Comprehensive genomic profiling (CGP) is useful for optimizing targeted therapy and immunotherapy strategies for thoracic malignancies. This study aimed to evaluate the clinical utility and diagnostic complementarity of the in-house sequencing platform Rapid-Neo. We retrospectively analyzed 110 patients with thoracic malignancies who underwent Rapid-Neo testing. The baseline characteristics, sequencing results, concordance with companion diagnostics (CDx), and clinical outcomes were assessed. Of 110 patients, 100 (90.9%) had primary lung cancer. Rapid-Neo identified at least one genomic alteration in 99.1% of cases and well-established driver alterations in 66.0% of lung cancer cases. TMB-high and MSI-high statuses were observed in 9.0% and 2.0% of cases, respectively. Among the 90 cases with prior CDx, Rapid-Neo identified driver alterations in 10.0% of the cases, including EGFR, KRAS, MET, RET, and ERBB2, suggesting its potential to overcome the limitations of conventional CDx tests. High concordance (96.8%) was observed between the Rapid-Neo and CDx results, finally. In EGFR-mutant lung adenocarcinoma, high tumor mutation burden (TMB) was associated with a significantly shorter progression-free survival (PFS) after EGFR-TKI therapy (HR = 2.58, p = 0.018) and remained an independent prognostic factor in multivariate analysis. Furthermore, among patients receiving immune checkpoint inhibitors (ICIs), favorable genomic markers such as TMB-high or MSI-high were associated with prolonged PFS. Rapid-Neo demonstrated high sensitivity and concordance with CDx, while also identifying actionable driver alterations missed by the initial CDx. Moreover, the genomic markers identified by Rapid-Neo may provide predictive values for both targeted therapy and immunotherapy responses, supporting their integration into routine clinical decision-making.
综合基因组谱分析(CGP)有助于优化胸部恶性肿瘤的靶向治疗和免疫治疗策略。本研究旨在评估内部测序平台Rapid-Neo的临床应用和诊断互补性。我们回顾性分析了110例胸部恶性肿瘤患者,他们接受了快速neo检测。评估基线特征、测序结果、与伴随诊断(CDx)的一致性和临床结果。110例患者中,100例(90.9%)为原发性肺癌。Rapid-Neo在99.1%的病例中发现了至少一个基因组改变,在66.0%的肺癌病例中确定了驱动因素改变。tmb -高和msi -高分别占9.0%和2.0%。在90例既往CDx患者中,Rapid-Neo在10.0%的病例中发现了驱动因素改变,包括EGFR、KRAS、MET、RET和ERBB2,这表明它有潜力克服传统CDx检测的局限性。最后,Rapid-Neo与CDx结果高度一致(96.8%)。在egfr突变型肺腺癌中,高肿瘤突变负担(TMB)与EGFR-TKI治疗后显著缩短的无进展生存期(PFS)相关(HR = 2.58, p = 0.018),并且在多因素分析中仍然是一个独立的预后因素。此外,在接受免疫检查点抑制剂(ICIs)治疗的患者中,有利的基因组标记如tmb -高或msi -高与延长的PFS相关。Rapid-Neo显示了与CDx的高灵敏度和一致性,同时也识别了初始CDx遗漏的可操作的驱动改变。此外,Rapid-Neo识别的基因组标记可以为靶向治疗和免疫治疗反应提供预测价值,支持将其纳入常规临床决策。
{"title":"Clinical Application of In-House Comprehensive Genomic Profiling for Thoracic Cancer: Insights From a Japanese Hospital","authors":"Hatsuyo Takaoka, Hideki Terai, Kohei Nakamura, Takaaki Mizuno, Ryutaro Kawano, Katsura Emoto, Yutaka Kurebayashi, Nao Takada, Kenta Hamabe, Kazuhito Horie, Akihiko Ogata, Katsuhito Kinoshita, Lisa Shigematsu, Fumimaro Ito, Masahiko Okada, Takahiro Fukushima, Shigenari Nukaga, Testuo Tani, Keiko Ohgino, Kaoru Kaseda, Shinnosuke Ikemura, Hiroyuki Yasuda, Keisuke Asakura, Hajime Okita, Hiroshi Nishihara, Koichi Fukunaga","doi":"10.1111/cas.70168","DOIUrl":"10.1111/cas.70168","url":null,"abstract":"<p>Comprehensive genomic profiling (CGP) is useful for optimizing targeted therapy and immunotherapy strategies for thoracic malignancies. This study aimed to evaluate the clinical utility and diagnostic complementarity of the in-house sequencing platform Rapid-Neo. We retrospectively analyzed 110 patients with thoracic