Treatment of malignant pleural mesothelioma by fibroblast activation protein-specific re-directed T cells

Petra C Schuberth, Christian Hagedorn, Shawn M Jensen, Pratiksha Gulati, Maries van den Broek, Axel Mischo, Alex Soltermann, Astrid Jüngel, Osiris Marroquin Belaunzaran, Rolf Stahel, Christoph Renner, Ulf Petrausch, Petra C Schuberth, Christian Hagedorn, Shawn M Jensen, Pratiksha Gulati, Maries van den Broek, Axel Mischo, Alex Soltermann, Astrid Jüngel, Osiris Marroquin Belaunzaran, Rolf Stahel, Christoph Renner, Ulf Petrausch

Abstract

Introduction: Malignant pleural mesothelioma (MPM) is an incurable malignant disease, which results from chronic exposition to asbestos in at least 70% of the cases. Fibroblast activation protein (FAP) is predominantly expressed on the surface of reactive tumor-associated fibroblasts as well as on particular cancer types. Because of its expression on the cell surface, FAP is an attractive target for adoptive T cell therapy. T cells can be re-directed by retroviral transfer of chimeric antigen receptors (CAR) against tumor-associated antigens (TAA) and therefore represent a therapeutic strategy of adoptive immunotherapy.

Methods: To evaluate FAP expression immunohistochemistry was performed in tumor tissue from MPM patients. CD8+ human T cells were retrovirally transduced with an anti-FAP-F19-∆CD28/CD3ζ-CAR. T cell function was evaluated in vitro by cytokine release and cytotoxicity assays. In vivo function was tested with an intraperitoneal xenograft tumor model in immunodeficient mice.

Results: FAP was found to be expressed in all subtypes of MPM. Additionally, FAP expression was evaluated in healthy adult tissue samples and was only detected in specific areas in the pancreas, the placenta and very weakly for cervix and uterus. Expression of the anti-FAP-F19-∆CD28/CD3ζ-CAR in CD8+ T cells resulted in antigen-specific IFNγ release. Additionally, FAP-specific re-directed T cells lysed FAP positive mesothelioma cells and inflammatory fibroblasts in an antigen-specific manner in vitro. Furthermore, FAP-specific re-directed T cells inhibited the growth of FAP positive human tumor cells in the peritoneal cavity of mice and significantly prolonged survival of mice.

Conclusion: FAP re-directed CD8+ T cells showed antigen-specific functionality in vitro and in vivo. Furthermore, FAP expression was verified in all MPM histotypes. Therefore, our data support performing a phase I clinical trial in which MPM patients are treated with adoptively transferred FAP-specific re-directed T cells.

