{"title":"Next-generation EGFR tyrosine kinase inhibitors to overcome C797S mutation in non-small cell lung cancer (2019–2024)","authors":"Debasis Das, Lingzhi Xie and Jian Hong","doi":"10.1039/D4MD00384E","DOIUrl":"10.1039/D4MD00384E","url":null,"abstract":"<p >Lung cancer is a leading cause of cancer-related deaths worldwide. Non-small cell lung cancer (NSCLC) accounts for the major portion (80–85%) of all lung cancer cases. Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) are commonly used as the targeted therapy for <em>EGFR</em>-mutated NSCLC. The FDA has approved first-, second- and third-generation EGFR-TKIs as therapeutics options. Osimertinib, the third-generation irreversible EGFR-TKI, has been approved for the treatment of NSCLC patients with the EGFR<small><sup>T790M</sup></small> mutation. However, due to the EGFR<small><sup>C797S</sup></small> mutation in the kinase domain of EGFR, resistance to osimertinib is observed and that limits the long-term effectiveness of the drug. The C797S mutation is one of the major causes of drug resistance against the third-generation EGFR TKIs. The C797S mutations including EGFR double mutations (19Del/C797S or L858R/C797S) and or EGFR triple mutations (19Del/T790M/C797S or L858R/T790M/C797S) cause major resistance to the third-generation EGFR-TKIs. Therefore, the discovery and development of fourth-generation EGFR-TKIs to target triple mutant EGFR with C797S mutation is a challenging topic in medicinal chemistry research. In this review, we discuss the discovery of novel fourth-generation EGFR TKIs, medicinal chemistry approaches and the strategies to overcome the C797S mutations. <em>In vitro</em> activities of EGFR-TKIs (2019–2024) against mutant EGFR TK, anti-proliferative activities, structural modifications, binding modes of the inhibitors and <em>in vivo</em> efficacies in animal models are discussed here.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" 10","pages":" 3371-3394"},"PeriodicalIF":4.1,"publicationDate":"2024-08-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142154864","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Abdrrahman Shemsu Surur, Chin Fung Chan, Frieda-Marie Bartz, Iris L. K. Wong, Van T. D. Nguyen, Lukas Schulig, Andreas Link, Tak Hang Chan, Larry M. C. Chow and Patrick J. Bednarski
{"title":"Fexinidazole optimization: enhancing anti-leishmanial profile, metabolic stability and hERG safety†","authors":"Abdrrahman Shemsu Surur, Chin Fung Chan, Frieda-Marie Bartz, Iris L. K. Wong, Van T. D. Nguyen, Lukas Schulig, Andreas Link, Tak Hang Chan, Larry M. C. Chow and Patrick J. Bednarski","doi":"10.1039/D4MD00426D","DOIUrl":"10.1039/D4MD00426D","url":null,"abstract":"<p >The lack of adequate anti-leishmanial therapies has led to the continued suffering of millions of people from developing nations. Moreover, optimism for a therapeutic intervention by fexinidazole was dashed due to the inability to maintain cures and control unwanted side effects. To solve these shortcomings, the structural elements of fexinidazole responsible for anti-leishmanial activity and toxicities were explored. Accordingly, a systematic analog design approach was taken for the synthesis of 24 novel analogs. We established the structural features important for activity and identified modifications that improved the hERG receptor safety and liver microsomal metabolic stability. Compared to fexinidazole, the <em>S</em>-configured imidazolooxazole analog <strong>51</strong> exhibited 25-fold greater potency against miltefosine resistant <em>L. donovani</em> amastigotes, greater metabolic stability and little hERG receptor inhibition. Replacement of the toxicophore nitro group for a cyano group resulted in a complete loss of anti-leishmanial activity. The SAR findings should be useful in the further development of this important class of anti-leishmanial agents.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" 11","pages":" 3837-3852"},"PeriodicalIF":4.1,"publicationDate":"2024-08-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142252041","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Raitis Bobrovs, Svetlana Terentjeva, Ninni Elise Olafsen, Zilvinas Dambrauskas, Antanas Gulbinas, Toivo Maimets, Indrek Teino, Aigars Jirgensons, Jason Matthews and Kristaps Jaudzems
