Transfusionsmedizin 2014; 4(4): 179-183
DOI: 10.1055/s-0034-1383140
Übersicht
Georg Thieme Verlag KG Stuttgart · New York

Designer-T-Zellen: Können T-Zellen mit definierter Spezifität die adoptive Immuntherapie zum Erfolg führen?

Designer T Cells: Can T Cells with Defined Specificity Make Adoptive Cell Therapy Successful?
H. Abken
1   Tumorgenetik, Klinik I für Innere Medizin, Uniklinik Köln
2   Zentrum für Molekulare Medizin Köln (ZMMK), Universität zu Köln
› Author Affiliations
Further Information

Publication History

Publication Date:
21 November 2014 (online)

Zusammenfassung

Die adoptive Zelltherapie mit modifizierten T-Zellen hat kürzlich spektakuläre Erfolge in der Behandlung refraktärer Leukämien erzielt. Die Strategie beruht darauf, dass Patienten-T-Zellen durch einen chimären Antigenrezeptor (CAR) definierte Spezifität verliehen wird, sodass sie Zielzellen im Patienten gezielt aufspüren können. Der CAR erkennt mithilfe einer antikörperabgeleiteten Bindedomäne seine Zielstruktur unabhängig vom Histokompatibilitätskomplex und aktiviert die T-Zelle, was zur Zytokinfreisetzung und Lyse der Zielzelle führt. In der klinischen Anwendung werden die T-Zellen aus dem peripheren Blut des Patienten isoliert, ex vivo mit einem viralen Vektor zur Expression des CAR modifiziert, zu therapeutischen Zellzahlen amplifiziert und dem Patienten transfundiert. Die CAR-T-Zellen akkumulieren in Geweben mit der CAR-Zielstruktur, eliminieren die erkannten Tumorzellen und bilden ein spezifisches immunologisches Gedächtnis aus. Aufgrund dieser Eigenschaften hat die CAR-T-Zell-Strategie besonderes Interesse in der Onkologie erlangt. Jüngste Phase-I-Studien unterstreichen die Hoffnung, dass patienteneigene T-Zellen mit einem tumorspezifischen CAR eine Remission erzielen und den Tumor langfristig kontrollieren können.

Abstract

Adoptive cell therapy with modified T cells has recently gained spectacular success in the treatment of refractory leukemia. The strategy is based on the engraftment of defined specificity to patientʼs T cells by a chimeric antigen receptor (CAR) in order to specifically retrieve targeted cells. The CAR recognizes its target by an antibody-derived binding domain independently of the major histocompatibility complex and activates the T cell resulting in cytokine release and lysis of target cells. For clinical application T cells are isolated from patientʼs blood, modified ex vivo by a viral vector to express the CAR, amplified to therapeutic cell numbers and administered to the patient. CAR T cells accumulate in tissues with the CAR target, eliminate targeted tumor cells and build up an immunologic memory. Based on these properties the CAR T cell strategy has gained particular interest in oncology. Recent phase I trials sustain hope that patientʼs T cells engineered with a tumor-specific CAR induce remission and control tumors in the long term.

 
  • Literatur

  • 1 Kalos M, Levine BL, Porter DL et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med 2011; 3: 95ra73
  • 2 Porter DL, Levine BL, Kalos M et al. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med 2011; 365: 725-733
  • 3 Grupp SA, Kalos M, Barrett D et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med 2013; 368: 1509-1518
  • 4 Eshhar Z. The T-body approach: redirecting T cells with antibody specificity. Handb Exp Pharmacol 2008; 181: 329-342
  • 5 Hombach A, Abken H. Costimulation tunes tumor-specific activation of redirected T cells in adoptive immunotherapy. Cancer Immunol Immunother 2007; 56: 731-737
  • 6 James SE, Greenberg PD, Jensen MC et al. Antigen sensitivity of CD22-specific chimeric TCR is modulated by target epitope distance from the cell membrane. J Immunol 2008; 180: 7028-7038
  • 7 Till BG, Jensen MC, Wang J et al. CD20-specific adoptive immunotherapy for lymphoma using a chimeric antigen receptor with both CD28 and 4-1BB domains: pilot clinical trial results. Blood 2012; 119: 3940-3950
  • 8 Chmielewski M, Hombach A, Heuser C et al. T cell activation by antibody-like immunoreceptors: increase in affinity of the single-chain fragment domain above threshold does not increase T cell activation against antigen-positive target cells but decreases selectivity. J Immunol 2004; 173: 7647-7653
  • 9 Zhong S, Malecek K, Johnson LA et al. T-cell receptor affinity and avidity defines antitumor response and autoimmunity in T-cell immunotherapy. Proc Natl Acad Sci U S A 2013; 110: 6973-6978
  • 10 Mezzanzanica D, Canevari S, Mazzoni A et al. Transfer of chimeric receptor gene made of variable regions of tumor-specific antibody confers anticarbohydrate specificity on T cells. Cancer Gene Ther 1998; 5: 401-407
  • 11 Hombach A, Heuser C, Sircar R et al. T cell targeting of TAG72+ tumor cells by a chimeric receptor with antibody-like specificity for a carbohydrate epitope. Gastroenterology 1997; 113: 1163-1170
  • 12 Westwood JA, Smyth MJ, Teng MW et al. Adoptive transfer of T cells modified with a humanized chimeric receptor gene inhibits growth of Lewis-Y-expressing tumors in mice. Proc Natl Acad Sci U S A 2005; 102: 19051-19056
  • 13 Hombach AA, Schildgen V, Heuser C et al. T cell activation by antibody-like immunoreceptors: the position of the binding epitope within the target molecule determines the efficiency of activation of redirected T cells. J Immunol 2007; 178: 4650-4657
  • 14 Hombach AA, Abken H. Of chimeric antigen receptors and antibodies: OX40 and 41BB costimulation sharpen up T cell-based immunotherapy of cancer. Immunotherapy 2013; 5: 677-681
  • 15 Beecham EJ, Ma Q, Ripley R et al. Coupling CD28 co-stimulation to immunoglobulin T-cell receptor molecules: the dynamics of T-cell proliferation and death. J Immunother 2000; 23: 631-642
  • 16 Klebanoff CA, Gattinoni L, Torabi-Parizi P et al. Central memory self/tumor-reactive CD8+ T cells confer superior antitumor immunity compared with effector memory T cells. Proc Natl Acad Sci U S A 2005; 102: 9571-9576
  • 17 Kershaw MH, Westwood JA, Parker LL et al. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res 2006; 12: 6106-6115
  • 18 Brentjens RJ, Davila ML, Riviere I et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med 2013; 5: 177ra138
  • 19 Chmielewski M, Hombach AA, Abken H. Of CARs and TRUCKs: chimeric antigen receptor (CAR) T cells engineered with an inducible cytokine to modulate the tumor stroma. Immunol Rev 2014; 257: 83-90
  • 20 Singh H, Figliola MJ, Dawson MJ et al. Manufacture of clinical-grade CD19-specific T cells stably expressing chimeric antigen receptor using Sleeping Beauty system and artificial antigen presenting cells. PLoS One 2013; 8: e64138
  • 21 Zhao Y, Moon E, Carpenito C et al. Multiple injections of electroporated autologous T cells expressing a chimeric antigen receptor mediate regression of human disseminated tumor. Cancer Res 2010; 70: 9053-9061
  • 22 Kaneko S, Mastaglio S, Bondanza A et al. IL-7 and IL-15 allow the generation of suicide gene-modified alloreactive self-renewing central memory human T lymphocytes. Blood 2009; 113: 1006-1015
  • 23 Morgan RA, Yang JC, Kitano M et al. 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
  • 24 Parente-Pereira AC, Burnet J, Ellison D et al. Trafficking of CAR-engineered human T cells following regional or systemic adoptive transfer in SCID beige mice. J Clin Immunol 2011; 31: 710-718
  • 25 Weber J, Atkins M, Hwu P et al. White paper on adoptive cell therapy for cancer with tumor-infiltrating lymphocytes: a report of the CTEP subcommittee on adoptive cell therapy. Clin Cancer Res 2011; 17: 1664-1673
  • 26 Hombach AA, Chmielewski M, Rappl G et al. Adoptive immunotherapy with redirected T cells produces CCR7- cells that are trapped in the periphery and benefit from combined CD28-OX40 costimulation. Hum Gene Ther 2013; 24: 259-269
  • 27 Song DG, Ye Q, Carpenito C et al. In vivo persistence, tumor localization, and antitumor activity of CAR-engineered T cells is enhanced by costimulatory signaling through CD137 (4-1BB). Cancer Res 2011; 71: 4617-4627
  • 28 Moon EK, Carpenito C, Sun J et al. Expression of a functional CCR2 receptor enhances tumor localization and tumor eradication by retargeted human T cells expressing a mesothelin-specific chimeric antibody receptor. Clin Cancer Res 2011; 17: 4719-4730
  • 29 Lamers CH, Sleijfer S, Vulto AG et al. Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience. J Clin Oncol 2006; 24: e20-e22
  • 30 Chmielewski M, Kopecky C, Hombach AA et al. IL-12 release by engineered T cells expressing chimeric antigen receptors can effectively Muster an antigen-independent macrophage response on tumor cells that have shut down tumor antigen expression. Cancer Res 2011; 71: 5697-5706