351
|
Kubota Y, Kawano M, Iwasaki T, Itonaga I, Kaku N, Ozaki T, Tanaka K. Current management of neurotrophic receptor tyrosine kinase fusion-positive sarcoma: an updated review. Jpn J Clin Oncol 2025; 55:313-326. [PMID: 39895082 PMCID: PMC11973637 DOI: 10.1093/jjco/hyaf015] [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: 10/31/2024] [Accepted: 01/15/2025] [Indexed: 02/04/2025] Open
Abstract
In recent years, pembrolizumab has demonstrated significant efficacy in treating tumors characterized by a high tumor mutational burden and high microsatellite instability. Tropomyosin receptor kinase (TRK) inhibitors have shown considerable efficacy against tumors harboring neurotrophic receptor tyrosine kinase (NTRK) fusion genes, highlighting the growing importance of personalized medicine in cancer treatment. Advanced sequencing technologies enable the rapid analysis of numerous genetic abnormalities in tumors, facilitating the identification of patients with positive biomarkers. These advances have increased the likelihood of providing effective, tailored treatments. NTRK fusion genes are present in various cancer types, including sarcomas, and the TRK inhibitors larotrectinib and entrectinib have been effectively used for these malignancies. Consequently, the treatment outcomes for NTRK fusion-positive tumors have improved significantly, reflecting a shift toward more personalized therapeutic approaches. This review focuses on NTRK fusion-positive sarcomas and comprehensively evaluates their epidemiology, clinical features, and radiological and histological characteristics. We also investigated the treatment landscape, including the latest methodologies involving TRK inhibitors, and discussed the long-term efficacy of these inhibitors, and their optimal order of use. Notably, larotrectinib has demonstrated a high response rate in infantile fibrosarcoma, and its efficacy has been confirmed even in advanced cases. However, further research is warranted to optimize treatment duration and subsequent management strategies. The accumulation of clinical cases worldwide will play a pivotal role in refining the treatment approaches for tumors associated with NTRK fusion genes.
Collapse
Affiliation(s)
- Yuta Kubota
- Department of Orthopaedic Surgery, Faculty of Medicine, Oita University, 1-1 Idaigaoka Hasama, Yufu City, Oita 879-5593, Japan
| | - Masanori Kawano
- Department of Orthopaedic Surgery, Faculty of Medicine, Oita University, 1-1 Idaigaoka Hasama, Yufu City, Oita 879-5593, Japan
| | - Tatsuya Iwasaki
- Department of Orthopaedic Surgery, Faculty of Medicine, Oita University, 1-1 Idaigaoka Hasama, Yufu City, Oita 879-5593, Japan
| | - Ichiro Itonaga
- Department of Orthopaedic Surgery, Faculty of Medicine, Oita University, 1-1 Idaigaoka Hasama, Yufu City, Oita 879-5593, Japan
| | - Nobuhiro Kaku
- Department of Orthopaedic Surgery, Faculty of Medicine, Oita University, 1-1 Idaigaoka Hasama, Yufu City, Oita 879-5593, Japan
| | - Toshifumi Ozaki
- Department of Orthopaedic Surgery, Science of Functional Recovery and Reconstruction, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Kazuhiro Tanaka
- Department of Orthopaedic Surgery, Faculty of Medicine, Oita University, 1-1 Idaigaoka Hasama, Yufu City, Oita 879-5593, Japan
- Department of Advanced Medical Sciences, Faculty of Medicine, Oita University, 1-1 Idaigaoka Hasama, Yufu City, Oita 879-5593, Japan
| |
Collapse
|
352
|
Yin Q, Zhang Y, Xie X, Hou M, Chen X, Ding J. Navigating the future of gastric cancer treatment: a review on the impact of antibody-drug conjugates. Cell Death Discov 2025; 11:144. [PMID: 40188055 PMCID: PMC11972320 DOI: 10.1038/s41420-025-02429-5] [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: 11/05/2024] [Revised: 03/07/2025] [Accepted: 03/21/2025] [Indexed: 04/07/2025] Open
Abstract
Gastric cancer, marked by its high incidence and poor prognosis, demands the urgent development of novel and effective treatment strategies, especially for patients ineligible for surgery or those who have had limited success with chemotherapy, radiotherapy and targeted therapies. Recently, antibody-drug conjugates (ADCs) have become a key area of investigation due to their high specificity and potent antitumor effects. These therapies combine monoclonal antibodies, designed to bind to tumor-specific antigens, with cytotoxic agents that selectively target and destroy malignant cells. ADCs have generated significant interest in clinical trials as a promising approach to improve both treatment efficacy and patient outcomes in gastric cancer. However, their clinical application is not without challenges and limitations that must be addressed. This review discusses the recent progress in the use of ADCs for gastric cancer treatment.
Collapse
Affiliation(s)
- Qingling Yin
- GuiZhou University Medical College, Guiyang, 550025, Guizhou, China
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Yanlong Zhang
- GuiZhou University Medical College, Guiyang, 550025, Guizhou, China
| | - Xueqing Xie
- GuiZhou University Medical College, Guiyang, 550025, Guizhou, China
| | - Meijun Hou
- Graduate School, Zunyi Medical University, Zunyi, Guizhou, 563006, China
| | - Xunsheng Chen
- Department of Gastrointestinal Surgery, Guizhou Provincial People's Hospital, Guiyang, 550002, Guiyang, China
| | - Jie Ding
- Department of Gastrointestinal Surgery, Guizhou Provincial People's Hospital, Guiyang, 550002, Guiyang, China.
| |
Collapse
|
353
|
He P, Li H, Yang Z, Zhang R, Ye Q, Deng T, Li W, He S, Dong G, Yu Z, Li Y. Discovery and preclinical evaluations of drug candidate DA-0157 capable of overcoming EGFR drug-resistant mutation C797S and EGFR/ALK co-mutations. Eur J Med Chem 2025; 287:117323. [PMID: 39892095 DOI: 10.1016/j.ejmech.2025.117323] [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: 11/21/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/03/2025]
Abstract
Activating mutations in the epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) are significant oncogenic drivers in non-small cell lung cancer (NSCLC) patients. Despite several approved EGFR and ALK inhibitors, drug-resistant mutations pose a major challenge. Especially, there is currently no approved EGFR inhibitors targeting the C797S mutation, a refractory mutation resistant to the third-generation EGFR inhibitors. Furthermore, an increasing number of patients with EGFR/ALK co-mutations have been identified in clinical practice, yet there are no effective therapeutic options available for them. In this study, we report the discovery and preclinical evaluations of a new small-molecule drug candidate, DA-0157, which is capable of overcoming EGFR drug-resistant mutation C797S and EGFR/ALK co-mutations. DA-0157 demonstrated excellent in vitro efficacy, significantly inhibiting various EGFRC797S mutants resistant to the third-generation EGFR inhibitors, ALK rearrangements, and EGFR/ALK co-mutations. In vivo studies revealed that DA-0157 substantially inhibited tumor growth in the LD1-0025-200717 EGFRDel19/T790M/C797S PDX model (40 mg/kg/d, TGI: 98.3 %), Ba/F3-EML-4-ALK-L1196 M CDX model (40 mg/kg/d, TGI: 125.2 %), and NCI-H1975 EGFRDel19/T790M/C797S & NCI-H3122 (EML4-ALK) dual-side implantation CDX model (40 mg/kg/d, TGI: 89.5 % & 113.9 %). DA-0157 demonstrates favorable pharmacokinetic properties and safety. Currently, DA-0157 (DAJH-1050766) is undergoing Phase I/II clinical trials.
Collapse
Affiliation(s)
- Peng He
- Chengdu DIAO Pharmaceutical Group Co., Ltd., Chengdu, 610041, China
| | - Haiyan Li
- Chengdu DIAO Pharmaceutical Group Co., Ltd., Chengdu, 610041, China
| | - Zhenyu Yang
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rui Zhang
- Chengdu DIAO Pharmaceutical Group Co., Ltd., Chengdu, 610041, China
| | - Qijun Ye
- Chengdu DIAO Pharmaceutical Group Co., Ltd., Chengdu, 610041, China
| | - Ta Deng
- Chengdu DIAO Pharmaceutical Group Co., Ltd., Chengdu, 610041, China
| | - Wenwen Li
- Chengdu DIAO Pharmaceutical Group Co., Ltd., Chengdu, 610041, China
| | - Shucheng He
- Chengdu DIAO Pharmaceutical Group Co., Ltd., Chengdu, 610041, China
| | - Guangxin Dong
- Chengdu DIAO Pharmaceutical Group Co., Ltd., Chengdu, 610041, China.
| | - Zhou Yu
- Chengdu DIAO Pharmaceutical Group Co., Ltd., Chengdu, 610041, China.
| | - Yi Li
- Chengdu DIAO Pharmaceutical Group Co., Ltd., Chengdu, 610041, China.
| |
Collapse
|
354
|
Farajzadeh M, Fathi M, Jalali P, Mahmoudsalehi Kheshti A, Khodayari S, Hojjat-Farsangi M, Jadidi F. Long noncoding RNAs in acute myeloid leukemia: biomarkers, prognostic indicators, and treatment potential. Cancer Cell Int 2025; 25:131. [PMID: 40188050 PMCID: PMC11972515 DOI: 10.1186/s12935-025-03763-5] [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: 01/20/2025] [Accepted: 03/20/2025] [Indexed: 04/07/2025] Open
Abstract
Long noncoding RNAs (lncRNAs) have been recognized as significant modulators of gene expression and are essential for various biological functions, even though they don't appear to have the ability to encode proteins. Originally considered dark matter, lncRNAs have been recognized as being dysregulated and contributing to the onset, progression, and resistance to treatment of acute myeloid leukemia (AML). AML is a prevalent type of leukemia characterized by the disruption of myeloid cell differentiation, leading to an increased number of immature myeloid progenitor cells. Currently, the need for novel biomarkers and treatment targets to enhance therapeutic alternatives has led to a focus on lncRNAs as possible indicators for prognostic, therapeutic, and diagnostic systems in various human cancers, including AML. Recent research has recognized a limited set of lncRNAs as possible prognostic biomarkers or diagnoses in AML. This review evaluates the key research that highlights the significance of lncRNAs in AML and discusses their roles and impacts on the disease. Furthermore, we intend to underscore the importance of lncRNAs as new and trustworthy markers for the diagnosis, prediction, drug resistance, and targets for treatment in AML.
Collapse
Affiliation(s)
- Maryam Farajzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehrdad Fathi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pooya Jalali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences,, Tehran, Iran
| | | | - Shahla Khodayari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Farhad Jadidi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
355
|
Stoop TF, Javed AA, Oba A, Koerkamp BG, Seufferlein T, Wilmink JW, Besselink MG. Pancreatic cancer. Lancet 2025; 405:1182-1202. [PMID: 40187844 DOI: 10.1016/s0140-6736(25)00261-2] [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: 10/02/2024] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 04/07/2025]
Abstract
Pancreatic cancer is frequently a lethal disease with an aggressive tumour biology often presenting with non-specific symptoms. Median survival is approximately 4 months with a 5-year survival of 13%. Surveillance is recommended in individuals with familial pancreatic cancer, specific mutations, and high-risk intraductal papillary mucinous neoplasm, as they are at high risk of developing pancreatic cancer. Chemotherapy combined with surgical resection remains the cornerstone of treatment. However, only a small subset of patients are candidates for surgery. Multi-agent chemotherapy has improved survival in the palliative setting for patients with metastatic disease, as (neo)adjuvant and induction therapy have in patients with borderline resectable and locally advanced pancreatic. Given that pancreatic cancer is predicted to become the second leading cause of cancer-related death by 2030, novel therapies are urgently needed.
Collapse
Affiliation(s)
- Thomas F Stoop
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Ammar A Javed
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands; Division of Surgical Oncology, Department of Surgery, New York University Medical Center, New York, NY, USA
| | - Atsushi Oba
- Department of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan; Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Division of Surgical Oncology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Thomas Seufferlein
- Department of International Medicine I, Ulm University Hospital, Ulm, Germany
| | - Johanna W Wilmink
- Department of Medical Oncology, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Marc G Besselink
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands.
| |
Collapse
|
356
|
Lu D, Huang L, Weng C. Unveiling the Novel Anti - Tumor Potential of Digitonin, a Steroidal Saponin, in Gastric Cancer: A Network Pharmacology and Experimental Validation Study. Drug Des Devel Ther 2025; 19:2653-2666. [PMID: 40206492 PMCID: PMC11980797 DOI: 10.2147/dddt.s504671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/15/2025] [Indexed: 04/11/2025] Open
Abstract
Background Gastric cancer (GC) remains a leading cause of cancer-related mortality, with limited effective treatment options for advanced stages. As a steroidal saponin with documented anti-neoplastic properties in multiple cancers, digitonin's mode of action in GC pathogenesis has yet to be fully elucidated. This research focused on exploring the potential of Digitonin in GC treatment using a combination of network pharmacology and experimental validation. Methods The inhibitory effects of Digitonin on the proliferation, invasion, and migration of gastric cancer cells were evaluated using CCK-8, colony formation, wound healing, and transwell assays. Key targets of Digitonin were identified through network pharmacology. Molecular docking and various experiments, including Western blot, immunofluorescence, and a subcutaneous xenograft model, were used for validation. Results Digitonin exhibited stronger cytotoxicity against GC cells and significantly inhibited GC cell proliferation, migration, and invasion. Network pharmacology analysis revealed that the core targets of Digitonin are involved in key cancer-related signaling pathways, including HIF-1α, Ras, and PI3K-Akt pathways, with HSP90AA1 and NFKB1 identified as central targets. Further molecular docking, Western blotting, and immunofluorescence experiments confirmed that Digitonin significantly suppressed the expression of HSP90AA1 and inhibited the nuclear translocation of NFKB1, inducing cell apoptosis. Additionally, a subcutaneous xenograft model of GC further validated that Digitonin effectively inhibited tumor growth. Conclusion Digitonin serves as a promising multi-target therapeutic agent for GC. This study underscores the potential of combining network pharmacology with traditional Chinese medicine to identify novel therapeutic targets and develop effective anti-cancer strategies. In addition, these findings suggest that digitonin could be a promising candidate for future clinical trials in GC treatment.
Collapse
MESH Headings
- Humans
- Stomach Neoplasms/drug therapy
- Stomach Neoplasms/pathology
- Stomach Neoplasms/metabolism
- Cell Proliferation/drug effects
- Saponins/pharmacology
- Saponins/chemistry
- Network Pharmacology
- Animals
- Drug Screening Assays, Antitumor
- Mice
- Dose-Response Relationship, Drug
- Cell Movement/drug effects
- Molecular Docking Simulation
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Mice, Inbred BALB C
- Mice, Nude
- Tumor Cells, Cultured
- Structure-Activity Relationship
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/metabolism
- Molecular Structure
- Cell Line, Tumor
- Antineoplastic Agents, Phytogenic/pharmacology
- Antineoplastic Agents, Phytogenic/chemistry
- Apoptosis/drug effects
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Dongdong Lu
- Department of Gastroenterology, Ningbo No. 2 Hospital, Ningbo, Zhejiang Province, 315000, People’s Republic of China
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, 310000, People’s Republic of China
| | - Leijie Huang
- Department of Gastroenterology, Ningbo No. 2 Hospital, Ningbo, Zhejiang Province, 315000, People’s Republic of China
| | - Chunyan Weng
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, 310000, People’s Republic of China
| |
Collapse
|
357
|
Chu T, Lim Y, Sun Y, Wirtz D, Wu PH. Accelerated Discovery of Cell Migration Regulators Using Label-Free Deep Learning-Based Automated Tracking. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.01.646705. [PMID: 40236190 PMCID: PMC11996530 DOI: 10.1101/2025.04.01.646705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Cell migration plays a key role in normal developmental programs and in disease, including immune responses, tissue repair, and metastasis. Unlike other cell functions, such as proliferation which can be studied using high-throughput assays, cell migration requires more sophisticated instruments and analysis, which decreases throughput and has led to more limited mechanistic advances in our understanding of cell migration. Current assays either preclude single-cell level analysis, require tedious manual tracking, or use fluorescently labeled cells, which greatly limit the number of extracellular conditions and molecular manipulations that can be studied in a reasonable amount of time. Using the migration of cancer cells as a testbed, we established a workflow that images large numbers of cells in real time, using a 96-well plate format. We developed and validated a machine-vision and deep-learning analysis method, DeepBIT, to automatically detect and track the migration of individual cells from time-lapsed videos without cell labeling and user bias. We demonstrate that our assay can examine cancer cell motility behavior in many conditions, using different small-molecule inhibitors of known and potential regulators of migration, different extracellular conditions such as different contents in extracellular matrix and growth factors, and different CRISPR-mediated knockouts. About 1500 cells per well were tracked in 840 different conditions, for a total of ~1.3M tracked cells, in 70h (5 min per condition). Manual tracking of these cells by a trained user would take ~5.5 years. This demonstration reveals previously unidentified molecular regulators of cancer cell migration and suggests that collagen content can change the sign of how cytoskeletal molecules can regulate cell migration.
Collapse
|
358
|
Xin X, Tang Y, Lu M, Huang J, Shang J, Yang L, Dai L, Yin J, Li J, Leng Q, Tang H, Zhong X. Prognostic value of diffusion-weighted imaging to cytoreductive surgery with or without hyperthermic intraperitoneal chemotherapy for patients with gastric cancer and peritoneal metastases. BMC Cancer 2025; 25:616. [PMID: 40188022 PMCID: PMC11972487 DOI: 10.1186/s12885-025-14008-z] [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: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND To investigate the prognostic value of the apparent diffusion coefficient (ADC) calculated from diffusion-weighted imaging (DWI) to cytoreductive surgery (CRS), with or without hyperthermic intraperitoneal chemotherapy (HIPEC), for gastric cancer (GC) patients with peritoneal metastasis (PM). METHODS Between May 2016 and December 2020, 95 newly diagnosed GC patients with PM who underwent CRS combined with HIPEC (CRS + HIPEC group, n = 61) and CRS alone (CRS group, n = 34) were retrospectively included. All patients underwent abdominal 3.0 T MRI scan, including DWI, and the mean ADC (ADCmean), minimum ADC (ADCmin), and maximum ADC (ADCmax) values of the whole-volume tumor were measured. The prognostic value of the ADC parameters and clinical and histopathological characteristics were investigated by univariate and multivariate Cox analyses. RESULTS The median overall survival (OS) periods of the CRS + HIPEC and CRS groups were 18 and 9 months, respectively ([hazard ratio (HR) = 0.44 [95% CI: 0.27-0.71], P<0.001). The ADCmean and ADCmin values were positively correlated with OS in all patients (Spearman's rho [R] = 0.361 and 0.470), as well as in the CRS + HIPEC (R = 0.369 and 0.417) and CRS (R = 0.192 and 0.409) groups. The multivariate Cox analysis demonstrated that the ADCmean ≤ 1.39 × 10- 3 mm2/s and ADCmin ≤ 0.77 × 10- 3 mm2/s were significantly associated with a negative prognosis in the total population (HR = 1.68 [95% CI: 1.02-2.75] and 2.48 [95% CI: 1.51-4.08], P all < 0.05) and the CRS + HIPEC group (HR = 2.22 [95% CI: 1.19-4.14] and 2.37 [95% CI: 1.26-4.37], P all < 0.05), along with pathologic T and N stages. Only the ADCmin ≤ 0.77 × 10- 3 mm2/s was identified as an independent prognostic factor in the CRS group (HR = 3.49 [95% CI: 1.19-10.20], P = 0.022). CONCLUSIONS The minimum ADC was identified as a strong independent prognostic factor for GC patients with PM who underwent CRS, with or without HIPEC.
Collapse
Affiliation(s)
- Xin Xin
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yongfang Tang
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Man Lu
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jie Huang
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jian Shang
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lidan Yang
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lihuan Dai
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jinxue Yin
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiansheng Li
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qibin Leng
- Department of Oncology Institute, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Hongsheng Tang
- Department of Abdominal Surgery, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Xi Zhong
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
359
|
Tandyk A, Christ T. Analysis of pembrolizumab in microsatellite instability high (MSI-H) and deficient mismatch repair (dMMR) non-colorectal patients with metastatic cancer. J Oncol Pharm Pract 2025:10781552251331958. [PMID: 40183381 DOI: 10.1177/10781552251331958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Pembrolizumab was approved for the treatment of MSI-H/dMMR solid malignancies in a tumor agnostic approval in 2017. Treatment records for patients harboring MSI-H/dMMR status at our institution treated with pembrolizumab were reviewed for outcomes of duration of treatment, progression free survival, and overall survival. Adverse events and immunotherapy biomarkers of PDL-1 status and tumor mutation burden (TMB) were also assessed. Thirty patients were included: 13 with endometrial cancer, 5 non-colorectal gastrointestinal cancer, 2 ovarian or uterine cancer, 2 breast cancer, and 8 with other malignancies. The median duration of treatment was 11 months (range 0-35). Progression free survival and overall survival were not reached. Five patients (16.7%) experienced immune related adverse events, resulting in discontinuation of pembrolizumab in 3. PDL-1 expression and TMB did not appear to correspond to treatment outcomes. This analysis provides a real-world dataset of MSI-H/dMMR solid tumor patients being treated with pembrolizumab, outcomes of which are consistent with registry trials.
Collapse
Affiliation(s)
- Alexis Tandyk
- Rush MD Anderson Cancer Center Pharmacy, Chicago, USA
- UCHealth University of Colorado Hospital, Department of Pharmacy, Aurora, USA
| | - Trevor Christ
- Rush MD Anderson Cancer Center Pharmacy, Chicago, USA
| |
Collapse
|
360
|
Braganca Xavier C, Guardia GDA, Alves JPB, Lopes CDH, Awni BM, Campos EF, Jardim DL, Galante PAF. Identifying predictors of overall survival among patients with TMB-low metastatic cancer treated with immune checkpoint inhibitors. Oncologist 2025; 30:oyaf078. [PMID: 40285678 PMCID: PMC12032576 DOI: 10.1093/oncolo/oyaf078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have significantly advanced cancer therapy, yet their efficacy in tumors with low tumor mutational burden (TMB) remains suboptimal. In this study, we aimed to elucidate the impact of somatic mutations on overall survival (OS) in TMB-low patients treated with ICIs and to explore the potential for personalized treatment selection through machine learning. METHODS We conducted a comprehensive analysis of 1172 TMB-low (TMB < 10 mutations per megabase) patients with cancer receiving ICIs, examining the association between specific gene mutations and OS. Additionally, we developed a decision tree model (DTM) to predict OS based on clinical features and tumor mutational profiles. RESULTS Our findings reveal that mutations in DAXX, HLA-A, H3C2, IGF1R, CTNNB1, SMARCA4, KMT2D, and TP53 are significantly associated with poorer survival outcomes in the multivariate analysis. Remarkably, for renal cell carcinoma (RCC) patients, VHL mutations predicted improved OS following ICI even when adjusted for age, sex, and microsatellite instability (MSI) status in both multivariate analysis and the DTM model. CONCLUSIONS These results reinforce the prevailing notion that TMB alone does not predict ICI response, highlighting the critical role of individual gene mutations in TMB-low tumors under ICI therapy. Furthermore, our study demonstrates the promise of machine learning models in optimizing ICI treatment decisions, paving the way for more precise and effective therapeutic strategies in this patient population.
Collapse
Affiliation(s)
- Camila Braganca Xavier
- MD Anderson Cancer Center, Houston, TX, 77030, United States
- Hospital Sírio-Libanês, São Paulo, SP, 01308-050, Brazil
| | | | | | | | - Beatriz M Awni
- Hospital Sírio-Libanês, São Paulo, SP, 01308-050, Brazil
| | | | - Denis L Jardim
- Hospital Sírio-Libanês, São Paulo, SP, 01308-050, Brazil
- Oncoclínicas&CO - Medica Scientia Innovation Research (MedSir), São Paulo, SP, 04538-132, Brazil
| | | |
Collapse
|
361
|
Liang M, Lu J, Wang X, Song P, Ai S, Cai D, Sun F, Lu X, Wang M, Fu S, Yu H, Guan W, Shen X. Expression Patterns of Immune Checkpoint Molecules and Their Clinical Values in Gastric Neuroendocrine Neoplasms. Clin Transl Gastroenterol 2025:01720094-990000000-00386. [PMID: 40183457 DOI: 10.14309/ctg.0000000000000842] [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: 10/10/2024] [Accepted: 03/27/2025] [Indexed: 04/05/2025] Open
Abstract
INTRODUCTION Gastric neuroendocrine neoplasms (g-NENs) are a rare type of stomach tumor. However, limited data exist about the expression and clinical significance of B7 family ligands/receptors in patients with g-NENs. Thus, we conducted this study to address this issue in a cohort of 112 patients with g-NENs. METHODS Using immunohistochemistry, we mapped and quantified the expression of the B7 family ligands/receptors in 112 g-NEN samples: programmed cell death ligand 1 and 2 (PD-L1 and PD-L2), B7-H3, B7-H4, recombinant human galectin-9 (LGALS9), and CD155. Associations between the marker levels, clinicopathological variables, and survival were evaluated. RESULTS The percentages of high expression of PD-L1, PD-L2, B7-H3, B7-H4, LGALS9, and CD155 in the cohort of 112 g-NEN cases were 37.5%, 55.4%, 46.4%, 37.5%, 46.4%, and 51.8%, respectively. Elevated expression of PD-L1, PD-L2, B7-H3, B7-H4, LGALS9, and CD155 was significantly associated with several clinicopathological characteristics. K-M analysis indicated that high expression levels of CD155, B7-H3, PD-L2, and LGALS9 were correlated with poor overall survival (OS) ( P < 0.0001, P = 0.0002, P = 0.0319 and P = 0.0120, respectively). Multivariate Cox regression analysis indicated that high CD155 expression, vasculature invasion, and worse World Health Organization pathological grade were independent prognostic factors for OS ( P = 0.007, P = 0.030, and P = 0.019, respectively). DISCUSSION We detected variable expression of the PD-L1, PD-L2, B7-H3, B7-H4, LGALS9, and CD155 proteins in g-NENs. These results suggest that the expression level of CD155 may be a vital indicator of OS in patients with g-NENs. B7 family ligands/receptors could be potential immunotherapeutic targets for g-NENs.
Collapse
Affiliation(s)
- Mengjie Liang
- Department of General Surgery, Division of Gastric Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University , Nanjing, China
| | - Junren Lu
- Department of General Surgery, Division of Gastric Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University , Nanjing, China
| | - Xingzhou Wang
- Department of General Surgery, Division of Gastric Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University , Nanjing, China
| | - Peng Song
- Department of General Surgery, Division of Gastric Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University , Nanjing, China
| | - Shichao Ai
- Department of General Surgery, Division of Gastric Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University , Nanjing, China
| | - Daming Cai
- Department of General Surgery, Division of Gastric Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University , Nanjing, China
| | - Feng Sun
- Department of General Surgery, Division of Gastric Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University , Nanjing, China
| | - Xiaofeng Lu
- Department of General Surgery, Division of Gastric Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University , Nanjing, China
| | - Meng Wang
- Department of General Surgery, Division of Gastric Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University , Nanjing, China
| | - Shuang Fu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Heng Yu
- Department of General Surgery, Division of Gastric Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University , Nanjing, China
| | - Wenxian Guan
- Department of General Surgery, Division of Gastric Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University , Nanjing, China
| | - Xiaofei Shen
- Department of General Surgery, Division of Gastric Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University , Nanjing, China
- Department of General Surgery, Division of Gastric Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
362
|
Ma Y, Li Z, Wei C, Zhang J, Fu Q, Zhang Z, Shang C, Wang J, Wan X, Zhang B, Zhang Y, Li J, Zhang H, Bie L, Xia Q, Luo S, Li N. Neoadjuvant camrelizumab plus trastuzumab and chemotherapy for HER2-positive gastric or gastroesophageal junction adenocarcinoma: a single-arm, phase 2 trial. Gastric Cancer 2025:10.1007/s10120-025-01606-w. [PMID: 40184004 DOI: 10.1007/s10120-025-01606-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 03/10/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND The impact of neoadjuvant combined chemotherapy, immunotherapy, and targeted therapy on pathologic responses and survival outcomes in HER2-positive locally advanced gastric cancer remains unclear. PATIENTS AND METHODS In this single-arm, phase 2 trial, patients with HER2-positive resectable cT4 and/or N + M0 gastric or gastroesophageal junction (G/GEJ) adenocarcinoma received four cycles of neoadjuvant camrelizumab plus trastuzumab and CapOx, followed by D2 gastrectomy and four cycles of CapOx. The primary endpoint was pathological complete response (pCR, ypT0N0) rate. RESULTS Twenty-five patients were enrolled and received neoadjuvant combination treatment. Of these patients, 11 (44%) were in cT3 and 14 (56%) in cT4a; all had positive nodal status. Of the 23 patients who underwent surgery, 5 (21.7%, 95% CI: 7.5-43.7) achieved pCR (ypT0N0), and 7 (30.4%, 95% CI: 13.2-52.9) achieved near pCR (ypT0). The R0 resection rate was 100%. During a median follow-up of 41.0 months, no patients with pCR had recurrence or death. In contrast, five of 18 patients with non-pCR had recurrence, and four of them died. The three-year disease-free survival rate was 78.3%. During neoadjuvant treatment, grade 3 adverse events were observed in 36% of patients, with no grade 4 or 5 adverse events reported. No treatment-related surgical delay or reoperation occurred. CONCLUSION Neoadjuvant camrelizumab plus trastuzumab and chemotherapy demonstrated favorable response and tolerable safety in HER2-positive G/GEJ adenocarcinoma.
Collapse
Affiliation(s)
- Yijie Ma
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450000, China
| | - Zhi Li
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450000, China
| | - Chen Wei
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450000, China
| | - Jian Zhang
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450000, China
| | - Qiang Fu
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450000, China
| | - Zhandong Zhang
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450000, China
| | - Chuang Shang
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450000, China
| | - Jinbang Wang
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450000, China
| | - Xiangbin Wan
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450000, China
| | - Bin Zhang
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450000, China
| | - Yongchao Zhang
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450000, China
| | - Jing Li
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450000, China
| | - He Zhang
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450000, China
| | - Liangyu Bie
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450000, China
| | - Qingxin Xia
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450000, China
| | - Suxia Luo
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450000, China
| | - Ning Li
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450000, China.
| |
Collapse
|
363
|
Ferraro DA, Bisig B, Rotzinger DC, Pareja F, Missiaglia E, Voutsadakis I, Homicsko K, Digklia A. Case Report: Lasting complete response to pembrolizumab in mismatch repair-deficient cardiac sarcoma: a genomic characterization. Front Oncol 2025; 15:1485386. [PMID: 40248199 PMCID: PMC12003144 DOI: 10.3389/fonc.2025.1485386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 03/10/2025] [Indexed: 04/19/2025] Open
Abstract
Sarcomas are traditionally considered "cold" tumors with poor response to immunotherapy. However, evidence accumulating over the last years shows that immune checkpoint inhibitors (ICIs) may have a role in selected sarcoma patients according to predictive markers. Here, we report the case of a woman diagnosed with a primary cardiac undifferentiated sarcoma. Following failure of standard first line chemotherapy, high-throughput sequencing (HTS) revealed a high tumor mutational burden (TMB), pathogenic mutations in FAT1 and NOTCH2 and a microsatellite instability (MSI)-associated signature. Immunohistochemistry confirmed mismatch repair-deficiency (MMRd) and abundant CD8+ tumor-infiltrating lymphocytes (TILs), in the absence of tertiary lymphoid structures. The patient was, therefore, treated with the ICI pembrolizumab, reaching a complete response that continues to persist at last follow-up, more than seven years from initial diagnosis and nearly six years from initiation of ICI treatment. This case illustrates the importance of performing HTS in rare sarcomas given the availability of efficient therapies, such as those for tumors displaying high TMB or MMRd/MSI. In agreement with other reports, it supports the contention that MMRd/MSI status and high numbers of TILs are valuable predictive markers of response to immunotherapy in sarcomas.
Collapse
Affiliation(s)
- Daniela A. Ferraro
- Department of Medical Oncology, CHUV University Hospital, Lausanne, Switzerland
| | - Bettina Bisig
- Institute of Pathology, Department of Laboratory Medicine and Pathology, CHUV University Hospital, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - David C. Rotzinger
- Department of Radiology, CHUV University Hospital, Lausanne, Switzerland
| | - Fresia Pareja
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Edoardo Missiaglia
- Institute of Pathology, Department of Laboratory Medicine and Pathology, CHUV University Hospital, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Ioannis Voutsadakis
- Algoma District Cancer Program, Sault Area Hospital, Sault Ste. Marie, ON, Canada
- Division of Clinical Sciences, Section of Internal Medicine, Northern Ontario School of Medicine, Sudbury, ON, Canada
| | - Krisztian Homicsko
- Department of Medical Oncology, CHUV University Hospital, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Antonia Digklia
- Department of Medical Oncology, CHUV University Hospital, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
364
|
Chen SY, He PL, Lu LY, Lin MC, Chan SH, Tsai JS, Luo WT, Wang LH, Li HJ. ST6GAL1-Mediated Sialylation of PECAM-1 Promotes a Transcellular Diapedesis-Like Process That Directs Lung Tropism of Metastatic Breast Cancer. Cancer Res 2025; 85:1199-1218. [PMID: 39786386 DOI: 10.1158/0008-5472.can-24-1550] [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: 05/10/2024] [Revised: 11/01/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025]
Abstract
Metastasis is the leading cause of mortality in breast cancer, with lung metastasis being particularly detrimental. Identification of the processes determining metastatic organotropism could enable the development of approaches to prevent and treat breast cancer metastasis. In this study, we found that lung-tropic and non-lung-tropic breast cancer cells differ in their response to sialic acids, affecting the sialylation of surface proteins. Lung-tropic cells showed higher levels of ST6GAL1, whereas non-lung-tropic cells had more ST3GAL1. ST6GAL1-mediated α-2,6-sialylation, unlike ST3GAL1-mediated α-2,3-sialylation, increased lung metastasis by promoting cancer cell migration through pulmonary endothelial layers and reducing junction protein levels. α-2,6-Sialylated platelet/endothelial cell adhesion molecule 1 (PECAM-1) on breast cancer cells facilitated extravasation through the pulmonary endothelium, a critical step in lung metastasis. Knockdown of ST6GAL1 or PECAM-1 significantly reduced lung metastasis. The human pulmonary endothelium displayed high PECAM-1 levels. Through transhomophilic interaction with pulmonary PECAM-1, α-2,6-sialylated PECAM-1 on ST6GAL1-positive cancer cells increased pulmonary extravasation in a diapedesis-like, cell-autonomous manner. Additionally, lung-tropic cells and their exosomes increased the permeability of pulmonary endothelial cells, promoting metastasis in a non-cell-autonomous manner. Analysis of human breast cancer samples showed a correlation between elevated ST6GAL1/PECAM-1 expression and lung metastasis. These results suggest that targeting ST6GAL1-mediated α-2,6-sialylation could be a potential therapeutic strategy to prevent lung metastasis in patients with breast cancer. Significance: ST6GAL1-mediated α-2,6-sialylation of PECAM-1 dictates lung-tropic metastasis of breast cancer, revealing that the pattern of sialylation of breast cancer cells is a determinant of metastatic organ tropism and a potential therapeutic target.
Collapse
Affiliation(s)
- Shih-Yin Chen
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Pei-Lin He
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Li-Yu Lu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Meng-Chieh Lin
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Shih-Hsuan Chan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Chinese Medicine Research Center, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Jia-Shiuan Tsai
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Wen-Ting Luo
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Lu-Hai Wang
- Chinese Medicine Research Center, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Hua-Jung Li
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
- Program in Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung City, Taiwan
| |
Collapse
|
365
|
Liu W, Li H, Botos I, Kumkhaek C, Zhu J, Rodgers GP. Olfactomedin 4 promotes gastric cancer cell G2/M progression and serves as a therapeutic target in gastric adenocarcinoma. Carcinogenesis 2025; 46:bgaf010. [PMID: 40056162 PMCID: PMC12013284 DOI: 10.1093/carcin/bgaf010] [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: 09/04/2024] [Revised: 01/24/2025] [Accepted: 03/04/2025] [Indexed: 03/10/2025] Open
Abstract
Olfactomedin 4 (OLFM4) is a member of the olfactomedin domain-containing olfactomedin glycoprotein family and plays important roles in innate immunity, inflammation, and cancer. It exhibits increased expression in gastric cancer patient tissues and has been shown to regulate proliferation and apoptosis in gastric cancer cells. However, the molecular mechanism(s) underlying OLFM4's role in gastric cancer remain unknown. In this study, we found that OLFM4 knockdown significantly inhibited YCC3 gastric cancer cell proliferation and induced G2/M cell cycle arrest. Yeast two-hybridization screening revealed that OLFM4 directly interacts with cyclin B1 interacting protein 1 (CCNB1IP1), an E3 ubiquitin protein ligase. In YCC3 cells, OLFM4 co-immunoprecipitated and colocalized with CCNB1IP1 and underwent cell cycle phase-specific nucleo-cytoplasmic shuttling. OLFM4 knockdown decreased both cyclin B1 protein levels and CDK1 activity in YCC3 cells. Screening of a cohort of OLFM4-targeted microRNAs (miRNAs) for their impact on cell proliferation identified several that significantly downregulated OLFM4 protein levels and inhibited YCC3 cell proliferation in vitro. Rescue experiments demonstrated that these miRNAs' inhibitory effect on cell proliferation was partially related to their downregulation of OLFM4. When three of these miRNAs were individually administered intratumorally to nude mice bearing YCC3 cell xenografts, tumor growth was significantly inhibited when compared with tumors treated with a negative control miRNA. These results suggest that OLFM4 promotes cell cycle progression and cell proliferation in gastric cancer cells and may have utility as a therapeutic target in gastric adenocarcinoma.
Collapse
Affiliation(s)
- Wenli Liu
- Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bldg. 10, Room 9N119, 9000 Rockville Pike, Bethesda, MD 20892, United States
| | - Hongzhen Li
- Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bldg. 10, Room 9N119, 9000 Rockville Pike, Bethesda, MD 20892, United States
| | - Istvan Botos
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bldg. 10, Room 9N119, 9000 Rockville Pike, Bethesda, MD 20892, United States
| | - Chutima Kumkhaek
- Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bldg. 10, Room 9N119, 9000 Rockville Pike, Bethesda, MD 20892, United States
| | - Jianqiong Zhu
- Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bldg. 10, Room 9N119, 9000 Rockville Pike, Bethesda, MD 20892, United States
| | - Griffin P Rodgers
- Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bldg. 10, Room 9N119, 9000 Rockville Pike, Bethesda, MD 20892, United States
| |
Collapse
|
366
|
Ambrosini M, Manca P, Nasca V, Sciortino C, Ghelardi F, Seligmann JF, Taieb J, Pietrantonio F. Epidemiology, pathogenesis, biology and evolving management of MSI-H/dMMR cancers. Nat Rev Clin Oncol 2025:10.1038/s41571-025-01015-z. [PMID: 40181086 DOI: 10.1038/s41571-025-01015-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2025] [Indexed: 04/05/2025]
Abstract
Deficiency in DNA mismatch repair (dMMR) is a common pathway of carcinogenesis across different tumour types and confers a characteristic microsatellite instability-high (MSI-H) molecular phenotype. The prevalence of the MSI-H/dMMR phenotype is highest in endometrial and colorectal cancers, and this phenotype is associated with a distinct tumour biology, prognosis and responsiveness to various anticancer treatments. In a minority of patients, MSI-H/dMMR cancers result from an inherited pathogenic variant in the context of Lynch syndrome, which has important implications for familial genetic screening. Whether these hereditary cancers have a different biology and clinical behaviour to their sporadic counterparts remains uncertain. Interest in this tumour molecular subtype has increased following the discovery of the high sensitivity of metastatic MSI-H/dMMR cancers to immune-checkpoint inhibitors (ICIs) in a histology-agnostic manner, which reflects the genomic hypermutation resulting from dMMR that renders these tumours highly immunogenic and immune infiltrated. This vulnerability is now also being exploited in early stage disease settings. Despite this common biological foundation, different MSI-H/dMMR cancers have histotype-specific features that correspond to their particular cell or tissue of origin, which might be associated with differences in prognosis and sensitivity to ICIs. In this Review, we provide an overview of the epidemiology, biology, pathogenesis, clinical diagnosis and treatment of MSI-H/dMMR tumours as a histology-agnostic cancer phenomenon. We also highlight peculiarities associated with specific pathogenetic alterations and histologies of MSI-H/dMMR tumours.
Collapse
Affiliation(s)
- Margherita Ambrosini
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Gastroenterology and Digestive Oncology, European Georges Pompidou Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Paolo Manca
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vincenzo Nasca
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Carolina Sciortino
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo Ghelardi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Jenny F Seligmann
- Division of Oncology, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Julien Taieb
- Department of Gastroenterology and Digestive Oncology, European Georges Pompidou Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- Paris-Cité University, SIRIC CARPEM Comprehensive Cancer Center, Paris, France
| | - Filippo Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| |
Collapse
|
367
|
Zhang H, Sun N, Li F, Wang Q, Sun Z, Zhang Y, Wang L, Zhao C, Fu Y. Construction of a modified TNM staging system and prediction model based on examined lymph node counts for gastric cancer patients at pathological stage N3. Front Oncol 2025; 15:1569736. [PMID: 40248208 PMCID: PMC12003143 DOI: 10.3389/fonc.2025.1569736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 03/13/2025] [Indexed: 04/19/2025] Open
Abstract
Background Examined lymph node (ELN) count is a critical factor affecting the number of metastatic lymph nodes (MLNs). The impact of the ELN number on survival and staging remains unclear. Methods This study included 4,291 stage N3 GC patients from the SEER database (training cohort) and 567 stage N3 GC patients from the FAHZZU database (validation cohort). The optimal ELN count and stage migration were investigated, and a modified TNM (mTNM) staging system including the ELN count was proposed. LASSO regression and random forest analyses were used to screen and evaluate the variables associated with survival, and an mTNM-based nomogram was constructed. The performance of the mTNM staging system and mTNM-based nomogram were compared with that of the 8th edition of the TNM staging system. Results The optimal threshold of the ELN count was identified as 21. An insufficient number of ELNs (≤ 21) was associated with poorer survival outcomes and led to stage migration in all N3 patients. A new mTNM staging system was proposed, integrating the ELN count into the TNM staging system (8th edition). LASSO regression analysis revealed that age, tumor size, adjuvant chemotherapy, adjuvant radiotherapy, and the mTNM system were associated with overall survival (OS) outcomes, and random forest analysis revealed that the mTNM system was the most important variable for predicting survival. An mTNM-based nomogram was constructed to predict 1-, 3-, and 5-year OS rates. Compared with the TNM staging system (8th edition), the mTNM staging system and mTNM-based nomogram showed superior prognosis discriminative ability, better predictive accuracy, and greater net improvement in survival outcomes. Conclusions The optimal ELN count for N3 GC patients was 21. The mTNM staging system and mTNM-based nomogram showed superior discriminative ability, predictive accuracy, and greater net benefit for OS outcomes.
Collapse
Affiliation(s)
- Hongyu Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Nan Sun
- Department of Plastic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Feng Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qiyang Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhao Sun
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yawei Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lei Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunlin Zhao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Fu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
368
|
Lin S, Sun B, Zhu Y, Huang Y, Qin Y, Yao N, Liu Y, Chen G. Natural product Pulsatilla saponin D sensitizes BRCA-proficient ovarian cancers to PARP inhibitors through inhibiting homologous recombination repair. J Pharm Pharmacol 2025; 77:511-523. [PMID: 40036611 DOI: 10.1093/jpp/rgaf006] [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: 04/21/2024] [Accepted: 01/27/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND As a strategy in the development of effective cancer therapeutics, synthetic lethality has been used in clinical practice. Poly adenosine diphosphate (ADP)-ribose polymerase inhibitors are the first approved drug utilized synthetic lethality and achieved promising therapeutic efficacy in cancer cells with BRCA1/2 mutation. Nonetheless, most cancer patients with wild-type BRCA1/2 gene are not qualified for PARPi therapy. To induce BRCAness phenotype in cancer cells with normal BRCA1/2 status, we identified Pulsatilla Saponin D (SB365), which efficiently inhibited recruitment of BRCA1 at DNA double-strand breaks, leading to homologous recombination repair deficiency. METHODS We utilized the HR repair reporter system. The reporter cells were treated with a natural compounds library to identify the agent that significantly decreased HR activity. Then, we detected the expression of HR related proteins using immunofluorescence and western blot. Colony formation and CCK8 was used to detect the inhibitory effect of Pulsatilla Saponin D on cell proliferation. Apoptosis was measured using Annexin V/PI staining. Comet assay kits were used to carry out the comet assay. Ovarian cancer xenograft model, immunohistochemical staining and Hematoxylin-Eosin staining was used to detect the antitumor efficacy and toxicity of Pulsatilla Saponin D. KEY FINDINGS Pulsatilla Saponin D greatly increased PARPi-induced DNA DSBs, growth inhibition and apoptosis in ovarian cancer cells. Combined administration of PARPi and Pulsatilla Saponin D induced synergistic anti-tumor effects in ovarian cancer cells and xenograft mouse model without obvious toxicity. CONCLUSIONS In summary, our study found Pulsatilla Saponin D is a novel HR repair inhibitor and would optimize clinical application of PARP inhibitors on cancer patients with WT BRCA1/2.
Collapse
Affiliation(s)
- Shengbin Lin
- School of Biopharmacy, China Pharmaceutical University, Nanjing, P.R. China
- Department of Clinical Laboratory, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Binghe Sun
- School of Biopharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Yin Zhu
- Department of Oncology, Jurong Hospital Affiliated to Jiangsu University, Zhenjiang, P.R. China
| | - Yi Huang
- School of Biopharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Yu Qin
- School of Biopharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Nan Yao
- School of Biopharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Yongzhu Liu
- Department of Gynecology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangdong, P.R. China
| | - Guo Chen
- School of Biopharmacy, China Pharmaceutical University, Nanjing, P.R. China
| |
Collapse
|
369
|
Pérol M, Li W, Pennell NA, Liu G, Ohe Y, De Braud F, Nagasaka M, Felip E, Xiong A, Zhang Y, Fan H, Wang X, Li S, Lai RK, Ran F, Zhang X, Chen W, Bazhenova L, Zhou C. Taletrectinib in ROS1+ Non-Small Cell Lung Cancer: TRUST. J Clin Oncol 2025:JCO2500275. [PMID: 40179330 DOI: 10.1200/jco-25-00275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 02/14/2025] [Accepted: 02/26/2025] [Indexed: 04/05/2025] Open
Abstract
PURPOSE Taletrectinib is an oral, potent, CNS-active, selective, next-generation ROS1 tyrosine kinase inhibitor (TKI). We report integrated efficacy and safety from registrational taletrectinib studies in ROS1+ non-small cell lung cancer. METHODS TRUST-I and TRUST-II were phase II, single-arm, open-label, nonrandomized, multicenter trials. Efficacy outcomes were pooled from TRUST-I and TRUST-II pivotal cohorts. The safety population comprised all patients treated with once-daily oral taletrectinib 600 mg pooled across the taletrectinib clinical program. The primary end point was independent review committee-assessed confirmed objective response rate (cORR). Secondary outcomes included intracranial (IC)-ORR, progression-free survival (PFS), duration of response (DOR), and safety. RESULTS As of June 7, 2024, the efficacy-evaluable population included 273 patients in TRUST-I and TRUST-II. Among TKI-naïve patients (n = 160), the cORR was 88.8% and the IC-cORR was 76.5%; in TKI-pretreated patients (n = 113), the cORR was 55.8% and the IC-cORR was 65.6%. In TKI-naïve patients, the median DOR and median PFS were 44.2 and 45.6 months, respectively. In TKI-pretreated patients, the median DOR and median PFS were 16.6 and 9.7 months. The cORR in patients with G2032R mutation was 61.5% (8 of 13). Among 352 patients treated with taletrectinib 600 mg once daily, the most frequent treatment-emergent adverse events (TEAEs) were GI events (88%) and elevated AST (72%) and ALT (68%); most were grade 1. Neurologic TEAEs were infrequent (dizziness, 21%; dysgeusia, 15%) and mostly grade 1. TEAEs leading to discontinuations (6.5%) were low. CONCLUSION Taletrectinib showed a high response rate with durable responses, robust IC activity, prolonged PFS, favorable safety, and low rates of neurologic adverse events in TKI-naïve and pretreated patients.
Collapse
Affiliation(s)
- Maurice Pérol
- Department of Medical Oncology, Léon Bérard Cancer Center, Lyon, France
| | - Wei Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | | | - Geoffrey Liu
- Princess Margaret Cancer Centre, Temerty School of Medicine, University of Toronto, Toronto, Canada
| | | | | | - Misako Nagasaka
- University of California Irvine School of Medicine and Chao Family Comprehensive Cancer Center, Orange, CA
| | | | - Anwen Xiong
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Yongchang Zhang
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Huijie Fan
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xicheng Wang
- The First Affiliated Hospital/School of Clinical Medicine Guangdong Pharmaceutical University, Guangzhou, China
| | | | | | | | | | | | | | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
- Department of Medical Oncology, East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
370
|
Thangaretnam K, Islam MO, Lv J, El-Rifai A, Perloff A, Soutto HL, Peng D, Chen Z. WEE1 inhibition in cancer therapy: Mechanisms, synergies, preclinical insights, and clinical trials. Crit Rev Oncol Hematol 2025; 211:104710. [PMID: 40187712 DOI: 10.1016/j.critrevonc.2025.104710] [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: 03/04/2025] [Accepted: 03/22/2025] [Indexed: 04/07/2025] Open
Abstract
WEE1 is a serine/threonine kinase that regulates the G2/M checkpoint by phosphorylating CDK1, preventing premature mitotic entry and maintaining genomic stability. Many cancers, particularly those with TP53 mutations, upregulate WEE1 to counteract replication stress and DNA damage, making it a key target for therapy. WEE1 inhibitors, especially adavosertib (AZD1775), have shown strong preclinical and clinical activity in ovarian, breast, gastrointestinal, and head and neck cancers. By inducing mitotic catastrophe and increasing DNA damage, WEE1 inhibition enhances the effectiveness of chemotherapies, including platinum-based agents, antimetabolites, and PARP inhibitors. It also synergizes with radiotherapy and immune checkpoint inhibitors, improving responses in tumors with immune evasion. However, challenges such as acquired resistance, toxicity, and patient selection remain obstacles to clinical implementation. Given the expanding role of WEE1 inhibitors in cancer treatment, a comprehensive review is needed to summarize their biological functions, structural regulation, and therapeutic applications. This review highlights key findings from preclinical and clinical studies, explores emerging biomarkers for patient stratification, and discusses strategies to overcome resistance and toxicity. By integrating current knowledge, we aim to provide insights into optimizing WEE1-targeted therapies and guiding future research to maximize their clinical impact in cancer treatment.
Collapse
Affiliation(s)
- Krishnapriya Thangaretnam
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Md Obaidul Islam
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Jialun Lv
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | | | - Ava Perloff
- College of Arts and Sciences, University of Miami, FL 33136, USA
| | - Houda L Soutto
- College of Arts and Sciences, University of Miami, FL 33136, USA
| | - Dunfa Peng
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Zheng Chen
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA.
| |
Collapse
|
371
|
Bénard F, Marcil S, Mack L, Deban M, Bildersheim M, Bouchard-Fortier A, Osman Y, Mercier F, Purich K, Haase E, Schiller D, Soucisse M, Sidéris L, Leblanc G, Dubé P, Boulanger-Gobeil C, Hamilton T, Gervais MK. Survival outcomes of cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in selected patients with stage IV gastric adenocarcinoma - A Canadian case series. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:110000. [PMID: 40288217 DOI: 10.1016/j.ejso.2025.110000] [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: 12/17/2024] [Revised: 02/25/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025]
Abstract
INTRODUCTION Despite advances in systemic therapy, metastatic gastric cancer is associated with a poor prognosis. As peritoneal disease is common, several studies looked at the potential benefits of hyperthermic intraperitoneal chemotherapy (HIPEC) in this context, with encouraging results. However, no Canadian data currently exists on the subject. MATERIALS AND METHODS This study aims to report characteristics and outcomes of Canadian patients who underwent cytoreductive surgery and HIPEC (CRS-HIPEC) for gastric cancer associated with peritoneal disease or positive peritoneal cytology. This multicenter retrospective study included patients 18 years or older with gastric cancer associated with isolated peritoneal involvement who underwent CRS-HIPEC in five tertiary centers from 2016 to 2022. RESULTS CRS-HIPEC was performed on 20 patients aged 34-69 years old, most of whom presented with poorly differentiated (90 %) adenocarcinoma, with synchronous peritoneal disease (95 %). Median PCI was 3 (0-13). The associated 90-day morbidity rate, defined as Clavien-Dindo grade III and above complications, was 10 %. At a mean follow-up of 23.3 months (range 4-48), 25 % of patients remained disease-free, with an estimated median overall survival of 24.2 months. CONCLUSION CRS-HIPEC for gastric cancer can achieve longer term survival in highly selected patients with low-burden peritoneal disease or positive cytology. Ongoing randomized trials will further clarify patients' selection criteria and benefits of this approach.
Collapse
Affiliation(s)
- Florence Bénard
- Division of Surgical Oncology, Department of Surgery, Centre Hospitalier de l'Université de Montréal (CHUM), 1051 Rue Sanguinet, Montréal, QC, Canada, H2X 3E4
| | - Stéphanie Marcil
- Division of Surgical Oncology, Department of Surgery, Hôpital Maisonneuve-Rosemont, 5415 Bd de l'Assomption, Montréal, QC, Canada, H1T 2M4
| | - Lloyd Mack
- Division of Surgical Oncology, Department of Surgery, Foothills Medical Center, 1403 29 St NW, Calgary, AB, Canada, T2N 2T9
| | - Melina Deban
- Division of Surgical Oncology, Department of Surgery, Foothills Medical Center, 1403 29 St NW, Calgary, AB, Canada, T2N 2T9
| | - Michael Bildersheim
- Division of Surgical Oncology, Department of Surgery, Foothills Medical Center, 1403 29 St NW, Calgary, AB, Canada, T2N 2T9
| | - Antoine Bouchard-Fortier
- Division of Surgical Oncology, Department of Surgery, Foothills Medical Center, 1403 29 St NW, Calgary, AB, Canada, T2N 2T9
| | - Yasmin Osman
- Division of Surgical Oncology, Department of Surgery, Hôpital Maisonneuve-Rosemont, 5415 Bd de l'Assomption, Montréal, QC, Canada, H1T 2M4
| | - Frédéric Mercier
- Division of Surgical Oncology, Department of Surgery, Centre Hospitalier de l'Université de Montréal (CHUM), 1051 Rue Sanguinet, Montréal, QC, Canada, H2X 3E4
| | - Kieran Purich
- Division of Surgical Oncology, Department of Surgery, Grey Nuns Community Hospital, 1100 Youville Dr W Northwest, Edmonton, AB, Canada, T6L 5X8
| | - Erika Haase
- Division of Surgical Oncology, Department of Surgery, Grey Nuns Community Hospital, 1100 Youville Dr W Northwest, Edmonton, AB, Canada, T6L 5X8
| | - Dan Schiller
- Division of Surgical Oncology, Department of Surgery, Grey Nuns Community Hospital, 1100 Youville Dr W Northwest, Edmonton, AB, Canada, T6L 5X8
| | - Mikael Soucisse
- Division of Surgical Oncology, Department of Surgery, Hôpital Maisonneuve-Rosemont, 5415 Bd de l'Assomption, Montréal, QC, Canada, H1T 2M4
| | - Lucas Sidéris
- Division of Surgical Oncology, Department of Surgery, Hôpital Maisonneuve-Rosemont, 5415 Bd de l'Assomption, Montréal, QC, Canada, H1T 2M4
| | - Guy Leblanc
- Division of Surgical Oncology, Department of Surgery, Hôpital Maisonneuve-Rosemont, 5415 Bd de l'Assomption, Montréal, QC, Canada, H1T 2M4
| | - Pierre Dubé
- Division of Surgical Oncology, Department of Surgery, Hôpital Maisonneuve-Rosemont, 5415 Bd de l'Assomption, Montréal, QC, Canada, H1T 2M4
| | - Cindy Boulanger-Gobeil
- Division of Surgical Oncology, Department of Surgery, Hôtel-Dieu de Québec, 11 Côte du Palais, Québec, Canada, G1R 2J6
| | - Trevor Hamilton
- Division of Surgical Oncology, Department of Surgery, Vancouver General Hospital, Jim Pattison Pavilion, 899 W 12th Ave, Vancouver, BC, Canada, V5Z 1M9
| | - Mai-Kim Gervais
- Division of Surgical Oncology, Department of Surgery, Hôpital Maisonneuve-Rosemont, 5415 Bd de l'Assomption, Montréal, QC, Canada, H1T 2M4.
| |
Collapse
|
372
|
Mao S, Li J, Huang J, Lv L, Zhang Q, Cheng Q, Liu X, Bi Z, Yao J. Therapeutic potential of microRNA-506 in cancer treatment: mechanisms and therapeutic implications. Front Oncol 2025; 15:1524763. [PMID: 40248198 PMCID: PMC12003368 DOI: 10.3389/fonc.2025.1524763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 03/17/2025] [Indexed: 04/19/2025] Open
Abstract
Cancer is a complex and highly lethal disease marked by unchecked cell proliferation, aggressive behavior, and a strong tendency to metastasize. Despite significant advancements in cancer diagnosis and treatment, challenges such as early detection difficulties, drug resistance, and adverse effects of radiotherapy or chemotherapy continue to threaten patient survival. MicroRNAs (miRNAs) have emerged as critical regulators in cancer biology, with miR-506 being extensively studied and recognized for its tumor-suppressive effects across multiple cancer types. This review examines the regulatory mechanisms of miR-506 in common cancers, focusing on its role in the competing endogenous RNA (ceRNA) network and its effects on cancer cell proliferation, apoptosis, and migration. We also discuss the potential of miR-506 as a therapeutic target and its role in overcoming drug resistance in cancer treatment. Overall, these insights underscore the therapeutic potential of miR-506 and its promise in developing novel cancer therapies.
Collapse
Affiliation(s)
- Shuzhen Mao
- Department of Pharmacy, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Junyan Li
- Department of Pathology, Second People’s Hospital of Ningyang, Taian, Shandong, China
| | - Jiahui Huang
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Lili Lv
- Department of Pathology, Second People’s Hospital of Ningyang, Taian, Shandong, China
| | - Qilian Zhang
- Department of Pathology, People’s Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Qing Cheng
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Xiaojing Liu
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Zhiwei Bi
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Jing Yao
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
373
|
Abate M, Stroobant E, Fei T, Lin YH, Shimada S, Drebin H, Chen E, Tang LH, Shah SP, Wolchok JD, Janjigian YY, Strong VE, Vardhana SA. Host Tissue Factors Predict Immune Surveillance and Therapeutic Outcomes in Gastric Cancer. Cancer Immunol Res 2025; 13:591-601. [PMID: 39786344 PMCID: PMC11964842 DOI: 10.1158/2326-6066.cir-23-0563] [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: 07/13/2023] [Revised: 10/25/2024] [Accepted: 01/07/2025] [Indexed: 01/12/2025]
Abstract
The immune composition of solid tumors is typically inferred from biomarkers, such as histologic and molecular classifications, somatic mutational burden, and PD-L1 expression. However, the extent to which these biomarkers predict the immune landscape in gastric adenocarcinoma-an aggressive cancer often linked to chronic inflammation-remains poorly understood. We leveraged high-dimensional spectral cytometry to generate a comprehensive single-cell immune landscape of tumors, normal tissue, and lymph nodes from patients in the Western Hemisphere with gastric adenocarcinoma. The immune composition of gastric tumors could not be predicted by traditional metrics such as tumor histology, molecular classification, mutational burden, or PD-L1 expression via IHC. Instead, our findings revealed that innate immune surveillance within tumors could be anticipated by the immune profile of the normal gastric mucosa. Additionally, distinct T-cell states in the lymph nodes were linked to the accumulation of activated and memory-like CD8+ tumor-infiltrating lymphocytes. Unbiased reclassification of patients based on tumor-specific immune infiltrate generated four distinct subtypes with varying immune compositions. Tumors with a T cell-dominant immune subtype, which spanned The Cancer Genome Atlas molecular subtypes, were exclusively associated with superior responses to immunotherapy. Parallel analysis of metastatic gastric cancer patients treated with immune checkpoint blockade showed that patients who responded to immunotherapy had a pretreatment tumor composition that corresponded to a T cell-dominant immune subtype from our analysis. Taken together, this work identifies key host-specific factors associated with intratumoral immune composition in gastric cancer and offers an immunological classification system that can effectively identify patients likely to benefit from immune-based therapies.
Collapse
Affiliation(s)
- Miseker Abate
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Surgery, New York-Presbyterian Hospital/Weill Cornell Medicine, New York, NY, USA
| | - Emily Stroobant
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Teng Fei
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ya-Hui Lin
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Shoji Shimada
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Digestive Disease Center, Showa University, Northern Yokohama Hospital, Yokohama, Japan
| | - Harrison Drebin
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Eunise Chen
- Department of Surgery, University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL, USA
| | - Laura H. Tang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sohrab P. Shah
- Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jedd D. Wolchok
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Yelena Y. Janjigian
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vivian E. Strong
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Santosha A. Vardhana
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
374
|
Yamane H, Yoshimitsu A, Akimoto E, Abe T, Yamane M. Real-World Insights: End-of-Life Care for Patients with Terminal Gastric Cancer at Home. Palliat Med Rep 2025; 6:137-143. [PMID: 40308717 PMCID: PMC12040547 DOI: 10.1089/pmr.2024.0104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2025] [Indexed: 05/02/2025] Open
Abstract
Background As the Japanese population continues to age, home health care provision has greater significance. However, the number of patients with terminal cancer receiving end-of-life care at home remains limited, and predicting the prognosis of patients with terminal-stage gastric cancer is challenging. Objective To analyze the real-world data of patients with terminal gastric cancer receiving end-of-life care at home to provide insights into their care and health outcomes. Design A two-center retrospective study. Setting/Subjects This study focused on terminal patients with gastric cancer who died at home between 2021 and 2024 in Japan. A total of 27 participants (14 males and 13 females) were included in the study, with a median age of 78 years. Measurements First, we analyzed the detailed clinical course of the patients during home care. Second, we performed a comparative analysis by dividing the patients into two groups based on median overall survival (OS). Results The median OS during home care was 22 days. The patients were divided into two groups: long OS (OS-L) and short OS (OS-S). Strong opioid use and home oxygen therapy were recorded in 22 and nine patients, respectively. In the OS-S group, oral intake was significantly reduced (25.0% vs. 66.7%, p = 0.032). There was a notable difference in serum albumin levels between the two groups (2.8 vs. 2.4 mg/dL, p = 0.038). The neutrophil-to-lymphocyte ratio (NLR)/albumin was significantly higher in the OS-S group compared to the OS-L group (1.0 vs. 2.3, p = 0.032). Conclusions Oral intake, serum albumin level, and NLR/albumin were important prognostic factors in end-of-life care at home of patients with terminal gastric cancer.
Collapse
Affiliation(s)
- Hiroaki Yamane
- Department of Surgery, Yamane Clinic, Hiroshima, Japan
- Department of Clinical Oncology, JA Hiroshima General Hospital, Hiroshima, Japan
| | | | | | - Tomoyuki Abe
- Department of Gastroenterological Surgery, National Hospital Organization Higashihiroshima Medical Center, Hiroshima, Japan
| | - Motoi Yamane
- Department of Surgery, Yamane Clinic, Hiroshima, Japan
| |
Collapse
|
375
|
Okano N, Pirozzi A, Abidoye O, Hoyek C, Eslinger C, Zheng-Lin B, Jamal F, Sahwan O, Sonbol MB, Uson Junior PLS, Hayashi M, Sato T, Nishioka M, Nagashima F, Bekaii-Saab T, Borad MJ, Hironaka S. Systemic therapy for pretreated advanced biliary tract cancer: past developments and recent advances. Jpn J Clin Oncol 2025:hyaf052. [PMID: 40173029 DOI: 10.1093/jjco/hyaf052] [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: 02/27/2025] [Accepted: 03/17/2025] [Indexed: 04/04/2025] Open
Abstract
Biliary tract cancer (BTC) remains among the most challenging malignancies with a poor prognosis and limited treatment options, particularly in pretreated patients. As most patients experience disease progression after first-line treatment, effective second-line and subsequent treatments are required. Although the addition of modified FOLFOX (fluorouracil, leucovorin, and oxaliplatin) to active symptom control improved the overall survival of patients with progressing advanced BTC despite gemcitabine plus cisplatin treatment, its efficacy was modest. Moreover, most clinical trials demonstrated modest efficacy of molecular-targeted agents for molecularly unselected pretreated advanced BTC. Patients with advanced BTC carry a relatively high druggable genetic alteration rate and have shown promising responses to molecular-matched therapies targeting gene alterations such as FGFR2 fusions/rearrangements, IDH1 mutation, and HER2 overexpression/amplification. Additionally, tumor-agnostic approaches, including BRAF V600E, NTRK fusion, and RET fusion, have expanded the treatment options for some patients. Immune checkpoint inhibitors have shown limited efficacy as mono- or combination therapy in patients with pretreated advanced BTC. Therefore, developmental efforts have shifted to immune checkpoint inhibitor and other combinations such as vascular endothelial growth factor receptor inhibitors or radiation. In addition to refining combination strategies to enhance the therapeutic potential of immune checkpoint inhibitor, future research should focus on elucidating the tumor microenvironment. This review delineates the evolution of systemic therapies in patients with pretreated advanced BTC. By examining past developments and recent advances through prospective trials, it highlights novel approaches that may improve outcomes in this challenging disease.
Collapse
Affiliation(s)
- Naohiro Okano
- Department of Medical Oncology, Kyorin University Faculty of Medicine, 6-20-2, Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan
- Division of Hematology and Oncology, Mayo Clinic, 5881 E Mayo Blvd, Phoenix, AZ 85054, United States
| | - Angelo Pirozzi
- Division of Hematology and Oncology, Mayo Clinic, 5881 E Mayo Blvd, Phoenix, AZ 85054, United States
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, Milan 20072, Italy
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan 20089, Italy
| | - Oluseyi Abidoye
- Division of Hematology and Oncology, Mayo Clinic, 5881 E Mayo Blvd, Phoenix, AZ 85054, United States
| | - Celine Hoyek
- Division of Hematology and Oncology, Mayo Clinic, 5881 E Mayo Blvd, Phoenix, AZ 85054, United States
| | - Cody Eslinger
- Division of Hematology and Oncology, Mayo Clinic, 5881 E Mayo Blvd, Phoenix, AZ 85054, United States
| | - Binbin Zheng-Lin
- Division of Hematology and Oncology, Mayo Clinic, 5881 E Mayo Blvd, Phoenix, AZ 85054, United States
| | - Fares Jamal
- Division of Hematology and Oncology, Mayo Clinic, 5881 E Mayo Blvd, Phoenix, AZ 85054, United States
| | - Oudai Sahwan
- Division of Hematology and Oncology, Mayo Clinic, 5881 E Mayo Blvd, Phoenix, AZ 85054, United States
| | - Mohamad Bassam Sonbol
- Division of Hematology and Oncology, Mayo Clinic, 5881 E Mayo Blvd, Phoenix, AZ 85054, United States
| | - Pedro Luiz Serrano Uson Junior
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, Avenida Albert Einstein 627, São Paulo 05652900, Brazil
| | - Masato Hayashi
- Department of Medical Oncology, Kyorin University Faculty of Medicine, 6-20-2, Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan
| | - Taro Sato
- Department of Medical Oncology, Kyorin University Faculty of Medicine, 6-20-2, Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan
- Department of Gastroenterology and Hepatology, Kyorin University Faculty of Medicine, 6-20-2, Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan
| | - Mariko Nishioka
- Department of Medical Oncology, Kyorin University Faculty of Medicine, 6-20-2, Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan
| | - Fumio Nagashima
- Department of Medical Oncology, Kyorin University Faculty of Medicine, 6-20-2, Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan
| | - Tanios Bekaii-Saab
- Division of Hematology and Oncology, Mayo Clinic, 5881 E Mayo Blvd, Phoenix, AZ 85054, United States
| | - Mitesh J Borad
- Division of Hematology and Oncology, Mayo Clinic, 5881 E Mayo Blvd, Phoenix, AZ 85054, United States
| | - Shuichi Hironaka
- Department of Medical Oncology, Kyorin University Faculty of Medicine, 6-20-2, Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan
| |
Collapse
|
376
|
Qiao XX, Jiang HG, Tang Y, Shi JM, Jiang LM, Yang L, Hou Q, Wang SL, Song YW, Liu YP, Fang H, Chen B, Lu NN, Qi SN, Li YX, Cao JZ, Zhou FX, Zhao DB, Li N, Jin J. Long-Term Prognostic Analysis of Chemoradiation Therapy Versus Chemotherapy after D2 Resection for High-Risk Gastric Cancer: Results From a Prospective Randomized Control Study. Int J Radiat Oncol Biol Phys 2025:S0360-3016(25)00304-9. [PMID: 40185210 DOI: 10.1016/j.ijrobp.2025.03.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/04/2025] [Accepted: 03/22/2025] [Indexed: 04/07/2025]
Abstract
PURPOSE To explore the population of patients who could benefit from adjuvant radiation therapy (RT) and investigated the value of postoperative chemoradiation therapy (CRT) in patients with gastric cancer after D2 resection. METHODS AND MATERIALS This randomized clinical trial enrolled patients between October 1, 2011, and December 31, 2019. Patients with gastric cancer who underwent D2 gastrectomy were randomized (1:1) to receive postoperative CRT or adjuvant chemotherapy after surgery. The adjuvant chemotherapy group received 8 cycles of SOX (S-1 + oxaliplatin) chemotherapy. RT was given after 4 to 6 cycles of SOX chemotherapy. RT comprised 45 Gy in 25 fractions of 1.8 Gy over 5 weeks by intensity modulated RT concurrently with S-1 chemotherapy. The primary endpoint was 3-year disease-free survival (DFS). RESULTS A total of 312 patients (median [IQR] age, 58 [50-64] years) were enrolled, including 157 patients randomized to the adjuvant chemotherapy group and 155 patients randomized to the adjuvant CRT group. The 3-year DFS was 66.7% for the control arm and 70.7% for the experimental arm (hazard ratio [HR], 0.82; 95% CI, 0.54-1.25; P = .35). We defined patients with pN stage ≥ N2 and extraperigastric lymph node metastasis as the high-risk group and the remaining patients as the low-risk group. The 3-year DFS rates for the high-risk group and the low-risk group were 59.3% and 76.8%, respectively (HR, 2.11; 95% CI, 1.39-3.22; P < .01). For high-risk patients, the 3-year DFS rates in the adjuvant chemotherapy and adjuvant CRT groups were 53.0% and 71.0%, respectively (HR, 0.53; 95% CI, 0.29-0.97; P < .05). More grade 3 and 4 acute toxic effects were observed in the adjuvant chemotherapy group than in the CRT group (41 patients [26.1%] vs 28 patients [18.5%]; P = .11), but the difference was not significant. CONCLUSIONS Subgroup analysis of this randomized clinical trial revealed that high-risk patients can benefit from adjuvant CRT.
Collapse
Affiliation(s)
- Xia-Xi Qiao
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huan-Gang Jiang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yuan Tang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jin-Ming Shi
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li-Ming Jiang
- State Key Laboratory of Molecular Oncology and Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Yang
- State Key Laboratory of Molecular Oncology and Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qing Hou
- Department of Radiotherapy, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Shu-Lian Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yong-Wen Song
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue-Ping Liu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui Fang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bo Chen
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ning-Ning Lu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shu-Nan Qi
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ye-Xiong Li
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian-Zhong Cao
- Department of Radiotherapy, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Fu-Xiang Zhou
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Dong-Bing Zhao
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Ning Li
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Radiotherapy, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China.
| | - Jing Jin
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China.
| |
Collapse
|
377
|
Tang M, Liu T, Zhang Y, Ding J. Efficacy and safety of pembrolizumab in the treatment of advanced hepatocellular carcinoma: a systematic review and meta-analysis. Eur J Clin Pharmacol 2025:10.1007/s00228-025-03829-3. [PMID: 40172662 DOI: 10.1007/s00228-025-03829-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/21/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND The efficacy and safety of pembrolizumab in treating advanced hepatocellular carcinoma (HCC) are inconsistent across studies. This study sheds light on the efficacy and safety of pembrolizumab in advanced HCC patients. METHODS Several databases were comprehensively searched up to January 13, 2025, to identify studies assessing pembrolizumab for advanced HCC. Outcome indicators included overall survival (OS), progression-free survival (PFS), objective response rate (ORR), rash, adverse events (AEs), and severe adverse events (SAEs). Pooled effects were estimated through hazard ratios (HRs) and odds ratios (ORs) with 95% confidence intervals (CIs). R 4.4.1. was employed for statistical analyses. RESULTS Twenty-two studies involving 2964 patients were encompassed. Meta-analysis indicated that pembrolizumab demonstrated an ORR of 28% in single-arm analyses. Pembrolizumab significantly improved ORR in comparison to placebo (OR = 2.57, 95% CI: 1.32-5.03) but showed no significant advantage over nivolumab. Pembrolizumab markedly enhanced PFS (HR = 0.76, 95% CI: 0.69-0.85) and OS (HR = 0.78, 95% CI: 0.70-0.88) compared to placebo, but no significant differences were observed when compared to nivolumab. Pembrolizumab significantly raised the risk of rash in comparison to placebo (OR = 2.27, 95% CI: 1.55-3.31) but showed no significant difference versus nivolumab. The pembrolizumab group showed a higher incidence of AEs (OR = 1.94, 95% CI: 1.42-2.64) and SAEs (OR = 2.10, 95% CI: 1.04-4.25) than the placebo group, with no significant difference between pembrolizumab and nivolumab. CONCLUSIONS This study proves that pembrolizumab may have promising therapeutic effects in patients with advanced HCC, although no clear advantage over nivolumab was observed. The occurrence of AEs warrants attention in clinical practice.
Collapse
Affiliation(s)
- Mingyang Tang
- Health Science Center, Hubei Minzu University, Enshi Hubei, 445000, China
| | - Tao Liu
- Hepatobiliary Pancreatic Spleen Surgery, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, No. 158 Wuyang Avenue, Wuyangba Street, Enshi Hubei, Hubei Province, 445000, China
| | - Yukun Zhang
- Abdominal Oncology Department, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Hubei, 445000, China
| | - Jun Ding
- Hepatobiliary Pancreatic Spleen Surgery, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, No. 158 Wuyang Avenue, Wuyangba Street, Enshi Hubei, Hubei Province, 445000, China.
| |
Collapse
|
378
|
Amatto PDPG, Chaves L, França SDC, Carvalho JCT, Carmona F, Pereira AMS. Efficacy of different pharmaceutical forms of Curcuma longa or curcumin in reducing oral mucositis severity and incidence in cancer patients: a systematic review and meta-analysis. Front Pharmacol 2025; 16:1560729. [PMID: 40242451 PMCID: PMC12000115 DOI: 10.3389/fphar.2025.1560729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Background Curcuma longa L. (turmeric, Zingiberaceae) has been traditionally used for its anti-inflammatory, wound-healing, and antimicrobial properties. These characteristics have made it a key component in managing inflammatory and ulcerative conditions like oral mucositis (OM). This study aimed to evaluate the effectiveness of various pharmaceutical formulations of C. longa or curcumin in reducing the OM severity, incidence and associated pain in patients undergoing chemotherapy and/or radiotherapy for cancer. Methods This systematic review and meta-analysis of randomized clinical trials was conducted according to PRISMA guidelines, registered in PROSPERO (#CRD42024504111). Searches were performed in PubMed, Embase, and Cochrane databases. Studies comparing C. longa or curcumin with placebo in cancer patients experiencing oral mucositis, reporting outcomes such as the World Health Organization Oral Mucositis Grading Scale, pain scores (visual analogue scale), or OM incidence were included. Risk ratios and weighted mean differences with 95% confidence intervals were calculated using fixed- or random-effects models. Results Six studies with 159 patients (mean age ∼50 years, 40% women) were included. C. longa extracts, curcumin, or nanocurcumin were administered in capsules, mouthwash, or gel formulations. The pooled analysis showed significant reductions in WHO scores and oral pain compared to placebo. OM incidence decreased by 6% overall, with a notable 37%-reduction observed in patients using curcumin-containing mouthwash during radiotherapy alone. Subgroup analyses revealed consistent benefits across all oncological treatments. Conclusion C. longa, curcumin, or nanocurcumin in various formulations, effectively reduce OM severity and pain while curcumin-containing mouthwash reduced OM incidence in cancer patients undergoing treatment. Systematic Review Registration identifier CRD 42024504111.
Collapse
Affiliation(s)
- Pedro de Padua G. Amatto
- Department of Biotechnology in Medicinal Plants, University of Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | - Lucas Chaves
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Suzelei de Castro França
- Department of Biotechnology in Medicinal Plants, University of Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | | | - Fabio Carmona
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Ana Maria Soares Pereira
- Department of Biotechnology in Medicinal Plants, University of Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
- Botanical Garden of Medicinal Plants Ordem e Progresso, Jardinópolis, Brazil
| |
Collapse
|
379
|
Zhou K, Liu Y, Tang C, Zhu H. Pancreatic Cancer: Pathogenesis and Clinical Studies. MedComm (Beijing) 2025; 6:e70162. [PMID: 40182139 PMCID: PMC11965705 DOI: 10.1002/mco2.70162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 03/08/2025] [Accepted: 03/12/2025] [Indexed: 04/05/2025] Open
Abstract
Pancreatic cancer (PC) is a highly lethal malignancy, with pancreatic ductal adenocarcinoma (PDAC) being the most common and aggressive subtype, characterized by late diagnosis, aggressive progression, and resistance to conventional therapies. Despite advances in understanding its pathogenesis, including the identification of common genetic mutations (e.g., KRAS, TP53, CDKN2A, SMAD4) and dysregulated signaling pathways (e.g., KRAS-MAPK, PI3K-AKT, and TGF-β pathways), effective therapeutic strategies remain limited. Current treatment modalities including chemotherapy, targeted therapy, immunotherapy, radiotherapy, and emerging therapies such as antibody-drug conjugates (ADCs), chimeric antigen receptor T (CAR-T) cells, oncolytic viruses (OVs), cancer vaccines, and bispecific antibodies (BsAbs), face significant challenges. This review comprehensively summarizes these treatment approaches, emphasizing their mechanisms, limitations, and potential solutions, to overcome these bottlenecks. By integrating recent advancements and outlining critical challenges, this review aims to provide insights into future directions and guide the development of more effective treatment strategies for PC, with a specific focus on PDAC. Our work underscores the urgency of addressing the unmet needs in PDAC therapy and highlights promising areas for innovation in this field.
Collapse
Affiliation(s)
- Kexun Zhou
- Department of Medical OncologyCancer CenterWest China HospitalSichuan UniversityChengduChina
| | - Yingping Liu
- Department of RadiotherapyCancer HospitalChinese Academy of Medical SciencesBeijingChina
| | - Chuanyun Tang
- The First Clinical Medical College of Nanchang UniversityNanchang UniversityNanchangChina
| | - Hong Zhu
- Department of Medical OncologyCancer CenterWest China HospitalSichuan UniversityChengduChina
- Division of Abdominal Tumor Multimodality TreatmentCancer CenterWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
380
|
Hu WT, Li M, Ma PJ, Yang D, Liu XD, Wang Y. A silence catalyst: CCL5-mediated intercellular communication in cancer. Arch Toxicol 2025:10.1007/s00204-025-04036-w. [PMID: 40167774 DOI: 10.1007/s00204-025-04036-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
Chemokine CCL5 (RANTES), as a key mediator of intercellular communication in cancers, and its role in cancer development, metastasis and immune escape has received increasing attention. CCL5 and its receptors are important components of the tumor microenvironment and play a tumor promoting role in different ways by triggering signaling pathways through binding to the primary receptor CCR5. CCL5 was viewed as indispensable "gate keepers" of immunity and inflammation, it remains unclear of CCL5-mediated intercellular communication. Therefore, in this review, we summarize the latest information on the origin, structure, and characterization of CCL5 and role of CCL5 in the tumor microenvironment. It includes CCL5-mediated intercellular communication through exosomes, microvesicles and others in breast, lung, and ovarian cancers. CCL5 has a multifaceted role in cancer and has potential applications as a biomarker for cancer diagnosis and prognosis, which provides theoretical bases and therapeutic targets for the development of new cancer therapeutic strategies.
Collapse
Affiliation(s)
- Wei-Ting Hu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Ming Li
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Pei-Jun Ma
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Ding Yang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Xiao-Dong Liu
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Yun Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China.
| |
Collapse
|
381
|
Nowotny R, Rhode P, Niebisch S, Chon SH, Alakus H, Thieme R, Gockel I, Plum PS. [Innovations in the Current Guideline on Gastric Carcinoma and Outlook for the Future]. Zentralbl Chir 2025; 150:163-166. [PMID: 40199374 DOI: 10.1055/a-2529-5300] [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: 04/10/2025]
Abstract
The third version of the national S3 guideline for gastric cancer is currently being finalised and will be published in the near future. Therefore, this review article primarily discusses the aspects of the current 2019 version and provides an outlook on the innovations in the upcoming version. Emphasis will be placed on the transfer of care to centres with appropriate expertise. Overall, previous revisions of the guideline have placed an increased focus on the genetic aspects of the disease and have specified corresponding risk groups for the occurrence of this neoplasia (including the presence of a CDH1 mutation or HNPCC carriers). In addition, more "borderline" cases for surgery, such as patients with oligometastasis or peritoneal carcinomatosis, have been included in the guidelines. However, for the time being, these patient groups should only be treated in trials. Finally, the guideline discusses the increasing use of immunotherapy in current and future treatment regimens, in addition to the current chemotherapeutic standard of care analogous to the FLOT regimen. New targeted therapeutic approaches such as monoclonal antibodies against claudin 18.2 will complement the treatment of gastric cancer and will certainly be included in the next version of the guideline.
Collapse
Affiliation(s)
- Robert Nowotny
- Klinik und Poliklinik für Viszeral-, Transplantations-, Thorax- und Gefäßchirurgie, Universitätsklinikum Leipzig, Leipzig, Deutschland
| | - Philipp Rhode
- Klinik und Poliklinik für Viszeral-, Transplantations-, Thorax- und Gefäßchirurgie, Universitätsklinikum Leipzig, Leipzig, Deutschland
| | - Stefan Niebisch
- Klinik und Poliklinik für Viszeral-, Transplantations-, Thorax- und Gefäßchirurgie, Universitätsklinikum Leipzig, Leipzig, Deutschland
| | - Seung-Hun Chon
- Klinik und Poliklinik für Allgemein-, Viszeral-, Thorax- und Transplantationschirurgie, Uniklinik Köln, Köln, Deutschland
| | - Hakan Alakus
- Klinik und Poliklinik für Allgemein-, Viszeral-, Thorax- und Transplantationschirurgie, Uniklinik Köln, Köln, Deutschland
| | - René Thieme
- Klinik und Poliklinik für Viszeral-, Transplantations-, Thorax- und Gefäßchirurgie, Universitätsklinikum Leipzig, Leipzig, Deutschland
| | | | - Patrick Sven Plum
- Klinik und Poliklinik für Viszeral-, Transplantations-, Thorax- und Gefäßchirurgie, Universitätsklinikum Leipzig, Leipzig, Deutschland
| |
Collapse
|
382
|
Viscuse P, Skelton WP, Devitt MM, Dreicer R. When You Get to the Fork in the Road, Take It: The Challenges in Managing Patients With Advanced Prostate Cancer. JCO Oncol Pract 2025; 21:467-475. [PMID: 39353159 DOI: 10.1200/op-24-00591] [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: 07/23/2024] [Revised: 09/03/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024] Open
Abstract
As is the case with most solid tumors, the heterogeneity of the disease biology of prostate cancer presents clinicians managing this disease with daily challenges. However, in contrast to other common cancers such as breast, lung, and colorectal cancers, there are unique challenges in prostate cancer management, including the variety of clinicians who manage aspects of the disease (urologists, medical oncologist, radiation oncologists) and the striking absence of prospective comparative data to inform the optimal sequence of systemic therapy in patients with metastatic castration-resistant disease. The purpose of this review is to attempt to assist practicing oncologists with sorting through the myriad of prostate cancer disease subsets and the challenges in making therapeutic decisions in multiple data-free zones given the absence of level 1 comparative clinical trials in the metastatic hormone-sensitive and castration-resistant states.
Collapse
Affiliation(s)
- Paul Viscuse
- University of Virginia Comprehensive Cancer Center, Charlottesville, VA
| | - William P Skelton
- University of Virginia Comprehensive Cancer Center, Charlottesville, VA
| | - Michael M Devitt
- University of Virginia Comprehensive Cancer Center, Charlottesville, VA
| | - Robert Dreicer
- University of Virginia Comprehensive Cancer Center, Charlottesville, VA
| |
Collapse
|
383
|
Inoue K, Nakamura Y, Caughey B, Zheng-Lin B, Ueno M, Furukawa M, Kawamoto Y, Itoh S, Umemoto K, Sudo K, Satoh T, Mizuno N, Kajiwara T, Fujisawa T, Bando H, Yoshino T, Strickler JH, Morizane C, Bekaii-Saab T, Ikeda M. Clinicomolecular Profile and Efficacy of Human Epidermal Growth Factor Receptor 2 (HER2)-Targeted Therapy for HER2-Amplified Advanced Biliary Tract Cancer. JCO Precis Oncol 2025; 9:e2400718. [PMID: 40209139 PMCID: PMC12005869 DOI: 10.1200/po-24-00718] [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: 10/07/2024] [Revised: 12/17/2024] [Accepted: 02/19/2025] [Indexed: 04/12/2025] Open
Abstract
PURPOSE This study aimed to investigate the clinicomolecular profiles and the efficacy of human epidermal growth factor receptor 2 (HER2)-targeted therapy in HER2-amplified biliary tract cancer (BTC). METHODS This study was an international collaboration that used combined data from the prospective SCRUM-Japan GOZILA and MONSTAR-SCREEN in Japan and retrospective reviews in the United States; patients with advanced BTC who had received systemic therapy were included. The clinicomolecular profiles were evaluated in an exploratory cohort, whereas the efficacy of HER2-targeted therapy was assessed in a biomarker-selected cohort. RESULTS Of the 439 patients in the exploratory cohort, 43 (10%) had HER2 amplification. The frequencies of coalterations were higher in patients with HER2 amplification versus patients without HER2 amplification including HER2 mutations (26% v 5%, P < .001), TP53 mutations (84% v 61%, P = .003), and BRAF amplification (9% v 2%, P = .030). There were no KRAS mutations identified in patients with HER2-amplified BTC. No significant difference in overall survival (OS) was observed between patients with and without HER2 amplification (median, 17.7 v 16.9 months; hazard ratio [HR], 0.95 [95% CI, 0.65 to 1.40]). Of the 60 patients with HER2-amplified BTC in the biomarker-selected cohort (43 from Japan and 17 from the United States), the OS was significantly longer in 29 patients who received HER2-targeted therapy than in those who did not receive HER2-targeted therapy (median, 24.3 v 12.1 months; HR, 0.39 [95% CI, 0.23 to 0.82]). Multivariate analysis identified HER2-targeted therapy as an independent prognostic factor for OS (HR, 0.29 [95% CI, 0.14 to 0.58]; P < .001). CONCLUSION HER2 amplification was found in 10% of advanced BTC and was not identified as an independent prognostic factor for OS. Patients with HER2-amplified BTC derive significant benefit from HER2-targeted therapy.
Collapse
Affiliation(s)
- Kanae Inoue
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshiaki Nakamura
- Translational Research Support Office, Division of Drug and Diagnostic Development Promotion, Department for the Promotion of Drug and Diagnostic Development, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Bennett Caughey
- Division of Hematology/Oncology, Massachusetts General Hospital, Boston, MA
| | - Binbin Zheng-Lin
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Masayuki Furukawa
- Department of Hepato-Biliary-Pancreatology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Yasuyuki Kawamoto
- Division of Cancer Center, Hokkaido University Hospital, Sapporo, Japan
| | - Shinji Itoh
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kumiko Umemoto
- Department of Clinical Oncology, St Marianna University School of Medicine, Kawasaki, Japan
| | - Kentaro Sudo
- Department of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Taroh Satoh
- Center for Cancer Genomics and Precision Medicine, Osaka University Hospital, Osaka, Japan
| | - Nobumasa Mizuno
- Department of Gastroenterology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Takeshi Kajiwara
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Takao Fujisawa
- Translational Research Support Office, Division of Drug and Diagnostic Development Promotion, Department for the Promotion of Drug and Diagnostic Development, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Head and Neck Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hideaki Bando
- Translational Research Support Office, Division of Drug and Diagnostic Development Promotion, Department for the Promotion of Drug and Diagnostic Development, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | | | - Chigusa Morizane
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | | | - Masafumi Ikeda
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
384
|
Bouhniz OE, Kenani A. Potential role of genetic polymorphisms in neoadjuvant chemotherapy response in breast cancer. J Chemother 2025; 37:97-111. [PMID: 38511398 DOI: 10.1080/1120009x.2024.2330241] [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: 10/27/2023] [Revised: 02/09/2024] [Accepted: 03/11/2024] [Indexed: 03/22/2024]
Abstract
Chemoresistance leads to treatment failure, which can arise through different mechanisms including patients' characteristics. Searching for genetic profiles as a predictor for drug response and toxicity has been extensively studied in pharmacogenomics, thus contributing to personalized medicine and providing alternative treatments. Numerous studies have demonstrated significant evidence of association between genetic polymorphisms and response to neoadjuvant chemotherapy (NAC) in breast cancer. In this review, we explored the potential impact of genetic polymorphisms in NAC primary resistance through selecting a specific clinical profile. The genetic variability within pharmacokinetics, pharmacodynamics, DNA synthesis and repair, and oncogenic signaling pathways genes could be predictive or prognostic markers for NAC resistance. The clinical implication of these results can help provide individualized treatment plans in the early stages of breast cancer treatment. Further studies are needed to determine the genetic hosts of primary chemoresistance mechanisms in order to further emphasize the implementation of genotypic approaches in personalized medicine.
Collapse
Affiliation(s)
- Om Elez Bouhniz
- Research Laboratory "Environment, Inflammation, Signaling and Pathologies" (LR18ES40), Faculty of Medicine of Monastir, University of Monastir, Monastir, Tunisia
| | - Abderraouf Kenani
- Research Laboratory "Environment, Inflammation, Signaling and Pathologies" (LR18ES40), Faculty of Medicine of Monastir, University of Monastir, Monastir, Tunisia
| |
Collapse
|
385
|
Lee JH, Kim J, Lee DG. The Influence of Lifestyle Behaviors and Body Mass Index Changes on Long-term Outcomes After Gastric Cancer Surgery: A Population-Based Cohort Study. J Gastric Cancer 2025; 25:356-369. [PMID: 40200878 PMCID: PMC11982505 DOI: 10.5230/jgc.2025.25.e18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/26/2024] [Accepted: 02/03/2025] [Indexed: 04/10/2025] Open
Abstract
PURPOSE The present study investigated the impact of lifestyle behaviors and body mass index (BMI) on late recurrence, gastric remnant cancer (GRC), and long-term survival after curative gastrectomy. MATERIALS AND METHODS This retrospective study utilized data from the Korean National Health Insurance claims database. Among 71,014 patients with gastric cancer who underwent curative gastrectomy between January 2009 and December 2012, 23,359 remained cancer-free for five years. Of these, 7,735 patients with health examination data within 2 years before surgery and 5 years after surgery were analyzed for lifestyle behaviors, including smoking, alcohol consumption, and physical activity. Multivariable analysis was used to evaluate the independent effects of these factors and changes in BMI on late recurrence, GRC, and long-term survival. RESULTS Late recurrence or GRC occurred among 628 patients (8.1%). Older age (≥60 years) and total gastrectomy were identified as risk factors. Although lifestyle behaviors and BMI changes did not directly affect recurrence, they significantly affected mortality. In the total gastrectomy group, current underweight status (hazard ratio [HR], 1.586) was associated with increased mortality. Among the partial gastrectomy group, continued smoking (HR, 1.366) and current underweight status (HR, 1.915) increased mortality risk. Conversely, regular physical activity (starting: HR, 0.674; continuing: HR, 0.699) and postoperative overweight or obesity (BMI >25 kg/m²) (HR, 0.713) were associated with reduced mortality. Changes in alcohol consumption showed inconsistent effects between the partial and total gastrectomy groups. CONCLUSIONS The long-term survival of post-gastrectomy patients improved with smoking cessation, regular physical activity, and maintenance of body weight.
Collapse
Affiliation(s)
- Ju-Hee Lee
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea.
| | - Jiyeong Kim
- Department of Pre-Medicine, College of Medicine, and Biostatistics Laboratory, Medical Research Collaborating Center, Hanyang University, Seoul, Korea
| | - Dong-Gyu Lee
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
| |
Collapse
|
386
|
Kim B, Park J, Na HY, Park S, Jin J, Jung K, Lee JC, Hwang JH, Seo M, Kim J. The origin of patient-derived cancer organoids from pathologically undiagnosed specimen in patients with pancreatobiliary cancers. Cell Oncol (Dordr) 2025; 48:523-535. [PMID: 39688793 PMCID: PMC11996933 DOI: 10.1007/s13402-024-01026-5] [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] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
PURPOSE Tissue confirmation of pancreatobiliary cancer is often difficult because of the location of the tumor and structure of the surrounding blood vessels. Patient-derived cancer organoids (PDCOs) reflect the genomic characteristics of individual cancers. Although diverse attempts to construct PDCOs for various pancreatobiliary cancer models are ongoing, no research results have yet confirmed the possibility of performing a precise diagnosis on PDCOs derived from pathologically negative patient samples. METHODS We obtained a total of nine samples, including pathologically negative samples, from four patients (three patients with pancreatic cancer and one patient with gallbladder cancer) using different tissue acquisition methods to establish PDCOs (success rate 75%). RESULTS We successfully verified whether the constructed PDCOs could represent the tissues of patients with pancreatobiliary cancer at each multi-omics level using tumor panel sequencing, single-cell RNA sequencing, hematoxylin and eosin, and immunohistochemical staining. PDCOs from pathologically negative samples showed expression patterns of malignant ductal cell-related biomarkers similar to those of other pathologically positive samples. Furthermore, the expression patterns at the single-cell level in PDCO from patients ultimately diagnosed with gallbladder cancer after surgery were different from those in patients with pancreatic cancer. CONCLUSION Therefore, our study implicated the potential of PDCOs as diagnostic and research tools, including for case involving limited tissue samples. Based on these results, we anticipate that this could be extended to more advanced studies, such as drug sensitivity testing, through large-scale trials in the near future.
Collapse
Affiliation(s)
- Bomi Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | | | - Hee Young Na
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Sinwoo Park
- Department of Computer and Information Science, Korea University, Sejong, Korea
| | | | - Kwangrok Jung
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Jong-Chan Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Jin-Hyeok Hwang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Minseok Seo
- Department of Computer and Information Science, Korea University, Sejong, Korea.
| | - Jaihwan Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea.
| |
Collapse
|
387
|
Mukherjee S, Fujiwara Y, Fountzilas C, Pattnaik H, Chatley S, Vadehra D, Kukar M, Attwood K, George A, Advani S, Yu H, Catalfamo K, Brown A, Spickard E, Fungtammasan A, George S, Liao C, Iyer R, Hatoum H. Trifluridine/Tipiracil and Oxaliplatin as Induction Chemotherapy in Resectable Esophageal and Gastroesophageal Junction Adenocarcinoma: A Phase II Study. Cancer Med 2025; 14:e70835. [PMID: 40200573 PMCID: PMC11978735 DOI: 10.1002/cam4.70835] [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: 03/12/2025] [Accepted: 03/18/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Preoperative chemoradiation (CRT) followed by surgery for localized esophageal and gastroesophageal junction adenocarcinoma (EGAC) is a standard of care with a pathologic complete response (pCR) rate of 20%. We evaluated a novel combination of trifluridine/tipiracil with oxaliplatin as induction chemotherapy (IC) followed by CRT. METHODS We enrolled patients with potentially resectable localized EGAC (T3, T4aN0, or node-positive disease) in this open-label, single-arm, multicenter, Phase II trial between January 2020 and October 2022. Patients received three cycles of IC with trifluridine/tipiracil and oxaliplatin and then underwent concurrent CRT with weekly carboplatin and paclitaxel followed by surgery. The primary objective was to evaluate the pCR rate. The secondary objectives were to evaluate 2-year progression-free survival (PFS), 2-year overall survival (OS), and toxicities. Circulating tumor DNA (ctDNA) was measured at prespecified intervals to assess its correlation with clinical outcomes. RESULTS Of the 22 enrolled patients, 19 (86.4%) were male and 20 (90.9%) were Caucasian. The median age was 61 years, and 12 (54.5%) had their primary disease at the gastroesophageal junction. Twenty (90.9%) patients had T3 disease, and 15 (68.2%) had node-positive disease. Only two patients had pCRs, and an additional five had near pCRs. Since we could not meet our predefined pCR rate at the interim analysis, the study was closed. After a median follow-up of 15.8 months, 2-year OS and PFS were 43% and 41%, respectively. ctDNA clearance was associated with a significantly higher OS rate (p = 0.012) and PFS rate (p = 0.008). Nausea (59.1%) and fatigue (59.1%) were common treatment-related adverse events (AEs); nine (40.9%) patients had Grade 3 or higher AEs. CONCLUSION IC with trifluridine/tipiracil and oxaliplatin followed by CRT did not improve pCR rate in resectable EGAC compared to pCR from previous reports with CRT alone. We found a correlation between ctDNA clearance and improved survival, which merits further investigation. CLINICAL TRIAL INFORMATION NCT04097028.
Collapse
Affiliation(s)
- Sarbajit Mukherjee
- Roswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
- Miami Cancer Institute, Baptist Health South FloridaMiamiFloridaUSA
| | - Yu Fujiwara
- Roswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | | | | | - Sarah Chatley
- Roswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | - Deepak Vadehra
- Roswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | - Moshim Kukar
- Roswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | | | - Anthony George
- Roswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | | | - Han Yu
- Roswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | | | - Alyson Brown
- Roswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | | | | | - Sagila George
- Stephenson Cancer Center – University of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
| | - Chih‐Yi Liao
- The University of Chicago Medical Center & Biological SciencesChicagoIllinoisUSA
| | - Renuka Iyer
- Roswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | - Hassan Hatoum
- Stephenson Cancer Center – University of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
| |
Collapse
|
388
|
Sun Y, Gao L, Zhou X, Wang Z, Li Y, Sun Q. Local Recurrence and Survival Outcomes of Multifocal/Multicentric Breast Cancer After Breast Conserving Therapy: A systematic Review and Meta-Analysis. Clin Breast Cancer 2025; 25:e229-e239.e9. [PMID: 39542811 DOI: 10.1016/j.clbc.2024.10.009] [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: 09/04/2024] [Revised: 10/10/2024] [Accepted: 10/13/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND The appropriateness of BCT for MF/MCBC is debated, with concerns about higher recurrence rates. This study aims to provide an updated systematic review and meta-analysis of LR and survival outcomes for MF/MCBC patients undergoing BCT. METHODS PubMed, Web of Science, Embase, and the Cochrane Library were searched up to May 2024. Eligible studies included original research articles comparing LR, DFS, or OS in patients with MF/MC or UF breast cancer undergoing BCT or mastectomy. Meta-analyses for LR were conducted using the Mantel-Haenszel method. Published Kaplan-Meier curves for DFS and OS were digitized and aggregated to estimate summary survival curves. RESULTS 21 studies were included in the meta-analysis for LR, comprising 28,589 participants, and 7 studies for survival analysis. The meta-analysis revealed that MF/MC breast cancer patients undergoing BCT had a significantly higher LR rate compared to UF patients (OR = 1.76, 95% CI: 1.24-2.49, P = .002), though recent studies indicated comparable LR rates. No significant difference in LR was found between MF/MC patients treated with BCT versus mastectomy (OR = 1.72, 95% CI: 0.96-3.10, P = .07). The estimated 3-, 5-, and 8-year DFS rates were 92.4%, 88.3%, and 84.5%, respectively, while the OS rates were 98.0%, 95.8%, and 91.8%. CONCLUSION BCT for MF/MC breast cancer was associated with higher LR rates compared to UF breast cancer, but the disparity was reducing in recent years. BCT offers comparable LR outcomes to mastectomy in MF/MC patients. Survival outcomes for MF/MC patients treated with BCT were favorable, affirming its oncological safety.
Collapse
Affiliation(s)
- Youshi Sun
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lu Gao
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xingtong Zhou
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zihao Wang
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Li
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of International Medical Service (Xidan Campus), Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Qiang Sun
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
389
|
Kicken MP, Deenen MJ, van der Wekken AJ, van den Borne BEEM, van den Heuvel MM, Ter Heine R. Opportunities for Precision Dosing of Cytotoxic Drugs in Non-Small Cell Lung Cancer: Bridging the Gap in Precision Medicine. Clin Pharmacokinet 2025; 64:511-531. [PMID: 40045151 PMCID: PMC12041064 DOI: 10.1007/s40262-025-01492-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2025] [Indexed: 04/30/2025]
Abstract
Precision dosing of classical cytotoxic drugs in oncology remains underdeveloped, especially in treating non-small cell lung cancer (NSCLC). Despite advancements in targeted therapy and immunotherapy, classical cytotoxic agents continue to play a critical role in NSCLC treatment. However, the current body surface area (BSA)-based dosing of these agents fails to adequately address interindividual variability in pharmacokinetics. By better considering patient characteristics, treatment outcomes can be improved, reducing risks of under-exposure and over-exposure. This narrative review explores opportunities for precision dosing for key cytotoxic agents used in NSCLC treatment: cisplatin, carboplatin, pemetrexed, docetaxel, (nab-)paclitaxel, gemcitabine, and vinorelbine. A comprehensive review of regulatory reports and an extensive literature search were conducted to evaluate current dosing practices, pharmacokinetics, pharmacodynamics, and exposure-response relationships. Our findings highlight promising developments in precision dosing, although the number of directly implementable strategies remains limited. The most compelling evidence supports using the biomarker cystatin C for more precise carboplatin dosing and adopting weekly dosing schedules for docetaxel, paclitaxel, and nab-paclitaxel. Additionally, we recommend direct implementation of therapeutic drug monitoring (TDM)-guided dosing for paclitaxel. This review stresses the urgent need to reassess conventional dosing paradigms for classical cytotoxic agents to better align with the principles of the precision dosing framework. Our recommendations show the potential of precision dosing to improve NSCLC treatment, addressing gaps in the current dosing of classical cytotoxic drugs. Given the large NSCLC patient population, optimising the dosing of these agents could significantly improve treatment outcomes and reduce toxicity for many patients.
Collapse
Affiliation(s)
- M P Kicken
- Department of Clinical Pharmacy, Catharina Hospital, Michelangelolaan 2, 5623 EJ, Eindhoven, The Netherlands.
- Department of Pharmacy, Radboudumc, Research Institute for Medical Innovation, Nijmegen, The Netherlands.
| | - M J Deenen
- Department of Clinical Pharmacy, Catharina Hospital, Michelangelolaan 2, 5623 EJ, Eindhoven, The Netherlands
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, The Netherlands
| | - A J van der Wekken
- Department of Pulmonology and Tuberculosis, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | | | - M M van den Heuvel
- Department of Pulmonology, Radboudumc, Research Institute for Medical Innovation, Nijmegen, The Netherlands
- Department of Pulmonology, University Medical Center, Utrecht, The Netherlands
| | - R Ter Heine
- Department of Pharmacy, Radboudumc, Research Institute for Medical Innovation, Nijmegen, The Netherlands
| |
Collapse
|
390
|
Oh SE, Suh YS, An JY, Ryu KW, Cho I, Kim SG, Park JH, Hur H, Kim HH, Ahn SH, Hwang SH, Yoon HM, Park KB, Kim HI, Kwon IG, Yang HK, Suh BJ, Jeong SH, Kim TH, Kwon OK, Ahn HS, Park JY, Yoon KY, Son MW, Kong SH, Son YG, Song GJ, Yun JH, Bae JM, Park DJ, Lee S, Yang JY, Seo KW, Jang YJ, Kang SH, Eom BW, Lee J, Lee HJ. Prospective Multicenter Observational Study on Postoperative Quality of Life According to Type of Gastrectomy for Gastric Cancer. J Gastric Cancer 2025; 25:382-399. [PMID: 40200880 PMCID: PMC11982510 DOI: 10.5230/jgc.2025.25.e26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 04/10/2025] Open
Abstract
PURPOSE This study evaluated the postoperative quality of life (QoL) after various types of gastrectomy for gastric cancer. MATERIALS AND METHODS A multicenter prospective observational study was conducted in Korea using the Korean Quality of Life in Stomach Cancer Patients Study (KOQUSS)-40, a new QoL assessment tool focusing on postgastrectomy syndrome. Overall, 496 patients with gastric cancer were enrolled, and QoL was assessed at 5 time points: preoperatively and at 1, 3, 6, and 12 months after surgery. RESULTS Distal gastrectomy (DG) and pylorus-preserving gastrectomy (PPG) showed significantly better outcomes than total gastrectomy (TG) and proximal gastrectomy (PG) with regard to total score, indigestion, and dysphagia. DG, PPG, and TG also showed significantly better outcomes than PG in terms of dumping syndrome and worry about cancer. Postoperative QoL did not differ significantly according to anastomosis type in DG, except for Billroth I anastomosis, which achieved better bowel habit change scores than the others. No domains differed significantly when comparing double tract reconstruction and esophagogastrostomy after PG. The total QoL score correlated significantly with postoperative body weight loss (more than 10%) and extent of resection (P<0.05 for both). Reflux as assessed by KOQUSS-40 did not correlate significantly with reflux observed on gastroscopy 1 year postoperatively (P=0.064). CONCLUSIONS Our prospective observation using KOQUSS-40 revealed that DG and PPG lead to better QoL than TG and PG. Further study is needed to compare postoperative QoL according to anastomosis type in DG and PG.
Collapse
Affiliation(s)
- Sung Eun Oh
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yun-Suhk Suh
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Ji Yeong An
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Keun Won Ryu
- Center for Gastric Cancer, National Cancer Center, Goyang, Korea
| | - In Cho
- Department of Surgery, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Sung Geun Kim
- Department of Surgery, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ji-Ho Park
- Department of Surgery, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju, Korea
| | - Hoon Hur
- Department of Surgery, Ajou University School of Medicine, Suwon, Korea
| | - Hyung-Ho Kim
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Sang-Hoon Ahn
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Sun-Hwi Hwang
- Department of Surgery, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, Korea
| | - Hong Man Yoon
- Center for Gastric Cancer, National Cancer Center, Goyang, Korea
| | - Ki Bum Park
- Department of Surgery, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Hyoung-Il Kim
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - In Gyu Kwon
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Han-Kwang Yang
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Byoung-Jo Suh
- Department of Surgery, Inje University Haeundae Paik Hospital, Busan, Korea
| | - Sang-Ho Jeong
- Department of Surgery, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon, Korea
| | - Tae-Han Kim
- Department of Surgery, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon, Korea
| | - Oh Kyoung Kwon
- Department of Surgery, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Hye Seong Ahn
- Department of Surgery, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Ji Yeon Park
- Department of Surgery, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Ki Young Yoon
- Department of Surgery, Kosin University College of Medicine, Busan, Korea
| | - Myoung Won Son
- Department of Surgery, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Seong-Ho Kong
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Young-Gil Son
- Department of Surgery, Keimyung University Dongsan Medical Center, Daegu, Korea
| | - Geum Jong Song
- Department of Surgery, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Jong Hyuk Yun
- Department of Surgery, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Jung-Min Bae
- Department of Surgery, Yeungnam University College of Medicine, Daegu, Korea
| | - Do Joong Park
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Sol Lee
- Department of Surgery, Seoul Medical Center, Seoul, Korea
| | - Jun-Young Yang
- Department of Surgery, Gachon University Gil Medical Center, Incheon, Korea
| | - Kyung Won Seo
- Department of Surgery, Kosin University College of Medicine, Busan, Korea
| | - You-Jin Jang
- Department of Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - So Hyun Kang
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Bang Wool Eom
- Center for Gastric Cancer, National Cancer Center, Goyang, Korea
| | - Joongyub Lee
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, Korea.
| | - Hyuk-Joon Lee
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
391
|
Cooper WA, Webster F, Butnor KJ, Calabrese F, Chou TY, Hwang DM, Kern I, Popat S, Sholl L, Yatabe Y, Nicholson AG. Data set for the reporting of lung cancer: recommendations from the International Collaboration on Cancer Reporting (ICCR). Histopathology 2025; 86:665-680. [PMID: 39438780 DOI: 10.1111/his.15313] [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] [Indexed: 10/25/2024]
Abstract
Lung cancer is the leading cause of cancer related deaths worldwide, although some patients with early-stage disease can be cured with surgical resection. Standardised reporting of all clinically relevant pathological parameters is essential for best patient care and is also important for ongoing data collection and refinement of important pathological features that impact patient prognosis, staging and clinical care. Using the established International Collaboration on Cancer Reporting (ICCR) procedure, a representative international expert panel of nine lung pathologists as well as an oncologist was convened. Essential core elements and suggested non-core elements were identified for inclusion in the resected lung cancer pathology data set based on predetermined levels of evidence as well as consensus expert opinion. A lung cancer histopathology reporting guide was developed that includes relevant clinical, macroscopic, microscopic and ancillary testing. Critical review and discussion of current evidence was incorporated into the new data set including changes from the 2021 World Health Organisation (WHO) Classification of Thoracic Tumours, fifth edition, new requirements for grading invasive non-mucinous adenocarcinomas, assessment of response to neoadjuvant therapy and requirements for molecular testing in early-stage resected lung carcinomas. This ICCR data set represents incorporation of all relevant parameters for histology reporting of lung cancer resection specimens. Routine use of this data set is recommended for all pathology reporting of resected lung cancer and it is freely available worldwide on the ICCR website (https://www.iccr-cancer.org/datasets/published-datasets/). Widespread implementation will help to ensure consistent and comprehensive pathology reporting and data collection essential for lung cancer patient care, clinical trials and other research.
Collapse
Affiliation(s)
- Wendy A Cooper
- Royal Prince Alfred Hospital, NSW Health Pathology, Camperdown, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | - Fleur Webster
- International Collaboration on Cancer Reporting, Surry Hills, NSW, Australia
| | - Kelly J Butnor
- Department of Pathology and Laboratory Medicine, The University of Vermont Medical Center, Burlington, Vermont, USA
| | - Fiorella Calabrese
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | | | - David M Hwang
- Department of Laboratory Medicine and Pathobiology, University of Toronto and Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Izidor Kern
- Cytology and Pathology Laboratory, University Clinic of Respiratory and Allergic Diseases, Golnik, Slovenia
| | | | - Lynette Sholl
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Yasushi Yatabe
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| | | |
Collapse
|
392
|
Nash A, DeBonis J, Murungi D, Castillo B, Kim B, Hu F, Chambers C, Nguyen A, Hernandez A, Wang Z, Rios PD, Ghani S, Joshi I, Isa D, Zheng N, Peng W, Igoshin OA, Oberholzer J, Hodges HC, Reticker-Flynn N, Veiseh O. IL-12-producing cytokine factories induce precursor exhausted T cells and elimination of primary and metastatic tumors. J Immunother Cancer 2025; 13:e010685. [PMID: 40169286 PMCID: PMC11962782 DOI: 10.1136/jitc-2024-010685] [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: 10/03/2024] [Accepted: 03/17/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Curative responses to immunotherapy require the generation of robust systemic immunity with limited toxicity. Recruitment of T cell populations such as precursor exhausted T cells (Tpex) from lymphoid tissues to tumors is a hallmark of effective treatment. However, the ability to efficiently induce this recruitment is lacking in current immunotherapy approaches. Furthermore, systemic administration of immunotherapies frequently results in dose-limiting toxicities, yielding an inadequate therapeutic window for eliciting durable responses. METHODS In this investigation, we evaluated the safety and antitumor efficacy of locally administered interleukin 12 (IL-12) using a clinically translatable cytokine delivery platform (NCT05538624) to identify Tpex recruitment capabilities at tolerable cytokine doses. RESULTS We show IL-12 cytokine factories can effectively treat a broad spectrum of cancer types. Single-cell RNA sequencing data suggests that the antitumor efficacy seen in our studies was due to retinal pigmented epithelial cells-mIL12 treatment inducing differentiation of Tpex cells within the tumor microenvironment. When administered in combination with checkpoint therapy, IL-12 cytokine factory treatment generated systemic abscopal immunity, preventing subcutaneous tumor outgrowth in 8/9 mice with colorectal cancer and lung metastasis in mice with melanoma. Furthermore, this platform was well tolerated in a non-human primate without signs of toxicity. CONCLUSIONS Our new immunotherapy approach provides a robust strategy for inducing Tpex recruitment and systemic immunity against a range of solid peritoneal malignancies, many incurable with current immunotherapy strategies. Notably, these features were achieved using IL-12, and by leveraging our technology, we avoided the toxicities that have prevented the translation of IL-12 to the clinic. Our findings provide a strong rationale for the clinical development of IL-12 cytokine factories.
Collapse
Affiliation(s)
- Amanda Nash
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Jonathon DeBonis
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Danna Murungi
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Bertha Castillo
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Boram Kim
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Fangheng Hu
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Courtney Chambers
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Annie Nguyen
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Andrea Hernandez
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Zeshi Wang
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | | | | | | | | | - Ningbo Zheng
- Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Weiyi Peng
- Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Oleg A Igoshin
- Department of Bioengineering, Rice University, Houston, Texas, USA
- Department of Biosciences, Rice University, Houston, Texas, USA
- Department of Chemistry, Rice University, Houston, Texas, USA
- Center for Theoretical Biological Physics, Rice University, HoustON, Texas, USA
| | - Jose Oberholzer
- Celltrans, Chicago, Illinois, USA
- Department of Visceral Surgery and Transplantation, University Hospital Zurich, Zurich, Switzerland
| | - H Courtney Hodges
- Department of Bioengineering, Rice University, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Nathan Reticker-Flynn
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, Stanford, California, USA
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, Texas, USA
| |
Collapse
|
393
|
Busakhala N, Atundo L, Kiprono H, Keitany K, Melly E, Ruto R, Wanja M, Chepsiror D, Rangoonwala H, Kipchirchir C, Chesori E, Oguda J, Opakas J, Loehrer PJ, Diero L, Morgan J. Characteristics and Associated Survival of Patients Diagnosed With Non-Small Cell Lung Cancer in a Designated Lung Cancer Program in Western Kenya. JCO Glob Oncol 2025; 11:e2400212. [PMID: 40184569 DOI: 10.1200/go.24.00212] [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: 05/16/2024] [Revised: 10/10/2024] [Accepted: 02/11/2025] [Indexed: 04/06/2025] Open
Abstract
PURPOSE Although lung cancer is a major cause of cancer incidence and mortality worldwide, lung cancer studies in sub-Saharan Africa are scarce. Here, we present outputs from a designated lung cancer program in western Kenya, part of the Multi-National Lung Cancer Control Program, which focused on case finding, diagnosis, and treatment. METHODS We retrospectively reviewed patients with pathologically confirmed non-small cell lung cancer (NSCLC) enrolled in this program at Moi Teaching and Referral Hospital from January 2018 to December 2022. Clinical data were analyzed using descriptive statistics, Kaplan-Meier methods, and proportional hazards regression model. RESULTS Two hundred forty-nine patients diagnosed with NSCLC were included with a median age at diagnosis of 61 (IQR, 52-70) years. Most patients were married (n = 177; 71%) and nonsmokers (n = 177; 71%) with 58 (23%) having received tuberculosis treatment and 93 (37%) having Eastern Cooperative Oncology Group (ECOG) performance status (PS) of ≥ 2. At diagnosis, adenocarcinoma was the prominent histology (n = 187; 75%) along with clinical stage IV (n = 195; 78% stage IV) or unstaged (n = 40; 16%) disease. Most patients received chemotherapy and radiotherapy (n = 176; 71%) with few palliative care referrals (n = 2; 0.8%). The median overall survival (OS) was only 3.7 months (IQR, 2.7-5.4). ECOG PS (3 or 4) and being unstaged were predictors of poor 1-year OS. CONCLUSION Patients with NSCLC enrolled in this program presented with advanced disease and poor survival. Despite a designated case finding effort, late diagnosis remained common and highlights a need for locally relevant interventions targeting community and provider education as well as innovative diagnostics that can improve early recognition of lung cancer. These interventions must also be paired with access to proven treatments including molecular therapies and palliative care which can extend lung cancer survival.
Collapse
Affiliation(s)
- Naftali Busakhala
- Academic Model for Providing Access to Healthcare (AMPATH), Eldoret, Kenya
- Moi University College of Health Sciences, Eldoret, Kenya
- Moi University Teaching and Referral Hospital, Eldoret, Kenya
| | - Lawrence Atundo
- Academic Model for Providing Access to Healthcare (AMPATH), Eldoret, Kenya
| | | | - Kibet Keitany
- Moi University Teaching and Referral Hospital, Eldoret, Kenya
| | - Elias Melly
- Kenya National Cancer Institute, Nairobi, Kenya
| | - Ruth Ruto
- Academic Model for Providing Access to Healthcare (AMPATH), Eldoret, Kenya
| | - Madrine Wanja
- Academic Model for Providing Access to Healthcare (AMPATH), Eldoret, Kenya
| | | | | | | | - Erick Chesori
- Moi University Teaching and Referral Hospital, Eldoret, Kenya
| | - John Oguda
- Indiana University School of Medicine, Indianapolis, IN
| | - Jesse Opakas
- Moi University Teaching and Referral Hospital, Eldoret, Kenya
| | - Patrick J Loehrer
- Academic Model for Providing Access to Healthcare (AMPATH), Eldoret, Kenya
- Indiana University School of Medicine, Indianapolis, IN
| | - Lameck Diero
- Academic Model for Providing Access to Healthcare (AMPATH), Eldoret, Kenya
- Moi University College of Health Sciences, Eldoret, Kenya
- Moi University Teaching and Referral Hospital, Eldoret, Kenya
| | - Jennifer Morgan
- Academic Model for Providing Access to Healthcare (AMPATH), Eldoret, Kenya
- Indiana University School of Medicine, Indianapolis, IN
- University of Minnesota School of Medicine, Minneapolis, MN
| |
Collapse
|
394
|
He L, Liu B, Wang Z, Han Q, Chen H. Evolving Landscape of HER2-Targeted Therapies for Gastric Cancer Patients. Curr Treat Options Oncol 2025; 26:260-277. [PMID: 40056280 DOI: 10.1007/s11864-025-01300-0] [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] [Accepted: 02/06/2025] [Indexed: 03/10/2025]
Abstract
OPINION STATEMENT Gastric cancer (GC) is a deadly disease worldwide, and trastuzumab in combination with chemotherapy has been the standard first-line treatment for HER2-positive GC following the TOGA trial. Besides adjuvant therapy, HER2-directed therapy is widely used as neoadjuvant or translational therapy, and survival benefit even surgical opportunities is seen in these patients. However, resistance is not rare in recent years, and the second-line treatment for trastuzumab beyond progression has received widespread attention in GC. Moreover, current evidence cannot recommend trastuzumab for patients with IHC1+ HER2 low expression GC yet. Researchers are currently investigating whether GC patients with low HER2 expression could also benefit from HER2-directed therapies. In addition to using HER2 as a target for targeted therapy, HER2-mediated targeted delivery of cytotoxic drugs and targeted immunity have made important contributions to overcoming trastuzumab resistance in recent trials. HER2/neu-derived peptide epitopes vaccination and HER2-specific chimeric antigen receptor (CAR) therapy focus on reestablishing anti-tumor immunity in different ways and show significant anti-tumor activity. Other antibodies that target different regions of the HER2 receptor or block key downstream pathways such as AKT or PI3K also offer potential anti-tumor activity against HER2. HER2 use in GC will not be hampered by resistance or low expression and will play a bigger role. We review the current efforts to enable GC patients with trastuzumab-resistant and HER2 low-expressing accessible to HER2 targeted therapy and present our consideration for future HER2 in GC.
Collapse
Affiliation(s)
- Lijuan He
- Lanzhou University Second Hospital, Lanzhou, 730030, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Ben Liu
- Lanzhou University Second Hospital, Lanzhou, 730030, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Zhuanfang Wang
- Lanzhou University Second Hospital, Lanzhou, 730030, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Qinying Han
- Lanzhou University Second Hospital, Lanzhou, 730030, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Hao Chen
- Lanzhou University Second Hospital, Lanzhou, 730030, China.
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, 730030, China.
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou, 730030, China.
- Humanized animal model laboratory, Lanzhou University Second Hospital, Lanzhou, 730030, China.
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China.
| |
Collapse
|
395
|
Tu DH, Qu R, Wen F, Zhou Q, Liu Q, Huang L, Chen T. Successful conversion surgery following tislelizumab with chemotherapy in a patient with stage IIIC lung adenocarcinoma harboring RET fusions: A case report and review of the literature. Exp Ther Med 2025; 29:70. [PMID: 39991722 PMCID: PMC11843209 DOI: 10.3892/etm.2025.12820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 01/28/2025] [Indexed: 02/25/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have emerged as a beacon of hope for most patients with stage III non-small cell lung cancer (NSCLC) who are no longer surgical candidates. However, the literature on the use of immunotherapy in patients with NSCLC with rearranged during transfection (RET) gene fusions is scant. The present study reports the case of a 61-year-old female patient, diagnosed with stage IIIC lung adenocarcinoma, exhibiting two RET gene fusions and high programmed death-ligand 1 expression. Following four treatment cycles of tislelizumab in combination with pemetrexed and cisplatin, the patient was successfully downstaged, enabling radical surgery. The post-operative pathology analysis indicated a major pathologic response. This case study contributes to the growing body of evidence supporting the use of ICIs in treating locally advanced NSCLC with RET gene fusions.
Collapse
Affiliation(s)
- De-Hao Tu
- Department of Thoracic Surgery, Yueyang Central Hospital, Yueyang, Hunan 414000, P.R. China
| | - Rirong Qu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Fang Wen
- Department of Oncology, Yueyang Central Hospital, Yueyang, Hunan 414000, P.R. China
| | - Qiang Zhou
- Department of Oncology, Yueyang Central Hospital, Yueyang, Hunan 414000, P.R. China
| | - Qianyun Liu
- Department of Medical Imaging, Yueyang Central Hospital, Yueyang, Hunan 414000, P.R. China
| | - Lingmei Huang
- Department of Pulmonary and Critical Care Medicine, Yueyang Central Hospital, Yueyang, Hunan 414000, P.R. China
| | - Tao Chen
- Department of Thoracic Surgery, Yueyang Central Hospital, Yueyang, Hunan 414000, P.R. China
| |
Collapse
|
396
|
Liu Y, Yan X, Qu C, Tang F, Wang Q, Li Y. The role of TMEM119 in gastric adenocarcinoma and its specific effects on immunity. J Int Med Res 2025; 53:3000605241306668. [PMID: 40219804 PMCID: PMC12033527 DOI: 10.1177/03000605241306668] [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: 03/20/2024] [Accepted: 11/25/2024] [Indexed: 04/14/2025] Open
Abstract
ObjectiveTo investigate the prognostic significance and immunological implication of transmembrane protein 119 (TMEM119) in stomach adenocarcinoma (STAD).MethodsThis study included STAD-associated data obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. In addition, TMEM119 expression levels were analysed by immunohistochemistry in tissue samples from patients with STAD (with microsatellite instability or microsatellite stability). Gene Set Enrichment Analysis (GSEA) was conducted to explore signalling pathways related to TMEM119 in STAD. Additionally, CIBERSORT and ESTIMATE algorithms were applied to examine the relationship between TMEM119 expression and tumour-infiltrating immune cells, as well as the tumour microenvironment.ResultsSurgical specimens from 100 patients with STAD (50 each with microsatellite stability or microsatellite instability); TCGA RNA-sequence and clinical data from 375 STAD tumour tissues and 32 paracancerous tissues; and two GEO datasets (GSE27342, comprising 80 paracancerous tissues and 80 tumour tissues; and GSE84437, comprising 433 tumour samples) were analysed. TMEM119 was found to be elevated in STAD, and associated with poor prognosis. Clinical gastric cancer tissues exhibited increased TMEM119 expression. TMEM119 was enriched in immune-related functions and pathways. TMEM119 correlated with immune checkpoint genes, tumour mutational burden, and microsatellite instability. TMEM119 was positively correlated with tumour-infiltrating immune cells, tumour microenvironment, mannose receptor C-type I (CD206), and programmed cell death-ligand 1 (PD-L1), while inversely related to nitric oxide synthase 2.ConclusionsTMEM119 may be a potential immune-related biomarker for STAD prognosis and therapeutic targeting.
Collapse
Affiliation(s)
- Yating Liu
- Department of Medical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Xin Yan
- Department of Medical Oncology, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou, China
| | - Caihao Qu
- Department of Medical Oncology, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou, China
| | - Futian Tang
- Department of Medical Oncology, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou, China
| | - Qian Wang
- Department of Medical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Yumin Li
- Department of Medical Oncology, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou, China
| |
Collapse
|
397
|
Ding Y, Yu Y. Therapeutic potential of flavonoids in gastrointestinal cancer: Focus on signaling pathways and improvement strategies (Review). Mol Med Rep 2025; 31:109. [PMID: 40017144 PMCID: PMC11884236 DOI: 10.3892/mmr.2025.13474] [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: 09/18/2024] [Accepted: 01/30/2025] [Indexed: 03/01/2025] Open
Abstract
Flavonoids are a group of polyphenolic compounds distributed in vegetables, fruits and other plants, which have considerable antioxidant, anti‑tumor and anti‑inflammatory activities. Several types of gastrointestinal (GI) cancer are the most common malignant tumors in the world. A large number of studies have shown that flavonoids have inhibitory effects on cancer, and they are recognized as a class of potential anti‑tumor drugs. Therefore, the present review investigated the molecular mechanisms of flavonoids in the treatment of different types of GI cancer and summarized the drug delivery systems commonly used to improve their bioavailability. First, the classification of flavonoids and the therapeutic effects of various flavonoids on human diseases were briefly introduced. Then, to clarify the mechanism of action of flavonoids on different types of GI cancer in the human body, the metabolic process of flavonoids in the human body and the associated signaling pathways causing five common types of GI cancer were discussed, as well as the corresponding therapeutic targets of flavonoids. Finally, in clinical settings, flavonoids have poor water solubility, low permeability and inferior stability, which lead to low absorption efficiency in vivo. Therefore, the three most widely used drug delivery systems were summarized. Suggestions for improving the bioavailability of flavonoids and the focus of the next stage of research were also put forward.
Collapse
Affiliation(s)
- Ye Ding
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yong Yu
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
398
|
Vickers AD. A Comparison of the Performance of 6 Surrogacy Models, Including Weighted Linear Regression, Meta-Regression, and Bivariate Meta-Analysis. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2025; 28:591-598. [PMID: 39880197 DOI: 10.1016/j.jval.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/31/2025]
Abstract
OBJECTIVES Several trial-level surrogate methods have been proposed in the literature. However, often only 1 method is presented in practice. By plotting trial-level associations between surrogate and final outcomes with prediction intervals and by presenting results from cross-validation procedures, this research demonstrates the value of comparing a range of model predictions. METHODS Two oncology data sets were used as examples. One contained 34 trials and had an overall moderate surrogate association; the other contained 14 trials and had an overall strong association. The models fitted included weighted linear regression, meta-regression, and Bayesian bivariate random-effects meta-analysis (BRMA). RESULTS Predictions from the models showed a high degree of variation when there was a moderate association (surrogate threshold effect of 0.413-0.906) and less variation when there was a strong association (surrogate threshold effect of 0.696-0.887). For both data sets, BRMA provided the most robust results, although informative priors for the heterogeneity distribution were needed for the smaller data set. Weighted linear regression models provided reasonable predictions in cases of moderate association. However, in the case of strong association, Bayesian BRMA demonstrated greater uncertainty in predictions. CONCLUSIONS Weighted linear regression provides a useful reference because prediction intervals represent 95% of variance in the data. However, the weights used in such a model must include information on follow-up time. In cases with small data sets, and in cases in which there appeared to be a strong association, Bayesian BRMA provided predictions that were more robust than those provided by weighted linear regression.
Collapse
|
399
|
Song J, Zhu J, Jiang Y, Guo Y, Liu S, Qiao Y, Du Y, Li J. Advancements in immunotherapy for gastric cancer: Unveiling the potential of immune checkpoint inhibitors and emerging strategies. Biochim Biophys Acta Rev Cancer 2025; 1880:189277. [PMID: 39938663 DOI: 10.1016/j.bbcan.2025.189277] [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: 06/10/2024] [Revised: 01/08/2025] [Accepted: 02/04/2025] [Indexed: 02/14/2025]
Abstract
Gastric cancer (GC) is linked to high morbidity and mortality rates. Approximately two-thirds of GC patients are diagnosed at an advanced or metastatic stage. Conventional treatments for GC, including surgery, radiotherapy, and chemotherapy, offer limited prognostic improvement. Recently, immunotherapy has gained attention for its promising therapeutic effects in various tumors. Immunotherapy functions by activating and regulating the patient's immune cells to target and eliminate tumor cells, thereby reducing the tumor burden in the body. Among immunotherapies, immune checkpoint inhibitors (ICIs) are the most advanced. ICIs disrupt the inhibitory protein-small molecule (PD-L1, CTLA4, VISTA, TIM-3 and LAG3) interactions produced by immune cells, reactivating these cells to recognize and attack tumor cells. However, adverse reactions and resistance to ICIs hinder their further clinical and experimental development. Therefore, a comprehensive understanding of the advancements in ICIs for GC is crucial. This article discusses the latest developments in clinical trials of ICIs for GC and examines combination therapies involving ICIs (targeted therapy, chemotherapy, radiotherapy), alongside ongoing clinical trials. Additionally, the review investigates the tumor immune microenvironment and its role in non-responsiveness to ICIs, highlighting the function of tumor immune cells in ICI efficacy. Finally, the article explores the prospects and limitations of new immunotherapy-related technologies, such as tumor vaccines, nanotechnologies, and emerging therapeutic strategies, aiming to advance research into personalized and optimized immunotherapy for patients with locally advanced gastric cancer.
Collapse
Affiliation(s)
- Jiawei Song
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China; Department of Experimental Surgery, Xijing Hospital, Xi'an 710038, China
| | - Jun Zhu
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China
| | - Yu Jiang
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China
| | - Yajie Guo
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China
| | - Shuai Liu
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China
| | - Yihuan Qiao
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China
| | - Yongtao Du
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China
| | - Jipeng Li
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China; Department of Experimental Surgery, Xijing Hospital, Xi'an 710038, China.
| |
Collapse
|
400
|
Wang X, Jiang Y, Yang S, Wang F, Zhang X, Wang W, Chen Y, Wu X, Xiang J, Li Y, Jiang X, Yuan W, Zhang J, Yu KH, Ward RL, Hawkins N, Jonnagaddala J, Li G, Li R. Foundation Model for Predicting Prognosis and Adjuvant Therapy Benefit From Digital Pathology in GI Cancers. J Clin Oncol 2025:JCO2401501. [PMID: 40168636 DOI: 10.1200/jco-24-01501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 12/21/2024] [Accepted: 02/06/2025] [Indexed: 04/03/2025] Open
Abstract
PURPOSE Artificial intelligence (AI) holds significant promise for improving cancer diagnosis and treatment. Here, we present a foundation AI model for prognosis prediction on the basis of standard hematoxylin and eosin-stained histopathology slides. METHODS In this multinational cohort study, we developed AI models to predict prognosis from histopathology images of patients with GI cancers. First, we trained a foundation model using over 130 million patches from 104,876 whole-slide images on the basis of self-supervised learning. Second, we fine-tuned deep learning models for predicting survival outcomes and validated them across seven cohorts, including 1,619 patients with gastric and esophageal cancers and 2,594 patients with colorectal cancer. We further assessed the model for predicting survival benefit from adjuvant chemotherapy. RESULTS The AI models predicted disease-free survival and disease-specific survival with a concordance index of 0.726-0.797 for gastric cancer and 0.714-0.757 for colorectal cancer in the validation cohorts. The models stratified patients into high-risk and low-risk groups, with 5-year survival rates of 49%-52% versus 76%-92% in gastric cancer and 43%-72% versus 81%-98% in colorectal cancer. In multivariable analysis, the AI risk scores remained an independent prognostic factor after adjusting for clinicopathologic variables. Compared with stage alone, an integrated model consisting of stage and image information improved prognosis prediction across all validation cohorts. Finally, adjuvant chemotherapy was associated with improved survival in the high-risk group but not in the low-risk group (treatment-model interaction P = .01 and .006) for stage II/III gastric and colorectal cancer, respectively. CONCLUSION The pathology foundation model can accurately predict survival outcomes and complement clinicopathologic factors in GI cancers. Pending prospective validation, it may be used to improve risk stratification and inform personalized adjuvant therapy.
Collapse
Affiliation(s)
- Xiyue Wang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Yuming Jiang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Sen Yang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Fang Wang
- Department of Pathology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Xiaoming Zhang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Wei Wang
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yijiang Chen
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Xiaoyan Wu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Jinxi Xiang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Yuchen Li
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Xiaofeng Jiang
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
| | - Wei Yuan
- College of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Jing Zhang
- College of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Kun-Hsing Yu
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA
| | - Robyn L Ward
- Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Nicholas Hawkins
- School of Medical Sciences, University of New South Wales, Sydney, Kensington, NSW, Australia
| | | | - Guoxin Li
- School of Clinical Medicine, Tsinghua University, Beijing Tsinghua Changgung Hospital, Beijing, China
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ruijiang Li
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
- Stanford Institute for Human-Centered Artificial Intelligence, Stanford, CA
| |
Collapse
|