Design of the AdCAR-T system
AdCAR-T comprises a two component signal transduction system based on a split recognition/activation design in which labeled AMs are applied to transmit antigen recognition into T-cell activation via an anti-label CAR. AdCAR-T mediated T-cell activation and target-cell lysis is the result of a two-step process (Fig. 1A), i) antigen-specific binding of the AM on the target cell and ii) binding of the AdCAR-expressing T cell to the AM. We used mAbs and mAb fragments as AMs to utilize their specific surface antigen-binding capacity. AMs are generated by biotinylation using a specific linker chemistry, resulting in a molecule comprising an antigen-binding moiety, a linker moiety and a label moiety (biotin) (Fig. 1A). The AdCAR is based on unique characteristics of the mAb mBio3. mBio3 binds to biotin in the context of a specific linker, referred to as a Linker-Label Epitope (LLE). We designed AdCAR-T by conjugating mBio3 derived scFvs on 2nd and 3rd generation CAR backbones consisting of different extracellular spacer domains, as indicated, CD8 transmembrane, 4-1BB or CD28 and 4-1BB co-stimulatory as well as CD3-ζ signaling domain. Truncated LNGFR (ΔLNGFR) was integrated downstream of a F2A site for detection and enrichment. In vitro testing was performed using the AdCAR-4-1BB-CD3-zeta, IgG4 hinge only, construct if not annotated otherwise. For initial evaluation, we LLE-conjugated the Fc-optimized CD19-4G7SDIE mAb (LLE-CD19 mAb) and the FDA/EMA-approved GD2 mAb ch14.18/dinutuximab beta (LLE-GD2 mAb), both in clinical use at our institution (26, 27).
AdCAR-T mediate highly specific target-cell lysis
AdCAR can be expressed on activated T cells by lentiviral transduction as determined by ΔLNGFR expression (fig. S1A). The applied expansion protocol consistently resulted in balanced CD4+ to CD8+ T-cell ratios (fig. S1B). AdCAR-T got activated exclusively in the presence of antigen-specific LLE-conjugated AMs and the corresponding antigen-expressing target cell, assessed by cytokine production and transition into an effector phenotype (Fig. 1B, fig. S1C). Importantly, AdCAR-T activation resulted in specific LLE-AM mediated target-cell lysis, demonstrated for LLE-CD19 mAb and LLE-GD2 mAb against the CD19+ BCP-ALL cell line NALM6 and the GD2+ neuroblastoma cell line LS. No specific lysis was observed in the presence of antigen-specific mAb without LLE conjugation or LLE-AMs of irrelevant specificity (Fig. 1, C-D). The molecular architecture of the different AdCAR variants is indicated in (fig. S2A). In these experiments the molecular variation was limited to the spacer length. A similar AdCAR expression level on T cells for the different AdCAR constructs is shown after LNGFR enrichment by microbeads in (fig. S2B). Comparing the impact of different extracellular spacer domains (23, 28, 29), we found superior performance of the short, IgG4-hinge (XS) construct independent of the i) target antigen CD19 in BCP-ALL targeting NALM6 (fig. S2C) or GD2 in neuroblastoma targeting LS (Fig. 2SE) and independent of the ii) the AM format, full-size LLE-CD19 mAb or fragments thereof [LLE-CD19 F(ab’)2, LLE-Fab], despite the different AM-size (fig. S2D) with regard to cytotoxicity (fig. S2, C-E) and cytokine production (fig. S2F). In vitro testing revealed no differences in effector function of 2nd versus 3rd generation AdCAR constructs in terms of cytotoxicity and repetitive cytotoxicity (fig. S3, A-B), however with regard to proliferative capacity, 3rd gen AdCAR-T were superior (fig. S3C).
AdCAR-T function is not impaired in the presence of biotin
One major obstacle of using biotin as a tag is the presence of free and protein-bound biotin in the human body. To test possible interference with free or protein-bound biotin, we assessed LLE-AM mediated target-cell lysis by AdCAR-T under increasing concentrations of biotin, up to 10000-fold of the physiological concentration (19), or in the presence of 50% human serum. Strikingly, there was only minor inhibition of AdCAR-T function at supraphysiologic free biotin concentrations, further underscoring the selective binding of the AdCAR to the LLE-tag (Fig. 2A, fig. S4B).
