201
|
Zheng W, Ge Z, Wu Q, Wan H, Sun J, Nai Y, Lv C. Olaparib Combined with Anti-PD1 Enhances Immunotherapy of Gastric Cancer Via NF-κB/c-Myc/PD-L1 Signaling. Dig Dis Sci 2025:10.1007/s10620-025-09021-y. [PMID: 40237904 DOI: 10.1007/s10620-025-09021-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND PARP inhibitors, effective in BRCA-mutated cancers, show potential in gastric cancer (GC) where homologous recombination defects (e.g., BRCA1/2 mutations) are common. Olaparib, a PARP inhibitor, upregulates PD-L1, suggesting synergy with PD-1 inhibitors for enhanced GC therapy. METHODS Using CCK-8 screening of 867 drugs, olaparib demonstrated potent GC cell inhibition. Western blot and qRT-PCR assessed PD-L1, c-MYC, COX-2, and NF-κB pathway proteins (p65/p-p65). Functional assays (Transwell, wound healing, colony formation) evaluated olaparib's effects on GC cell proliferation, migration, and invasion. A GC mouse model tested olaparib combined with anti-PD1. TCGA and Kaplan-Meier analyzed PARP expression-prognosis correlations. RESULTS Olaparib suppressed GC cell proliferation, migration, and invasion in vitro. Western blot revealed upregulated c-MYC, COX-2, p65, p-p65, and PD-L1, confirmed by qRT-PCR for PD-L1. Low PARP expression correlated with better GC patient survival. In vivo, olaparib synergized with anti-PD1 to enhance tumor suppression. CONCLUSION Olaparib activates the NF-κB/c-MYC pathway to elevate PD-L1, supporting its combination with PD-1 inhibitors as a promising GC therapeutic strategy.
Collapse
Affiliation(s)
- Wubin Zheng
- Department of General Surgery, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, 68 Changle Rd., Nanjing, 210006, Jiangsu, China
| | - Zhifa Ge
- Department of General Surgery, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, 68 Changle Rd., Nanjing, 210006, Jiangsu, China
| | - Qingwei Wu
- Department of General Surgery, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, 68 Changle Rd., Nanjing, 210006, Jiangsu, China
| | - Haoyue Wan
- Department of General Surgery, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, 68 Changle Rd., Nanjing, 210006, Jiangsu, China
| | - Junjie Sun
- Department of General Surgery, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, 68 Changle Rd., Nanjing, 210006, Jiangsu, China
| | - Yongjun Nai
- Department of General Surgery, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, 68 Changle Rd., Nanjing, 210006, Jiangsu, China.
| | - Chengyu Lv
- Department of General Surgery, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, 68 Changle Rd., Nanjing, 210006, Jiangsu, China
| |
Collapse
|
202
|
Ashaye A, Shi L, Aldoss I, Montesinos P, Vachhani P, Rocha V, Papayannidis C, Leonard JT, Baer MR, Ribera JM, McCloskey J, Wang J, Gao S, Rane D, Guo S. Patient-reported outcomes in Philadelphia chromosome-positive acute lymphoblastic leukemia patients treated with ponatinib or imatinib: results from the PhALLCON trial. Leukemia 2025:10.1038/s41375-025-02608-4. [PMID: 40240572 DOI: 10.1038/s41375-025-02608-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/27/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025]
Abstract
In the Phase 3 PhALLCON trial (NCT03589326), ponatinib demonstrated superior efficacy and comparable safety profile versus imatinib in adults with newly diagnosed Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL). Here we report patient-reported outcomes (PRO) from PhALLCON assessed as exploratory endpoints using the Functional Assessment of Cancer Therapy-Leukemia (FACT-Leu) and EQ-5D-5L. Primary PRO domains included FACT-G physical well-being, FACT-Leu subscale (FACT-LeuS), Trial Outcome Index (TOI), FACT-Leu total score, FACT-G total score, and EQ-5D visual analogue scale. Differences in least-squares mean score changes from baseline to the end of induction (EOI)/consolidation (EOC) and time to confirmed improvement/deterioration were analyzed. Overall treatment tolerability was assessed using the FACT-GP5. Analyses included 238 patients (ponatinib 159, imatinib 79) with ≥1 PRO assessment. Least-squares mean changes from baseline favored ponatinib, with significant and meaningful differences in FACT-LeuS, TOI, and FACT-Leu total score at EOI and across the primary domains except for FACT-LeuS at EOC. Median time to confirmed improvement was shorter with ponatinib versus imatinib for key measures. Ponatinib-treated patients tended to report being less bothered by treatment side effects as assessed by FACT-GP5. These findings highlight ponatinib's potentially favorable impact on health-related quality of life, supporting its use as frontline treatment for Ph+ ALL.
Collapse
Affiliation(s)
- Ajibade Ashaye
- Takeda Development Center Americas, Inc., Cambridge, MA, USA.
| | | | | | - Pau Montesinos
- Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | | | - Vanderson Rocha
- Instituto do Câncer do Estado de São Paulo, University of São Paulo, São Paulo, Brazil
| | - Cristina Papayannidis
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "L. e A. Seràgnoli", Bologna, Italy
| | | | - Maria R Baer
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Jose-Maria Ribera
- ICO-Hospital Germans Trias i Pujol, Josep Carreras Leukaemia Research Institute, Badalona, Spain
| | | | - Jianxiang Wang
- Institute of Hematology & Blood Diseases Hospital of CAMS & PUMC, Tianjin, China
| | - Sujun Gao
- The First Hospital of Jilin University, Changchun, China
| | - Deepali Rane
- Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | | |
Collapse
|
203
|
Zhang Y, Lang L, Guo X, Huang K, Yi J, Yuan Y, Zhu M, Zhang S, Hu B, Li X, Zhang Y. The association and impact of radiographic, pathological emphysema and spirometric airway obstruction on patients with resectable lung adenocarcinoma. Respir Res 2025; 26:151. [PMID: 40241184 PMCID: PMC12004668 DOI: 10.1186/s12931-025-03225-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 04/06/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Destruction of alveoli structure and lung function are interrelated, however, their correlation and clinical significance have been not well defined in patients with lung cancer. Thus, this study aimed to examine the association among radiographic, pathological emphysema and spirometric airway obstruction in patients with resectable lung cancer as well as explore their impact on postoperative pulmonary complications (PPCs) and long-term prognosis. METHODS Lung adenocarcinoma (LUAD) patients who performed chest CT, spirometry, and curative resection were included from a prospective three-institution database. CT-defined emphysema at baseline was assessed visually and quantitatively, pathological emphysema was reviewed on postoperative specimen. Multivariable regression models, propensity score matching, stratified analysis, and subgroup analysis were adopted to reduce selection bias. RESULTS Our cohort included 902 patients, with a median follow-up of 5.6 years. CT-defined emphysema was present in 163 patients (18.1%) and most of them (86.5%) were validated with pathological evidence. 169 had spirometric airway obstruction, while only 29.6% patients overlapped with CT-defined emphysema. Multivariable logistic regression models showed CT-defined emphysema, not airway obstruction, was associated with an increased risk of PPCs (adjusted odds ratio, 2.35; 95% CI, 1.40-3.93; P = 0.001). After adjusting for age, sex, body mass index, smoking history, tumour stage, vascular invasion, pleural invasion, multivariate cox analysis identified CT-defined emphysema, not airway obstruction, as an independent prognostic factor for OS (adjusted hazard ratio, 1.44; 95%CI, 1.05-1.97; P = 0.022). Patients with both radiographic and pathological emphysema experienced worse OS (log-rank P < 0.001). In the propensity score-matched cohort, stratified analysis, and never-smokers subgroup analysis, CT-defined emphysema remained a strong and statistically significant factor related to poor survival. CONCLUSIONS The presence of radiological and pathological emphysema in resectable LUAD was associated with frequent PPCs and decreased survival. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Yixiao Zhang
- Department of Pulmonary and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Lu Lang
- Department of Pulmonary and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xiaojuan Guo
- Department of Radiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Kewu Huang
- Department of Pulmonary and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jiawen Yi
- Department of Pulmonary and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yuan Yuan
- Department of Pulmonary and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Min Zhu
- Department of Pulmonary and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Shu Zhang
- Department of Pulmonary and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Bin Hu
- Department of Thoracic Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| | - Xue Li
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| | - Yuhui Zhang
- Department of Pulmonary and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
204
|
Imaoka H, Ikeda M, Ozaka M, Oshima K, Okano N, Shimizu S, Tsumura H, Komatsu Y, Yamashita T, Kataoka S, Nagano H, Hisano T, Sasaki M, Kobayashi S, Fukushima T, Mitsunaga S, Furukawa T, Hamauchi S, Ueno M, Furuse J. Phase 1/2 study of liposomal irinotecan plus S-1 for metastatic pancreatic cancer refractory to gemcitabine-based treatment. Eur J Cancer 2025; 222:115424. [PMID: 40252631 DOI: 10.1016/j.ejca.2025.115424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/03/2025] [Accepted: 04/11/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Liposomal irinotecan (nal-IRI) plus fluorouracil/folinic acid (5-FU/LV) improves survival in gemcitabine-refractory metastatic pancreatic cancer (PC) but requires a central venous port. S-1, an oral fluoropyrimidine with proven efficacy in PC, may replace 5-FU/LV in nal-IRI plus 5-FU/LV, potentially enhancing both convenience and antitumor effect. METHODS This single-arm, open-label, phase 1/2 study included patients with histologically or cytologically confirmed adenocarcinoma, aged 20-80 years, an Eastern Cooperative Oncology Group performance status of 0-1, with metastatic disease, and refractory to gemcitabine-based treatment. The primary endpoint in phase 1 part was the frequency of dose-limiting toxicity (DLT) to nal-IRI plus S-1. The primary endpoint in phase 2 part was overall survival. This trial was registered in the Japan Registry of Clinical Trials database (jRCTs031210040). RESULTS In phase 1 part, one patient with DLT was observed at nal-IRI 70 mg/m2 (day 1) with S-1 80 mg/m2/day (day 1-7) in a 2-week cycle, establishing this as the recommended phase 2 dose (RP2D). Forty-nine patients from phase 1 (n = 6) and phase 2 part (n = 43) were treated with the RP2D, and their results were pooled. Median overall survival was 10.3 months (95 % confidence interval, 8.1-12.0 months). A confirmed partial response was achieved in 10 patients (20.4 %). The most frequent treatment-emergent adverse events were hypoalbuminemia (98.0 %), anemia (98.0 %), and anorexia (81.6 %). There were no treatment-related deaths. CONCLUSIONS This study demonstrated that nal-IRI plus S-1 exhibited promising efficacy and an acceptable safety profile in patients with metastatic PC refractory to gemcitabine-based treatment.
Collapse
Affiliation(s)
- Hiroshi Imaoka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
| | - Masafumi Ikeda
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masato Ozaka
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kotoe Oshima
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Naohiro Okano
- Department of Medical Oncology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Satoshi Shimizu
- Department of Gastroenterology, Saitama Cancer Center, Saitama, Japan
| | - Hidetaka Tsumura
- Department of Gastroenterological Oncology, Hyogo Cancer Center, Akashi, Japan
| | - Yoshito Komatsu
- Division of Cancer Center, Hokkaido University Hospital, Sapporo, Japan
| | - Taro Yamashita
- Department of Gastroenterology, Kanazawa University Hospital, Kanazawa, Japan
| | - Shigeki Kataoka
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Terumasa Hisano
- Department of Hepato-Biliary-Pancreatology, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Mitsuhito Sasaki
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Satoshi Kobayashi
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Taito Fukushima
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Shuichi Mitsunaga
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takaaki Furukawa
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Satoshi Hamauchi
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Junji Furuse
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| |
Collapse
|
205
|
Tesch ME, Partridge AH. Can Improving Adjuvant Endocrine Therapy Persistence Improve Survival Outcomes for Patients With Early-Stage Breast Cancer? J Clin Oncol 2025:JCO2500371. [PMID: 40239125 DOI: 10.1200/jco-25-00371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Affiliation(s)
- Megan E Tesch
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Ann H Partridge
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
206
|
Han F, Qi G, Li R, Peng J, Yan S, Yuan C, Kong B, Ma H. USP28 promotes PARP inhibitor resistance by enhancing SOX9-mediated DNA damage repair in ovarian cancer. Cell Death Dis 2025; 16:305. [PMID: 40240356 PMCID: PMC12003857 DOI: 10.1038/s41419-025-07647-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 03/31/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025]
Abstract
PARP inhibitor (PARPi) resistance presents a significant challenge in ovarian cancer treatment, necessitating the development of effective therapeutic strategies to overcome this resistance and improve patient outcomes. Our study demonstrated that elevated expression of SRY-box 9 (SOX9) contributes to olaparib resistance in ovarian cancer. Mechanistically, the deubiquitinating enzyme USP28 was identified as a novel interacting partner of SOX9. USP28 inhibited the ubiquitination and subsequent degradation of SOX9, which is mediated by the E3 ubiquitin ligase FBXW7 during olaparib treatment. ChIP-Seq analysis revealed that SOX9 binds to the promoters of key DNA damage repair (DDR) genes (SMARCA4, UIMC1, and SLX4), thereby regulating DDR processes in ovarian cancer. Additionally, USP28 promoted olaparib resistance by stabilizing SOX9 protein and enhancing DNA damage repair. Furthermore, the USP28 specific inhibitor AZ1 reduced SOX9 protein stability and increased the sensitivity of ovarian cancer cells to olaparib. In conclusion, targeted inhibition of USP28 promoted ubiquitination-mediated degradation of SOX9, thereby impairing DNA damage repair capabilities and sensitizing ovarian cancer cells to PARPi. These findings elucidate the underlying mechanisms of PARPi resistance in ovarian cancer and suggest the potential efficacy of combining USP28 inhibitors with PARPi to overcome this resistance.
Collapse
Affiliation(s)
- Fang Han
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, China
| | - Gonghua Qi
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Rongrong Li
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Jiali Peng
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Shi Yan
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Cunzhong Yuan
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Beihua Kong
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Hanlin Ma
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China.
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
207
|
Qin Y, Mo Y, Li P, Liang X, Yu J, Chen D. Concurrent immunotherapy improves progression-free survival but increases toxicity in unresectable stage III NSCLC. Lung Cancer 2025; 203:108544. [PMID: 40253944 DOI: 10.1016/j.lungcan.2025.108544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/13/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND The PACIFIC trial established consolidation immunotherapy (IO) after concurrent chemoradiotherapy as the standard treatment for unresectable stage III non-small cell lung cancer (NSCLC) by improving survival. However, the optimal timing of IO remains debated. This study analyzes the survival benefits and risks of IO concurrent with radiotherapy (RT) versus IO following RT. METHODS A systematic search of multiple databases identified studies comparing IO concurrent with RT and IO following RT in unresectable stage III NSCLC. Data on overall survival (OS), progression-free survival (PFS), and adverse events (AEs) were analyzed using the "meta" package in R, along with a single-center cohort study. RESULTS The meta-analysis showed improved PFS with IO concurrent with RT, with significant differences at 1 year (69.5 % vs 57.6 %) and 1.5 years (56.3 % vs 45.7 %). OS was slightly better with IO following RT, with fewer severe AEs (≥grade 3: 52.6 % vs 37.2 %). A single-center cohort validated superior PFS for the concurrent group (HR = 2.039, 95 % CI: 1.014-4.322, P = 0.046). Shorter intervals between RT and IO were associated with better PFS in the following group (HR = 1.002, 95 % CI: 1.001-1.003, P = 0.002). CONCLUSION Concurrent IO with RT during chemoradiotherapy significantly improved PFS in unresectable stage III NSCLC, though OS did not benefit due to a higher incidence of severe AEs. Earlier IO initiation after RT was associated with better PFS. Our findings suggest the potential benefits of concurrent IO for selected patients.
Collapse
Affiliation(s)
- Yiwei Qin
- Department of Radiation Oncology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, People's Republic of China; Department of Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250000, People's Republic of China
| | - You Mo
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou University, Shantou, Guangdong 515000, People's Republic of China
| | - Pengwei Li
- Department of Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250000, People's Republic of China
| | - Xinyi Liang
- Department of Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250000, People's Republic of China; School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong 261000, People's Republic of China
| | - Jinming Yu
- Department of Radiation Oncology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, People's Republic of China; Department of Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250000, People's Republic of China.
| | - Dawei Chen
- Department of Radiation Oncology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, People's Republic of China; Department of Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250000, People's Republic of China.
| |
Collapse
|
208
|
Shen Y, Gao XJ, Zhang XX, Zhao JM, Hu FF, Han JL, Tian WY, Yang M, Wang YF, Lv JL, Zhan Q, An FM. Endoscopists and endoscopic assistants’ qualifications, but not their biopsy rates, improve gastric precancerous lesions detection rate. World J Gastrointest Endosc 2025; 17:104097. [DOI: 10.4253/wjge.v17.i4.104097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/27/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Detecting gastric precancerous lesions (GPLs) is critical for the early diagnosis and treatment of gastric cancer. Endoscopy combined with tissue examination is an important method for detecting GPLs. However, negative biopsy results often increase patients’ risks, economic burdens, and lead to additional healthcare costs. Improving the detection rate of GPLs and reducing the rate of negative biopsies is currently a key focus in endoscopic quality control.
