Yonsei Med J.  2014 Jul;55(4):1014-1027. 10.3349/ymj.2014.55.4.1014.

Identification of Pancreatic Cancer-Associated Tumor Antigen from HSP-Enriched Tumor Lysate-Pulsed Human Dendritic Cells

Affiliations
  • 1Innovative Cell and Gene Therapy Center, International St. Mary's Hospital, Incheon, Korea.
  • 2Center for Bioanalysis, Division of Metrology for Quality of Life, Korea Research Institute of Standards and Science, Daejeon, Korea.
  • 3Department of Chemistry, Yonsei University, Seoul, Korea. mhmoon@yonsei.ac.kr
  • 4Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Korea. hyung@hallym.or.kr

Abstract

PURPOSE
Vaccine strategies utilizing dendritic cells (DCs) to elicit anti-tumor immunity are the subject of intense research. Although we have shown that DCs pulsed with heat-treated tumor lysate (HTL) induced more potent anti-tumor immunity than DCs pulsed with conventional tumor lysate (TL), the underlying molecular mechanism is unclear. In order to explore the molecular basis of this approach and to identify potential antigenic peptides from pancreatic cancer, we analyzed and compared the major histocompatibility complex (MHC) ligands derived from TL- and HTL-pulsed dendritic cells by mass spectrophotometry.
MATERIALS AND METHODS
Human monocyte-derived dendritic cells were pulsed with TL or HTL prior to maturation induction. To delineate differences of MHC-bound peptide repertoire eluted from DCs pulsed with TL or HTL, nanoflow liquid chromatography-electrospray ionization-tandem mass spectrometry (nLC-ESI-MS-MS) was employed.
RESULTS
HTL, but not TL, significantly induced DC function, assessed by phenotypic maturation, allostimulation capacity and IFN-gamma secretion by stimulated allogeneic T cells. DCs pulsed with TL or HTL displayed pancreas or pancreatic cancer-related peptides in context of MHC class I and II molecules. Some of the identified peptides had not been previously reported as expressed in pancreatic cancer or cancer of other tissue types.
CONCLUSION
Our partial lists of MHC-associated peptides revealed the differences between peptide profiles eluted from HTL-and TL-loaded DCs, implying that induced heat shock proteins in HTL chaperone tumor-derived peptides enhanced their delivery to DCs and promoted cross-presentation by DC. These findings may aid in identifying novel tumor antigens or biomarkers and in designing future vaccination strategies.

Keyword

Tumor lysate; heat shock proteins; dendritic cells; tumor antigens; nanoflow LC-MS-MS

MeSH Terms

Antigens, Neoplasm/*immunology
Cell Line, Tumor
Dendritic Cells/*immunology
Humans
Pancreatic Neoplasms/*immunology
Antigens, Neoplasm
Pancreatic Carcinoma

Figure

  • Fig. 1 Western blot analysis for HSP family protein expression by TL and HTL. Human gastric cancer cell line (N87) and human pancreatic ductal adenocarcinoma cell line (Panc-1) were exposed to 42℃ for 1 hr, followed by recovery for 24 hr at 37℃. Tumor cell lysates from untreated tumor lysate (TL) or tumor lysate preparation after heat treatment (HTL) were prepared as described in the Materials and Methods section and immunoblotted with indicated antibodies. HTL, heat-treated tumor lysate; TL, tumor lysate; HSP, heat shock protein.

  • Fig. 2 Phenotypic maturation of dendritic cells induced by HTL, HSP-enriched tumor lysate prepared from heat-treated tumor cells. Human dendritic cells (5×105/mL) were incubated with 4 mg/mL of TL or HTL for 4 hr. After washing, cells were cultured in the presence of GM-CSF and IL-4 (iDC) or in the presence of GM-CSF, IL-4 with cytokine cocktail, composed of TNF-α, IL-1β, IL-6, and PGE2 (mDC), for 48 hr. DCs were collected and stained with monoclonal antibodies specific to HLA-DR, CD40, CD86, CD80, CD1a, and CD83. Gray histograms indicate untreated DCs, fine-line histograms, TL-pulsed DCs; and thick line histograms, HTL-pulsed DCs. Histograms show the expression of CD1a, CD40, CD80, CD86, HLA-DR, and CD83, which represent one representative experiment of four performed. Numbers denote MFI values. HTL, heat-treated tumor lysate; TL, tumor lysate; HSP, heat shock protein; DC, dendritic cell; iDC, immature DC; MFI, median fluorescence intensity; mDC, mature DC.

