HER2 as a promising target for cytotoxicity T cells in human melanoma therapy

Juan Ma, Huamin Han, Deruo Liu, Wei Li, Hongxiang Feng, Xin Xue, Xiaoran Wu, Ge Niu, Ge Zhang, Yunfeng Zhao, Changzhen Liu, Hua Tao, Bin Gao, Juan Ma, Huamin Han, Deruo Liu, Wei Li, Hongxiang Feng, Xin Xue, Xiaoran Wu, Ge Niu, Ge Zhang, Yunfeng Zhao, Changzhen Liu, Hua Tao, Bin Gao

Abstract

Anti-HER2/neu antibody therapy has been reported to mediate tumor regression of HER2/ neu(+) tumors. Here we demonstrated the expression of HER2 in a wide range of human melanoma cells including a primary culture and seven cell lines, and we further investigated whether HER2 could be served as a target for T cell mediated immunotherapy of human melanoma. Specific cytolytic activity of activated T cells (ATC) armed with anti-CD3 x anti-HER2 bispecific antibody (HER2Bi-Ab) against Malme-3M-luc cells was evaluated by bioluminescent signal generated by luciferase reporter which did not alter HER2 expression or proliferation ability of Malme-3M cells. Contrast with unarmed ATC, increased cytotoxic activity of HER2Bi-armed ATC against Malme-3M-luc cells was observed at effector/target (E/T) ratios of 1:1, 5:1, and 20:1. Moreover, HER2Bi-armed ATC expressed higher level of activation marker CD69 and secreted significantly higher level of IFN-γ than unarmed ATC counterpart at the E/T ratio of 20:1. In addition, compared with anti-HER2 mAb (Herceptin®) or unarmed ATC, HER2Bi-armed ATC showed remarkable suppression effect on Malme-3M-luc tumor cells. Furthermore, in melanoma tumor cell xenograft mice, infusion of HER2Bi-armed ATC successfully inhibited the growth of melanoma tumors. The anti-tumor effect of HER2Bi-armed ATC may provide a promising immunotherapy for melanoma in the future.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Expression of HER2 in different…
Figure 1. Expression of HER2 in different human melanoma cells.
(A) Surface expression of HER2 was evaluated by flow cytometry on seven melanoma cell lines (OCM-1, OMM-1, 92-1, Malme-3M, Mel888, SK Mel28 cell, and Mel624) and a primary melanoma (PM) cell. Shaded histograms represent cells stained with Herceptin® and open histograms represent cells stained with a non-related human IgG as control. (B) Western blot analysis showed HER2 protein band (185 kD) in cell lysates of human melanoma cells. β-actin was used as an internal control. A HER2 non-expression cell line K562 was used as a negative control.
Figure 2. Generation of Malme-3M-luc cells.
Figure 2. Generation of Malme-3M-luc cells.
(A) Schematic representation of the lentivirus vectors 3.7EF-1α-ER-△LNGFR-CMV-luciferase and 3.7CMV-ER-BirA. (B) Malme-3M cells were transduced with both 3.7lnluc and 3.7BirA lentiviral vectors and luc-positive cells were isolated by streptavidin beads. (C) FACS analysis of LNGFR expression in isolated Malme-3M-luc cells. Cells were labeled with anti-LNGFR monoclonal antibody CD271 followed by goat-anti-mouse-PE-CY5. Histogram indicates LNGFR expression, in which the shaded histogram represents purified Malme-3M-luc cells, and the black line represents un-transduced Malme-3M cells. (D) The bioluminescence image signal was measured with the Xenogen IVIS system, and serial dilution of Malme-3M-luc cells were plated in black 96-well plate in PBS containing D-luciferin substrate at final concentration of 0.15 mg/ml. (E) Pearson’s correlation coefficient analysis. The correlation coefficient (R) between the luciferase quantity and living cell number is 0.999.
Figure 3. No effect of the luciferase…
Figure 3. No effect of the luciferase gene on HER2 expression and the proliferation of Malme-3M cells and the binding of anti-CD3×anti-Her2 Bispecific antibody (HER2Bi-Ab) to both HER2 positive and CD3 positive cells.
(A) Proliferation of Malme-3M and Malme-3M-luc cells was assessed by CCK8 assay. The experiments were triplicated and the data are shown as the mean absorbance with SD (at OD 450 nm). Statistical analysis was conducted using Student’s t-test. (B) Surface expression of HER2 on Malme-3M-luc cells was analyzed by flow cytometry. Shaded histogram represents cells stained with Herceptin®, and open histogram represents cells stained with a control human IgG. (C) Malme-3M-luc cells were incubated with HER2Bi-Ab (1μg/ml, shaded histogram) or a combination of OKT3 and Herceptin® (1 μg/ml, open histogram) for 30 minutes at room temperature. HER2Bi-Ab binding was analyzed by flow cytometry using FITC- goat-anti-mouse IgG to detect the anti-CD3 moiety of the HER2Bi-Ab. (D) Activated T cells (ATC) were incubated with HER2Bi-Ab (1μg/ml, shaded histogram) or a combination of OKT3 and Herceptin® (1 μg/ml, open histogram) for 30 minutes at room temperature. HER2Bi-Ab binding was analyzed by flow cytometry using FITC- goat-anti-human IgG to detect the anti-HER2 moiety of the HER2Bi-Ab. (E) Malme-3M-luc cells were incubated with HER2Bi-Ab (1μg/ml, shaded histogram, OKT3 (1μg/ml, open histogram), or Herceptin® (1μg/ml, dot histogram) for 30 minutes at room temperature. HER2Bi-Ab binding was analyzed by flow cytometry using FITC- goat-anti-human IgG to detect the anti-HER2 moiety of the HER2Bi-Ab. (F) ATC were incubated with HER2Bi-Ab (1μg/ml, shaded histogram), Herceptin® (1μg/ml, open histogram), or OKT3 (1μg/ml, dot histogram) for 30 minutes at room temperature. HER2Bi-Ab binding was analyzed by flow cytometry using FITC- goat-anti-mouse IgG to detect the anti-CD3 moiety of the HER2Bi-Ab.
Figure 4. Growth inhibition of melanoma cells…
Figure 4. Growth inhibition of melanoma cells by HER2Bi-armed ATC.
Malme-3M-luc cells were seeded (2 x104/well) into 96-well round bottom microplate in triplicates overnight. On the following day, the medium was removed, and fresh medium alone or containing the unconjugated mAbs OKT3 or Herceptin (1μg/ml), ATC (2 x105/well), HER2Bi-armed ATC (2 x105/well, armed with 50 ng/HER2Bi/106 ATC) or a combination of OKT3 and Herceptin® with ATC (unarmed ATC) for 18 hours. (A) Real-time photographs were taken at 200x magnification. a. Malme-3M-luc targets alone; b. Malme-3M-luc with OKT3; c. Malme-3M-luc with Herceptin; d. Malme-3M-luc with ATC; e. Malme-3M-luc with unarmed-ATC; f. Malme-3M-luc with HER2Bi-armed ATC. HER2Bi-armed ATC, but not ATC or unarmed ATC aggregated with Malme-3M-luc tumor cells in culture after 18 hours incubation. (B) The proliferation of Malme-3M-luc was assessed by CCK8 assay with the OD value measured at 450 nm. The data are mean ± SD of triplicate experiments, and a representative experiment of three was shown. Statistical analysis was conducted using Student’s t-test. *: P<0.01, HER2Bi-armed ATC compared with combination of the OKT3 and Herceptin with ATC under similar conditions.
Figure 5. The lytic activity of HER2Bi-armed…
Figure 5. The lytic activity of HER2Bi-armed ATC against Malme-3M-luc cells
(A) Bioluminescence images of Malme-3M-luc cells after incubation with HER2Bi-armed ATC or unarmed ATC at different E/T ratio (1:1, 5:1, and 20:1) for 18 hours. (B) Bioluminescence image signal in photons per second was converted into living cell number and the cytotoxicity assay was measured at the indicated E/T. (C) The lytic activity of HER2Bi-armed ATC against B16-luc cells. (D) IFN-γ secretion by HER2Bi-armed ATC against Malme-3M–luc cells. Supernatants of co-cultures at indicated E/T were harvested at 18 hours and analyzed for IFN-γ level using IFN-γ ELISA Kit. (E) Expression of CD69 on HER2Bi-armed ATC or unarmed ATC was detected by flow cytometry after 18 hours co-culture with Malme-3M-luc cell at E/T ratio of 5:1. For B and C, the data are mean ± SD of triplicate experiments. Statistical analysis was conducted using Student’s t-test. A representative experiment of at least two was shown. *: P<0.01, HER2Bi-armed ATC compared with unarmed ATC under similar conditions.
Figure 6. In vivo cytotoxicity of HER2Bi-armed…
Figure 6. In vivo cytotoxicity of HER2Bi-armed ATC against Malme-3M-luc cells.
SCID-Beige mice were inoculated with Malme-3M-luc cells (1 x106/mouse) alone, or a mixture of Malme-3M-luc cells with un-armed ATC (250 ng Herceptin® and OKT3, combined with 1 x107ATC/mouse), or with HER2Bi-ATC (1 x107 /mouse, armed with 50 ng/HER2Bi/106 ATC) on day 0. Each group contained 5 mice. (A) Bioluminescence images of a representative mouse from each group on day 2, 7 and 14 were shown. (B) Images were analyzed using Living Image software and tumor values were represented as total flux measurements in photons/second. Mean values of tumor growth curves were shown. The significance between different groups was compared by one-way analysis of variance (ANOVA) followed by LSD for multiple comparison. **P<0.01, *P<0.05.

References

    1. Dillman RO, Cornforth AN, Depriest C, McClay EF, Amatruda TT et al. (2012) Tumor Stem Cell Antigens as Consolidative Active Specific Immunotherapy: A Randomized Phase II Trial of Dendritic Cells Versus Tumor Cells in Patients With Metastatic Melanoma. J Immunother 35: 641-649. doi:. PubMed: .
    1. Ullenhag GJ, Sadeghi AM, Carlsson B, Ahlström H, Mosavi F et al. (2012) Adoptive T-cell therapy for malignant melanoma patients with TILs obtained by ultrasound-guided needle biopsy. Cancer Immunol Immunother 61: 725-732. doi:. PubMed: .
    1. Murer K, Urosevic M, Willers J, Selvam P, Laine E et al. (2004) Expression of Melan- A/MART-1 in primary melanoma cell cultures has prognostic implication in metastatic melanoma patients. Melanoma Res 14: 257-262. doi:. PubMed: .
    1. Patel D, Bassi R, Hooper AT, Sun H, Huber J et al. (2008) Enhanced suppression of melanoma tumor growth and metastasis by combined therapy with anti-VEGF receptor and anti-TYRP-1/gp75 monoclonal antibodies. Anticancer Res 28: 2679-2686. PubMed: .
    1. Yang B, O’Herrin SM, Wu J, Reagan-Shaw S, Ma Y et al. (2007) MAGE-A, mMage-b, and MAGE-C proteins form complexes with KAP1 and suppress p53-dependent apoptosis in MAGE-positive cell lines. Cancer Res 67: 9954-9962. doi:. PubMed: .
    1. Talebian F, Liu JQ, Liu Z, Khattabi M, He Y et al. (2012) Melanoma cell expression of CD200 inhibits tumor formation and lung metastasis via inhibition of myeloid cell functions. PLOS ONE 7: e31442. doi:. PubMed: .
    1. Weber J (2011) Immunotherapy for melanoma. Curr Opin Oncol 23: 163-169. doi:. PubMed: .
    1. Ribas A (2012) Tumor immunotherapy directed at PD-1. N Engl J Med 366: 2517-2519. doi:. PubMed: .
    1. Hansel DE, Ashfaq R, Rahman A, Wanzer D, Yeo CJ et al. (2005) A subset of pancreatic adenocarcinomas demonstrates coamplification of topoisomerase IIalpha and HER2/neu: use of immunolabeling and multicolor FISH for potential patient screening andtreatment. Am J Clin Pathol 123: 28-35. doi:. PubMed: .
    1. Marín AP, Arranz EE, Sánchez AR, Auñón PZ, Barón MG (2010) Role of anti-Her-2 therapy in bladder carcinoma. J Cancer Res Clin Oncol 136: 1915-1920. doi:. PubMed: .
    1. Glisson B, Colevas AD, Haddad R, Krane J, El-Naggar A et al. (2004) HER2 expression in salivary gland carcinomas: dependence on histological subtype. Clin Cancer Res 10: 944-946. doi:. PubMed: .
    1. Pagni F, Zannella S, Ronchi S, Garanzini C, Leone BE (2013) HER2 Status of Gastric Carcinoma and Corresponding Lymph Node Metastasis. Pathol Oncol Res 19: 103-109. doi:. PubMed: .
    1. Jørgensen JT, Hersom M (2012) HER2 as a Prognostic Marker in Gastric Cancer - A Systematic Analysis of Data from the Literature. J Cancer 3: 137-144. doi:. PubMed: .
    1. Gilcrease MZ, Woodward WA, Nicolas MM, Corley LJ, Fuller GN et al. (2009) Even low-level HER2 expression may be associated with worse outcome in node-positive breast cancer. Am J Surg Pathol 33: 759-767. doi:. PubMed: .
    1. Arnould L, Gelly M, Penault-Llorca F, Benoit L, Bonnetain F et al. (2006) Trastuzumab-based treatment of HER2-positive breast cancer: an antibody-dependent cellular cytotoxicity mechanism? Br J Cancer 94: 259-267. doi:. PubMed: .
    1. Hudis CA (2007) Trastuzumab - Mechanism of action and use - Reply. N Engl J Med 357: 1665-1666.
    1. Potti A, Hille RC, Koch M (2003) Immunohistochemical determination of HER-2/neu overexpression in malignant melanoma reveals no prognostic value, while c-Kit (CD117) overexpression exhibits potential therapeutic implications. J Carcinog 2: 8. doi:. PubMed: .
    1. Helmke BM, Zahel T, Breinig M, Schirmacher P, Aulmann S et al. (2010) Her2 overexpression is a rare event in anorectal melanoma. Melanoma Res 20: 431-434. PubMed: .
    1. Kluger HM, DiVito K, Berger AJ, Halaban R, Ariyan S et al. (2004) Her2/neu is not a commonly, expressed therapeutic target in melanoma - a large cohort tissue microarray study. Melanoma Res 14: 207-210. doi:. PubMed: .
    1. Inman JL, Kute T, White W, Pettenati M, Levine EA (2003) Absence of HER2 overexpression in metastatic malignant melanoma. J Surg Oncol 84: 82-88. doi:. PubMed: .
    1. Persons DL, Arber DA, Sosman JA, Borelli KA, Slovak ML (2000) Amplification and overexpression of HER-2/neu are uncommon in advanced stage melanoma. Anticancer Res 20: 1965-1968. PubMed: .
    1. Easty DJ, Herlyn M, Bennett DC (1995) Abnormal Protein-Tyrosine Kinase Gene-Expression during Melanoma Progression and Metastasis. Int J Cancer 60: 129-136. doi:. PubMed: .
    1. Chenevix-Trench G, Martin NG, Ellem KAO (1990) Gene-Expression in Melanoma Cell-Lines and Cultured Melanocytes - Correlation between Levels of C-Src-1, C-Myc and P53. Oncogene 5: 1187-1193. PubMed: .
    1. Bodey B, Bodey B, Gröger AM, Luck JV, Siegel SE et al. (1997) Clinical and prognostic significance of the expression of the c-erbB-2 and c-erbB-3 oncoproteins in primary and metastatic malignant melanomas and breast carcinomas. Anticancer Res 17: 1319-1330. PubMed: .
    1. Ma J, Usui Y, Kezuka T, Okunuki Y, Zhang L et al. (2011) Costimulatory molecule expression on human uveal melanoma cells: functional analysis of CD40 and B7-H1. Exp Eye Res 96: 98-106. PubMed: .
    1. Han H, Liu Q, He W, Ong K, Liu X et al. (2011) An efficient vector system to modify cells genetically. PLOS ONE 6: e26380. doi:. PubMed: .
    1. Clay TM, Custer MC, Sachs J, Hwu P, Rosenberg SA et al. (1999) Efficient transfer of a tumor antigen-reactive TCR to human peripheral blood lymphocytes confers anti-tumor reactivity. J Immunol 163: 507-513. PubMed: .
    1. Fu X, Tao L, Rivera A, Williamson S, Song XT et al. (2010) A simple and sensitive method for measuring tumor-specific T cell cytotoxicity. PLOS ONE 5: e11867. doi:. PubMed: .
    1. Brown CE, Wright CL, Naranjo A, Vishwanath RP, Chang WC et al. (2005) Biophotonic cytotoxicity assay for high-throughput screening of cytolytic killing. J Immunol Methods 297: 39-52. doi:. PubMed: .
    1. Tiffen JC, Bailey CG, Ng C, Rasko JE, Holst J (2010) Luciferase expression and bioluminescence does not affect tumor cell growth in vitro or in vivo. Mol Cancer 9: 299. doi:. PubMed: .
    1. Molina MA, Codony-Servat J, Albanell J, Rojo F, Arribas J et al. (2001) Trastuzumab (herceptin), a humanized anti-Her2 receptor monoclonal antibody, inhibits basal and activated Her2 ectodomain cleavage in breast cancer cells. Cancer Res 61: 4744-4749. PubMed: .
    1. Seimetz D, Lindhofer H, Bokemeyer C (2010) Development and approval of the trifunctional antibody catumaxomab (anti-EpCAM x anti-CD3) as a targeted cancer immunotherapy. Cancer Treat Rev 36: 458-467. doi:. PubMed: .
    1. Park S, Jiang Z, Mortenson ED, Deng L, Radkevich-Brown O et al. (2010) The therapeutic effect of anti-HER2/neu antibody depends on both innate and adaptive immunity. Cancer Cell 18: 160-170. doi:. PubMed: .
    1. Clynes RA, Towers TL, Presta LG, Ravetch JV (2000) Inhibitory Fc receptors modulate in vivo cytotoxicity against tumor targets. Nat Med 6: 443-446. doi:. PubMed: .
    1. Grabert RC, Cousens LP, Smith JA, Olson S, Gall J et al. (2006) Human T cells armed with Her2/neu bispecific antibodies divide, are cytotoxic, and secrete cytokines with repeated stimulation. Clin Cancer Res 12: 569-576. doi:. PubMed: .

Source: PubMed

3
Iratkozz fel