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 Apr 12;21(1):255.
doi: 10.1186/s12967-023-04080-z.

FAP-targeted CAR-T suppresses MDSCs recruitment to improve the antitumor efficacy of claudin18.2-targeted CAR-T against pancreatic cancer

Affiliations

FAP-targeted CAR-T suppresses MDSCs recruitment to improve the antitumor efficacy of claudin18.2-targeted CAR-T against pancreatic cancer

Yifan Liu et al. J Transl Med. .

Abstract

Purpose: The claudin 18.2 (CLDN18.2) antigen is frequently expressed in malignant tumors, including pancreatic ductal adenocarcinoma (PDAC). Although CLDN18.2-targeted CAR-T cells demonstrated some therapeutic efficacy in PDAC patients, further improvement is needed. One of the major obstacles might be the abundant cancer-associated fibroblasts (CAFs) in the PDAC tumor microenvironment (TME). Targeting fibroblast activation protein (FAP), a vital characteristic of CAFs provides a potential way to overcome this obstacle. In this study, we explored the combined antitumor activity of FAP-targeted and CLDN18.2-targeted CAR-T cells against PDAC.

Methods: Novel FAP-targeted CAR-T cells were developed. Sequential treatment of FAP-targeted and CLDN18.2-targeted CAR-T cells as well as the corresponding mechanism were explored in immunocompetent mouse models of PDAC.

Results: The results indicated that the priorly FAP-targeted CAR-T cells infusion could significantly eliminate CAFs and enhance the anti-PDAC efficacy of subsequently CLDN18.2-targeted CAR-T cells in vivo. Interestingly, we observed that FAP-targeted CAR-T cells could suppress the recruitment of myeloid-derived suppressor cells (MDSCs) and promote the survival of CD8+ T cells and CAR-T cells in tumor tissue.

Conclusion: In summary, our finding demonstrated that FAP-targeted CAR-T cells could increase the antitumor activities of sequential CAR-T therapy via remodeling TME, at least partially through inhibiting MDSCs recruitment. Sequential infusion of FAP-targeted and CLDN18.2-targeted CAR-T cells might be a feasible approach to enhance the clinical outcome of PDAC.

PubMed Disclaimer

Conflict of interest statement

Dr. Hua Jiang and Dr. Zonghai Li has ownership interests of CAR-T cells relating to this work and is a stockholder of CARsgen Therapeutics, Inc. The other authors declare that they have no conflicts of interest.

