研究者業績

髙見 真理子

Mariko Takami

基本情報

所属
千葉大学 大学院医学研究院 免疫細胞医学 助教
学位
PhD(Loyola University Chicago Stritch School of Medicine)

研究者番号
60770906
J-GLOBAL ID
201801000984055925
researchmap会員ID
B000310255

受賞

 2

主要な論文

 23
  • Mariko Takami, Takahiro Aoki, Katsuhiro Nishimura, Hidekazu Tanaka, Atsushi Onodera, Shinichiro Motohashi
    Cancer research communications 4(2) 446-459 2024年2月19日  査読有り筆頭著者
    UNLABELLED: Invariant natural killer T (iNKT) cells play an essential role in antitumor immunity by exerting cytotoxicity and producing massive amounts of cytokines. iNKT cells express invariant T-cell receptors (TCR) to recognize their cognate glycolipid antigens such as α-galactosylceramide (α-GalCer) presented on CD1d. We recently reported that iNKT cells recognize CD1d-negative leukemia cell line K562 in a TCR-dependent manner. However, it remains controversial how iNKT cells use TCRs to recognize and exhibit cytotoxic activity toward CD1d-negative tumors cells without CD1d restriction. Here, we report that iNKT cells exerted cytotoxicity toward K562 cells via a carried over anti-Vα24 TCR mAb from positive selection by magnetic bead sorting. We found that addition of the anti-Vα24Jα18 TCR mAb (6B11 mAb) rendered iNKT cells cytotoxic to K562 cells in an FcγRII (CD32)-dependent manner. Moreover, iNKT cells treated with 6B11 mAb became cytotoxic to other CD32+ cell lines (U937 and Daudi). In addition, iNKT cells treated with 6B11 mAb suppressed K562 cell growth in a murine xenograft model in vivo. These data suggest that anti-iNKT TCR mAb treatment of iNKT cells can be applied as a therapeutic strategy to treat CD32+ cancers such as leukemia, lymphoma, and lung cancer. SIGNIFICANCE: Our findings unveiled that iNKT cells recognize and kill CD1d-negative target tumors via the anti-iNKT TCR mAb bound to CD32 at the tumor site, thereby bridging iNKT cells and CD1d-negative tumors. These findings shed light on the therapeutic potential of anti-iNKT TCR mAbs in NKT cell-based immunotherapy to treat CD1d-negative CD32+ cancers.
  • Ayaka Hara, Ryo Koyama-Nasu, Mariko Takami, Takahide Toyoda, Takahiro Aoki, Fumie Ihara, Masayoshi Kobayashi, Seiichiro Hirono, Tomoo Matsutani, Toshinori Nakayama, Yasuo Iwadate, Shinichiro Motohashi
    Cancer immunology, immunotherapy : CII 2020年10月31日  査読有り
    Glioblastoma is the most common and aggressive type of brain tumor with high recurrence and fatality rates. Although various therapeutic strategies have been explored, there is currently no effective treatment for glioblastoma. Recently, the number of immunotherapeutic strategies has been tested for malignant brain tumors. Invariant natural killer T (iNKT) cells play an important role in anti-tumor immunity. To address if iNKT cells can target glioblastoma to exert anti-tumor activity, we assessed the expression of CD1d, an antigen-presenting molecule for iNKT cells, on glioblastoma cells. Glioblastoma cells from 10 of 15 patients expressed CD1d, and CD1d-positive glioblastoma cells pulsed with glycolipid ligand induced iNKT cell-mediated cytotoxicity in vitro. Although CD1d expression was low on glioblastoma stem-like cells, retinoic acid, which is the most common differentiating agent, upregulated CD1d expression in these cells and induced iNKT cell-mediated cytotoxicity. Moreover, intracranial administration of human iNKT cells induced tumor regression of CD1d-positive glioblastoma in orthotopic xenografts in NOD/Shi-scid IL-2RγKO (NOG) mice. Thus, CD1d expression represents a novel target for NKT cell-based immunotherapy for glioblastoma patients.
  • Aoki, T, Takami, M, Takatani, T, Motoyoshi, K, Ishii, A, Hara, A, Toyoda, T, Okada, R, Hino, M, Koyama-Nasu, R, Kiuchi, M, Hirahara, K, Kimura, M, Nakayama, T, Shimojo, N, Motohashi, S
