Next Article in Journal
Submicron Topographically Patterned 3D Substrates Enhance Directional Axon Outgrowth of Dorsal Root Ganglia Cultured Ex Vivo
Next Article in Special Issue
Lipotoxicity as a Barrier for T Cell-Based Therapies
Previous Article in Journal
Electrospun Core–Sheath Nanofibers with Variable Shell Thickness for Modifying Curcumin Release to Achieve a Better Antibacterial Performance
Previous Article in Special Issue
A New Serum Macrophage Checkpoint Biomarker for Innate Immunotherapy: Soluble Signal-Regulatory Protein Alpha (sSIRPα)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Identification of the Antigens Recognised by Colorectal Cancer Patients Using Sera from Patients Who Exhibit a Crohn’s-like Lymphoid Reaction

by
Viktoriya B. Boncheva
1,†,
Michael Linnebacher
2,
Said Kdimati
2,
Hannah Draper
3,
Laurence Orchard
4,
Ken I. Mills
5,
Gerald O’Sullivan
6,‡,
Mark Tangney
6,7,8 and
Barbara-ann Guinn
9,*
1
Department of Life Sciences, University of Bedfordshire, Park Square, Luton LU1 3JU, UK
2
Department of General Surgery, Molecular Oncology and Immunotherapy, University Medical Center Rostock, Schillingallee 69, 18057 Rostock, Germany
3
Hull York Medical School, University of Hull, Cottingham Road, Kingston-upon-Hull HU6 7RX, UK
4
Cancer Sciences, Southampton University Hospitals Trust, University of Southampton, Southampton SO16 6YD, UK
5
Patrick G. Johnson Centre for Cancer Research, Queen’s University Belfast, Lisburn Road, Belfast BT9 7AE, UK
6
Cancer Research@UCC, Cork, Ireland
7
SynBioCentre, University College Cork, Cork, Ireland
8
APC Microbiome Ireland, University College Cork, Cork, Ireland
9
Department of Biomedical Sciences, University of Hull, Cottingham Road, Kingston-upon-Hull HU6 7RX, UK
*
Author to whom correspondence should be addressed.
Current address: NHS Greater Glasgow and Clyde, Glasgow G12 0XH, UK.
Deceased.
Biomolecules 2022, 12(8), 1058; https://doi.org/10.3390/biom12081058
Submission received: 18 June 2022 / Revised: 21 July 2022 / Accepted: 25 July 2022 / Published: 29 July 2022
(This article belongs to the Special Issue Immunotherapy and Cancer)

Abstract

:
A Crohn’s-like lymphoid reaction (CLR) is observed in about 15% of colorectal cancer (CRC) patients and is associated with favourable outcomes. To identify the immune targets recognised by CRC CLR patient sera, we immunoscreened a testes cDNA library with sera from three patients. Immunoscreening of the 18 antigens identified by SEREX with sera from normal donors showed that only the heavy chain of IgG3 (IGHG3) and a novel antigen we named UOB-COL-7, were solely recognised by sera from CRC CLR patients. ELISA showed an elevation in IgG3 levels in patients with CRC (p = 0.01). To extend our studies we analysed the expression of our SEREX-identified antigens using the RNA-sequencing dataset (GSE5206). We found that the transcript levels of multiple IGHG probesets were highly significant (p < 0.001) in their association with clinical features of CRC while above median levels of DAPK1 (p = 0.005) and below median levels of GTF2H5 (p = 0.004) and SH3RF2 (p = 0.02) were associated with improved overall survival. Our findings demonstrate the potential of SEREX-identified CRC CLR antigens to act as biomarkers for CRC and provide a rationale for their further characterization and validation.

Graphical Abstract

1. Introduction

Colorectal cancer (CRC) is one of the most commonly diagnosed cancers worldwide. It affects the bowel and rectum, is rare in people under 40, with almost 85 per cent of cases being diagnosed in persons over 65 years of age. One in every twenty people in the UK develops CRC, with only half of them surviving beyond five years, mainly because CRC is often detected once well-established and after it has spread beyond the bowel. The disease stage at the time of diagnosis governs both the choice of treatment and the prognosis. The most commonly used staging system for CRC is the American Joint Committee on Cancer (AJCC), also known as the tumour, node, metastases (TNM) system [1], which reflects how far the cancer has spread, and whether it has reached nearby structures such as lymph nodes and/or distant organs.
Stage I CRC is an asymptomatic malignancy, developing slowly through the progressive accumulation of genetic mutations within precancerous bowel lesions and polyps. Diagnosis at this stage reduces the risk of death from CRC, giving a 90% chance of survival beyond five years [2] and a significantly decreased frequency of disease recurrence. However, most cases of CRC are detected once the cancerous cells have moved beyond the middle layers of the colon (Stage II). Treatment generally involves surgery, chemotherapy and/or radiotherapy, with little in the way of immunotherapy except for the approximately 15% of CRCs that show mismatch repair deficiency or microsatellite instability (MSI). In these patients, there have been promising results with immune checkpoint inhibitor (ICI) treatment, utilising pembrolizumab and nivolumab to block programmed cell death 1, resulting in improved survival, even in metastatic CRC (reviewed in [3]). However, the vast majority of CRC patients have been shown to be largely unresponsive to ICI treatment. Current research focusses on how to improve the immune response to ICIs in patients that are microsatellite stable/DNA mismatch repair proficient (reviewed in [4]). Currently, groups are examining ways to make microsatellite stable CRC an “immune hot” tumour through the examination of components of the immune system as well as studies of the potential of bispecific antibodies, cellular therapies, vaccines and cytokines to stimulate an anti-tumour immune response.
The Crohn’s-like reaction (CLR) is a CRC specific ectopic lymphoid reaction characterised by peritumoural aggregates at the advancing edge of the tumour (reviewed in [5]). Early in CLR development CD4+ T cells cluster mostly with mature dendritic cells but as the CLR matures, B cell numbers increase and follicular dendritic cells are recruited and create lymphoid follicles. CLR CRC was first described in 1990 [6] and is associated with improved survival among CRC patients. It should be noted that CLR has no biological relationship to Crohn’s disease and is in fact a CRC-specific ectopic or tertiary lymphoid reaction, with similar structures found in other non-CRC tumours. CRC tumours deficient in the DNA mismatch repair system have MSI and a high mutational burden associated with the presence and density of CLR but it is solely the subgroup of CRC known to have CLRs that we have focused on in this study.
With the aim of trying to find new immune targets for CRC therapy, we wanted to determine which antigens were being recognised by sera from patients with CLR-CRC, who showed an improved overall survival and were able to clear post-operative micrometastasis [7]. We focused on this group in the hope of identifying antigens that could be used as targets for CRC immunotherapy in the future.

2. Materials and Methods

2.1. Patient and Healthy Volunteer (HV) Serum Samples

Diagnostic and clinical investigations for staging all CRC patients who attended the Mercy Hospital Cork were performed in accordance with the guidelines of the human ethics committee for clinical research, National University of Ireland, Cork. Histological examination showed tumour extension beyond the muscularis propria and absence of nodal metastases. Three Dukes’ B non-synchronous CRC patients who had no concurrent inflammatory bowel disease or familial polyposis coli had been treated surgically with radical resection of the primary tumour including mesentary of the bowel in a 3-month period during 2007 and were followed up for micrometastases in a dedicated clinic for a mean period of 3 years post-surgery. CC005 (male, aged 67), CC010 (male, aged 77) and CC014 (female, aged 88) each met the criteria described by Murphy et al. 2000 [7] as having Jass and “Crohn’- like” lymphoid reactions during follow-up appointments. The other 12 Dukes’ B/stage II CRC samples collected during this same period in 2007 did not meet the CLR criteria and, thus, were not used for immunoscreening in our study but were used in ELISAs.
HV blood samples were obtained from The National Blood Service, North London Blood Transfusion Centre and used for immunoscreening. Serum was collected from clotted peripheral blood samples following centrifugation at 1200× g rpm for 10 min, aliquoted and frozen at −80 °C until required. The testes library was prepared from cDNAs from five HVs aged between 19 and 65 (Stratagene Europe, Amsterdam, The Netherlands) [8].
The clinical and pathological characteristics of the CRC samples used to examine IgG3 levels by immunohistochemistry (IHC) are summarised in Table S1. The process of establishing patient-derived cell lines has been reported previously [9], and primary cell lines, directly established from human fresh tumour tissues, are indicated with the prefix HROC and the ID number of the patient, e.g., HROC60. All patients signed the written informed consent and the procedures were approved by the Ethics Committee of the University Hospital of Rostock (reference number II HV 43/2004 and A 45/2007) in accordance with the declaration of Helsinki.

2.2. Immunoscreening of the Testes cDNA Library and Identification of Positive Clones

Serological analysis of recombinant cDNA expression libraries (SEREX) was performed as described previously [10] with minor modifications [11]. Briefly, the testes cDNA library was prepared following the manufacturer’s instructions [8] from five normal donors aged between 19 and 65 (Stratagene Europe, Amsterdam, The Netherlands). Phage containing cDNA inserts were plated at 104 pfu per 132 mm Petri dish onto a lawn of XL1 Blue MRF’ E. coli. Sera was cleared using E. coli XL1 Blue MRF’ lysates, with and without phage lacking a cDNA insert, either bound to CNBr activated sepharose 4B beads (Amersham Pharmacia Biotech, Buckinghamshire, UK) or 132 mm 0.2 µm circular nitrocellulose membranes (PALL Corporation, New York, NY, USA). The reactivity of serum was confirmed through agglutination tests [12] using 30 µL sheep and/or pigs blood and 15 µL CRC sera diluted 1:10 in phosphate-buffered saline. Pre-cleared CRC sera were diluted in TBS/0.05% sodium azide and used for primary and secondary immunoscreening (the latter to confirm sero-positivity). All positive plaques were isolated to monoclonality, eluted, in vivo excised (following Stratagenes ‘ZAP Express cDNA Synthesis kit’ manufacturers’ guidelines, Cat #200451) and plated as phagemids in E. coli on selective media plates. Single colonies were expanded by overnight culture and plasmid DNA was isolated using a Qiagen plasmid mini kit (Qiagen Ltd., Sussex, UK, Cat #12123).

