1
|
Wozniak S, Janson G, Feig M. Accurate Predictions of Molecular Properties of Proteins via Graph Neural Networks and Transfer Learning. J Chem Theory Comput 2025; 21:4830-4845. [PMID: 40270304 PMCID: PMC12080100 DOI: 10.1021/acs.jctc.4c01682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/14/2025] [Accepted: 04/15/2025] [Indexed: 04/25/2025]
Abstract
Machine learning has emerged as a promising approach for predicting molecular properties of proteins, as it addresses limitations of experimental and traditional computational methods. Here, we introduce GSnet, a graph neural network (GNN) trained to predict physicochemical and geometric properties including solvation-free energies, diffusion constants, and hydrodynamic radii, based on three-dimensional protein structures. By leveraging transfer learning, pretrained GSnet embeddings were adapted to predict solvent-accessible surface area (SASA) and residue-specific pKa values, achieving high accuracy and generalizability. Notably, GSnet outperformed existing protein embeddings for SASA prediction and a locally charge-aware variant, aLCnet, approached the accuracy of simulation-based and empirical methods for pKa prediction. Our GNN framework demonstrated robustness across diverse data sets, including intrinsically disordered peptides, and scalability for high-throughput applications. These results highlight the potential of GNN-based embeddings and transfer learning to advance protein structure analysis, providing a foundation for integrating predictive models into proteome-wide studies and structural biology pipelines.
Collapse
Affiliation(s)
- Spencer Wozniak
- Department of Biochemistry
and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Giacomo Janson
- Department of Biochemistry
and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Michael Feig
- Department of Biochemistry
and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
2
|
Mao Y, Shangguan D, Huang Q, Xiao L, Cao D, Zhou H, Wang YK. Emerging artificial intelligence-driven precision therapies in tumor drug resistance: recent advances, opportunities, and challenges. Mol Cancer 2025; 24:123. [PMID: 40269930 PMCID: PMC12016295 DOI: 10.1186/s12943-025-02321-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/02/2025] [Indexed: 04/25/2025] Open
Abstract
Drug resistance is one of the main reasons for cancer treatment failure, leading to a rapid recurrence/disease progression of the cancer. Recently, artificial intelligence (AI) has empowered physicians to use its powerful data processing and pattern recognition capabilities to extract and mine valuable drug resistance information from large amounts of clinical or omics data, to study drug resistance mechanisms, to evaluate and predict drug resistance, and to develop innovative therapeutic strategies to reduce drug resistance. In this review, we proposed a feasible workflow for incorporating AI into tumor drug resistance research, highlighted current AI-driven tumor drug resistance applications, and discussed the opportunities and challenges encountered in the process. Based on a comprehensive literature analysis, we systematically summarized the role of AI in tumor drug resistance research, including drug development, resistance mechanism elucidation, drug sensitivity prediction, combination therapy optimization, resistance phenotype identification, and clinical biomarker discovery. With the continuous advancement of AI technology and rigorous validation of clinical data, AI models are expected to fuel the development of precision oncology by improving efficacy, guiding therapeutic decisions, and optimizing patient prognosis. In summary, by leveraging clinical and omics data, AI models are expected to pioneer new therapy strategies to mitigate tumor drug resistance, improve efficacy and patient survival, and provide novel perspectives and tools for oncology treatment.
Collapse
Affiliation(s)
- Yuan Mao
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Lymphoma and Hematology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China
| | - Dangang Shangguan
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Qi Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ling Xiao
- Department of Histology and Embryology of Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China
| | - Dongsheng Cao
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Hui Zhou
- Department of Lymphoma and Hematology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China.
- Department of Lymphoma and Hematology, Hunan Cancer Hospital, Changsha, Hunan, People's Republic of China.
| | - Yi-Kun Wang
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
3
|
Zhang L, Wang Y, Li Y, Chen ZS, Hu C. Advanced materials for cancer treatment and beyond. Front Pharmacol 2025; 16:1557155. [PMID: 40110134 PMCID: PMC11920709 DOI: 10.3389/fphar.2025.1557155] [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: 01/08/2025] [Accepted: 02/13/2025] [Indexed: 03/22/2025] Open
Abstract
Conservative anti-cancer treatment represented by chemotherapy and surgery lacks tumor-specificity and could hardly resolve the problems associated with multidrug resistance (MDR) in cancers. Novel therapeutic materials in cancer treatment, such as those with anti-MDR or controllable treatment features, represent a significant trend due to their advantages of high and specific efficacy and timely intervention of cancer progress. In addition to their excellent biocompatibility and specificity, they can be utilized in therapies that require ease of operation, provided they are designed with high detection sensitivity. In this review, we summarize a series of recently developed materials that exhibit these advantages, including immune-enhancing and tumor microenvironment (TME)- responsive materials, and those with integrated therapeutic and imaging capabilities. We also introduce advanced modification approaches that can impart essential targeting functionalities to these materials.
Collapse
Affiliation(s)
- Lei Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, China
| | - Yanan Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yangjia Li
- National Engineering Research Center for Sugarcane, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Chaohua Hu
- National Engineering Research Center for Sugarcane, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
4
|
Xue J, Li Q, Wang Y, Yin R, Zhang J. Insight into the structure, oligomerization, and the role in drug resistance of human UDP-glucuronosyltransferases. Arch Toxicol 2025; 99:1153-1165. [PMID: 39812829 DOI: 10.1007/s00204-024-03929-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 11/28/2024] [Indexed: 01/16/2025]
Abstract
Human UDP-glucuronosyltransferases (UGTs) are pivotal phase II metabolic enzymes facilitating the transfer of glucuronic acid from UDP-glucuronic acid (UDPGA) to various substrates. UGTs are classic type I transmembrane glycoproteins, mainly localized in the endoplasmic reticulum (ER) membrane. This review comprehensively explores UGTs, encompassing gene expression, functional characteristics, substrate specificity, and metabolic mechanisms. A recent analysis of C-terminal structures, compared with original data, underscores the pivotal role of α3, α4, and β4 functional domains in selectively recognizing diverse glycosyl donors. Accumulating evidence suggests that UGTs function as homo- and heterodimers, with oligomers likely stabilizing UGTs and modulating their activity. The review sheds light on the implications of UGT oligomerization on substrate glucuronidation and the interplay between protein-protein interaction and glucuronidation activity. UGT-mediated drug resistance, often underestimated, emerges as a clinically relevant form of chemical resistance, with delineated outcomes in tumors and other diseases. This review provides a multifaceted exploration of the physiological significance of UGTs, spanning genetics, proteins, oligomerization, drug resistance, and more, offering insights into their metabolic mechanisms. Understanding interactions between UGT isoforms is crucial for predicting drug-drug interactions, preventing drug toxicity, and enabling precision treatment.
Collapse
Affiliation(s)
- Jia Xue
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiuyi Li
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yao Wang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ruxi Yin
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jian Zhang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
5
|
Kerestély M, Keresztes D, Szarka L, Kovács BM, Schulc K, Veres DV, Csermely P. System level network data and models attack cancer drug resistance. Br J Pharmacol 2025. [PMID: 39909489 DOI: 10.1111/bph.17469] [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: 12/03/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 02/07/2025] Open
Abstract
Drug resistance is responsible for >90% of cancer related deaths. Cancer drug resistance is a system level network phenomenon covering the entire cell. Small-scale interactomes and signalling network models of drug resistance guide directed drug development. Recently, proteome-wide human interactome and signalling network data have become available, which have been extended by drug-target interactions, drug resistance-inducing mutations, as well as by several cancer and drug resistance-related multi-omics datasets. System level signalling network models have become available examining therapy resistance, performing in silico clinical trials, and conducting large, in silico drug combination screens. Drug resistance network data and models have become interoperable and reliable. These advances paved the road for building proteome-wide drug resistance models.
