1. Introduction
Gastric cancer (GC) is the third leading cause of cancer-related deaths globally [
1]. The GC incidence is higher in Eastern Asia than in Western countries [
1]. The vast majority of GC are adenocarcinomas, which can be divided into intestinal-type gastric cancer (IGC), diffuse-type gastric cancer (DGC), and mixed histology according to the Lauren classification [
2]. The World Health Organization classifies gastric adenocarcinomas into papillary, tubular, mucinous, and poorly cohesive carcinomas [
3]. Furthermore, next-generation sequencing defined four molecular subtypes, including Epstein–Barr virus-positive, microsatellite instability, genomically stable, and chromosomally unstable types [
4]. The analysis also revealed the alterations in the GC genome and provided treatment options with anti-human epidermal growth factor receptor 2 (HER2) therapy [
5] or immune checkpoint inhibitor therapy [
6]. However, the benefit of those therapies is limited to a small subset of patients. In patients with advanced GC, especially those with DGC, peritoneal metastasis and subsequent development of malignant ascites are the most frequent cause of death. Tanaka
et al., therefore, performed a comprehensive multi-omic analysis of malignant ascitic samples and their corresponding tumor cell lines [
7]. They identified the genomic alterations and significant amplification of known cancer driver genes, such as
KRAS,
FGFR2,
MET,
ERBB2,
EGFR, MYC,
CCND1, and
CD44 in GC with peritoneal metastasis [
7]. Among them, the cell surface antigens (FGFR2, MET, HER2, EGFR, and CD44) are potentially treatable with monoclonal antibody (mAb) therapy. Compared to the first four antigens, mAb therapy or diagnosis against CD44 has not been established.
CD44 plays important roles in the tumor progression and has various isoforms, which are generated by the alternative splicing of CD44 mRNA [
8]. The mRNA of CD44 standard (CD44s) isoform is produced by constant region exons including the first five (1 to 5) and the last five (16 to 20) [
9]. The mRNAs of CD44 variant (CD44v) isoform are produced by the assembling of variant exons (v1–v10) with the constant region exons of CD44s [
10]. CD44s and CD44v receive the post-translational modifications, such as
N-glycosylation and
O-glycosylation. Therefore, the molecular weight of CD44s is reached 80~100 kDa, and CD44v has various molecular weights (100~250 kDa) due to a variety of glycosylation [
11]. Both CD44s and CD44v (pan-CD44) can attach to hyaluronic acid (HA), which is important for cellular adhesion, homing, and motility [
12].
CD44v is overexpressed in tumors [
13] and promotes tumor malignant progression through the binding to growth factors, and the acquisition of invasiveness, stemness, and drug resistance [
14,
15,
16]. These were mediated by the unique functions of the variant’s exon-encoded region. The v3-encoded region can recruit heparin-binding growth factors to their receptor, and promote the signal transduction [
17,
18]. The v6-encoded region forms a ternary complex with hepatocyte growth factor and its receptor MET, which is essential for the activation [
19]. Furthermore, the v8–10-encoded region binds to and stabilizes a cystine–glutamate transporter (xCT), which enhances cystine uptake and glutathione synthesis [
20]. The elevation of reduced glutathione (GSH) mediates the defense to reactive oxygen species (ROS) [
20] and chemotherapeutic drugs [
21]. The expression of CD44v8–10 is associated with the function of xCT and intracellular redox status, which is associated with the poor prognosis [
22]. Therefore, the establishment of CD44v-specific mAbs are essential for CD44-targeting tumor diagnosis and therapy. However, the roles of the variant 8-encoded region in tumor development have not been fully elucidated.
Our group previously established an anti-pan-CD44 mAb, C
44Mab-5 (IgG
1, kappa) [
23] using the Cell-Based Immunization and Screening (CBIS) method. Moreover, another anti-pan-CD44 mAb, C
44Mab-46 [
24] was developed by immunizing mice with CD44v3–10 ectodomain. Both C
44Mab-5 and C
44Mab-46 have epitopes within the standard exon 2 and 5-encoding regions, respectively [
25,
26,
27]. We further showed that both C
44Mab-5 and C
44Mab-46 are applicable to flow cytometry and immunohistochemical analyses in oral squamous cell carcinomas (OSCC) [
23] and esophageal SCC [
24]. Furthermore, we produced a class-switched and a defucosylated version of C
44Mab-5 (5-mG
2a-f) using fucosyltransferase 8-deficient ExpiCHO-S cells and evaluated the antitumor effects of 5-mG
2a-f in OSCC xenograft bearing mice [
28]. We have developed various anti-CD44v mAbs, including anti-CD44v4 (C
44Mab-108) [
29], anti-CD44v5 (C
44Mab-3) [
30], anti-CD44v6 (C
44Mab-9) [
31], anti-CD44v7/8 (C
44Mab-34) [
32], and anti-CD44v9 (C
44Mab-1) [
33].
