Sezen Gül, Juliette Vergnaud, François Fay, Elias Fattal
{"title":"Precision engineering of macrophage reprogramming with RNA interference-loaded lipid nanoparticles: a game-changer in cancer immunotherapy.","authors":"Sezen Gül, Juliette Vergnaud, François Fay, Elias Fattal","doi":"10.1007/s13346-025-01970-1","DOIUrl":"https://doi.org/10.1007/s13346-025-01970-1","url":null,"abstract":"<p><p>Tumor-associated macrophages (TAMs) represent solid tumors' most prevalent immune cell subset. These cells primarily adopt an immunosuppressive phenotype in the tumor microenvironment, promoting tumor initiation and progression. Their ability to shift between distinct activation states identifies TAMs as ideal targets for cancer treatment. Consequently, reprogramming TAMs from an immunosuppressive to an immunostimulatory state has emerged as a promising therapeutic approach to fight cancer. RNA interference has gained significant attention as a therapeutic modality due to its potential to selectively inhibit the expression of one or several critical proteins for the pro-tumorous activities of TAMs. However, the efficiency of RNA interference is limited by its susceptibility to nuclease degradation, rapid clearance from the body, and poor cellular uptake. These limitations necessitate the development of delivery systems to enhance their therapeutic potential. Among the nanocarriers we discuss in this review, lipid nanoparticles (LNPs) have been widely recognized as the most effective for siRNA or miRNA, providing stability, high gene silencing efficiency, and biocompatibility. The clinical application of LNPs has been further advanced by recent progress in microfluidics, enabling reproducible and scalable production of LNPs with high encapsulation efficiency. The increasing number of preclinical studies shows the growing interest in cancer immunotherapy using RNA interference-LNPs. In this review, we summarize the current knowledge on macrophage biology and its role in cancer, explore advancements in RNA interference-LNP technology, review ongoing research efforts, and discuss key translational challenges that must be addressed for the clinical success of RNA interference-LNP-based macrophage reprogramming.</p>","PeriodicalId":11357,"journal":{"name":"Drug Delivery and Translational Research","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145079948","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Gitika Kumari, Sourabh Kundu, Paras Famta, Krishna K Kalahasti, Saurabh Srivastava, Geereddy Bhanuprakash Reddy, Dadi A Srinivasarao
{"title":"Core-shell nanotherapeutics for diabetic cataract and retinopathy: current state-of-the-art and translational challenges.","authors":"Gitika Kumari, Sourabh Kundu, Paras Famta, Krishna K Kalahasti, Saurabh Srivastava, Geereddy Bhanuprakash Reddy, Dadi A Srinivasarao","doi":"10.1007/s13346-025-01971-0","DOIUrl":"https://doi.org/10.1007/s13346-025-01971-0","url":null,"abstract":"<p><p>Diabetes associated ocular complications, such as diabetic retinopathy (DR) and diabetic cataract (DC), constitute a substantial number of blind cases worldwide. Contemporary therapeutic interventions include intravitreal administration of anti-VEGF drugs for DR and surgical interventions for DC. However, these interventions suffer from drawbacks such as low patient compliance, invasiveness, and elicitation of unintended effects on sensitive ocular tissues, ultimately leading to vision loss. Therefore, there is a need for the development of advanced therapeutic interventions that are less invasive. The advent of nanotechnology has brought a paradigm shift in the treatment of ophthalmic diseases, including DR and DC. Nanoparticles, due to their high aspect ratio, offer several advantages such as quick internalization, ease of surface modification, and amenability for attachment of targeting ligands, thereby offering improved bioavailability. Moreover, the development of core-shell nanoparticles, with varying surface and bulk composition, offers additional advantages such as tissue tropism, controlled drug release, stimuli-responsiveness, and amenability for multiple drug loading, improved stability, permeability, and intra-ocular accumulation while showing minimal toxicity, ultimately offering spatio-temporally controlled drug delivery. In this review, we discussed various ocular tissue barriers while emphasizing the importance of different pristine and core-shell polymeric nanoparticles with respect to tissue tropism. Further, characterization techniques and drug release mechanisms of core-shell nanoparticles were discussed. We also presented preclinical studies that demonstrated improved therapeutic efficacy of nanoparticulate delivery systems. Finally, we presented our perspectives on challenges for scale-up and clinical translation of core-shell nanoparticles.</p>","PeriodicalId":11357,"journal":{"name":"Drug Delivery and Translational Research","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145085438","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Riyad F Alzhrani, Rawan A Fitahi, Majed A Majrashi, Yu Zhang, Mohammed Maniruzzaman
{"title":"Toward a harmonized regulatory framework for 3D-printed pharmaceutical products: the role of critical feedstock materials and process parameters.","authors":"Riyad F Alzhrani, Rawan A Fitahi, Majed A Majrashi, Yu Zhang, Mohammed Maniruzzaman","doi":"10.1007/s13346-025-01966-x","DOIUrl":"https://doi.org/10.1007/s13346-025-01966-x","url":null,"abstract":"<p><p>The commercialization of additive manufacturing (AM) in pharmaceuticals manufacturing has attracted significant attention for its potential to produce customized products. However, the process is slow and hindered by the lack of designated regulatory guidelines tailored to 3D-printed pharmaceutical products (3DPPs). The 3D-printing technology has paved the way for personalized medicine, enabled treatment of rare genetic disorders, and offered many other possibilities for patients. Despite the US Food and Drug Administration (FDA) approval of Spritam<sup>®</sup>, a clear regulatory framework for licensing 3DPPs by the FDA or EMA remains unavailable. The current practice considers all products the same, regardless of their manufacturing method and/or complexity. While this approach has been generally accepted, it frequently fails to evaluate the unique quality attributes of 3DPPs. The lack of a harmonized regulatory framework tailored to the 3DPPs presents a major barrier to the widespread adoption of AM and other innovative technologies. To bridge this gap, this review highlights the most critical parameters related to the feedstock materials and 3D-printing processes, emphasizing their impact on the quality attributes of finished 3DPPs. Numerous scenarios have been proposed to encourage regulatory authorities to establish robust regulatory guidance for the 3D-printing technology at either industrial or point-of-care (PoC) settings. Coordinated efforts between regulatory authorities, industry partners and other stakeholders are necessary to define product specifications and identify appropriate analytical techniques for evaluating finished 3DPPs. By developing a harmonized regulatory framework and establishing quality control measures, the full potential of AM can be realized. This will ultimately ensure that novel 3DPPs and personalized medicines adhere to rigorous regulatory standards of quality, safety and efficacy.</p>","PeriodicalId":11357,"journal":{"name":"Drug Delivery and Translational Research","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145069355","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Austin Stellpflug, Amit Joshi, Shue Wang, Linxia Gu, Rongxue Wu, Bo Wang
{"title":"Image-guided in vivo evaluation and comparison of bone-targeting peptides for therapeutic intervention.","authors":"Austin Stellpflug, Amit Joshi, Shue Wang, Linxia Gu, Rongxue Wu, Bo Wang","doi":"10.1007/s13346-025-01968-9","DOIUrl":"10.1007/s13346-025-01968-9","url":null,"abstract":"<p><p>Hydroxyapatite (HA)-binding peptides hold strong potential for bone-targeted therapies due to their high affinity for mineralized tissues. However, most existing studies have primarily focused on in vitro binding characterization, offering limited insight into their in vivo biodistribution and bone-binding performance. In this study, we evaluated the in vivo behavior of four HA-binding peptides, including D8, E8, YD8, and YE8, using fluorescence imaging to assess biodistribution in both healthy and pathological bone environments. In healthy animals, D8 showed the strongest bone-binding capacity, with prominent localization in the skull, femur, and tibia, while YD8 exhibited moderate binding. E8 and YE8 showed more limited localization, influenced by peptide dosage and binding kinetics. In pathological models, including tibial defects and osteogenesis imperfecta (OIM) mice, D8 and YD8 preferentially accumulated in compromised bone regions, highlighting their potential utility in targeting diseased bone microenvironments. Fluorescence imaging combined with spectral unmixing algorithms enabled effective visualization and quantification of peptide localization and distribution. These findings emphasize the value of in vivo studies for advancing the therapeutic and diagnostic applications of HA-binding peptides. The results provide a foundation for optimizing peptide design to improve specificity and efficacy in bone repair and regeneration.</p>","PeriodicalId":11357,"journal":{"name":"Drug Delivery and Translational Research","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-09-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145069376","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Application and development prospects of bio-based smart responsive hydrogels in insulin delivery.","authors":"Huijuan Su, Xiaolian Chen, Dongyi Huang, Tingting Huang, Weiju Liao, Runfang Deng, Xingzhen Huang","doi":"10.1007/s13346-025-01967-w","DOIUrl":"https://doi.org/10.1007/s13346-025-01967-w","url":null,"abstract":"<p><p>The increasing prevalence of diabetes has made insulin formulations a cornerstone in the management of the disease. However, traditional insulin delivery methods, such as subcutaneous and intravenous injections, face several challenges, including the need for frequent administration, insulin resistance, and the risk of hypoglycemia. This review examines the use of hydrogels in insulin delivery, with focus on recent advancements in the precise control of insulin release through smart response mechanisms, including pH, glucose, and temperature. The crosslinking methods and polymer compositions of hydrogels are pivotal in enhancing their biocompatibility, mechanical properties, and release regulation. Moreover, composite systems incorporating various carriers, such as nanoparticles, liposomes, and microneedles, offer significant improvements in insulin loading capacity and stable release profiles. The review also explores the potential applications of bioinspired designs and machine learning techniques in optimizing hydrogel material properties, reducing the cost of production, and enhancing the safety and efficacy of insulin delivery. Hydrogels have tremendous development space in drug delivery systems, but they face translational challenges. Through analyzing the storage, scale production and regulatory hurdles of hydrogels, this review offers some exploratory solutions. These strategies aim to promote the clinical application of insulin hydrogels and provide safer and more effective treatment options for patients with diabetes.</p>","PeriodicalId":11357,"journal":{"name":"Drug Delivery and Translational Research","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145039381","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Hanging drop culture reprograms mesenchymal stem cell transcriptome to enhance cell delivery efficiency via attenuated pulmonary entrapment.","authors":"Hsiang-Tzu Lee, Sheng-Yung Fu, Wei-Han Weng, Wei-Chen Chao, Yu-Pao Hsu, Nan-Ping Yang, Yu-Hsu Chen, Shau-Kwaun Chen, Chien-Wen Chang","doi":"10.1007/s13346-025-01927-4","DOIUrl":"https://doi.org/10.1007/s13346-025-01927-4","url":null,"abstract":"<p><p>The three-dimensional (3D) culture system has emerged as an indispensable platform for modulating stem cell function in biomedicine, drug screening, and cell therapy. Despite a few studies confirming the functionality of 3D culture, the molecular factors underlying this process remain obscure. Here, we have utilized a hanging drop method to generate 3D spheroid-derived mesenchymal stem cells (3D MSCs) and compared them to conventionally 2D-cultured MSCs. The results showed that 3D MSCs exhibited distinct phenotypic features than 2D-cultured MSCs and expressed different transcriptional responses, as found from RNA-Seq analysis. Gene ontology (GO) annotations and KEGG pathway mapping pinpointed that the 3D MSCs responded more actively to incoming signals as they upregulated receptors and cytokine production while downregulating proteolysis-, cytoskeletal-, extracellular matrix-, and adhesion-related genes. Functionally, these MSCs also displayed enhanced chemotaxis and improved pulmonary transgression post-intravenous injection. This study provides mechanistic insights on addressing a significant limitation of MSC therapy: pulmonary entrapment after systemic delivery. Moreover, upregulated pluripotency-associated genes, such as Oct4, Sox2, and Nanog, suggest that the 3D MSCs possessed enhanced stemness and regenerative capacity. The results indicate that 3D spheroid culture reshapes MSC transcriptomic and functional profiles and emerges as a promising strategy for improving their therapeutic potential in regenerative medicine.</p>","PeriodicalId":11357,"journal":{"name":"Drug Delivery and Translational Research","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-09-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145008144","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Riti Chatterjee, Vishal Sangita Babasaheb Galave, Anil B Jindal
{"title":"Current status of Liraglutide delivery systems for the management of type 2 diabetes mellitus.","authors":"Riti Chatterjee, Vishal Sangita Babasaheb Galave, Anil B Jindal","doi":"10.1007/s13346-025-01965-y","DOIUrl":"https://doi.org/10.1007/s13346-025-01965-y","url":null,"abstract":"<p><p>Diabetes is a metabolic disorder of increasing global concern. Characterized by constantly elevated levels of glucose, severe β-cell dysfunction, and insulin resistance, it is the cause of a major burden on patients if not managed with therapeutic and lifestyle changes. The human body is slowly developing tolerance to many marketed antidiabetic drugs and the quest for the discovery of newer molecules continues. Liraglutide is a prominent GLP-1 receptor agonist which is administered daily via subcutaneous injection. In addition to lowering HbA1c levels, it is also known for promoting weight loss and improving cardiovascular outcomes. A variety of novel formulation strategies have been explored to improve its bioavailability and patient compliance. To address these limitations, various advanced drug delivery systems have been investigated, including polymeric nanoparticles, lipid-based nanocarriers, biodegradable microparticles, hydrogels, and dissolvable microneedles. These systems aim to prolong drug release, enhance mucosal penetration, increase stability, and reduce dosing frequency. While many of these platforms show promise in preclinical and early clinical studies, critical translational barriers remain. These include challenges in large-scale manufacturing, ensuring formulation sterility, achieving regulatory approval, and maintaining stability during storage and distribution.This review provides a comprehensive overview of the formulation strategies developed for liraglutide delivery, critically examines their pharmacokinetic and pharmacodynamic profiles, and evaluates the current state of clinical translation. By identifying both the potential and the limitations of these delivery technologies, the article aims to inform future research directions in GLP-1-based therapeutics and improve treatment outcomes for patients with T2DM.</p>","PeriodicalId":11357,"journal":{"name":"Drug Delivery and Translational Research","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-09-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145008164","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Mahitab Elsayed, Aya Essawy, Radwa M Ismail, Yasmine Gamil, Mohamed G Hamed, Dalia Elsabaawy, Eman Abdelhakeem, Doaa Hegazy, Radwa M A Abd-Elal
{"title":"Exploring topical atorvastatin hyalurosomal gel as an adjuvant for reducing systemic corticosteroid dosage: a randomized clinical trial in severe oral lichen planus patients.","authors":"Mahitab Elsayed, Aya Essawy, Radwa M Ismail, Yasmine Gamil, Mohamed G Hamed, Dalia Elsabaawy, Eman Abdelhakeem, Doaa Hegazy, Radwa M A Abd-Elal","doi":"10.1007/s13346-025-01956-z","DOIUrl":"https://doi.org/10.1007/s13346-025-01956-z","url":null,"abstract":"<p><p>Oral lichen planus (OLP) is a chronic inflammatory disorder with limited topical treatment options and long-term corticosteroid dependency. This study investigates a novel atorvastatin-loaded hyalurosomal gel (ATV-Hyalugel) as a topical adjuvant to reduce systemic corticosteroid use in severe OLP. The objective of the study is to develop, optimize, characterize ATV-Hyalugel and evaluate its clinical efficacy in a randomized controlled clinical trial. ATV-loaded hyalurosomes (ATV-HAs) were prepared via thin-film hydration and optimized using an I-optimal mixture design (independent variables: phospholipid, Tween 80, and hyaluronic acid; responses: entrapment efficiency (EE%), particle size (PS), and zeta potential (ZP). The optimal formulation was incorporated into a chitosan gel, which was characterized for its pH, rheological behavior, and in-vitro drug release. Four weeks randomized controlled trial (n = 90) compared: group one received standard prednisolone (40 mg/day) while group two received half-dose prednisolone (20 mg/day) in combination with ATV-Hyalugel (topically, three times daily). Pain and ulcer scores were recorded weekly. Between-group comparisons were performed using the Mann-Whitney U test (non-parametric; α = 0.05), and within-group improvement from baseline to Week 4 was assessed using the Kruskal-Wallis test. Optimized ATV-HAs demonstrated high EE% (79.1 ± 0.4%), uniform PS (221.2 ± 5.1 nm), and stable ZP (-31.6 ± 0.2 mV). ATV-Hyalugel exhibited mucosa-compatible pH (6.48 ± 0.2), pseudoplastic rheology, and a sustained release profile dominated by diffusion-driven kinetics. Clinically, group two achieved therapeutic equivalence to group one by Week 2 (p > 0.05), despite receiving 50% less corticosteroid. Both groups showed significant symptom reduction from baseline to Week four (p < 0.0001, Kruskal-Wallis). No adverse events were reported with ATV-Hyalugel. ATV-Hyalugel enables a 50% corticosteroid dose reduction while maintaining clinical efficacy. Its favorable release kinetics and safety profile support its use as an innovative adjuvant therapy for severe OLP.</p>","PeriodicalId":11357,"journal":{"name":"Drug Delivery and Translational Research","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145005958","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Targeted delivery of IFN-α-anti-GPC3 fusion protein via mRNA-LNP platform elicits potent anti-tumor immunity in hepatocellular carcinoma.","authors":"Yajie Pan, Ruyue Chen, Xueyan Lv, Yuehang Wang, Hongyu Zhang","doi":"10.1007/s13346-025-01911-y","DOIUrl":"https://doi.org/10.1007/s13346-025-01911-y","url":null,"abstract":"<p><p>This study aimed to utilize the mRNA-lipid nanoparticle (mRNA-LNP) platform to achieve in situ hepatic expression of an interferon-α (IFN-α)/anti-glypican-3 (anti-GPC3) fusion protein (GPA01), enhancing IFN-α targeting and antitumor activity to provide a precision therapy strategy for GPC3-positive hepatocellular carcinoma (HCC). mRNA encoding a GPC-3/IFN-α bispecific fusion protein was designed and synthesized, encapsulated in lipid nanoparticles, and transfected into HCC cell lines (HepG2) for in vitro characterization of protein expression, binding activity, and gene induction. Orthotopic HCC models (HepG2-luc) and subcutaneous tumor model (Hepa 1-6/hGPC3-hi) were established in mice to evaluate tumor growth, survival, and immune cell infiltration following treatment with mRNA-LNP or control agents. Safety was assessed in human IFNAR transgenic mice. In vitro experiments demonstrated successful transfection and bioactive fusion protein expression by mRNA-LNP, with transfected supernatants showing specific GPC3 binding and interferon-stimulated gene (ISG) induction. In vivo studies revealed that GPC-3/IFN-α mRNA-LNP significantly inhibited tumor growth, prolonged median survival, and increased intratumoral CD8⁺ T cell and NK cell infiltration compared to controls, with favorable safety profiles. Combination therapy with PD-1 antibody (PD-1 Ab) exerted synergistic antitumor effects, primarily dependent on CD8⁺ T cell infiltration. Safety evaluations in human IFNAR transgenic mice showed good tolerability at single doses of 1-10 mpk, with transient changes in select biomarkers. Repeated dosing (6 or 10 mpk) identified a maximum tolerated dose (MTD) of 6 mpk, at least 40-fold higher than the minimal effective dose (MED, 0.15 mpk). mRNA-LNP-mediated delivery of IFN-α-anti-GPC3 fusion protein achieves targeted in situ hepatic expression, significantly enhancing antitumor activity with a broad therapeutic window. This strategy offers a novel approach for precision immunotherapy in HCC, holding substantial potential for clinical translation.</p>","PeriodicalId":11357,"journal":{"name":"Drug Delivery and Translational Research","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144999938","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}