malignancies who underwent Rapid-Neo testing. The baseline characteristics, sequencing results, concordance with companion diagnostics (CDx), and clinical outcomes were assessed. Of 110 patients, 100 (90.9%) had primary lung cancer. Rapid-Neo identified at least one genomic alteration in 99.1% of cases and well-established driver alterations in 66.0% of lung cancer cases. TMB-high and MSI-high statuses were observed in 9.0% and 2.0% of cases, respectively. Among the 90 cases with prior CDx, Rapid-Neo identified driver alterations in 10.0% of the cases, including <i>EGFR</i>, <i>KRAS</i>, <i>MET</i>, <i>RET</i>, and <i>ERBB2</i>, suggesting its potential to overcome the limitations of conventional CDx tests. High concordance (96.8%) was observed between the Rapid-Neo and CDx results, finally. In EGFR-mutant lung adenocarcinoma, high tumor mutation burden (TMB) was associated with a significantly shorter progression-free survival (PFS) after EGFR-TKI therapy (HR = 2.58, <i>p</i> = 0.018) and remained an independent prognostic factor in multivariate analysis. Furthermore, among patients receiving immune checkpoint inhibitors (ICIs), favorable genomic markers such as TMB-high or MSI-high were associated with prolonged PFS. Rapid-Neo demonstrated high sensitivity and concordance with CDx, while also identifying actionable driver alterations missed by the initial CDx. Moreover, the genomic markers identified by Rapid-Neo may provide predictive values for both targeted therapy and immunotherapy responses, supporting their integration into routine clinical decision-making.</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"116 10","pages":"2819-2830"},"PeriodicalIF":4.3,"publicationDate":"2025-08-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cas.70168","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144776713","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Infiltration of resident memory T cells (TRMs) in the main tumor has been reported as a favorable prognostic factor. However, the role of TRMs in the lymph nodes (LNs) remains unclear. Thus, we examined the prognostic impact of TRMs infiltration within LNs of patients with gastric cancer (GC). Among 151 patients with metastasis to LN station No. 3, we classified them into two groups (CD103hi and CD103lo) based on the number of CD103+ T cells using immunohistochemical staining and analyzed the association between these groups and survival outcomes. We also examined the phenotype of CD8+ CD103+ T cells in the metastatic LNs using flow cytometry. Among patients with LN metastasis, metastasis to LN station No. 3 was significantly associated with a poor prognosis. There was a significant correlation between the number of CD8+ CD103+ T cells between the main lesion and the metastatic LNs. CD103hi was associated with a favorable prognosis (5-year overall survival [OS], log-rank p = 0.001; 5-year recurrence free survival [RFS], log-rank p = 0.001). Among adjuvant chemotherapy cases, patients with CD103hi exhibited significantly better OS and RFS than those with CD103lo (OS, log-rank p < 0.001; RFS, log-rank p < 0.001). Flow cytometry revealed that PD-1 expression in CD8+ CD103+ T cells was higher in metastatic than in normal LNs. Among patients with CD103hi, those with high PD-1 expression exhibited significantly better OS than those with low PD-1 expression. In conclusion, the infiltration of TRMs into LNs is a critical prognostic factor in GC.