  • Fig. 3 HTL-pulsed DCs enhanced T cell proliferation, IL-12 production and IFN-γ production. (A) DC exhibits enhanced allostimulatory capacity after pulsing with HTL compared to that of TL-pulsed DC. Gamma-irradiated DC were added to CD3+ T cells at the indicated DC/T cell ratio and incubated for an 5 days before the addition of [3H] thymidine to assess T cell proliferation. Data represent one of three independent experiments with similar results. The error bars are the SEM of triplicate determinations. (B) The production of IL-12p70 was slightly increased by HTL-pulsing. Mature DCs were generated by stimulating immature DCs (iDC) with cytokine cocktail for 48 hr. The error bars are the SD of triplicate determinations. (C) Measurement of IFN-γ in supernatants of allogeneic T cells stimulated by unpulsed, TL-pulsed or HTL-pulsed mature DCs. Allogeneic T cells were stimulated as described in Materials and Methods. The error bars are the SD of triplicate determinations. *p<0.05, **p<0.01. HTL, heat-treated tumor lysate; TL, tumor lysate; DC, dendritic cell.

  • Fig. 4 Base peak chromatograms of naturally processed MHC class I- and II-associated peptides derived from TL-DC (A) and HTL-DC (B). Peptides were eluted from DCs by treatment with citrate-phosphate buffer (pH 3.3) and peptides were separated using a homemade pulled tip capillary column by binary gradient elution at 200 nL/min (see details in Materials and Method section). TL, tumor lysate; DC, dendritic cell; HTL, heat-treated tumor lysate; MHC, major histocompatibility complex.

  • Fig. 5 Identification of tumor-associated peptides eluted from TL-DC or HTL-DC. The MS-MS spectra of m/z 625.94 ([M+3H]3+) from HTL-DC (A) and of m/z 828.32 ([M+2H]2+) from TL (B), which were found to originate from the same protein, human S100 calcium binding protein A4 (residues 94-110). HTL, heat-treated tumor lysate; TL, tumor lysate; DC, dendritic cell.


