Combined blockade of HER2 and VEGF exerts greater growth inhibition of HER2-overexpressing gastric cancer xenografts than individual blockade

Rohit Singh, Woo Jin Kim, Pyeung-Hyeun Kim, Hyo Jeong Hong, Rohit Singh, Woo Jin Kim, Pyeung-Hyeun Kim, Hyo Jeong Hong

Abstract

Gastric cancer overexpressing the human epidermal growth factor 2 (HER2) protein has a poor outcome, although a combination of chemotherapy and the anti-HER2 antibody trastuzumab has been approved for the treatment of advanced gastric cancer. Vascular endothelial growth factor (VEGF) expression in gastric cancer is correlated with recurrence and poor prognosis; however, the anti-VEGF antibody bevacizumab has shown limited efficacy against gastric cancer in clinical trials. In this study, we evaluated the antitumor effects of trastuzumab; VEGF-Trap binding to VEGF-A, VEGF-B and placental growth factor (PlGF); and a combination of trastuzumab and VEGF-Trap in a gastric cancer xenograft model. Although trastuzumab and VEGF-Trap each moderately inhibited tumor growth, the combination of these agents exerted greater inhibition compared with either agent alone. Immunohistochemical analyses indicated that the reduction in tumor growth was associated with decreased proliferation and increased apoptosis of tumor cells and decreased tumor vascular density. The combined treatment resulted in fewer proliferating tumor cells, more apoptotic cells and reduced tumor vascular density compared with treatment with trastuzumab or VEGF-Trap alone, indicating that trastuzumab and VEGF-Trap had additive inhibitory effects on the tumor growth and angiogenesis of the gastric cancer xenografts. These data suggest that trastuzumab in combination with VEGF-Trap may represent an effective approach to treating HER2-overexpressing gastric cancer.

Figures

Figure 1
Figure 1
Human epidermal growth factor 2 (HER2) and vascular endothelial growth factor (VEGF) expression levels in gastric cancer cells. NCI-N87Luc+ cells express HER2 (a) and VEGF (b), as detected by flow cytometry. (c) Detection of the expression levels of VEGF and VEGF-related receptors (VEGFR) by reverse transcriptase-polymerase chain reaction (RT-PCR). GAPDH, glyceraldehyde 3-phosphate dehydrogenase; HUVEC, human umbilical vein endothelial cell; NRP-1, neuropilin-1.
Figure 2
Figure 2
Analysis of bromodeoxyuridine (BrdU) incorporation and total DNA content. (a and b) Epidermal growth factor (EGF)-induced cell cycle progression in NCI-N87Luc+ cells. Cells were serum-starved overnight and treated with vascular endothelial growth factor (VEGF) or EGF for 24 h. The cells were then pulsed with BrdU for 6 h, fixed and stained with a allophycocyanin (APC)-coupled anti-BrdU antibody and 7-aminoactinomycin D (7-AAD). (c and d) Cells (70–80% confluence) were treated with immunoglobulin G (IgG), trastuzumab or VEGF-Trap for 48 h and pulsed with BrdU for 6 h. The cells were then fixed and stained with an APC-coupled anti-BrdU antibody and 7-AAD. The stained cells were analyzed by a FACSAria flow cytometer (n=3; *P<0.05 versus control).
Figure 3
Figure 3
Antitumor and antiangiogenic activity of trastuzumab (Tr) in vitro. (a) In vitro proliferation (WST-1 assay) of NCI-N87Luc+ cells. (b) Inhibition of Akt and Erk 1/2 phosphorylation by trastuzumab. (c) Measurement of vascular endothelial growth factor (VEGF) mRNA in response to trastuzumab treatment in vitro by reverse transcriptase-polymerase chain reaction (RT-PCR). (d) In vitro trastuzumab-mediated antibody-dependent cell-mediated cytotoxicity (ADCC) against human epidermal growth factor 2 (HER2)-positive gastric cancer cells. GAPDH, glyceraldehyde 3-phosphate dehydrogenase.
Figure 4
Figure 4
The combined inhibition of human epidermal growth factor 2 (HER2) and vascular endothelial growth factor (VEGF) reduces tumor growth more efficiently than single-agent inhibition. (a) Mice bearing NCI-N87Luc+ tumors were divided into four groups and treated with an isotype-control antibody, VEGF-Trap (VT), trastuzumab (Tr) or a combination of VEGF-Trap and trastuzumab (VT+Tr) for 28 days. (b) Changes in body weight. No significant change in body weight was observed among the treatment groups. (c) Dissected tumor weight after 28 days (n=7; *P<0.05 versus isotype control; #P<0.05 versus VEGF-Trap; $P<0.05 versus trastuzumab). IgG, immunoglobulin G.
Figure 5
Figure 5
Viable and necrotic areas of tumors at the end of treatment. (a) Viable (green) and necrotic (black) areas, as differentiated by YO-PRO-1 staining and quantified by the ImageJ software program. The scale bar represents 1 mm. Viable (b) and necrotic tumor areas (c) after 28 days of treatment. (d) Viable and necrotic tumor areas are presented as a ratio (n=7; *P<0.05 versus isotype control; #P<0.05 versus vascular endothelial growth factor (VEGF)-Trap (VT); $P<0.05 versus trastuzumab (Tr)). IgG, immunoglobulin G.
Figure 6
Figure 6
The effect of treatment on tumor cell proliferation and apoptosis. Immunohistochemical staining of phospho (p)-histone-H3 (a and c) and activated caspase-3 (b and d) after 28 days of treatment. The scale bar represents 100 μm. The inset high-magnification ( × 60 objective lens) image shows a YO-PRO-1 (green)-stained nucleus and phospho-histone-H3 or activated caspase-3 (red) (n=7; *P<0.05 versus isotype control; #P<0.05 versus vascular endothelial growth factor (VEGF)-Trap (VT); $P<0.05 versus trastuzumab (Tr)).
Figure 7
Figure 7
The effect of treatment on vascular density. Two-dimensional images constructed from Z-stack images showing CD31-positive blood vessels in the peritumoural (a and c) and intratumoral (a and d) regions. The black dotted line indicates the boundary of the tumors. The scale bar represents 100 μm (n=7; *P<0.05 versus isotype control; #P<0.05 versus vascular endothelial growth factor (VEGF)-Trap (VT); $P<0.05 versus trastuzumab (Tr)). IgG, immunoglobulin G.

References

    1. Holbro T, Civenni G, Hynes NE. The ErbB receptors and their role in cancer progression. Exp Cell Res. 2003;284:99–110.
    1. Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol. 2001;2:127–137.
    1. Laughner E, Taghavi P, Chiles K, Mahon PC, Semenza GL. HER2 (neu) signaling increases the rate of hypoxia-inducible factor 1alpha (HIF-1alpha) synthesis: novel mechanism for HIF-1-mediated vascular endothelial growth factor expression. Mol Cell Biol. 2001;21:3995–4004.
    1. Wen XF, Yang G, Mao W, Thornton A, Liu J, Bast RC, Jr, et al. HER2 signaling modulates the equilibrium between pro- and antiangiogenic factors via distinct pathways: implications for HER2-targeted antibody therapy. Oncogene. 2006;25:6986–6996.
    1. Burstein HJ. The distinctive nature of HER2-positive breast cancers. N Engl J Med. 2005;353:1652–1654.
    1. Gravalos C, Jimeno A. HER2 in gastric cancer: a new prognostic factor and a novel therapeutic target. Ann Oncol. 2008;19:1523–1529.
    1. Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61:69–90.
    1. Cobleigh MA, Vogel CL, Tripathy D, Robert NJ, Scholl S, Fehrenbacher L, et al. Multinational study of the efficacy and safety of humanized anti-HER2 monoclonal antibody in women who have HER2-overexpressing metastatic breast cancer that has progressed after chemotherapy for metastatic disease. J Clin Oncol. 1999;17:2639–2639.
    1. Vogel CL, Cobleigh MA, Tripathy D, Gutheil JC, Harris LN, Fehrenbacher L, et al. Efficacy and safety of trastuzumab as a single agent in first-line treatment of HER2-overexpressing metastatic breast cancer. J Clin Oncol. 2002;20:719–726.
    1. Nahta R, Esteva FJ. Herceptin: mechanisms of action and resistance. Cancer Lett. 2006;232:123–138.
    1. Fukumura D, Xavier R, Sugiura T, Chen Y, Park E, Lu N, et al. Tumor induction of VEGF promoter activity in stromal cells. Cell. 1998;94:715–725.
    1. Harmey J, Dimitriadis E, Kay E, Redmond HP, Bouchier-Hayes D. Regulation of macrophage production of vascular endothelial growth factor (VEGF) by hypoxia and transforming growth factor beta-1. Ann Surg Oncol. 1998;5:271–278.
    1. Izumi Y, Xu L, di Tomaso E, Fukumura D, Jain RK. Tumour biology: herceptin acts as an anti-angiogenic cocktail. Nature. 2002;416:279–280.
    1. Jain RK. Molecular regulation of vessel maturation. Nat Med. 2003;9:685–693.
    1. Figg WD, Folkman J. Angiogenesis: An Integrative Approach from Science to Medicine. Springer: New York, NY, USA; 2008.
    1. Le X, Mao W, Lu C, Thornton A, Heymach JV, Sood AK, et al. Specific blockade of VEGF and HER2 pathways results in greater growth inhibition of breast cancer xenografts that overexpress HER2. Cell Cycle. 2008;7:3747–3758.
    1. Alami N, Sun Y, De P, Benmassaoud AM, Wang Y, Leyland-Jones B. Combination effects of herceptin, pertuzumab and bevacizumab in a HER2-overexpressing breast cancer xenograft model. Cancer Res. 2009;69:4058(abstract).
    1. Pegram M, Yeon C, Ku N, Gaudreault J, Slamon D. Phase I combined biological therapy of breast cancer using two humanized monoclonal antibodies directed against HER2 proto-oncogene and vascular endothelial growth factor (VEGF) Breast Cancer Res Treat. 2004;88:S124–S125. (abstract 3039).
    1. Pegram M, Chan D, Dichmann R, Tan-Chiu E, Yeon C, Durna L, et al. Phase II combined biological therapy targeting the HER2 proto-oncogene and the vascular endothelial growth factor using trastuzumab (T) and bevacizumab (B) as first line treatment of HER2-amplified breast cancer. Breast Cancer Res Treat. 2006;100:S28–S29. (abstract 301).
    1. Pierga J, Petit T, Delozier T, Ferrero J, Campone M, Gligorov J, et al. Neoadjuvant bevacizumab, trastuzumab, and chemotherapy for primary inflammatory HER2-positive breast cancer (BEVERLY-2): an open-label, single-arm phase 2 study. Lancet Oncol. 2012;13:375–384.
    1. Dvorak HF. Vascular permeability factor/vascular endothelial growth factor: a critical cytokine in tumor angiogenesis and a potential target for diagnosis and therapy. J Clin Oncol. 2002;20:4368–4380.
    1. Yancopoulos GD, Davis S, Gale NW, Rudge JS, Wiegand SJ, Holash J. Vascular-specific growth factors and blood vessel formation. Nature. 2000;407:242–248.
    1. Ferrara N, Gerber H, LeCouter J. The biology of VEGF and its receptors. Nat Med. 2003;9:669–676.
    1. Ellis LM, Hicklin DJ. VEGF-targeted therapy: mechanisms of anti-tumour activity. Nat Rev Cancer. 2008;8:579–591.
    1. Miao H, Lee P, Lin H, Soker S, Klagsbrun M. Neuropilin-1 expression by tumor cells promotes tumor angiogenesis and progression. FASEB J. 2000;14:2532–2539.
    1. Lohela M, Bry M, Tammela T, Alitalo K. VEGFs and receptors involved in angiogenesis versus lymphangiogenesis. Curr Opin Cell Biol. 2009;21:154–165.
    1. Donnini S, Machein MR, Plate KH, Weich HA. Expression and localization of placenta growth factor and P1GF receptors in human meningiomas. J Pathol. 1999;189:66–71.
    1. André T, Kotelevets L, Vaillant JC, Coudray AM, Weber L, Prévot S, et al. VEGF, VEGF-B, VEGF-C, and their receptors KDR, FLT-1 and FLT-4 during the neoplastic progression of human colonic mucosa. Int J Cancer. 2000;86:174–181.
    1. Fischer C, Mazzone M, Jonckx B, Carmeliet P. FLT1 and its ligands VEGFB and PlGF: drug targets for anti-angiogenic therapy. Nat Rev Cancer. 2008;8:942–956.
    1. Rini BI, Michaelson MD, Rosenberg JE, Bukowski RM, Sosman JA, Stadler WM, et al. Antitumor activity and biomarker analysis of sunitinib in patients with bevacizumab-refractory metastatic renal cell carcinoma. J Clin Oncol. 2008;26:3743–3748.
    1. Willett CG, Duda DG, Di Tomaso E, Boucher Y, Ancukiewicz M, Sahani DV, et al. Efficacy, safety, and biomarkers of neoadjuvant bevacizumab, radiation therapy, and fluorouracil in rectal cancer: a multidisciplinary phase II study. J Clin Oncol. 2009;27:3020–3026.
    1. Zhang F, Tang Z, Hou X, Lennartsson J, Li Y, Koch AW, et al. VEGF-B is dispensable for blood vessel growth but critical for their survival, and VEGF-B targeting inhibits pathological angiogenesis. Proc Natl Acad Sci USA. 2009;106:6152–6157.
    1. Jussila L, Alitalo K. Vascular growth factors and lymphangiogenesis. Physiol Rev. 2002;82:673–700.
    1. Lieu C, Tran H, Jiang Z, Mao M, Overman M, Eng C, et al. The association of alternate VEGF ligands with resistance to anti-VEGF therapy in metastatic colorectal cancer. J Clin Oncol. 2011;29:3533.
    1. Ebos JM, Kerbel RS. Antiangiogenic therapy: impact on invasion, disease progression, and metastasis. Nat Rev Clin Oncol. 2011;8:210–221.
    1. Kumar R, Crouthamel M, Rominger D, Gontarek R, Tummino P, Levin R, et al. Myelosuppression and kinase selectivity of multikinase angiogenesis inhibitors. Br J Cancer. 2009;101:1717–1723.
    1. Bergers G, Hanahan D. Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer. 2008;8:592–603.
    1. Gianni L, Romieu GH, Lichinitser M, Serrano SV, Mansutti M, Pivot X, et al. AVEREL: A Randomized Phase III Trial evaluating bevacizumab in combination with docetaxel and trastuzumab as first-line therapy for HER2-positive locally recurrent/metastatic breast cancer. J Clin Oncol. 2013;31:1719–1725.
    1. Lin N, Seah D, Gelman R, Desantis S, Mayer E, Isakoff S, et al. A phase II study of bevacizumab in combination with vinorelbine and trastuzumab in HER2-positive metastatic breast cancer. Breast Cancer Res Treat. 2013;139:403–410.
    1. Holash J, Davis S, Papadopoulos N, Croll SD, Ho L, Russell M, et al. VEGF-Trap: a VEGF blocker with potent antitumor effects. Proc Natl Acad Sci USA. 2002;99:11393–11398.
    1. Papadopoulos N, Martin J, Ruan Q, Rafique A, Rosconi MP, Shi E, et al. Binding and neutralization of vascular endothelial growth factor (VEGF) and related ligands by VEGF Trap, ranibizumab and bevacizumab. Angiogenesis. 2012;15:171–185.
    1. Gaya A, Tse V. A preclinical and clinical review of aflibercept for the management of cancer. Cancer Treat Rev. 2012;38:484–493.
    1. Okines AF, Cunningham D. Trastuzumab in gastric cancer. Eur J Cancer. 2010;46:1949–1959.
    1. Bang Y, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet. 2010;376:687–697.
    1. Kanai T, Konno H, Tanaka T, Baba M, Matsumoto K, Nakamura S, et al. Anti-tumor and anti-metastatic effects of human-vascular-endothelial-growth-factor-neutralizing antibody on human colon and gastric carcinoma xenotransplanted orthotopically into nude mice. Int J Cancer. 1998;77:933–936.
    1. Ohtsu A, Shah MA, Van Cutsem E, Rha SY, Sawaki A, Park SR, et al. Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a Randomized, Double-Blind, Placebo-Controlled Phase III Study. J Clin Oncol. 2011;29:3968–3976.
    1. Kim SY, Kim HP, Kim YJ, Oh do Y, Im SA, Lee D, et al. Trastuzumab inhibits the growth of human gastric cancer cell lines with HER2 amplification synergistically with cisplatin. Int J Oncol. 2008;32:89–95.
    1. Hashizume H, Falcón BL, Kuroda T, Baluk P, Coxon A, Yu D, et al. Complementary actions of inhibitors of angiopoietin-2 and VEGF on tumor angiogenesis and growth. Cancer Res. 2010;70:2213–2223.
    1. Klos KS, Zhou X, Lee S, Zhang L, Yang W, Nagata Y, et al. Combined trastuzumab and paclitaxel treatment better inhibits ErbB-2-mediated angiogenesis in breast carcinoma through a more effective inhibition of Akt than either treatment alone. Cancer. 2003;98:1377–1385.
    1. Hynes NE, MacDonald G. ErbB receptors and signaling pathways in cancer. Curr Opin Cell Biol. 2009;21:177–184.
    1. du Manoir JM, Francia G, Man S, Mossoba M, Medin JA, Viloria-Petit A, et al. Strategies for delaying or treating in vivo acquired resistance to trastuzumab in human breast cancer xenografts. Clin Cancer Res. 2006;12:904–916.
    1. Van Cutsem E, de Haas S, Kang YK, Ohtsu A, Tebbutt NC, Ming Xu J, et al. Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a biomarker evaluation from the AVAGAST randomized phase III trial. J Clin Oncol. 2012;30:2119–2127.
    1. Van Cutsem E, Jayson G, Dive C, Dilba P, de Haas S, Wild N, et al. Analysis of blood plasma factors in the AVITA Phase III Randomized Study of bevacizumab (bev) with gemcitabine-erlotinib (ge) in patients (pts) with metastatic pancreatic cancer (mPC) Eur J Cancer. 2011;47:(abstract 803).

Source: PubMed

3
Předplatit