1
|
Sharma R, Kumar S, Komal K, Ghosh R, Thakur S, Pal RR, Kumar M. Comprehensive insights into pancreatic cancer treatment approaches and cutting-edge nanocarrier solutions: from pathology to nanomedicine. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04094-y. [PMID: 40202672 DOI: 10.1007/s00210-025-04094-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 03/21/2025] [Indexed: 04/10/2025]
Abstract
Pancreatic cancer is one of the most lethal malignancies worldwide. It is characterized by poor prognosis, high mortality, and recurrence rates. Various modifiable and non-modifiable risk factors are associated with pancreatic cancer incidence. Available treatments for pancreatic cancer include surgery, chemotherapy, radiotherapy, photodynamic therapy, supportive care, targeted therapy, and immunotherapy. However, the survival rates for PC are very low. Regrettably, despite efforts to enhance prognosis, the survival rate of pancreatic cancer remains relatively low. Therefore, it is essential to investigate new approaches to improve pancreatic cancer treatment. By synthesizing current knowledge and identifying existing gaps, this article provides a comprehensive overview of risk factors, pathology, conventional treatments, targeted therapies, and recent advancements in nanocarriers for its treatment, along with various clinical trials and patents that justify the safety and efficacy of innovative carriers for drug delivery systems. Ultimately, this review underscores the potential of these innovative formulations to improve outcomes and contribute significantly to the advancement of Pancreatic Cancer treatment. Together, these insights highlight nano-formulations as a promising frontier for effectively treating Pancreatic Cancer.
Collapse
Affiliation(s)
- Rohit Sharma
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Sourabh Kumar
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Kumari Komal
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Rashmi Ghosh
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Shubham Thakur
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Ravi Raj Pal
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Manish Kumar
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India.
| |
Collapse
|
2
|
Jafarnezhad-Ansariha F, Contran N, Cristofori C, Simonato M, Davanzo V, Moz S, Galozzi P, Fogar P, Nordi E, Padoan A, Aita A, Fassan M, Fantin A, Sartori A, Sperti C, Correani A, Carnielli V, Cogo P, Basso D. Cystic Fluid Total Proteins, Low-Density Lipoprotein Cholesterol, Lipid Metabolites, and Lymphocytes: Worrisome Biomarkers for Intraductal Papillary Mucinous Neoplasms. Cancers (Basel) 2025; 17:643. [PMID: 40002238 PMCID: PMC11853297 DOI: 10.3390/cancers17040643] [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: 12/30/2024] [Revised: 02/06/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Objectives: Pancreatic cystic neoplasms (PCNs), particularly intraductal papillary mucinous neoplasms (IPMNs), present a challenge for their potential malignancy. Despite promising biomarkers like CEA, amylase, and glucose, our study investigates whether metabolic indices in blood and cystic fluids (CFs), in addition to lymphocyte subsets and hematopoietic stem/progenitor cells (HSPCs), can effectively differentiate between high- and low-risk PCNs. Materials and Methods: A total of 26 patients (11 males, mean age 69.5 ± 9 years) undergoing Endoscopic Ultrasound-guided Fine Needle Aspiration were consecutively enrolled. Analyses included blood, serum, and CF, assessing glucose, CEA, cholesterol (total, HDL, and LDL), and total proteins. Flow cytometry examined immunophenotyping in peripheral blood and cystic fluids. Mass spectrometry was used for the metabolomic analysis of CF. Sensitivity, specificity, and ROC analyses evaluated discriminatory power. Results: A total of 25 out of 26 patients had IPMN. Patients were categorized as low or high risk based on multidisciplinary evaluation of clinical, radiological, and endoscopic data. High-risk patients showed lower CF total proteins and LDL cholesterol (p = 0.005 and p = 0.031), with a marked reduction in CF lymphocytes (p = 0.005). HSCPs were absent in CF. In blood, high-risk patients showed increased non-MHC-restricted cytotoxic T cells (p = 0.019). The metabolomic analysis revealed significantly reduced middle and long-chain acyl carnitines (AcCa) and tryptophan metabolites in high-risk patients. ROC curves indicated comparable discriminant abilities for CF lymphocytes (AUC 0.868), CF total proteins (AUC 0.859), and CF LDL cholesterol (AUC 0.795). The highest performance was achieved by the AcCa 14:2 and 16:0 (AUC: 0.9221 and 0.8857, respectively). Conclusions: CF levels of glucose, CEA, LDL cholesterol, and total proteins together with lymphocyte counts are easy translational biomarkers that may support risk stratification of PCNs in IPMN patients and might be endorsed by metabolomic analysis. Further studies are required for potential clinical integration.
