A novel strategy for treating oncogene-mutated tumors by targeting tumor microenvironment and synergistically enhancing anti-PD-1 immunotherapy

IF 20.1 1区 医学 Q1 ONCOLOGY Cancer Communications Pub Date : 2024-02-09 DOI:10.1002/cac2.12521
Yingqiang Liu, Linjiang Tong, Mengge Zhang, Qi Zhang, Qiupei Liu, Fang Feng, Yan Li, Mengzhen Lai, Haotian Tang, Yi Chen, Meiyu Geng, Wenhu Duan, Jian Ding, Hua Xie
{"title":"A novel strategy for treating oncogene-mutated tumors by targeting tumor microenvironment and synergistically enhancing anti-PD-1 immunotherapy","authors":"Yingqiang Liu,&nbsp;Linjiang Tong,&nbsp;Mengge Zhang,&nbsp;Qi Zhang,&nbsp;Qiupei Liu,&nbsp;Fang Feng,&nbsp;Yan Li,&nbsp;Mengzhen Lai,&nbsp;Haotian Tang,&nbsp;Yi Chen,&nbsp;Meiyu Geng,&nbsp;Wenhu Duan,&nbsp;Jian Ding,&nbsp;Hua Xie","doi":"10.1002/cac2.12521","DOIUrl":null,"url":null,"abstract":"<p>Oncogenes are critical factors in tumorigenesis of diverse cancer types and play essential roles in tumor immune escape. Mutations in Kirsten rat sarcoma viral oncogene homolog (<i>KRAS</i>) and epidermal growth factor receptor (<i>EGFR</i>) are among the most frequent gain-of-function alterations [<span>1</span>]. After many years of in-depth research, inhibitors targeting <i>EGFR</i> or <i>KRAS</i> mutations have been successfully developed, however, their clinical benefit is relatively limited, and they will inevitably encounter the challenge of drug resistance. The emergence of resistance is attributed to secondary mutations in driver genes and other complicated factors. It is worth noting that approved treatment strategies are currently lacking for tumors with different types of <i>KRAS</i> or <i>EGFR</i> mutations, including <i>KRAS<sup>G12D</sup></i>, <i>KRAS<sup>G13D</sup></i>, and <i>EGFR<sup>C797S</sup></i> mutations that are common in tumors [<span>2</span>]. Additionally, oncogene mutations could trigger a cascade of tumor microenvironment changes, ultimately resulting in tumor progression or resistance to programmed death-1 (PD-1) antibody therapy [<span>3, 4</span>]. SYHA1813, a novel vascular endothelial growth factor receptor (VEGFR) and colony-stimulating factor 1 receptor (CSF1R) dual inhibitor, exhibited potent preclinical anti-glioma activity by inhibiting angiogenesis and promoting tumor immunity and showed promising efficacy in an ongoing clinical study (ChiCTR2100045380) [<span>5, 6</span>]. Here, we determined SYHA1813's antitumor activity in tumor models bearing <i>KRAS</i> or <i>EGFR</i> mutations.</p><p>We first examined the effects of SYHA1813 against cell line-derived xenograft (CDX) tumor models containing <i>KRAS<sup>G12C</sup></i> mutation (NCI-H358 lung cancer), <i>KRAS<sup>G12D</sup></i> mutation (PANC-1 pancreatic cancer) and wild-type <i>KRAS</i> (<i>KRAS<sup>WT</sup></i>) (HT-29 colorectal cancer). The results demonstrated that oral administration of SYHA1813 at a dose of 10 mg/kg significantly reduced tumor growth in the NCI-H358 xenograft model, with comparable efficacy to the US Food and Drug Administration (FDA) approved KRAS<sup>G12C</sup> inhibitor sotorasib (AMG510) (Figure 1A). SYHA1813 treatment also resulted in tumor regression in PANC-1 and HT-29 xenograft models (Figure 1B-C). No significant body weight loss was observed in all groups (Supplementary Figure S1). Moreover, considering the emergence of drug resistance as a significant challenge of AMG510, we established a drug resistance model of AMG510 (designated as AMG510R). We found that although AMG510 exhibited attenuated efficacy against the AMG510R model compared to the NCI-H358 model, SYHA1813 could still suppress the growth of drug-resistant tumors at the same dose (Figure 1D). Furthermore, SYHA1813 was evaluated in two patient-derived xenograft (PDX) models, including gastric tumor model GC-1-005 (<i>KRAS<sup>G13D</sup></i>) and colorectal tumor model CRC-1-003 (<i>KRAS<sup>WT</sup></i>) (Figure 1E). SYHA1813 also exhibited potent antitumor activity in these PDX models (Figure 1F-G). Given that SYHA1813 exerted antitumor effects by modulating angiogenesis and macrophages, we further analyzed the expression of the markers associated with angiogenesis, macrophages, and proliferation in NCI-H358 and HT-29 tumor tissues. The immunohistochemistry (IHC) results showed that SYHA1813 significantly reduced the expression of angiogenic marker CD31, macrophage marker F4/80, M2-phenotype macrophage marker CD206 and arginase-1 (ARG1), and the tumor proliferation marker Ki67 (Figure 1H and Supplementary Figure S2). In summary, SYHA1813 exhibited robust antitumor activities across a panel of genetically and histologically heterogeneous <i>KRAS</i>-mutated tumor models, including the model resistant to AMG510.</p><p>We then asked whether SYHA1813 could suppress the growth of tumors carrying <i>EGFR</i> mutations or resistant to EGFR tyrosine kinase inhibitors (TKIs). To address this question, we initially employed the NCI-H1975 model containing <i>EGFR<sup>L858R/T790M</sup></i> mutation, which is resistant to the first-generation EGFR TKIs, and found that SYHA1813 significantly suppressed the tumor growth (Figure 1I). Then, we evaluated the activity of SYHA1813 against the tumor model carrying <i>EGFR<sup>C797S</sup></i> triple mutation (PC-9-OR) that is resistant to third-generation EGFR TKIs [<span>7</span>]. As shown in Figure 1J, osimertinib (AZD9291), the first FDA approved third-generation EGFR TKI, failed to inhibit tumor growth even at the dose of 10 mg/kg. However, we observed a significant deceleration with SYHA1813 in tumor growth rate at doses of 5 and 10 mg/kg (Figure 1J). Furthermore, we employed the osimertinib-resistant tumor (designated as AZDR) [<span>8</span>] and also observed significant suppression upon SYHA1813 monotherapy (Figure 1K). Similarly, in another resistant model (designated as 67R) to the third-generation EGFR TKI ASK120067 [<span>8</span>], SYHA1813 monotherapy inhibited tumor growth (Figure 1L). The IHC results also demonstrated a reduction in angiogenic and macrophage markers in tumor tissues following SYHA1813 administration (Supplementary Figure S3). These findings demonstrated that SYHA1813 exhibited potent antitumor activity in different EGFR TKI-resistant models.