{"title":"Discovery and optimisation of pyrazolo[1,5-a]pyrimidines as aryl hydrocarbon receptor antagonists†","authors":"Raitis Bobrovs, Svetlana Terentjeva, Ninni Elise Olafsen, Zilvinas Dambrauskas, Antanas Gulbinas, Toivo Maimets, Indrek Teino, Aigars Jirgensons, Jason Matthews and Kristaps Jaudzems","doi":"10.1039/D4MD00266K","DOIUrl":"10.1039/D4MD00266K","url":null,"abstract":"<p >The aryl hydrocarbon receptor (AHR) is a versatile ligand-dependent transcription factor involved in diverse biological processes, from metabolic adaptations to immune system regulation. Recognising its pivotal role in cancer immunology, AHR has become a promising target for cancer therapy. Here we report the discovery and structure–activity relationship studies of novel AHR antagonists. The potential AHR antagonists were identified <em>via</em> homology model-based high-throughput virtual screening and were experimentally verified in a luciferase reporter gene assay. The identified pyrazolo[1,5-<em>a</em>]pyrimidine-based AHR antagonist <strong>7</strong> (IC<small><sub>50</sub></small> = 650 nM) was systematically optimised to elucidate structure–activity relationships and reach low nanomolar AHR antagonistic potency (<strong>7a</strong>, IC<small><sub>50</sub></small> = 31 nM). Overall, the findings presented here provide new starting points for AHR antagonist development and offer insightful information on AHR antagonist structure–activity relationships.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" 10","pages":" 3477-3484"},"PeriodicalIF":4.1,"publicationDate":"2024-08-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142154862","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"A photoswitchable CENP-E inhibitor with single blue-green light to control chromosome positioning in mitotic cells†","authors":"Kazuya Matsuo, Takashi Kikukawa, Tomonori Waku, Akio Kobori and Nobuyuki Tamaoki","doi":"10.1039/D4MD00458B","DOIUrl":"10.1039/D4MD00458B","url":null,"abstract":"<p >Reversibly photoswitchable chemical tools have aided in the development of novel approaches in the biomedical field. The visible region of light should be ideal for the biological application of this approach because of its low phototoxicity and deep penetration depth compared to ultraviolet light. Herein, we report a photoswitchable centromere-associated protein E (CENP-E) inhibitor, which is controllable with low-energy blue-green light (around 500 nm) illumination. This photoswitchable tool enabled us to control CENP-E-driven chromosome movements and positioning at subcellular resolutions with low phototoxic effects. This study can contribute to the development of a unique technique for chromosome engineering.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" 11","pages":" 3795-3799"},"PeriodicalIF":4.1,"publicationDate":"2024-08-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142268777","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Sara C. Silva-Reis, Vera M. Costa, Daniela Correia da Silva, David M. Pereira, Xavier Cruz Correia, Xerardo García-Mera, José E. Rodríguez-Borges and Ivo E. Sampaio-Dias
{"title":"Discovery of hybrid Glypromate conjugates with neuroprotective activity against paraquat-induced toxicity†","authors":"Sara C. Silva-Reis, Vera M. Costa, Daniela Correia da Silva, David M. Pereira, Xavier Cruz Correia, Xerardo García-Mera, José E. Rodríguez-Borges and Ivo E. Sampaio-Dias","doi":"10.1039/D4MD00584H","DOIUrl":"10.1039/D4MD00584H","url":null,"abstract":"<p >Neurodegenerative disorders comprise a series of heterogeneous conditions that affect millions of people worldwide, representing a significant health burden in both developed and developing countries. Without disease-modifying treatments currently available, the development of effective neurotherapeutics is a health priority. In this work, a new series of peptide-conjugates of the Glypromate neuropeptide is reported to determine the interplay of annular constriction and neuroprotective activity. To this end, (1<em>R</em>,3<em>S</em>,4<em>S</em>)-2-azanorbornane-3-carboxylic acid was used as an <small>L</small>-proline and <small>L</small>-pipecolic acid surrogate in addition to functionalization of the glutamate residue with relevant active pharmaceutical ingredients (APIs), namely amantadine, memantine, and (<em>R</em>)-1-aminoindane. Using non-differentiated SH-SY5Y cells, conjugates <strong>14a</strong> and <strong>15a</strong>, functionalized with amantadine, significantly reduced protein aggregation, with <strong>15a</strong> outperforming both Glypromate (2-fold enhancement, <em>p</em> < 0.05) and an equimolar mixture of Glypromate and amantadine (<em>p</em> < 0.0001). On the other hand, in SH-SY5Y differentiated cells, conjugate <strong>18c</strong> functionalized with (<em>R</em>)-1-aminoindane counteracted the toxicity elicited by paraquat (<em>p</em> < 0.0001), while Glypromate was found to exacerbate the neurotoxicity. Altogether, this work adds new insights into Glypromate research by demonstrating that chemical conjugation and annular constriction are effective strategies to tune neuroprotective responses against different neurotoxic stimuli, paving the way for the development of new neurotherapeutics.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" 11","pages":" 3711-3727"},"PeriodicalIF":4.1,"publicationDate":"2024-08-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142252044","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ahmed M. Badawy, Enas E. Eltamany, Rodina M. Hussien, Osama G. Mohamed, Mayada M. El-Ayouty, Mohamed S. Nafie, Ashootosh Tripathi and Safwat A. Ahmed
{"title":"Cornulacin: a new isoflavone from Cornulaca monacantha and its isolation, structure elucidation and cytotoxicity through EGFR-mediated apoptosis†","authors":"Ahmed M. Badawy, Enas E. Eltamany, Rodina M. Hussien, Osama G. Mohamed, Mayada M. El-Ayouty, Mohamed S. Nafie, Ashootosh Tripathi and Safwat A. Ahmed","doi":"10.1039/D4MD00524D","DOIUrl":"10.1039/D4MD00524D","url":null,"abstract":"<p >Chemical investigation of the methanolic extract of <em>Cornulaca monacantha</em> (Amaranthaceae), an annual wild herb collected from North Sinai, Egypt, yielded a new isoflavone cornulacin <strong>1</strong> and five known compounds: <em>N-trans</em>-feruloyltyramine <strong>2</strong>, <em>N-trans</em>-feruloyl-3′-methoxytyramine <strong>3</strong>, <em>N-trans</em>-caffeoyl tyramine <strong>4</strong>, Cannabisin F <strong>5</strong> and (2a<em>S</em>, 3a<em>S</em>) lyciumamide D <strong>6</strong>. Using MTT assay, the isolated compounds were evaluated for their <em>in vitro</em> cytotoxicity against pancreatic (Panc1) and ovarian (A2780) cancer cell lines. Compounds <strong>1</strong>, <strong>2</strong>, <strong>3</strong>, and <strong>4</strong> exhibited promising cytotoxic activity against the tested cells, among which compound <strong>1</strong> (IC<small><sub>50</sub></small> of 2.1 ± 0.21 μM) was the most active one against A2780 cells, whereas compound <strong>2</strong> (IC<small><sub>50</sub></small> of 3.4 ± 0.11 μM) was the most effective compound against Panc1 cells. Accordingly, compound <strong>1</strong> was further investigated for its apoptotic induction in A2780 cancer cells using Annexin V/PI staining. Compound <strong>1</strong> significantly stimulated apoptotic ovarian A2780 cancer cells by 45.9-fold and arrested cell proliferation in the S-phase. Such activity was mediated through the upregulation of proapoptotic genes Bax; P53; and caspase 3, 8, and 9 besides the downregulation of the Bcl-2 gene, the anti-apoptotic one. Furthermore, molecular docking investigation demonstrated the strong binding affinity of compound <strong>1</strong> with EGFR active sites, which validated its experimental EGFR enzyme inhibition activity.