1. Introduction
Pancreatic cancer has become the third leading cause of death in men and women combined in United States, 2022 [
1]. The development of pancreatic cancer has been explained by four common oncogenic events, including
KRAS, CDKN2A, SMAD4, and
TP53 [
2,
3]. However, pancreatic cancer shows the heterogeneity in drug response and clinical outcomes [
4]. Therefore, the detailed understanding of pancreatic cancers has been required to improve patient selection for current therapies, and to develop novel therapeutic strategies. An integrated genomic analysis of pancreatic ductal adenocarcinomas (PDAC) was performed and defined four subtypes, including squamous, pancreatic progenitor, immunogenic, and aberrantly differentiated endocrine exocrine (ADEX), which correspond to the histopathological characteristics [
5]. Additionally, various marker proteins have been investigated for early diagnostic and drug responses of pancreatic cancers [
6,
7]. Studies have suggested that CD44 plays important roles in malignant progression of tumors through its cancer stemness and metastasis-promoting properties [
8].
CD44 is a type I transmembrane glycoprotein, and is expressed as a wide variety of isoforms in various type of cells. [
9]. The variety of isoforms is produced by the alternative splicing of CD44 mRNA. The CD44 standard isoform (CD44s) is the smallest isoform of CD44 (85–95 kDa), which presents on the membrane of most vertebrate cells. CD44s mRNA is assembled by the first five and the last five constant region exons [
10]. The CD44 variant isoforms (CD44v) are produced by the alternative splicing of middle variant exons (v1–v10) and the standard exons of CD44s [
11]. Both CD44s and CD44v (pan-CD44) are known as hyaluronic acid (HA) receptor, which mediates cellular homing, migration, adhesion, and proliferation [
12,
13].
CD44v isoforms were overexpressed in carcinomas and induced metastatic properties [
14,
15]. A growing body of evidence suggests that CD44v plays critical roles in the promotion of tumor invasion, metastasis, cancer-initiating properties [
16], and resistance to chemo- and radiotherapy [
17,
18]. Reports indicated the important functions of each variant exon-encoded region. The v3-encoded region acts as a co-receptor of receptor tyrosine kinases [
19]. Since the v3-encoded region possesses heparan sulfate moieties, it can recruit to fibroblast growth factors (FGFs) and heparin-binding epidermal growth factor-like growth factor (HB-EGF). Furthermore, the v6-encoded region forms ternary complex with HGF and its receptor c-MET, which is essential for the activation [
20]. Additionally, the oxidative stress resistance is mediated by the v8–10-encoded region through binding with a cystine–glutamate transporter (xCT) subunit [
21]. Therefore, establishment and characterization of mAbs, which recognize each CD44v, are thought to be essential for understanding each variant function and development of CD44-targeting tumor diagnosis and therapy. However, the function and distribution of the variant 5-encoded region in tumors have not been fully understood.
Our group established the novel anti-pan-CD44 mAbs, C
44Mab-5 (IgG
1, kappa) [
22] and C
44Mab-46 (IgG
1, kappa) [
23] using the Cell-Based Immunization and Screening (CBIS) method and immunization of CD44v3–10 ectodomain, respectively. Both C
44Mab-5 and C
44Mab-46 have epitopes within the standard exons (1 to 5)-encoding sequences [
24,
25,
26]. Furthermore, we showed that both C
44Mab-5 and C
44Mab-46 are applicable to flow cytometry and immunohistochemistry in oral [
22] and esophageal squamous cell carcinomas (SCC) [
23]. We have also investigated the antitumor effects of core-fucose deficient C
44Mab-5 in mouse xenograft models of oral SCC [
27]. Here, we developed a novel anti-CD44v5 mAb, C
44Mab-3 (IgG
1, kappa) by CBIS method, and assessed its applications, including flow cytometry, western blotting, and immunohistochemical analyses.
