リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「Development of a novel miR-34-regulated oncolytic coxsackievirus B3 with minimal toxicity and strong anti-tumor activity」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

Development of a novel miR-34-regulated oncolytic coxsackievirus B3 with minimal toxicity and strong anti-tumor activity

賈, 楊 東京大学 DOI:10.15083/0002002447

2021.10.15

概要

Lung cancer became the major cause of cancer deaths among Japanese, making up approximately 25% of all cancer deaths in men and 14% in women. Five-year survival of lung cancer patients is 39.1% for all stages, despite the use of intensive combined therapies and recent advances in molecular targeting therapies. To improve this poor prognosis, new therapeutic modalities are urgently required.

 Oncolytic viruses (OVs) have emerged as new modalities for cancer treatment depending on tumor cell lysis by preferential replication of OVs in tumor cells followed by activation of host’s anti-tumor immunity. Very recently, talimogene laherparepvec (T-Vec), a modified herpes simplex virus type 1 encoding granulocyte-macrophage colony-stimulating factor (GM-CSF), is the first oncolytic virus approved by US Food and Drug Administration for the advanced melanoma. The success of this therapy encourages scientists to make further research in this new field. Till now, other types of OVs, such as adenoviruses, reoviruses, measles viruses, and Newcastle disease viruses, have been developed and pre-clinical and clinical studies for many cancers have been performed. However, tumor cells often become resistant to OVs due to downregulation of their receptors, resulting in insufficient proliferation and transmission of viruses. Therefore, novel OVs targeting different tumor-expressing receptors with strong proliferative ability would be useful in the clinical setting.

 In order to obtain a novel OV, our lab previously screened 28 enterovirus strains and found that coxsackievirus B3 (CVB3), which uses coxsackievirus and adenovirus receptor (CAR) and decay- accelerating factor (DAF) as its receptors, was a novel OV with a strong ability to lyse human non–small cell lung cancer cells (NSCLC). CVB3 is a small, nonenveloped, single-stranded, positive-sense RNA virus and belongs to the Enterovirus genus within the Picornaviridae family. Its rapid replication and the broad expression of its receptors are considered to broaden the range of target cancers potentially susceptible to CVB3 therapy, resulting in a strong and sustaining anti-tumor effect. However, during subsequent characterization of CVB3 toward clinical translation of the virus, several non-negligible toxicities were observed in mouse tumor models due to its broad tropism, particularly in pancreas. Consequently, elimination of these virulent features of CVB3 is essential for developing CVB3-based OVs in the clinical setting.

 To inhibit viral replication-induced cytotoxicity in non-tumor cells, I adopted gene silencing mechanism of micro RNAs (miRNAs) by inserting target sequences of tumor suppressor miRNAs, which are abundantly expressed in non-tumor cells, in the genome of oncolytic CVB3. miRNAs mainly cause degradation or translational inhibition of mRNAs by guiding the RNA-induced silencing complex (RISC) to target sites. Among these miRNAs, I focused on miRNA-34 family, which consists of only three members of miR-34a, miR-34b and miR-34c and was reported to be broadly down-regulated in many tumor cells including NSCLC, to decrease off target replication of CVB3 in normal organ cells and keep vigorous virus replication in tumor cells.

 Additionally, the insertable location of miRNA target sites (miRTs) is another important issue to suppress viral replication in normal organ cells, while maintaining oncolytic activity in cancer cells. Based on previous reports about insertion-tolerant sites of picornaviruses, I predicted that miRTs could be inserted in the 5’ region immediately upstream of the start codon, as well as in the 3’region just downstream of the stop codon. Furthermore, insertion of four copies of miRT in viral vector genomes was reported to inhibit virus replication by complementary miRNAs. Therefore, I engineered miRNA-regulated CVB3s (miRT- CVBs) by inserting four tandem miR-34a target sites (miR-34aT) or miR-34c target sites (miR-34cT) into the 5’UTR (5-CVBs) or 3’UTR (3-CVBs) of the CVB3 genome.

 First, I transfected synthetic miR-34a or miR-34c mimics to H1299 cells, which were expressing low levels of miR-34a/c compared to normal cells, and then inoculated the cells with miRT-CVBs or wild type (WT-) CVBs. After 72 h, the miR34a or miR-34c mimics-transfected cells exhibited much less cell lysis when infected with miRT-CVBs harboring complimentary miRTs, while in untransfected H1299 cells, all miRT-CVBs induced massive cell lysis as did in WT-CVB. These results indicate that insertion of miRTs made CVB3 less toxic only in cells expressing miR-34a/c.

