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 「The therapeutic potential of multiclonal tumoricidal T cells derived from tumor infiltrating lymphocyte-derived iPS cells」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

The therapeutic potential of multiclonal tumoricidal T cells derived from tumor infiltrating lymphocyte-derived iPS cells

Ito, Takeshi Kawai, Yohei Yasui, Yutaka Iriguchi, Shoichi Minagawa, Atsutaka Ishii, Tomoko Miyoshi, Hiroyuki Taketo, Mark, M. Kawada, Kenji Obama, Kazutaka Sakai, Yoshiharu Kaneko, Shin 京都大学 DOI:10.1038/s42003-021-02195-x

2021

概要

Tumor-infiltrating lymphocytes (TIL), which include tumor-specific T lymphocytes with frequency, are used for adoptive cell transfer therapy (ACT) in clinical practice. The optimization of TIL preparation has been investigated to reduce the senescence and increase the abundance of TIL, as both the quality and quantity of the transferred cells have great influence on the outcome of TIL-based ACT (TIL-ACT). Considering the effects of cell reprogramming on senescence, we expected that the anti-tumor effect could be enhanced by TIL regeneration. To confirm this hypothesis, we established tumor-specific TIL-derived iPS cells (TIL-iPSC) with human colorectal cancer specimens. T cells differentiated from TIL-iPSC (TIL-iPS-T) retained not only intrinsic T cell functions and tumor specificity, but also exhibited improved proliferation capacity and additional killing activity. Moreover, less differentiated profiles and prolonged persistency were seen in TIL-iPS-T compared with primary cells. Our findings imply that iPSC technology has great potential for TIL-ACT.

この論文で使われている画像

関連論文

参考文献

1.

2.

3.

4.

5.

6.

7.

8.

9.

Alexandrov, L. B., Nik-Zainal, S., Wedge, D. C. & Aparicio, S. A. J. R.

Signatures of mutational processes in human cancer. Nature 500, 415–421

(2013).

Hause, R. J., Pritchard, C. C., Shendure, J. & Salipante, S. J. Classification and

characterization of microsatellite instability across 18 cancer types. Nat. Med.

22, 1342–1350 (2016).

Yarchoan, M., Johnson, B. A., Lutz, E. R., Laheru, D. A. & Jaffee, E. M.

Targeting neoantigens to augment antitumour immunity. Nat. Rev. Cancer 17,

209–222 (2017).

Yarchoan, M., Hopkins, A. & Jaffee, E. M. Tumor mutational burden and

response rate to PD-1 inhibition. N. Engl. J. Med. 377, 2500–2501 (2017).

Le, D. T. et al. Mismatch repair deficiency predicts response of solid tumors to

PD-1 blockade. Science 357, 409–413 (2017).

Lemery, S., Keegan, P. & Pazdur, R. First FDA approval agnostic of cancer site

—when a biomarker defines the indication. N. Engl. J. Med. 337, 1409–1412

(2017).

Lauss, M. et al. Mutational and putative neoantigen load predict clinical

benefit of adoptive T cell therapy in melanoma. Nat. Commun. 8, 1–11

(2017).

Rosenberg, S. A., Spiess, P. & Lafreniere, R. A new approach to the adoptive

immunotherapy of cancer with tumor-infiltrating lymphocytes. Science 233,

1318–1321 (1986).

Topalian, S. L., Muul, L. M., Solomon, D. & Rosenberg, S. A. Expansion of

human tumor infiltrating lymphocytes for use in immunotherapy trials. J.

Immunol. Methods 102, 127–141 (1987).

COMMUNICATIONS BIOLOGY | (2021)4:694 | https://doi.org/10.1038/s42003-021-02195-x | www.nature.com/commsbio

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02195-x

10. Rosenberg, S. A. et al. Use of tumor-infiltrating lymphocytes and interleukin-2

in the immunotherapy of patients with metastatic melanoma. N. Engl. J. Med.

319, 1676–1680 (1988).

11. Dudley, M. E., Wunderlich, J. R., Shelton, T. E., Even, J. & Rosenberg, S. A.

Generation of tumor-infiltrating lymphocyte cultures for use in adoptive

transfer therapy for melanoma patients. J. Immunother. 26, 332–342 (2008).

12. Dudley, M. E. et al. Adoptive cell transfer therapy following nonmyeloablative but lymphodepleting chemotherapy for the treatment of

patients with refractory metastatic melanoma. J. Clin. Oncol. 23, 2346–2357

(2005).

13. Dudley, M. E. et al. Adoptive cell therapy for patients with metastatic

melanoma: evaluation of intensive myeloablative chemoradiation preparative

regimens. J. Clin. Oncol. 26, 5233–5239 (2008).

14. Besser, M. J. et al. Clinical responses in a phase II study using adoptive transfer

of short-term cultured tumor infiltration lymphocytes in metastatic melanoma

patients. Clin. Cancer Res. 16, 2646–2655 (2010).

15. Rosenberg, S. A. et al. Durable complete responses in heavily pretreated

patients with metastatic melanoma using T-cell transfer immunotherapy. Clin.

Cancer Res. 17, 4550–4557 (2011).

16. Radvanyi, L. G. et al. Specific lymphocyte subsets predict response to adoptive

cell therapy using expanded autologous tumor-infiltrating lymphocytes in

metastatic melanoma patients. Clin. Cancer Res. 18, 6758–6770 (2012).

17. Besser, M. J. et al. Adoptive transfer of tumor-infiltrating lymphocytes in

patients with metastatic melanoma: intent-to-treat analysis and efficacy after

failure to prior immunotherapies. Clin. Cancer Res. 19, 4792–4800 (2013).

18. Goff, S. L. et al. Randomized, prospective evaluation comparing intensity of

lymphodepletion before adoptive transfer of tumor-infiltrating lymphocytes

for patients with metastatic melanoma. J. Clin. Oncol. 34, 2389–2397 (2016).

19. Andersen, R. et al. Long-lasting complete responses in patients with metastatic

melanoma after adoptive cell therapy with tumor-infiltrating lymphocytes and

an attenuated IL2 regimen. Clin. Cancer Res. 22, 3734–3745 (2016).

20. Tran, E. et al. Cancer immunotherapy based on mutation-specific CD4+

T cells in a patient with epithelial cancer. Science 344, 641–646 (2014).

21. Tran, E. et al. T-cell transfer therapy targeting mutant KRAS in cancer. N.

Engl. J. Med. 375, 2255–2262 (2016).

22. Stevanović, S. et al. Landscape of immunogenic tumor antigens in successful

immunotherapy of virally induced epithelial cancer. Science 356, 200–205

(2017).

23. Zacharakis, N. et al. Immune recognition of somatic mutations leading to

complete durable regression in metastatic breast cancer. Nat. Med. 24,

724–730 (2018).

24. Dafni, U. et al. Efficacy of adoptive therapy with tumor-infiltrating

lymphocytes and recombinant interleukin-2 in advanced cutaneous

melanoma: a systematic review and meta-analysis. Ann. Oncol. 30, 1902–1913

(2019).

25. Itzhaki, O. et al. Establishment and large-scale expansion of minimally

cultured ‘young’ tumor infiltrating lymphocytes for adoptive transfer therapy.

J. Immunother. 34, 212–220 (2011).

26. Robbins, P. F. et al. Cutting edge: persistence of transferred lymphocyte

clonotypes correlates with cancer regression in patients receiving cell transfer

therapy. J. Immunol. 173, 7125–7130 (2004).

27. Dudley, M. E. et al. CD8+ enriched ‘Young’ tumor infiltrating lymphocytes

can mediate regression of metastatic melanoma. Clin. Cancer Res. 16,

6122–6131 (2010).