Reference

1. Berger TG, Feuerstein B, Strasser E, Hirsch U, Schreiner D, Schuler G, et al. Large-scale generation of mature monocyte-derived dendritic cells for clinical application in cell factories. J Immunol Methods. 2002; 268:131–140.
Article
2. Kim S, Kim HO, Baek EJ, Choi Y, Kim HS, Lee MG. Monocyte enrichment from leukapheresis products by using the Elutra cell separator. Transfusion. 2007; 47:2290–2296.
Article
3. Schreurs MW, Eggert AA, de Boer AJ, Vissers JL, van Hall T, Offringa R, et al. Dendritic cells break tolerance and induce protective immunity against a melanocyte differentiation antigen in an autologous melanoma model. Cancer Res. 2000; 60:6995–7001.
4. Hofmann S, Glückmann M, Kausche S, Schmidt A, Corvey C, Lichtenfels R, et al. Rapid and sensitive identification of major histocompatibility complex class I-associated tumor peptides by Nano-LC MALDI MS/MS. Mol Cell Proteomics. 2005; 4:1888–1897.
Article
5. Ridge JP, Di Rosa F, Matzinger P. A conditioned dendritic cell can be a temporal bridge between a CD4+ T-helper and a T-killer cell. Nature. 1998; 393:474–478.
Article
6. Hu HM, Winter H, Urba WJ, Fox BA. Divergent roles for CD4+ T cells in the priming and effector/memory phases of adoptive immunotherapy. J Immunol. 2000; 165:4246–4253.
Article
7. Wang RF. The role of MHC class II-restricted tumor antigens and CD4+ T cells in antitumor immunity. Trends Immunol. 2001; 22:269–276.
Article
8. Schuurhuis DH, Laban S, Toes RE, Ricciardi-Castagnoli P, Kleijmeer MJ, van der Voort EI, et al. Immature dendritic cells acquire CD8(+) cytotoxic T lymphocyte priming capacity upon activation by T helper cell-independent or -dependent stimuli. J Exp Med. 2000; 192:145–150.
Article
9. Melief CJ, Van Der Burg SH, Toes RE, Ossendorp F, Offringa R. Effective therapeutic anticancer vaccines based on precision guiding of cytolytic T lymphocytes. Immunol Rev. 2002; 188:177–182.
Article
10. Dolan BP, Gibbs KD Jr, Ostrand-Rosenberg S. Tumor-specific CD4+ T cells are activated by "cross-dressed" dendritic cells presenting peptide-MHC class II complexes acquired from cell-based cancer vaccines. J Immunol. 2006; 176:1447–1455.
Article
11. Parmiani G, Castelli C, Dalerba P, Mortarini R, Rivoltini L, Marincola FM, et al. Cancer immunotherapy with peptide-based vaccines: what have we achieved? Where are we going? J Natl Cancer Inst. 2002; 94:805–818.
Article
12. Admon A, Barnea E, Ziv T. Tumor antigens and proteomics from the point of view of the major histocompatibility complex peptides. Mol Cell Proteomics. 2003; 2:388–398.
Article
13. Fields RC, Shimizu K, Mulé JJ. Murine dendritic cells pulsed with whole tumor lysates mediate potent antitumor immune responses in vitro and in vivo. Proc Natl Acad Sci U S A. 1998; 95:9482–9487.
Article
14. Nestle FO, Alijagic S, Gilliet M, Sun Y, Grabbe S, Dummer R, et al. Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nat Med. 1998; 4:328–332.
Article
15. Schnurr M, Galambos P, Scholz C, Then F, Dauer M, Endres S, et al. Tumor cell lysate-pulsed human dendritic cells induce a T-cell response against pancreatic carcinoma cells: an in vitro model for the assessment of tumor vaccines. Cancer Res. 2001; 61:6445–6450.
16. Yu JS, Liu G, Ying H, Yong WH, Black KL, Wheeler CJ. Vaccination with tumor lysate-pulsed dendritic cells elicits antigen-specific, cytotoxic T-cells in patients with malignant glioma. Cancer Res. 2004; 64:4973–4979.
Article
