Skip to main content
Cancer Immunology, Immunotherapy : CII logoLink to Cancer Immunology, Immunotherapy : CII
. 2023 Feb 16;72(7):2057–2065. doi: 10.1007/s00262-023-03388-5

Cisplatin resistance driver claspin is a target for immunotherapy in urothelial carcinoma

Shuhei Yamada 1,2, Haruka Miyata 2, Makoto Isono 3, Kanta Hori 1,2, Junko Yanagawa 1, Aiko Murai 1, Tomoyuki Minowa 1,4, Yuka Mizue 1, Kenta Sasaki 1,5, Kenji Murata 1, Serina Tokita 1, Munehide Nakatsugawa 6, Sadahiro Iwabuchi 7, Shinichi Hashimoto 7, Terufumi Kubo 1, Takayuki Kanaseki 1, Tomohide Tsukahara 1, Takashige Abe 2, Nobuo Shinohara 2, Yoshihiko Hirohashi 1,, Toshihiko Torigoe 1,
PMCID: PMC10992486  PMID: 36795123

Abstract

Bladder cancer is a major and fatal urological disease. Cisplatin is a key drug for the treatment of bladder cancer, especially in muscle-invasive cases. In most cases of bladder cancer, cisplatin is effective; however, resistance to cisplatin has a significant negative impact on prognosis. Thus, a treatment strategy for cisplatin-resistant bladder cancer is essential to improve the prognosis. In this study, we established a cisplatin-resistant (CR) bladder cancer cell line using an urothelial carcinoma cell lines (UM-UC-3 and J82). We screened for potential targets in CR cells and found that claspin (CLSPN) was overexpressed. CLSPN mRNA knockdown revealed that CLSPN had a role in cisplatin resistance in CR cells. In our previous study, we identified human leukocyte antigen (HLA)-A*02:01-restricted CLSPN peptide by HLA ligandome analysis. Thus, we generated a CLSPN peptide-specific cytotoxic T lymphocyte clone that recognized CR cells at a higher level than wild-type UM-UC-3 cells. These findings indicate that CLSPN is a driver of cisplatin resistance and CLSPN peptide-specific immunotherapy may be effective for cisplatin-resistant cases.

Supplementary Information

The online version contains supplementary material available at 10.1007/s00262-023-03388-5.

Keywords: Urothelial carcinoma, Claspin, Cisplatin resistance, Immunotherapy

Introduction

Bladder cancer is the 10th most common cancer worldwide, with 550,000 new cases and more than 200,000 deaths per year worldwide [1, 2]. Approximately 70% of new bladder cancer cases are non-invasive, and while most of these cases can be controlled surgically, the recurrence rate is high. The remaining 30% of new cases are muscle-invasive bladder cancer, and many of them have distant metastasis. Patients with distant metastasis have an extremely poor prognosis, with a 5 year survival rate of approximately 5%. For many years, the mainstay of treatment for bladder cancer with local invasion or distant metastasis has been chemotherapy, mainly cisplatin (CDDP), and although the response rate is not low, many patients eventually become resistant to treatment and the prognosis for advanced bladder cancer remains poor [1, 3]. CDDP-based chemotherapy in neoadjuvant and adjuvant settings improves the overall survival of patients with muscle-invasive bladder cancer; therefore, the development of treatment resistance, especially to CDDP, has a direct impact on prognosis. Unlike many other cancers, the survival rate of bladder cancer has not improved in 30 years [4]. Therefore, it is essential to analyze the mechanism underlying resistance to CDDP to improve the outcome of current bladder cancer therapy.

Bladder cancer is an immunogenic malignancy and cancer immunotherapy using BCG has been approved for decades [5]. Immune checkpoint blockade has also been approved for bladder cancer [69]. However, these immunotherapies are based on the non-specific augmentation of the immune response; thus, analysis of antigen-specificity in bladder cancer remains essential for further innovation in immunotherapy.

In this study, we established a novel CDDP-resistant bladder cancer subline and identified the novel immunotherapy target claspin (CLSPN) as a driver of CDDP resistance.

Materials and methods

Cell lines and cell culture

The following human cell lines were used in this study: human bladder carcinoma lines (UM-UC-3 and J82), erythroleukemia (K562), and TAP-deficient (T2) cells (American Type Culture Collection, Rockville, MD). The cell lines were maintained in Dulbecco’s-modified Eagle’s medium (DMEM) or RPMI (Sigma-Aldrich, St. Louis, MO) supplemented with 10% fetal bovine serum (FBS) and 1% penicillin–streptomycin (5 mg/mL penicillin, 5 mg/mL streptomycin; Thermo Fisher Scientific, Waltham, MA). The cells were cultured at 37 ℃ in an incubator containing humidified air and 5% CO2.

Establishment of a UM-UC3 CDDP-resistant (CR) cell line

The CR cell line was derived from the original parental UM-UC-3 cell line and J82 cell line by continuous exposure to CDDP at stepwise increasing concentrations. Initially, exponentially growing cells were exposed to 0.5 μmol/L CDDP. These cells were maintained in CDDP-containing DMEM supplemented with 10% FBS and 1% penicillin/streptomycin, and passaged upon reaching 70–80% confluency. At this point, the CDDP concentration was increased by approximately 1.5-fold and the above process was repeated continuously over a period of approximately 6 months. The final concentration of CDDP was 5 μmol/L. To address the transient response to CDDP, the cells were treated with CDDP at several concentrations for quantitative real-time (qRT)-PCR and 1 μmol/L for an interferon gamma (IFNγ) enzyme-linked immunospot (ELISPOT) assay for 2 days.

Aldefluor assay, sphere-formation assay, and CDDP sensitivity assay

An ALDEFLUOR assay (STEMCELL Technologies, Vancouver, Canada) using wild-type (WT) UM-UC-3 cells and CR cells was performed as previously described [10]. The stained cells were analyzed by a FACSCanto (BD Biosciences, Bedford, MA). A sphere-formation assay and estimation of cancer stem-like cells (CSCs)/cancer-initiating cells (CICs) were performed as previously described [10]. The number of CSCs/CICs was estimated using the Extreme Limiting Dilution Analysis (ELDA) website (http://bioinf.wehi.edu.au/software/elda/) [11]. Resistance to CDDP was assessed as previously described [12], using a WST-8 assay (Dojindo Molecular Technologies, Kumamoto, Japan).

Small interfering RNA (siRNA)-mediated knockdown

The cells were transfected with siRNA targeting CLSPN (Thermo Fisher Scientific, Waltham, MA) using the Lipofectamine RNAi MAX reagent (Thermo Fisher Scientific). Non-targeting siRNA (Thermo Fisher Scientific) was used as a negative control. CLSPN knockdown was confirmed by qRT-PCR.

Western blotting and qRT-PCR

Western blotting was performed as previously described [10]. Briefly, 5.0 × 105 WT UM-UC-3 or CR cells were lysed in 100 μL radio-immunoprecipitation assay buffer. An anti-CLSPN rabbit polyclonal antibody (Cell Signaling Technology, Danvers, MA) was used at a dilution of 1:1,000. An anti-MDR1 rabbit monoclonal antibody (Cell Signaling Technology) was used at a dilution of 1:1,000. An anti-β-actin mouse monoclonal antibody (Sigma-Aldrich) was used as a loading control at a dilution of 1:1,000. Anti-mouse IgG and anti-rabbit IgG secondary antibodies (KPL, Gaithersburg, MD) were used at a dilution of 1:2,000. The membrane was visualized with Chemiluminescent HRP Substrate (Millipore, Billerica, MA), and images were taken by an Odyssey Fc imaging System (LI-COR, Lincoln, NE). qRT-PCR was performed as previously described [13].

Cap analysis of gene expression (CAGE) analysis

CAGE library preparation, sequencing, mapping, gene expression, motif discovery analysis, and Gene Ontology enrichment analysis were performed by DNAFORM (Yokohama, Japan) as previously described [12].

CAGE tag count data were clustered by CAGEr [14] using the Paraclu algorithm [15] with default parameters. Differentially expressed genes (CPM > 1 and fold-change < 0.25, or CPM > 1 and fold-change > 4) were detected with the DESeq2 package (version 1.20.0) and used for Gene Ontology enrichment analysis by the clusterProfiler package [16]. Raw and processed CAGE data have been deposited in the NCBI GEO database (GSE217413).

Cytotoxic T lymphocyte (CTL) induction, ELISPOT assay, and establishment of a CLSPN peptide-specific CTL clone

Induction of CTL and establishment of a CTL clone were performed as previously described, with some modifications [17]. Briefly, peripheral blood mononuclear cells (PBMCs) isolated from a human leukocyte antigen (HLA)-A*02:01-positive donor were stimulated for 1 week with 10 μg/mL CLSPN peptide (Cosmo Bio, Inc., Tokyo, Japan). The PBMCs were stimulated with artificial antigen-presenting cells (aAPCs) that were established by transduction with CD80, CD83, and HLA-A*02:01 cDNAs, as previously described [18]. For CTL culture, interleukin (IL)-2 and IL-15 (PeproTech, Cranbury, NJ) were used at final concentrations of 10 U/mL and 10 ng/mL, respectively. An IFNγ ELISPOT assay was performed as previously described [16]. CLSPN peptide-reactive wells were stained with CLSPN-HLA-A2 tetramer (MBL, Nagoya, Japan), and tetramer-positive cells were sorted into 96-well plates (Corning, Inc., Corning, NY) with a FACSAria II (BD Biosciences) and co-cultured with 100 Gy-irradiated PBMCs as feeder cells for 2 weeks as previously described [13, 19]. The wells with growing cells were evaluated by staining with CLSPN-HLA-A2 tetramer.

Lactate dehydrogenase (LDH) cytotoxicity assay

To evaluate the function of the CTL clone, an LDH release assay was performed using a CyQUANT LDH Cytotoxicity Assay Kit (Thermo Fisher Scientific). WT UM-UC-3 and CR cells were plated in 96-well plates at a density of 2.0 × 104 cells/well in DMEM containing 10% FBS. After overnight culture, the medium was replaced with control medium (serum-free AIM-V) and the CLSPN-specific CTL clone yc3 was added at an effector-target ratio of 9:1, 3:1, or 1:1 and co-cultured for 8 h. The release of LDH was then measured according to the manufacturer’s instructions.

Statistical analysis

Student’s t test was used to compare two groups; p < 0.05 was considered to be statistically significant. Differences in the estimated frequency of CSCs/CICs were analyzed by the chi-square test.

Results

Establishment of CR cells

In this study, we aimed to identify potential immunological targets that are expressed in CDDP-resistant subpopulations of urothelial carcinoma cells. We cultured the UM-UC-3 urothelial carcinoma cell line in CDDP-containing medium, and we increased the concentration of CDDP gradually over a period of approximately 6 months to 5 μM (Fig. 1A) to establish a CR cell line. CR cells showed resistance to CDDP compared with WT cells (Fig. 1B). A previous report indicated that chemoresistance is related to CSCs [20]; thus, we examined whether CSCs are enriched in CR cells. An ALDEFLUOR assay revealed that the aldehyde dehydrogenase-high (ALDHhigh) population was increased, indicating that CSCs were enriched in CR cells (Fig. 1C). A sphere-formation assay revealed that the frequency of sphere-forming CSCs was increased in CR cells compared with WT cells (Fig. 1D).

Fig. 1.

Fig. 1

Establishment and characterization of a CR subline. A Schematic summary of the culture strategy for establishing a CR cell line. UM-UC-3 cells were cultured in medium containing CDDP at 0.5 μmol/L. The concentration of CDDP was increased over a period of approximately 6 months. B Sensitivity of WT and CR cells to CDDP. The cells were cultured in medium containing CDDP at several concentrations for 48 h. Cell viability was assessed by a WST-8 assay. Each value is the mean ± standard deviation (SD). Statistical analysis was performed using Student’s t-test. C ALDEFLUOR assay. WT UM-UC-3 and CR cells were stained with BODIPY®-aminoacetaldehyde and analyzed by FACS. The ALDHhigh-positive gate was defined by a sample treated with the ALDH1 inhibitor DEAB. Numerical data indicate ALDHhigh rates. D Sphere-formation assay. The sphere-forming ability of WT UM-UC-3 and CR cells was assessed. The cells were seeded at 1.0 × 103, 102, 101, and 100 cells/well in an ultra-low attachment plate and cultured for 1 week. Sphere-forming wells were counted. Stem cell frequency was analyzed using the ELDA web site. CI, confidence interval. Differences of the estimated frequencies of CSCs/CICs were analyzed by a chi-square test

CLSPN expression is upregulated in CR cells

To identify a potential immunological target, we screened the genes that were overexpressed in CR cells compared with WT cells. We performed CAGE analysis and found that 2284 genes were upregulated in CR cells (Supplemental Table S1). CDDP resistance responsive gene MDR1/ABCB1 was 59.5-fold increased and the protein expression was confirmed by a Western blot (Fig. 2D). A previous study revealed that MDR1/ABCB1 is related to CDDP [21] and the results in this study was consistent with previous study. To narrow down the candidate genes, we referenced HLA ligandome analysis and a public database of gene expression (GTExPortal: https://www.gtexportal.org). Previously, we performed HLA ligandome analysis using UM-UC-3 cells to identify HLA-A2-bound antigenic peptides [20]. Thus, we screened genes that were upregulated in CR cells, that encoded HLA-A2-bound antigenic peptides identified by HLA ligandome analysis, and whose expression in normal organs was less than 2 transcripts per million. Finally, we found that the CLSPN gene was overexpressed in CR cells and the candidate peptide sequence encoded by CLSPN was SLLNQPKAV (Fig. 2A). CLSPN mRNA expression in normal organs was low or undetectable according to qRT-PCR (Fig. 2B). CLSPN mRNA expression was higher in CR cells than in WT cells (Fig. 2C). Western blot analysis revealed that CLSPN protein was expressed in CR and WT cells (Fig. 2D). To generalize the results, we established another CDDP-resistant line using an urothelial carcinoma cell J82. J82 CR cells also showed resistance to CDDP compared with that in J82 WT cells (Supplemental Figure S1A). qRT-PCR analysis and Western blot analysis revealed that CLSPN mRNA and protein were overexpressed in J82 CR cells compared with that in J82 WT cells.

Fig. 2.

Fig. 2

CLSPN is overexpressed in CR cells. A CAGE analysis of WT UM-UC-3 cells vs. CR cells. Gene expression in CR cells was assessed by CAGE analysis. The mRNA expression of WT and CR cells was analyzed in duplicate and fold-change was plotted in a scatter plot (left panel). The CLSPN plot is indicated. To narrow down candidate genes, (1) CR overexpressed genes, (2) expression in normal organs < 2 (transcripts per million), and (3) HLA-A2-bound peptides were used as filters. Finally, CLSPN and its coding antigenic peptide SLLNQPKAV were identified. B qRT-PCR of CLSPN in normal organs. CLSPN expression in normal organs was assessed by qRT-PCR. Each value is the relative mean ± SD. C qRT-PCR of CLSPN in WT UM-UC-3 and CR cells. CLSPN expression in WT UM-UC-3 and CR cells was assessed by qRT-PCR. Each value is the relative mean ± SD. D Western blot analysis of WT UM-UC-3 and CR cells. CLSPN and MDR1 expression in WT UM-UC-3 and CR cells was assessed by western blot analysis. β-Actin was used as an internal positive control

CLSPN is a driver of CDDP resistance

CLSPN was expressed at higher levels in CR cells than in WT cells, and a previous study indicated that CLSPN is related to CSCs [22]. CLSPN is expressed in the G1/S phase of the cell cycle and has a role in the DNA checkpoint response [23]. Thus, we hypothesized that CLSPN might have a role in CDDP resistance. To assess the functions of CLSPN, we knocked down CLSPN mRNA using siRNA; CLSPN mRNA knockdown was confirmed by qRT-PCR (data not shown). siRNA knockdown of CLSPN mRNA increased the sensitivity of CR cells to CDDP (Fig. 3A). CDDP induced the expression of CLSPN mRNA in CR and WT cells (Fig. 3B).

Fig. 3.

Fig. 3

CLSPN has a role in CDDP resistance and its expression is induced by CDDP. A Resistance to CDDP. Resistance to CDDP was assessed by a WST-8 assay. WT UM-UC-3 and CR cells were cultured in medium containing serial concentrations of CDDP for 48 h, and then cell viability was examined by a WST-8 assay. Each value is the relative mean ± SD. B CLSPN mRNA induction by CDDP treatment. WT UM-UC-3 and CR cells were cultured in medium containing serial concentrations of CDDP for 48 h, and then CLSPN expression in normal organs was assessed by qRT-PCR. Each value is the relative mean ± SD

Establishment and characterization of a CLSPN peptide-specific CTL clone

To generate CLSPN peptide-specific CTLs, we stimulated HLA-A*02:01-positive PBMCs with HLA-A*02:01-positive aAPCs pulsed with CLSPN peptide (Fig. 4A). After stimulation for 5 times, the cells were evaluated by an IFNγ ELISPOT assay. The number of IFNγ spots was increased in CLSPN peptide-pulsed T2 cells compared with peptide non-pulsed T2 cells (Fig. 4B). Flow cytometry revealed the presence of CLSPN tetramer-positive cells (Fig. 4B). To analyze the CTLs at the clonal level, we established the CLSPN-specific CTL clone yc3 by CLSPN tetramer-positive single cell sorting (Fig. 4B). Seven wells showed cell growth and we tested the cells by staining with CLSPN-HLA-A2 tetramer, finding that the yc3 clone was specific for CLSPN tetramer. Thus, we performed further analyses using the yc3 clone.

Fig. 4.

Fig. 4

Induction and characterization of a CLSPN peptide-specific CTL clone. A A schematic summary of CTL induction. PBMCs were obtained from an HLA-A*02:01-positive donor and CD8+ T cells were then isolated. CD8+ T cells were stimulated with HLA-A*02:01+ aAPCs (K562 cells transduced with CD80, CD83, and HLA-A*02:01 cDNAs) pulsed with CLSPN peptide (SLLNQPKAV) at day 1, 8, 15, 22, and 29. At day 34 the CD8+ T cells were assessed by an ELISPOT assay and FACS. B CTL cloning strategy. Stimulated CD8+ T cells were analyzed by an IFNγ ELISPOT assay and FACS with CLSPN tetramer staining. For the IFNγ ELISPOT assay, T2 cells pulsed with CLSPN peptide and non-pulsed cells were used as targets. For FACS analysis, HIV-tetramer was used as a negative control. CLSPN tetramer-positive cells were single cell sorted and cultured. Cells grew in seven wells and one clone (yc3) showed reactivity to CLSPN peptide according to an IFNγ ELISPOT assay and FACS. C CLSPN peptide-specific CTL clone recognizes CR cells. The CLSPN peptide-specific CTL yc3 clone was evaluated for reactivity to WT UM-UC-3 and CR cells by an IFNγ ELISPOT assay. CLSPN peptide-pulsed T2 cells were used as a positive control and peptide non-pulsed T2 cells were used as a negative control. Each value is the mean ± SD. D CLSPN peptide-specific CTL clone recognizes CDDP-treated WT cells. The CLSPN peptide-specific CTL yc3 clone was evaluated for reactivity to WT UM-UC-3 cells and CDDP-treated WT cells by an IFNγ ELISPOT assay. WT cells were cultured in CDDP-containing medium (5 μM) for 48 h, and then used for assay. T2 cells were used as a negative control. Each value is the mean ± SD. E CLSPN knockdown by siRNA decreases the CTL response. WT UM-UC-3 cells were transfected with CLSPN siRNA, and 48 h later, the cells were evaluated by an IFNγ ELISPOT assay with the CLSPN peptide-specific CTL yc3 clone. T2 cells were used as a negative control. Each value is the mean ± SD

An IFNγ ELISPOT assay revealed that the yc3 clone recognized WT and CR cells (Fig. 4C). Although the yc3 clone recognized CR cells more strongly than WT cells, the difference did not reach statistical significance (p = 0.101) (Fig. 4C). Because CLSPN expression was induced by CDDP (Fig. 3B), we examined whether CDDP stimulation enhanced the reactivity of the yc3 clone. The addition of CDDP significantly increased the number of IFNγ spots compared with CDDP non-treated WT cells (Fig. 4D). To confirm the specificity of the yc3 clone, we knocked down CLSPN mRNA with siRNA and assessed the reactivity of the yc3 clone. Knockdown of CLSPN mRNA significantly decreased the number of IFNγ spots compared with WT cells (Fig. 4E). Finally, to confirm the cytotoxic activity of the yc3 clone, we examined cytotoxicity using an LDH release assay. The LDH release assay revealed that the yc3 clone showed higher cytotoxicity to CLSPN peptide-pulsed T2 cells than to control peptide-pulsed T2 cells, peptide non-pulsed T2 cells, and negative control K562 cells (Fig. 5A). Furthermore, the yc3 clone showed higher cytotoxicity to CR cells than to WT cells (Fig. 5B). These results indicate that CLSPN peptide is endogenously expressed by CR cells, and the yc3 clone recognizes and kills CR cells in a CLSPN peptide-dependent manner.

Fig. 5.

Fig. 5

Cytotoxicity assay using the CLSPN peptide-specific CTL clone. A Cytotoxicity of the yc3 clone for CLSPN peptide-pulsed targets. The cytotoxicity of the yc3 clone was assessed by an LDH release assay. Target cells were co-cultured with the yc3 clone for 8 h at different effector/target ratios. HIV peptide-pulsed T2 cells, peptide non-pulsed T2 cells, and K562 cells were used as negative controls. Each value is the mean ± SD. B Cytotoxicity of the yc3 clone to WT UM-UC-3 and CR cells. The cytotoxicity of the yc3 clone was assessed by an LDH release assay. WT UM-UC-3 and CR cells were used as targets. Target cells were co-cultured with the yc3 clone for 8 h at different effector/target ratios. Each value is the mean ± SD

Discussion

In this study, we established a novel CDDP-resistant UM-UC-3 urothelial carcinoma cell line that showed a CSC-like phenotype. CSCs are resistant to chemotherapy and radiotherapy by several mechanisms [24, 25]. In contrast, CR cells showed a CSC-enriched phenotype with a high rate of ALDHhigh cells and high sphere-forming ability. In a previous study, a chemotherapy-resistant lung cancer subline showed a CSC-like phenotype with a high rate of ALDHhigh cells [20]. Thus, treatment resistance and CSCs might be an overlapping population in heterogenous cancer cells. Indeed, we found that CLSPN is overexpressed in UM-UC-3 CR cells, and our previous study revealed that CLSPN peptide is expressed in UM-UC-3 H10 cells, a CSC-like clone derived from UM-UC-3 cells [24]. CSCs are resistant to treatment by the expression of anti-apoptosis proteins or transporters [26]. CR cells also showed higher expression levels of the anti-apoptotic protein BIRC5 and transporter proteins including ABCB1, ABCB9, ABCB10, and ABCC1 than WT cells (Supplemental Table S1). A previous study revealed that CDDP-resistant or gemcitabine-resistant bladder cancer lines show high MDR1/ABCB1 expression and also cross-resistance to other chemotherapeutic reagents [27]. Thus, CR cells were resistant to CDDP and might be multi-drug resistant due to high ABCB1 expression; however, they were sensitive to CTLs. Thus, CR cells should be sensitive to granzyme B and perforin, which are the molecules used by CTLs for cytotoxicity, suggesting that the molecular mechanisms related to resistance for CDDP and CTLs are different.

We did not address the relationship between antiapoptotic or transporter proteins and resistance to CDDP. However, CLSPN knockdown increased sensitivity to CDDP; thus, CLSPN is a driver of CDDP resistance. A previous study revealed that CLSPN is preferentially expressed in CSCs of gastric cancer [22]. Thus, CLSPN might be related to CDDP resistance and the CSC phenotype. However, CLSPN knockdown did not reduce the ALDHhigh population in our study (data not shown); thus, CLSPN might not have role in the maintenance of ALDHhigh cells.

CLSPN is reported to mediate the phosphorylation and activation of CHK1 by ATR during DNA replicative stress in the S phase of the cell cycle [23]. In addition, p38 stress-activated kinase phosphorylates CLSPN and protects cells from DNA damage during the S phase [29, 30]. However, CLSPN was shown to be related to DNA replication stress and to protect cancer cells in an ATR-CHK1 checkpoint-independent manner [28]. Higher CLSPN mRNA expression is related to a poorer prognosis in lung cancer [28]. Other studies have revealed that CLSPN is overexpressed in gastric cancer, renal cell carcinoma, prostate cancer, bladder cancer, and low-grade glioma [22, 2932]. CLSPN protein expression is highest in the S phase, and in the G2/M phase, CLSPN is phosphorylated by PLK1 to induce protein degradation by the ubiquitin–proteasome system [23, 33]. Regarding this protein expression regulatory system, CLSPN protein expression is very low in normal tissues; however, it can be detected at higher levels in cancer tissues by immunohistochemical staining [22, 2931]. These findings suggest that CLSPN protein might be relatively more stable in cancer cells compared with normal cells. Considering its common molecular function in cell biology, CLSPN might be expressed in a wide variety of malignancies and CLSPN-targeting immunotherapy might be applicable for several malignancies.

Antigenic peptides are produced by proteasomal antigen protein processing [3436]. Fusion of the ubiquitin gene to an antigen gene enhances the CTL response [37]. Thus, CLSPN protein is a reasonable target for immunotherapy because it is degraded by the ubiquitin–proteasome system [23, 33]. In this study, we demonstrated that CLSPN expression was induced by 2 day stimulation with CDDP. This quick response will enable us to design further potential combination immunotherapies of CDDP and CLSPN-targeting immunotherapy including peptide vaccination, mRNA vaccination, and engineered T cell therapy. Furthermore, CDDP stimulation induces the expression of the immune checkpoint molecule PD-L1 in oral squamous cell carcinoma cells [37]. Thus, combination immunotherapy with CDDP might require PD1/PD-L1 blockade.

Taken together, these findings suggest that CLSPN is a driver of CDDP resistance in UM-UC-3 bladder cancer cells, and CLSPN can be targeted by CLSPN peptide-specific CTLs. This study provides support for the use of immunotherapy targeting CDDP-resistant cases and combination therapy of CDDP and immunotherapy.

Supplementary Information

Below is the link to the electronic supplementary material.

Acknowledgements

This work was supported by a KAKENHI grant from the Japan Society for the Promotion of Science (17H01540 to T. Torigoe and 20H03460 to Y. Hirohashi). This work was also supported by grants from the Japan Agency for Medical Research and Development’s Project for Cancer Research and Therapeutic Evolution (16770510 to T. Torigoe and 20cm0106352h0002 to T. Kanaseki), the Project for Promotion of Cancer Research and Therapeutic Evolution (22ama221317h0001 to Y. Hirohashi), and the Japan Science and Technology Agency’s CREST program (JPMJCR15G3 to S. Hashimoto).

Author contributions

All authors contributed to the study conception and design. Material preparation, data collection, and analysis were performed by SYHM, MI, KHYH, JY, AM, and ST. Data analysis was also performed by KM, TM, MN, SI, and SH. TA, TK, TT, TK, NS, and TT supervised the investigation. The first draft of the manuscript was written by SY, YH, NS, and TT, and all authors commented on previous versions of the manuscript. All authors read and approved the final manuscript.

Declarations

Conflicts of interest

The authors have no financial conflicts of interest to disclose.

Footnotes

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Contributor Information

Yoshihiko Hirohashi, Email: hirohash@sapmed.ac.jp.

Toshihiko Torigoe, Email: torigoe@sapmed.ac.jp.

References

  • 1.Saginala K, Barsouk A, Aluru JS, et al. Epidemiology of bladder cancer. Med Sci. 2020 doi: 10.3390/medsci8010015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209–249. doi: 10.3322/caac.21660. [DOI] [PubMed] [Google Scholar]
  • 3.Patel VG, Oh WK, Galsky MD. Treatment of muscle-invasive and advanced bladder cancer in 2020. CA Cancer J Clin. 2020;70:404–423. doi: 10.3322/caac.21631. [DOI] [PubMed] [Google Scholar]
  • 4.Berdik C. Unlocking bladder cancer. Nature. 2017;551:S34–S35. doi: 10.1038/551S34a. [DOI] [PubMed] [Google Scholar]
  • 5.Guallar-Garrido S, Julián E. Bacillus calmette-guérin (BCG) therapy for bladder cancer: an update. Immunotargets Ther. 2020;9:1–11. doi: 10.2147/ITT.S202006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Bellmunt J, de Wit R, Vaughn DJ, et al. Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N Engl J Med. 2017;376:1015–1026. doi: 10.1056/NEJMoa1613683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Galsky MD, Arija JÁA, Bamias A, et al. Atezolizumab with or without chemotherapy in metastatic urothelial cancer (IMvigor130): a multicentre, randomised, placebo-controlled phase 3 trial. Lancet. 2020;395:1547–1557. doi: 10.1016/S0140-6736(20)30230-0. [DOI] [PubMed] [Google Scholar]
  • 8.Powles T, Park SH, Voog E, et al. Avelumab maintenance therapy for advanced or metastatic urothelial carcinoma. N Engl J Med. 2020;383:1218–1230. doi: 10.1056/NEJMoa2002788. [DOI] [PubMed] [Google Scholar]
  • 9.Bajorin DF, Witjes JA, Gschwend JE, et al. Adjuvant nivolumab versus placebo in muscle-invasive urothelial carcinoma. N Engl J Med. 2021;384:2102–2114. doi: 10.1056/NEJMoa2034442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Inoue R, Hirohashi Y, Kitamura H, et al. GRIK2 has a role in the maintenance of urothelial carcinoma stem-like cells, and its expression is associated with poorer prognosis. Oncotarget. 2017;8:28826–28839. doi: 10.18632/oncotarget.16259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Hu Y, Smyth GK. ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. J Immunol Methods. 2009;347:70–78. doi: 10.1016/j.jim.2009.06.008. [DOI] [PubMed] [Google Scholar]
  • 12.Miyata H, Hirohashi Y, Yamada S, et al. GRIK2 is a target for bladder cancer stem-like cell-targeting immunotherapy. Cancer Immunol Immunother. 2022;71:795–806. doi: 10.1007/s00262-021-03025-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Morita R, Hirohashi Y, Torigoe T, et al. Olfactory receptor family 7 subfamily c member 1 is a novel marker of colon cancer-initiating cells and is a potent target of immunotherapy. Clin Cancer Res. 2016;22:3298–3309. doi: 10.1158/1078-0432.CCR-15-1709. [DOI] [PubMed] [Google Scholar]
  • 14.Haberle V, Forrest ARR, Hayashizaki Y, et al. CAGEr: precise TSS data retrieval and high-resolution promoterome mining for integrative analyses. Nucleic Acids Res. 2015;43:e51. doi: 10.1093/nar/gkv054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Frith MC, Valen E, Krogh A, et al. A code for transcription initiation in mammalian genomes. Genome Res. 2008;18:1–12. doi: 10.1101/gr.6831208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Yu G, Wang L-G, Han Y, He Q-Y. Clusterprofiler: an R package for comparing biological themes among gene clusters. OMICS. 2012;16:284–287. doi: 10.1089/omi.2011.0118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Morita R, Hirohashi Y, Nakatsugawa M, et al. Production of multiple CTL epitopes from multiple tumor-associated antigens. Methods Mol Biol. 2014;1139:345–355. doi: 10.1007/978-1-4939-0345-0_28. [DOI] [PubMed] [Google Scholar]
  • 18.Hirano N, Butler MO, Xia Z, et al. Engagement of CD83 ligand induces prolonged expansion of CD8+ T cells and preferential enrichment for antigen specificity. Blood. 2006;107:1528–1536. doi: 10.1182/blood-2005-05-2073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Matsuki M, Hirohashi Y, Nakatsugawa M, et al. Tumor-infiltrating CD8+ T cells recognize a heterogeneously expressed functional neoantigen in clear cell renal cell carcinoma. Cancer Immunol Immunother. 2022;71:905–918. doi: 10.1007/s00262-021-03048-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Barr MP, Gray SG, Hoffmann AC, et al. Generation and characterisation of cisplatin-resistant non-small cell lung cancer cell lines displaying a stem-like signature. PLoS ONE. 2013;8:e54193. doi: 10.1371/journal.pone.0054193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Vallo S, Michaelis M, Rothweiler F, et al. Drug-resistant urothelial cancer cell lines display diverse sensitivity profiles to potential second-line therapeutics. Transl Oncol. 2015;8:210–216. doi: 10.1016/j.tranon.2015.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Kobayashi G, Sentani K, Hattori T, et al. Clinicopathological significance of claspin overexpression and its association with spheroid formation in gastric cancer. Hum Pathol. 2019;84:8–17. doi: 10.1016/j.humpath.2018.09.001. [DOI] [PubMed] [Google Scholar]
  • 23.Peschiaroli A, Dorrello NV, Guardavaccaro D, et al. SCFbetaTrCP-mediated degradation of claspin regulates recovery from the DNA replication checkpoint response. Mol Cell. 2006;23:319–329. doi: 10.1016/j.molcel.2006.06.013. [DOI] [PubMed] [Google Scholar]
  • 24.Hirohashi Y, Torigoe T, Tsukahara T, et al. Immune responses to human cancer stem-like cells/cancer-initiating cells. Cancer Sci. 2016;107:12–17. doi: 10.1111/cas.12830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Prager BC, Xie Q, Bao S, Rich JN. Cancer stem cells: the architects of the tumor ecosystem. Cell Stem Cell. 2019;24:41–53. doi: 10.1016/j.stem.2018.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Park CY, Tseng D, Weissman IL. Cancer stem cell-directed therapies: recent data from the laboratory and clinic. Mol Ther. 2009;17:219–230. doi: 10.1038/mt.2008.254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Ulsamer A, Martínez-Limón A, Bader S, et al. Regulation of claspin by the p38 stress-activated protein kinase protects cells from DNA damage. Cell Rep. 2022;40:111375. doi: 10.1016/j.celrep.2022.111375. [DOI] [PubMed] [Google Scholar]
  • 28.Bianco JN, Bergoglio V, Lin Y-L, et al. Overexpression of claspin and timeless protects cancer cells from replication stress in a checkpoint-independent manner. Nat Commun. 2019;10:910. doi: 10.1038/s41467-019-08886-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Kobayashi G, Sentani K, Babasaki T, et al. Claspin overexpression is associated with high-grade histology and poor prognosis in renal cell carcinoma. Cancer Sci. 2020;111:1020–1027. doi: 10.1111/cas.14299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kobayashi G, Hayashi T, Sentani K, et al. Clinicopathological significance of claspin overexpression and its efficacy as a novel biomarker for the diagnosis of urothelial carcinoma. Virchows Arch. 2021 doi: 10.1007/s00428-021-03239-7. [DOI] [PubMed] [Google Scholar]
  • 31.Babasaki T, Sentani K, Sekino Y, et al. Overexpression of claspin promotes docetaxel resistance and is associated with prostate-specific antigen recurrence in prostate cancer. Cancer Med. 2021;10:5574–5588. doi: 10.1002/cam4.4113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Jia Y, Cheng X, Liang W, et al. CLSPN is a potential biomarker associated with poor prognosis in low-grade gliomas based on a multi-database analysis. Curr Res Transl Med. 2022;70:103345. doi: 10.1016/j.retram.2022.103345. [DOI] [PubMed] [Google Scholar]
  • 33.Smits VAJ, Cabrera E, Freire R, Gillespie DA. Claspin-checkpoint adaptor and DNA replication factor. FEBS J. 2019;286:441–455. doi: 10.1111/febs.14594. [DOI] [PubMed] [Google Scholar]
  • 34.Kloetzel PM. Antigen processing by the proteasome. Nat Rev Mol Cell Biol. 2001;2:179–187. doi: 10.1038/35056572. [DOI] [PubMed] [Google Scholar]
  • 35.Andersson HA, Barry MA. Maximizing antigen targeting to the proteasome for gene-based vaccines. Mol Ther. 2004;10:432–446. doi: 10.1016/j.ymthe.2004.05.035. [DOI] [PubMed] [Google Scholar]
  • 36.Park M-J, Kim E-K, Han J-Y, et al. Fusion of the human cytomegalovirus pp65 antigen with both ubiquitin and ornithine decarboxylase additively enhances antigen presentation to CD8(+) T cells in human dendritic cells. Hum Gene Ther. 2010;21:957–967. doi: 10.1089/hum.2009.216. [DOI] [PubMed] [Google Scholar]
  • 37.Sasaya T, Kubo T, Murata K, et al. Cisplatin-induced HSF1-HSP90 axis enhances the expression of functional PD-L1 in oral squamous cell carcinoma. Cancer Med. 2022 doi: 10.1002/cam4.5310. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials


Articles from Cancer Immunology, Immunotherapy : CII are provided here courtesy of Springer

RESOURCES