1
|
Yay F, Ayan D. Bioinformatic analysis of neuropeptide related genes in patients diagnosed with invasive breast carcinoma. Comput Biol Med 2024; 183:109304. [PMID: 39437604 DOI: 10.1016/j.compbiomed.2024.109304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/28/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
PURPOSE Neuropeptide receptors are expressed in many malignancies. Effectors involved in the action mechanisms of HCRTR1, HCRTR2, NPY4R (PPYR1) may be related to breast cancer (BC). Genes encoding these receptors and PPY and PTPN11 genes were aimed to examine via bioinformatics tools in the BRCA cohort. To our knowledge, this is the first study in which these receptor genes and PP, which have not found much research in BC, are examined together with PTPN11 and analyzed comprehensively in large patient cohorts from public databases. METHODS cBioPortal was used for gene alteration analyses, GeneMania for association analyses with other genes, Kaplan-Meier Plotter for Overall Survival (OS) and Relapse Free Survival (RFS) analyses, UALCAN for methylation analyses, TIMER2.0 for expression analyses, The Human Protein Atlas database for expression validations, TIMER for immune infiltration analyses, WEKA 3.8.6 for diagnostic classification performances of the genes based on Random Forest Classifier and Enrichr-KG for Gene Ontology (GO) Biological Process (BP) and KEGG analysis. RESULTS 19 (1.9 %) nucleotide changes were found in 996 cases. Missense mutation is most common. Decreased expression levels of the HCRTR1 gene were associated with shorter OS and RFS, but decreased expression levels of the PTPN11 gene were associated with longer OS and RFS. Decreased expression levels NPY4R (PPYR1) gene were associated with shorter RFS. Increased expression levels of HCRTR2 and PPY genes were associated with longer RFS. HCRTR1 and NPY4R (PPYR1) genes were statistically hypermethylated; conversely HCRTR2 and PPY genes were hypomethylated. There was no significant change in PTPN11 gene promoter methylation level. HCRTR1, NPY4R (PPYR1) and PTPN11 gene expressions were downregulated; conversely, HCRTR2 and PPY gene expressions upregulated. Weak correlations were observed between NPY4R (PPYR1) gene expression and CD4+ T Cell, Neutrophil, Dendritic Cell and between PTPN11 gene expression and CD8+ T Cell, Macrophage, Neutrophil, Dendritic Cell infiltrations. Area under the receiver operating characteristics curve values of the 10-fold cross-validation and by splitting the dataset in a ratio of 80:20 models were 0.930 and 0.963 respectively. HCRTR2 and HCRTR1 belong to regulation of cytosolic calcium ion concentration, cellular calcium ion homeostasis GO BPs. CONCLUSION In BC patients, increases in HCRTR2 and PPY gene expressions could be considered as positive prognostic factors. Decreases in HCRTR1 and NPY4R (PPYR1) gene expressions could be considered as negative prognostic factors. Decreased expression of PTPN11 gene may have a positive prognostic factor. Changes in existing genes are likely to be both a biomarker and therapeutic target for BC. However, experimental and clinical studies are needed to elucidate the mechanisms underlying these neuropeptide receptors in terms of breast carcinogenesis.
Collapse
Affiliation(s)
- Fatih Yay
- Nigde Omer Halisdemir University Training and Research Hospital, Clinical Biochemistry Laboratory, Nigde, Turkey.
| | - Durmus Ayan
- Nigde Omer Halisdemir University Training and Research Hospital, Clinical Biochemistry Laboratory, Nigde, Turkey; Nigde Omer Halisdemir University, Faculty of Medicine, Medical Biochemistry, Nigde, Turkey.