Figures

Figure 1
Figure 1
IHC of fresh-frozen malignant pleural mesothelioma patient sections. A, c and e were stained with the FAP recognizing murine F19 antibody, whereas b, d, f represent the secondary system only. All three histosubtypes of MPM have been analyzed: epithelioid (a, b), sarcomatoid (c, d) and bi-phasic (e, f). Regions marked with * indicate areas of predominant tumor cell location.
Figure 2
Figure 2
Transduction efficacy and purity of CD8+ T cells (a-c) and functional activity of CAR transduced T cells in vitro (d, e, f). Transduction efficacy was assessed by staining the CH2/3 immunoglobulin linker domain with anti-hIgG mAb and with anti-CD8 mAb (n = 10). A shows non-transduced T cells, whereas b displays T cells transduced with the FAP-specific anti-FAP-F19-∆CD28/CD3ζ CAR and c with the NY-ESO-1 specific anti-NY-ESO-1-T1-∆CD28/CD3ζ CAR. 24 h co-cultivation of re-directed T cells with the target cell lines HT1080FAP-luc and T2-1B was analyzed for IFNγ release (n = 5) (d). Comparison of IL-2 production of anti-FAP-F19-∆CD28/CD3ζ and anti-FAP-F19-CD28/CD3ζ after 24 h of co-cultivation with HT1080FAP-luc (n = 2) (e). Proliferation of anti-FAP-F19-∆CD28/CD3ζ re-directed T cells after stimulation with recombinant human FAP (1 μg/ml) in the absence and presence of IL-2 (100 IU/ml) at indicated time points (n = 2) (f). Error bars were calculated as mean + SD (***p < 0.001, **p < 0.01 as calculated by Student’s unpaired 2 tailed t-tests).
Figure 3
Figure 3
Antigen-specific cytolysis of anti-FAP-F19-CD28/CD3ζ re-directed T cells. Re-directed T cells recognizing FAP (anti-FAP-F19-∆CD28/CD3ζ (square) and NY-ESO-1157-165 peptide in the HLA-A*02:01 context (anti-NY-ESO-1-T1-∆CD28/CD3ζ) (circle) were cultured with MSTO-211H cells (a, FAP positive), HT1080FAP-luc cells (b, FAP positive), primary fibroblasts (c, FAP positive), HT1080PA-luc cells (d, FAP negative) and T2-1B cells (e, FAP negative but HLA-A*02:01, NY-ESO-1157-165 peptide positive) at different effector to target ratios and analyzed after 4 h of incubation in a europium release assay (n = 2). Error bars were calculated as mean + SD.
Figure 4
Figure 4
Xenograft model of disseminated i.p. tumors. Anti-FAP-F19-∆CD28/CD3ζ or anti-NY-ESO-1-T1-∆CD28/CD3ζ re-directed T cells were co-injected i.p. with 1*106 HT1080FAP-luc tumor cells at an effector to target ratio of 5:1. HT1080FAP-luc tumor cells were recorded by bioluminescence imaging in the same mouse at the indicated days starting day 4 after cell injection (a). A total of 11 mice per experimental setting were treated with re-directed T cells from 2 different donors (donor 1: 6 mice per group, donor 2: 5 mice per group). The median survival of mice was determined by Kaplan-Meier analysis (b). Significance between cohorts was determined by log rank test.