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" 9","pages":" 3228-3238"},"PeriodicalIF":4.1,"publicationDate":"2024-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142056375","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Xiaoqin Luo, Jin Wang, Ruichang Wang, Jiabing Lian, Mengnan Guo, Hongrui Zhou, Mengxue Zhang, Zhe Yang, Xiaolong Li, Xianran He and Xiuli Bi
{"title":"SLL-1A-16 suppresses proliferation and induces autophagy in non-small-cell lung cancer cells via the AKT/mTOR signaling pathway†","authors":"Xiaoqin Luo, Jin Wang, Ruichang Wang, Jiabing Lian, Mengnan Guo, Hongrui Zhou, Mengxue Zhang, Zhe Yang, Xiaolong Li, Xianran He and Xiuli Bi","doi":"10.1039/D4MD00405A","DOIUrl":"10.1039/D4MD00405A","url":null,"abstract":"<p >Non-small-cell lung cancer (NSCLC), which accounts for approximately eighty-five percent of lung cancer diagnoses worldwide, is a malignancy with high incidence and mortality rates. Among the various antitumor compounds, organic selenium-containing compounds have emerged as a promising class of therapeutic agents for cancer treatment. In the present study, SLL-1A-16, a new organoselenium small molecule, was discovered to exhibit antiproliferative activity against NSCLC both <em>in vitro</em> and <em>in vivo</em>. Treatment with SLL-1A-16 significantly inhibited NSCLC cell proliferation and induced apoptosis and autophagy. Mechanistically, SLL-1A-16 inhibited cell proliferation through G1-S phase arrest by reducing cyclin D1 and CDK4 expression. Additionally, SLL-1A-16 significantly induced apoptosis by upregulating cleaved caspase 3 and Bax expression, while downregulating Bcl-2 levels. Our study also demonstrated that SLL-1A-16 induced autophagy in NSCLC cells by inhibiting the Akt/mTOR pathway. Overall, our findings suggest that SLL-1A-16 could induce cell cycle arrest, apoptosis and autophagy in NSCLC cells by inhibiting the Akt/mTOR signaling pathways, providing a theoretical basis for the potential clinical application of SLL-1A-16 as a chemotherapeutic agent in NSCLC treatment.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" 10","pages":" 3460-3468"},"PeriodicalIF":4.1,"publicationDate":"2024-08-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142154877","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Inhibiting SARS-CoV-2 viral entry by targeting spike:ACE2 interaction with O-modified quercetin derivatives†","authors":"Reuben James Z. Rosal and Monissa C. Paderes","doi":"10.1039/D4MD00286E","DOIUrl":"10.1039/D4MD00286E","url":null,"abstract":"<p >The cell entry of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is mediated by the interaction between the receptor-binding domain of its spike (S) protein and human angiotensin-converting enzyme 2 (ACE2). Quercetin, a flavonoid found abundantly in plants, shows potential as a SARS-CoV-2 S:ACE2 inhibitor but is known to have low bioavailability. Modification of quercetin by capping its hydroxyl moieties could enhance the metabolic stability, solubility, and bioavailability, and reduce toxicity. In this study, sixteen (16) <em>O</em>-modified quercetin derivatives were synthesized by incorporating alkyl and acyl moieties of varying lengths, sizes, and polarities to the hydroxyl groups. The SARS-CoV-2 S:ACE2 inhibitory activity and toxicity of the synthesized derivatives were assessed <em>in vitro</em>, and their physicochemical properties, pharmacokinetics, and drug-likeness were predicted and evaluated using the SwissADME web tool. Results showed that functionalization of the hydroxyl moieties of quercetin generally resulted in more potent inhibitors (>50% inhibition). Five (5) derivatives displayed a dose-dependent inhibition against the SARS-CoV-2 S:ACE2 interaction with promising IC<small><sub>50</sub></small> values (<em>i.e.</em>, <strong>2e</strong> (IC<small><sub>50</sub></small> = 7.52 μM), <strong>3a</strong> (IC<small><sub>50</sub></small> = 5.00 μM), <strong>3b</strong> (IC<small><sub>50</sub></small> = 25.70 μM), <strong>3c</strong> (IC<small><sub>50</sub></small> = 2.22 μM), and <strong>4b</strong> (IC<small><sub>50</sub></small> = 3.28 μM)). Moreover, these compounds exhibited low hepato-, nephro-, and cardiotoxicity, and their SwissADME profiles indicated favorable physicochemical, pharmacokinetic, and drug-like properties, suggesting their potential as promising lead SARS-CoV-2 S:ACE2 inhibitors.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" 9","pages":" 3212-3222"},"PeriodicalIF":4.1,"publicationDate":"2024-08-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142009366","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jinhua Ning, Nini Zhan, Zhanpan Wu, Yuzhe Li, Die Zhang, Yadian Shi, Yingxun Zhou, Chuan-Huizi Chen and Wenbin Jin
{"title":"In vitro identification of oridonin hybrids as potential anti-TNBC agents inducing cell cycle arrest and apoptosis by regulation of p21, γH2AX and cleaved PARP†","authors":"Jinhua Ning, Nini Zhan, Zhanpan Wu, Yuzhe Li, Die Zhang, Yadian Shi, Yingxun Zhou, Chuan-Huizi Chen and Wenbin Jin","doi":"10.1039/D4MD00580E","DOIUrl":"10.1039/D4MD00580E","url":null,"abstract":"<p >TNBC has been recognized as the most highly aggressive breast cancer without chemotherapeutic drugs. A collection of oridonin hybrids consisting of conventional antitumor pharmacophores including nitrogen mustards and adamantane-1-carboxylic acid were synthesized by deletion or blockade of multiple hydroxyl groups and structural rearrangement. Compound <strong>11a</strong> showed the most promising anti-TNBC activity with nearly 15-fold more potent antiproliferative effects than oridonin against MDA-MB-231 and HCC1806. Moreover, <strong>11a</strong> significantly inhibited HCC1806, MDA-MB-231 and MDA-MB-468 cell proliferation by arresting cells at the G2/M phase in a dose-dependent manner. Furthermore, <strong>11a</strong> could trigger dose-dependently early and late apoptosis in those indicated cell lines. More importantly, <strong>11a</strong> could significantly increase p21, γH2AX and cleaved PARP accumulation in a dose-dependent manner. Furthermore, compound <strong>11a</strong> exhibited better stability than oridonin in a plasma assay. Taken together, all results demonstrated that <strong>11a</strong> may warrant further investigation as a promising anticancer drug candidate for the treatment of TNBC.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" 11","pages":" 3674-3694"},"PeriodicalIF":4.1,"publicationDate":"2024-08-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142154858","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Synthetic modification of protein surfaces to mediate induced-proximity pharmacology","authors":"Lyn H. Jones","doi":"10.1039/D4MD00388H","DOIUrl":"10.1039/D4MD00388H","url":null,"abstract":"<p >Molecular glues and bifunctional small molecules, such as targeted protein degraders, induce protein proximity to mediate gain-of-function pharmacology. Emerging technologies that synthetically manipulate protein surfaces to create neoproteins, and the development of covalent chemical probes for intra- and inter-protein surface labeling are described. Ligand-directed protein surface modification strategies have the potential to enhance the induced-proximity pharmacology toolkit and expand the druggable proteome, and this Opinion considers the opportunities and challenges that lie ahead.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" 9","pages":" 2974-2979"},"PeriodicalIF":4.1,"publicationDate":"2024-08-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142056381","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}