Collapse
Affiliation(s)
- Márk Kerestély
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Dávid Keresztes
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Levente Szarka
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Borbála M Kovács
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Klára Schulc
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
- Department of Internal Medicine and Oncology, Division of Oncology, Semmelweis University, Budapest, Hungary
| | - Dániel V Veres
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
- Turbine Simulated Cell Technologies, Budapest, Hungary
| | - Peter Csermely
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
6
|
Keresztes D, Kerestély M, Szarka L, Kovács BM, Schulc K, Veres DV, Csermely P. Cancer drug resistance as learning of signaling networks. Biomed Pharmacother 2025; 183:117880. [PMID: 39884030 DOI: 10.1016/j.biopha.2025.117880] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/08/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025] Open
Abstract
Drug resistance is a major cause of tumor mortality. Signaling networks became useful tools for driving pharmacological interventions against cancer drug resistance. Signaling datasets now cover the entire human cell. Recently, network adaptation became understood as a learning process. We review rapidly increasing evidence showing that the development of cancer drug resistance can be described as learning of signaling networks. During drug adaptation, the network forgets drug-affected pathways by desensitization and relearns by strengthening alternative pathways. Thus, resistant cancer cells develop a drug resistance memory. We show that all key players of cellular learning (i.e., IDPs, protein translocation, microRNAs/lncRNAs, scaffolding proteins and epigenetic/chromatin memory) have important roles in the development of cancer drug resistance. Moreover, all of them are central components of the epithelial-mesenchymal transition leading to metastases and resistance. Phenotypic plasticity was recently listed as a hallmark of cancer. We review how network plasticity induces rare, pre-existent drug-resistant cells in the absence of drug treatment. Key network methods assessing the development of drug resistance and network pharmacological interventions against drug resistance are summarized. Finally, we highlight the class of cellular memory drugs affecting cellular learning and forgetting, and we summarize current challenges to prevent or break drug resistance using network models.
Collapse
Affiliation(s)
- Dávid Keresztes
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Márk Kerestély
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Levente Szarka
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Borbála M Kovács
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Klára Schulc
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary; Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
| | - Dániel V Veres
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary; Turbine Simulated Cell Technologies, Budapest, Hungary
| | - Peter Csermely
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
7
|
Lin JY, Lai JK, Chen JY, Cai JY, Yang ZD, Yang LQ, Zheng ZT, Guo XG. Global insights into MRSA bacteremia: a bibliometric analysis and future outlook. Front Microbiol 2025; 15:1516584. [PMID: 39911705 PMCID: PMC11794302 DOI: 10.3389/fmicb.2024.1516584] [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: 10/29/2024] [Accepted: 12/23/2024] [Indexed: 02/07/2025] Open
Abstract
Background Methicillin-resistant Staphylococcus aureus (MRSA) bloodstream infections (BSIs) pose a significant challenge to global public health, characterized by high morbidity and mortality rates, particularly in immunocompromised patients. Despite extensive research, the rapid development of MRSA antibiotic resistance has outpaced current treatment methods, increasing the difficulty of treatment. Therefore, reviewing research on MRSA BSIs is crucial. Methods This study conducted a bibliometric analysis, retrieving and analyzing 1,621 publications related to MRSA BSIs from 2006 to 2024. The literature was sourced from the Web of Science Core Collection (WoSCC), and data visualization and trend analysis were performed using VOSviewer, CiteSpace, and Bibliometrix software packages. Results The bibliometric analysis showed that research on MRSA BSIs was primarily concentrated in the United States, China, and Japan. The United States leads in research output and influence, with significant contributions from institutions such as the University of California system and the University of Texas system. The journal with the most publications is Antimicrobial Agents and Chemotherapy, while the most cited global publication is Vincent JL's article "Sepsis in European Intensive Care Units: Results of the SOAP Study" published in Critical Care Medicine in 2006. Cosgrove SE's article "Comparison of Mortality Associated with Methicillin-Resistant and Methicillin-Susceptible Staphylococcus aureus Bacteremia: A Meta-analysis" had the most co-citations. Key trends in the research include MRSA's antibiotic resistance mechanisms, the application of new diagnostic technologies, and the impact of COVID-19 on MRSA studies. Additionally, artificial intelligence (AI) and machine learning are increasingly applied in MRSA diagnosis and treatment, and phage therapy and vaccine development have become future research hotspots. Conclusion Methicillin-resistant Staphylococcus aureus BSIs remain a major global public health challenge, especially with the increasing severity of antibiotic resistance. Although progress has been made in new treatments and diagnostic technologies, further validation is required. Future research will rely on integrating genomics, AI, and machine learning to drive personalized treatment. Strengthening global cooperation, particularly in resource-limited countries, will be key to effectively addressing MRSA BSIs.
Collapse
Affiliation(s)
- Jia-Yi Lin
- Department of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Guangzhou Medical University, Guangzhou, China
| | - Jia-Kai Lai
- Department of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Biomedical Engineering, The School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, China
| | - Jian-Yi Chen
- Department of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Guangzhou Medical University, Guangzhou, China
| | - Jia-Yu Cai
- Department of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Clinical Medicine, The Third School of Clinical Medicine, Guangzhou Medical University, Guangzhou, China
| | - Zhan-Dong Yang
- Department of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Guangzhou Medical University, Guangzhou, China
| | - Liu-Qingqing Yang
- Department of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Ze-Tao Zheng
- Department of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Guangzhou Medical University, Guangzhou, China
| | - Xu-Guang Guo
- Department of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Clinical Medicine, The Third School of Clinical Medicine, Guangzhou Medical University, Guangzhou, China
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
8
|
Wozniak S, Janson G, Feig M. Accurate Predictions of Molecular Properties of Proteins via Graph Neural Networks and Transfer Learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.10.627714. [PMID: 39713395 PMCID: PMC11661272 DOI: 10.1101/2024.12.10.627714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Machine learning has emerged as a promising approach for predicting molecular properties of proteins, as it addresses limitations of experimental and traditional computational methods. Here, we introduce GSnet, a graph neural network (GNN) trained to predict physicochemical and geometric properties including solvation free energies, diffusion constants, and hydrodynamic radii, based on three-dimensional protein structures. By leveraging transfer learning, pre-trained GSnet embeddings were adapted to predict solvent-accessible surface area (SASA) and residue-specific pKa values, achieving high accuracy and generalizability. Notably, GSnet outperformed existing protein embeddings for SASA prediction, and a locally charge-aware variant, aLCnet, approached the accuracy of simulation-based and empirical methods for pKa prediction. Our GNN framework demonstrated robustness across diverse datasets, including intrinsically disordered peptides, and scalability for high-throughput applications. These results highlight the potential of GNN-based embeddings and transfer learning to advance protein structure analysis, providing a foundation for integrating predictive models into proteome-wide studies and structural biology pipelines.
