Expression of IL10R and CD33 in bone marrow mononuclear cells of AML patients
Using flow cytometry (FCM), we assessed the expression of CD33, IL10RA and IL10RB in bone marrow mononuclear cells (BMMNCs) of newly diagnosed AML patients through a CD45/SSC double-parameter scatter plot gating method (Figure S1). The results showed that although the mentioned markers were co-expressed in the primary cells, the positive expression rates of IL10RA [(8.677 ± 8.124)%] and IL10RB [(33.38 ± 31.65)%] were lower than those of CD33 [(92.88 ± 8.214)%; vs IL10RA, p < 0.0001; vs IL10RB, p = 0.0019] (Fig. 1b). More notably, this underscores the significance of epitope spreading through targeting CD33 over the simplistic approach of solely targeting IL10R.
Construction of CAR.BsAb-T and verification of CD33 bsAb secretion
We fused the anti-human CD33 single-chain variable fragment (scFv) with the anti-human CD3 scFv derived from our previous study, and inserted them into our previously constructed second-generation IL 10R CAR, leading to the generation of the CAR.BsAb expression plasmid. As controls, we included vectors featuring the second-generation CAR but lacking the extracellular scFv (4-1BB Vector) (Fig. 1c).
T cells were transduced with the expression vectors, and the infection efficiency of Vector-T, IL10R CAR-T, and CAR.BsAb-T was assessed (Fig. 1d). Although the CAR-T antigen-binding region exhibited stable expression on the cell surface, CAR.BsAb-T cells demonstrated a lower mean fluorescence intensity (MFI) level of CAR compared to IL10R CAR-T cells (Fig. 1e).
Supernatant from Vector-T and CAR.BsAb-T was collected and purified using His-tag affinity chromatography. The result of Western Blot confirmed the secretion of CD33 bsAb (56kDa) (Fig. 1f). In addition, using FCM, the surface expression of a His-tag on Molm13 cells or T cells was detected after co-incubation with purified protein, which validate the binding characteristics of the bsAb with CD3 and CD33 antigens (Fig. 1g). The surface binding of CAR.BsAb-T cells to His-tag or APC-labeled human siglec-3/CD33 protein was detected by FCM, confirming the antigen-specific binding capability of secreted CD33 bsAb (Figure S2).
Additionally, we used ELISA to measure CD33 bsAb concentration in the supernatant. It showed an increase over time post-transduction, with the concentration stabilizing by day 8 (Fig. 1h).
FCM was used to gate AML blasts based on CD45dim and SSClow; subsequently, the expression of IL-10RA, IL-10RB, and CD33 in primary cells was analyzed.
FCM detected surface binding of CAR.BsAb-T cells to either His-tag or APC-labeled human Siglec-3/CD33 protein.
In vitro efficacy of CAR.BsAb-T in killing AML cells
To assess the activation initiation of CAR.BsAb-T, T cells from different groups were co-cultured with Molm 13. The activation and proliferation markers, CD69 at 6 hours (Fig. 2a) and CD25 at 24 hours (Fig. 2b), were evaluated using FCM. Compared to the control vector and IL10R CAR-T groups, the levels of early and late activation markers CD69 and CD25 in CAR.BsAb-T group increased, indicating the effective activation of CAR.BsAb-T cells.
Subsequently, co-cultures of leukemia cells, including THP-1, U937, MV4-11 and Molm 13 cells with Vector-T, IL10R CAR-T, and CAR.BsAb-T at varying effector-to-target (E:T) ratios were performed to evaluate antigen-specific cytotoxicity (Fig. 2c-d). These findings demonstrate that CAR.BsAb-T cells do effectively respond to antigen-positive myeloid leukemia cell lines. Moreover, at different time points, they enhance the specific killing effect of IL10R CAR-T to some extent.
The expression proportion of CD107a on the surface of T cells was detected 5h after co-culture at a 1:1 E:T ratio, demonstrating that CAR.BsAb-T induced a more intense degranulation effect (Fig. 2e).
The supernatant was collected after co-culturing at an E:T ratio of 1:1 for 48h, and the secretion of cytokines was detected by FCM (Fig. 2f). More strikingly, in co-culture with Molm13 leukemia cells, CAR.BsAb-T significantly increased the levels of IL-2, TNF-α and IFN-γ, but the level of IL-6, an important factor in cytokine release syndrome (CRS), showed no significant difference from that of IL10R CAR-T, indicating that CAR.BsAb-T increased the killing effect on target cells without causing significant side effects.
Effectiveness of CAR.BsAb-T in vitro lysis of primary AML cells
Primary BMMNCs were randomly selected from six AML patients as target cells (Fig. 3a, Table 1), thereby enabling a further evaluation of the cytotoxicity of CAR.BsAb-T cells against primary leukemia cells. In alignment with prior in vitro cytotoxicity assays on AML cell lines, CAR.BsAb-T cells demonstrated a specific lysis capability that correlated with antigen expression levels (Fig. 3b), and exhibited elevated degranulation levels in comparison to that of IL10R CAR-T cells (Fig. 3c).
In comparison to that of IL10R CAR-T, there was an obvious increase in IL-2 release, while the release of IFN-γ, TNF-α, and IL-6 showed no significant differences (Fig. 3d). FCM analysis showed a marked reduction in the proportion of the abnormal CD34+ cell population (Figure S3) within the residual target cells, underscoring the potential clinical applicability of CAR.BsAb-T cells in targeting leukemia stem cells in AML patients.
Table 1
Patient ID
|
Disease
|
Sample type
|
CD33(%)
|
IL10RA(%)
|
P1
|
AML
|
BM
|
97.77
|
13.03
|
P2
|
AML
|
BM
|
99
|
12.14
|
P3
|
AML
|
BM
|
87.44
|
19
|
P4
|
AML
|
BM
|
93.17
|
14.21
|
P5
|
AML
|
BM
|
61.96
|
13.98
|
P6
|
AML
|
BM
|
47.36
|
5.66
|
Role of CAR.BsAb-T in immune escape
In AML cell lines, Kasumi-1 cells displayed lower surface expression levels of IL10RA and IL10RB compared to the previously mentioned cell lines (Fig. 4a). Co-culturing effector cells with Kasumi-1 at various E:T ratios revealed that CAR.BsAb-T enhanced the cytotoxic effect of IL10R CAR-T (Fig. 4b). The assessment of cytokine secretion in the culture system (Fig. 4c) further demonstrated that the combined strategy effectively addressed immune escape issues arising from lower antigen expression levels.
To model immune escape resulting from the loss of CD33, Thp-1 and U937 cell lines with CD33 knockout (Thp-1CD33KO, U937CD33KO) were constructed (Fig. 4d). Vector-T, IL10R CAR-T, CD33 bsAb-T, and CAR.BsAb-T were co-cultured with the aforementioned target cells at different E:T ratios. It was observed that CAR.BsAb-T partially improved the cytotoxic effect and cytokine release of CD33 bsAb-T, but was less potent than IL10R CAR-T (Fig. 4e-f), possibly due to lower surface CAR expression levels compared to IL10R CAR-T.
Myeloid-derived suppressor cells (MDSCs) are immature myeloid lineage cells with immunosuppressive properties that directly contribute to immune escape and promote tumor invasion through various non-immunologic mechanisms [34]. Early-stage MDSCs (eMDSCs) are characterized as lin− (including CD3, CD14, CD15, CD19, CD56) HLA-DR− CD11b+ [35]. By co-culturing effector cells with primary BMMNCs obtained from AML patients, the eradication effects of CAR.BsAb-T on eMDSCs could be observed within 36h. (Figure S4)
BMMNCs were cultured with effector T cells at a 1:1 E:T ratio for 36 hours, and representative FCM analysis of the residual proportion of eMDSCs [lin− (including CD3, CD14, CD15, CD19, CD56) HLA-DR− CD11b+] was conducted.
Sustained killing activity of CAR.BsAb-T against tumor cells through bystander T cells
To evaluate the effects of the released CD33 bsAb, as previously reported, cell-impenetrable transwell membranes (0.4 µm pore size) were utilized.[28] Effector cells were added to the upper chamber and human primary T cells to the lower chamber (Fig. 5a). After co-culturing for 72 hours, the lower chamber human primary T cells, stained with CFSE, exhibited a decrease in fluorescence intensity coinciding with the appearance of proliferation peaks. This suggests that T cells undergo proliferation and activation in response to CD33 bsAb stimulation. (Fig. 5b).
Next, we speculated whether the untransduced primary T (UTD-T) cells could be induced to exhibit cytotoxic activity. Therefore, the UTD-T cells and target cells Molm 13, U937, or U937CD33KO at a 1:1 ET ratio were added to the lower chamber (Fig. 5c). It was observed that in the co-culture systems with Molm 13 and U937, these bystander UTD-T cells in the lower chamber could utilize the CD33 bsAb secreted in the upper chamber to exert significant specific cytotoxic effects, while no corresponding killing effect was observed in the U937CD33KO co-culture system (Fig. 5d).
Cytokine levels in the co-culture system were examined, and showed a notable increase in Th1 cytokines IFN-γ and TNF-α, while no significant difference was observed in IL-2, indicating the antigen-specific cytotoxic effects of bystander UTD-T cells (Fig. 5e).
In order to simulate tumor recurrence in vivo, a rechallenge model involving repeated stimulation of tumor cells every 48 hours for a total of three rounds was implemented. The purpose of this model was to thoroughly evaluate the cytotoxic effects of the aforementioned CAR-T cells during disease recurrence (Fig. 5f). The results indicated that although a decrease in cytotoxicity was observed in CAR.BsAb-T after repeated antigen stimulation, it continued to enhance the enduring killing capacity of IL10R CAR-T. Particularly noteworthy is that, even after the third round of stimulation, the proportion of residual target cells decreased (Fig. 5g).
In vivo Efficacy of CAR.BsAb-T in anti-AML
NSG mice bearing cell line-derived xenograft (CDX) tumors were used to evaluate the in vivo efficacy of the aforementioned CAR-T constructs. These mice were injected with 1×106 Molm 13 luciferase cells (Molm13luc) on day − 3, followed by an intravenous injection of 1×107 T cells on day 1. The regimen of in vivo anti-tumor effects of the CAR-T constructs was shown in Fig. 6a. The dynamic body weight changes of mice monitored on a weekly basis revealed that, from day 7 onwards, the control group exhibited a decline in body weight, leading to cachexia and rapid progression to death. In contrast, the CAR-T and CAR.BsAb treated experimental groups of mice maintained a stable trend. (Fig. 6b)
Imaging conducted at day 0, 7, 14, 21, and 28 post-transplantation using bioluminescence showed the in vivo infiltration of leukemia cells (Fig. 6c-d), with CAR.BsAb-T contributing to further reduction in the leukemia burden.
Analysis of the difference in survival time demonstrated that, compared to 4-1BB Vector-T, CAR.BsAb-T significantly extended the survival of mice (Fig. 6e).
To further investigated whether CAR.BsAb-T could stimulate bystander T cells to exert tumor killing effects in vivo just as observed in vitro study shown in Fig. 5D, a PDX mouse model was established using 1×106 Molm 13luc cells and then treated with 5×106 different CAR-T cells. On the third day following this treatment, 5×106 UTD-T cells were infused. Meanwhile, a CAR BsAb-T control group was established and only CAR.BsAb-T treatment (without UTD-T cell infusion) was given (Fig. 7a). Notably, by the 14th day, the 4-1BB Vector group exhibited a significant decrease in body weight and increased mortality due to tumor burden (Fig. 7b-c). At the same time, in the CAR.BsAb-T sequential administration with UTD-T group, a significant reduction in tumor burden and an extended survival period were observed, while no bystander effects were observed in the IL10R CAR-T and CAR.BsAb-T treatment only groups (Fig. 7d-e).