In this study, we established a novel anti-CD44v8 mAb, C44Mab-94 (IgG1, kappa) using the CBIS method and evaluated its applications.
2. Materials and Methods
2.1. Cell Lines
The human OSCC cell line (HSC-3) and the human gastric cancer cell lines (MKN45 and NUGC-4) were obtained from the Japanese Collection of Research Bioresources (Osaka, Japan). The human pancreatic cancer cell line (PANC-1) was obtained from the Cell Resource Center for Biomedical Research Institute of Development, Aging, and Cancer at Tohoku University (Sendai, Japan). Chinese hamster ovary (CHO)-K1 and P3X63Ag8U.1 (P3U1; a mouse multiple myeloma) cell lines were obtained from the American Type Culture Collection (ATCC, Manassas, VA, USA). HSC-3 was cultured in DMEM medium (Nacalai Tesque, Inc., Kyoto, Japan), supplemented with 100 μg/mL streptomycin, 100 U/mL penicillin, and 0.25 μg/mL amphotericin B (Nacalai Tesque, Inc.), and 10% (v/v) heat-inactivated fetal bovine serum (FBS; Thermo Fisher Scientific, Inc., Waltham, MA, USA). The cell lines (MKN45, NUGC-4, PANC-1, CHO-K1, and P3U1) were cultured in RPMI-1640 medium (Nacalai Tesque, Inc.), supplemented as indicated above. All cells were cultured using a humidified incubator at 37°C, in an atmosphere of 5% CO2 and 95% air.
2.2. Construction of plasmid DNA and Establishment of Stable Transfectants
The cDNAs of CD44s and CD44v3–10 were obtained as described previously [
23]. The cDNAs were cloned into pCAG-ssPA16 vectors with a signal sequence and N-terminal PA16 tag (GLEGGVAMPGAEDDVV) [
23,
34,
35,
36,
37], which can be detected by NZ-1 mAb, (anti-human podoplanin [PDPN] mAb) [
38,
39,
40,
41,
42,
43,
44,
45,
46,
47,
48,
49,
50,
51,
52,
53]. N-terminal PA16-tagged CD44v3–10 deletion mutants (dN224, dN266, dN304, dN343, dN386, dN430, dN464, dN494, and dN562) were amplified using a HotStar HiFidelity Polymerase Kit (Qiagen Inc., Hilden, Germany), and subcloned into the pCAG-ssPA16 vector. The pCAG- ssPA16-CD44s, pCAG-ssPA16-CD44v3–10, and pCAG-ssPA16-CD44v3–10 deletion mutant vectors were transfected into CHO-K1 cells. The pCAG-ssPA16-CD44v3–10 vector was transfected into PANC-1 cells. The transfection was performed using a Neon transfection system (Thermo Fisher Scientific, Inc.). By the limiting dilution method, stable transfectants PANC-1/CD44v3–10, CHO/CD44s, CHO/CD44v3–10, and several deletion mutants of CHO/CD44v3–10 (dN224, dN266, dN304, dN343, dN386, dN430, dN464, dN494, and dN562) were finally established.
2.3. Production of Hybridomas
The 6-week-old female BALB/c mice (CLEA Japan, Tokyo, Japan) were intraperitoneally immunized with PANC-1/CD44v3–10 (1 × 10
8 cells) and Imject Alum (Thermo Fisher Scientific Inc.). Additional immunizations of PANC-1/CD44v3–10 (1 × 10
8 cells, three times) and a booster injection of PANC-1/CD44v3–10 (1 × 10
8 cells) 2 days before the sacrifice were performed. Hybridomas were produced as described previously [
30]. The supernatants, which are positive for CHO/CD44v3–10 cells and negative for CHO-K1 cells, were selected by flow cytometry, SA3800 Cell Analyzers (Sony Corp. Tokyo, Japan).