Collapse
Affiliation(s)
- Spencer Wozniak
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| | - Giacomo Janson
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| | - Michael Feig
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
9
|
Asfa SS, Arshinchi Bonab R, Önder O, Uça Apaydın M, Döşeme H, Küçük C, Georgakilas AG, Stadler BM, Logotheti S, Kale S, Pavlopoulou A. Computer-Aided Identification and Design of Ligands for Multi-Targeting Inhibition of a Molecular Acute Myeloid Leukemia Network. Cancers (Basel) 2024; 16:3607. [PMID: 39518047 PMCID: PMC11544916 DOI: 10.3390/cancers16213607] [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: 08/29/2024] [Revised: 10/07/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES Acute myeloid leukemia (AML) is characterized by therapeutic failure and long-term risk for disease relapses. As several therapeutic targets participate in networks, they can rewire to eventually evade single-target drugs. Hence, multi-targeting approaches are considered on the expectation that interference with many different components could synergistically hinder activation of alternative pathways and demolish the network one-off, leading to complete disease remission. METHODS Herein, we established a network-based, computer-aided approach for the rational design of drug combinations and de novo agents that interact with many AML network components simultaneously. RESULTS A reconstructed AML network guided the selection of suitable protein hubs and corresponding multi-targeting strategies. For proteins responsive to existing drugs, a greedy algorithm identified the minimum amount of compounds targeting the maximum number of hubs. We predicted permissible combinations of amiodarone, artenimol, fostamatinib, ponatinib, procaine, and vismodegib that interfere with 3-8 hubs, and we elucidated the pharmacological mode of action of procaine on DNMT3A. For proteins that do not respond to any approved drugs, namely cyclins A1, D2, and E1, we used structure-based de novo drug design to generate a novel triple-targeting compound of the chemical formula C15H15NO5, with favorable pharmacological and drug-like properties. CONCLUSIONS Overall, by integrating network and structural pharmacology with molecular modeling, we determined two complementary strategies with the potential to annihilate the AML network, one in the form of repurposable drug combinations and the other as a de novo synthesized triple-targeting agent. These target-drug interactions could be prioritized for preclinical and clinical testing toward precision medicine for AML.
Collapse
Affiliation(s)
- Seyedeh Sadaf Asfa
- Izmir Biomedicine and Genome Center, 35340 Balçova, İzmir, Türkiye; (S.S.A.); (R.A.B.); (O.Ö.); (M.U.A.); (H.D.); (S.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, 35340 Balçova, İzmir, Türkiye
- Department of Pharmacology and Therapeutics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R3E 0W2, Canada
| | - Reza Arshinchi Bonab
- Izmir Biomedicine and Genome Center, 35340 Balçova, İzmir, Türkiye; (S.S.A.); (R.A.B.); (O.Ö.); (M.U.A.); (H.D.); (S.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, 35340 Balçova, İzmir, Türkiye
- Department of Pharmacology and Therapeutics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R3E 0W2, Canada
| | - Onur Önder
- Izmir Biomedicine and Genome Center, 35340 Balçova, İzmir, Türkiye; (S.S.A.); (R.A.B.); (O.Ö.); (M.U.A.); (H.D.); (S.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, 35340 Balçova, İzmir, Türkiye
| | - Merve Uça Apaydın
- Izmir Biomedicine and Genome Center, 35340 Balçova, İzmir, Türkiye; (S.S.A.); (R.A.B.); (O.Ö.); (M.U.A.); (H.D.); (S.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, 35340 Balçova, İzmir, Türkiye
| | - Hatice Döşeme
- Izmir Biomedicine and Genome Center, 35340 Balçova, İzmir, Türkiye; (S.S.A.); (R.A.B.); (O.Ö.); (M.U.A.); (H.D.); (S.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, 35340 Balçova, İzmir, Türkiye
| | - Can Küçük
- Department of Medical Biology, Faculty of Medicine, Dokuz Eylül University, 35330 Balçova, İzmir, Türkiye;
| | - Alexandros G. Georgakilas
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou Campous, 15780 Athens, Greece;
| | - Bernhard M. Stadler
- Technische Hochschule Nürnberg, Faculty of Applied Chemistry, 90489 Nuremberg, Germany;
| | - Stella Logotheti
- Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
| | - Seyit Kale
- Izmir Biomedicine and Genome Center, 35340 Balçova, İzmir, Türkiye; (S.S.A.); (R.A.B.); (O.Ö.); (M.U.A.); (H.D.); (S.K.)
- Department of Biophysics, Faculty of Medicine, Izmir Katip Çelebi University, 35330 Çiğli, İzmir, Türkiye
| | - Athanasia Pavlopoulou
- Izmir Biomedicine and Genome Center, 35340 Balçova, İzmir, Türkiye; (S.S.A.); (R.A.B.); (O.Ö.); (M.U.A.); (H.D.); (S.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, 35340 Balçova, İzmir, Türkiye
| |
Collapse
|
10
|
Yu QX, Wu RC, Wang J, Tuo ZT, Yang J, Zhang YP, Jin J, Yuan Q, Wang CN, Feng DC, Li DX. Exploring the role of ADAMTSL2 across multiple cancer types: A pan-cancer analysis and validated in colorectal cancer. Discov Oncol 2024; 15:538. [PMID: 39384622 PMCID: PMC11465020 DOI: 10.1007/s12672-024-01401-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/25/2024] [Indexed: 10/11/2024] Open
Abstract
BACKGROUND Recent studies have established a correlation between ADAMTSL2 (ADAMTS-like 2) and the development of various cancers. This study aims to conduct a comprehensive pan-cancer analysis in 37 cancer types and investigate its potential role in colon and rectal adenocarcinoma (COADREAD). METHOD Pan-cancer and mutation data were sourced from The Cancer Genome Atlas (TCGA) database and analyzed using Sangerbox analysis platform. We explored the expression patterns and prognostic implications of ADAMTSL2, and investigated its relationships with tumor heterogeneity, stemness, immune checkpoint genes, immune cell infiltration, RNA modifications, and mutational profiles across different cancers. Additionally, with Ethics Committee approval, we conducted immunohistochemical (IHC) analysis on 120 COADEAD samples to evaluate ADAMTSL2 expression and its association with clinicopathological parameters. RESULTS ADAMTSL2 expression was positively correlated with the hazard ratio of OS, DSS, DFI and PFI for ESCA and COADREAD. A negative correlation was observed between ADAMTSL2 expression and NEO levels in COAD. Gene alterations in ADAMTSL2 were observed, with a mutation frequency of 5.0% in COAD. There is a significant correlation between ADAMTSL2 expression and immune cell infiltration in a variety of cancers. The expression level of ADAMTSL2 protein was associated with T stage, N stage, M stage (p < 0.05). Kaplan‒Meier survival curves demonstrated that the high ADAMTSL2 group had a shorter OS time (p = 0.047) and progression free survival time (p = 0.026) than the low ADAMTSL2 group. CONCLUSION In summary, we conducted a comprehensive pan-cancer analysis of ADAMTSL2 and we demonstrated that ADAMTSL2 may serve as a novel prognostic biomarker and immunotherapy target in COADREAD.
Collapse
Affiliation(s)
- Qing-Xin Yu
- Department of pathology, Ningbo Clinical Pathology Diagnosis center, Ningbo, 315211, Zhejiang, China
- Department of pathology, Ningbo Medical Centre Lihuili Hospital, Ningbo, 315040, Zhejiang, China
| | - Rui-Cheng Wu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jie Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zhou-Ting Tuo
- Department of Urological Surgery, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Jun Yang
- Department of pathology, Ningbo Clinical Pathology Diagnosis center, Ningbo, 315211, Zhejiang, China
- Department of pathology, Ningbo Medical Centre Lihuili Hospital, Ningbo, 315040, Zhejiang, China
| | - Yong-Ping Zhang
- Department of pathology, Ningbo Clinical Pathology Diagnosis center, Ningbo, 315211, Zhejiang, China
| | - Jing Jin
- Department of pathology, Ningbo Clinical Pathology Diagnosis center, Ningbo, 315211, Zhejiang, China
| | - Quan Yuan
- Department of pathology, Ningbo Clinical Pathology Diagnosis center, Ningbo, 315211, Zhejiang, China
| | - Chun-Nian Wang
- Department of pathology, Ningbo Clinical Pathology Diagnosis center, Ningbo, 315211, Zhejiang, China.
- Department of pathology, Ningbo Medical Centre Lihuili Hospital, Ningbo, 315040, Zhejiang, China.
| | - De-Chao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
- Division of Surgery & Interventional Science, University College London, London, W1W 7TS, UK.
| | - Deng-Xiong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
11
|
Hakami MA. Harnessing machine learning potential for personalised drug design and overcoming drug resistance. J Drug Target 2024; 32:918-930. [PMID: 38842417 DOI: 10.1080/1061186x.2024.2365934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/01/2024] [Accepted: 06/04/2024] [Indexed: 06/07/2024]
Abstract
Drug resistance in cancer treatment presents a significant challenge, necessitating innovative approaches to improve therapeutic efficacy. Integrating machine learning (ML) in cancer research is promising as ML algorithms outrival in analysing complex datasets, identifying patterns, and predicting treatment outcomes. Leveraging diverse data sources such as genomic profiles, clinical records, and drug response assays, ML uncovers molecular mechanisms of drug resistance, enabling personalised treatment, maximising efficacy and minimising adverse effects. Various ML algorithms contribute to the drug discovery process - Random Forest and Decision Trees predict drug-target interactions and aid in virtual screening, and SVM classify leads on bioactivity data. Neural Networks model QSAR to optimise lead compounds and K-means clustering group compounds with similar chemical properties aiding compound selection. Gaussian Processes predict drug responses, Bayesian Networks infer causal relationships, Autoencoders generate novel compounds, and Genetic Algorithms optimise molecular structures. These algorithms collectively enhance efficiency and success rates in drug design endeavours, from lead identification to optimisation and are cost-effective, empowering clinicians with real-time treatment monitoring and improving patient outcomes. This review highlights the immense potential of ML in revolutionising cancer care through effective drug design to reduce drug resistance, and we have also discussed various limitations and research gaps to understand better.
Collapse
Affiliation(s)
- Mohammed Ageeli Hakami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Al-Quwayiyah, Riyadh, Saudi Arabia
| |
Collapse
|
12
|
Amorim AM, Piochi LF, Gaspar AT, Preto A, Rosário-Ferreira N, Moreira IS. Advancing Drug Safety in Drug Development: Bridging Computational Predictions for Enhanced Toxicity Prediction. Chem Res Toxicol 2024; 37:827-849. [PMID: 38758610 PMCID: PMC11187637 DOI: 10.1021/acs.chemrestox.3c00352] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 04/29/2024] [Accepted: 05/07/2024] [Indexed: 05/19/2024]
Abstract
The attrition rate of drugs in clinical trials is generally quite high, with estimates suggesting that approximately 90% of drugs fail to make it through the process. The identification of unexpected toxicity issues during preclinical stages is a significant factor contributing to this high rate of failure. These issues can have a major impact on the success of a drug and must be carefully considered throughout the development process. These late-stage rejections or withdrawals of drug candidates significantly increase the costs associated with drug development, particularly when toxicity is detected during clinical trials or after market release. Understanding drug-biological target interactions is essential for evaluating compound toxicity and safety, as well as predicting therapeutic effects and potential off-target effects that could lead to toxicity. This will enable scientists to predict and assess the safety profiles of drug candidates more accurately. Evaluation of toxicity and safety is a critical aspect of drug development, and biomolecules, particularly proteins, play vital roles in complex biological networks and often serve as targets for various chemicals. Therefore, a better understanding of these interactions is crucial for the advancement of drug development. The development of computational methods for evaluating protein-ligand interactions and predicting toxicity is emerging as a promising approach that adheres to the 3Rs principles (replace, reduce, and refine) and has garnered significant attention in recent years. In this review, we present a thorough examination of the latest breakthroughs in drug toxicity prediction, highlighting the significance of drug-target binding affinity in anticipating and mitigating possible adverse effects. In doing so, we aim to contribute to the development of more effective and secure drugs.
Collapse
Affiliation(s)
- Ana M.
B. Amorim
- Department
of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CNC-UC—Center
for Neuroscience and Cell Biology, University
of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CIBB—Centre
for Innovative Biomedicine and Biotechnology, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- PhD
Programme in Biosciences, Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- PURR.AI,
Rua Pedro Nunes, IPN Incubadora, Ed C, 3030-199 Coimbra, Portugal
| | - Luiz F. Piochi
- Department
of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CNC-UC—Center
for Neuroscience and Cell Biology, University
of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CIBB—Centre
for Innovative Biomedicine and Biotechnology, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
| | - Ana T. Gaspar
- Department
of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CNC-UC—Center
for Neuroscience and Cell Biology, University
of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CIBB—Centre
for Innovative Biomedicine and Biotechnology, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
| | - António
J. Preto
- CNC-UC—Center
for Neuroscience and Cell Biology, University
of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CIBB—Centre
for Innovative Biomedicine and Biotechnology, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- PhD Programme
in Experimental Biology and Biomedicine, Institute for Interdisciplinary
Research (IIIUC), University of Coimbra, Casa Costa Alemão, 3030-789 Coimbra, Portugal
| | - Nícia Rosário-Ferreira
- CNC-UC—Center
for Neuroscience and Cell Biology, University
of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CIBB—Centre
for Innovative Biomedicine and Biotechnology, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
| | - Irina S. Moreira
- Department
of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CNC-UC—Center
for Neuroscience and Cell Biology, University
of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CIBB—Centre
for Innovative Biomedicine and Biotechnology, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
| |
Collapse
|
13
|
Cai Z, Peng H, Sun S, He J, Luo F, Huang Y. DeepKa Web Server: High-Throughput Protein p Ka Prediction. J Chem Inf Model 2024; 64:2933-2940. [PMID: 38530291 DOI: 10.1021/acs.jcim.3c02013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
DeepKa is a deep-learning-based protein pKa predictor proposed in our previous work. In this study, a web server was developed that enables online protein pKa prediction driven by DeepKa. The web server provides a user-friendly interface where a single step of entering a valid PDB code or uploading a PDB format file is required to submit a job. Two case studies have been attached in order to explain how pKa's calculated by the web server could be utilized by users. Finally, combining the web server with post processing as described in case studies, this work suggests a quick workflow of investigating the relationship between protein structure and function that are pH dependent. The web server of DeepKa is freely available at http://www.computbiophys.com/DeepKa/main.
Collapse
Affiliation(s)
- Zhitao Cai
- College of Computer Engineering, Jimei University, Xiamen 361021, China
| | - Hao Peng
- National Pilot School of Software, Yunnan University, Kunming 650504, China
| | - Shuo Sun
- College of Computer Engineering, Jimei University, Xiamen 361021, China
| | - Jiahao He
- College of Computer Engineering, Jimei University, Xiamen 361021, China
| | - Fangfang Luo
- College of Computer Engineering, Jimei University, Xiamen 361021, China
| | - Yandong Huang
- College of Computer Engineering, Jimei University, Xiamen 361021, China
| |
Collapse
|
14
|
Podolski-Renić A, Čipak Gašparović A, Valente A, López Ó, Bormio Nunes JH, Kowol CR, Heffeter P, Filipović NR. Schiff bases and their metal complexes to target and overcome (multidrug) resistance in cancer. Eur J Med Chem 2024; 270:116363. [PMID: 38593587 DOI: 10.1016/j.ejmech.2024.116363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/15/2024] [Accepted: 03/25/2024] [Indexed: 04/11/2024]
Abstract
Overcoming multidrug resistance (MDR) is one of the major challenges in cancer therapy. In this respect, Schiff base-related compounds (bearing a R1R2CNR3 bond) gained high interest during the past decades. Schiff bases are considered privileged ligands for various reasons, including the easiness of their preparation and the possibility to form complexes with almost all transition metal ions. Schiff bases and their metal complexes exhibit many types of biological activities and are used for the treatment and diagnosis of various diseases. Until now, 13 Schiff bases have been investigated in clinical trials for cancer treatment and hypoxia imaging. This review represents the first collection of Schiff bases and their complexes which demonstrated MDR-reversal activity. The areas of drug resistance covered in this article involve: 1) Modulation of ABC transporter function, 2) Targeting lysosomal ABCB1 overexpression, 3) Circumvention of ABC transporter-mediated drug efflux by alternative routes of drug uptake, 4) Selective activity against MDR cancer models (collateral sensitivity), 5) Targeting GSH-detoxifying systems, 6) Overcoming apoptosis resistance by inducing necrosis and paraptosis, 7) Reactivation of mutated p53, 8) Restoration of sensitivity to DNA-damaging anticancer therapy, and 9) Overcoming drug resistance through modulation of the immune system. Through this approach, we would like to draw attention to Schiff bases and their metal complexes representing highly interesting anticancer drug candidates with the ability to overcome MDR.
Collapse
Affiliation(s)
- Ana Podolski-Renić
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Serbia
| | | | - Andreia Valente
- Centro de Química Estrutural and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, Lisboa, Portugal
| | - Óscar López
- Departamento de Química Organica, Facultad de Química, Universidad de Sevilla, Sevilla, Spain
| | - Julia H Bormio Nunes
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria; Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Christian R Kowol
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Petra Heffeter
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| | - Nenad R Filipović
- Department of Chemistry and Biochemistry, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
15
|
Jiang D, Qian Q, Yang X, Zeng Y, Liu H. Machine learning based on optimal VOI of multi-sequence MR images to predict lymphovascular invasion in invasive breast cancer. Heliyon 2024; 10:e29267. [PMID: 38623213 PMCID: PMC11016709 DOI: 10.1016/j.heliyon.2024.e29267] [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: 12/08/2023] [Revised: 03/24/2024] [Accepted: 04/03/2024] [Indexed: 04/17/2024] Open
Abstract
Objectives Lymphovascular invasion serves as a crucial prognostic indicator in invasive breast cancer, influencing treatment decisions. We aimed to develop a machine learning model utilizing optimal volumes of interest extracted from multisequence magnetic resonance images to predict lymphovascular invasion in patients with invasive breast cancer. Materials and methods This study comprised 191 patients postoperatively diagnosed with invasive breast cancer through multi-sequence magnetic resonance imaging. Independent predictors were identified through univariate and multivariate logistic regression analyses, culminating in the construction of a clinical model. Radiomic features were extracted from multi-sequence magnetic resonance imaging images across various volume of interest scales (-2 mm, entire, +2 mm, +4 mm, and +6 mm). Subsequently, various radiomic models were developed using machine learning model algorithms, including logistic regression, support vector machine, k-nearest neighbor, gradient boosting machine, classification and regression tree, and random forest. A hybrid model was then formulated, amalgamating optimal radiomic and clinical models. Results The area under the curve of the clinical model was 0.757. Among the radiomic models, the most efficient diagnosis was achieved by the k-nearest neighbor-based radiomics-volume of interest (+2 mm), resulting in an area under the curve of 0.780. The hybrid model, integrating the k-nearest neighbor-based radiomics-volume of interest (+2 mm), and the clinical model surpassed the individual clinical and radiomics models, exhibiting a superior area under the curve of 0.864. Conclusion Utilizing a hybrid approach integrating clinical data and multi-sequence magnetic resonance imaging-derived radiomics models based on the multiscale tumor region volume of interest (+2 mm) proved effective in determining lymphovascular invasion status in patients with invasive breast cancer. This innovative methodology may offer valuable insights for treatment planning and disease management.
Collapse
Affiliation(s)
- Dengke Jiang
- Department of Radiology, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410005, China
| | - Qiuqin Qian
- Department of Radiology, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410005, China
| | - Xiuqi Yang
- Department of Radiology, Xiangtan Central Hospital, Xiangtan, Hunan, 411100, China
| | - Ying Zeng
- Department of Radiology, Xiangtan Central Hospital, Xiangtan, Hunan, 411100, China
| | - Haibo Liu
- Department of Radiology, Xiangtan Central Hospital, Xiangtan, Hunan, 411100, China
| |
Collapse
|
16
|
Toledo B, Deiana C, Scianò F, Brandi G, Marchal JA, Perán M, Giovannetti E. Treatment resistance in pancreatic and biliary tract cancer: molecular and clinical pharmacology perspectives. Expert Rev Clin Pharmacol 2024; 17:323-347. [PMID: 38413373 DOI: 10.1080/17512433.2024.2319340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/12/2024] [Indexed: 02/29/2024]
Abstract
INTRODUCTION Treatment resistance poses a significant obstacle in oncology, especially in biliary tract cancer (BTC) and pancreatic cancer (PC). Current therapeutic options include chemotherapy, targeted therapy, and immunotherapy. Resistance to these treatments may arise due to diverse molecular mechanisms, such as genetic and epigenetic modifications, altered drug metabolism and efflux, and changes in the tumor microenvironment. Identifying and overcoming these mechanisms is a major focus of research: strategies being explored include combination therapies, modulation of the tumor microenvironment, and personalized approaches. AREAS COVERED We provide a current overview and discussion of the most relevant mechanisms of resistance to chemotherapy, target therapy, and immunotherapy in both BTC and PC. Furthermore, we compare the different strategies that are being implemented to overcome these obstacles. EXPERT OPINION So far there is no unified theory on drug resistance and progress is limited. To overcome this issue, individualized patient approaches, possibly through liquid biopsies or single-cell transcriptome studies, are suggested, along with the potential use of artificial intelligence, to guide effective treatment strategies. Furthermore, we provide insights into what we consider the most promising areas of research, and we speculate on the future of managing treatment resistance to improve patient outcomes.
Collapse
Affiliation(s)
- Belén Toledo
- Department of Health Sciences, University of Jaén, Jaén, Spain
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), Amsterdam, The Netherlands
| | - Chiara Deiana
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Fabio Scianò
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), Amsterdam, The Netherlands
- Lumobiotics GmbH, Karlsruhe, Germany
| | - Giovanni Brandi
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Sanitaria ibs. GRANADA, Hospitales Universitarios de Granada-Universidad de Granada, Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, Spain
| | - Macarena Perán
- Department of Health Sciences, University of Jaén, Jaén, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, Spain
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), Amsterdam, The Netherlands
- Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, Pisa, Italy
- Cancer Pharmacology Lab, Associazione Italiana per la Ricerca sul Cancro (AIRC) Start-Up Unit, Fondazione Pisana per la Scienza, University of Pisa, Pisa, Italy
| |
Collapse
|
17
|
Wilczak M, Surman M, Przybyło M. The Role of Intracellular and Extracellular Vesicles in the Development of Therapy Resistance in Cancer. Curr Pharm Des 2024; 30:2765-2784. [PMID: 39113303 DOI: 10.2174/0113816128326325240723051625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 06/19/2024] [Indexed: 10/22/2024]
Abstract
Cancer is the second leading cause of global mortality and claims approximately 10 million lives annually. Despite advances in treatments such as surgery, chemotherapy, and immunotherapy, resistance to these methods has emerged. Multidrug resistance (MDR), where cancer cells resist diverse treatments, undermines therapy effectiveness, escalating mortality rates. MDR mechanisms include, among others, drug inactivation, reduced drug uptake, enhanced DNA repair, and activation of survival pathways such as autophagy. Moreover, MDR mechanisms can confer resistance to other therapies like radiotherapy. Understanding these mechanisms is crucial for improving treatment efficacy and identifying new therapeutic targets. Extracellular vesicles (EVs) have gathered attention for their role in cancer progression, including MDR development through protein transfer and genetic reprogramming. Autophagy, a process balancing cellular resources, also influences MDR. The intersection of EVs and autophagy further complicates the understanding of MDR. Both extracellular (exosomes, microvesicles) and intracellular (autophagic) vesicles contribute to therapy resistance by regulating the tumor microenvironment, facilitating cell communication, and modulating drug processing. While much is known about these pathways, there is still a need to explore their potential for predicting treatment responses and understanding tumor heterogeneity.
Collapse
Affiliation(s)
- Magdalena Wilczak
- Department of Glycoconjugate Biochemistry, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland
| | - Magdalena Surman
- Department of Glycoconjugate Biochemistry, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Małgorzata Przybyło
- Department of Glycoconjugate Biochemistry, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| |
Collapse
|
18
|
Jadranin M, Savić D, Lupšić E, Podolski-Renić A, Pešić M, Tešević V, Milosavljević S, Krstić G. LC-ESI QToF MS Non-Targeted Screening of Latex Extracts of Euphorbia seguieriana ssp. seguieriana Necker and Euphorbia cyparissias and Determination of Their Potential Anticancer Activity. PLANTS (BASEL, SWITZERLAND) 2023; 12:4181. [PMID: 38140508 PMCID: PMC10747863 DOI: 10.3390/plants12244181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/22/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023]
Abstract
Euphorbia seguieriana ssp. seguieriana Necker (ES) and Euphorbia cyparissias (EC) with a habitat in the Deliblato Sands were the subject of this examination. The latexes of these so far insufficiently investigated species of the Euphorbia genus are used in traditional medicine for the treatment of wounds and warts on the skin. To determine their chemical composition, non-targeted screening of the latexes' chloroform extracts was performed using liquid chromatography coupled with quadrupole time-of-flight mass spectrometry employing an electrospray ionization source (LC-ESI QTOF MS). The analysis of the obtained results showed that the latexes of ES and EC represent rich sources of diterpenes, tentatively identified as jatrophanes, ingenanes, tiglianes, myrsinanes, premyrsinanes, and others. Examination of the anticancer activity of the ES and EC latex extracts showed that both extracts significantly inhibited the growth of the non-small cell lung carcinoma NCI-H460 and glioblastoma U87 cell lines as well as of their corresponding multi-drug resistant (MDR) cell lines, NCI-H460/R and U87-TxR. The obtained results also revealed that the ES and EC extracts inhibited the function of P-glycoprotein (P-gp) in MDR cancer cells, whose overexpression is one of the main mechanisms underlying MDR.
Collapse
Affiliation(s)
- Milka Jadranin
- University of Belgrade—Institute of Chemistry, Technology and Metallurgy, Department of Chemistry, Njegoševa 12, 11000 Belgrade, Serbia;
| | - Danica Savić
- University of Belgrade—Institute of Chemistry, Technology and Metallurgy, Department of Chemistry, Njegoševa 12, 11000 Belgrade, Serbia;
| | - Ema Lupšić
- Department of Neurobiology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11108 Belgrade, Serbia; (E.L.); (A.P.-R.); (M.P.)
| | - Ana Podolski-Renić
- Department of Neurobiology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11108 Belgrade, Serbia; (E.L.); (A.P.-R.); (M.P.)
| | - Milica Pešić
- Department of Neurobiology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11108 Belgrade, Serbia; (E.L.); (A.P.-R.); (M.P.)
| | - Vele Tešević
- University of Belgrade—Faculty of Chemistry, Studentski trg 12–16, 11000 Belgrade, Serbia; (V.T.); (S.M.)
| | - Slobodan Milosavljević
- University of Belgrade—Faculty of Chemistry, Studentski trg 12–16, 11000 Belgrade, Serbia; (V.T.); (S.M.)
- Serbian Academy of Science and Arts, Kneza Mihaila 35, 11000 Belgrade, Serbia
| | - Gordana Krstić
- University of Belgrade—Faculty of Chemistry, Studentski trg 12–16, 11000 Belgrade, Serbia; (V.T.); (S.M.)
| |
Collapse
|
19
|
Milković L, Mlinarić M, Lučić I, Čipak Gašparović A. The Involvement of Peroxiporins and Antioxidant Transcription Factors in Breast Cancer Therapy Resistance. Cancers (Basel) 2023; 15:5747. [PMID: 38136293 PMCID: PMC10741870 DOI: 10.3390/cancers15245747] [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: 09/05/2023] [Revised: 11/16/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Breast cancer is still the leading cause of death in women of all ages. The reason for this is therapy resistance, which leads to the progression of the disease and the formation of metastases. Multidrug resistance (MDR) is a multifactorial process that leads to therapy failure. MDR involves multiple processes and many signaling pathways that support each other, making it difficult to overcome once established. Here, we discuss cellular-oxidative-stress-modulating factors focusing on transcription factors NRF2, FOXO family, and peroxiporins, as well as their possible contribution to MDR. This is significant because oxidative stress is a consequence of radiotherapy, chemotherapy, and immunotherapy, and the activation of detoxification pathways could modulate the cellular response to therapy and could support MDR. These proteins are not directly responsible for MDR, but they support the survival of cancer cells under stress conditions.
Collapse
Affiliation(s)
| | | | | | - Ana Čipak Gašparović
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (L.M.); (M.M.); (I.L.)
| |
Collapse
|
20
|
Srivastava D, Patra N. Enhanced Uptake of Anticancer C6-Pep Dimer in a Model Membrane Caused by Differential p Ka in Acidic Microenvironment. J Phys Chem B 2023; 127:9747-9758. [PMID: 37776281 DOI: 10.1021/acs.jpcb.3c04217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/02/2023]
Abstract
Acidic tumor microenvironment (TME) presents a challenge for the action of antitumor drugs by acting as an additional barrier for the passive crossing of the cell membrane by chemotherapic agents playing a critical role in the proliferation of tumor cells. Anticancer lipopeptide C6-Pep dimer containing the leucine zipper motif shows an increased uptake into the model tumor membrane in TME, and application of external heat might lead to the uncoiling of the zipper, which could result in cell lysis. This work investigated the cause of this increased uptake of C6-Pep dimer into the bilayer model in TME. Accurate protonation states of all the titratable residues of the C6-Pep dimer in TME were determined using constant pH molecular dynamics. In TME, except for two terminal Glu5 residues, all other Glu residues in the C6-Pep dimer were permanently protonated. The remaining Glu5 residues had differential pKa values, leading to the construction of four possible dimers with different fixed protonation states, and molecular dynamics was used to study their interaction with the anionic bilayer. Except for the dimer at a physiological pH, the other dimers were positively charged and could readily adsorb on the membrane surface. The free energy of insertion of these dimers in the bilayer was lower for single and double protonated Glu5-containing dimers than for the others. After the insertion of the lipopeptides into the membrane, thinning of the bilayer in the vicinity of dimers and an increase in area per lipid of the bilayer were observed for all systems, indicating destabilization of the bilayer due to this intercalation. This study shows that the anticancer lipopeptide C6-Pep utilizes the TME around a tumor cell for insertion into the membrane.
Collapse
Affiliation(s)
- Diship Srivastava
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (ISM) Dhanbad, Dhanbad 826004, India
| | - Niladri Patra
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (ISM) Dhanbad, Dhanbad 826004, India
| |
Collapse
|
21
|
Zhang W, Fan Y, Zhang J, Shi D, Yuan J, Ashrafizadeh M, Li W, Hu M, Abd El-Aty AM, Hacimuftuoglu A, Linnebacher M, Cheng Y, Li W, Fang S, Gong P, Zhang X. Cell membrane-camouflaged bufalin targets NOD2 and overcomes multidrug resistance in pancreatic cancer. Drug Resist Updat 2023; 71:101005. [PMID: 37647746 DOI: 10.1016/j.drup.2023.101005] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 09/01/2023]
Abstract
AIMS Multidrug resistance in pancreatic cancer poses a significant challenge in clinical treatment. Bufalin (BA), a compound found in secretions from the glands of toads, may help overcome this problem. However, severe cardiotoxicity thus far has hindered its clinical application. Hence, the present study aimed to develop a cell membrane-camouflaged and BA-loaded polylactic-co-glycolic acid nanoparticle (CBAP) and assess its potential to counter chemoresistance in pancreatic cancer. METHODS The toxicity of CBAP was evaluated by electrocardiogram, body weight, distress score, and nesting behavior of mice. In addition, the anticarcinoma activity and underlying mechanism were investigated both in vitro and in vivo. RESULTS CBAP significantly mitigated BA-mediated acute cardiotoxicity and enhanced the sensitivity of pancreatic cancer to several clinical drugs, such as gemcitabine, 5-fluorouracil, and FOLFIRINOX. Mechanistically, CBAP directly bound to nucleotide-binding and oligomerization domain containing protein 2 (NOD2) and inhibited the expression of nuclear factor kappa-light-chain-enhancer of activated B cells. This inhibits the expression of ATP-binding cassette transporters, which are responsible for chemoresistance in cancer cells. CONCLUSIONS Our findings indicate that CBAP directly inhibits NOD2. Combining CBAP with standard-of-care chemotherapeutics represents a safe and efficient strategy for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Wei Zhang
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, Guangdong 518060, China; International Association for Diagnosis and Treatment of Cancer, Shenzhen, Guangdong 518055, China
| | - Yibao Fan
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China; School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Jinze Zhang
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Dan Shi
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Jiahui Yuan
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Milad Ashrafizadeh
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Wei Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, China
| | - Man Hu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China
| | - A M Abd El-Aty
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, 12211 Giza, Egypt; Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum 25070, Turkey
| | - Ahmet Hacimuftuoglu
- Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum 25070, Turkey
| | - Michael Linnebacher
- Clinic of General Surgery, Molecular Oncology and Immunotherapy, Rostock University Medical Center, Rostock 18059, Germany
| | - Yongxian Cheng
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Weiguang Li
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, 999077, China.
| | - Shuo Fang
- Department of Oncology, The Seventh Affiliated Hospital Sun Yat-sen University, Shenzhen, Guangdong 518107, China.
| | - Peng Gong
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China; International Association for Diagnosis and Treatment of Cancer, Shenzhen, Guangdong 518055, China.
| | - Xianbin Zhang
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China; International Association for Diagnosis and Treatment of Cancer, Shenzhen, Guangdong 518055, China.
| |
Collapse
|
22
|
Bai Z, Dou J, Saleh T, Xu J, Zhu W. Editorial: Different cell death modes in cancer treatment. Front Pharmacol 2023; 14:1291585. [PMID: 37829304 PMCID: PMC10565464 DOI: 10.3389/fphar.2023.1291585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 09/18/2023] [Indexed: 10/14/2023] Open
Affiliation(s)
- Zhaoshi Bai
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jie Dou
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Tareq Saleh
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - Jingwen Xu
- Guangdong Pharmaceutical University, Guangzhou, China
| | - Wufu Zhu
- Jiangxi Science and Technology Normal University, Nanchang, China
| |
Collapse
|
23
|
Bai Z, Zhou Y, Peng Y, Ye X, Ma L. Perspectives and mechanisms for targeting mitotic catastrophe in cancer treatment. Biochim Biophys Acta Rev Cancer 2023; 1878:188965. [PMID: 37625527 DOI: 10.1016/j.bbcan.2023.188965] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/14/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023]
Abstract
Mitotic catastrophe is distinct from other cell death modes due to unique nuclear alterations characterized as multi and/or micronucleation. Mitotic catastrophe is a common and virtually unavoidable consequence during cancer therapy. However, a comprehensive understanding of mitotic catastrophe remains lacking. Herein, we summarize the anticancer drugs that induce mitotic catastrophe, including microtubule-targeting agents, spindle assembly checkpoint kinase inhibitors, DNA damage agents and DNA damage response inhibitors. Based on the relationships between mitotic catastrophe and other cell death modes, we thoroughly evaluated the roles played by mitotic catastrophe in cancer treatment as well as its advantages and disadvantages. Some strategies for overcoming its shortcomings while fully utilizing its advantages are summarized and proposed in this review. We also review how mitotic catastrophe regulates cancer immunotherapy. These summarized findings suggest that the induction of mitotic catastrophe can serve as a promising new therapeutic approach for overcoming apoptosis resistance and strengthening cancer immunotherapy.
Collapse
Affiliation(s)
- Zhaoshi Bai
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, China.
| | - Yiran Zhou
- Department of General Surgery, Rui Jin Hospital, Research Institute of Pancreatic Diseases, School of Medicine, Shanghai JiaoTong University, Shanghai 200025, China
| | - Yaling Peng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Xinyue Ye
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Lingman Ma
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China.
| |
Collapse
|
24
|
Lei Z, Tian Q, Teng Q, Wurpel JND, Zeng L, Pan Y, Chen Z. Understanding and targeting resistance mechanisms in cancer. MedComm (Beijing) 2023; 4:e265. [PMID: 37229486 PMCID: PMC10203373 DOI: 10.1002/mco2.265] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/05/2023] [Accepted: 03/23/2023] [Indexed: 05/27/2023] Open
Abstract
Resistance to cancer therapies has been a commonly observed phenomenon in clinical practice, which is one of the major causes of treatment failure and poor patient survival. The reduced responsiveness of cancer cells is a multifaceted phenomenon that can arise from genetic, epigenetic, and microenvironmental factors. Various mechanisms have been discovered and extensively studied, including drug inactivation, reduced intracellular drug accumulation by reduced uptake or increased efflux, drug target alteration, activation of compensatory pathways for cell survival, regulation of DNA repair and cell death, tumor plasticity, and the regulation from tumor microenvironments (TMEs). To overcome cancer resistance, a variety of strategies have been proposed, which are designed to enhance the effectiveness of cancer treatment or reduce drug resistance. These include identifying biomarkers that can predict drug response and resistance, identifying new targets, developing new targeted drugs, combination therapies targeting multiple signaling pathways, and modulating the TME. The present article focuses on the different mechanisms of drug resistance in cancer and the corresponding tackling approaches with recent updates. Perspectives on polytherapy targeting multiple resistance mechanisms, novel nanoparticle delivery systems, and advanced drug design tools for overcoming resistance are also reviewed.
Collapse
Affiliation(s)
- Zi‐Ning Lei
- PrecisionMedicine CenterScientific Research CenterThe Seventh Affiliated HospitalSun Yat‐Sen UniversityShenzhenP. R. China
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's UniversityQueensNew YorkUSA
| | - Qin Tian
- PrecisionMedicine CenterScientific Research CenterThe Seventh Affiliated HospitalSun Yat‐Sen UniversityShenzhenP. R. China
| | - Qiu‐Xu Teng
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's UniversityQueensNew YorkUSA
| | - John N. D. Wurpel
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's UniversityQueensNew YorkUSA
| | - Leli Zeng
- PrecisionMedicine CenterScientific Research CenterThe Seventh Affiliated HospitalSun Yat‐Sen UniversityShenzhenP. R. China
| | - Yihang Pan
- PrecisionMedicine CenterScientific Research CenterThe Seventh Affiliated HospitalSun Yat‐Sen UniversityShenzhenP. R. China
| | - Zhe‐Sheng Chen
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's UniversityQueensNew YorkUSA
| |
Collapse
|
25
|
Cai Z, Liu T, Lin Q, He J, Lei X, Luo F, Huang Y. Basis for Accurate Protein p Ka Prediction with Machine Learning. J Chem Inf Model 2023; 63:2936-2947. [PMID: 37146199 DOI: 10.1021/acs.jcim.3c00254] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
pH regulates protein structures and the associated functions in many biological processes via protonation and deprotonation of ionizable side chains where the titration equilibria are determined by pKa's. To accelerate pH-dependent molecular mechanism research in the life sciences or industrial protein and drug designs, fast and accurate pKa prediction is crucial. Here we present a theoretical pKa data set PHMD549, which was successfully applied to four distinct machine learning methods, including DeepKa, which was proposed in our previous work. To reach a valid comparison, EXP67S was selected as the test set. Encouragingly, DeepKa was improved significantly and outperforms other state-of-the-art methods, except for the constant-pH molecular dynamics, which was utilized to create PHMD549. More importantly, DeepKa reproduced experimental pKa orders of acidic dyads in five enzyme catalytic sites. Apart from structural proteins, DeepKa was found applicable to intrinsically disordered peptides. Further, in combination with solvent exposures, it is revealed that DeepKa offers the most accurate prediction under the challenging circumstance that hydrogen bonding or salt bridge interaction is partly compensated by desolvation for a buried side chain. Finally, our benchmark data qualify PHMD549 and EXP67S as the basis for future developments of protein pKa prediction tools driven by artificial intelligence. In addition, DeepKa built on PHMD549 has been proven an efficient protein pKa predictor and thus can be applied immediately to, for example, pKa database construction, protein design, drug discovery, and so on.
Collapse
Affiliation(s)
- Zhitao Cai
- College of Computer Engineering, Jimei University, Xiamen 361021, China
| | - Tengzi Liu
- College of Computer Engineering, Jimei University, Xiamen 361021, China
| | - Qiaoling Lin
- College of Computer Engineering, Jimei University, Xiamen 361021, China
| | - Jiahao He
- College of Computer Engineering, Jimei University, Xiamen 361021, China
| | - Xiaowei Lei
- College of Computer Engineering, Jimei University, Xiamen 361021, China
| | - Fangfang Luo
- College of Computer Engineering, Jimei University, Xiamen 361021, China
| | - Yandong Huang
- College of Computer Engineering, Jimei University, Xiamen 361021, China
| |
Collapse
|
26
|
Das T, Kaur H, Gour P, Prasad K, Lynn AM, Prakash A, Kumar V. Intersection of network medicine and machine learning towards investigating the key biomarkers and pathways underlying amyotrophic lateral sclerosis: a systematic review. Brief Bioinform 2022; 23:6780269. [PMID: 36411673 DOI: 10.1093/bib/bbac442] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/12/2022] [Accepted: 09/13/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Network medicine is an emerging area of research that focuses on delving into the molecular complexity of the disease, leading to the discovery of network biomarkers and therapeutic target discovery. Amyotrophic lateral sclerosis (ALS) is a complicated rare disease with unknown pathogenesis and no available treatment. In ALS, network properties appear to be potential biomarkers that can be beneficial in disease-related applications when explored independently or in tandem with machine learning (ML) techniques. OBJECTIVE This systematic literature review explores recent trends in network medicine and implementations of network-based ML algorithms in ALS. We aim to provide an overview of the identified primary studies and gather details on identifying the potential biomarkers and delineated pathways. METHODS The current study consists of searching for and investigating primary studies from PubMed and Dimensions.ai, published between 2018 and 2022 that reported network medicine perspectives and the coupling of ML techniques. Each abstract and full-text study was individually evaluated, and the relevant studies were finally included in the review for discussion once they met the inclusion and exclusion criteria. RESULTS We identified 109 eligible publications from primary studies representing this systematic review. The data coalesced into two themes: application of network science to identify disease modules and promising biomarkers in ALS, along with network-based ML approaches. Conclusion This systematic review gives an overview of the network medicine approaches and implementations of network-based ML algorithms in ALS to determine new disease genes, and identify critical pathways and therapeutic target discovery for personalized treatment.
Collapse
Affiliation(s)
- Trishala Das
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi-110067, India
| | - Harbinder Kaur
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi-110067, India
| | - Pratibha Gour
- Dept. of Plant Molecular Biology, University of Delhi, South Campus, New Delhi-110021, India
| | - Kartikay Prasad
- Amity Institute of Neuropsychology & Neurosciences (AINN), Amity University, Noida, UP-201303, India
| | - Andrew M Lynn
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi-110067, India
| | - Amresh Prakash
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Gurgaon-122413, India
| | - Vijay Kumar
- Amity Institute of Neuropsychology & Neurosciences (AINN), Amity University, Noida, UP-201303, India
| |
Collapse
|
27
|
From single-omics to interactomics: How can ligand-induced perturbations modulate single-cell phenotypes? ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 131:45-83. [PMID: 35871896 DOI: 10.1016/bs.apcsb.2022.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Cells suffer from perturbations by different stimuli, which, consequently, rise to individual alterations in their profile and function that may end up affecting the tissue as a whole. This is no different if we consider the effect of a therapeutic agent on a biological system. As cells are exposed to external ligands their profile can change at different single-omics levels. Detecting how these changes take place through different sequencing technologies is key to a better understanding of the effects of therapeutic agents. Single-cell RNA-sequencing stands out as one of the most common approaches for cell profiling and perturbation analysis. As a result, single-cell transcriptomics data can be integrated with other omics data sources, such as proteomics and epigenomics data, to clarify the perturbation effects and mechanism at the cell level. Appropriate computational tools are key to process and integrate the available information. This chapter focuses on the recent advances on ligand-induced perturbation and single-cell omics computational tools and algorithms, their current limitations, and how the deluge of data can be used to improve the current process of drug research and development.
Collapse
|