Purpose:

The development of safe and effective chimeric antigen receptor (CAR) T-cell therapy for acute myeloid leukemia (AML) has largely been limited by the concomitant expression of most AML-associated surface antigens on normal myeloid progenitors and by the potential prolonged disruption of normal hematopoiesis by the immunotargeting of these antigens. The purpose of this study was to evaluate B7-homolog 3 (B7-H3) as a potential target for AML-directed CAR T-cell therapy. B7-H3, a coreceptor belonging to the B7 family of immune checkpoint molecules, is overexpressed on the leukemic blasts of a significant subset of patients with AML and may overcome these limitations as a potential target antigen for AML-directed CAR-T therapy.

Experimental Design:

B7-H3 expression was evaluated on AML cell lines, primary AML blasts, and normal bone marrow progenitor populations. The antileukemia efficacy of B7-H3–specific CAR-T cells (B7-H3.CAR-T) was evaluated using in vitro coculture models and xenograft models of disseminated AML, including patient-derived xenograft models. The potential hematopoietic toxicity of B7-H3.CAR-Ts was evaluated in vitro using colony formation assays and in vivo in a humanized mouse model.

Results:

B7-H3 is expressed on monocytic AML cell lines and on primary AML blasts from patients with monocytic AML, but is not significantly expressed on normal bone marrow progenitor populations. B7-H3.CAR-Ts exhibit efficient antigen-dependent cytotoxicity in vitro and in xenograft models of AML, and are unlikely to cause unacceptable hematopoietic toxicity.

Conclusions:

B7-H3 is a promising target for AML-directed CAR-T therapy. B7-H3.CAR-Ts control AML and have a favorable safety profile in preclinical models.

Translational Relevance

Because of its high expression on malignant cells and its restricted distribution in normal tissues, B7-homolog 3 (B7-H3) is an attractive target for antibody-based cancer immunotherapy. We have previously generated B7-H3–specific chimeric antigen receptor (CAR) T cells (B7-H3.CAR-T), demonstrated antitumor efficacy in several solid tumor models, and demonstrated lack of evident toxicities in a syngeneic tumor model. Our current results suggest that B7-H3 is expressed on a subset of acute myeloid leukemia (AML), but is not detectable on normal bone marrow progenitor populations. We have demonstrated that B7-H3.CAR-Ts exhibit efficient antigen-dependent cytotoxicity in vitro and in xenograft models of AML, and are unlikely to cause unacceptable hematopoietic toxicity. B7-H3 is thus a viable target antigen for AML-directed CAR T-cell therapy. Our results support the clinical development of B7-H3.CAR-Ts for the treatment of patients with relapsed/refractory B7-H3–positive AML.

Acute myeloid leukemia (AML) accounts for approximately 80% of acute leukemias in adults and has an incidence of at least 20,000 cases per year in the United States (1). Despite advances in risk stratification, optimization of chemotherapy regimens, improvement in stem cell transplantation techniques, and the advent of multiple targeted therapies, prognosis remains poor. These clinical findings emphasize the need for novel treatment strategies (1). Adoptive cellular therapy with chimeric antigen receptor (CAR) T cells (CAR-T) has emerged as a highly effective form of immunotherapy for B-cell acute lymphoblastic leukemia (ALL) and certain other B-cell malignancies (2); however, safe and effective CAR-T therapy for AML remains elusive. The success of CAR-T therapy in B-cell malignancies is predicated on the tolerability of the protracted B-cell aplasia that often accompanies CD19-directed CAR-T therapy. In contrast, prolonged disruption of normal myelopoiesis (especially neutropenia) would not be tolerable, and the development of CAR-T therapy for AML has been largely limited by the fact that most AML-associated surface antigens are also expressed on normal myeloid progenitor cells (3).

While it is difficult to envision the identification of cell surface antigens that are absent on all normal myeloid progenitors and yet expressed on all subtypes of AML, it seems plausible that some antigens absent on early myeloid lineage cells may be preferentially overexpressed in certain AML subtypes. We have identified B7-homolog 3 (B7-H3) as one such candidate. B7-H3 (or CD276) is a coreceptor belonging to the B7 family of immune checkpoint molecules. Its structure is that of a 110-kDa type I transmembrane protein with four Ig-like domains, a transmembrane domain, and a short cytoplasmic tail (4). The receptor for B7-H3 has yet to be definitively identified (5, 6), and the nature of its signaling also remains controversial (7). Although B7-H3 mRNA is expressed in many normal tissues, corresponding protein expression is limited (7). In multiple human cancers, however, B7-H3 protein is overexpressed. Examples include various solid tumors (glioblastoma, lung, breast, colon, pancreatic, renal, ovarian, prostate, and melanoma), as well as a subset of patients with AML (7, 8). B7-H3 expression in AML appears to be higher in AML with a monocytic immunophenotype (8), which is often associated with aggressive clinical features (9, 10). To evaluate the potential safety of B7-H3 as a target for CAR-T therapy, we have previously performed a comprehensive evaluation of B7-H3 expression in normal human tissues and demonstrated a lack of major organ toxicity in a syngeneic tumor model (11). We describe here the preclinical assessment of B7-H3–specific CAR T cells (B7-H3.CAR-T) as a possible therapy for a subset of patients with AML and our evaluation of the potential hematopoietic toxicity of this approach.

Cell lines and human samples

Deidentified cryopreserved samples of peripheral blood and bone marrow aspirate from patients with newly diagnosed AML were obtained through the University of North Carolina (UNC) Tissue Procurement Facility (UNC Hospital, Chapel Hill, NC). Primary AML blasts for use in patient-derived xenograft (PDX) models were obtained from the Public Repository of Xenografts (PRoXe, Dana-Farber Cancer Institute, Boston, MA). See Supplementary Materials and Methods for additional details, descriptions of other cell lines used, and tissue culture conditions.

Carboxyfluorescein diacetate succinimidyl ester assays

T cells were labeled with 1.5 μmol/L carboxyfluorescein diacetate succinimidyl ester (CFSE; Invitrogen) as described previously (12), followed by coculture with irradiated B7-H3+ or B7-H3 cell lines. After 5 days, T-cell proliferation was assessed via flow cytometry analysis.

Generation of retroviral vectors and activated B7-H3.CAR-Ts

The B7-H3–specific single-chain variable fragment (scFv) from the murine mAb, 376.96 (13), was cloned from the 376.96 mouse hybridoma as described previously (11). Generation of retroviral vectors and activated B7-H3.CAR-Ts has been described previously (11, 14, 15) and is summarized further in the Supplementary Materials and Methods.

Flow cytometry

Flow cytometry data acquisition was performed using a BD FACSCanto II or BD FACS Fortessa Flow Cytometer and BD Diva Software (BD Biosciences). Data analysis was performed using FlowJo Software versions 9.3 and 10.5 (Tree Star). See Supplementary Materials and Methods for a list of mAbs used.

In vitro analysis of B7-H3.CAR-T activity against AML cell lines

The activity of B7-H3.CAR-Ts was evaluated in coculture models with B7-H3+ AML cell lines or primary patient-derived monocytic AML blasts. For cell lines, 0.5 × 106 tumor cells were plated in a 24-well plate in 2 mL of media (without any exogenous cytokines added). For primary AML blasts, 1 × 106 cells from cryopreserved bone marrow aspirates were plated in a 24-well plate. Control T cells or B7-H3.CAR-Ts were then added at a 1:5 effector-to-target (E:T) ratio. Twenty-four hours later, 500 μL of coculture supernatant was collected and the concentration of IFNγ and IL2 was measured by ELISA (R&D Systems). On days 4–7 of coculture, cells were collected, and the percentage of T cells and residual tumor cells was assessed via flow cytometry. Monocytic cell lines and primary AML blasts were identified via staining for CD33 and T cells via staining for CD3.

Colony formation assay of normal hematopoietic progenitors

CD34+ cells were isolated via Magnetic Microbead (Miltenyi Biotec) selection from umbilical cord blood units obtained from healthy donors. CD34+ cells were coincubated with B7-H3.CAR-28-Ts, CD19.CAR-28-Ts, or control T cells [derived from donor peripheral blood mononuclear cells (PBMC)] at an E:T ratio of 10:1 (T-cell concentration of 5 × 105/mL) for 6 hours, after which the coculture was diluted 1:11 (without depletion of T cells) in methylcellulose-containing medium supplemented with recombinant cytokines (MethoCult H4434 Classic; StemCell Technologies) and plated in duplicate as described previously (16). After 2 weeks in culture, granulocyte-macrophage progenitor (GMP) and common myeloid progenitor (CMP) colonies were scored in a blinded fashion using a high-quality inverted microscope.

Xenograft mouse models using AML cell lines

For mouse xenograft models, monocytic AML cell lines (THP1 and OCI-AML2) were transduced with a retroviral vector encoding the eGFP firefly luciferase (eGFP-FFluc) gene (17). For the THP1 model, a single-cell clone was first selected on the basis of high eGFP expression and high in vitro FFluc activity (18). eGFP-FFluc-THP1 or FFluc-OCI-AML2 cells were injected into the lateral tail vein of 6- to 8-week-old female NOD scid IL2Rγ−/− (NSG) mice. Seven days later, mice were injected intravenously with CAR-T cells. Further details of each experiment are described below. Tumor growth was monitored via bioluminescence imaging using an IVIS Lumina II In Vivo Imaging System (PerkinElmer; ref. 16). Mice were monitored and euthanized according to UNC (Chapel Hill, NC) Institutional Animal Care and Use Committee (IACUC) standards and/or when the luciferase signal (total flux) reached a predetermined threshold (1 × 1010 photons/second for eGFP-FFLuc-OCI-AML2 model and 1 × 1011 photons/second for eGFP-FFluc-THP1 model due to increased FFluc intensity compared with eGFP-FFLuc-OCI-AML2).

PDX models

Patient-derived AML blasts were obtained from PRoXe (samples DFAM-6855 and CBAM-44728) and expanded in vivo in NSG mice. For the first model (DFAM-6855), AML blasts were injected intravenously into 6- to 8-week-old female NSG mice and engraftment was monitored via weekly flow cytometry analysis of peripheral blood CD45+CD33+ cells. When the majority of mice had >20% peripheral blood AML blasts, they were randomized and injected intravenously with either B7-H3.CAR-28-Ts or control T cells. Eight days later, all mice were sacrificed and flow cytometry was used to quantify AML blasts and T cells in blood, bone marrow, and spleen. In an additional experiment, blasts were retrovirally transduced to express the fusion protein, eGFP-FFLuc, expanded in vivo in NSG mice, sorted by flow cytometry on the basis of eGFP expression, expanded again in vivo, and then similarly injected into female NSG mice. After 4 days, mice were randomized and injected intravenously with either B7-H3.CAR-28-Ts or CD19.CAR-28-Ts. Tumor growth was assessed via bioluminescence imaging using an IVIS Lumina II In Vivo Imaging System (PerkinElmer; ref. 16), unless otherwise indicated. Mice were monitored and euthanized according to UNC IACUC standards. In a second model (CBAM-44728), AML blasts were expanded in vivo and then injected intravenously in 6- to 8-week-old female NSG mice. After 4 days, mice were injected intravenously with either B7-H3.CAR-28-Ts or CD19.CAR-28-Ts. All mice were sacrificed on day 63 after T-cell injection and flow cytometry analysis was used to quantify AML blasts and CAR-Ts in the bone marrow, spleen, and blood.

Humanized mouse models

Hematopoietic stem cells (HSC) were isolated from human fetal liver tissue specimens (obtained from medically indicated or elective pregnancy termination) and were injected intrahepatically into 2- to 6-day-old NSG mice (19). Engraftment of human leukocytes was monitored in the peripheral blood beginning at 6 weeks. Following reconstitution of 40%–60% human leukocytes, 2 mice were sacrificed and Magnetic Microbeads (Miltenyi Biotec) were used to isolate human T cells (CD4+ or CD8+ cells) from the murine spleens. After activation with CD3 and CD28 antibodies, a subset of T cells was retrovirally transduced to generate B7-H3.CARs-Ts as described above. B7-H3.CAR-28-Ts or control T cells were then injected intravenously into the remaining mice on days 0 and 16. After the initial T-cell injection, mice were monitored regularly for the development of graft-versus-host disease or systemic inflammatory response syndrome. Levels of circulating human CD45+CD3+, CD45+CD19+, CD45+CD33+, and CD45+CD14+ cells were monitored via flow cytometry on a weekly basis. Mice were sacrificed on day 40 and flow cytometry was used to evaluate these populations in the bone marrow and spleen, as well as levels of human CD34+CD38+ and CD34+CD38 cells in the bone marrow.

B7-H3 is expressed on monocytic AML cell lines and expression compares favorably with other potential targets for AML-directed CAR-T therapy

Human monocytic AML cell lines (THP1, U937, OCI-AML2, and OCI-AML3) were stained with the B7-H3–specific mAbs, 376.96 and 7-517, demonstrating high expression of B7-H3 (Fig. 1A). In addition to staining with the B7-H3–specific mAb 7-517, the same cell lines, as well as a B7-H3 acute promyelocytic leukemia cell line (HL60), and healthy donor peripheral blood monocytes (CD3CD19CD20CD56HLA-DR+CD14+) were stained with mAbs specific for other recently identified candidate surface antigens for AML-directed CAR-T therapy (ADGRE2, CCR1, CD70, LILRB2, and CLEC12A; ref. 20). As shown in Fig. 1B, the lack of B7-H3 expression on normal human monocytes and high expression on monocytic AML cell lines were similar to the expression pattern of CD70, whereas ADGRE2, CCR1, LILRB2, and CLEC12A were all found to be expressed on normal monocytes and had variable expression on monocytic AML cell lines.

Figure 1.

B7-H3 is expressed on monocytic AML cell lines and primary AML blasts obtained from patients with monocytic AML, but not on bone marrow progenitor cells. A, Expression of B7-H3 in four human monocytic AML cell lines stained with the 376.96 or 7-517 mAbs (black lines) versus isotype control (gray fill) as assessed by flow cytometry. B, Expression of B7-H3 and other potential target antigens for AML-directed CAR-T therapy (20) as assessed by flow cytometry on five human AML cell lines and on classical CD14+CD16 peripheral blood monocytes from normal donors. C, Representative flow cytometry plots demonstrating B7-H3 expression on primary AML blasts. D, B7-H3 expression on primary AML blasts from patients (n = 10) with monocytic AML stained with 376.96 or 7-517 mAbs (box plot indicates median and interquartile range). E, Expression of monocytic and myeloid markers (as assessed by flow cytometry) on the AML blasts and lymphocytes from the same cohort of patient samples. F and G, Expression of B7-H3 as assessed by flow cytometry staining with 7-517 mAb on the indicated hematopoietic progenitor populations in normal bone marrow. F, Gating strategy. G, Staining in the indicated populations with B7-H3 mAb 7-517 (black) versus isotype control (gray fill). H and I, Expression of B7-H3 as assessed by flow cytometry staining with mAb 376.96 on the indicated hematopoietic progenitor populations in normal bone marrow. H, Gating strategy. I, Staining in the indicated populations with B7-H3 mAb 376.96 (black) versus secondary antibody alone (gray fill). THP1 is shown as positive staining control. CMP, common myeloid progenitors; GMP, granulocyte-macrophage progenitors. Error bars indicate SEM.

Figure 1.

B7-H3 is expressed on monocytic AML cell lines and primary AML blasts obtained from patients with monocytic AML, but not on bone marrow progenitor cells. A, Expression of B7-H3 in four human monocytic AML cell lines stained with the 376.96 or 7-517 mAbs (black lines) versus isotype control (gray fill) as assessed by flow cytometry. B, Expression of B7-H3 and other potential target antigens for AML-directed CAR-T therapy (20) as assessed by flow cytometry on five human AML cell lines and on classical CD14+CD16 peripheral blood monocytes from normal donors. C, Representative flow cytometry plots demonstrating B7-H3 expression on primary AML blasts. D, B7-H3 expression on primary AML blasts from patients (n = 10) with monocytic AML stained with 376.96 or 7-517 mAbs (box plot indicates median and interquartile range). E, Expression of monocytic and myeloid markers (as assessed by flow cytometry) on the AML blasts and lymphocytes from the same cohort of patient samples. F and G, Expression of B7-H3 as assessed by flow cytometry staining with 7-517 mAb on the indicated hematopoietic progenitor populations in normal bone marrow. F, Gating strategy. G, Staining in the indicated populations with B7-H3 mAb 7-517 (black) versus isotype control (gray fill). H and I, Expression of B7-H3 as assessed by flow cytometry staining with mAb 376.96 on the indicated hematopoietic progenitor populations in normal bone marrow. H, Gating strategy. I, Staining in the indicated populations with B7-H3 mAb 376.96 (black) versus secondary antibody alone (gray fill). THP1 is shown as positive staining control. CMP, common myeloid progenitors; GMP, granulocyte-macrophage progenitors. Error bars indicate SEM.

Close modal

B7-H3 is expressed on the surface of primary AML blasts from patients with monocytic AML, but not in normal bone marrow progenitor populations

We obtained deidentified cryopreserved pretreatment bone marrow aspirates (n = 10) from patients with monocytic AML (see Fig. 1E for expression of myeloid and monocytic markers; see Supplementary Table S1 for clinical annotation associated with these samples) and demonstrated that blast populations (identified on the basis of CD45-SSC gating; Fig. 1C) had positive staining with the B7-H3–specific mAbs, 376.96 and 7-517 (Fig. 1D). B7-H3 expression varied across samples, with some samples displaying uniformly high B7-H3 expression.

With the caveat that B7-H3 is likely regulated at the posttranscriptional level (7), we also analyzed a previously published RNA sequencing dataset (21) including 200 patients with untreated, de novo AML. B7-H3 expression was higher in patients with monocytic AML [French-American-British (FAB) M5; P < 0.0001] and patients with acute promyelocytic leukemia (FAB M3 or documented PML-RARA fusion; P < 0.0001 and P = 0.0004, respectively), whereas B7-H3 expression was lower in FAB M2 AML (P < 0.0001; Supplementary Fig. S1A, S1C, and S1D). B7-H3 expression was higher in patients with mutations in NPM1 (P = 0.0224) and lower in patients with mutations in IDH1/IDH2 (P = 0.0460), CEBPA (P < 0.0001), and WT1 (P = 0.0001; Supplementary Fig. S1E). There was also a positive correlation of B7-H3 expression with CD33 (Supplementary Fig. S1F) and CD11b (Supplementary Fig. S1G) expression, and a negative correlation between the expression of B7-H3 and CD34 (Supplementary Fig. S1H). When stratified by B7-H3 z-score of <0 and ≥0, the median overall survival (mOS) was 26.3 and 11.8 months, respectively, although the difference was not significant (P = 0.2005; Supplementary Fig. S1I). When acute promyelocytic leukemia was excluded, mOS was 21.5 and 9.9 months (P = 0.0432; Supplementary Fig. S1J). In a separate dataset including samples from 562 patients with both newly diagnosed and previously treated AML (22), we again found significantly higher B7-H3 expression in patients with FAB M3 (P = 0.0026) and M5 (P = 0.0119) AML, and lower expression in patients with FAB M2 AML (P = 0.0018; Supplementary Fig. S1K). In the same dataset (22), we found higher B7-H3 expression in patients with relapsed versus untreated AML (P = 0.0281; Supplementary Fig. S1L).

We also evaluated the expression of B7-H3 on normal myeloid progenitor populations using cryopreserved normal bone marrow aspirates. Bone marrow aspirates were analyzed by flow cytometry using the gating strategy shown in Fig. 1F to identify the following hematopoietic progenitor populations: megakaryocyte-erythroid progenitors (MEP), common myeloid progenitors (CMP), granulocyte-macrophage progenitors (GMP), HSCs, and multipotent progenitor cells (MPP). We demonstrated no significant staining with the B7-H3–specific mAb 7-517 on any of these populations (Fig. 1G). Similarly, using the gating strategy shown in Fig. 1H, we evaluated the CD45+LineageCD34+CD38+ and CD45+LineageCD34+CD38 populations, and demonstrated no significant staining with the B7-H3–specific mAb 376.96 (Fig. 1I).

B7-H3.CAR-Ts target B7-H3+ AML cell lines and primary AML samples

Utilizing the scFv from the B7-H3 mAb 376.96 (13, 23), we have previously generated retroviral constructs encoding B7-H3.CAR-28 (incorporating the CD28 endodomain) and B7-H3.CAR-BB (incorporating the 4-1BB endodomain; ref. 11). Following activation with CD3/CD28, donor PBMCs were retrovirally transduced with the constructs mentioned above to generate B7-H3.CAR-Ts (Fig. 2A). CD19.CAR-28-Ts were used as a control. Median (range) transduction efficiencies for the B7-H3.CAR-28, B7-H3.CAR-BB, and CD19.CAR-28 constructs among eight representative donors were 77% (66%–87%), 70% (55%–87%), and 73% (65%–84%), respectively (Fig. 2B). The antileukemia efficacy of B7-H3.CAR-Ts was evaluated in coculture assays. B7-H3.CAR-Ts, but not control T cells, eliminated B7-H3+ cell lines (OCI-AML2, OCI-AML3, THP1, and U937; Fig. 2C and D). In the same experiments, we demonstrated significant levels of IFNγ (Fig. 2E) and IL2 (Fig. 2F) in the media harvested, following a 24-hour incubation, from cocultures with B7-H3.CAR-Ts, but not control T cells. To further assess antigen specificity of B7-H3.CAR-Ts, we stained B7-H3.CAR-Ts with CFSE and demonstrated B7-H3–specific proliferation of B7-H3.CAR-Ts (but not control T cells) in coculture with B7-H3+ cell lines (THP1 and U937), but not B7-H3 cell lines (Kasumi and HL60; Fig. 2G).

Figure 2.

B7-H3.CAR-Ts target B7-H3+ AML cell lines. A, Representative flow cytometry histograms indicating transduction efficiency of donor T cells with the indicated CAR constructs (empty fill) compared with nontransduced T cells (gray fill). B, Transduction efficiencies of donor T cells (n = 8) with each of the indicated CAR constructs. Representative flow cytometry plots of OCI-AML2, OCI-AML3, THP1, or U937 cell lines (CD3CD33+) cocultured with B7-H3.CAR-Ts or control T cells (CD3+CD33; C), and percentage of CD33+ tumor cells after 5–7 days in coculture (n = 3–5 donors; D). For all cell lines, P < 0.0001 for comparison of B7-H3.CAR-Ts with control T cells (one-way ANOVA with Holm–Sidak test adjusted P value). Summary of the concentration of IFNγ (E) and IL2 (F) in the coculture media after 24 hours as measured by ELISA (n = 3–5). G, Representative CFSE dilution assay demonstrating proliferation of B7-H3.CAR-Ts (dashed lines), compared with control T cells (solid lines with gray fill), when cocultured with THP1 or U937 cells (B7-H3+). No significant proliferation was observed when T cells were cocultured with Kasumi or HL60 cells (B7-H3). *, P < 0.05; **, P < 0.01; ***, P < 0.001, one-way ANOVA with Holm–Sidak test adjusted P value. Error bars indicate SEM. ns, not significant.

Figure 2.

B7-H3.CAR-Ts target B7-H3+ AML cell lines. A, Representative flow cytometry histograms indicating transduction efficiency of donor T cells with the indicated CAR constructs (empty fill) compared with nontransduced T cells (gray fill). B, Transduction efficiencies of donor T cells (n = 8) with each of the indicated CAR constructs. Representative flow cytometry plots of OCI-AML2, OCI-AML3, THP1, or U937 cell lines (CD3CD33+) cocultured with B7-H3.CAR-Ts or control T cells (CD3+CD33; C), and percentage of CD33+ tumor cells after 5–7 days in coculture (n = 3–5 donors; D). For all cell lines, P < 0.0001 for comparison of B7-H3.CAR-Ts with control T cells (one-way ANOVA with Holm–Sidak test adjusted P value). Summary of the concentration of IFNγ (E) and IL2 (F) in the coculture media after 24 hours as measured by ELISA (n = 3–5). G, Representative CFSE dilution assay demonstrating proliferation of B7-H3.CAR-Ts (dashed lines), compared with control T cells (solid lines with gray fill), when cocultured with THP1 or U937 cells (B7-H3+). No significant proliferation was observed when T cells were cocultured with Kasumi or HL60 cells (B7-H3). *, P < 0.05; **, P < 0.01; ***, P < 0.001, one-way ANOVA with Holm–Sidak test adjusted P value. Error bars indicate SEM. ns, not significant.

Close modal

To assess the activity of B7-H3.CAR-Ts against primary AML blasts, we cocultured B7-H3.CAR-Ts derived from healthy donors with primary AML blasts obtained from patients with monocytic AML. When cocultured at a 1:5 E:T ratio for 48 hours, B7-H3.CAR-Ts, but not control T cells, displayed significant cytotoxicity against primary monocytic AML blasts (Fig. 3A and B). We also demonstrated significant release of IFNγ and IL2 by B7-H3.CAR-Ts as measured in the media harvested from cocultures following a 24-hour incubation (Fig. 3C and D). The B7-H3 expression for the same patient samples is shown in Fig. 1D. B7-H3.CAR-Ts appeared to have more cytotoxic activity against AML blasts with higher B7-H3 expression, although this difference was not statistically significant (Supplementary Fig. S3). Given the shorter incubation time for these cocultures as compared with the cocultures with AML cell lines, shown in Fig. 2C–F, we demonstrated that under coculture conditions comparable with those used in Fig. 3A–D and H–K, healthy donor–derived B7-H3.CAR-Ts (n = 3) exhibited similar cytotoxicity (P < 0.0001) against the AML cell line, THP1 (Fig. 3M).

Figure 3.

