文献检索文档翻译深度研究
Suppr Zotero 插件Zotero 插件
邀请有礼套餐&价格历史记录

新学期,新优惠

限时优惠:9月1日-9月22日

30天高级会员仅需29元

1天体验卡首发特惠仅需5.99元

了解详情
不再提醒
插件&应用
Suppr Zotero 插件Zotero 插件浏览器插件Mac 客户端Windows 客户端微信小程序
高级版
套餐订阅购买积分包
AI 工具
文献检索文档翻译深度研究
关于我们
关于 Suppr公司介绍联系我们用户协议隐私条款
关注我们

Suppr 超能文献

核心技术专利:CN118964589B侵权必究
粤ICP备2023148730 号-1Suppr @ 2025

卡巴他赛对巨噬细胞的作用改善了针对 CD47 的免疫疗法在三阴性乳腺癌中的疗效。

Effect of cabazitaxel on macrophages improves CD47-targeted immunotherapy for triple-negative breast cancer.

机构信息

Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, California, USA.

Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA.

出版信息

J Immunother Cancer. 2021 Mar;9(3). doi: 10.1136/jitc-2020-002022.


DOI:10.1136/jitc-2020-002022
PMID:33753567
原文链接:https://pmc.ncbi.nlm.nih.gov/articles/PMC7986678/
Abstract

BACKGROUND: Limited therapeutic options are available for triple-negative breast cancer (TNBC), emphasizing an urgent need for more effective treatment approaches. The development of strategies by targeting tumor-associated macrophages (TAMs) to stimulate their ability of Programmed Cell Removal (PrCR) provides a promising new immunotherapy for TNBC treatment. METHODS: CD47 is a critical self-protective "don't eat me" signal on multiple human cancers against macrophage immunosurveillance. Using human and mouse TNBC preclinical models, we evaluated the efficacy of PrCR-based immunotherapy by blocking CD47. We performed high-throughput screens on FDA-approved anti-cancer small molecule compounds for agents potentiating PrCR and enhancing the efficacy of CD47-targeted therapy for TNBC treatment. RESULTS: We showed that CD47 was widely expressed on TNBC cells and TAMs represented the most abundant immune cell population in TNBC tumors. Blockade of CD47 enabled PrCR of TNBC cells, but the efficacy was not satisfactory. Our high-throughput screens identified cabazitaxel in enhancing PrCR-based immunotherapy. A combination of CD47 blockade and cabazitaxel treatment yielded a highly effective treatment strategy, promoting PrCR of TNBC cells and inhibiting tumor development and metastasis in preclinical models. We demonstrated that cabazitaxel potentiated PrCR by activating macrophages, independent of its cytotoxicity toward cancer cells. When treated with cabazitaxel, the molecular and phenotypic signatures of macrophages were polarized toward M1 state, and the NF-kB signaling pathway became activated. CONCLUSION: The combination of CD47 blockade and macrophage activation by cabazitaxel synergizes to vastly enhance the elimination of TNBC cells. Our results show that targeting macrophages is a promising and effective strategy for TNBC treatment.

摘要

背景:三阴性乳腺癌(TNBC)的治疗选择有限,这凸显了迫切需要更有效的治疗方法。通过靶向肿瘤相关巨噬细胞(TAMs)来刺激其程序性细胞清除(PrCR)能力的策略为 TNBC 的治疗提供了一种很有前途的新免疫疗法。

方法:CD47 是多种人类癌症针对巨噬细胞免疫监视的关键自我保护“别吃我”信号。我们使用人类和小鼠 TNBC 临床前模型,通过阻断 CD47 来评估基于 PrCR 的免疫疗法的疗效。我们对 FDA 批准的抗癌小分子化合物进行了高通量筛选,以寻找增强 PrCR 并提高 CD47 靶向治疗 TNBC 疗效的药物。

结果:我们表明,CD47 在 TNBC 细胞上广泛表达,TAMs 是 TNBC 肿瘤中最丰富的免疫细胞群体。阻断 CD47 可使 TNBC 细胞发生 PrCR,但疗效并不理想。我们的高通量筛选鉴定出卡巴他赛可增强基于 PrCR 的免疫疗法。CD47 阻断与卡巴他赛联合治疗产生了一种非常有效的治疗策略,可促进 TNBC 细胞的 PrCR,并抑制临床前模型中的肿瘤发展和转移。我们证明,卡巴他赛通过激活巨噬细胞增强 PrCR,而不依赖于其对癌细胞的细胞毒性。用卡巴他赛处理后,巨噬细胞的分子和表型特征向 M1 状态极化,NF-kB 信号通路被激活。

结论:CD47 阻断与卡巴他赛激活巨噬细胞的联合作用可极大增强 TNBC 细胞的清除。我们的研究结果表明,靶向巨噬细胞是一种很有前途且有效的 TNBC 治疗策略。

https://cdn.ncbi.nlm.nih.gov/pmc/blobs/5b97/7986678/1356fd67e689/jitc-2020-002022f06.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/5b97/7986678/ebf48b07c673/jitc-2020-002022f01.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/5b97/7986678/ea86856f6ac4/jitc-2020-002022f02.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/5b97/7986678/9cd3a68d3214/jitc-2020-002022f03.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/5b97/7986678/8a191a5fc750/jitc-2020-002022f04.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/5b97/7986678/f4d099a71466/jitc-2020-002022f05.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/5b97/7986678/1356fd67e689/jitc-2020-002022f06.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/5b97/7986678/ebf48b07c673/jitc-2020-002022f01.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/5b97/7986678/ea86856f6ac4/jitc-2020-002022f02.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/5b97/7986678/9cd3a68d3214/jitc-2020-002022f03.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/5b97/7986678/8a191a5fc750/jitc-2020-002022f04.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/5b97/7986678/f4d099a71466/jitc-2020-002022f05.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/5b97/7986678/1356fd67e689/jitc-2020-002022f06.jpg

相似文献

[1]
Effect of cabazitaxel on macrophages improves CD47-targeted immunotherapy for triple-negative breast cancer.

J Immunother Cancer. 2021-3

[2]
A polymeric nanoplatform enhances the cGAS-STING pathway in macrophages to potentiate phagocytosis for cancer immunotherapy.

J Control Release. 2024-9

[3]
Supermolecular nanovehicles co-delivering TLR7/8-agonist and anti-CD47 siRNA for enhanced tumor immunotherapy.

Int J Biol Macromol. 2023-11-1

[4]
Taraxacum mongolicum extract inhibited malignant phenotype of triple-negative breast cancer cells in tumor-associated macrophages microenvironment through suppressing IL-10 / STAT3 / PD-L1 signaling pathways.

J Ethnopharmacol. 2021-6-28

[5]
Targeting CD47 in Anaplastic Thyroid Carcinoma Enhances Tumor Phagocytosis by Macrophages and Is a Promising Therapeutic Strategy.

Thyroid. 2019-5-10

[6]
Novel insights into paclitaxel's role on tumor-associated macrophages in enhancing PD-1 blockade in breast cancer treatment.

J Immunother Cancer. 2024-7-15

[7]
Targeting a cell surface vitamin D receptor on tumor-associated macrophages in triple-negative breast cancer.

Elife. 2021-6-1

[8]
Targeting tumor cell-to-macrophage communication by blocking Vtn-C1qbp interaction inhibits tumor progression via enhancing macrophage phagocytosis.

Theranostics. 2024

[9]
IL1R2 Blockade Alleviates Immunosuppression and Potentiates Anti-PD-1 Efficacy in Triple-Negative Breast Cancer.

Cancer Res. 2024-7-15

[10]
Modulating macrophage-mediated programmed cell removal: An attractive strategy for cancer therapy.

Biochim Biophys Acta Rev Cancer. 2024-9

引用本文的文献

[1]
Macrophage polarization in disease therapy: insights from astragaloside IV and cycloastragenol.

Front Pharmacol. 2025-5-23

[2]
Mechanisms of tumor-associated macrophages in breast cancer and treatment strategy.

Front Immunol. 2025-2-28

[3]
Triple-Negative Breast Cancer Systemic Treatment: Disruptive Early-Stage Developments for Overcoming Stagnation in the Advanced Pipeline.

Cancers (Basel). 2025-2-13

[4]
Phosphoproteomic Profiling Reveals mTOR Signaling in Sustaining Macrophage Phagocytosis of Cancer Cells.

Cancers (Basel). 2024-12-19

[5]
A protocol for high-throughput screening for small chemicals promoting macrophage-mediated tumor cell phagocytosis in mice.

STAR Protoc. 2025-3-21

[6]
Bruton Tyrosine Kinase Inhibition Decreases Inflammation and Differentially Impacts Phagocytosis and Cellular Metabolism in Mouse- and Human-derived Myeloid Cells.

Immunohorizons. 2024-9-1

[7]
Targeting the CD47/SIRPα pathway in malignancies: recent progress, difficulties and future perspectives.

Front Oncol. 2024-7-5

[8]
Enhanced treatment of breast cancer brain metastases with oncolytic virus expressing anti-CD47 antibody and temozolomide.

Mol Ther Oncol. 2024-6-5

[9]
Beyond Anti-PD-1/PD-L1: Improving Immune Checkpoint Inhibitor Responses in Triple-Negative Breast Cancer.

Cancers (Basel). 2024-6-11

[10]
Prediction of Tumor-Associated Macrophages and Immunotherapy Benefits Using Weakly Supervised Contrastive Learning in Breast Cancer Pathology Images.

J Imaging Inform Med. 2024-12

本文引用的文献

[1]
Phase 2 study of cabazitaxel as second-line treatment in patients with HER2-negative metastatic breast cancer previously treated with taxanes-a Hellenic Cooperative Oncology Group (HeCOG) Trial.

Br J Cancer. 2020-8

[2]
Visualizing and interpreting cancer genomics data via the Xena platform.

Nat Biotechnol. 2020-6

[3]
Macrophage checkpoint blockade: results from initial clinical trials, binding analyses, and CD47-SIRPα structure-function.

Antib Ther. 2020-4

[4]
FDA Approval Summary: Atezolizumab Plus Paclitaxel Protein-bound for the Treatment of Patients with Advanced or Metastatic TNBC Whose Tumors Express PD-L1.

Clin Cancer Res. 2020-5-15

[5]
Cabazitaxel versus Abiraterone or Enzalutamide in Metastatic Prostate Cancer.

N Engl J Med. 2019-9-30

[6]
Diversity, Mechanisms, and Significance of Macrophage Plasticity.

Annu Rev Pathol. 2019-9-17

[7]
Phagocytosis checkpoints as new targets for cancer immunotherapy.

Nat Rev Cancer. 2019-8-28

[8]
High expression of CD47 in triple negative breast cancer is associated with epithelial-mesenchymal transition and poor prognosis.

Oncol Lett. 2019-9

[9]
CD24 signalling through macrophage Siglec-10 is a target for cancer immunotherapy.

Nature. 2019-7-31

[10]
Recent advances in triple negative breast cancer: the immunotherapy era.

BMC Med. 2019-5-9

文献AI研究员

20分钟写一篇综述,助力文献阅读效率提升50倍

立即体验

用中文搜PubMed

大模型驱动的PubMed中文搜索引擎

马上搜索

推荐工具

医学文档翻译智能文献检索