AdCAR-T mediate highly efficient and tightly controllable target-cell lysis
AdCAR-T sufficiently lysed target cells in vitro even at low E:T ratios (1:10), demonstrating serial killing capacity comparable to conventional CD19- and GD2-CAR-T cells (Fig. 2, B-C). Target-cell lysis was equally mediated by biotinylated full-size LLE-mAb, LLE-F(ab’)2 and LLE-Fab (fig. S2D). AdCAR-T efficiently lysed target cells at LLE-AM concentrations starting as low as 1 pg/mL (EC50 of LLE-CD19 mAb vs. NALM6 = 7.9 pg/mL) with a coverage of 5 log-levels at highly potent activity, ensuring a wide therapeutic range with optimal effector function > 1 ng/mL LLE-AM (Fig. 2D, fig. S4A). LLE-AM titration enabled finely tunable “ON”-switch function. Moreover, LLE-AM dependent AdCAR-T activity could also be terminated by addition of antigen-specific mAbs without LLE-tag, functioning as an “OFF”-switch (Fig. 2E), underscoring the specificity and controllability of AdCAR-T system.
AdCAR-T eradicate disseminated lymphoma in vivo
Having proven functionality of the AdCAR system in vitro, we next set out to investigate efficacy in vivo. First we tested the general in vitro stability of the LLE-conjugation of full length mAb in human whole blood for 24 hours (Fig. 3A). Then we evaluated mouse plasma LLE-mAb levels 24 hours after intravenous (iv) versus intraperitoneal (ip) application of 5 µg LLE-mAb and found no clear benefit of iv-application of the LLE-mAb compared to ip-application (Fig. 3B). Consequently, we proceeded with ip-application of LLE-mAb in NSG mice in all subsequent in vivo experiments. The plasma levels of LLE-CD19 mAb were measured after 1 week and after two weeks in non-tumor bearing mice (Fig. 3C) and revealed a plasma level half-life of t1/2=3.19 days at 50 µg ip application of LLE-CD19 mAb (fig. S4C). Consequently, we applied LLE-mAb twice per week. Further we tested the impact of coadministration of 10 mg human IgG ip to saturate Fc-receptors and thus to mitigate Fc-receptor mediated effector function. There was no impact of LLE-mAb mouse plasma levels without hIgG compared to the condition with coadministration of 10 mg hIgG (fig. S4D).
In vitro functional testing elucidated a significant higher proliferative capacity of 3rd generation AdCAR-T compared to 2nd generation AdCAR-T (fig. S3C) and in vivo comparison of 2nd versus 3rd generation AdCAR-T confirmed the importance of CAR-T cell proliferation to eradicate leukemia in a NALM6 tumor model utilizing the LLE-CD19 4G7SDIE mAb as AM. The superiority of 3rd versus 2nd generation CAR systems in adapter CAR technologies has also been previously reported by other groups (30). Conventional CD19 CAR-T cells served as positive controls and cleared NALM6 from NSG mice more rapidly than the 3rd gen AdCAR-T (fig. S3D). In consequence, for subsequent in vivo evaluation of the AdCAR technology we utilized the 3rd generation AdCAR-CD28-4-1BB-CD3-zeta construct.
To demonstrate the operational versatility and the potential for straight clinical translation of the AdCAR-T technology, we chose the FDA/EMA approved CD20 mAb rituximab for in vivo evaluation. Rituximab was LLE-conjugated, achieving a purity of > 95% LLE-rituximab (average 2 LLE/mAb). AdCAR-T were generated on the CliniMACS Prodigy™, allowing a GMP-compliant closed-system processing for cell enrichment, activation, transduction, washing and expansion (31, 32). To underscore in vivo efficacy, we decided to use a rapidly progressive xenograft model of Burkitt’s lymphoma (Raji cell line) (Fig. 3D, fig. S5A) (31). After confirmation of homogeneous engraftment on day − 1 (fig. S5B), mice were grouped and the treatment according to indicated conditions was initiated. LLE-rituximab was injected intraperitoneally at 50 µg twice weekly starting on day − 1 and suspended on day + 23. AdCAR-T were injected intravenously on day 0, 6.23 × 106 AdCAR-T and a total of 20 × 106 human T cells per mouse (Fig. 3D). Conventional CD20-CAR-T (31), 6.07 × 106 cells per mouse, served as a positive control. Tumor burden was assessed by in vivo bioluminescence imaging (BLI). Strikingly, AdCAR-T in combination with LLE-rituximab completely eradicated disseminated lymphoma, as efficient as conventional CD20-CAR-T, although with slightly delayed kinetics. Mice remained in complete remission, demonstrated by BLI and flow cytometry of bone marrow, even after LLE-rituximab administration was terminated. In contrast, neither AdCAR-T nor LLE-rituximab alone mediated a significant effect on tumor burden (Fig. 3, E-F, fig. S5A). Solid engraftment of AdCAR-T was confirmed in the bone marrow on day + 44 (fig. S5C).
In a NHL JeKo-1 tumor model 1 × 107 AdCAR-T were challenged to control and eradicate tumor burden at two different dose levels of either LLE-CD19 4G7SDIE mAb or LLE-CD20 rituximab applied ip twice per week (DL1 50 µg, DL2 5 µg). AdCAR-T plus CD19 mAb and CD20 mAb without LLE conjugation served as negative controls. At DL1 either with LLE-CD19 mAb or LLE-CD20 mAb, AdCAR-T controlled tumor growth, whereas at DL2 tumor growth kinetics were delayed in 3 mice and one mouse only achieved tumor clearance (fig. S3, E-F). Collectively, these data clearly underscore high in vivo therapeutic efficacy of the AdCAR-T system and warrant that clinical investigation should be initiated.
AdCAR-T mediate target-cell lysis against a variety of antigens proportionately to the frequency of antigen positive expressing target cells
One key feature of the adapter approach is the possibility for versatile and universal targeting. To test this hypothesis, we generated LLE-mAbs targeting antigens, associated with myeloid malignancies: CD15, CD32, CD33, CD38, CD117, CD123, CD133, CD135, CD276, CD371.
LLE-mAb specific target-cell lysis was evaluated in the acute myeloid leukemia (AML) cell lines HL-60 and Kasumi-1, harboring inter- and intratumoral heterogeneity of target antigen expression. We found highly efficient target-cell lysis for 9 out of 10 evaluated LLE-AMs. Importantly, the percentage of specific lysis correlated well with the percentage of antigen-expressing cells in general, indicating the ability to specifically eliminate antigen-positive cells, while sparing antigen-negative cells (Fig. 4, A-B).
Simultaneous or sequential targeting might prevent selection of antigen-loss variants
Therapy failure due to antigen evasion is one of the major clinical challenges in CAR-T therapy (9, 10). To address this issue, we generated antigen-loss variants for CD19, CD20 and CD19/CD20 of the non-Hodgkin Lymphoma (NHL) cell line JeKo-1 by CRISPR/Cas9 knockout. All JeKo-1 variants showed similar growth kinetics, allowing simultaneous evaluation (fig. S6A). To test, whether dual or multiple targeting might prevent the specific selection of antigen-loss variants, we exposed AdCAR-T with a mix of wild type and knockout variants (Fig. 4C, fig. S6B). Single targeting by either LLE-CD19 mAb or LLE-CD20 mAb eliminated target antigen positive cells with high selectivity, while sparing antigen negative variants (Fig. 4, D-E, fig. S6, C-D). Moreover, dual targeting led to the selection of double negative Jeko-1 cells (Fig. 4F, fig. S6E). Addition of a 3rd AM, LLE-ROR-1 mAb, led to additional reduction of the double negative subset underscoring the capability for multiple “OR”-gating by AdCAR-T (Fig. 4G). Monotargeting using the LLE-ROR-1 mAb significantly reduced the cell number in all four JeKo-1 variants comparably (fig. S6F). Moreover, AdCAR-T were demonstrated to be capable and flexible in sequential targeting. Thus sequential use of LLE-mAbs of different specificity (Fig. 5A) led to the anticipated reduction of the corresponding positive JeKo-1 subset.. Both treatment strategies were successful in sequential targeting. Either starting with LLE-CD19 mAb and in a second step by adding LLE-CD20 mAb after 48 h or vice versa. Solely, the corresponding positive tumor cell subsets were sequentially eliminated by addition of the LLE-mAb. The repetitive and sequential targeting demonstrates the serial killing capacity under various circumstances indicating the cellular functional integrity of AdCAR-T (Fig. 5A, fig. S3B).
AdCAR-T function as an “AND”-gate
In LLE-AM titration experiments, we revealed that a certain threshold concentration of LLE-mAb bound to the target cell surface [LLE density (Tρ)] expressed on the target cell is required to activate AdCAR-T (Fig. 2D, fig. S4A). AdCAR-T specifically bind to LLE-tags, independently of the targeted antigen (Fig. 6A). Thus, we hypothesized that Tρ can either be reached by increasing concentrations of one single specific LLE-AM (Fig. 6B, left panel) and likewise as the result of the assembly of LLE-AMs targeted to different antigens (Σρ = ρA + ρB + ρC+…+ ρX) on the target cell surface at below activation threshold concentrations (Fig. 6B, right panel), referred to as “surface activation matrix” (SAM). To test this hypothesis, we performed titration experiments with single LLE-AMs and combinations thereof. The antigen expression of CD19 and CD20 in JeKo-1 is displayed in (Fig. 6C). Monotargeting in JeKo-1 showed parallel lysis curves for LLE-CD19 mAb and LLE-CD20 mAb. Combining these two LLE-AMs vs. JeKo-1 at equimolar concentrations led to additive lysis (Fig. 6, D-E). Further, titration of monotargeting was evaluated in NALM6 for LLE-CD19 mAb, LLE-CD10 mAb, LLE-CD138 mAb and LLE-CD22 mAb as well as the combination of these four LLE-mAbs at below threshold concentrations leading to significantly increased lysis (fig. S9A). The mechanism of additively reaching ρT (SAM) was confirmed for combining three LLE-AMs targeted to CD19, CD10 and CD138 vs. NALM6 in BCP-ALL, in neuroblastoma targeting GD2, CD81 and CD276 vs. LS and in triple negative breast cancer targeting EGFR, CD47 and CD276 vs. Hs578T (Fig. 6, F-H, fig. S7, A-B, D, fig. S9A). The expression level of CD32, CD33, CD38, CD305 and CD371 in the AML cell lines U937, HL-60 and MOLM13 is displayed in (Fig. 7, A, D-E). Encoded in the same colors, the lysis for the monotargeting is shown. To further potentiate the combinatorial effect, we combined 5 different AMs targeting the above listed target antigens at below threshold concentrations at the same time. Unprecedented, we found complete target elimination in combination of 5 AMs without a significant cytotoxic effect mediated by single targeting, demonstrating that AdCAR-T can function as an “AND”-gate (Fig. 7, B-E, fig. S7, C-D).
AdCAR-T selectively lyse target cells by integration of multiplex expression profiles
If AdCAR-T can identify target cells by multi-variant antigen expression profiles, they should be capable to differentiate between cancerous and healthy tissue by rational AM selection. As a proof-of-concept, we co-cultured AdCAR-T with NALM6 cells, expressing CD10, CD19 and CD138, together with freshly isolated peripheral B cells expressing solely CD19 or unmanipulated PBMCs containing B cells expressing solely CD19. Expression level of CD10, CD19 and CD138 is illustrated in (Fig. 8A). Importantly, for B-cell positive selection CD19 REAlease® beads were used to guarantee no occupation of the CD19 extracellular domain by CD19 directed microbeads. Titration experiments showed a high intrinsic susceptibility of NALM6 to AdCAR mediated lysis and a combinatorial effect at fascinating low AM concentrations (fig. S8). Addition of an AM combination targeted to CD10, CD19 and CD138 at below threshold concentration for single targeting led to the elimination of NALM6, without significant lysis of healthy B-cells. In contrast, higher concentrations of LLE-CD19 mAb above the threshold concentration for monotargeting with LLE-CD19 mAb sufficiently eliminated both, NALM6 and healthy B cells (Fig. 8, B-C). Our results underscore the potential of selective cytotoxic pressure and elimination of cell subsets by AdCAR-T cells in combination with LLE-AMs at below threshold concentration based on distinct antigen expression profiles which are characteristic in malignant transformed leukemic blasts of the B lineage but also in other cancers, that do partly share potential target antigens. AdCAR mediated cytotoxicity might be reduced by distribution and switch of AM combinations more likely to hit cancerous versus healthy tissues (fig. S10).