Collapse
Affiliation(s)
- Fahimeh Jafarnezhad-Ansariha
- Department of Surgery, Oncology and Gastroenterology-DISCOG, University of Padua, 35128 Padua, Italy; (F.J.-A.); (C.S.)
| | - Nicole Contran
- Laboratory Medicine, University-Hospital of Padua, 35128 Padua, Italy; (N.C.); (V.D.); (S.M.); (P.F.); (E.N.); (A.P.); (A.A.); (D.B.)
| | - Chiara Cristofori
- Department of Gastroenterology, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (C.C.); (A.F.)
| | - Manuela Simonato
- Department of Medicine—DIMED, University of Padua, 35128 Padua, Italy; (M.S.); (M.F.)
- Pediatric Research Institute “Citta’ della Speranza”, Critical Care Biology and PCare Laboratories, 35127 Padua, Italy;
| | - Veronica Davanzo
- Laboratory Medicine, University-Hospital of Padua, 35128 Padua, Italy; (N.C.); (V.D.); (S.M.); (P.F.); (E.N.); (A.P.); (A.A.); (D.B.)
| | - Stefania Moz
- Laboratory Medicine, University-Hospital of Padua, 35128 Padua, Italy; (N.C.); (V.D.); (S.M.); (P.F.); (E.N.); (A.P.); (A.A.); (D.B.)
| | - Paola Galozzi
- Department of Medicine—DIMED, University of Padua, 35128 Padua, Italy; (M.S.); (M.F.)
| | - Paola Fogar
- Laboratory Medicine, University-Hospital of Padua, 35128 Padua, Italy; (N.C.); (V.D.); (S.M.); (P.F.); (E.N.); (A.P.); (A.A.); (D.B.)
| | - Evelyn Nordi
- Laboratory Medicine, University-Hospital of Padua, 35128 Padua, Italy; (N.C.); (V.D.); (S.M.); (P.F.); (E.N.); (A.P.); (A.A.); (D.B.)
| | - Andrea Padoan
- Laboratory Medicine, University-Hospital of Padua, 35128 Padua, Italy; (N.C.); (V.D.); (S.M.); (P.F.); (E.N.); (A.P.); (A.A.); (D.B.)
- Department of Medicine—DIMED, University of Padua, 35128 Padua, Italy; (M.S.); (M.F.)
| | - Ada Aita
- Laboratory Medicine, University-Hospital of Padua, 35128 Padua, Italy; (N.C.); (V.D.); (S.M.); (P.F.); (E.N.); (A.P.); (A.A.); (D.B.)
- Department of Medicine—DIMED, University of Padua, 35128 Padua, Italy; (M.S.); (M.F.)
| | - Matteo Fassan
- Department of Medicine—DIMED, University of Padua, 35128 Padua, Italy; (M.S.); (M.F.)
| | - Alberto Fantin
- Department of Gastroenterology, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (C.C.); (A.F.)
| | - Anna Sartori
- Pediatric Research Institute “Citta’ della Speranza”, Critical Care Biology and PCare Laboratories, 35127 Padua, Italy;
| | - Cosimo Sperti
- Department of Surgery, Oncology and Gastroenterology-DISCOG, University of Padua, 35128 Padua, Italy; (F.J.-A.); (C.S.)
| | - Alessio Correani
- Department of Odontostomatologic and Specialized Clinical Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (A.C.); (V.C.)
| | - Virgilio Carnielli
- Department of Odontostomatologic and Specialized Clinical Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (A.C.); (V.C.)
- Division of Neonatology, Mother and Child Department, G. Salesi University Hospital, 60123 Ancona, Italy
| | - Paola Cogo
- Department of Medicine, Division of Pediatrics, S. Maria della Misericordia University Hospital, University of Udine, 33100 Udine, Italy;
| | - Daniela Basso
- Laboratory Medicine, University-Hospital of Padua, 35128 Padua, Italy; (N.C.); (V.D.); (S.M.); (P.F.); (E.N.); (A.P.); (A.A.); (D.B.)
