Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2023 Jun 12;21(1):383.
doi: 10.1186/s12967-023-04206-3.

Inhalable CAR-T cell-derived exosomes as paclitaxel carriers for treating lung cancer

Affiliations

Inhalable CAR-T cell-derived exosomes as paclitaxel carriers for treating lung cancer

Wei Zheng et al. J Transl Med. .

Abstract

Background: Non-small cell lung cancer (NSCLC) is a worldwide health threat with high annual morbidity and mortality. Chemotherapeutic drugs such as paclitaxel (PTX) have been widely applied clinically. However, systemic toxicity due to the non-specific circulation of PTX often leads to multi-organ damage, including to the liver and kidney. Thus, it is necessary to develop a novel strategy to enhance the targeted antitumor effects of PTX.

Methods: Here, we engineered exosomes derived from T cells expressing the chimeric antigen receptor (CAR-Exos), which targeted mesothelin (MSLN)-expressing Lewis lung cancer (MSLN-LLC) through the anti-MSLN single-chain variable fragment (scFv) of CAR-Exos. PTX was encapsulated into CAR-Exos (PTX@CAR-Exos) and administered via inhalation to an orthotopic lung cancer mouse model.

Results: Inhaled PTX@CAR-Exos accumulated within the tumor area, reduced tumor size, and prolonged survival with little toxicity. In addition, PTX@CAR-Exos reprogrammed the tumor microenvironment and reversed the immunosuppression, which was attributed to infiltrating CD8+ T cells and elevated IFN-γ and TNF-α levels.

Conclusions: Our study provides a nanovesicle-based delivery platform to promote the efficacy of chemotherapeutic drugs with fewer side effects. This novel strategy may ameliorate the present obstacles to the clinical treatment of lung cancer.

Keywords: CAR-T; Exosomes; Inhalation; Lung cancer; Paclitaxel; Targeted delivery.

PubMed Disclaimer

Conflict of interest statement

The authors declare that they have no known financial and personal relationships with other people or organizations that can inappropriately influence our work, there is no professional or other personal interest of any nature or kind in any product, service and/or company that could be construed as influencing the position presented in, or the review of, the manuscript entitled.

Figures

Scheme 1.
Scheme 1.
Schematic illustration of inhalable CAR exosomes derived from CAR-T cells as PTX carriers for treating lung cancer. A Preparation process of PTX@CAR-Exo. B Antitumor effect of inhalable PTX@CAR-Exo against lung cancer. CAR-T cell-derived exosomes (CAR-Exos) inherited the targeted cytolytic effects from parental cells by releasing granzyme B and perforins. The loaded PTX was delivered into the tumor area to stimulate immune responses. In contrast to conventional CAR-T cell therapy and intravenous administration of PTX, the inhalable PTX@CAR-Exos used in an orthotopic lung cancer model exhibited superior antitumor effects and improved the bioavailability of both CAR-Exos and PTX, with decreased systemic toxicity
Fig. 1
Fig. 1
Preparation and characterization of CAR-T cells. A Molecular design of anti-MSLN mCAR. B The transfection efficiency of lentiviral in T cells was assessed by GFP expression using fluorescence microscopy. Scale bar: 20 μm. C Anti-MSLN CAR expression was assessed by Protein L staining and flow cytometry. Representative Protein L staining results are shown for mock transduced T cells and MSLN CAR-T cells. D Fluorescence images of LLC cells stably expressing GFP-MSLN after puromycin selection. Scale bar: 20 μm. E Representative flow cytometry histogram of the expression level of MSLN on LLC cells. An anti-MSLN antibody was used to confirm MSLN expression. F Cytolysis of CAR-T cells against MSLN-LLC cells. The cytotoxic activity of CAR-T and control T cells against cancer cell lines was assessed by an LDH-release assay at the indicated effector-to-target (E:T) ratios. G The level of IFN-γ evaluated by ELISA after co-culture of CAR-T cells with MSLN-LLC cells for 24 h. Data are presented as the mean ± SD of three biological replicates. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, ns: not significant
Fig. 2
Fig. 2
Preparation and characterization of CAR-Exos. A Representative TEM image of T-Exo and CAR-Exo. Scale bar: 100 nm. B Size distribution and zeta potential of T-Exos and CAR-Exos measured by NTA and DLS. Data are presented as the mean ± SD of three biological replicates. C The expression of CAR protein and exosomal markers in CAR-Exos or whole-cell lysates of CAR-T cells were analyzed by Western blotting. D Flow cytometric analyses of CAR-Exos conjugated to latex beads or CAR-T cells. The histograms shown isotype controls (black) and positive expression (red). E Representative Confocal laser scanning microscopy (CLSM) images of CAR-Exos bound to the surface of MSLN-LLC cells (green). T-Exos and CAR-Exos were labeled with DiI (red) and DAPI (blue) labeled nuclei of MSLN-LLC cells. CAR-Exos showed a higher binding affinity to the MSLN protein than T-Exos. Scale bar: 10 μm
Fig. 3
Fig. 3
The enhanced antitumor effect of PTX@CAR-Exos in vitro. A Flow cytometric analyses of granzyme B and perforin expression of CAR exosomes conjugated to latex beads or CAR-T cells. The histograms shown isotype controls (black) and positive expression (red). B The expression of granzyme B and perforin in CAR-Exos or whole-cell lysates of CAR-T cells were analyzed by western blotting. C The cytotoxic activity of different concentrations of CAR exosomes against MSLN-LLC cells. D In vitro drug release profile of paclitaxel-loaded CAR exosomes was evaluated in PBS at pH 7.4 and different temperatures. The percentage of drug release (%) = OD value of the PTX released from the CAR exosomes /OD value of the total PTX in the CAR Exosomes × 100%. E Cytotoxicity and Calcein-AM/PI staining analysis in vivo. Fluorescent microscopic imaging of MSLN-LLC cells after treatment with PBS, T-Exos, CAR-Exos, PTX, PTX@T-Exos, or PTX@CAR-Exos for 24 h, cells were stained with Calcein-AM/PI (original magnification, 10 ×). Scale bar: 100 μm. F Quantitative analysis of cell death rate analyzed by Calcein-AM/PI staining. Data are presented as the mean ± SD of three biological replicates. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, ns: not significant
Fig. 4
Fig. 4
The biodistribution of DiR-labeled CAR-Exos via inhalation in vivo. A The biodistribution of CAR-Exos in vivo via intravenous injection or inhalation. B The quantification of DiR intensity from A. C The intensity of inhaled CAR-Exos at different time points in vivo. D The quantification of DiR intensity at different time points. E The intensity of inhaled CAR-Exos at different concentrations of exosomes in vivo. F The quantification of DiR intensity at different doses. Data are presented as the mean ± SD of three biological replicates
Fig. 5
Fig. 5
The therapeutic efficacy of PTX@CAR-Exos in orthotopic lung tumor-bearing mice. A The designed diagram for evaluating the efficacy of PBS, T-Exos, CAR-Exos, or PTX@CAR-Exos treated orthotopic lung tumor-bearing mice. Mice were given inhaled therapy via nebulizer for 2 weeks, starting on day 7 after establishment of the orthotopic model; IVIS imaging was performed on days 14 and 21 post implantation. B Respective bioluminescence images of MSLN-LLC cells in mice on days 7, 14, and 21 post implantation. C Statistical analysis of the signal intensity from B. D Survival analysis of tumor bearing mice after different treatments. E Representative images of tumor nodules in different groups and H&E-stained slice images of the lungs. Scale bar: 100 μm. Data are presented as the mean ± SD of three biological replicates. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, ns: not significant
Fig. 6
Fig. 6
Improvement in the tumor microenvironment following PTX@CAR-Exos treatment in vivo. A, C Representative flow cytometry plots and statistical analysis of CD4+ T cells and CD8+ T cells infiltration at the tumor site. B, D Representative flow cytometry plots and statistical analysis of infiltrated CD8+GzmB+ T cells at the tumor site. Cytokine concentrations E TNF-α and F IFN-γ in tumor tissues as quantified by ELISA assay. Data are presented the mean ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, ns: not significant

Similar articles

Cited by

References

    1. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72:7–33. doi: 10.3322/caac.21708. - DOI - PubMed
    1. Herbst RS, Morgensztern D, Boshoff C. The biology and management of non-small cell lung cancer. Nature. 2018;553:446–454. doi: 10.1038/nature25183. - DOI - PubMed
    1. Planchard D, Popat S, Kerr K, Novello S, Smit EF, Faivre-Finn C, Mok TS, Reck M, Van Schil PE, Hellmann MD, et al. Metastatic non-small cell lung cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2018;29:iv192–iv237. doi: 10.1093/annonc/mdy275. - DOI - PubMed
    1. Weiss RB, Donehower RC, Wiernik PH, Ohnuma T, Gralla RJ, Trump DL, Baker JR, Jr, Van Echo DA, Von Hoff DD, Leyland-Jones B. Hypersensitivity reactions from taxol. J Clin Oncol. 1990;8:1263–1268. doi: 10.1200/JCO.1990.8.7.1263. - DOI - PubMed
    1. Schiff PB, Fant J, Horwitz SB. Promotion of microtubule assembly in vitro by taxol. Nature. 1979;277:665–667. doi: 10.1038/277665a0. - DOI - PubMed

Publication types