17. Geiger JD, Hutchinson RJ, Hohenkirk LF, McKenna EA, Yanik GA, Levine JE, et al. Vaccination of pediatric solid tumor patients with tumor lysate-pulsed dendritic cells can expand specific T cells and mediate tumor regression. Cancer Res. 2001; 61:8513–8519.
18. Höltl L, Zelle-Rieser C, Gander H, Papesh C, Ramoner R, Bartsch G, et al. Immunotherapy of metastatic renal cell carcinoma with tumor lysate-pulsed autologous dendritic cells. Clin Cancer Res. 2002; 8:3369–3376.
19. Hatfield P, Merrick AE, West E, O'Donnell D, Selby P, Vile R, et al. Optimization of dendritic cell loading with tumor cell lysates for cancer immunotherapy. J Immunother. 2008; 31:620–632.
Article
20. Sauter B, Albert ML, Francisco L, Larsson M, Somersan S, Bhardwaj N. Consequences of cell death: exposure to necrotic tumor cells, but not primary tissue cells or apoptotic cells, induces the maturation of immunostimulatory dendritic cells. J Exp Med. 2000; 191:423–434.
21. Kotera Y, Shimizu K, Mulé JJ. Comparative analysis of necrotic and apoptotic tumor cells as a source of antigen(s) in dendritic cell-based immunization. Cancer Res. 2001; 61:8105–8109.
22. Galea-Lauri J, Wells JW, Darling D, Harrison P, Farzaneh F. Strategies for antigen choice and priming of dendritic cells influence the polarization and efficacy of antitumor T-cell responses in dendritic cell-based cancer vaccination. Cancer Immunol Immunother. 2004; 53:963–977.
Article
23. Steinman RM, Turley S, Mellman I, Inaba K. The induction of tolerance by dendritic cells that have captured apoptotic cells. J Exp Med. 2000; 191:411–416.
Article
24. Kim HS, Choo YS, Koo T, Bang S, Oh TY, Wen J, et al. Enhancement of antitumor immunity of dendritic cells pulsed with heat-treated tumor lysate in murine pancreatic cancer. Immunol Lett. 2006; 103:142–148.
Article
25. Qiu J, Li GW, Sui YF, Song HP, Si SY, Ge W. Heat-shocked tumor cell lysate-pulsed dendritic cells induce effective anti-tumor immune response in vivo. World J Gastroenterol. 2006; 12:473–478.
Article
26. Bachleitner-Hofmann T, Stift A, Friedl J, Pfragner R, Radelbauer K, Dubsky P, et al. Stimulation of autologous antitumor T-cell responses against medullary thyroid carcinoma using tumor lysate-pulsed dendritic cells. J Clin Endocrinol Metab. 2002; 87:1098–1104.
Article
27. Arnold-Schild D, Hanau D, Spehner D, Schmid C, Rammensee HG, de la Salle H, et al. Cutting edge: receptor-mediated endocytosis of heat shock proteins by professional antigen-presenting cells. J Immunol. 1999; 162:3757–3760.
28. Babatz J, Röllig C, Oelschlägel U, Zhao S, Ehninger G, Schmitz M, et al. Large-scale immunomagnetic selection of CD14+ monocytes to generate dendritic cells for cancer immunotherapy: a phase I study. J Hematother Stem Cell Res. 2003; 12:515–523.
Article
29. Santin AD, Bellone S, Ravaggi A, Pecorelli S, Cannon MJ, Parham GP. Induction of ovarian tumor-specific CD8+ cytotoxic T lymphocytes by acid-eluted peptide-pulsed autologous dendritic cells. Obstet Gynecol. 2000; 96:422–430.
Article
30. Kang D, Moon MH. Development of non-gel-based two-dimensional separation of intact proteins by an on-line hyphenation of capillary isoelectric focusing and hollow fiber flow field-flow fractionation. Anal Chem. 2006; 78:5789–5798.
Article
31. Kang D, Ji ES, Moon MH, Yoo JS. Lectin-based enrichment method for glycoproteomics using hollow fiber flow field-flow fractionation: application to Streptococcus pyogenes. J Proteome Res. 2010; 9:2855–2862.
Article
32. Srivastava PK. Immunotherapy for human cancer using heat shock protein-peptide complexes. Curr Oncol Rep. 2005; 7:104–108.
Article
