MedCommPub Date : 2025-10-05eCollection Date: 2025-10-01DOI: 10.1002/mco2.70412
Hengyi Chen, Hongling Yang, Lu Guo, Qingxiang Sun
{"title":"The Role of Immune Checkpoint Inhibitors in Cancer Therapy: Mechanism and Therapeutic Advances.","authors":"Hengyi Chen, Hongling Yang, Lu Guo, Qingxiang Sun","doi":"10.1002/mco2.70412","DOIUrl":"https://doi.org/10.1002/mco2.70412","url":null,"abstract":"<p><p>The rapid development of immune checkpoint inhibitors has fundamentally changed the landscape of cancer treatment. These agents restore T cell-mediated antitumor immune responses by targeting key immune checkpoint molecules, thereby suppressing or eliminating tumors. However, their clinical application still faces multiple challenges, mainly including efficacy heterogeneity, drug resistance, immune-related adverse events. Furthermore, there is still a lack of reliable biomarkers for predicting efficacy and toxicity. More critically, there is absence of precise predictive models that can systematically integrate multiomics features, dynamic tumor microenvironment evolution, and patient individual differences to comprehensively address the above issues. This review systematically summarizes the latest advancements in this field. The main contents include emerging targets like lymphocyte activation gene 3, T cell immunoreceptor with immunoglobulin and tyrosine-based inhibitory motif domain, and mucin-domain-containing-3, combination strategies, and the current research status and limitations of various predictive biomarkers. Moreover, it focuses on the potential of microbiome regulation, metabolic reprogramming, and artificial intelligence-driven multiomics analysis technologies in achieving dynamic patient stratification and personalized treatment. By integrating the frontier research results and clinical insights, the review aims to provide a systematical theory framework and future directions for advancing precision immunotherapy.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":"6 10","pages":"e70412"},"PeriodicalIF":10.7,"publicationDate":"2025-10-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12497686/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145246089","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
MedCommPub Date : 2025-10-05eCollection Date: 2025-10-01DOI: 10.1002/mco2.70413
Long Zhao, Haoyue Cui, Yutong Li, Yingjiang Ye, Zhanlong Shen
{"title":"Metabolite to Modifier: Lactate and Lactylation in the Evolution of Tumors.","authors":"Long Zhao, Haoyue Cui, Yutong Li, Yingjiang Ye, Zhanlong Shen","doi":"10.1002/mco2.70413","DOIUrl":"https://doi.org/10.1002/mco2.70413","url":null,"abstract":"<p><p>Lactate, once dismissed as a mere by-product of cancer metabolism, has emerged as a pivotal factor in tumor progression, exerting diverse effects on metabolic reprogramming and immune modulation. Lactate enhances tumor cell adaptability through sustained glycolysis and concurrently shapes the tumor microenvironment by modulating immune, stromal, and endothelial cell function. This review highlights the evolving understanding of lactate's role, extending beyond the Warburg effect to its regulatory capacity via lactylation, a recently identified post-translational modification. The complex interaction between lactate and tumor biology is examined, emphasizing its influence on the tumor microenvironment and immune dynamics. Additionally, potential therapeutic strategies targeting lactate metabolism and transport are explored, along with lactylation regulation by histone-modifying enzymes. Inhibitors targeting lactate production and transport, especially those against lactate dehydrogenase (LDH) and monocarboxylate transporters (MCTs), have shown considerable potential in preclinical and early clinical studies. Recent advancements are discussed, underscoring the potential of integrating metabolic regulation with immunotherapies, thereby offering a dual pathway in cancer treatment. These insights establish lactate and lactylation as pivotal modulators of tumor biology and highlight their potential as targets in precision oncology.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":"6 10","pages":"e70413"},"PeriodicalIF":10.7,"publicationDate":"2025-10-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12497687/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145246006","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
MedCommPub Date : 2025-10-05eCollection Date: 2025-10-01DOI: 10.1002/mco2.70414
Xiaochi Li, Junli Li, Jiazheng Wei, Weixin Du, Cheng Su, Xiaobin Shen, Aihua Zhao, Miao Xu
{"title":"Design Strategies for Novel Lipid Nanoparticle for mRNA Vaccine and Therapeutics: Current Understandings and Future Perspectives.","authors":"Xiaochi Li, Junli Li, Jiazheng Wei, Weixin Du, Cheng Su, Xiaobin Shen, Aihua Zhao, Miao Xu","doi":"10.1002/mco2.70414","DOIUrl":"https://doi.org/10.1002/mco2.70414","url":null,"abstract":"<p><p>Messenger RNA (mRNA) vaccines have revolutionized infectious disease prevention and cancer immunotherapy due to their rapid development, potent immunogenicity, and flexible design. Central to the clinical success of mRNA vaccines, lipid nanoparticles (LNPs) function as efficient, nonviral delivery systems capable of protecting mRNA and facilitating its uptake by target cells. Recent advances have demonstrated that LNP-formulated mRNA vaccines and therapeutics elicit robust immune responses and confer effective protection against a broad spectrum of pathogens, including viruses and bacteria. Moreover, LNP-based therapies have shown promising therapeutic efficacy in various cancers and rare diseases, as evidenced by both preclinical models and clinical trials. This review provides a comprehensive overview of the key components, structural features, and preparation technologies of LNPs. It further discusses ongoing challenges in LNP design, such as delivery efficiency, tissue targeting, and safety, and proposes rational strategies to address these limitations. Additionally, recent progress in the analytical methods used to characterize the critical quality attributes of LNPs is highlighted. This review aims to guide the rational design of next-generation LNPs and to support the broader application of mRNA-based vaccines and therapeutics.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":"6 10","pages":"e70414"},"PeriodicalIF":10.7,"publicationDate":"2025-10-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12497691/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145246011","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
MedCommPub Date : 2025-10-04DOI: 10.1002/mco2.70407
Mingchuan Liu, Tingwei Peng, Rui Yu, Kexin Wang, Di Wang, Xiaojie Jia, Yan Zhang, Jianqiang Hu, Bingchao Qi, Yan Li
{"title":"Heart Regeneration and Repair: Molecular Mechanism and Therapeutic Targets","authors":"Mingchuan Liu, Tingwei Peng, Rui Yu, Kexin Wang, Di Wang, Xiaojie Jia, Yan Zhang, Jianqiang Hu, Bingchao Qi, Yan Li","doi":"10.1002/mco2.70407","DOIUrl":"https://doi.org/10.1002/mco2.70407","url":null,"abstract":"<p>The substantial loss of cardiomyocytes resulting from myocardial infarction leads to pathological remodeling of the heart and the onset of heart failure. Promoting heart regeneration is therefore a critical therapeutic goal for repairing damaged cardiac tissue. Over the past two decades, the utilization of cardiac stem cells for heart regeneration has emerged as a focal point of research. However, the related mechanisms and efficacy remain constrained by poor integration and survival. Concurrently, genetic lineage tracing has definitively shown that the adult mammalian heart lacks significant endogenous stem cells. It is now widely accepted that heart regeneration primarily arises from the proliferation of pre-existing adult cardiomyocytes. This review systematically summarizes the physiological and microenvironmental changes during the developmental process of cardiomyocytes, elucidates the intrinsic and extrinsic molecular biological mechanisms that regulate cardiomyocyte proliferation, and discusses exogenous cell transplantation therapy, potentially endogenous pharmacological and genetic approaches, as well as promising bioengineering and cross-disciplinary methods. By synthesizing these multifaceted advances, this review aims to clarify important issues that require further elucidation in this field, thereby advancing the depth of research on heart regeneration and its clinical translational applications.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":"6 10","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mco2.70407","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145224363","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
MedCommPub Date : 2025-10-04DOI: 10.1002/mco2.70404
Md Abdus Samad, Iftikhar Ahmad, Mohammad Rashid Khan, Mohd Suhail, Torki A. Zughaibi, Fahad A. Al-Abbasi, Khaled A. Alhosaini, Mohd Shahnawaz Khan, Ajoy Kumer, Shams Tabrez
{"title":"Breast Cancer: Molecular Pathogenesis and Targeted Therapy","authors":"Md Abdus Samad, Iftikhar Ahmad, Mohammad Rashid Khan, Mohd Suhail, Torki A. Zughaibi, Fahad A. Al-Abbasi, Khaled A. Alhosaini, Mohd Shahnawaz Khan, Ajoy Kumer, Shams Tabrez","doi":"10.1002/mco2.70404","DOIUrl":"https://doi.org/10.1002/mco2.70404","url":null,"abstract":"<p>Breast cancer (BC) is the most prevalent cancer in women and remains the leading cause of cancer-related mortality globally. Its development is influenced by multiple factors, including genetics, environmental, aging, and modulation of various signaling pathways. The heterogeneity of BC together with the emergence of treatment resistance and recurrence have prompted researchers to explore and develop new therapeutic approaches. Recently, oncology research has primarily focused on the development of targeted therapies against molecular abnormalities in BC. These therapies include monoclonal antibodies, tyrosine kinase inhibitors, antibody–drug conjugates, PI3K/Akt/mTOR pathway inhibitors, CDK 4/6 inhibitors, PARP inhibitors, antiangiogenic agents, and various other targeted drugs. Immunomodulatory strategies, including immune checkpoint inhibitors (anti-PD-1/PD-L1), CTLA-4 blockers, adoptive T-cell therapy, and cancer vaccines, stimulate immune response against cancer cells. Epigenetic therapies like DNMT and HDAC inhibitors have also shown promise in BC treatment. This review highlights how innovative approaches like targeting intratumoral heterogeneity, liquid biopsy for resistance mutation detection, bypass mechanisms (<i>FGFR1</i> activation following CDK4/6 inhibition), artificial intelligence-based drug discovery, patient-derived organoids, and adaptive trial designs are shaping BC treatment. By combining molecular insights with precision therapeutics, these advancements offer significant potential to address resistance, improve efficacy, and enhance patient outcomes.