- Department of Medicine—DIMED, University of Padua, 35128 Padua, Italy; (M.S.); (M.F.)
| |
Collapse
|
3
|
Philipson E, Jabbar K, Bratlie SO, Hansson G, Persson J, Vilhav C, Wennerblom J, Sadik R, Naredi P, Bourghardt Fagman J, Engström C. Adjunct mucin biomarkers MUC2+MUC5AC and MUC5AC+PSCA in a clinical setting identify and may improve correct selection of high-risk pancreatic lesions for surgery. HPB (Oxford) 2025; 27:214-221. [PMID: 39562183 DOI: 10.1016/j.hpb.2024.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 10/29/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND Pancreatic cancer has dismal prognosis with a 5-year survival of 12 %. Cystic lesions have been identified as premalignant lesions. The challenge is to identify lesions with high risk of malignant progression, to offer patients prophylactic curative pancreatic surgery. Previous studies have identified mucin biomarker panels (MUCPs) as potential discriminators of pre- and malignant pancreatic cystic lesions. The present study assessed whether MUCPs contribute to more accurate identification of patients with high-risk pancreatic lesions and improve selection for surgery. METHODS This retrospective crossover study included 88 patients referred to endoscopic ultrasound because of unclear pancreatic cystic lesions. Clinical management and surgical decision-making with and without MUCP values were assessed by two expert teams with access to patient medical history, radiology, fine-needle aspirates, cytology, and cystic fluid carcinoembryonic antigen. RESULTS The adjunct of MUCPs improved decision-making in 2 of 21 cases with surgical pathology, identifying one cancer that otherwise would have been missed and sparing one patient from unnecessary surgery. CONCLUSION Access to MUCPs in a clinical setting improved correct selection of high-risk pancreatic lesions for surgery in single cases. A higher number of incorrect recommendations for surgery with the adjunct of MUCPs was also noted, which calls for caution.
Collapse
Affiliation(s)
- Eva Philipson
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Karolina Jabbar
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden; Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Sweden
| | - Svein-Olav Bratlie
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Gunnar Hansson
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Sweden
| | - Jan Persson
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Caroline Vilhav
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Johanna Wennerblom
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Riadh Sadik
- Department of Medicine, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg and Sahlgrenska University Hospital, Sweden
| | - Peter Naredi
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Johan Bourghardt Fagman
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Cecilia Engström
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
4
|
Luan X, Wang X, Bian G, Li X, Gao Z, Liu Z, Zhang Z, Han T, Zhao J, Zhao H, Luan X, Zhu W, Dong L, Guo F. Exosome applications for the diagnosis and treatment of pancreatic ductal adenocarcinoma: An update (Review). Oncol Rep 2025; 53:13. [PMID: 39575479 PMCID: PMC11605277 DOI: 10.3892/or.2024.8846] [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: 08/08/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a malignant neoplasm that typically manifests with subtle clinical manifestations in its early stages and frequently eludes diagnosis until the advanced phases of the disease. The limited therapeutic options available for PDAC significantly contribute to its high mortality rate, highlighting the urgent need for novel biomarkers capable of effectively identifying early clinical manifestations and facilitating precise diagnosis. The pivotal role of cellular exosomes in both the pathogenesis and therapeutic interventions for PDAC has been underscored. Furthermore, researchers have acknowledged the potential of exosomes as targeted drug carriers against regulatory cells in treating PDAC. The present article aims to provide a comprehensive review encompassing recent advancements in utilizing exosomes for elucidating mechanisms underlying disease development, patterns of metastasis, diagnostic techniques and treatment strategies associated with PDAC.