B7-H3.CAR-Ts target primary AML blasts obtained from patients with monocytic AML. A, Representative flow cytometry plots of primary AML blasts (CD3CD33+) cocultured with donor-derived B7-H3.CAR-Ts or control T cells (CD3+CD33). B, Percentage of CD33+ cells after 48 hours in coculture of primary AML blasts (n = 10) with donor-derived (n = 2–3 donors) B7-H3.CAR-Ts or control T cells (P < 0.0001 for one-way ANOVA of means of replicates; P < 0.0001 for comparisons of both B7-H3.CAR-28-Ts and B7-H3.CAR-BB-Ts with control T cells using Holm-Sidak multiple comparison test). Summary of the concentration of IFNγ (P = 0.0059, one-way ANOVA of means of replicates; P = 0.0206 for comparison of B7-H3.CAR-28-Ts with control T cells and P = 0.0082 for comparison of B7-H3.CAR-BB-Ts with control T cells; Holm-Sidak multiple comparison test; C) and IL2 (P = 0.0031, one way ANOVA of means of replicates; P = 0.0156 for comparison of B7-H3.CAR-28-Ts with control T cells and P = 0.0064 for comparison of B7-H3.CAR-BB-Ts with control T cells; Holm-Sidak test adjusted P value; D) in the coculture media after 24 hours as measured by ELISA (n = 10 patient samples, T cells from 2–3 donors). E, B7-H3 expression, based on staining with the B7-H3 mAbs 376.96 (performed for three of the four samples) and 7-517, on primary AML blasts used for autologous cocultures summarized in H–K. F, Representative flow cytometry histograms (data included in G) indicating transduction efficiency of bone marrow–derived T cells (from patients with AML) with the indicated CAR constructs (empty fill) compared with nontransduced T cells (gray fill). G, Transduction efficiencies of patient bone marrow–derived T cells used in H–L. H, Representative flow cytometry plots of primary AML blasts (data included in I) cocultured with autologous patient-derived B7-H3.CAR-Ts or control T cells (CD3+CD33). Coculture of OCI-AML3 cells with the same CAR T cells is shown as a control. I, Percentage of CD33+ cells after 48 hours in coculture of primary AML blasts (n = 4) with autologous B7-H3.CAR-Ts or control T cells. IFNγ (J) and IL2 (K) in the media of the coculture experiments shown in H–I, as measured by ELISA after 24 hours (n = 4, P values as shown). L, Percentage of CD33+ cells after coculture of U937 cells (CD33+) with AML patient–derived (n = 3) B7-H3.CAR-Ts or control T cells for 48 hours. M, Percentage CD33+ cells after coculture of THP1 cells (CD33+) with healthy donor–derived B7-H3.CAR-Ts or control T cells for 48 hours. **, P < 0.01; ****, P < 0.0001, one-way ANOVA with Holm-Sidak test adjusted P value. Error bars indicate SEM. ns, not significant.

Figure 3.

B7-H3.CAR-Ts target primary AML blasts obtained from patients with monocytic AML. A, Representative flow cytometry plots of primary AML blasts (CD3CD33+) cocultured with donor-derived B7-H3.CAR-Ts or control T cells (CD3+CD33). B, Percentage of CD33+ cells after 48 hours in coculture of primary AML blasts (n = 10) with donor-derived (n = 2–3 donors) B7-H3.CAR-Ts or control T cells (P < 0.0001 for one-way ANOVA of means of replicates; P < 0.0001 for comparisons of both B7-H3.CAR-28-Ts and B7-H3.CAR-BB-Ts with control T cells using Holm-Sidak multiple comparison test). Summary of the concentration of IFNγ (P = 0.0059, one-way ANOVA of means of replicates; P = 0.0206 for comparison of B7-H3.CAR-28-Ts with control T cells and P = 0.0082 for comparison of B7-H3.CAR-BB-Ts with control T cells; Holm-Sidak multiple comparison test; C) and IL2 (P = 0.0031, one way ANOVA of means of replicates; P = 0.0156 for comparison of B7-H3.CAR-28-Ts with control T cells and P = 0.0064 for comparison of B7-H3.CAR-BB-Ts with control T cells; Holm-Sidak test adjusted P value; D) in the coculture media after 24 hours as measured by ELISA (n = 10 patient samples, T cells from 2–3 donors). E, B7-H3 expression, based on staining with the B7-H3 mAbs 376.96 (performed for three of the four samples) and 7-517, on primary AML blasts used for autologous cocultures summarized in H–K. F, Representative flow cytometry histograms (data included in G) indicating transduction efficiency of bone marrow–derived T cells (from patients with AML) with the indicated CAR constructs (empty fill) compared with nontransduced T cells (gray fill). G, Transduction efficiencies of patient bone marrow–derived T cells used in H–L. H, Representative flow cytometry plots of primary AML blasts (data included in I) cocultured with autologous patient-derived B7-H3.CAR-Ts or control T cells (CD3+CD33). Coculture of OCI-AML3 cells with the same CAR T cells is shown as a control. I, Percentage of CD33+ cells after 48 hours in coculture of primary AML blasts (n = 4) with autologous B7-H3.CAR-Ts or control T cells. IFNγ (J) and IL2 (K) in the media of the coculture experiments shown in H–I, as measured by ELISA after 24 hours (n = 4, P values as shown). L, Percentage of CD33+ cells after coculture of U937 cells (CD33+) with AML patient–derived (n = 3) B7-H3.CAR-Ts or control T cells for 48 hours. M, Percentage CD33+ cells after coculture of THP1 cells (CD33+) with healthy donor–derived B7-H3.CAR-Ts or control T cells for 48 hours. **, P < 0.01; ****, P < 0.0001, one-way ANOVA with Holm-Sidak test adjusted P value. Error bars indicate SEM. ns, not significant.

Close modal

We utilized cryopreserved bone marrow aspirates from patients (n = 4) with AML with variable B7-H3 expression (Fig. 3E) to generate B7-H3.CAR-Ts and activated control T cells. Median (range) transduction efficiencies of B7-H3.CAR-28 and B7-H3.CAR-BB were 56% (49%–68%) and 56% (46%–59%), respectively (Fig. 3F and G). Utilizing a second cryopreserved bone marrow aspirate sample from the same patient, we demonstrated that autologous B7-H3.CAR-28-Ts (P = 0.0085) and B7-H3.CAR-BB-Ts (P = 0.0085) both displayed significant cytotoxicity against primary AML blasts when cocultured at a 1:5 E:T ratio for 48 hours (Fig. 3H and I). On the basis of clinical annotation, the median blast percentage of the bone marrow aspirates used for these cocultures was 86% (range, 78%–92%). For coculture analysis, blasts were identified as CD45+/CD33+ cells. Release of IFNγ and IL2 by B7-H3.CAR-Ts and control T cells was measured in the media harvested from cocultures following a 24-hour incubation period. IFNγ and IL2 release was detected from B7-H3.CAR-Ts and was mostly undetectable in the media of control T cells (P = 0.0750 and P = 0.1524 for IFNγ and IL2, respectively; Fig. 3J and K). Under identical coculture conditions, we also demonstrated the ability of AML patient–derived (n = 3) B7-H3.CAR-28-Ts (P = 0.0049) and B7-H3.CAR-BB-Ts (P = 0.0049) to efficiently kill the AML cell line, U937 (Fig. 3L).

B7-H3.CAR-Ts show antitumor activity in xenograft mouse models of disseminated AML

We evaluated the in vivo efficacy of B7-H3.CAR-Ts in two murine models of disseminated AML as described above. In the first model, eGFP-FFLuc-OCI-AML2 cells (2 × 106 cells/mouse) were injected intravenously into NSG mice on day −7, followed by injection (1 × 107 cells/mouse) of B7-H3.CAR-28-Ts (n = 10), B7-H3.CAR-BB-Ts (n = 10), or CD19.CAR-28-Ts (n = 7) on days 0 and +20. Leukemia proliferation was assessed via weekly bioluminescence imaging. Tumor proliferation in mice treated with B7-H3.CAR-Ts was significantly delayed, as compared with mice treated with CD19.CAR-28-Ts (P < 0.0001; Fig. 4A–D). We elected to perform a second CAR-T injection in this model because of data from prior experiments in which a higher eGFP-FFLuc-OCI-AML2 dose (5 × 106 cells/mouse) was injected intravenously in NSG mice at day −7, followed by a single injection (1 × 107 cells/mouse) of B7-H3.CAR-28-Ts, B7-H3.CAR-BB-Ts, or CD19.CAR-28-Ts (n = 5 per group) at day 0 (Supplementary Fig. S4A–S4D). In this experiment, we observed a significant prolongation of survival (P = 0.0008) in B7-H3.CAR-T–treated mice, but all mice ultimately relapsed prior to day 40. We hypothesized that this may have been related to the kinetics of the tumor cell growth compared with CAR T-cell expansion in vivo, and that a second CAR T-cell injection could potentially overcome this limitation. In a pilot experiment, we also observed that in mice initially engrafted with eGFP-FFLuc-OCI-AML2 cells on day −7 (5 × 106 cells/mouse), followed by CD19.CAR-28-T injection (1 × 107 cells/mouse) on day 0, tumor growth could be controlled after injection of B7-H3.CAR-28-Ts (1 × 107 cells/mouse) on day 20 (Supplementary Fig. S4E–S4G).

Figure 4.

B7-H3.CAR-Ts control leukemia growth in xenograft models. A–D, OCI-AML2 cells labeled with eGFP-FFLuc were injected intravenously into NSG mice on day −7 followed by injection of B7-H3.CAR-Ts (n = 10 mice/construct, T cells from two donors) or CD19.CAR-28-Ts (n = 8 mice and two donors) on days 0 and 20. A, Representative tumor bioluminescence images at the specified timepoints. B, Experimental schema. C, Tumor bioluminescence intensity (BLI) of each mouse plotted over the course of the experiment. D, KaplanMeier survival curves indicating time to death or documented BLI greater than specified threshold. E–H, THP1 cells labeled with eGFP-FFLuc were injected intravenously into NSG mice on day −7, followed by injection of B7-H3.CAR-Ts (n = 10 mice/construct, T cells from two donors) or CD19.CAR-28-Ts (n = 8 mice and two donors) on day 0. E, Representative tumor bioluminescence images at the specified timepoints. F, Experimental schema. G, Tumor BLI of each mouse plotted over the course of the experiment. H, KaplanMeier survival curves indicating time to death or documented BLI greater than specified threshold. For D and H, P values indicate log-rank test. i.v., intravenous.

Figure 4.

B7-H3.CAR-Ts control leukemia growth in xenograft models. A–D, OCI-AML2 cells labeled with eGFP-FFLuc were injected intravenously into NSG mice on day −7 followed by injection of B7-H3.CAR-Ts (n = 10 mice/construct, T cells from two donors) or CD19.CAR-28-Ts (n = 8 mice and two donors) on days 0 and 20. A, Representative tumor bioluminescence images at the specified timepoints. B, Experimental schema. C, Tumor bioluminescence intensity (BLI) of each mouse plotted over the course of the experiment. D, KaplanMeier survival curves indicating time to death or documented BLI greater than specified threshold. E–H, THP1 cells labeled with eGFP-FFLuc were injected intravenously into NSG mice on day −7, followed by injection of B7-H3.CAR-Ts (n = 10 mice/construct, T cells from two donors) or CD19.CAR-28-Ts (n = 8 mice and two donors) on day 0. E, Representative tumor bioluminescence images at the specified timepoints. F, Experimental schema. G, Tumor BLI of each mouse plotted over the course of the experiment. H, KaplanMeier survival curves indicating time to death or documented BLI greater than specified threshold. For D and H, P values indicate log-rank test. i.v., intravenous.

Close modal

In the second model, eGFP-FFLuc–expressing THP1 cells (5 × 106 cells/mouse) were similarly injected intravenously into NSG mice on day −7, followed by intravenous injection (1 × 107 cells/mouse) of B7-H3.CAR-Ts (n = 10 mice/construct) or CD19.CAR-28-Ts (n = 8 mice). Tumor growth was similarly assessed via weekly bioluminescence imaging. Both B7-H3.CAR-28-Ts and B7-H3.CAR-BB-Ts, but not CD19.CAR-28-Ts, effectively controlled the growth OCI-AML2 cells in vivo (P = 0.0004; Fig. 4E–H). Surface B7-H3 expression was found to be preserved on eGFP-FFluc-THP1 cells isolated from mice that relapsed after treatment with B7-H3.CAR-Ts (Supplementary Fig. S2A and S2B).

B7-H3.CAR-Ts show antitumor activity in a PDX model of AML

To evaluate the activity of B7-H3.CAR-Ts against human primary AML blasts, we utilized PDX mouse models of AML. For these experiments, because of the limited availability of tumor cells, we conducted the experiment using only B7-H3.CAR-Ts carrying the CD28 costimulation. For the first PDX model, we initially confirmed B7-H3 expression on primary AML blasts from sample DFAM-6855 (99.1% B7-H3+ using mAb 7-517; Fig. 5A) and confirmed the in vitro activity of B7-H3.CAR-28-Ts against these cells (Fig. 5B). NSG mice were then injected (1 × 106 cells/mouse) on day −21 with primary AML blasts (Fig. 5C). After confirmation of engraftment (Fig. 5D), B7-H3.CAR-28-Ts (n = 7) or control T cells (n = 5) were injected (1 × 107 cells/mouse) on day 0. Mice were sacrificed on day 8 and peripheral blood, bone marrow, and spleens were harvested (Fig. 5C). Compared with mice treated with control T cells, we demonstrated significantly decreased huCD45+CD33+ AML blasts in the peripheral blood (P = 0.0016) and spleen (P = 0.007; Fig. 5E). Also, compared with control mice, we demonstrated significantly increased CD8+ T-cell expansion in the spleens of B7-H3-CAR-28-T–treated mice (Fig. 5F).

Figure 5.

B7-H3.CAR-Ts control leukemia growth in a PDX model of AML. A, Expression of B7-H3 on patient-derived AML blasts based on staining with mAb 7-517 (black lines) versus isotype control (gray fill). B, Patient-derived AML blasts were cocultured in vitro with B7-H3.CAR-Ts or control T cells. C, Unlabeled patient-derived AML blasts were injected into the lateral tail veins of 6- to 8-week-old NSG mice on day −21, followed by injection of B7-H3.CAR-28-Ts or control T cells at day 0. D, Engraftment in all mice was confirmed via peripheral blood flow cytometry analysis on day 0 (representative flow cytometry plots on left). E, On day 8, all mice were sacrificed, and the CD45+CD33+ cells in blood, bone marrow, and spleen, were quantified by flow cytometry. F, CD4+ and CD8+ cells were quantified in blood and spleen. G, Patient-derived AML blasts were labeled with eGFP-FFLuc, expanded in vivo, then injected intravenously into 6- to 8-week-old NSG mice on day −4, followed by injection of B7-H3.CAR-28-Ts (n = 7 mice) or CD19.CAR-28-Ts (n = 3 mice) on day 0. H, Representative tumor bioluminescence images at the specified timepoints. I, Tumor bioluminescence intensity (BLI) of each mouse plotted over time. For days 14, 17, 21, and 26, P < 0.0001, t test of BLI with Holm-Sidak test adjusted P values. J, KaplanMeier survival curves indicating time to death (P = 0.0027, log-rank test). Patient-derived AML blasts are from sample DFAM-6855 (PRoXe). For D–F, groups were compared using unpaired and nonparametric Mann–Whitney tests with two-tailed P values as shown. Error bars indicate SEM. i.v., intravenous.

Figure 5.

B7-H3.CAR-Ts control leukemia growth in a PDX model of AML. A, Expression of B7-H3 on patient-derived AML blasts based on staining with mAb 7-517 (black lines) versus isotype control (gray fill). B, Patient-derived AML blasts were cocultured in vitro with B7-H3.CAR-Ts or control T cells. C, Unlabeled patient-derived AML blasts were injected into the lateral tail veins of 6- to 8-week-old NSG mice on day −21, followed by injection of B7-H3.CAR-28-Ts or control T cells at day 0. D, Engraftment in all mice was confirmed via peripheral blood flow cytometry analysis on day 0 (representative flow cytometry plots on left). E, On day 8, all mice were sacrificed, and the CD45+CD33+ cells in blood, bone marrow, and spleen, were quantified by flow cytometry. F, CD4+ and CD8+ cells were quantified in blood and spleen. G, Patient-derived AML blasts were labeled with eGFP-FFLuc, expanded in vivo, then injected intravenously into 6- to 8-week-old NSG mice on day −4, followed by injection of B7-H3.CAR-28-Ts (n = 7 mice) or CD19.CAR-28-Ts (n = 3 mice) on day 0. H, Representative tumor bioluminescence images at the specified timepoints. I, Tumor bioluminescence intensity (BLI) of each mouse plotted over time. For days 14, 17, 21, and 26, P < 0.0001, t test of BLI with Holm-Sidak test adjusted P values. J, KaplanMeier survival curves indicating time to death (P = 0.0027, log-rank test). Patient-derived AML blasts are from sample DFAM-6855 (PRoXe). For D–F, groups were compared using unpaired and nonparametric Mann–Whitney tests with two-tailed P values as shown. Error bars indicate SEM. i.v., intravenous.

Close modal

In an additional experiment, the same patient-derived AML blasts were transduced to express eGFP-FFLuc and were injected intravenously (1 × 106 cells/mouse) into NSG mice on day −4, followed by injection (1 × 107 cells/mouse) of B7-H3.CAR-28-Ts (n = 7 mice) or CD19.CAR-28-Ts (n = 3 mice) on day 0 (Fig. 5G). Leukemia proliferation was assessed via weekly bioluminescence imaging. Compared with CD19.CAR-28-T–treated mice, B7-H3.CAR-28-T–treated mice experienced significantly delayed tumor growth (P < 0.0001; Fig. 5H and I) and significantly prolonged survival (P = 0.0027; Fig. 5J). B7-H3 expression was also found to be preserved on patient-derived AML blasts recovered from the spleen after relapse following treatment with B7-H3.CAR-28-Ts (Supplementary Fig. S2E).

In a second PDX model, we used patient-derived AML blasts from sample CBAM-44728 as described above. After confirming B7-H3 expression (Supplementary Fig. S5B), blasts were injected intravenously (8 × 105 cells/mouse) into NSG mice on day −4, followed by intravenous injection (1 × 107 cells/mouse) of B7-H3.CAR-28-Ts (n = 6 mice) or CD19.CAR-28-Ts (n = 5 mice) on day 0 (Supplementary Fig. S5C). All mice were sacrificed on day 63, followed by analysis of patient-derived AML blasts and CAR-Ts in the bone marrow, spleen, and/or blood. AML blasts were significantly decreased (undetectable in 4/6 mice; 2 mice with ≤100 blasts/femur) in the bone marrow of B7-H3.CAR-28-T- compared with CD19.CAR-28-T–treated mice (P = 0.0043) and undetectable in the spleens of B7-H3.CAR-28-T–treated mice (P = 0.0022; Supplementary Fig. S5D). Both B7-H3.CAR-28-Ts and CD19.CAR-28-Ts were detectable in the blood and/or spleens of all mice (Supplementary Fig. S5E).

