1. Introduction
CD44 is involved in the tumor malignant progression through the promotion of tumor cell proliferation, migration, invasiveness, and stemness [
1]. The variety of CD44 function is mediated by the alternative splicing of 20 exons [
2]. CD44 standard (CD44s), the shortest isoform of CD44, is generated by the first five (1–5) and the last five (16–20) exons and expressed in a broad range of tissues. The central (6–15) exons are alternatively spliced and inserted between the first and last five exons of CD44s. The variant exon-containing CD44 is designated as the CD44 variant (CD44v) isoform [
3].
The CD44 ectodomain includes a hyaluronic acid (HA)-binding domain (HABD) [
4]. The HABD is present in both CD44s and CD44v isoforms. Upon HA binding, CD44s and CD44v transduce the intracellular signaling through the cytoplasmic domain, which promotes cell migration and proliferation [
5]. The variant exons-encoding regions possess a variety of functions. The v3-encoded region can be attached to a heparan sulfate side chain, which recruits heparin-binding growth factors and stimulates the signal transduction through activation of the receptors [
6,
7]. The v6-encoded region potentiates the MET signaling pathway through the formation of a ternary complex with its ligand, hepatocyte growth factor [
8,
9]. Moreover, the v8–10-encoded region regulates the intracellular reduced glutathione levels through the promotion of a cystine–glutamate transporter function [
10]. These functions are essential for tumor cell proliferation, invasiveness, and survival to oxidative stress [
10] and chemotherapeutic drugs [
11]. Therefore, CD44 has been considered as an essential target for tumor therapy [
4].
Monoclonal antibodies (mAbs) against CD44 have been evaluated in clinical trials [
12,
13]. RG7356, a humanized anti-pan-CD44 mAb, exhibited the antitumor effect for B cell leukemia but no cytotoxicity on normal B cells [
14]. In human chronic lymphocytic leukemia-engrafted mouse model, RG7356 administration resulted in complete clearance of engrafted leukemia cells [
14]. In acute myeloid leukemia [
15] and advanced CD44-positive solid tumor [
16], phase I clinical trials were conducted. Although RG7356 exhibited an acceptable safety profile, the studies were terminated due to the lack of dose-response relationship with RG7356 in both clinical and pharmacodynamic aspects [
16].
Since CD44v expression is restricted in epithelial tissue and carcinomas, anti-CD44v mAbs were developed and evaluated in clinical studies. Humanized anti-CD44v6 mAbs (BIWA-4 and BIWA-8) labeled with
186Re showed antitumor efficacy in head and neck squamous cell carcinoma (SCC) xenograft-bearing mice [
17]. Moreover, the antibody-drug conjugate (ADC) of BIWA-4, bivatuzumab-mertansine, was developed and evaluated in clinical trials [
13]. However, the clinical trials were terminated due to severe toxicity in the skin, probably due to the efficient accumulation of mertansine in the skin [
13,
18]. Therefore, anti-CD44 mAbs with more potent efficacy and lower toxicity to normal cells are desired.
The Fc region of therapeutic mAb binds to FcγRs on dendritic cells, macrophages, and neutrophils, which influences the adaptive immune responses through antigen presentation and cytokine production [
19]. Moreover, the FcγR binding results in the activation of natural killer (NK) cells and macrophages, which mediates antibody-dependent cellular cytotoxicity (ADCC) [
19]. The complement-dependent cellular cytotoxicity (CDC) is also considered as an essential effector function in tumor immunotherapy [
20]. The Fc region of therapeutic mAb binds to complement C1q, facilitating the assembly of active C1 complex (C1q, C1r, and C1s). The reaction of the complement cascade finally promotes the assembly of the pore-forming membrane attack complex (MAC or C5b–C9) on the tumor cell membrane, which results in the terminal cell lysis [
20]. The involvement of CDC in the antitumor effect was first recognized in the treatment of B cell lymphomas by an anti-CD20 mAb, rituximab [
21,
22]. Furthermore, the cytolytic capacity by complements has been shown in anti-CD38 and CD52 immunotherapies for multiple myeloma and chronic lymphocytic leukemia, respectively [
22,
23,
24]. Moreover, a growing body of evidence suggests that complement plays critical roles in not only tumor cell lysis but also several immunologic functions in antitumor immunity [
25,
26]. In the immunotherapy against solid tumors, an anti-HER2 bispecific and biparatopic antibody, zanidatamab, exerted more potent CDC against HER2-positive breast cancers compared with clinically approved anti-HER2 mAb, trastuzumab [
27].