Figures

Fig. 1
Fig. 1
Bioinformatics analysis of FAP biological and tumor microenvironment characteristics in pancreatic cancer. a Pan-cancer analysis of FAP expression. b Identification of optimal FAP expression cutoff dividing PDAC cohort. The upper scatter plot shows each cutoff point’s standardized log-rank statistic value. The lower Kaplan-Meier plot shows overall survival for patients divided by the optimal cutoff. cf Biology and TME characteristics in high-FAP vs. low-FAP groups of PDAC cohort. c Density distribution of tumor purity. d Enrichment score of HALLMARK gene-sets related to high FAP expression. e Volcano map of the chemokines. f Immune suppressive characteristics estimated by IOBR. g IHC staining of FAP protein expression in 9 pancreatic cancer samples
Fig. 2
Fig. 2
CAFs impair the antitumor activity of CAR-T cells in vivo. a IHC staining of CAFs in 4T1 and E0771 breast and PANC02 and KPC1199 pancreatic tumor-bearing mice. b-f In vivo experimental of Antigen-Positive E0771 allografts. b Experimental scheme. Antigen-Positive E0771 tumor cells alone or combined with NIH 3T3 fibroblast were in situ inoculated into C57BL/6 mice, treated i.p. injection with CPA, and then given CAR-T cells (i.v.; n = 5 mice per group). c The tumor volume of tumors of each treatment group. d The tumor growth inhibition of each treatment group. e The tumor weight at the end point of the animal experiment. f CAR copy numbers in genomic DNA of residual tumors after therapy were measured by qRT-PCR (TaqMan probe). The images were obtained under original magnification 200×. Scale bars, 100 μm. All data are presented as the mean ± SEM of triplicate experiments. *p < 0.05, **p < 0.01, ***p < 0.001
Fig. 3
Fig. 3
Generation of FAP-targeted CAR-T cells and CLDN18.2-targeted CAR-T cells. a Binding specificity of the anti-FAP antibody to mFAP-transfected 3T3 or huFAP-transfected HT-1080. b Affinity measurements of anti-FAP antibody binding to FAP through BiacoreSurface Plasmon resonance (GE Healthcare). The upper shows its binding to mu-FAP and, the lower shows its binding to hu-FAP. c The construction of 8E3-mBBZ and 8E5-mBBZ CAR-T cells. This construct includes an extracellular antigen recognition region (8E5 targeting CLDN18.2 and 8E3 targeting FAP), a hinge, a transmembrane domain, an intracellular region of mouse 41-BB costimulatory molecules, and a mouse CD3-ζ chain. d The transduction efficiency of FAP-mBBZ and CLDN18.2-BBZ CAR-T CAR on splenic T cells derived from C57BL/6 was determined by flow cytometry. UTD cells served as negative controls. e Western blot of CD3-ζ in CAR-T. Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) served as a loading control. f The expression of CLDN18.2 on KPC1199, PANC02, and PANC02-A2. Cells incubated with a mouse anti-mouse IgG antibody as a negative control. g The establishment of mFAP NIH 3T3 mouse fibroblast cells. NIH 3T3 was transduced by pWPT-GFP-mFAP lentivirus and further sorted by flow cytometry. h-i CAR-T cells were incubated with tumor or fibroblast cells at three effector: target (E: T) ratios for 18 h. Cell lysis was tested using a standard nonradioactive cytotoxicity assay. h Cytotoxic activities of CLDN18.2-mBBZ CAR-T cells on CLDN18.2-positive or CLDN18.2-negative tumor cells. i Cytotoxic activities of FAP-mBBZ CAR-T cells on FAP-positive or FAP-negative fibroblast cells
Fig. 4
Fig. 4
Sequential therapy of FAP-targeted and CLDN18.2-targeted cells dramatically improved antitumor efficacy against CLDN18.2-positive tumor-bearing mice. a-d In vivo experimental design. C57BL/6 mice were injected s.c. with PANC02-A2 cells and allowed to establish for 10 days. Mice were assigned to four experimental groups. The first and second CAR-T cells were injected on day 10 and day 18, respectively (i.v.; n = 5 mice per group). b The tumor growth inhibition of each treatment group. c The volume of tumors of each treatment group. d The body weight of mice of each treatment group. e-h In vivo experimental design. C57BL/6 mice were injected s.c. with KPC1199 cells and allowed to establish for 12 days. Mice were assigned to four experimental groups. The first and second CAR-T cells were injected on day 12 and day 19, respectively (i.v.; n = 5 mice per group). f The tumor growth inhibition of each treatment group. g The volume of tumors of each treatment group. h The body weight of mice of each treatment group. All data are presented as the mean ± SEM of triplicate experiments. *p < 0.05, **p < 0.01, ***p < 0.001
Fig. 5
Fig. 5
Sequential therapy of FAP-targeted and CLDN18.2-targeted CAR-T cells dramatically increase the accumulation of CD8+ T and CAR-T cells. a–c Tumor tissues were harvested from the mice at the end of therapy in the PANC02 model. a The representative images of tumor-infiltrating T cells immunostained with anti-CD4 and anti-CD8 from each treatment group. b Histograms show the quantification of T cells in tumor tissues. c CAR copy numbers in genomic DNA of residual tumors from each treatment group were measured by qRT-PCR (TaqMan probe). df Tumor tissues were harvested from the mice at the end of therapy in the KPC1199 model. d The representative images of tumor-infiltrating T cells immunostained with anti-CD4 and anti-CD8 from each treatment group. e Histograms show the quantification of T cells in tumor tissues. f CAR copy numbers in genomic DNA of residual tumors from each treatment group were measured by qRT-PCR (TaqMan probe). The images were obtained under original magnification 200×. Scale bars, 100 μm. All data are presented as the mean ± SEM of triplicate experiments. *p < 0.05, **p < 0.01
Fig. 6
Fig. 6
FAP-targeted CAR-T cells eliminate CAFs and change tumor microenvironment in vivo. C57BL/6 mice were injected s.c. with PANC02-A2 or KPC1199 cells and treated with CAR-T cells after the tumor was established. To exclude the influence of tumor volume difference by distinct CAR-T cells, tumor tissue was harvested 7 days after treatment. ac Analysis of the CAFs in tumor tissues of PANC02-A2 and KPC1199 allografts. a The representative images of CAFs immunostained with α-SMA in tumor tissues from each treatment group. b q-PCR measured the mRNA expression of CXCL12 in tumor tissues from each treatment group. c PET image labeling FAP by FAPI in PANC02-A2 allografts 7 days after the treatment of FAP-mBBZ or UTD T cells.  d–f  Analysis of the immune cell in tumor tissues of PANC02-A2 allografts. d The representative flow cytometry plots showing the frequencies of tumor-infiltrating CD8+ and CD4+ cells in CD3+ T of each treatment group. e The representative flow cytometry plots showing the frequencies of tumor-infiltrating CD45+ immune cells and MDSCs of each treatment group. f Flow cytometry quantitation of tumor-infiltrating immune cells of each treatment group. gi Analysis of the immune cell in tumor tissues of KPC1199 allografts. g The representative flow cytometry plots showing the frequencies of tumor-infiltrating CD8+ and CD4+ cells in CD3+ T of each treatment group. h The representative flow cytometry plots showing the frequencies of tumor-infiltrating CD45+ immune cells and MDSCs of each treatment group. i Flow cytometry quantitation of tumor-infiltrating immune cells of each treatment group. The images were obtained under original magnification 200×. Scale bars, 100 μm. All data are presented as the mean ± SEM of triplicate experiments. *p < 0.05, **p < 0.01, *** p < 0.001
Fig. 7
Fig. 7
Schema: Sequential Therapy enhances the antitumor ability of CAR-T by suppressing the infiltration of MDSCs. Left: CAFs are crucial in TME, forming an immune surveillance barrier and perpetuating tumor-promoting. The CAFs could also promote recruit monocytes from the bone marrow, such as MDSCs, to form a tumor-suppressing microenvironment, suppressing CAR-T cell function. Right: FAP-targeted CAR-T cells eliminate CAFs via specific recognition of FAP on CAFs in the tumor microenvironment. Further, CLDN18.2-targeted CAR-T cells could also increase cytotoxic T cells and inhibit the recruiting of MDSCs. With an improved immune suppressive microenvironment, the antitumor effect of the sequential infusion of CLDN18.2-targeted CAR-T cells were enhanced

Similar articles

Cited by

References

    1. Mizrahi JD, Surana R, Valle JW, Shroff RT. Pancreatic cancer. Lancet. 2020;395(10242):2008–20. doi: 10.1016/S0140-6736(20)30974-0. - DOI - PubMed
    1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49. doi: 10.3322/caac.21660. - DOI - PubMed
    1. Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Miklos DB, Jacobson CA, et al. Axicabtagene ciloleucel CAR T-Cell therapy in refractory large B-Cell lymphoma. N Engl J Med. 2017;377(26):2531–44. doi: 10.1056/NEJMoa1707447. - DOI - PMC - PubMed
    1. Jiang H, Shi Z, Wang P, Wang C, Yang L, Du G, et al. Claudin18.2-Specific chimeric Antigen receptor Engineered T cells for the treatment of gastric Cancer. J Natl Cancer Inst. 2019;111(4):409–18. doi: 10.1093/jnci/djy134. - DOI - PubMed
    1. Qi C, Gong J, Li J, Liu D, Qin Y, Ge S, et al. Claudin18.2-specific CAR T cells in gastrointestinal cancers: phase 1 trial interim results. Nat Med. 2022;28(6):1189–98. doi: 10.1038/s41591-022-01800-8. - DOI - PMC - PubMed

Publication types

Substances