2.3. Sequence Analysis of cDNA Inserts

T3 and T7 primers (5′-GCAATTAACCCTCACTAAAGG-3′ and 5′-TAATACGACTCACTATAGGG-3′, respectively) bind regions that flank the multiple cloning site. They were used to amplify the cDNA inserts in the pBK-CMV phagemid of immunoresponsive clones with ReadyMix Taq PCR Reaction Mix with MgCl2 (Sigma Aldrich, UK; Cat #P4600). PCR products were gel purified using a QIAQuick gel extraction kit (Qiagen Ltd., Manchester, YORK, UK; Cat #28704) and sent to the Department of Biochemistry at the University of Cambridge [13] for Sanger sequencing. Nucleotide sequences generated by Applied Biosystems’ Sequencing Analysis Software and predicted amino acid sequences were compared with known sequences in the gene, expressed sequence tag (EST) and protein databases, including the National Centre for Biotechnology Information (NCBI) BLAST, Baylor College of Medicine (BCM) Search launcher, the U.K. human genome mapping project (hgmp) resource centre and The Institute for Genomic Research (TIGR) web-based facilities.

2.4. Immunoscreening of Testes Responsive Clones with Sera from Immune-Responsive CRC Patients and HVs

Every unique antigen (named UOB-COL-1-18) was immunoscreened with sera from each CRC patient with CLR (CC005, CC010 and CC014), while UOB-COL-1 to -15 were also immunoscreened with eight age- and sex-matched HV sera.

2.5. Microarray Analysis

Utilising existing microarray data, chosen by virtue of their sample size, availability of clinical details and suitability of the controls, we compared gene transcript levels to clinical features, as well as above and below median expression and its relationship to overall survival. The GSE5206 dataset [14] was generated from the analyses of 100 human CRC samples encompassing all clinical stages [15] and five normal colon tissues. We examined the levels of expression of each antigen in CRC patients versus normal donors and any correlation between levels of expression and clinical features. We additionally analysed IGHG2 and IGHG3 gene expression using The Cancer Genome Atlas (TCGA) Colon Adenomarcinoma (COAD) dataset [16] which assesses 571 CRC patients by RNA-sequencing. This dataset is publicly available through xenabrowser.net and the dataset was chosen because it is one of the largest and most annotated datasets available.

2.6. IHC

Immunolabelling of IgG3 was performed using 3.5 μg/mL monoclonal rabbit anti-human IgG3 (clone RM119, Sigma-Aldrich, Cat# SAB5600212, St. Louis, MO, USA) with a human tonsil as a positive control. Clone RM119 reacts to the heavy chain of IgG3 and not to any other IgG subclasses (IgG1, IgG2 or IgG4), nor does it show any cross reactivity with IgM, IgA, IgD or IgE. IgG antibody was also used as a control for the presence of other IgG subclasses. Immunolabelling was detected using the EnVision®+ Dual link system (DAB+), HRP-labelled anti-rabbit polymere (Agilent Technologies Deutschland GmbH, Waldbronn, Germany; Cat#K4063), following the manufacturer’s instructions. Blocking and dilution buffers contained 2% bovine serum albumin (Sigma-Aldrich, Cat#A9418).

2.7. ELISA

We detected IgG3 levels in sera from 12 CRC patients (CC001-CC012) and 12 normal donors using a pre-coated solid-phase sandwich ELISA kit IgG3 (Invitrogen Cat#BMS2094, Waltham, MA, USA) and following the manufacturer’s instructions.

3. Results

3.1. Eighteen Antigens Were Recognised by CLR-CRC Patient Sera

Testes have many features similar to cancer, including rapid proliferation and global promoter hypomethylations, offering an opportunity to identify cancer-testis antigens (recently reviewed in [17]) and immunoscreen a broad range of proteins [18]. Primary immunoscreening was performed following the SEREX technique on 821,644 plaque forming units (pfu) using either CC005 (1:2000 and 1:200 dilutions), CC010 (1:200 dilution) or CC014 (1:200 dilution) serum. Each of these patients had been identified as meeting the criteria for CLR-CRC as described by Murphy et al. 2000 [7]. A total of154 phages were identified as potentially sero-positive and secondary immunoscreening confirmed 32 phages harboured immune reactive cDNA inserts using the same sera that they were identified with.
Following amplification of cDNA inserts in the pBK-CMV vector using T3-T7 primers, products were isolated from 1% agarose gel using QIAquick gel extraction and Nanodrop analysis performed to quantify the cDNA. Agarose gel electrophoresis demonstrated that a range of cDNA insert sizes had been sero-recognised by CRC CLR sera in the testes cDNA library (data not shown), indicating the cDNA library were representative of a breadth of cDNA transcripts (as shown in [8]).
In total, 32 sero-responsive clones that encoded 18 independent antigens (Table 1) were named UOB-COL-1 to UOB-COL-18 following SEREX convention [19]. The nine individual cDNA fragments that were mapped to corresponding genes did so with maximum identity values ranging between 90% and 99%. The differences between the query sequence (UOB-COL cDNA insert) and the subject were single nucleotides and most likely due to errors in the sequencing process. FASTA sequences are already available for these sequences [8].

3.2. Two Proteins Were Recognised by CLR-CRC but Not HV Patient Sera

To determine which antigens were solely recognised by CLR-CRC sera, we immunoscreened each unique cDNA with sera from each CLR-CRC patient and eight HVs. Fifteen of the eighteen immunoresponsive sequences were identified and confirmed with serum CC014, nine with CC010 and ten with serum CC005 (Table 2). None of the HV sera reacted against UOB-COL-1, an antigen found to be strongly responsive with all three of the sera from patients with CLR-CRC, or UOB-COL-7. UOB-COL-1 encoded the immunoglobulin heavy constant gamma 3 (G3m Marker) (IGHG3), while the sequence of UOB-COL-7 did not map onto a known gene. Each HV sera reacted with at least six of the antigens and in many cases reacted with up to 12 of the UOB-COL antigens.

3.3. Confirmation of Ig Heavy Chain Recognition by Patient Sera

To check the validity of the identification of the IGHG3 and IGHG2 proteins (identified as UOB-COL-1 and 2, respectively, in our study), we immunoscreened part of the testes cDNA library with anti-IgG (secondary) antibody only. No positives were found following the immunoscreening of more than 300 pfu and the same antigens had not been identified during previous immunoscreening studies by our group [20]. This suggests that the heavy chain fragments (IGHG2 and IGHG3) were not identified as a product of contamination but due to recognition of IgG heavy chains by the responsive CRC patient sera. In addition, UOB-COL-1 was not sero-recognised by eight HV sera, suggesting the result was not an artefact of the sero-screening method.