The Cell-Based Immunization and Screening method is a strategy to obtain mAbs against a membrane protein comprehensively. We immunized mice with the CD44v3–10-overexpressed cells or CD44v3–10 ectodomain and established various anti-CD44 mAbs and determined their epitopes. C
44Mab-5 [
28] and C
44Mab-46 [
29] are anti-pan-CD44 mAbs, which have the epitopes within the constant exon 2- and 5-encoded sequences, respectively [
30,
31,
32]. We also established various CD44v-specific mAbs. C
44Mab-6 recognizes variant exon 3-encoded sequences and is referred to as anti-CD44v3 mAb [
33]. Furthermore, C
44Mab-3 (an anti-CD44v5 mAb) [
34], C
44Mab-9 (an anti-CD44v6 mAb) [
35], C
44Mab-34 (an anti-CD44v7/8 mAb) [
36], C
44Mab-1 (an anti-CD44v9 mAb) [
37], and C
44Mab-18 (an anti-CD44v10 mAb) [
38] were established. C
44Mab-108 is an anti-CD44v4 mAb, established by peptide immunization [
39]. These mAbs cover almost all variant exons and are applicable to flow cytometry, western blotting, and immunohistochemistry.
In this study, we produced an IgG2a-type C44Mab-18 (C44Mab-18-mG2a) and investigated the antitumor efficacy against CD44v3–10 and oral SCC (OSCC) xenografts by comparing anti-pan-CD44 mAb, C44Mab-46-mG2a.
3. Discussion
Among CD44v, v10-containing isoforms include the most abundant CD44v, such as CD44v3–10, CD44v6–10, and CD44v8–10 [
1,
41]. Therefore, anti-CD44v10 mAbs including C
44Mab-18-mG
2a can target the broad range of CD44v-expressing tumor cells. In this study, we evaluated the antitumor activities against CD44-positive cells compared with an anti-pan-CD44 mAb, C
44Mab-46-mG
2a. C
44Mab-18-mG
2a exhibited the higher reactivity to CHO/CD44v3–10 and OSCC cells compared with C
44Mab-46-mG
2a (
Figure 1 and
Figure 2). C
44Mab-18-mG
2a exhibited a superior ADCC against CD44v3–10 (
Figure 3) and OSCC (
Figure 5) cells. In contrast, C
44Mab-46-mG
2a showed a superior CDC against those cells. Furthermore, C
44Mab-18-mG
2a or C
44Mab-46-mG
2a similarly inhibited CD44v3–10 and OSCC xenograft growth compared with the control mouse IgG
2a (
Figure 4 and
Figure 6). These results indicate that C
44Mab-18-mG
2a could be a promising therapeutic regimen for CD44v10-positive tumors.
In CHO/CD44v3–10 cells, C
44Mab-18-mG
2a and C
44Mab-46-mG
2a recognized common target, but mainly exerted ADCC and CDC activity, respectively (
Figure 3). The epitope of C
44Mab-46 was previously determined as
174-TDDDV
-178 sequence in the constant exon 5-encoded region [
30] which is relatively apart from transmembrane domain compared to variant exon 10-encoded region recognized by C
44Mab-18 [
38]. To activate the classical pathway of complement, an ordered hexamer formation of IgG mAb is required to bind to the hexavalent complement C1q [
42,
43]. The structure of C
44Mab-46-mG
2a–CD44v3–10 complex may provide the appropriate space to form the hexameric structure of mAb–C1q complex and recruit the pore-forming membrane attack complex to exert CDC. In contrast, C
44Mab-18-mG
2a showed a higher reactivity to CHO/CD44v3–10 compared with C
44Mab-46-mG
2a in flow cytometry (
Figure 1). The difference of the reactivity and the epitope would influence the ADCC activity. Further studies are required to reveal the relationship among the affinity of mAb, epitope, and ADCC activity
Near-infrared photoimmunotherapy (NIR-PIT) uses a targeted mAb conjugated with a photoactivatable dye such as IRDye700DX (IR700) [
44]. When the mAb binds to the antigen-expressed target cells, IR700 induces plasma membrane rupture and immunogenic cell death by NIR-light exposure. Preclinical studies of anti-pan-CD44 mAb-based NIR-PIT (IM7-IR700) were conducted. In the syngeneic mouse model of OSCC, IM7-IR700 administration and the NIR light exposure to OSCC tumors resulted in a significant reduction but failed to induce durable antitumor responses [
45]. Because IM7 is a pan-CD44 mAb, IM7 might target not only tumor cells, but also CD44s-positive immune cells which involve in the antitumor immunity. The expression of CD44v is low in hematopoietic cells compared with CD44s [
1]. We previously showed that C
44Mab-18 can distinguish tumor cells from stromal tissues in immunohistochemistry. In contrast, C
44Mab-46 stained both tumor and stromal tissue including fibroblasts and leukocytes [
38]. Therefore, anti-CD44v10 mAb such as C
44Mab-18 might be a promising mAb for NIR-PIT without affecting the host immune cells in the tumor microenvironment.
Since both CD44s and CD44v are expressed in normal cells, there is a concern of adverse effects due to the recognition of normal cells by mAbs. In fact, clinical trials of the anti-CD44v6 mAb–ADC to advanced solid tumors were discontinued because of the skin toxicities [
13,
18]. Therefore, cancer specific mAbs (CasMabs) are desired to reduce the adverse effects. A CasMab against CD44v6 (clone 4C8) recognizes aberrantly
O-glycosylated Tn (GalNAcα1-
O-Ser/Thr) antigen in variant exon 6-encoded region. The 4C8 mAb was further developed to chimeric antigen receptor (CAR)-T cells, which exhibited target-specific in vitro cytotoxicity and significant tumor regression
in vivo [
46]. We have developed CasMabs against various tumor antigens, including HER2 (clones H
2Mab-214 [
47] and H
2Mab-250 [
48]) and reported the antitumor effect in mouse xenograft models using recombinant mouse IgG
2a or human IgG
1 mAbs [
49,
50]. These anti-HER2 mAbs were screened by the reactivity to cancer and normal cells in flow cytometry. H
2Mab-214 was revealed to recognize a locally misfolded structure in the Cys-rich HER2 extracellular domain 4, which usually forms a β-sheet [
47]. H
2Mab-250 also shows a specific reactivity against HER2-positive tumor cells, which has been developed as CAR-T cell therapy. The phase I study has been conducted in the US (NCT06241456). We have developed CasMabs against CD44s or CD44v by comparing the reactivity against tumor and normal cells. The anti-CD44 CasMabs could contribute to developing novel modalities such as ADCs and CAR-T cells.
4. Materials and Methods
4.1. Cell Lines and Cell Culture
CHO-K1 was obtained from the American Type Culture Collection (ATCC, Manassas, VA, USA). OSCC cell lines, HSC-2 and SAS were obtained from the Japanese Collection of Research Bioresources (Osaka, Japan). CHO/CD44s and CHO/CD44v3–10 were previously established by transfecting pCAG-Ble/PA16-CD44s and pCAG-Ble/PA16-CD44v3–10 into CHO-K1 cells using a Neon transfection system (Thermo Fisher Scientific, Inc., Waltham, MA, USA) [
29,
38].
HSC-2 and SAS was cultured in Dulbecco’s Modified Eagle Medium (DMEM, Nacalai Tesque, Inc.) containing 10% (v/v) heat−inactivated fetal bovine serum (FBS, Thermo Fisher Scientific Inc.), 100 U/mL of penicillin (Nacalai Tesque, Inc.), 100 μg/mL streptomycin (Nacalai Tesque, Inc.), and 0.25 μg/mL amphotericin B (Nacalai Tesque, Inc.).
