Adult T-Cell Leukemia/Lymphoma Cells Were Eliminated By T Cells With Anti-CD38 Chimeric Antigen Receptor Through CD38 Expression Induced By All-Trans Retinoic Acid

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1824-1824
Author(s):  
Tetsumi Yoshida ◽  
Keichiro Mihara ◽  
Akira Kitanaka ◽  
Kazuya Shimoda ◽  
Kazuhiro Morishita ◽  
...  

Abstract Patients with adult T-cell leukemia and lymphoma (ATLL) often succumb to death even though multi-anti-cancer drugs are used. Thus, it is essential for establishing a novel therapeutic strategy for ATLL. We have previously developed a chimeric antigen receptor against CD38 (anti-CD38-CAR) and showed powerful cytotoxicity of anti-CD38-CAR to B-cell lymphoma cells as well as to myeloma cells expressing CD38. Unfortunately, as CD38 is poorly expressed on the cell surface of ATLL cells, it is required to induce CD38 to apply our anti-CD38-CAR. Here, we investigated cytotoxicity of T cells transduced with anti-CD38-CAR against ATLL cell lines and cells obtained from ATLL patients through CD38 induction by all-trans retinoic acid (ATRA), which is clinically available for acute promyelocytic leukemia. We evaluated an effect of ATRA on cytotoxicity of T cells bearing anti-CD38-CAR against ATLL cells through flow cytometry. We firstly confirmed the expression of anti-CD38-CAR on human T cells retrovirally transduced (10-70%). Then, secondly, we prepared ATLL cell lines (MT-2, MT-4, S1T, Hut102, and Su9T: >95%, <5%, <5%, 15%, and <5% at CD38 expression, respectively). We co-incubated CD38-specific T cells with ATLL cell line cells for 3 days. MT-2 cells were entirely abrogated by T cells harboring anti-CD38-CAR. However, others were restrictedly succumbed to death after 3-day co-culture with T cells carrying anti-CD38-CAR. Next, we investigated whether ATRA could enhance CD38 expression on the cell surface of ATLL cell lines and exert a cytotoxicity of T cells with anti-CD38-CAR. Intriguingly, even 10nM of ATRA augmented CD38 expression in MT-4, S1T, and Hut102 cells (>80%), but not in Su9T cells. Co-culture experiments in the presence of ATRA showed that MT-4, S1T, and Hut102 but Su9T cells were efficiently eliminated by T cells bearing anti-CD38-CAR, leading to a positive correlation of cytotoxicity with CD38 expression level. We tested whether ATLL cells obtained from 3 patients were disrupted by T cells bearing anti-CD38-CAR. CD38 was expressed in the cells from patients at a variety of expression ratio (0-30%). Intriguingly, CD38 expression was significantly enhanced in ATLL cells from 2 of 3 individual patients with ATRA (>50%). And resultantly, T cells bearing anti-CD38-CAR exerted more powerful cytotoxicity against ATLL cells with CD38 enhanced by ATRA (cytotoxicity of T cells with anti-CD38-CAR in CD38-positive ATLL fraction: >90%). ATRA exerted enhancing effect on the cytotoxicity of T cells bearing anti-CD38-CAR against ATLL cells through the augmentation of CD38 expression. These results may provide us a rationale for novel clinical settings of T cells carrying anti-CD38-CAR on patients with ATLL using ATRA. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 591-591
Author(s):  
Keichiro Mihara ◽  
Tetsumi Yoshida ◽  
Seiko Ishida ◽  
Yoshifumi Takei ◽  
Akira Kitanaka ◽  
...  

Abstract The survival of patients with adult T-cell leukemia (ATL) has been improved by the introduction of anti-CCR4 monoclonal antibody and the expanding use of allogeneic hematopoietic stem cell transplantation. However, not all patients benefit from these treatment modalities, warranting development of a novel therapeutic strategy. CD38, a cell surface ectoenzyme that functions as cyclic ADP ribose hydrolase, is an attractive target of chimeric antigen receptor (CAR) therapy for lymphoid neoplasms because it is widely expressed on the cells of B- or T-lymphoid malignancies. We have previously demonstrated the prominent cytotoxicity of T cells engineered to express an anti-CD38-CAR against B-lymphoma cells and myeloma cells expressing CD38. To expand the applicability of anti-CD38-CAR against ATL cells that usually express undetectable or low levels of CD38, notably, we were successfully able to induce cell surface CD38 expression in HTLV-1-infected cell lines with all-trans retinoic acid (ATRA) (Yoshida T, et al. 2013 ASH Meeting). In ATL cells freshly isolated obtained from the patients, we were able to induce CD38 with ATRA in 60-80% of the cells; the remaining cells survived under the anti-CD38-CAR treatment. We hereby report our attempts in improving the efficacy of anti-CD38-CAR T cells against ATL cells from the patients through the expression of CD38 enhanced with the entry of agents, which are clinically used. Firstly, we investigated whether ATL cells from patients could be transduced with anti-CD38-CAR and what is the efficiency of transduction into T cells in our settings. ATL cells (CD4+ CD25+ GFP+) transduced with retroviral vector were little detected. CD4- CD25- GFP+ T cells alone were detected in our transduction methods. Transduction efficiency was over 40%. To increase the expression of CD38 on ATL cells, we took notice of the CD38 gene upstream region that contains binding sites for interferon regulatory factor-1 (IRF-1) and peroxisome proliferator-activated receptor (PPAR). We thus investigated whether IFN-α, IFN-γ or troglitazone, which is a PPAR-α and -γ agonist, could enhance CD38 expression in ATL cell lines (MT-4, Su9T, ED, and S1T cells), which are negative for CD38. IFN-α and IFN-γ efficiently enhanced CD38 expression in MT-4 cells in a dose-dependent manner but not in Su9T, ED, and S1T cells. As little as 2.5U/ml of IFN-α induced CD38 expression in MT-4 cells for 18 hours in vitr o (>95% at positivity of CD38). 10-25% increase in CD38 expression was observed in ED cells with 125-250 pM troglitazone after 18 hours of treatment, but not in MT-4, Su9T, and S1T cells. Prolonged exposure to troglitazone was toxic to cells. Combined treatment with 10nM ATRA and IFN-α, which induced higher expression of CD38 than IFN-γ, synergistically enhanced CD38 expression of ATL cells from the patients (>90%at positivity of CD38). We next co-cultured ATL cells form three patients with T cells transduced with mock or anti-CD38-CAR in the presence of both ATRA and IFN-α at effector (E): target (T) ratio of 1: 2 for 3 days. The treatment eradicated more than 95% of these ATL cells, demonstrating that ATL cells can be eliminated by T cells harboring anti-CD38-CAR in the presence of ATRA and IFN-α, which is actively used for ATL patients. CD38 targeting therapy is a feasible method, because an anti-CD38 antibody, daratuzumab, has been used to treat plasma cell myeloma. The safety regarding the clinical use of T cells bearing anti-CD38-CAR still needs to be established. As CAR therapy reportedly causes cytokine storm and can potentially be lethal, we envision an inducible immunotherapy with CAR to be a preferred modality with increased efficacy and safety. Our results provide a rationale for a novel therapeutic strategy involving T cells carrying anti-CD38-CAR in combination with ATRA and IFN-α for patients with ATL. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 4803-4803
Author(s):  
Yasuhiro Maeda ◽  
Terufumi Yamaguchi ◽  
Yasuki Hijikata ◽  
Yasuyoshi Morita ◽  
Chikara Hirase ◽  
...  

Abstract We previously reported that all-trans retinoic acid (ATRA) inhibits growth in HTLV-1-positive T-cell lines and fresh cells from patients with adult T-cell leukemia. However, the mechanism of this inhibition is not clear. In the present study, we observed that NF-κB transcriptional activity as well as cell growth decreased significantly in HTLV-1-positive T-cell lines in the presence of ATRA. Furthermore, we observed that ATRA reduced HTLV-1 proviral DNA, HTLV-1 genes (gag, tax or pol mRNA) using the real time quantitative polymerase chain reaction. SIL-2R was reduced by ATRA in both protein level (culture supernantant) and mRNA level in HTLV-1-positive T-cell lines. Interestingly, ATRA significantly inhibited RT activity similar to azidothimidine (AZT) in HTLV-1-positive T-cell lines. Moreover, AZT was inhibitory of proviral DNA but not NF-kB transcriptional activity and sIL-2R on HTLV-1, however ATRA was inhibitory of NF-kB, proviral DNA and sIL-2R on HTLV-1. These results suggested that the decrease of sIL-2R induced by ATRA may be caused by the actions of a NF-kB inhibitor acting on the NF-kB/sIL-2R signal pathway. These results suggested that ATRA could have two roles, as a NF-kB inhibitor and as a RT inhibitor.


2007 ◽  
Vol 134 (6) ◽  
pp. 673-677 ◽  
Author(s):  
Yasuhiro Maeda ◽  
Terufumi Yamaguchi ◽  
Yasuki Hijikata ◽  
Miyako Tanaka ◽  
Chikara Hirase ◽  
...  

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1901-1901
Author(s):  
Keichiro Mihara ◽  
Joyeeta Bhattacharyya ◽  
Yoshihiro Takihara ◽  
Akiro Kimura

Abstract Abstract 1901 Majority of AML blasts express CD38, while human hematopoietic stem cells express CD34 but not CD38. Accordingly, we focused on CD38 for a therapeutic target and developed a cellular immunotherapy by using T cells bearing anti-CD38-chimeric antigen receptor (CAR). We recently reported T cells with anti-CD38-CAR efficiently eliminated B-cell lymphoma cells expressing CD38 in vitro and in vivo. However, intensity of CD38 in lymphoma cells is much higher than that in AML cells. Additionally, leukemic stem cells(LSCs) phenotypically express CD34+CD38−. Thus, to fully employ anti-CD38-CAR against AML blasts including LSCs, intensity of CD38 expression need to be raised for the clinical application. All-trans retinoic acid (ATRA) is widely used for treatment of patients with acute promyelocytic leukemia (APL). Interestingly, it has been reported that ATRA enhances CD38 expression on the surface of AML cells. In this study, we investigated whether human peripheral T cells retrovirally transduced with anti-CD38-CAR kills AML cells and furthermore, whether the cytotoxicity is enhanced in the presence of ATRA. First of all, we confirmed that T cells bearing anti-CD38-CAR expressed GFP as well as the anti-goat mouse IgG-PerCP, whose expression is consistent with the expression of anti-CD38-CAR on the T cells. Next, we evaluated the cytotoxic effect of CD38-specific T cells against two AML cell lines (THP-1 and CMK), which highly express CD38 (>99%). Lactate dehydrogenase (LDH) releasing assay and flow cytometry were performed for cytotoxicity. We co-incubated THP-1 or CMK cells with T cells transduced at a variety of effector (E): target (T) ratios for 3 consecutive days in vitro. Interestingly, these assays showed that T cells bearing anti-CD38-CAR were cytotoxic against THP-1 and CMK cells in dose- and time-dependent manners. However, in the cases of KG1a, U937 and HL60 cells which partially express CD38, killing effect was restricted to CD38+ cells by T cells bearing anti-CD38-CAR, and CD38− AML cells remained alive. These results suggested that augmentation of CD38 expression is essential for the sufficient cytotoxicity against AML cells not expressing CD38 by T cells bearing anti-CD38-CAR. We, then, investigated whether ATRA augmented CD38 expression and resultantly enhanced the cytotoxicity against AML cell lines. Interestingly, ATRA augmented CD38 expression in HL60 cells as well as even in KG1a and U937. However, ATRA by itself exerted no effect on cytotoxicity or proliferating activity in KG1, U937 and even in HL60 cells. These results showed that ATRA contributed to enhancement of CD38 expression, but not to cytotoxicity and proliferation on AML cell lines. Next, we attempted to examine cytotoxic effect of T cells with anti-CD38-CAR on KG1, U937 or HL60 cells following ATRA treatment. AML cell lines were co-cultured with the effector T cells transduced for 3 days in the presence of ATRA. The killing effect of T cells bearing anti-CD38-CAR against AML cell lines was limited in the absence of ATRA. Intriguingly, T cells with anti-CD38-CAR exerted enhanced cytotoxic effect on AML cells in the presence of ATRA. Next, we applied our settings above to AML cells freshly isolated from AML patients. Firstly, we confirmed that CD38 expression was enhanced by ATRA in AML cells from patients as observed in AML cell lines. Furthermore, 3-day incubation of patients' AML cells with T cells bearing anti-CD38-CAR abrogated AML cells in the presence of ATRA. These results indicated that T cells expressing anti-CD38-CAR efficiently eliminated AML cells from patients as well as AML cell line cells through the enhancement of CD38 expression by ATRA. Here, we propose that pre-treatment of ATRA in patient with AML enhances CD38 expression on the leukemic cell surface, which augments the cytotoxic effect of T cells bearing CAR to eradicate leukemic cells including LSCs. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 82-82
Author(s):  
Tomoko Kohno ◽  
Yasuaki Yamada ◽  
Norihiko Akamatsu ◽  
Shimeru Kamihira ◽  
Masao Tomonaga ◽  
...  

