Identification of novel therapeutic targets in microdissected clear cell ovarian cancers

Michael P Stany, Vinod Vathipadiekal, Laurent Ozbun, Rebecca L Stone, Samuel C Mok, Hui Xue, Takashi Kagami, Yuwei Wang, Jessica N McAlpine, David Bowtell, Peter W Gout, Dianne M Miller, C Blake Gilks, David G Huntsman, Susan L Ellard, Yu-Zhuo Wang, Pablo Vivas-Mejia, Gabriel Lopez-Berestein, Anil K Sood, Michael J Birrer, Michael P Stany, Vinod Vathipadiekal, Laurent Ozbun, Rebecca L Stone, Samuel C Mok, Hui Xue, Takashi Kagami, Yuwei Wang, Jessica N McAlpine, David Bowtell, Peter W Gout, Dianne M Miller, C Blake Gilks, David G Huntsman, Susan L Ellard, Yu-Zhuo Wang, Pablo Vivas-Mejia, Gabriel Lopez-Berestein, Anil K Sood, Michael J Birrer

Abstract

Clear cell ovarian cancer is an epithelial ovarian cancer histotype that is less responsive to chemotherapy and carries poorer prognosis than serous and endometrioid histotypes. Despite this, patients with these tumors are treated in a similar fashion as all other ovarian cancers. Previous genomic analysis has suggested that clear cell cancers represent a unique tumor subtype. Here we generated the first whole genomic expression profiling using epithelial component of clear cell ovarian cancers and normal ovarian surface specimens isolated by laser capture microdissection. All the arrays were analyzed using BRB ArrayTools and PathwayStudio software to identify the signaling pathways. Identified pathways validated using serous, clear cell cancer cell lines and RNAi technology. In vivo validations carried out using an orthotopic mouse model and liposomal encapsulated siRNA. Patient-derived clear cell and serous ovarian tumors were grafted under the renal capsule of NOD-SCID mice to evaluate the therapeutic potential of the identified pathway. We identified major activated pathways in clear cells involving in hypoxic cell growth, angiogenesis, and glucose metabolism not seen in other histotypes. Knockdown of key genes in these pathways sensitized clear cell ovarian cancer cell lines to hypoxia/glucose deprivation. In vivo experiments using patient derived tumors demonstrate that clear cell tumors are exquisitely sensitive to antiangiogenesis therapy (i.e. sunitinib) compared with serous tumors. We generated a histotype specific, gene signature associated with clear cell ovarian cancer which identifies important activated pathways critical for their clinicopathologic characteristics. These results provide a rational basis for a radically different treatment for ovarian clear cell patients.

Conflict of interest statement

Competing Interests: Pfizer Canada is a healthcare company. SLE and YZW received Investigator-initiated project funding from Pfizer Canada. This does not alter the authors' adherence to all the PLoS ONE policies on sharing data and materials.

Figures

Figure 1. Whole genome expression profiling of…
Figure 1. Whole genome expression profiling of clear cell ovarian tumors.
(a) Graphic representation of whole genome expression profiling of the Clear Cell Ovarian Cancer Specimen (CCOC) and Ovarian Surface Epithelium (OSE). (b) and (c) Comparison of qRT-PCR data and microarray data of the six overexpressed and four underexpressed genes used for validation (d) Pathway analysis of differentially regulated genes identified in the clear cell ovarian cancer microarray. Genes included in the analysis were required to have a fold change ≥1.5. Multiple probe sets were averaged for each gene. Red, gene is up-regulated. Blue, gene is down-regulated.
Figure 2. Clear cell ovarian cancer cell…
Figure 2. Clear cell ovarian cancer cell lines were more resistant to hypoxia/glucose deprivation than serous ovarian cancer cell lines.
(a) Cellular proliferation assays of ovarian cancer cell lines of clear cell and serous origin under different oxygen and glucose conditions. The doubling times of the cell lines were compared, and the fold change in doubling time between the conditions of normal oxygen/normal glucose (NN) and hypoxia/glucose deprivation (HG) were calculated. (b) The bar graph demonstrates a statistically significant difference when the fold change in doubling times is averaged by histotype, showing that the three clear cell ovarian cancer cell lines were less affected by hypoxia/glucose deprivation than the six serous cell lines (p = 0.0037). (c)Trypan blue exclusion assay of OVCA-420 and ES2 cells grown in normal oxygen/normal glucose (NN) and hypoxia/ glucose deprivation (HG) for 72 hours. (d) Proliferation assay of OVCA-420 cells incubated in NN, HG, and HG with Z-VAD-FAK. The addition of Z-VAD-FAK did not alter the growth inhibition of HG. (e) Necrosis assay of OVCA-420 cells demonstrated a statistically significant increase in necrosis when incubated in HG (p = 0.003).
Figure 3. Knockdown of HIF1α and ENO1…
Figure 3. Knockdown of HIF1α and ENO1 in three clear cell ovarian cancer cell lines.
(a) Knockdown efficiency of siRNA molecules targeting HIF1α and ENO1 in three clear cell ovarian cancer cell lines, assessed by quantitative real-time PCR. (b) The percent of growth inhibition after transfection of siRNA molecules targeting HIF1α and ENO1. Growth was assessed after 24 hours of hypoxia/glucose deprivation *p = 0.12, **p<0.05,***p = <0.0001. (c, d) Knockdown efficiency of siRNA molecules targeting HIF1α and ENO1 in OVCA429 and OVCA420 and the percent of growth inhibition after transfection of siRNA molecules targeting HIF1α and ENO1. (e, f) Effect of HIF1α and ENO1 knockdown on proliferation of ES-2 and TOV-21G clear cell cell lines (***p = <0.0001).
Figure 4. Effect of Enolase or HIF1…
Figure 4. Effect of Enolase or HIF1 α siRNA-DOPC ± sunitinib on ovarian clear cell tumor progression in the female athymic nude mouse model.
(a) Tumor weight: ES2 cells (1×106 cells/mouse) were injected intraperitoneal into mice. The mice were treated with control, Enolase or HIF1 α siRNA-DOPC ± sunitinib (n = 10/group). Immunohistochemical staining for CD31 antigen was performed on frozen slides of tumor to evaluate the number of tumor nodules (b) and tumor microvessel density (MVD) (c). (See Materials and Methods section for details).
Figure 5. Effect of sunitinib on the…
Figure 5. Effect of sunitinib on the growth of patient-derived CCOC tissue xenografts.
(a.) Effect of a two-week treatment with sunitinib on growth of subrenal capsule xenografts in NOD-SCID mice (6 mice/group; 2 grafts per kidney) of transplantable serous (LTL237, 247 and 259) and clear cell (LTL175) ovarian carcinoma tissue lines derived from patients' cancers. Growth of the xenografts is expressed as % tumor volume determined at necropsy by measurement with calipers. Data are presented as means ± S.E.M. (b, c) Effect of sunitinib on VEGFR2 and PDGFRβ tyrosine phosphorylation as shown by Western blot analysis. Subrenal capsule xenografts of ovarian LTL247 serous carcinoma tissue and LTL175 clear cell carcinoma tissue in mice, treated for 2 weeks with sunitinib or vehicle (control), were lysed and processed for Western blot analysis of VEGFR2 (b) and PDGFRβ (c) tyrosine residues. The results are representative of 3 experiments. (d) Representative H&E-stained tumor sections of control (d.a), and sunitinib-treated (d.b) LTL175 tissue. (e) Effect of sunitinib on microvessel density of subrenal capsule xenografts in NOD-SCID mice of serous (LTL237, 247, 259) and clear cell (LTL175) ovarian carcinoma tissue lines. Data are presented as the average number of blood vessels per ×400 microscopic field ± S.E.M. (f) Effect of sunitinib on apoptosis in LTL175 xenografts in NOD-SCID mice treated with sunitinib for 2 weeks. Representative tissue sections of control (f. a) and sunitinib-treated (f. c) TUNEL-stained tumor tissue and counterstained with DAPI (f. b, control; f. d, sunitinib-treated); (g), percentage of apoptotic cells determined via microscopic analysis using a 400× microscopic field; data presented as means ± S.E.M.

References

    1. Schiller W. Mesonephroma ovarii. Am J Cancer. 1939;35:1–21.
    1. Genton CY. Ultrastructure of clear cell carcinoma of the ovary. Case report and review of the literature. Virchows Arch A Pathol Anat Histol. 1979;385:77–91.
    1. Silverberg SG. Ultrastructure and histogenesis of clear cell carcinoma of the ovary. Am J Obstet Gynecol. 1973;115:394–400.
    1. Jenison EL, Montag AG, Griffiths CT, Welch WR, Lavin PT, et al. Clear cell adenocarcinoma of the ovary: a clinical analysis and comparison with serous carcinoma. Gynecol Oncol. 1989;32:65–71.
    1. Kennedy AW, Biscotti CV, Hart WR, Webster KD. Ovarian clear cell adenocarcinoma. Gynecol Oncol. 1989;32:342–349.
    1. Crozier MA, Copeland LJ, Silva EG, Gershenson DM, Stringer CA. Clear cell carcinoma of the ovary: a study of 59 cases. Gynecol Oncol. 1989;35:199–203.
    1. Kobel M, Kalloger SE, Huntsman DG, Santos JL, Swenerton KD, et al. Differences in tumor type in low-stage versus high-stage ovarian carcinomas. Int J Gynecol Pathol. 2010;29:203–211.
    1. Sugiyama T, Kamura T, Kigawa J, Terakawa N, Kikuchi Y, et al. Clinical characteristics of clear cell carcinoma of the ovary: a distinct histologic type with poor prognosis and resistance to platinum-based chemotherapy. Cancer. 2000;88:2584–2589.
    1. Pectasides D, Pectasides E, Psyrri A, Economopoulos T. Treatment issues in clear cell carcinoma of the ovary: a different entity? Oncologist. 2006;11:1089–1094.
    1. Matsuura Y, Robertson G, Marsden DE, Kim SN, Gebski V, et al. Thromboembolic complications in patients with clear cell carcinoma of the ovary. Gynecol Oncol. 2007;104:406–410.
    1. Nordback I, Lauslahti K. Clinicopathologic and histochemical study of ovarian clear cell carcinoma. Int J Gynaecol Obstet. 1980;18:85–89.
    1. Dickersin GR, Welch WR, Erlandson R, Robboy SJ. Ultrastructure of 16 cases of clear cell adenocarcinoma of the vagina and cervix in young women. Cancer. 1980;45:1615–1624.
    1. Zorn KK, Bonome T, Gangi L, Chandramouli GV, Awtrey CS, et al. Gene expression profiles of serous, endometrioid, and clear cell subtypes of ovarian and endometrial cancer. Clin Cancer Res. 2005;11:6422–6430.
    1. Schwartz DR, Kardia SL, Shedden KA, Kuick R, Michailidis G, et al. Gene expression in ovarian cancer reflects both morphology and biological behavior, distinguishing clear cell from other poor-prognosis ovarian carcinomas. Cancer Res. 2002;62:4722–4729.
    1. Zorn KK, Jazaeri AA, Awtrey CS, Gardner GJ, Mok SC, et al. Choice of normal ovarian control influences determination of differentially expressed genes in ovarian cancer expression profiling studies. Clin Cancer Res. 2003;9:4811–4818.
    1. Bonome T, Lee JY, Park DC, Radonovich M, Pise-Masison C, et al. Expression profiling of serous low malignant potential, low-grade, and high-grade tumors of the ovary. Cancer Res. 2005;65:10602–10612.
    1. Donninger H, Bonome T, Radonovich M, Pise-Masison CA, Brady J, et al. Whole genome expression profiling of advance stage papillary serous ovarian cancer reveals activated pathways. Oncogene. 2004;23:8065–8077.
    1. Aponte M, Jiang W, Lakkis M, Li MJ, Edwards D, et al. Activation of platelet-activating factor receptor and pleiotropic effects on tyrosine phospho-EGFR/Src/FAK/paxillin in ovarian cancer. Cancer Res. 2008;68:5839–5848.
    1. Landen CN, Jr, Chavez-Reyes A, Bucana C, Schmandt R, Deavers MT, et al. Therapeutic EphA2 gene targeting in vivo using neutral liposomal small interfering RNA delivery. Cancer Res. 2005;65:6910–6918.
    1. Halder J, Kamat AA, Landen CN, Jr, Han LY, Lutgendorf SK, et al. Focal adhesion kinase targeting using in vivo short interfering RNA delivery in neutral liposomes for ovarian carcinoma therapy. Clin Cancer Res. 2006;12:4916–4924.
    1. Landen CN, Jr, Lin YG, Armaiz Pena GN, Das PD, Arevalo JM, et al. Neuroendocrine modulation of signal transducer and activator of transcription-3 in ovarian cancer. Cancer Res. 2007;67:10389–10396.
    1. Ebos JM, Lee CR, Christensen JG, Mutsaers AJ, Kerbel RS. Multiple circulating proangiogenic factors induced by sunitinib malate are tumor-independent and correlate with antitumor efficacy. Proc Natl Acad Sci U S A. 2007;104:17069–17074.
    1. Sun L, Liang C, Shirazian S, Zhou Y, Miller T, et al. Discovery of 5-[5-fluoro-2-oxo-1,2- dihydroindol-(3Z)-ylidenemethyl]-2,4- dimethyl-1H-pyrrole-3-carboxylic acid (2-diethylaminoethyl)amide, a novel tyrosine kinase inhibitor targeting vascular endothelial and platelet-derived growth factor receptor tyrosine kinase. J Med Chem. 2003;46:1116–1119.
    1. Lee CH, Xue H, Sutcliffe M, Gout PW, Huntsman DG, et al. Establishment of subrenal capsule xenografts of primary human ovarian tumors in SCID mice: potential models. Gynecol Oncol. 2005;96:48–55.
    1. Press JZ, Kenyon JA, Xue H, Miller MA, De Luca A, et al. Xenografts of primary human gynecological tumors grown under the renal capsule of NOD/SCID mice show genetic stability during serial transplantation and respond to cytotoxic chemotherapy. Gynecol Oncol. 2008;110:256–264.
    1. Mendel DB, Laird AD, Xin X, Louie SG, Christensen JG, et al. In vivo antitumor activity of SU11248, a novel tyrosine kinase inhibitor targeting vascular endothelial growth factor and platelet-derived growth factor receptors: determination of a pharmacokinetic/pharmacodynamic relationship. Clin Cancer Res. 2003;9:327–337.
    1. Semenza GL, Jiang BH, Leung SW, Passantino R, Concordet JP, et al. Hypoxia response elements in the aldolase A, enolase 1, and lactate dehydrogenase A gene promoters contain essential binding sites for hypoxia-inducible factor 1. J Biol Chem. 1996;271:32529–32537.
    1. Kilic M, Kasperczyk H, Fulda S, Debatin KM. Role of hypoxia inducible factor-1 alpha in modulation of apoptosis resistance. Oncogene. 2007;26:2027–2038.
    1. Kelly BD, Hackett SF, Hirota K, Oshima Y, Cai Z, et al. Cell type-specific regulation of angiogenic growth factor gene expression and induction of angiogenesis in nonischemic tissue by a constitutively active form of hypoxia-inducible factor 1. Circ Res. 2003;93:1074–1081.
    1. Tsao MS, Liu N, Nicklee T, Shepherd F, Viallet J. Angiogenesis correlates with vascular endothelial growth factor expression but not with Ki-ras oncogene activation in non-small cell lung carcinoma. Clin Cancer Res. 1997;3:1807–1814.
    1. Nevo O, Soleymanlou N, Wu Y, Xu J, Kingdom J, et al. Increased expression of sFlt-1 in in vivo and in vitro models of human placental hypoxia is mediated by HIF-1. Am J Physiol Regul Integr Comp Physiol. 2006;291:R1085–1093.
    1. Cuadrado MJ, Buendia P, Velasco F, Aguirre MA, Barbarroja N, et al. Vascular endothelial growth factor expression in monocytes from patients with primary antiphospholipid syndrome. J Thromb Haemost. 2006;4:2461–2469.
    1. Crutchley DJ, Que BG. Copper-induced tissue factor expression in human monocytic THP-1 cells and its inhibition by antioxidants. Circulation. 1995;92:238–243.
    1. Wang Z, Banerjee S, Li Y, Rahman KM, Zhang Y, et al. Down-regulation of notch-1 inhibits invasion by inactivation of nuclear factor-kappaB, vascular endothelial growth factor, and matrix metalloproteinase-9 in pancreatic cancer cells. Cancer Res. 2006;66:2778–2784.
    1. Ciofani M, Zuniga-Pflucker JC. Notch promotes survival of pre-T cells at the beta-selection checkpoint by regulating cellular metabolism. Nat Immunol. 2005;6:881–888.
    1. Wu R, Hendrix-Lucas N, Kuick R, Zhai Y, Schwartz DR, et al. Mouse model of human ovarian endometrioid adenocarcinoma based on somatic defects in the Wnt/beta-catenin and PI3K/Pten signaling pathways. Cancer Cell. 2007;11:321–333.
    1. Yamaguchi K, Mandai M, Oura T, Matsumura N, Hamanishi J, et al. Identification of an ovarian clear cell carcinoma gene signature that reflects inherent disease biology and the carcinogenic processes. Oncogene. 2010;29:1741–1752.
    1. Mok SC, Bonome T, Vathipadiekal V, Bell A, Johnson ME, et al. A gene signature predictive for outcome in advanced ovarian cancer identifies a survival factor: microfibril-associated glycoprotein 2. Cancer Cell. 2009;16:521–532.
    1. Sprecher CA, Kisiel W, Mathewes S, Foster DC. Molecular cloning, expression, and partial characterization of a second human tissue-factor-pathway inhibitor. Proc Natl Acad Sci U S A. 1994;91:3353–3357.
    1. Cappellini MD. Coagulation in the pathophysiology of hemolytic anemias. Hematology Am Soc Hematol Educ Program. 2007. pp. 74–78.
    1. Medina P, Navarro S, Estelles A, Espana F. Polymorphisms in the endothelial protein C receptor gene and thrombophilia. Thromb Haemost. 2007;98:564–569.
    1. Maeshima K, Maeshima A, Hayashi Y, Kishi S, Kojima I. Crucial role of activin a in tubulogenesis of endothelial cells induced by vascular endothelial growth factor. Endocrinology. 2004;145:3739–3745.
    1. Poulaki V, Joussen AM, Mitsiades N, Mitsiades CS, Iliaki EF, et al. Insulin-like growth factor-I plays a pathogenetic role in diabetic retinopathy. Am J Pathol. 2004;165:457–469.
    1. Frede S, Freitag P, Otto T, Heilmaier C, Fandrey J. The proinflammatory cytokine interleukin 1beta and hypoxia cooperatively induce the expression of adrenomedullin in ovarian carcinoma cells through hypoxia inducible factor 1 activation. Cancer Res. 2005;65:4690–4697.
    1. Ito N, Huang K, Claesson-Welsh L. Signal transduction by VEGF receptor-1 wild type and mutant proteins. Cell Signal. 2001;13:849–854.
    1. Pollard PJ, El-Bahrawy M, Poulsom R, Elia G, Killick P, et al. Expression of HIF-1alpha, HIF-2alpha (EPAS1), and their target genes in paraganglioma and pheochromocytoma with VHL and SDH mutations. J Clin Endocrinol Metab. 2006;91:4593–4598.
    1. Isaacs JS, Jung YJ, Mimnaugh EG, Martinez A, Cuttitta F, et al. Hsp90 regulates a von Hippel Lindau-independent hypoxia-inducible factor-1 alpha-degradative pathway. J Biol Chem. 2002;277:29936–29944.
    1. Gwak GY, Yoon JH, Kim KM, Lee HS, Chung JW, et al. Hypoxia stimulates proliferation of human hepatoma cells through the induction of hexokinase II expression. J Hepatol. 2005;42:358–364.
    1. Welsh S, Williams R, Kirkpatrick L, Paine-Murrieta G, Powis G. Antitumor activity and pharmacodynamic properties of PX-478, an inhibitor of hypoxia-inducible factor-1alpha. Mol Cancer Ther. 2004;3:233–244.
    1. Chen J, Zhao S, Nakada K, Kuge Y, Tamaki N, et al. Dominant-negative hypoxia-inducible factor-1 alpha reduces tumorigenicity of pancreatic cancer cells through the suppression of glucose metabolism. Am J Pathol. 2003;162:1283–1291.
    1. Gitlits VM, Toh BH, Sentry JW. Disease association, origin, and clinical relevance of autoantibodies to the glycolytic enzyme enolase. J Investig Med. 2001;49:138–145.
    1. Walter M, Berg H, Leidenberger FA, Schweppe KW, Northemann W. Autoreactive epitopes within the human alpha-enolase and their recognition by sera from patients with endometriosis. J Autoimmun. 1995;8:931–945.
    1. Wu MH, Chen KF, Lin SC, Lgu CW, Tsai SJ. Aberrant expression of leptin in human endometriotic stromal cells is induced by elevated levels of hypoxia inducible factor-1alpha. Am J Pathol. 2007;170:590–598.

Source: PubMed

3
Tilaa