Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Johannes W. Pedersen is active.

Publication


Featured researches published by Johannes W. Pedersen.


Cancer Research | 2010

Cancer Biomarkers Defined by Autoantibody Signatures to Aberrant O-Glycopeptide Epitopes

Hans H. Wandall; Ola Blixt; Mads Agervig Tarp; Johannes W. Pedersen; Eric P. Bennett; Ulla Mandel; Govind Ragupathi; Phil O. Livingston; Michael A. Hollingsworth; Joyce Taylor-Papadimitriou; Joy Burchell; Henrik Clausen

Autoantibodies to cancer antigens hold promise as biomarkers for early detection of cancer. Proteins that are aberrantly processed in cancer cells are likely to present autoantibody targets. The extracellular mucin MUC1 is overexpressed and aberrantly glycosylated in many cancers; thus, we evaluated whether autoantibodies generated to aberrant O-glycoforms of MUC1 might serve as sensitive diagnostic biomarkers for cancer. Using an antibody-based glycoprofiling ELISA assay, we documented that aberrant truncated glycoforms were not detected in sera of cancer patients. An O-glycopeptide microarray was developed that detected IgG antibodies to aberrant O-glycopeptide epitopes in patients vaccinated with a keyhole limpet hemocyanin-conjugated truncated MUC1 peptide. We detected cancer-associated IgG autoantibodies in sera from breast, ovarian, and prostate cancer patients against different aberrent O-glycopeptide epitopes derived from MUC1. These autoantibodies represent a previously unaddressed source of sensitive biomarkers for early detection of cancer. The methods we have developed for chemoenzymatic synthesis of O-glycopeptides on microarrays may allow for broader mining of the entire cancer O-glycopeptidome.


International Journal of Cancer | 2011

Seromic profiling of colorectal cancer patients with novel glycopeptide microarray

Johannes W. Pedersen; Ola Blixt; Eric P. Bennett; Mads Agervig Tarp; Imran Dar; Ulla Mandel; Steen Seier Poulsen; Anders Elm Pedersen; Susanne Rasmussen; Per Jess; Henrik Clausen; Hans H. Wandall

Cancer‐associated autoantibodies hold promise as sensitive biomarkers for early detection of cancer. Aberrant post‐translational variants of proteins are likely to induce autoantibodies, and changes in O‐linked glycosylation represent one of the most important cancer‐associated post‐translational modifications (PTMs). Short aberrant O‐glycans on proteins may introduce novel glycopeptide epitopes that can elicit autoantibodies because of lack of tolerance. Technical barriers, however, have hampered detection of such glycopeptide‐specific autoantibodies. Here, we have constructed an expanded glycopeptide array displaying a comprehensive library of glycopeptides and glycoproteins derived from a panel of human mucins (MUC1, MUC2, MUC4, MUC5AC, MUC6 and MUC7) known to have altered glycosylation and expression in cancer. Seromic profiling of patients with colorectal cancer identified cancer‐associated autoantibodies to a set of aberrant glycopeptides derived from MUC1 and MUC4. The cumulative sensitivity of the array analysis was 79% with a specificity of 92%. The most prevalent of the identified autoantibody targets were validated as authentic cancer immunogens by showing expression of the epitopes in cancer using novel monoclonal antibodies. Our study provides evidence for the value of glycopeptides and other PTM‐peptide arrays in diagnostic measures.


Clinical Cancer Research | 2011

Serum Galectin-2, -4, and -8 Are Greatly Increased in Colon and Breast Cancer Patients and Promote Cancer Cell Adhesion to Blood Vascular Endothelium

Hannah Barrow; Xiu-Li Guo; Hans H. Wandall; Johannes W. Pedersen; Bo Fu; Qicheng Zhao; Chen Chen; Jonathan Rhodes; Lu-Gang Yu

