Your browser doesn't support javascript.
loading
Microdissection tools to generate organoids for modeling the tumor immune microenvironment.
Cordts, Seth C; Yuki, Kanako; Henao Echeverri, Maria F; Narasimhan, Balasubramanian; Kuo, Calvin J; Tang, Sindy K Y.
Afiliação
  • Cordts SC; Department of Mechanical Engineering, Stanford University, Stanford, CA, USA.
  • Yuki K; Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA.
  • Henao Echeverri MF; Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA.
  • Narasimhan B; Department of Statistics, Stanford University, Stanford, USA.
  • Kuo CJ; Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA.
  • Tang SKY; Department of Mechanical Engineering, Stanford University, Stanford, CA, USA. sindy@stanford.edu.
Microsyst Nanoeng ; 10(1): 126, 2024 Sep 10.
Article em En | MEDLINE | ID: mdl-39251611
ABSTRACT
Patient-derived tumor organoids have emerged as promising models for predicting personalized drug responses in cancer therapy, but they typically lack immune components. Preserving the in vivo association between tumor cells and endogenous immune cells is critical for accurate testing of cancer immunotherapies. Mechanical dissection of tumor specimens into tumor fragments, as opposed to enzymatic digestion into single cells, is essential for maintaining these native tumor-immune cell spatial relationships. However, conventional mechanical dissection relying on manual mincing is time-consuming and irreproducible. This study describes two microdissection devices, the µDicer and µGrater, to facilitate the generation of intact tumor fragments from mouse B16 melanoma, a common model of human melanoma. The µDicer- and µGrater-cut tumor fragments were used to generate air‒liquid interface (ALI) organoids that copreserve tumor cells with infiltrating immune subsets without artificial reconstitution. The µDicer, consisting of a hexagonal array of silicon microblades, was employed to investigate the effect of organoid size. The viability of ALI organoid immune cells appeared insensitive to organoid sizes exceeding ~400 µm but diminished in organoids ~200 µm in size. The µGrater, consisting of an array of submillimeter holes in stainless steel, was employed to accelerate dissection. For the samples studied, the µGrater was 4.5 times faster than manual mincing. Compared with those generated by manual mincing, ALI organoids generated by the µGrater demonstrated similar viability, immune cell composition, and responses to anti-PD-1 immunotherapy. With further optimization, the µGrater holds potential for integration into clinical workflows to support the advancement of personalized cancer immunotherapy.

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2024 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2024 Tipo de documento: Article