2.4. Enzyme-Linked Immunosorbent Assay (ELISA)
Four peptides, covering from v7, v8, and v9 regions of CD44v3–10, were obtained from Sigma-Aldrich Corp. (St. Louis, MO, USA).
The peptide sequences were as follows.
CD44p421–440 (GHQAGRRMDMDSSHSTTLQP); v7/v8,
CD44p431–450 (DSSHSTTLQPTANPNTGLVE); v8,
CD44p441–460 (TANPNTGLVEDLDRTGPLSM); v8,
CD44p451–470 (DLDRTGPLSMTTQQSNSQSF); v8/v9.
The peptides (10 µg/mL) were immobilized on 96-well immunoplates (Nunc Maxisorp; Thermo Fisher Scientific Inc). The blocking was performed with 1% (
w/v) bovine serum albumin (BSA) in PBST. C
44Mab-94 (10 µg/mL) or blocking buffer was added to the peptides-coated wells. The detection was performed as described previously [
30].
2.5. Flow Cytometry
In the CBIS screening and epitope mapping, the hybridoma supernatants were treated with CHO-K1, CHO/CD44v3–10, or CHO/CD44v3–10 deletion mutants. In the dose-dependent assay, CHO-K1, CHO/CD44s, CHO/CD44v3–10, MKN45, and NUGC-4 were incubated with C44Mab-94, C44Mab-46, or control blocking buffer (0.1% BSA in PBS). Then, the cells were treated with anti-mouse IgG conjugated with Alexa Fluor 488 (1:2000; Cell Signaling Technology, Inc., Danvers, MA, USA). The data were analyzed using the EC800 Cell Analyzer or the SA3800 Cell Analyzer (Sony Corp.).
2.6. Determination of Dissociation Constant (KD) by Flow Cytometry
The diluted C44Mab-94 (from 1300 to 0.08 nM) was suspended with CHO/CD44v3–10, MKN45, and NUGC-4 cells. Then, the cells were treated with anti-mouse IgG conjugated with Alexa Fluor 488 (1:200). Fluorescence data were analyzed and the apparent KD was determined by the fitting binding isotherms to built-in one-site binding models of GraphPad Prism 8 (GraphPad Software, Inc., La Jolla, CA, USA).
2.7. Western Blot Analysis
Cell lysates were prepared as described previously [
33], and were denatured in sodium dodecyl sulfate (SDS) sample buffer (Nacalai Tesque, Inc.). The 10 μg of proteins were subjected to electrophoresis using polyacrylamide gels (5–20%; FUJIFILM Wako Pure Chemical Corporation, Osaka, Japan) and transferred onto polyvinylidene difluoride membranes (Merck KGaA, Darmstadt, Germany). The membranes were blocked with 4% skim milk (Nacalai Tesque, Inc.) in PBST, and were incubated with 10 μg/mL of C
44Mab-94, 10 μg/mL of C
44Mab-46, or 0.5 μg/mL of an anti-β-actin mAb (clone AC-15; Sigma-Aldrich Corp.). The detection was performed as described previously [
31].
2.8. Immunohistochemical Analysis
Formalin-fixed paraffin-embedded (FFPE) gastric carcinoma (BS01012e and BS01011b) and OSCC (OR601c) tissue arrays were obtained from US Biomax Inc. (Rockville, MD, USA). The tissue arrays were autoclaved in citrate buffer (pH 6.0; Nichirei Biosciences, Inc., Tokyo, Japan) for 20 min. The blocking was performed using SuperBlock T20 (Thermo Fisher Scientific, Inc.). The sections were incubated with C
44Mab-94 (5 μg/mL) and C
44Mab-46 (5 μg/mL). The detection was performed as described previously [
30].
4. Discussion
The VFF series anti-CD44v mAbs were previously established by the immunization of bacterial-expressed CD44v3–10 and glutathione
S-transferase fusion protein [
56,
57]. The clones, VFF-8 (v5), VFF-18 (v6), VFF-9 (v7), VFF-18(v7/8), and VFF14 (v10) have been used for various applications [
58]. Furthermore, VFF18 was humanized as BIWA-4 [
59], and developed to bivatuzumab-mertansine, an antibody-drug conjugate (ADC), for clinical trials [
60,
61]. An anti-CD44v3 mAb (clone 3G5) [
62] and an anti-CD44v9 mAb (clone RV3) [
20] were also developed and widely used for researches. However, a CD44v8-specific mAb has not been developed.
In this study, we developed a novel anti-CD44v8 mAb, C
44Mab-94 using the CBIS method (
Figure 1). We determined the epitope as a v8-encoded region using deletion mutants of CD44 (
Figure 2), and synthetic peptides (
Figure 3). We have established anti-CD44 mAbs using CHO/CD44v3–10 [
23,
30,
31,
33], PANC-1/CD44v3–10 (in this study), or purified CD44v3–10 ectodomain [
24,
32] as antigens. We listed them in our original "Antibody Bank" (see
Supplementary Materials). However, clones which recognize the v8-encoded region were rare, suggesting that the region has low antigenicity and/or locates the inside of CD44v3–10 protein. Although the affinity of C
44Mab-94 is low against target cells, C
44Mab-94 can be applied to various applications, including flow cytometry (
Figure 4), western blotting (
Figure 5), and immunohistochemistry (
Figure 6).
We confirmed that C
44Mab-94 recognizes a synthetic peptide of v8-encoded region (DSSHSTTLQPTANPNTGLVE), but not border regions (v7/v8 and v8/v9) by ELISA (
Figure 3). The epitope region possesses multiple confirmed and predicted
O-glycosylation sites [
63]. C
44Mab-94 recognized a ~75-kDa band in the lysate of CHO/CD44v3–10 (
Figure 5A), which is similar to the predicted molecular size from the amino acids of CD44v3–10. Therefore, C
44Mab-94 could recognize CD44v3–10 regardless of the glycosylation. The detailed epitope analysis and the influence of glycosylation on C
44Mab-94 recognition are requied in future studies.
In a GC cell line, the major transcripts of CD44v, including CD44v3, 8–10, CD44v6–10, CD44v8–10, and CD44v3, 8 were identified [
54] (
Figure 1A). C
44Mab-94 can cover all products of the transcripts, and detect the broad CD44v-expressing GC. Since CD44 v8–10 plays critical roles in the regulation of ROS defense and GC progression [
20], an anti-CD44v9 mAb (clone RV3) was mainly used in immunohistochemistry to date. Several studies revealed that CD44v9 is a predictive marker for the recurrence of GC [
64] and a biomarker for GC patient selection and efficacy of xCT inhibitors, sulfasalazine [
65]. Further investigations are required to reveal the relationship between CD44v8 expression and clinical factors using C
44Mab-94. Additionally, C
44Mab-94 recognized both IGC (
Figure 6A) and DGC (
Figure 6E) in immunohistochemistry. It is worthwhile to investigate whether CD44v8 is expressed in a specific molecular subtype of GC [
4] in the future study.
A comprehensive analysis of malignant ascites identified the amplifications of cancer driver genes including
CD44 [
7]. Although the expression pattern of CD44v is not identified, CD44v8 is thought to be an important target for mAb therapy due to the commonly included region in GC [
54]. We have shown the antitumor activity using class-switched and defucosylated IgG
2a recombinant mAbs [
28,
66,
67,
68,
69,
70,
71,
72]. The defucosylated IgG
2a mAbs can be produced by CHO-K1 lacking fucosyltransferases 8, and exhibited potent ADCC activity
in vitro, and suppressed the growth of xenograft [
28,
66,
67,
68,
69,
70,
71,
72]. Therefore, the production of defucosylated C
44Mab-94 is one of the strategies to evaluate the antitumor effect on GC with peritoneal metastasis in the preclinical model.
Clinical applications of a humanized anti-CD44v6 mAb (BIWA-4) bivatuzumab−mertansine drug conjugate to solid tumors failed because of the skin toxicities [
60,
61]. The accumulation of mertansine drug was thought to be a cause of the toxicity [
60,
61]. Human acute myeloid leukemia (AML) cells also express high levels of CD44 mRNA due to suppression of CpG islands methylation in the promoter [
73]. Furthermore, higher expression of CD44v6 was observed in AML patients with
FLT3 or
DNMT3A mutations. Therefore, a mutated version of BIWA-4, called BIWA-8 was engineered to develop chimeric antigen receptors (CARs) for AML. The CD44v6 CAR-T cells exhibited potent anti-leukemic effects [
73], indicating that CD44v6 is a rational target of CAR-T therapy for AML harboring
FLT3 or
DNMT3A mutations. Additionally, the CD44v6 CAR-T also showed an antitumor effect in lung and ovarian cancer xenograft models [
74], which is expected for a wider development toward solid tumors.
