101
|
Zhu C, Yang J, Liu L, Li B, Sun T, Sheng W, He Q. Bibliometric analysis of glycolysis and prostate cancer research from 2004 to 2024. Discov Oncol 2025; 16:34. [PMID: 39800812 PMCID: PMC11725561 DOI: 10.1007/s12672-025-01790-2] [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: 08/20/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Prostate cancer (PCa) ranks as the second most common disease among men and the fourth most prevalent cancer worldwide. Enhanced glycolysis and excessive lactate secretion are recognized as critical factors driving the progression of various cancers. This study systematically investigated the research trends associated with glycolysis in PCa through bibliometric analysis. METHOD In this study, we conducted a systematic search of the Web of Science and PubMed databases for literature pertaining to the glycolysis of PCa that was published between January 1, 2004, and June 30, 2024. To achieve this objective, we employed CiteSpace software to generate visualizations that illustrate countries/regions, institutions, journals, and keywords. Additionally, we extracted pertinent quantitative data. Furthermore, we utilized VOSviewer software to create a collaboration network map among various journals. RESULTS Between 2004 and 2024, a total of 408 research articles on glycolysis in PCa were published, indicating a consistent upward trend in the annual publication rate. In this field, the United States not only leads in the volume of research papers but also has the highest degree of centrality. The journal "Cancer Research" is recognized as the most influential in the field, whereas "Prostate and Cancer" serves as a significant platform for disseminating research related to glycolysis in PCa. Keyword analysis has identified four primary research directions that have dominated this field over the past two decades. The role of glycolysis and its associated enzymes in PCa underpins this research. Glycolysis has also demonstrated significant clinical value in the diagnosis and prognosis of PCa. Moreover, drugs targeting glycolytic inhibitors and natural products have exhibited therapeutic potential against this disease. By modulating glycolytic mechanisms, there is potential to increase resistance in PCa. Currently, leading research in this area encompasses the application of nanotechnology to PCa glycolysis, the roles of long noncoding RNAs (lncRNAs) and microRNAs (miRNAs) in this metabolic pathway, and the interactions between glycolysis and other biological processes in PCa. CONCLUSION This study employs bibliometric analysis to provide a comprehensive overview of research on glycolysis in PCa over the past two decades. It highlights the current state of knowledge in this field, identifies key research hotspots, and explores emerging frontiers, particularly nanotechnology, lncRNA, and miRNA, which are driving innovative research directions.
Collapse
Affiliation(s)
- Congxu Zhu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, No. 300 Bachelor's Road, Changsha, 410208, China
| | - Jingjing Yang
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, No. 300 Bachelor's Road, Changsha, 410208, China
| | - Lumei Liu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, No. 300 Bachelor's Road, Changsha, 410208, China
| | - Bonan Li
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, No. 300 Bachelor's Road, Changsha, 410208, China
- Hunan Normal University Affiliated Changsha Hospital, No. 200 North Jinxing Road, Changsha, 410023, China
| | - Tiansong Sun
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, No. 300 Bachelor's Road, Changsha, 410208, China
| | - Wen Sheng
- School of Rehabilitation Medicine and Health Care, Hunan University of Medicine, No. 492 Jinxi South Road, Huaihua, 418000, China.
| | - Qinghu He
- School of Rehabilitation Medicine and Health Care, Hunan University of Medicine, No. 492 Jinxi South Road, Huaihua, 418000, China.
| |
Collapse
|
102
|
Holbrook KL, Lee WY. Volatile Organic Metabolites as Potential Biomarkers for Genitourinary Cancers: Review of the Applications and Detection Methods. Metabolites 2025; 15:37. [PMID: 39852380 PMCID: PMC11767221 DOI: 10.3390/metabo15010037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/26/2025] Open
Abstract
Cancer is one of the leading causes of death globally, and is ranked second in the United States. Early detection is crucial for more effective treatment and a higher chance of survival rates, reducing burdens on individuals and societies. Genitourinary cancers, in particular, face significant challenges in early detection. Finding new and cost-effective diagnostic methods is of clinical need. Metabolomic-based approaches, notably volatile organic compound (VOC) analysis, have shown promise in detecting cancer. VOCs are small organic metabolites involved in biological processes and disease development. They can be detected in urine, breath, and blood samples, making them potential candidates for sensitive and non-invasive alternatives for early cancer detection. However, developing robust VOC detection methods remains a hurdle. This review outlines the current landscape of major genitourinary cancers (kidney, prostate, bladder, and testicular), including epidemiology, risk factors, and current diagnostic tools. Furthermore, it explores the applications of using VOCs as cancer biomarkers, various analytical techniques, and comparisons of extraction and detection methods across different biospecimens. The potential use of VOCs in detection, monitoring disease progression, and treatment responses in the field of genitourinary oncology is examined.
Collapse
Affiliation(s)
| | - Wen-Yee Lee
- Department of Chemistry and Biochemistry, University of Texas at El Paso, El Paso, TX 79968, USA;
| |
Collapse
|
103
|
Ding L, Qian J, Dai R, Zhang H, Miao J, Wang J, Yu M, Tan X, Li Y. The hidden impact: social isolation and inflammation's role in pancreatic cancer risk among those with diabetes. BMC Cancer 2025; 25:58. [PMID: 39794805 PMCID: PMC11720300 DOI: 10.1186/s12885-025-13470-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: 03/26/2024] [Accepted: 01/07/2025] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Pancreatic cancer poses a significant challenge in individuals with diabetes, prompting a reevaluation of established risk factors beyond conventional glycemic control measures. OBJECTIVES To explore the complex interplay of metabolic and psychosocial determinants in pancreatic cancer risk among individuals with diabetes, challenging prevailing perspectives and advocating for a comprehensive approach. METHODS A total of 21,945 UK Biobank participants with baseline diabetes diagnosis were analyzed. Social isolation was assessed through a questionnaire capturing five factors: household size, social activities, friend/family visits, loneliness, and confiding in others. Incident pancreatic cancer was identified using ICD codes. Baseline characteristics, insulin use, and other relevant factors were analyzed. Hazard ratios and mediation analyses were conducted to determine the relationship between social isolation, inflammation, and pancreatic cancer risk. RESULTS Individuals with high social isolation were more likely to be male, smokers, non-drinkers, and have shorter sleep duration. They also had an increased risk of pancreatic cancer (HR = 2.65, 95% CI = 1.12-6.24) compared to those with low social isolation. Mediation analyses highlighted inflammation as a crucial mediator, with the proportion mediated by inflammation being 19.44% for insulin use, 10.34% for smoking, and 8.33% for social isolation. CONCLUSIONS Our findings highlight the importance of psychosocial factors in pancreatic cancer risk and underscore the need for further research to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Lilu Ding
- Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, 481 Binwen Road, Hangzhou, 310053, China
| | - Jing Qian
- Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, 481 Binwen Road, Hangzhou, 310053, China
| | - Ruoqi Dai
- Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, 481 Binwen Road, Hangzhou, 310053, China
| | - Hui Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, 481 Binwen Road, Hangzhou, 310053, China
| | - Jingyou Miao
- Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, 481 Binwen Road, Hangzhou, 310053, China
| | - Jing Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, 481 Binwen Road, Hangzhou, 310053, China
| | - Min Yu
- Zhejiang Provincial Center for Disease Control and Prevention, 3399 Binsheng Road, Hangzhou, 310051, China
| | - Xiao Tan
- Department of Big Data in Health Science, Zhejiang University School of Public Health and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
| | - Yingjun Li
- Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, 481 Binwen Road, Hangzhou, 310053, China.
| |
Collapse
|
104
|
Bi Y, Yv H, Ma X, Chen S. Case report: A successful case of targeted immunotherapy for locally advanced pancreatic cancer under non-surgical conditions. Front Immunol 2025; 15:1519186. [PMID: 39867905 PMCID: PMC11760604 DOI: 10.3389/fimmu.2024.1519186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/23/2024] [Indexed: 01/30/2025] Open
Abstract
Introduction Locally advanced pancreatic cancer (LAPC) is a borderline unresectable malignancy that presents significant treatment challenges. The management of LAPC remains a complex issue, particularly in patients who are not eligible for surgical resection. Case Here, we report the case of a 60-year-old woman diagnosed with LAPC through pathological biopsy who subsequently underwent targeted immunotherapy following the failure of a gemcitabine, oxaliplatin, and S-1 (G&S) chemotherapy regimen. Intervention Based on next-generation sequencing (NGS), the patient's treatment regimen was adjusted to include albumin-bound paclitaxel and capecitabine chemotherapy, along with the PD-1 inhibitor camrelizumab (200 mg/cycle) for six cycles. Throughout the treatment period, the patient consistently declined surgical intervention. Imaging studies, including an upper abdominal computed tomography (CT), revealed the formation of a calcified layer surrounding the cancerous tissue in the pancreatic head. Remarkably, the patient has shown stable disease and no evidence of metastasis since the initiation of targeted immunotherapy. Conclusion This case highlights the potential of targeted immunotherapy for the treatment of LAPC, particularly in non-surgical patients. A personalized approach guided by NGS, combined with immunotherapy, is an effective alternative to traditional treatment strategies for managing this challenging malignancy.
Collapse
Affiliation(s)
- Yuanbo Bi
- Department of Hepatobiliary Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Haotian Yv
- Department of Hepatobiliary Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaopeng Ma
- Department of Oncology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shengxiong Chen
- Department of Hepatobiliary Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
105
|
Shi CW, VanderMeer TJ, Pudusseri A. Metastatic pancreatic cancer now in remission: a case report and literature review. Discov Oncol 2025; 16:26. [PMID: 39786582 PMCID: PMC11717766 DOI: 10.1007/s12672-025-01756-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025] Open
Abstract
Pancreatic cancer is a highly aggressive malignancy with the majority of patients presenting at a late stage with unresectable or metastatic disease. Even with first line treatment, median survival is approximately 11 months in patients with advanced PDAC. This report details the unique case of a patient that presented with peritoneal metastases from an adenocarcinoma of the body of the pancreas, had a remarkable response to palliative chemotherapy and is alive without evidence of disease 12 months following cessation of all active treatment. The initial diagnosis was 4 years ago and extensive resection of the primary was completed 2 years ago. The patient was started on standard FOLFIRINOX chemotherapy regimen, completed 6 cycles, but stopped Oxaliplatin and Irinotecan due to neuropathy and fatigue, on November 5, 2020, and transitioned to 5-fluorouracil (5-FU) and leucovorin. There was radiographic response and a notable decrease in tumor marker CA 19-9. On July 12, 2022 he underwent a multivisceral resection that included a radical anterograde modular pancreatico-splenectomy, partial gastrectomy, and splenic flexure colectomy with primary anastomosis due to tumor involvement of the posterior stomach and splenic flexure. Surgical pathology noted a moderately differentiated, grade 2 tumor staged ypT2 N0 M0. He continued the same adjuvant regimen of 5-FU and leucovorin for approximately 9 months with no new or recurrent disease on imaging. His CA 19-9 decreased within normal range after surgery and has remained within the normal limits. He remains on active surveillance. Overall, barring clear availability for targeted therapies, a metastatic PDAC of the tail may be considered to have a better prognosis than previously considered. FOLFIRINOX is the ideal treatment if the patient has a high-performance status, and PRODIGE 35 recommends 8 minimum cycles. However, in our case, the patient only tolerated 6 cycles and was still highly responsive. Despite a stage IV diagnosis, the primary tumor was resected in order to mitigate the risk for mutation and progression. Although rare, greater hope for patients with PDAC of the tail with favorable tumor biology responsive to FOLFIRINOX may contribute to increased surgical resection rates and improve survival rates.
Collapse
|
106
|
Pian LL, Song MH, Wang TF, Qi L, Peng TL, Xie KP. Identification and analysis of pancreatic intraepithelial neoplasia: opportunities and challenges. Front Endocrinol (Lausanne) 2025; 15:1401829. [PMID: 39839479 PMCID: PMC11746065 DOI: 10.3389/fendo.2024.1401829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 12/17/2024] [Indexed: 01/23/2025] Open
Abstract
Pancreatic intraepithelial neoplasia (PanIN) is the most common precursor lesion of pancreatic ductal adenocarcinoma (PDAC), which has poor prognosis with a short median overall survival of 6-12 months and a low 5-year survival rate of approximately 3%. It is crucial to remove PanIN lesions to prevent the development of invasive PDAC, as PDAC spreads rapidly outside the pancreas. This review aims to provide the latest knowledge on PanIN risk, pathology, cellular origin, genetic susceptibility, and diagnosis, while identifying research gaps that require further investigation in this understudied area of precancerous lesions. PanINs are classified into PanIN 1, PanIN 2, and PanIN 3, with PanIN 3 having the highest likelihood of developing into invasive PDAC. Differentiating between PanIN 2 and PanIN 3 is clinically significant. Genetic alterations found in PDAC are also present in PanIN and increase with the grade of PanIN. Imaging methods alone are insufficient for distinguishing PanIN, necessitating the use of genetic and molecular tests for identification. In addition, metabolomics technologies and miRNAs are playing an increasingly important role in the field of cancer diagnosis, offering more possibilities for efficient identification of PanIN. Although detecting and stratifying the risk of PanIN poses challenges, the combined utilization of imaging, genetics, and metabolomics holds promise for improving patient survival in this field.
Collapse
Affiliation(s)
- Ling-ling Pian
- School of Medicine, The South China University of Technology, Guangzhou, Guangdong, China
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
| | - Mei-hui Song
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
| | - Teng-fei Wang
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, China
| | - Ling Qi
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
| | - Tie-li Peng
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
| | - Ke-ping Xie
- School of Medicine, The South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
107
|
Takemura K, Ikeda K, Miyake H, Sogame Y, Yasuda H, Okada N, Iwata K, Sakagami J, Yamaguchi K, Itoh Y, Umemura A. Epithelial-Mesenchymal Transition Suppression by ML210 Enhances Gemcitabine Anti-Tumor Effects on PDAC Cells. Biomolecules 2025; 15:70. [PMID: 39858464 PMCID: PMC11763895 DOI: 10.3390/biom15010070] [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: 09/27/2024] [Revised: 12/23/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers in the world. Neoadjuvant chemotherapy (NAC) has become a standard treatment for patients scheduled for surgical resection, but the high rate of postoperative recurrence is a critical problem. Optimization of NAC is desirable to reduce postoperative recurrence and achieve long-term survival. However, if a patient's general condition deteriorates due to NAC toxicity, surgical outcomes may be compromised. Therefore, we aimed to identify drug(s) that can be used in combination with gemcitabine (GEM), a drug widely used for the treatment of PDAC, to inhibit distant metastatic recurrence, particularly after surgery. After several screening steps, ML210, a low molecular weight chemical, was found to suppress the epithelial-mesenchymal transition (EMT) in PDAC cells in combination with GEM. Specifically, low dose ML210 in combination with GEM was sufficient for cell migration without apparent toxicity or cell death. Mechanistically, ML210, which was developed as a glutathione peroxidase 4 (GPX4) inhibitor to induce lipid peroxidation, increased the oxidized lipid concentrations in PDAC cells. The oxidization of the cell membrane lipids may suppress EMT, including cell migration. Since EMT is a major malignant phenotype of PDAC, our findings may lead to the advancement of PDAC therapy, especially in the prevention of postoperative recurrence.
Collapse
Affiliation(s)
- Keisuke Takemura
- Department of Pharmacology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (K.T.); (K.I.); (N.O.); (K.I.)
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (H.M.); (Y.S.); (H.Y.); (J.S.); (K.Y.); (Y.I.)
| | - Kyohei Ikeda
- Department of Pharmacology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (K.T.); (K.I.); (N.O.); (K.I.)
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (H.M.); (Y.S.); (H.Y.); (J.S.); (K.Y.); (Y.I.)
| | - Hayato Miyake
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (H.M.); (Y.S.); (H.Y.); (J.S.); (K.Y.); (Y.I.)
| | - Yoshio Sogame
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (H.M.); (Y.S.); (H.Y.); (J.S.); (K.Y.); (Y.I.)
| | - Hiroaki Yasuda
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (H.M.); (Y.S.); (H.Y.); (J.S.); (K.Y.); (Y.I.)
