of 343 common targets were obtained by the intersection of drug targets and disease targets. protein-protein interaction protein interaction network analysis showed that PIK3CA, PIK3R1, MAPK1, SRC, AKT1, and so on may be the core targets of the compound Hongjingshen recipe in the treatment of pulmonary fibrosis. Gene Ontology (GO) enrichment analysis identified 3463 items, and KEGG pathway enrichment analysis identified 181 related signaling pathways, including the PI3K-Akt signaling pathway, HCMV pathway, Hb pathway, PGs pathway, and KSHV signaling pathway. CONCLUSION Compound Hongginshen Decoction has the characteristics of a multichannel and multitargeted effect in the treatment of pulmonary fibrosis. Radix Ophiopogonis and Dendrobium officinale play a key role in the treatment of pulmonary fibrosis. The whole compound prescription may play a therapeutic role by affecting cell metabolism, being anti-inflammatory, regulating the immune system, promoting angiogenesis, and improving anaerobic metabolism.
Collapse
Affiliation(s)
- Haixu Chen
- Department of Basic Medicine, Sichuan Vocational College of Health and Rehabilitation, Zigong, Sichuan, China
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Yu Lin
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- Department of Clinical and Medical Technology, Sichuan Vocational College of Health and Rehabilitation, Zigong, Sichuan, China
| | - Lianlin Zeng
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- * Correspondence: Shiwei Liu, Department of Rehabilitation Medicine, Zigong No.4 People’s Hospital, Sichuan 643000, China (e-mail: ) and Lianlin Zeng, Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China (e-mail: )
| | - Shiwei Liu
- Department of Rehabilitation Medicine, Zigong No.4 People’s Hospital, Sichuan, China
- * Correspondence: Shiwei Liu, Department of Rehabilitation Medicine, Zigong No.4 People’s Hospital, Sichuan 643000, China (e-mail: ) and Lianlin Zeng, Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China (e-mail: )
| |
Collapse
|
23
|
Xu W, Wang M, Bai Y, Chen Y, Ma X, Yang Z, Zhao L, Li Y. The role of microfibrillar‐associated protein 2 in cancer. Front Oncol 2022; 12:1002036. [DOI: 10.3389/fonc.2022.1002036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 11/03/2022] [Indexed: 12/05/2022] Open
Abstract
Microfibrillar-associated protein 2 (MFAP2), a component of the extracellular matrix, is important in controlling growth factor signal transduction. Recent studies have shown that MFAP2, an effective prognostic molecule for various tumors, is associated with tumor occurrence and development and may be involved in remodeling the extracellular matrix and regulating proliferation, apoptosis, invasion, tumor cell metastasis, and tumor angiogenesis. However, MFAP2’s specific mechanism in these tumor processes remains unclear. This article reviewed the possible mechanism of MFAP2 in tumorigenesis and progression and provided a reference for the clinical prognosis of patients with cancer and new therapeutic target discovery.
Collapse
|