28. Zhou, J. et al. Telomere length of transferred lymphocytes correlates with

in vivo persistence and tumor regression in melanoma patients receiving cell

transfer therapy. J. Immunol. 175, 7046–7052 (2005).

29. Butler, M. O. et al. Establishment of antitumor memory in humans using in

vitro-educated CD8+ T cells. Sci. Transl. Med. 3, 80ra34-80ra34 (2011).

30. Crompton, J. G. et al. Akt inhibition enhances expansion of potent tumorspecific lymphocytes with memory cell characteristics. Cancer Res. 75,

296–305 (2015).

31. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse

embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676

(2006).

32. Takahashi, K. et al. Induction of pluripotent stem cells from adult human

fibroblasts by defined factors. Cell 131, 861–872 (2007).

33. Seki, T. et al. Generation of induced pluripotent stem cells from human

terminally differentiated circulating t cells. Cell Stem Cell 7, 11–13 (2010).

34. Loh, Y. H. et al. Reprogramming of T cells from human peripheral blood. Cell

Stem Cell 7, 15–19 (2010).

35. Staerk, J. et al. Reprogramming of human peripheral blood cells to induced

pluripotent stem cells. Cell Stem Cell 7, 20–24 (2010).

36. Seki, T., Yuasa, S. & Fukuda, K. Generation of induced pluripotent stem cells

from a small amount of human peripheral blood using a combination of

activated T cells and Sendai virus. Nat. Protoc. 7, 718–728 (2012).

37. Vizcardo, R. et al. Regeneration of human tumor antigen-specific T cells from

iPSCs derived from mature CD8+ T cells. Cell Stem Cell 12, 31–36 (2013).

ARTICLE

38. Nishimura, T. et al. Generation of rejuvenated antigen-specific T cells by

reprogramming to pluripotency and redifferentiation. Cell Stem Cell 12,

114–126 (2013).

39. Miyoshi, H. & Stappenbeck, T. S. In vitro expansion and genetic modification

of gastrointestinal stem cells in spheroid culture. Nat. Protoc. 8, 2471–2482

(2013).

40. Miyoshi, H. et al. An improved method for culturing patient-derived

colorectal cancer spheroids. Oncotarget 9, 21950–21964 (2018).

41. Turcotte, S. et al. Phenotype and function of T cells infiltrating visceral

metastases from gastrointestinal cancers and melanoma: implications for

adoptive cell transfer therapy. J. Immunol. 191, 2217–2225 (2013).

42. Hall, M. L. et al. Expansion of tumor-infiltrating lymphocytes (TIL) from

human pancreatic tumors. J. Immunother. Cancer 4, 1–12 (2016).

43. Lee, H. J. et al. Expansion of tumor-infiltrating lymphocytes and their

potential for application as adoptive cell transfer therapy in human breast

cancer. Oncotarget 8, 113345–113359 (2017).

44. Savas, P. et al. Single-cell profiling of breast cancer T cells reveals a tissueresident memory subset associated with improved prognosis. Nat. Med. 24,

986–993 (2018).

45. Datar, I. et al. Expression analysis and significance of PD-1, LAG-3, and TIM3 in human non-small cell lung cancer using spatially resolved and

multiparametric single-cell analysis. Clin. Cancer Res. 25, 4663–4673

(2019).

46. Friese, C. et al. CTLA-4 blockade boosts the expansion of tumor-reactive CD8

+ tumor-infiltrating lymphocytes in ovarian cancer. Sci. Rep. 10, 1–12 (2020).

47. Rosenberg, S. A. et al. Treatment of patients with metastatic melanoma with

autologous tumor-infiltrating lymphocytes and interleukin 2. J. Natl Cancer

Inst. 86, 1159–1166 (1994).

48. Santegoets, S. J. A. M. et al. IL-21 promotes the expansion of CD27 + CD28+

tumor infiltrating lymphocytes with high cytotoxic potential and low collateral

expansion of regulatory T cells. J. Transl. Med. 11, 1–10 (2013).

49. Liu, Z. et al. Tumor-infiltrating lymphocytes (TILs) from patients with glioma.

Oncoimmunology 6, e1252894 (2017).

50. Tavera, R. J. et al. Utilizing T-cell activation signals 1, 2, and 3 for tumorinfiltrating lymphocytes (TIL) expansion: the advantage over the sole use of

interleukin-2 in cutaneous and uveal melanoma. J. Immunother. 41, 399–405

(2018).

51. Simoni, Y. et al. Bystander CD8+ T cells are abundant and phenotypically

distinct in human tumour infiltrates. Nature https://doi.org/10.1038/s41586018-0130-2 (2018).

52. Scheper, W. et al. Low and variable tumor reactivity of the intratumoral TCR

repertoire in human cancers. Nat. Med. 25, 89–94 (2019).

53. Ahmadzadeh, M. et al. Tumor antigen-specific CD8 T cells infiltrating the

tumor express high levels of PD-1 and are functionally impaired. Blood 114,

1537–1544 (2009).

54. Ye, Q. et al. CD137 accurately identifies and enriches for naturally occurring

tumor-reactive T cells in tumor. Clin. Cancer Res. 20, 44–55 (2014).

55. Betts, M. R. et al. Sensitive and viable identification of antigen-specific CD8+

T cells by a flow cytometric assay for degranulation. J. Immunol. Methods 281,

65–78 (2003).

56. Wolfl, M. et al. Activation-induced expression of CD137 permits detection,

isolation, and expansion of the full repertoire of CD8+ T cells responding to

antigen without requiring knowledge of epitope specificities. Blood 110,

201–210 (2007).

57. Saito, H., Okita, K., Chang, A. E. & Ito, F. Adoptive transfer of CD8+ T cells

generated from induced pluripotent stem cells triggers regressions of large

tumors along with immunological memory. Cancer Res. 76, 3473–3483

(2016).

58. Maeda, T. et al. Regeneration of CD8αβ T cells from T-cell-derived iPSC

imparts potent tumor antigen-specific cytotoxicity. Cancer Res. 76, 6839–6850

(2016).

59. Minagawa, A. et al. Enhancing T cell receptor stability in rejuvenated iPSCderived T cells improves their use in cancer immunotherapy. Cell Stem Cell 23,

850–858.e4 (2018).

60. Saito, H. et al. Reprogramming of melanoma tumor-infiltrating lymphocytes

to induced pluripotent stem cells. Stem Cells Int. 2016, 1–11 (2016).

61. Yasui, Y., Hitoshi, Y. & Kaneko, S. In vitro differentiation of T cell: from

human iPS cells in feeder-free condition. Methods Mol. Biol. 2048, 77–80

(2019).

62. Iriguchi, S. et al. A clinically applicable and scalable method to regenerate Tcells from iPSCs for off-the-shelf T-cell immunotherapy. Nat. Commun. 12,

1–15 (2021).

63. Ueda, N. et al. Generation of TCR-expressing innate lymphoid-like helper

cells that induce cytotoxic T cell-mediated anti-leukemic cell response. Stem

Cell Rep. 10, 1935–1946 (2018).

64. Menk, A. V. et al. 4-1BB costimulation induces T cell mitochondrial function

and biogenesis enabling cancer immunotherapeutic responses. J. Exp. Med.

215, jem.20171068 (2018).

COMMUNICATIONS BIOLOGY | (2021)4:694 | https://doi.org/10.1038/s42003-021-02195-x | www.nature.com/commsbio

15

ARTICLE

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02195-x

65. Tran, E. et al. Immunogenicity of somatic mutations in human

gastrointestinal cancers. Science 350, 1387–1391 (2015).

66. Chatani, P. D. & Yang, J. C. Mutated RaS: targeting the “untargetable” with

T cells. Clin. Cancer Res. 26, 537–544 (2020).

67. Dijkstra, K. K. et al. Generation of tumor-reactive T cells by co-culture of

peripheral blood lymphocytes and tumor organoids. Cell 174, 1586–1598

(2018).