</p><p>Given that the response rates to immunotherapy are quite limited across most tumor types, including those carrying <i>KRAS</i> mutations, and importantly, angiogenesis and macrophages have been identified as key factors involved in the resistance to immunotherapy [<span>9</span>], we then validated whether SYHA1813 could enhance the antitumor activity of anti-PD-1 therapy in immunocompetent mice models bearing CT26 (<i>KRAS<sup>G12D</sup></i> mutation) and MC38 (<i>KRAS<sup>WT</sup></i>) colon cancer (Figure 1M-N). In both models, monotherapy with anti-PD-1 antibody or SYHA1813 yielded moderate to marked suppression of tumor growth, while the combination of SYHA1813 and anti-PD-1 antibody conferred significant advantages over each monotherapy group. In the CT26 and MC38 models, the combinational ratio was 1.88 and 1.42, respectively, both exceeding 1, indicative of a synergistic effect (Supplementary Table S1). Flow cytometry analysis revealed that the combination therapy significantly reduced F4/80<sup>+</sup> and CD206<sup>+</sup> macrophages (Figure 1O). Additionally, a noticeable increase of CD8<sup>+</sup> T cells was observed in the combination treatment group, along with an upregulation of granzyme B expression (Figure 1P), indicating an enhanced cytotoxic response against tumor cells.</p><p>Our study discovered the activities of SYHA1813 in <i>KRAS</i>- or <i>EGFR</i>-mutated tumors, providing support for further investigation of SYHA1813. Given potential variations in the response of subcutaneous tumors and other tumor types to angiogenic agents, further experiments in orthotopic models will be conducted. The findings in the current work highlight the feasibility of a therapeutic strategy targeting the tumor microenvironment, which can be applied to treat oncogene-driven cancers and overcome the challenge of drug resistance to targeted therapy.</p><p>Hua Xie, Jian Ding, Wenhu Duan, and Meiyu Geng supervised the project. Yingqiang Liu, Linjiang Tong, Mengge Zhang, Mengzhen Lai, Fang Feng, Haotian Tang, Yi Chen, and Yan Li conducted the experiments. Yingqiang Liu, Mengge Zhang, Qi Zhang and Qiupei Liu analyzed the data. Yingqiang Liu, Qiupei Liu, Hua Xie, Jian Ding, and Wenhu Duan wrote the manuscript.</p><p>The authors declare no potential conflicts of interest.</p><p>This research was supported by grants from the Natural Science Foundation of China for Innovation Research Group (81821005), the National Natural Science Foundation of China (82273948, 81573271 and 81903638), High-level Innovative Research Institute (2021B0909050003), State Key Laboratory of Drug Research (SKLDR-2023-TT-01 and SIMM2205KF-09), Lingang Laboratory (LG202103-02-02), and Institutes for Drug Discovery and Development, Chinese Academy of Sciences (CASIMM0120225003-1 and -2).</p><p>Animal experiments containing cell line-derived xenografts and patient-derived xenografts were carried out in accordance with the Guide for the Care and Use of Laboratory Animals and approved by the Institutional Animal Care and Use Committee of Shanghai Institute of Materia Medica (approval number 2021-04-DJ-59).</p><p>Not applicable.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1000,"publicationDate":"2024-02-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cac2.12521","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cancer Communications","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/cac2.12521","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Oncogenes are critical factors in tumorigenesis of diverse cancer types and play essential roles in tumor immune escape. Mutations in Kirsten rat sarcoma viral oncogene homolog (KRAS) and epidermal growth factor receptor (EGFR) are among the most frequent gain-of-function alterations [1]. After many years of in-depth research, inhibitors targeting EGFR or KRAS mutations have been successfully developed, however, their clinical benefit is relatively limited, and they will inevitably encounter the challenge of drug resistance. The emergence of resistance is attributed to secondary mutations in driver genes and other complicated factors. It is worth noting that approved treatment strategies are currently lacking for tumors with different types of KRAS or EGFR mutations, including KRASG12D, KRASG13D, and EGFRC797S mutations that are common in tumors [2]. Additionally, oncogene mutations could trigger a cascade of tumor microenvironment changes, ultimately resulting in tumor progression or resistance to programmed death-1 (PD-1) antibody therapy [3, 4]. SYHA1813, a novel vascular endothelial growth factor receptor (VEGFR) and colony-stimulating factor 1 receptor (CSF1R) dual inhibitor, exhibited potent preclinical anti-glioma activity by inhibiting angiogenesis and promoting tumor immunity and showed promising efficacy in an ongoing clinical study (ChiCTR2100045380) [5, 6]. Here, we determined SYHA1813's antitumor activity in tumor models bearing KRAS or EGFR mutations.

We first examined the effects of SYHA1813 against cell line-derived xenograft (CDX) tumor models containing KRASG12C mutation (NCI-H358 lung cancer), KRASG12D mutation (PANC-1 pancreatic cancer) and wild-type KRAS (KRASWT) (HT-29 colorectal cancer). The results demonstrated that oral administration of SYHA1813 at a dose of 10 mg/kg significantly reduced tumor growth in the NCI-H358 xenograft model, with comparable efficacy to the US Food and Drug Administration (FDA) approved KRASG12C inhibitor sotorasib (AMG510) (Figure 1A). SYHA1813 treatment also resulted in tumor regression in PANC-1 and HT-29 xenograft models (Figure 1B-C). No significant body weight loss was observed in all groups (Supplementary Figure S1). Moreover, considering the emergence of drug resistance as a significant challenge of AMG510, we established a drug resistance model of AMG510 (designated as AMG510R). We found that although AMG510 exhibited attenuated efficacy against the AMG510R model compared to the NCI-H358 model, SYHA1813 could still suppress the growth of drug-resistant tumors at the same dose (Figure 1D). Furthermore, SYHA1813 was evaluated in two patient-derived xenograft (PDX) models, including gastric tumor model GC-1-005 (KRASG13D) and colorectal tumor model CRC-1-003 (KRASWT) (Figure 1E). SYHA1813 also exhibited potent antitumor activity in these PDX models (Figure 1F-G). Given that SYHA1813 exerted antitumor effects by modulating angiogenesis and macrophages, we further analyzed the expression of the markers associated with angiogenesis, macrophages, and proliferation in NCI-H358 and HT-29 tumor tissues. The immunohistochemistry (IHC) results showed that SYHA1813 significantly reduced the expression of angiogenic marker CD31, macrophage marker F4/80, M2-phenotype macrophage marker CD206 and arginase-1 (ARG1), and the tumor proliferation marker Ki67 (Figure 1H and Supplementary Figure S2). In summary, SYHA1813 exhibited robust antitumor activities across a panel of genetically and histologically heterogeneous KRAS-mutated tumor models, including the model resistant to AMG510.