Because CD44 mRNA is elevated in AML, other CD44 variants could be transcribed in AML. Furthermore, CD44v8–10 was elevated during chronic myeloid leukemia (CML) progression from chronic phase to blast crisis in a humanized mouse model, which is required for the maintenance of stemness of CML [
75]. Therefore, we will investigate the reactivity of C
44Mab-94 against hematopoietic malignancy. Further studies are required to investigate the selective expression of CD44v8 in leukemia cells, but not in hematopoietic stem cells to certify its safety as a CAR-T antigen.
In this study, we used tumor cell-expressed CD44v3–10 as an immunogen. This strategy is important for the establishment of cancer-specific mAbs (CasMabs). We previously developed PDPN-targeting CasMabs [
76,
77,
78,
79] and podocalyxin-targeting CasMabs [
80], which recognize cancer-type aberrant glycosylation of the targets [
81]. Anti-PDPN-CasMabs are currently applied to CAR-T therapy in preclinical models [
48,
82,
83]. For CasMab development, we should do further screening of our established anti-CD44 mAbs by comparing the reactivity against normal cells. Anti-CD44 CasMabs could be applicable for designing the modalities including ADCs and CAR-T.
Figure 1.
Anti-human CD44 mAbs production. (
A) The structure of CD44s and CD44v. The mRNA of CD44s is assembled by the constant exons (1–5) and (16–20), and produces the standard isoform, CD44s. The mRNAs of CD44v are generated by the alternative splicing of variant exons. CD44v3–10 is an immunogen. CD44v3, 8–10, CD44v6–10, CD44v8–10, and CD44v3, 8 are detected in GC cell line [
54] (
B) PANC1/CD44v3–10 was used as an immunogen. (
C) The hybridomas were produced by fusion with splenocytes and P3U1 cells. (
D) The screening was performed using parental CHO-K1 and CHO/CD44v3–10 cells by flow cytometry. (
E) A clone C
44Mab-94 was established. Furthermore, the binding epitope was determined by flow cytometry using CD44 deletion mutant-expressed CHO-K1 cells and ELISA.
Figure 1.
Anti-human CD44 mAbs production. (
A) The structure of CD44s and CD44v. The mRNA of CD44s is assembled by the constant exons (1–5) and (16–20), and produces the standard isoform, CD44s. The mRNAs of CD44v are generated by the alternative splicing of variant exons. CD44v3–10 is an immunogen. CD44v3, 8–10, CD44v6–10, CD44v8–10, and CD44v3, 8 are detected in GC cell line [
54] (
B) PANC1/CD44v3–10 was used as an immunogen. (
C) The hybridomas were produced by fusion with splenocytes and P3U1 cells. (
D) The screening was performed using parental CHO-K1 and CHO/CD44v3–10 cells by flow cytometry. (
E) A clone C
44Mab-94 was established. Furthermore, the binding epitope was determined by flow cytometry using CD44 deletion mutant-expressed CHO-K1 cells and ELISA.
Figure 2.
Epitope determination of C44Mab-94 using deletion mutants of CD44v3–10. (A) The CD44v3–10 deletion mutants-expressed on CHO-K1 cells. (B) The CD44v3–10 mutants-expressed CHO-K1 cells were incubated with C44Mab-94 (B, red line) or an anti-PA tag mAb, NZ-1 (C, red line), followed by secondary antibodies. The black line represents the negative control (blocking buffer).
Figure 2.
Epitope determination of C44Mab-94 using deletion mutants of CD44v3–10. (A) The CD44v3–10 deletion mutants-expressed on CHO-K1 cells. (B) The CD44v3–10 mutants-expressed CHO-K1 cells were incubated with C44Mab-94 (B, red line) or an anti-PA tag mAb, NZ-1 (C, red line), followed by secondary antibodies. The black line represents the negative control (blocking buffer).
Figure 3.
Determination of C44Mab-94 epitope by ELISA. Four synthesized peptides, which cover the CD44v7 to v9 region, were immobilized on immunoplates. The plates were incubated with C44Mab-94 or buffer control (–1st Ab), followed by incubation with peroxidase-conjugated anti-mouse immunoglobulins. Optical density was measured at 655 nm. NC, negative control (solvent; DMSO in PBS). Error bars represent means ± SDs.
Figure 3.
Determination of C44Mab-94 epitope by ELISA. Four synthesized peptides, which cover the CD44v7 to v9 region, were immobilized on immunoplates. The plates were incubated with C44Mab-94 or buffer control (–1st Ab), followed by incubation with peroxidase-conjugated anti-mouse immunoglobulins. Optical density was measured at 655 nm. NC, negative control (solvent; DMSO in PBS). Error bars represent means ± SDs.
Figure 4.
Flow cytometric analysis of C44Mab-94. CHO/CD44v3–10 (A), CHO/CD44s (B), CHO-K1 (C), HSC-3 (D), MKN45 (E), and NUGC-4 (F) cells were incubated with 0.01–10 µg/mL of C44Mab-94. Then, the cells were treated with anti-mouse IgG conjugated with Alexa Fluor 488 (Red line). The black line represents the negative control (blocking buffer).
Figure 4.
Flow cytometric analysis of C44Mab-94. CHO/CD44v3–10 (A), CHO/CD44s (B), CHO-K1 (C), HSC-3 (D), MKN45 (E), and NUGC-4 (F) cells were incubated with 0.01–10 µg/mL of C44Mab-94. Then, the cells were treated with anti-mouse IgG conjugated with Alexa Fluor 488 (Red line). The black line represents the negative control (blocking buffer).
Figure 5.
Western blot analysis using C44Mab-94. The cell lysates of CHO-K1, CHO/CD44s, CHO/CD44v3–10, HSC-3, MKN45, and NUGC-4 (10 µg) were electrophoresed and transferred onto polyvinylidene fluoride membranes. The membranes were incubated with 10 µg/mL of C44Mab-94 (A), 10 µg/mL of C44Mab-46 (B), and 0.5 µg/mL of an anti-β-actin mAb (C). Then, the membranes were incubated with anti-mouse immunoglobulins conjugated with peroxidase. The black arrows indicate CD44s (~75 kDa). The red arrows indicate CD44v3–10 (>180 kDa). CD44v8 was broadly detected in HSC-3, MKN45, and NUGC-4 lysates. The white arrow indicates ~75-kDa band recognized by C44Mab-94 in CHO/CD44v3–10 lysate.
Figure 5.
Western blot analysis using C44Mab-94. The cell lysates of CHO-K1, CHO/CD44s, CHO/CD44v3–10, HSC-3, MKN45, and NUGC-4 (10 µg) were electrophoresed and transferred onto polyvinylidene fluoride membranes. The membranes were incubated with 10 µg/mL of C44Mab-94 (A), 10 µg/mL of C44Mab-46 (B), and 0.5 µg/mL of an anti-β-actin mAb (C). Then, the membranes were incubated with anti-mouse immunoglobulins conjugated with peroxidase. The black arrows indicate CD44s (~75 kDa). The red arrows indicate CD44v3–10 (>180 kDa). CD44v8 was broadly detected in HSC-3, MKN45, and NUGC-4 lysates. The white arrow indicates ~75-kDa band recognized by C44Mab-94 in CHO/CD44v3–10 lysate.
Figure 6.
Immunohistochemistry using C44Mab-94 and C44Mab-46 against GC. (A–F) The serial sections of GC tissue arrays (BS01011b) were treated with 5 µg/mL of C44Mab-94 or 5 µg/mL of C44Mab-46, followed by treatment with the Envision+ kit. The chromogenic reaction was performed using DAB, and the sections were counterstained with hematoxylin. Scale bar = 100 µm.
Figure 6.
Immunohistochemistry using C44Mab-94 and C44Mab-46 against GC. (A–F) The serial sections of GC tissue arrays (BS01011b) were treated with 5 µg/mL of C44Mab-94 or 5 µg/mL of C44Mab-46, followed by treatment with the Envision+ kit. The chromogenic reaction was performed using DAB, and the sections were counterstained with hematoxylin. Scale bar = 100 µm.
Table 1.
Immunohistochemical analysis using C44Mab-94 and C44Mab-46 against GC tissue array (BS01011b).
Table 1.
Immunohistochemical analysis using C44Mab-94 and C44Mab-46 against GC tissue array (BS01011b).
No. |
Age |
Sex |
Pathology diagnosis |
TNM |
Grade |
Stage |
C44Mab-94 |
C44Mab-46 |
1 |
55 |
F |
Adenocarcinoma |
T2N0M0 |
1 |
IB |
- |
+ |
2 |
51 |
F |
Adenocarcinoma |
T2N0M0 |
- |
IB |
- |
- |
3 |
71 |
M |
Adenocarcinoma |
T3N1M0 |
1 |
IIB |
- |
++ |
4 |
63 |
M |
Adenocarcinoma |
T3N0M0 |
1 |
IIA |
- |
- |
5 |
61 |
M |
Adenocarcinoma |
T2N0M0 |
1 |
IB |
- |
- |
6 |
61 |
M |
Adenocarcinoma |
T2N0M0 |
1 |
IB |
- |
+ |
7 |
60 |
M |
Adenocarcinoma |
T3N2M0 |
1 |
IIIA |
- |
- |
8 |
54 |
M |
Adenocarcinoma |
T3N2M0 |
1 |
IIIA |
+ |
++ |
9 |
46 |
F |
Adenocarcinoma |
T3N0M0 |
1 |
IIA |
- |
+ |
10 |
66 |
M |
Mucinous adenocarcinoma |
T3N0M0 |
2-3 |
IIA |
- |
+ |
11 |
56 |
M |
Adenocarcinoma |
T2N0M0 |
2 |
IB |
++ |
+ |
12 |
52 |
F |
Adenocarcinoma |
T3N0M0 |
2 |
IIA |
+ |
+ |
13 |
70 |
M |
Adenocarcinoma |
T3N0M0 |
2 |
IIA |
- |
+ |
14 |
71 |
M |
Adenocarcinoma |
T2N0M0 |
2 |
IB |
- |
++ |
15 |
61 |
M |
Adenocarcinoma |
T3N0M0 |
2 |
IIA |
- |
- |
16 |
75 |
M |
Adenocarcinoma |
T3N1M0 |
2 |
IIB |
- |
- |
17 |
72 |
F |
Adenocarcinoma |
T3N0M0 |
2 |
IIA |
+ |
+ |
18 |
60 |
M |
Adenocarcinoma |
T3N0M0 |
2 |
IIA |
- |
- |
19 |
63 |
F |
Adenocarcinoma |
T3N0M0 |
2 |
IIA |
- |
- |
20 |
69 |
M |
Adenocarcinoma |
T2N0M0 |
2 |
IB |
- |
- |
21 |
54 |
F |
Adenocarcinoma |
T3N0M0 |
2 |
IIA |
- |
- |
22 |
50 |
F |
Adenocarcinoma |
T3N0M0 |
3 |
IIA |
- |
- |
23 |
64 |
M |
Adenocarcinoma |
T3N0M0 |
3 |
IIA |
- |
++ |
24 |
59 |
M |
Adenocarcinoma |
T2N0M0 |
2 |
IB |
++ |
+++ |
25 |
59 |
M |
Adenocarcinoma |
T2N0M0 |
2 |
IB |
- |
- |
26 |
44 |
M |
Adenocarcinoma |
T3N0M0 |
2 |
IIA |
- |
- |
27 |
76 |
M |
Adenocarcinoma |
T3N0M0 |
2 |
IIA |
- |
+ |
28 |
56 |
M |
Adenocarcinoma |
T3N0M0 |
2 |
IIA |
- |
+ |
29 |
56 |
M |
Adenocarcinoma |
T2N0M0 |
2 |
IB |
+ |
+ |
30 |
58 |
M |
Adenocarcinoma |
T3N0M0 |
2 |
IIA |
- |
+ |
31 |
94 |
M |
Adenocarcinoma |
T2N0M0 |
2 |
IB |
+ |
+ |
32 |
56 |
F |
Adenocarcinoma |
T2N0M0 |
3 |
IB |
+ |
+ |
33 |
56 |
M |
Adenocarcinoma |
T4N1M0 |
3 |
IIIA |
+ |
+ |
34 |
51 |
F |
Adenocarcinoma |
T3N0M0 |
2 |
IIA |
+ |
+ |
35 |
67 |
M |
Adenocarcinoma |
T3N0M0 |
2 |
IIA |
+ |
+ |
36 |
53 |
M |
Adenocarcinoma |
T3N0M0 |
2-3 |
IIA |
+ |
+ |
37 |
48 |
F |
Adenocarcinoma |
T2N1M0 |
3 |
IIA |
++ |
++ |
38 |
58 |
M |
Adenocarcinoma |
T2N0M0 |
2 |
IB |
- |
- |
39 |
61 |
M |
Adenocarcinoma |
T2N0M0 |
3 |
IB |
- |
+ |
40 |
62 |
M |
Adenocarcinoma |
T2N0M0 |
3 |
IB |
- |
+ |
41 |
65 |
M |
Adenocarcinoma |
T2N0M0 |
3 |
IB |
+ |
+ |
42 |
47 |
F |
Adenocarcinoma |
T3N1M0 |
3 |
IIB |
- |
- |
43 |
65 |
M |
Adenocarcinoma |
T2N0M0 |
- |
IB |
- |
- |
44 |
52 |
F |
Adenocarcinoma |
T2N0M0 |
3 |
IB |
- |
- |
45 |
72 |
M |
Adenocarcinoma |
T2N0M0 |
3 |
IB |
- |
+ |
46 |
68 |
F |
Adenocarcinoma |
T3N0M0 |
3 |
IIA |
- |
++ |
47 |
56 |
M |
Adenocarcinoma |
T3N0M0 |
3 |
IIA |
++ |
++ |
48 |
59 |
M |
Adenocarcinoma |
T3N1M0 |
3 |
IIB |
+ |
+ |
49 |
62 |
M |
Adenocarcinoma |
T3N1M0 |
3 |
IIB |
- |
- |
50 |
60 |
M |
Adenocarcinoma |
T3N1M0 |
3 |
IIB |
+ |
+++ |
51 |
64 |
M |
Adenocarcinoma |
T2N0M0 |
3 |
IB |
+ |
+ |
52 |
69 |
M |
Adenocarcinoma |
T2N0M0 |
3 |
IB |
+ |
++ |
53 |
75 |
M |
Adenocarcinoma |
T2N0M0 |
3 |
IB |
+ |
++ |
54 |
48 |
M |
Adenocarcinoma |
T2N0M0 |
3 |
IB |
- |
- |
55 |
59 |
M |
Adenocarcinoma |
T2N0M0 |
3 |
IB |
- |
- |
56 |
64 |
M |
Adenocarcinoma |
T3N0M0 |
3 |
IIA |
+ |
+ |
57 |
55 |
M |
Adenocarcinoma |
T2N0M0 |
3 |
IB |
++ |
++ |
58 |
58 |
M |
Adenocarcinoma |
T3N0M0 |
3 |
IIA |
+ |
+ |
59 |
64 |
M |
Adenocarcinoma |
T3N0M0 |
3 |
IIA |
+ |
+ |
60 |
67 |
M |
Adenocarcinoma |
T3N1M0 |
3 |
IIB |
- |
+ |
61 |
49 |
M |
Adenocarcinoma |
T2N0M0 |
3 |
IB |
- |
- |
62 |
35 |
M |
Adenocarcinoma |
T3N1M0 |
3 |
IIB |
- |
- |
63 |
45 |
F |
Adenocarcinoma |
T4N0M1 |
3 |
IV |
+ |
++ |
64 |
43 |
M |
Adenocarcinoma |
T2N0M0 |
3 |
IB |
- |
+ |
65 |
56 |
M |
Adenocarcinoma |
T2N0M0 |
3 |
IB |
- |
- |
66 |
66 |
M |
Adenocarcinoma |
T2N0M0 |
3 |
IB |
+ |
+ |
67 |
60 |
M |
Adenocarcinoma |
T3N0M0 |
3 |
IIA |
- |
- |
68 |
74 |
M |
Adenocarcinoma |
T2N0M0 |
3 |
IB |
+ |
++ |
69 |
58 |
M |
Adenocarcinoma |
T2N0M0 |
3 |
IB |
- |
- |
70 |
68 |
M |
Mucinous adenocarcinoma |
T2N0M0 |
2 |
IB |
+ |
+ |
71 |
50 |
M |
Mucinous adenocarcinoma |
T3N0M0 |
3 |
IIA |
- |
- |
72 |
51 |
M |
Papillary adenocarcinoma |
T2N0M0 |
2 |
IB |
- |
+ |