Collapse
Affiliation(s)
- Xinchi Luan
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Xuezhe Wang
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Gang Bian
- Department of Gastroenterology, Affiliated Qingdao Third People's Hospital, Qingdao University, Qingdao, Shandong 266041, P.R. China
| | - Xiaoxuan Li
- Department of Oncology, Key Laboratory of Cancer Molecular and Translational Research, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266031, P.R. China
| | - Ziru Gao
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Zijiao Liu
- School of Clinical and Basic Medicine and Institute of Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Zhishang Zhang
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Tianyue Han
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Jinpeng Zhao
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Hongjiao Zhao
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Xinyue Luan
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Wuhui Zhu
- Department of Hepatobiliary surgery, Affiliated Qingdao Third People's Hospital, Qingdao University, Qingdao, Shandong 266041, P.R. China
| | - Lili Dong
- Department of Gastroenterology, Affiliated Qingdao Third People's Hospital, Qingdao University, Qingdao, Shandong 266041, P.R. China
| | - Feifei Guo
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| |
Collapse
|
5
|
Chen C, Li P, Fan G, Yang E, Jing S, Shi Y, Gong Y, Zhang L, Wang Z. Role of TRIP13 in human cancer development. Mol Biol Rep 2024; 51:1088. [PMID: 39436503 DOI: 10.1007/s11033-024-10012-x] [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: 08/21/2024] [Accepted: 10/11/2024] [Indexed: 10/23/2024]
Abstract
As an AAA + ATPase, thyroid hormone receptor interacting protein 13 (TRIP13) primarily functions in DNA double-strand break repair, chromosome recombination, and cell cycle checkpoint regulation; aberrant expression of TRIP13 can result in chromosomal instability (CIN). According to recent research, TRIP13 is aberrantly expressed in a variety of cancers, and a patient's poor prognosis and tumor stage are strongly correlated with high expression of TRIP13. Tumor cell and subcutaneous xenograft growth can be markedly inhibited by TRIP13 knockdown or TRIP13 inhibitor administration. In the initiation and advancement of human malignancies, TRIP13 seems to function as an oncogene. Based on available data, TRIP13 may function as a biological target and biomarker for cancer. The creation of inhibitors that specifically target TRIP13 may present novel approaches to treating cancer.
Collapse
Affiliation(s)
- Chaohu Chen
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Pan Li
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Guangrui Fan
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Enguang Yang
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Suoshi Jing
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Yibo Shi
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Yuwen Gong
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Luyang Zhang
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Zhiping Wang
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China.
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China.
| |
Collapse
|
6
|
Dong F, Yan J, Zhang X, Zhang Y, Liu D, Pan X, Xue L, Liu Y. Artificial intelligence-based predictive model for guidance on treatment strategy selection in oral and maxillofacial surgery. Heliyon 2024; 10:e35742. [PMID: 39170321 PMCID: PMC11336844 DOI: 10.1016/j.heliyon.2024.e35742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/27/2024] [Accepted: 08/02/2024] [Indexed: 08/23/2024] Open
Abstract
Application of deep learning (DL) and machine learning (ML) is rapidly increasing in the medical field. DL is gaining significance for medical image analysis, particularly, in oral and maxillofacial surgeries. Owing to the ability to accurately identify and categorize both diseased and normal soft- and hard-tissue structures, DL has high application potential in the diagnosis and treatment of tumors and in orthognathic surgeries. Moreover, DL and ML can be used to develop prediction models that can aid surgeons to assess prognosis by analyzing the patient's medical history, imaging data, and surgical records, develop more effective treatment strategies, select appropriate surgical modalities, and evaluate the risk of postoperative complications. Such prediction models can play a crucial role in the selection of treatment strategies for oral and maxillofacial surgeries. Their practical application can improve the utilization of medical staff, increase the treatment accuracy and efficiency, reduce surgical risks, and provide an enhanced treatment experience to patients. However, DL and ML face limitations, such as data drift, unstable model results, and vulnerable social trust. With the advancement of social concepts and technologies, the use of these models in oral and maxillofacial surgery is anticipated to become more comprehensive and extensive.
Collapse
Affiliation(s)
- Fanqiao Dong
- School of Stomatology, China Medical University, Shenyang, China
| | - Jingjing Yan
- Hospital of Stomatology, China Medical University, Shenyang, China
| | - Xiyue Zhang
- School of Stomatology, China Medical University, Shenyang, China
| | - Yikun Zhang
- School of Stomatology, China Medical University, Shenyang, China
| | - Di Liu
- School of Stomatology, China Medical University, Shenyang, China
| | - Xiyun Pan
- School of Stomatology, China Medical University, Shenyang, China
| | - Lei Xue
- School of Stomatology, China Medical University, Shenyang, China
- Hospital of Stomatology, China Medical University, Shenyang, China
| | - Yu Liu
- First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
7
|
Șolea SF, Brisc MC, Orășeanu A, Venter FC, Brisc CM, Șolea RM, Davidescu L, Venter A, Brisc C. Revolutionizing the Pancreatic Tumor Diagnosis: Emerging Trends in Imaging Technologies: A Systematic Review. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:695. [PMID: 38792878 PMCID: PMC11122838 DOI: 10.3390/medicina60050695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/19/2024] [Accepted: 04/21/2024] [Indexed: 05/26/2024]
Abstract
Background and Objectives: The pancreas, ensconced within the abdominal cavity, requires a plethora of sophisticated imaging modalities for its comprehensive evaluation, with ultrasonography serving as a primary investigative technique. A myriad of pancreatic pathologies, encompassing pancreatic neoplasia and a spectrum of inflammatory diseases, are detectable through these imaging strategies. Nevertheless, the intricate anatomical confluence and the pancreas's deep-seated topography render the visualization and accurate diagnosis of its pathologies a formidable endeavor. The objective of our paper is to review the best diagnostic imagistic tools for the pancreas. Materials and Methods: we have gathered several articles using Prisma guidelines to determine the best imagistic methods. The imperative of pancreatic scanning transcends its diagnostic utility, proving to be a pivotal element in a multitude of clinical specialties, notably surgical oncology. Within this domain, multidetector computed tomography (MDCT) of the pancreas holds the distinction of being the paramount imaging modality, endorsed for its unrivaled capacity to delineate the staging and progression of pancreatic carcinoma. In synergy with MDCT, there has been a notable advent of avant-garde imaging techniques in recent years. These advanced methodologies, including ultrasonography, endoscopic ultrasonography, contrast-enhanced ultrasonography, and magnetic resonance imaging (MRI) conjoined with magnetic resonance cholangiopancreatography (MRCP), have broadened the horizon of tumor characterization, offering unparalleled depth and precision in oncological assessment. Other emerging diagnostic techniques, such as elastography, also hold a lot of potential and promise for the future of pancreatic imaging. Fine needle aspiration (FNA) is a quick, minimally invasive procedure to evaluate lumps using a thin needle to extract tissue for analysis. It is less invasive than surgical biopsies and usually performed as an outpatient with quick recovery. Its accuracy depends on sample quality, and the risks include minimal bleeding or discomfort. Results, guiding further treatment, are typically available within a week. Elastography is a non-invasive medical imaging technique that maps the elastic properties and stiffness of soft tissue. This method, often used in conjunction with ultrasound or MRI, helps differentiate between hard and soft areas in tissue, providing valuable diagnostic information. It is particularly useful for assessing liver fibrosis, thyroid nodules, breast lumps, and musculoskeletal conditions. The technique is painless and involves applying gentle pressure to the area being examined. The resulting images show tissue stiffness, indicating potential abnormalities. Elastography is advantageous for its ability to detect diseases in early stages and monitor treatment effectiveness. The procedure is quick, safe, and requires no special preparation, with results typically available immediately. Results: The assembled and gathered data shows the efficacy of various techniques in discerning the nature and extent of neoplastic lesions within the pancreas. Conclusions: The most common imaging modalities currently used in diagnosing pancreatic neoplasms are multidetector computed tomography (MDCT), endoscopic ultrasound (EUS), and magnetic resonance imaging (MRI), alongside new technologies, such as elastography.
Collapse
Affiliation(s)
- Sabina Florina Șolea
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; (S.F.Ș.); (A.O.); (F.C.V.); (R.M.Ș.); (A.V.); (C.B.)
- Bihor Clinical County Emergency Hospital, 410169 Oradea, Romania
| | - Mihaela Cristina Brisc
- Bihor Clinical County Emergency Hospital, 410169 Oradea, Romania
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Alexandra Orășeanu
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; (S.F.Ș.); (A.O.); (F.C.V.); (R.M.Ș.); (A.V.); (C.B.)
- Bihor Clinical County Emergency Hospital, 410169 Oradea, Romania
| | - Florian Ciprian Venter
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; (S.F.Ș.); (A.O.); (F.C.V.); (R.M.Ș.); (A.V.); (C.B.)