    Cancer Science 111(7) 2223-2233 2020年4月23日  査読有り
  • Takahide Toyoda, Toshiko Kamata, Kazuhisa Tanaka, Fumie Ihara, Mariko Takami, Hidemi Suzuki, Takahiro Nakajima, Takayuki Ikeuchi, Yohei Kawasaki, Hideki Hanaoka, Toshinori Nakayama, Ichiro Yoshino, Shinichiro Motohashi
    Journal for immunotherapy of cancer 8(1) 2020年3月  査読有り
    BACKGROUND: Invariant natural killer T (iNKT) cells produce copious amounts of cytokines in response to specific glycolipid antigens such as α-galactosylceramide (αGalCer) presented by CD1d-expressing antigen-presenting cells (APCs), thus orchestrating other immune cells to fight tumors. Because of their ability to induce strong antitumor responses activated by αGalCer, iNKT cells have been studied for their application in cancer immunotherapy. In our previous phase I/II trial in non-small cell lung cancer (NSCLC) patients who had completed the standard treatment, we showed a relatively long median survival time without severe treatment-related adverse events. Based on these results, we performed a phase II trial to evaluate clinical responses, safety profiles and immune responses as a second-line treatment for advanced NSCLC. METHODS: Patients with advanced or recurrent NSCLC refractory to first-line chemotherapy were eligible. αGalCer-pulsed APCs were intravenously administered four times. Overall survival time was evaluated as the primary endpoint. The safety profile and immune responses after APC injection were also monitored. This study was an open label, single-arm, phase II clinical trial performed at Chiba University Hospital, Japan. RESULTS: Thirty-five patients were enrolled in this study, of which 32 (91.4%) completed the trial. No severe adverse events related to the treatment were observed. The estimated median survival time of the 35 cases was 21.9 months (95% CI, 14.8 to 26.0). One case (2.9%) showed a partial response, 14 cases (40.0%) remained as stable disease, and 19 cases (54.3%) were evaluated as progressive disease. The geometric mean number of iNKT cells in all cases was significantly decreased and the mean numbers of natural killer (NK) cells, interferon-γ-producing cells in response to αGalCer, and effector CD8+ T cells were significantly increased after the administration of αGalCer-pulsed APCs. CONCLUSIONS: The intravenous administration of αGalCer-pulsed APCs was well-tolerated and was accompanied by prolonged overall survival. These results are encouraging and warrant further evaluation in a randomized phase III trial to demonstrate the survival benefit of this immunotherapy. TRIAL REGISTRATION NUMBER: UMIN000007321.
  • Toshiko Kamata, Shigetoshi Yoshida, Mariko Takami, Fumie Ihara, Hiroko Yoshizawa, Takahide Toyoda, Yuichiro Takeshita, Seiichi Nobuyama, Yukiko Kanetsuna, Tetsuo Sato, Ichiro Yoshino, Shinichiro Motohashi
    Cancer science 111(1) 288-296 2020年1月  査読有り
    The role of immune checkpoint inhibitors in metastatic lung cancer has been established in recent years and the pretherapeutic profiles of the tumor microenvironment in responders have been increasingly reported. The role of salvage surgery and the immune profiles of the posttherapeutic specimens in patients achieving an objective response have rarely been studied. We report a case of metastatic lung cancer treated by anti-programmed death-1 Ab followed by surgical resection. The immune status of the tumor was assessed, showing germinal center formation, memory B cell infiltration, and a high frequency of interferon gamma -secreting T cells.
  • Fumie Ihara, Daiju Sakurai, Mariko Takami, Toshiko Kamata, Naoki Kunii, Kazuki Yamasaki, Tomohisa Iinuma, Toshinori Nakayama, Shinichiro Motohashi, Yoshitaka Okamoto