Abstract Adult T cell leukemia/lymphoma (ATLL) is a lymphoproliferative disorder caused by a retrovirus, human T-lymphotropic virus type 1 (HTLV-1). ATLL is subclassified into four subtypes, smoldering, chronic, acute, and lymphoma. The acute type progresses rapidly and is usually resistant to conventional chemotherapy. In contrast, the chronic type shows an indolent clinical course and the patients survive for several years, even without chemotherapy. Irrespective of the subtypes, however, ATLL patients are in a severely immune-suppressed condition and can easily acquire opportunistic infections such as Pneumocystis Carinii pneumonitis. Suppression of cell-mediated immunity has also been reported in HTLV-1 carriers. Although ATLL cells show the activated helper/inducer T-cell phenotypes, CD4+ and CD25+, they exhibit strong immune-suppressive activity in vitro. The recent notion of CD4+ CD25+ regulatory T cells (Treg) prompted us to investigate the origin of ATLL cells from the standpoint of Treg. Forkhead/winged helix transcription factor (Foxp3) is a functional marker of Treg, which plays a central role in their generation. There are other marker molecules for Treg, including glucocorticoid-induced TNFR family-related protein (GITR) and the chemokine receptors CCR4 and CCR8. In the present study, we examined primary ATLL cells from 48 patients: 36 patients with acute type and 12 patients with chronic type. We also examined ATLL cell lines, HTLV-1-infected T-cell lines and peripheral blood mononuclear cells (PBMC) from healthy adults as control cells. We used RT-PCR for detection of Foxp3, GITR, CCR4, and CCR8 mRNA expression. Foxp3 and/or GITR mRNA were detected in over 90% of the patients, and 50% of the patients expressed both. There was no difference between subtypes. In contrast, Foxp3 and GITR mRNA were scarcely detected in the PBMC from healthy adults. Furthermore, we confirmed GITR expression at the protein level by flow cytometry. CCR4 and CCR8 mRNA were also detected in almost all ATLL samples, at significantly higher levels than in the normal PBMC. Corresponding to the results of the primary cells, ATLL cell lines and HTLV-1-infected T-cell lines also expressed GITR mRNA, although HTLV-1-negative cell lines, Jurkat and Molt4, completely lack it. Next, we examined whether GITR affects ATLL cell proliferation using a GITR- expressing IL-2-dependent ATLL cell line, KK1. We found that GITR ligand induced proliferation of KK1 cells in an IL-2-negative condition. Thus, these results indicate the Treg origin of ATLL cells and show that GITR expression is possibly involved in the development of ATLL.


Leukemia ◽  
2005 ◽  
Vol 19 (6) ◽  
pp. 1010-1017 ◽  
Author(s):  
T Yamaguchi ◽  
Y Maeda ◽  
S Ueda ◽  
Y Hijikata ◽  
Y Morita ◽  
...  

Leukemia ◽  
2004 ◽  
Vol 18 (6) ◽  
pp. 1159-1160 ◽  
Author(s):  
Y Maeda ◽  
T Yamaguchi ◽  
S Ueda ◽  
H Miyazato ◽  
M Matsuda ◽  
...  

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2096-2096 ◽  
Author(s):  
Inger S. Nijhof ◽  
Henk M. Lokhorst ◽  
Berris van Kessel ◽  
Richard W.J. Groen ◽  
Anton C. Martens ◽  
...  

Abstract INTRODUCTION Daratumumab is an anti-CD38 monoclonal antibody (mAb) with lytic activity against multiple myeloma (MM) cells, including ADCC (antibody-dependent cell-mediated cytotoxicity) and CDC (complement-dependent cytotoxicity). In current clinical phase I/II trials, daratumumab induced anti-MM activity; however, the depth of the response varied between patients. Up till now it is unknown what determines the intrinsic sensitivity of MM cells towards daratumumab-mediated ADCC and CDC. We examined potential determinants of daratumumab sensitivity including CD38 levels, the frequency of effector cells, and expression levels of the complement inhibitory proteins, CD46, CD55, and CD59, which interfere with the different steps of complement activation. RESULTS In bone marrow samples from MM patients, we observed a significant correlation between CD38 expression and daratumumab-mediated ADCC (127 patients; R = 0.428; P < 0.0001) as well as CDC (56 patients; R = 0.338; P = 0.011). Similarly, experiments with isogenic MM cell lines expressing different levels of CD38, revealed that the level of CD38 expression correlates with the extent of daratumumab-mediated ADCC and CDC. Other determinants of daratumumab susceptibility include the effector:target ratio for ADCC, and levels of the complement-inhibitory proteins CD55 and CD59 for CDC. Our data suggest that upregulation of CD38 expression may improve the anti-MM activity of daratumumab. Since, interaction of all-trans retinoic acid (ATRA) with nuclear retinoic acid receptors results in altered expression of target genes including induction of CD38 expression, we evaluated the combination of ATRA and daratumumab. As little as 10 nM ATRA was sufficient to induce a 1.9 – 4.4-fold increase in CD38 expression on the MM cell lines RPMI8226, UM9, and XG1, which resulted in a significant improvement of daratumumab-mediated ADCC and CDC. Importantly, 10 nM ATRA alone resulted in no or only a minimal increase in MM cell death. In addition, ATRA induced a 1.0 – 26.5 (median 1.7) fold increase in CD38 expression on primary MM cells from 26 patients. Also in these primary MM cells, pretreatment with ATRA resulted in a significant increase in their susceptibility to daratumumab-mediated CDC in 13 out of 16 patients as well as ADCC in 8 out of 11 patients. ATRA also enhanced the efficacy of daratumumab in MM cells which are completely resistant to daratumumab-mediated CDC and/or ADCC. Pooled results of these patients show that ATRA improved CDC mediated by 10 µg/mL daratumumab from 16.1 % to 43.9 % (P < 0.0001), and ADCC from 25.1 % to 39.5 % (P = 0.0315). Importantly, expression levels of CD55 and CD59 on MM cells were also significantly reduced by ATRA, which may explain that ATRA improves CDC to a higher extent than ADCC. CONCLUSION Our results provide evidence that CD38 expression levels may predict response to daratumumab. Furthermore, we show that ATRA increases CD38 expression on MM cells, resulting in enhanced daratumumab-mediated lysis of MM cells. Our results provide the preclinical rationale for further evaluation of daratumumab combined with ATRA in MM patients. Disclosures Lokhorst: Celgene: Research Funding; J&J: Research Funding; Genmab: Research Funding. Martens:J&J: Research Funding. Doshi:Janssen R&D: Employment. Mutis:Genmab BV: Research Funding; J&J: Research Funding; Celgene: Research Funding. Sasser:Janssen R&D: Employment. van de Donk:Genmab BV: Research Funding; J&J: Research Funding; Celgene: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document