3.4. Only IGHG2 and IGHG3 Had Elevated Transcription in CRC Patient Tissue

We examined the expression of the nine known antigens identified by SEREX in this study ((IGHG2, IGHG3, ZNF465, CYB5R3, SLC34A2, RPL37A, SH3RF2, DAPK1 and GTF2H5) using the publicly available RNA-sequencing dataset (GSE5206 [14]). The only probesets with elevated levels (>2 fold or p < 0.05) in patients when compared with healthy donor tissues were those that included IGHG2 or IGHG3 (Table 3).
Using an independent dataset (TCGA-COAD) The Cancer Genome Atlas (TCGA) Colon Adenomarcinoma (COAD) dataset [16] which annotates 571 CRC patient samples by RNA-sequencing, we did not observe a unique sub-population of CRC patients with elevated IGHG3 expression in their tumours (Figure 1). However, we did find a significant association between the expression of IGHG2 and IGHG3, suggesting that the transcription of these constant regions have similar expression profiles in CRC patients (p = 8.25 × 10−145).
Table 1. BLAST results for the nine previously identified antigens identified by CRC CLR patients.
Table 1. BLAST results for the nine previously identified antigens identified by CRC CLR patients.
NameGene SymbolChromosome LocalisationGeneral Function of the Encoded Protein #SEREX
Identified Antigen
Significance in Different Types of Cancer
UOB-COL-1IGHG314q32.33Involvement in a number of molecular, biological and cell-signalling pathwaysNoPotential: Diagnostic marker in malignant mesothelioma [21]; Diagnostic and prognostic marker in prostate cancer [22]; Prognostic marker in breast cancer [23]; overexpressed in non-small cell lung carcinoma [24].
UOB-COL-2IGHG214q32.33Involvement in a number of molecular, biological and cell-signalling pathwaysNoNo known tumour-associated properties.
UOB-COL-3AZFP2q21.2Novel family of genes with unidentified general functionYesEncodes a novel transmembrane zinc finger protein with a KRAB box domain. Found to be overexpressed in a patient with acute myeloid leukaemia and detected with autologous serum, SEREX id: GKT-AML8 [25]. Also found to be overexpressed in CML patients and several cancer cell lines but not in normal donor blood cells.
UOB-COL-4CYB5R322q13.2Desaturation and elongation of fatty acids, cholesterol biosynthesis, drug metabolism, and, in erythrocyte, methemoglobin reductionNoPolymorphism associated with increased breast cancer risk [26].
UOB-COL-5SLC34A24p15.2pH-sensitive sodium-dependent phosphate transporterNoDownregulation in A549 (lung adenocarcinoma cells) promotes tumour development [27]; Overexpressed in breast cancer and could act as potential therapeutic target [28]; Its encoded protein (NaPi2b) is targeted by Rebmab200 (humanised monoclonal antibody) in cancer [29]; Potential diagnostic marker in ovarian cancer [30].
UOB-COL-11RPL37A2q35Structural constituent of ribosomeYes *Showed to predict response to neoadjuvant doxorubicin and cyclophosphamide in breast cancer patients (as part of a panel of genes) [31].
UOB-COL-16SH3RF25q32Promotes cell survival and apoptosis. Inhibits PPP1CA phosphatase activity.NoOverexpressed in human cancers and regulates PAK4 in colon cancer. Acts as an oncogene and may represent an effective therapeutic target for cancer treatment [32].
UOB-COL-17GTF2H56q25.3Functions in gene transcription and DNA repairNoLikely to be involved in carcinogenesis [33].
UOB-COL-18DAPK19q21.33Calcium/calmodulin-dependent serine/threonine kinase involved in multiple cellular signalling pathways that trigger cell survival, apoptosis, and autophagyNoDAPK promoter methylation may be involved in NSCLC carcinogenesis [34]; DAPK promoter methylation and abnormal expression of DAPK mRNA in acute leukaemia patients [35]; DAPK as potential therapeutic target [36].
# Information obtained from Genecards.og. * Identified in cell line from patient with melanoma by SEREX (Cancer Immunome Database).
Table 2. Immunoscreening of antigens with sera from each patient with CRC CLR and HVs indicated that UOB-COL-1 and UOB-COL-7 were only recognised by patient sera.
Table 2. Immunoscreening of antigens with sera from each patient with CRC CLR and HVs indicated that UOB-COL-1 and UOB-COL-7 were only recognised by patient sera.
SEREX IDRecognition by CRC-CLR Sera during ImmunoscreeningHV Sero-Screening
CC005CC010CC014Total12345678Total
UOB-COL-1+++3/30/8
UOB-COL-2++2/3++++++6/8
UOB-COL-3+1/3++++++++8/8
UOB-COL-4+++3/3+++++++7/8
UOB-COL-5++2/3++++++6/8
UOB-COL-6+++3/3++2/8
UOB-COL-7+++3/30/8
UOB-COL-8++2/3+++++++7/8
UOB-COL-9++2/3+++++++7/8
UOB-COL-10++2/3++++++++8/8
UOB-COL-11+1/3++++++++8/8
UOB-COL-12+1/3++++++6/8
UOB-COL-13+1/3++++++++8/8
UOB-COL-14+1/3++++++++8/8
UOB-COL-15++2/36/1411/1412/148/1411/1411/1412/1411/1412/14
UOB-COL-16+ND+2/2
UOB-COL-17++ND2/2
UOB-COL-18+NDND1/1
Total number of antigens recognised by reactive sera10/189/1615/16
ND: not done.
Table 3. Analysis of antigen transcription in 100 CRC patients compared with five normal donors using publicly available RNA-seq data. Nine probesets, each including IGHG2 and IGHG3, had >2-fold increased transcription of the heavy chain constant regions of IgG2 and IgG3 in tumour versus normal tissue.
Table 3. Analysis of antigen transcription in 100 CRC patients compared with five normal donors using publicly available RNA-seq data. Nine probesets, each including IGHG2 and IGHG3, had >2-fold increased transcription of the heavy chain constant regions of IgG2 and IgG3 in tumour versus normal tissue.
Probeset IDGene Symbolp-ValueFold-Change
211868_x_atIGH///IGHA1///IGHA2///IGHD///IGHG1///IGHG2///IGHG3///IGHM///IGHV4-312.13 × 10−52.69
211641_x_atIGHA1///IGHA2///IGHD///IGHG1///IGHG3///IGHM///IGHV4-310.0012.47
211637_x_atIGH///IGHA1///IGHA2///IGHD///IGHG1///IGHG3///IGHG4///IGHM///IGHV3-23///IGHV4-310.0013.09
214916_x_atIGHA1///IGHA2///IGHG1///IGHG3///IGHM///IGHV3-23///IGHV4-310.0022.62
211650_x_atIGH///IGHA1///IGHD///IGHG1///IGHG3///IGHM///IGHV3-23///IGHV4-310.0022.59
211639_x_atIGH///IGHA1///IGHA2///IGHD///IGHG1///IGHG3///IGHG4///IGHM///IGHV4-310.0062.09
217281_x_atIGH///IGHA1///IGHA2///IGHG1///IGHG2///IGHG3///IGHM///IGHV4-310.00672.49
216557_x_atIGHA1///IGHD///IGHG1///IGHG3///IGHM///IGHV4-310.0072.49
211635_x_atIGHA1///IGHA2///IGHD///IGHG1///IGHG3///IGHG4///IGHM///IGHV4-310.0182.13

3.5. IgG3 Protein Was Not Detected in CRC Primary Tumour Cell Lines

To determine whether IgG heavy chain fragments were being expressed by the tumour tissue, in the same way that other epithelial cancers including CRC have been shown to previously [37], or whether antibody recognition of IGHG3 products were due to infiltration of the adaptive immune response into the tumour as part of the CLR reaction, we examined the expression of IgG3 in cell lines and Hansestadt Rostock colorectal cancer (HROC) patient samples. The epithelial-derived SW480 cell line (CVCL_0546) from a patient with Dukes’ B [38] and the epithelial-like HCT116 [39] cell line (CVCL_0291) were used as positive controls for intracellular IgG2 and IgG3 protein production (data not shown). In addition, we examined 20 HROC samples from 18 patients that represented with adenocarcinoma of the colon from a broad range of anatomic sites, grading and staging types, and molecular classes (Table S1). We predominantly observed single IgG3 positive cells in stroma and parenchyma and weak staining in the stroma of some CRC tissues (Figure 2; Table 4). Despite two of the patients having high lymphocytic stroma reaction and CLR, neither showed high levels of intracellular IgG3 expression.

3.6. IgG3 Levels Were Elevated in CRC Patient Sera

We found there was an increase in the levels of IgG3 protein in the sera of patients with CRC when compared to healthy donors (p = 0.01; Figure 3). However, the levels of IgG3 in the CLR-CRC patients (CC005 and CC010) used for immunoscreening fell within the range of the CRC group.

3.7. IGH Transcription Was Indicative of Clinical Features of CRC

Gene expression analysis of 100 CRC samples in the GSE5206 dataset indicated associations between clinical features and expression of the genes of interest identified in this study (Table S2). Only columns/rows of data that contained a >two-fold change in gene transcript levels are shown in Table 5, Table 6, Table 7, Table 8, Table 9 and Table S3. With regard to Dukes’ stage, only three probesets, each IGH family members (211650_x_at; 214916_x_at; 216557_x_at), showed a >two-fold difference in expression, each in Dukes’ stage B down versus in situ (IS). IGH, family members were increased in 0 versus 2 and 0 versus 2B, each representing increased expression in less aggressive stages of CRC and reinforcing the idea that IGH family member transcription decreases with CRC progression, especially in AJCC stage 2 and 2B.
When considering the TNM staging of CRC, in the T stage, RPL37A was increased in 1 versus 3, 4 and X (Table 6 and Table S3B). The N stage showed no two-fold differences, while three probesets (211650_x_at, 216557_x_at, 217281_x_at), each IGH family members, had fold changes that were >two in M stage (2.05, 2.18 and 2.12, respectively) for 0 up versus X. Four probesets (211650_x_at, 216557_x_at, 217281_x_at and 211430_s_at) had fold changes >2 (2.18, 2.06, 2.12 and 2.08, respectively) in 1 up versus X in M stage.
Transcription of IGH family members demonstrated >two-fold increase in colon (ascending) versus colon (NOS), colon ascending (down) versus hepatic flexure, colon (cecum) up versus colon (NOS), colon (cecum) down versus hepatic flexure and colon descending up versus colon (NOS) (Table 7), while a number of IGH probesets were decreased in colon (ascending) down versus colon (splenic flexure), colon (cecum) down versus colon (splenic flexure), colon (descending) down versus colon (splenic flexure) and colon (NOS) down versus colon (sigmoid; splenic flexure and transverse). p-values for this data are shown in Table S3C.
With regard to recurrence type, CYB5R3 showed a >two-fold decrease in transcript levels in distance of recurrence to peritoneum or ascites versus distance from site of recurrence, behaviour NOS; versus local recurrence, behaviour NOS; versus never disease free, since diagnosis and versus regional recurrence, behaviour NOS (Table 8). p-values for this data are shown in Table S3D. IGH family members probesets were decreased by 9–12-fold in distance of recurrence to peritoneum or ascites versus distance to site of recurrence, behaviour NOS down (13 of 16 IGH probesets) and versus local recurrence (12 of 16 IGH probesets). Two-fold increases in transcription in IGH family member probesets was observed in local recurrence, behaviour NOS up versus never disease free since diagnosis; versus regional recurrence, behaviour NOS and versus regional tissue recurrence of invasive cancer.

3.8. Expression of Three CLR CRC Antigens Indicated Overall Survival in CRC Patients

Below median levels of death-associated protein kinase 1 (DAPK1) and above median levels of SH3 domain containing ring finger 2 (SH3RF2) and General transcription factor IIH subunit 5 (GTF2H5) were associated with improved overall survival in the GSE17537 dataset (n = 232 CRC patients) [40] (Figure 4). This was not the case for members of the IGH family despite their preponderant association with clinical features.

4. Discussion

Through immunoscreening, we identified 18 antigens that were recognised by sera from three CRC patients with CLR; nine were novel and have yet to be identified as transcribed genes through sequence analysis. However, eight of the known antigens (the exception being IGHG2) had been shown to play a role in various tumour types previously. UOB-COL-1, encoding the immunoglobulin heavy constant 3 (G3m marker), had been shown to be increased in inflamed colonic tissue, including ulcerative colitis and Crohn’s disease [41]. Indeed, immunoglobulins have been found to be intrinsically produced by a range of, often epithelial, cancer types [37,42,43,44], having been shown to play a role in cancer initiation, proliferation, invasion, metastasis and survival [45,46,47,48,49]. Immunoglobulins have been shown to be expressed in CRC tissue by a number of groups [42,50], while Geng et al. [51] recently reported that the immunoglobulins expressed by CRC tend to be IgG, rather than other classes of IgH, and show unique VHDJH patterns and somatic hypermutation hotspots. IGHG3 specifically has also been shown to be overexpressed in a number of cancers, again predominantly epithelial tumours, including non-squamous non-small cell lung cancer [24,48], breast cancer [23,52], prostate cancer [22], malignant mesothelioma [21] and CRC [51,53]. Recently Xu et al. [54] showed that mouse IgG could be used to treat and prevent melanoma, breast and colon cancer in syngeneic animal models, while treatment of patients with autoimmunity with anti-IgG was incidentally shown to reduce tumour growth in patients [55,56,57]. The HROC collection of low passage CRC cell lines have been shown to produce IgG previously, and the intrinsic levels are significantly higher than those found in human T cells (used as controls) [58]. However, levels of IgG were very low in the medium, suggesting that although colon cancer cells can generate IgG, unlike B cells, they do not secrete it. We found that few of the HROC tissues we tested expressed notable levels of IgG3, suggesting that the samples we had performed immunoscreening on had in fact demonstrated an infiltration of the tumour by IgG3 secreting B cells rather than intrinsically producing the heavy chain of IgG3. Although Mon et al. showed that IGHG3 expression correlates with non-small cell lung cancer patients who respond to chemotherapy [59] and all three CLR patients tested by SEREX reacted to the IGHG3 gene product, IgG3 protein levels in low passage CRC samples appeared to be very low. Indeed, IgG3 levels in the CRC patient sera were elevated compared with normal donors, again suggesting that the antibody response to IGHG3 products was to infiltrating B cells and/or antibodies rather than IGHG3 production within the tumour cells.
Of the other antigens identified using CLR sera from CRC patients, SH3RF2 was one of two proteins that has been directly linked to CRC through its activity as an oncogene. SH3RF2 has previously been shown to have elevated expression in 159 CRC tissues and to correlate with poor prognostic indicators [32]. In contrast, we found above-median levels of SH3RF2 to correlate with improved overall survival (p = 0.02). Although SH3RF2 has been shown to have high expression in colon cancer, with moderate membranous positivity, SH3RF2 is described as having low cancer specificity and is not prognostic. CYB5R3 (identified as UOB-COL-4 in our study) has been shown to be downregulated in breast cancer, negating its ability to detoxify the responsive hydroxylamine metabolites of known mammary carcinogens [60]. Blanke et al. [26] demonstrated that a particular polymorphism in the CYB5R3 gene was associated with breast cancer risk in women. Their study suggested that CYB5R3 may be directly involved in the development of other tumours associated with aromatic and heterocyclic amine exposures, such as CRC. When CYB5R3 siRNA was injected intravenously into mice they showed a decrease in lung cancer burden [26], with cellular effects including signalling alterations associated with extravasation, transforming growth factor beta (TGFβ) and hypoxia-inducible factor 1-alpha (HIFα) pathways, and apoptosis [61].
RPL37A (named UOB-COL-11 in our study) has also been found to be upregulated in high grade astrocytomas [62] and to have a general association with lifetime and overall glioblastoma survival (p-value < 0.05 in both cases) [63]. In breast cancer, RPL37A expression, in conjunction with metastases suppressor 1 (MTSS1), as well as SET and MYND domain-containing protein 2 (SMYD2), were shown to predict the response of breast cancer patients to neoadjuvant doxorubicin and cyclophosphamide [31]. We found that above-median levels of RPL37A in CRC patients significantly correlated with improved disease-free survival (p = 0.0055), supporting the belief that RPL37A may play a role in cell death, as suggested in glioblastoma [63]. SLC34A2 (named UOB-COL-5 in our study) is a sodium-dependent phosphate transporter that is overexpressed in CRC, and its expression was significantly correlated with N stage in CRC [64]. SLC34A2 can be used to stratify patient prognosis in stage II and III CRC, while high levels of SLC34A2 can be correlated with higher post-operative metastases rates and act as an independent adverse factor affecting patient prognosis. Knock-down experiments showed that the absence of SLC34A2 inhibited cell proliferation, colony formation, induced apoptosis and arrested the cell cycle, while in in vivo studies, SLC34A2 knock-down prevented xenograft growth and promoted apoptosis.
DAPK1, identified as UOB-COL-18 in this study, is known to be an important serine/threonine kinase that acts as a positive regulator of programmed cell death and is also involved in the regulation of autophagy and cell migration [36]. The DAPK promoter has been reported to be methylated in several types of cancer [34,65,66], thus disrupting the process of programmed cell death. However, Satoh et al. [65] have demonstrated that in CRC, methylation alone is not responsible for silencing DAPK and histone deacetylation is also required. Inhibition of histone deacetylation acts synergistically with the inhibition of DNA methylation to induce DAPK gene expression. Therefore, DAPK1 may be a suitable target for the treatment of CRC patients through the activation of apoptosis using histone deacetylase inhibitors, with the frequency of DAPK1 expression dictating feasibility. Models of colon cancer [67] have shown that the loss of DAPK1 expression has been shown to impact migratory capacity [68], tumour cell dissemination and increase invasiveness. We found that above-median levels of DAPK1 were associated with poorer overall survival (p = 0.005), which supports its potential as a target for CRC therapy.
Zinc finger 465 (ZNF465; named UOB-COL-3 in this study) has previously been identified through the autologous immunoscreening of a library made from a patient with acute myeloid leukaemia (AML) [25]. ZNF465 is a Krüppel-type zinc finger transmembrane protein with a 5′ Krüppel-associated box domain that is typical of negative regulators of gene transcription [69]. We found ZNF465 to be expressed in AML and chronic myeloid leukaemia but not HVs. To date, most Krüppel-type zinc finger proteins are found to be transcriptional repressor proteins involved in a variety of biological processes [69,70]. General Transcription Factor IIH, polypeptide 5 (GTF2H5), is a small protein that stabilizes the multi-subunit transcription repair factor IIH (TFIIH). TFIIH plays a key role in a nuclear excision repair in healthy tissues and was identified as one of six most significantly mutated genes in squamous cell carcinoma related to carcinogenesis [33]. Above-median levels of GTF2H5 have been associated with improved overall survival and reduced disease-free survival, with a much higher level of significance in its inverse association with disease free survival. This may be explained by its role in nuclear excision repair, whereby its greater expression leads to genetic repair and removes neo-antigens that would otherwise act as immune targets following chemotherapy and/or surgery.
The limitations of this study included the small number of sera samples from CRC CLR patients that were used for immunoscreening. Only IGHG2 and IGHG3 had elevated expression in CRC patients compared with healthy donors, as determined by RNA-seq analysis in an independent dataset, but protein levels were not determined as tissue was not available within the patient samples we had collected and intrinsic IGHG3 expression was not found in the CRC primary cell lines. There is not a linear relationship between RNA transcription and protein levels in higher eukaryotes [71] due to various biological and technical factors, including codon bias, RNA secondary structure, as well as protein abundance and turnover; however, the presence of the proteins in patients samples would have provided confirmation of their presence, even though most did not correlate with disease (except IGHG2 and IGHG3) or overall survival (except SHR3RF2, GTF2H5 and DAPK1).
The aim of this study was to determine which antigens were recognised by patients with CRC CLR and whether these same antigens could provide insights into why some patients respond well to treatment and others do not. In addition, we were hoping to find novel targets for future treatments of CRC patients, many of whom lack MSI and fail to respond to ICI treatment. We identified two antigens that were recognised by all three CRC CLR patients. Although one of the antigens was novel (UOB-COL-7) with unknown function, the other, IGHG3, showed a number of associations with the clinical features of CRC in a large dataset study. Future studies will determine which antigen(s) are being recognised by these CRC-associated antibodies and whether blocking these antibodies could reduce tumour growth as described previously [53]. Of note was the identification of DAPK1, SH3RF2 and GTF2H5, whose expression provides a clear indication of overall survival and targets worthy of further investigation for CRC treatment.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/biom12081058/s1, Table S1: Patient information for samples analysed by immunohistochemistry (DOI: 10.5281/zenodo.6660264); Table S2: Differential expression of SEREX-defined antigens in colorectal cancer when comparing clinical features using the GSE5206 dataset (DOI: 10.5281/zenodo.5535023), Table S3: Differentially expressed antigens that showed >two-fold difference in expression between (A) AJCC stage (B) T stage (C) collection sites and (D) recurrence type. p-values shown (DOI: 10.5281/zenodo.5535019).

Author Contributions

V.B.B. and B.-a.G. contributed to the writing of the manuscript.; G.O., M.T. and B.-a.G.: Conception of the study; V.B.B., M.L. and B.-a.G.: Design of the study; G.O. and L.O.: recruitment of samples for the study; V.B.B., S.K., H.D. and K.I.M.: Accuracy of the data; H.D. and K.I.M.: Statistical analysis; G.O., M.T. and B.-a.G.: Resources and funding, B.-a.G.: Study supervision, and the guarantor of the manuscript and takes responsibility for the integrity of the work as a whole. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by Breakthrough Cancer Research (previously known as Cork Cancer Research Centre) while the University of Bedfordshire awarded a fee waiver to V.B.B. for her MPhil studies that formed the basis of this publication.

Institutional Review Board Statement

Samples were collected following the Declaration of Helsinki guidelines and local ethical approval, as appropriate to the time at which the study was performed. This study used acellular products (serum) and no genetic material, while data generated from gene expression studies have been cited.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

All data supporting reported results can be found in the supplementary data.

Acknowledgments

Thank you to Aleksandra Kasznicka for her input into the writing of this manuscript.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Amin, M.B.; Greene, F.L.; Edge, S.B.; Compton, C.C.; Gershenwald, J.E.; Brookland, R.K.; Meyer, L.; Gress, D.M.; Byrd, D.R.; Winchester, D.P. The Eighth Edition AJCC Cancer Staging Manual: Continuing to build a bridge from a population-based to a more “personalized” approach to cancer staging. CA Cancer J. Clin. 2017, 67, 93–99. [Google Scholar] [CrossRef] [PubMed]
  2. Cancer Research UK. Bowel Cancer Statistics. 2020. Available online: https://www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/bowel-cancer (accessed on 4 February 2020).
  3. Ganesh, K.; Stadler, Z.K.; Cercek, A.; Mendelsohn, R.B.; Shia, J.; Segal, N.H.; Diaz, L.A., Jr. Immunotherapy in colorectal cancer: Rationale, challenges and potential. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 361–375. [Google Scholar] [CrossRef] [PubMed]
  4. Sahin, I.; Ciombor, K.K.; Diaz, L.A.; Yu, J.; Kim, R. Immunotherapy for Microsatellite Stable Colorectal Cancers: Challenges and Novel Therapeutic Avenues. Am. Soc. Clin. Oncol. Educ. Book 2022, 42, 242–253. [Google Scholar] [CrossRef] [PubMed]
  5. Maoz, A.; Dennis, M.; Greenson, J.K. The Crohn’s-Like Lymphoid Reaction to Colorectal Cancer-Tertiary Lymphoid Structures with Immunologic and Potentially Therapeutic Relevance in Colorectal Cancer. Front. Immunol. 2019, 10, 1884. [Google Scholar] [CrossRef] [Green Version]
  6. Graham, D.M.; Appelman, H.D. Crohn’s-like lymphoid reaction and colorectal carcinoma: A potential histologic prognosticator. Mod. Pathol. 1990, 3, 332–335. [Google Scholar]
  7. Murphy, J.; O’Sullivan, G.C.; Lee, G.; Madden, M.; Shanahan, F.; Collins, J.K.; Talbot, I.C. The inflammatory response within Dukes’ B colorectal cancers: Implications for progression of micrometastases and patient survival. Am. J. Gastroenterol. 2000, 95, 3607–3614. [Google Scholar] [CrossRef]
  8. Boncheva, V.B. The Identification of Tumour Antigens Recognised by Patients with Dukes’ B (Stage II) Reactive Colorectal Cancers Using SEREX. Master’s Thesis, University of Bedfordshire, Luton, UK, 2013. [Google Scholar]
  9. Mullins, C.S.; Micheel, B.; Matschos, S.; Leuchter, M.; Burtin, F.; Krohn, M.; Huhns, M.; Klar, E.; Prall, F.; Linnebacher, M. Integrated Biobanking and Tumor Model Establishment of Human Colorectal Carcinoma Provides Excellent Tools for Preclinical Research. Cancers 2019, 11, 1520. [Google Scholar] [CrossRef] [Green Version]
  10. Sahin, U.; Tureci, O.; Schmitt, H.; Cochlovius, B.; Johannes, T.; Schmits, R.; Stenner, F.; Luo, G.; Schobert, I.; Pfreundschuh, M. Human neoplasms elicit multiple specific immune responses in the autologous host. Proc. Natl. Acad. Sci. USA 1995, 92, 11810–11813. [Google Scholar] [CrossRef] [Green Version]
  11. Liggins, A.P.; Guinn, B.A.; Banham, A.H. Identification of lymphoma-associated antigens using SEREX. Methods Mol. Med. 2005, 115, 109–128. [Google Scholar]
  12. Landsteiner, K. On agglutination of normal human blood. Transfusion 1961, 1, 5–8. [Google Scholar] [CrossRef]
  13. Department of Biochemistry, University of Cambridge. Available online: https://www.bioc.cam.ac.uk/dnasequencing/sanger-sequencing (accessed on 14 April 2016).
  14. Large-Scale Deployment of Embryonic Gene Programming in Human and Murine Colon Cancer: A New Target for Intervention. Available online: https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE5206 (accessed on 14 April 2016).
  15. Kaiser, S.; Park, Y.K.; Franklin, J.L.; Halberg, R.B.; Yu, M.; Jessen, W.J.; Freudenberg, J.; Chen, X.; Haigis, K.; Jegga, A.G.; et al. Transcriptional recapitulation and subversion of embryonic colon development by mouse colon tumor models and human colon cancer. Genome Biol. 2007, 8, R131. [Google Scholar] [CrossRef] [Green Version]
  16. The Cancer Genome Atlas (TCGA) Research Network. Comprehensive molecular characterization of human colon and rectal cancer. Nature 2012, 487, 330–337. [Google Scholar] [CrossRef] [Green Version]
  17. Li, X.F.; Ren, P.; Shen, W.Z.; Jin, X.; Zhang, J. The expression, modulation and use of cancer-testis antigens as potential biomarkers for cancer immunotherapy. Am. J. Transl. Res. 2020, 12, 7002–7019. [Google Scholar] [PubMed]
  18. Ramskold, D.; Wang, E.T.; Burge, C.B.; Sandberg, R. An abundance of ubiquitously expressed genes revealed by tissue transcriptome sequence data. PLoS Comput. Biol. 2009, 5, e1000598. [Google Scholar] [CrossRef]
  19. Chen, Y.T. The journey from autologous typing to SEREX, NY-ESO-1, and cancer/testis antigens. Cancer Immun. 2012, 12, 8. [Google Scholar]
  20. Guinn, B.A.; Bland, E.A.; Lodi, U.; Liggins, A.P.; Tobal, K.; Petters, S.; Wells, J.W.; Banham, A.H.; Mufti, G.J. Humoral detection of leukaemia-associated antigens in presentation acute myeloid leukaemia. Biochem. Biophys. Res. Commun. 2005, 335, 1293–1304. [Google Scholar] [CrossRef]
  21. Zhang, X.; Shen, W.; Dong, X.; Fan, J.; Liu, L.; Gao, X.; Kernstine, K.H.; Zhong, L. Identification of novel autoantibodies for detection of malignant mesothelioma. PLoS ONE 2013, 8, e72458. [Google Scholar] [CrossRef] [Green Version]
  22. Ledet, E.M.; Hu, X.; Sartor, O.; Rayford, W.; Li, M.; Mandal, D. Characterization of germline copy number variation in high-risk African American families with prostate cancer. Prostate 2013, 73, 614–623. [Google Scholar] [CrossRef]
  23. Bin Amer, S.M.; Maqbool, Z.; Nirmal, M.S.; Qattan, A.T.; Hussain, S.S.; Jeprel, H.A.; Tulbah, A.M.; Malik, O.A.; Al-Tweigeri, T.A. Gene expression profiling in women with breast cancer in a Saudi population. Saudi Med. J. 2008, 29, 507–513. [Google Scholar]
  24. Remmelink, M.; Mijatovic, T.; Gustin, A.; Mathieu, A.; Rombaut, K.; Kiss, R.; Salmon, I.; Decaestecker, C. Identification by means of cDNA microarray analyses of gene expression modifications in squamous non-small cell lung cancers as compared to normal bronchial epithelial tissue. Int. J. Oncol. 2005, 26, 247–258. [Google Scholar] [CrossRef]
  25. Collin, J.F.; Wells, J.W.; Czepulkowski, B.; Lyne, L.; Duriez, P.J.; Banham, A.H.; Mufti, G.J.; Guinn, B.A. A novel zinc finger gene, ZNF465, is inappropriately expressed in acute myeloid leukaemia cells. Genes Chromosomes Cancer 2015, 54, 288–302. [Google Scholar] [CrossRef] [PubMed]
  26. Blanke, K.L.; Sacco, J.C.; Millikan, R.C.; Olshan, A.F.; Luo, J.; Trepanier, L.A. Polymorphisms in the carcinogen detoxification genes CYB5A and CYB5R3 and breast cancer risk in African American women. Cancer Causes Control 2014, 25, 1513–1521. [Google Scholar] [CrossRef] [PubMed]
  27. Yang, W.; Wang, Y.; Pu, Q.; Ye, S.; Ma, Q.; Ren, J.; Zhong, G.; Liu, L.; Zhu, W. Elevated expression of SLC34A2 inhibits the viability and invasion of A549 cells. Mol. Med. Rep. 2014, 10, 1205–1214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Chen, Z.; Li, M.; Yuan, Y.; Wang, Q.; Yan, L.; Gu, J. Cancer/testis antigens and clinical risk factors for liver metastasis of colorectal cancer: A predictive panel. Dis. Colon. Rectum 2010, 53, 31–38. [Google Scholar] [CrossRef]
  29. Lopes dos Santos, M.; Yeda, F.P.; Tsuruta, L.R.; Horta, B.B.; Pimenta, A.A., Jr.; Degaki, T.L.; Soares, I.C.; Tuma, M.C.; Okamoto, O.K.; Alves, V.A.; et al. Rebmab200, a humanized monoclonal antibody targeting the sodium phosphate transporter NaPi2b displays strong immune mediated cytotoxicity against cancer: A novel reagent for targeted antibody therapy of cancer. PLoS ONE 2013, 8, e70332. [Google Scholar] [CrossRef] [Green Version]
  30. Shyian, M.; Gryshkova, V.; Kostianets, O.; Gorshkov, V.; Gogolev, Y.; Goncharuk, I.; Nespryadko, S.; Vorobjova, L.; Filonenko, V.; Kiyamova, R. Quantitative analysis of SLC34A2 expression in different types of ovarian tumors. Exp. Oncol. 2011, 33, 94–98. [Google Scholar]
