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 Dec;9(48):eadg9721.
doi: 10.1126/sciadv.adg9721. Epub 2023 Dec 1.

EpCAM-targeting CAR-T cell immunotherapy is safe and efficacious for epithelial tumors

Affiliations

EpCAM-targeting CAR-T cell immunotherapy is safe and efficacious for epithelial tumors

Dan Li et al. Sci Adv. 2023 Dec.

Abstract

The efficacy of CAR-T cells for solid tumors is unsatisfactory. EpCAM is a biomarker of epithelial tumors, but the clinical feasibility of CAR-T therapy targeting EpCAM is lacking. Here, we report pre- and clinical investigations of EpCAM-CAR-T cells for solid tumors. We demonstrated that EpCAM-CAR-T cells costimulated by Dectin-1 exhibited robust antitumor activity without adverse effects in xenograft mouse models and EpCAM-humanized mice. Notably, in clinical trials for epithelial tumors (NCT02915445), 6 (50%) of the 12 enrolled patients experienced self-remitted grade 1/2 toxicities, 1 patient (8.3%) experienced reversible grade 3 leukopenia, and no higher-grade toxicity reported. Efficacy analysis determined two patients as partial response. Three patients showed >23 months of progression-free survival, among whom one patient experienced 2-year progress-free survival with detectable CAR-T cells 200 days after infusion. These data demonstrate the feasibility and tolerability of EpCAM-CAR-T therapy.

PubMed Disclaimer

Figures

Fig. 1.
Fig. 1.. The Dectin-1–stimulated EpCAM-specific CAR has a distinct effect on T cell in self-activation, differentiation, and exhaustion compared with that of CARs with the CD28 or 4-1BB costimulatory domain.
(A) Schematic of EpCAM-specific CARs carrying CD28, 4-1BB, or Dectin-1 costimulatory domain. (B) Expression of different EpCAM-specific CARs on T cells, respectively. (C and D) Expression of activation-related markers on CAR-T cells after 9 days of CAR transduction. n = 5 different donors. Cells were pregated for the CD3+CAR+ subset. (E) Expression of exhaustion-associated receptors on CAR-T cells after 9 days of lentivirus transduction. The color lines are represented as mean. (F) Fold change of EpCAM–CAR-T cells differentiation in central memory and effector phenotypes of five different donors. The data are displayed as mean. (G) Schematic of the xenograft model. B-NDG mice were intraperitoneally injected with 2 × 105 HT-29–Luc cells on day −7. After that, the mice were intraperitoneally infused with three doses of Mock-T or CAR-T cells. n = 6 mice for each group. After 21 days of tumor inoculation, mice were euthanized to analyze CAR-T cell persistence. (H and I) Bioluminescence images and statistical results of tumor burden. (J and K) The percent of splenetic Mock-T cells and CAR-T cells after 21 days of adoptive T cell therapy. (L) Differentiation of splenetic CD4+ and CD8+ CAR-T cells in central memory and effector phenotypes in the xenograft model. (M and N) Exhaustion-associated receptors expression on splenetic CAR-T cells after 21 days of adoptively T cells administration. The data are presented as mean ± SD. *P < 0.05 and **P < 0.01. P < 0.05 was considered statistically significant. ns, no significance. Wilcoxon test was used for (D) to (F). Two-tailed unpaired Student’s t test was used for differences analysis in (K), (L), and (N). TM, transmembrane domain; Teff, effector T cell; Tcm, central memory T cells.
Fig. 2.
Fig. 2.. Intraperitoneal and intravenous infusion of EpCAM–CAR-T cells induced colon cancer remission in a xenograft mouse model.
(A) Schematic of the animal experiments. B-NDG mice were intraperitoneally injected with 2 × 105 Luc-expressing HT-29–Luc cells on day −7. After that, the mice were randomly assigned to different groups and were intraperitoneally or intravenously infused with three doses of Mock-T or CAR-T cells. n = 5 mice for each group. (B and C) Bioluminescence images and statistical result of tumor burden after intravenous administration of CAR-T cells. (D and E) Tumor weight at the end point of the experiment and the survival of mice who received intravenous CAR-T infusion. (F and G) Bioluminescence images and statistical result of tumor burden after intraperitoneal administration of CAR-T cells. (H and I) Tumor weight at the end point of the experiment and the survival of mice who received intraperitoneal CAR-T infusion. (J to M) Flow cytometry analysis of EpCAM–CAR-T cell persistence in the spleens of mice with intravenous (J) and (K) or intraperitoneal (L) and (M) T cell infusion. The Mock-T cell group was examined on day 21, and the CAR-T cell groups were examined when euthanized. n = 4 mice in the 1 × 107 CAR-T cells intravenously treated group and n = 5 mice in the other group. (N and O) EpCAM–CAR-T cells persisted in the peripheral blood of mice that received intravenous infusion of T cells. (P and Q) Representative results showing tumor-infiltrated human CD3+ T cells in mice that received intravenous injection of T cells, as determined by immunohistochemistry (P) and statistical results shown in (Q). Scale bars, 200 μm (white) and 50 μm (black). The data are expressed as the means ± SD. Log-rank test for (E) and (I) and Mann-Whitney U test for (C), (D), (G), (H), (K), (M), and (O). *P < 0.05, **P < 0.01, and ****P < 0.0001. P < 0.05 was considered statistically significant.
Fig. 3.
Fig. 3.. EpCAM–CAR-T cells were well tolerated by EpCAM-humanized mice.
(A) Genetic engineering strategy for EpCAM-humanized C57BL/6 mice. The murine EpCAM gene locus was truncated, and a homologous human EpCAM gene locus was inserted from exon 2 to exon 7. Purple, exon of mouse EpCAM; green, exon of human EpCAM; E, exon. (B) Experimental schematic showing the safety evaluation in the C57BL/6-EpCAMtml EpCAM mouse model. The mice were randomly grouped; n = 3 per group. All mice were preconditioned with cyclophosphamide (100 mg/kg). Two days later, a single dose of Mock-T cells (1 × 106) or EpCAM–CAR-T cells (1 × 106, 5 × 106, and 1 × 107) was intravenously administered. (C) Percentage of body weight change was normalized to that on day 0 after T cell infusion. (D) Serum cytokine levels in peripheral blood at the indicated points after T cell infusion (means ± SD). (E and F) Representative immunohistochemical (E) and quantitative real-time PCR (F) analysis of mouse CD3+ cells (E) and EpCAM-CAR gene (F) distribution in the spleen, lung, and other organs 7 days after T cell transfer. Scale bars, 50 μm. (G to J) Organ damage-related serum biomarkers were measured in the liver (G), kidney (H), heart (I), and pancreas (J) on day 7 after T cell transfer. The data are presented as the means ± SD, and data analysis was performed by one-way analysis of variance (ANOVA). P < 0.05 was considered statistically significant. AST, aspartate aminotransferase; ALT, alanine aminotransferase; CRE, creatinine; BUN, blood urea nitrogen; LDH, lactate dehydrogenase; CK-MB, creatine kinase-MB; AMY, amylase.
Fig. 4.
Fig. 4.. EpCAM–CAR-T cell clinical protocol design and cohort diagram.
(A) Clinical protocol schematic for patient screening, CAR-T cell manufacture, and time points of the clinical study. Cy, cyclophosphamide; Flu, fludarabine; CT c/a/p, computed tomography of chest/abdomen/pelvis. (B) Number of patients screened, enrolled, and treated with autologous EpCAM–CAR-T cells in the clinical study. (C) Waterfall plot summarizing the adverse effects of CAR-T therapy in the clinical study. (D) Swimming plot showing patients’ disease development and substantial therapy and present status. IMRT, intensity-modulated radiotherapy.
Fig. 5.
Fig. 5.. EpCAM–CAR-T cell persistence, cytokine secretion and radiologic evaluation of patients after EpCAM–CAR-T cell infusion.
(A) EpCAM-CAR gene copies in genomic DNA of peripheral blood from patients at the indicated times following CAR-T cell transfer. (B to D) Representative serum IL-6 (B), TNF-α (C), and CRP (D) levels in patients treated with EpCAM–CAR-T cells. Cohort 1: violet; cohort 2: blue; cohort 3: orange. (E) Computed tomography scans showing the tumor response in patient 10 and patient 3 after EpCAM–CAR-T cell transfer. Red arrows indicate the tumor site.

References

    1. S. J. Schuster, M. R. Bishop, C. S. Tam, E. K. Waller, P. Borchmann, J. P. McGuirk, U. Jäger, S. Jaglowski, C. Andreadis, J. R. Westin, I. Fleury, V. Bachanova, S. R. Foley, P. J. Ho, S. Mielke, J. M. Magenau, H. Holte, S. Pantano, L. B. Pacaud, R. Awasthi, J. Chu, Ö. Anak, G. Salles, R. T. Maziarz; JULIET Investigators , Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N. Engl. J. Med. 380, 45–56 (2019). - PubMed
    1. E. A. Chong, M. Ruella, S. J. Schuster; Lymphoma Program Investigators at the University of Pennsylvania , Five-year outcomes for refractory B-cell lymphomas with CAR T-cell therapy. N. Engl. J. Med. 384, 673–674 (2021). - PubMed
    1. D. L. Porter, B. L. Levine, M. Kalos, A. Bagg, C. H. June, Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N. Engl. J. Med. 365, 725–733 (2011). - PMC - PubMed
    1. S. Rafiq, C. S. Hackett, R. J. Brentjens, Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat. Rev. Clin. Oncol. 17, 147–167 (2020). - PMC - PubMed
    1. R. A. Morgan, J. C. Yang, M. Kitano, M. E. Dudley, C. M. Laurencot, S. A. Rosenberg, Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol. Ther. 18, 843–851 (2010). - PMC - PubMed

Substances