Mapping PAM4 (clivatuzumab), a monoclonal antibody in clinical trials for early detection and therapy of pancreatic ductal adenocarcinoma, to MUC5AC mucin

David V Gold, Guy Newsome, Donglin Liu, David M Goldenberg, David V Gold, Guy Newsome, Donglin Liu, David M Goldenberg

Abstract

Background: PAM4, an antibody that has high specificity for pancreatic ductal adenocarcinoma (PDAC), compared to normal pancreas, benign lesions of the pancreas, and cancers originating from other tissues, is being investigated as a biomarker for early detection, as well as antibody-targeted imaging and therapy. Therefore, the identity of the antigen bound by this monoclonal antibody (MAb) can provide information leading to improved use of the antibody. Prior results suggested the antigen is a mucin-type glycoprotein rich in cysteine disulfide bridges that provide stable conformation for the PAM4-epitope.

Methods: Indirect and sandwich enzyme immunoassays (EIA) were performed to compare and contrast the reactivity of PAM4 with several anti-mucin antibodies having known reactivity to specific mucin species (e.g., MUC1, MUC4, MUC5AC, etc.). Studies designed to block reactivity of PAM4 with its specific antigen also were performed.

Results: We demonstrate that MAbs 2-11 M1 and 45 M1, each reactive with MUC5AC, are able to provide signal in a heterologous sandwich immunoassay where PAM4 is the capture antibody. Further, we identify MAbs 21 M1, 62 M1, and 463 M1, each reactive with MUC5AC, as inhibiting the reaction of PAM4 with its specific epitope. MAbs directed to MUC1, MUC3, MUC4, MUC16 and CEACAM6 are not reactive with PAM4-captured antigen, nor are they able to block the reaction of PAM4 with its antigen.

Conclusions: These data implicate MUC5AC as a specific mucin species to which PAM4 is reactive. Furthermore, this realization may allow for the improvement of the current PAM4 serum-based immunoassay for detection of early-stage PDAC by the application of anti-MUC5AC MAbs as probes in this sandwich EIA.

Figures

Figure 1
Figure 1
Reactivity of several anti-mucin MAbs with a high molecular weight mucin containing fraction (CPM1) isolated from the Capan-1 human pancreatic adenocarcinoma. MAbs are identified by clone name with reactive species of mucin indicated by horizontal bars beneath MAb clone names (MUC1, etc.). In addition to PAM4, substantial reactions were observed for anti-MUC1, -MUC5AC, and -CEACAM6 antibodies. All MAbs were employed at a concentration of 10 μg/mL.
Figure 2
Figure 2
Reaction of several anti-mucin MAbs with PAM4-captured antigen. Mucin antigens were captured on hPAM4 coated plates, and then probed with several murine anti-mucin MAbs for reaction signal. Both anti-MUC5AC MAbs (2-11 M1 and 45 M1) bound to the hPAM4-captured mucin, whereas the anti-MUC1 MAbs (MA5 and KC4) did not bind. The homologous hPAM4/mPAM4, capture/probe immunoassay gave no signal, suggesting the density of PAM4 epitopes within the mucin may be low, possibly only a single site. A rabbit polyclonal anti-CPM1 IgG was used as a positive control for reaction with hPAM4-captured antigen.
Figure 3
Figure 3
Inhibition of hPAM4/antigen binding reaction by murine anti-mucin MAbs. A) Anti-mucin mMAbs (purified IgG) were added to CPM1-coated plates as potential inhibitors prior to addition of hPAM4. mPAM4 provided almost complete inhibition of the reaction between hPAM4 and antigen with the 45 M1 anti-MUC5AC providing limited inhibitory effect (ICmax = 25.5%). Neither 2-11 M1, anti-MUC5AC nor MA5 and KC4, anti-MUC1 MAbs were able to inhibit the specific hPAM4/antigen reaction. B) A similar inhibition study was performed with several anti-MUC5AC MAbs obtained as ascites fluids. MAbs 21 M1, 62 M1, and 463 M1, anti-MUC5AC provided substantial inhibitory effect, similar to that observed with mPAM4 IgG self-inhibition. The ascites form of 45 M1 yielded an inhibitory effect similar to that of the purified IgG. Ascites containing anti-alpha fetoprotein was employed as a negative control.
Figure 4
Figure 4
Representation of the domains of the MUC5AC glycoprotein with reactive epitopes indicated for several anti-MUC5AC MAbs. (Derived from references [22,23,28]). Data derived by transfection with plasmid vectors containing the cDNA of the 3’-end of MUC5AC, along with derivative cDNA vectors obtained by restriction enzyme digestion, have identified the location of specific epitopes for anti-MUC5AC MAbs employed in the current studies. Specific blocking studies (Figures 3A and B) suggest the PAM4-epitope resides within the cysteine-rich C-terminus domain.