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":"6 10","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mco2.70404","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145224359","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
MedCommPub Date : 2025-10-04DOI: 10.1002/mco2.70401
Gang Fan, Shilin Chen, Qingping Zhang, Na Yu, Ziyang Shen, Zhaoji Liu, Weiming Guo, Zhihan Tang, Jing Yang, Miao Liu
{"title":"Proteolysis-Targeting Chimera (PROTAC): Current Applications and Future Directions","authors":"Gang Fan, Shilin Chen, Qingping Zhang, Na Yu, Ziyang Shen, Zhaoji Liu, Weiming Guo, Zhihan Tang, Jing Yang, Miao Liu","doi":"10.1002/mco2.70401","DOIUrl":"https://doi.org/10.1002/mco2.70401","url":null,"abstract":"<p>Targeted protein degradation (TPD) represents a paradigm shift in drug discovery, moving beyond traditional binding-based inhibition toward active removal of disease-driving proteins. This approach has unlocked therapeutic possibilities for previously “undruggable” targets, including transcription factors like MYC and STAT3, mutant oncoproteins such as KRAS G12C, and scaffolding molecules lacking conventional binding pockets. Among TPD strategies, proteolysis-targeting chimeras (PROTACs) have emerged as the leading clinical platform, with the first molecule entering trials in 2019 and progression to Phase III completion by 2024. This comprehensive review examines PROTAC development across diverse therapeutic areas, analyzing key targets including kinases, hormone receptors, antiapoptotic proteins, and epigenetic modulators. We evaluate clinical progression of breakthrough candidates such as ARV-110 for prostate cancer, ARV-471 for breast cancer, and BTK degraders, while discussing critical challenges including the “hook effect” and oral bioavailability limitations. The review explores future directions encompassing innovative delivery strategies, tissue-specific degrader design, and approaches for expanding E3 ligase repertoires and overcoming resistance. This review provides essential foundations for rational target selection, molecular optimization, and clinical translation strategies. By integrating mechanistic insights with clinical realities, this analysis offers perspectives on PROTAC technology advancement and identifies opportunities for transforming treatment of complex diseases resistant to conventional therapies.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":"6 10","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mco2.70401","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145224329","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Hypertrophic Scarring and Keloids: Epidemiology, Molecular Pathogenesis, and Therapeutic Interventions","authors":"Xiaowan Fang, Yuxiang Wang, Hao Chen, Zhenzhen Yan, Shunxin Jin, Yixin Wu, Futing Shu, Shichu Xiao","doi":"10.1002/mco2.70381","DOIUrl":"https://doi.org/10.1002/mco2.70381","url":null,"abstract":"<p>Wound healing is a complex, multicellular process that is essential for restoring tissue integrity after injury. In a subset of individuals, however, this process becomes dysregulated, culminating in hypertrophic scars or keloids—fibroproliferative disorders marked by excessive extracellular matrix deposition and prolonged inflammation. Although these lesions differ clinically, both share overlapping molecular mechanisms involving aberrant activation of the TGF-β, Intergrin-FAK, and Wnt/β-catenin pathways. Recent insights from single-cell and multiomics technologies have revealed profound heterogeneity within scar-forming fibroblast populations and highlighted the modulatory roles of immune cells, genetic predispositions, and anatomical tension. However, despite increasing mechanistic understanding, current interventions—including surgery, corticosteroids, and laser therapy—are limited by high recurrence rates and variable efficacy. Emerging strategies now target fibroblast plasticity, inflammatory circuits, and biomechanical feedback via tools such as gene editing, immune modulation, and smart biomaterials. This review integrates advances across epidemiology, molecular signaling, and therapeutic innovation, underscoring the need for personalized, multitargeted approaches. Ultimately, transforming pathological scarring from a persistent clinical burden into a regenerative opportunity will depend on interdisciplinary collaboration and the continued translation of benchside discovery into bedside care.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":"6 10","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mco2.70381","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145224364","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