33. Salio M, Cerundolo V, Lanzavecchia A. Dendritic cell maturation is induced by mycoplasma infection but not by necrotic cells. Eur J Immunol. 2000; 30:705–708.
Article
34. Reynolds JL, Mahajan SD, Aalinkeel R, Nair B, Sykes DE, Schwartz SA. Proteomic analyses of the effects of drugs of abuse on monocyte-derived mature dendritic cells. Immunol Invest. 2009; 38:526–550.
Article
35. Reynolds JL, Mahajan SD, Sykes DE, Schwartz SA, Nair MP. Proteomic analyses of methamphetamine (METH)-induced differential protein expression by immature dendritic cells (IDC). Biochim Biophys Acta. 2007; 1774:433–442.
Article
36. Horlock C, Shakib F, Mahdavi J, Jones NS, Sewell HF, Ghaemmaghami AM. Analysis of proteomic profiles and functional properties of human peripheral blood myeloid dendritic cells, monocyte-derived dendritic cells and the dendritic cell-like KG-1 cells reveals distinct characteristics. Genome Biol. 2007; 8:R30.
Article
37. Watarai H, Hinohara A, Nagafune J, Nakayama T, Taniguchi M, Yamaguchi Y. Plasma membrane-focused proteomics: dramatic changes in surface expression during the maturation of human dendritic cells. Proteomics. 2005; 5:4001–4011.
Article
38. McIlroy D, Tanguy-Royer S, Le Meur N, Guisle I, Royer PJ, Léger J, et al. Profiling dendritic cell maturation with dedicated microarrays. J Leukoc Biol. 2005; 78:794–803.
Article
39. Pereira SR, Faça VM, Gomes GG, Chammas R, Fontes AM, Covas DT, et al. Changes in the proteomic profile during differentiation and maturation of human monocyte-derived dendritic cells stimulated with granulocyte macrophage colony stimulating factor/interleukin-4 and lipopolysaccharide. Proteomics. 2005; 5:1186–1198.
Article
40. Rivollier A, Perrin-Cocon L, Luche S, Diemer H, Strub JM, Hanau D, et al. High expression of antioxidant proteins in dendritic cells: possible implications in atherosclerosis. Mol Cell Proteomics. 2006; 5:726–736.
41. Ferret-Bernard S, Curwen RS, Mountford AP. Proteomic profiling reveals that Th2-inducing dendritic cells stimulated with helminth antigens have a 'limited maturation' phenotype. Proteomics. 2008; 8:980–993.
Article
42. Ferreira GB, van Etten E, Lage K, Hansen DA, Moreau Y, Workman CT, et al. Proteome analysis demonstrates profound alterations in human dendritic cell nature by TX527, an analogue of vitamin D. Proteomics. 2009; 9:3752–3764.
Article
43. Gundacker NC, Haudek VJ, Wimmer H, Slany A, Griss J, Bochkov V, et al. Cytoplasmic proteome and secretome profiles of differently stimulated human dendritic cells. J Proteome Res. 2009; 8:2799–2811.
Article
44. Shiwa M, Nishimura Y, Wakatabe R, Fukawa A, Arikuni H, Ota H, et al. Rapid discovery and identification of a tissue-specific tumor biomarker from 39 human cancer cell lines using the SELDI ProteinChip platform. Biochem Biophys Res Commun. 2003; 309:18–25.
Article
45. Park T, Chen ZP, Leavitt J. Activation of the leukocyte plastin gene occurs in most human cancer cells. Cancer Res. 1994; 54:1775–1781.
46. Shimada H, Shiratori T, Yasuraoka M, Kagaya A, Kuboshima M, Nomura F, et al. Identification of Makorin 1 as a novel SEREX antigen of esophageal squamous cell carcinoma. BMC Cancer. 2009; 9:232.
Article
47. Li YN, Zhang L, Li XL, Cui DJ, Zheng HD, Yang SY, et al. Glycoprotein nonmetastatic B as a prognostic indicator in small cell lung cancer. APMIS. 2014; 122:140–146.
Article
48. Kabbage M, Chahed K, Hamrita B, Guillier CL, Trimeche M, Remadi S, et al. Protein alterations in infiltrating ductal carcinomas of the breast as detected by nonequilibrium pH gradient electrophoresis and mass spectrometry. J Biomed Biotechnol. 2008; 2008:564127.