References

    1. Hollingsworth MA, Swanson BJ. Mucins in cancer: protection and control of the cell surface. Nat Rev Cancer. 2004;12(1):45–60. doi: 10.1038/nrc1251.
    1. Kufe DW. Mucins in cancer: function, prognosis and therapy. Nat Rev Cancer. 2009;12(12):874–885. doi: 10.1038/nrc2761.
    1. Rachagani S, Torres MP, Moniaux N, Batra SK. Current status of mucins in the diagnosis and therapy of cancer. Biofactors. 2009;12(6):509–527. doi: 10.1002/biof.64.
    1. Ringel J, Lohr M. The MUC gene family: their role in the diagnosis and early detection of pancreatic cancer. Mol Cancer. 2003;12:9–13. doi: 10.1186/1476-4598-2-9.
    1. Andrianifahanana M, Moniaux N, Schmied BM, Ringel J, Friess H, Hollingsworth MA, Buchler MW, Aubert JP, Batra SK. Mucin (MUC) gene expression in human pancreatic adenocarcinoma and chronic pancreatitis: a potential role of MUC4 as a tumor marker of diagnostic significance. Clin Cancer Res. 2001;12(12):4033–4040.
    1. Torres MP, Chakraborty S, Souchek J, Batra SK. Mucin-based targeted pancreatic cancer therapy. Curr Pharm Des. 2012;12(17):2472–2481. doi: 10.2174/13816128112092472.
    1. Schmid BC, Rudas M, Fabjani G, Speiser P, Kaserer K, Leodolter S, Zeillinger R. Evaluation of MUC1 splice variants as prognostic markers in patients with ductal carcinoma in situ of the breast. Oncol Rep. 2003;12(6):1981–1985.
    1. Brockhausen I. Mucin-type O-glycans in human colon and breast cancer: glycodynamics and functions. EMBO Rep. 2006;12(6):599–604. doi: 10.1038/sj.embor.7400705.
    1. Yue T, Goldstein IJ, Hollingsworth MA, Kaul K, Brand RE, Haab BB. The prevalence and nature of glycan alterations on specific proteins in pancreatic cancer patients revealed using antibody-lectin sandwich arrays. Mol Cell Proteomics. 2009;12(7):1697–1707. doi: 10.1074/mcp.M900135-MCP200.
    1. Haab BB, Porter A, Yue T, Li L, Scheiman J, Anderson MA, Barnes D, Schmidt CM, Feng Z, Simeone DM. Glycosylation variants of mucins and CEACAMs as candidate biomarkers for the diagnosis of pancreatic cystic neoplasms. Ann Surg. 2010;12(5):937–945. doi: 10.1097/SLA.0b013e3181d7738d.
    1. Jass JR, Allison LJ, Edgar SG. Distribution of sialosyl Tn and Tn antigens within normal and malignant colorectal epithelium. J Pathol. 1995;12(2):143–149. doi: 10.1002/path.1711760207.
    1. Cao Y, Schlag PM, Karsten U. Immunodetection of epithelial mucin (MUC1, MUC3) and mucin-associated glycotopes (TF, Tn, and sialosyl-Tn) in benign and malignant lesions of colonic epithelium: apolar localization corresponds to malignant transformation. Virchows Arch. 1997;12(3):159–166. doi: 10.1007/s004280050083.
    1. Gold DV, Hollingsworth P, Kremer T, Nelson D. Identification of a human pancreatic duct tissue-specific antigen. Cancer Res. 1983;12(1):235–238.
    1. Gold DV, Lew K, Maliniak R, Hernandez M, Cardillo T. Characterization of monoclonal antibody PAM4 reactive with a pancreatic cancer mucin. Int J Cancer. 1994;12(2):204–210. doi: 10.1002/ijc.2910570213.
    1. Gold DV, Karanjawala Z, Modrak DE, Goldenberg DM, Hruban RH. PAM4-reactive MUC1 is a biomarker for early pancreatic adenocarcinoma. Clin Cancer Res. 2007;12(24):7380–7387. doi: 10.1158/1078-0432.CCR-07-1488.
    1. Gold DV, Goggins M, Modrak DE, Newsome G, Liu M, Shi C, Hruban RH, Goldenberg DM. Detection of early-stage pancreatic adenocarcinoma. Cancer Epidemiol Biomarkers Prev. 2010;12(11):2786–2794. doi: 10.1158/1055-9965.EPI-10-0667.
    1. Gold DV, Modrak DE, Ying Z, Cardillo TM, Sharkey RM, Goldenberg DM. New MUC1 serum immunoassay differentiates pancreatic cancer from pancreatitis. J Clin Oncol. 2006;12(2):252–258.
    1. Gold DV, Gaedcke J, Ghadimi BM, Goggins M, Hruban RH, Liu M, Newsome G, Goldenberg DM. PAM4 enzyme immunoassay alone and in combination with CA 19–9 for the detection of pancreatic adenocarcinoma. Cancer. 2013;12(3):522–528. doi: 10.1002/cncr.27762.
    1. Gulec SA, Cohen SJ, Pennington KL, Zuckier LS, Hauke RJ, Horne H, Wegener WA, Teoh N, Gold DV, Sharkey RM, Goldenberg DM. Treatment of advanced pancreatic carcinoma with 90Y-clivatuzumab tetraxetan: a phase I single-dose escalation trial. Clin Cancer Res. 2011;12(12):4091–4100. doi: 10.1158/1078-0432.CCR-10-2579.
    1. Ocean AJ, Pennington KL, Guarino MJ, Sheikh A, Bekaii-Saab T, Serafini AN, Lee D, Sung MW, Gulec SA, Goldsmith SJ, Manzone T, Holt M, O'Neil BH, Hall N, Montero AJ, Kauh J, Gold DV, Horne H, Wegener WA, Goldenberg DM. Fractionated radioimmunotherapy with 90Y-clivatuzumab tetraxetan and low-dose gemcitabine is active in advanced pancreatic cancer: a phase 1 trial. Cancer. 2012;12(22):5497–5506. doi: 10.1002/cncr.27592.
    1. Reis CA, David L, Nielsen PA, Clausen H, Mirgorodskaya K, Roepstorff P, Sobrinho-Simoes M. Immunohistochemical study of MUC5AC expression in human gastric carcinomas using a novel monoclonal antibody. Int J Cancer. 1997;12(1):112–121. doi: 10.1002/(SICI)1097-0215(19970220)74:1<112::AID-IJC19>;2-H.
    1. Nollet S, Forgue-Lafitte ME, Kirkham P, Bara J. Mapping of two new epitopes on the apomucin encoded by MUC5AC gene: expression in normal GI tract and colon tumors. Int J Cancer. 2002;12(3):336–343. doi: 10.1002/ijc.10335.
    1. Nollet S, Escande F, Buisine MP, Forgue-Lafitte ME, Kirkham P, Okada Y, Bara J. Mapping of SOMU1 and M1 epitopes on the apomucin encoded by the 5' end of the MUC5AC gene. Hybrid Hybridomics. 2004;12(2):93–99. doi: 10.1089/153685904774129694.
    1. Lidell ME, Bara J, Hansson GC. Mapping of the 45 M1 epitope to the C-terminal cysteine-rich part of the human MUC5AC mucin. FEBS J. 2008;12(3):481–489. doi: 10.1111/j.1742-4658.2007.06215.x.
    1. Gold DV. Reply to the Letter to the Editor by BARA et al. Clin Cancer Res. 2008;12(16):5306–5307.
    1. Sipos B, Möser S, Kalthoff H, Török V, Löhr M, Klöppel G. A comprehensive characterization of pancreatic ductal carcinoma cell lines: towards the establishment of an in vitro research platform. Virchows Arch. 2003;12(5):444–452.
    1. Bara J, Gautier R, Mouradian P, Decaens C, Daher N. Oncofetal mucin M1 epitope family: characterization and expression during colonic carcinogenesis. Int J Cancer. 1991;12(2):304–310. doi: 10.1002/ijc.2910470222.
    1. Lidell ME, Hansson GC. Cleavage in the GDPH sequence of the C-terminal cysteine-rich part of the human MUC5AC mucin. Biochem J. 2006;12(1):121–129. doi: 10.1042/BJ20060443.

Source: PubMed

3
購読する