  31. Barros Filho, M.C.; Katayama, M.L.; Brentani, H.; Abreu, A.P.; Barbosa, E.M.; Oliveira, C.T.; Goes, J.C.; Brentani, M.M.; Folgueira, M.A. Gene trio signatures as molecular markers to predict response to doxorubicin cyclophosphamide neoadjuvant chemotherapy in breast cancer patients. Braz. J. Med. Biol. Res. 2010, 43, 1225–1231. [Google Scholar] [CrossRef] [Green Version]
  32. Kim, T.W.; Kang, Y.K.; Park, Z.Y.; Kim, Y.H.; Hong, S.W.; Oh, S.J.; Sohn, H.A.; Yang, S.J.; Jang, Y.J.; Lee, D.C.; et al. SH3RF2 functions as an oncogene by mediating PAK4 protein stability. Carcinogenesis 2014, 35, 624–634. [Google Scholar] [CrossRef]
  33. Zhang, Q.; Zhang, J.; Jin, H.; Sheng, S. Whole transcriptome sequencing identifies tumor-specific mutations in human oral squamous cell carcinoma. BMC Med. Genom. 2013, 6, 28. [Google Scholar] [CrossRef] [Green Version]
  34. Li, F.F.; Yang, Y.; Wang, X.L.; Hong, Y.Y.; Wang, N.F.; Chen, Z.D. Promoter methylation of DAPK gene may contribute to the pathogenesis of nonsmall cell lung cancer: A meta-analysis. Tumour Biol. 2014, 35, 6011–6020. [Google Scholar] [CrossRef]
  35. Niu, Y.M.; Wang, P.P.; Wang, Y.; Wang, Y.Z.; Cai, D.L.; Li, Y. Expression of death-associated protein kinase gene and methylation status of promoter region in acute leukemia. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2014, 22, 30–34. [Google Scholar] [CrossRef]
  36. Huang, Y.; Chen, L.; Guo, L.; Hupp, T.R.; Lin, Y. Evaluating DAPK as a therapeutic target. Apoptosis 2014, 19, 371–386. [Google Scholar] [CrossRef]
  37. Zheng, H.; Li, M.; Ren, W.; Zeng, L.; Liu, H.D.; Hu, D.; Deng, X.; Tang, M.; Shi, Y.; Gong, J.; et al. Expression and secretion of immunoglobulin alpha heavy chain with diverse VDJ recombinations by human epithelial cancer cells. Mol. Immunol. 2007, 44, 2221–2227. [Google Scholar] [CrossRef]
  38. Leibovitz, A.; Stinson, J.C.; McCombs, W.B., III; McCoy, C.E.; Mazur, K.C.; Mabry, N.D. Classification of human colorectal adenocarcinoma cell lines. Cancer Res. 1976, 36, 4562–4569. [Google Scholar]
  39. Brattain, M.G.; Brattain, D.E.; Fine, W.D.; Khaled, F.M.; Marks, M.E.; Kimball, P.M.; Arcolano, L.A.; Danbury, B.H. Initiation and characterization of cultures of human colonic carcinoma with different biological characteristics utilizing feeder layers of confluent fibroblasts. Oncodevelopmental Biol. Med. 1981, 2, 355–366. [Google Scholar]
  40. Smith, J.J.; Deane, N.G.; Wu, F.; Merchant, N.B.; Zhang, B.; Jiang, A.; Lu, P.; Johnson, J.C.; Schmidt, C.; Bailey, C.E.; et al. Experimentally derived metastasis gene expression profile predicts recurrence and death in patients with colon cancer. Gastroenterology 2010, 138, 958–968. [Google Scholar] [CrossRef] [Green Version]
  41. Lawrance, I.C.; Fiocchi, C.; Chakravarti, S. Ulcerative colitis and Crohn’s disease: Distinctive gene expression profiles and novel susceptibility candidate genes. Hum. Mol. Genet. 2001, 10, 445–456. [Google Scholar] [CrossRef] [Green Version]
  42. Qiu, X.; Zhu, X.; Zhang, L.; Mao, Y.; Zhang, J.; Hao, P.; Li, G.; Lv, P.; Li, Z.; Sun, X.; et al. Human epithelial cancers secrete immunoglobulin g with unidentified specificity to promote growth and survival of tumor cells. Cancer Res. 2003, 63, 6488–6495. [Google Scholar]
  43. Li, M.; Feng, D.Y.; Ren, W.; Zheng, L.; Zheng, H.; Tang, M.; Cao, Y. Expression of immunoglobulin kappa light chain constant region in abnormal human cervical epithelial cells. Int. J. Biochem. Cell Biol. 2004, 36, 2250–2257. [Google Scholar] [CrossRef]
  44. Chen, Z.; Gu, J. Immunoglobulin G expression in carcinomas and cancer cell lines. FASEB J. 2007, 21, 2931–2938. [Google Scholar] [CrossRef]
  45. Wang, J.; Lin, D.; Peng, H.; Huang, Y.; Huang, J.; Gu, J. Cancer-derived immunoglobulin G promotes tumor cell growth and proliferation through inducing production of reactive oxygen species. Cell Death Dis. 2013, 4, e945. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Jiang, C.; Huang, T.; Wang, Y.; Huang, G.; Wan, X.; Gu, J. Immunoglobulin G expression in lung cancer and its effects on metastasis. PLoS ONE 2014, 9, e97359. [Google Scholar] [CrossRef] [PubMed]
  47. Liao, Q.; Liu, W.; Liu, Y.; Wang, F.; Wang, C.; Zhang, J.; Chu, M.; Jiang, D.; Xiao, L.; Shao, W.; et al. Aberrant high expression of immunoglobulin G in epithelial stem/progenitor-like cells contributes to tumor initiation and metastasis. Oncotarget 2015, 6, 40081–40094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Tang, J.; Zhang, J.; Liu, Y.; Liao, Q.; Huang, J.; Geng, Z.; Xu, W.; Sheng, Z.; Lee, G.; Zhang, Y.; et al. Lung squamous cell carcinoma cells express non-canonically glycosylated IgG that activates integrin-FAK signaling. Cancer Lett. 2018, 430, 148–159. [Google Scholar] [CrossRef]
  49. Sheng, Z.; Liu, Y.; Qin, C.; Liu, Z.; Yuan, Y.; Hu, F.; Du, Y.; Yin, H.; Qiu, X.; Xu, T. IgG is involved in the migration and invasion of clear cell renal cell carcinoma. J. Clin. Pathol. 2016, 69, 497–504. [Google Scholar] [CrossRef] [Green Version]
  50. Barbera-Guillem, E.; Nyhus, J.K.; Wolford, C.C.; Friece, C.R.; Sampsel, J.W. Vascular endothelial growth factor secretion by tumor-infiltrating macrophages essentially supports tumor angiogenesis, and IgG immune complexes potentiate the process. Cancer Res. 2002, 62, 7042–7049. [Google Scholar]
  51. Geng, Z.H.; Ye, C.X.; Huang, Y.; Jiang, H.P.; Ye, Y.J.; Wang, S.; Zhou, Y.; Shen, Z.L.; Qiu, X.Y. Human colorectal cancer cells frequently express IgG and display unique Ig repertoire. World J. Gastrointest. Oncol. 2019, 11, 195–207. [Google Scholar] [CrossRef]
  52. Hsu, H.M.; Chu, C.M.; Chang, Y.J.; Yu, J.C.; Chen, C.T.; Jian, C.E.; Lee, C.Y.; Chiang, Y.T.; Chang, C.W.; Chang, Y.T. Six novel immunoglobulin genes as biomarkers for better prognosis in triple-negative breast cancer by gene co-expression network analysis. Sci. Rep. 2019, 9, 4484. [Google Scholar] [CrossRef] [Green Version]
  53. Niu, N.; Zhang, J.; Huang, T.; Sun, Y.; Chen, Z.; Yi, W.; Korteweg, C.; Wang, J.; Gu, J. IgG expression in human colorectal cancer and its relationship to cancer cell behaviors. PLoS ONE 2012, 7, e47362. [Google Scholar] [CrossRef] [Green Version]
  54. Xu, Q.; Zhang, Z.; Chen, Z.; Zhang, B.; Zhao, C.; Zhang, Y.; Zhao, C.; Deng, X.; Zhou, Y.; Wu, Y.; et al. Nonspecific immunoglobulin G is effective in preventing and treating cancer in mice. Cancer Manag. Res. 2019, 11, 2073–2085. [Google Scholar] [CrossRef] [Green Version]
  55. Shoenfeld, Y.; Katz, U. IVIg therapy in autoimmunity and related disorders: Our experience with a large cohort of patients. Autoimmunity 2005, 38, 123–137. [Google Scholar] [CrossRef]
  56. Krause, I.; Shoenfeld, Y. Intravenous immunoglobulin treatment for fibrosis, atherosclerosis, and malignant conditions. Methods Mol. Med. 2005, 109, 403–408. [Google Scholar] [CrossRef]
  57. Sobieszczanska, M.; Tubek, S.; Poplicha, D.; Grabelus, A.; Pawelczak, J. Henoch-Schonlein purpura (HSP) and high-dose immunoglobulin treatment in patient with familiar prostatic adenocarcinoma. Hum. Vaccines Immunother. 2014, 10, 358–359. [Google Scholar] [CrossRef]
  58. Shang, Y.; Zhang, X.; Lu, L.; Jiang, K.; Krohn, M.; Matschos, S.; Mullins, C.S.; Vollmar, B.; Zechner, D.; Gong, P.; et al. Pharmaceutical immunoglobulin G impairs anti-carcinoma activity of oxaliplatin in colon cancer cells. Br. J. Cancer 2021, 124, 1411–1420. [Google Scholar] [CrossRef]
  59. Mon, M.M.; Srisomsap, C.; Chokchaichamnankit, D.; Watcharatanyatip, K.; Weeraphan, C.; Svasti, J.; Maneechai, K.; Thongsuksai, P.; Raungrut, P. Serum Proteomic Profiling Reveals Differentially Expressed IGHG3 and A1AG1 as Potential Predictors of Chemotherapeutic Response in Advanced Non-small Cell Lung Cancer. Anticancer Res. 2021, 41, 1871–1882. [Google Scholar] [CrossRef]
  60. Rhoads, K.; Sacco, J.C.; Drescher, N.; Wong, A.; Trepanier, L.A. Individual variability in the detoxification of carcinogenic arylhydroxylamines in human breast. Toxicol. Sci. 2011, 121, 245–256. [Google Scholar] [CrossRef] [Green Version]