MedCommPub Date : 2025-10-03DOI: 10.1002/mco2.70391
Zhaoyun Liu, Jiao Lai, Ziyu Ma, Jianhua Pan, Chun Yang, Rong Fu
{"title":"Targeting the Ubiquitin–Proteasome System for Cancer","authors":"Zhaoyun Liu, Jiao Lai, Ziyu Ma, Jianhua Pan, Chun Yang, Rong Fu","doi":"10.1002/mco2.70391","DOIUrl":"https://doi.org/10.1002/mco2.70391","url":null,"abstract":"<p>Ubiquitin is a highly conserved small molecule that exists in large quantities in eukaryotic cells and plays a crucial role in protein quality control by phagocytosis and degradation of ubiquitin-modified proteins. The abnormal expression of the ubiquitin–proteasome system (UPS) in cancer leads to the abnormal expression of ubiquitin ligases and ubiquitin-binding enzymes, resulting in the abnormal accumulation of ubiquitinated proteins. Consequently, UPS dysregulation can contribute to tumor initiation, progression, and resistance to therapy. While proteasome inhibitors have shown clinical success, comprehensive reviews integrating upstream UPS components and their therapeutic potential are lacking. This paper reviews the composition of the UPS, its tumor-promoting mechanisms, as well as the small molecule inhibitors and proteasome inhibitors based on this system, including their mechanisms of action and adverse effects, and explores their clinical advances in the treatment of cancer. This review provides a valuable framework for developing next-generation anti-cancer therapies and establishes the UPS as a critical therapeutic target for precision oncology.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":"6 10","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mco2.70391","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145224073","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
MedCommPub Date : 2025-09-30DOI: 10.1002/mco2.70397
{"title":"Correction to “RNA Modifications in Cancer”","authors":"","doi":"10.1002/mco2.70397","DOIUrl":"10.1002/mco2.70397","url":null,"abstract":"<p>Han Wu, Shi Chen, Xiang Li, Yuyang Li, Yiwen Qin, Bohe Shi, Yifei Tang, Zhuoyi Yan, Yang Hao, Dongxu Wang, Weiwei Liu.</p><p><i>RNA modifications in cancer</i>. MedComm. 2024; [doi: http://doi.org/10.1002/mco2.70042].</p><p>In the published version of this article, the name of the fifth author was incorrectly listed as <b>“Yiwen Qing”</b>. The correct spelling of the author's name is <b>“Yiwen Qin”</b>.</p><p>The author order and affiliations remain unchanged. All authors have approved this correction.</p><p>We apologize for this error.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":"6 10","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12483941/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145214923","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
MedCommPub Date : 2025-09-29DOI: 10.1002/mco2.70403
Xiaowen Liang, Yue Yin, Yuanlong Lin, Shiman Chen, Qi Qian, Jing Yuan, Liuqing Yang, Yang Yang
{"title":"Molecular and Cellular Mechanisms of Respiratory Syncytial Viral Infection: Its Implications for Prophylactic and Therapeutic Pharmaceuticals","authors":"Xiaowen Liang, Yue Yin, Yuanlong Lin, Shiman Chen, Qi Qian, Jing Yuan, Liuqing Yang, Yang Yang","doi":"10.1002/mco2.70403","DOIUrl":"10.1002/mco2.70403","url":null,"abstract":"<p>Respiratory syncytial virus (RSV) is a notorious pathogen that serves as the leading cause of lower respiratory tract infections (LRTI) among infants, the elderly, and immunocompromised individuals. Its widespread prevalence exerts a considerable burden on global healthcare systems and economies, owing to the high rates of hospitalization and the potential for long-term health complications. Significant progress has been achieved in RSV prevention strategies during recent years, with three vaccines currently approved worldwide for active immunization in adults aged 60 years and older, as well as pregnant women. Furthermore, the monoclonal antibody nirsevimab has been approved for the prevention of RSV infections in infants. However, effective antiviral treatments for postinfection cases remain an unmet clinical need. This review comprehensively elaborates the molecular and cellular mechanisms of RSV infection, including viral structure, replication cycle, and pathogenic mechanisms. Meanwhile, we systematically summarize the latest advances in preventive and therapeutic agents and analyze the practical applications and existing limitations of current immunization strategies. Furthermore, we discuss and propose the challenges and future directions in drug development. The review provide insights for developing novel and effective prevention and treatment strategies against RSV infection.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":"6 10","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-09-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12477444/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145202567","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}