Purpose: Adhesion of disseminating tumor cells to the blood vascular endothelium is a pivotal step in metastasis. Previous investigations have shown that galectin-3 concentrations are increased in the bloodstream of patients with cancer and that galectin-3 promotes adhesion of disseminating tumor cells to vascular endothelium in vitro and experimental metastasis in vivo. This study determined the levels of galectin-1, -2, -3, -4, -8, and -9 in the sera of healthy people and patients with colon and breast cancer and assessed the influence of these galectins on cancer-endothelium adhesion. Experimental Design: Serum galectins and auto–anti-MUC1 antibodies were assessed using ELISA and mucin protein (MUC1) glycan microarrays, and cancer-endothelium adhesion was determined using monolayers of human microvascular lung endothelial cells. Results: The levels of serum galectin-2, -3, -4, and -8 were significantly increased up to 31-fold in patients with cancer and, in particular, those with metastases. As previously shown for galectin-3, the presence of these galectins enhances cancer-endothelium adhesion by interaction with the Thomsen-Friedenreich (TF; Galβ1,3GalNAcα-) disaccharide on cancer-associated MUC1. This causes MUC1 cell surface polarization, thus exposing underlying adhesion molecules that promote cancer-endothelium adhesion. Elevated circulating galectin-2 levels were associated with increased mortality in patients with colorectal cancer, but this association was suppressed when anti-MUC1 antibodies with specificity for the TF epitope of MUC1 were also present in the circulation. Conclusions: Increased circulation of several members of the galectin family is common in patients with cancer and these may, like circulating galectin-3, also be involved in metastasis promotion. Clin Cancer Res; 17(22); 7035–46. ©2011 AACR.


British Journal of Cancer | 2013

Early detection of cancer in the general population: a blinded case-control study of p53 autoantibodies in colorectal cancer.

Johannes W. Pedersen; A Gentry-Maharaj; Evangelia-Ourania Fourkala; Anne Dawnay; Matthew Burnell; Alexey Zaikin; Anders Elm Pedersen; Ian Jacobs; Usha Menon; Hans H. Wandall

Background:Recent reports from cancer screening trials in high-risk populations suggest that autoantibodies can be detected before clinical diagnosis. However, there is minimal data on the role of autoantibody signatures in cancer screening in the general population.Methods:Informative p53 peptides were identified in sera from patients with colorectal cancer using an autoantibody microarray with 15-mer overlapping peptides covering the complete p53 sequence. The selected peptides were evaluated in a blinded case–control study using stored serum from the multimodal arm of the United Kingdom Collaborative Trial of Ovarian Cancer Screening where women gave annual blood samples. Cases were postmenopausal women who developed colorectal cancer following recruitment, with 2 or more serum samples preceding diagnosis. Controls were age-matched women with no history of cancer.Results:The 50 640 women randomised to the multimodal group were followed up for a median of 6.8 (inter-quartile range 5.9–8.4) years. Colorectal cancer notification was received in 101 women with serial samples of whom 97 (297 samples) had given consent for secondary studies. They were matched 1 : 1 with 97 controls (296 serial samples). The four most informative peptides identified 25.8% of colorectal cancer patients with a specificity of 95%. The median lead time was 1.4 (range 0.12–3.8) years before clinical diagnosis.Conclusion:Our findings suggest that in the general population, autoantibody signatures are detectable during preclinical disease and may be of value in cancer screening. In colorectal cancer screening in particular, where the current need is to improve compliance, it suggests that p53 autoantibodies may contribute towards risk stratification.


British Journal of Cancer | 2013

Autoantibodies to MUC1 glycopeptides cannot be used as a screening assay for early detection of breast, ovarian, lung or pancreatic cancer.

Brian Burford; A Gentry-Maharaj; Rosalind Graham; Diane S. Allen; Johannes W. Pedersen; A S Nudelman; Ola Blixt; Evangelia-Ourania Fourkala; D Bueti; Anne Dawnay; Jeremy Ford; R Desai; Leonor David; P Trinder; Bruce Acres; T Schwientek; Alexander Gammerman; Celso A. Reis; Luis F. Santos Silva; Hugo Osório; Rachel Hallett; Hans H. Wandall; Ulla Mandel; Michael A. Hollingsworth; Ian Jacobs; Ian S. Fentiman; Henrik Clausen; Joyce Taylor-Papadimitriou; Usha Menon; Joy Burchell

Background:Autoantibodies have been detected in sera before diagnosis of cancer leading to interest in their potential as screening/early detection biomarkers. As we have found autoantibodies to MUC1 glycopeptides to be elevated in early-stage breast cancer patients, in this study we analysed these autoantibodies in large population cohorts of sera taken before cancer diagnosis.Methods:Serum samples from women who subsequently developed breast cancer, and aged-matched controls, were identified from UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS) and Guernsey serum banks to formed discovery and validation sets. These were screened on a microarray platform of 60mer MUC1 glycopeptides and recombinant MUC1 containing 16 tandem repeats. Additional case–control sets comprised of women who subsequently developed ovarian, pancreatic and lung cancer were also screened on the arrays.Results:In the discovery (273 cases, 273 controls) and the two validation sets (UKCTOCS 426 cases, 426 controls; Guernsey 303 cases and 606 controls), no differences were found in autoantibody reactivity to MUC1 tandem repeat peptide or glycoforms between cases and controls. Furthermore, no differences were observed between ovarian, pancreatic and lung cancer cases and controls.Conclusion:This robust, validated study shows autoantibodies to MUC1 peptide or glycopeptides cannot be used for breast, ovarian, lung or pancreatic cancer screening. This has significant implications for research on the use of MUC1 in cancer detection.


Journal of Biological Chemistry | 2011

Lectin domains of polypeptide GalNAc-transferases exhibit glycopeptide binding specificity

Johannes W. Pedersen; Eric P. Bennett; Katrine T. Schjoldager; Morten Meldal; Andreas P. Holmér; Ola Blixt; Emiliano Cló; Steven B. Levery; Henrik Clausen; Hans H. Wandall

UDP-GalNAc:polypeptide α-N-acetylgalactosaminyltransferases (GalNAc-Ts) constitute a family of up to 20 transferases that initiate mucin-type O-glycosylation. The transferases are structurally composed of catalytic and lectin domains. Two modes have been identified for the selection of glycosylation sites by GalNAc-Ts: confined sequence recognition by the catalytic domain alone, and concerted recognition of acceptor sites and adjacent GalNAc-glycosylated sites by the catalytic and lectin domains, respectively. Thus far, only the catalytic domain has been shown to have peptide sequence specificity, whereas the primary function of the lectin domain is to increase affinity to previously glycosylated substrates. Whether the lectin domain also has peptide sequence selectivity has remained unclear. Using a glycopeptide array with a library of synthetic and recombinant glycopeptides based on sequences of mucins MUC1, MUC2, MUC4, MUC5AC, MUC6, and MUC7 as well as a random glycopeptide bead library, we examined the binding properties of four different lectin domains. The lectin domains of GalNAc-T1, -T2, -T3, and -T4 bound different subsets of small glycopeptides. These results indicate an additional level of complexity in the initiation step of O-glycosylation by GalNAc-Ts.


International Journal of Cancer | 2014

Cancer‐associated autoantibodies to MUC1 and MUC4—A blinded case–control study of colorectal cancer in UK collaborative trial of ovarian cancer screening

Johannes W. Pedersen; Aleksandra Gentry-Maharaj; Alexander Nøstdal; Evangelia-Ourania Fourkala; Anne Dawnay; Matthew Burnell; Alexey Zaikin; Joy Burchell; Joyce Taylor Papadimitriou; Henrik Clausen; Ian Jacobs; Usha Menon; Hans H. Wandall

Recent reports suggest that autoantibodies directed to aberrantly glycosylated mucins, in particular MUC1 and MUC4, are found in patients with colorectal cancer. There is, however, limited information on the autoantibody levels before clinical diagnosis, and their utility in cancer screening in the general population. In our study, we have generated O‐glycosylated synthetic MUC1 and MUC4 peptides in vitro, to mimic cancer‐associated glycoforms, and displayed these on microarrays. The assays performance was tested through an initial screening of serum samples taken from patients at the time of colorectal cancer diagnosis and healthy controls. Subsequently, the selected biomarkers were evaluated in a blinded nested case–control study using stored serum samples from among the 50,640 women randomized to the multimodal arm of the UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS), where women gave annual blood samples for several years. Cases were 97 postmenopausal women who developed colorectal cancer after recruitment and were age‐matched to 97 women without any history of cancer. MUC1‐STn and MUC1‐Core3 IgG autoantibodies identified cases with 8.2 and 13.4% sensitivity, respectively, at 95% specificity. IgA to MUC4 glycoforms were unable to discriminate between cases and controls in the UKCTOCS sera. Additional analysis was undertaken by combining the data of MUC1‐STn and MUC1‐Core3 with previously generated data on autoantibodies to p53 peptides, which increased the sensitivity to 32.0% at 95% specificity. These findings suggest that a combination of antibody signatures may have a role as part of a biomarker panel for the early detection of colorectal cancer.


Labmedicine | 2011

Autoantibodies as Biomarkers in Cancer

Johannes W. Pedersen; Hans H. Wandall

Circulating autoantibodies produced by the patient’s own immune system after exposure to cancer proteins are emerging as promising biomarkers for the early detection of cancer. An advantage of autoantibodies in cancer detection is their production in large quantities, despite the presence of a relatively small amount of the corresponding antigen. Autoantibodies are also expected to have persistent concentrations and long half-lives due to limited proteolysis and clearance from the circulation. Here, we review current methods for the broad screening of cancer-specific autoantibody targets and the use of such targets to develop clinically relevant assays for the detection of cancer.


Glycoconjugate Journal | 2010

Characterization of an immunodominant cancer-specific O-glycopeptide epitope in murine podoplanin (OTS8)

Catharina Steentoft; Katrine T. Schjoldager; Emiliano Cló; Ulla Mandel; Steven B. Levery; Johannes W. Pedersen; Knud J. Jensen; Ola Blixt; Henrik Clausen

Auto-antibodies induced by cancer represent promising sensitive biomarkers and probes to identify immunotherapeutic targets without immunological tolerance. Surprisingly few epitopes for such auto-antibodies have been identified to date. Recently, a cancer-specific syngeneic murine monoclonal antibody 237, developed to a spontaneous murine fibrosarcoma, was shown to be directed to murine podoplanin (OTS8) with truncated Tn O-glycans. Our understanding of such cancer-specific auto-antibodies to truncated glycoforms of glycoproteins is limited. Here we have investigated immunogenicity of a chemoenzymatically produced Tn-glycopeptide derived from the putative murine podoplanin O-glycopeptide epitope. We found that the Tn O-glycopeptide was highly immunogenic in mice and produced a Tn-glycoform specific response with no reactivity against unglycosylated peptides or the O-glycopeptide with extended O-glycan (STn and T glycoforms). The immunodominant epitope was strictly dependent on the peptide sequence, required Tn at a specific single Thr residue (Thr77), and antibodies to the epitope were not found in naive mice. We further tested a Tn O-glycopeptide library derived from human podoplanin by microarray analysis and demonstrated that the epitope was not conserved in man. We also tested human cancer sera for potential auto-antibodies to similar epitopes, but did not detect such antibodies to the Tn-library of podoplanin. The reagents and methods developed will be valuable for further studies of the nature and timing of induction of auto-antibodies to distinct O-glycopeptide epitopes induced by cancer. The results demonstrate that truncated O-glycopeptides constitute highly distinct antibody epitopes with great potential as targets for biomarkers and immunotherapeutics.


Journal of Immunotherapy | 2011

Wildtype p53-specific antibody and T-cell responses in cancer patients.

Anders Elm Pedersen; Anette Stryhn; Sune Justesen; Mikkel Harndahl; Susanne Rasmussen; Frede Donskov; Mogens H. Claesson; Johannes W. Pedersen; Hans H. Wandall; Inge Marie Svane; Søren Buus

Mutation in the p53 gene based on single amino acid substitutions is a frequent event in human cancer. Accumulated mutant p53 protein is released to antigen presenting cells of the immune system and anti-p53 immune responses even against wt p53 is induced and observed in a number of human cancer patients. Detection of antibodies against wt p53 protein has been used as a diagnostic and prognostic marker and discovery of new T-cell epitopes has enabled design of cancer vaccination protocols with promising results. Here, we identified wt p53-specific antibodies in various cancer patients and identified a broad range of responses against wt p53 protein and 15-mer peptides using a novel print array technology. Likewise, using bioinformatic tools in silico, we identified CD8+ T-cell specificity or reactivity against HLA-A*02:01 binding peptides wt p53(65–73), wt p53(187–197), and wt p53(264–272) in breast cancer patients and against HLA-A*01:01 binding peptide wt p53(226–234) and HLA-B*07:02 binding peptide wt p53(74–82) in renal cell cancer and breast cancer patients, respectively. Finally, we analyzed antibody and T-cell responses against wt p53 15-mer peptides in patients with metastatic renal cell carcinoma who were alive with no evidence of disease after a follow-up period of minimum 5 years after treatment with IL-2±IFN-&agr;±histamine containing immunotherapy to identify novel epitopes for use in immunotherapy and for potential response biomarkers. However, none of the wt p53 reactivity observed justified use of 15-mer or was related to survival in this rare patient population.

Collaboration


Dive into the Johannes W. Pedersen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Henrik Clausen

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar

Ulla Mandel

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar

Ola Blixt

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anne Dawnay

University College London

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge