All-trans retinoic acid and interferon-α increase CD38 expression on adult T-cell leukemia cells and sensitize them to T cells bearing anti-CD38 chimeric antigen receptors

نویسندگان

  • K Mihara
  • T Yoshida
  • S Ishida
  • Y Takei
  • A Kitanaka
  • K Shimoda
  • K Morishita
  • Y Takihara
  • T Ichinohe
چکیده

Survival of patients with adult T-cell leukemia (ATL), which is caused by human T-cell lymphotropic virus type-1 (HTLV-1), has been improved by the introduction of anti-CCR4 monoclonal antibody and allogeneic hematopoietic stem cell transplantation. However, not all patients benefit from these modalities, necessitating a novel therapeutic strategy. Recently, an adoptive T-cell immunotherapy with chimeric antigen receptor (CAR) is clinically promising for patients with refractory blood diseases. Thus, CD38 is an attractive target of CAR therapy for lymphoid neoplasms because it is widely expressed on cells of Band T-lymphoid malignancies. We previously demonstrated marked cytotoxicity of T cells engineered to express anti-CD38-CAR against B-lymphoma cells and myeloma cells expressing CD38. To expand anti-CD38-CAR applicability against ATL cells that usually express undetectable or low CD38 levels, we must induce CD38 on the ATL cell surface. Interestingly, all-trans retinoic acid (ATRA), which is clinically used to treat patients with acute promyelocytic leukemia (APL), enhances CD38 expression on HL60 cells. Moreover, the upstream sequence of the CD38 gene contains an interferon regulatory factor-1 (IRF-1)-binding site. Here we show the marked cytotoxicity of anti-CD38-CAR T cells in HTLV-1-transformed cell lines as well as in cells from patients with ATL through the induction of CD38 expression by treatment with both ATRA and interferon (IFN)-α. HTLV-1-transformed cell lines MT-2, MT-4, S1T, Su9T and ED were from Miyazaki University. Hut102 cells were obtained from the Cell Research Center for Biomedical Research (Sendai, Japan). Cells were cultured in RPMI-1640 medium containing 10% fetal calf serum and L-glutamine (Sigma, St Louis, MO, USA). ATL cells (acute type) from bone marrow, accounting for over 65% of mononuclear cells and peripheral blood, were provided after obtaining informed consent. These ATL cells and donors’ cells were examined for approval by the institutional review board of Hiroshima University. A retroviral vector consisting of green fluorescent protein (GFP), CD8α, and 4-1BB, CD3ζ and anti-CD38 scFv was previously developed. Peripheral blood mononuclear cells were stimulated for 48 h with 7 μg/ml PHA-M (Sigma) and 200 IU/ml interleukin-2 (PeproTech, London, UK). T cells were transduced in an RD114-pseudotyped retrovirus-containing medium with 4 μg/ml polybrene (Sigma) in a retronectin-coated tube (Takara-Bio, Otsu, Japan) by spinoculation. An anti-CD38 antibody (CPK-H; MBL, Nagoya, Japan) was added to protect transduced T cells from autolysis through cross-linkage of anti-CD38-CAR with intrinsic CD38, as described previously. To detect anti-CD38-CAR, cells were stained with a goat anti-mouse (Fab’)2 antibody-biotin (Jackson ImmunoResearch, West Grove, PA, USA), followed by PerCP–streptavidin (BD, Franklin Lakes, NJ, USA). Antibody staining was detected using a FACSCalibur flow cytometer (BD). For lactate dehydrogenase (LDH)-releasing cytotoxicity assay, cells (1 × 10 cells per ml) were incubated with transduced T cells (1 × 10 cells per ml) for 18 h at 37 °C in Opti-MEM medium (Invitrogen, Carlsbad, CA, USA). Solution containing tetrazolium salt and diaphorase was added to the supernatant collected before measuring absorbance using the LDH Cytotoxicity Detection Kit (Takara-Bio). To evaluate cytotoxicity of anti-CD38-CAR T cells, co-cultured cells were collected and stained with an antiCD38 antibody-APC (BD). The specific cytotoxicity of anti-CD38-CAR T cells against CD38 ATL cells treated with ATRA (Sigma) and/or IFNα (PeproTech) was evaluated by flow cytometry. We first examined anti-CD38-CAR expression on retrovirally transduced human T cells from healthy donors using goat antimouse-IgG-PerCP, which cross-reacts with CAR. We confirmed that PerCP and GFP contained in the vector were co-expressed in transduced T cells (transduction efficiency: 61.26±10.66% (n=5)). Next, we investigated whether patients’ ATL cells could be transduced with anti-CD38-CAR. GFP-positive T cells were negative for CD4 and CD25, indicating that ATL cells were not transduced with anti-CD38-CAR (Figure 1a). These results agreed with a previous observation that CD8 T cells were markedly expanded and transduced with our methods. The transduction efficiency was 40.31±2.40% (n=4). Next, we evaluated the cytotoxicity of transduced T cells using the LDH releasing assay by co-incubating anti-CD38-CAR T cells with HTLV-1-transformed cell lines. As expected, MT-2 cells, with expression of CD38 being the highest among the six cell lines tested (97.06%), were efficiently abrogated by anti-CD38-CAR T cells (75.36±0.11% (n=3); Table 1). However, the other HTLV-I-transformed cell lines (MT-4, S1T, Hut102, Su9T and ED) lacking CD38 expression mostly survived after co-incubation with anti-CD38-CAR T cells (Table 1). Therefore, augmentation of CD38 expression was required to induce anti-CD38-CAR T-cell cytotoxicity against HTLV-1-transformed cell lines. We investigated whether ATRA enhanced cytotoxicity of anti-CD38CAR T cells by inducing CD38 expression on HTLV-1-transformed cell lines. As little as 10 nM of ATRA compared with an effective blood concentration for treating patients with APL, increased CD38 expression by over 80% in MT-4, S1T and Hut102 cells, but not in Su9T and ED cells (Figure 1b; Supplementary Figure 1a; Table 1). Three-day co-incubation of anti-CD38-CAR T cells with these cell lines at an effector (E): target (T) ratio of 1:2 in ATRA presence resulted in efficient elimination of MT-4, S1T and Hut102 cells according to the increased levels of CD38 expression (Figure 1b; Table 1). Cytotoxicity against cell lines was dependent on the number of T cells with anti-CD38-CAR in ATRA presence (Supplementary Figures 1b and c). Alternatively, ATRA withdrawal led to the basal level of CD38 expression of MT-4 cells before ATRA administration for 10 days (data not shown). However, CD38 induction by ATRA was insufficient to completely eliminate HTLV-1-transformed cells because 10–20% of S1T and Hut102 cells did not express CD38 in ATRA presence. To further improve the killing of HTLV-1-transformed cells and primary ATL cells by anti-CD38-CAR T cells through induced CD38 expression, we examined whether IFN-α and/or IFN-γ could enhance expression of the CD38 gene, whose upstream contains binding sites for IRF-1. IFN-α and IFN-γ efficiently enhanced CD38 expression in MT-4 cells even at a concentration below the therapeutic level, but not in Su9T, ED or S1T cells (Supplementary Figures 1a and c). As low as 2.5 U/ml of IFN-α induced CD38 expression in MT-4 cells for 18 h Citation: Blood Cancer Journal (2016) 6, e421; doi:10.1038/bcj.2016.30

برای دانلود متن کامل این مقاله و بیش از 32 میلیون مقاله دیگر ابتدا ثبت نام کنید

ثبت نام

اگر عضو سایت هستید لطفا وارد حساب کاربری خود شوید

منابع مشابه

All-trans retinoic acid enhances cytotoxic effect of T cells with an anti-CD38 chimeric antigen receptor in acute myeloid leukemia

We reported that T cells with anti-CD38-chimeric antigen receptors (CAR) eliminated B-cell lymphoma cells expressing CD38. To employ anti-CD38-CAR against acute myeloid leukemia (AML) blasts not expressing CD38, it is necessary to induce or increase the intensity of CD38 expression. A lactate dehydrogenase (LDH)-releasing assay and flow cytometry showed that anti-CD38-CAR T cells were cytotoxic...

متن کامل

T cells bearing anti-CD19 and/or anti-CD38 chimeric antigen receptors effectively abrogate primary double-hit lymphoma cells

Patients with B cell lymphomas bearing MYC translocation combined with translocation involving other genes, such as BCL2, BCL3, or BCL6, defined as double-hit lymphoma (DHL), have a poor prognosis. Recent studies expanded the concept to include double-expressing lymphoma (DEL) that co-overexpresses MYC protein with either of those proteins. Accordingly, we defined cytogenetic DHL and DEL as pri...

متن کامل

Pre-clinical evaluation of CD38 chimeric antigen receptor engineered T cells for the treatment of multiple myeloma.

Adoptive transfer of chimeric antigen receptor-transduced T cells is a promising strategy for cancer immunotherapy. The CD38 molecule, with its high expression on multiple myeloma cells, appears a suitable target for antibody therapy. Prompted by this, we used three different CD38 antibody sequences to generate second-generation retroviral CD38-chimeric antigen receptor constructs with which we...

متن کامل

Retinoic acid-induced CD38 antigen as a target for immunotoxin-mediated killing of leukemia cells.

A major obstacle in the successful delivery of antibody-based therapeutics to tumor cells is the heterogeneity of target antigen expression. We reported previously that retinoic acid (RA) is a potent and selective inducer of the cell-surface antigen CD38 in myeloid leukemia cells. The purpose of this study was to determine whether the RA-induced CD38 antigen could be a target for an anti-CD38-b...

متن کامل

T-cell immunotherapy with a chimeric receptor against CD38 is effective in eradicating chemotherapy-resistant B-cell lymphoma cells overexpressing survivin induced by BMI-1

The expression of BMI-1 (B lymphoma Mo-MLV insertion region 1 homolog), a member of the polycomb-group genes (PcG), is well correlated with a poor prognosis and treatment failure among patients with malignancies such as myelodysplastic syndrome, chronic myeloid leukemia, acute myeloid leukemia and lymphoma. Recently, we found that BMI-1 renders B-cell lymphoma cells refractory to several anti-c...

متن کامل

ذخیره در منابع من


  با ذخیره ی این منبع در منابع من، دسترسی به آن را برای استفاده های بعدی آسان تر کنید

برای دانلود متن کامل این مقاله و بیش از 32 میلیون مقاله دیگر ابتدا ثبت نام کنید

ثبت نام

اگر عضو سایت هستید لطفا وارد حساب کاربری خود شوید

عنوان ژورنال:

دوره 6  شماره 

صفحات  -

تاریخ انتشار 2016