References

    1. Raja S, Murthy SC, Mason DP. Malignant pleural mesothelioma. Curr Oncol Rep. 2011;13:259–264. doi: 10.1007/s11912-011-0177-9.
    1. Teta MJ, Mink PJ, Lau E, Sceurman BK, Foster ED. US mesothelioma patterns 1973–2002: indicators of change and insights into background rates. Eur J Cancer Prev. 2008;17:525–534. doi: 10.1097/CEJ.0b013e3282f0c0a2.
    1. Opitz I, Kestenholz P, Lardinois D, Muller M, Rousson V, Schneiter D, Stahel R, Weder W. Incidence and management of complications after neoadjuvant chemotherapy followed by extrapleural pneumonectomy for malignant pleural mesothelioma. Eur J Cardiothorac Surg. 2006;29:579–584. doi: 10.1016/j.ejcts.2006.01.015.
    1. Weder W, Opitz I, Stahel R. Multimodality strategies in malignant pleural mesothelioma. Semin Thorac Cardiovasc Surg. 2009;21:172–176. doi: 10.1053/j.semtcvs.2009.07.004.
    1. Sugarbaker DJ, Garcia JP, Richards WG, Harpole DH Jr, Healy-Baldini E, DeCamp MM Jr, Mentzer SJ, Liptay MJ, Strauss GM, Swanson SJ. Extrapleural pneumonectomy in the multimodality therapy of malignant pleural mesothelioma. Results in 120 consecutive patients. Ann Surg. 1996;224:288–294. doi: 10.1097/00000658-199609000-00005. discussion 294–286.
    1. Ertl HC, Zaia J, Rosenberg SA, June CH, Dotti G, Kahn J, Cooper LJ, Corrigan-Curay J, Strome SE. Considerations for the clinical application of chimeric antigen receptor T cells: observations from a recombinant DNA advisory committee symposium held June 15, 2010. Cancer Res. 2011;71:3175–3181. doi: 10.1158/0008-5472.CAN-10-4035.
    1. Kershaw MH, Teng MW, Smyth MJ, Darcy PK. Supernatural T cells: genetic modification of T cells for cancer therapy. Nat Rev Immunol. 2005;5:928–940. doi: 10.1038/nri1729.
    1. Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365:725–733. doi: 10.1056/NEJMoa1103849.
    1. Kershaw MH, Westwood JA, Parker LL, Wang G, Eshhar Z, Mavroukakis SA, White DE, Wunderlich JR, Canevari S, Rogers-Freezer L. et al.A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Canc Res. 2006;12:6106–6115. doi: 10.1158/1078-0432.CCR-06-1183.
    1. Louis CU, Savoldo B, Dotti G, Pule M, Yvon E, Myers GD, Rossig C, Russell HV, Diouf O, Liu E. et al.Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood. 2011;118:6050–6056. doi: 10.1182/blood-2011-05-354449.
    1. Stewart-Jones G, Wadle A, Hombach A, Shenderov E, Held G, Fischer E, Kleber S, Nuber N, Stenner-Liewen F, Bauer S. et al.Rational development of high-affinity T-cell receptor-like antibodies. Proc Natl Acad Sci U S A. 2009;106:5784–5788. doi: 10.1073/pnas.0901425106.
    1. Schuberth PC, Jakka G, Jensen SM, Wadle A, Gautschi F, Haley D, Haile S, Mischo A, Held G, Thiel M. et al.Effector memory and central memory NY-ESO-1-specific re-directed T cells for treatment of multiple myeloma. Gene Ther. 2012;20:386–395.
    1. Garin-Chesa P, Old LJ, Rettig WJ. Cell surface glycoprotein of reactive stromal fibroblasts as a potential antibody target in human epithelial cancers. Proc Natl Acad Sci U S A. 1990;87:7235–7239. doi: 10.1073/pnas.87.18.7235.
    1. Rettig WJ, Garin-Chesa P, Healey JH, Su SL, Ozer HL, Schwab M, Albino AP, Old LJ. Regulation and heteromeric structure of the fibroblast activation protein in normal and transformed cells of mesenchymal and neuroectodermal origin. Canc Res. 1993;53:3327–3335.
    1. Park JE, Lenter MC, Zimmermann RN, Garin-Chesa P, Old LJ, Rettig WJ. Fibroblast activation protein, a dual specificity serine protease expressed in reactive human tumor stromal fibroblasts. J Biol Chem. 1999;274:36505–36512. doi: 10.1074/jbc.274.51.36505.
    1. Dohi O, Ohtani H, Hatori M, Sato E, Hosaka M, Nagura H, Itoi E, Kokubun S. Histogenesis-specific expression of fibroblast activation protein and dipeptidylpeptidase-IV in human bone and soft tissue tumours. Histopathology. 2009;55:432–440. doi: 10.1111/j.1365-2559.2009.03399.x.
    1. Scanlan MJ, Raj BK, Calvo B, Garin-Chesa P, Sanz-Moncasi MP, Healey JH, Old LJ, Rettig WJ. Molecular cloning of fibroblast activation protein alpha, a member of the serine protease family selectively expressed in stromal fibroblasts of epithelial cancers. Proc Natl Acad Sci U S A. 1994;91:5657–5661. doi: 10.1073/pnas.91.12.5657.
    1. Jacob M, Chang L, Pure E. Fibroblast activation protein in remodeling tissues. Curr Mol Med. 2012;12:1220–1243. doi: 10.2174/156652412803833607.
    1. Kraman M, Bambrough PJ, Arnold JN, Roberts EW, Magiera L, Jones JO, Gopinathan A, Tuveson DA, Fearon DT. Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein-alpha. Science. 2010;330:827–830. doi: 10.1126/science.1195300.
    1. Scott AM, Wiseman G, Welt S, Adjei A, Lee FT, Hopkins W, Divgi CR, Hanson LH, Mitchell P, Gansen DN. et al.A phase I dose-escalation study of sibrotuzumab in patients with advanced or metastatic fibroblast activation protein-positive cancer. Clin Canc Res. 2003;9:1639–1647.
    1. Hofheinz RD, al-Batran SE, Hartmann F, Hartung G, Jager D, Renner C, Tanswell P, Kunz U, Amelsberg A, Kuthan H, Stehle G. Stromal antigen targeting by a humanised monoclonal antibody: an early phase II trial of sibrotuzumab in patients with metastatic colorectal cancer. Onkologie. 2003;26:44–48. doi: 10.1159/000069863.
    1. Welt S, Divgi CR, Scott AM, Garin-Chesa P, Finn RD, Graham M, Carswell EA, Cohen A, Larson SM, Old LJ. et al.Antibody targeting in metastatic colon cancer: a phase I study of monoclonal antibody F19 against a cell-surface protein of reactive tumor stromal fibroblasts. J Clin Oncol. 1994;12:1193–1203.
    1. Kofler DM, Chmielewski M, Rappl G, Hombach A, Riet T, Schmidt A, Hombach AA, Wendtner CM, Abken H. CD28 costimulation Impairs the efficacy of a redirected t-cell antitumor attack in the presence of regulatory t cells which can be overcome by preventing Lck activation. Mol Ther. 2011;19:760–767. doi: 10.1038/mt.2011.9.
    1. Willemsen RA, Ronteltap C, Chames P, Debets R, Bolhuis RL. T cell retargeting with MHC class I-restricted antibodies: the CD28 costimulatory domain enhances antigen-specific cytotoxicity and cytokine production. J Immunol. 2005;174:7853–7858.
    1. Bauer S, Adrian N, Williamson B, Panousis C, Fadle N, Smerd J, Fettah I, Scott AM, Pfreundschuh M, Renner C. Targeted bioactivity of membrane-anchored TNF by an antibody-derived TNF fusion protein. J Immunol. 2004;172:3930–3939.
    1. Held G, Matsuo M, Epel M, Gnjatic S, Ritter G, Lee SY, Tai TY, Cohen CJ, Old LJ, Pfreundschuh M. et al.Dissecting cytotoxic T cell responses towards the NY-ESO-1 protein by peptide/MHC-specific antibody fragments. Eur J Immunol. 2004;34:2919–2929. doi: 10.1002/eji.200425297.
    1. Ospelt C, Mertens JC, Jungel A, Brentano F, Maciejewska-Rodriguez H, Huber LC, Hemmatazad H, Wuest T, Knuth A, Gay RE. et al.Inhibition of fibroblast activation protein and dipeptidylpeptidase 4 increases cartilage invasion by rheumatoid arthritis synovial fibroblasts. Arthritis Rheum. 2010;62:1224–1235. doi: 10.1002/art.27395.
    1. Payeli SK, Kollnberger S, Marroquin Belaunzaran O, Thiel M, McHugh K, Giles J, Shaw J, Kleber S, Ridley A, Wong-Baeza I. et al.Inhibiting HLA-B27 homodimer-driven immune cell inflammation in spondylarthritis. Arthritis Rheum. 2012;64:3139–3149. doi: 10.1002/art.34538.
    1. Weijtens ME, Willemsen RA, van Krimpen BA, Bolhuis RL. Chimeric scFv/gamma receptor-mediated T-cell lysis of tumor cells is coregulated by adhesion and accessory molecules. Int J Canc. 1998;77:181–187. doi: 10.1002/(SICI)1097-0215(19980717)77:2<181::AID-IJC2>;2-M.
    1. Hombach A, Hombach AA, Abken H. Adoptive immunotherapy with genetically engineered T cells: modification of the IgG1 Fc 'spacer' domain in the extracellular moiety of chimeric antigen receptors avoids 'off-target' activation and unintended initiation of an innate immune response. Gene Ther. 2010;17:1206–1213. doi: 10.1038/gt.2010.91.
    1. Petrausch U, Schuberth PC, Hagedorn C, Soltermann A, Tomaszek S, Stahel R, Weder W, Renner C. Re-directed T cells for the treatment of fibroblast activation protein (FAP)-positive malignant pleural mesothelioma (FAPME-1) BMC Canc. 2012;12:615. doi: 10.1186/1471-2407-12-615.
    1. Stahel RA, Felley-Bosco E, Opitz I, Weder W. Malignant pleural mesothelioma. Future Oncol. 2009;5:391–402. doi: 10.2217/fon.09.7.
    1. Jakobsen JN, Sorensen JB. Review on clinical trials of targeted treatments in malignant mesothelioma. Canc Chemother Pharmacol. 2011;68:1–15. doi: 10.1007/s00280-011-1655-3.
    1. Izzi V, Masuelli L, Tresoldi I, Foti C, Modesti A, Bei R. Immunity and malignant mesothelioma: from mesothelial cell damage to tumor development and immune response-based therapies. Canc Lett. 2012;322:18–34. doi: 10.1016/j.canlet.2012.02.034.
    1. Gattinoni L, Lugli E, Ji Y, Pos Z, Paulos CM, Quigley MF, Almeida JR, Gostick E, Yu Z, Carpenito C. et al.A human memory T cell subset with stem cell-like properties. Nat Med. 2011;17:1290–1297. doi: 10.1038/nm.2446.
    1. Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, Rosenberg SA. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther. 2010;18:843–851. doi: 10.1038/mt.2010.24.
    1. Bauer S, Jendro MC, Wadle A, Kleber S, Stenner F, Dinser R, Reich A, Faccin E, Godde S, Dinges H. et al.Fibroblast activation protein is expressed by rheumatoid myofibroblast-like synoviocytes. Arthritis Res Ther. 2006;8:R171. doi: 10.1186/ar2080.
    1. Brokopp CE, Richards P, Bauer S, Aikawa E, Graves K, Vogt P, Renner C, Luescher TF, Hoerstrup SP, Matter CM. Fibroblast activation protein is induced by inflammation and degrades collagen in vulnerable human atherosclerotic plaques. European Heart Journal. 2010;31:829–829.
    1. Brokopp CE, Schoenauer R, Richards P, Bauer S, Lohmann C, Emmert MY, Weber B, Winnik S, Aikawa E, Graves K. et al.Fibroblast activation protein is induced by inflammation and degrades type I collagen in thin-cap fibroatheromata. Eur Heart J. 2011;32:2713–2722. doi: 10.1093/eurheartj/ehq519.
    1. Friedmann-Morvinski D, Bendavid A, Waks T, Schindler D, Eshhar Z. Redirected primary T cells harboring a chimeric receptor require costimulation for their antigen-specific activation. Blood. 2005;105:3087–3093. doi: 10.1182/blood-2004-09-3737.
    1. Krause A, Guo HF, Latouche JB, Tan C, Cheung NK, Sadelain M. Antigen-dependent CD28 signaling selectively enhances survival and proliferation in genetically modified activated human primary T lymphocytes. J Exp Med. 1998;188:619–626. doi: 10.1084/jem.188.4.619.
    1. Cooper D, Bansal-Pakala P, Croft M. 4-1BB (CD137) controls the clonal expansion and survival of CD8 T cells in vivo but does not contribute to the development of cytotoxicity. Eur J Immunol. 2002;32:521–529. doi: 10.1002/1521-4141(200202)32:2<521::AID-IMMU521>;2-X.
    1. Hacein-Bey-Abina S, Hauer J, Lim A, Picard C, Wang GP, Berry CC, Martinache C, Rieux-Laucat F, Latour S, Belohradsky BH. et al.Efficacy of gene therapy for X-linked severe combined immunodeficiency. N Engl J Med. 2010;363:355–364. doi: 10.1056/NEJMoa1000164.
    1. Scholler J, Brady TL, Binder-Scholl G, Hwang WT, Plesa G, Hege KM, Vogel AN, Kalos M, Riley JL, Deeks SG. et al.Decade-long safety and function of retroviral-modified chimeric antigen receptor T cells. Sci Transl Med. 2012;4:132ra153.
    1. Parente-Pereira AC, Burnet J, Ellison D, Foster J, Davies DM, van der Stegen S, Burbridge S, Chiapero-Stanke L, Wilkie S, Mather S, Maher J. Trafficking of CAR-engineered human T cells following regional or systemic adoptive transfer in SCID beige mice. J Clin Immunol. 2011;31:710–718. doi: 10.1007/s10875-011-9532-8.

Source: PubMed

3
구독하다