We then asked whether SYHA1813 could suppress the growth of tumors carrying EGFR mutations or resistant to EGFR tyrosine kinase inhibitors (TKIs). To address this question, we initially employed the NCI-H1975 model containing EGFRL858R/T790M mutation, which is resistant to the first-generation EGFR TKIs, and found that SYHA1813 significantly suppressed the tumor growth (Figure 1I). Then, we evaluated the activity of SYHA1813 against the tumor model carrying EGFRC797S triple mutation (PC-9-OR) that is resistant to third-generation EGFR TKIs [7]. As shown in Figure 1J, osimertinib (AZD9291), the first FDA approved third-generation EGFR TKI, failed to inhibit tumor growth even at the dose of 10 mg/kg. However, we observed a significant deceleration with SYHA1813 in tumor growth rate at doses of 5 and 10 mg/kg (Figure 1J). Furthermore, we employed the osimertinib-resistant tumor (designated as AZDR) [8] and also observed significant suppression upon SYHA1813 monotherapy (Figure 1K). Similarly, in another resistant model (designated as 67R) to the third-generation EGFR TKI ASK120067 [8], SYHA1813 monotherapy inhibited tumor growth (Figure 1L). The IHC results also demonstrated a reduction in angiogenic and macrophage markers in tumor tissues following SYHA1813 administration (Supplementary Figure S3). These findings demonstrated that SYHA1813 exhibited potent antitumor activity in different EGFR TKI-resistant models.

Given that the response rates to immunotherapy are quite limited across most tumor types, including those carrying KRAS mutations, and importantly, angiogenesis and macrophages have been identified as key factors involved in the resistance to immunotherapy [9], we then validated whether SYHA1813 could enhance the antitumor activity of anti-PD-1 therapy in immunocompetent mice models bearing CT26 (KRASG12D mutation) and MC38 (KRASWT) colon cancer (Figure 1M-N). In both models, monotherapy with anti-PD-1 antibody or SYHA1813 yielded moderate to marked suppression of tumor growth, while the combination of SYHA1813 and anti-PD-1 antibody conferred significant advantages over each monotherapy group. In the CT26 and MC38 models, the combinational ratio was 1.88 and 1.42, respectively, both exceeding 1, indicative of a synergistic effect (Supplementary Table S1). Flow cytometry analysis revealed that the combination therapy significantly reduced F4/80+ and CD206+ macrophages (Figure 1O). Additionally, a noticeable increase of CD8+ T cells was observed in the combination treatment group, along with an upregulation of granzyme B expression (Figure 1P), indicating an enhanced cytotoxic response against tumor cells.