| |
Collapse
|
2
|
Kojja V, Rudraram V, Kancharla B, Siva H, Tangutur AD, Nayak PK. Identification of phytoestrogens as sirtuin inhibitor against breast cancer: Multitargeted approach. Comput Biol Chem 2024; 112:108168. [PMID: 39127010 DOI: 10.1016/j.compbiolchem.2024.108168] [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: 06/04/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024]
Abstract
Despite progress in diagnosis and treatment strategies, breast cancer remains a primary risk to female health as indicated by second most cancer-deaths globally caused by this cancer. High risk mutation is linked to prognosis of breast cancer. Due to high resistance of breast cancer against current therapies, there is necessity of novel treatment strategies. Sirtuins are signaling proteins belonging to histone deacetylase class III family, known to control several cellular processes. Therefore, targeting sirtuins could be one of the approaches to treat breast cancer. Several plants synthesize phytoestrogens which exhibit structural and physiological similarities to estrogens and have been recognized to possess anticancer activity. In our study, we investigated several phytoestrogens for sirtuin inhibition by conducting molecular docking studies, and in-vitro studies against breast cancer cell lines. In molecular docking studies, we identified coumestrol possessing high binding energy with sirtuin proteins 1-3 as compared to other phytoestrogens. The molecular dynamic studies showed stable interaction of ligand and protein with higher affinity at sirtuin proteins 1-3 binding sites. In cell proliferation assay and colony formation assay using breast cancer cell lines (MCF-7 and MDAMB-231) coumestrol caused significant reduction in cell proliferation and number of colonies formed. Further, the flow cytometric analysis showed that coumestrol induces intracellular reactive oxygen species and the western blot analysis revealed reduction in the level of SIRT-1 expression in breast cancer cell lines. In conclusion, in-silico data and in-vitro studies suggest that the phytoestrogen coumestrol has sirtuin inhibitory activity against breast cancer.
Collapse
Affiliation(s)
- Venkateswarlu Kojja
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, India
| | - Vanitha Rudraram
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Bhanukiran Kancharla
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, India
| | - Hemalatha Siva
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, India
| | - Anjana Devi Tangutur
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India.
| | - Prasanta Kumar Nayak
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
3
|
Salem MG, Alqahtani AM, Mali SN, Alshwyeh HA, Jawarkar RD, Altamimi AS, Alshawwa SZ, Al-Olayan E, Saied EM, Youssef MF. Synthesis and antiproliferative evaluation of novel 3,5,8-trisubstituted coumarins against breast cancer. Future Med Chem 2024; 16:1053-1073. [PMID: 38708686 PMCID: PMC11216633 DOI: 10.4155/fmc-2023-0375] [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: 12/11/2023] [Accepted: 02/13/2024] [Indexed: 05/07/2024] Open
Abstract
Aim: This study focused on designing and synthesizing novel derivatives of 3,5,8-trisubstituted coumarin. Results: The synthesized compounds, particularly compound 5, exhibited significant cytotoxic effects on MCF-7 cells, surpassing staurosporine, and reduced toxicity toward MCF-10A cells, highlighting potential pharmacological advantages. Further, compound 5 altered the cell cycle and significantly increased apoptosis in MCF-7 cells, involving both early (41.7-fold) and late stages (33-fold), while moderately affecting necrotic signaling. The antitumor activity was linked to a notable reduction (4.78-fold) in topoisomerase IIβ expression. Molecular modeling indicated compound 5's strong affinity for EGFR, human EGF2 and topoisomerase II proteins. Conclusion: These findings highlight compound 5 as a multifaceted antitumor agent for breast cancer.
