Dichotomy of All-Trans Retinoic Acid Inducing Signals for Adult T-Cell Leukemia.

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.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4623-4623
Author(s):  
Terufumi Yamaguchi ◽  
Yasuhiro Maeda ◽  
Satomi Ueda ◽  
Yasuki Hijikata ◽  
Mitsuhiro Matsuda ◽  
...  

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 the NF-κB transcriptional activity as well as cell growth in HTLV-1-positive T-cell lines decreased significantly in these cells in the presence of ATRA. However, no significant growth inhibition was observed after treatment with IFN-γ, TNF-α, and TGF-β of growth inhibitory cytokines induced by ATRA. Thereafter, we observed that ATRA reduced HTLV-1 proviral DNA, gag and tax mRNA load using a real time quantitative polymerase chain reaction and soluble IL-2 receptor in the culture supernatant using ELISA in HTLV-1-positive T-cell lines. Interestingly, ATRA significantly inhibited reverse transcriptase activity similar to azidothymidine in HTLV-1-positive T-cell lines. These results suggested that ATRA could inhibit reverse transcriptase activity in the growth inhibition of ATL cells. Additionally, an effect of ATRA on replication of the human immunodeficiency virus (HIV) was also observed. ATRA significantly reduced the HIV proviral DNA load of both a HIV-1-positive cell line and HIV-1-infected patients. These results indicated that ATRA may be useful for HIV treatment in a clinical setting.


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

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2525-2525
Author(s):  
Tetsuro Nakazato ◽  
Chie Ishikawa ◽  
Taeko Okudaira ◽  
Mariko Tomita ◽  
Naoki Mori

Abstract Adult T-cell leukemia (ATL) is caused by human T-cell leukemia virus type I (HTLV-I) and remains incurable. Retinoid is a collective term for compounds, which bind to and activate retinoic acid receptors (RARα, β, γ and RXRα, β, γ), members of nuclear hormone receptor superfamily. It is involved in cell differentiation, morphogenesis, proliferation, and anti-neoplastic processes. The most important endogenous retinoid is all-trans-retinoic acid (ATRA), which is an RARα, β, and γ ligand. ATRA and its mimics have been in clinical use for treatment of acute promyelocytic leukemia (APL) and adult T-cell leukemia (ATL). Many synthetic retinoids have been developed and attempts to improve their medicinal properties have been made. Among them, a novel synthetic retinoid, Am80 (Tamibarotene) is an RARα- and RARβ-specific (but RARγ- and RXRs-nonbinding) synthetic retinoid that is expected to overcome ATRA resistance, because of several times more potent differentiation activity than ATRA and sustained plasma level during continuous administration due to a lower affinity for cellular retinoic acid binding protein. On this background, we examined the inhibitory effect of Am80 on HTLV-I-infected T-cell lines and primary ATL cells. Am80 showed little growth inhibition of peripheral blood mononuclear cells, but it markedly inhibited the growth of both HTLV-I-infected T-cell lines and primary ATL cells. Am 80 could arrest cells in the G1 phase of the cell cycle and induced apoptosis in HTLV-I-infected T-cell lines. The NF-κB pathway is critical for the immortalization and survival of HTLV-I-infected T cells. Therefore, NF-κB pathway was examined as potential targets of Am80 signaling. Am80 significantly inhibited phosphorylation of IκBα and NF-κB-DNA binding, in conjunction with the reduction of expression of proteins involved in the G1-S cell cycle transition and apoptosis. Furthermore, in animal studies, treatment with Am80 produced partial inhibition of growth of tumors of an HTLV-I-infected T-cell line transplanted subcutaneously in severe combined immunodeficient mice. These findings clearly demonstrate that Am80 is a potential inhibitor of NF-κB in ATL cells, and might be a useful therapeutic agent against ATL.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4985-4985
Author(s):  
Yasuhiro Maeda ◽  
Terufumi Yamaguchi ◽  
Chikara Hirase ◽  
Akihisa Kanamaru

Abstract We previously reported that all-trans retinoic acid (ATRA) inhibited growth in HTLV-I- positive T-cell lines and fresh cells from patients with adult T-cell leukemia. We here confirmed the clinical effects of ATRA in 20 patients with ATL. Twenty patients (n=20) with median age of 56 years (range 35–68 years) diagnosed with ATL received ATRA orally. ATRA was administered for a median of 25.7 days (range 14–56 days). Efficacy was described below; no CR case, PR case was 55%, NR case was 45%. In 7 acute cases, PR case was 4 (20%) and NR case was 3 (15%). In 3 lymphoma cases, no NR case and 3 PR cases (15%) was found. In 4 chronic cases, PR case was 1 (4%) and NR case was 3 (15%). In 6 skin type. PR case was 3 (15 %) and NR case was 3 (15%). Major side effects were headache (n=5), transient liver dysfunction (n=2), hyperlipidemia (n=2) and anorexia (n=1). No major toxicity was observed. These results indicated that ATRA might be a useful agent for skin involvement of ATL with safety.


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

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.


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

1997 ◽  
Vol 21 (3) ◽  
pp. 211-216 ◽  
Author(s):  
Kakushi Matsushita ◽  
Naomichi Arima ◽  
Hideo Ohtsubo ◽  
Hiroshi Fujiwara ◽  
Shiroh Hidaka ◽  
...  

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.


Sign in / Sign up

Export Citation Format

Share Document