HBOC alleviated tumour hypoxia during radiotherapy more intensely in large solid tumours than regular ones.

IF 4.5 3区 生物学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Artificial Cells, Nanomedicine, and Biotechnology Pub Date : 2024-12-01 Epub Date: 2023-11-23 DOI:10.1080/21691401.2023.2276768
Yingcan Xu, Kehui Zhu, Jiakang Wu, Shifan Zheng, Rui Zhong, Wentao Zhou, Ye Cao, Jiaxin Liu, Hong Wang
{"title":"HBOC alleviated tumour hypoxia during radiotherapy more intensely in large solid tumours than regular ones.","authors":"Yingcan Xu, Kehui Zhu, Jiakang Wu, Shifan Zheng, Rui Zhong, Wentao Zhou, Ye Cao, Jiaxin Liu, Hong Wang","doi":"10.1080/21691401.2023.2276768","DOIUrl":null,"url":null,"abstract":"<p><p>Radiotherapy (RT) is a highly valuable method in cancer therapy, but its therapeutic efficacy is limited by its side effects and tumour radiation resistance. The resistance is mainly induced by hypoxia in the tumour microenvironment (TME). As a nano-oxygen carrier, Haemoglobin-based oxygen carriers (HBOCs) administration is a promising strategy to alleviate tumour hypoxia which may remodel TME to ameliorate radiation resistance and enable RT more effective. In this study, we administered fractionated RT combined with HBOC to treat Miapaca-2 cell and Hela cell xenografts on nude mice. The study found that HBOC relieved hypoxic environment and down-regulate expression of hypoxia-inducible factor-1α (Hif-1α) both in regular (100 mm<sup>3</sup>) and large (360/400 mm<sup>3</sup>) tumours. The proliferation and metastasis of tumour tissue also decreased after HBOC application. Nevertheless, <i>in vivo</i> RT combined with HBOC performed more effectively to suppress tumour growth in large tumours than in regular tumours. This is due to more severe hypoxic regions exist in the large solid tumours compared to the regular counterparts, and HBOC administration may be more effective in alleviating hypoxia in large tumours. Thus, HBOC sensitization therapy is more suitable for large solid tumours.</p>","PeriodicalId":8736,"journal":{"name":"Artificial Cells, Nanomedicine, and Biotechnology","volume":null,"pages":null},"PeriodicalIF":4.5000,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Artificial Cells, Nanomedicine, and Biotechnology","FirstCategoryId":"5","ListUrlMain":"https://doi.org/10.1080/21691401.2023.2276768","RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2023/11/23 0:00:00","PubModel":"Epub","JCR":"Q1","JCRName":"BIOTECHNOLOGY & APPLIED MICROBIOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Radiotherapy (RT) is a highly valuable method in cancer therapy, but its therapeutic efficacy is limited by its side effects and tumour radiation resistance. The resistance is mainly induced by hypoxia in the tumour microenvironment (TME). As a nano-oxygen carrier, Haemoglobin-based oxygen carriers (HBOCs) administration is a promising strategy to alleviate tumour hypoxia which may remodel TME to ameliorate radiation resistance and enable RT more effective. In this study, we administered fractionated RT combined with HBOC to treat Miapaca-2 cell and Hela cell xenografts on nude mice. The study found that HBOC relieved hypoxic environment and down-regulate expression of hypoxia-inducible factor-1α (Hif-1α) both in regular (100 mm3) and large (360/400 mm3) tumours. The proliferation and metastasis of tumour tissue also decreased after HBOC application. Nevertheless, in vivo RT combined with HBOC performed more effectively to suppress tumour growth in large tumours than in regular tumours. This is due to more severe hypoxic regions exist in the large solid tumours compared to the regular counterparts, and HBOC administration may be more effective in alleviating hypoxia in large tumours. Thus, HBOC sensitization therapy is more suitable for large solid tumours.

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
HBOC对大实体瘤放疗期间肿瘤缺氧的缓解作用强于普通实体瘤。
放射治疗是一种非常有价值的癌症治疗方法,但其副作用和肿瘤的放射耐药性限制了其治疗效果。耐药主要由肿瘤微环境(TME)缺氧引起。作为一种纳米氧载体,血红蛋白基氧载体(HBOCs)给药是一种很有前景的缓解肿瘤缺氧的策略,它可能会重塑TME,改善辐射抵抗,使RT更有效。在本研究中,我们采用分馏RT联合HBOC治疗裸鼠Miapaca-2细胞和Hela细胞异种移植。研究发现,HBOC在常规肿瘤(100 mm3)和大肿瘤(360/400 mm3)中均能缓解缺氧环境,下调缺氧诱导因子-1α (Hif-1α)的表达。应用HBOC后,肿瘤组织的增殖和转移也有所减少。然而,体内RT联合HBOC在抑制大肿瘤肿瘤生长方面比在常规肿瘤中更有效。这是由于与常规肿瘤相比,大实体肿瘤中存在更严重的缺氧区域,HBOC给药可能更有效地缓解大肿瘤中的缺氧。因此,HBOC增敏治疗更适合于大型实体瘤。
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
Artificial Cells, Nanomedicine, and Biotechnology
Artificial Cells, Nanomedicine, and Biotechnology BIOTECHNOLOGY & APPLIED MICROBIOLOGY-ENGINEERING, BIOMEDICAL
CiteScore
10.90
自引率
0.00%
发文量
48
审稿时长
20 weeks
期刊介绍: Artificial Cells, Nanomedicine and Biotechnology covers the frontiers of interdisciplinary research and application, combining artificial cells, nanotechnology, nanobiotechnology, biotechnology, molecular biology, bioencapsulation, novel carriers, stem cells and tissue engineering. Emphasis is on basic research, applied research, and clinical and industrial applications of the following topics:artificial cellsblood substitutes and oxygen therapeuticsnanotechnology, nanobiotecnology, nanomedicinetissue engineeringstem cellsbioencapsulationmicroencapsulation and nanoencapsulationmicroparticles and nanoparticlesliposomescell therapy and gene therapyenzyme therapydrug delivery systemsbiodegradable and biocompatible polymers for scaffolds and carriersbiosensorsimmobilized enzymes and their usesother biotechnological and nanobiotechnological approachesRapid progress in modern research cannot be carried out in isolation and is based on the combined use of the different novel approaches. The interdisciplinary research involving novel approaches, as discussed above, has revolutionized this field resulting in rapid developments. This journal serves to bring these different, modern and futuristic approaches together for the academic, clinical and industrial communities to allow for even greater developments of this highly interdisciplinary area.
期刊最新文献
Zingiberis rhizoma-based carbon dots alter serum oestradiol and follicle-stimulating hormone levels in female mice. HBOC alleviated tumour hypoxia during radiotherapy more intensely in large solid tumours than regular ones. Inhibitory effects of Curcumae Radix carbonisata-based carbon dots against liver fibrosis induced by carbon tetrachloride in mice. Haemostatic potency of sodium alginate/aloe vera/sericin composite scaffolds - preparation, characterisation, and evaluation. Investigation of pharmacokinetics and immunogenicity of magnetosomes.
×
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
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