A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer

Michael H Kershaw, Jennifer A Westwood, Linda L Parker, Gang Wang, Zelig Eshhar, Sharon A Mavroukakis, Donald E White, John R Wunderlich, Silvana Canevari, Linda Rogers-Freezer, Clara C Chen, James C Yang, Steven A Rosenberg, Patrick Hwu, Michael H Kershaw, Jennifer A Westwood, Linda L Parker, Gang Wang, Zelig Eshhar, Sharon A Mavroukakis, Donald E White, John R Wunderlich, Silvana Canevari, Linda Rogers-Freezer, Clara C Chen, James C Yang, Steven A Rosenberg, Patrick Hwu

Abstract

Purpose: A phase I study was conducted to assess the safety of adoptive immunotherapy using gene-modified autologous T cells for the treatment of metastatic ovarian cancer.

Experimental design: T cells with reactivity against the ovarian cancer-associated antigen alpha-folate receptor (FR) were generated by genetic modification of autologous T cells with a chimeric gene incorporating an anti-FR single-chain antibody linked to the signaling domain of the Fc receptor gamma chain. Patients were assigned to one of two cohorts in the study. Eight patients in cohort 1 received a dose escalation of T cells in combination with high-dose interleukin-2, and six patients in cohort 2 received dual-specific T cells (reactive with both FR and allogeneic cells) followed by immunization with allogeneic peripheral blood mononuclear cells.

Results: Five patients in cohort 1 experienced some grade 3 to 4 treatment-related toxicity that was probably due to interleukin-2 administration, which could be managed using standard measures. Patients in cohort 2 experienced relatively mild side effects with grade 1 to 2 symptoms. No reduction in tumor burden was seen in any patient. Tracking 111In-labeled adoptively transferred T cells in cohort 1 revealed a lack of specific localization of T cells to tumor except in one patient where some signal was detected in a peritoneal deposit. PCR analysis showed that gene-modified T cells were present in the circulation in large numbers for the first 2 days after transfer, but these quickly declined to be barely detectable 1 month later in most patients. An inhibitory factor developed in the serum of three of six patients tested over the period of treatment, which significantly reduced the ability of gene-modified T cells to respond against FR+ tumor cells.

Conclusions: Large numbers of gene-modified tumor-reactive T cells can be safely given to patients, but these cells do not persist in large numbers long term. Future studies need to employ strategies to extend T cell persistence. This report is the first to document the use of genetically redirected T cells for the treatment of ovarian cancer.

Figures

Fig. 1
Fig. 1
Growth and phenotype of gene-modified T cells. A, following allogeneic stimulation, 8 × 107 Tcells were transduced with retroviral vector encoding the MOv-γ receptor and maintained at 1to 2 × 106/mL in media containing IL-2. Transduced T cells were restimulated with allogeneic PBMCs on day 21, which resulted in further expansion of Tcells. Using this method, large numbers of dual-specificTcells could be generated. T cell expansion depicted is for patient 9. Representative of all six patients in cohort 2.The phenotype of transduced Tcells from cohort 2 of the study was determined with respect to T cell subset markers and chimeric receptor expression using specific antibodies and flow cytometry. B, although the relative proportions of CD4+ and CD8+ T cells varied between patients, the culture was made up predominantly of CD4+ and CD8+ cells as seen in the representative plot. C, expression of the chimeric MOv-γ receptor was evident following staining with anti-idiotype antibody (thick line) compared with isotype control antibody (thin line). Representative of all patients.
Fig. 2
Fig. 2
Biodistribution of radiolabeled T cells. Patients received up to 7.5 × 109 111In-labeledTcells, and imaging was done using a gamma camera at intervals followingTcell transfer. A, representative image of four transfers that were done withTcells that received a single stimulation with OKT3. B, representative image of three transfers that were done usingTcells that had received two stimulations with OKT3. Radioisotope signal was detected in lungs, liver, and spleen. Radiolabeled cells were preferentially retained in lungs of patients that received twice stimulated Tcells. C, anteroposterior image of the abdomen of patient 4 at 48 hours after receiving T cells in cycle 2 with evidence of T cell localization to a peritoneal tumor (bottom) in addition to localization to liver (top).
Fig. 3
Fig. 3
Preincubation of patient serum with protein G removed inhibitory activity. MOv-γ-transduced T cells were incubated overnight with IGROV-1tumor cells, and antitumor activity was determined by the amount of IFN-γ secreted. T cell activity was inhibited in the presence of serum taken from patient 9 afterTcell transfer. This inhibitory activity of posttreatment serum was removed by pretreatment of serum with protein G.

References

    1. Rosenberg SA, Spiess P, Lafreniere R. A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. Science. 1986;233:1318–21.
    1. Dudley ME, Wunderlich JR, Yang JC, et al. Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. J Clin Oncol. 2005;23:2346–57.
    1. Verri E, Guglielmini P, Puntoni M, et al. HER2/neu oncoprotein overexpression in epithelial ovarian cancer: evaluation of its prevalence and prognostic significance. Clinical study Oncology. 2005;68:154–61.
    1. Thor A, Ohuchi N, Szpak CA, Johnston WW, Schlom J. Distribution of oncofetal antigen tumor-associated glycoprotein-72 defined by monoclonal antibody B723. Cancer Res. 1986;46:3118–24.
    1. Yin BW, Finstad CL, Kitamura K, et al. Serological and immunochemical analysis of Lewis y (Ley) blood group antigen expression in epithelial ovarian cancer. Int J Cancer. 1996;65:406–12.
    1. Coney LR, Tomassetti A, Carayannopoulos L, et al. Cloning of a tumor-associated antigen: MOv18 and MOv19 antibodies recognize a folate-binding protein. Cancer Res. 1991;51:6125–32.
    1. Haynes NM, Trapani JA, Teng MW, et al. Single-chain antigen recognition receptors that costimulate potent rejection of established experimental tumors. Blood. 2002;100:3155–63.
    1. Pinthus JH, Waks T, Malina V, et al. Adoptive immuno-therapy of prostate cancer bone lesions using redirected effector lymphocytes. J Clin Invest. 2004;114:1774–81.
    1. Patel SD, Ge Y, Moskalenko M, McArthur JG. Anti-Tumor CC49-zeta CD4 T cells possess both cytolytic and helper functions. J Immunother. 2000;23:661–8.
    1. Gade TP, Hassen W, Santos E, et al. Targeted elimination of prostate cancer by genetically directed human T lymphocytes. Cancer Res. 2005;65:9080–8.
    1. Haynes NM, Trapani JA, Teng MW, et al. Rejection of syngeneic colon carcinoma by CTLs expressing single-chain antibody receptors codelivering CD28 costimulation. J Immunol. 2002;169:5780–6.
    1. Ho WY, Blattman JN, Dossett ML, Yee C, Greenberg PD. Adoptive immunotherapy: engineering T cell responses as biologic weapons for tumor mass destruction. Cancer Cell. 2003;3:431–7.
    1. Kahlon KS, Brown C, Cooper LJ, Raubitschek A, Forman SJ, Jensen MC. Specific recognition and killing of glioblastoma multiforme by interleukin 13-zetakine redirected cytolytic T cells. Cancer Res. 2004;64:9160–6.
    1. Kershaw MH, Teng MW, Smyth MJ, Darcy PK. Supernatural T cells: genetic modification of T cells for cancer therapy. Nat Rev Immunol. 2005;5:928–40.
    1. Ma Q, Safar M, Holmes E, Wang Y, Boynton AL, Junghans RP. Anti-prostate specific membrane antigen designer T cells for prostate cancer therapy. Prostate. 2004;61:12–25.
    1. Mansoor W, Gilham DE, Thistlethwaite FC, Hawkins RE. Engineering T cells for cancer therapy. Br J Cancer. 2005;93:1085–91.
    1. Rossig C, Brenner MK. Genetic modification of T lymphocytes for adoptive immunotherapy. Mol Ther. 2004;10:5–18.
    1. Miotti S, Canevari S, Menard S, et al. Characterization of human ovarian carcinoma-associated antigens defined by novel monoclonal antibodies with tumor-restricted specificity. Int J Cancer. 1987;39:297–303.
    1. Hwu P, Shafer GE, Treisman J, et al. Lysis of ovarian cancer cells by human lymphocytes redirected with a chimeric gene composed of an antibody variable region and the Fc receptor γ chain. J Exp Med. 1993;178:361–6.
    1. Hwu P, Yang JC, Cowherd R, et al. In vivo antitumor activity of Tcells redirected with chimeric antibody/T-cell receptor genes. Cancer Res. 1995;55:3369–73.
    1. Kershaw MH, Westwood JA, Hwu P. Dual-specific T cells combine proliferation and antitumor activity. Nat Biotechnol. 2002;20:1221–27.
    1. Parker LL, Do MT, Westwood JA, et al. Expansion and characterization of T cells transduced with a chimeric receptor against ovarian cancer. Hum Gene Ther. 2000;11:2377–87.
    1. Benard J, Da Silva J, De Blois MC, et al. Characterization of a human ovarian adenocarcinoma line, IGROV1, in tissue culture and in nude mice. Cancer Res. 1985;45:4970–9.
    1. Mantovani L, Menard S, Mezzanzanica D, Miotti S, Pupa SM, Colnaghi MI. Evaluation of the immunore-active fraction of an anti-tumour monoclonal antibody. Br J Cancer Suppl. 1990;10:15–7.
    1. Mantovani LT, Miotti S, Menard S, et al. Folatebinding protein distribution innormal tissues andbiological fluids from ovarian carcinoma patients as detected by the monoclonal antibodies MOv18 and MOv19. Eur J Cancer. 1994;30A:363–9.
    1. Griffith KD, Read EJ, Carrasquillo JA, et al. In vivo distribution of adoptively transferred indium-111-labeled tumor infiltrating lymphocytes and peripheral blood lymphocytes in patients with metastatic melanoma. J Natl Cancer Inst. 1989;81:1709–17.
    1. Pockaj BA, Sherry RM, Wei JP, et al. Localization of 111Indium-labeled tumor infiltrating lymphocytes to tumor in patients receiving adoptive immunotherapy. Augmentation with cyclophosphamide and correlation with response. Cancer. 1994;73:1731–7.
    1. Miotti S, Negri DR, Valota O, et al. Level of anti-mouse-antibody response induced by bi-specific monoclonal antibody OC/TR in ovarian-carcinoma patients is associated with longer survival. Int J Cancer. 1999;84:62–8.
    1. Dubey P, Su H, Adonai N, et al. Quantitative imaging of the T cell antitumor response by positron-emission tomography. Proc Natl Acad Sci U S A. 2003;100:1232–7.
    1. Freedman RS, Platsoucas CD. Immunotherapy for peritoneal ovarian carcinoma metastasis using ex vivo expanded tumor infiltrating lymphocytes. Cancer Treat Res. 1996;82:115–46.
    1. Gattinoni L, Klebanoff CA, Palmer DC, et al. Acquisition of full effect nor function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ Tcells. JClin Invest. 2005;115:1616–26.
    1. Maher J, Brentjens RJ, Gunset G, Riviere I, Sadelain M. Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRzeta/CD28 receptor. Nat Biotechnol. 2002;20:70–5.
    1. Taylor DD, Edwards RP, Case CR, Gercel-Taylor C. Modulation of CD3-zeta as a marker of clinical response to IL-2 therapy in ovarian cancer patients. Gynecol Oncol. 2004;94:54–60.
    1. Figini M, Obici L, Mezzanzanica D, et al. Panning phage antibody libraries on cells: isolation of human Fab fragments against ovarian carcinoma using guided selection. Cancer Res. 1998;58:991–6.

Source: PubMed

3
Se inscrever