AIM To explore the relationships between the endoscopist biopsy rate (EBR), qualifications of endoscopists and endoscopic assistants, and detection rate of GPLs.
METHODS EBR, endoscopists, and endoscopic assistants were divided into four groups: Low, moderate, high, and very high levels. Multivariable logistic regression analysis was used to analyze the relationships between EBR and the qualifications of endoscopists with respect to the detection rate of positive lesions. Pearson and Spearman correlation analyses were used to evaluate the correlation between EBR, endoscopist or endoscopic assistant qualifications, and the detection rate of positive lesions.
RESULTS Compared with those in the low EBR group, the odds ratio (OR) values for detecting positive lesions in the moderate, high, and very high EBR groups were 1.12 [95% confidence interval (CI): 1.06-1.19, P < 0.001], 1.22 (95%CI: 1.14-1.31, P < 0.001), and 1.38 (95%CI: 1.29-1.47, P < 0.001), respectively. EBR was positively correlated with the detection rate of gastric precancerous conditions (atrophic gastritis/intestinal metaplasia) (ρ = 0.465, P = 0.004). In contrast, the qualifications of the endoscopists were positively correlated with GPLs detection (ρ = 0.448, P = 0.005). Compared to endoscopists with low qualification levels, those with moderate, high, and very high qualification levels endoscopists demonstrated increased detection rates of GPLs by 13% (OR = 1.13, 95%CI: 0.98-1.31), 20% (OR = 1.20, 95%CI: 1.03-1.39), and 32% (OR = 1.32, 95%CI: 1.15-1.52), respectively. Further analysis revealed that the qualifications of endoscopists were positively correlated with the detection rates of GPLs in the cardia (ρ = 0.350, P = 0.034), angularis (ρ = 0.396, P = 0.015) and gastric body (ρ = 0.453, P = 0.005) but not in the antrum (ρ = 0.292, P = 0.079). Moreover, the experience of endoscopic assistants was positively correlated with the detection rate of precancerous lesions by endoscopists with low or moderate qualifications (ρ = 0.427, P = 0.015).
CONCLUSION Endoscopists and endoscopic assistants with high/very high qualifications, but not EBR, can improve the detection rate of GPLs. These results provide reliable evidence for the development of gastroscopic quality control indicators.
Collapse
Affiliation(s)
- Yao Shen
- Department of Gastroenterology, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, National Clinical Research Center for Digestive Diseases (Xi’an) Jiangsu Branch, Wuxi 214023, Jiangsu Province, China
| | - Xiao-Juan Gao
- Department of Gastroenterology, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, National Clinical Research Center for Digestive Diseases (Xi’an) Jiangsu Branch, Wuxi 214023, Jiangsu Province, China
| | - Xiao-Xue Zhang
- Department of Gastroenterology, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, National Clinical Research Center for Digestive Diseases (Xi’an) Jiangsu Branch, Wuxi 214023, Jiangsu Province, China
| | - Jia-Min Zhao
- Department of Gastroenterology, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, National Clinical Research Center for Digestive Diseases (Xi’an) Jiangsu Branch, Wuxi 214023, Jiangsu Province, China
| | - Fei-Fan Hu
- Department of Gastroenterology, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, National Clinical Research Center for Digestive Diseases (Xi’an) Jiangsu Branch, Wuxi 214023, Jiangsu Province, China
| | - Jing-Lue Han
- Department of Gastroenterology, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, National Clinical Research Center for Digestive Diseases (Xi’an) Jiangsu Branch, Wuxi 214023, Jiangsu Province, China
| | - Wen-Ying Tian
- Department of Gastroenterology, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, National Clinical Research Center for Digestive Diseases (Xi’an) Jiangsu Branch, Wuxi 214023, Jiangsu Province, China
| | - Mei Yang
- Department of Gastroenterology, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, National Clinical Research Center for Digestive Diseases (Xi’an) Jiangsu Branch, Wuxi 214023, Jiangsu Province, China
| | - Yun-Fei Wang
- Department of Gastroenterology, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, National Clinical Research Center for Digestive Diseases (Xi’an) Jiangsu Branch, Wuxi 214023, Jiangsu Province, China
| | - Jia-Le Lv
- Department of Gastroenterology, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, National Clinical Research Center for Digestive Diseases (Xi’an) Jiangsu Branch, Wuxi 214023, Jiangsu Province, China
| | - Qiang Zhan
- Department of Gastroenterology, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, National Clinical Research Center for Digestive Diseases (Xi’an) Jiangsu Branch, Wuxi 214023, Jiangsu Province, China
| | - Fang-Mei An
- Department of Gastroenterology, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, National Clinical Research Center for Digestive Diseases (Xi’an) Jiangsu Branch, Wuxi 214023, Jiangsu Province, China
| |
Collapse
|
209
|
Huang R, Kee L, Gont A, Meens J, Ferens FG, Irwin MS, Ailles L, Yuzwa SA, Robinson CM, Ohh M. Comparative single-cell transcriptomic profiling of patient-derived renal carcinoma cells in cellular and animal models of kidney cancer. FEBS Open Bio 2025. [PMID: 40241258 DOI: 10.1002/2211-5463.70022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 02/26/2025] [Accepted: 03/06/2025] [Indexed: 04/18/2025] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common form of kidney cancer that often displays resistance to conventional cancer therapies, including chemotherapy and radiation therapy. Targeted treatments, including immunotherapies and small molecular inhibitors, have been associated with improved outcomes. However, variations in the patient response and the development of resistance suggest that more models that better recapitulate the pathogenesis and metastatic mechanisms of ccRCC are required to improve our understanding and disease management. Here, we examined the transcriptional landscapes of in vitro cell culture as well as in vivo orthotopic and metastatic NOD/SCID-γ mouse models of ccRCC using a single patient-derived RCC243 cell line to allow unambiguous comparison between models. In our mouse model assays, RCC243 cells formed metastatic tumors, and all tumors retained clear cell morphology irrespective of model type. Notably, gene expression profiles differed markedly between the RCC243 tumor models-cell culture, orthotopic tumors, and metastatic tumors-suggesting an impact of the experimental model system and whether the tumor was orthotopic or metastatic. Furthermore, we found conserved prognostic markers between RCC243 tumor models and human ccRCC patient datasets, and genes upregulated in metastatic RCC243 were associated with worse patient outcomes.
Collapse
Affiliation(s)
- Richard Huang
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada
| | - Lynn Kee
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Alexander Gont
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Jalna Meens
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Fraser G Ferens
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada
| | - Meredith S Irwin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Canada
- Department of Paediatrics, The Hospital for Sick Children, Toronto, Canada
| | - Laurie Ailles
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Canada
| | - Scott A Yuzwa
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada
| | - Claire M Robinson
- School of Medicine, Health Sciences Centre, University College Dublin, Dublin 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Michael Ohh
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada
- Department of Biochemistry, University of Toronto, Canada
| |
Collapse
|
210
|
Sammallahti H, Rezasoltani S, Pekkala S, Kokkola A, Asadzadeh Agdaei H, Azizmohhammad Looha M, Ghanbari R, Zamani F, Sadeghi A, Sarhadi VK, Tiirola M, Puolakkainen P, Knuutila S. Fecal profiling reveals a common microbial signature for pancreatic cancer in Finnish and Iranian cohorts. Gut Pathog 2025; 17:24. [PMID: 40241224 PMCID: PMC12001732 DOI: 10.1186/s13099-025-00698-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Pancreatic cancer (PC) presents a significant challenge in oncology because of its late-stage diagnosis and limited treatment options. The inadequacy of current screening methods has prompted investigations into stool-based assays and microbial classifiers as potential early detection markers. The gut microbiota composition of PC patients may be influenced by population differences, thereby impacting the accuracy of disease prediction. However, comprehensive profiling of the PC gut microbiota and analysis of these cofactors remain limited. Therefore, we analyzed the stool microbiota of 33 Finnish and 50 Iranian PC patients along with 35 Finnish and 34 Iranian healthy controls using 16S rRNA gene sequencing. We assessed similarities and differences of PC gut microbiota in both populations while considering sociocultural impacts and generated a statistical model for disease prediction based on microbial classifiers. Our aim was to expand the current understanding of the PC gut microbiota, discuss the impact of population differences, and contribute to the development of early PC diagnosis through microbial biomarkers. RESULTS Compared with healthy controls, PC patients presented reduced microbial diversity, with discernible microbial profiles influenced by factors such as ethnicity, demographics, and lifestyle. PC was marked by significantly higher abundances of facultative pathogens including Enterobacteriaceae, Enterococcaceae, and Fusobacteriaceae, and significantly lower abundances of beneficial bacteria. In particular, bacteria belonging to the Clostridia class, such as butyrate-producing Lachnospiraceae, Butyricicoccaceae, and Ruminococcaceae, were depleted. A microbial classifier for the prediction of pancreatic ductal adenocarcinoma (PDAC) was developed in the Iranian cohort and evaluated in the Finnish cohort, where it yielded a respectable AUC of 0.88 (95% CI 0.78, 0.97). CONCLUSIONS This study highlights the potential of gut microbes as biomarkers for noninvasive PC screening and the development of targeted therapies, emphasizing the need for further research to validate these findings in diverse populations. A comprehensive understanding of the role of the gut microbiome in PC could significantly enhance early detection efforts and improve patient outcomes.
Collapse
Affiliation(s)
- Heidelinde Sammallahti
- Department of Pathology, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland
- Department of Surgery, Abdominal Center, University of Helsinki, Helsinki University Hospital, 00290, Helsinki, Finland
| | - Sama Rezasoltani
- Division of Oral Microbiology and Immunology, Department of Operative Dentistry, Periodontology and Preventive Dentistry, Rheinisch-Westfälische Technische Hochschule (RWTH) University Hospital, 52074, Aachen, Germany
| | - Satu Pekkala
- Faculty of Sport and Health Sciences, University of Jyväskylä, 40014, Jyväskylä, Finland
| | - Arto Kokkola
- Department of Surgery, University of Helsinki and Helsinki University Hospital, 00290, Helsinki, Finland
| | - Hamid Asadzadeh Agdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, P.O. Box 1985717411, Tehran, Iran
| | - Mehdi Azizmohhammad Looha
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, P.O. Box 1985717411, Tehran, Iran
| | - Reza Ghanbari
- Gene Therapy Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Zamani
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Virinder Kaur Sarhadi
- Department of Oral and Maxillofacial Diseases, Helsinki University Hospital and University of Helsinki, 00290, Helsinki, Finland
| | - Marja Tiirola
- Department of Environmental and Biological Sciences, Nanoscience Center, University of Jyväskylä, 40014, Jyväskylä, Finland
- BiopSense Oy, Eeronkatu 10, 40720, Jyväskylä, Finland
| | - Pauli Puolakkainen
- Department of Surgery, Abdominal Center, University of Helsinki, Helsinki University Hospital, 00290, Helsinki, Finland
| | - Sakari Knuutila
- Department of Pathology, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland.
| |
Collapse
|
211
|
Tao ZH, Han JX, Xu J, Zhao E, Wang M, Wang Z, Lin XL, Xiao XY, Hong J, Chen H, Chen YX, Chen HM, Fang JY. Screening of patient-derived organoids identifies mitophagy as a cell-intrinsic vulnerability in colorectal cancer during statin treatment. Cell Rep Med 2025; 6:102039. [PMID: 40154491 DOI: 10.1016/j.xcrm.2025.102039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 01/26/2025] [Accepted: 03/03/2025] [Indexed: 04/01/2025]
Abstract
Statins, commonly used to lower cholesterol, are associated with improved prognosis in colorectal cancer (CRC), though their effectiveness varies. This study investigates the anti-cancer effects of atorvastatin in CRC using patient-derived organoids (PDOs) and PDO-derived xenograft (PDOX) models. Our findings reveal that atorvastatin induces mitochondrial dysfunction, leading to apoptosis in cancer cells. In response, cancer cells induce mitophagy to clear damaged mitochondria, enhancing survival and reducing statin efficacy. Analysis of a clinical cohort confirms mitophagy's role in diminishing statin effectiveness. Importantly, inhibiting mitophagy significantly enhances the anti-cancer effects of atorvastatin in CRC PDOs, xenograft models, and azoxymethane (AOM)-dextran sulfate sodium (DSS) mouse models. These findings identify mitophagy as a critical pro-survival mechanism in CRC during statin treatment, providing insights into the variable responses observed in epidemiological studies. Targeting this vulnerability through combination therapy can elicit potent therapeutic responses.
Collapse
Affiliation(s)
- Zhi-Hang Tao
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ji-Xuan Han
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jia Xu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Enhao Zhao
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ming Wang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zheng Wang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Lin Lin
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiu-Ying Xiao
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Hong
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Haoyan Chen
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying-Xuan Chen
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hui-Min Chen
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
212
|
DeLiberty JM, Roach MK, Stalnecker CA, Robb R, Schechter EG, Pieper NL, Taylor KE, Pita LM, Yang R, Bang S, Drizyte-Miller K, Ackermann SE, Peña SRN, Baldelli E, Min SM, Drewry DH, Petricoin EF, Morris JP, Der CJ, Cox AD, Bryant KL. Concurrent Inhibition of the RAS-MAPK Pathway and PIKfyve Is a Therapeutic Strategy for Pancreatic Cancer. Cancer Res 2025; 85:1479-1495. [PMID: 39932818 PMCID: PMC11999774 DOI: 10.1158/0008-5472.can-24-1757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 11/14/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by KRAS- and autophagy-dependent growth. Inhibition of the KRAS-RAF-MEK-ERK pathway enhances autophagic flux and dependency, and concurrent treatment with the nonspecific autophagy inhibitor chloroquine (CQ) and ERK-MAPK pathway inhibitors can synergistically block PDAC growth. However, CQ is limited in terms of specificity and potency. To find alternative anti-autophagy strategies, in this study, we performed a CRISPR-Cas9 loss-of-function screen in PDAC cell lines that identified the lipid kinase phosphatidylinositol-3-phosphate 5-kinase (PIKfyve) as a growth-promoting gene. PIKfyve inhibition by the small molecule apilimod resulted in durable growth suppression, with much greater potency than CQ treatment. PIKfyve inhibition caused lysosomal dysfunction, reduced autophagic flux, and led to the accumulation of autophagy-related proteins. Furthermore, PIKfyve inhibition blocked the compensatory increases in autophagic flux associated both with MEK inhibition and with direct RAS inhibition. Accordingly, combined inhibition of PIKfyve and the RAS-MAPK pathway showed robust growth suppression across a panel of KRAS-mutant PDAC models. Growth suppression was due, in part, to potentiated cell-cycle arrest and induction of apoptosis following loss of inhibitor of apoptosis proteins. These findings indicate that concurrent inhibition of RAS and PIKfyve is a synergistic, cytotoxic combination that may represent a therapeutic strategy for PDAC. Significance: PIKfyve inhibition effectively blocks autophagy in multiple models of KRAS-mutant pancreatic cancer and can synergize with inhibitors of members of the RAS-MAPK pathway, providing an effective combination strategy for pancreatic cancer.
Collapse
Affiliation(s)
| | - Mallory K. Roach
- Department of Pharmacology, George Mason University, Manassas, VA, USA
| | - Clint A. Stalnecker
- Lineberger Comprehensive Cancer Center, George Mason University, Manassas, VA, USA
| | - Ryan Robb
- Department of Pharmacology, George Mason University, Manassas, VA, USA
| | - Elyse G. Schechter
- Lineberger Comprehensive Cancer Center, George Mason University, Manassas, VA, USA
| | - Noah L. Pieper
- Lineberger Comprehensive Cancer Center, George Mason University, Manassas, VA, USA
| | - Khalilah E. Taylor
- Lineberger Comprehensive Cancer Center, George Mason University, Manassas, VA, USA
| | - Lily M. Pita
- Lineberger Comprehensive Cancer Center, George Mason University, Manassas, VA, USA
| | - Runying Yang
- Lineberger Comprehensive Cancer Center, George Mason University, Manassas, VA, USA
| | - Scott Bang
- Department of Pharmacology, George Mason University, Manassas, VA, USA
| | | | | | | | - Elisa Baldelli
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Sophia M. Min
- Structural Genomics Consortium and Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David H. Drewry
- Lineberger Comprehensive Cancer Center, George Mason University, Manassas, VA, USA
- Structural Genomics Consortium and Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Emanuel F. Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - John P. Morris
- Department of Pharmacology, George Mason University, Manassas, VA, USA
- Lineberger Comprehensive Cancer Center, George Mason University, Manassas, VA, USA
| | - Channing J. Der
- Department of Pharmacology, George Mason University, Manassas, VA, USA
- Lineberger Comprehensive Cancer Center, George Mason University, Manassas, VA, USA
| | - Adrienne D. Cox
- Department of Pharmacology, George Mason University, Manassas, VA, USA
- Lineberger Comprehensive Cancer Center, George Mason University, Manassas, VA, USA
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, NC, USA
| | - Kirsten L. Bryant
- Department of Pharmacology, George Mason University, Manassas, VA, USA
- Lineberger Comprehensive Cancer Center, George Mason University, Manassas, VA, USA
| |
Collapse
|
213
|
Hu X, Li Y, Ma H, Xiong L, Tan J, Jin Y. Psychometric properties and measurement invariance of the health behavior scale for cancer patients in Chinese cancer population. Health Qual Life Outcomes 2025; 23:39. [PMID: 40234935 PMCID: PMC12001631 DOI: 10.1186/s12955-025-02368-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 04/07/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Health behavior plays a major role in the development, progression, and prognosis of cancer. The Health Behavior Scale for Cancer Patients (HBSCP) can be used to assess the level of health behavior in cancer patients. This study aimed to explore its psychometric properties and Measurement Invariance (MI) in the Chinese cancer population. METHODS A longitudinal study was conducted with 567 cancer patients across two hospitals, and 428 participants underwent a second assessment three months later. Analyses were performed to evaluate reliability (internal consistency), validity (structural validity, convergent validity, and criterion-related validity), and MI of the Chinese version of the HBSCP. RESULTS The two-factor structural model of the 9-item scale demonstrated an excellent fit in Confirmatory Factor Analysis (CFA). Psychometric analyses indicated strong internal consistency, with Cronbach's alpha coefficients ranging from 0.846 to 0.899 and McDonald's Omega values between 0.847 and 0.897. Convergent validity was supported by Composite Reliability (CR > 0.70) and Average Variance Extracted (AVE > 0.50). Criterion-related validity was established via significant correlations with the Health-Promoting Lifestyle Profile II (HPLP-II; r = 0.653 ~ 0.760). Multi-group CFA further confirmed MI across cancer types (ΔCFI&TLI < 0.01; ΔRMSEA < 0.015) and time groups (3-month interval). CONCLUSIONS This study provides longitudinal evidence supporting the adequate psychometric properties and temporal stability of the Chinese version of the HBSCP, thus validating its utility for measuring health behavior in Chinese cancer populations. The Chinese HBSCP can serve as a tool for healthcare providers to assess the current status and changes in health behavior among cancer patients.
Collapse
Affiliation(s)
- Xiaoxiao Hu
- School of Nursing, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu, China
| | - Yang Li
- Department of nursing, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, 300121, Tianjin, China
| | - Hongwen Ma
- Department of nursing, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, 300121, Tianjin, China
| | - Lina Xiong
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Jiangxi, 330200, Nanchang, China
| | - Jiping Tan
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, 158 Wuyang Road, Enshi, 445099, Hubei, China.
| | - Yanfei Jin
- School of Nursing, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu, China.
| |
Collapse
|
214
|
Zhang P, Chen T, Yang M. Clinical feature and gene expression analysis in low prostate-specific antigen, high-grade prostate cancer. PLoS One 2025; 20:e0321728. [PMID: 40233079 PMCID: PMC11999122 DOI: 10.1371/journal.pone.0321728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 03/11/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Prostate cancer (PCa) patients with low prostate-specific antigen (PSA) levels can occasionally present high-grade disease. These patients often exhibit resistance to androgen deprivation therapy and have poor outcomes. The mechanisms underlying these observations remain poorly understood. This study aimed to investigate the clinical characteristics and potential gene expression mechanisms in this subgroup. PATIENTS AND METHODS Clinical data from 365,558 PCa patients were categorized into four groups based on PSA levels and Gleason score (GS): Group 1 (PSA ≤ 2.5 ng/mL, GS < 8), Group 2 (PSA ≤ 2.5 ng/mL, GS ≥ 8), Group 3 (PSA > 2.5 ng/mL, GS < 8), and Group 4 (PSA > 2.5 ng/mL, GS ≥ 8). Clinical characteristics were compared using Kruskal-Wallis H and Pearson's chi-squared tests. Competing-risks regression assessed prostate cancer-specific mortality (PCSM). Gene set enrichment analysis (GSEA) was performed on 219 PCa patients to compare Group A (PSA ≤ 2.5 ng/mL, GS ≥ 8) with Group B (PSA > 2.5 ng/mL, GS ≥ 8). RESULTS Group 2 had a significantly higher tumor stage (p < 0.001) and increased hazard ratio for PCSM (p < 0.001). GSEA in Group A identified 156 upregulated gene sets and highlighted several enriched pathways, including the polycomb repressive complex 2, the epidermal growth factor receptor family, retrograde axonal transport, the tumor necrosis factor/nuclear factor-κB pathway, the Rho guanine nucleotide exchange factor/RhoA pathway, and the phosphoinositide 3-kinase signaling pathways (p < 0.05, false discovery rate-adjusted p < 0.25). CONCLUSION PCa patients with low PSA levels and high GS demonstrated an increased risk of PCSM. They were characterized by the aberrant activation of multiple signaling pathways. Targeted therapeutic strategies aimed at these pathways warrant further investigation for their potential to improve outcomes in this aggressive PCa subtype.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Urology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Tieding Chen
- Department of Urology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Ming Yang
- Department of Urology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| |
Collapse
|
215
|
Zhang N, Li J, Ren Y, Xu Y. Comprehensive pan-cancer analysis of PPP1R3G reveals its diagnostic, prognostic, and immunotherapeutic implications. Discov Oncol 2025; 16:530. [PMID: 40232629 PMCID: PMC12000506 DOI: 10.1007/s12672-025-02361-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 04/10/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND PPP1R3G, a regulatory subunit of protein phosphatase 1, plays a critical role in glycogen metabolism and has been implicated in various cancers. This study provides a comprehensive pan-cancer analysis of PPP1R3G, evaluating its expression, diagnostic and prognostic significance, and potential as a therapeutic target. METHODS We performed an extensive pan-cancer analysis of PPP1R3G using several databases to assess its expression and investigate its correlations with clinical outcomes. Our investigation included assessing PPP1R3G's impact on survival, its correlation with immune checkpoints and tumor stemness scores, and its prognostic significance. We also explored its relationship with immunomodulators, genomic profiles, and immunological characteristics, as well as its response to immunotherapy and involvement in various biological pathways. RESULTS PPP1R3G expression varied significantly across different cancers and correlated with both diagnostic and prognostic outcomes. Moreover, PPP1R3G was significantly linked to immune checkpoints, immunomodulators, prognosis, immunoregulatory genes, tumor stemness, cellular function, and immune infiltration across numerous cancer types. Further analysis of PPP1R3G-related gene enrichment, mutation profiles, RNA modifications, and genomic heterogeneity revealed that missense mutations were the predominant alteration affecting PPP1R3G. CONCLUSIONS Overall, the expression of PPP1R3G is closely associated with various cancers and may serve as a potential biomarker for cancer detection.
Collapse
Affiliation(s)
- Nie Zhang
- Graduate School of Anhui Medical University, Hefei, China
- Key Laboratory of Gametes and Abnormal Reproductive Tract of National Health Commission, Anhui Medical University, Hefei, China
| | - Jiaoyu Li
- Graduate School of Anhui Medical University, Hefei, China
- Key Laboratory of Gametes and Abnormal Reproductive Tract of National Health Commission, Anhui Medical University, Hefei, China
| | - Yanzhi Ren
- Department of Cardiology, Shizhong District People's Hospital, Zaozhuang, China
| | - Yahui Xu
- Graduate School of Anhui Medical University, Hefei, China.
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
216
|
Yang Y, Ji X, Li S, Gao X, Wang Y, Huang Y. Ultrasound-based radiomics for predicting the five major histological subtypes of epithelial ovarian cancer. BMC Med Imaging 2025; 25:122. [PMID: 40234786 PMCID: PMC12001453 DOI: 10.1186/s12880-025-01624-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 03/03/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Computational approaches have been proposed using radiomics in order to assess tumour heterogeneity, which is motivated by the concept that biomedical images may contain underlying pathophysiology information and has the potential to quantitatively measure the heterogeneity of intra- and intertumours. Ovarian cancer has the highest mortality among malignant tumours of female reproductive system and can be further divided into many subtypes with different management strategies and prognosis. The purpose of our study is to develop and validate ultrasound-based radiomics models to distinguish the five major histological subtypes of epithelial ovarian cancer. METHODS From January 2018 to August 2022, 1209 eligible ovarian cancer patients were enrolled. There were two subjects in this study: all patients (n = 1209) and patients with the five major histological subtypes (n = 1039). After image segmentation manually, radiomics features were extracted and some clinical characteristics were added. Nine feature selection methods were used to select the optimal predictive features. Seven classifiers were carried out to construct models. Choose the combination with the best predictive performance as the final result. RESULTS As for low-grade serous carcinoma, endometrioid carcinoma, and clear cell carcinoma, the models yields AUCs below 0.80 in the 10-fold cross-validation in the two groups. As for mucinous carcinoma, the AUCs were 0.83(95%CI, 0.74-0.93) and 0.89(95%CI, 0.83-0.95) in the validation cohorts and 0.80(95%CI, 0.73-0.87) and 0.86(95%CI, 0.78-0.94) in the 10-fold cross-validation in the two groups, respectively. As for high-grade serous carcinoma (HGSC), the models showed AUCs of 0.87(95%CI, 0.83-0.91) and 0.85(95%CI, 0.81-0.89) in the validation cohorts and 0.87(95%CI, 0.85-0.89) and 0.84(95%CI, 0.81-0.87) in the 10-fold cross-validation in the two groups, respectively, and exhibited high consistency between the predicted results and the actual outcomes, and brought great net benefits for patients. CONCLUSIONS The ultrasound-based radiomics models in discriminating HGSC and non-HGSC showed good predictive performance, as well as high consistency between the predicted results and the actual outcomes, and brought significant net benefits for patients.
Collapse
Affiliation(s)
- Yang Yang
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xinyu Ji
- Department of Ultrasound, Shengjing Hospital, China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning Province, 110004, China
| | - Sen Li
- Yizhun Medical AI, Beijing, China
| | - Xuemeng Gao
- Department of Ultrasound, Shengjing Hospital, China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning Province, 110004, China
| | - Yitong Wang
- Department of Ultrasound, Shengjing Hospital, China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning Province, 110004, China
| | - Ying Huang
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Department of Ultrasound, Shengjing Hospital, China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning Province, 110004, China.
| |
Collapse
|
217
|
Shin AE, Sugiura K, Kariuki SW, Cohen DA, Flashner SP, Klein-Szanto AJ, Nishiwaki N, De D, Vasan N, Gabre JT, Lengner CJ, Sims PA, Rustgi AK. LIN28B-mediated PI3K/AKT pathway activation promotes metastasis in colorectal cancer models. J Clin Invest 2025; 135:e186035. [PMID: 39808497 PMCID: PMC11996871 DOI: 10.1172/jci186035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer death because of metastatic spread. LIN28B is overexpressed in 30% of CRCs and promotes metastasis, yet its mechanisms remain unclear. In this study, we genetically modified CRC cell lines to overexpress LIN28B, resulting in enhanced PI3K/AKT pathway activation and liver metastasis in mice. We developed genetically modified mouse models with constitutively active Pik3ca that form intestinal tumors progressing to liver metastases with an intact immune system, addressing the limitations of previous Pik3ca-mutant models, including long tumor latency, mixed histology, and lack of distant metastases. The PI3Kα-specific inhibitor alpelisib reduced migration and invasion in vitro and metastasis in vivo. We present a comprehensive analysis of vertical inhibition of the PI3K/AKT pathway in CRC using the FDA-approved drugs alpelisib and capivasertib (an AKT inhibitor) in combination with LY2584702 (a ribosomal protein S6 kinase inhibitor) in CRC cell lines and mouse- and patient-derived organoids. Tissue microarrays from patients with CRC verified that LIN28B and PI3K/AKT pathway activation correlate with CRC progression. These findings highlight the critical role of the LIN28B-mediated PI3K/AKT pathway in CRC metastasis, the therapeutic potential of targeted inhibition, and the promise of patient-derived organoids in precision medicine in metastatic CRC.
Collapse
Affiliation(s)
- Alice E. Shin
- Division of Digestive and Liver Diseases, Department of Medicine, and
| | - Kensuke Sugiura
- Division of Digestive and Liver Diseases, Department of Medicine, and
| | | | - David A. Cohen
- Department of Surgery, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons; Columbia University Irving Medical Center, New York, New York, USA
| | | | | | | | - Dechokyab De
- Division of Digestive and Liver Diseases, Department of Medicine, and
| | - Neil Vasan
- Division of Hematology and Oncology, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - Joel T. Gabre
- Division of Digestive and Liver Diseases, Department of Medicine, and
| | - Christopher J. Lengner
- Department of Biomedical Sciences, School of Veterinary Medicine, and Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Peter A. Sims
- Department of Systems Biology, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - Anil K. Rustgi
- Division of Digestive and Liver Diseases, Department of Medicine, and
| |
Collapse
|
218
|
Zhi B, Zhao P, Duan L, Liu Y, Zhu Z, Zhang X. Association of cancer with overactive bladder and impact of overactive bladder on mortality among cancer survivors: NHANES 1999-2018. PLoS One 2025; 20:e0320491. [PMID: 40233025 PMCID: PMC11999114 DOI: 10.1371/journal.pone.0320491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 02/20/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Cancer is one of the leading causes of death worldwide. There is little knowledge on the association between cancer and risk of overactive bladder (OAB). Additionally, the impact of OAB on mortality among cancer survivors remains unknown. We aimed to examine the association between cancer and risk of OAB and investigate OAB associations with mortality outcomes in cancer survivors. METHODS We analyzed data of the National Health and Nutrition Examination Survey from 1999 to 2018. Cancer status was identified through the NHANES questionnaire. OAB was diagnosed with a cumulative OAB Symptom Score ≥ 3. Multivariable logistic analysis was performed to examine the relationship between cancer and OAB. Cox regression analysis was carried out to investigate the relationship between OAB and mortality in subjects with cancer. RESULTS In total, this study included a cohort of 32,166 participants. Within the study population, the occurrence of cancer was observed in 9.606%, and OAB was prevalent in 20.167%. The findings from the logistic regression analyses indicated a heightened risk of OAB among individuals with cancer in comparison to those devoid of cancer. Results from subgroup analyses also revealed affirmative correlations between cancer and OAB. Furthermore, the diagnosis of cancer, irrespective of whether it was genitourinary, non-genitourinary, pelvic, or non-pelvic in nature, was found to be correlated with an increased risk of OAB. Within the cohort comprising 3,090 participants diagnosed with cancer, a total of 850 all-cause deaths were recorded during a median follow-up duration of 76 months. After accounting for multiple covariates, OAB was linked to an elevated risk of all-cause and cancer-specific mortality. Moreover, the influence of OAB on mortality from all causes was observed across various cancer types, including both genitourinary and non-genitourinary, as well as pelvic and non-pelvic cancers. CONCLUSIONS The investigation identified a noteworthy positive correlation between cancer and the likelihood of OAB. Moreover, OAB was significantly correlated with an elevated risk of mortality among individuals who had received a cancer diagnosis.
Collapse
Affiliation(s)
- Bin Zhi
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - Pin Zhao
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - Liyuan Duan
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - Yuan Liu
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - Zhaowei Zhu
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - Xuepei Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| |
Collapse
|
219
|
Wang Y, Sun X, Lu B, Zhang D, Yin Y, Liu S, Chen L, Zhang Z. Current applications, future Perspectives and challenges of Organoid technology in oral cancer research. Eur J Pharmacol 2025; 993:177368. [PMID: 39947346 DOI: 10.1016/j.ejphar.2025.177368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/31/2025] [Accepted: 02/10/2025] [Indexed: 02/20/2025]
Abstract
Oral cancer poses significant health risks with an increasing incidence annually. Despite advancements in treatment methods, their efficacy is frequently constrained by cancer heterogeneity and drug resistance, leading to minimal improvement in the 5-year survival rate. Therefore, there is a critical need for new treatment methods leaded by representative preclinical research models. Compared to other models, organoids can more precisely simulate the tissue structure, genetic characteristics, and tumor microenvironment (TME) of in vivo tumors, exhibiting high tumor specificity. This makes organoid technology a valuable tool in investigating tumor development, mechanisms of metastasis, drug screening, prediction of clinical responses, and personalized patient treatment. Moreover, integrating organoid technology with other biotechnologies could expand its applications in tissue regeneration. Although organoid technology is increasingly utilized in oral cancer research, a systematic review in this field is absent. This paper is to bridge the gap by reviewing the development and current status of organoid research, highlighting its applications, future prospects, and challenges in oral cancer. It aims to provide novel insights into the role of organoids in precision treatment and regenerative medicine for oral cancer.
Collapse
Affiliation(s)
- Yunyi Wang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Xiang Sun
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Bingxu Lu
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Danya Zhang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Yaping Yin
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Shuguang Liu
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China.
| | - Lei Chen
- Department of Burn, Wound Repair & Reconstruction, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, Guangzhou, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Zhaoqiang Zhang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China.
| |
Collapse
|
220
|
Luo L, Cheng Y, Wang H, Li L, Niu H, Yang Y, Zhou Q, He J, Xu J. Lidocaine-A Promising Candidate for the Treatment of Cancer-Induced Bone Pain: A Narrative Review. Adv Ther 2025:10.1007/s12325-025-03192-w. [PMID: 40232625 DOI: 10.1007/s12325-025-03192-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 03/27/2025] [Indexed: 04/16/2025]
Abstract
Pain is one of the most common symptoms in patients with cancer, with cancer-induced bone pain (CIBP) significantly affecting their quality of life. Opioids are commonly used as first-line treatments for cancer pain, but their use requires caution due to non-mechanistic analgesia and significant side effects. As a result, there is a need for new non-opioid drugs that target cancer pain through specific mechanisms. Recent studies on the anticancer effects of lidocaine have highlighted its potential benefits in both treating cancer and alleviating cancer-induced pain. This article discusses the mechanism of action and clinical applications of lidocaine in cancer pain management, and suggests new treatment approaches for patients with CIBP.
Collapse
Affiliation(s)
- Lihan Luo
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Yuqi Cheng
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Hanxi Wang
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Li Li
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Hanyun Niu
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Yuzhu Yang
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Qianqian Zhou
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Jiannan He
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
| | - Jianhong Xu
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
| |
Collapse
|
221
|
Zhang Y, Zhu WL, Wu M, Gao TY, Hu HX, Xu ZY. Using bioinformatics methods to elucidate fatty acid-binding protein 4 as a potential biomarker for colon adenocarcinoma. World J Gastrointest Oncol 2025; 17:103113. [PMID: 40235911 PMCID: PMC11995352 DOI: 10.4251/wjgo.v17.i4.103113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/07/2025] [Accepted: 02/14/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Colon adenocarcinoma (COAD) ranks second in terms of cancer-related deaths. We found that fatty acid-binding protein 4 (FABP4), which is related to cell adhesion and immunity, affects the occurrence and development of COAD. This study focused on the possibility of using FABP4 as a biomarker for COAD and constructed a nomogram for predicting the survival of COAD patients. AIM To verify the possibility of using FABP4 as a biomarker for COAD. METHODS A total of 453 COAD tissue samples, along with 41 normal tissue samples, were obtained from The Cancer Genome Atlas database. The difference in FABP4 expression between COAD tissues and normal tissues was analyzed, and the results were verified by immunohistochemistry. The WGCNA algorithm links FABP4 expression with an enrichment analysis and with immune cell infiltration pathways. The biological functions of FABP4 and its coexpressed genes were explored through enrichment analyses. The ESTIMATE, CIBERSORT and ssGSEA methods were used for the immune infiltration analysis. Finally, risk scores were calculated by a Cox analysis. A nomogram was constructed by combining risk scores with routine clinicopathological factors. We assessed the accuracy of survival predictions based on the C-index. The C-index ranges from 0.5 to 1.0, and in general, a C-index value greater than 0.65 indicates a reasonable estimate. The results were validated using the Gene Expression Omnibus (GEO) database. RESULTS FABP4 was significantly differentially expressed in COAD. It is a promising auxiliary biomarker for screening and diagnosis. Enrichment analyses suggested that FABP4 may influence the invasion and progression of COAD through cell adhesion. The immunological analysis revealed that FABP4 expression in COAD was significantly positively correlated with immune cell infiltration. Moreover, a nomogram to predict the survival of COAD patients was successfully constructed by integrating the calculated risk scores of 15 candidate genes and routine clinicopathological factors. This nomogram could effectively predict 1-year, 3-year, and 5-year survival (C-index = 0.786) and was verified (C-index = 0.73). CONCLUSION This study established FABP4 as an effective biomarker for screening, assisting in the diagnosis and determining the prognosis.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Medical Engineering, Wannan Medical College, Wuhu 241002, Anhui Province, China
| | - Wen-Li Zhu
- Seven Inpatient Ward, The Fourth People's Hospital of Wuhu, Wuhu 241002, Anhui Province, China
| | - Min Wu
- Sixteen Inpatient Ward, The Fourth People's Hospital of Wuhu, Wuhu 241002, Anhui Province, China
| | - Tian-Yuan Gao
- Department of Pathology, The Second Affiliated Hospital of Wannan Medical College, Wuhu 241000, Anhui Province, China
| | - Hui-Xian Hu
- Department of Medical Engineering, Wannan Medical College, Wuhu 241002, Anhui Province, China
| | - Zheng-Yuan Xu
- Department of Medical Engineering, Wannan Medical College, Wuhu 241002, Anhui Province, China
| |
Collapse
|
222
|
Zhu B, Wei Y, Zheng X, Tang C, Xie X, Lv Y. Enhanced Breath Aldehyde Analysis by Dual-Membrane-Assisted Charge Tagging, Enrichment, and Onsite Elution NanoESI-MS. Anal Chem 2025; 97:8021-8029. [PMID: 40165712 DOI: 10.1021/acs.analchem.5c00434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Aldehydes, crucial volatile organic compounds present in exhaled breath, have been established as promising biomarkers for cancer diagnosis. However, their rapid and sensitive detection through widely employed spray-based ionization mass spectrometry is still challenging. To address this, we introduce a charged "iridium isotopic signature" probe tailored for efficient capture and unambiguous identification of ubiquitous aldehydes in the gas phase. This 191/193Ir-tagged mass spectrometric probe, equipped with a reactive amine moiety capable of interacting with aldehydes, is immobilized on the porous Nylon-6 membrane that facilitates efficient gas transport and enriches aldehydes from the complex breath matrix. Following a rapid solvent extraction, the Ir-tagging aldehyde derivatives were successfully eluted with efficient removal of excess probes by the oxidized cellulose membrane, yielding a purified sample ideally suited for direct, rapid, and ultrasensitive (with a detection limit below 0.1 ppt) nanoelectrospray ionization mass spectrometry (nanoESI-MS) analysis. By utilizing an analogous iridium complex as an internal standard, our method precisely identified and quantified 12 aldehydes in exhaled breath (EB), with several exhibiting significant elevations in esophageal cancer patients compared with healthy controls. This highlights its efficacy as a rapid and accurate tool for detecting breath aldehyde biomarkers, offering promising avenues for cancer diagnosis.
Collapse
Affiliation(s)
- Beichen Zhu
- Analytical & Testing Center, Sichuan University, Chengdu 610064, China
| | - Yifan Wei
- Analytical & Testing Center, Sichuan University, Chengdu 610064, China
| | - Xiumei Zheng
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu 610040, China
| | - Chengxi Tang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu 610040, China
| | - Xiaobo Xie
- Analytical & Testing Center, Sichuan University, Chengdu 610064, China
| | - Yi Lv
- Analytical & Testing Center, Sichuan University, Chengdu 610064, China
- Key Laboratory of Green Chemistry & Technology, Ministry of Education, College of Chemistry, Sichuan University, Chengdu, Sichuan 610064, China
| |
Collapse
|
223
|
Huang Y, Huang J, Zhan J, Chen M, Zheng J, He J, Fang W, Zhang L, Li J. CCN1 is a therapeutic target upregulated in EML4-ALK mutant lung adenocarcinoma reversibly resistant to alectinib. Cell Death Dis 2025; 16:303. [PMID: 40234387 PMCID: PMC12000322 DOI: 10.1038/s41419-025-07601-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 03/11/2025] [Accepted: 03/27/2025] [Indexed: 04/17/2025]
Abstract
There is limited understanding of the phenomenon of reversible drug resistance, which is characterized by tumor cells regaining sensitivity when the drug is changed or withdrawn after a period of drug resistance. This phenomenon is usually not associated with genetic alterations of tumor cells. In this study, reversible resistant state was induced by alectinib in EML4-ALK mutant lung cancer cell. By performing RNA sequencing on reversible drug-resistant cell line to examine changes in transcriptional profile, significant change in CCN1 was detected after withdrawal and repeated administration of alectinib. Targeting CCN1 resulted in inhibition of tumor cell proliferation and angiogenesis, and restoration of sensitivity to alectinib in reversible drug-resistant cells. Further studies revealed that CCN1 could affect the expression of VEGFA by affecting AKT phosphorylation, and the change of NF-κB could impact the activation of CCN1-AKT-VEGFA pathway. Suppressing NF-κB or CCN1 receptor could improve the sensitivity to alectinib, further suggesting that NF-κB and CCN1 might play a key role in overcoming reversible drug resistance.
Collapse
Affiliation(s)
- Yihua Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China
- Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jie Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Jianhua Zhan
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Maojian Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jiani Zheng
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Junyi He
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Wenfeng Fang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China.
| | - Li Zhang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China.
| | - Jing Li
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
224
|
Lan JX, Huang LJ, Kang SS, Hao-Huang, Liu SL, Dai W, Xu XL, Wang JY, Shu GZ, Hou W. Design, synthesis, biological evaluation, and mechanism of action of new pyrazines as anticancer agents in vitro and in vivo. Bioorg Med Chem 2025; 121:118108. [PMID: 39955800 DOI: 10.1016/j.bmc.2025.118108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 02/02/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025]
Abstract
Cancer is the second leading cause of mortality worldwide. The development of innovative antitumor pharmaceuticals is urgently needed to alter this circumstance. N-heterocycles, pyrazines for example are prevalent pharmacophores in the architecture of anticancer medicines. This research involved the design and synthesis of seventy-seven new pyrazine derivatives, followed by an evaluation of their anticancer activity in vitro and in vivo. Several new pyrazines exhibiting remarkable antiproliferative activity and selectivity were identified. The links between structure and function were analyzed, and the mechanisms of action were examined. Our mechanistic investigations indicated that these chemicals triggered mitochondria-associated apoptosis in cancer cells. Moreover, they suppressed the phosphorylation of STAT3, concomitant with the down-regulation of BcL-2, BcL-XL, c-Myc, XIAP, GLI1, TAZ, MCL1, JAK1, JAK2 and up-regulation of Bax, p21. Furthermore, the lead compounds B-11 and C-27 demonstrated significant anticancer activity in vivo in the SKOV3 xenograft nude mouse model. Our research establishes a basis for the identification of pyrazines as JAK/STAT3 inhibition based anticancer lead compounds.
Collapse
Affiliation(s)
- Jin-Xia Lan
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China; School of Public Health and Health Management, Gannan Medical University, Ganzhou 341000 PR China
| | - Le-Jun Huang
- School of Rehabilitation, Gannan Medical University, Ganzhou 341000 PR China
| | - Si-Shuang Kang
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China
| | - Hao-Huang
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China
| | - Sheng-Lan Liu
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China
| | - Wei Dai
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China
| | - Xin-Liang Xu
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China
| | - Jin-Yang Wang
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China
| | - Guang-Zhao Shu
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China
| | - Wen Hou
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China.
| |
Collapse
|
225
|
Chen G, Wang S, Mo X, Zhu W, Wang R, Song X. Leukocyte telomere length serves as the novel prognostic biomarker for the resectable NSCLC. Sci Rep 2025; 15:13002. [PMID: 40234460 PMCID: PMC12000439 DOI: 10.1038/s41598-025-95569-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/21/2025] [Indexed: 04/17/2025] Open
Abstract
The relationship of Leukocyte telomere length (LTL) dynamic changes with resectable NSCLC progression remains unclear. This study aims to reveal its clinical utility for prognosis of the resectable NSCLC. LTL was measured in 76 resectable NSCLC patients and 80 healthy controls using peripheral blood samples. Pre-operation LTL (Pre-LTL) and post-operation LTL (Po-LTL) were analyzed in relation to TNM stage, metastasis, and survival outcomes. The prognostic value was evaluated by disease-free survival (DFS) and overall survival (OS). NSCLC patients had significantly shorter LTL compared to controls, with LTL inversely correlated to disease stage. Po-LTL increased significantly and was associated with better OS. Combining Po-LTL with TNM stage improved prognostic prediction for OS and DFS. LTL is a promising biomarker for predicting prognosis in resectable NSCLC. Po-LTL, as well as in combination with TNM stage, enhances predictive accuracy for OS and DFS.
Collapse
Affiliation(s)
- Guanxuan Chen
- Oncology Department, Affiliated Zhongshan Hospital of Dalian University, No.6 Jiefang Street, Zhongshan District, Dalian, 116001, Liaoning, PR China
| | - Shiwen Wang
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, PR China
- Department of Pathology, Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Xinkai Mo
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, PR China
| | - Wanqi Zhu
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, PR China
| | - Ruoyu Wang
- Oncology Department, Affiliated Zhongshan Hospital of Dalian University, No.6 Jiefang Street, Zhongshan District, Dalian, 116001, Liaoning, PR China.
| | - Xingguo Song
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, PR China.
- Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, 250117, Shandong Province, PR China.
| |
Collapse
|
226
|
Ding P, Wu H, Wu J, Li T, Gu R, Zhang L, Niu X, He J, Yang J, Yang P, Guo H, Tian Y, Meng N, Li X, Guo Z, Meng L, Zhao Q. Non-invasive liquid biopsy based on transcriptomic profiling for early diagnosis of occult peritoneal metastases in locally advanced gastric cancer. NPJ Precis Oncol 2025; 9:109. [PMID: 40234664 PMCID: PMC12000504 DOI: 10.1038/s41698-025-00875-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 03/10/2025] [Indexed: 04/17/2025] Open
Abstract
This study proposes a novel non-invasive diagnostic approach utilizing transcriptomic profiling of liquid biopsy samples for the early detection of occult peritoneal metastases in locally advanced gastric cancer (LAGC). By analyzing RNA expression patterns of cancer cells, this method identifies specific gene signatures associated with peritoneal spread, potentially offering a more sensitive and comprehensive diagnostic tool compared to conventional imaging techniques. A 4-mRNA panel (BUB1, SPC25, CT83, MMP3) integrated with clinical features was developed into a Risk Stratification Assessment (RSA) model, demonstrating superior predictive accuracy in multiple cohorts with an area under the curve (AUC) of 0.836 in training and 0.882 in validation. This approach offers a promising alternative for early diagnosis, improving treatment decisions and clinical outcomes for gastric cancer patients, while enabling a shift from tissue-based testing to non-invasive blood-based diagnostics.
Collapse
Affiliation(s)
- Ping'an Ding
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Haotian Wu
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Jiaxiang Wu
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Tongkun Li
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Renjun Gu
- School of Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
- Department of Gastroenterology and Hepatology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Lilong Zhang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, 430065, Hubei, China
| | - Xiaoman Niu
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Jinchen He
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Jiaxuan Yang
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Peigang Yang
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Honghai Guo
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Yuan Tian
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Ning Meng
- Department of General Surgery, Shijiazhuang People's Hospital, Shijiazhuang, 050050, Hebei, China
| | - Xiaolong Li
- Department of General Surgery, Baoding Central Hospital, Baoding, 071030, Hebei, China
| | - Zhenjiang Guo
- Department of General Surgery, Hengshui People's Hospital, Hengshui, 053099, Hebei, China
| | - Lingjiao Meng
- Research Center and Tumor Research Institute of the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China.
| | - Qun Zhao
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China.
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China.
- Big data analysis and mining application for precise diagnosis and treatment of gastric cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China.
| |
Collapse
|
227
|
Li R, Sun X, Yu Z, Li P, Zhao X. Identification of predictors for lymph node metastasis in T2 colorectal cancer: retrospective cohort study from a high-volume hospital. BMC Cancer 2025; 25:700. [PMID: 40234815 PMCID: PMC12001727 DOI: 10.1186/s12885-025-14104-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 04/07/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the most prevalent malignant tumor of the digestive system globally, ranking third in incidence and second in mortality. In previous studies, the rate of lymph node metastasis (LNM) in T2 CRC ranged from 18.0 to 28.0%. We aim to identify T2 CRC patients without LNM and thereby mitigate the complications and potential impact on the quality of life associated with surgery. METHODS In this retrospective study, 787 cases with T2 CRC were selected. The preoperative and postoperative clinicopathological features were retrospectively studied. Univariate analysis and multivariate analysis were performed using binary logistic regression to determine the predictive factor for LNM. Odds ratio (OR) and 95% confidence interval (CI) were conducted. RESULTS 184 (23.4%) patients were diagnosed with LNM, including 144 (78.3%) patients with N1stage and 40 (21.7%) patients with N2 stage. According to univariate analysis and multivariate analysis, poorly differentiated tumors (p = 0.003, OR = 4.405, 95%CI: 1.632-11.893), perineural invasion (p = 0.001, OR = 4.789, 95%CI: 1.958-11.716), and lymphovascular invasion (p = 0.001, OR = 2.779, 95%CI: 1.497-5.159) were independent risk factors of LNM, while male (p = 0.017, OR = 0.652, 95%CI: 0.459-0.926) and elevated preoperative PLR (p = 0.048, OR = 0.996, 95%CI: 0.993-1.000) seemed to be independent protective factors. Larger tumor size did not show significant association with LNM. CONCLUSIONS Approximately three-quarters of T2 CRC patients are likely to avoid unnecessary surgery. Female, poorly differentiated tumors, perineural invasion, and lymphovascular invasion are expected to be used as predictors of LNM in T2 CRC.
Collapse
Affiliation(s)
- Rui Li
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, China
| | - Xu Sun
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, China
| | - Zhiyuan Yu
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, China
| | - Peiyu Li
- Medical School of Chinese PLA, Beijing, China.
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China.
- School of Medicine, Nankai University, Tianjin, China.
| | - Xudong Zhao
- Medical School of Chinese PLA, Beijing, China.
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China.
| |
Collapse
|
228
|
Tong Y, Chen Z, Wu J, Huang Q, He Y, Shang H, Xia D, Peng E, Wang Z, Liang X, Tang K. METTL3 promotes an immunosuppressive microenvironment in bladder cancer via m6A-dependent CXCL5/CCL5 regulation. J Immunother Cancer 2025; 13:e011108. [PMID: 40234090 PMCID: PMC12001370 DOI: 10.1136/jitc-2024-011108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/17/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Bladder cancer (BLCA) is a challenging malignancy with a poor prognosis, particularly in muscle-invasive cases. Despite recent advancements in immunotherapy, response rates remain suboptimal. This study investigates the role of METTL3, an m6A RNA methylation "writer," in regulating the immune microenvironment of BLCA. METHODS Through bioinformatics analysis, we identified METTL3 as being associated with the formation of an immunosuppressive microenvironment in BLCA and poor response to immunotherapy. Subsequently, we silenced METTL3 expression in BLCA cells using short hairpin RNA (shRNA) or inhibited its function with STM2457. The effectiveness of these interventions in remodeling the BLCA tumor microenvironment (TME) was confirmed through animal experiments and flow cytometry. Mechanistically, RNA sequencing and methylated RNA immunoprecipitation (MeRIP) sequencing revealed the molecular pathways by which METTL3 regulates the TME. This was further validated using in vitro cell co-culture, immunoprecipitation, ELISA, and RNA degradation assays. The synergistic effect of METTL3 with anti-Programmed Cell Death Protein 1 (PD-1) treatment in BLCA was confirmed in both orthotopic and ectopic BLCA animal models. RESULTS METTL3 was found to increase CXCL5 levels and suppress CCL5 expression in an m6A-dependent manner, leading to increased recruitment of myeloid-derived suppressor cells (MDSCs) and reduced infiltration of CD8+T cells. Silencing METTL3 or inhibiting its function restored immune cell balance and significantly enhanced the efficacy of anti-PD-1 therapy. Clinically, METTL3 overexpression correlated with poor complete response rate to immune checkpoint inhibitors (ICIs) therapy, associated with an immunosuppressive microenvironment characterized by elevated MDSC levels and reduced CD8+T cell infiltration. CONCLUSIONS These findings highlight METTL3 as a key regulator of the immune microenvironment in BLCA and a promising therapeutic target to improve immunotherapy outcomes. Targeting METTL3 could potentially enhance the efficacy of ICIs in patients with BLCA.
Collapse
Affiliation(s)
- Yonghua Tong
- Department of Urology, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiqiang Chen
- Department of Urology, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Wu
- Department of Urology, Huazhong University of Science and Technology, Wuhan, China
| | - Qiu Huang
- Department of Urology, Huazhong University of Science and Technology, Wuhan, China
| | - Yu He
- Department of Urology, Huazhong University of Science and Technology, Wuhan, China
| | - Haojie Shang
- Department of Urology, Huazhong University of Science and Technology, Wuhan, China
| | - Ding Xia
- Department of Urology, Huazhong University of Science and Technology, Wuhan, China
| | - Ejun Peng
- Department of Urology, Huazhong University of Science and Technology, Wuhan, China
| | - Zhihua Wang
- Department of Urology, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Liang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, Guangdong, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Kun Tang
- Department of Urology, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
229
|
Zhang L, Liu Y, Wen X, Zhang X, Fan P, Cao X. Integrated bioinformatics analysis reveals that OPRK1 inhibits ferroptosis and induces enzalutamide resistance in prostate cancer. Eur J Med Res 2025; 30:279. [PMID: 40229787 PMCID: PMC11998335 DOI: 10.1186/s40001-025-02484-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 03/20/2025] [Indexed: 04/16/2025] Open
Abstract
Enzalutamide (Enz) is employed in the management of castration-resistant prostate cancer (CRPC). However, a substantial subset of patients may develop resistance to Enz, thereby reducing its therapeutic effectiveness. The underlying mechanisms contributing to the development of Enz resistance in PCa, whether arising from androgen deprivation or the burden of Enz treatment, remain inadequately understood. OPRK1 plays a key role in Enz resistance through ferroptosis inhibition, which is detected by the analysis of Gene Expression Omnibus (GEO) databases. Silencing OPRK1 via small interfering RNA (siRNA) resulted in the activation of ferroptosis signaling in LNCaP cells. These findings indicate that OPRK1 significantly contributes to Enz resistance in PCa and may serve as a promising therapeutic target for resistant patients.
Collapse
Affiliation(s)
- Liangrong Zhang
- Department of Urology, The First Hospital of Shanxi Medical University, 85 Jiefang South Street, Yingze District, Taiyuan, 030001, Shanxi, People's Republic of China
- Department of Urology, Shanxi Provincial People's Hospital, Taiyuan, 030012, Shanxi, People's Republic of China
| | - Yanqin Liu
- Department of Nephrology, Shanxi Provincial People's Hospital, Taiyuan, 030012, Shanxi, People's Republic of China
| | - Xiaodong Wen
- Department of Urology, Shanxi Provincial People's Hospital, Taiyuan, 030012, Shanxi, People's Republic of China
| | - Xiangkai Zhang
- Department of Urology, Shanxi Provincial People's Hospital, Taiyuan, 030012, Shanxi, People's Republic of China
| | - Peng Fan
- Department of Radiology, Shanxi Provincial People's Hospital, Taiyuan, 030012, Shanxi, People's Republic of China
| | - Xiaoming Cao
- Department of Urology, The First Hospital of Shanxi Medical University, 85 Jiefang South Street, Yingze District, Taiyuan, 030001, Shanxi, People's Republic of China.
| |
Collapse
|
230
|
Qi J, Jiang T, Liu B, Hu Q, Chen J, Ma N, Xu Y, Song H, Song J. LINC02167 stabilizes KSR1 mRNA in an m 5C-dependent manner to regulate the ERK/MAPK signaling pathway and promotes colorectal cancer metastasis. J Exp Clin Cancer Res 2025; 44:121. [PMID: 40234937 PMCID: PMC11998267 DOI: 10.1186/s13046-025-03368-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/17/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Metastasis is a leading cause of colorectal cancer (CRC)-related mortality, yet its molecular mechanisms remain poorly understood. Long noncoding RNAs (lncRNAs) have emerged as critical regulators of CRC metastasis, but their specific roles are not fully elucidated. This study identifies and characterizes a novel lncRNA LINC02167 as a critical regulator of CRC metastasis. METHODS LINC02167 expression was analyzed in CRC tissues via real-time quantitative polymerase chain reaction and fluorescence in situ hybridization. Functional assays evaluated its role in CRC cell migration, invasion, and metastasis in vitro and in vivo. Mechanistic exploration involves a combination of techniques, including RNA sequencing, mass spectrometry, RNA pull-down, RNA immunoprecipitation, chromatin immunoprecipitation, luciferase reporter assays, RNA stability assays, and bioinformatics analysis, to uncover the molecular interactions and pathways regulated by LINC02167. RESULTS LINC02167 is markedly upregulated in CRC tissues and strongly correlates with advanced clinical features and poor prognosis. Functional analyses reveal that LINC02167 enhances CRC cell migration and invasion in vitro and promotes metastasis in vivo. Mechanistically, LINC02167 serves as a molecular scaffold, forming a complex with YBX1 and ILF3 to facilitate YBX1 binding to NSUN2-mediated m5C modification sites on KSR1 mRNA, thereby stabilizing KSR1 mRNA and activating the ERK/MAPK signaling pathway to drive CRC metastasis. Additionally, MYC-driven transcriptional activation leads to the upregulation of LINC02167 in CRC. CONCLUSIONS This study uncovers a novel mechanism through which LINC02167 promotes the ERK/MAPK pathway and CRC metastasis via m5C modification, underscoring its potential as a promising therapeutic target for metastatic CRC treatment.
Collapse
Affiliation(s)
- Junwen Qi
- Affiliated First Clinical College, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Tao Jiang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
- Affiliated First Clinical College, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Bowen Liu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Qihang Hu
- Affiliated First Clinical College, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Junnan Chen
- Affiliated First Clinical College, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Ning Ma
- Affiliated First Clinical College, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Yixin Xu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Hu Song
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China.
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China.
| | - Jun Song
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China.
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China.
| |
Collapse
|
231
|
Davis MB, Martini R. Precision oncology and genetic ancestry: The science behind population-based cancer disparities. Cancer Cell 2025; 43:619-622. [PMID: 40233717 DOI: 10.1016/j.ccell.2025.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 03/16/2025] [Accepted: 03/17/2025] [Indexed: 04/17/2025]
Abstract
Precision oncology has transformed cancer care but has barely benefited patients of diverse ancestry. Historically, self-reported race/ethnicity has served as a surrogate for biological differences, but genetic ancestry provides a more precise framework for understanding genetic drivers of cancer disparities, including associations between ancestry and tumor subtypes, and genetic variants affecting drug metabolism and treatment response. To improve precision oncology for all patients and reduce cancer disparities, we propose expanding ancestry-inclusive genomic data, reevaluating disease-associated variants within ancestrally diverse cohorts, and standardizing data-sharing practices.
Collapse
Affiliation(s)
- Melissa Boneta Davis
- Institute of Translational Genomic Medicine, Morehouse School of Medicine, Atlanta, GA, USA; Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, USA; Georgia Research Alliance, Atlanta, GA, USA.
| | - Rachel Martini
- Institute of Translational Genomic Medicine, Morehouse School of Medicine, Atlanta, GA, USA; Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| |
Collapse
|
232
|
Li F, Wan X, Li Z, Zhou L. The NR3C2-SIRT1 signaling axis promotes autophagy and inhibits epithelial mesenchymal transition in colorectal cancer. Cell Death Dis 2025; 16:295. [PMID: 40229278 PMCID: PMC11997134 DOI: 10.1038/s41419-025-07575-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 02/23/2025] [Accepted: 03/18/2025] [Indexed: 04/16/2025]
Abstract
Colorectal cancer (CRC) is one of the most aggressive and lethal cancers with a complex pathogenesis, there is an urgent need to find new drug therapeutic targets. This study highlights the important role of the NR3C2-SIRT1 signaling axis in the metastasis mechanism of CRC. Our findings revealed that the expression of NR3C2 in CRC tissues was lower than that in adjacent non-cancerous tissues, and was negatively correlated with N stage by bioanalysis, IHC, western blot and qRT-PCR. NR3C2 overexpression / knockdown can significantly inhibit / promote the migration and invasion of CRC cells, at the same time inhibit / promote EMT. Mechanically, the regulatory molecule SIRT1 was identified by RNA-seq, bioinformatics analysis, western blot and ChIP. SIRT1 was also involved in the metastasis process of CRC, and NR3C2 was found to regulate the expression of LC3B and SQSTM1/p62 in a SIRT1-dependent manner. Therefore, NR3C2 forms a signaling axis with SIRT1, which can directly promote autophagy and inhibit EMT process in vivo and in vitro. Collectively, our findings suggest that NR3C2 - SIRT1 signal axis promote autophagy and inhibit EMT, ultimately inhibits lung metastasis of CRC.
Collapse
Affiliation(s)
- Feng Li
- Department of Pharmacology, West China School of Basic Science and Forensic Medicine, Sichuan University, Chengdu, China
| | - Xing Wan
- Department of Pharmacology, West China School of Basic Science and Forensic Medicine, Sichuan University, Chengdu, China
| | - Zhigui Li
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China.
| | - Liming Zhou
- Department of Pharmacology, West China School of Basic Science and Forensic Medicine, Sichuan University, Chengdu, China.
| |
Collapse
|
233
|
Galsky MD, Autio KA, Cabanski CR, Wentzel K, Graff JN, Friedlander TW, Howes TR, Shotts KM, Densmore J, Spasic M, Da Silva DM, Chen RO, Lata J, Skolnik J, Keler T, Yellin MJ, LaVallee TM, Fairchild J, Boffo S, O’Donnell-Tormey J, Dugan U, Bhardwaj N, Subudhi SK, Fong L. Clinical and Translational Results from PORTER, a Multicohort Phase I Platform Trial of Combination Immunotherapy in Metastatic Castration-Resistant Prostate Cancer. Clin Cancer Res 2025; 31:1463-1475. [PMID: 39964352 PMCID: PMC11995007 DOI: 10.1158/1078-0432.ccr-24-3693] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/03/2025] [Accepted: 02/14/2025] [Indexed: 04/15/2025]
Abstract
PURPOSE Current immune checkpoint therapies offer limited benefits for metastatic castration-resistant prostate cancer. Novel combinations may enhance immunotherapy efficacy. PATIENTS AND METHODS We conducted an open-label, noncomparative platform trial (NCT03835533) in metastatic castration-resistant prostate cancer to assess nivolumab-based combinations. The cohorts were as follows: (A) bempegaldesleukin 0.006 mg/kg and nivolumab 360 mg i.v. every 3 weeks; (B) stereotactic body radiotherapy 30 to 50 Gy, CDX-301 75 μg/kg s.c. for 5 days, poly-ICLC 1 mg intramuscularly weekly twice for 3 weeks, and nivolumab 480 mg every 4 weeks; and (C) CDX-301 75 μg/kg for 10 days, INO-5151 3 mg intramuscularly on lead-in day 8, day 1 of cycles 1 to 3, and then every 12 weeks, and nivolumab 480 mg every 4 weeks. The primary endpoint was safety; secondary endpoints included composite response rate (radiographic, PSA, or circulating tumor cell responses), 6-month disease control rate, progression-free survival, and overall survival. Serial blood and tissue samples were analyzed for pharmacodynamics and association with disease control. RESULTS A total of 43 patients were enrolled (n = 14, 15, and 14 in cohorts A, B, and C, respectively). Grade 3 to 4 treatment-related adverse events occurred in 10 (71%), 2 (13%), and 2 (14%) patients, respectively, with one grade 5 treatment-related adverse event in cohort A. Composite response rates were 7% (1/14), 33% (5/15), and 7% (1/14). Across cohorts, 6-month disease control was associated with preexisting memory/regulatory T cells, TNFα, and other inflammatory pathways. CONCLUSIONS Cohort B, which combined radiotherapy with CDX-301, poly-ICLC, and nivolumab, demonstrated encouraging clinical activity. Preexisting rather than treatment-induced immune activation was associated with clinical benefit across cohorts, highlighting the importance of baseline immune fitness.
Collapse
Affiliation(s)
- Matthew D. Galsky
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Karen A. Autio
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| | | | | | - Julie N. Graff
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
- VA Portland Health Care System, Portland, Oregon
| | - Terence W. Friedlander
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Timothy R. Howes
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Kristin M. Shotts
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Julie Densmore
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Marko Spasic
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Diane M. Da Silva
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | | | - Jennifer Lata
- Inovio Pharmaceuticals, Plymouth Meeting, Pennsylvania
| | | | - Tibor Keler
- Celldex Therapeutics, Inc., Hampton, New Jersey
| | | | | | - Justin Fairchild
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | | | | | - Ute Dugan
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Nina Bhardwaj
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Sumit K. Subudhi
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lawrence Fong
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, California
- Immunotherapy Integrated Research Center, Fred Hutchison Cancer Center, Seattle, Washington
| |
Collapse
|
234
|
Atuati SF, Becker G, Oliveira SM. Diosmetin Attenuates Painful Symptoms Caused by Antineoplastics Paclitaxel and Anastrozole in Mice. Mol Neurobiol 2025:10.1007/s12035-025-04939-w. [PMID: 40229455 DOI: 10.1007/s12035-025-04939-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 04/08/2025] [Indexed: 04/16/2025]
Abstract
Cancer patients often experience painful symptoms as an adverse effect resulting from various factors, including antineoplastic therapy. Paclitaxel is a chemotherapeutic agent used to treat solid tumours and, unfortunately, causes acute and chronic peripheral neuropathic pain in patients. Anastrozole is an antineoplastic used to treat hormone receptor-positive breast cancer and causes musculoskeletal pain symptoms. The therapies used to control these pain symptoms have limited efficacy and cause adverse effects, highlighting the importance of finding new treatments. Diosmetin is a flavonoid that has shown efficacy in neuropathic, nociplastic, and inflammatory pain models. Therefore, we verified the antinociceptive effects of diosmetin on nociceptive parameters induced by paclitaxel and anastrozole. We investigated the effects of diosmetin (0.015, 0.15 and 1.5 mg/kg; oral route (p.o.)) on nociceptive parameters (mechanical and cold allodynia and pain affective‑motivational behaviour) induced by a single and repeated paclitaxel (1 mg/kg, intraperitoneal (i.p.)) administrations, as well as on the pain-like symptoms (mechanical allodynia, muscle strength, and pain affective‑motivational behaviour) of a single administration of anastrozole (0.2 mg/kg, p.o.) and the combination of both antineoplastics in mice. Diosmetin reduced the nociceptive parameters induced by paclitaxel and anastrozole. In addition, we observed that anastrozole exacerbated the mechanical allodynia induced by paclitaxel, an effect that was also reversed by diosmetin. These results suggest that diosmetin may be an effective therapeutic alternative for the treatment of painful symptoms caused by antineoplastic drugs, such as paclitaxel and anastrozole.
Collapse
Affiliation(s)
- Samuel Felipe Atuati
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Centre of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Gabriela Becker
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Centre of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Sara Marchesan Oliveira
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Centre of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil.
- Department of Biochemistry and Molecular Biology, Centre of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil.
| |
Collapse
|
235
|
Martin SD, Thornton S, Chow C, Milne K, de Barros JS, Morris KA, Leung S, Jamieson A, Nelson BH, Cochrane DR, Huntsman DG, Gilks CB, Hoang L, McAlpine JN, Zhang AW. Activated immune infiltrates expand opportunities for targeted therapy in p53-abnormal endometrial carcinoma. J Pathol 2025. [PMID: 40223796 DOI: 10.1002/path.6429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 02/12/2025] [Accepted: 03/17/2025] [Indexed: 04/15/2025]
Abstract
Tumor protein p53 mutated/abnormal (p53abn) endometrial carcinomas account for over 50% of deaths but comprise only 15% of all endometrial carcinomas. Most patients show limited response to standard-of-care chemotherapy with or without radiotherapy, and only a minority of cases are amenable to targeted therapies like poly-ADP ribose polymerase (PARP) inhibitors and HER2-directed therapies. Recent immunotherapy clinical trials have demonstrated remarkable efficacy, not only in mismatch repair deficient (MMRd) tumors but also in a subset of mismatch repair-proficient (MMRp) tumors. However, the immune microenvironment and its relationship to other therapeutic targets in MMRp endometrial carcinoma remains poorly understood. Here, we characterize the immune microenvironment of p53abn endometrial carcinoma, the most clinically aggressive subtype of MMRp endometrial carcinoma, and correlate antitumor immune signatures with other targetable alterations. We accrued 256 treatment-naïve p53abn endometrial carcinomas and systemically profiled T-cell, B-cell, myeloid, and tumor-cell populations with multiplex immunofluorescence to assess the tissue localization and functional status of immune cells. Shallow whole-genome sequencing was performed on a subset of 126 cases. Patterns of immune infiltration were compared to survival outcomes and mutational signatures. Mixture modeling divided p53abn endometrial carcinoma into tumor-infiltrating lymphocyte (TIL)-rich and TIL-poor subsets. Over 50% of tumors were TIL-rich. TIL-rich cases overexpressed targetable immune evasion molecules and were associated with longer overall and disease-specific survival in multivariate analysis. This effect was particularly pronounced in advanced stage disease and in patients who did not receive adjuvant chemotherapy. TIL did not associate with homologous recombination deficient mutational signatures or HER2 amplification. Our findings demonstrate a biological rationale for immunotherapy in a substantial subset of patients with p53abn endometrial cancer and may help inform combination therapies with immune checkpoint inhibition, PARP inhibitors, and anti-HER2 agents. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Spencer D Martin
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| | - Shelby Thornton
- Molecular and Advanced Pathology Core (MAPcore), The University of British Columbia, Vancouver, Canada
| | - Christine Chow
- Molecular and Advanced Pathology Core (MAPcore), The University of British Columbia, Vancouver, Canada
| | - Katy Milne
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada
| | - Juliana Sobral de Barros
- Department of Molecular Oncology, British Columbia Cancer Agency, The University of British Columbia, Vancouver, Canada
| | - Kayleigh A Morris
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada
| | - Samuel Leung
- Department of Molecular Oncology, British Columbia Cancer Agency, The University of British Columbia, Vancouver, Canada
| | - Amy Jamieson
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, The University of British Columbia, Vancouver, Canada
| | - Brad H Nelson
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada
- Department of Medical Genetics, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| | - Dawn R Cochrane
- Department of Molecular Oncology, British Columbia Cancer Agency, The University of British Columbia, Vancouver, Canada
| | - David G Huntsman
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| | - C Blake Gilks
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| | - Lien Hoang
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| | - Jessica N McAlpine
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, The University of British Columbia, Vancouver, Canada
| | - Allen W Zhang
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| |
Collapse
|
236
|
Smith-Bindman R, Chu PW, Azman Firdaus H, Stewart C, Malekhedayat M, Alber S, Bolch WE, Mahendra M, Berrington de González A, Miglioretti DL. Projected Lifetime Cancer Risks From Current Computed Tomography Imaging. JAMA Intern Med 2025:2832778. [PMID: 40227719 PMCID: PMC11997853 DOI: 10.1001/jamainternmed.2025.0505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/11/2025] [Indexed: 04/15/2025]
Abstract
Importance Approximately 93 million computed tomography (CT) examinations are performed on 62 million patients annually in the United States, and ionizing radiation from CT is a known carcinogen. Objective To project the number of future lifetime cancers in the US population associated with CT imaging in 2023. Design, Setting, and Participants This risk model used a multicenter sample of CT examinations prospectively assembled between January 2018 and December 2020 from the University of California San Francisco International CT Dose Registry. Data analysis was conducted from October 2023 to October 2024. Main Outcomes and Measures Distributions of CT examinations and associated organ-specific radiation doses were estimated by patient age, sex, and CT category and scaled to the US population based on the number of examinations in 2023, quantified by the IMV national survey. Lifetime radiation-induced cancer incidence and 90% uncertainty limits (UL) were estimated by age, sex, and CT category using National Cancer Institute software based on the National Research Council's Biological Effects of Ionizing Radiation VII models and projected to the US population using scaled examination counts. Results An estimated 61 510 000 patients underwent 93 000 000 CT examinations in 2023, including 2 570 000 (4.2%) children, 58 940 000 (95.8%) adults, 32 600 000 (53.0%) female patients, and 28 910 000 (47.0%) male patients. Approximately 103 000 (90% UL, 96 400-109 500) radiation-induced cancers were projected to result from these examinations. Estimated radiation-induced cancer risks were higher in children and adolescents, yet higher CT utilization in adults accounted for most (93 000; 90% UL, 86 900-99 600 [91%]) radiation-induced cancers. The most common cancers were lung cancer (22 400 cases; 90% UL, 20 200-25 000 cases), colon cancer (8700 cases; 90% UL, 7800-9700 cases), leukemia (7900 cases; 90% UL, 6700-9500 cases), and bladder cancer (7100 cases, 90% UL, 6000-8500 cases) overall, while in female patients, breast was second most common (5700 cases; 90% UL, 5000-6500 cases). The largest number of cancers was projected to result from abdomen and pelvis CT in adults, reflecting 37 500 of 103 000 cancers (37%) and 30 million of 93 million CT examinations (32%), followed by chest CT (21 500 cancers [21%]; 20 million examinations [21%]). Estimates remained large over a variety of sensitivity analyses, which resulted in a range of 80 000 to 127 000 projected cancers across analyses. Conclusions and Relevance This study found that at current utilization and radiation dose levels, CT examinations in 2023 were projected to result in approximately 103 000 future cancers over the course of the lifetime of exposed patients. If current practices persist, CT-associated cancer could eventually account for 5% of all new cancer diagnoses annually.
Collapse
Affiliation(s)
- Rebecca Smith-Bindman
- Department of Epidemiology and Biostatistics, University of California, San Francisco
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco
- Philip R. Lee Institute for Health Policy Studies, University of California, San Francisco
| | - Philip W. Chu
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Hana Azman Firdaus
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Carly Stewart
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Matthew Malekhedayat
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Susan Alber
- Department of Public Health Sciences, University of California, Davis
| | - Wesley E. Bolch
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville
| | - Malini Mahendra
- Philip R. Lee Institute for Health Policy Studies, University of California, San Francisco
- Division of Pediatric Critical Care, Department of Pediatrics, UCSF Benioff Children’s Hospital, University of California, San Francisco
| | | | - Diana L. Miglioretti
- Department of Public Health Sciences, University of California, Davis
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle
| |
Collapse
|
237
|
Wang M, Ding Q, Su W, Luo M, Yang R, Chen G, Wang Q, Zhang N, Gao J, Wang X, Huang T, Liu P, Fu D, Hong X, Zeng X, Wei Y, Xiao Y. A Mitochondrion-Targeted NIR-II Modulator for Synergistic Ferroptosis-Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2501397. [PMID: 40223477 DOI: 10.1002/smll.202501397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/19/2025] [Indexed: 04/15/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have limited clinical efficacy against gastric cancer (GC) due to the nonimmunogenic tumor microenvironment. Therefore, inducing immunogenic cell death (ICD) to reprogram the immunogenic landscape is essential. This study develops HD-FA nanoparticles by encapsulating a novel mitochondrion-targeted NIR-II modulator, HD, within DSPE-PEG-FA. HD-FA exhibits superior spatiotemporal resolution, robust tumor accumulation, and minimal adverse effects. Upon 808 nm laser irradiation, HD-FA generates reactive oxygen species, leading to ferroptosis and oxidative stress damage in GC cells by inhibiting the SLC7A11/GSH/GPX4 axis. HD-FA triggers ICD, resulting in antitumor activity not only in primary tumors but also in distant tumors. Moreover, HD-FA promotes dendritic cell maturation, increases the effector-memory T-cell frequency, and reduces the presence of myeloid-derived suppressor cells, thereby fostering enhanced antitumor immunity. This study presents the first report of a novel NIR-II modulator for GC immunogenic synergistic therapy with ICIs, marking significant advancements in the fight against GC.
Collapse
Affiliation(s)
- Miao Wang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Hubei Cancer Clinical Study Center & Hubei Key Laboratory of Tumor Biological Behaviors, School of Pharmaceutical Sciences, Wuhan, 430071, China
| | - Qihang Ding
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Hubei Cancer Clinical Study Center & Hubei Key Laboratory of Tumor Biological Behaviors, School of Pharmaceutical Sciences, Wuhan, 430071, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| | - Wuyue Su
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Hubei Cancer Clinical Study Center & Hubei Key Laboratory of Tumor Biological Behaviors, School of Pharmaceutical Sciences, Wuhan, 430071, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
- Shenzhen Institute of Wuhan University, Shenzhen, 518057, China
| | - Min Luo
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Hubei Cancer Clinical Study Center & Hubei Key Laboratory of Tumor Biological Behaviors, School of Pharmaceutical Sciences, Wuhan, 430071, China
| | - Runping Yang
- Department of Dermatology, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Guopeng Chen
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Qian Wang
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Nan Zhang
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jialu Gao
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Hubei Cancer Clinical Study Center & Hubei Key Laboratory of Tumor Biological Behaviors, School of Pharmaceutical Sciences, Wuhan, 430071, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
- Shenzhen Institute of Wuhan University, Shenzhen, 518057, China
| | - Xiaofen Wang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Hubei Cancer Clinical Study Center & Hubei Key Laboratory of Tumor Biological Behaviors, School of Pharmaceutical Sciences, Wuhan, 430071, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
- Shenzhen Institute of Wuhan University, Shenzhen, 518057, China
| | - Tianhe Huang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Hubei Cancer Clinical Study Center & Hubei Key Laboratory of Tumor Biological Behaviors, School of Pharmaceutical Sciences, Wuhan, 430071, China
| | - Pan Liu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Hubei Cancer Clinical Study Center & Hubei Key Laboratory of Tumor Biological Behaviors, School of Pharmaceutical Sciences, Wuhan, 430071, China
| | - Dujiang Fu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Hubei Cancer Clinical Study Center & Hubei Key Laboratory of Tumor Biological Behaviors, School of Pharmaceutical Sciences, Wuhan, 430071, China
| | - Xuechuan Hong
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Hubei Cancer Clinical Study Center & Hubei Key Laboratory of Tumor Biological Behaviors, School of Pharmaceutical Sciences, Wuhan, 430071, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
- Shenzhen Institute of Wuhan University, Shenzhen, 518057, China
| | - Xiaodong Zeng
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Hubei Cancer Clinical Study Center & Hubei Key Laboratory of Tumor Biological Behaviors, School of Pharmaceutical Sciences, Wuhan, 430071, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
| | - Yongchang Wei
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Hubei Cancer Clinical Study Center & Hubei Key Laboratory of Tumor Biological Behaviors, School of Pharmaceutical Sciences, Wuhan, 430071, China
| | - Yuling Xiao
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Hubei Cancer Clinical Study Center & Hubei Key Laboratory of Tumor Biological Behaviors, School of Pharmaceutical Sciences, Wuhan, 430071, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
| |
Collapse
|
238
|
Ha A, Woolman M, Waas M, Govindarajan M, Kislinger T. Recent implementations of data-independent acquisition for cancer biomarker discovery in biological fluids. Expert Rev Proteomics 2025. [PMID: 40227112 DOI: 10.1080/14789450.2025.2491355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/26/2025] [Accepted: 04/06/2025] [Indexed: 04/15/2025]
Abstract
INTRODUCTION Cancer is the second leading cause of death worldwide and accurate biomarkers for early detection and disease monitoring are needed to improve outcomes. Biological fluids, such as blood and urine, are ideal samples for biomarker measurements as they can be routinely collected with relatively minimally invasive methods. However, proteomics analysis of fluids has been a challenge due to the high dynamic range of its protein content. Advances in data-independent acquisition (DIA) mass spectrometry-based proteomics can address some of the technical challenges in the analysis of biofluids, thus enabling the ability for mass spectrometry to propel large-scale biomarker discovery. AREAS COVERED We reviewed principles of DIA and its recent applications in cancer biomarker discovery using biofluids. We summarized DIA proteomics studies using biological fluids in the context of cancer research over the past decade, and provided a comprehensive overview of the benefits and challenges of DIA-MS. EXPERT OPINION Various studies showed the potential of DIA-MS in identifying putative cancer biomarkers in a high-throughput manner. However, the lack of proper study design and standardization of methods across platforms still need to be addressed to fully utilize the benefits of DIA-MS to accelerate the biomarker discovery and verification processes.
Collapse
Affiliation(s)
- Annie Ha
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Michael Woolman
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Matthew Waas
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Meinusha Govindarajan
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Thomas Kislinger
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| |
Collapse
|
239
|
Zhan J, Cai Y, Cheng P, Zheng L, Pu K. Body fluid diagnostics using activatable optical probes. Chem Soc Rev 2025; 54:3906-3929. [PMID: 40084539 DOI: 10.1039/d4cs01315h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
In vitro diagnostics often detects biomarkers in body fluids (such as blood, urine, sputum, and cerebrospinal fluids) to identify life-threatening diseases at an early stage, monitor overall health, or provide information to help cure, treat, or prevent diseases. Most clinically used optical in vitro diagnostic tests utilize dye-labeled biomolecules for biomarker recognition and signal readout, which typically involve complex steps and long processing times. Activatable optical probes (AOPs), which spontaneously activate their optical signals only in the presence of disease biomarkers, offer higher signal-to-background ratios and improved detection specificity. They also have the potential to simplify detection procedures by eliminating multiple washing steps. In this review, we summarize recent advances in the use of AOPs for pre-clinical and clinical body fluid diagnostics across various diseases, including cancer, nephro-urological disorders, infectious diseases, and digestive diseases. We begin by discussing the molecular design strategies of AOPs to achieve different optical signal readouts and biomarker specificity. We then highlight their diagnostic applications in various disease models and body fluids. Finally, we address the challenges and future perspectives of AOPs in enhancing body fluid diagnostics and advancing precision medicine.
Collapse
Affiliation(s)
- Jie Zhan
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Yanbin Cai
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Penghui Cheng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, China.
| | - Lei Zheng
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, 636921, Singapore, Singapore
| |
Collapse
|
240
|
Song T, Li S, Zhao K, Zou D, Zhang M, Wang H. Comprehensive analysis of prognosis and tumor immune microenvironment of cuproptosis-related gene CDKN2A in lung adenocarcinoma. BMC Pulm Med 2025; 25:179. [PMID: 40229853 PMCID: PMC11998297 DOI: 10.1186/s12890-025-03631-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 03/27/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Recent research has increasingly highlighted the significance of various forms of cell death in contributing to tumor heterogeneity and modulating anti-tumor immunity. However, the potential implications of cuproptosis-related genes (CRGs) in lung adenocarcinoma (LUAD) remains poorly explored. METHODS We conducted a comprehensive analysis of the expression profiles of 19 CRGs in LUAD based on The Cancer Genome Atlas (TCGA). Utilizing consensus clustering, we stratified the TCGA cohort into two distinct LUAD subtypes (Cluster 1 and Cluster 2). The expression of CDKN2A was further validated across multiple datasets, including TCGA, GEO, Cancer Cell Line Encyclopedia (CCLE), and the Human Protein Atlas (HPA). The prognostic value of the CDKN2A was evaluated through univariate, multivariate, and survival analyses. Gene set enrichment analysis (GSEA) was performed to elucidate the molecular mechanisms associated with the CDKN2A. Additionally, we assessed the levels of immune cell infiltration in LUAD using the CIBERSORT, ESTIMATE, and XCELL algorithms. RESULTS By systematically analyzing the genetic alterations of 19 CRGs in LUAD, we found 15 differentially expressed genes between LUAD and adjacent normal tissues. Subsequently, using the consensus clustering method, we classified LUAD patients into two molecular subtypes and cluster 2 had a poor prognosis. CDKN2A emerged as a key gene of interest, exhibiting elevated expression in LUAD and correlating with adverse patient outcomes. Moreover, immunoinfiltration analysis revealed differential levels of immune cell infiltration between the CDKN2A high and CDKN2A low expression groups. CONCLUSIONS Our findings indicate that CDKN2A may serve as an effective prognostic biomarker for LUAD and may offer valuable insights into potential immunotherapeutic strategies for these patients. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Teng Song
- Department of Oncology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, 300121, China
- The Institute of Translational Medicine, Tianjin Union Medical Center, NanKai University, Tianjin, 300121, China
- Tianjin Cancer Institute of lntegrative Traditional Chinese and Western Medicine, Tianjin, 300121, China
| | - Shuping Li
- Department of Oncology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, 300121, China
- The Institute of Translational Medicine, Tianjin Union Medical Center, NanKai University, Tianjin, 300121, China
- Tianjin Cancer Institute of lntegrative Traditional Chinese and Western Medicine, Tianjin, 300121, China
| | - Ke Zhao
- Department of Oncology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, 300121, China
- The Institute of Translational Medicine, Tianjin Union Medical Center, NanKai University, Tianjin, 300121, China
- Tianjin Cancer Institute of lntegrative Traditional Chinese and Western Medicine, Tianjin, 300121, China
| | - Dandan Zou
- Department of Oncology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, 300121, China
- The Institute of Translational Medicine, Tianjin Union Medical Center, NanKai University, Tianjin, 300121, China
- Tianjin Cancer Institute of lntegrative Traditional Chinese and Western Medicine, Tianjin, 300121, China
| | - Miao Zhang
- Department of Oncology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, 300121, China.
- The Institute of Translational Medicine, Tianjin Union Medical Center, NanKai University, Tianjin, 300121, China.
- Tianjin Cancer Institute of lntegrative Traditional Chinese and Western Medicine, Tianjin, 300121, China.
| | - Huaqing Wang
- Department of Oncology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, 300121, China.
- The Institute of Translational Medicine, Tianjin Union Medical Center, NanKai University, Tianjin, 300121, China.
- Tianjin Cancer Institute of lntegrative Traditional Chinese and Western Medicine, Tianjin, 300121, China.
| |
Collapse
|
241
|
Ciombor KK, Bae SW, Whisenant JG, Ayers GD, Sheng Q, Peterson TE, Smith GT, Lin K, Chowdhury S, Kanikarla Marie P, Sorokin A, Cohen AS, Goff LW, Cardin DB, Shen JP, Kopetz S, Eng C, Shyr Y, Berlin J, Manning HC. Results of the Phase I/II Study and Preliminary B-cell Gene Signature of Combined Inhibition of Glutamine Metabolism and EGFR in Colorectal Cancer. Clin Cancer Res 2025; 31:1437-1448. [PMID: 39927885 PMCID: PMC11996605 DOI: 10.1158/1078-0432.ccr-24-3133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/20/2024] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
PURPOSE EGFR-targeting mAbs are essential for managing rat sarcoma virus wild-type metastatic colorectal cancer (mCRC), but their limited efficacy necessitates exploring immunologic and metabolic factors influencing response. This study evaluated glutamine metabolism targeting with EGFR inhibition to identify response biomarkers in patients with prior anti-EGFR treatment progression. PATIENTS AND METHODS We conducted a phase I/II trial in patients with KRAS wild-type mCRC, combining panitumumab (6 mg/kg) and CB-839 (600 mg/kg or 800 mg/kg), hypothesizing that the dual inhibition of glutamine metabolism and MAPK signaling would enhance outcomes. As study correlatives, we investigated the B-cell activation signature "B-score" and glutamine PET as potential treatment response biomarkers. RESULTS The combination of panitumumab and CB-839 was tolerable with manageable side effects, including grade 4 hypomagnesemia in four patients, a known panitumumab-related event. Two patients achieved partial response, and five had stable disease, with a 41% disease control rate. Median progression-free survival and overall survival were 1.84 and 8.87 months, respectively. A positive correlation between "B-score" and lesion size reduction suggested its association with clinical benefit (partial response and stable disease). Lower "B-score" correlated with greater tumor avidity for glutamine by PET, indicating B-cell activation sensitivity to glutamine depletion. CONCLUSIONS The combination of CB-839 and panitumumab showed safety and promising preliminary responses, but the study closed early due to CB-839 development termination. The B-cell activation signature "B-score" emerged as a potential biomarker for EGFR and glutaminase inhibition in mCRC, warranting further studies. These findings suggest opportunities to improve immune response and therapies in glutaminolysis-dependent tumors.
Collapse
Affiliation(s)
- Kristen K Ciombor
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Seong-Woo Bae
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jennifer G Whisenant
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Gregory D Ayers
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Todd E Peterson
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Gary T Smith
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Kangyu Lin
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Saikat Chowdhury
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Preeti Kanikarla Marie
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alexey Sorokin
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Allison S Cohen
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Laura W Goff
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Dana B Cardin
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - John Paul Shen
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cathy Eng
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Yu Shyr
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jordan Berlin
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - H Charles Manning
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
242
|
Chladek M, Meza MV, Wang J, Sae-Hau M, Buenfil A, Turnbull J, Zaman F, Despiegel N. Understanding Patients' Experiences in Newly Diagnosed Adult B Cell Acute Lymphoblastic Leukemia: Qualitative Interviews to Develop a Patient-Centric Conceptual Model. Oncol Ther 2025:10.1007/s40487-025-00336-4. [PMID: 40221946 DOI: 10.1007/s40487-025-00336-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 03/26/2025] [Indexed: 04/15/2025] Open
Abstract
INTRODUCTION Treatment outcomes for older adults with B cell acute lymphoblastic leukemia (B cell ALL) are poor, partially because of poor tolerance to intense chemotherapy. Information on patient experience-an important consideration in drug development-is lacking. We investigated the signs, symptoms, and impacts of B cell ALL on older patients (or those with comorbidities that may reduce chemotherapy tolerance). METHODS This observational study involved teleconference-based, qualitative, semi-structured interviews with patients newly diagnosed with B cell ALL, aged ≥ 55 years, or 30-54 years with ≥ 1 comorbidity. Participants described their B cell ALL experience, including signs, symptoms, and impacts, and how bothersome/disturbing these were from 0 (not at all) to 10 (greatly) at three timepoints (around diagnosis, at worst, and at interview). Salient signs/symptoms were those reported by ≥ 40%, with average disturbance ratings of ≥ 4. A conceptual model of key disease- and treatment-related signs, symptoms, and impacts was developed. RESULTS Interviews with 20 participants (mean age 57.9 years; 80% diagnosed within 18 months) revealed 63 signs/symptoms and 37 impacts. All reported fatigue-related symptoms, and most reported gastrointestinal (n = 18, 90%), central/peripheral nervous system (n = 16, 80%), and pain-, respiratory-, blood-, and mouth-related (all n = 14, 70%) symptoms. Eight signs/symptoms were salient around diagnosis (fatigue, tiredness, weakness, exhaustion, shortness of breath, sweating, general pain, and diarrhea) and 16 were salient "at worst"; four remained salient at interview (all fatigue-related). All participants reported emotional impacts, and most reported physical and social impacts (both n = 16, 80%). The most frequent impact was inability to do previous hobbies/activities (n = 15, 75%), followed by decreased ability for activities of daily living and worry/fear/nervousness (both n = 12, 60%). CONCLUSION This study provides insight into patients' experience with newly diagnosed B cell ALL among older patients or those with clinically significant comorbidities. This enhances understanding of what matters most to patients and informs future treatment development and clinical care.
Collapse
Affiliation(s)
| | | | - Jessie Wang
- Amgen Inc, 1 Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - Maria Sae-Hau
- The Leukemia & Lymphoma Society, Washington, DC, USA
| | | | | | - Faraz Zaman
- Amgen Inc, 1 Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | | |
Collapse
|
243
|
Goetz MP, Wander SA, Bachelot T, de Nonneville A, Gal-Yam EN, Sammons SL, Shen S, Twelves C, Boruta G, Portman DJ, Damodaran S. ELAINE 3: phase 3 study of lasofoxifene plus abemaciclib to treat ER+/HER2-, ESR1-mutated, metastatic breast cancer. Future Oncol 2025:1-8. [PMID: 40222048 DOI: 10.1080/14796694.2025.2481825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 03/17/2025] [Indexed: 04/15/2025] Open
Abstract
Endocrine therapy (ET) is recommended for patients with estrogen receptor-positive (ER+) metastatic breast cancer (mBC), but most patients will develop treatment resistance, often due to mutations in the ER-α-coding gene, ESR1. Therapeutic options are limited for endocrine-resistant mBC, particularly following treatment with a cyclin-dependent kinase 4/6 inhibitor (CDK4/6i). Lasofoxifene had anti-tumor activity in two separate phase 2, open-label studies (ELAINE 1 and 2) when given as monotherapy or combined with abemaciclib. The phase 3, randomized ELAINE 3 trial will evaluate the efficacy and safety of lasofoxifene/abemaciclib versus fulvestrant/abemaciclib for locally advanced or metastatic, ER+/HER2- breast cancer with an ESR1 mutation that progressed after ET-CDK4/6i treatment. Enrollment is planned for up to 500 patients to evaluate progression-free survival as the primary endpoint.Clinical trial registration: www.clinicaltrials.gov identifier is NCT05696626.
Collapse
Affiliation(s)
| | - Seth A Wander
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Thomas Bachelot
- Department of Medical Oncology, Centre Leon Berard, Lyon, France
| | - Alexandre de Nonneville
- Department of Medical Oncology, Institut Paoli Calmettes, Aix Marseille Université, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm U1068, CNRS U7258, Marseille, France
| | - Einav Nili Gal-Yam
- Oncology Department, Breast Oncology Institute Sheba Medical Center, Ramat Gan, Israel
| | - Sarah L Sammons
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Sherry Shen
- Department of Medicine, Memorial Sloan Kettering Cancer Center/Evelyn H. Lauder Breast and Imaging Center, New York, NY, USA
| | - Chris Twelves
- Department of Medical Oncology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | | | | | - Senthil Damodaran
- Department of Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
244
|
Liu Y, Cui H, Sun C. The supramolecular polymer-related signature predicts prognosis and indicates immune microenvironment infiltration in gastric cancer. Clinics (Sao Paulo) 2025; 80:100641. [PMID: 40228435 DOI: 10.1016/j.clinsp.2025.100641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/28/2025] [Accepted: 03/21/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Gastric Cancer (GC) remains a leading global cause of cancer mortality, underscoring the urgent need for advanced prognostic tools. This study aimed to construct and evaluate a prognostic risk signature based on Supramolecular Polymer-Related Genes (SPRGs) in gastric cancer. METHODS The authors downloaded data from TCGA-STAD, GEO, and CCLE databases for patients with GC and validation cohorts. Through consensus clustering, Cox proportional hazards models, LASSO Cox regression, and nomogram development, the authors identified and constructed a GC Prognostic risk Index (SPI). Additionally, the authors conducted drug sensitivity analysis and immune landscape assessment. Functional evaluations were conducted through colony formation, transwell invasion, and wound healing assays. RESULTS The authors identified that 182 SPRGs were significantly upregulated and 226 were downregulated in gastric cancer. Consensus clustering revealed two molecular subtypes, with cluster 1 having significantly lower overall survival compared to cluster 2. SPI effectively distinguished high-risk and low-risk patients across all cohorts. Furthermore, SPI was associated with tumor stage, lymph node metastasis, and tumor size, and could predict drug sensitivity in GC patients. Immune landscape analysis showed higher infiltration of naïve B cells, M2 macrophages, and activated NK cells in high-SPI patients. A nomogram model for GC prognosis using SPI and patient age was developed. KLC1 knockdown significantly suppressed GC cell proliferation, while markedly attenuating metastatic potential and invasion capacity. CONCLUSION This study constructed a prognostic risk signature based on SPRGs in gastric cancer, which is closely related to clinical pathological features, drug sensitivity, and immune landscape, providing new insights for personalized treatment.
Collapse
Affiliation(s)
- Yan Liu
- Department of Gastroenterology, Ningbo Haishu People's Hospital, Ningbo, PR China.
| | - Hongyao Cui
- Department of Gastroenterology, Ningbo Haishu People's Hospital, Ningbo, PR China
| | - Chuan Sun
- Department of Gastroenterology, Ningbo Haishu People's Hospital, Ningbo, PR China
| |
Collapse
|
245
|
Deng H, Wu D, He Y, Yu X, Liu J, Zhang Y, Leng B, Yuan X, Xiao L. E2F1-driven EXOSC10 transcription promotes hepatocellular carcinoma growth and stemness: a potential therapeutic target. Hereditas 2025; 162:60. [PMID: 40221814 PMCID: PMC11992873 DOI: 10.1186/s41065-025-00430-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND E2F Transcription Factor 1 (E2F1) is a transcription factor that plays a crucial role in the growth of many cancers, including hepatocellular carcinoma (HCC). Herein, this study probed the functions and underlying mechanisms of E2F1 in HCC tumorigenesis. METHODS The expression profiles of E2F1 and Exosome Component 10 (EXOSC10) were detected using qRT-PCR and western blotting. Functional experiments were carried out using 5-ethynyl-2'-deoxyuridine (EdU), flow cytometry, tube formation, and sphere formation assays in vitro, as well as xenograft experiments in vivo, respectively. Stemness-related proteins were assayed using western blotting. The interaction between E2F1 and EXOSC10 was verified using bioinformatics analysis and dual-luciferase reporter assay. RESULTS E2F1 was highly expressed in HCC tissues and cells, and was associated with advanced TNM stage, distant metastasis, and short survival rate. Functionally, knockdown of E2F1 suppressed HCC cell proliferation, angiogenesis, and stemness, and induced cell apoptosis. Mechanistically, E2F1 directly bound to the promoter region of EXOSC10 to up-regulate its expression. EXOSC10 silencing impaired HCC cell proliferation, angiogenesis, and stemness. Moreover, the anticancer effects of E2F1 knockdown were reversed by EXOSC10 elevation. In vivo assay, E2F1 deficiency suppressed HCC tumor growth and eliminated cancer stemness, while these effects were abolished by EXOSC10 up-regulation. CONCLUSION E2F1 promotes EXOSC10 transcription and then facilitates HCC growth and cancer stemness, revealing a potential target for HCC therapy.
Collapse
Affiliation(s)
- Haoyue Deng
- Department of Pathology, Suining Central Hospital, Suining, 629000, Sichuan, China
| | - Dingyong Wu
- Department of Oncology, Songshan General Hospital, Chongqing, 401120, China
| | - Yongpeng He
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Inaffiliationidualized Treatment, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, 400030, China
| | - Xiaolei Yu
- Department of Oncology, Fengning Manchu Autonomous County Hospital, No.737 Binhe Road, Chengde, 067000, Hebei, China
| | - Jifei Liu
- Department of Oncology, Fengning Manchu Autonomous County Hospital, No.737 Binhe Road, Chengde, 067000, Hebei, China
| | - Yanrui Zhang
- Department of Oncology, Fengning Manchu Autonomous County Hospital, No.737 Binhe Road, Chengde, 067000, Hebei, China
| | - Bing Leng
- Department of Oncology, Fengning Manchu Autonomous County Hospital, No.737 Binhe Road, Chengde, 067000, Hebei, China
| | - Xiaofeng Yuan
- Department of Oncology, Fengning Manchu Autonomous County Hospital, No.737 Binhe Road, Chengde, 067000, Hebei, China.
| | - Liguo Xiao
- Department of Oncology, Fengning Manchu Autonomous County Hospital, No.737 Binhe Road, Chengde, 067000, Hebei, China.
| |
Collapse
|
246
|
Aladelokun O, Benitez K, Wang Y, Jain A, Berardi D, Maroun G, Shen X, Roper J, Gibson J, Sumigray K, Khan SA, Johnson CH. Sex-specific effects of exogenous asparagine on colorectal tumor growth, 17β-estradiol levels, and aromatase. Pharmacol Res 2025; 215:107736. [PMID: 40228761 DOI: 10.1016/j.phrs.2025.107736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
Sex-related differences in asparagine metabolism are associated with cancer prognosis. However, the effect of exogenous asparagine on colorectal cancer (CRC) growth in men and women remains unclear. This study aims to understand the relationship between exogenous asparagine supplementation and 17β-estradiol levels and to elucidate mechanisms underlying sex-dependent signaling during CRC development. We administered asparagine intraperitoneally to tumor-bearing male and female immunodeficient Rag2/Il2RG (R2G2) mice. Asparagine supplementation caused a significant increase in tumor asparagine levels in both the tumor-bearing male and female R2G2 mice but increased serum estradiol levels and suppressed tumor growth in female R2G2 mice only. Additionally, we combined transcriptome, metabolome, and immunochemical analyses, and found that intraperitoneal asparagine treatment induced sex-dependent intra-tumoral metabolic changes to asparagine, aspartate, glutamine and glutamate levels. We observed that in females, exogenous asparagine exerts a negative feed-back effect on de novo asparagine synthesis and is associated with the activation of a sub-population of macrophages that may secrete 17β-estradiol via an aromatase or cytochrome P450 family 19 (CYP19)-dependent mechanism. Conversely, in male mice, asparagine treatment augments tumor growth, and is related to decreased numbers of macrophages, and a reduction in CYP19-mediated 17β-estradiol secretion . Overall, our results reveal a novel and sex-specific role for exogenous asparagine during cancer progression and underscores the importance of understanding mechanisms that control asparagine biosynthesis.
Collapse
Affiliation(s)
- Oladimeji Aladelokun
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Katherine Benitez
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA; Program in Translational Biomedicine, Yale University School of Medicine, New Haven, CT, USA
| | - Yuying Wang
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Abhishek Jain
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Domenica Berardi
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Georgio Maroun
- Department of Molecular Biochemistry and Biophysics, Yale University, New Haven, CT, USA
| | - Xinyi Shen
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Jatin Roper
- Division of Gastroenterology, Department of Medicine, Duke University School of Medicine, NC, USA
| | - Joanna Gibson
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Kaelyn Sumigray
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Sajid A Khan
- Division of Surgical Oncology, Department of Surgery, Yale School of Medicine, New Haven, CT, USA.
| | - Caroline H Johnson
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA.
| |
Collapse
|
247
|
Chen C, Wen L, Chen G, Yang F, Chen Z, Ji J, Gu J. Pan-cancer analysis of ITGB3 as a potential prognostic and immunological biomarker. Discov Oncol 2025; 16:522. [PMID: 40220261 PMCID: PMC11993531 DOI: 10.1007/s12672-025-02300-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
Integrin β3 (ITGB3) acts as a crucial regulator and target within the tumor immune microenvironment (TIME) and is highly expressed in the TIME of various tumors. The TIMER, TCGA, GTEx, and CCLE databases were utilized to comprehensively analyze the differential expression of ITGB3 in tumor tissues. Kaplan-Meier analysis, forest plots, and univariate and multivariate Cox regression were used to assess the genetic alterations, clinicopathological characteristics, and prognostic value of ITGB3. Additionally, the R software package was used to evaluate the relationship between ITGB3 expression and immune cell infiltration, immunomodulatory genes, and immune checkpoints, and potential signaling pathways were examined through differential expression and enrichment analysis. We found that the high expression of ITGB3 is a significant risk factor for six types of cancer, including adrenocortical carcinoma (ACC), and is closely associated with a lower survival rate. Anti-tumor immune cells (CD8 + T cells, CD4 + Th1 cells, and NKT cells) were significantly reduced. By contrast, pro-tumor immune cells (Tregs and CD4 + Th2 cells), immune checkpoints (CTLA4 and PD-CD1), and negatively regulated co-stimulators of T-cell activation (CTLA4, PD-CD1, and IL10) were significantly elevated in most types of cancer with high ITGB3 expression. Overall, our preliminary results indicate that ITGB3 plays an important role in immunosuppression in the tumor microenvironment. Elevated levels of ITGB3 inhibit tumor immunity, facilitate tumor immune escape, and affect patient prognosis, and it may be a prognostic biomarker.
Collapse
Affiliation(s)
- Changshun Chen
- Department of Orthopedics and Trauma Surgery, Affiliated Hospital of Yunnan University, Kunming, 650032, China
| | - Lei Wen
- Department of Orthopedics and Trauma Surgery, Affiliated Hospital of Yunnan University, Kunming, 650032, China
| | - Ge Chen
- Department of Orthopedics and Trauma Surgery, Affiliated Hospital of Yunnan University, Kunming, 650032, China
| | - Fei Yang
- Department of Orthopedics, Nanchong Central Hospital, Nanchong, 637000, China
| | - Zhong Chen
- Department of Orthopedics and Trauma Surgery, Affiliated Hospital of Yunnan University, Kunming, 650032, China
| | - Jianhua Ji
- Department of Orthopedics and Trauma Surgery, Affiliated Hospital of Yunnan University, Kunming, 650032, China.
| | - Jinyi Gu
- Clinical Laboratory of Affiliated Hospital of Yunnan University, Kunming, 650032, China.
| |
Collapse
|
248
|
Li P, Pang KL, Chen SJ, Yang D, Nai AT, He GC, Fang Z, Yang Q, Cai MB, He JY. ADORA2B promotes proliferation and migration in head and neck squamous cell carcinoma and is associated with immune infiltration. BMC Cancer 2025; 25:673. [PMID: 40221657 PMCID: PMC11992813 DOI: 10.1186/s12885-025-14102-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Adenosine A2B receptor (ADORA2B), a G protein-coupled receptor, is implicated in tumor progression and immune regulation in various cancers. However, its specific role in head and neck squamous cell carcinoma (HNSC) remains largely unexplored. This study aims to elucidate the expression profile, prognostic value, immune modulatory role, and therapeutic potential of ADORA2B in HNSC. METHODS Comprehensive bioinformatics analyses were performed using TCGA and GEO datasets to evaluate ADORA2B expression, clinical correlations, and prognostic significance in HNSC. Weighted gene co-expression network analysis (WGCNA) and functional enrichment analyses were conducted to explore ADORA2B-associated pathways. Immune infiltration was assessed via ESTIMATE and single-sample gene set enrichment analysis (ssGSEA). Immune checkpoint blockade (ICB) therapy sensitivity and drug sensitivity were analyzed using the IMvigor210 and NCI-60 databases, respectively. In vitro experiments, including siRNA-mediated ADORA2B knockdown, CCK-8 assays, colony formation, and wound healing assays, were performed to validate the oncogenic role of ADORA2B. RESULTS ADORA2B was significantly overexpressed in HNSC tumor tissues compared to adjacent normal tissues, and its expression correlated with advanced clinical stage as well as poor overall survival (OS) and progression-free survival (PFS). Functional enrichment analyses revealed significant downregulation of immune-related pathways in high ADORA2B expression groups. High ADORA2B expression was associated with a more immunosuppressive tumor microenvironment (TME), characterized by lower immune and stromal scores and reduced immune cell infiltration. Immunotherapy response analysis demonstrated that patients with high ADORA2B expression exhibited poorer outcomes following ICB therapy. Drug sensitivity analysis identified several agents, including Ixazomib citrate, Masitinib, and others, as potential therapeutic candidates for high ADORA2B expression patients. In vitro experiments confirmed that ADORA2B knockdown significantly inhibited HNSC cell proliferation, colony formation, and migration, underscoring its critical role in tumor progression. CONCLUSION ADORA2B is a key oncogenic driver in HNSC, contributing to tumor proliferation, migration, and an immunosuppressive TME. Its high expression is associated with poor prognosis and reduced immunotherapy efficacy. Targeting ADORA2B may enhance therapeutic outcomes and overcome treatment resistance, highlighting its potential as a diagnostic, prognostic, and therapeutic biomarker.
Collapse
Affiliation(s)
- Pian Li
- Department of Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ke-Ling Pang
- Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Shuang-Jing Chen
- The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Dong Yang
- Department of Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ai-Tao Nai
- Department of Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Gui-Cheng He
- Department of Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhe Fang
- Department of Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Qiao Yang
- Department of Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Man-Bo Cai
- Department of Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| | - Jun-Yan He
- Department of Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
249
|
Cruz FB, Maschio A, Alves ASBZ, Santos WS, Neves LP, Perini AP. Numerical dosimetry of stereotactic radiosurgery treatments in pediatric patients. Appl Radiat Isot 2025; 221:111840. [PMID: 40239357 DOI: 10.1016/j.apradiso.2025.111840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 03/04/2025] [Accepted: 04/11/2025] [Indexed: 04/18/2025]
Abstract
Brain and nervous system neoplasms account for 25 % of childhood cancer cases. In these instances, radiotherapy treatments increase survival rates, but the high radiosensitivity of pediatric patients raises concerns about the potential adverse effects of radiation. Thus, dose delivery precision becomes crucial in treatment planning, and stereotactic radiosurgeries, such as those performed with Gamma Knife equipment, are the leading-edge techniques in precision radiotherapy. This study aims to evaluate conversion coefficients for equivalent doses (CC[HT]) for out-of-field organs using pediatric mesh-type reference computational phantoms aged 1-, 5 -, 10-, and 15-years during stereotactic radiosurgery through computational simulations with the MCNP 6.3.0 code. Higher CC[HT] values were found for organs closer to the target organ of the treatment, such as the eyes, salivary glands, and thyroid, which received an average of 33.6 %, 6.4 %, and 2.6 % of the treatment dose, respectively. As the age of the pediatric phantom increased, CC[HT] values in organs farther from the target region decreased due to the increase in body length. We also determined the conversion coefficients for effective doses CC[E] and observed higher CC[E] values for the computational phantom with the lowest BMI. Thus, computational simulations showed to be meaningful tool for estimating out-of-field CC[HT] values in pediatric patients and CC[E] values, given the challenges of performing organ-level dosimetry.
Collapse
Affiliation(s)
- Felipe B Cruz
- Postgraduate Program in Biomedical Engineering (PPGEB), Faculty of Electrical Engineering, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - Alessa Maschio
- Postgraduate Program in Biomedical Engineering (PPGEB), Faculty of Electrical Engineering, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - Arthur S B Z Alves
- Postgraduate Program in Biomedical Engineering (PPGEB), Faculty of Electrical Engineering, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - William S Santos
- Postgraduate Program in Biomedical Engineering (PPGEB), Faculty of Electrical Engineering, Federal University of Uberlândia, Uberlândia, MG, Brazil; Department of Physics, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | - Lucio P Neves
- Postgraduate Program in Biomedical Engineering (PPGEB), Faculty of Electrical Engineering, Federal University of Uberlândia, Uberlândia, MG, Brazil; Institute of Physics, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - Ana P Perini
- Postgraduate Program in Biomedical Engineering (PPGEB), Faculty of Electrical Engineering, Federal University of Uberlândia, Uberlândia, MG, Brazil; Institute of Physics, Federal University of Uberlândia, Uberlândia, MG, Brazil.
| |
Collapse
|
250
|
Zhou S, Zhu S, Li Z, He Z, Xu W, Pan X, Wu W, Chen L. Segmentectomy versus lobectomy for intersegmental small-sized non-small cell lung cancer. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:110051. [PMID: 40239272 DOI: 10.1016/j.ejso.2025.110051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/29/2025] [Accepted: 04/10/2025] [Indexed: 04/18/2025]
Abstract
OBJECTIVES Intersegmental non-small cell lung cancer (NSCLC) represents a type of lesion involving at least two segments. This study aimed to demonstrate the characteristics of intersegmental NSCLC and compare the prognosis of segmentectomy with lobectomy for intersegmental NSCLC. METHODS A retrospective study was conducted on patients with small-sized NSCLC who underwent segmentectomy or lobectomy between January 2012 and December 2020. Intersegmental and non-intersegmental nodules were determined by three-dimensional computed tomographic bronchography and angiography. The log-rank test and Cox hazard proportional regression were adopted for prognostic analyses. RESULTS Totally, 1702 patients with 425 intersegmental nodules (25 %) and 1277 non-intersegmental nodules (75 %) were enrolled. Among eligible patients, 1086 patients accepted segmentectomy (intersegmental n = 188, non-intersegmental n = 898) and 616 patients underwent lobectomy (intersegmental n = 237, non-intersegmental n = 379). Intersegmental nodules were associated with larger tumor size and more lymph node metastasis (5.4 % vs. 2.1 %, P = 0.001), while intersegmental NSCLC achieved a similar prognosis to non-intersegmental NSCLC. Segmentectomy for intersegmental nodules required longer operative times, resection of more subsegments (4.06 vs. 2.91, P < 0.001), and larger resected lung volumes compared to that for non-intersegmental nodules. For patients with intersegmental nodules, the lobectomy group had larger tumor sizes, more solid nodules, and more lymph node metastasis than the segmentectomy group. After propensity score-matching, segmentectomy (n = 75) yielded comparable 5-year overall survival and disease-free survival with the lobectomy (n = 75). CONCLUSION Intersegmental nodules demonstrate a prognosis similar to non-intersegmental nodules, despite showing a higher rate of lymph node metastasis. Segmentectomy achieved long-term outcomes comparable to lobectomy for selected intersegmental NSCLC.
Collapse
Affiliation(s)
- Shengzhe Zhou
- Department of Thoracic Surgery, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shusheng Zhu
- Department of Thoracic Surgery, Taizhou Hospital of Traditional Chinese Medicine, Taizhou, China
| | - Zhihua Li
- Department of Thoracic Surgery, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhicheng He
- Department of Thoracic Surgery, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wenzheng Xu
- Department of Thoracic Surgery, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xianglong Pan
- Department of Thoracic Surgery, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Weibing Wu
- Department of Thoracic Surgery, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Liang Chen
- Department of Thoracic Surgery, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China.
| |
Collapse
|