  61. Lund, R.R.; Leth-Larsen, R.; Caterino, T.D.; Terp, M.G.; Nissen, J.; Laenkholm, A.V.; Jensen, O.N.; Ditzel, H.J. NADH-Cytochrome b5 Reductase 3 Promotes Colonization and Metastasis Formation and Is a Prognostic Marker of Disease-Free and Overall Survival in Estrogen Receptor-Negative Breast Cancer. Mol. Cell Proteom. 2015, 14, 2988–2999. [Google Scholar] [CrossRef] [Green Version]
  62. MacDonald, T.J.; Pollack, I.F.; Okada, H.; Bhattacharya, S.; Lyons-Weiler, J. Progression-associated genes in astrocytoma identified by novel microarray gene expression data reanalysis. Methods Mol. Biol. 2007, 377, 203–222. [Google Scholar] [CrossRef]
  63. Serao, N.V.; Delfino, K.R.; Southey, B.R.; Beever, J.E.; Rodriguez-Zas, S.L. Cell cycle and aging, morphogenesis, and response to stimuli genes are individualized biomarkers of glioblastoma progression and survival. BMC Med. Genom. 2011, 4, 49. [Google Scholar] [CrossRef] [Green Version]
  64. Liu, L.; Yang, Y.; Zhou, X.; Yan, X.; Wu, Z. Solute carrier family 34 member 2 overexpression contributes to tumor growth and poor patient survival in colorectal cancer. Biomed. Pharmacother. 2018, 99, 645–654. [Google Scholar] [CrossRef]
  65. Satoh, A.; Toyota, M.; Itoh, F.; Kikuchi, T.; Obata, T.; Sasaki, Y.; Suzuki, H.; Yawata, A.; Kusano, M.; Fujita, M.; et al. DNA methylation and histone deacetylation associated with silencing DAP kinase gene expression in colorectal and gastric cancers. Br. J. Cancer 2002, 86, 1817–1823. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Zhao, W.-H.; Meng, F.Y.; Lai, Y.-R.; Peng, Z.-G.; Ma, J. Promoter methylation and expression of death-associated protein kinase gene in acute leukemia. Nan Fang Yi Ke Da Xue Xue Bao (Natl. Libr. Med.) 2017, 37, 407–410. [Google Scholar] [CrossRef]
  67. Steinmann, S.; Kunze, P.; Hampel, C.; Eckstein, M.; Bertram Bramsen, J.; Muenzner, J.K.; Carle, B.; Ndreshkjana, B.; Kemenes, S.; Gasparini, P.; et al. DAPK1 loss triggers tumor invasion in colorectal tumor cells. Cell Death Dis. 2019, 10, 895. [Google Scholar] [CrossRef] [PubMed]
  68. Ivanovska, J.; Zlobec, I.; Forster, S.; Karamitopoulou, E.; Dawson, H.; Koelzer, V.H.; Agaimy, A.; Garreis, F.; Soder, S.; Laqua, W.; et al. DAPK loss in colon cancer tumor buds: Implications for migration capacity of disseminating tumor cells. Oncotarget 2015, 6, 36774–36788. [Google Scholar] [CrossRef]
  69. Urrutia, R. KRAB-containing zinc-finger repressor proteins. Genome Biol. 2003, 4, 231. [Google Scholar] [CrossRef] [Green Version]
  70. Yang, H.; Yuan, W.; Wang, Y.; Zhu, C.; Liu, B.; Wang, Y.; Yang, D.; Li, Y.; Wang, C.; Wu, X.; et al. ZNF649, a novel Kruppel type zinc-finger protein, functions as a transcriptional suppressor. Biochem. Biophys. Res. Commun. 2005, 333, 206–215. [Google Scholar] [CrossRef]
  71. Maier, T.; Guell, M.; Serrano, L. Correlation of mRNA and protein in complex biological samples. FEBS Lett. 2009, 583, 3966–3973. [Google Scholar] [CrossRef] [Green Version]
Figure 1. IGHG2 and IGHG3 expression levels were examined in 571 CRC patient samples following RNA-seq (TCGA-COAD dataset). There was no evidence of a unique sub-population of patients with elevated IGHG3 expression within the CRC population. However, we did find a significant association between the levels of IGHG2 (x-axis) and IGHG3 (y-axis) expression suggesting the transcription of these constant regions have similar expression profiles in CRC patients (p = 8.25 × 10−145).
Figure 1. IGHG2 and IGHG3 expression levels were examined in 571 CRC patient samples following RNA-seq (TCGA-COAD dataset). There was no evidence of a unique sub-population of patients with elevated IGHG3 expression within the CRC population. However, we did find a significant association between the levels of IGHG2 (x-axis) and IGHG3 (y-axis) expression suggesting the transcription of these constant regions have similar expression profiles in CRC patients (p = 8.25 × 10−145).
Biomolecules 12 01058 g001
Figure 2. Immunolabelling of intracellular IgG3 in CRC samples. Examples of staining include single cells, strong in stroma (HROC086) and parenchyma (HROC0252_Tu2) and very slight background staining (HROC117).
Figure 2. Immunolabelling of intracellular IgG3 in CRC samples. Examples of staining include single cells, strong in stroma (HROC086) and parenchyma (HROC0252_Tu2) and very slight background staining (HROC117).
Biomolecules 12 01058 g002
Figure 3. IgG3 levels in CRC patients. Levels of IgG3 were elevated in the sera of CRC patients used for immunoscreening when compared to healthy donors (p = 0.01), however within this group the IgG3 sera levels in CC005 and CC010 (shown as red squares) were not elevated above the rest of the group of CRC patients.
Figure 3. IgG3 levels in CRC patients. Levels of IgG3 were elevated in the sera of CRC patients used for immunoscreening when compared to healthy donors (p = 0.01), however within this group the IgG3 sera levels in CC005 and CC010 (shown as red squares) were not elevated above the rest of the group of CRC patients.
Biomolecules 12 01058 g003
Figure 4. Gene expression analysis was used to determine the impact of above and below median antigen expression on survival during follow-up. The probability of overall survival is shown on the y-axis, while time in months is shown on the x-axis. Below median levels of (i) DAPK1 and above median levels of (ii) SH3RF2 and (iii) GTF2H5 were associated with improved 5-year overall survival in CRC patients.
Figure 4. Gene expression analysis was used to determine the impact of above and below median antigen expression on survival during follow-up. The probability of overall survival is shown on the y-axis, while time in months is shown on the x-axis. Below median levels of (i) DAPK1 and above median levels of (ii) SH3RF2 and (iii) GTF2H5 were associated with improved 5-year overall survival in CRC patients.
Biomolecules 12 01058 g004
Table 4. IgG3 expression in CRC samples.
Table 4. IgG3 expression in CRC samples.
Sample IDMolecular SubtypeControlIgG3Observations Regarding IgG3 Staining
HROC40CIMP-H, non MSI−/+Very slight background staining
HROC54CIMP-H, non MSI+Very slight background staining
HROC60CIMP-H, non MSI+Very slight background staining
HROC62spStdDiffusely in necrosis
HROC86spStd++Single cells strong in stroma
HROC117CIMP-H, non MSI++Very slight background staining
HROC126spStdWeak and diffusely in necrosis
HROC131spMSI-H
HROC155spStdSingle cells moderate in stroma
HROC159spMSI-H+Moderate staining in stroma
HROC169CIMP-H, non MSI
HROC212spMSI-H+Single cells in stroma
HROC252_Tu1Lynch+Diffusely IgG3 in stroma
HROC252_Tu2++Single cells in stroma, also in parenchyma
HROC252_Tu3+Single cells in stroma, low in parenchyma
HROC257spMSI-H+Weak and diffusely IgG3 in necrosis, weak in stroma
HROC260spStdSingle cells weak in stroma
HROC269spMSI-HWeak in necrosis
HROC315Lynch+Necrotic tumour, IgG3 strong in single cells in stroma
HROC324LynchWeak in necrosis
Tonsil +++/++++Regions of heavily brown staining
CIMP-H: CpG island methylator phenotype-high; Lynch: Lynch syndrome, also known as hereditary non-polyposis colorectal cancer (HNPCC); MSI: microsatellite instability; sp: sporadic; spStd: sporadic standard type.
Table 5. Fold change in antigen transcript levels that achieved significance, when comparing AJCC stage. Light green cells indicate values that are increased by greater than 2-fold, yellow cells indicate values that are decreased by greater than 2-fold.
Table 5. Fold change in antigen transcript levels that achieved significance, when comparing AJCC stage. Light green cells indicate values that are increased by greater than 2-fold, yellow cells indicate values that are decreased by greater than 2-fold.
Probeset IDGene SymbolStage 0 down vs. StageStage 2B down vs. Stage 3B
22B3B4
213459_atRPL37A1.422.411.041.27−2.31
211650_x_atIGH family members2.252.731.771.89−1.54
214916_x_at2.192.861.632.05−1.75
216557_x_at2.142.111.601.77−1.32
217281_x_at1.912.041.501.60−1.36
Table 6. Fold change in antigen transcript levels that achieved significance, when comparing T stage. Light green cells indicate values that are increased by greater than 2-fold, yellow cells indicate values that are decreased by greater than 2-fold.
Table 6. Fold change in antigen transcript levels that achieved significance, when comparing T stage. Light green cells indicate values that are increased by greater than 2-fold, yellow cells indicate values that are decreased by greater than 2-fold.
Probeset IDGene SymbolStage Comparisons
1 vs. 31 vs. 41 vs. IS1 vs. X3 vs. IS
213459_atRPL37A2.572.381.772.52−1.45
211639_x_atIGH family members−1.24−1.26−2.20−1.32−1.77
211650_x_at1.131.01−1.781.04−2.00
214916_x_at1.161.11−1.77−1.01−2.04
Table 7. Fold change in antigen transcript levels that achieved significance, when comparing collection sites (colon ascending, descending and nos). Light green cells indicate values that are increased by greater than 2-fold, yellow cells indicate values that are decreased by greater than 2-fold.
Table 7. Fold change in antigen transcript levels that achieved significance, when comparing collection sites (colon ascending, descending and nos). Light green cells indicate values that are increased by greater than 2-fold, yellow cells indicate values that are decreased by greater than 2-fold.
Probeset IDGene SymbolCOLON (ASCENDING) vs. COLON (NOS)COLON (ASCENDING)COLON (DESCENDING)COLON (DESCENDING)COLON (NOS)
vs. COLON (SPLENIC FLEXURE)vs. HEPATIC FLEXUREvs. COLON (NOS)vs. RECTUM (NOS)vs. COLON (SPLENIC FLEXURE)vs. HEPATIC FLEXUREvs. COLON (SIGMOID)vs. COLON (SPLENIC FLEXURE)vs COLON (TRANSVERSE)vs. HEPATIC FLEXUREvs. RECTOSIGMOID JUNCTION
211430_s_atIGH family members2.26−1.81−1.21.29−2.06−3.19−2.11−2.38−4.1−1.5−2.71−2.00
211635_x_at1.91−2.171.161.26−1.64−3.3−1.31−2.37−4.15−1.56−1.65−1.46
211637_x_at4.23−2.562.142.89−1.63−3.751.46−3.86−10.8−3.6−1.98−2.74
211639_x_at2.62−1.941.662.2−1.37−2.311.39−2.86−5.08−2.46−1.58−2.08
211641_x_at1.62−1.871.821.38−1.27−2.191.55−1.59−3.03−1.371.12−1.18
211650_x_at3.04−2.132.272.53−1.5−2.561.89−3.01−6.47−2.58−1.34−2.26
211868_x_at2.5−1.541.431.91−1.35−2.021.09−2.52−3.86−2.26−1.75−2.20
214916_x_at2.8−1.962.761.98−1.76−2.781.95−2.73−5.5−2.16−1.01−1.92
216557_x_at3.68−2.222.342.26−1.86−3.611.44−3.31−8.15−2.93−1.57−2.47
217236_x_at1.33−1.671.971.16−1.19−1.911.72−1.31−2.22−1.191.48−1.12
217281_x_at2.95−2.122.561.98−1.69−3.151.72−2.81−6.26−2.3−1.15−2.23
217360_x_at1.33−1.751.331.34−1.09−1.731.34−1.4−2.33−1.211.0−1.16
Table 8. Fold change in antigen transcript levels that achieved significance, when comparing collection sites including colon (cecum, nos, splenic flexure, sigmoid, transverse) and rectum (nos). Light green cells indicate values that are increased by greater than 2-fold, yellow cells indicate values that are decreased by greater than 2-fold.
Table 8. Fold change in antigen transcript levels that achieved significance, when comparing collection sites including colon (cecum, nos, splenic flexure, sigmoid, transverse) and rectum (nos). Light green cells indicate values that are increased by greater than 2-fold, yellow cells indicate values that are decreased by greater than 2-fold.
Probeset IDGene SymbolCOLON (CECUM) vs. COLON (NOS)COLON (CECUM)COLON (CECUM) vs. COLON (NOS)COLON (NOS) vs. RECTUM (NOS)COLON (SIGMOID)COLON (SPLENIC FLEXURE)COLON (TRANSVERSE) vs. HEPATIC FLEXUREHEPATIC FLEXURE vs. RECTUM (NOS)
vs. COLON (SPLENIC FLEXURE)vs. COLON (SPLENIC FLEXURE)vs. HEPATIC FLEXUREvs. COLON (TRANSVERSE)vs. HEPATIC FLEXUREvs. RECTOSIGMOID JUNCTIONvs. RECTUM (NOS)
211430_s_atIGH family members2.46−1.672.46−2.65−1.72−1.142.741.512.051.55−1.811.02
211635_x_at1.95−2.131.95−2.06−1.751.442.672.512.842.02−1.06−1.25
211637_x_at3.85−2.813.85−4.71−2.811.953.015.473.952.301.82−2.38
211639_x_at2.66−1.912.66−3.02−1.781.812.073.222.451.681.55−1.91
211641_x_at1.49−2.031.49−1.75−1.901.782.213.402.561.731.54−1.96
211650_x_at2.87−2.252.87−3.79−2.152.262.514.852.871.711.93−2.84
211868_x_at2.29−1.692.29−2.58−1.531.441.702.201.751.501.29−1.47
214916_x_at2.34−2.352.34−3.49−2.022.692.555.432.871.582.12−3.44
216557_x_at3.29−2.483.29−4.20−2.462.112.795.193.311.941.86−2.68
217236_x_at1.28−1.731.28−1.39−1.691.941.873.291.971.601.76−2.06
217281_x_at2.75−2.272.75−3.36−2.232.442.715.432.801.862.00−2.92
217360_x_at1.33−1.751.33−1.47−1.671.401.932.332.011.581.21−1.47
Table 9. Fold change in antigen transcript levels that achieved significance, when comparing recurrence type. Light green cells indicate values that are increased by greater than 2-fold, yellow cells indicate values that are decreased by greater than 2-fold.
Table 9. Fold change in antigen transcript levels that achieved significance, when comparing recurrence type. Light green cells indicate values that are increased by greater than 2-fold, yellow cells indicate values that are decreased by greater than 2-fold.
Probset IDGene SymbolDIST RECUR TO PERITONEUM OR ASCITESDIST SITE OF RECUR, BEHAV NOS (40)DIST SITE OF RECUR, BEHAV NOS (40)LOCAL RECURRENCE, BEHAV NOS (10)NEVER DISEASE FREE SINCE DX (70)REGIONAL RECURRENCE, BEHAV NOS (20)
vs. DIST SITE OF RECUR, BEHAV NOS (40)vs. LOCAL RECURRENCE, BEHAV NOS (10)vs. NEVER DISEASE FREE SINCE DX (70)vs. REGIONAL RECURRENCE, BEHAV NOS (20)vs. REGIONAL TISSUE RECUR OF INVAS CA (21)vs LOCAL RECURRENCE, BEHAV NOS (10)vs REGIONAL RECURRENCE, BEHAV NOS (20)vs REGIONAL TISSUE RECUR OF INVAS CA (21)vs NEVER DISEASE FREE SINCE DX (70)vs REGIONAL RECURRENCE, BEHAV NOS (20)vs REGIONAL TISSUE RECUR OF INVAS CA (21)vs REGIONAL TISSUE RECUR OF INVAS CA (21)vs REGIONAL TISSUE RECUR OF INVAS CA (21)
1554574_a_atCYB5R3−2.31−2.78−2.58−2.02−1.85−1.21.151.251.081.381.51.391.09
211430_s_atIGH family members1.06−21.38−1.383.2−2.11−1.463.032.761.456.42.324.42
211635_x_at−1.02−10.57−1.32−1.61−2.26−10.35−1.58−2.217.996.574.68−1.7−1.4
211637_x_at−1.39−10.91−1.92−2.70−2.02−7.83−1.94−1.455.694.035.4−1.051.34
211639_x_at−1.09−3.63−1.25−1.73−1.47−3.31−1.58−1.342.92.12.47−1.171.18
211641_x_at−1.26−4.86−1.34−1.74−1.59−3.86−1.38−1.263.622.83.06−1.181.09
211650_x_at−1.39−9.01−1.45−2.01−2.01−6.48−1.44−1.456.24.494.48−1.38−1.00
211868_x_at1.20−2.471.09−1.251.08−2.97−1.51−1.122.691.972.66−1.011.35
214916_x_at−1.2−11.5−1.40−1.84−1.88−9.58−1.53−1.578.196.266.1−1.34−1.03
216557_x_at−1.47−12.9−1.67−2.55−1.82−8.38−1.74−1.247.364.826.75−1.091.4
217236_x_at−1.3−2.13−1.43−1.59−1.71−1.63−1.22−1.311.491.331.24−1.2−1.07
217281_x_at−1.18−10.01−1.32−1.99−1.82−8.5−1.69−1.557.595.045.49−1.381.09
217360_x_at−1.17−3.25−1.06−1.57−1.75−2.78−1.34−1.493.072.071.86−1.65−1.11
234419_x_at−1.02−2.24−1.04−1.24−1.6−2.21−1.22−1.582.151.81.4−1.54−1.29
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Boncheva, V.B.; Linnebacher, M.; Kdimati, S.; Draper, H.; Orchard, L.; Mills, K.I.; O’Sullivan, G.; Tangney, M.; Guinn, B.-a. Identification of the Antigens Recognised by Colorectal Cancer Patients Using Sera from Patients Who Exhibit a Crohn’s-like Lymphoid Reaction. Biomolecules 2022, 12, 1058. https://doi.org/10.3390/biom12081058

AMA Style

Boncheva VB, Linnebacher M, Kdimati S, Draper H, Orchard L, Mills KI, O’Sullivan G, Tangney M, Guinn B-a. Identification of the Antigens Recognised by Colorectal Cancer Patients Using Sera from Patients Who Exhibit a Crohn’s-like Lymphoid Reaction. Biomolecules. 2022; 12(8):1058. https://doi.org/10.3390/biom12081058

Chicago/Turabian Style

Boncheva, Viktoriya B., Michael Linnebacher, Said Kdimati, Hannah Draper, Laurence Orchard, Ken I. Mills, Gerald O’Sullivan, Mark Tangney, and Barbara-ann Guinn. 2022. "Identification of the Antigens Recognised by Colorectal Cancer Patients Using Sera from Patients Who Exhibit a Crohn’s-like Lymphoid Reaction" Biomolecules 12, no. 8: 1058. https://doi.org/10.3390/biom12081058

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop