Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2019, Vol. 13 Issue (1) : 57-68    https://doi.org/10.1007/s11684-019-0683-y
RESEARCH ARTICLE
Chimeric antigen receptor T cell targeting EGFRvIII for metastatic lung cancer therapy
Zhao Zhang1,2, Jun Jiang3, Xiaodong Wu1,2, Mengyao Zhang4, Dan Luo1,2, Renyu Zhang1,2, Shiyou Li3(), Youwen He5(), Huijie Bian1,2(), Zhinan Chen1,2()
1. National Translational Science Center for Molecular Medicine, Xi’an 710032, China
2. Department of Cell Biology, Fourth Military Medical University, Xi’an 710032, China
3. Beijing Institute of Genomics, Chinese Academy of Science, Beijing 100101, China
4. Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing 100142, China
5. Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
 Download: PDF(5211 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Lung cancer is the most common incident cancer and the leading cause of cancer death. In recent years, the development of tumor immunotherapy especially chimeric antigen receptor T (CAR-T) cell has shown a promising future. Epidermal growth factor receptor variant III (EGFRvIII) is a tumor-specific mutation expressed in various types of tumors and has been detected in non-small cell lung cancer with a mutation rate of 10%. Thus, EGFRvIII is a potential antigen for targeted lung cancer therapy. In this study, CAR vectors were constructed and transfected into virus-packaging cells. Then, activated T cells were infected with retrovirus harvested from stable virus-producing single clone cell lines. CAR expression on the surfaces of the T cells was detected by flow cytometry and Western blot. The function of CAR-T targeting EGFRvIII was then evaluated. The EGFRvIII-CAR vector was successfully constructed and confirmed by DNA sequencing. A stable virus-producing cell line was produced from a single clone by limited dilution. The culture conditions for the cell line, including cell density, temperature, and culture medium were optimized. After infection with retrovirus, CAR was expressed on more than 90% of the T cells. The proliferation of CAR-T cells were induced by cytokine and specific antigen in vitro. More importantly, EGFRvIII-CART specifically and efficiently recognized and killed A549-EGFRvIII cells with an effector/target ratio of 10:1 by expressing and releasing cytokines, including perforin, granzyme B, IFN-g, and TNF-α. The in vivo study indicated that the metastasis of A549-EGFRvIII cells in mice were inhibited by EGFRvIII-CART cells, and the survival of the mice was significantly prolonged with no serious side effects. EGFRvIII-CART showed significantly efficient antitumor activity against lung cancer cells expressing EGFRvIII in vivo and in vitro. Therefore, CAR-T targeting EGFRvIII is a potential therapeutic strategy in preventing recurrence and metastasis of lung cancer after surgery.

Keywords chimeric antigen receptor T cells      epidermal growth factor receptor      lung cancer      immunotherapy      tumor immunology     
Corresponding Author(s): Shiyou Li,Youwen He,Huijie Bian,Zhinan Chen   
Just Accepted Date: 29 December 2018   Online First Date: 18 February 2019    Issue Date: 12 March 2019
 Cite this article:   
Zhao Zhang,Jun Jiang,Xiaodong Wu, et al. Chimeric antigen receptor T cell targeting EGFRvIII for metastatic lung cancer therapy[J]. Front. Med., 2019, 13(1): 57-68.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-019-0683-y
https://academic.hep.com.cn/fmd/EN/Y2019/V13/I1/57
Fig.1  Expression of chimeric antigen receptor (CAR) of cell membrane. EGFRvIII-CAR was constructed by the extracellular domain of single chain antibody against EGFRvIII, and the intracellular domain consisting of CD28, 4-1BB, and CD3z. MOCK-CAR only contains the transmembrane and intracellular domains.
Fig.2  Establishment of retrovirus-producing cell line and optimization of culture conditions. (A) CAR expression plasmid was transfected into Phoenix Eco cells by lipotamine 2000 (scale bar= 50 μm). (B) PT67 cells were infected by the supernatant of the Phoenix cells (scale bar= 50 μm). (C - E) Virus titer of the producing cell line harvested from RV-E70 cultured in different conditions. (F) Virus titers of RV-E70 and MOCK-C291 virus producing cell line after cultured and passaged for 10 generations. *P<0.05, **P <0.01.
Fig.3  Isolation and infection of PBMCs. (A) Ratio of CD3+CD4+ and CD3+CD8+ T cells in PBMCs before (above) and after (below) cultured in vitro. (B) Expression of CD25 and CD69 before (above) and after (below) T cells were activated by OKT-3 in vitro. (C) Expression of EGFRvIII scFv on uninfected PBMCs (above) and infected PBMCs were detected by FACS analysis. (D) Western blot analysis indicated both wild type CD3ζand chimeric antigen receptor CD3ζ were expressed in CAR-T cells.
Fig.4  Proliferation of PBMCs, MOCK-CART and EGFRvIII-CART in vitro. (A) Cytokine dependent cell proliferation. (B) Antigen dependent cell proliferation. n = 8, **P<0.01.
Fig.5  Cytotoxicity of PBMCs, MOCK-CART and EGFRvIII-CART in vitro. (A) Percentage of target cell lysis at different E:T ratios cocultured with tumor cells in vitro detected by LDH release assay. n = 8, **P<0.01 vs. other groups, ##P<0.01 vs. A549-blank and A549-EGFRvIII-blank groups. (B) Cytotoxicity of PBMCs, MOCK-CART and EGFRvIII-CART analyzed by RTCA system at the E:T ratio of 10:1. n = 3.
Fig.6  Expression and release of cytokines of effector cells cocultured with target cells in vitro. (A) Representative results of expression of IFN-γ, TNF-α, granzyme B and perforin in PBMCs, MOCK-CART and EGFRvIII-CART detected by intracellular staining. (B) Cytokines released from effector cells in the supernatant analyzed by ELISA. n = 6, **P <0.01.
Fig.7  Antitumor response of EGFRvIII-CART cells in vivo. (A) NPG mice were randomized into four groups and received i.v. injections of A549 tumor cells on day 0. Effector cells were injected for 3 times on day 7, 9, and 11 after tumor cell injection. PBMCs were collected once a week, and mice were observed for 90 days. (B) Lung tissue samples of mice in different groups. Normal lung tissue was pink and smooth and had no lesions, and tumor tissues appeared dark red and had granular nodules. (C) Weight of lung tissue in each group. n = 8, **P<0.01. (D) Survival curve of mice in each group. (E) Percentage of human CD3+ T cells in peripheral blood of mice in each groups analyzed by FACS. n = 8.
1 WChen, R Zheng, SZhang, PZhao, H Zeng, XZou, JHe. Annual report on status of cancer in China, 2010. Chin J Cancer Res 2014; 26(1): 48–58
https://doi.org/10.3978/j.issn.1000-9604.2014.01.08 pmid: 24653626
2 WChen, R Zheng, HZeng, SZhang, JHe. Annual report on status of cancer in China, 2011. Chin J Cancer Res 2015; 27(1): 2–12
https://doi.org/10.3978/j.issn.1000-9604.2015.01.06 pmid: 25717220
3 WChen, R Zheng, PDBaade, SZhang, HZeng, F Bray, AJemal, XQYu, J He. Cancer statistics in China, 2015. CA Cancer J Clin 2016; 66(2): 115–132
https://doi.org/10.3322/caac.21338 pmid: 26808342
4 MReck, KF Rabe. Precision diagnosis and treatment for advanced non-small-cell lung cancer. N Engl J Med 2017; 377(9): 849–861
https://doi.org/10.1056/NEJMra1703413 pmid: 28854088
5 NARizvi, S Peters. Immunotherapy for unresectable stage III non-small-cell lung cancer. N Engl J Med 2017; 377(20): 1986–1988
https://doi.org/10.1056/NEJMe1711430 pmid: 29141165
6 ARibas. Adaptive immune resistance: how cancer protects from immune attack. Cancer Discov 2015; 5(9): 915–919
https://doi.org/10.1158/2159-8290.CD-15-0563 pmid: 26272491
7 SARosenberg, MT Lotze, LMMuul, SLeitman, AEChang, SEEttinghausen, YLMatory, JMSkibber, EShiloni, JTVetto, CASeipp, CSimpson, CMReichert. Observations on the systemic administration of autologous lymphokine-activated killer cells and recombinant interleukin-2 to patients with metastatic cancer. N Engl J Med 1985; 313(23): 1485–1492
https://doi.org/10.1056/NEJM198512053132327 pmid: 3903508
8 GGross, T Waks, ZEshhar. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc Natl Acad Sci USA 1989; 86(24): 10024–10028
https://doi.org/10.1073/pnas.86.24.10024 pmid: 2513569
9 SLMaude, N Frey, PAShaw, RAplenc, DMBarrett, NJBunin, AChew, VE Gonzalez, ZZheng, SFLacey, YDMahnke, JJMelenhorst, SRRheingold, AShen, DT Teachey, BLLevine, CHJune, DLPorter, SAGrupp. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med 2014; 371(16): 1507–1517
https://doi.org/10.1056/NEJMoa1407222 pmid: 25317870
10 JHPark, I Rivière, MGonen, XWang, B Sénéchal, KJCurran, CSauter, YWang, B Santomasso, EMead, MRoshal, PMaslak, MDavila, RJBrentjens, MSadelain. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N Engl J Med 2018; 378(5): 449–459