常驻记忆T细胞(TRMs)在主要肿瘤的浸润已被报道为一个有利的预后因素。然而,TRMs在淋巴结(LNs)中的作用尚不清楚。因此,我们研究了胃癌(GC)患者ln内TRMs浸润对预后的影响。在151例LN 3号站转移患者中,我们采用免疫组化染色方法,根据CD103+ T细胞的数量将其分为CD103hi和CD103lo两组,并分析这两组患者与生存结果的关系。我们还使用流式细胞术检测了转移性LNs中CD8+ CD103+ T细胞的表型。在有淋巴结转移的患者中,转移到淋巴结3号站与预后不良显著相关。CD8+ CD103+ T细胞数量在主病变与转移灶之间有显著相关性。CD103hi与良好的预后相关(5年总生存率[OS], log-rank p = 0.001;5年无复发生存率(RFS), log-rank p = 0.001)。在辅助化疗病例中,CD103hi患者的OS和RFS明显优于CD103lo患者(OS, log-rank p + CD103+ T细胞的转移率高于正常LNs)。在CD103hi患者中,PD-1高表达患者的OS明显优于PD-1低表达患者。总之,TRMs向LNs的浸润是GC的关键预后因素。
{"title":"Resident Memory T Cell in Metastatic Lymph Nodes Is Associated With Favorable Prognosis in Gastric Cancer Patients","authors":"Masaki Nishiyama, Yuichiro Miki, Hiroaki Tanaka, Seiji Natsuki, Kenji Kuroda, Mami Yoshii, Tatsuro Tamura, Takahiro Toyokawa, Shigeru Lee, Kiyoshi Maeda","doi":"10.1111/cas.70163","DOIUrl":"10.1111/cas.70163","url":null,"abstract":"<p>Infiltration of resident memory T cells (TRMs) in the main tumor has been reported as a favorable prognostic factor. However, the role of TRMs in the lymph nodes (LNs) remains unclear. Thus, we examined the prognostic impact of TRMs infiltration within LNs of patients with gastric cancer (GC). Among 151 patients with metastasis to LN station No. 3, we classified them into two groups (CD103<sup>hi</sup> and CD103<sup>lo</sup>) based on the number of CD103<sup>+</sup> T cells using immunohistochemical staining and analyzed the association between these groups and survival outcomes. We also examined the phenotype of CD8<sup>+</sup> CD103<sup>+</sup> T cells in the metastatic LNs using flow cytometry. Among patients with LN metastasis, metastasis to LN station No. 3 was significantly associated with a poor prognosis. There was a significant correlation between the number of CD8<sup>+</sup> CD103<sup>+</sup> T cells between the main lesion and the metastatic LNs. CD103<sup>hi</sup> was associated with a favorable prognosis (5-year overall survival [OS], log-rank <i>p</i> = 0.001; 5-year recurrence free survival [RFS], log-rank <i>p</i> = 0.001). Among adjuvant chemotherapy cases, patients with CD103<sup>hi</sup> exhibited significantly better OS and RFS than those with CD103<sup>lo</sup> (OS, log-rank <i>p</i> < 0.001; RFS, log-rank <i>p</i> < 0.001). Flow cytometry revealed that PD-1 expression in CD8<sup>+</sup> CD103<sup>+</sup> T cells was higher in metastatic than in normal LNs. Among patients with CD103<sup>hi</sup>, those with high PD-1 expression exhibited significantly better OS than those with low PD-1 expression. In conclusion, the infiltration of TRMs into LNs is a critical prognostic factor in GC.</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"116 10","pages":"2688-2698"},"PeriodicalIF":4.3,"publicationDate":"2025-08-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cas.70163","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144776714","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Lung adenocarcinomas (LUADs) in never-smokers exhibit distinct molecular profiles from those of smokers, and their driver mutations are quite divergent. We aimed to evaluate the utility of RNA-seq for the molecular profiling of LUAD in Japanese never or light smokers. A hybridization capture-based RNA panel (TOP2-RNA) was used to confirm the validity of mutational and expression analyses of the panel in 122 Japanese LUAD cases. For the discovery cohort, 270 primary LUADs were molecularly profiled using TOP2-RNA. Whole transcriptome sequencing (WTS) was conducted for the samples without any oncogenic driver mutations. A risk score was developed using TOP2-RNA expression data to predict the prognosis of surgically resected LUAD. Driver oncogenes were identified in 180 cases (66.7%) of the discovery cohort. The frequency of MET ex14 skipping was high (12.6%) among cases without EGFR mutations. Actionable novel fusions of RDX-RASGRF1, PRKCI-RASGRF2, and OCLN-RASGRF2 were identified in three never-smoker cases by WTS. A functional assay identified that the expression of RASGRF fusions transformed the cells through phosphorylation of MEK, which was inhibited by cobimetinib treatment. High-risk patients defined by the risk score based on the four-gene signature had significantly worse RFS and OS for all stages and stage I patients in the discovery and validation cohorts. This study identified novel RASGRF1/2 fusions that might be targetable by MEK inhibitors. RNA-based molecular profiling could identify actionable mutations and assess the prognostic biomarkers for patient stratification to determine the optimal treatment based on the molecular profiling of individual LUAD cases.
{"title":"Discovery of Novel RASGRF2 Fusions as a Therapeutic Target in Lung Adenocarcinoma of Never or Light Smokers","authors":"Yuki Terashima, Soohwan Park, Hiroshi Ikeuchi, Takuo Hayashi, Shinya Kojima, Toshihide Ueno, Masachika Ikegami, Rina Kitada, Yoshiyuki Suehara, Shinya Tanaka, Kenji Suzuki, Hiroyuki Mano, Kazuya Takamochi, Shinji Kohsaka","doi":"10.1111/cas.70142","DOIUrl":"10.1111/cas.70142","url":null,"abstract":"<p>Lung adenocarcinomas (LUADs) in never-smokers exhibit distinct molecular profiles from those of smokers, and their driver mutations are quite divergent. We aimed to evaluate the utility of RNA-seq for the molecular profiling of LUAD in Japanese never or light smokers. A hybridization capture-based RNA panel (TOP2-RNA) was used to confirm the validity of mutational and expression analyses of the panel in 122 Japanese LUAD cases. For the discovery cohort, 270 primary LUADs were molecularly profiled using TOP2-RNA. Whole transcriptome sequencing (WTS) was conducted for the samples without any oncogenic driver mutations. A risk score was developed using TOP2-RNA expression data to predict the prognosis of surgically resected LUAD. Driver oncogenes were identified in 180 cases (66.7%) of the discovery cohort. The frequency of <i>MET</i> ex14 skipping was high (12.6%) among cases without <i>EGFR</i> mutations. Actionable novel fusions of <i>RDX-RASGRF1</i>, <i>PRKCI-RASGRF2,</i> and <i>OCLN-RASGRF2</i> were identified in three never-smoker cases by WTS. A functional assay identified that the expression of <i>RASGRF</i> fusions transformed the cells through phosphorylation of MEK, which was inhibited by cobimetinib treatment. High-risk patients defined by the risk score based on the four-gene signature had significantly worse RFS and OS for all stages and stage I patients in the discovery and validation cohorts. This study identified novel <i>RASGRF1/2</i> fusions that might be targetable by MEK inhibitors. RNA-based molecular profiling could identify actionable mutations and assess the prognostic biomarkers for patient stratification to determine the optimal treatment based on the molecular profiling of individual LUAD cases.</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"116 10","pages":"2868-2881"},"PeriodicalIF":4.3,"publicationDate":"2025-07-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cas.70142","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144745613","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The nucleolus, a prominent membrane-less nuclear compartment, is organized around ribosomal RNA (rRNA) gene (rDNA) clusters, known as nucleolar organizing regions (NORs), located on the short arms of acrocentric chromosomes. It serves as the primary site for ribosome biogenesis, an energy-intensive process crucial for cell growth and proliferation. This involves RNA polymerase I (Pol I)-mediated transcription of 47S precursor rRNA (pre-rRNA), pre-rRNA processing, and ribosomal subunit assembly, reflected in its tripartite structure maintained by liquid–liquid phase separation. Recent evidence indicates that only about 30% of nucleolar proteins are exclusively involved in ribosome production. The remaining proteome participates in diverse cellular functions, establishing the nucleolus as a multifunctional organelle. It functions as a critical stress sensor and signaling hub, responding to various intracellular insults such as nutrient starvation, DNA damage, and viral infection. Many chemotherapeutic agents also induce the response called nucleolar stress via disruption of the nucleolar structure or function, potentially leading to rDNA instability. Nucleolar stress frequently leads to dynamic transition of nucleolar proteins, inducing nucleolar reorganization. Of these, the stress induced by transcriptional changes leads to the unique nucleolar structures termed nucleolar caps and nucleolar necklaces. In this review, we summarize the recent findings about the molecular mechanism of nucleolar changes upon stresses and discuss the possible relationship between rDNA instability and cancer.
{"title":"Nucleolar Organization in Response to Transcriptional Stress","authors":"Rikiya Imamura, Takaaki Yasuhara","doi":"10.1111/cas.70164","DOIUrl":"10.1111/cas.70164","url":null,"abstract":"<p>The nucleolus, a prominent membrane-less nuclear compartment, is organized around ribosomal RNA (rRNA) gene (rDNA) clusters, known as nucleolar organizing regions (NORs), located on the short arms of acrocentric chromosomes. It serves as the primary site for ribosome biogenesis, an energy-intensive process crucial for cell growth and proliferation. This involves RNA polymerase I (Pol I)-mediated transcription of 47S precursor rRNA (pre-rRNA), pre-rRNA processing, and ribosomal subunit assembly, reflected in its tripartite structure maintained by liquid–liquid phase separation. Recent evidence indicates that only about 30% of nucleolar proteins are exclusively involved in ribosome production. The remaining proteome participates in diverse cellular functions, establishing the nucleolus as a multifunctional organelle. It functions as a critical stress sensor and signaling hub, responding to various intracellular insults such as nutrient starvation, DNA damage, and viral infection. Many chemotherapeutic agents also induce the response called nucleolar stress via disruption of the nucleolar structure or function, potentially leading to rDNA instability. Nucleolar stress frequently leads to dynamic transition of nucleolar proteins, inducing nucleolar reorganization. Of these, the stress induced by transcriptional changes leads to the unique nucleolar structures termed nucleolar caps and nucleolar necklaces. In this review, we summarize the recent findings about the molecular mechanism of nucleolar changes upon stresses and discuss the possible relationship between rDNA instability and cancer.</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"116 10","pages":"2649-2656"},"PeriodicalIF":4.3,"publicationDate":"2025-07-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cas.70164","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144734525","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
A low molecular compound originally developed as an anexelekto inhibitor, TP-0903, has been highlighted as a promising therapeutic agent for treating chronic lymphocytic leukemia, solid tumors, and drug-resistant AML. We investigated the in vitro effects of TP-0903 on a myelodysplastic syndrome (MDS)-derived cell line (MDS-L) and two myeloid leukemia cell lines. TP-0903 effectively inhibited cell proliferation and induced apoptosis in all three cell lines. In MDS-L cells, the PI3K/AKT and JAK/STAT3 pathways were inhibited, suggesting that this may be partly due to decreased direct interactions with hepatocyte growth factor receptor, commonly known as MET. Regarding its effect on the cell cycle, TP-0903 was found to impact the DNA damage response and cell cycle-related factors, particularly those centered around Aurora kinases. In MDS-L cells, inhibition of Aurora A phosphorylation led to decreased levels of BORA, which in turn suppressed polo-like kinase 1 activation. This suppression hindered mitosis initiation, resulting in cell cycle arrest at the G2/M phase. Additionally, chromosomal misregulation caused by Aurora A inhibition appeared to impair cell division and contribute to cell death. Gene expression profiling of MDS-L revealed changes in the ferroptosis-related genes, including HMOX1 and transferrin, along with elevated levels of reactive oxygen species and intracellular iron accumulation. These findings suggest the activation of an atypical ferroptosis pathway mediated through the TGF-β1/SMAD3 signaling pathway. Overall, these data indicate that TP-0903 may offer a novel therapeutic strategy for the treatment of refractory hematological malignancies.
{"title":"TP-0903 Suppresses Aurora A–PLK1 Signaling to Inhibit Proliferation of a Myelodysplastic Syndrome-Derived Cell Line","authors":"Tomoko Kimura-Hyoda, Mikuri Ryu, Ryosuke Yuta, Saori Fukumoto, Kentaro Hosokawa, Hisayuki Yao, Yoko Yasuda, Koichi Miura, Kaoru Tohyama, Fumio Arai, Takeshi Uchiumi","doi":"10.1111/cas.70151","DOIUrl":"10.1111/cas.70151","url":null,"abstract":"<p>A low molecular compound originally developed as an anexelekto inhibitor, TP-0903, has been highlighted as a promising therapeutic agent for treating chronic lymphocytic leukemia, solid tumors, and drug-resistant AML. We investigated the in vitro effects of TP-0903 on a myelodysplastic syndrome (MDS)-derived cell line (MDS-L) and two myeloid leukemia cell lines. TP-0903 effectively inhibited cell proliferation and induced apoptosis in all three cell lines. In MDS-L cells, the PI3K/AKT and JAK/STAT3 pathways were inhibited, suggesting that this may be partly due to decreased direct interactions with hepatocyte growth factor receptor, commonly known as MET. Regarding its effect on the cell cycle, TP-0903 was found to impact the DNA damage response and cell cycle-related factors, particularly those centered around Aurora kinases. In MDS-L cells, inhibition of Aurora A phosphorylation led to decreased levels of BORA, which in turn suppressed polo-like kinase 1 activation. This suppression hindered mitosis initiation, resulting in cell cycle arrest at the G2/M phase. Additionally, chromosomal misregulation caused by Aurora A inhibition appeared to impair cell division and contribute to cell death. Gene expression profiling of MDS-L revealed changes in the ferroptosis-related genes, including <i>HMOX1</i> and <i>transferrin</i>, along with elevated levels of reactive oxygen species and intracellular iron accumulation. These findings suggest the activation of an atypical ferroptosis pathway mediated through the TGF-β1/SMAD3 signaling pathway. Overall, these data indicate that TP-0903 may offer a novel therapeutic strategy for the treatment of refractory hematological malignancies.</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"116 10","pages":"2831-2845"},"PeriodicalIF":4.3,"publicationDate":"2025-07-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cas.70151","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144734526","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Photodynamic therapy (PDT) is a noninvasive anticancer treatment that uses a photosensitizer and light irradiation. PDT generates reactive oxygen species (ROS), thereby inducing tumor cell death. Stimulation of the interferon gene (STING) activation is highlighted as an immunotherapeutic strategy for cancer treatment. However, the role of STING and ROS in cancer therapy remains unclear. We hypothesized that STING regulates ROS generation in PDT, and that STING loss alters ROS homeostasis and causes therapeutic resistance. We established STING knockout (KO) HCT116 cells and compared the therapeutic efficacy of talaporfin sodium (TS)-PDT in KO and parental cells. Cell death induction was analyzed by 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide assay. ROS induction was analyzed using 2′,7′-dichlorofluorescin diacetate assay. STING-regulated gene activation was assessed by western blotting. Furthermore, the efficacy of STING agonist (2′-3′-cyclic GMP–AMP sodium and ADU-S100) and TS-PDT combination was assessed in a xenograft tumor model. STING KO suppressed cell death induced by TS-PDT (IC50 16.58 [±1.03] vs. 19.21 [±1.38] μmol/L). STING KO suppressed ROS generation of TS-PDT (mean fluorescence intensity, 4240 [±517.4] vs. 2234 [±551.9]). STING-dependent signaling was enhanced by TS-PDT, and these effects were eliminated by STING loss. The combination of STING agonist and TS-PDT exhibited significantly greater tumor growth inhibition than single therapy alone. STING is an important regulator of cellular ROS homeostasis and tumor cell susceptibility to ROS in PDT. Combining a STING agonist with PDT could enhance its therapeutic efficacy and may have potential for future clinical applications.
{"title":"cGAS–STING Pathway Activation Enhances Antitumor Effect of Talaporfin Photodynamic Therapy Through ROS Production","authors":"Makiko Sasaki, Mamoru Tanaka, Yasunari Sasaki, Yuki Kojima, Taketo Suzuki, Hirotada Nishie, Shigeki Fukusada, Naomi Sugimura, Keiji Ozeki, Takaya Shimura, Eiji Kubota, Hiromi Kataoka","doi":"10.1111/cas.70162","DOIUrl":"10.1111/cas.70162","url":null,"abstract":"<p>Photodynamic therapy (PDT) is a noninvasive anticancer treatment that uses a photosensitizer and light irradiation. PDT generates reactive oxygen species (ROS), thereby inducing tumor cell death. Stimulation of the interferon gene (STING) activation is highlighted as an immunotherapeutic strategy for cancer treatment. However, the role of STING and ROS in cancer therapy remains unclear. We hypothesized that STING regulates ROS generation in PDT, and that STING loss alters ROS homeostasis and causes therapeutic resistance. We established STING knockout (KO) HCT116 cells and compared the therapeutic efficacy of talaporfin sodium (TS)-PDT in KO and parental cells. Cell death induction was analyzed by 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide assay. ROS induction was analyzed using 2′,7′-dichlorofluorescin diacetate assay. STING-regulated gene activation was assessed by western blotting. Furthermore, the efficacy of STING agonist (2′-3′-cyclic GMP–AMP sodium and ADU-S100) and TS-PDT combination was assessed in a xenograft tumor model. STING KO suppressed cell death induced by TS-PDT (IC<sub>50</sub> 16.58 [±1.03] vs. 19.21 [±1.38] μmol/L). STING KO suppressed ROS generation of TS-PDT (mean fluorescence intensity, 4240 [±517.4] vs. 2234 [±551.9]). STING-dependent signaling was enhanced by TS-PDT, and these effects were eliminated by STING loss. The combination of STING agonist and TS-PDT exhibited significantly greater tumor growth inhibition than single therapy alone. STING is an important regulator of cellular ROS homeostasis and tumor cell susceptibility to ROS in PDT. Combining a STING agonist with PDT could enhance its therapeutic efficacy and may have potential for future clinical applications.</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"116 10","pages":"2677-2687"},"PeriodicalIF":4.3,"publicationDate":"2025-07-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cas.70162","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144734524","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
This letter argues that Japan's dependence on a single foreign company for classifying germline variants is delaying critical treatment for cancer patients. The author proposes establishing a national “Germline Expert Panel” to provide independent, timely oversight, and ensure patients receive appropriate care.