Article
49. Gongoll S, Peters G, Mengel M, Piso P, Klempnauer J, Kreipe H, et al. Prognostic significance of calcium-binding protein S100A4 in colorectal cancer. Gastroenterology. 2002; 123:1478–1484.
Article
50. Shen J, Person MD, Zhu J, Abbruzzese JL, Li D. Protein expression profiles in pancreatic adenocarcinoma compared with normal pancreatic tissue and tissue affected by pancreatitis as detected by two-dimensional gel electrophoresis and mass spectrometry. Cancer Res. 2004; 64:9018–9026.
Article
51. Melle C, Ernst G, Escher N, Hartmann D, Schimmel B, Bleul A, et al. Protein profiling of microdissected pancreas carcinoma and identification of HSP27 as a potential serum marker. Clin Chem. 2007; 53:629–635.
Article
52. Nakatsura T, Senju S, Ito M, Nishimura Y, Itoh K. Cellular and humoral immune responses to a human pancreatic cancer antigen, coactosin-like protein, originally defined by the SEREX method. Eur J Immunol. 2002; 32:826–836.
Article
53. Wang B, Sun J, Kitamoto S, Yang M, Grubb A, Chapman HA, et al. Cathepsin S controls angiogenesis and tumor growth via matrix-derived angiogenic factors. J Biol Chem. 2006; 281:6020–6029.
Article
54. Rosty C, Ueki T, Argani P, Jansen M, Yeo CJ, Cameron JL, et al. Overexpression of S100A4 in pancreatic ductal adenocarcinomas is associated with poor differentiation and DNA hypomethylation. Am J Pathol. 2002; 160:45–50.
Article
55. Celluzzi CM, Mayordomo JI, Storkus WJ, Lotze MT, Falo LD Jr. Peptide-pulsed dendritic cells induce antigen-specific CTL-mediated protective tumor immunity. J Exp Med. 1996; 183:283–287.
Article
56. Hoffmann TK, Meidenbauer N, Dworacki G, Kanaya H, Whiteside TL. Generation of tumor-specific T-lymphocytes by cross-priming with human dendritic cells ingesting apoptotic tumor cells. Cancer Res. 2000; 60:3542–3549.
57. Gong J, Avigan D, Chen D, Wu Z, Koido S, Kashiwaba M, et al. Activation of antitumor cytotoxic T lymphocytes by fusions of human dendritic cells and breast carcinoma cells. Proc Natl Acad Sci U S A. 2000; 97:2715–2718.
Article
58. Ashley DM, Faiola B, Nair S, Hale LP, Bigner DD, Gilboa E. Bone marrow-generated dendritic cells pulsed with tumor extracts or tumor RNA induce antitumor immunity against central nervous system tumors. J Exp Med. 1997; 186:1177–1182.
Article
59. Cranmer LD, Trevor KT, Hersh EM. Clinical applications of dendritic cell vaccination in the treatment of cancer. Cancer Immunol Immunother. 2004; 53:275–306.
Article
60. Kammerer R, Stober D, Riedl P, Oehninger C, Schirmbeck R, Reimann J. Noncovalent association with stress protein facilitates cross-priming of CD8+ T cells to tumor cell antigens by dendritic cells. J Immunol. 2002; 168:108–117.
Article
61. Delneste Y. Scavenger receptors and heat-shock protein-mediated antigen cross-presentation. Biochem Soc Trans. 2004; 32(Pt 4):633–635.
Article
62. Becker T, Hartl FU, Wieland F. CD40, an extracellular receptor for binding and uptake of Hsp70-peptide complexes. J Cell Biol. 2002; 158:1277–1285.
Article
63. Basu S, Binder RJ, Ramalingam T, Srivastava PK. CD91 is a common receptor for heat shock proteins gp96, hsp90, hsp70, and calreticulin. Immunity. 2001; 14:303–313.
Article
64. Doody AD, Kovalchin JT, Mihalyo MA, Hagymasi AT, Drake CG, Adler AJ. Glycoprotein 96 can chaperone both MHC class I- and class II-restricted epitopes for in vivo presentation, but selectively primes CD8+ T cell effector function. J Immunol. 2004; 172:6087–6092.
Article