https://doi.org/10.1056/NEJMoa1709919 pmid: 29385376
11 VJindal, E Arora, SGupta. Challenges and prospects of chimeric antigen receptor T cell therapy in solid tumors. Med Oncol 2018; 35(6): 87
https://doi.org/10.1007/s12032-018-1149-9 pmid: 29730801
12 CJWikstrand, LP Hale, SKBatra, MLHill, PAHumphrey, SNKurpad, REMcLendon, DMoscatello, CNPegram, CJReist, STTraweek, AJWong, MRZalutsky and DDBigner. Monoclonal antibodies against EGFRvIII are tumor specific and react with breast and lung carcinomas and malignant gliomas. Cancer Res 1995; 55(14): 3140–3148
pmid: 7606735
13 RNishikawa, XD Ji, RCHarmon, CSLazar, GNGill, WKCavenee, HJHuang. A mutant epidermal growth factor receptor common in human glioma confers enhanced tumorigenicity. Proc Natl Acad Sci USA 1994; 91(16): 7727–7731
https://doi.org/10.1073/pnas.91.16.7727 pmid: 8052651
14 HYu, X Gong, XLuo, WHan, G Hong, BSingh, CKTang. Co-expression of EGFRvIII with ErbB-2 enhances tumorigenesis: EGFRvIII mediated constitutively activated and sustained signaling pathways, whereas EGF-induced a transient effect on EGFR-mediated signaling pathways. Cancer Biol Ther 2008; 7(11): 1818–1828
https://doi.org/10.4161/cbt.7.11.6847 pmid: 18787418
15 RZeineldin, M Rosenberg, DOrtega, CBuhr, MG Chavez, MSStack, DFKusewitt, LGHudson. Mesenchymal transformation in epithelial ovarian tumor cells expressing epidermal growth factor receptor variant III. Mol Carcinog 2006; 45(11): 851–860
https://doi.org/10.1002/mc.20237 pmid: 16788982
16 IEGarcia de Palazzo, GPAdams, PSundareshan, AJWong, JRTesta, DDBigner, LMWeiner. Expression of mutated epidermal growth factor receptor by non-small cell lung carcinomas. Cancer Res 1993; 53(14): 3217–3220
pmid: 8391918
17 IOkamoto, LC Kenyon, DREmlet, TMori, J Sasaki, SHirosako, YIchikawa, HKishi, AKGodwin, MYoshioka, MSuga, M Matsumoto, AJWong. Expression of constitutively activated EGFRvIII in non-small cell lung cancer. Cancer Sci 2003; 94(1): 50–56
https://doi.org/10.1111/j.1349-7006.2003.tb01351.x pmid: 12708474
18 JDuan, Z Wang, HBai, TAn, M Zhuo, MWu, YWang, L Yang, JWang. Epidermal growth factor receptor variant III mutation in Chinese patients with squamous cell cancer of the lung. Thorac Cancer 2015; 6(3): 319–326
https://doi.org/10.1111/1759-7714.12204 pmid: 26273378
19 MOnodera, A Yachie, DMNelson, HWelchlin, RAMorgan, RMBlaese. A simple and reliable method for screening retroviral producer clones without selectable markers. Hum Gene Ther 1997; 8(10): 1189–1194
https://doi.org/10.1089/hum.1997.8.10-1189 pmid: 9215736
20 JSBridgeman, RE Hawkins, SBagley, MBlaylock, MHolland, DEGilham. The optimal antigen response of chimeric antigen receptors harboring the CD3ζ transmembrane domain is dependent upon incorporation of the receptor into the endogenous TCR/CD3 complex. J Immunol 2010; 184(12): 6938–6949
https://doi.org/10.4049/jimmunol.0901766 pmid: 20483753
21 HEHeslop. Safer CARS. Mol Ther 2010; 18(4): 661–662
https://doi.org/10.1038/mt.2010.42 pmid: 20357776
22 CCarpenito, MC Milone, RHassan, JCSimonet, MLakhal, MMSuhoski, AVarela-Rohena, KMHaines, DFHeitjan, SMAlbelda, RGCarroll, JLRiley, IPastan, CHJune. Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains. Proc Natl Acad Sci USA 2009; 106(9): 3360–3365
https://doi.org/10.1073/pnas.0813101106 pmid: 19211796
23 LAlvarez-Vallina, RE Hawkins. Antigen-specific targeting of CD28-mediated T cell co-stimulation using chimeric single-chain antibody variable fragment-CD28 receptors. Eur J Immunol 1996; 26(10): 2304–2309
https://doi.org/10.1002/eji.1830261006 pmid: 8898938
24 MCGong, JB Latouche, AKrause, WDHeston, NHBander, MSadelain. Cancer patient T cells genetically targeted to prostate-specific membrane antigen specifically lyse prostate cancer cells and release cytokines in response to prostate-specific membrane antigen. Neoplasia 1999; 1(2): 123–127
https://doi.org/10.1038/sj.neo.7900018 pmid: 10933046
25 RAMorgan, JC Yang, MKitano, MEDudley, CMLaurencot, SARosenberg. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther 2010; 18(4): 843–851
https://doi.org/10.1038/mt.2010.24 pmid: 20179677
26 KJHatanpaa, S Burma, DZhao, AAHabib. Epidermal growth factor receptor in glioma: signal transduction, neuropathology, imaging, and radioresistance. Neoplasia 2010; 12(9): 675–684
https://doi.org/10.1593/neo.10688 pmid: 20824044
27 LAJohnson, J Scholler, TOhkuri, AKosaka, PRPatel, SEMcGettigan, AKNace, TDentchev, PThekkat, ALoew, AC Boesteanu, APCogdill, TChen, JA Fraietta, CCKloss, ADPosey Jr, BEngels, RSingh, TEzell, NIdamakanti, MHRamones, NLi, L Zhou, GPlesa, JTSeykora, HOkada, CHJune, JLBrogdon, MVMaus. Rational development and characterization of humanized anti-EGFR variant III chimeric antigen receptor T cells for glioblastoma. Sci Transl Med 2015; 7(275): 275ra22
https://doi.org/10.1126/scitranslmed.aaa4963 pmid: 25696001
28 MOhno, T Ohkuri, AKosaka, KTanahashi, CHJune, ANatsume, HOkada. Expression of miR-17-92 enhances anti-tumor activity of T-cells transduced with the anti-EGFRvIII chimeric antigen receptor in mice bearing human GBM xenografts. J Immunother Cancer 2013; 1(1): 21
https://doi.org/10.1186/2051-1426-1-21 pmid: 24829757
29 CHsu, SA Jones, CJCohen, ZZheng, KKerstann, JZhou, PF Robbins, PDPeng, XShen, TJ Gomes, CEDunbar, DJMunroe, CStewart, KCornetta, DWangsa, TRied, SA Rosenberg, RAMorgan. Cytokine-independent growth and clonal expansion of a primary human CD8+ T-cell clone following retroviral transduction with the IL-15 gene. Blood 2007; 109(12): 5168–5177
https://doi.org/10.1182/blood-2006-06-029173 pmid: 17353346
30 SNagaraj, C Ziske, IGSchmidt-Wolf. Human cytokine-induced killer cells have enhanced in vitro cytolytic activity via non-viral interleukin-2 gene transfer. Genet Vaccines Ther 2004; 2(1): 12
https://doi.org/10.1186/1479-0556-2-12 pmid: 15329148
31 JCMarkley, M Sadelain. IL-7 and IL-21 are superior to IL-2 and IL-15 in promoting human T cell-mediated rejection of systemic lymphoma in immunodeficient mice. Blood 2010; 115(17): 3508–3519
https://doi.org/10.1182/blood-2009-09-241398 pmid: 20190192
32 CHsu, MS Hughes, ZZheng, RBBray, SARosenberg, RAMorgan. Primary human T lymphocytes engineered with a codon-optimized IL-15 gene resist cytokine withdrawal-induced apoptosis and persist long-term in the absence of exogenous cytokine. J Immunol 2005; 175(11): 7226–7234
https://doi.org/10.4049/jimmunol.175.11.7226 pmid: 16301627
33 LGattinoni, SE Finkelstein, CAKlebanoff, PAAntony, DCPalmer, PJSpiess, LNHwang, ZYu, C Wrzesinski, DMHeimann, CDSurh, SARosenberg, NPRestifo. Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J Exp Med 2005; 202(7): 907–912
https://doi.org/10.1084/jem.20050732 pmid: 16203864
34 DMO’Rourke, MP Nasrallah, ADesai, JJMelenhorst, KMansfield, JJDMorrissette, MMartinez-Lage, SBrem, E Maloney, AShen, RIsaacs, SMohan, GPlesa, SFLacey, JMNavenot, ZZheng, BLLevine, HOkada, CHJune, JLBrogdon, MVMaus. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci Transl Med 2017; 9(399): eaaa0984
https://doi.org/10.1126/scitranslmed.aaa0984 pmid: 28724573
35 NChen, A Morello, ZTano, PSAdusumilli. CAR T-cell intrinsic PD-1 checkpoint blockade: a two-in-one approach for solid tumor immunotherapy. OncoImmunology 2017; 6(2): e1273302
https://doi.org/10.1080/2162402X.2016.1273302 pmid: 28344886
36 QHuang, J Xia, LWang, XWang, X Ma, QDeng, YLu, M Kumar, ZZhou, LLi, Z Zeng, KHYoung, QYi, M Zhang, YLi. miR-153 suppresses IDO1 expression and enhances CAR T cell immunotherapy. J Hematol Oncol 2018; 11(1): 58
https://doi.org/10.1186/s13045-018-0600-x pmid: 29685162
[1] Yong Fan, Yan Geng, Lin Shen, Zhuoli Zhang. Advances on immune-related adverse events associated with immune checkpoint inhibitors[J]. Front. Med., 2021, 15(1): 33-42.
[2] Hongnan Mo, Binghe Xu. Progress in systemic therapy for triple-negative breast cancer[J]. Front. Med., 2021, 15(1): 1-10.
[3] Ping Li, Ningxin Dong, Yu Zeng, Jie Liu, Xiaochen Tang, Junbang Wang, Wenjun Zhang, Shiguang Ye, Lili Zhou, Alex Hongsheng Chang, Aibin Liang. Chimeric antigen receptor T-cell therapy: a promising treatment modality for relapsed/refractory mantle cell lymphoma[J]. Front. Med., 2020, 14(6): 811-815.
[4] Ching-Hon Pui. Precision medicine in acute lymphoblastic leukemia[J]. Front. Med., 2020, 14(6): 689-700.
[5] Qiaoshuai Lan, Shuai Xia, Qian Wang, Wei Xu, Haiyan Huang, Shibo Jiang, Lu Lu. Development of oncolytic virotherapy: from genetic modification to combination therapy[J]. Front. Med., 2020, 14(2): 160-184.
[6] Jiahui Xu, Qianqian Wang, Elaine Lai Han Leung, Ying Li, Xingxing Fan, Qibiao Wu, Xiaojun Yao, Liang Liu. Compound C620-0696, a new potent inhibitor targeting BPTF, the chromatin-remodeling factor in non-small-cell lung cancer[J]. Front. Med., 2020, 14(1): 60-67.
[7] Yumeng Wang, Guiling Li. PD-1/PD-L1 blockade in cervical cancer: current studies and perspectives[J]. Front. Med., 2019, 13(4): 438-450.
[8] Synat Kang, Yanyan Li, Yifeng Bao, Yi Li. High-affinity T cell receptors redirect cytokine-activated T cells (CAT) to kill cancer cells[J]. Front. Med., 2019, 13(1): 69-82.
[9] Renyu Zhang, Zhao Zhang, Zekun Liu, Ding Wei, Xiaodong Wu, Huijie Bian, Zhinan Chen. Adoptive cell transfer therapy for hepatocellular carcinoma[J]. Front. Med., 2019, 13(1): 3-11.
[10] Kohei Miyazono, Yoko Katsuno, Daizo Koinuma, Shogo Ehata, Masato Morikawa. Intracellular and extracellular TGF-β signaling in cancer: some recent topics[J]. Front. Med., 2018, 12(4): 387-411.
[11] Chia-Wei Li, Yun-Ju Lai, Jennifer L. Hsu, Mien-Chie Hung. Activation of phagocytosis by immune checkpoint blockade[J]. Front. Med., 2018, 12(4): 473-480.
[12] Fang Fang, Weihua Xiao, Zhigang Tian. Challenges of NK cell-based immunotherapy in the new era[J]. Front. Med., 2018, 12(4): 440-450.
[13] Dalin Zhang, Liwei Qu, Bo Zhou, Guizhen Wang, Guangbiao Zhou. Genomic variations in the counterpart normal controls of lung squamous cell carcinomas[J]. Front. Med., 2018, 12(3): 280-288.
[14] Hongbing Shen. Low-dose CT for lung cancer screening: opportunities and challenges[J]. Front. Med., 2018, 12(1): 116-121.
[15] Min Yu, Zonghai Li. Natural killer cells in hepatocellular carcinoma: current status and perspectives for future immunotherapeutic approaches[J]. Front. Med., 2017, 11(4): 509-521.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed