International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Cancer of unknown primary (CUP) is defined as histologically confirmed metastatic cancer in which the primary site cannot be found even after extensive standard investigations (1). At present, the favorable risk subgroup for CUP includes patients with neuroendocrine carcinomas of unknown primary, peritoneal adenocarcinomatosis of a serous papillary subtype, isolated axillary nodal metastases in female patients, squamous cell carcinoma involving non-supraclavicular cervical lymph nodes (LNs), single metastatic deposit from unknown primary, and men with blastic bone metastases and prostate-specific antigen expression (2). Novel favorable subsets of CUP have emerged, including colorectal, lung and renal CUP, which underlines the importance of cancer-specific treatments (2). Patients with CUP who are categorized into the unfavorable subset (~80% of patients) receive empiric chemotherapy with a platinum-taxane regimen. However, the prognosis remains poor, with a median survival period of 6–12 months (3,4). The poor prognosis of CUP is attributable to its clinical heterogeneity originating from various types of cancers, which makes a single empiric regimen inefficient (5).
Our previous randomized controlled study (6) assessed whether site-specific therapy based on prediction of the primary site may improve the outcomes in untreated patients with CUP. Genome-wide gene expression profiling of CUP was performed in the study using a microarray to predict the primary site in each patient with CUP (6). However, substantial shortcomings have been identified in the existing research comparing site-specific treatment and empiric chemotherapy. These deficiencies include patient accrual problems (oversampling treatment-resistant tumor types and long recruitment), study design limitations (observational and problematic trials), heterogeneity among the CUP classifiers (epigenetic vs. transcriptomic profiling) and incomparable therapies (7). Treatment based on the prediction of the primary site did not lead to any improvement of the overall survival compared with empiric chemotherapy in our previous study (6); however, gene profiling analyses of patients with CUP and comparison of the gene expression patterns in these patients with those in tumors from 24 known primary sites revealed several genes that were uniquely upregulated in CUP (8). Of the ~22,000 genes mounted on the microarray, 44 genes that were upregulated in CUPs were identified in our previous study (8).
The early metastasis in the natural history of CUP remains poorly understood. The present study identified genes that could serve a role in the metastasis in CUP. Small interfering RNA (siRNA) knockdown screens were used to assess the genes upregulated in the present CUP cohort to determine their impact on the migration of cancer cells and further characterize candidate genes using an in vivo metastasis model.
The A549 human lung cancer cell line (cat. no. CCL-185) and the MDA231 breast cancer cell line (cat. no. HTB-26) were procured from American Type Culture Collection, and cultured in DMEM (Thermo Fisher Scientific, Inc.) with 10% FBS (MilliporeSigma) in accordance with the instructions provided by the suppliers. The cells were maintained in a 5% CO2 humidified atmosphere at 37°C.
The proliferation rate of cultured cells was analyzed using MTT (MilliporeSigma) as previously described (9). The assay was performed in triplicate where applicable.
Total RNA (1 µg) from cells, extracted using ISOGEN (Nippon Gene Co., Ltd.), was converted into cDNA using a GeneAmp RNA-PCR kit (Thermo Fisher Scientific, Inc.). Reverse transcription was performed at 37°C for 2 h. qPCR was performed using TB Green Premix Ex Taq II (Takara Bio, Inc.), including TB Green as an intercalator that emits fluorescence when bound to double-stranded DNA. The thermocycling conditions were as follows: 95°C for 1 min and 50 cycles of 95°C for 5 sec and 60°C for 30 sec. Quantification based on the cycle threshold (Ct) values (2−ΔΔCq method) (10) was performed using an Applied Biosystems 7900 HT Fast Real-time PCR System (Thermo Fisher Scientific, Inc.) as previously described (11). The primers used to amplify the target genes are listed in Table SI. GAPDH was used to normalize the expression levels in quantitative analyses. For the siRNAs listed in Table I, the analyses were conducted once, but in triplicate in case the migration rate was <50%. For a few siRNAs that impaired the viability of the cells, it was not possible to perform the analyses (see below; Table I).
Total proteins were extracted from cells using RIPA buffer (FUJIFILM Wako Pure Chemical Corporation) containing protease inhibitor mix Complete™ (Roche Diagnostics). The protein concentrations were determined using a BCA Protein Assay Kit (Pierce; Thermo Fisher Scientific, Inc.). The proteins were boiled at 100°C, loaded (20 mg per lane), separated by SDS-PAGE (5–20%) and transferred to PVDF membranes. After 1 h of blocking with 3% (w/v) BSA (Merck KGaA) at room temperature, the membranes were incubated overnight with primary antibodies at 4°C. After being rinsed twice with TBS buffer (pH 8.0) containing 0.1% Tween-20, the membranes were incubated with HRP-labeled secondary antibodies against mouse IgG (100-fold dilution; cat. no. 7076; Cell Signaling Technology, Inc.) at room temperature for 1 h. An enhanced chemiluminescence solution (GE Healthcare) was used for color development. β-actin was used as the internal standard. The experiment was performed in triplicate. Antibodies against protein kinase DNA-activated catalytic subunit (PRKDC)/DNA-dependent protein kinase catalytic subunit (DNA-PK) (1,000-fold dilution; cat. no. 12311; Cell Signaling Technology, Inc.), proteasome subunit β type-4 (PSMB4) (200-fold dilution; cat. no. sc-390878; Santa Cruz Biotechnology, Inc.) and β-actin (200-fold dilution; cat. no. sc-47778; Santa Cruz Biotechnology, Inc.) were used. ImageJ software ver. 1.54 g (National Institutes of Health) was used for densitometry.
The migration assay was performed using the Boyden chamber method using polycarbonate membranes with a pore size of 8 µm (Chemotaxicell; Kurabo Bio-Medical Department; Kurabo Industries, Ltd.) as previously described (11). The membranes were coated with fibronectin (50 µg/ml) on the outer side at room temperature for 1 h and dried for 2 h at room temperature. The cells (2×104 cells/well) were then seeded into the upper chambers containing 500 µl migrating medium (DMEM containing 0.5% FBS), and the upper chambers were placed into the lower chambers containing 700 µl DMEM with 10% FBS. After incubation at 37°C for 24 h, the media in the upper chambers were aspirated, and the non-migrated cells on the inner sides of the membranes were removed using a cotton swab. The cells that had migrated to the outer side of the membranes were fixed with 100% ethanol at room temperature for 5 min, stained with 0.1% crystal violet solution at room temperature for 15 min and observed under a light microscope. The number of migrated cells in five fields per chamber was averaged. The migration rate was verified in triplicate for each gene where applicable (Table I).
The selection of genes was based on our previous expression microarray analysis for CUP (GSE42392) (8), which was compared with microarray datasets for several cancer types obtained from the Gene Expression Omnibus database (https://www.ncbi.nlm.nih.gov/geo/) (12). The datasets used were GSE781 (13), GSE1456 (14), GSE2109 (15), GSE2742 (16), GSE3149 (17), GSE3167 (18), GSE4127 (19), GSE4176 (20), GSE5787 (21), GSE6791 (22) and GSE8218 (23).
Predesigned siRNAs (MISSION® siRNA) targeting 50 genes and a nonspecific target (MISSION® siRNA Universal Negative Control; Table I) were purchased from MilliporeSigma. Cells were transfected with each siRNA (10 nM) using RNAiMAX (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions as previously described (24), incubated at 37°C for 48 h, and then trypsinized and seeded immediately for the migration assay.
shRNAs targeting PRKDC or PSMB4 were constructed using oligonucleotides encoding siRNA directed against the respective genes (5′-CCTGAAGTCTTTACAACATATCTC-3′ and 5′-CCGCAACATCTCTCGCATTATCTC-3′ for PRKDC and PSMB4 shRNA, respectively). The oligonucleotides were cloned into RNAi-Ready pSIREN RetroQZsGreen (Takara Bio, Inc.), which is a self-inactivating retroviral expression vector. This dual expression vector encodes green fluorescent protein (GFP) under the control of the cytomegalovirus promotor and shRNA under the control of the U6 promoter. The viral vectors constructed were designated as pSIREN-shPRKDC, pSIREN-shPSMB4 and a control vector (pSIREN-RetroQZsGreen vector without oligonucleotide inserted). Each of the pSIREN-RetroQZsGreen constructs (45 µg) was co-transfected with a pVSV-G vector (Takara Bio, Inc.) constituting the viral envelope (molar ratio of 3:1 for pSIREN-RetroQZsGreen constructs:pVSV-G vector) into GP2-293 packaging cells (2×107 cells; Takara Bio, Inc.) using the FuGENE6 transfection reagent (Roche Diagnostics). After 48 h at 37°C, the culture medium was collected, and the viral particles were concentrated by centrifugation at 15,000 × g for 3 h at 4°C. The viral pellet was then resuspended in fresh DMEM. The viral vector titer was calculated using a Retrovirus Titer Set (Takara Bio, Inc.) according to the manufacturer's instructions. The minimal MOI was then determined to be 4 for all the constructs by counting the GFP-positive A549 cells infected by serial dilutions of the virus-containing media. At 3 days after the infection with the virus in A549 cells (1×105 cells), the cells were trypsinized, propagated for 1 week and resuspended in DMEM (5×106 cells/ml). Subsequently, 1 ml of the resuspended solution was processed for cell sorting using a FACSCalibur Flow Cytometer (BD Biosciences). This device consists of a flow cytometer and a fluorescence activated cell sorter (FACS) in which GFP-positive cells excited by the 488 nm laser line emit green fluorescence at 509 nm, which is optimally detected by the FL-1 detector, and the GFP-positive cells were captured by a catcher tube for sorting (25). The software used for analysis was CellQuest software Ver. 3.3 (BD Biosciences). The subsequent experiment (LN metastasis assay) was performed immediately after the sorted cells reached the number required for inoculation into mice (1×107 cells for each construct) during the culture for ~1 week.
A total of 60 nude mice (BALB/c nu/nu; 5-week-old; female; mean weight, 17.8 g; CLEA Japan, Inc.) were used for the in vivo experiments. Experimemts were approved by the Institutional Animal Care Committee of the Kindai University Faculty of Medicine (approval no. KDMS-25-019; Osaka-Sayama, Japan). The mice were housed at the Animal Laboratory, Kindai University Faculty of Medicine (Osaka-Sayama, Japan), and maintained in a specific pathogen-free vivarium at 18–23°C with 50% humidity under a 12/12 h dark/light cycle. The animals had free access to drinking water and were fed a pelleted basal diet ad libitum.
The in vivo study consisted of two parts that estimated two effects on the potential of the cells to metastasize from footpad to popliteal LN: Experiment 1, the effect of downregulation of PSMB4 and PRKDC; and experiment 2, the effect of inhibitors of PSMB4 and PRKDC. For each of them, a single experiment was performed and 30 mice were used. For experiment 1, the cells (A549 cells bearing a control vector, pSIREN-shPRKDC, or pSIREN-shPSMB4) were resuspended in Hanks' Balanced Salt Solution (MilliporeSigma) and injected subcutaneously (1×106 cells) into the hind footpads of mice (10 mice per group) on day 0 in accordance with the method described for a previous study (26). On the final day (day 20), mice were euthanized using CO2 flowing from a compressed cylinder into a chamber (under ambient conditions) containing the mice so that 100% CO2 gradually filled the chamber (30%/min) (27). Next, the primary tumors and popliteal LNs were visualized by fluorescence imaging and images were captured using a macro-imaging station consisting of a SBIG cooled CCD camera model ST-7XME (Santa Barbara Imaging) mounted onto a dark box. The integrated density of the fluorescence spots was quantified using ImageJ software ver. 1.54 g (National Institutes of Health). For experiment 2, the DNA-PK inhibitor NU7441 (10 mg/kg; FUJIFILM Wako Pure Chemical Corporation) (28) and the proteasome inhibitor bortezomib (1 mg/kg; Takeda Pharmaceutical Company, Ltd.) (29), and vehicle (DMSO) were administered intravenously alone (9–10 mice/group) 1 week after inoculation of A549 cells bearing a control vector, and this administration was repeated twice a week for 3 weeks (2nd to 4th weeks). The LNs were resected (on day 35), and their maximum diameters were measured. Fluorescence images were also captured for the primary tumors and popliteal LNs. The animal details and protocol were otherwise as aforementioned.
Data are presented as the mean ± SD, unless otherwise noted. An unpaired t-test was used for comparisons between two groups. Comparisons among multiple groups were conducted using one-way ANOVA, followed by the Student-Newman-Keuls test. These analyses were performed as a single experiment. Statistical analyses were performed using SigmaPlot v13.0 (Grafiti LLC). P<0.05 was considered to indicate a statistically significant difference.
In our previous microarray study, 44 genes that were upregulated in CUPs compared with non-CUPs as a whole were identified (8). Of the 44 genes, 40 were selected as candidates for the next screening using the migration assay (Table I). Three pseudogene-like genes (LOC392501, LOC442171 and LOC646417) with unknown functions and one gene without an available siRNA (LAPTM5) were excluded. As additional candidates for the screening, 10 genes that were screened out by the comparison of gene expression profiles between CUP and individual cancer types of known primary site were selected (8). These 10 genes were most highly expressed in CUP of all cancer types examined, and each of the genes showed ≥1.5-fold enriched expression in CUP compared with the second most enriched cancer type for each gene (Table SII). A total of 50 genes were selected (Table I) for the migration assay.
For all 50 aforementioned genes, specific siRNAs were obtained from the same commercial source (MISSION® siRNA; Table I). For screening, A549 cells were transfected with siRNAs, and genes were evaluated for suppressed migration after siRNA knockdown. Images for cell migration assays are shown in Figs. 1 and S1. The migration assay showed that A549 cells transfected with some siRNAs exhibited poor migration, meaning the number of A549 cells migrating to the outer side of the porous membrane in the Boyden chamber was decreased compared with that of the cells transfected with control siRNA (Fig. 1A). For each of the 50 siRNAs transfected into A549 cells, the present study aimed to determine the cell viability, the fold-change in the expression of the targeted gene and the cell migration rate (Table I). The results revealed that transfection with siRNAs targeting SERF2, PRKDC, SH3GLB1, CAPNS1, PSMB4, RPS7 and RPL11 reduced the migration of A549 cells by >50% (Fig. 1A; Table I).
Our previous study identified six genes encoding ribosomal proteins (RPLP2, RPL18A, RPL36, RPS10, RPS7 and RPL11) as CUP-associated (−upregulated) genes (8). siRNAs against four of these ribosomal protein genes (RPLP2, RPL18A, RPL36 and RPS10) almost completely impaired the viability of A549 cells, and it was not possible to perform the expression assay and migration assay using siRNAs of these genes (Table I). The other two siRNAs (targeting RPS7 and RPL11) abolished the viability of MDA231 cells (Table I), indicating that reduced expression of any single ribosomal protein molecule could be critical for cell survival.
The present study subsequently focused on the siRNAs against the five genes (SERF2, PRKDC, SH3GLB1, CAPNS1 and PSMB4) based on their ability to reduce migration to less than half of that of the control without affecting the cell viability (>50% of the control; Table I). The present study further analyzed their influence on the migration of another cell line (MDA231). Only siRNAs specific for PRKDC and PSMB4 markedly reduced the migration of the cells (Fig. 1B; Table I). These siRNAs did not affect the viability of MDA231 cells, resulting in a cell viability rate of 75.2% for PRKDC-specific siRNA and >90% for PSMB4-specific siRNA. Furthermore, a marked reduction in the expression of the respective genes was observed. The siRNAs targeting PRKDC and PSMB4 reduced the expression levels of the corresponding genes (0.107- and 0.033-fold changes, respectively), which was similar to the trends observed for A549 cells (Table I). Western blotting confirmed that the expression of PRKDC and PSMB4 was reduced by the siRNA-induced mRNA knockdown (Fig. 2).
To determine whether downregulation of the two genes, PRKDC and PSMB4, might affect the potential of the cells to metastasize to the LNs in vivo, cell lines that stably expressed both GFP and shRNA targeting the respective genes were engineered. A control cell line transfected with a control vector that expressed GFP alone was also established. The stable cell lines for these constructs were isolated by FACS using GFP as a reporter (Fig. S2A-C) and they showed stable shRNA-based knockdown of PRKDC and PSMB4 during the passage culture for ≥3 months. Western blotting confirmed that the PRKDC and PSMB4 proteins expressed in A549 cells were reduced for the respective constructs compared with the control vector (Fig. S3A). The growth rate remained unaltered in the A549 cells transfected with shRNA targeting PSMB4 (>90% of the rate in the control cells), but the cells transfected with shRNA targeting PRKDC exhibited an ~25% reduction in the growth rate relative to that in the control cells at 72 h (Fig. S3B). To evaluate the metastasis-promoting potential of these two genes, cells (1×106) containing the respective shRNA constructs were subcutaneously injected into the footpads of BALB/c nude mice. On day 20, the fluorescence intensities of GFP expressed in the footpad and popliteal LNs were measured (Fig. 3A). The fluorescence intensities were lower after injection of cells expressing PRKDC-shRNA than after injection of cells transfected with a control vector (~14.8%), whereas fluorescence intensities following injection of the cells expressing PSMB4-shRNA were comparable to those following injection of cells containing the control vector (Fig. 3B and C). A proportionate reduction in fluorescence intensities was also observed in the popliteal LNs in both the PRKDC-shRNA and PSMB4-shRNA groups, with popliteal LN/footpad fluorescence intensity ratios of 39.4 and 45.5%, respectively, compared with the control group (Fig. 3C). It was attempted to perform a similar assay using MDA231-shRNA constructs; however, this cell line failed to become engrafted into the footpads of the mice (data not shown).
The present study subsequently examined whether known inhibitors of PRKDC (DNA-PK) or PSMB4 (proteasome) could alter the migration of cells. The inhibitors were injected intravenously into the tails of mice that had been inoculated with A549 cells harboring a control vector expressing GFP. NU7441, a DNA-PK inhibitor, did not significantly change the fluorescence intensity in either the footpads or popliteal LNs (Fig. 4A and B). By contrast, bortezomib, a proteasome inhibitor, markedly retarded the migration of the cells to the popliteal LNs, although the cell growth rate in the footpads was comparable to that in the control animals, resulting in a lower popliteal/footpad fluorescence intensity ratio compared with that in the control group (P=0.093) and in the animals injected with NU7441 (P=0.017) (Fig. 4B).
A marked difference in the size of the metastatic LNs was observed between the groups that were and were not treated with bortezomib (Fig. 4C). Compared with the control group (vehicle), the bortezomib-treated group showed significantly smaller (undeveloped) tumors at the metastatic sites (LNs; P<10−4). The mean LN maximum diameters in the group treated with vehicle and in the group treated with NU7447 (DNA-PK inhibitor) were 6.3- and 4.5-fold greater than the diameter of intact LNs, whereas they were only 1.4-fold greater in the group treated with bortezomib.
Our previous study analyzed tumor mRNA samples from 60 patients with CUP using microarray analysis and constructed a normalized gene expression profile specific to CUPs, which identified a number of genes that were upregulated in the CUP (8).
In the present study, to further narrow down the genes closely related to the development of CUP among these candidate genes, cell-based siRNA screening was performed in vitro. A549 and MDA231 cells were selected for the screening because they are among the most widely used cell lines as models for research on the metastasis of lung cancer and breast cancer, respectively (30,31). Furthermore, our previous study demonstrated that the gene expression profile of CUP closely resembled that of lung adenocarcinoma (8).
Individual knockdown of several candidate genes in A549 or MDA231 cells using specific siRNAs resulted in restricted migration of the cells. siRNAs against PRKDC and PSMB4 restricted the migration in both cell lines, suggesting that these genes might be involved in the metastatic ability of CUP. Furthermore, shRNAs for PRKDC and PSMB4 also suppressed the migration of A549 cells in vivo, with significance observed for shRNA for PRKDC, whereas knockdown of PRKDC tended to slow the growth of the cells as well. The PRKDC gene encodes DNA-PKcs, a large subunit (~469 KDa) of DNA-PK, which is an abundantly expressed kinase in higher eukaryotes (32). As a DNA damage repair protein, it drives several pathways that promote metastasis and tumor growth (33,34). In melanoma cells, DNA-PKcs may control the secretion of numerous proteins involved in metastasis, such as matrix metalloproteinases, thereby regulating the tumor microenvironment (35). A similar regulation in favor of metastasis could occur in CUPs exhibiting upregulation of DNA-PKcs (8).
PSMB4 encodes the β7 subunit of proteasome 20S. The 20S proteasome is the catalytic core of the proteasome complex with a concentric circular structure, including two α rings and two β rings, each ring consisting of 7 subunits, α1-7 and β1-7, respectively (36). Proteasomes affect tumor development by regulating tumor signaling pathways, including NF-κB signaling. They recognize and degrade ubiquitinated IκB, an inhibitor of NF-κB, thereby activating NF-κB signaling (37). Proteasomes are also required to maintain homeostasis in proliferating tumor cells by eliminating the accumulation of misfolded proteins (38). In multiple myeloma (MM), plasma cells secrete several immunoglobulins, which are macromolecules that are synthesized and folded in the endoplasmic reticulum (ER) (36). Therefore, proteasome inhibitors that are widely used in the treatment of MM can block the degradation of IκB and trigger the accumulation of misfolded proteins, inhibiting NF-κB activity and inducing ER stress, respectively, ultimately leading to cell death of the MM cells (39,40). Furthermore, DNA-PKcs is an additional target that is cleaved and inactivated by proteasome inhibitors in MM cells (40,41).
In the present study, bortezomib, the first proteasome inhibitor developed as a chemotherapy drug (29), markedly restricted the metastatic ability of A549 cells in vivo. The target of this drug has been identified as the β5 subunit of proteasome 20S (39). Subunits other than the β7 subunit (encoded by PSMB4) include the β5 subunit, which was also upregulated in both CUPs (GSE42392) (8) and A549 cells compared with small airway epithelial cells (GSE4824) (42) (Table SIII). The trend was also noted in other tumors located in the liver or head and neck regions (Table SIII), with each of these tumors identified as a potential treatment target for bortezomib (36,37).
The development of immune checkpoint inhibitors (ICIs) has markedly changed the treatment paradigms for numerous cancer types. Among patients with CUP, 28% exhibit one or more predictive biomarkers for ICIs. For example, programmed death-ligand 1 is expressed on ≥5% tumor cells in 22.5% or on lymphocytes in 58.7% of such patients. Microsatellite instability-high is observed in 1.8% and tumor mutational burden (TMB) ≥17 mutations per megabase of the tumor genome is present in 11.8% of these patients (43).
Although these biomarkers have not yet been validated in patients with CUP, those with CUP with TMB >10 mutations per megabase generally experience improved outcomes when treated with ICIs (43), and our previous study recently demonstrated the clinical benefits of nivolumab in patients with CUP (44). The present study demonstrated a putative role for the proteasome in the progression of CUP that could be prevented by proteasome-targeted therapy. Proteasome inhibitors combined with ICIs may be an additional therapeutic option for CUP, for which there are limited treatment options.
The authors would like to thank Mrs. Tomoko Kitayama (Department of Genome Biology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan) and Mr. Kentaro Egawa (Center for Animal Experiment, Kindai University Faculty of Medicine, Osaka-Sayama, Japan) for help with the animal experiments.
The present study was supported by the Grant-in-Aid for Scientific Research (C) (grant no. 17K07204) of Ministry of Education, Culture, Sports, Science and Technology of Japan, and by the Japan Agency for Medical Research and Development (grant no. 201438137A).
The data generated in the present study may be requested from the corresponding author.
YF designed the study and drafted the manuscript. YF and MADV performed the experiments and collected data. YF, MADV, HH, KNa and KNi analyzed and interpreted the data, and revised the manuscript. YF and MADV confirmed the authenticity of all the raw data. All authors have read and approved the final version of the manuscript.
The present study was approved by the Institutional Animal Care Committee of the Kindai University Faculty of Medicine (approval no. KDMS-25-019; Osaka-Sayama, Japan) and carried out in compliance with the standards for the use of laboratory animals.
Not applicable.
The authors declare that they have no competing interests.
|
CUP |
cancer of unknown primary |
|
PRKDC |
protein kinase DNA-activated catalytic subunit |
|
PSMB4 |
proteasome subunit β type-4 |
|
Pavlidis N and Pentheroudakis G: Cancer of unknown primary site. Lancet. 379:1428–1435. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Rassy E, Parent P, Lefort F, Boussios S, Baciarello G and Pavlidis N: New rising entities in cancer of unknown primary: Is there a real therapeutic benefit? Crit Rev Oncol Hematol. 147:1028822020. View Article : Google Scholar : PubMed/NCBI | |
|
Greco FA and Pavlidis N: Treatment for patients with unknown primary carcinoma and unfavorable prognostic factors. Semin Oncol. 36:65–74. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Pavlidis N, Khaled H and Gaafar R: A mini review on cancer of unknown primary site: A clinical puzzle for the oncologists. J Adv Res. 6:375–382. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Fizazi K, Greco FA, Pavlidis N, Daugaard G, Oien K and Pentheroudakis G; ESMO Guidelines Committee, : Cancers of unknown primary site: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 26 (Suppl 5):v133–v138. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Hayashi H, Kurata T, Takiguchi Y, Arai M, Takeda K, Akiyoshi K, Matsumoto K, Onoe T, Mukai H, Matsubara N, et al: Randomized phase II trial comparing site-specific treatment based on gene expression profiling with carboplatin and paclitaxel for patients with cancer of unknown primary site. J Clin Oncol. 37:570–579. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Rassy E, Labaki C, Chebel R, Boussios S, Smith-Gagen J, Greco FA and Pavlidis N: Systematic review of the CUP trials characteristics and perspectives for next-generation studies. Cancer Treat Rev. 107:1024072022. View Article : Google Scholar : PubMed/NCBI | |
|
Kurahashi I, Fujita Y, Arao T, Kurata T, Koh Y, Sakai K, Matsumoto K, Tanioka M, Takeda K, Takiguchi Y, et al: A microarray-based gene expression analysis to identify diagnostic biomarkers for unknown primary cancer. PLoS One. 8:e632492013. View Article : Google Scholar : PubMed/NCBI | |
|
Arao T, Fukumoto H, Takeda M, Tamura T, Saijo N and Nishio K: Small in-frame deletion in the epidermal growth factor receptor as a target for ZD6474. Cancer Res. 64:9101–9104. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Tanaka K, Arao T, Maegawa M, Matsumoto K, Kaneda H, Kudo K, Fujita Y, Yokote H, Yanagihara K, Yamada Y, et al: SRPX2 is overexpressed in gastric cancer and promotes cellular migration and adhesion. Int J Cancer. 124:1072–1080. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Clough E and Barrett T: The gene expression omnibus database. Methods Mol Biol. 1418:93–110. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Lenburg ME, Liou LS, Gerry NP, Frampton GM, Cohen HT and Christman MF: Previously unidentified changes in renal cell carcinoma gene expression identified by parametric analysis of microarray data. BMC Cancer. 3:312003. View Article : Google Scholar : PubMed/NCBI | |
|
Pawitan Y, Bjöhle J, Amler L, Borg AL, Egyhazi S, Hall P, Han X, Holmberg L, Huang F, Klaar S, et al: Gene expression profiling spares early breast cancer patients from adjuvant therapy: Derived and validated in two population-based cohorts. Breast Cancer Res. 7:R953–R964. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
International Genetics Consortium, . Expression project for oncology (expO). Gene Expression Omnibus, GSE2109. 2005.Available from:. http://www.intgen.org/ | |
|
Luesch H, Chanda SK, Raya RM, DeJesus PD, Orth AP, Walker JR, Belmonte JCI and Schultz PG: A functional genomics approach to the mode of action of apratoxin A. Nat Chem Biol. 2:158–167. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Bild AH, Yao G, Chang JT, Wang Q, Potti A, Chasse D, Joshi NR, Harpole D, Lancaster JM, Berchuck A, et al: Oncogenic pathway signatures in human cancers as a guide to targeted therapies. Nature. 439:353–357. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Dyrskjøt L, Kruhøffer M, Thykjaer T, Marcussen N, Jensen JL, Møller K and Ørntoft TF: Gene expression in the urinary bladder: A common carcinoma in situ gene expression signature exists disregarding histopathological classification. Cancer Res. 64:4040–4048. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Gemma A, Li C, Sugiyama Y, Matsuda K, Seike Y, Kosaihira S, Minegishi Y, Noro R, Nara M, Seike M, et al: Anticancer drug clustering in lung cancer based on gene expression profiles and sensitivity database. BMC Cancer. 6:1742006. View Article : Google Scholar : PubMed/NCBI | |
|
Rinaldi A, Kwee I, Taborelli M, Largo C, Uccella S, Martin V, Poretti G, Gaidano G, Calabrese G, Martinelli G, et al: Genomic and expression profiling identifies the B-cell associated tyrosine kinase Syk as a possible therapeutic target in mantle cell lymphoma. Br J Haematol. 132:303–316. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Bachtiary B, Boutros PC, Pintilie M, Shi W, Bastianutto C, Li JH, Schwock J, Zhang W, Penn LZ, Jurisica I, et al: Gene expression profiling in cervical cancer: An exploration of intratumor heterogeneity. Clin Cancer Res. 12:5632–5640. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Pyeon D, Newton MA, Lambert PF, den Boon JA, Sengupta S, Marsit CJ, Woodworth CD, Connor JP, Haugen TH, Smith EM, et al: Fundamental differences in cell cycle deregulation in human papillomavirus-positive and human papillomavirus-negative head/neck and cervical cancers. Cancer Res. 67:4605–4619. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, Xia XQ, Jia Z, Sawyers A, Yao H, Wang-Rodriquez J, Mercola D and McClelland M: In silico estimates of tissue components in surgical samples based on expression profiling data. Cancer Res. 70:6448–6455. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Kaneda H, Arao T, Tanaka K, Tamura D, Aomatsu K, Kudo K, Sakai K, De Velasco MA, Matsumoto K, Fujita Y, et al: FOXQ1 is overexpressed in colorectal cancer and enhances tumorigenicity and tumor growth. Cancer Res. 70:2053–2063. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Picot J, Guerin CL, Le Van Kim C and Boulanger CM: Flow cytometry: Retrospective, fundamentals and recent instrumentation. Cytotechnology. 64:109–130. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Chen Z, Zhuo W, Wang Y, Ao X and An J: Down-regulation of layilin, a novel hyaluronan receptor, via RNA interference, inhibits invasion and lymphatic metastasis of human lung A549 cells. Biotechnol Appl Biochem. 50:89–96. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Kura Y, De Velasco MA, Sakai K, Uemura H, Fujita K and Nishio K: Exploring the relationship between ulcerative colitis, colorectal cancer, and prostate cancer. Hum Cell. 37:1706–1718. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao Y, Thomas HD, Batey A, Cowell IG, Richardson CJ, Griffin RJ, Calvert AH, Newell DR, Smith GCM and Curtin NJ: Preclinical evaluation of a potent novel DNA-dependent protein kinase inhibitor NU7441. Cancer Res. 66:5354–5362. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Chen D, Frezza M, Schmitt S, Kanwar J and Dou QP: Bortezomib as the first proteasome inhibitor anticancer drug: Current status and future perspectives. Curr Cancer Drug Targets. 11:239–253. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Lengrand J, Pastushenko I, Vanuytven S, Song Y, Venet D, Sarate RM, Bellina M, Moers V, Boinet A, Sifrim A, et al: Pharmacological targeting netrin-1 inhibits EMT in cancer. Nature. 620:402–408. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Liu K, Newbury PA, Glicksberg BS, Zeng WZD, Paithankar S, Andrechek ER and Chen B: Evaluating cell lines as models for metastatic breast cancer through integrative analysis of genomic data. Nat Commun. 10:21382019. View Article : Google Scholar : PubMed/NCBI | |
|
Dylgjeri E and Knudsen KE: DNA-PKcs: A targetable protumorigenic protein kinase. Cancer Res. 82:523–533. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Xiang Z, Hou G, Zheng S, Lu M, Li T, Lin Q, Liu H, Wang X, Guan T, Wei Y, et al: ER-associated degradation ligase HRD1 links ER stress to DNA damage repair by modulating the activity of DNA-PKcs. Proc Natl Acad Sci USA. 121:e24030381212024. View Article : Google Scholar : PubMed/NCBI | |
|
Caron P, Pankotai T, Wiegant WW, Tollenaere MAX, Furst A, Bonhomme C, Helfricht A, de Groot A, Pastink A, Vertegaal ACO, et al: WWP2 ubiquitylates RNA polymerase II for DNA-PK-dependent transcription arrest and repair at DNA breaks. Genes Dev. 33:684–704. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Kotula E, Berthault N, Agrario C, Lienafa MC, Simon A, Dingli F, Loew D, Sibut V, Saule S and Dutreix M: DNA-PKcs plays role in cancer metastasis through regulation of secreted proteins involved in migration and invasion. Cell Cycle. 14:1961–1972. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou X, Xu R, Wu Y, Zhou L and Xiang T: The role of proteasomes in tumorigenesis. Genes Dis. 11:1010702023. View Article : Google Scholar : PubMed/NCBI | |
|
Narayanan S, Cai CY, Assaraf YG, Guo HQ, Cui Q, Wei L, Huang JJ, Ashby CR Jr and Chen ZS: Targeting the ubiquitin-proteasome pathway to overcome anti-cancer drug resistance. Drug Resist Updat. 48:1006632020. View Article : Google Scholar : PubMed/NCBI | |
|
Chhabra S: Novel proteasome inhibitors and histone deacetylase inhibitors: Progress in myeloma therapeutics. Pharmaceuticals (Basel). 10:402017. View Article : Google Scholar : PubMed/NCBI | |
|
Gandolfi S, Laubach JP, Hideshima T, Chauhan D, Anderson KC and Richardson PG: The proteasome and proteasome inhibitors in multiple myeloma. Cancer Metastasis Rev. 36:561–584. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Hideshima T and Anderson KC: Biologic impact of proteasome inhibition in multiple myeloma cells-from the aspects of preclinical studies. Semin Hematol. 49:223–227. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Wu YH, Hong CW, Wang YC, Huang WJ, Yeh YL, Wang BJ, Wang YJ and Chiu HW: A novel histone deacetylase inhibitor TMU-35435 enhances etoposide cytotoxicity through the proteasomal degradation of DNA-PKcs in triple-negative breast cancer. Cancer Lett. 400:79–88. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Byers LA, Diao L, Wang J, Saintigny P, Girard L, Peyton M, Shen L, Fan Y, Giri U, Tumula PK, et al: An epithelial-mesenchymal transition gene signature predicts resistance to EGFR and PI3K inhibitors and identifies Axl as a therapeutic target for overcoming EGFR inhibitor resistance. Clin Cancer Res. 19:279–290. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Rassy E, Boussios S and Pavlidis N: Genomic correlates of response and resistance to immune checkpoint inhibitors in carcinomas of unknown primary. Eur J Clin Invest. 51:e135832021. View Article : Google Scholar : PubMed/NCBI | |
|
Tanizaki J, Yonemori K, Akiyoshi K, Minami H, Ueda H, Takiguchi Y, Miura Y, Segawa Y, Takahashi S, Iwamoto Y, et al: Open-label phase II study of the efficacy of nivolumab for cancer of unknown primary. Ann Oncol. 33:216–226. 2022. View Article : Google Scholar : PubMed/NCBI |