68. Miyazaki, K. et al. The transcription factor E2A activates multiple enhancers

that drive Rag expression in developing T and B cells. Sci. Immunol. 5,

eabb1455 (2020).

69. Maekawa, H. et al. A chemosensitivity study of colorectal cancer using

xenografts of patient-derived tumor-initiating cells. Mol. Cancer Ther. 17,

2187–2196 (2018).

70. Etxeberria, I. et al. Intratumor adoptive transfer of IL-12 mRNA transiently

engineered antitumor CD8+ T cells. Cancer Cell 36, 613–629 (2019).

provided the spheroids and instructed on the spheroids culture methods. K.K., K.O., and

Y.S. provided the human colorectal cancer specimens. S.K. supervised the study. T. Ito

and S.K. wrote the manuscript.

Competing interests

S.K. is a founder, shareholder, and chief scientific officer at Thyas Co., Ltd. and received

research funding from Thyas Co., Ltd., Takeda Pharmaceutical Co., Ltd., Kirin Co., Ltd.,

TERUMO Co., Ltd., Astellas Pharma Co., Ltd. and Tosoh Co. Ltd. The remaining

authors declare no competing interests.

Additional information

Supplementary information The online version contains supplementary material

available at https://doi.org/10.1038/s42003-021-02195-x.

Correspondence and requests for materials should be addressed to S.K.

Acknowledgements

Reprints and permission information is available at http://www.nature.com/reprints

We thank Shinya Yamanaka for providing critical advice; Takehito Yamamoto for

providing the spheroids; Kanae Mitsunaga for technical assistance with the flow cytometry analysis; Shunsuke Kihara for technical assistance with the time-lapse imaging;

Yoriko Indo, Daisuke Seki and Akito Tanaka for supporting the animal experiments;

Norikazu Saiki, Kohei Ohara, Eri Imai, Kaede Makino, Sanae Kamibayashi, Munehiro

Yoshida, Kengo Nakagoshi, Shuichi Kitayama, Tadayo Miyasaka, Sayaka Okamoto,

Ayako Kumagai, Yoshihiro Iwamoto, Yoshitaka Ishiguro, Masahiro Tanaka, Tatsuki

Ueda, Masazumi Waseda, Akihiro Ishikawa, Bo Wang, Sara Shiina, Reiko Saikawa, Yuta

Mishima, Hitomi Takakubo, Yukie Seto, and Katsura Noda for technical assistance; and

Peter Karagiannis for editing the manuscript. This study was supported in part by a

Grant-in-Aid for Scientific Research (KAKENHI) and collaborative research grant of

Thyas Co., Ltd.

Author contributions

T. Ito and S.K. designed the concept and study. T. Ito performed the experiments and

acquired the data. Y.Y. cooperated with the iPSC establishment from TIL. T. Ito and S.K.

analyzed and interpreted the data. Y.K., S.I., and A.M. provided protocols and advice for

the experiments. T. Ishii cooperated with the in vivo experiments. H.M. and M.M.T.

16

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in

published maps and institutional affiliations.

Open Access This article is licensed under a Creative Commons

Attribution 4.0 International License, which permits use, sharing,

adaptation, distribution and reproduction in any medium or format, as long as you give

appropriate credit to the original author(s) and the source, provide a link to the Creative

Commons license, and indicate if changes were made. The images or other third party

material in this article are included in the article’s Creative Commons license, unless

indicated otherwise in a credit line to the material. If material is not included in the

article’s Creative Commons license and your intended use is not permitted by statutory

regulation or exceeds the permitted use, you will need to obtain permission directly from

the copyright holder. To view a copy of this license, visit http://creativecommons.org/

licenses/by/4.0/.

© The Author(s) 2021

COMMUNICATIONS BIOLOGY | (2021)4:694 | https://doi.org/10.1038/s42003-021-02195-x | www.nature.com/commsbio

...

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