Our study discovered the activities of SYHA1813 in KRAS- or EGFR-mutated tumors, providing support for further investigation of SYHA1813. Given potential variations in the response of subcutaneous tumors and other tumor types to angiogenic agents, further experiments in orthotopic models will be conducted. The findings in the current work highlight the feasibility of a therapeutic strategy targeting the tumor microenvironment, which can be applied to treat oncogene-driven cancers and overcome the challenge of drug resistance to targeted therapy.

Hua Xie, Jian Ding, Wenhu Duan, and Meiyu Geng supervised the project. Yingqiang Liu, Linjiang Tong, Mengge Zhang, Mengzhen Lai, Fang Feng, Haotian Tang, Yi Chen, and Yan Li conducted the experiments. Yingqiang Liu, Mengge Zhang, Qi Zhang and Qiupei Liu analyzed the data. Yingqiang Liu, Qiupei Liu, Hua Xie, Jian Ding, and Wenhu Duan wrote the manuscript.

The authors declare no potential conflicts of interest.

This research was supported by grants from the Natural Science Foundation of China for Innovation Research Group (81821005), the National Natural Science Foundation of China (82273948, 81573271 and 81903638), High-level Innovative Research Institute (2021B0909050003), State Key Laboratory of Drug Research (SKLDR-2023-TT-01 and SIMM2205KF-09), Lingang Laboratory (LG202103-02-02), and Institutes for Drug Discovery and Development, Chinese Academy of Sciences (CASIMM0120225003-1 and -2).

Animal experiments containing cell line-derived xenografts and patient-derived xenografts were carried out in accordance with the Guide for the Care and Use of Laboratory Animals and approved by the Institutional Animal Care and Use Committee of Shanghai Institute of Materia Medica (approval number 2021-04-DJ-59).

Not applicable.

Abstract Image

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
通过靶向肿瘤微环境和协同增强抗 PD-1 免疫疗法治疗癌基因突变肿瘤的新策略。
癌基因是多种癌症类型肿瘤发生的关键因素,在肿瘤免疫逃逸中发挥着重要作用。Kirsten 大鼠肉瘤病毒癌基因同源物(KRAS)和表皮生长因子受体(EGFR)的突变是最常见的功能获得性改变[1]。经过多年的深入研究,针对表皮生长因子受体(EGFR)或 KRAS 突变的抑制剂已被成功开发出来,但其临床疗效相对有限,且不可避免地会遇到耐药性的挑战。耐药性的出现归因于驱动基因的二次突变和其他复杂因素。值得注意的是,对于不同类型的 KRAS 或 EGFR 突变的肿瘤,包括肿瘤中常见的 KRASG12D、KRASG13D 和 EGFRC797S 突变,目前尚缺乏获批的治疗策略[2]。此外,癌基因突变可能引发一连串的肿瘤微环境变化,最终导致肿瘤进展或对程序性死亡-1(PD-1)抗体疗法产生抗药性[3, 4]。SYHA1813是一种新型血管内皮生长因子受体(VEGFR)和集落刺激因子1受体(CSF1R)双重抑制剂,在临床前通过抑制血管生成和促进肿瘤免疫表现出强大的抗胶质瘤活性,并在一项正在进行的临床研究(ChiCTR2100045380)中显示出良好的疗效[5, 6]。我们首先研究了SYHA1813对含有KRASG12C突变(NCI-H358肺癌)、KRASG12D突变(PANC-1胰腺癌)和野生型KRAS(KRASWT)(HT-29结直肠癌)的细胞系衍生异种移植(CDX)肿瘤模型的作用。结果表明,在NCI-H358异种移植模型中,口服10毫克/千克剂量的SYHA1813可显著降低肿瘤生长,其疗效与美国食品药品管理局(FDA)批准的KRASG12C抑制剂索托拉西布(AMG510)相当(图1A)。SYHA1813治疗还导致PANC-1和HT-29异种移植模型中的肿瘤消退(图1B-C)。所有组均未观察到明显的体重下降(补充图 S1)。此外,考虑到耐药性的出现是 AMG510 面临的一个重大挑战,我们建立了 AMG510 的耐药模型(命名为 AMG510R)。我们发现,虽然与 NCI-H358 模型相比,AMG510 对 AMG510R 模型的疗效有所减弱,但 SYHA1813 仍能在相同剂量下抑制耐药肿瘤的生长(图 1D)。此外,SYHA1813还在两种患者来源异种移植(PDX)模型中进行了评估,包括胃肿瘤模型GC-1-005(KRASG13D)和结直肠肿瘤模型CRC-1-003(KRASWT)(图1E)。SYHA1813 在这些 PDX 模型中也表现出了强大的抗肿瘤活性(图 1F-G)。鉴于 SYHA1813 通过调节血管生成和巨噬细胞发挥抗肿瘤作用,我们进一步分析了 NCI-H358 和 HT-29 肿瘤组织中与血管生成、巨噬细胞和增殖相关的标记物的表达。免疫组化(IHC)结果显示,SYHA1813能显著降低血管生成标志物CD31、巨噬细胞标志物F4/80、M2型巨噬细胞标志物CD206和精氨酸酶-1(ARG1)以及肿瘤增殖标志物Ki67的表达(图1H和补充图S2)。总之,SYHA1813在一组基因和组织学异质性的KRAS突变肿瘤模型(包括对AMG510耐药的模型)中表现出强大的抗肿瘤活性。为了解决这个问题,我们首先采用了含有EGFRL858R/T790M突变的NCI-H1975模型,该模型对第一代表皮生长因子受体TKIs耐药,结果发现SYHA1813能显著抑制肿瘤的生长(图1I)。然后,我们评估了 SYHA1813 对携带 EGFRC797S 三重突变的肿瘤模型(PC-9-OR)的活性,该模型对第三代 EGFR TKIs 具有耐药性[7]。如图 1J 所示,美国 FDA 批准的第一种第三代 EGFR TKI--奥西替尼(AZD9291)即使剂量为 10 mg/kg,也无法抑制肿瘤生长。然而,我们观察到 SYHA1813 在 5 毫克/公斤和 10 毫克/公斤剂量下的肿瘤生长速度明显减慢(图 1J)。此外,我们还采用了奥希替尼耐药肿瘤(命名为 AZDR)[8],也观察到 SYHA1813 单药治疗后肿瘤生长速度明显下降(图 1K)。同样,在另一个对第三代表皮生长因子受体 TKI ASK120067 [8]耐药的模型(命名为 67R)中,SYHA1813 单药治疗也抑制了肿瘤生长(图 1L)。IHC 结果还显示,服用 SYHA1813 后,肿瘤组织中的血管生成标记物和巨噬细胞标记物减少(补充图 S3)。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
Cancer Communications
Cancer Communications Biochemistry, Genetics and Molecular Biology-Cancer Research
CiteScore
25.50
自引率
4.30%
发文量
153
审稿时长
4 weeks
期刊介绍: Cancer Communications is an open access, peer-reviewed online journal that encompasses basic, clinical, and translational cancer research. The journal welcomes submissions concerning clinical trials, epidemiology, molecular and cellular biology, and genetics.
期刊最新文献
Perioperative toripalimab plus neoadjuvant chemotherapy might improve outcomes in resectable esophageal cancer: an interim analysis of a phase III randomized clinical trial. Tryptophan 2,3-dioxygenase-positive matrix fibroblasts fuel breast cancer lung metastasis via kynurenine-mediated ferroptosis resistance of metastatic cells and T cell dysfunction. Targeting SRSF10 might inhibit M2 macrophage polarization and potentiate anti-PD-1 therapy in hepatocellular carcinoma. Unraveling the routes to distant metastases in colorectal cancer: Tumor deposits and lymph node metastases as the gateway. Issue Information
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1