 The cytotoxic activity of miRT-CVBs was also evaluated using several human cancer cell lines and a human normal lung cell line, BEAS-2B, by MTS assay. The results showed that infection with miRT-CVBs in cancer cells retained its abundant viral replication in comparison with the parental WT-CVB, while its attenuated replication was observed in BEAS-2B expressing high level of miR-34a and miR-34c, depending on intracellular miR-34a or miR-34c levels.

 Next, to investigate the anti-tumor activity of miRT-CVBs in vivo, I injected miRT-CVBs and WT-CVB into H1299 tumors in BALB/c nude mice, and monitored the tumor growth. In contrast to control mice showing continuous tumor growth, all WT- or miRT-CVB-treated groups exhibited complete tumor regression with no death during the observation period, indicating that miRT-CVBs preserved the anti- tumor activity of WT-CVB.

 To address in vivo toxicity of those CVBs, blood biochemistry tests and pathological examination were performed for CVB-treated tumor-harboring mice. In WT-CVB treated mice, serum AST, ALT, LDH, and amylase levels were significantly increased, and histological signs of pancreatic injury were observed. While 5-CVBs failed to reduce the CVB3-associated pathogenicity, 3-CVB3 did not significantly increase the enzyme levels except ALT and induced less pancreatic injury. Because elevated LDH reflected the damage of many organs, the reduced LDH elevation in 3-CVBs mice were considered to be due to restricted virus replication in various organs. Between miR-34a and miR-34c target sites, miR-34a seemed to work better to reduce toxicities in mimics-transfected H1299 cells and in normal tissues, possibly because all organs including heart and pancreas, which are susceptible to CVB3 expressed miR-34a, while miR-34c expressed only in lung and brain. Another possibility is the difference in seed-matched sites of miRNA. Although miR-34a and miR-34c belong to the same family and share the same seed region, the type of miR-34aT, which contains an A at position 1 named 8mer-type site, is thought to induce miRNA inhibition much stronger than other types of sites. Although miR-34aT or miRT-34cT was successfully inserted in the 5’UTR and 3’UTR of CVB3 genome without losing anti-tumor effect of the original virus, 3-CVBs exhibited significantly lower pathogenicity than 5-CVBs. Stronger inhibition by inserting miRTs in 3’UTR was consistent with previous reports in which miRNA can inhibit a mRNA more efficiently when the target sequence is in the 3’UTR than in the 5’UTR. These results indicated that insertion of miRTs reduced WT- CVB-associated pathogenicity in normal cells without attenuating the anti-tumor activity, and the 3a-CVB (miR-34a inserted into 3’UTR of virus) was the best miRT-CVB in terms of anti-tumor effect and safety, while this virus still induced slight side effect, mild ALT elevation and less pancreas injury.

 To further improve the safety of 3a-CVB, four tandem miR-34aT were inserted not only in 3’ UTR but also in 5’UTR of CVB3 genome (53a-CVB) to increase miR-34a-sensitive sites. 53a-CVB showed the same level of cytotoxicity in H1299 cells as WT-CVB but significant lower toxicity in miR34a-transfected H1299 cells. As expected, the decrease of cytotoxicity was more significant than that of 5a-CVB (miR-34a inserted into 5’UTR of virus) or 3a-CVB. While 53a-CVB preserved the same level of anti-tumor effect in the xenograft mouse model, 53a-CVB did not cause significant elevation of all enzymes including ALT, which was elevated in 3a-CVB-treated mice. Histopathological examination also showed no findings of organ injuries by 53a-CVB treatment, suggesting nearly perfect suppressed CVB-induced organ toxicity by double insertion of miR34aT in both UTRs. To confirm the suppressed CVB replication in normal organs, copy numbers of CVB genomes in each mouse organ were determined by RT-qPCR. In mice treated with WT-CVB, pancreas was most heavily infected besides tumors, followed by spleen, heart, and lung. On the other hand, virus copies of all organs including pancreas were much less in 53a-CVB-treated mice than WT-CVB. Of note is that tumor of 53a-treated mice had similar virus load to that of WT-CVB-treated mice. These data were computable with the findings that 53a-CVB not only preserved anti-tumor effect but also obtained very low toxicity. To further evaluate the safety, I addressed effect of immunity on CVB3-relating toxicity by using immunocompetent C57BL/6J mouse model transplanted with syngeneic mouse lung cancer TC-1 cells. There were no obvious signs of organ injuries by 53a-CVB treatment as observed in xenograft nude mice model suggesting that 53a-CVB would be a very efficient and safe and useful OV in the clinical setting.

 To date, the target sequences of let-7 miRNA family members, which are another tumor suppressor miRNAs, have been used to reduce toxicity of OVs in normal tissues. Insertion of the let-7 targets in OVs, however, often decreases not only normal tissue toxicity but also anti-tumor activity due to the extensive overlap in target sequences among its various family members. Thus, I tried to find other miRNA-regulating systems with stronger on-target effects and milder off-target effects. In contrast to let-7, the miR-34 family consists of only three members, suggesting less interference among the family members. In this study, there was almost no reduction in the cytotoxicity when H1299 cells transfected with miR34a/c were infected with CVBs including different miR34a/cT, also suggesting excellence of miR34a/c-regulating system for controlling CVB replication.

 Finally, I addressed why miR34aT-inserted CVB (miR34aT-CVB) could efficiently kill some tumor cells with high miR-34a. Although miR-34a was expressed more strongly in A549 cells than in normal lung cell line, 53a-CVB still exerted stronger cytotoxicity in these tumor cells with much lower titers than in non- tumor cells. To demonstrate mechanisms of this phenomenon, I focused on PI3K/Akt and MAP/ERK/MEK pathway because CVB3 replication depended on these pathways, which were often activated in cancer cells. In the presence of inhibitors for those pathways, cytotoxicity was significantly reduced by miR34aT-CVB in miR34a-high A549 cells but not in miR34a-low AsPC cells, suggesting that miR-34-induced inhibition of miR-34aT-CVB replication could be overridden by aberrantly activated PI3K/Akt and/or MAP/ERK/MEK pathways in tumor cells.

 In conclusion, I successfully developed the first miR-34a/c–regulated CVB3 by inserting miR-34aT or miR-34cT in the 5’UTR or 3’UTR of the CVB3 genome, without attenuating its oncolytic activity. miRT- CVBs exerted significantly lower toxicity in normal tissues where miR-34a/c were highly expressed. CVB3 containing miR-34aT had less pathogenicity than CVB3 containing miR-34T, and insertion of the miR- 34a/cT into the 3’UTR reduced pathogenicity to a greater extent than insertion into the 5’UTR. Finally, 53a-CVB, a double miR34aT-inserted CVB, was minimally toxic to normal tissues while maintaining full oncolytic activity in mouse xenograft model of lung cancer. This study successfully indicated that 53a-CVB is a promising OV that could be useful in the clinical setting due to its minimal toxicity with strong oncolytic effects.

この論文で使われている画像

参考文献

1. S. Miyamoto et al., Coxsackievirus B3 Is an Oncolytic Virus with Immunostimulatory Properties That Is Active against Lung Adenocarcinoma. Cancer Research 72, 2609-2621 (2012).

2. F. f. t. P. o. C. Research., "Cancer Statistics in Japan, 2015," (March, 2016.).

3. S. N. Lester, K. Li, Toll-Like Receptors in Antiviral Innate Immunity. Journal of Molecular Biology 426, 1246-1264 (2014).

4. S. B. Fleming, Viral Inhibition of the IFN-Induced JAK/STAT Signalling Pathway: Development of Live Attenuated Vaccines by Mutation of Viral- Encoded IFN-Antagonists. Vaccines 4, (2016).

5. M. A. Garcia et al., Impact of protein kinase PKR in cell biology: from antiviral to antiproliferative action. Microbiology and Molecular Biology Reviews 70, 1032-+ (2006).

6. M. J. Clemens, Targets and mechanisms for the regulation of translation in malignant transformation. Oncogene 23, 3180-3188 (2004).

7. Y. H. Huang, S. Goel, D. G. Duda, D. Fukumura, R. K. Jain, Vascular Normalization as an Emerging Strategy to Enhance Cancer Immunotherapy. Cancer Research 73, 2943-2948 (2013).

8. C. J. Breitbach et al., Targeting Tumor Vasculature With an Oncolytic Virus. Molecular Therapy 19, 886-894 (2011).

9. M. T. Bejarano, J. R. Merchan, Targeting tumor vasculature through oncolytic virotherapy: recent advances. Oncolytic Virotherapy 4, 169-181 (2015).

10. F. A. Angarita, S. A. Acuna, K. Ottolino-Perry, S. Zerhouni, J. A. McCart, Mounting a strategic offense: fighting tumor vasculature with oncolytic viruses. Trends in Molecular Medicine 19, 378-392 (2013).

11. S. J. Ries, C. H. Brandts, Oncolytic viruses for the treatment of cancer: current strategies and clinical trials. Drug Discovery Today 9, 759-768 (2004).

12. H. L. Kaufman, F. J. Kohlhapp, A. Zloza, Oncolytic viruses: a new class of immunotherapy drugs (vol 14, pg 642, 2015). Nature Reviews Drug Discovery 15, (2016).

13. S. J. Russell, K. W. Peng, Oncolytic Virotherapy: A Contest between Apples and Oranges. Molecular Therapy 25, 1107-1116 (2017).

14. R. H. I. Andtbacka et al., Talimogene Laherparepvec Improves Durable Response Rate in Patients With Advanced Melanoma. Journal of Clinical Oncology 33, 2780-U2798 (2015).

15. K. Twumasi-Boateng, J. L. Pettigrew, Y. Y. E. Kwok, J. C. Bell, B. H. Nelson, Oncolytic viruses as engineering platforms for combination immunotherapy. Nature Reviews Cancer 18, 419-432 (2018).

16. C. Heise et al., Efficacy of a replication-selective adenovirus against ovarian carcinomatosis is dependent on tumor burden, viral replication and p53 status. Gene Therapy 7, 1925-1929 (2000).

17. K. Fukuda et al., E1A, E1B double-restricted adenovirus for oncolytic gene therapy of gallbladder cancer. Cancer Research 63, 4434-4440 (2003).

18. M. A. Strassburg, THE GLOBAL ERADICATION OF SMALLPOX. American Journal of Infection Control 10, 53-59 (1982).

19. G. L. Smith, A. Vanderplasschen, M. Law, The formation and function of extracellular enveloped vaccinia virus. Journal of General Virology 83, 2915- 2931 (2002).

20. B. He et al., Suppression of the phenotype of gamma(1)34.5(-) herpes simplex virus-1: Failure of activated RNA-dependent protein kinase to shut off protein synthesis is associated with a deletion in the domain of the alpha 47 gene. Journal of Virology 71, 6049-6054 (1997).

21. K. Fruh et al., A VIRAL INHIBITOR OF PEPTIDE TRANSPORTERS FOR ANTIGEN PRESENTATION. Nature 375, 415-418 (1995).

22. H. L. Kaufman, C. E. Ruby, T. Hughes, C. L. Slingluff, Jr., Current status of granulocyte-macrophage colony-stimulating factor in the immunotherapy of melanoma. Journal for immunotherapy of cancer 2, 11-11 (2014).

23. T. J. Tuthill, E. Groppelli, J. M. Hogle, D. J. Rowlands, Picornaviruses. Cell Entry by Non-Enveloped Viruses 343, 43-89 (2010).

24. L. van der Linden, K. C. Wolthers, F. J. M. van Kuppeveld, Replication and Inhibitors of Enteroviruses and Parechoviruses. Viruses-Basel 7, 4529-4562 (2015).

25. D. R. Shafren, D. T. Williams, R. D. Barry, A decay-accelerating factor-binding strain of coxsackievirus B3 requires the coxsackievirus-adenovirus receptor protein to mediate lytic infection of rhabdomyosarcoma cells. Journal of Virology 71, 9844-9848 (1997).

26. K. M. Bedard, B. L. Semler, Regulation of picornavirus gene expression. Microbes and Infection 6, 702-713 (2004).

27. F. S. Garmaroudi et al., Coxsackievirus B3 replication and pathogenesis. Future Microbiology 10, 629-652 (2015).

28. R. E. Rhoades, J. M. Tabor-Godwin, G. Tsueng, R. Feuer, Enterovirus infections of the central nervous system. Virology 411, 288-305 (2011).

29. K. R. Spindler, T. H. Hsu, Viral disruption of the blood brain barrier. Trends in Microbiology 20, 282-290 (2012).

30. V. Ambros, The functions of animal microRNAs. Nature 431, 350-355 (2004).

31. D. P. Bartel, MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 116, 281-297 (2004).

32. M. V. Iorio, C. M. Croce, microRNA involvement in human cancer. Carcinogenesis 33, 1126-1133 (2012).

33. E. J. Kelly, E. M. Hadac, S. Greiner, S. J. Russell, Engineering microRNA responsiveness to decrease virus pathogenicity. Nature Medicine 14, 1278-1283 (2008).

34. M. F. Leber et al., MicroRNA-sensitive Oncolytic Measles Viruses for Cancer- specific Vector Tropism. Molecular Therapy 19, 1097-1106 (2011).

35. A. J. Ruiz, E. M. Hadac, R. A. Nace, S. J. Russell, MicroRNA-Detargeted Mengovirus for Oncolytic Virotherapy. Journal of Virology 90, 4078-4092 (2016).

36. M. Shayestehpour et al., Targeting human breast cancer cells by an oncolytic adenovirus using microRNA-targeting strategy. Virus Research 240, 207-214 (2017).

37. L. He et al., A microRNA component of the p53 tumour suppressor network. Nature 447, 1130-U1116 (2007).

38. M. Rokavec, H. H. Li, L. C. Jiang, H. Hermeking, The p53/miR-34 axis in development and disease. Journal of Molecular Cell Biology 6, 214-230 (2014).

39. D. G. Zhang, J. N. Zheng, D. S. Pei, P53/microRNA-34-induced metabolic regulation: new opportunities in anticancer therapy. Molecular Cancer 13, (2014).

40. H. Hermeking, The miR-34 family in cancer and apoptosis. Cell Death and Differentiation 17, 193-199 (2010).

41. R. P. Tomko, R. L. Xu, L. Philipson, HCAR and MCAR: The human and mouse cellular receptors for subgroup C adenoviruses and group B coxsackieviruses. Proceedings of the National Academy of Sciences of the United States of America 94, 3352-3356 (1997).

42. C. B. Coyne, J. M. Bergelson, CAR: A virus receptor within the tight junction. Advanced Drug Delivery Reviews 57, 869-882 (2005).

43. J. Y. Liu, S. M. Wang, I. C. Chen, C. K. Yu, C. C. Liu, Hepatic damage caused by coxsackievirus B3 is dependent on age-related tissue tropisms associated with the coxsackievirus-adenovirus receptor. Pathogens and Disease 68, 52-60 (2013).

44. J. B. Rohll, D. H. Moon, D. J. Evans, J. W. Almond, THE 3'-UNTRANSLATED REGION OF PICORNAVIRUS RNA - FEATURES REQUIRED FOR EFFICIENT GENOME REPLICATION. Journal of Virology 69, 7835-7844 (1995).

45. F. He et al., Coxsackievirus B3 engineered to contain microRNA targets for muscle-specific microRNAs displays attenuated cardiotropic virulence in mice. J Virol 89, 908-916 (2015).

46. B. D. Brown et al., Endogenous microRNA can be broadly exploited to regulate transgene expression according to tissue, lineage and differentiation state. Nature Biotechnology 25, 1457-1467 (2007).

47. K. Fuse et al., Myeloid differentiation factor-88 plays a crucial role in the pathogenesis of coxsackievirus B3-induced myocarditis and influences type I interferon production. Circulation 112, 2276-2285 (2005).

48. S. Pinkert et al., Virus-Host Coevolution in a Persistently Coxsackievirus B3- Infected Cardiomyocyte Cell Line. Journal of Virology 85, 13409-13419 (2011).

49. P. K. M. Cheung et al., Specific interactions of mouse organ proteins with the 5'untranslated region of coxsackievirus B3: Potential determinants of viral tissue tropism. Journal of Medical Virology 77, 414-424 (2005).

50. L. R. Zou et al., A cluster of coxsackievirus A21 associated acute respiratory illness: the evidence of efficient transmission of CVA21. Archives of Virology 162, 1057-1059 (2017).

51. A. J. Ruiz, S. J. Russell, MicroRNAs and oncolytic viruses. Current Opinion in Virology 13, 40-48 (2015).

52. Z. L. Ma et al., MicroRNA-34a inhibits the proliferation and promotes the apoptosis of non-small cell lung cancer H1299 cell line by targeting TGF beta R2. Tumor Biology 36, 2481-2490 (2015).

53. H. B. Sun, J. Tian, W. H. Xian, T. T. Xie, X. D. Yang, miR-34a Inhibits Proliferation and Invasion of Bladder Cancer Cells by Targeting Orphan Nuclear Receptor HNF4G. Disease Markers, (2015).

54. D. Y. Kim et al., A novel miR-34a target, protein kinase D1, stimulates cancer stemness and drug resistance through GSK3/beta-catenin signaling in breast cancer. Oncotarget 7, 14791-14802 (2016).

55. J. Q. Liang et al., LEF1 Targeting EMT in Prostate Cancer Invasion Is Regulated by miR-34a. Molecular Cancer Research 13, 681-688 (2015).

56. E. Gallardo et al., miR-34a as a prognostic marker of relapse in surgically resected non-small-cell lung cancer. Carcinogenesis 30, 1903-1909 (2009).

57. C. S. Song et al., miR-34a sensitizes lung cancer cells to cisplatin via p53/miR- 34a/MYCN axis. Biochemical and Biophysical Research Communications 482, 22-27 (2017).

58. J. F. Wiggins et al., Development of a Lung Cancer Therapeutic Based on the Tumor Suppressor MicroRNA-34. Cancer Research 70, 5923-5930 (2010).

59. D. P. Bartel, MicroRNAs: Target Recognition and Regulatory Functions. Cell 136, 215-233 (2009).

60. B. P. Lewis, C. B. Burge, D. P. Bartel, Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 120, 15-20 (2005).

61. V. Agarwal, G. W. Bell, J. W. Nam, D. P. Bartel, Predicting effective microRNA target sites in mammalian mRNAs. Elife 4, (2015).

62. W. L. Xu, A. San Lucas, Z. X. Wang, Y. Liu, Identifying microRNA targets in different gene regions. Bmc Bioinformatics 15, (2014).

63. D. Barnes, M. Kunitomi, M. Vignuzzi, K. Saksela, R. Andino, Harnessing endogenous miRNAs to control virus tissue tropism as a strategy for developing attenuated virus vaccines. Cell Host & Microbe 4, 239-248 (2008).

64. C. L. Jopling, M. K. Yi, A. M. Lancaster, S. M. Lemon, P. Sarnow, Modulation of hepatitis C virus RNA abundance by a liver-specific microRNA. Science 309, 1577-1581 (2005).

65. Z. W. Liu et al., Structural and functional analysis of the 5 ' untranslated region of coxsackievirus B3 RNA: In vivo translational and infectivity studies of full- length mutants. Virology 265, 206-217 (1999).

66. A. Grimson et al., MicroRNA targeting specificity in mammals: Determinants beyond seed pairing. Molecular Cell 27, 91-105 (2007).

67. K. K. H. Farh et al., The widespread impact of mammalian microRNAs on mRNA repression and evolution. Science 310, 1817-1821 (2005).

68. L. P. Lim et al., Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs. Nature 433, 769-773 (2005).

69. H. Hamzeiy, J. Allmer, M. Yousef, Computational Methods for MicroRNA Target Prediction. Mirnomics: Microrna Biology and Computational Analysis 1107, 207-221 (2014).

70. H. C. Martin et al., Imperfect centered miRNA binding sites are common and can mediate repression of target mRNAs. Genome Biology 15, (2014).

71. P. A. Lodge, M. Herzum, J. Olszewski, S. A. Huber, COXSACKIEVIRUS-B-3 MYOCARDITIS - ACUTE AND CHRONIC FORMS OF THE DISEASE CAUSED BY DIFFERENT IMMUNOPATHOGENIC MECHANISMS. American Journal of Pathology 128, 455-463 (1987).

72. C. Fountzilas, S. Patel, D. Mahalingam, Review: Oncolytic virotherapy, updates and future directions. Oncotarget 8, 102617-102639 (2017).

73. I. Mena et al., Coxsackievirus infection of the pancreas: Evaluation of receptor expression, pathogenesis, and immunopathology. Virology 271, 276-288 (2000).

74. M. T. Barger, J. E. Craighead, IMMUNOMODULATION OF ENCEPHALOMYOCARDITIS VIRUS-INDUCED DISEASE IN A/J MICE. Journal of Virology 65, 2676-2681 (1991).

参考文献をもっと見る

全国の大学の
卒論・修論・学位論文

一発検索!

この論文の関連論文を見る