Cargando…

Rapid Profiling of Tumor‐Immune Interaction Using Acoustically Assembled Patient‐Derived Cell Clusters

Tumor microenvironment crosstalk, in particular interactions between cancer cells, T cells, and myeloid‐derived suppressor cells (MDSCs), mediates tumor initiation, progression, and response to treatment. However, current patient‐derived models such as tumor organoids and 2D cultures lack some essen...

Descripción completa

Detalles Bibliográficos
Autores principales: Ao, Zheng, Wu, Zhuhao, Cai, Hongwei, Hu, Liya, Li, Xiang, Kaurich, Connor, Chang, Jackson, Gu, Mingxia, Cheng, Liang, Lu, Xin, Guo, Feng
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9353481/
https://www.ncbi.nlm.nih.gov/pubmed/35611994
http://dx.doi.org/10.1002/advs.202201478
_version_ 1784762872904024064
author Ao, Zheng
Wu, Zhuhao
Cai, Hongwei
Hu, Liya
Li, Xiang
Kaurich, Connor
Chang, Jackson
Gu, Mingxia
Cheng, Liang
Lu, Xin
Guo, Feng
author_facet Ao, Zheng
Wu, Zhuhao
Cai, Hongwei
Hu, Liya
Li, Xiang
Kaurich, Connor
Chang, Jackson
Gu, Mingxia
Cheng, Liang
Lu, Xin
Guo, Feng
author_sort Ao, Zheng
collection PubMed
description Tumor microenvironment crosstalk, in particular interactions between cancer cells, T cells, and myeloid‐derived suppressor cells (MDSCs), mediates tumor initiation, progression, and response to treatment. However, current patient‐derived models such as tumor organoids and 2D cultures lack some essential niche cell types (e.g., MDSCs) and fail to model complex tumor‐immune interactions. Here, the authors present the novel acoustically assembled patient‐derived cell clusters (APCCs) that can preserve original tumor/immune cell compositions, model their interactions in 3D microenvironments, and test the treatment responses of primary tumors in a rapid, scalable, and user‐friendly manner. By incorporating a large array of 3D acoustic trappings within the extracellular matrix, hundreds of APCCs can be assembled within a petri dish within 2 min. Moreover, the APCCs can preserve sensitive and short‐lived (≈1 to 2‐day lifespan in vivo) tumor‐induced MDSCs and model their dynamic suppression of T cell tumor toxicity for up to 24 h. Finally, using the APCCs, the authors succesully model the combinational therapeutic effect of a multi‐kinase inhibitor targeting MDSCs (cabozantinib) and an anti‐PD‐1 immune checkpoint inhibitor (pembrolizumab). The novel APCCs may hold promising potential in predicting treatment response for personalized cancer adjuvant therapy as well as screening novel cancer immunotherapy and combinational therapy.
format Online
Article
Text
id pubmed-9353481
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-93534812022-08-09 Rapid Profiling of Tumor‐Immune Interaction Using Acoustically Assembled Patient‐Derived Cell Clusters Ao, Zheng Wu, Zhuhao Cai, Hongwei Hu, Liya Li, Xiang Kaurich, Connor Chang, Jackson Gu, Mingxia Cheng, Liang Lu, Xin Guo, Feng Adv Sci (Weinh) Research Articles Tumor microenvironment crosstalk, in particular interactions between cancer cells, T cells, and myeloid‐derived suppressor cells (MDSCs), mediates tumor initiation, progression, and response to treatment. However, current patient‐derived models such as tumor organoids and 2D cultures lack some essential niche cell types (e.g., MDSCs) and fail to model complex tumor‐immune interactions. Here, the authors present the novel acoustically assembled patient‐derived cell clusters (APCCs) that can preserve original tumor/immune cell compositions, model their interactions in 3D microenvironments, and test the treatment responses of primary tumors in a rapid, scalable, and user‐friendly manner. By incorporating a large array of 3D acoustic trappings within the extracellular matrix, hundreds of APCCs can be assembled within a petri dish within 2 min. Moreover, the APCCs can preserve sensitive and short‐lived (≈1 to 2‐day lifespan in vivo) tumor‐induced MDSCs and model their dynamic suppression of T cell tumor toxicity for up to 24 h. Finally, using the APCCs, the authors succesully model the combinational therapeutic effect of a multi‐kinase inhibitor targeting MDSCs (cabozantinib) and an anti‐PD‐1 immune checkpoint inhibitor (pembrolizumab). The novel APCCs may hold promising potential in predicting treatment response for personalized cancer adjuvant therapy as well as screening novel cancer immunotherapy and combinational therapy. John Wiley and Sons Inc. 2022-05-25 /pmc/articles/PMC9353481/ /pubmed/35611994 http://dx.doi.org/10.1002/advs.202201478 Text en © 2022 The Authors. Advanced Science published by Wiley‐VCH GmbH https://creativecommons.org/licenses/by/4.0/This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research Articles
Ao, Zheng
Wu, Zhuhao
Cai, Hongwei
Hu, Liya
Li, Xiang
Kaurich, Connor
Chang, Jackson
Gu, Mingxia
Cheng, Liang
Lu, Xin
Guo, Feng
Rapid Profiling of Tumor‐Immune Interaction Using Acoustically Assembled Patient‐Derived Cell Clusters
title Rapid Profiling of Tumor‐Immune Interaction Using Acoustically Assembled Patient‐Derived Cell Clusters
title_full Rapid Profiling of Tumor‐Immune Interaction Using Acoustically Assembled Patient‐Derived Cell Clusters
title_fullStr Rapid Profiling of Tumor‐Immune Interaction Using Acoustically Assembled Patient‐Derived Cell Clusters
title_full_unstemmed Rapid Profiling of Tumor‐Immune Interaction Using Acoustically Assembled Patient‐Derived Cell Clusters
title_short Rapid Profiling of Tumor‐Immune Interaction Using Acoustically Assembled Patient‐Derived Cell Clusters
title_sort rapid profiling of tumor‐immune interaction using acoustically assembled patient‐derived cell clusters
topic Research Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9353481/
https://www.ncbi.nlm.nih.gov/pubmed/35611994
http://dx.doi.org/10.1002/advs.202201478
work_keys_str_mv AT aozheng rapidprofilingoftumorimmuneinteractionusingacousticallyassembledpatientderivedcellclusters
AT wuzhuhao rapidprofilingoftumorimmuneinteractionusingacousticallyassembledpatientderivedcellclusters
AT caihongwei rapidprofilingoftumorimmuneinteractionusingacousticallyassembledpatientderivedcellclusters
AT huliya rapidprofilingoftumorimmuneinteractionusingacousticallyassembledpatientderivedcellclusters
AT lixiang rapidprofilingoftumorimmuneinteractionusingacousticallyassembledpatientderivedcellclusters
AT kaurichconnor rapidprofilingoftumorimmuneinteractionusingacousticallyassembledpatientderivedcellclusters
AT changjackson rapidprofilingoftumorimmuneinteractionusingacousticallyassembledpatientderivedcellclusters
AT gumingxia rapidprofilingoftumorimmuneinteractionusingacousticallyassembledpatientderivedcellclusters
AT chengliang rapidprofilingoftumorimmuneinteractionusingacousticallyassembledpatientderivedcellclusters
AT luxin rapidprofilingoftumorimmuneinteractionusingacousticallyassembledpatientderivedcellclusters
AT guofeng rapidprofilingoftumorimmuneinteractionusingacousticallyassembledpatientderivedcellclusters