GPC3-7-19-CAR-T cells exert superior cytotoxic effects on GPC3-positive HCC cells in vitro
To profile the characteristics of fourth-generation GPC3-CAR-T cells expressing the costimulatory molecules IL7 and CCL19 (GPC3-7-19-CAR-T cells), we constructed three other types of CARs in parallel in this study. Among them, Vector-CAR-T cells served as a negative control, while GPC3-CAR-T cells were used as conventional CAR-T cells specifically targeting GPC3, and 7-19-CAR-T cells expressed with IL7 and CCL19 but lacked GPC3 targeting (Fig. 1A). The GPC3-CAR-T cells were made to express an enhanced green fluorescence protein (EGFP) to enable evaluation of viral transfection efficiency. Most activated T-cell clusters expressed EGFP after 5 days of culture, as observed under a fluorescence microscope (Supplementary Fig. 1A, B). Approximately 48%-62% of CAR-positive T cells in each group were identified as positive by flow cytometry (Fig. 1A, B). To optimize the expansion efficiency of CAR-T cells, 10 ng/mL rhIL-15 was added to culture media with 10 ng/mL rhIL-2 beginning on the second day after T-cell stimulation. CAR-T cells proliferated rapidly with approximately 200-fold expansion within 12 days, which was much higher than that of the conventional T cells that were only given IL-2 (Supplementary Fig. 1C, D).
Moreover, GPC3-7-19-CAR-T cells secreted the highest levels of IL-7 and CCL-19 when cocultured with GPC3-positive HCC cell lines. Approximately 480 pg/mL IL-7 and 17000 pg/mL CCL-19 were detected when GPC3-7-19-CAR-T cells were cocultured with HepG2 cells. 7-19-CAR-T cells showed moderate levels of IL-7 (300 pg/mL) and CCL-19 (1500 pg/mL) secretion, whereas GPC3-CAR-T cells and vector-CAR-T cells secreted minimal amounts (Fig. 1D, E).
To assess the specific antitumor effects of GPC3-7-19-CAR-T cells against GPC3-positive HCC cells, we first evaluated GPC3 expression levels in several HCC cell lines by flow cytometry. GPC3 was primarily expressed in HepG2 and Hep3B cells (97.5% and 90.1% of cells with positive expression, respectively) and expressed at low levels in SMMC7721 cells (5.9% of cells with positive expression, Supplementary Fig. 1E, F).
Next, CAR-T cells from each group were incubated with HCC cells at an effector-to-target (E/T) ratio of 1:1. HepG2 cell growth was monitored continuously for 72 hours by measuring impedance using the RTCA SP xCELLigence system (Roche)[13]. GPC3-7-19-CAR-T cells showed 20% (35% vs. 15%) higher cytotoxicity toward HepG2 cells than GPC3-CAR-T cells and other T cells (Fig. 2B, C).
To further verify the results, a nonradioactive LDH assay was employed. GPC3-7-19-CAR-T cells exhibited at least a 1.5-fold increased killing rate of HepG2 and Hep3B cells compared with GPC3-CAR-T cells at an E:T ratio of 1:1 or 2:1 (Fig. 1H, I), but there was no increased killing of SMMC7721 cells (Fig. 1J). As seen on optical microscopy, HepG2 cells were clearly surrounded by GPC3-7-19-CAR-T cells in clusters after a 6-hour coculture, suggesting that activated GPC3-7-19-CAR-T cells exerted strong and specific killing effects against GPC3-positive HCC cells (Fig. 1K).
Gpc3-7-19-car-t Cells Exhibit Excellent Activity Against Hcc Xenografts In Humanized Mouse Models
To better examine the in vivo safety and efficacy of GPC3-7-19-CAR-T cells, a subcutaneous xenograft model of HepG2-EGFP-Luci cells was established in humanized NSG mice, in which the microenvironment was similar to that of patients who have received CAR-T-cell infusion into peripheral blood[14]. Each mouse received two doses of CAR-T cells at day 0 and day 4 via tail vein injection. The tumors and vital organs were collected at the endpoint to analyze cytokine secretion and tumor-infiltrating lymphocytes, and immunohistochemistry (IHC) staining was performed. A detailed schedule is shown in Fig. 2A.
To confirm the successful establishment of the humanized mouse model, baseline lymphocytes in the peripheral blood of all mice were tested by flow cytometry. The human CD45 population represented approximately 70% of all cells on average in each humanized mouse. Statistically, human pan-T cells (CD3+) represented approximately 19% of all cells, of which the percentages of human Th cells (CD3+CD4+), Ts cells (CD3+CD8+), natural killer (NK) cells (CD3−CD56+), dendritic cells (CD11c+) and monocytes (CD11b+) were approximately 30%, 54%, 3.6%, 3.0% and 7.9%, respectively (Fig. 2B-E).
Then, tumor growth was monitored and recorded every other day. On day 76, by intravital fluorescence imaging, the tumor volume of GPC3-7-19-CAR-T-cell-treated mice was significantly smaller than that of GPC3-CAR-T-cell-treated mice. Of note, 2 xenograft tumors in GPC3-7-19-CAR-T-cell-treated mice disappeared. In contrast, the largest subcutaneous tumor in GPC3-CAR-T-cell-treated mice was about 2 cm3, and the tumorigenesis rate was 60% in GPC3-7-19-CAR-T-cell-treated mice compared with 100% in GPC3-CAR-T-cell-treated mice (n = 5, Fig. 2F, G, H). Furthermore, the general condition and body weight of GPC3-7-19-CAR-T-cell-treated mice was better and higher than those of GPC3-CAR-T-cell-treated mice, indicating the safety of GPC3-IL7-CCL19-CAR-T-cell infusion (Fig. 2I). In summary, GPC3-7-19-CAR-T cells exhibited excellent antitumor effects against GPC3-positive HCC cells compared with conventional GPC3-CAR-T cells.
The CD4 + T-cell subset plays a dominant role in the enhanced antitumor activity of GPC3-7-19-CAR-T cells
To monitor the in vivo expansion of CAR-T cells in humanized mice, peripheral blood samples of mice were collected at days 0, 1, 4, 7, 14, 28, 56, and 73 to determine CAR copy numbers by q-PCR. As shown, the CAR copies of both GPC3-CAR-T and GPC3-7-19-CAR-T cells peaked around day 14. Then, there was a rapid decrease in CAR copies of GPC3-CAR-T cells and a sustained higher level of GPC3-7-19-CAR-T cells until day 56 (Fig. 3A). On day 28, higher levels of human IFN-γ and IL-2 but not TNF-α were observed in GPC3-7-19-CAR-T-cell-treated mice than in traditional GPC3-CAR-T-cell treated mice (Fig. 3B).
To explore the underlying mechanism of GPC3-7-19-CAR-T-cell-mediated antitumor effects in humanized NSG mice, human cell populations were analyzed in mouse peripheral blood, liver and tumor tissues by flow cytometry on day 76. A significant increase in human CD4+ T cells was detected in mouse liver and tumor tissues in GPC3-7-19-CAR-T-cell treated mice compared to GPC3-CAR-T-cell treated mice. Furthermore, two populations of human CD4+ and CD8+ T cells with central (TCM cells, CD45RA−CCR7+) and effector (TEM cells, CD45RA−CCR7−) memory phenotypes were further evaluated. The total CD4+ T cell and CD4+ TEM cell subset levels in mouse liver and tumor tissues in GPC3-IL7-CCL19-CAR-T-cell treated mice were dramatically higher than those in GPC3-CAR-T-cell treated mice. Notably, the levels of both the CD8+ TCM and CD4+ TEM cell subsets, but not the CD8+ TEM cell subset, in the tumor tissues of GPC3-IL7-CCL19-CAR-T-cell-treated mice were significantly elevated compared with those in GPC3-CAR-T-cell treated mice (Fig. 3C-F). Then, CD4+ T and CD8+ T cells in tumors were assessed once more by IHC and immunofluorescence (IF) staining, and more CD4+ T cells were found in tumor tissue in GPC3-7-19-CAR-T-cell-treated mice (Fig. 3G-J). All of the above results indicated that CD4+ T cells, especially CD4+ TEM cell subsets, play a key role in the enhanced antitumor activity of GPC3-7-19-CAR-T cells in vivo. In addition, the increased level of CD8+ TCM cells might also lead to a better tumor-suppressive effect of GPC3-7-19-CAR-T cells.
Gpc3-7-19-car-t Cells Remodel The Tumor Immune Microenvironment
Next, the tumor immune microenvironment (TIME) was evaluated after GPC3-7-19-CAR-T-cell infusion. First, bone marrow myeloid-derived suppressor cells (MDSCs) were assessed in xenograft tumors and mouse organs based on their surface markers outlined in Fig. 4A. Statistically, the percentages of MDSCs in all myeloid cells (CD45+CD11b+ HLA-DR−) in GPC3-IL7-CCL19-CAR-T-cell-treated mice were at least 2-fold lower than those in GPC3-CAR-T-cell-treated mice, especially in tumor and liver tissue (Fig. 4B).
In pedigree analysis, MDSCs can be classified into PMN-MDSCs (CD11b+ HLA-DR−, CD15+) and monocyte-like MDSCs (M-MDSCs, CD11b+ HLA-DR−, CD14+, Arg1+, Fig. 4A). We identified MDSC subsets in mice treated with CAR-T-cell therapy, and a 40% lower proportion of M-MDSCs was found in GPC3-7-19-CAR-T-cell treated mice, especially in the xenograft tumors and in most organs except the bone marrow. (Fig. 4C).
In the TME, Treg cells might also play an inhibitory role in adoptive CAR-T-cell therapy. Treg cells can be rapidly induced or recruited by TGF-β and IL-10 derived from MDSCs. In line with this, the levels of Treg cells in the peripheral blood, liver and tumor tissue of GPC3-7-19-CAR-T-cell-treated mice were significantly less than those of GPC3-CAR-T-cell-treated mice (Fig. 4D, E). All of the above data demonstrated that GPC3-7-19-CAR-T cells reversed the inhibitory TME by decreasing MDSC and Treg cell infiltration in tumors, thus achieving better therapeutic effects in HCC xenografts models.
Dcs Are Recruited To Assist The Killing Activities Of Gpc3-7-19-car-t Cells
CCL19 is thought to be a key chemokine that recruits CCR7+ immunocompetent cells, such as DCs, T and B cells, and macrophages, to tumor tissue. A remarkable increase in human DCs was found in tumor tissues in GPC3-7-19-CAR-T-cell-treated mice (Fig. 5F, G). To confirm the results, mature HLA-DR+CD11b+CD86+ DCs (Fig. 5A, B) were cocultured with either GPC3-7-19-CAR-T cells or GPC3-CAR-T cells in a Transwell system. The migrated DCs in the lower compartment were then counted (Fig. 5C). Statistically, the numbers of migrated DCs were 10% higher in the GPC3-7-19-CAR-T-cell coculture (Fig. 6D, E). IHC staining of DCs in humanized mouse liver and tumor tissue showed significantly higher DC infiltration in mouse liver and tumor tissue after GPC3-7-19-CAR-T-cell treatment compared with GPC3-CAR-T-cell treatment (Fig. 5F, G). Overall, these results confirmed that GPC3-7-19-CAR-T cells induce enhanced antitumor effects by recruiting DCs.
Potential Clinical Application Of Gpc3-7-19-car-t Cells As Treatment For Gpc3-positive Hcc Patients
After confirming the safety and efficacy of GPC3-IL7-CCL19-CAR-T cells in in vitro and in vivo mouse models, their utility in clinical application was further evaluated with primary HCC cells derived from 3 patients. Single primary GPC3-positive HCC cells were prepared by enzyme digestion and Ficoll-Paque separation. After 16–20 days, primary HCC cells were cocultured with CAR-T cells for 24 hours, and the killing efficiency was determined by flow cytometry. GPC3-IL7-CCL19-CAR-T cells killed approximately 70% of primary HCC cells, 25% higher than what GPC3-CAR-T cells achieved at an E/T ratio of 5:1 (Fig. 6A, B).
In addition, a 3D microfluidic tumor model was employed to evaluate the response of GPC3-IL7-CCL19-CAR-T cells to primary GPC3-positive HCC cells (Fig. 6C). In this system, before exerting their cytotoxicity on targets, CAR-T cells need to migrate from medium-filled channels (side) into a collagen-filled central region (tumor site). Over a 12-hour coculture, more CAR-T cells migrated into the tumor channel to kill tumor cells, and more dead cells in the central 3D matrix were observed, especially in the GPC3-7-9-CAR-T-cell group (Fig. 6D). Simultaneously, secreted factors were quantified in media channel supernatants. As expected, 100 pg/mL and 400 pg/mL IL-7 and 5000 pg/mL and 10000 pg/mL CCL19 were detected in the supernatants of GPC3-CAR-T cells and GPC3-7-19-CAR-T cells, respectively. In addition, 1300 pg/mL TNF-α and 4500 pg/mL IFN-γ were also detected in the supernatant of GPC3-7-19-CAR-T cells, which was 2-fold higher than those detected in GPC3-CAR-T-cell and 7–9 cell supernatants (Fig. 6E). In summary, GPC3-7-19-CAR-T cells displayed a higher migration ability and cytotoxic activity against primary HCC cells, indicating their potential in clinical applications in HCC patients.
Gpc3-7-19-car-t Cells Show Remarkable Antitumor Activity In One Patient With Hcc With Gpc3 Expression
Based on the preclinical results, we conducted a phase I clinical trial in advanced HCC patients with positive GPC3 expression to further explore the clinical safety and feasibility of GPC3-7-19-CAR-T-cell therapy. Patients were enrolled based on the inclusion and exclusion criteria. One patient was administered 5×106 GPC3-7-19-CAR-T cells/kg (CAR-positive rate 69%, Supplementary Fig. 2B) after lymphodepletion according to the regimen. Metastatic lesions at baseline and follow-up were assessed by computed tomography (CT). The size of the metastatic retroperitoneal lymph nodule had decreased significantly (22.1×15.6 mm vs. 15.1× 11.7 mm) and the mediastinal lymph nodule had almost disappeared by day 56 (Supplementary Fig. 2A). The patient did not suffer any toxic effects, and CT-based staging evaluation revealed stable response (SD) according to Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1. Blood was collected for pharmacokinetic (PK) and pharmacodynamic (PD) analyses. At day 56, the proportion of CD8 + T cells was much higher than that at baseline (67.9% vs. 16.6%, Supplementary Fig. 2C). Furthermore, TNF-α and IFN-γ secretion was significantly increased (Supplementary Fig. 2D, E). It is worth noting that the tumor biomarkers serum AFP and CEA were decreased by approximately 50% (Supplementary Fig. 2F). In addition, hepatic dysfunction marker enzymes such as γ-GT, AST, ALT, and ALP were obviously decreased (Supplementary Fig. 2G). Every 7 days, the lymphocyte populations of the patient were profiled. Compared with that before GPC3-7-19-CAR-Tcell infusion, the level of CD3 + T cells was higher on day 56. CD8 + T-cell subsets were sustainably increased and CD4 + T-cell subsets were decreased after CAR-T-cell treatment (Supplementary Fig. 2H).