    Cancer immunology, immunotherapy : CII 68(12) 1935-1947 2019年12月  査読有り
    BACKGROUND: Due to the strong tumoricidal activities of activated natural killer T (NKT) cells, invariant NKT cell-based immunotherapy has shown promising clinical efficacy. However, suppressive factors, such as regulatory T cells (Tregs), may be obstacles in the use of NKT cell-based cancer immunotherapy for advanced cancer patients. Here, we investigated the suppressive effects of Tregs on NKT cells and the underlying mechanisms with the aim to improve the antitumor activities of NKT cells. METHODS: Peripheral blood samples were obtained from healthy donors, patients with benign tumors, and patients with head and neck squamous cell carcinoma (HNSCC). NKT cells, induced with α-galactosylceramide (α-GalCer), and monocyte-derived dendritic cells (DCs) were co-cultured with naïve CD4+ T cell-derived Tregs to investigate the mechanism of the Treg suppressive effect on NKT cell cytotoxic function. The functions and phenotypes of NKT cells were evaluated with flow cytometry and cytometric bead array. RESULTS: Treg suppression on NKT cell function required cell-to-cell contact and was mediated via impaired DC maturation. NKT cells cultured under Treg-enriched conditions showed a decrease in CD4- NKT cell frequency, which exert strong tumoricidal responsiveness upon α-GalCer stimulation. The same results were observed in HNSCC patients with significantly increased effector Tregs. CONCLUSION: Tregs exert suppressive effects on NKT cell tumoricidal function by inducing more CD4- NKT cell anergy and less CD4+ NKT cell anergy. Both Treg depletion and NKT cell recovery from the anergy state may be important for improving the clinical efficacy of NKT cell-based immunotherapy in patients with advanced cancers.
  • Kazuaki Harada, Fumie Ihara, Mariko Takami, Toshiko Kamata, Naoko Mise, Hiroko Yoshizawa, Tomoro Hishiki, Takeshi Saito, Keita Terui, Mitsuyuki Nakata, Shugo Komatsu, Takayuki Ikeuchi, Toshinori Nakayama, Hideo Yoshida, Shinichiro Motohashi
    Cancer science 110(3) 888-902 2019年3月  査読有り
    Dendritic cells (DC) play a key role in the initiation of both antitumor immunity and immunological tolerance. It has been demonstrated that exposure to soluble factors produced by tumor cells modulates DC functions and induces tolerogenic DC differentiation. In this study, we investigated the effects of neuroblastoma cell line-derived soluble factors on DC differentiation. Monocytes isolated from healthy volunteers were incubated with interleukin (IL)-4 and granulocyte-macrophage colony-stimulating factor in the presence of culture supernatants from neuroblastoma cell lines. The culture supernatants from neuroblastoma cell lines, such as NLF and GOTO, partially blocked both downregulation of CD14 and upregulation of CD1a, and dramatically decreased IL-12 and tumor necrosis factor (TNF)-α production from mature DC, while no effect of SH-SY5Y cell supernatant was noted. In addition, IL-6 and IL-10 production from monocytes was increased by the supernatants of NLF and GOTO cells at 24 hours after incubation. Furthermore, we evaluated DC functions through stimulation of invariant natural killer T (iNKT) cells. α-Galactosylceramide-pulsed DC co-cultured with supernatants of NLF cells were unable to sufficiently stimulate iNKT cells. The decreased ability of iNKT cells to produce interferon (IFN)-γ after stimulation with neuroblastoma cell line supernatant-cultured DC was reversed by addition of IL-12. CD40 expression and IL-12 production in NLF-sup-treated DC were increased by addition of exogenous IFN-γ. These results indicate that tolerogenic DC are induced in the neuroblastoma tumor microenvironment and attenuate the antitumor effects of iNKT cells. Interactions between iNKT cells and αGalCer-pulsed DC have the potential to restore the immunosuppression of tolerogenic DC through IFN-γ production.
  • Takami M, Cunha C, Motohashi S, Nakayama T, Iwashima M
    European journal of immunology 48(12) 1938-1943 2018年12月  査読有り筆頭著者
  • Takami, M, Ihara, F, Motohashi, S
    Front. Immunol. 9 2021 2018年9月  査読有り筆頭著者
  • Toshiko Kamata, Akane Suzuki, Naoko Mise, Fumie Ihara, Mariko Takami, Yuji Makita, Atsushi Horinaka, Kazuaki Harada, Naoki Kunii, Shigetoshi Yoshida, Ichiro Yoshino, Toshinori Nakayama, Shinichiro Motohashi
    CANCER IMMUNOLOGY IMMUNOTHERAPY 65(12) 1477-1489 2016年12月  査読有り
    The role of invariant natural killer T (iNKT) cells in antitumor immunity has been studied extensively, and clinical trials in patients with advanced cancer have revealed a prolonged survival in some cases. In recent years, humanized blocking antibodies against co-stimulatory molecules such as PD-1 have been developed. The enhancement of T cell function is reported to improve antitumor immunity, leading to positive clinical effects. However, there are limited data on the role of PD-1/programmed death ligand (PDL) molecules in human iNKT cells. In this study, we investigated the interaction between PD-1 on iNKT cells and PDL on antigen-presenting cells (APCs) in the context of iNKT cell stimulation. The blockade of PDL1 at the time of stimulation resulted in increased release of helper T cell (Th) 1 cytokines from iNKT cells, leading to the activation of NK cells. The direct antitumor function of iNKT cells was also enhanced after stimulation with anti-PDL1 antibody-treated APCs. According to these results, we conclude that the co-administration of anti-PDL1 antibody and alpha-galactosylceramide (alpha GalCer)-pulsed APCs enhances iNKT cell-mediated antitumor immunity.
  • Naoko Mise, Mariko Takami, Akane Suzuki, Toshiko Kamata, Kazuaki Harada, Tomoro Hishiki, Takeshi Saito, Keita Terui, Tetsuya Mitsunaga, Mitsuyuki Nakata, Takayuki Ikeuchi, Toshinori Nakayama, Hideo Yoshida, Shinichiro Motohashi
    CANCER SCIENCE 107(3) 233-241 2016年3月  査読有り
    Anti-ganglioside GD2 antibodies mainly work through antibody-dependent cellular cytotoxicity (ADCC) and have demonstrated clinical benefit for children with neuroblastoma. However, high-risk neuroblastoma still has a high recurrence rate. For further improvement in patient outcomes, ways to maximize the cytotoxic effects of anti-GD2 therapies with minimal toxicity are required. Activated invariant natural killer T (iNKT) cells enhance both innate and typeI acquired anti-tumor immunity by producing several kinds of cytokines. In this report, we investigated the feasibility of combination therapy using iNKT cells and an anti-GD2 antibody. Although some of the expanded iNKT cells expressed natural killer (NK) cell markers, including FcR, iNKT cells were not directly associated with ADCC. When co-cultured with activated iNKT cells, granzyme A, granzyme B and interferon gamma (IFN) production from NK cells were upregulated, and the cytotoxicity of NK cells treated with anti-GD2 antibodies was increased. Not only cytokines produced by activated iNKT cells, but also NK-NKT cell contact or NK cell-dendritic cell contact contributed to the increase in NK cell cytotoxicity and further IFN production by iNKT cells and NK cells. In conclusion, iNKT cell-based immunotherapy could be an appropriate candidate for anti-GD2 antibody therapy for neuroblastoma.
  • Mariko Takami, Kotaro Fujimaki, Michael I. Nishimura, Makio Iwashima
    JOURNAL OF IMMUNOLOGY 195(6) 2520-2523 2015年9月  査読有り筆頭著者
    The immunoregulatory functions of vitamin D have been well documented in various immunological disorders, including multiple sclerosis, arthritis, and asthma. IL-10 is considered a chief effector molecule that promotes the vitamin D-induced immunosuppressive states of T cells and accessory cells. In this article, we demonstrate that the active form of vitamin D, 1,25-dihydroxyvitamin D-3 (calcitriol), has a profound inhibitory effect on the development of human Th9, a CD4 T cell subset that is highly associated with asthma, in an IL-10-independent manner. Our data show that calcitriol represses the expression of BATF, a transcription factor essential for Th9, via suppressing the expression of aryl hydrocarbon receptor, without an increase in IL-10. The data show a novel link between vitamin D and two key transcription factors involved in T cell differentiation.
  • Takami M, Love RB, Iwashima M
    Journal of immunology (Baltimore, Md. : 1950) 188(9) 4369-4375 2012年5月  査読有り筆頭著者
  • Nagendra Singh, Mutsumi Yamamoto, Mariko Takami, Yoichi Seki, Mayuko Takezaki, Andrew L. Mellor, Makio Iwashima
    JOURNAL OF IMMUNOLOGY 184(1) 94-104 2010年1月  査読有り
    Ag receptor stimulation of preactivated T cells causes rapid cell death in an IL-2- and Fas-dependent manner. This phenomenon, known as activation-induced cell death (AICD), plays a pivotal role in the removal of Ag-reactive T cells after initial expansion. In this study, we report a novel form of T cell apoptosis that is distinct from classic AICD. When peripheral T cells were activated with anti-CD3 and anti-CD28 Abs precoated onto plastic plates, CD4(+)CD25(-) and CD8 T cells initially expanded but underwent massive apoptosis after 4 d. Unlike classic AICD, this type of T cell apoptosis pathway requires engagement of CD28 and expression of p53, a tumor-suppressor gene. The most striking feature of this form of apoptosis was regulatory T cell resistance. Under the same stimulating conditions, CD4(+)CD25(+) T cells grew continuously beyond 4 d. Consequently, when the entire CD4 population was cultured with plate-bound anti-CD3 plus anti-CD28 Ab, CD4(+)CD25(+)FoxP3(+) regulatory T cells outgrew non-regulatory T cells and expanded > 7000-fold after 11 d. The data presented herein demonstrate a novel process of Ag-induced T cell death by sustained TCR and CD28 engagement and represent a simple and efficient procedure for the expansion of regulatory T cells in vitro. The Journal of Immunology, 2010, 184: 94-104.
  • Nagendra Singh, Yoichi Seki, Mariko Takami, Babak Baban, Phil R. Chandler, Davood Khosravi, Xiangjian Zheng, Mayuko Takezaki, Jeffrey R. Lee, Andrew L. Mellor, Wendy B. Bollag, Makio Iwashima
    NATURE METHODS 3(8) 629-636 2006年8月  査読有り
    Antigen stimulation of lymphocytes induces upregulation of phospholipase D (PLD) activity, but the biological significance of PLD-mediated signaling in T cells has not been well established. Here we demonstrate that PLD signaling is essential for proliferation of mouse CD8(+) T cells and CD4(+)CD25(-) T cells, but is not required for proliferation of CD4(+)CD25(+) regulatory T cells. We exploited this observation to develop an efficient method to enrich for regulatory T cells starting from preparations of total CD4(+) T lymphocytes. Inhibition of PLD signaling blocked effector T-cell proliferation after T cell-antigen receptor (TCR) engagement, but had no significant effect on the proliferation of CD4(+)CD25(+) T cells with regulatory functions. Consequently, cells expanded in vitro for one week by antigen receptor stimulation with PLD signal inhibition were markedly enriched for regulatory T cells.

MISC

 9
  • 豊田 行英, 本橋 新一郎, 山本 高義, 森本 淳一, 坂入 祐一, 和田 啓伸, 鈴木 秀海, 中島 崇裕, 吉野 一郎, 伊原 史英, 高見 真理子
    千葉医学雑誌 95(6) 198-198 2019年12月  
  • Takahiro Aoki, Mariko Takami, Tomozumi Takatani, Kiwamu Motoyoshi, Ayana Ishii, Ayaka Hara, Takahide Toyoda, Reona Okada, Moeko Hino, Ryo Koyama-Nasu, Masahiro Kiuchi, Kiyoshi Hirahara, Toshinori Nakayama, Naoki Shimojo, Shinichiro Motohashi
    Blood 134(Supplement_1) 3225-3225 2019年11月13日  
    Background: Invariant natural killer T (iNKT) cells are known as CD1d-restricted T cells that express the invariant T-cell receptors (TCR) Vα24 and Vβ11 in humans and specifically recognize glycolipid antigens such as α-galactosylceramide (αGalCer) presented by CD1d. iNKT cells show direct cytotoxicity toward CD1d-positive tumor cells presenting glycolipid antigens and indirect cytotoxicity by activating other cytotoxic immune cells or regulating CD1d-positive immunosuppressive cells in the tumor microenvironment. Although we previously reported that αGalCer-activated NKT cells exert a potent perforin-dependent cytotoxic activity against a wide variety of human tumor cell lines, the direct recognition of CD1d-negative tumors is controversial and the mechanism is unknown. Here we clarify whether iNKT cells recognize and exhibit cytotoxicity toward leukemia cells in a CD1d-independent manner and identify the molecule that recognizes CD1d-negative leukemia cells. Methods: Purified iNKT cells were generated from peripheral blood mononuclear cells (PBMCs) of healthy adult volunteer donors. PBMCs were cultured in complete RPMI 1640 medium for 9-14 days in the presence of 100 U/mL of recombinant human IL-2 and 200 ng/mL of αGalCer. The iNKT cells were then isolated with an autoMACS Pro separator using FITC-labeled anti-Vα24 antibody (clone, C15) and anti-FITC microbeads. We evaluated the cytotoxic activity of iNKT cells toward CD1d-negative leukemia cells within four days after isolation using a CD107a assay for degranulation, cytometric bead array for cytokine production, and cytotoxicity assay in vitro and in vivo. For in vivo cytotoxicity assays, NOG mice were inoculated with 1 × 106 K562-luc cells on day 0 and with 4 × 106 human iNKT cells on day 1. Gene knock-out (KO) was performed using a CRISPR/Cas9 system. T-cell or NK receptor-KO iNKT cells were used for experiments three or four days after electroporation of the Cas9 protein and guide RNA CRISPR ribonucleoprotein complex. Patient-derived leukemia cells were obtained from PBMCs or bone marrow mononuclear cells of pre-treatment pediatric patients. All studies were approved by the institutional review board and the Animal Care and Use Committee of Chiba University. Results: We observed that iNKT cells degranulated and released Th1 cytokines when co-cultured with CD1d-negative leukemia cells (K562, HL-60, REH, and CD1d-KO U937) as well as αGalCer-loaded CD1d-positive leukemia cells (Jurkat), and showed in vitro cytotoxicity toward these CD1d-negative leukemia cells. This CD1d-independent degranulation decreased over time after isolation and was not restored with re-stimulation by αGalCer. The cytotoxicity of iNKT cells toward K562 cells was confirmed in vivo by comparsion with survival curves of K562-inoculated NOG mice given iNKT cells or PBS alone (log-rank, p= 0.016). To identify the receptors contributing to the CD1d-independent recognition and cytotoxicity against CD1d-negative leukemia cells, we first focused on costimulatory receptors, which are also known as activating NK receptors and are expressed on iNKT cells such as NKG2D, DNAM-1, 2B4, LFA-1, and CD2, and analyzed cytotoxicity after blocking these receptors with antibodies. We found that all costimulatory receptors that we assessed contributed to cytotoxicity toward CD1d-negative leukemia cells. Next, we analyzed cytotoxicity of TCR-KO iNKT cells toward CD1d-negative leukemia cells to confirm the contribution of TCR to CD1d-independent recognition. Notably, TCR-KO iNKT cells showed decreased degranulation, Th1 cytokine release, and cytotoxicity toward K562 cells more so than iNKT cells with KO of NK receptors such as LFA-1(CD11a) or CD2. To assess the clinical application potential of adoptive iNKT cell immunotherapy for leukemia treatment, we analyzed degranulation of iNKT cells using patient-derived leukemia cells. We found iNKT cells degranulation using cells from four out of five myeloid leukemia cases, but only one out of eight BCP-ALL cases (p = 0.032). Conclusion: Primary iNKT cells activated by αGalCer can recognize and show anti-tumor effects toward leukemia cells in an unrestricted manner via CD1d. The TCR also has an important role in recognizing CD1d-negative leukemia cells and multiple NK receptors assist in cytotoxicity. Adoptive iNKT cell immunotherapy may be effective in treating myeloid leukemia. Disclosures No relevant conflicts of interest to declare.
  • 原 彩佳, 那須 亮, 髙見 真理子, 廣野 誠一郎, 松谷 智郎, 中山 俊憲, 岩立 康男, 本橋 新一郎
    日本がん免疫学会総会プログラム・抄録集 23回 94-94 2019年7月  
  • 原田 和明, 齋藤 武, 照井 慶太, 中田 光政, 小松 秀吾, 秦 佳孝, 吉澤 比呂子, 工藤 渉, 古金 遼也, 髙見 真理子
    日本小児外科学会雑誌 55(3) 605-605 2019年5月  
  • 豊田 行英, 本橋 新一郎, 坂入 祐一, 田村 創, 和田 啓伸, 藤原 大樹, 鈴木 秀海, 中島 崇裕, 山田 義人, 千代 雅子, 吉野 一郎, 高見 真理子
    千葉医学雑誌 94(4) 168-168 2018年8月  

書籍等出版物

 1

講演・口頭発表等

 34

共同研究・競争的資金等の研究課題

 4