CHO/CD44v3–10 was cultured in Roswell Park Memorial Institute (RPMI)-1640 medium (Nacalai Tesque, Inc.) supplemented with 10% (v/v) FBS, antibiotics as mentioned above, and 5 mg/mL Zeocin (InvivoGen, San Diego, CA, USA). All cells were grown in a humidified incubator at 37°C with 5% CO2.
4.2. Antibodies
An anti-pan-CD44 mAb (C
44Mab-46) and an anti-CD44v10 mAb (C
44Mab-18) were previously established [
29,
38]. A recombinant mouse IgG
2a-type mAb, C
44Mab-46-mG
2a, was generated previously [
40]. To generate a recombinant mouse IgG
2a-type mAb from C
44Mab-18 (IgM, kappa), V
H cDNAs of C
44Mab-18 and C
H of mouse IgG
2a were cloned into the pCAG-Ble vector (FUJIFILM Wako Pure Chemical Corporation, Osaka, Japan). V
L and mouse kappa light chain (C
L) cDNA of C
44Mab-18 was also cloned into the pCAG-Neo vector (FUJIFILM Wako Pure Chemical Corporation). Using the ExpiCHO Expression System (Thermo Fisher Scientific, Inc), the vectors were transfected into BINDS-09 cells (
http://www.med-tohoku-antibody.com/topics/001_paper_cell.htm) [
51] and the supernatants were collected. C
44Mab-18-mG
2a were purified using Ab-Capcher (ProteNova Co., Ltd., Kagawa, Japan). PMab-231 (a control mouse IgG
2a) was previously described [
49].
4.3. Flow Cytometry
CHO/CD44v3–10, HSC-2, and SAS were obtained using 1 mM ethylenediamine tetraacetic acid (EDTA; Nacalai Tesque, Inc.) and 0.25% trypsin treatment. The cells were treated with C44Mab-18-mG2a, C44Mab-46-mG2a, PMab-231, or blocking buffer [0.1% bovine serum albumin (BSA; Nacalai Tesque, Inc.) in phosphate-buffered saline (PBS)] (control) for 30 min at 4˚C. Subsequently, the cells were incubated in Alexa Fluor 488-conjugated anti-mouse IgG (1:2,000; Cell Signaling Technology, Inc., Danvers, MA, USA) for 30 min at 4˚C. Fluorescence data were collected using the SA3800 Cell Analyzer (Sony Corp., Tokyo, Japan) and analyzed using SA3800 software ver. 2.05 (Sony Corp.).
4.4. ADCC
The ADCC activity of C44Mab-18-mG2a and C44Mab-46-mG2a was measured as follows. The target cells (CHO/CD44v3–10, HSC-2, and SAS) were labeled with 10 µg/mL Calcein AM (Thermo Fisher Scientific, Inc.) and plated in 96-well plates (1 × 104 cells/well). The calcein-labeled target cells were mixed with the effector splenocyte (effector to target ratio, 100:1) from female BALB/c nude mice (Jackson Laboratory Japan, Inc., Kanagawa, Japan) with 100 μg/ml of C44Mab-18-mG2a, C44Mab-46-mG2a, or control mouse IgG2a (PMab-231). After a 4.5-hour incubation at 37°C, the calcein release into the medium was measured using a microplate reader (Power Scan HT; BioTek Instruments, Inc., Winooski, VT).
The cytolyticity (% lysis) was determined: % lysis is calculated as (E - S)/(M - S) x 100, where “E” indicates the fluorescence in effector and target cell cultures, “S” means the spontaneous fluorescence of only target cells, and “M” indicates the maximum fluorescence after treatment with a lysis buffer [10 mM Tris-HCl (pH 7.4), 10 mM EDTA, and 0.5% Triton X-100]. Statistical analyses were performed using GraphPad PRISM 6 software (GraphPad Software, Inc., La Jolla, CA, USA). All data are shown as mean ± standard error of the mean (SEM). One-way ANOVA with Tukey's multiple comparisons test was used for the statistical analyses. p < 0.05 was considered to indicate a statistically significant difference.
4.5. CDC
The calcein-labeled target cells were mixed with rabbit complement (final dilution 1:10, Low-Tox-M Rabbit Complement; Cedarlane Laboratories, Hornby, ON, Canada) and 100 μg/ml of C44Mab-18-mG2a, C44Mab-46-mG2a, or control mouse IgG2a (PMab-231). After a 4-hour incubation at 37°C, the calcein release into the medium was measured. Statistical analyses were performed as described above.
4.6. Antitumor Activity of C44Mab-18-mG2a and C44Mab-46-mG2a in Xenografts of CHO/CD44v3–10, HSC-2, and SAS
The animal study protocol was approved (approval nos. 2024-013 and 2024-038) by the Institutional Committee for Experiments of the Institute of Microbial Chemistry (Numazu, Japan). CHO/CD44v3–10, HSC-2, or SAS cells (5 × 106 cells) suspended with BD Matrigel Matrix Growth Factor Reduced (BD Biosciences, Franklin Lakes, NJ, USA) were inoculated into the left flank of female BALB/c nude mice subcutaneously. On day 7 after the inoculation, 100 μg of C44Mab-18-mG2a (n=8), C44Mab-46-mG2a (n=8), or control mouse IgG2a (PMab-231) (n=8) in 100 μl PBS were injected intraperitoneally. Additional antibody injections were performed on days 14 and 21. The tumor volume was measured on the indicated days.
In the HSC-2, or SAS xenograft experiment, 500 μg of C44Mab-18-mG2a (n=8), C44Mab-46-mG2a (n=8), or control mouse IgG2a (PMab-231) (n=8) in 100 μl PBS were injected intraperitoneally on days 7 and 14 after the inoculation. The tumor volume was measured on the indicated days. The xenograft tumors were carefully removed from the sacrificed mice and weighed immediately.
All data are expressed as the mean ± standard error of the mean (SEM). In tumor weight measurement, one-way ANOVA with Tukey's multiple comparisons test was conducted. Two-way ANOVA with Tukey's multiple comparisons test was utilized for tumor volume and mice weight. GraphPad Prism 6 (GraphPad Software, Inc.) was used for all calculations. p < 0.05 was considered to indicate a statistically significant difference.
Author Contributions
For research articles with several authors, a short paragraph specifying their individual contributions must be provided. The following statements should be used “Conceptualization, M.K., M.K.K., and Y.Kato; methodology, T.O.; formal analysis, T.T.; investigation, K.I., H.S., T.O., G.L., and T.T.; data curation, H.S. and Y.Kato; writing—original draft preparation, K.I. and H.S.; writing—review and editing, Y.Kato; supervision, A.O. and Y.Katori; project administration, Y.Kato.; funding acquisition, H.S., T.T., M.K.K., and Y.Kato. All authors have read and agreed to the published version of the manuscript.”
Figure 1.
Flow cytometry using C44Mab-18-mG2a and C44Mab-46-mG2a. (A) Class-switched mouse IgG2a mAbs, C44Mab-18-mG2a, and C44Mab-46-mG2a were generated from C44Mab-18 (mouse IgM) and C44Mab-46 (mouse IgG1), respectively. CHO/CD44v3–10 (B) and CHO/CD44s (C) cells were treated with buffer control (black) or 10-0.01 µg/mL of C44Mab-18-mG2a and C44Mab-46-mG2a (red). The cells were further treated with Alexa Fluor 488-conjugated anti-mouse IgG. Fluorescence data were analyzed using the SA3800 Cell Analyzer.
Figure 1.
Flow cytometry using C44Mab-18-mG2a and C44Mab-46-mG2a. (A) Class-switched mouse IgG2a mAbs, C44Mab-18-mG2a, and C44Mab-46-mG2a were generated from C44Mab-18 (mouse IgM) and C44Mab-46 (mouse IgG1), respectively. CHO/CD44v3–10 (B) and CHO/CD44s (C) cells were treated with buffer control (black) or 10-0.01 µg/mL of C44Mab-18-mG2a and C44Mab-46-mG2a (red). The cells were further treated with Alexa Fluor 488-conjugated anti-mouse IgG. Fluorescence data were analyzed using the SA3800 Cell Analyzer.