65. Wu Y, Wan T, Zhou X, Wang B, Yang F, Li N, et al. Hsp70-like protein 1 fusion protein enhances induction of carcinoembryonic antigen-specific CD8+ CTL response by dendritic cell vaccine. Cancer Res. 2005; 65:4947–4954.
Article
66. Kurotaki T, Tamura Y, Ueda G, Oura J, Kutomi G, Hirohashi Y, et al. Efficient cross-presentation by heat shock protein 90-peptide complex-loaded dendritic cells via an endosomal pathway. J Immunol. 2007; 179:1803–1813.
Article
67. Basu S, Binder RJ, Suto R, Anderson KM, Srivastava PK. Necrotic but not apoptotic cell death releases heat shock proteins, which deliver a partial maturation signal to dendritic cells and activate the NF-kappa B pathway. Int Immunol. 2000; 12:1539–1546.
Article
68. Somersan S, Larsson M, Fonteneau JF, Basu S, Srivastava P, Bhardwaj N. Primary tumor tissue lysates are enriched in heat shock proteins and induce the maturation of human dendritic cells. J Immunol. 2001; 167:4844–4852.
Article
69. Takakura Y, Takemoto S, Nishikawa M. Hsp-based tumor vaccines: state-of-the-art and future directions. Curr Opin Mol Ther. 2007; 9:385–391.
70. Feng H, Zeng Y, Whitesell L, Katsanis E. Stressed apoptotic tumor cells express heat shock proteins and elicit tumor-specific immunity. Blood. 2001; 97:3505–3512.
Article
71. Hashemi SM, Hassan ZM, Soudi S, Ghazanfari T, Kheirandish M, Shahabi S. Evaluation of anti-tumor effects of tumor cell lysate enriched by HSP-70 against fibrosarcoma tumor in BALB/c mice. Int Immunopharmacol. 2007; 7:920–927.
Article
72. Urban RG, Chicz RM, Lane WS, Strominger JL, Rehm A, Kenter MJ, et al. A subset of HLA-B27 molecules contains peptides much longer than nonamers. Proc Natl Acad Sci U S A. 1994; 91:1534–1538.
Article
73. Stryhn A, Pedersen LO, Holm A, Buus S. Longer peptide can be accommodated in the MHC class I binding site by a protrusion mechanism. Eur J Immunol. 2000; 30:3089–3099.
Article
74. Weinschenk T, Gouttefangeas C, Schirle M, Obermayr F, Walter S, Schoor O, et al. Integrated functional genomics approach for the design of patient-individual antitumor vaccines. Cancer Res. 2002; 62:5818–5827.
75. Hickman HD, Luis AD, Buchli R, Few SR, Sathiamurthy M, VanGundy RS, et al. Toward a definition of self: proteomic evaluation of the class I peptide repertoire. J Immunol. 2004; 172:2944–2952.
Article
76. Alldinger I, Dittert D, Peiper M, Fusco A, Chiappetta G, Staub E, et al. Gene expression analysis of pancreatic cell lines reveals genes overexpressed in pancreatic cancer. Pancreatology. 2005; 5:370–379.
Article
77. Grønborg M, Kristiansen TZ, Iwahori A, Chang R, Reddy R, Sato N, et al. Biomarker discovery from pancreatic cancer secretome using a differential proteomic approach. Mol Cell Proteomics. 2006; 5:157–171.
Article
78. Uozumi N, Gao C, Yoshioka T, Nakano M, Moriwaki K, Nakagawa T, et al. Identification of a novel type of CA19-9 carrier in human bile and sera of cancer patients: an implication of the involvement in nonsecretory exocytosis. J Proteome Res. 2010; 9:6345–6353.
Article
79. Storr SJ, Zaitoun AM, Arora A, Durrant LG, Lobo DN, Madhusudan S, et al. Calpain system protein expression in carcinomas of the pancreas, bile duct and ampulla. BMC Cancer. 2012; 12:511.
Article
80. Ohnami S, Matsumoto N, Nakano M, Aoki K, Nagasaki K, Sugimura T, et al. Identification of genes showing differential expression in antisense K-ras-transduced pancreatic cancer cells with suppressed tumorigenicity. Cancer Res. 1999; 59:5565–5571.
Full Text Links
  • YMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr