Targeting and killing of glioblastoma with activated T cells armed with bispecific antibodies

Ian M Zitron, Archana Thakur, Oxana Norkina, Geoffrey R Barger, Lawrence G Lum, Sandeep Mittal, Ian M Zitron, Archana Thakur, Oxana Norkina, Geoffrey R Barger, Lawrence G Lum, Sandeep Mittal

Abstract

Background: Since most glioblastomas express both wild-type EGFR and EGFRvIII as well as HER2/neu, they are excellent targets for activated T cells (ATC) armed with bispecific antibodies (BiAbs) that target EGFR and HER2.

Methods: ATC were generated from PBMC activated for 14 days with anti-CD3 monoclonal antibody in the presence of interleukin-2 and armed with chemically heteroconjugated anti-CD3 × anti-HER2/neu (HER2Bi) and/or anti-CD3 × anti-EGFR (EGFRBi). HER2Bi- and/or EGFRBi-armed ATC were examined for in vitro cytotoxicity using MTT and 51Cr-release assays against malignant glioma lines (U87MG, U118MG, and U251MG) and primary glioblastoma lines.

Results: EGFRBi-armed ATC killed up to 85% of U87, U118, and U251 targets at effector:target ratios (E:T) ranging from 1:1 to 25:1. Engagement of tumor by EGFRBi-armed ATC induced Th1 and Th2 cytokine secretion by armed ATC. HER2Bi-armed ATC exhibited comparable cytotoxicity against U118 and U251, but did not kill HER2-negative U87 cells. HER2Bi- or EGFRBi-armed ATC exhibited 50--80% cytotoxicity against four primary glioblastoma lines as well as a temozolomide (TMZ)-resistant variant of U251. Both CD133- and CD133+ subpopulations were killed by armed ATC. Targeting both HER2Bi and EGFRBi simultaneously showed enhanced efficacy than arming with a single BiAb. Armed ATC maintained effectiveness after irradiation and in the presence of TMZ at a therapeutic concentration and were capable of killing multiple targets.

Conclusion: High-grade gliomas are suitable for specific targeting by armed ATC. These data, together with additional animal studies, may provide the preclinical support for the use of armed ATC as a valuable addition to current treatment regimens.

Figures

Figure 1
Figure 1
Linearity of MTT and 51Cr responses of glioma lines. Upper panel: A representative data showing a linear correlation between OD and cell numbers of U251MG glioma cell line. The indicated numbers of U251MG cells were incubated with MTT for 4 hours under standard conditions. The assay was completed by the solubilization of formazan, the A570 and A650 nm measured, and the difference calculated. Lower panel: Shows correlation between counts per minute (CPM) and % specific cytotoxicity using 51Cr release assay.
Figure 2
Figure 2
BiAb-armed ATC specifically kill long term glioma cell lines. 2A: Non-irradiated or irradiated ATC were armed with the indicated amounts (ng/106 cells) of either EGFRBi (circles) or HER2Bi (triangles) and used as effector cells at an E:T of 5:1. The figure shows the results from one of two donors, with U118MG cells as targets. Both MTT and 51Cr assays were used as readouts. 2B: Shows the data for U87MG, U118MG, and U251MG cells using non-irradiated unarmed ATC or armed ATC at one arming dose of 50 ng for EGFRBi, HER2Bi and CD20Bi at E/T of 5:1. Both MTT and 51Cr assays were used as readouts. 2C: U87MG, U118MG, and U251MG cells were exposed overnight to the indicated effector cells at an E:T of 5:1 and residual viability determined by MTT assay. U87MG expresses only EGFR whereas U118MG and U251MG express both HER2/neu and EGFR. Data were analyzed by 1-way ANOVA: for all 3 glioma lines, overall p < 0.0001; for all 3 lines, unarmed vs. CD20Bi p value is non-significant (p > 0.05) (n.s.). 2C (lower panel): Shows the expression of EGFR and HER2 (blue) compared to isotype control (red), and plots for percent positive cells and mean fluorescence intensity (MFI). For U87MG, Unarmed vs. HER2Bi, p > 0.05 (n.s.); Unarmed vs. EGFRBi, p < 0.001 (***). For U118MG, Unarmed vs. HER2Bi and Unarmed vs. EGFRBi, p < 0.001 (***). For U251MG, Unarmed vs. HER2Bi and Unarmed vs. EGFRBi, p < 0.001 (***).
Figure 3
Figure 3
Tumor cells in primary culture are specifically killed by BiAb-armed ATC. Ex vivo glioma cells expressing both HER2/neu and EGFR were incubated overnight with unarmed ATC or ATC armed with HER2Bi, EGFRBi or CD20Bi. The data are pooled from 4 experiments, using 4 different ATC donors and a range of E:T. Residual viabilities (mean ± SEM) based upon MTT assays are shown. The targets are killed by HER2Bi- and EGFRBi-armed ATC, whereas unarmed and CD20Bi-armed ATC fail to kill. Mean of 4 donors (± SEM), when compared with unarmed ATC, the residual viable cells after HER2Bi- or EGFRBi-ATC were statistically significant (p < 0.001, 1-way ANOVA; ***). Arming with CD20BiAb showed no statistically significant difference from unarmed ATC (p > 0.05, 1-way ANOVA; n.s.). Lower panel: Shows the HER2 (7.15%) and EGFR (9.99%) expression as percent positive cells in this ex vivo cell line.
Figure 4
Figure 4
Targeting ATC to two molecules on the tumor cells’ surfaces enhances killing. Targets were ex vivo cells from two patients with glioblastoma (08–32 and 08–33); both ex vivo lines expressed both HER2/neu and EGFR. The targets were exposed overnight (E:T = 5:1) to unarmed ATC or ATC populations armed with single BiAb (ATC-HER2Bi, ATC-EGFRBi, ATC-CD20Bi), a mixture of equal numbers of singly-armed HER2Bi- and EGFRBi-ATC (ATC-HER2Bi+ATC-EGFRBi) and a population of ATC simultaneously armed with HER2Bi and EGFRBi (ATC-HER2Bi,EGFRBi). Mean residual viability was determined by MTT assay. For both 08-32 and 08-33 target cells, overall analysis by 1-way ANOVA, p < 0.0001. Unarmed or CD20Bi-armed vs. HER2Bi-, EGFRBi and both doubly armed ATC, p < 0.0001 (***). HER2Bi vs. either doubly-armed ATC, p < 0.05 (only for 8-33). EGFRBi vs. HER2Bi+EGFRBi, p < 0.01 (**). EGFRBi vs. HER2Bi,EGFRBi, p < 0.05 (*). Comparisons between individual effector cells performed using Bonferroni multiple comparison test.
Figure 5
Figure 5
Both CD133 enriched and CD133– glioma cells are susceptible targets for BiAb-armed ATC. Cells from an ex vivo glioblastoma were separated into CD133– and CD133 enriched populations using magnetic separation (Miltenyi Biotec). The separated populations, stained with a CD133/2-specific MAb are shown in the inset. Unfractionated (Unfx) cells and CD133– and CD133 enriched populations were incubated overnight with unarmed ATC, or ATC armed with HER2Bi or CD20Bi, at an E:T of 3:1. The mean (± SEM) residual viability was determined using the MTT assay. Lower panel: Show the expression of HER2 and EGFR in CD133 enriched population from two ex vivo primary cells. Overall analysis by 1-way ANOVA, p < 0.0001. Individual comparison (Tukey-Kramer test): unarmed unfx vs. HER2Bi unfx p < 0.001 (***); unarmed unfx vs. CD20Bi unfx p > 0.05 ; unarmed CD133– vs. HER2Bi CD133–p < 0.001 (***); unarmed CD133– vs. CD20Bi CD133–p < 0.001; unarmed CD133+ vs. HER2Bi CD133+p < 0.05 (*); unarmed CD133+ vs. CD20Bi CD133+p < 0.01.
Figure 6
Figure 6
Killing by armed ATC is resistant to both radiation and temozolomide. TMZ-resistant U251MG cells were used as target cells for overnight killing by unarmed, HER2Bi-, and EGFRBi-armed ATC. The upper panel shows the MTT data and the lower panel shows 51Cr release data. Each panel has three sets of 4 columns. The E:T was 10:1. The individual sets represent the effects of unarmed, HER2Bi-armed, and EGFRBi-armed ATC, respectively. Within each data set, the 4 columns show the viability or cytotoxicity, respectively, for type of treatment with and without irradiation or TMZ (final 100 μM). The four sets of effector cells were untreated (Radiation– TMZ–), irradiated only (Radiation+ TMZ–), exposed to TMZ only (100 μM in the assay medium) (Radiation– TMZ+) and both irradiated and exposed to TMZ (Radiation+ TMZ+). Two assay plates were set up in parallel. The MTT data are shown as mean (± SEM) residual percent viable cells and the mean (± SEM) 51Cr release data as percent cytotoxicity. Two-way ANOVA of the MTT data show highly significant effects of both treatment and arming (p < 0.0001 for each), but no effect of interaction (p = 0.27). For the 51Cr data, treatment (p < 0.0001), arming (p < 0.0001) and the interaction of the two (p = 0.0003) are all highly significant.
Figure 7
Figure 7
Armed ATC show sequential killing and effector function is unaffected by contact with glioma cells. In the first overnight culture (Culture 1) unarmed ATC or HER2Bi-armed ATC were incubated with SKBR3 or U251MG target cells (first kill) (E:T = 10:1). The effector cells were removed and MTT added to the wells to determine residual viability compared to control target cells to which no effectors had been added. Data are means (± SEM) of 9–11 cultures. After overnight incubation, without any additional IL-2, the unarmed or HER2Bi-armed ATC were used in a second culture (Culture 2), in which they were added to fresh targets or irrelevant targets (E:T = 10:1). After overnight incubation (second kill), the effectors were removed and MTT added to the wells to determine residual viability of the targets. Data are shown as mean (± SEM) of 4–7 cultures in each group.
Figure 8
Figure 8
Both Th1 and Th2 cytokines are secreted by armed ATC. Unarmed or EGFRBi-armed ATC (107) were incubated overnight alone or with U251MG cells (E:T = 25:1). The values are reported as pg/106/24 hours in 1 ml cultures. The culture supernatant was assayed by Bio-Plex assay for the presence of Th1 and Th2 cytokines. Data shown are for each of the ATC donors individually; the horizontal line indicates the median value. The figure shows the concentrations only for those cytokines which showed differences between effectors alone and effectors plus targets.

References

    1. Dolecek TA, Propp JM, Stroup NE, Kruchko C. CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2005-2009. Neuro Oncol. 2012;14(5):v1–49. doi: 10.1093/neuonc/nos218.
    1. Weiner LM, Clark JI, Davey M, Li WS, Ring DB, Alpaugh RK. Garcia de Palazzo I. Phase I trial of 2B1, a bispecific monoclonal antibody targeting c-erbB-2 and Fc gamma RIII. Cancer Res. 1995;55:4586–4593.
    1. Ren-Heidenreich L, Davol PA, Kouttab NM, Elfenbein GJ, Lum LG. Redirected T-cell cytotoxicity to epithelial cell adhesion molecule-overexpressing adenocarcinomas by a novel recombinant antibody, E3Bi, in vitro and in an animal model. Cancer. 2004;100:1095–1103. doi: 10.1002/cncr.20060.
    1. Reusch U, Sundaram M, Davol PA, Olson SD, Davis JB, Demel K, Nissim J, Rathore R, Liu PY, Lum LG. Anti-CD3 x anti-epidermal growth factor receptor (EGFR) bispecific antibody redirects T-cell cytolytic activity to EGFR-positive cancers in vitro and in an animal model. Clin Cancer Res. 2006;12:183–190. doi: 10.1158/1078-0432.CCR-05-1855.
    1. Chan JK, Hamilton CA, Cheung MK, Karimi M, Baker J, Gall JM, Schulz S, Thorne SH, Teng NN, Contag CH. Enhanced killing of primary ovarian cancer by retargeting autologous cytokine-induced killer cells with bispecific antibodies: a preclinical study. Clin Cancer Res. 2006;12:1859–1867. doi: 10.1158/1078-0432.CCR-05-2019.
    1. Sen M, Wankowski DM, Garlie NK, Siebenlist RE, Van Epps D, LeFever AV, Lum LG. Use of anti-CD3 x anti-HER2/neu bispecific antibody for redirecting cytotoxicity of activated T cells toward HER2/neu+ tumors. J Hematother Stem Cell Res. 2001;10:247–260. doi: 10.1089/15258160151134944.
    1. Gall JM, Davol PA, Grabert RC, Deaver M, Lum LG. T cells armed with anti-CD3 x anti-CD20 bispecific antibody enhance killing of CD20+ malignant B cells and bypass complement-mediated rituximab resistance in vitro. Exp Hematol. 2005;33:452–459. doi: 10.1016/j.exphem.2005.01.007.
    1. Lum HE, Miller M, Davol PA, Grabert RC, Davis JB, Lum LG. Preclinical studies comparing different bispecific antibodies for redirecting T cell cytotoxicity to extracellular antigens on prostate carcinomas. Anticancer Res. 2005;25:43–52.
    1. Yankelevich M, Kondadasula SV, Thakur A, Buck S, Cheung NK, Lum LG. Anti-CD3 x anti-GD2 bispecific antibody redirects T-cell cytolytic activity to neuroblastoma targets. Pediatr Blood Cancer. 2012;59:1198–1205. doi: 10.1002/pbc.24237.
    1. Grabert RC, Cousens LP, Smith JA, Olson S, Gall J, Young WB, Davol PA, Lum LG. Human T cells armed with Her2/neu bispecific antibodies divide, are cytotoxic, and secrete cytokines with repeated stimulation. Clin Cancer Res. 2006;12:569–576. doi: 10.1158/1078-0432.CCR-05-2005.
    1. Davol PA, Smith JA, Kouttab N, Elfenbein GJ, Lum LG. Anti-CD3 x anti-HER2 bispecific antibody effectively redirects armed T cells to inhibit tumor development and growth in hormone-refractory prostate cancer-bearing severe combined immunodeficient beige mice. Clin Prostate Cancer. 2004;3:112–121.
    1. Nitta T, Sato K, Yagita H, Okumura K, Ishii S. Preliminary trial of specific targeting therapy against malignant glioma. Lancet. 1990;335:368–371. doi: 10.1016/0140-6736(90)90205-J.
    1. Yuan X, Curtin J, Xiong Y, Liu G, Waschsmann-Hogiu S, Farkas DL, Black KL, Yu JS. Isolation of cancer stem cells from adult glioblastoma multiforme. Oncogene. 2004;23:9392–9400. doi: 10.1038/sj.onc.1208311.
    1. Lum LG, Ramesh M, Thakur A, Mitra S, Deol A, Uberti JP, Pellett PE. Targeting cytomegalovirus-infected cells using T cells armed with anti-CD3 x anti-CMV bispecific antibody. Biol Blood Marrow Transplant. 2012;18:1012–1022. doi: 10.1016/j.bbmt.2012.01.022.
    1. Thakur A, Norkina O, Lum LG. In vitro synthesis of primary specific anti-breast cancer antibodies by normal human peripheral blood mononuclear cells. Cancer Immunol Immunother. 2011;60:1707–1720. doi: 10.1007/s00262-011-1056-9.
    1. Thakur A, Lum LG. Cancer therapy with bispecific antibodies: Clinical experience. Curr Opin Mol Ther. 2010;12:340–349.
    1. Thakur A, Lum LG, Schalk D, Azmi A, Banerjee S, Sarkar FH, Mohommad R. Pan-Bcl-2 inhibitor AT-101 enhances tumor cell killing by EGFR targeted T cells. PLoS One. 2012;7:e47520. doi: 10.1371/journal.pone.0047520.
    1. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA. 2003;100:3983–3988. doi: 10.1073/pnas.0530291100.
    1. Singh SK, Clarke ID, Hide T, Dirks PB. Cancer stem cells in nervous system tumors. Oncogene. 2004;23:7267–7273. doi: 10.1038/sj.onc.1207946.
    1. Ogden AT, Waziri AE, Lochhead RA, Fusco D, Lopez K, Ellis JA, Kang J, Assanah M, McKhann GM, Sisti MB. Identification of A2B5+CD133- tumor-initiating cells in adult human gliomas. Neurosurgery. 2008;62:505–514. doi: 10.1227/01.neu.0000316019.28421.95.
    1. Mineo JF, Bordron A, Quintin-Roue I, Loisel S, Ster KL, Buhe V, Lagarde N, Berthou C. Recombinant humanised anti-HER2/neu antibody (Herceptin) induces cellular death of glioblastomas. Br J Cancer. 2004;91:1195–1199.
    1. Kolenko V, Wang Q, Riedy MC, O’Shea J, Ritz J, Cathcart MK, Rayman P, Tubbs R, Edinger M, Novick A. Tumor-induced suppression of T lymphocyte proliferation coincides with inhibition of Jak3 expression and IL-2 receptor signaling: role of soluble products from human renal cell carcinomas. J Immunol. 1997;159:3057–3067.
    1. Yamauchi A, Taga K, Mostowski HS, Bloom ET. Target cell-induced apoptosis of interleukin-2-activated human natural killer cells: roles of cell surface molecules and intracellular events. Blood. 1996;87:5127–5135.
    1. Shiraki K, Tsuji N, Shioda T, Isselbacher KJ, Takahashi H. Expression of Fas ligand in liver metastases of human colonic adenocarcinomas. Proc Natl Acad Sci USA. 1997;94:6420–6425. doi: 10.1073/pnas.94.12.6420.
    1. Saas P, Walker PR, Hahne M, Quiquerez AL, Schnuriger V, Perrin G, French L, Van Meir EG, de Tribolet N, Tschopp J, Dietrich PY. Fas ligand expression by astrocytoma in vivo: maintaining immune privilege in the brain? J Clin Invest. 1997;99:1173–1178. doi: 10.1172/JCI119273.
    1. O’Connell J, O’Sullivan GC, Collins JK, Shanahan F. The Fas counterattack: Fas-mediated T cell killing by colon cancer cells expressing Fas ligand. J Exp Med. 1996;184:1075–1082. doi: 10.1084/jem.184.3.1075.
    1. Niehans GA, Brunner T, Frizelle SP, Liston JC, Salerno CT, Knapp DJ, Green DR, Kratzke RA. Human lung carcinomas express Fas ligand. Cancer Res. 1997;57:1007–1012.
    1. Husain N, Chiocca EA, Rainov N, Louis DN, Zervas NT. Co-expression of Fas and Fas ligand in malignant glial tumors and cell lines. Acta Neuropathol. 1998;95:287–290. doi: 10.1007/s004010050799.
    1. Hahne M, Rimoldi D, Schroter M, Romero P, Schreier M, French LE, Schneider P, Bornand T, Fontana A, Lienard D. Melanoma cell expression of Fas(Apo-1/CD95) ligand: implications for tumor immune escape. Science. 1996;274:1363–1366. doi: 10.1126/science.274.5291.1363.
    1. Nam KO, Shin SM, Lee HW. Cross-linking of 4-1BB up-regulates IL-13 expression in CD8(+) T lymphocytes. Cytokine. 2006;33:87–94. doi: 10.1016/j.cyto.2005.12.003.
    1. Kawakami M, Leland P, Kawakami K, Puri RK. Mutation and functional analysis of IL-13 receptors in human malignant glioma cells. Oncol Res. 2001;12:459–467.
    1. Kapp U, Yeh WC, Patterson B, Elia AJ, Kagi D, Ho A, Hessel A, Tipsword M, Williams A, Mirtsos C. Interleukin 13 is secreted by and stimulates the growth of Hodgkin and Reed-Sternberg cells. J Exp Med. 1999;189:1939–1946. doi: 10.1084/jem.189.12.1939.
    1. Oshima Y, Puri RK. Suppression of an IL-13 autocrine growth loop in a human Hodgkin/Reed-Sternberg tumor cell line by a novel IL-13 antagonist. Cell Immunol. 2001;211:37–42. doi: 10.1006/cimm.2001.1828.
    1. Ohshima K, Akaiwa M, Umeshita R, Suzumiya J, Izuhara K, Kikuchi M. Interleukin-13 and interleukin-13 receptor in Hodgkin’s disease: possible autocrine mechanism and involvement in fibrosis. Histopathology. 2001;38:368–375. doi: 10.1046/j.1365-2559.2001.01083.x.
    1. Skinnider BF, Elia AJ, Gascoyne RD, Trumper LH, von Bonin F, Kapp U, Patterson B, Snow BE, Mak TW. Interleukin 13 and interleukin 13 receptor are frequently expressed by Hodgkin and Reed-Sternberg cells of Hodgkin lymphoma. Blood. 2001;97:250–255. doi: 10.1182/blood.V97.1.250.
    1. Formentini A, Prokopchuk O, Strater J, Kleeff J, Grochola LF, Leder G, Henne-Bruns D, Korc M, Kornmann M. Interleukin-13 exerts autocrine growth-promoting effects on human pancreatic cancer, and its expression correlates with a propensity for lymph node metastases. Int J Colorectal Dis. 2009;24:57–67. doi: 10.1007/s00384-008-0550-9.
    1. Heimberger AB, Sampson JH. Immunotherapy coming of age: what will it take to make it standard of care for glioblastoma? Neuro Oncol. 2011;13:3–13. doi: 10.1093/neuonc/noq169.
    1. Jacobs SK, Wilson DJ, Melin G, Parham CW, Holcomb B, Kornblith PL, Grimm EA. Interleukin-2 and lymphokine activated killer (LAK) cells in the treatment of malignant glioma: clinical and experimental studies. Neurol Res. 1986;8:81–87.
    1. Dillman RO, Duma CM, Schiltz PM, DePriest C, Ellis RA, Okamoto K, Beutel LD, De Leon C, Chico S. Intracavitary placement of autologous lymphokine-activated killer (LAK) cells after resection of recurrent glioblastoma. J Immunother. 2004;27:398–404. doi: 10.1097/00002371-200409000-00009.
    1. Hida T, Koike K, Sekido Y, Nishida K, Sugiura T, Ariyoshi Y, Takahashi T, Ueda R. Epitope analysis of cluster 1 and NK cell-related monoclonal antibodies. Br J Cancer Suppl. 1991;14:24–28.
    1. Pfosser A, Brandl M, Salih H, Grosse-Hovest L, Jung G. Role of target antigen in bispecific-antibody-mediated killing of human glioblastoma cells: a pre-clinical study. Int J Cancer. 1999;80:612–616. doi: 10.1002/(SICI)1097-0215(19990209)80:4<612::AID-IJC21>;2-K.
    1. Jung G, Brandl M, Eisner W, Fraunberger P, Reifenberger G, Schlegel U, Wiestler OD, Reulen HJ, Wilmanns W. Local immunotherapy of glioma patients with a combination of 2 bispecific antibody fragments and resting autologous lymphocytes: evidence for in situ t-cell activation and therapeutic efficacy. Int J Cancer. 2001;91:225–230. doi: 10.1002/1097-0215(200002)9999:9999<::AID-IJC1038>;2-7.
    1. Nishimura T, Nakamura Y, Takeuchi Y, Gao XH, Tokuda Y, Okumura K, Habu S. Bispecific antibody-directed antitumor activity of human CD4+ helper/killer T cells induced by anti-CD3 monoclonal antibody plus interleukin 2. Jpn J Cancer Res. 1991;82:1207–1210. doi: 10.1111/j.1349-7006.1991.tb01782.x.
    1. Nishimura T, Nakamura Y, Takeuchi Y, Tokuda Y, Iwasawa M, Kawasaki A, Okumura K, Habu S. Generation propagation, and targeting of human CD4+ helper/killer T cells induced by anti-CD3 monoclonal antibody plus recombinant IL-2. An efficient strategy for adoptive tumor immunotherapy. J Immunol. 1992;148:285–291.
    1. Hishii M, Nitta T, Ebato M, Okumura K, Sato K. Targeting therapy for glioma by LAK cells coupled with bispecific antibodies. J Clin Neurosci. 1994;1:261–265. doi: 10.1016/0967-5868(94)90067-1.
    1. Davico Bonino L, De Monte LB, Spagnoli GC, Vola R, Mariani M, Barone D, Moro AM, Riva P, Nicotra MR, Natali PG. Bispecific monoclonal antibody anti-CD3 x anti-tenascin: an immunotherapeutic agent for human glioma. Int J Cancer. 1995;61:509–515. doi: 10.1002/ijc.2910610414.

Source: PubMed

3
Prenumerera