Collapse
Affiliation(s)
- Manar G Salem
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| | - Alaa M Alqahtani
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
| | - Suraj N Mali
- School of Pharmacy, DY Patil Deemed to be University Sector 7, Nerul, Navi Mumbai, 400706, India
| | - Hussah Abdullah Alshwyeh
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, Dammam, 31441, Saudi Arabia
- Basic & Applied Scientific Research Centre, Imam Abdulrahman Bin Faisal University, PO Box 1982, Dammam, 31441, Saudi Arabia
| | - Rahul D Jawarkar
- Department of Medicinal Chemistry & Drug Discovery, Dr. Rajendra Gode Institute of Pharmacy, University Mardi Road, Amravati, 444603, India
| | - Abdulmalik S Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, PO Box 173, Alkharj, 11942, Saudi Arabia
| | - Samar Z Alshawwa
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, PO Box 84428, Riyadh, 11671, Saudi Arabia
| | - Ebtesam Al-Olayan
- Department of Zoology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Essa M Saied
- Chemistry Department (Biochemistry Division), Faculty of Science, Suez Canal University, Ismailia, 41522, Egypt
- Institute for Chemistry, Humboldt Universität zu Berlin, Brook-Taylor-Str. 2, Berlin, 12489, Germany
| | - Mohamed F Youssef
- Chemistry Department (Organic Chemistry Division), Faculty of Science, Suez Canal University, Ismailia, 41522, Egypt
| |
Collapse
|
4
|
Liu X, Wang LL, Duan CY, Rong YR, Liang YQ, Zhu QX, Hao GP, Wang FZ. Daurisoline inhibits proliferation, induces apoptosis, and enhances TRAIL sensitivity of breast cancer cells by upregulating DR5. Cell Biol Int 2024. [PMID: 38563483 DOI: 10.1002/cbin.12162] [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: 11/25/2023] [Revised: 02/27/2024] [Accepted: 03/17/2024] [Indexed: 04/04/2024]
Abstract
Daurisoline (DS) is an isoquinoline alkaloid that exerts anticancer activities in various cancer cells. However, the underlying mechanisms through which DS affects the survival of breast cancer cells remain poorly understood. Therefore, the present study was undertaken to investigate the potential anticancer effect of DS on breast cancer cells and reveal the mechanism underlying the enhanced tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis by DS. Cell counting kit-8 (CCK-8) and 5-ethynyl-2-deoxyuridine (EdU) assay were used to evaluate the ability of cell proliferation. Flow cytometry was selected to examine the cell cycle distribution. TUNEL assay was used to detect the cell apoptosis. The protein expression was measured by Western blot analysis. DS was found to reduce the cell viability and suppress the proliferation of MCF-7 and MDA-MB-231 cells by causing G1 phase cell cycle arrest. DS could trigger apoptosis by promoting the cleavage of caspase-8 and PARP. The phosphorylation of ERK, JNK, and p38MAPK was upregulated clearly following DS treatment. Notably, SP600125 (JNK inhibitor) pretreatment significantly abrogated DS-induced PARP cleavage. DS inactivated Akt/mTOR and Wnt/β-catenin signaling pathway and upregulated the expression of ER stress-related proteins. Additionally, DS amplified TRAIL-caused viability reduction and apoptosis in breast cancer cells. Mechanismly, DS upregulated the protein level of DR4 and DR5, and knockdown of DR5 attenuated the cotreatment-induced cleavage of PARP. Inhibition of JNK could block DS-induced upregulation of DR5. This study provides valuable insights into the mechanisms of DS inhibiting cell proliferation, triggering apoptosis, and enhancing TRAIL sensitivity of breast cancer cells.
Collapse
Affiliation(s)
- Xin Liu
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, People's Republic of China
| | - Lin-Lin Wang
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, People's Republic of China
| | - Cun-Yu Duan
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, People's Republic of China
| | - Yan-Ru Rong
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, People's Republic of China
| | - Ya-Qi Liang
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, People's Republic of China
| | - Qing-Xiang Zhu
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, People's Republic of China
| | - Gang-Ping Hao
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, People's Republic of China
| | - Feng-Ze Wang
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, People's Republic of China
- Center Laboratory, The Second Affiliated Hospital of Shandong First Medical University, Taian, People's Republic of China
| |
Collapse
|
5
|
Shahab M, Ziyu P, Waqas M, Zheng G, Bin Jardan YA, Fentahun Wondmie G, Bouhrhia M. Targeting human progesterone receptor (PR), through pharmacophore-based screening and molecular simulation revealed potent inhibitors against breast cancer. Sci Rep 2024; 14:6768. [PMID: 38514638 PMCID: PMC10958019 DOI: 10.1038/s41598-024-55321-0] [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: 09/13/2023] [Accepted: 02/22/2024] [Indexed: 03/23/2024] Open
Abstract
Breast cancer, the prevailing malignant tumor among women, is linked to progesterone and its receptor (PR) in both tumorigenesis and treatment responsiveness. Despite thorough investigation, the precise molecular mechanisms of progesterone in breast cancer remain unclear. The human progesterone receptor (PR) serves as an essential therapeutic target for breast cancer treatment, warranting the rapid design of small molecule therapeutics that can effectively inhibit HPR. By employing cutting-edge computational techniques like molecular screening, simulation, and free energy calculation, the process of identifying potential lead molecules from natural products has been significantly expedited. In this study, we employed pharmacophore-based virtual screening and molecular simulations to identify natural product-based inhibitors of human progesterone receptor (PR) in breast cancer treatment. High-throughput molecular screening of traditional Chinese medicine (TCM) and zinc databases was performed, leading to the identification of potential lead compounds. The analysis of binding modes for the top five compounds from both database provides valuable structural insights into the inhibition of HPR for breast cancer treatment. The top five hits exhibited enhanced stability and compactness compared to the reference compound. In conclusion, our study provides valuable insights for identifying and refining lead compounds as HPR inhibitors.
Collapse
Affiliation(s)
- Muhammad Shahab
- State Key Laboratories of Chemical Resources Engineering, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Peng Ziyu
- School of chemistry and chemical engineering, Wuhan University of Science and Technology, Wuhan, 430081, People's Republic of China
| | - Muhammad Waqas
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat Al-Mouz, 616, Nizwa, Oman
| | - Guojun Zheng
- State Key Laboratories of Chemical Resources Engineering, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China.
| | - Yousef A Bin Jardan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P. O. BOX 2455, 11451, Riyadh, Saudi Arabia
| | | | - Mohammed Bouhrhia
- Laboratory of Biotechnology and Natural Resources Valorization, Faculty of Sciences, Ibn Zohr University, 80060, Agadir, Morocco
| |
Collapse
|
6
|
Al Moteri M, Mahesh TR, Thakur A, Vinoth Kumar V, Khan SB, Alojail M. Enhancing accessibility for improved diagnosis with modified EfficientNetV2-S and cyclic learning rate strategy in women with disabilities and breast cancer. Front Med (Lausanne) 2024; 11:1373244. [PMID: 38515985 PMCID: PMC10954891 DOI: 10.3389/fmed.2024.1373244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 02/27/2024] [Indexed: 03/23/2024] Open
Abstract
Breast cancer, a prevalent cancer among women worldwide, necessitates precise and prompt detection for successful treatment. While conventional histopathological examination is the benchmark, it is a lengthy process and prone to variations among different observers. Employing machine learning to automate the diagnosis of breast cancer presents a viable option, striving to improve both precision and speed. Previous studies have primarily focused on applying various machine learning and deep learning models for the classification of breast cancer images. These methodologies leverage convolutional neural networks (CNNs) and other advanced algorithms to differentiate between benign and malignant tumors from histopathological images. Current models, despite their potential, encounter obstacles related to generalizability, computational performance, and managing datasets with imbalances. Additionally, a significant number of these models do not possess the requisite transparency and interpretability, which are vital for medical diagnostic purposes. To address these limitations, our study introduces an advanced machine learning model based on EfficientNetV2. This model incorporates state-of-the-art techniques in image processing and neural network architecture, aiming to improve accuracy, efficiency, and robustness in classification. We employed the EfficientNetV2 model, fine-tuned for the specific task of breast cancer image classification. Our model underwent rigorous training and validation using the BreakHis dataset, which includes diverse histopathological images. Advanced data preprocessing, augmentation techniques, and a cyclical learning rate strategy were implemented to enhance model performance. The introduced model exhibited remarkable efficacy, attaining an accuracy rate of 99.68%, balanced precision and recall as indicated by a significant F1 score, and a considerable Cohen's Kappa value. These indicators highlight the model's proficiency in correctly categorizing histopathological images, surpassing current techniques in reliability and effectiveness. The research emphasizes improved accessibility, catering to individuals with disabilities and the elderly. By enhancing visual representation and interpretability, the proposed approach aims to make strides in inclusive medical image interpretation, ensuring equitable access to diagnostic information.
Collapse
Affiliation(s)
- Moteeb Al Moteri
- Department of Management Information Systems, College of Business Administration, King Saud University, Riyadh, Saudi Arabia
| | - T. R. Mahesh
- Department of Computer Science and Engineering, Faculty of Engineering and Technology, JAIN (Deemed-to-be University), Bangalore, India
| | - Arastu Thakur
- Department of Computer Science and Engineering, Faculty of Engineering and Technology, JAIN (Deemed-to-be University), Bangalore, India
| | - V. Vinoth Kumar
- School of Computer Science Engineering and Information Systems, Vellore Institute of Technology, Vellore, India
| | - Surbhi Bhatia Khan
- Department of Data Science, School of Science Engineering and Environment, University of Salford, Manchester, United Kingdom
- Department of Electrical and Computer Engineering, Lebanese American University, Byblos, Lebanon
| | - Mohammed Alojail
- Department of Management Information Systems, College of Business Administration, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
7
|
Mishra Y, Ranjan A, Mishra V, Chattaraj A, Aljabali AAA, El-Tanani M, Hromić-Jahjefendić A, Uversky VN, Tambuwala MM. The role of the gut microbiome in gastrointestinal cancers. Cell Signal 2024; 115:111013. [PMID: 38113978 DOI: 10.1016/j.cellsig.2023.111013] [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: 09/11/2023] [Revised: 12/06/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023]
Abstract
The gut microbiota present in the human digestive system is incredibly varied and is home to trillions of microorganisms. The gut microbiome is shaped at birth, while numerous genetic, dietary, and environmental variables primarily influence the microbiome composition. The importance of gut microbiota on host health is becoming more widely acknowledged. Digestion, intestinal permeability, and immunological and metabolism responses can all be affected by changes in the composition and function of the gut microbiota. There is mounting evidence that the microbial population's complex traits are important biomarkers and indicators of patient outcomes in cancer and its therapies. Numerous studies have demonstrated that changed commensal gut microorganisms contribute to the development and spread of cancer through various routes. Despite the ongoing controversy surrounding the gut microbiome and gastrointestinal cancer, accumulating evidence points to a potentially far more intricate connection than a simple cause-and-effect relationship. SIMPLE SUMMARY: Due to their high frequency and fatality rate, gastrointestinal cancers are regarded as a severe public health issue with complex medical and economic burdens. The gut microbiota may directly or indirectly interact with existing therapies like immunotherapy and chemotherapy, affecting how well a treatment works. The gut microbiome influences the immune response's activity, function, and development. Generally, certain gut bacteria impact the antitumor actions during cancer by creating particular metabolites or triggering T-cell responses. Yet, certain bacterial species have been found to promote cellular proliferation and metastasis in cancer, and comprehending these interactions in the context of cancer may help identify possible treatment targets. Notwithstanding the improvements in the field, additional research is still required to comprehend the underlying processes, examine the effects on existing therapies, and pinpoint certain bacteria and immune cells that can cause this interaction.
Collapse
Affiliation(s)
- Yachana Mishra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Abhigyan Ranjan
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Aditi Chattaraj
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Alaa A A Aljabali
- Department of Pharmaceutical Sciences, Yarmouk University, Irbid, Jordan
| | - Mohamed El-Tanani
- College of Pharmacy, Ras Alkhama Medical and Health Sciences University, United Arab Emirates
| | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka cesta 15, Sarajevo 71000, Bosnia and Herzegovina
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool, Lincoln LN6 7TS, England, United Kingdom.
| |
Collapse
|
8
|
Jabeen K, Khan MA, Hameed MA, Alqahtani O, Alouane MTH, Masood A. A novel fusion framework of deep bottleneck residual convolutional neural network for breast cancer classification from mammogram images. Front Oncol 2024; 14:1347856. [PMID: 38454931 PMCID: PMC10917916 DOI: 10.3389/fonc.2024.1347856] [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: 12/05/2023] [Accepted: 02/05/2024] [Indexed: 03/09/2024] Open
Abstract
With over 2.1 million new cases of breast cancer diagnosed annually, the incidence and mortality rate of this disease pose severe global health issues for women. Identifying the disease's influence is the only practical way to lessen it immediately. Numerous research works have developed automated methods using different medical imaging to identify BC. Still, the precision of each strategy differs based on the available resources, the issue's nature, and the dataset being used. We proposed a novel deep bottleneck convolutional neural network with a quantum optimization algorithm for breast cancer classification and diagnosis from mammogram images. Two novel deep architectures named three-residual blocks bottleneck and four-residual blocks bottle have been proposed with parallel and single paths. Bayesian Optimization (BO) has been employed to initialize hyperparameter values and train the architectures on the selected dataset. Deep features are extracted from the global average pool layer of both models. After that, a kernel-based canonical correlation analysis and entropy technique is proposed for the extracted deep features fusion. The fused feature set is further refined using an optimization technique named quantum generalized normal distribution optimization. The selected features are finally classified using several neural network classifiers, such as bi-layered and wide-neural networks. The experimental process was conducted on a publicly available mammogram imaging dataset named INbreast, and a maximum accuracy of 96.5% was obtained. Moreover, for the proposed method, the sensitivity rate is 96.45, the precision rate is 96.5, the F1 score value is 96.64, the MCC value is 92.97%, and the Kappa value is 92.97%, respectively. The proposed architectures are further utilized for the diagnosis process of infected regions. In addition, a detailed comparison has been conducted with a few recent techniques showing the proposed framework's higher accuracy and precision rate.
Collapse
Affiliation(s)
- Kiran Jabeen
- Department of Computer Science, HITEC University, Taxila, Pakistan
| | - Muhammad Attique Khan
- Department of Computer Science, HITEC University, Taxila, Pakistan
- Department of Computer Science and Mathematics, Lebanese American University, Beirut, Lebanon
| | - Mohamed Abdel Hameed
- Department of Computer Science, Faculty of Computers and Information, Luxor University, Luxor, Egypt
| | - Omar Alqahtani
- College of Computer Science, King Khalid University, Abha, Saudi Arabia
| | | | - Anum Masood
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
9
|
Kashyap MK, Mangrulkar SV, Kushwaha S, Ved A, Kale MB, Wankhede NL, Taksande BG, Upaganlawar AB, Umekar MJ, Koppula S, Kopalli SR. Recent Perspectives on Cardiovascular Toxicity Associated with Colorectal Cancer Drug Therapy. Pharmaceuticals (Basel) 2023; 16:1441. [PMID: 37895912 PMCID: PMC10610064 DOI: 10.3390/ph16101441] [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: 09/08/2023] [Revised: 10/06/2023] [Accepted: 10/08/2023] [Indexed: 10/29/2023] Open
Abstract
Cardiotoxicity is a well-known adverse effect of cancer-related therapy that has a significant influence on patient outcomes and quality of life. The use of antineoplastic drugs to treat colorectal cancers (CRCs) is associated with a number of undesirable side effects including cardiac complications. For both sexes, CRC ranks second and accounts for four out of every ten cancer deaths. According to the reports, almost 39% of patients with colorectal cancer who underwent first-line chemotherapy suffered cardiovascular impairment. Although 5-fluorouracil is still the backbone of chemotherapy regimen for colorectal, gastric, and breast cancers, cardiotoxicity caused by 5-fluorouracil might affect anywhere from 1.5% to 18% of patients. The precise mechanisms underlying cardiotoxicity associated with CRC treatment are complex and may involve the modulation of various signaling pathways crucial for maintaining cardiac health including TKI ErbB2 or NRG-1, VEGF, PDGF, BRAF/Ras/Raf/MEK/ERK, and the PI3/ERK/AMPK/mTOR pathway, resulting in oxidative stress, mitochondrial dysfunction, inflammation, and apoptosis, ultimately damaging cardiac tissue. Thus, the identification and management of cardiotoxicity associated with CRC drug therapy while minimizing the negative impact have become increasingly important. The purpose of this review is to catalog the potential cardiotoxicities caused by anticancer drugs and targeted therapy used to treat colorectal cancer as well as strategies focused on early diagnosing, prevention, and treatment of cardiotoxicity associated with anticancer drugs used in CRC therapy.
Collapse
Affiliation(s)
- Monu Kumar Kashyap
- Goel Institute of Pharmaceutical Sciences, Faizabad Road, Lucknow 226028, Uttar Pradesh, India;
- Dr. A. P. J. Abdul Kalam Technical University, Lucknow 222001, Uttar Pradesh, India;
| | - Shubhada V. Mangrulkar
- Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur 441002, Maharashtra, India; (S.V.M.); (M.B.K.); (N.L.W.)
| | - Sapana Kushwaha
- National Institute of Pharmaceutical Education and Research, Raebareli 229010, Uttar Pradesh, India
| | - Akash Ved
- Dr. A. P. J. Abdul Kalam Technical University, Lucknow 222001, Uttar Pradesh, India;
| | - Mayur B. Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur 441002, Maharashtra, India; (S.V.M.); (M.B.K.); (N.L.W.)
| | - Nitu L. Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur 441002, Maharashtra, India; (S.V.M.); (M.B.K.); (N.L.W.)
| | - Brijesh G. Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur 441002, Maharashtra, India; (S.V.M.); (M.B.K.); (N.L.W.)
| | - Aman B. Upaganlawar
- SNJB’s Shriman Sureshdada Jain Collge of Pharmacy, Neminagar, Chandwad, Nadik 423101, Maharashtra, India;
| | - Milind J. Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur 441002, Maharashtra, India; (S.V.M.); (M.B.K.); (N.L.W.)
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si 27478, Chungcheongbuk Do, Republic of Korea
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| |
Collapse
|
10
|
Ye F, Dewanjee S, Li Y, Jha NK, Chen ZS, Kumar A, Vishakha, Behl T, Jha SK, Tang H. Advancements in clinical aspects of targeted therapy and immunotherapy in breast cancer. Mol Cancer 2023; 22:105. [PMID: 37415164 PMCID: PMC10324146 DOI: 10.1186/s12943-023-01805-y] [Citation(s) in RCA: 98] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/08/2023] [Indexed: 07/08/2023] Open
Abstract
Breast cancer is the second leading cause of death for women worldwide. The heterogeneity of this disease presents a big challenge in its therapeutic management. However, recent advances in molecular biology and immunology enable to develop highly targeted therapies for many forms of breast cancer. The primary objective of targeted therapy is to inhibit a specific target/molecule that supports tumor progression. Ak strain transforming, cyclin-dependent kinases, poly (ADP-ribose) polymerase, and different growth factors have emerged as potential therapeutic targets for specific breast cancer subtypes. Many targeted drugs are currently undergoing clinical trials, and some have already received the FDA approval as monotherapy or in combination with other drugs for the treatment of different forms of breast cancer. However, the targeted drugs have yet to achieve therapeutic promise against triple-negative breast cancer (TNBC). In this aspect, immune therapy has come up as a promising therapeutic approach specifically for TNBC patients. Different immunotherapeutic modalities including immune-checkpoint blockade, vaccination, and adoptive cell transfer have been extensively studied in the clinical setting of breast cancer, especially in TNBC patients. The FDA has already approved some immune-checkpoint blockers in combination with chemotherapeutic drugs to treat TNBC and several trials are ongoing. This review provides an overview of clinical developments and recent advancements in targeted therapies and immunotherapies for breast cancer treatment. The successes, challenges, and prospects were critically discussed to portray their profound prospects.
Collapse
Affiliation(s)
- Feng Ye
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Yuehua Li
- Department of Medical Oncology, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
- School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, 144411, India
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, 11439, USA
| | - Ankush Kumar
- Pharmaceutical and Health Sciences, Career Point University, Hamirpur, Himachal Pradesh, India
| | - Vishakha
- Pharmaceutical and Health Sciences, Career Point University, Hamirpur, Himachal Pradesh, India
| | - Tapan Behl
- School of Health Sciences and Technology, University of Petroleum and Energy Studies, Bidholi, Dehradun, Uttarakhand, India.
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India.
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, 140413, India.
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun, 248007, India.
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
11
|
Xie Y, Chen Y, Wang Q, Li B, Shang H, Jing H. Early Prediction of Response to Neoadjuvant Chemotherapy Using Quantitative Parameters on Automated Breast Ultrasound Combined with Contrast-Enhanced Ultrasound in Breast Cancer. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:1638-1646. [PMID: 37100671 DOI: 10.1016/j.ultrasmedbio.2023.03.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 03/15/2023] [Accepted: 03/23/2023] [Indexed: 05/17/2023]
Abstract
OBJECTIVE This prospective study was aimed at evaluating the role of automated breast ultrasound (ABUS) and contrast-enhanced ultrasound (CEUS) in the early prediction of treatment response to neoadjuvant chemotherapy (NAC) in patients with breast cancer. METHODS Forty-three patients with pathologically confirmed invasive breast cancer treated with NAC were included. The standard for evaluation of response to NAC was based on surgery within 21 d of completing treatment. The patients were classified as having a pathological complete response (pCR) and a non-pCR. All patients underwent CEUS and ABUS 1 wk before receiving NAC and after two treatment cycles. The rising time (RT), time to peak (TTP), peak intensity (PI), wash-in slope (WIS) and wash-in area under the curve (Wi-AUC) were measured on the CEUS images before and after NAC. The maximum tumor diameters in the coronal and sagittal planes were measured on ABUS, and the tumor volume (V) was calculated. The difference (∆) in each parameter between the two treatment time points was compared. Binary logistic regression analysis was used to identify the predictive value of each parameter. RESULTS ∆V, ∆TTP and ∆PI were independent predictors of pCR. The CEUS-ABUS model achieved the highest AUC (0.950), followed by those based on CEUS (0.918) and ABUS (0.891) alone. CONCLUSION The CEUS-ABUS model could be used clinically to optimize the treatment of patients with breast cancer.
Collapse
Affiliation(s)
- Yongwei Xie
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yu Chen
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
| | - Qiucheng Wang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
| | - Bo Li
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
| | - Haitao Shang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hui Jing
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
12
|
Mishra Y, Chattaraj A, Mishra V, Ranjan A, Tambuwala MM. Aptamers Versus Vascular Endothelial Growth Factor (VEGF): A New Battle against Ovarian Cancer. Pharmaceuticals (Basel) 2023; 16:849. [PMID: 37375796 DOI: 10.3390/ph16060849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/28/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Cancer is one of the diseases that causes a high mortality as it involves unregulated and abnormal cell growth proliferation that can manifest in any body region. One of the typical ovarian cancer symptoms is damage to the female reproductive system. The death rate can be reduced through early detection of the ovarian cancer. Promising probes that can detect ovarian cancer are suitable aptamers. Aptamers, i.e., so-called chemical antibodies, have a strong affinity for the target biomarker and can typically be identified starting from a random library of oligonucleotides. Compared with other probes, ovarian cancer targeting using aptamers has demonstrated superior detection effectiveness. Various aptamers have been selected to detect the ovarian tumor biomarker, vascular endothelial growth factor (VEGF). The present review highlights the development of particular aptamers that target VEGF and detect ovarian cancer at its earliest stages. The therapeutic efficacy of aptamers in ovarian cancer treatment is also discussed.
Collapse
Affiliation(s)
- Yachana Mishra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Aditi Chattaraj
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Abhigyan Ranjan
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool, Lincoln LN6 7TS, UK
| |
Collapse
|