2. Materials and Methods
2.1. Cell Lines
CHO-K1 and mouse multiple myeloma P3X63Ag8U.1 (P3U1) cell lines were obtained from the American Type Culture Collection (ATCC, Manassas, VA, USA). Human pancreas cancer cell lines, PK-1 and PK-8 were obtained from the Cell Resource Center for Biomedical Research Institute of Development, Aging and Cancer at Tohoku University. These cells were cultured in Roswell Park Memorial Institute (RPMI)-1640 medium (Nacalai Tesque, Inc., Kyoto, Japan), supplemented with 100 U/mL penicillin, 100 μg/mL streptomycin, 0.25 μg/mL amphotericin B (Nacalai Tesque, Inc.), and 10% heat-inactivated fetal bovine serum (FBS; Thermo Fisher Scientific, Inc., Waltham, MA, USA). All the cells were grown in a humidified incubator at 37 °C with 5% CO2.
2.2. Plasmid Construction and Establishment of Stable Transfectants
CD44v3–10 open reading frame was obtained from the RIKEN BRC through the National Bio-Resource Project of the MEXT, Japan. CD44s cDNA was amplified using HotStar HiFidelity Polymerase Kit (Qiagen Inc., Hilden, Germany) using LN229 (a glioblastoma cell line) cDNA as a template. CD44v3–10 and CD44 cDNAs were cloned into pCAG-Ble-ssPA16 vector with signal sequence and N-terminal PA16 tag of 16 amino acids (GLEGGVAMPGAEDDVV) [
22,
28,
29,
30,
31], which is detected by NZ-1, which was originally developed against human podoplanin [
32,
33,
34,
35,
36,
37,
38,
39,
40,
41,
42,
43,
44,
45,
46,
47]. The pCAG-Ble/PA16-CD44s and pCAG-Ble/PA16-CD44v3–10 vectors were transfected into CHO-K1 cells using a Neon transfection system (Thermo Fisher Scientific, Inc.), which offers an innovative electroporation method that utilizes a proprietary biologically compatible pipette tip chamber to generate a more uniform electric field for a significant increase in transfection efficiency and cell viability. By limiting dilution method, CHO/CD44s and CHO/CD44v3–10 clones were finally established.
2.3. Hybridomas
The female BALB/c mice were purchased from CLEA Japan (Tokyo, Japan). Animals were housed under specific pathogen-free conditions. All animal experiments were also conducted according to relevant guidelines and regulations to minimize animal suffering and distress in the laboratory. The Animal Care and Use Committee of Tohoku University (Permit number: 2019NiA-001) approved animal experiments. The mice were monitored daily for health during the full four-week duration of the experiment. A reduction of more than 25% of the total body weight was defined as a humane endpoint. During sacrifice, the mice were euthanized through cervical dislocation, after which death was verified through respiratory and cardiac arrest. BALB/c mice were intraperitoneally immunized with CHO/CD44v3–10 (1 × 108 cells) and Imject Alum (Thermo Fisher Scientific Inc.) as an adjuvant, which stimulates a nonspecific immune response for mixed antigens using this formulation of aluminum hydroxide and magnesium hydroxide. After the three additional immunizations per week, a booster injection was performed two days before harvesting the spleen cells of immunized mice. The hybridomas were established by the fusion of splenocytes and P3U1 cells using polyethylene glycol 1500 (PEG1500; Roche Diagnostics, Indianapolis, IN, USA). RPMI-1640 supplemented with hypoxanthine, aminopterin, and thymidine (HAT; Thermo Fisher Scientific Inc.) was used for the selection of hybridomas. The supernatants, which are positive for CHO/CD44v3–10 cells and negative for CHO-K1 cells, were selected by the flow cytometry-based high throughput screening using SA3800 Cell Analyzers (Sony Corp. Tokyo, Japan).
2.4. Enzyme-Linked Immunosorbent Assay (ELISA)
Fifty-eight synthesized peptides, which cover the CD44v3–10 extracellular domain [
24], were synthesized by Sigma-Aldrich Corp. (St. Louis, MO, USA) The peptides (1 µg/mL) were immobilized on Nunc Maxisorp 96-well immunoplates (Thermo Fisher Scientific Inc) for 30 min at 37 °C. After washing with phosphate-buffered saline (PBS) containing 0.05% (
v/v) Tween 20 (PBST; Nacalai Tesque, Inc.) using Microplate Washer, HydroSpeed (Tecan, Zürich, Switzerland), wells were blocked with 1% (
w/v) bovine serum albumin (BSA)-containing PBST for 30 min at 37 °C. C
44Mab-3 (10 µg/mL) were added to each well, and then incubated with anti-mouse immunoglobulins conjugated to peroxidase (1:2000 diluted; Agilent Technologies Inc., Santa Clara, CA, USA). Enzymatic reactions were performed using 1-Step Ultra TMB (Thermo Fisher Scientific Inc.). The optical density (655 nm) was mesured using an iMark microplate reader (Bio-Rad Laboratories, Inc., Berkeley, CA, USA).