Figure 2.
Flow cytometry using C44Mab-18-mG2a and C44Mab-46-mG2a against oral squamous cell carcinoma (OSCC) cell lines. HSC-2 (A) and SAS (B) cells were treated with buffer control (black) or 10-0.01 µg/mL of C44Mab-18-mG2a and C44Mab-46-mG2a (red). The cells were further treated with Alexa Fluor 488-conjugated anti-mouse IgG. Fluorescence data were analyzed using the SA3800 Cell Analyzer.
Figure 2.
Flow cytometry using C44Mab-18-mG2a and C44Mab-46-mG2a against oral squamous cell carcinoma (OSCC) cell lines. HSC-2 (A) and SAS (B) cells were treated with buffer control (black) or 10-0.01 µg/mL of C44Mab-18-mG2a and C44Mab-46-mG2a (red). The cells were further treated with Alexa Fluor 488-conjugated anti-mouse IgG. Fluorescence data were analyzed using the SA3800 Cell Analyzer.
Figure 3.
Evaluation of ADCC and CDC activity of C44Mab-18-mG2a and C44Mab-46-mG2a against CHO/CD44v3–10 cells. The ADCC (A) and CDC (B) induced by C44Mab-18-mG2a, C44Mab-46-mG2a, or control mouse IgG2a (mIgG2a, PMab-231) against CHO/CD44v3–10 cells. Values are shown as mean ± SEM. Asterisks indicate statistical significance (**p < 0.01 and *p < 0.05; one-way ANOVA with Tukey's multiple comparisons test).
Figure 3.
Evaluation of ADCC and CDC activity of C44Mab-18-mG2a and C44Mab-46-mG2a against CHO/CD44v3–10 cells. The ADCC (A) and CDC (B) induced by C44Mab-18-mG2a, C44Mab-46-mG2a, or control mouse IgG2a (mIgG2a, PMab-231) against CHO/CD44v3–10 cells. Values are shown as mean ± SEM. Asterisks indicate statistical significance (**p < 0.01 and *p < 0.05; one-way ANOVA with Tukey's multiple comparisons test).
Figure 4.
Antitumor activity of C44Mab-18-mG2a and C44Mab-46-mG2a against CHO/CD44v3–10 xenograft. (A) Tumor volume of CHO/CD44v3–10 xenograft. CHO/CD44v3–10 cells (5 × 106 cells) were injected into mice subcutaneously. On days 7, 14, and 21, 100 μg of C44Mab-18-mG2a (n=8), C44Mab-46-mG2a (n=8) or control mouse IgG2a (mIgG2a, PMab-231) (n=8) were injected into mice intraperitoneally (arrows). The tumor volume was measured on days 7, 14, 17, 21, 24, and 28 following the inoculation. Values are presented as the mean ± SEM. *p < 0.05 and **p < 0.01 (two-way ANOVA with Tukey's multiple comparisons test). The weight (B) and appearance (C) of excised CHO/CD44v3–10 xenografts on day 28. Values are presented as the mean ± SEM. *p < 0.05 (one-way ANOVA with Tukey's multiple comparisons test). The body weight (D) and appearance (E) of CHO/CD44v3–10 xenograft-bearing mice treated with C44Mab-18-mG2a, C44Mab-46-mG2a, or control mIgG2a. Scale bar, 1 cm.
Figure 4.
Antitumor activity of C44Mab-18-mG2a and C44Mab-46-mG2a against CHO/CD44v3–10 xenograft. (A) Tumor volume of CHO/CD44v3–10 xenograft. CHO/CD44v3–10 cells (5 × 106 cells) were injected into mice subcutaneously. On days 7, 14, and 21, 100 μg of C44Mab-18-mG2a (n=8), C44Mab-46-mG2a (n=8) or control mouse IgG2a (mIgG2a, PMab-231) (n=8) were injected into mice intraperitoneally (arrows). The tumor volume was measured on days 7, 14, 17, 21, 24, and 28 following the inoculation. Values are presented as the mean ± SEM. *p < 0.05 and **p < 0.01 (two-way ANOVA with Tukey's multiple comparisons test). The weight (B) and appearance (C) of excised CHO/CD44v3–10 xenografts on day 28. Values are presented as the mean ± SEM. *p < 0.05 (one-way ANOVA with Tukey's multiple comparisons test). The body weight (D) and appearance (E) of CHO/CD44v3–10 xenograft-bearing mice treated with C44Mab-18-mG2a, C44Mab-46-mG2a, or control mIgG2a. Scale bar, 1 cm.
Figure 5.
Evaluation of ADCC and CDC activity of C44Mab-18-mG2a and C44Mab-46-mG2a against HSC-2 and SAS cells. The ADCC (A,C) and CDC (B,D) induced by C44Mab-18-mG2a, C44Mab-46-mG2a, or control mouse IgG2a (mIgG2a, PMab-231) against HSC-2 (A,B) and SAS (C,D) cells. Values are shown as mean ± SEM. Asterisks indicate statistical significance (**p < 0.01 and *p < 0.05; one-way ANOVA with Tukey's multiple comparisons test).
Figure 5.
Evaluation of ADCC and CDC activity of C44Mab-18-mG2a and C44Mab-46-mG2a against HSC-2 and SAS cells. The ADCC (A,C) and CDC (B,D) induced by C44Mab-18-mG2a, C44Mab-46-mG2a, or control mouse IgG2a (mIgG2a, PMab-231) against HSC-2 (A,B) and SAS (C,D) cells. Values are shown as mean ± SEM. Asterisks indicate statistical significance (**p < 0.01 and *p < 0.05; one-way ANOVA with Tukey's multiple comparisons test).
Figure 6.
Antitumor activity of C44Mab-18-mG2a and C44Mab-46-mG2a against HSC-2 and SAS xenograft. (A,B) Tumor volume in HSC-2 (A) and SAS (B) xenograft. HSC-2 and SAS cells (5 × 106 cells) were injected into mice subcutaneously. On days 7 and 14, 500 μg of C44Mab-18-mG2a (n=8), C44Mab-46-mG2a (n=8), or control mouse IgG2a (mIgG2a, PMab-231) (n=8) were injected into mice intraperitoneally (arrows). The tumor volume was measured on days 7, 14, and 17 following the inoculation. Values are presented as the mean ± SEM. **p < 0.01 (two-way ANOVA with Tukey's multiple comparisons test). (C,D) The weight and appearance of the excised HSC-2 (C) and SAS (D) xenografts on day 17. Values are presented as the mean ± SEM. * P < 0.05 and **p < 0.01 (one-way ANOVA with Tukey's multiple comparisons test). (E,F) The body weight of HSC-2 (E) and SAS (F) xenograft-bearing mice treated with C44Mab-18-mG2a, C44Mab-46-mG2a, or control mIgG2a. Scale bar, 1 cm.
Figure 6.
Antitumor activity of C44Mab-18-mG2a and C44Mab-46-mG2a against HSC-2 and SAS xenograft. (A,B) Tumor volume in HSC-2 (A) and SAS (B) xenograft. HSC-2 and SAS cells (5 × 106 cells) were injected into mice subcutaneously. On days 7 and 14, 500 μg of C44Mab-18-mG2a (n=8), C44Mab-46-mG2a (n=8), or control mouse IgG2a (mIgG2a, PMab-231) (n=8) were injected into mice intraperitoneally (arrows). The tumor volume was measured on days 7, 14, and 17 following the inoculation. Values are presented as the mean ± SEM. **p < 0.01 (two-way ANOVA with Tukey's multiple comparisons test). (C,D) The weight and appearance of the excised HSC-2 (C) and SAS (D) xenografts on day 17. Values are presented as the mean ± SEM. * P < 0.05 and **p < 0.01 (one-way ANOVA with Tukey's multiple comparisons test). (E,F) The body weight of HSC-2 (E) and SAS (F) xenograft-bearing mice treated with C44Mab-18-mG2a, C44Mab-46-mG2a, or control mIgG2a. Scale bar, 1 cm.