B7-H3.CAR-Ts do not exhibit toxicity against normal myeloid progenitor populations in vitro and in vivo in a humanized mouse model

To evaluate the potential toxicity of B7-H3.CAR-Ts against normal myeloid progenitor populations, we first utilized an in vitro CFU assay. Umbilical cord blood–derived CD34+ cells were coincubated with B7-H3.CAR-28-Ts, CD19.CAR-28-Ts, or control T cells derived from donor PMBCs (n = 4 cord blood donors, each cocultured with T cells from two PBMC donors), followed by culture in semisolid methylcellulose medium. No significant differences were observed in the numbers of granulocyte-macrophage progenitor (GMP), common myeloid progenitor (CMP), or total colonies (P = 0.75, P = 0.73, and P = 0.96, respectively; Fig. 6A).

Figure 6.

B7-H3.CAR-Ts do not exhibit unacceptable toxicity against normal myeloid progenitor populations. A, Human umbilical cord blood–derived CD34+ cells were coincubated with B7-H3.CAR-28-Ts, CD19.CAR-28-Ts, or control T cells, followed by transfer to semisolid medium for 2 weeks, after which granulocyte-macrophage progenitor (GMP) and common myeloid progenitor (CMP) colonies were scored. No significant differences were observed between groups for either colony type (one-way ANOVA). B–H, Humanized mouse model to evaluate the effects of B7-H3.CAR-Ts on normal hematopoiesis. B, Schema demonstrating generation of humanized mouse model. NSG mice injected intrahepatically with human fetal liver–derived CD34+ cells. Following engraftment, subset of mice (n = 2) were sacrificed. Human CD3+ (huCD3+) cells were isolated from murine spleens and used to generate CAR-Ts. C, Flow cytometry histograms indicating transduction efficiency of B7-H3.CAR-28-Ts and CD19.CAR-28-Ts. D, Flow cytometry analysis of THP1 (B7-H3+) or BV173 (B7-H3) cells cocultured with B7-H3.CAR-28-Ts or control T cells (generated as shown in B) for 4 days. E, Humanized mice shown in B were injected intravenously with 5 × 106 (day 0) and 10 × 106 (day 16) B7-H3.CAR-28-Ts (n = 5), CD19.CAR-28-Ts (n = 5), or control (n = 4) T cells (normalized for transduction efficiency). Percentage (top) and absolute numbers (bottom) of human CD3+, CD14+, and CD33+ cells in the peripheral blood of humanized mice are shown at the indicated timepoints. Vertical dotted lines indicate injections of B7-H3.CAR-28-Ts, CD19.CAR-28-Ts, or control T cells as described in the text. F, Human CD3+, CD14+, and CD33+ cells in murine spleens at the conclusion of the experiment. G, Human CD3+, CD14+, and CD33+ cells in murine bone marrow. H, Human CD34+CD38+ and CD34+CD38 populations in murine bone marrow. *, P < 0.05, one-way ANOVA with Holm-Sidak test adjusted P value. Error bars indicate SEM. ns, not significant.

Figure 6.

B7-H3.CAR-Ts do not exhibit unacceptable toxicity against normal myeloid progenitor populations. A, Human umbilical cord blood–derived CD34+ cells were coincubated with B7-H3.CAR-28-Ts, CD19.CAR-28-Ts, or control T cells, followed by transfer to semisolid medium for 2 weeks, after which granulocyte-macrophage progenitor (GMP) and common myeloid progenitor (CMP) colonies were scored. No significant differences were observed between groups for either colony type (one-way ANOVA). B–H, Humanized mouse model to evaluate the effects of B7-H3.CAR-Ts on normal hematopoiesis. B, Schema demonstrating generation of humanized mouse model. NSG mice injected intrahepatically with human fetal liver–derived CD34+ cells. Following engraftment, subset of mice (n = 2) were sacrificed. Human CD3+ (huCD3+) cells were isolated from murine spleens and used to generate CAR-Ts. C, Flow cytometry histograms indicating transduction efficiency of B7-H3.CAR-28-Ts and CD19.CAR-28-Ts. D, Flow cytometry analysis of THP1 (B7-H3+) or BV173 (B7-H3) cells cocultured with B7-H3.CAR-28-Ts or control T cells (generated as shown in B) for 4 days. E, Humanized mice shown in B were injected intravenously with 5 × 106 (day 0) and 10 × 106 (day 16) B7-H3.CAR-28-Ts (n = 5), CD19.CAR-28-Ts (n = 5), or control (n = 4) T cells (normalized for transduction efficiency). Percentage (top) and absolute numbers (bottom) of human CD3+, CD14+, and CD33+ cells in the peripheral blood of humanized mice are shown at the indicated timepoints. Vertical dotted lines indicate injections of B7-H3.CAR-28-Ts, CD19.CAR-28-Ts, or control T cells as described in the text. F, Human CD3+, CD14+, and CD33+ cells in murine spleens at the conclusion of the experiment. G, Human CD3+, CD14+, and CD33+ cells in murine bone marrow. H, Human CD34+CD38+ and CD34+CD38 populations in murine bone marrow. *, P < 0.05, one-way ANOVA with Holm-Sidak test adjusted P value. Error bars indicate SEM. ns, not significant.

Close modal

To assess the effects of B7-H3.CAR-Ts on hematopoietic progenitors, we utilized a murine model of normal human hematopoiesis. Human fetal liver–derived CD34+ cells were engrafted in NSG mice (n = 16) as described above. When the median human CD45+ cell percentage was 47%, 2 mice were sacrificed and human T cells were isolated and used to generate B7-H3.CAR-28-Ts and CD19-CAR-28-Ts (transduction efficiencies 69% and 79%, respectively; Fig. 6B and C). B7-H3.CAR-28-T function was confirmed via in vitro coculture assay with the THP1 (B7-H3+) and BV173 (B7-H3) cell lines (Fig. 6D). The remaining mice (n = 14) were injected intravenously with either B7-H3.CAR-28-Ts (n = 5), CD19.CAR-28-Ts (n = 5), or control T cells (n = 4) at days 0 and 16 (5 × 106 cells/mouse at each timepoint). Human CD3+, CD33+, and CD14+ cells in the peripheral blood were assessed at baseline and every 7–9 days thereafter. Compared with mice treated with CD19.CAR-28-Ts and control T cells, mice treated with B7-H3.CAR-28-Ts had no significant decreases in the levels of circulating CD14+ or CD33+ cells (Fig. 6E). Mice were sacrificed at the conclusion of the experiment and spleen and bone marrow were evaluated via flow cytometry. Compared with mice treated with control T cells, absolute numbers of CD14+ and CD33+ cells were moderately decreased, but not eliminated, in the spleens of B7-H3.CAR-28-T–treated mice (P = 0.0196 and 0.0359, respectively; Fig. 6F). No significant differences in the level of CD14+ and CD33+ cells were detected in the bone marrow (Fig. 6G).

Myeloid progenitor populations were subsequently evaluated in the bone marrow by gating on live, huCD45+, lineage (CD3CD14CD16CD19CD20CD56), CD34+CD38+, and CD34+CD38 populations. Compared with mice treated with control T cells, there was a significant decrease in the percentage of CD34+CD38+ cells in mice treated with B7-H3.CAR-28-Ts (P = 0.0169). However, this effect did not appear to be specific to B7-H3.CAR-Ts as a similar decrease in this population was seen in mice treated with CD19.CAR-28-Ts (P = 0.0206); as discussed below, we hypothesize that this is related to nonspecific CAR-T activation (Fig. 6H). Apparent decreases in the percentage of CD34+CD38 cells in B7-H3.CAR-28-T- and CD19.CAR-28-T–treated mice were not statistically significant (P = 0.0705; Fig. 6H).

We have previously generated B7-H3–specific CAR-T cells, demonstrated antitumor efficacy in several solid tumor models, confirmed the absence of significant B7-H3 expression in most normal human tissues, and, by taking advantage of the cross-reactivity of B7-H3.CAR-Ts with the murine form of B7-H3, demonstrated a lack of evident toxicities in a syngeneic tumor model, further supporting the potential clinical translation of B7-H3.CAR-Ts (11). In this study, we investigated the potential of B7-H3 as a clinically relevant target antigen for AML-directed CAR-T therapy. We have demonstrated that B7-H3.CAR-Ts could represent a safe and effective therapy for a subset of patients with AML, and that such therapy is unlikely to cause unacceptable hematopoietic toxicity.

In addition to direct antitumor effects, it is also possible that B7-H3.CAR-Ts could augment adaptive immune responses in AML (7, 24, 25). B7-H3 expression on tumor cells, and on myeloid-derived suppressor cells in the tumor microenvironment, is likely to play an immunosuppressive role, and may drive immune escape in multiple cancer types (7, 24–27). Patients with solid tumor treated with an anti-B7-H3 mAb were shown to have significant posttreatment increases in T-cell receptor clonality (26). B7-H3 and other costimulatory ligands (e.g., PD-L1 and CD80) are also upregulated in patients with AML who relapse following allogeneic stem cell transplantation, suggesting that B7-H3 may contribute to impaired allorecognition by donor T cells in this setting (28).

B7-H3 is thus a promising therapeutic target for human cancers, and multiple clinical trials are evaluating anti-B7-H3 mAbs, including a bispecific antibody targeting B7-H3 and CD3 (7, 27–34). In this study, we have provided efficacy and additional safety data to support the clinical assessment B7-H3.CAR-Ts as a treatment for AML. Our data suggest that B7-H3.CAR-Ts could potentially overcome the prolonged hematopoietic toxicity that has limited the development of CAR-Ts targeting certain other AML-associated antigens (35, 36). In the humanized mouse model described above, compared with mice treated with control T cells, we observed a similar decrease in myeloid progenitor populations in mice treated with both B7-H3.CAR-28-Ts and CD19.CAR-28-Ts. We suspect that this is explained by nonspecific activation of the CAR-Ts, rather than antigen-specific toxicity against myeloid progenitors. Although plasma cytokine levels were not measured in this experiment, in a similar humanized mouse model, our group previously demonstrated significantly increased levels of IFNγ, TNFα, and IL6 in the plasma of mice treated with CD19.CAR-Ts as compared with control T cells (37). Our data, together with the lack of reported myeloid toxicity in patients infused with a B7-H3–specific mAb (33), suggest that targeting B7-H3 will be associated with a better safety profile compared with other pan-myeloid markers. Nonetheless, for patients enrolled in a phase I clinical study of B7-H3.CAR-Ts, it would be recommended to have the option of HSC rescue, if needed.

Although we did not observe B7-H3 antigen loss in cells isolated from relapsed mice in either our xenograft mouse models using AML cell lines or in the PDX models, it remains to be seen whether antigen escape will limit the clinical efficacy of B7-H3.CAR-Ts in patients with AML. It is also possible that increased adaptive immune responses could overcome this potential limitation. If antigen escape does occur, B7-H3 could still represent an attractive potential target antigen for use in a combinatorial approach. Finally, we recognize that this study also does not fully characterize the expression of B7-H3 on AML leukemic stem cell (LSC) populations. AML LSCs are most well defined in the approximately 75% of AML samples that are positive for CD34, and typically reside in the CD34+CD38 population of AML blasts. In patients with CD34 AML, the majority of LSCs are thought to reside in the CD34 population; however, LSCs in this subset of AML are less well studied (30). Indeed, recent studies evaluating potential LSC targets for CAR-T therapy in AML have focused only on the CD34+CD38 blast population (32). We have found that most B7-H3+ AMLs are negative for CD34 (which is consistent with a monocytic immunophenotype), and it is thus more challenging to reliably evaluate the LSC expression of B7-H3. It is notable, however, that B7-H3.CAR-Ts demonstrated in vitro cytotoxicity against patient-derived AML samples that were also shown to have the ability to reliably engraft in NSG mice. Nonetheless, in the absence of serial transfer experiments in vivo, it is not possible to definitively conclude whether there is differential activity of B7-H3.CAR-Ts against LSCs versus healthy hematopoietic progenitor cells.

The optimal choice of costimulatory domain for CAR-T therapy remains unclear, and likely differs depending on factors related to the tumor growth kinetics, target antigen characteristics, and the scFv used. CD28-containing CARs are known to activate faster, to produce larger magnitude changes in protein phosphorylation, and to lead to a more effector T-cell–like phenotype as compared with 4-1BB–containing CARs (31). Prior studies suggest that 4-1BB–containing CARs may be better able to maintain effector functions in the setting of chronic antigen stimulation (31). Reduced levels of cytokine production with 4-1BB–containing CARs may also correlate with a lower potential for cytokine release syndrome or neurotoxicity (31), and the only FDA-approved CAR-T therapy for ALL incorporates the 4-1BB signaling domain. Prior work by our group in a pancreatic adenocarcinoma model has shown that 4-1BB–containing B7-H3.CAR-Ts expressed lower levels of PD-1 and showed greater antitumor efficacy against pancreatic ductal adenocarcinoma (PDAC) cell lines transduced to constitutively express PD-L1 (11). However, the kinetics of PDAC tumor growth are clearly different than that of AML, and these advantages may not necessarily translate to the treatment of AML. In this study, most experiments were designed to evaluate B7-H3.CAR-28-Ts and B7-H3.CAR-BB-Ts in parallel. Although there was a trend toward greater efficacy of B7-H3.CAR-28-Ts, our data generally suggest similar efficacy between the two constructs, both in vitro and in mouse xenograft models. Any potential advantage of B7-H3.CAR-28-Ts is likely explained by the rapid kinetics of AML cell line expansion in the model systems used. While the optimal costimulatory domain remains unclear and may depend, in part, on the burden of disease at the time of CAR-T infusion, we have elected to pursue B7-H3.CAR-28-Ts for initial clinical evaluation in AML.

In conclusion, our data support further evaluation of B7-H3 as a target for AML-directed CAR-T therapy, demonstrate that B7-H3.CAR-Ts are able to control the proliferation of B7-H3+ AML both in vitro and in xenograft models, and with the limitations discussed above, suggest that B7-H3.CAR-Ts are unlikely to cause significant hematopoietic toxicity.

E.I. Lichtman reports grants from ASCO/Conquer Cancer Foundation, NIH/NCI, and North Carolina University Cancer Research Fund and other from NIH/NCI during the conduct of the study; E.I. Lichtman served (without compensation) on an advisory board for Seattle Genetics. H. Du reports a patent for B7-H3–specific CAR issued, licensed, and with royalties paid from bluebird Bio. B. Savoldo reports personal fees and other from Tessa Therapeutics and grants from bluebird bio, NIH, and Cell Medica outside the submitted work. G. Dotti reports grants from Cell Medica, Bellicum Pharmaceutical, and bluebird bio and personal fees from Catamaran, Bellicum Pharmaceutical, and Tessa Therapeutics outside the submitted work, and has been issued a patent on the B7-H3.CAR. No disclosures were reported by the other authors.

E.I. Lichtman: Conceptualization, resources, data curation, formal analysis, supervision, funding acquisition, validation, investigation, visualization, methodology, writing–original draft, writing–review and editing. H. Du: Conceptualization, investigation, methodology, writing–review and editing. P. Shou: Conceptualization, investigation, methodology, writing–review and editing. F. Song: Investigation, methodology. K. Suzuki: Methodology. S. Ahn: Investigation, methodology, writing–review and editing. G. Li: Investigation, methodology, writing–review and editing. S. Ferrone: Resources, methodology, writing–review and editing. L. Su: Resources, methodology, writing–review and editing. B. Savoldo: Conceptualization, resources, supervision, investigation, methodology, writing–review and editing. G. Dotti: Conceptualization, resources, data curation, supervision, funding acquisition, methodology, writing–review and editing.