- Saiseikai Shiga Hospital, 2-4-1 Ohashi, Ritto 520-3046, Shiga, Japan
| | - Nobuhiro Okada
- Department of Pharmacology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (K.T.); (K.I.); (N.O.); (K.I.)
| | - Kazumi Iwata
- Department of Pharmacology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (K.T.); (K.I.); (N.O.); (K.I.)
| | - Junichi Sakagami
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (H.M.); (Y.S.); (H.Y.); (J.S.); (K.Y.); (Y.I.)
- Fukuchiyama City Hospital, 231 Atsunaka-cho, Fukuchiyama 620-8505, Kyoto, Japan
| | - Kanji Yamaguchi
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (H.M.); (Y.S.); (H.Y.); (J.S.); (K.Y.); (Y.I.)
| | - Yoshito Itoh
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (H.M.); (Y.S.); (H.Y.); (J.S.); (K.Y.); (Y.I.)
| | - Atsushi Umemura
- Department of Pharmacology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (K.T.); (K.I.); (N.O.); (K.I.)
| |
Collapse
|
108
|
Bloemberg J, van Wees S, Kortman VG, Sakes A. Design of a wasp-inspired biopsy needle capable of self-propulsion and friction-based tissue transport. Front Bioeng Biotechnol 2025; 12:1497221. [PMID: 39834634 PMCID: PMC11743259 DOI: 10.3389/fbioe.2024.1497221] [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: 09/16/2024] [Accepted: 12/02/2024] [Indexed: 01/22/2025] Open
Abstract
Percutaneous pancreatic core biopsy is conclusive but challenging due to large-diameter needles, while smaller-diameter needles used in aspiration methods suffer from buckling and clogging. Inspired by the ovipositor of parasitic wasps, which resists buckling through self-propulsion and prevents clogging via friction-based transport, research has led to the integration of these functionalities into multi-segment needle designs or tissue transport system designs. This study aimed to combine these wasp-inspired functionalities into a single biopsy needle by changing the interconnection of the needle segments. The resulting biopsy needle features six parallel needle segments interconnected by a ring passing through slots along the length of the needle segments, enabling a wasp-inspired reciprocating motion. Actuation employs a cam and follower mechanism for controlled translation of the segments. The needle prototype, constructed from nitinol rods and stainless steel rings, measures 3 mm in outer diameter and 1 mm in inner diameter. Testing in gelatin phantoms demonstrated efficient gelatin core transport (up to 69.9% ± 9.1% transport efficiency) and self-propulsion (0.842 ± 0.042 slip ratio). Future iterations should aim to reduce the outer diameter while maintaining tissue yield. The design offers a promising new avenue for wasp-inspired medical tools, potentially enhancing early pancreatic cancer detection, thus reducing healthcare costs and patient complications.
Collapse
Affiliation(s)
- Jette Bloemberg
- Department of BioMechanical Engineering, Bio-Inspired Technology Group, Faculty of Mechanical Engineering, Delft University of Technology, Delft, Netherlands
| | - Suzanne van Wees
- Department of BioMechanical Engineering, Bio-Inspired Technology Group, Faculty of Mechanical Engineering, Delft University of Technology, Delft, Netherlands
- Department of Biomedical Engineering, Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
| | - Vera G. Kortman
- Department of BioMechanical Engineering, Bio-Inspired Technology Group, Faculty of Mechanical Engineering, Delft University of Technology, Delft, Netherlands
| | - Aimée Sakes
- Department of BioMechanical Engineering, Bio-Inspired Technology Group, Faculty of Mechanical Engineering, Delft University of Technology, Delft, Netherlands
| |
Collapse
|
109
|
Athanasiou A, Kureshi N, Wittig A, Sterner M, Huber R, Palma NA, King T, Schiess R. Biomarker Discovery for Early Detection of Pancreatic Ductal Adenocarcinoma (PDAC) Using Multiplex Proteomics Technology. J Proteome Res 2025; 24:315-322. [PMID: 39699878 PMCID: PMC11705213 DOI: 10.1021/acs.jproteome.4c00752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/27/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
Early detection of pancreatic ductal adenocarcinoma (PDAC) can improve survival but is hampered by the absence of early disease symptoms. Imaging remains key for surveillance but is cumbersome and may lack sensitivity to detect small tumors. CA19-9, the only FDA-approved blood biomarker for PDAC, is insufficiently sensitive and specific to be recommended for surveillance. We aimed to discover a blood-based protein signature to improve PDAC detection in our main target population consisting of stage I or II PDAC patients (n = 75) and various controls including healthy controls (n = 50), individuals at high risk (genetic and familial) for PDAC (n = 47), or those under surveillance for an intraductal papillary mucinous neoplasm (n = 36). Roughly 3000 proteins were measured using Olink multiplex technology and conventional immunoassays. Machine learning combined biomarker candidates into 4- to 6-plex signatures. These signatures significantly (p < 0.001) outperformed CA19-9 with 84% sensitivity at 95% specificity, compared to CA19-9's sensitivity of 53% in the target population. Exploratory analysis was performed in new-onset diabetes (n = 81) and chronic pancreatitis (n = 50) patients. In conclusion, 41 promising biomarker candidates across multiple signatures were identified using proteomics technology and will be further tested in an independent cohort.
Collapse
Affiliation(s)
| | - Natasha Kureshi
- Immunovia
Inc., 26 Forest Street,
Suite 110, Marlborough, Massachusetts 01752, United States
| | - Anja Wittig
- Proteomedix
AG, Wagistrasse 23, CH-8952 Schlieren, Switzerland
| | - Maria Sterner
- Immunovia
AB, Medicon Village,
Scheelevägen 8, SE-223 63 Lund, Sweden
| | - Ramy Huber
- Proteomedix
AG, Wagistrasse 23, CH-8952 Schlieren, Switzerland
| | - Norma A. Palma
- Immunovia
Inc., 26 Forest Street,
Suite 110, Marlborough, Massachusetts 01752, United States
| | - Thomas King
- Immunovia
Inc., 26 Forest Street,
Suite 110, Marlborough, Massachusetts 01752, United States
| | - Ralph Schiess
- Proteomedix
AG, Wagistrasse 23, CH-8952 Schlieren, Switzerland
| |
Collapse
|
110
|
Assawasirisin C, Fagenholz P, Qadan M, Hernandez-Barco Y, Aimprasittichai S, Kambadakone A, Mino-Kenudson M, Ike A, Chen SY, Sheng C, Brugge W, Warshaw AL, Lillemoe KD, Fernández-Del Castillo C. Unraveling the Long-term Natural History of Branch Duct Intraductal Papillary Mucinous Neoplasm: Beyond 10 years. Ann Surg 2025; 281:154-160. [PMID: 39253809 DOI: 10.1097/sla.0000000000006535] [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: 09/11/2024]
Abstract
OBJECTIVE To describe the long-term natural history of branch duct intraductal papillary mucinous neoplasm (BD-IPMN). BACKGROUND The BD-IPMN is a known precursor of pancreatic cancer, yet its long-term natural history is largely unknown. METHODS We retrospectively reviewed patients with BD-IPMN who were followed at the Massachusetts General Hospital for at least 10 years without surgical intervention. Patient and cyst characteristics, development of worrisome features (WF), need for surgery, and malignancy were recorded. The risk of pancreatic cancer in this cohort was compared with the general population by determining the standardized incidence ratio. RESULTS Three hundred sixteen patients with BD-IPMN who were followed for at least 10 years without intervention were identified. The median age was 63 years, and the median follow-up was 13.5 years (range: 10-28.8 years). Median cyst size at diagnosis was 1.2 cm (IQR: 0.8-1.7), was 1.8 cm (IQR: 1.2-2.6) at 10 years, and increased to 2.0 cm (IQR: 1.3-3.0) by the end of surveillance. At the 10-year mark, 24% of patients had WF, and by the end of surveillance, an additional 20% had developed WF or high-risk stigmata. 8.2% of patients developed pancreatic malignancy (high-grade dysplasia or invasive cancer). The standardized incidence ratio for pancreatic cancer was 9.28 (95% CI: 5.82-14.06), with almost two-thirds of invasive cancers occurring within the pancreatic cyst. CONCLUSIONS After 10 years of surveillance for BD-IPMN without intervention, the disease continues to progress, and 1 of every 12 patients will develop malignancy. The risk of pancreatic cancer appears to be 9 times higher than in the comparable age-matched population.
Collapse
Affiliation(s)
- Charnwit Assawasirisin
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Peter Fagenholz
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Motaz Qadan
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Yasmin Hernandez-Barco
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Satita Aimprasittichai
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Avinash Kambadakone
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Mari Mino-Kenudson
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Amarachi Ike
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Sheng-Yin Chen
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Chen Sheng
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - William Brugge
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Andrew L Warshaw
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Keith D Lillemoe
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | | |
Collapse
|
111
|
Saraswat I, Goel A, Gupta J. An In-depth Review on Argemone mexicana in the Management of Liver Health and Liver Cancer. Anticancer Agents Med Chem 2025; 25:24-34. [PMID: 39225208 DOI: 10.2174/0118715206307964240821051756] [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: 03/11/2024] [Revised: 05/25/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION Argemone mexicana, commonly known as the Mexican prickly poppy, has been historically employed in traditional medicine for various ailments, including liver disorders. Given the rising prevalence of liver diseases, including cancer, investigating the potential efficacy of Argemone mexicana in promoting liver health is of paramount importance. This review aims to provide a comprehensive analysis of the existing literature on the hepatoprotective and anticancer properties of Argemone mexicana. METHODOLOGY A systematic literature search was conducted across PubMed, Google Scholar, and relevant botanical and pharmacological databases. Studies from various sources, including in vitro experiments, animal models, and clinical trials, were included in the review. The search focused on articles published up to 2010-2023, encompassing research that explored the botanical characteristics, chemical composition, traditional uses, and pharmacological properties of Argemone mexicana, specifically emphasizing its impact on liver health and cancer. RESULTS The review revealed a wealth of studies highlighting the diverse pharmacological properties of Argemone mexicana. The botanical composition includes compounds with antioxidant and anti-inflammatory potential, suggesting hepatoprotective effects. Studies using in vitro and in vivo models demonstrated promising outcomes regarding liver function improvement and inhibition of liver cancer cell proliferation. While some clinical studies supported the traditional uses of Argemone mexicana, further well-designed trials are warranted to establish its clinical efficacy. CONCLUSION In conclusion, Argemone mexicana shows promise as a natural agent for promoting liver health and combating liver cancer. Bioactive compounds with antioxidant and anti-inflammatory properties suggest potential hepatoprotective effects. However, translating these findings into clinical practice requires further rigorous investigation, including well-designed clinical trials. This review provides a foundation for future research efforts aimed at elucidating the full therapeutic potential of Argemone mexicana in liver health and cancer management.
Collapse
Affiliation(s)
- Istuti Saraswat
- Department of Biotechnology, GLA University, 17km Stone, NH-2 Mathura-Delhi Road Mathura, Chaumuhan, Mathura, Uttar Pradesh, India
| | - Anjana Goel
- Department of Biotechnology, GLA University, 17km Stone, NH-2 Mathura-Delhi Road Mathura, Chaumuhan, Mathura, Uttar Pradesh, India
| | - Jyoti Gupta
- Department of Biotechnology, GLA University, 17km Stone, NH-2 Mathura-Delhi Road Mathura, Chaumuhan, Mathura, Uttar Pradesh, India
| |
Collapse
|
112
|
Coperchini F, Greco A, Petrosino E, Croce L, Teliti M, Marchesi N, Pascale A, Calì B, Pignatti P, Magri F, Uddin M, Rotondi M. Selective anti-CXCR2 receptor blockade by AZD5069 inhibits CXCL8-mediated pro-tumorigenic activity in human thyroid cancer cells in vitro. J Endocrinol Invest 2025; 48:53-65. [PMID: 38900374 PMCID: PMC11729135 DOI: 10.1007/s40618-024-02410-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Thyroid cancer is the most common endocrine malignancy. Current therapies are successful, however some patients progress to therapeutically refractive disease. The immunotherapeutic potential of the CXCL8-chemokine/CXCR2-chemokine-receptor system is currently being explored in numerous human cancers. This study aimed to evaluate if the targeting of CXCR2 by its selective antagonist, AZD5069, could modulate CXCL8-mediated pro-tumorigenic effects in thyroid-cancer (TC) cells in vitro. METHODS Normal human primary thyroid cells (NHT) and TC cell lines TPC-1 (RET/PTC), BCPAP, 8505C and 8305C (BRAFV600e) were treated with AZD5069 (100 pM-10 µM) over a time-course. Viability and proliferation were assessed by WST-1 and crystal violet assays. CXCL8 and CXCR2 mRNA were evaluated by RT-PCR. CXCL8-protein concentrations were measured in cell culture supernatants by ELISA. CXCR2 on cell surface was evaluated by flow-cytometry. Cell-migration was assessed by trans-well-migration chamber-system. RESULTS AZD5069 exerted negligible effects on cell proliferation or viability. AZD5069 significantly reduced CXCR2, (but not CXCL8) mRNAs in all cell types. CXCR2 was reduced on the membrane of some TC cell lines. A significant reduction of the CXCL8 secretion was found in TPC-1 cells (basal-secretion) and NHT (TNFα-induced secretion). AZD5069 significantly reduced basal and CXCL8-induced migration in NHT and different TC cells. CONCLUSIONS Our findings confirm the involvement of the CXCL8/CXCR2-axis in promoting pro-tumorigenic effects in TC cells, further demonstrating its immunotherapeutic significance in human cancer.
Collapse
Affiliation(s)
- F Coperchini
- Department of Internal Medicine and Therapeutics, University of Pavia, Via S. Maugeri 4, 27100, Pavia, Italy
| | - A Greco
- Department of Internal Medicine and Therapeutics, University of Pavia, Via S. Maugeri 4, 27100, Pavia, Italy
| | - E Petrosino
- Department of Internal Medicine and Therapeutics, University of Pavia, Via S. Maugeri 4, 27100, Pavia, Italy
| | - L Croce
- Department of Internal Medicine and Therapeutics, University of Pavia, Via S. Maugeri 4, 27100, Pavia, Italy
- Unit of Endocrinology and Metabolism, Laboratory for Endocrine Disruptors, Istituti Clinici Scientifici Maugeri IRCCS, 27100, Pavia, Italy
| | - M Teliti
- Department of Internal Medicine and Therapeutics, University of Pavia, Via S. Maugeri 4, 27100, Pavia, Italy
- Unit of Endocrinology and Metabolism, Laboratory for Endocrine Disruptors, Istituti Clinici Scientifici Maugeri IRCCS, 27100, Pavia, Italy
| | - N Marchesi
- Unit of Pharmacology, Department of Drug Sciences, University of Pavia, 27100, Pavia, Italy
| | - A Pascale
- Unit of Pharmacology, Department of Drug Sciences, University of Pavia, 27100, Pavia, Italy
| | - B Calì
- Department of General and Minimally Invasive Surgery, Istituti Clinici Scientifici Maugeri IRCCS, 27100, Pavia (PV), Italy
| | - P Pignatti
- Allergy and Immunology Unit, Istituti Clinici Scientifici Maugeri IRCCS, 27100, Pavia, Italy
| | - F Magri
- Department of Internal Medicine and Therapeutics, University of Pavia, Via S. Maugeri 4, 27100, Pavia, Italy
- Unit of Endocrinology and Metabolism, Laboratory for Endocrine Disruptors, Istituti Clinici Scientifici Maugeri IRCCS, 27100, Pavia, Italy
| | - M Uddin
- AstraZeneca Gothenburg, Biopharmaceuticals R&D, Mӧlndal, Sweden
| | - M Rotondi
- Department of Internal Medicine and Therapeutics, University of Pavia, Via S. Maugeri 4, 27100, Pavia, Italy.
- Unit of Endocrinology and Metabolism, Laboratory for Endocrine Disruptors, Istituti Clinici Scientifici Maugeri IRCCS, 27100, Pavia, Italy.
| |
Collapse
|
113
|
Madani SP, Mohseni A, Mirza-Aghazadeh-Attari M, Shahbazian H, Afyouni S, Borhani A, Zandieh G, Laheru D, Kamel IR. Role of volumetric tumor enhancement on CT in predicting overall survival in patients with unresectable pancreatic ductal adenocarcinoma. Clin Imaging 2025; 117:110365. [PMID: 39613522 DOI: 10.1016/j.clinimag.2024.110365] [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: 07/26/2024] [Revised: 11/06/2024] [Accepted: 11/18/2024] [Indexed: 12/01/2024]
Abstract
PURPOSE To assess the utility of volumetric tumor enhancement on CT to predict tumor treatment response and the overall survival (OS) of patients with PDAC undergoing FOLFIRINOX-based systemic chemotherapy. Additionally, we aim to explore the performance of a novel model that incorporates relevant volumetric CT-derived parameters to the established RECIST 1.1 in predicting both treatment response and OS. MATERIAL AND METHODS In this retrospective single-institution study, 127 patients with PDAC who received FOLFIRINOX neoadjuvant chemotherapy between December 2012 and November 2021 were included. Manual volumetric segmentation of the single largest tumor was performed on portal venous phase images. Total and enhancing tumor volumes were calculated. Response by RECIST 1.1 was compared to response by tumor volume and enhancing tumor volume on follow-up CT. RESULTS There was no association between overall survival and RECIST 1.1 (p-value = 0.284), volumetric RECIST (p-value = 0.402), and other volumetric CT variables, except for a percentage reduction in enhancing tumor volume (p-value = 0.043). Using univariate survival analysis for categorical thresholds defined by CART, the percentage change in enhancing tumor volume was associated with OS (p-value = 0.018). There was also a significant association between baseline enhancing tumor volume and OS (p-value <0.0001). Using these two categories, we defined a multivariable model associated with OS (p-value <0.0001). CONCLUSION Percentage reduction in enhancing tumor volume was related to OS in non-surgical PDAC patients treated with FOLFIRINOX chemotherapy and could potentially be incorporated into patient survival prediction models.
Collapse
Affiliation(s)
- Seyedeh Panid Madani
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Alireza Mohseni
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, MD, USA
| | | | - Haneyeh Shahbazian
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Shadi Afyouni
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Ali Borhani
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Ghazal Zandieh
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Daniel Laheru
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Ihab R Kamel
- Department of Radiology, University of Colorado, Aurora, CO, USA.
| |
Collapse
|
114
|
Jain A, Keesari PR, Pulakurthi YS, Katamreddy R, Dhar M, Desai R. Pancreatic Cancer Risk in Prediabetes: A Systematic Meta-analysis Approach. Pancreas 2025; 54:e51-e56. [PMID: 39324961 DOI: 10.1097/mpa.0000000000002391] [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] [Indexed: 09/27/2024]
Abstract
OBJECTIVES Pancreatic cancer and prediabetes pose significant public health challenges. Given the lack of strong evidence we performed a meta-analysis to assess the risk of pancreatic cancer in prediabetes. MATERIALS AND METHODS We performed a thorough search of the major databases over the last 10 years to identify relevant articles. The pooled odds ratio (OR) and hazard ratio (HR) were combined to calculate the effect size (ES). RESULTS We analyzed 5 studies including 5,425,111 prediabetic individuals and 16,096,467 normoglycemic population across 5 countries with a median follow-up of 8.5 years. We identified a noteworthy association between prediabetes and pancreatic cancer, reporting an unadjusted ES of 1.36 (95% confidence interval [CI] 1.05-1.77, P = 0.02) and an adjusted ES of 1.40 (1.23-1.59, P < 0.01). Subgroup analyses by age revealed variations in risk, with studies involving participants aged 60 and above exhibiting a higher ES (ES 1.83, 95% CI 1.28-2.62, P < 0.01). Geographical differences were also observed, with Japanese studies reporting a higher risk (ES 1.89, 95% CI 1.15-3.10, P < 0.01) compared with those from the United States (ES 1.32, 95% CI 1.13-1.53, P < 0.01). CONCLUSIONS We identified 40% higher risk of pancreatic cancer in patients with prediabetes than those with normal blood glucose necessitating urgent attention for further research and policy change.
Collapse
Affiliation(s)
- Akhil Jain
- From the University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | - Meekoo Dhar
- Staten Island University Hospital, Staten Island, NY
| | | |
Collapse
|
115
|
Buderath P, Dang TM, Kimmig R. Cancer-field surgery for endometrial cancer by robotic peritoneal mesometrial resection and targeted compartmental lymphadenectomy (PMMR+TCL). J Gynecol Oncol 2025; 36:e13. [PMID: 38945527 PMCID: PMC11790992 DOI: 10.3802/jgo.2025.36.e13] [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: 09/30/2023] [Revised: 01/18/2024] [Accepted: 05/21/2024] [Indexed: 07/02/2024] Open
Abstract
OBJECTIVE Cancer-field surgery by peritoneal mesometrial resection and targeted compartmental lymphadenectomy (PMMR+TCL) for the treatment of endometrial cancer (EC) aims at optimal locoregional tumor control without the need for adjuvant radiotherapy. In a previous publication we could demonstrate the feasibility of the method and presented encouraging first oncologic data. METHODS Following up our 2021 publication, we present data on the treatment of EC by PMMR+TCL in much larger cohort and with longer follow-up. RESULTS One hundred and thirty-five patients with EC International Federation of Gynecology and Obstetrics (FIGO) I-IV (75.6% FIGO I) underwent cancer field surgery via PMMR+TCL for EC in the years 2016-2023. Mean follow-up in our cohort was 27.5 months (0, 83; 19.7). The procedure was feasible and safe with favorable intra-and postoperative complication rates. Even though 50.4% of patients had an indication for postoperative radiotherapy following national and international guidelines, the rate of postoperative irradiation administered was 10.4%. The overall recurrence rate was 8.1% and we observed 2 (1.5%) isolated locoregional recurrences. CONCLUSION Our results confirm the feasibility and safety of PMMR+TCL in EC patients. Oncologic data are very encouraging and hint at a superior locoregional control without adjuvant irradiation. Larger studies with longer follow-up will be needed to confirm these results.
Collapse
Affiliation(s)
- Paul Buderath
- Department for Gynecology and Obstetrics, University Hospital Essen, Essen, Germany.
| | - Tra My Dang
- Department for Gynecology and Obstetrics, University Hospital Essen, Essen, Germany
| | - Rainer Kimmig
- Department for Gynecology and Obstetrics, University Hospital Essen, Essen, Germany
| |
Collapse
|
116
|
Reni M, Giordano G, Audisio M, Orsi G, Macchini M, Gobba SM, Rapposelli I, Lucenti A, Luchena G, Faloppi L, Zustovich F, Ricci V, Cergnul M, Formica V, Procaccio L, Baccolini V, Briccolani A, Cascinu S, Peretti U. Exploring external validity of chemotherapy for pancreatic ductal adenocarcinoma in real life. Dig Liver Dis 2025; 57:104-110. [PMID: 39003164 DOI: 10.1016/j.dld.2024.06.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/23/2024] [Accepted: 06/25/2024] [Indexed: 07/15/2024]
Abstract
INTRODUCTION Cisplatin, nab-paclitaxel, capecitabine, and gemcitabine (PAXG) regimen activity was assessed in a single institution phase II trial (PACT-19) on pancreatic ductal adenocarcinoma (PDAC). The PACT-31 study explored the external validity of PACT-19 results. MATERIALS AND METHODS Patients aged ≥18 and ≤75 years with KPS ≥70, and PDAC diagnosis receiving PAXG in the participating institutions were eligible and categorized as follows: A) PACT-19; B) PACT-31-HSR; C) PACT-31-non-HSR. With a sample of 175 patients, assuming a target 1-year overall survival of 60 % for metastatic and of 80 % for non-metastatic patients, the trial will be considered successful with the 1-year OS falling into the 95 % CI. RESULTS Data from 68 PACT-19 and 168 PACT-31 patients were retrieved. After 124 events, 1yOS was 52.5 % (95 %CI: 44.6-60.4 %) for metastatic and 80.5 % (95 %CI: 71.9-89.1 %) for non-metastatic patients. Survival overlapped between PACT-19 and PACT-31-HSR (median 17.6 and 17.4 months, p = 0.21) and was significantly shorter in PACT-31-non-HSR (median 11.3 months; p = 0.03). Differences of dose-intensity, use of maintenance therapy, and treatment after progression between PACT-31-HSR and non-HSR were evidenced. DISCUSSION PACT-19 results have external validity. The outcome difference between HSR and non-HSR centers endorses the need of creating a hub-and-spoke network aimed at sharing the expertise on rare-diseases.
Collapse
Affiliation(s)
- Michele Reni
- Department of Medical Oncology IRCCS Ospedale San Raffaele, Vita e Salute University, Via Olgettina 60, 20132 Milan, Italy.
| | - Guido Giordano
- Unit of Medical Oncology and Biomolecular Therapy, Policlinico Ospedali Riuniti di Foggia, Department of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 251, 71122 Foggia, Italy
| | - Marco Audisio
- Department of Oncology, University of Turin, AO Ordine Mauriziano, Medical Oncology Unit, ASL Torino 4, Ivrea, Turin, Italy
| | - Giulia Orsi
- Department of Medical Oncology IRCCS Ospedale San Raffaele, Vita e Salute University, Via Olgettina 60, 20132 Milan, Italy
| | - Marina Macchini
- Department of Medical Oncology IRCCS Ospedale San Raffaele, Vita e Salute University, Via Olgettina 60, 20132 Milan, Italy
| | - Stefania Maria Gobba
- Department of Medical Oncology, ASST-Settelaghi, Ospedale di Circolo, Viale Luigi Borri, 57, 21100 Varese, Italy
| | - Ilario Rapposelli
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via Piero Maroncelli, 40, 47014 Meldola, (FC), Italy
| | - Antonio Lucenti
- U.O.C. Oncologia Medica Ospedale Maria Paternò Arezzo, 97100, Maria Paternò E Arezzo, Ragusa, Italy
| | - Giovanna Luchena
- U.O.C. Oncologia, Ospedale S. Anna ASST-LARIANA, Via Ravona, 1, 22020 San Fermo della Battaglia, Como, Italy
| | - Luca Faloppi
- Medical Oncology Unit, Azienda Sanitaria Territoriale di Macerata, Via Santa Lucia, 2, Macerata, Italy
| | - Fable Zustovich
- Department of Medical Oncology, AULSS 1 Dolomiti, Ospedale S.Martino, V.le Europa, 22, 32100 Belluno, Italy
| | - Vincenzo Ricci
- Medical Oncology Unit, Azienda Ospedaliera di Rilievo Nazionale 'San Pio', Via Dell'Angelo 1, Benevento, Italy
| | - Massimiliano Cergnul
- Ospedale Civile di Legnano, ASST OVEST MILANESE, Via Candiani, 2, 20025 Legnano, Italy
| | - Vincenzo Formica
- Medical Oncology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Viale Oxford, 81, 00133 Roma, Italy
| | - Letizia Procaccio
- Dept of Oncology Veneto Institute of Oncology IOV, IRCCS, Via Gattamelata, 64, 35128 Padova, Italy
| | - Valeria Baccolini
- Department of Medical Oncology IRCCS Ospedale San Raffaele, Vita e Salute University, Via Olgettina 60, 20132 Milan, Italy
| | - Assunta Briccolani
- Department of Medical Oncology IRCCS Ospedale San Raffaele, Vita e Salute University, Via Olgettina 60, 20132 Milan, Italy
| | - Stefano Cascinu
- Department of Medical Oncology IRCCS Ospedale San Raffaele, Vita e Salute University, Via Olgettina 60, 20132 Milan, Italy
| | - Umberto Peretti
- Department of Medical Oncology IRCCS Ospedale San Raffaele, Vita e Salute University, Via Olgettina 60, 20132 Milan, Italy
| |
Collapse
|
117
|
Matsumoto M, Shirai Y, Tsunematsu M, Okui N, Gocho T, Onda S, Furukawa K, Haruki K, Uwagawa T, Ikegami T. Changes in Skeletal Muscle Volume During Preoperative Chemotherapy Affect the Outcome of Pancreatic Cancer. Am Surg 2025; 91:115-125. [PMID: 39180397 DOI: 10.1177/00031348241278021] [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: 08/26/2024]
Abstract
BACKGROUND This study aimed to investigate the effects of changes in clinicopathological factors during preoperative chemotherapy for pancreatic cancer, including skeletal muscle volume, on recurrence and prognosis after pancreatectomy. METHODS Data from 41 patients who underwent resection for pancreatic cancer after preoperative chemotherapy from 2012 to 2021 were retrospectively reviewed. Skeletal muscle volume was substituted for the psoas muscle area (PMA) at the level of the third lumbar vertebra. We investigated the relationship of clinicopathological factors during preoperative chemotherapy with disease-free survival (DFS) and overall survival (OS). The association between clinicopathological factors and a decrease in PMA was investigated. RESULTS In the multivariate analyses for DFS and OS, the factors associated with recurrence were as follows: decrease in PMA (P = 0.003) and the absence of adjuvant therapy (P = 0.03), and the factors associated with poor prognosis were as follows: decrease in PMA (P = 0.04) and the absence of adjuvant therapy (P = 0.008), and the resectability of borderline resectable and unresectable-locally advanced tumors (P = 0.033). All patients with partial response according to the Response Evaluation Criteria in Solid Tumors (version 1.1) had no decrease in PMA (P = 0.01). The proportion of patients with Evans classification ≥ II was significantly higher in the group without a decrease in PMA (P = 0.02). The proportion of patients with an average relative dose intensity of adjuvant therapy ≥0.6 was significantly higher in the group without a decrease in PMA (P = 0.02). CONCLUSION Changes in preoperative skeletal muscle volume during preoperative chemotherapy for pancreatic cancer is a potential predictor of recurrence and prognosis after pancreatectomy.
Collapse
Affiliation(s)
- Michinori Matsumoto
- Department of Surgery, The Jikei University School of Medicine, Minato-ku, Japan
| | - Yoshihiro Shirai
- Department of Surgery, The Jikei University School of Medicine, Minato-ku, Japan
| | - Masashi Tsunematsu
- Department of Surgery, The Jikei University School of Medicine, Minato-ku, Japan
| | - Norimitsu Okui
- Department of Surgery, The Jikei University School of Medicine, Minato-ku, Japan
| | - Takeshi Gocho
- Department of Surgery, The Jikei University School of Medicine, Minato-ku, Japan
| | - Shinji Onda
- Department of Surgery, The Jikei University School of Medicine, Minato-ku, Japan
| | - Kenei Furukawa
- Department of Surgery, The Jikei University School of Medicine, Minato-ku, Japan
| | - Koichiro Haruki
- Department of Surgery, The Jikei University School of Medicine, Minato-ku, Japan
| | - Tadashi Uwagawa
- Department of Surgery, The Jikei University School of Medicine, Minato-ku, Japan
| | - Toru Ikegami
- Department of Surgery, The Jikei University School of Medicine, Minato-ku, Japan
| |
Collapse
|
118
|
Abdelrasoul H. B-Cell Co-culture in the Tumor Microenvironment of Solid Tumors Using Pancreatic Cancer as a Tumor Model. Methods Mol Biol 2025; 2909:225-244. [PMID: 40029525 DOI: 10.1007/978-1-0716-4442-3_15] [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: 03/05/2025]
Abstract
B cells play a diverse role in the tumor microenvironment (TME) of solid tumors, influencing immune responses, tumor progression, and therapeutic outcomes. Depending on their activation state and interactions with other tumor components, they can exert both pro- and anti-tumor functions. They can promote tumor growth by secreting immunosuppressive cytokines or boost antitumor immunity by producing antitumor antibodies, presenting antigens, and activating T cells. Studying the effect of the TME on the phenotype and functionality of B cells is of great importance as their interactions with other TME components, such as cancer cells and stromal cells shape the immune landscape of the tumor. Gaining insight into these interactions sheds light on the mechanisms used by the tumor to evade immune detection. In this chapter, I summarize our current 3D co-culture system designed to investigate the effects of the TME on the functionality of B lymphocytes, using pancreatic ductal adenocarcinoma as a solid tumor model.
Collapse
Affiliation(s)
- Hend Abdelrasoul
- Department of General and Visceral Surgery, University Hospital Ulm, Ulm, Germany.
| |
Collapse
|
119
|
Nagarajan Y, Chandrasekaran N, Deepa Parvathi V. Functionalized Nanomaterials In Pancreatic Cancer Theranostics And Molecular Imaging. ChemistryOpen 2025; 14:e202400232. [PMID: 39434498 PMCID: PMC11726697 DOI: 10.1002/open.202400232] [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: 07/09/2024] [Revised: 08/13/2024] [Indexed: 10/23/2024] Open
Abstract
Pancreatic cancer (PC) is one of the most fatal malignancies in the world. This lethality persists due to lack of effective and efficient treatment strategies. Pancreatic ductal adenocarcinoma (PDAC) is an aggressive epithelial malignancy which has a high incidence rate and contributes to overall cancer fatalities. As of 2022, pancreatic cancer contributes to about 3 % of all cancers globally. Over the years, research has characterised germline predisposition, the origin cell, precursor lesions, genetic alterations, structural alterations, transcriptional changes, tumour heterogeneity, metastatic progression, and the tumour microenvironment, which has improved the understanding of PDAC carcinogenesis. By using molecular-based target therapies, these fundamental advancements support primary prevention, screening, early detection, and treatment. The focus of this review is the use of targeted nanoparticles as an alternative to conventional pancreatic cancer treatment due to the various side effects of the latter. The principles of nanoparticle based cancer therapy is efficient targeting of tumour cells via enhanced permeability and retention (EPR) effects and decrease the chemotherapy side effects due to their non-specificity. To increase the efficiency of existing therapies and modify target nanoparticles, several molecular markers of pancreatic cancer cells have been identified. Thus pancreatic cancer cells can be detected using appropriately functionalized nanoparticles with specific signalling molecules. Once cancer has been identified, these nanoparticles can kill the tumour by inducing hyperthermia, medication delivery, immunotherapy or gene therapy. As potent co-delivery methods for adjuvants and tumor-associated antigens; nanoparticles (NPs) have demonstrated significant promise as delivery vehicles in cancer therapy. This ensures the precise internalization of the functionalized nanoparticle and thus also activates the immune system effectively against tumor cells. This review also discusses the immunological factors behind the uptake of functionalized nanoparticles in cancer therapies. Theranostics, which combine imaging and therapeutic chemicals in a single nanocarrier, are the next generation of medicines. Pancreatic cancer treatment may be revolutionised by the development of a tailored nanocarrier with diagnostic, therapeutic, and imaging capabilities. It is extremely difficult to incorporate various therapeutic modalities into a single nanocarrier without compromising the individual functionalities. Surface modification of nanocarriers with antibodies or proteins will enable to attain multifunctionality which increases the efficiency of pancreatic cancer therapy.
Collapse
Affiliation(s)
- Yoghalakshmi Nagarajan
- Department of Biomedical SciencesFaculty of Biomedical Sciences & TechnologySri Ramachandra Institute of Higher Education and Research (SRIHER)Tamil NaduChennai600116India
| | - Natarajan Chandrasekaran
- Senior Professor & Former DirectorCentre for NanobiotechnologyVellore Institute of Technology (VIT)Vellore Campus, Tiruvalam roadTamil NaduKatpadiVellore 632014
| | - Venkatachalam Deepa Parvathi
- Department of Biomedical SciencesFaculty of Biomedical Sciences & TechnologySri Ramachandra Institute of Higher Education and Research (SRIHER)Tamil NaduChennai600116India
| |
Collapse
|
120
|
Burke L, Hinkson A, Haghnejad V, Jones R, Parker R, Rowe IA. Hepatocellular carcinoma risk scores for non-viral liver disease: A systematic review and meta-analysis. JHEP Rep 2025; 7:101227. [PMID: 39655093 PMCID: PMC11625341 DOI: 10.1016/j.jhepr.2024.101227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 12/12/2024] Open
Abstract
Background & Aims Hepatocellular carcinoma (HCC) risk prediction models may provide a more personalised approach to surveillance for HCC among patients with cirrhosis. This systematic review aims to summarise the performance of HCC prediction models in patients with non-viral chronic liver disease. Method The study was prospectively registered with PROSPERO (ID: CRD42022370078) and reported in accordance with PRISMA guidelines. MEDLINE and Embase databases were searched using a validated search filter for prediction model studies. Two reviewers independently assessed studies for inclusion and risk of bias. Measures of model performance (discrimination and calibration) to assess the risk of HCC at specified time points were identified. A random effects meta-analysis was performed on a subset of studies that reported performance of the same model. Results A total of 7,854 studies were identified. After review, 14 studies with a total of 94,014 participants were included; 45% of patients had viral hepatitis, 27% ALD (alcohol-related liver disease) and 19% MASLD (metabolic dysfunction-associated steatotic liver disease). Follow-up ranged from 15.1-138 months. Only one model was developed using a competing risk approach. Age (7 models) and sex (6 models) were the most frequently included predictors. Model discrimination (AUROC or c-statistic) ranged from 0.61-0.947. Only the 'aMAP' score (age, male sex, albumin, bilirubin, and platelets) had sufficient external validation for quantitative analysis, with a pooled c-statistic of 0.81 (95% CI 0.80-0.83). Calibration was reported in only 9 of 14 studies. All studies were rated at high risk of bias. Conclusion Studies describing risk prediction of HCC in non-viral chronic liver disease are poorly reported, have a high risk of bias and do not account for competing risk events. Patients with ALD and MASLD are underrepresented in development and validation cohorts. These factors remain barriers to the clinical utility and uptake of HCC risk models for those with the most common liver diseases. Impact and implications The recent EASL policy statement emphasises the potential of risk-based surveillance to reduce both hepatocellular carcinoma (HCC)-related deaths and surveillance costs. This study addresses the gap in understanding the performance of current HCC risk models in patients with non-viral liver diseases, reflecting the epidemiological landscape of liver disease in Western countries. In our review of these models we identified several key concerns regarding reporting standards and risk of bias and confirmed that patients with alcohol-related liver disease and metabolic dysfunction-associated steatotic liver disease are underrepresented in model development and validation cohorts. Additionally, most models fail to account for the significant risk of competing events, leading to potential overestimation of true HCC risk. This study highlights these critical issues that may hinder the implementation of risk models in clinical practice and offers key recommendations for future model development studies.
Collapse
Affiliation(s)
- Laura Burke
- Leeds Institute for Medical Research, University of Leeds, Leeds, United Kingdom
- Leeds Liver Unit, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Alexander Hinkson
- Leeds Institute for Medical Research, University of Leeds, Leeds, United Kingdom
| | - Vincent Haghnejad
- Leeds Liver Unit, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
- Department of Hepatology and Gastroenterology, University Hospital of Nancy, Nancy, France
| | - Rebecca Jones
- Leeds Institute for Medical Research, University of Leeds, Leeds, United Kingdom
| | - Richard Parker
- Leeds Institute for Medical Research, University of Leeds, Leeds, United Kingdom
| | - Ian A. Rowe
- Leeds Institute for Medical Research, University of Leeds, Leeds, United Kingdom
- Leeds Liver Unit, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| |
Collapse
|
121
|
Lauri G, Apadula L, Archibugi L, Lazzano P, Ponz de Leon Pisani R, Cobreros M, Tacelli M, Marchegiani G, Crippa S, Falconi M, Arcidiacono PG, Capurso G. Association of Smoking with progression from low-risk to high-risk intraductal papillary mucinous neoplasms and pancreatic cancer. Dig Liver Dis 2025; 57:38-43. [PMID: 38825412 DOI: 10.1016/j.dld.2024.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/29/2024] [Accepted: 05/16/2024] [Indexed: 06/04/2024]
Abstract
BACKGROUND Factors associated with the risk of pancreatic adenocarcinoma (PDAC) may play a role in the development and progression of Intraductal Papillary Mucinous Neoplasms (IPMNs). However, data are limited. AIM To compare exposome factors in three groups of patients with "high or low-risk" IPMNs, as assessed at diagnosis and during a 24-months follow-up, and with PDAC. METHODS Patients were matched (same sex, age ±5) 1:1. Exposure variables were compared across groups using Kruskal-Wallis, ANOVA, or Chi-square tests with Bonferroni correction. RESULTS A total of 151 patients were enrolled in each of the three groups (453 overall). The proportion of current smokers was progressively higher in "low-risk", "high-risk" IPMNs and PDAC patients (8.1 %, 11.2 %, 23.3 %; p = 0.0002). The three groups did not differ in terms of ever or heavy smoking, BMI, history of diabetes, cancer, cholecystectomy or chronic pancreatitis, use of statins or aspirin, and family history of cancer. A history of peptic ulcer was more common in PDAC (7.2 %) than in either "low-risk" (2.0 %) or "high-risk" (2.6%) IPMNs (p = 0.02, not significant after Bonferroni correction). CONCLUSION Active smoking seems associated with the progression of IPMNs to malignancy, and cessation of active smoking might be advised in patients with IPMN.
Collapse
Affiliation(s)
- Gaetano Lauri
- Pancreatico-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Laura Apadula
- Pancreatico-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Livia Archibugi
- Pancreatico-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Pilar Lazzano
- Pancreatico-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ruggero Ponz de Leon Pisani
- Pancreatico-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marina Cobreros
- Digestive Diseases Department, Marques de Valdecilla University Hospital, Instituto de investigacion sanitaria Valdecilla (IDIVAL), Santander, Spain
| | - Matteo Tacelli
- Pancreatico-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giovanni Marchegiani
- Department of Surgical, Oncological and Gastroenterological Sciences (DiSCOG)), University of Padua, Padua, Italy
| | - Stefano Crippa
- Pancreatic Surgery and Transplantation Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Massimo Falconi
- Pancreatic Surgery and Transplantation Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Paolo Giorgio Arcidiacono
- Pancreatico-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Gabriele Capurso
- Pancreatico-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
122
|
Fu X, Ma J, Ma F, Guo S, Wang X, Li Y, Tang Y, Qi J, Zhang W, Ye L. MISP-mediated enhancement of pancreatic cancer growth through the Wnt/β-catenin signaling pathway is suppressed by Fisetin. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167515. [PMID: 39278512 DOI: 10.1016/j.bbadis.2024.167515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 09/04/2024] [Accepted: 09/10/2024] [Indexed: 09/18/2024]
Abstract
Pancreatic cancer is a highly malignant tumor characterized by high mortality and low survival rates. The mitotic interactor and substrate of Plk1 (MISP) is a cancer-associated protein that regulates mitotic spindle localization and is highly expressed in several malignant tumors, contributing to tumor development. However, the function and regulatory mechanisms of MISP in pancreatic cancer remain unclear. In this study, we analyzed RNA sequencing data related to pancreatic cancer from the TCGA and GEO databases, identifying MISP as a potential prognostic marker for the disease. MISP was significantly upregulated in pancreatic cancer cells and tissues compared to normal pancreatic cells and tissues. Notably, in pancreatic cancer cells, high MISP protein expression promoted cell proliferation and growth. Mechanistically, the upregulation of MISP facilitated the nuclear accumulation of β-catenin, thereby activating the Wnt/β-catenin signaling pathway and promoting pancreatic cancer growth. In search of effective inhibitors of MISP expression, we screened an FDA-approved drug library and identified Fisetin as a potential suppressor of MISP expression. Fisetin was found to downregulate the transcription factor MYB, thereby reducing MISP expression. Further experiments demonstrated that Fisetin effectively inhibited the in vitro and in vivo growth of pancreatic cancer by suppressing the MISP/Wnt/β-catenin signaling axis. In summary, our research has identified MISP as a novel therapeutic target in pancreatic cancer and uncovered its associated regulatory mechanisms.
Collapse
Affiliation(s)
- Xueli Fu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jiaqi Ma
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Fangyuan Ma
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Shiman Guo
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xue Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Ye Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yanxin Tang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jingwei Qi
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90001, USA
| | - Weiying Zhang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China.
| | - Lihong Ye
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China.
| |
Collapse
|
123
|
Ma F, Huang J. Nomograph of cancer-specific survival in elderly patients with endometrial cancer based on SEER database. J Obstet Gynaecol Res 2025; 51:e16214. [PMID: 39832800 DOI: 10.1111/jog.16214] [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: 07/31/2024] [Accepted: 01/05/2025] [Indexed: 01/22/2025]
Abstract
OBJECTIVE This study aims to identify prognostic factors for elderly patients with endometrial cancer and to develop a nomogram for predicting cancer-specific survival in this population. METHODS Clinicopathological data of elderly patients diagnosed with endometrial cancer between 2004 and 2015 were extracted from the SEER database. Patients were randomly assigned to either a training cohort or a validation cohort at a ratio of 7:3. Univariate and multivariable Cox regression analyses were performed to identify independent prognostic factors. A nomogram was then constructed based on these factors, and its predictive accuracy and discriminative ability were assessed using the C-index, receiver operating characteristic (ROC), and calibration curve. RESULTS Multivariate analysis identified age, marital status, grade, Federation of International of Gynecology and Obstetrics, surgery, chemotherapy, radiation, and tumor size as independent prognostic factors for elderly patients with endometrial carcinoma. Nomograms derived from these factors demonstrated excellent calibration and discrimination. The C-indexes were 0.83 for the training set and 0.82 for the validation set. The area under the curve (AUC) values for the training set were 0.88, 0.87, and 0.86 at 1, 3, and 5 year respectively. Corresponding AUC values for the validation set were 0.89, 0.86, and 0.86. Calibration curves for both cohorts demonstrated close alignment with the diagonal, indicating robust agreement between nomogram predictions and actual outcomes. CONCLUSION A novel nomogram has been developed for personalized prognosis assessment in elderly patients with endometrial carcinoma, aiming to enhance tailored treatment strategies and clinical management.
Collapse
Affiliation(s)
- Fanghua Ma
- Department of ultrasound, The First People's Hospital of Linping District, Hangzhou, Zhejiang, China
| | - Jiesheng Huang
- Department of ultrasound, The First People's Hospital of Linping District, Hangzhou, Zhejiang, China
| |
Collapse
|
124
|
Gayibov E, Sychra T, Spálenková A, Souček P, Oliverius M. The use of patient-derived xenografts and patient-derived organoids in the search for new therapeutic regimens for pancreatic carcinoma. A review. Biomed Pharmacother 2025; 182:117750. [PMID: 39689516 DOI: 10.1016/j.biopha.2024.117750] [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: 09/06/2024] [Revised: 12/02/2024] [Accepted: 12/08/2024] [Indexed: 12/19/2024] Open
Abstract
Patient-derived organoids (PDOs) and xenografts (PDXs) are powerful tools for personalized medicine in pancreatic cancer (PC) research. This study explores the complementary strengths of PDOs and PDXs in terms of practicality, genetic fidelity, cost, and labor considerations. Among other models like 2D cell cultures, spheroids, cancer-on-chip systems, cell line-derived xenografts (CDX), and genetically engineered mouse models (GEMMs), PDOs and PDXs uniquely balance genetic fidelity and personalized medicine potential, offering distinct advantages over the simplicity of 2D cultures and the advanced, but often resource-intensive, GEMMs and cancer-on-chip systems. PDOs excel in high-throughput drug screening due to their ease of use, lower cost, and shorter experimental timelines. However, they lack a complete tumor microenvironment. Conversely, PDXs offer a more complex microenvironment that closely reflects patient tumors, potentially leading to more clinically relevant results. Despite limitations in size, number of specimens, and engraftment success, PDXs demonstrate significant concordance with patient responses to treatment, highlighting their value in personalized medicine. Both models exhibit significant genetic fidelity, making them suitable for drug sensitivity testing. The choice between PDOs and PDXs depends on the research focus, resource availability, and desired level of microenvironment complexity. PDOs are advantageous for high-throughput screening of a diverse array of potential therapeutic agents due to their relative ease of culture and scalability. PDXs, on the other hand, offer a more physiologically relevant model, allowing for a comprehensive evaluation of drug efficacy and mechanisms of action.
Collapse
Affiliation(s)
- Emin Gayibov
- 3rd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Tomáš Sychra
- 3rd Faculty of Medicine, Charles University, Prague, Czech Republic; Centre of Toxicology and Health Safety, National Institute of Public Health, Prague, Czech Republic; Department of General Surgery, 3rd Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czech Republic
| | - Alžběta Spálenková
- Centre of Toxicology and Health Safety, National Institute of Public Health, Prague, Czech Republic; Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Pavel Souček
- Centre of Toxicology and Health Safety, National Institute of Public Health, Prague, Czech Republic; Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic.
| | - Martin Oliverius
- 3rd Faculty of Medicine, Charles University, Prague, Czech Republic; Department of General Surgery, 3rd Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czech Republic.
| |
Collapse
|
125
|
Du Q, Zhang M, Gao A, He T, Guo M. Epigenetic silencing ZSCAN23 promotes pancreatic cancer growth by activating Wnt signaling. Cancer Biol Ther 2024; 25:2302924. [PMID: 38226836 PMCID: PMC10793710 DOI: 10.1080/15384047.2024.2302924] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/04/2024] [Indexed: 01/17/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most malignant tumor. Zinc finger and SCAN domain-containing protein 23 (ZSCAN23) is a new member of the SCAN domain family. The expression regulation and biological function remain to be elucidated. In this study, we explored the epigenetic regulation and the function of ZSCAN23 in PDAC. ZSCAN23 was methylated in 60.21% (171/284) of PDAC and its expression was regulated by promoter region methylation. The expression of ZSCAN23 inhibited cell proliferation, colony formation, migration, invasion, and induced apoptosis and G1/S phase arrest. ZSCAN23 suppressed Panc10.05 cell xenograft growth in mice. Mechanistically, ZSCAN23 inhibited Wnt signaling by interacting with myosin heavy chain 9 (MYH9) in pancreatic cancer cells. ZSCAN23 is frequently methylated in PDAC and may serve as a detective marker. ZSCAN23 suppresses PDAC cell growth both in vitro and in vivo.
Collapse
Affiliation(s)
- Qian Du
- Department of Gastroenterology and Hepatology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
- Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Meiying Zhang
- Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Aiai Gao
- Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Tao He
- Department of Pathology, Characteristic Medical Center of the Chinese People's Armed Police Force, Tianjin, People's Republic of China
| | - Mingzhou Guo
- Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
- National Key Laboratory of Kidney Diseases, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| |
Collapse
|
126
|
Torres C, Mancinelli G, Chen JWE, Cordoba-Chacon J, Pins D, Saeed S, McKinney R, Castellanos K, Orsi G, Singhal M, Patel A, Acebedo J, Coleman A, Heneche J, Yalagala PCR, Subbaiah PV, Leal C, Grimaldo S, Ortuno FM, Bishehsari F, Grippo PJ. Cell Membrane Fatty Acids and PIPs Modulate the Etiology of Pancreatic Cancer by Regulating AKT. Nutrients 2024; 17:150. [PMID: 39796583 PMCID: PMC11722924 DOI: 10.3390/nu17010150] [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: 10/24/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025] Open
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is one of the worst solid malignancies in regard to outcomes and metabolic dysfunction leading to cachexia. It is alarming that PDAC incidence rates continue to increase and warrant the need for innovative approaches to combat this disease. Due to its relatively slow progression (10-20 years), prevention strategies represent an effective means to improve outcomes. One of the risk factors for many cancers and for pancreatic cancer in particular is diet. Hence, our objective is to understand how a diet rich in ω3 and ω6 polyunsaturated fatty acids affects the progression of this disease. Methods: We investigated polyunsaturated fatty acid (PUFA) effects on disease progression employing both in vitro (PDAC cell lines) and in vivo (EL-Kras and KC mice) approaches. Also, we gathered data from the National Health and Nutrition Examination Survey (NHANES) and the National Cancer Institute (NCI) from 1999 to 2017 for a retrospective observational study. Results: The consumption of PUFAs in a patient population correlates with increased PDAC incidence, particularly when the ω3 intake increases to a lesser extent than ω6. Our data demonstrate dietary PUFAs can be incorporated into plasma membrane lipids affecting PI3K/AKT signaling and support the emergence of membrane-targeted therapies. Moreover, we show that the phospholipid composition of a lipid nanoparticle (LNP) can impact the cell membrane integrity and, ultimately, cell viability after administration of these LNPs. Conclusions: Cancer prevention is impactful particularly for those with very poor prognosis, including pancreatic cancer. Our results point to the importance of dietary intervention in this disease when detected early and the potential to improve the antiproliferative effect of drug efficacy when combined with these regimens in later stages of pancreatic cancer.
Collapse
Affiliation(s)
- Carolina Torres
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto de Investigacion Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| | - Georgina Mancinelli
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Jee-Wei Emily Chen
- Department of Materials Science & Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA; (J.-W.E.C.)
| | - Jose Cordoba-Chacon
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Danielle Pins
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Sara Saeed
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Ronald McKinney
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Karla Castellanos
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | | | - Megha Singhal
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Akshar Patel
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Jose Acebedo
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Adonis Coleman
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Jorge Heneche
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Poorna Chandra Rao Yalagala
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Papasani V. Subbaiah
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Cecilia Leal
- Department of Materials Science & Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA; (J.-W.E.C.)
| | - Sam Grimaldo
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
| | - Francisco M. Ortuno
- Department of Computer Architecture and Computer Technology, University of Granada, 18071 Granada, Spain
| | - Faraz Bishehsari
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Paul J. Grippo
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (G.M.); (S.S.); (R.M.); (A.P.)
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Illinois Chicago, 840 S. Wood Street, CSB 708, Chicago, IL 60612, USA
| |
Collapse
|
127
|
Zhang W, Chen J, Zhang W, Xu M. Advances in Endoscopic Ultrasound in Pancreatic Cancer Screening, Diagnosis, and Palliative Care. Biomedicines 2024; 13:76. [PMID: 39857661 PMCID: PMC11762820 DOI: 10.3390/biomedicines13010076] [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: 12/09/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
Pancreatic cancer is a highly aggressive malignancy with a profoundly poor prognosis. Clinically, the condition most frequently manifests with symptoms including painless jaundice, abdominal discomfort, and back pain. Early diagnosis and the implementation of effective therapeutic strategies are critical for improving patient survival outcomes. However, merely 10-20% of patients are diagnosed at an early stage, with the majority presenting at advanced stages, often with metastasis. Consequently, early detection and intervention are crucial for enhancing prognosis. The widespread adoption of endoscopic ultrasonography (EUS) technology in recent years has significantly enhanced the diagnostic accuracy for pancreatic space-occupying lesions. EUS is increasingly recognized for its pivotal role in alleviating malignant biliary obstruction (MBO), gastric outlet obstruction (GOO), and refractory pain in advanced pancreatic cancer. This article aims to provide an overall review of the current applications of EUS in the diagnosis and treatment of pancreatic cancer, exploring its advantages and limitations in early screening, diagnosis, and palliative care. Furthermore, this review explores potential future directions in the field, aiming to provide valuable insights to inform and enhance the clinical management of pancreatic cancer.
Collapse
Affiliation(s)
- Wenyu Zhang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Jingzheng Chen
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Wei Zhang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
- Department of Gastroenterology, Digestive Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Min Xu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
- Department of Gastroenterology, Digestive Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| |
Collapse
|
128
|
D’Antonio DL, Zenoniani A, Umme S, Piattelli A, Curia MC. Intratumoral Fusobacterium nucleatum in Pancreatic Cancer: Current and Future Perspectives. Pathogens 2024; 14:2. [PMID: 39860963 PMCID: PMC11768203 DOI: 10.3390/pathogens14010002] [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: 12/02/2024] [Revised: 12/22/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
The intratumoral microbiome plays a significant role in many cancers, such as lung, pancreatic, and colorectal cancer. Pancreatic cancer (PC) is one of the most lethal malignancies and is often diagnosed at advanced stages. Fusobacterium nucleatum (Fn), an anaerobic Gram-negative bacterium primarily residing in the oral cavity, has garnered significant attention for its emerging role in several extra-oral human diseases and, lately, in pancreatic cancer progression and prognosis. It is now recognized as oncobacterium. Fn engages in pancreatic tumorigenesis and metastasis through multifaceted mechanisms, including immune response modulation, virulence factors, control of cell proliferation, intestinal metabolite interactions, DNA damage, and epithelial-mesenchymal transition. Additionally, compelling research suggests that Fn may exert detrimental effects on cancer treatment outcomes. This paper extends the perspective to pancreatic cancer associated with Fn. The central focus is to unravel the oncogenomic changes driven by Fn in colonization, initiation, and promotion of pancreatic cancer development. The presence of Fusobacterium species can be considered a prognostic marker of PC, and it is also correlated to chemoresistance. Furthermore, this review underscores the clinical research significance of Fn as a potential tumor biomarker and therapeutic target, offering a novel outlook on its applicability in cancer detection and prognostic assessment. It is thought that given the role of Fn in tumor formation and metastasis processes via its FadA, FapA, Fap2, and RadD, new therapies for tumor treatment targeting Fn will be developed.
Collapse
Affiliation(s)
- Domenica Lucia D’Antonio
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (D.L.D.); (A.Z.); (S.U.)
| | - Anna Zenoniani
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (D.L.D.); (A.Z.); (S.U.)
| | - Samia Umme
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (D.L.D.); (A.Z.); (S.U.)
- Department of Neuroscience, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy
| | - Adriano Piattelli
- School of Dentistry, Saint Camillus International University of Health and Medical Sciences (UniCamillus), 00131 Rome, Italy;
- Facultad de Medicina, UCAM Universidad Católica San Antonio de Murcia, 30107 Murcia, Spain
| | - Maria Cristina Curia
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (D.L.D.); (A.Z.); (S.U.)
| |
Collapse
|
129
|
Du H, Chen HB, Zhao Y. Exploring a new chapter in traditional Chinese medicine: The potential of Calculus bovis in liver cancer treatment. World J Clin Oncol 2024; 15:1520-1527. [PMID: 39720650 PMCID: PMC11514369 DOI: 10.5306/wjco.v15.i12.1520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/19/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024] Open
Abstract
In the ongoing quest for new treatments in medicine, traditional Chinese medicine offers unique insights and potential. Recently, studies on the ability of Calculus bovis to inhibit M2-type tumour-associated macrophage polarisation by modulating the Wnt/β-catenin signalling pathway to suppress liver cancer have undoubtedly revealed new benefits and hope for this field of research. The purpose of this article is to comment on this study and explore its strengths and weaknesses, thereby providing ideas for the future treatment of liver cancer.
Collapse
Affiliation(s)
- Huang Du
- Department of Gastroenterology, Minqing County General Hospital, Fuzhou 350800, Fujian Province, China
| | - Hong-Bin Chen
- Department of Gastroenterology I, Sanming First Hospital, Fujian Medical University, Sanming 365000, Fujian Province, China
| | - Yu Zhao
- Department of Gastroenterology, Hannover Medical School, Carl-Neuberg-Straße 1, Hannover 30625, Lower Saxony, Germany
| |
Collapse
|
130
|
Khoshandam M, Sideris N, Ahmadieh-Yazdi A, Sheykhhasan M, Manoochehri H, Tanzadehpanah H, Mahaki H, Ghadam M, Lak S, Kalhor N, Rabiei M, Al-Musawi S, Dama P. The functional role of LncRNA HOXA-AS2 in multiple human cancers. Pathol Res Pract 2024; 266:155795. [PMID: 39756105 DOI: 10.1016/j.prp.2024.155795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 01/07/2025]
Abstract
Humans have more than 270,000 lncRNAs. Among these, lncRNA HOXA-AS2 is considered a transformative gene involved in various cellular processes, including cell proliferation, apoptosis, migration, and invasion. Thus, it can be regarded as a potential tumor marker for both diagnosis and prognosis. Aberrant expression of lncRNAs is associated with many cancers, including hepatocellular carcinoma (HCC), gallbladder carcinoma (GBC), acute promyelocytic leukemia (APL), lung cancer (LC), prostate cancer (PC), osteosarcoma (OS), colorectal cancer (CRC), cervical cancer (CC), and acute myeloid leukemia (AML). Targeting lncRNAs could be a promising strategy to complement or replace current cancer treatments. As a non-coding oncogene, lncRNA HOXA-AS2 is implicated in multiple cancers and could serve as a potential biomarker for various malignancies. The tumor size and disease stage of several cancers are correlated with HOXA-AS2 expression. Silencing HOXA-AS2 effectively suppresses tumor cell proliferation and promotes apoptosis, thereby inhibiting the progression of multiple cancer types. The regulatory mechanisms of HOXA-AS2 include inducing epithelial-mesenchymal transition (EMT), overexpressing B-cell lymphoma-2 (Bcl-2) and MYC proto-oncogene (c-Myc), gene silencing, activating AKT-MMP signaling pathways, EZH2 and LSD1, and functioning within a competing endogenous RNA (ceRNA) regulatory network by competitively binding miRNAs. This review surveys recent research on the structure, biological functions, abnormal expression, regulatory mechanisms, and diagnostic and therapeutic potential of HOXA-AS2 in various cancers.
Collapse
Affiliation(s)
- Mohadeseh Khoshandam
- Department of Reproductive Biology, Academic Center for Education, Culture and Research, Qom Branch, Qom, Iran
| | - Nikolaos Sideris
- Research Fellow School of Life Sciences, University of Sussex, Brighton, UK.
| | - Amirhossein Ahmadieh-Yazdi
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohsen Sheykhhasan
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
| | - Hamed Manoochehri
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Hamid Tanzadehpanah
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hanie Mahaki
- Vascular & Endovascular Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Ghadam
- National Institute of genetic engineering and biotechnology (NIGEB), Tehran, Iran
| | - Shermin Lak
- National Institute of genetic engineering and biotechnology (NIGEB), Tehran, Iran
| | - Naser Kalhor
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research, Qom, Iran
| | | | | | - Paola Dama
- Research Fellow School of Life Sciences, University of Sussex, Brighton, UK.
| |
Collapse
|
131
|
Alka K, Oyeniyi JF, Mohammad G, Zhao Y, Marcus S, Chinnaiyan P. The RAGE Inhibitor TTP488 (Azeliragon) Demonstrates Anti-Tumor Activity and Enhances the Efficacy of Radiation Therapy in Pancreatic Cancer Cell Lines. Cancers (Basel) 2024; 17:17. [PMID: 39796649 PMCID: PMC11718873 DOI: 10.3390/cancers17010017] [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: 11/28/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025] Open
Abstract
Pancreatic cancer is the third leading cause of cancer-related mortality in the United States, with rising incidence and mortality. The receptor for advanced glycation end products (RAGE) and its ligands significantly contribute to pancreatic cancer progression by enhancing cell proliferation, fostering treatment resistance, and promoting a pro-tumor microenvironment via activation of the nuclear factor-kappa B (NF-κB) signaling pathways. This study validated pathway activation in human pancreatic cancer and evaluated the therapeutic efficacy of TTP488 (Azeliragon), a small-molecule RAGE inhibitor, alone and in combination with radiation therapy (RT) in preclinical models of pancreatic cancer. Human (Panc1) and murine (Pan02) pancreatic cancer cell lines exhibited elevated levels of RAGE and its ligands compared to normal pancreatic tissue. In vitro, Azeliragon inhibited RAGE-mediated NF-κB activation and ligand-mediated cell proliferation in pancreatic cancer cell lines. Target engagement of Azeliragon was confirmed in vivo, as determined by decreased NF-κB activation. Azeliragon demonstrated significant growth delay in mouse models of pancreatic cancer and additive effects when combined with RT. Additionally, Azeliragon modulated the immune suppressive tumor microenvironment in pancreatic cancer by reducing immunosuppressive cells, including M2 macrophages, regulatory T cells, and myeloid-derived suppressor cells, while enhancing CD8+ T cell infiltration. These findings suggest that Azeliragon, by inhibiting RAGE-mediated signaling and modulating immune response, may serve as an effective anti-cancer agent in pancreatic cancer.
Collapse
Affiliation(s)
- Kumari Alka
- Department of Radiation Oncology, Corewell Health William Beaumont University Hospital, Royal Oak, MI 48076, USA; (K.A.); (J.F.O.); (G.M.); (Y.Z.)
| | - Jacob F. Oyeniyi
- Department of Radiation Oncology, Corewell Health William Beaumont University Hospital, Royal Oak, MI 48076, USA; (K.A.); (J.F.O.); (G.M.); (Y.Z.)
| | - Ghulam Mohammad
- Department of Radiation Oncology, Corewell Health William Beaumont University Hospital, Royal Oak, MI 48076, USA; (K.A.); (J.F.O.); (G.M.); (Y.Z.)
| | - Yi Zhao
- Department of Radiation Oncology, Corewell Health William Beaumont University Hospital, Royal Oak, MI 48076, USA; (K.A.); (J.F.O.); (G.M.); (Y.Z.)
| | | | - Prakash Chinnaiyan
- Department of Radiation Oncology, Corewell Health William Beaumont University Hospital, Royal Oak, MI 48076, USA; (K.A.); (J.F.O.); (G.M.); (Y.Z.)
- Radiation Oncology, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
| |
Collapse
|
132
|
Hu CY, Yin YF, Xu DP, Xu Y, Yang JY, Xu YN, Hua R. Construction and validation of immunogenic cell death-related molecular clusters, signature, and immune landscape in pancreatic cancer. Clin Exp Med 2024; 25:19. [PMID: 39708151 DOI: 10.1007/s10238-024-01533-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024]
Abstract
Pancreatic cancer (PC) is a malignancy of the gastrointestinal tract that is characterized by a poor prognosis. This study investigates the roles of immunogenic cell death (ICD) genes in the prognosis and progression of PC. Expression data for PC patients were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets, while ICD genes were sourced from published literature. We explored the expression patterns and identified two distinct clusters based on ICD genes. Kaplan-Meier analysis, differential expression analysis, tumor mutational burden analysis, and immune cell infiltration analysis were performed on these clusters. An ICD gene-based risk model was developed, categorizing samples from the TCGA and GEO datasets into low- and high-risk groups. Additionally, we investigated the expression levels of the genes included in the risk model within the TCGA cohort and our own samples. Finally, a loss-of-function assay was conducted to assess the role of MYD88 in PC. Two clusters of PC samples were identified, patients in the ICD-low cluster exhibited a higher degree of immune cell enrichment. The survival time of patients in the low-risk group was longer than that of those in the high-risk group. The genes included in the risk model (CASP1, MYD88, and PIK3CA) showed upregulated expression levels in tumor samples. Furthermore, the predictive accuracy of our risk model was validated using our own samples. Genetic inhibition of MYD88 led to significantly decreased proliferation and migration of PC cells in the loss-of-function assay. There were disparities in survival time and tumor immune microenvironment (TIME) between two ICD gene clusters. Additionally, we developed an ICD-related risk model that was validated as an independent prognostic indicator for patients with PC.
Collapse
Affiliation(s)
- Cheng-Yu Hu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Yi-Fan Yin
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Da-Peng Xu
- Shanghai Key Laboratory for Cancer Systems Regulation and Clinical Translation, Department of General Surgery, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, People's Republic of China.
| | - Yu Xu
- Department of Hepatopancreatobiliary Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Jian-Yu Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Yan-Nan Xu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Rong Hua
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China.
| |
Collapse
|
133
|
Pham TD, Becker JH, Metropulos AE, Mubin N, Spaulding C, Bentrem DJ, Munshi HG. Regorafenib induces DNA damage and enhances PARP inhibitor efficacy in pancreatic ductal carcinoma. BMC Cancer 2024; 24:1562. [PMID: 39707244 DOI: 10.1186/s12885-024-13334-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND There is increasing interest in enhancing the response of the PARP inhibitor olaparib, which is currently approved for pancreatic ductal adenocarcinoma (PDAC) patients with defects in DNA damage repair associated with germline BRCA1/2 mutations. Moreover, agents that can mimic these defects in the absence of germline BRCA1/2 mutations are an area of active research in hopes of increasing the number of patients eligible for treatment with PARP inhibitors. The extent to which regorafenib, an FDA-approved tyrosine kinase inhibitor, can be used to enhance the efficacy of PARP inhibitors in PDAC cells without known BRCA1/2 mutations remains to be investigated. METHODS Comet assay, cell cycle analysis, western blotting, and immunofluorescent detection of H2AXS139 were used to evaluate the extent to which regorafenib induces DNA damage in PDAC cell lines. The effects of regorafenib, either alone or in combination with PARPi inhibitors, on PDAC cell death were assessed by Annexin V/PI co-staining assay in cell lines and by immunohistochemistry staining for cleaved caspase-3 in mouse tumors and in ex vivo slice cultures of human PDAC tumors. Flow cytometry-based analysis was used to evaluate the ability of regorafenib to reprogram PDAC tumor microenvironment. RESULTS We now show that regorafenib, a tyrosine-kinase inhibitor with efficacy in several gastrointestinal malignancies, can enhance the response of olaparib in pancreatic cancer. While regorafenib induces DNA damage and limits the ability of PDAC cells to resolve the damage, regorafenib by itself does not induce apoptosis. However, regorafenib in combination with olaparib further induces DNA damage in vitro, in tumor-bearing mice, and in ex vivo slice cultures of human PDAC tumors, resulting in increased apoptosis compared to olaparib alone. Notably, we show that the efficacy of the combination treatment is not dependent on cytolytic T cells. CONCLUSIONS Together, these findings demonstrate that regorafenib can attenuate DNA damage response and potentiate the efficacy of PARP inhibitors in PDAC tumors.
Collapse
Affiliation(s)
- Thao D Pham
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- The Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA.
| | - Jeffrey H Becker
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Anastasia E Metropulos
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Nida Mubin
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Christina Spaulding
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - David J Bentrem
- Jesse Brown VA Medical Center, Chicago, IL, USA
- The Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hidayatullah G Munshi
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Jesse Brown VA Medical Center, Chicago, IL, USA.
- The Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA.
| |
Collapse
|
134
|
Bockorny B, Muthuswamy L, Huang L, Hadisurya M, Maria Lim C, Tsai LL, Gill RR, Wei JL, Bullock AJ, Grossman JE, Besaw RJ, Narasimhan S, Tao WA, Perea S, Sawhney MS, Freedman SD, Hildago M, Iliuk A, Muthuswamy SK. A large-scale proteomics resource of circulating extracellular vesicles for biomarker discovery in pancreatic cancer. eLife 2024; 12:RP87369. [PMID: 39693144 DOI: 10.7554/elife.87369] [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] [Indexed: 12/19/2024] Open
Abstract
Pancreatic cancer has the worst prognosis of all common tumors. Earlier cancer diagnosis could increase survival rates and better assessment of metastatic disease could improve patient care. As such, there is an urgent need to develop biomarkers to diagnose this deadly malignancy. Analyzing circulating extracellular vesicles (cEVs) using 'liquid biopsies' offers an attractive approach to diagnose and monitor disease status. However, it is important to differentiate EV-associated proteins enriched in patients with pancreatic ductal adenocarcinoma (PDAC) from those with benign pancreatic diseases such as chronic pancreatitis and intraductal papillary mucinous neoplasm (IPMN). To meet this need, we combined the novel EVtrap method for highly efficient isolation of EVs from plasma and conducted proteomics analysis of samples from 124 individuals, including patients with PDAC, benign pancreatic diseases and controls. On average, 912 EV proteins were identified per 100 µL of plasma. EVs containing high levels of PDCD6IP, SERPINA12, and RUVBL2 were associated with PDAC compared to the benign diseases in both discovery and validation cohorts. EVs with PSMB4, RUVBL2, and ANKAR were associated with metastasis, and those with CRP, RALB, and CD55 correlated with poor clinical prognosis. Finally, we validated a seven EV protein PDAC signature against a background of benign pancreatic diseases that yielded an 89% prediction accuracy for the diagnosis of PDAC. To our knowledge, our study represents the largest proteomics profiling of circulating EVs ever conducted in pancreatic cancer and provides a valuable open-source atlas to the scientific community with a comprehensive catalogue of novel cEVs that may assist in the development of biomarkers and improve the outcomes of patients with PDAC.
Collapse
Affiliation(s)
- Bruno Bockorny
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, United States
- Harvard Medical School, Boston, United States
| | | | - Ling Huang
- Henry Ford Cancer Institute, Detroit, United States
| | - Marco Hadisurya
- Department of Biochemistry, Purdue University West Lafayette, West Lafayette, United States
| | | | - Leo L Tsai
- Harvard Medical School, Boston, United States
- Department of Radiology, Beth Israel Deaconess Medical Center, Boston, United States
| | - Ritu R Gill
- Harvard Medical School, Boston, United States
- Department of Radiology, Beth Israel Deaconess Medical Center, Boston, United States
| | - Jesse L Wei
- Harvard Medical School, Boston, United States
- Department of Radiology, Beth Israel Deaconess Medical Center, Boston, United States
| | - Andrea J Bullock
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, United States
- Harvard Medical School, Boston, United States
| | | | - Robert J Besaw
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, United States
| | | | - Weiguo Andy Tao
- Department of Biochemistry, Purdue University West Lafayette, West Lafayette, United States
| | - Sofia Perea
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, United States
| | - Mandeep S Sawhney
- Harvard Medical School, Boston, United States
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, United States
| | - Steven D Freedman
- Harvard Medical School, Boston, United States
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, United States
| | - Manuel Hildago
- Division of Hematology-Oncology, Weill Cornell Medical College, New York, United States
- New York-Presbyterian Hospital, New York, United States
| | - Anton Iliuk
- Tymora Analytical Operations, West Lafayette, United States
| | | |
Collapse
|
135
|
Ouyang Y, Zhou B, Chu L, Chen X, Hao Q, Lei J. Causal associations of tea consumption on risk of pancreatic adenocarcinoma and the mediating role of vascular endothelial growth factor D levels. Br J Nutr 2024; 132:1503-1512. [PMID: 39501829 DOI: 10.1017/s0007114524002393] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Tea is one of the most widely consumed beverages in the world. However, the association between tea and risk of pancreatic adenocarcinoma remains controversial. This study aimed to investigate the causal relationship between tea consumption and risk of pancreatic adenocarcinoma and to explore their mediating effects. The two-sample Mendelian randomisation (MR) analysis showed an inverse causal relationship between tea intake and pancreatic adenocarcinoma (OR: 0·111 (0·02, 0·85), P < 0·04). To examine the mediating effects, we explored the potential mechanisms by which tea intake reduces the risk of pancreatic adenocarcinoma. Based on the oral bioavailability and drug-like properties in Traditional Chinese Medicine Systems Pharmacology database, we selected the main active ingredients of tea. We screened out the fifteen representative targeted genes by Pharmmapper database, and the gene ontology enrichment analysis showed that these targeted genes were related to vascular endothelial growth factor (VEGF) pathway. The two-step MR analysis of results showed that only VEGF-D played a mediating role, with a mediation ratio of 0·230 (0·066, 0·394). In conclusion, the findings suggest that VEGF-D mediates the effect of tea intake on the risk of pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Yonghao Ouyang
- Research Institute of General Surgery, Jinling Hospital, Nanjing210000, People's Republic of China
| | - Beini Zhou
- Jiangxi Modern polytechnic college, Nanchang330000, People's Republic of China
| | - Lihua Chu
- Jinggangshan University, Ji'an3343000, People's Republic of China
| | - Xin Chen
- Jiangxi University Of Traditional Chinese Medicine, Nanchang330000, People's Republic of China
| | - Qiang Hao
- Research Institute of General Surgery, Jinling Hospital, Nanjing210000, People's Republic of China
| | - Jiajia Lei
- College of Food Science & Project Engineering, Wuhan Polytechnic University, Wuhan430023, People's Republic of China
| |
Collapse
|
136
|
Yang H, Yu P, Gong J. Prognostic biomarker MICAL2 and associates with proliferation, migration and immune infiltration in pancreatic adenocarcinoma. J Appl Genet 2024:10.1007/s13353-024-00919-3. [PMID: 39661267 DOI: 10.1007/s13353-024-00919-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 12/12/2024]
Abstract
To elucidate the crucial function of MICAL2 as a potential immunotherapeutic target and a predictive biomarker in PAAD. The expression of MICAL2 in pan-cancer was investigated using public database, and the expression of MICAL2 in PAAD was validated using tissue samples. The diagnostic and prognostic significance of MICAL2 in PAAD was assessed through the application of ROC curves and Kaplan-Meier curves. The correlation between MICAL2 and infiltrating immune cells and immune checkpoints in PAAD was researched using the TIMER and TCGA databases. In vitro studies involved the evaluation of the biological functions of MICAL2 in human PAAD cells through the knockdown of MICAL2 expression using shRNA. Compared to corresponding normal tissues, the expression of MICAL2 exhibits significant differences in various cancers. Specifically, the level of MICAL2 expression is significantly increased in PAAD. Moreover, MICAL2 demonstrates considerable diagnostic potential in PAAD patients, and its elevated expression is indicative of an unfavorable prognosis. The differential expression of MICAL2 is related to infiltrating immune cells, immune cell markers, and immune checkpoints in PAAD. In ASPC-1 and PANC-1 cells, when MICAL2 was knocked down, there was a notable suppression of proliferation, migration, and invasion. MICAL2 functions as a significant predictor and promising immunotherapeutic target for prognosis assessment in PAAD. It has a pivotal function in fostering the growth and migration of PAAD cells.
Collapse
Affiliation(s)
- Huachao Yang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Department of Breast, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400021, China
| | - Pingping Yu
- Department of Health Management, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Jianping Gong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
137
|
Salgado D, Kang J, Costa AF. Mimics of pancreatic neoplasms at cross-sectional imaging: Pearls for characterization and diagnostic work-up. Curr Probl Diagn Radiol 2024:S0363-0188(24)00230-5. [PMID: 39701879 DOI: 10.1067/j.cpradiol.2024.12.002] [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: 06/06/2024] [Revised: 11/13/2024] [Accepted: 12/09/2024] [Indexed: 12/21/2024]
Abstract
Interpreting imaging examinations of the pancreas can be a challenge. Several different entities can mimic or mask pancreatic neoplasms, including normal anatomic variants, non-pancreatic lesions, and both acute and chronic pancreatitis. It is important to distinguish these entities from pancreatic neoplasms, as the management and prognosis of a pancreatic neoplasm, particularly adenocarcinoma, have considerable impact on patients. Normal pancreatic variants that mimic a focal lesion include focal fatty atrophy, annular pancreas, and ectopic pancreas. Extra-pancreatic lesions that can mimic a primary pancreatic neoplasm include vascular lesions, such as arteriovenous malformations and pseudoaneurysms, duodenal diverticula, and intra-pancreatic accessory spleen. Both acute and chronic pancreatitis can mimic or mask a pancreatic neoplasm and are also associated with pancreatic ductal adenocarcinoma. Awareness of these entities and their imaging features will enable the radiologist to narrow the differential diagnosis, provide recommendations that expedite diagnosis and avoid unnecessary work-up or delays in patient care.
Collapse
Affiliation(s)
- David Salgado
- Department of Diagnostic Radiology, Queen Elizabeth II Health Sciences Centre and Dalhousie University. Victoria General Building, 3rd floor, 1276 South Park Street, Halifax, Nova Scotia B3H 2Y9, Canada.
| | - Jessie Kang
- Department of Diagnostic Radiology, Queen Elizabeth II Health Sciences Centre and Dalhousie University. Victoria General Building, 3rd floor, 1276 South Park Street, Halifax, Nova Scotia B3H 2Y9, Canada.
| | - Andreu F Costa
- Department of Diagnostic Radiology, Queen Elizabeth II Health Sciences Centre and Dalhousie University. Victoria General Building, 3rd floor, 1276 South Park Street, Halifax, Nova Scotia B3H 2Y9, Canada.
| |
Collapse
|
138
|
Harne PS, Harne V, Wray C, Thosani N. Endoscopic innovations in diagnosis and management of pancreatic cancer: a narrative review and future directions. Therap Adv Gastroenterol 2024; 17:17562848241297434. [PMID: 39664230 PMCID: PMC11632891 DOI: 10.1177/17562848241297434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/15/2024] [Indexed: 12/13/2024] Open
Abstract
Pancreatic cancer serves as the third leading cause of cancer-associated morbidity and mortality in the United States, with a 5-year survival rate of only 12% with an expected increase in incidence and mortality in the coming years. Pancreatic ductal adenocarcinomas constitute most pancreatic malignancies. Certain genetic syndromes, including Lynch syndrome, hereditary breast and ovarian cancer syndrome, hereditary pancreatitis, familial adenomatous polyposis, Peutz-Jeghers syndrome, familial pancreatic cancer mutation, and ataxia telangiectasia, confer a significantly higher risk. Screening for pancreatic malignancies currently targets patients with germline mutations or those with significant family history. Screening the general population is not currently viable owing to overall low incidence and lack of specific tests. Endoscopic ultrasound (EUS) and its applied advances are increasingly being used for surveillance, diagnosis, and management of pancreatic malignancies and have now become an indispensable tool in their management. For patients with risk factors, EUS in combination with magnetic resonance imaging/magnetic resonance cholangiopancreatography is used for screening. The role of endoscopic modalities has been expanding with the increased utilization of endoscopic retrograde cholangiopancreatography, EUS-directed therapies include EUS-guided fine-needle aspiration and EUS-fine-needle biopsy (FNB). EUS combined with FNB has the highest specificity and sensitivity for detecting pancreatic cancer amongst available modalities. Studies also recognize that artificial intelligence assisted EUS in the early detection of pancreatic cancer. At the same time, surgical resection has been historically considered the only curative treatment for pancreatic cancer, over 80% of patients present with unresectable disease. We also discuss EUS-guided therapies of physicochemicals (radiofrequency ablation, brachytherapy, and intratumor chemotherapy), biological agents (gene therapies and oncolytic viruses), and immunotherapy. We aim to perform a detailed review of the current burden, risk factors, role of screening, diagnosis, and endoscopic advances in the treatment modalities available for pancreatic cancer.
Collapse
Affiliation(s)
- Prateek Suresh Harne
- Division of Gastroenterology, Allegheny Health Network, Pittsburgh, PA 15212, USA
| | - Vaishali Harne
- Division of Pediatric Gastroenterology, The University of Texas
- Health Science Center and McGovern School of Medicine, Houston, TX, USA
| | - Curtis Wray
- Department of Surgery, The University of Texas Health Science Center and McGovern School of Medicine, Houston, TX, USA
| | - Nirav Thosani
- Department of Surgery and Interventional Gastroenterology, The University of Texas
- Health Science Center and McGovern School of Medicine, Houston, TX, USA
| |
Collapse
|
139
|
Antoniou A, Tatsis D, Papadopoulou S, Pazaitou-Panayiotou K, Vahtsevanos K. Metachronous metastasis of papillary thyroid carcinoma to the parotid gland: a case report and review of the literature. Hormones (Athens) 2024:10.1007/s42000-024-00619-x. [PMID: 39648230 DOI: 10.1007/s42000-024-00619-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 11/26/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND We present a case of metachronous metastasis of papillary thyroid carcinoma to the parotid gland, this being an extremely rare metastasis, and a literature review. CASE REPORT A 56-year-old female patient presented with a history of a slowly growing mass on the left side of the neck. The patient reported a medical history of thyroidectomy due to papillary thyroid carcinoma 23 years previously and neck dissection for lymph node metastases 10 years previously, with additional therapy using radioactive iodine in both cases. Computed tomography (CT) of the neck revealed a single nodular solid mass in the tail of the left parotid gland which showed heterogeneous intake of contrast agent. FNA biopsy of the left parotid gland revealed cells typical of papillary thyroid carcinoma with positive immunochemistry for TTF-1. Due to this new metastasis, a total parotidectomy with preservation of the facial nerve was performed and additional therapy with radioactive iodine was administered. CONCLUSION Despite the fact that papillary thyroid carcinoma has a low incidence of regional and distant metastases, there are a few rare cases with distant metastases reported in the literature. Thus, awareness, especially among endocrinologists, and a multidisciplinary approach are crucial to ensure early detection and efficient treatment of these rare cases, distant metastases being the main cause of mortality and of reduction of overall survival rate among these patients.
Collapse
Affiliation(s)
- Asterios Antoniou
- Oral and Maxillofacial Surgery Department, Aristotle University of Thessaloniki, General Hospital G. Papanikolaou, Papanikolaou Avenue, Thessaloniki, 57010, Greece.
| | - Dimitris Tatsis
- Oral and Maxillofacial Surgery Department, Aristotle University of Thessaloniki, General Hospital G. Papanikolaou, Papanikolaou Avenue, Thessaloniki, 57010, Greece
| | | | | | - Konstantinos Vahtsevanos
- Oral and Maxillofacial Surgery Department, Aristotle University of Thessaloniki, General Hospital G. Papanikolaou, Papanikolaou Avenue, Thessaloniki, 57010, Greece
| |
Collapse
|
140
|
Park MA, Gumpper-Fedus K, Krishna SG, Genilo-Delgado MC, Brantley S, Hart PA, Dillhoff ME, Gomez MF, Basinski TL, Mok SR, Luthra AK, Fleming JB, Mohammadi A, Centeno BA, Jiang K, Karolak A, Jeong D, Chen DT, Stewart PA, Teer JK, Cruz-Monserrate Z, Permuth JB. Molecular Pathway and Immune Profile Analysis of IPMN-Derived Versus PanIN-Derived Pancreatic Ductal Adenocarcinomas. Int J Mol Sci 2024; 25:13164. [PMID: 39684873 DOI: 10.3390/ijms252313164] [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: 09/23/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
Intraductal papillary mucinous neoplasms (IPMN) are commonly detected pancreatic cysts that may transform into pancreatic ductal adenocarcinoma (PDAC). Predicting which IPMNs will progress to PDAC remains a clinical challenge. Moreover, identifying those clinically evident IPMNs for which a surveillance approach is best is a dire clinical need. Therefore, we aimed to identify molecular signatures that distinguished between PDAC with and without clinical evidence of an IPMN to identify novel molecular pathways related to IPMN-derived PDAC that could help guide biomarker development. Data from the Oncology Research Information Exchange Network (ORIEN) multi-institute sequencing project were utilized to analyze 66 PDAC cases from Moffitt Cancer Center and The Ohio State University Wexner Medical Center, for which tumor whole transcriptome sequencing datasets were generated. Cases were classified based on whether a tumor had originated from an IPMN (n = 16) or presumably through the pancreatic intraepithelial neoplasia (PanIN) pathway (n = 50). We then performed differential expression and pathway analysis using Gene-Set Enrichment Analysis (GSEA) and Pathway Analysis with Down-weighted Genes (PADOG) algorithms. We also analyzed immune profiles using the Tumor-Immune Microenvironment Deconvolution web portal for Bulk Transcriptomics (TIMEx). Both GSEA and TIMEx indicate that PanIN-derived PDAC tumors enrich inflammatory pathways (complement, hedgehog signaling, coagulation, inflammatory response, apical surface, IL-2/STAT5, IL-6/STAT3, EMT, KRAS signaling, apical junction, IFN-gamma, allograft rejection) and are comparatively richer in almost all immune cell types than those from IPMN-derived PDAC. IPMN-derived tumors were enriched for metabolic and energy-generating pathways (oxidative phosphorylation, unfolded protein response, pancreas beta cells, adipogenesis, fatty acid metabolism, protein secretion), and the most significantly upregulated genes (padj < 0.001) included mucin 2 (MUC2) and gastrokine-2 (GKN2). Further, the metabolic-linked gene signature enriched in the IPMN-derived samples is associated with a cluster of early-stage and long-survival (top 4th quartile) PDAC cases from The Cancer Genome Atlas (TCGA) expression database. Our data suggest that IPMN-derived and PanIN-derived PDACs differ in the expression of immune profiles and metabolic pathways. These initial findings warrant validation and follow-up to develop biomarker-based strategies for early PDAC detection and treatment.
Collapse
Affiliation(s)
- Margaret A Park
- Department of Gastrointestinal (GI) Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Kristyn Gumpper-Fedus
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Somashekar G Krishna
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Maria C Genilo-Delgado
- Department of Gastrointestinal (GI) Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Stephen Brantley
- Department of Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Phil A Hart
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Mary E Dillhoff
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Maria F Gomez
- Department of Gastrointestinal (GI) Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Toni L Basinski
- Department of Gastrointestinal (GI) Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Shaffer R Mok
- Department of Gastrointestinal (GI) Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Anjuli K Luthra
- Department of Gastrointestinal (GI) Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Jason B Fleming
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Amir Mohammadi
- Department of Gastrointestinal (GI) Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Barbara A Centeno
- Department of Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Kun Jiang
- Department of Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Aleksandra Karolak
- Department of Machine Learning, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Daniel Jeong
- Department of Radiology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Dung-Tsa Chen
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Paul A Stewart
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Jamie K Teer
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Zobeida Cruz-Monserrate
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Jennifer B Permuth
- Department of Gastrointestinal (GI) Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL 33612, USA
| |
Collapse
|
141
|
Qin D, Xi P, Huang K, Jiang L, Yao Z, Wei R, Li S. Nomogram for predicting post-progression-free survival in patients with recurrent pancreatic ductal adenocarcinoma after radical surgery: a retrospective analysis. Front Med (Lausanne) 2024; 11:1486750. [PMID: 39712186 PMCID: PMC11659012 DOI: 10.3389/fmed.2024.1486750] [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: 08/26/2024] [Accepted: 11/20/2024] [Indexed: 12/24/2024] Open
Abstract
Background Radical resection is the only curative method for patients with pancreatic adenocarcinoma (PDAC). However, nearly 85% of PDAC patients suffer from local or distant recurrence within 5 years after curative resection. The progression of recurrent lesions accelerates the mortality rate in PDAC patients. However, the influence of clinicopathological factors on post-progression-free survival (PPFS), defined as the period from tumor recurrence to the timing of the progression of recurrent lesions, has rarely been discussed. The present study aimed to explore the independent prognostic factors for PPFS and construct a nomogram for PPFS prediction. Materials and methods The 200 recurrent PDAC patients were divided into training and validation groups by leave-one-out cross-validation. The patients' clinicopathological characteristics were compared through a chi-square test. Meanwhile, these factors were enrolled in the univariate and multivariate COX regression to find the independent prognostic factors of PPFS. Moreover, the Kaplan-Meier survival analysis based on the independent prognostic factors was performed. Finally, we constructed a nomogram model for PPFS prediction, followed by an effectiveness examination. Results PDAC patients who received multi-agent chemotherapy after surgery showed a longer PPFS than the single-agent chemotherapy group. PDAC patients who received multi-agent chemotherapy after recurrence showed a similar PPFS compared to the single-agent chemotherapy group. Local recurrence with distant metastases, early recurrence, lympho-vascular invasion, higher T stage, and higher N stage predicted shorter PPFS in recurrent PDAC patients. Finally, a nomogram to indicate the progression of recurrent lesions was constructed. Conclusion Multi-agent chemotherapy is recommended for PDAC patients after surgery. Meanwhile, single-agent chemotherapy also deserves consideration after tumor recurrence. Moreover, the nomogram could be used in PPFS prediction.
Collapse
Affiliation(s)
| | | | | | | | | | - Ran Wei
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shengping Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
142
|
Zhuo E, Yang W, Wang Y, Tang Y, Wang W, Zhou L, Chen Y, Li P, Chen B, Gao W, Liu W. Global trends in machine learning applied to clinical research in liver cancer: Bibliometric and visualization analysis (2001-2024). Medicine (Baltimore) 2024; 103:e40790. [PMID: 39654222 PMCID: PMC11631000 DOI: 10.1097/md.0000000000040790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/09/2024] [Accepted: 11/14/2024] [Indexed: 12/12/2024] Open
Abstract
This study explores the intersection of liver cancer and machine learning through bibliometric analysis. The aim is to identify highly cited papers in the field and examine the current research landscape, highlighting emerging trends and key areas of focus in liver cancer and machine learning. By analyzing citation patterns, this study sheds light on the evolving role of machine learning in liver cancer research and its potential for future advancements.
Collapse
Affiliation(s)
- Enba Zhuo
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wenzhi Yang
- First Clinical College, Anhui Medical University, Hefei, China
| | - Yafen Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yanchao Tang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wanrong Wang
- First Clinical College, Anhui Medical University, Hefei, China
| | - Lingyan Zhou
- First Clinical College, Anhui Medical University, Hefei, China
| | - Yanjun Chen
- First Clinical College, Anhui Medical University, Hefei, China
| | - Pengman Li
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Bangjie Chen
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Weimin Gao
- First Clinical College, Anhui Medical University, Hefei, China
| | - Wang Liu
- Department of General Surgery, Sanya Central Hospital (The Third People’s Hospital of Hainan Province), Sanya, China
| |
Collapse
|
143
|
Hashimoto A, Hashimoto S. Plasticity and Tumor Microenvironment in Pancreatic Cancer: Genetic, Metabolic, and Immune Perspectives. Cancers (Basel) 2024; 16:4094. [PMID: 39682280 DOI: 10.3390/cancers16234094] [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: 11/09/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Cancer has long been believed to be a genetic disease caused by the accumulation of mutations in key genes involved in cellular processes. However, recent advances in sequencing technology have demonstrated that cells with cancer driver mutations are also present in normal tissues in response to aging, environmental damage, and chronic inflammation, suggesting that not only intrinsic factors within cancer cells, but also environmental alterations are important key factors in cancer development and progression. Pancreatic cancer tissue is mostly comprised of stromal cells and immune cells. The desmoplasmic microenvironment characteristic of pancreatic cancer is hypoxic and hypotrophic. Pancreatic cancer cells may adapt to this environment by rewiring their metabolism through epigenomic changes, enhancing intrinsic plasticity, creating an acidic and immunosuppressive tumor microenvironment, and inducing noncancerous cells to become tumor-promoting. In addition, pancreatic cancer has often metastasized to local and distant sites by the time of diagnosis, suggesting that a similar mechanism is operating from the precancerous stage. Here, we review key recent findings on how pancreatic cancers acquire plasticity, undergo metabolic reprogramming, and promote immunosuppressive microenvironment formation during their evolution. Furthermore, we present the following two signaling pathways that we have identified: one based on the small G-protein ARF6 driven by KRAS/TP53 mutations, and the other based on the RNA-binding protein Arid5a mediated by inflammatory cytokines, which promote both metabolic reprogramming and immune evasion in pancreatic cancer. Finally, the striking diversity among pancreatic cancers in the relative importance of mutational burden and the tumor microenvironment, their clinical relevance, and the potential for novel therapeutic strategies will be discussed.
Collapse
Affiliation(s)
- Ari Hashimoto
- Department of Molecular Biology, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Shigeru Hashimoto
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0818, Japan
| |
Collapse
|
144
|
Robinson RM, Reyes L, Christopher BN, Duncan RM, Burge RA, Siegel J, Nasarre P, Wang P, O'Bryan JP, Hobbs GA, Klauber-DeMore N, Dolloff NG. A High-Affinity Monoclonal Antibody Against the Pancreatic Ductal Adenocarcinoma Target, Anterior Gradient-2 (AGR2/PDIA17). Antibodies (Basel) 2024; 13:101. [PMID: 39727484 DOI: 10.3390/antib13040101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/07/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Anterior Gradient-2 (AGR2/PDIA17) is a member of the protein disulfide isomerase (PDI) family of oxidoreductases. AGR2 is up-regulated in several solid tumors, including pancreatic ductal adenocarcinoma (PDAC). Given the dire need for new therapeutic options for PDAC patients, we investigated the expression and function of AGR2 in PDAC and developed a novel series of affinity-matured AGR2-specific single-chain variable fragments (scFvs) and monoclonal antibodies. RESULTS We found that AGR2 was expressed in approximately 90% of PDAC but not normal pancreas biopsies, and the level of AGR2 expression correlated with increasing disease stage. AGR2 expression was inversely related to SMAD4 status in PDAC and colorectal cancer cell models and was secreted from cells into their media. In normal tissues, a high density of AGR2 was detected in the epithelium of cells in the digestive tract but was lacking in most other normal tissue systems. The addition of recombinant AGR2 to cell culture and genetic overexpression of AGR2 increased the adhesion, motility, and invasiveness of both human and mouse PDAC cells. Human phage display library screening led to the discovery of multiple AGR2-specific scFv clones that were affinity-matured to produce monoclonal antibody (MAb) clones with low picomolar binding affinity (S31R/A53F/Y). These high-affinity MAbs inhibited AGR2-mediated cell adhesion, migration, and binding to LYPD3, which is a putative cell surface binding partner of AGR2. CONCLUSIONS Our study provides novel, high-affinity, fully human, anti-AGR2 MAbs that neutralize the pro-tumor effects of extracellular AGR2 in PDAC.
Collapse
Affiliation(s)
- Reeder M Robinson
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Leticia Reyes
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Benjamin N Christopher
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ravyn M Duncan
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Rachel A Burge
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Julie Siegel
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Patrick Nasarre
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | - John P O'Bryan
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - G Aaron Hobbs
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Nancy Klauber-DeMore
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
- MUSC Hollings Cancer Center, Charleston, SC 29425, USA
| | - Nathan G Dolloff
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
- MUSC Hollings Cancer Center, Charleston, SC 29425, USA
- Zucker Institute for Innovation Commercialization, Charleston, SC 29425, USA
| |
Collapse
|
145
|
Feng Z, Wang Y, Liang Y, Gu X, Yang Y, Zhang Y, Peng Q. Development and validation of a prognostic risk score model for hepatocellular carcinoma in the Asian population based on immunogenic cell death-related genes. Discov Oncol 2024; 15:744. [PMID: 39630208 PMCID: PMC11618282 DOI: 10.1007/s12672-024-01630-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 11/25/2024] [Indexed: 12/08/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC), the predominant form of liver cancer, is marked by limited therapeutic success and unfavorable prognoses. Its etiology varies regionally, with hepatitis B virus (HBV) being the predominant cause in most of Asia. Immunogenic cell death (ICD), a specific type of cell death, has been extensively linked to HCC treatment in numerous studies. This research aims to explore the significance of ICD-related genes in the Asian HCC cohort, potentially offering novel approaches for HCC management. METHODS We initially obtained transcriptomic and clinical data pertinent to Asian HCC from the TCGA database. Subsequently, we classified the samples into distinct subgroups according to ICD gene expression levels and conducted analyses of the tumor microenvironment and enrichment. Furthermore, we randomly allocated the samples into training and testing cohorts, thereafter developing and validating an ICD gene-based prognostic model tailored for the Asian HCC population. RESULTS The Asian HCC samples were categorized into two subgroups: high and low ICD expression. In the low ICD expression group, we observed diminished infiltration of immune and stromal cells, increased tumor purity, and improved prognosis. Moreover, we devised a 5-gene risk-score prognostic model comprising BAX, CASP8, HMGB1, HSP90AA1, and IL6, demonstrating efficacy in prognostic predictions for the Asian HCC cohort. CONCLUSION Our investigation unveils new perspectives on the influence of ICDs within Asian HCC populations. The derived 5-gene risk-score prognostic model, based on ICDs, not only serves as a tool for assessing prognosis in Asian HCC cases but also suggests potential therapeutic targets for HCC treatment.
Collapse
Affiliation(s)
- Zhengyang Feng
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Yanjie Wang
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yong Liang
- Department of Oncology, The Fifth People's Hospital of Huai'an, Huai'an, China
| | - Xuhao Gu
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yinyin Yang
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yusong Zhang
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Qiliang Peng
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
146
|
Wang Q, Liu J, Yang Z. Global, regional, and national burden of pancreatic cancer from 1990 to 2021, with projections for 25 years: a systematic analysis for the Global Burden of Disease Study 2021. Eur J Cancer Prev 2024:00008469-990000000-00192. [PMID: 39718214 DOI: 10.1097/cej.0000000000000942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
This study examines the global burden of pancreatic cancer from 1990 to 2021 and projects future trends, aiming to provide insights for health policy and resource allocation to mitigate the disease's impact. We assessed the pancreatic cancer burden globally and by subgroups, employing linear regression models to analyze trends from 1990 to 2021. Cluster analysis was used to evaluate burden patterns across Global Burden of Disease regions. Forecasting was conducted using the age-period-cohort model and its Bayesian variant. Additionally, we evaluated risk factor contributions to the pancreatic cancer burden and used frontier analysis to explore the relationship between sociodemographic advancements and cancer rates. In 2021, pancreatic cancer accounted for 508 533 new cases, 439 001 prevalent cases, 505 752 deaths, and 11 316 963 disability-adjusted life years (DALYs). High-risk groups included males and middle-aged to older adults, with high-risk areas identified in regions with higher sociodemographic index (SDI). From 1990 to 2021, both pancreatic cancer cases and age-standardized rates (ASR) increased. Notably, high fasting plasma glucose surpassed tobacco as a leading risk factor for pancreatic cancer. Frontier analysis revealed an inverse relationship between SDI and pancreatic cancer ASR, plateauing at an SDI of 0.60. The global burden of pancreatic cancer continues to rise, with significant disparities across demographic and geographic segments. These findings highlight the need for targeted interventions and resource allocations to address this growing public health challenge.
Collapse
Affiliation(s)
- Qihong Wang
- Department of Digestive Endoscopy, General Hospital of Northern Theater Command, Shenyang, Liaoning Province, China
| | | | | |
Collapse
|
147
|
Perkins CM, Mao Y, Jiang J, Wilkie DJ, Han B, Chen QY, Luesch H, Ali J, Schmittgen TD. Small molecular weight epigenetic inhibitors modulate the extracellular matrix during pancreatic acinar ductal metaplasia. Biochem Biophys Res Commun 2024; 736:150496. [PMID: 39128264 DOI: 10.1016/j.bbrc.2024.150496] [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: 07/24/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/13/2024]
Abstract
The pancreatic ductal adenocarcinoma (PDAC) tumor microenvironment is distinguished by a high degree of fibrosis and inflammation, known as desmoplasia. Desmoplasia increases the stromal deposition and extracellular matrix (ECM) stiffness observed in the tumor microenvironment, contributing to the dampened penetration of pharmacological agents. The molecular and biophysical composition of the ECM during the earliest cellular changes in the development of PDAC, i.e. acinar ductal metaplasia (ADM), has not been extensively explored. We report that the mRNA expression of key protein components of the ECM increases during ADM in p48Cre/+;LSL-KrasG12D (KC) mouse acinar organoids cultured in Matrigel. Treatment of the organoids with small molecular weight epigenetic modulating compounds that inhibit or reverse ADM (largazole, FK228 and chaetocin) dramatically reduced the tissue mRNA expression of collagens, hyaluronan synthase, laminin and fibronectin. The storage moduli, determined by video tracking of fluorescent nanoparticles embedded into the Matrigel, increased during ADM and was reduced following treatment with the epigenetic modulating compounds. We report that the ECM of mouse organoids stiffens during ADM and is further enhanced by the presence of mutant Kras. Moreover, select HDAC and HMT inhibitors reduced the mRNA expression of ECM components and ECM stiffness during inhibition and reversal of ADM, suggesting that these compounds may be useful as adjuvants to enhance the tumor penetration of agents used to treat PDAC.
Collapse
Affiliation(s)
- Corey M Perkins
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA; Florida-California Cancer Research Education and Engagement (CaRE (2)) Health Equity Center, USA
| | - Yating Mao
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL, USA; National High Magnetic Field Laboratory, Tallahassee, FL, USA; Florida-California Cancer Research Education and Engagement (CaRE (2)) Health Equity Center, USA
| | - Jinmai Jiang
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA; Florida-California Cancer Research Education and Engagement (CaRE (2)) Health Equity Center, USA
| | - Diana J Wilkie
- Department of Behavioral Nursing Science, College of Nursing, University of Florida, Gainesville, FL, USA; Florida-California Cancer Research Education and Engagement (CaRE (2)) Health Equity Center, USA
| | - Bo Han
- Department of Surgery, University of Southern California, Los Angeles, CA, USA; Florida-California Cancer Research Education and Engagement (CaRE (2)) Health Equity Center, USA
| | - Qi-Yin Chen
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA; Center for Natural Products, Drug Discovery and Development, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Hendrik Luesch
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA; Center for Natural Products, Drug Discovery and Development, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Jamel Ali
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL, USA; National High Magnetic Field Laboratory, Tallahassee, FL, USA; Florida-California Cancer Research Education and Engagement (CaRE (2)) Health Equity Center, USA.
| | - Thomas D Schmittgen
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA; Florida-California Cancer Research Education and Engagement (CaRE (2)) Health Equity Center, USA.
| |
Collapse
|
148
|
Citarella A, Petrella S, Moi D, Dimasi A, Braga T, Ruberto L, Pieraccini S, Sironi M, Micale N, Schirmeister T, Damia G, Fasano V, Silvani A, Giannini C, Passarella D. Synthesis of α-fluorocinnamate derivatives as novel cathepsin S inhibitors with in vitro antiproliferative activity against pancreatic cancer cells. Bioorg Med Chem 2024; 115:117987. [PMID: 39509759 DOI: 10.1016/j.bmc.2024.117987] [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: 06/24/2024] [Revised: 10/14/2024] [Accepted: 11/02/2024] [Indexed: 11/15/2024]
Abstract
Cathepsins, key members of the papain-like family of cysteine proteases, are crucial for proteolysis processes within human cells, including osteolysis, immunomodulation and apoptosis. Recent research has highlighted the significant role of cathepsins, particularly the L, S, K, and B subtypes, in pancreatic cancer. This has driven the development of novel cathepsin inhibitors as potential treatments to inhibit tumor progression, migration and invasion. Targeting cathepsin S (CatS) has shown promise in reducing tumor progression and enhancing the efficacy of chemotherapeutic agents in preclinical models. Building on our previous work where we employed ethyl p-aminocinnamate ester derivatives for covalent inhibition of cysteine proteases, herein we have designed and synthesized three new derivatives basing on an isosteric replacement (H-F) at the level of cinnamate moiety. These derivatives emerged as potent covalent inhibitors of CatS (1.8-2.6 µM) with 2F showing also weak inhibition activity against CatL (20 %) and CatB (29 %). In vitro assays of 2F against pancreatic cancer cell lines BXPC3 and CAPAN1 revealed significant antiproliferative activity, with IC50 = 5.79 µM and 20.75 µM, respectively. These findings underscore the potential of α-fluorocinnamate-based cysteine protease inhibitors as promising candidates for further development in targeting CatS and CatL with the aim to reduce pancreatic cancer cell proliferation.
Collapse
Affiliation(s)
- Andrea Citarella
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy.
| | - Serena Petrella
- Laboratory of Gynecological Preclinical Oncology, Experimental Oncology Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via M. Negri 2, 20156 Milan, Italy
| | - Davide Moi
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Alessandro Dimasi
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Tommaso Braga
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Lorenzo Ruberto
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Stefano Pieraccini
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Maurizio Sironi
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Nicola Micale
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres 31, I-98166 Messina, Italy
| | - Tanja Schirmeister
- Department of Medicinal Chemistry, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Giovanna Damia
- Laboratory of Gynecological Preclinical Oncology, Experimental Oncology Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via M. Negri 2, 20156 Milan, Italy
| | - Valerio Fasano
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Alessandra Silvani
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Clelia Giannini
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Daniele Passarella
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| |
Collapse
|
149
|
Ni T, Zhao RH, Wu JF, Li CY, Xue G, Lin X. KLK7, KLK10, and KLK11 in Papillary Thyroid Cancer: Bioinformatic Analysis and Experimental Validation. Biochem Genet 2024; 62:4446-4471. [PMID: 38316654 DOI: 10.1007/s10528-024-10679-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 01/02/2024] [Indexed: 02/07/2024]
Abstract
Despite many studies on papillary thyroid carcinoma (PTC) in the past few decades, some critical and significant genes remain undiscovered. To explore genes that may play crucial roles in PTC, a detailed analysis of the expression levels, mutations, and clinical significance of Kallikrein-related peptidases (KLKs) family genes in PTC was undertaken to provide new targets for the precise treatment of the disease. A comprehensive analysis of KLK family genes was performed using various online tools, such as GEPIA, Kaplan-Meier Plotter, LinkedOmics, GSCA, TIMER, and Cluego. KLK7, KLK10, and KLK11 were critical factors of KLK family genes. Then, functional assays were carried out on KLK7/10/11 to determine their proliferation, migration, and invasion capabilities in PTC. The mRNA expression levels of KLK7, KLK10, KLK11, and KLK13 were significantly elevated in thyroid carcinoma, while KLK1, KLK2, KLK3 and KLK4 mRNA levels were decreased compared to normal tissues. Correlations between KLK2/7-12/15 expression levels and tumor stage were also observed in thyroid carcinoma. Survival analysis demonstrated that KLK4/5/7/9-12/14 was associated with overall survival in patients with thyroid cancer. Not only were KLK genes strongly associated with cancer-related pathways, but also KLK7/10/11 was associated with immune-cell infiltration. Finally, silencing KLK7/10/11 impaired human papillary thyroid carcinoma cells' growth, migration ability, and invasiveness. The increased expression of KLK7, KLK10, and KLK11 may serve as molecular markers to identify PTC patients. KLK7, KLK10, and KLK11 could be potential prognostic indicators and targets for precision therapy against PTC.
Collapse
Affiliation(s)
- Tao Ni
- Department of Otorhinolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, China
| | - Ru-Hua Zhao
- Department of Morphology Laboratory, Hebei North University, Zhangjiakou, 075000, China
| | - Jing-Fang Wu
- Department of Morphology Laboratory, Hebei North University, Zhangjiakou, 075000, China
| | - Chao-You Li
- Department of Otorhinolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, China
| | - Gang Xue
- Department of Otorhinolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, China.
| | - Xu Lin
- Department of Morphology Laboratory, Hebei North University, Zhangjiakou, 075000, China.
| |
Collapse
|
150
|
Radlinski MJ, Zaver HB, Shami VM. Diagnostic Endoscopic Ultrasound. Gastroenterol Clin North Am 2024; 53:663-681. [PMID: 39489581 DOI: 10.1016/j.gtc.2024.08.015] [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: 11/05/2024]
Abstract
The last two decades have seen the emergence of endoscopic technologies and techniques allowing for minimally invasive modalities for assessing and sampling lesions inside and outside of the gastrointestinal lumen, including the chest, abdomen, and pelvis. Incorporating these new endoscopic approaches has revolutionized the diagnosis and staging of intraluminal and extraluminal lesions and enabled more accessible and safer tissue acquisition.
Collapse
Affiliation(s)
- Mark J Radlinski
- Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, 1301 Medical Center Drive, 160 TVC, Nashville, TN 37232, USA
| | - Himesh B Zaver
- Division of Gastroenterology and Hepatology, University of Virginia Health System, Box 800708, Charlottesville, VA 22908, USA
| | - Vanessa M Shami
- Division of Gastroenterology and Hepatology, University of Virginia Health System, Box 800708, Charlottesville, VA 22908, USA.
| |
Collapse
|