2.5. Flow Cytometry
CHO-K1, CHO/CD44v3–10, PK-1, and PK-8 were harvested using 0.25% trypsin and 1 mM ethylenediamine tetraacetic acid (EDTA; Nacalai Tesque, Inc.). The cells were treated with C44Mab-3, C44Mab-46, or blocking buffer (control) (0.1% bovine serum albumin [BSA; Nacalai Tesque, Inc.] in PBS) for 30 min at 4˚C. Then, the cells were treated with Alexa Fluor 488-conjugated anti-mouse IgG (1:2000; Cell Signaling Technology, Inc.) for 30 min at 4˚C. Fluorescence data were collected using the SA3800 Cell Analyzer and analyzed using SA3800 software ver. 2.05 (Sony Corporation).
2.6. Determination of Dissociation Constant (KD) via Flow Cytometry
Serially diluted C44Mab-3 was treated with CHO/CD44v3–10 and PK-1 cells. The cells were further incubated with Alexa Fluor 488-conjugated anti-mouse IgG (1:200). Fluorescence data were collected using BD FACSLyric and analyzed using BD FACSuite software version 1.3 (BD Biosciences). The dissociation constant (KD) was determined by GraphPad Prism 8 (the fitting binding isotherms to built-in one-site binding models; GraphPad Software, Inc., La Jolla, CA, USA).
2.7. Western Blot Analysis
The total cell lysates (10 μg of protein) were subjected to electrophoresis on polyacrylamide gels (5%–20% gel, FUJIFILM Wako Pure Chemical Corporation, Osaka, Japan). The separated proteins were transferred onto polyvinylidene difluoride (PVDF) membranes (Merck KGaA, Darmstadt, Germany). The blocking was performed using 4% skim milk (Nacalai Tesque, Inc.) in PBS with 0.05% Tween 20. The membranes were incubated with 10 μg/mL of C44Mab-3, 10 μg/mL of C44Mab-46, 0.5 μg/mL of NZ-1, or 1 μg/mL of an anti-β-actin mAb (clone AC-15; Sigma-Aldrich Corp.), and then incubated with peroxidase-conjugated anti-mouse immunoglobulins (diluted 1:1000; Agilent Technologies, Inc.) for C44Mab-3, C44Mab-46, and anti-β-actin. The anti-rat immunoglobulins (diluted 1:1000; Agilent Technologies, Inc.) conjugated with peroxidase was used for NZ-1. The chemiluminescence signals were obtained with ImmunoStar LD (FUJIFILM Wako Pure Chemical Corporation), and detected using a Sayaca-Imager (DRC Co. Ltd., Tokyo, Japan).
2.8. Immunohistochemical Analysis
One formalin-fixed paraffin-embedded (FFPE) oral SCC tissue was obtained from Tokyo Medical and Dental University [
48]. FFPE sections of pancreatic carcinoma tissue arrays (Catalog number: PA241c and PA484) were purchased from US Biomax Inc. (Rockville, MD, USA). Pancreas adenocarcinoma tissue microarray with adjacent normal pancreas tissue (PA241c) contains 6 cases of pancreas adenocarcinoma with matched adjacent normal pancreas tissue, quadruple cores per case. Pancreas adenocarcinoma tissue microarray (PA484) contains 18 cases of pancreas adenocarcinoma, 1 each of pancreas islet cell tumor, mucinous adenocarcinoma and undifferentiated carcinoma, plus 3 normal pancreas tissue, duplicate cores per case. One oral SCC tissue was autoclaved in citrate buffer (pH 6.0; Nichirei biosciences, Inc., Tokyo, Japan), and pancreatic carcinoma tissue arrays were autoclaved in EnVision FLEX Target Retrieval Solution High pH (Agilent Technologies, Inc.) for 20 min. After blocking with SuperBlock T20 (Thermo Fisher Scientific, Inc.), the sections were incubated with C
44Mab-3 (1 μg/mL) and C
44Mab-46 (1 μg/mL) for 1 h at room temperature and then treated with the EnVision+ Kit for mouse (Agilent Technologies Inc.) for 30 min. The color was developed using 3,3′-diaminobenzidine tetrahydrochloride (DAB; Agilent Technologies Inc.). Hematoxylin (FUJIFILM Wako Pure Chemical Corporation) was used for the counterstaining. Leica DMD108 (Leica Microsystems GmbH, Wetzlar, Germany) was used to examine the sections and obtain images.
4. Discussion
Pancreatic cancer is the third leading cause of cancer death in United States [
1]. PDAC is the most common type of pancreatic cancer, and exhibits the extremely poor prognosis with a 5-year survival rate of approximately 10% [
49]. Advances in therapy have only achieved incremental improvements in overall outcome, but can provide notable benefit for undefined subgroups of patients. PDACs are heterogenous neoplasms with various histology [
4] and heterogenous molecular landscapes [
5]. Therefore, the identification of early diagnostic markers and therapeutic targets in each group has been desired. In this study, we developed C
44Mab-3 using the CBIS method (
Figure 1), and determined its epitope as variant 5-encoded region of CD44 (
Supplementary Table S1). Then, we showed the usefulness of C
44Mab-3 for multiple applications, including flow cytometry (
Figure 2 and
Figure 3), western blotting (
Figure 4), and immunohistochemistry of PDAC (
Figure 5).
An anti-CD44v5 mAb (clone VFF-8) was previously developed and mainly used for the immunohistochemical analyses of tumors [
50]. The epitope of VFF-8 was determined as IHHEHHEEEETPHSTST in v5-encoded region by ELISA [
51]. As shown in
Supplementary Table S1, C
44Mab-3 recognized both CD44p311–330 and CD44p321–340 peptides, which commonly possess HPPLIHHEHH sequence. The epitope of C
44Mab-3 partially shares that of VFF-8. Further investigation of the detailed epitope mapping is required. In addition, CD44 is known to be heavily glycosylated [
52], and the glycosylation pattern is thought to depend on the host cells. Since the epitope of C
44Mab-3 never contains serine or threonine, the recognition of C
44Mab-3 is thought to be independent of the glycosylation.
Immunohistochemistry using VFF-8 and conventional RT-PCR analyses were performed against PDAC [
50]. VFF-8 recognized PDAC, but not normal pancreas. Furthermore, RT-PCR analysis revealed that the exon v5 appeared in the chain containing at least v4–10 in 80% of PDAC and the cell lines tested. The authors discussed that one of the major differences between normal and PDAC was the linkage of CD44v5 to the CD44v6-containing chain [
50]. Our immunohistochemical analysis also support the finding (
Figure 5A, C, and G). Furthermore, we found that C
44Mab-3 could detect atypical type of PDAC, including metaplastic and diffusely invaded tumor cells (
Figure 5A and C). In contrast, C
44Mab-3 never stained a typical ductal structure of PDAC (
Figure 5E) and normal pancreatic epithelial cells (
Figure 5G). In addition to conventional PDAC, the World Health Organization classified nine histological subtypes of PDAC, which further highlight the morphologic heterogeneity of PDAC [
4]. It is worthwhile to investigate whether CD44v5 is expressed in a specific subtype of PDAC in the future study.
Large scale genomic analyses of PDACs defined 4 subtypes: (1) squamous; (2) pancreatic progenitor; (3) immunogenic; and (4) ADEX, that correlate with histopathological characteristics [
5]. Among them, squamous subtype is characterized as enriched for
TP53 and
KDM6A mutations, upregulation of the ∆Np63 transcriptional network, hypermethylation of pancreatic endodermal determinant genes and have a poor prognosis [
5]. ∆Np63 is known as a marker of basal cells of stratified epithelium and SCC [
53], and also reported to regulate HA metabolism and signaling [
54]. Especially, ΔNp63 directly regulates the expression of CD44 through p63-binding sites, which are located in the promoter region and in the first intron of CD44 gene, respectively [
54]. Therefore, CD44 transcription could be upregulated in ΔNp63-positive PDAC. However, the mechanism of the variant 5 inclusion during alternative splicing remains to be determined.
Clinical trials of anti-pan CD44 and variant-specific CD44 mAbs have been conducted [
55]. An anti-pan CD44 mAb, RG7356, was well tolerated in patients with advanced CD44-positive solid tumors. However, the study was terminated because of no evidence of a clinical and dose-response relationship with RG7356 [
56]. A clinical trial of humanized anti-CD44v6 mAb bivatuzumab−mertansine drug conjugate was conducted. However, it failed due to the severe skin toxicities [
57,
58]. The efficient accumulation of mertansine was most likely responsible for the high toxicity [
57,
58]. Although CD44v5 is never detected in normal pancreatic epithelium by C
44Mab-3 (this study) and VFF-8 [
50], CD44v5 could be detected in normal lung, skin, gastric, and bladder epithelium by VFF-8 [
51]. For the development of therapeutic use of C
44Mab-3, further investigations are required to reduce the toxicity to above tissues.
Our group converted mouse IgG
1 subclass of mAbs into a IgG
2a, and produced a defucosylated mAbs using fucosyltransferases 8-deficient CHO-K1 cells. The defucosylated IgG
2a mAbs showed potent antibody-dependent cellular cytotoxicity
in vitro, and suppressed the tumor xenograft growth [
27,
59,
60,
61,
62,
63,
64,
65]. Therefore, the production of a class-switched and defucosylated version of C
44Mab-3 is required to evaluate the antitumor activity
in vivo.
Figure 1.
A schematic illustration of ant-human CD44 mAbs production. (A) CHO/CD44v3–10 cells were intraperitoneally injected into BALB/c mice. (B) Hybridomas were produced by fusion of the splenocytes and P3U1 cells (C) The screening was performed by flow cytometry using parental CHO-K1 and CHO/CD44v3–10 cells. (D) After cloning and additional screening, a clone C44Mab-3 (IgG1, kappa) was established. Furthermore, the binding epitopes were determined by enzyme-linked immunosorbent assay (ELISA) using peptides, which cover the extracellular domain of CD44v3–10.
Figure 1.
A schematic illustration of ant-human CD44 mAbs production. (A) CHO/CD44v3–10 cells were intraperitoneally injected into BALB/c mice. (B) Hybridomas were produced by fusion of the splenocytes and P3U1 cells (C) The screening was performed by flow cytometry using parental CHO-K1 and CHO/CD44v3–10 cells. (D) After cloning and additional screening, a clone C44Mab-3 (IgG1, kappa) was established. Furthermore, the binding epitopes were determined by enzyme-linked immunosorbent assay (ELISA) using peptides, which cover the extracellular domain of CD44v3–10.
Figure 2.
Flow cytometry using C44Mab-3 against CD44-expressing cells. CHO/CD44v3–10 (A), CHO/CD44s (B), CHO-K1 (C), PK-1 (D), and PK-8 (E) cells were treated with 0.01-10 µg/mL of C44Mab-3, followed by treatment with Alexa Fluor 488-conjugated anti-mouse IgG (Red line). The black line represents the negative control (blocking buffer).
Figure 2.
Flow cytometry using C44Mab-3 against CD44-expressing cells. CHO/CD44v3–10 (A), CHO/CD44s (B), CHO-K1 (C), PK-1 (D), and PK-8 (E) cells were treated with 0.01-10 µg/mL of C44Mab-3, followed by treatment with Alexa Fluor 488-conjugated anti-mouse IgG (Red line). The black line represents the negative control (blocking buffer).
Figure 3.
The binding affinity of C44Mab-3 to CD44-expressing cells. CHO/CD44v3–10 (A) and PK-1 (B) cells were suspended in 100 µL of serially diluted C44Mab-3 (0.0006–10 µg/mL). The cells were incubated with Alexa Fluor 488-conjugated anti-mouse IgG. The apparent dissociation constant (KD) was measured by GraphPad PRISM 8 using the fluorescence data.
Figure 3.
The binding affinity of C44Mab-3 to CD44-expressing cells. CHO/CD44v3–10 (A) and PK-1 (B) cells were suspended in 100 µL of serially diluted C44Mab-3 (0.0006–10 µg/mL). The cells were incubated with Alexa Fluor 488-conjugated anti-mouse IgG. The apparent dissociation constant (KD) was measured by GraphPad PRISM 8 using the fluorescence data.
Figure 4.
Western blot analysis using C44Mab-3. The cell lysates of CHO-K1, CHO/CD44s, CHO/CD44v3–10, PK-1, and PK-8 (10 µg) were separated and transferred onto polyvinylidene fluoride membranes. The membranes were incubated with 10 µg/mL of C44Mab-46 (A), 10 µg/mL of C44Mab-3 (B), 0.5 µg/mL of anti-PA16 tag mAb (NZ-1) (C), and 1 µg/mL of an anti-β-actin mAb (D). Then, the membranes were incubated with anti-mouse immunoglobulins conjugated with peroxidase for C44Mab-46, C44Mab-3, and anti-β-actin. The anti-rat immunoglobulins conjugated with peroxidase was used for NZ-1. The red arrows indicate the CD44s (~75 kDa). The black arrows indicate the CD44v3–10 or CD44v5 (>180 kDa).
Figure 4.
Western blot analysis using C44Mab-3. The cell lysates of CHO-K1, CHO/CD44s, CHO/CD44v3–10, PK-1, and PK-8 (10 µg) were separated and transferred onto polyvinylidene fluoride membranes. The membranes were incubated with 10 µg/mL of C44Mab-46 (A), 10 µg/mL of C44Mab-3 (B), 0.5 µg/mL of anti-PA16 tag mAb (NZ-1) (C), and 1 µg/mL of an anti-β-actin mAb (D). Then, the membranes were incubated with anti-mouse immunoglobulins conjugated with peroxidase for C44Mab-46, C44Mab-3, and anti-β-actin. The anti-rat immunoglobulins conjugated with peroxidase was used for NZ-1. The red arrows indicate the CD44s (~75 kDa). The black arrows indicate the CD44v3–10 or CD44v5 (>180 kDa).
Figure 5.
Immunohistochemical analysis using C44Mab-3 and C44Mab-46 against pancreatic adenocarcinomas and normal pancreatic tissues. After antigen retrieval, serial sections of pancreatic carcinoma tissue arrays (Catalog number: PA484) were incubated with 1 µg/mL of C44Mab-3 or C44Mab-46 followed by treatment with the Envision+ kit. The color was developed using 3,3’-diaminobenzidine tetrahydrochloride (DAB), and the sections were counterstained with hematoxylin. Scale bar = 100 µm. (A–F) pancreatic adenocarcinomas; (G, H) normal pancreas tissues.
Figure 5.
Immunohistochemical analysis using C44Mab-3 and C44Mab-46 against pancreatic adenocarcinomas and normal pancreatic tissues. After antigen retrieval, serial sections of pancreatic carcinoma tissue arrays (Catalog number: PA484) were incubated with 1 µg/mL of C44Mab-3 or C44Mab-46 followed by treatment with the Envision+ kit. The color was developed using 3,3’-diaminobenzidine tetrahydrochloride (DAB), and the sections were counterstained with hematoxylin. Scale bar = 100 µm. (A–F) pancreatic adenocarcinomas; (G, H) normal pancreas tissues.
Table 1.
Immunohistochemical analysis using C44Mab-3 and C44Mab-46 against pancreatic carcinoma tissue arrays.
Table 1.
Immunohistochemical analysis using C44Mab-3 and C44Mab-46 against pancreatic carcinoma tissue arrays.
Tissue array |
Age |
Sex |
Organ |
Pathology diagnosis |
TNM |
Grade |
Stage |
Type |
C44Mab-3 |
PA241c |
66 |
F |
Pancreas |
Adenocarcinoma |
T2N0M0 |
1 |
I |
malignant |
+ |
|
66 |
F |
Pancreas |
Adjacent normal pancreas tissue |
|
|
|
|
– |
|
54 |
F |
Pancreas |
Adenocarcinoma |
T3N0M0 |
2 |
II |
malignant |
– |
|
54 |
F |
Pancreas |
Adjacent normal pancreas tissue |
|
|
|
|
– |
|
44 |
M |
Pancreas |
Adenocarcinoma |
T3N0M0 |
2 |
II |
malignant |
– |
|
44 |
M |
Pancreas |
Adjacent normal pancreas tissue |
|
|
|
|
– |
|
59 |
M |
Pancreas |
Adenocarcinoma |
T2N0M0 |
3 |
I |
malignant |
– |
|
59 |
M |
Pancreas |
Adjacent normal pancreas tissue |
|
|
|
|
– |
|
63 |
F |
Pancreas |
Adenocarcinoma |
T2N0M0 |
3 |
I |
malignant |
+ |
|
63 |
F |
Pancreas |
Adjacent normal pancreas tissue |
|
|
|
|
– |
|
53 |
F |
Pancreas |
Adenocarcinoma |
T3N0M0 |
3 |
II |
malignant |
– |
|
53 |
F |
Pancreas |
Adjacent normal pancreas tissue |
|
|
|
|
– |
PA484 |
35 |
M |
Pancreas |
Normal pancreas tissue |
- |
- |
- |
normal |
– |
|
38 |
F |
Pancreas |
Normal pancreas tissue |
- |
- |
- |
normal |
– |
|
38 |
M |
Pancreas |
Normal pancreas tissue |
- |
- |
- |
normal |
– |
|
60 |
M |
Pancreas |
Adenocarcinoma |
T3N0M0 |
2 |
II |
malignant |
– |
|
68 |
F |
Pancreas |
Adenocarcinoma |
T2N0M0 |
2 |
I |
malignant |
+ |
|
54 |
F |
Pancreas |
Adenocarcinoma |
T3N0M0 |
2 |
II |
malignant |
– |
|
42 |
F |
Pancreas |
Adenocarcinoma |
T3N0M0 |
2 |
II |
malignant |
– |
|
65 |
M |
Pancreas |
Adenocarcinoma |
T3N0M0 |
2 |
II |
malignant |
– |
|
75 |
F |
Pancreas |
Adenocarcinoma |
T3N0M1 |
2 |
IV |
malignant |
– |
|
57 |
M |
Pancreas |
Adenocarcinoma |
T3N0M0 |
3 |
II |
malignant |
+ |
|
44 |
M |
Pancreas |
Adenocarcinoma |
T3N0M0 |
3 |
II |
malignant |
– |
|
47 |
M |
Pancreas |
Adenocarcinoma |
T3N0M0 |
- |
II |
malignant |
– |
|
41 |
M |
Pancreas |
Adenocarcinoma |
T4N1M0 |
2 |
III |
malignant |
– |
|
64 |
F |
Pancreas |
Adenocarcinoma |
T3N0M0 |
2 |
II |
malignant |
– |
|
58 |
F |
Pancreas |
Adenocarcinoma |
T3N0M0 |
3 |
II |
malignant |
– |
|
47 |
F |
Pancreas |
Adenocarcinoma |
T3N1M0 |
3 |
III |
malignant |
+ |
|
78 |
M |
Pancreas |
Adenocarcinoma |
T2N0M0 |
3 |
I |
malignant |
+ |
|
49 |
M |
Pancreas |
Adenocarcinoma |
T3N0M0 |
2 |
II |
malignant |
+ |
|
53 |
F |
Pancreas |
Adenocarcinoma |
T3N0M0 |
3 |
II |
malignant |
+ |
|
60 |
M |
Pancreas |
Adenocarcinoma |
T2N0M0 |
3 |
I |
malignant |
+ |
|
57 |
F |
Pancreas |
Adenocarcinoma |
T2N0M0 |
3 |
I |
malignant |
– |
|
61 |
M |
Pancreas |
Mucinous adenocarcinoma |
T3N0M1 |
2 |
IV |
malignant |
– |
|
69 |
M |
Pancreas |
Undifferentiated carcinoma |
T2N0M0 |
- |
I |
malignant |
+ |