This work was partially supported by the University Cancer Research Fund at the University of North Carolina (to G. Dotti). E.I. Lichtman was supported by a Conquer Cancer Foundation of the American Society of Clinical Oncology Young Investigator Award and NCI grant 1T32CA211056-01A1 (to principal investigator, Jonathan Serody). S. Ferrone was supported by W81XWH-16-1-0500DOD, R01DE028172, and R03CA223886. The small-animal imaging core and flow cytometry core facilities were supported, in part, by an NCI Cancer Core grant (P30-CA016086-40). Cryopreserved human AML samples were provided by the Tissue Procurement Facility at UNC-LCCC.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
De Kouchkovsky
I
,
Abdul-Hay
M
. 
‘Acute myeloid leukemia: a comprehensive review and 2016 update’
.
Blood Cancer J
2016
;
6
:
e441
.
2.
Lichtman
EI
,
Dotti
G
. 
Chimeric antigen receptor T-cells for B-cell malignancies
.
Transl Res
2017
;
187
:
59
82
.
3.
Gill
S
. 
Chimeric antigen receptor T cell therapy in AML: how close are we? Best practice & research
Best Pract Res Clin Haematol
2016
;
29
:
329
33
.
4.
Steinberger
P
,
Majdic
O
,
Derdak
SV
,
Pfistershammer
K
,
Kirchberger
S
,
Klauser
C
, et al
Molecular characterization of human 4Ig-B7-H3, a member of the B7 family with four Ig-like domains
.
J Immunol
2004
;
172
:
2352
9
.
5.
Leitner
J
,
Klauser
C
,
Pickl
WF
,
Stockl
J
,
Majdic
O
,
Bardet
AF
, et al
B7-H3 is a potent inhibitor of human T-cell activation: no evidence for B7-H3 and TREML2 interaction
.
Eur J Immunol
2009
;
39
:
1754
64
.
6.
Hashiguchi
M
,
Kobori
H
,
Ritprajak
P
,
Kamimura
Y
,
Kozono
H
,
Azuma
M
. 
Triggering receptor expressed on myeloid cell-like transcript 2 (TLT-2) is a counter-receptor for B7-H3 and enhances T cell responses
.
Proc Natl Acad Sci U S A
2008
;
105
:
10495
500
.
7.
Picarda
E
,
Ohaegbulam
KC
,
Zang
X
. 
Molecular pathways: targeting B7-H3 (CD276) for human cancer immunotherapy
.
Clin Cancer Res
2016
;
22
:
3425
31
.
8.
Guery
T
,
Roumier
C
,
Berthon
C
,
Renneville
A
,
Preudhomme
C
,
Quesnel
B
. 
B7-H3 protein expression in acute myeloid leukemia
.
Cancer Med
2015
;
4
:
1879
83
.
9.
Dobrowolska
H
,
Gill
KZ
,
Serban
G
,
Ivan
E
,
Li
Q
,
Qiao
P
, et al
Expression of immune inhibitory receptor ILT3 in acute myeloid leukemia with monocytic differentiation
.
Cytometry B Clin Cytom
2013
;
84
:
21
9
.
10.
Dohner
H
,
Estey
EH
,
Amadori
S
,
Appelbaum
FR
,
Buchner
T
,
Burnett
AK
, et al
Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet
.
Blood
2010
;
115
:
453
74
.
11.
Du
H
,
Hirabayashi
K
,
Ahn
S
,
Kren
NP
,
Montgomery
SA
,
Wang
X
, et al
Antitumor responses in the absence of toxicity in solid tumors by targeting B7-H3 via chimeric antigen receptor T Cells
.
Cancer Cell
2019
;
35
:
221
237
.
12.
Hoyos
V
,
Savoldo
B
,
Quintarelli
C
,
Mahendravada
A
,
Zhang
M
,
Vera
J
, et al
Engineering CD19-specific T lymphocytes with interleukin-15 and a suicide gene to enhance their anti-lymphoma/leukemia effects and safety
.
Leukemia
2010
;
24
:
1160
70
.
13.
Imai
K
,
Wilson
BS
,
Bigotti
A
,
Natali
PG
,
Ferrone
S
. 
A 94,000-dalton glycoprotein expressed by human melanoma and carcinoma cells
.
J Natl Cancer Inst
1982
;
68
:
761
9
.
14.
Dotti
G
,
Savoldo
B
,
Pule
M
,
Straathof
KC
,
Biagi
E
,
Yvon
E
, et al
Human cytotoxic T lymphocytes with reduced sensitivity to Fas-induced apoptosis
.
Blood
2005
;
105
:
4677
84
.
15.
Krenciute
G
,
Krebs
S
,
Torres
D
,
Wu
MF
,
Liu
H
,
Dotti
G
, et al
Characterization and functional analysis of scfv-based chimeric antigen receptors to redirect T cells to IL13Rα2-positive glioma
.
Mol Ther
2016
;
24
:
354
63
.
16.
Arber
C
,
Feng
X
,
Abhyankar
H
,
Romero
E
,
Wu
MF
,
Heslop
HE
, et al
Survivin-specific T cell receptor targets tumor but not T cells
.
J Clin Invest
2015
;
125
:
157
68
.
17.
Vera
J
,
Savoldo
B
,
Vigouroux
S
,
Biagi
E
,
Pule
M
,
Rossig
C
, et al
T lymphocytes redirected against the kappa light chain of human immunoglobulin efficiently kill mature B lymphocyte-derived malignant cells
.
Blood
2006
;
108
:
3890
7
.
18.
Pule
MA
,
Straathof
KC
,
Dotti
G
,
Heslop
HE
,
Rooney
CM
,
Brenner
MK
. 
A chimeric T cell antigen receptor that augments cytokine release and supports clonal expansion of primary human T cells
.
Mol Ther
2005
;
12
:
933
41
.
19.
Li
G
,
Cheng
M
,
Nunoya
J
,
Cheng
L
,
Guo
H
,
Yu
H
, et al
Plasmacytoid dendritic cells suppress HIV-1 replication but contribute to HIV-1 induced immunopathogenesis in humanized mice
.
PLoS Pathog
2014
;
10
:
e1004291
.
20.
Perna
F
,
Berman
SH
,
Soni
RK
,
Mansilla-Soto
J
,
Eyquem
J
,
Hamieh
M
, et al
Integrating proteomics and transcriptomics for systematic combinatorial chimeric antigen receptor therapy of AML
.
Cancer Cell
2017
;
32
:
506
19
.
21.
Ley
TJ
,
Miller
C
,
Ding
L
,
Raphael
BJ
,
Mungall
AJ
,
Robertson
A
, et al
Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia
.
N Engl J Med
2013
;
368
:
2059
74
.
22.
Tyner
JW
,
Tognon
CE
,
Bottomly
D
,
Wilmot
B
,
Kurtz
SE
,
Savage
SL
, et al
Functional genomic landscape of acute myeloid leukaemia
.
Nature
2018
;
562
:
526
31
.
23.
Kasten
BB
,
Arend
RC
,
Katre
AA
,
Kim
H
,
Fan
J
,
Ferrone
S
, et al
B7-H3-targeted (212)Pb radioimmunotherapy of ovarian cancer in preclinical models
.
Nucl Med Biol
2017
;
47
:
23
30
.
24.
Chen
C
,
Shen
Y
,
Qu
QX
,
Chen
XQ
,
Zhang
XG
,
Huang
JA
. 
Induced expression of B7-H3 on the lung cancer cells and macrophages suppresses T-cell mediating anti-tumor immune response
.
Exp Cell Res
2013
;
319
:
96
102
.
25.
Lee
YH
,
Martin-Orozco
N
,
Zheng
P
,
Li
J
,
Zhang
P
,
Tan
H
, et al
Inhibition of the B7-H3 immune checkpoint limits tumor growth by enhancing cytotoxic lymphocyte function
.
Cell Res
2017
;
27
:
1034
45
.
26.
Powderly
J
,
Cote
G
,
Flaherty
K
,
Szmulewitz
R
,
Ribas
A
,
Weber
J
, et al
Interim results of an ongoing phase I, dose escalation study of MGA271 (Fc-optimized humanized anti-B7-H3 monoclonal antibody) in patients with refractory B7-H3-expressing neoplasms or neoplasms whose vasculature expresses B7-H3
.
J Immunotherapy Cancer
2015
;
3
:
O8
.
27.
Rizvi
N
,
Loo
D
,
Baughman
J
,
Yu
S
,
Chen
F
,
Moore
P
, et al
A phase I study of enoblituzumab in combination with pembrolizumab in patients with advanced B7-H3-expressing cancers
.
J Clin Oncol
34
:
15s
, 
2016
(
suppl; abstr TPS3104
).
28.
Toffalori
C
,
Zito
L
,
Gambacorta
V
,
Riba
M
,
Oliveira
G
,
Bucci
G
, et al
Immune signature drives leukemia escape and relapse after hematopoietic cell transplantation
.
Nat Med
2019
;
25
:
603
11
.
29.
Pollyea
DA
,
Jordan
CT
. 
Therapeutic targeting of acute myeloid leukemia stem cells
.
Blood
2017
;
129
:
1627
35
.
30.
Thomas
D
,
Majeti
R
. 
Biology and relevance of human acute myeloid leukemia stem cells
.
Blood
2017
;
129
:
1577
85
.
31.
Salter
AI
,
Ivey
RG
,
Kennedy
JJ
,
Voillet
V
,
Rajan
A
,
Alderman
EJ
, et al
Phosphoproteomic analysis of chimeric antigen receptor signaling reveals kinetic and quantitative differences that affect cell function
.
Sci Signal
2018
;
11
:
eaat6753
.
32.
Haubner
S
,
Perna
F
,
Kohnke
T
,
Schmidt
C
,
Berman
S
,
Augsberger
C
, et al
Coexpression profile of leukemic stem cell markers for combinatorial targeted therapy in AML
.
Leukemia
2019
;
33
:
64
74
.
33.
Powderly
J
,
Cote
G
,
Flaherty
K
,
Szmulewitz
R
,
Ribas
A
,
Weber
J
, et al
Interim results of an ongoing phase I, dose escalation study of MGA271 (Fc-optimized humanized anti-B7-H3 monoclonal antibody) in patients with refractory B7-H3-expressing neoplasms or neoplasms whose vasculature expresses B7-H3
.
J Immunother Cancer
2015
;
3
:
O8
.
34.
Tolcher
A
,
Alley
E
,
Chichili
G
,
Baughman
J
,
Moore
P
,
Bonvini
E
, et al
Phase 1, first-in-human, open label, dose escalation study of MGD009, a humanized B7-H3 x CD3 dual-affinity re-targeting (DART) protein in patients with B7-H3-expressing neoplasms or B7-H3 expressing tumor vasculature
.
J Clin Oncol
34
:
15s
, 
2016
(
suppl; abstr TPS3105
).
35.
Kenderian
SS
,
Ruella
M
,
Shestova
O
,
Klichinsky
M
,
Aikawa
V
,
Morrissette
JJ
, et al
CD33-specific chimeric antigen receptor T cells exhibit potent preclinical activity against human acute myeloid leukemia
.
Leukemia
2015
;
29
:
1637
47
.
36.
Gill
S
,
Tasian
SK
,
Ruella
M
,
Shestova
O
,
Li
Y
,
Porter
DL
, et al
Preclinical targeting of human acute myeloid leukemia and myeloablation using chimeric antigen receptor-modified T cells
.
Blood
2014
;
123
:
2343
54
.
37.
Diaconu
I
,
Ballard
B
,
Zhang
M
,
Chen
Y
,
West
J
,
Dotti
G
, et al
Inducible caspase-9 selectively modulates the toxicities of CD19-specific chimeric antigen receptor-modified T Cells
.
Mol Ther
2017
;
25
:
580
92
.