{"title":"Targeted LNPs deliver IL-15 superagonists mRNA for precision cancer therapy.","authors":"Juntao Yu, Qian Li, Chaoting Zhang, Qiu Wang, Shenggen Luo, Xiaona Wang, Rongkuan Hu, Qiang Cheng","doi":"10.1016/j.biomaterials.2024.123047","DOIUrl":"https://doi.org/10.1016/j.biomaterials.2024.123047","url":null,"abstract":"<p><p>Interleukin-15 (IL-15) emerges as a promising immunotherapeutic candidate, but the therapeutic utility remains concern due to the unexpected systematic stress. Here, we propose that the mRNA lipid nanoparticle (mRNA-LNP) system can balance the issue through targeted delivery to increase IL-15 concentration in the tumor area and reduce leakage into the circulation. In the established Structure-driven TARgeting (STAR) platform, the LNP<sup>Local</sup> and LNP<sup>Lung</sup> can effectively and selectively deliver optimized IL-15 superagonists mRNAs to local and lungs, respectively, in relevant tumor models. As a result, such superagonists exhibited well-balanced efficacy and side-effects, demonstrating the better anti-tumor activity, less systematic exposure, and less cytokine related risks. We finally verified the selective delivery and well tolerability of LNP<sup>Lung</sup> in non-human primates (NHPs), confirming the potential for clinical application. This finding provides new potentials for cancers treatment on lung cancers or lung metastasis cancers.</p>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"317 ","pages":"123047"},"PeriodicalIF":12.8,"publicationDate":"2024-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142913458","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
BiomaterialsPub Date : 2024-12-22DOI: 10.1016/j.biomaterials.2024.123039
Lai Wei, Wandi Zhu, Chunhong Dong, Joo Kyung Kim, Yao Ma, Timothy L Denning, Sang-Moo Kang, Bao-Zhong Wang
{"title":"Lipid nanoparticles encapsulating both adjuvant and antigen mRNA improve influenza immune cross-protection in mice.","authors":"Lai Wei, Wandi Zhu, Chunhong Dong, Joo Kyung Kim, Yao Ma, Timothy L Denning, Sang-Moo Kang, Bao-Zhong Wang","doi":"10.1016/j.biomaterials.2024.123039","DOIUrl":"https://doi.org/10.1016/j.biomaterials.2024.123039","url":null,"abstract":"<p><p>The rapid approval of SARS-CoV-2 mRNA lipid nanoparticle (LNP) vaccines indicates the versatility of mRNA LNPs in an urgent vaccine need. However, the mRNA vaccines do not induce mucosal cellular responses or broad protection against recent variants. To improve cross-protection of mRNA vaccines, here we engineered a pioneered mRNA LNP encapsulating with mRNA constructs encoding cytokine adjuvant and influenza A hemagglutinin (HA) antigen for intradermal vaccination. The adjuvant mRNA encodes a novel fusion cytokine GIFT4 comprising GM-CSF and IL-4. We found that the adjuvanted mRNA LNP vaccine induced high levels of humoral antibodies and systemic T cell responses against heterologous influenza antigens and protected immunized mice against influenza A viral infections. Also, the adjuvanted mRNA LNP vaccine elicited early germinal center reactions in draining lymph nodes and promoted antibody-secreting B cell responses. In addition, we generated another adjuvant mRNA encoding CCL27, which enhanced systemic immune responses. We found the two adjuvant mRNAs both showed effective adjuvanticity in enhancing humoral and cellular responses in mice. Interestingly, intradermal immunizations of GIFT4 or CCL27 mRNA adjuvanted mRNA LNP vaccines induced significant lung tissue-resident T cells. Our findings demonstrate that the cytokine mRNA can be a promising adjuvant flexibly formulated into mRNA LNP vaccines to provoke strong immunity against viral variants.</p>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"317 ","pages":"123039"},"PeriodicalIF":12.8,"publicationDate":"2024-12-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142890827","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
BiomaterialsPub Date : 2024-12-22DOI: 10.1016/j.biomaterials.2024.123041
Hadi Sudarjat, Chaolong Qin, Diane Ingabire, Aji Alex Moothedathu Raynold, Rudra Pangeni, Adam Pearcy, Tuo Meng, Long Zhao, Michelle Arriaga, Woon N Chow, Jennifer L Puetzer, Xiuling Lu, F Gerard Moeller, Matthew S Halquist, Charles O'Keeffe, Matthew L Banks, Qingguo Xu
{"title":"Janus LAAM-loaded electrospun fibrous buccal films for treating opioid use disorder.","authors":"Hadi Sudarjat, Chaolong Qin, Diane Ingabire, Aji Alex Moothedathu Raynold, Rudra Pangeni, Adam Pearcy, Tuo Meng, Long Zhao, Michelle Arriaga, Woon N Chow, Jennifer L Puetzer, Xiuling Lu, F Gerard Moeller, Matthew S Halquist, Charles O'Keeffe, Matthew L Banks, Qingguo Xu","doi":"10.1016/j.biomaterials.2024.123041","DOIUrl":"https://doi.org/10.1016/j.biomaterials.2024.123041","url":null,"abstract":"<p><p>The opioid crisis has claimed approximately one million lives in the United States since 1999, underscoring a significant public health concern. This surge in opioid use disorder (OUD) fatalities necessitates improved therapeutic options. Current OUD therapies often require daily clinical visits, leading to poor patient compliance and high costs to the health systems. Levo-alpha-acetylmethadol (LAAM) is a long-lasting OUD drug, and the thrice-weekly oral LAAM solution can offer better patient compliance compared to the traditional daily methadone therapies. However, LAAM is FDA-approved but withdrawn from the market. As part of the NIH HEAL Initiative, we aim to reintroduce LAAM back to the market to improve OUD therapeutic options by developing a novel Janus LAAM-loaded fibrous buccal film (LFBF) formulation made of a drug-containing electrospun fibrous layer and a backing layer. The buccal administration of LFBF exhibited superior transmucosal delivery of LAAM to systemic circulation with a nearly 4-fold higher drug bioavailability than the conventional oral LAAM solution in rabbits. Furthermore, upon buccal administration in an opioid-dependent rat model, the LFBF significantly decreased fentanyl choice in the fentanyl-dependent rats, while the conventional oral LAAM solution did not at the same dose. Both the buccal film and oral solution of LAAM reduced somatic withdrawal signs in the experimental animals. These findings highlight the buccal delivery of LAAM using electrospun fibers as a promising strategy with improved drug bioavailability. Furthermore, it sheds light on future clinical applications aiming for enhanced treatment outcomes in the battle against the current opioid crisis.</p>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"317 ","pages":"123041"},"PeriodicalIF":12.8,"publicationDate":"2024-12-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142925850","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Passivated hydrogel interface: Armor against foreign body response and inflammation in small-diameter vascular grafts.","authors":"Mengxue Zhou, Zihao Wang, Mengyu Li, Qi Chen, Shengmin Zhang, Jianglin Wang","doi":"10.1016/j.biomaterials.2024.123010","DOIUrl":"https://doi.org/10.1016/j.biomaterials.2024.123010","url":null,"abstract":"<p><p>The development of small-diameter vascular grafts (SDVGs) still faces significant challenges, particularly in overcoming blockages within vessels. A key issue is the foreign-body response (FBR) triggered by the implants, which impairs the integration between grafts and native vessels. In this study, we applied an interfacial infiltration strategy to create a stable, hydrophilic, and passivated hydrogel coating on SDVGs. This coating effectively resisted FBR and improved integration between the grafts and host tissue. We also incorporated anthocyanins, an antioxidant, into the hydrogel network to mitigate oxidative stress and promote endothelialization. The hydrogel coating exhibited excellent stability, retaining its integrity during continuous flushing over 15 days. Anthocyanins were released in response to reactive oxygen species (ROS), reducing inflammation and enhancing vascularization in a mouse subcutaneous implantation model. In a rabbit carotid artery replacement model, the SDVGs exhibited rapid endothelialization, guided vascular remodeling, and inhibited calcification, showing strong potential for clinical application. This study presents a straightforward and effective approach to improve the patency rate, endothelialization, and anti-calcification properties of SDVGs by equipping them with a protective anti-FBR and anti-inflammation hydrogel layer.</p>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"317 ","pages":"123010"},"PeriodicalIF":12.8,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142890828","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Tertiary amine modification enables triterpene nanoparticles to target the mitochondria and treat glioblastoma via pyroptosis induction.","authors":"Xingchun Gao, Xiangjun Tang, Zewei Tu, Jiang Yu, Youmei Bao, Gretchen Long, Wendy C Sheu, Haoan Wu, Jia Liu, Jiangbing Zhou","doi":"10.1016/j.biomaterials.2024.123035","DOIUrl":"https://doi.org/10.1016/j.biomaterials.2024.123035","url":null,"abstract":"<p><p>Glioblastoma (GBM), the most common primary brain tumor, lacks effective treatments. Emerging evidence suggests mitochondria as a promising therapeutic target, albeit successfully targeting represents a major challenge. Recently, we discovered a group of triterpenes that can self-assemble into nanoparticles (NPs) for cancer treatment. However, unmodified triterpene NPs lack affinity for mitochondria. In this study, using oleanolic acid (OA) as an example, we demonstrated that tertiary amine modification enabled triterpene NPs to selectively target the mitochondria through interaction with translocase of outer mitochondrial membrane 70 (TOM70) leading to effective killing of GBM cells via pyroptosis. We showed that the NPs could be engineered for preferentially penetrating brain tumors through surface conjugation of iRGD, and treatment with the resulting NPs significantly prolonged the survival of tumor-bearing mice. We found that the efficacy could be further improved by encapsulating lonidamine, a mitochondrial hexokinase inhibitor. Furthermore, the observed mitochondria targeting effect through tertiary amine modification could be extended to other triterpenes, including lupeol and glycyrrhetinic acid. Collectively, this study reveals a novel strategy for targeting the mitochondria through tertiary amine modification of triterpenes, offering a promising avenue for the effective treatment of GBM.</p>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"317 ","pages":"123035"},"PeriodicalIF":12.8,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142890832","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"In situ production and precise release of bioactive GM-CSF and siRNA by engineered bacteria for macrophage reprogramming in cancer immunotherapy.","authors":"Yaxin Wang, Yali Fan, Xinyu Zhang, Jing Liu, Dawei Sun, Lianyue Li, Guijie Bai, Xinyu Liu, Jun Kang, Yingying Zhang, Hanjie Wang","doi":"10.1016/j.biomaterials.2024.123037","DOIUrl":"https://doi.org/10.1016/j.biomaterials.2024.123037","url":null,"abstract":"<p><p>In the immunosuppressive tumor microenvironment (TME), tumor-associated macrophages (TAMs) predominantly exhibit an immunosuppressive M2 phenotype, which facilitates tumor proliferation and metastasis. Although current strategies aimed at reprogramming TAMs hold promise, their sustainability and effectiveness are limited due to repeated injections. Herein, a bacterial therapy platform containing two engineered strains was developed. One strain was engineered to produce and secrete granulocyte-macrophage colony-stimulating factor (GM-CSF) to promote M2-like TAMs repolarization to M1-like TAMs, while the other strain was designed to secrete small interfering RNA (siRNA) targeting signal regulatory protein α (SIRPα). The two strains can continuously and efficiently produce bioactive therapeutic agents in situ, exerting a sustained and synergistic therapeutic effect in TAMs to inhibit tumor growth. To enhance treatment efficacy, optogenetic strategy was implemented to effectively control the production of GM-CSF, and outer membrane vesicles (OMVs) produced by engineered bacteria were utilized to protect the siRNA from degradation in the external environment. The experimental results indicated that the bacterial therapy platform could continuously produce and release bioactive GM-CSF and SIRPα siRNA, exhibiting significant therapeutic activity. In vivo experiments further demonstrated that this platform showed more sustained and stable therapeutic effects compared to conventional drug therapies. Additionally, the combination of these two engineered strains yielded the highest ratio of M1/M2 TAMs (0.80) and the lowest ratio of F4/80<sup>+</sup>SIRPα<sup>+</sup>TAMs (3.46 %) than single strain therapy. Our study expanded the potential of engineered bacteria as pharmaceutical factories for in vivo therapeutic applications.</p>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"317 ","pages":"123037"},"PeriodicalIF":12.8,"publicationDate":"2024-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142890804","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Gelatin-DOPA-knob/fibrinogen hydrogel inspired by fibrin polymerization and mussel adhesion for rapid and robust hemostatic sealing.","authors":"Zhaodi Liu, Yihang Ding, Yuan Ding, Weilin Wang, Zhengwei Mao, Lisha Yu","doi":"10.1016/j.biomaterials.2024.123038","DOIUrl":"https://doi.org/10.1016/j.biomaterials.2024.123038","url":null,"abstract":"<p><p>Tissue adhesives have attracted significant interest in the field of hemostasis. However, challenges including weak tissue adhesion, inadequate biocompatibility, and instability limit their clinical applications. Here, we have developed a gelatin-DOPA-knob/fibrinogen hydrogel inspired by the fibrin polymerization and mussel adhesion, resulting in a biocompatible bioadhesive with outstanding adhesion performance and great storage stability. This strategy involves modifying gelatin with knob peptides and catechol groups inducing crosslinking with fibrinogen to form a hydrogel via knob-hole interactions, and enhancing interfacial adhesion performance by interacting with the blood-covered tissue through catechol groups and knob peptides. This hydrogel exhibits rapid gelation, enhanced mechanical strength and adhesion properties, compared to the commonly used fibrin glue in surgery. The hydrogel significantly reduces the time required to hemostasis and the amount of blood loss in severe hemorrhage models. It ensures superior hemostatic efficacy, excellent biocompatibility, and long-term storage stability, which holds significant promise in medical settings.</p>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"317 ","pages":"123038"},"PeriodicalIF":12.8,"publicationDate":"2024-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142890802","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Breast implant silicone exposure induces immunogenic response and autoimmune markers in human periprosthetic tissue.","authors":"Isabelle Pluvy, Eve Randrianaridera, Ismail Tahmaz, Martine Melin, Florelle Gindraux, Céline Keime, Arnaud Ponche, Tatiana Petithory, Laurent Pieuchot, Karine Anselme, Isabelle Brigaud","doi":"10.1016/j.biomaterials.2024.123025","DOIUrl":"https://doi.org/10.1016/j.biomaterials.2024.123025","url":null,"abstract":"<p><p>Silicone-based breast implants are commonly used, but there are concerns about their long-term safety. While implantation results in the formation of a periprosthetic tissue that isolates the implant from the rest of the host body, silicone can leak and reach surrounding tissues. We combined histological analysis and gene expression profiling (RNA sequencing) of samples from human patients with silicone breast implants with different fillers (silicone or serum), surface topographies and/or shell rupture, and performed systematic cross-comparisons. Our study shows that exposure to silicone gel filler, even in clinically asymptomatic cases, induces an immune response. This response includes the expression of markers associated with various autoimmune diseases. This study provides the first biological evidence of an association between silicone implants and autoimmune markers, highlighting the need for further research and stricter implant safety regulations. We suggest that implant design factors, such as filler type and surface texture, may influence the inflammatory response. Re-evaluation of existing clinical trials is warranted to investigate the association between implant characteristics and potential health risks.</p>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"317 ","pages":"123025"},"PeriodicalIF":12.8,"publicationDate":"2024-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142884873","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Effects of cell shape and nucleus shape on epithelial-mesenchymal transition revealed using chimeric micropatterns.","authors":"Hongyu Wang, Ruili Liu, Yue Yu, Hongrui Xue, Runjia Shen, Yanshuang Zhang, Jiandong Ding","doi":"10.1016/j.biomaterials.2024.123013","DOIUrl":"https://doi.org/10.1016/j.biomaterials.2024.123013","url":null,"abstract":"<p><p>Epithelial-mesenchymal transition (EMT) is a key phenotypic switch in cancer metastasis, leading to fatal consequences for patients. Under geometric constraints, the morphology of cancer cells changes in both cellular and subcellular levels, whose effects on EMT are, however, not fully understood. Herein, we designed and fabricated chimeric micropatterns of polystyrene (PS) with adhesion contrast to reveal the impacts of cell shapes and nuclear shapes on EMT in a decoupled way. Cell elongation was modulated via microwell aspect ratios (ARs), and nuclear deformation was generated through a micropillar array in the microwell. Human non-small cell lung cancer cells (A549) were cultured on the quasi-three dimensional micropatterned surfaces, and transforming growth factor-β1 (TGF-β1) was added to induce EMT. We found that chimeric micropatterns upregulated EMT with an increase of cellular AR and nuclear indentation under given TGF-β1. The subsequent assessment of the contractility and oriented assembly of microfilaments elucidated the key role of mechanotransduction in cell elongation and EMT, as proved by myosin inhibition, while it was obstructed by micropillars in the chimeric micropattern. Hence, the micropillar array possessed a nonmonotonic influence, enhancing the EMT of cells with AR of 1, but hindering the EMT with an impact more significant on microwells with large ARs due to the impeded cytoskeleton assembly. This fundamental research has illustrated the complex of cellular and subcellular geometries on cell behaviors including phenotype transition in cancer metastasis.</p>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"317 ","pages":"123013"},"PeriodicalIF":12.8,"publicationDate":"2024-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142902275","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
BiomaterialsPub Date : 2024-12-15DOI: 10.1016/j.biomaterials.2024.123022
Xing Huang, Shengzhong Hou, Yinggang Li, Gang Xu, Ning Xia, Zhenyu Duan, Kui Luo, Bole Tian
{"title":"Targeting lipid metabolism via nanomedicine: A prospective strategy for cancer therapy.","authors":"Xing Huang, Shengzhong Hou, Yinggang Li, Gang Xu, Ning Xia, Zhenyu Duan, Kui Luo, Bole Tian","doi":"10.1016/j.biomaterials.2024.123022","DOIUrl":"https://doi.org/10.1016/j.biomaterials.2024.123022","url":null,"abstract":"<p><p>Lipid metabolism has been increasingly recognized to play an influencing role in tumor initiation, progression, metastasis, and therapeutic drug resistance. Targeting lipid metabolic reprogramming represents a promising therapeutic strategy. Despite their structural complexity and poor targeting efficacy, lipid-metabolizing drugs, either used alone or in combination with chemotherapeutic agents, have been employed in clinical practice. The advent of nanotechnology offers new approaches to enhancing therapeutic effects, includingthe targeted delivery and integration of lipid metabolic reprogramming with chemotherapy, photodynamic therapy (PDT), and immunotherapy. The integrated nanoformulation, nanomedicine, could significantly advance the field of lipid metabolism therapy. In this review, we will briefly introduce the concept of cancer lipid metabolism reprogramming, then elaborate the latest advances in engineered nanomedicine for targeting lipid metabolism during cancer treatment, and finally provide our insights into future perspectives of nanomedicine for interference with lipid metabolism in the tumor microenvironment.</p>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"317 ","pages":"123022"},"PeriodicalIF":12.8,"publicationDate":"2024-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142925859","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}