- Bihor Clinical County Emergency Hospital, 410169 Oradea, Romania
| | - Ciprian Mihai Brisc
- Faculty of Medicine and Pharmacy, University of Oradea, 410068 Oradea, Romania; (C.M.B.); (L.D.)
| | - Răzvan Mihai Șolea
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; (S.F.Ș.); (A.O.); (F.C.V.); (R.M.Ș.); (A.V.); (C.B.)
| | - Lavinia Davidescu
- Faculty of Medicine and Pharmacy, University of Oradea, 410068 Oradea, Romania; (C.M.B.); (L.D.)
| | - Amina Venter
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; (S.F.Ș.); (A.O.); (F.C.V.); (R.M.Ș.); (A.V.); (C.B.)
| | - Ciprian Brisc
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; (S.F.Ș.); (A.O.); (F.C.V.); (R.M.Ș.); (A.V.); (C.B.)
- Bihor Clinical County Emergency Hospital, 410169 Oradea, Romania
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
- Faculty of Medicine and Pharmacy, University of Oradea, 410068 Oradea, Romania; (C.M.B.); (L.D.)
| |
Collapse
|
8
|
Huang J, Gao G, Ge Y, Liu J, Cui H, Zheng R, Wang J, Wang S, Go VL, Hu S, Liu Y, Yang M, Sun Y, Shang D, Tian Y, Zhang Z, Xiang Z, Wang H, Guo J, Xiao GG. Development of a Serum-Based MicroRNA Signature for Early Detection of Pancreatic Cancer: A Multicenter Cohort Study. Dig Dis Sci 2024; 69:1263-1273. [PMID: 38451429 PMCID: PMC11026211 DOI: 10.1007/s10620-024-08338-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/05/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND A grim prognosis of pancreatic cancer (PCa) was attributed to the difficulty in early diagnosis of the disease. AIMS Identifying novel biomarkers for early detection of PCa is thus urgent to improve the overall survival rates of patients. METHODS The study was performed firstly by identification of candidate microRNAs (miRNAs) in formalin-fixed, paraffin-embedded tissues using microarray profiles, and followed by validation in a serum-based cohort study to assess clinical utility of the candidates. In the cohorts, a total of 1273 participants from four centers were retrospectively recruited as two cohorts including training and validation cohort. The collected serum specimens were analyzed by real-time polymerase chain reaction. RESULTS We identified 27 miRNAs expressed differentially in PCa tissues as compared to the benign. Of which, the top-four was selected as a panel whose diagnostic efficacy was fully assessed in the serum specimens. The panel exhibited superior to CA19-9, CA125, CEA and CA242 in discriminating patients with early stage PCa from healthy controls or non-PCa including chronic pancreatitis as well as pancreatic cystic neoplasms, with the area under the curves (AUC) of 0.971 (95% CI 0.956-0.987) and 0.924 (95% CI 0.899-0.949), respectively. Moreover, the panel eliminated interference from other digestive tumors with a specificity of 90.2%. CONCLUSIONS A panel of four serum miRNAs was developed showing remarkably discriminative ability of early stage PCa from either healthy controls or other pancreatic diseases, suggesting it may be developed as a novel, noninvasive approach for early screening of PCa in clinic.
Collapse
Affiliation(s)
- Jing Huang
- National Key Laboratory of Fine Chemical Engineering and Department of Pharmacology, School of Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Ge Gao
- Department of Laboratory Medicine, The Second & The Third Xiangya Hospitals, Central South University, Changsha, China
| | - Yang Ge
- Department of Food Safety and Toxicology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianzhou Liu
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongtu Cui
- School of Biomedical Engineering, Dalian University of Technology, Dalian, China
| | - Ren Zheng
- National Key Laboratory of Fine Chemical Engineering and Department of Pharmacology, School of Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Jialin Wang
- National Key Laboratory of Fine Chemical Engineering and Department of Pharmacology, School of Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Si Wang
- National Key Laboratory of Fine Chemical Engineering and Department of Pharmacology, School of Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Vay Liang Go
- The UCLA Agi Hirshberg Center for Pancreatic Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Shen Hu
- The UCLA Agi Hirshberg Center for Pancreatic Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Yefu Liu
- Department of Hepatopancreatobiliary Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, China
| | - Minwei Yang
- Department of Biliary-Pancreatic Surgery, School of Medicine, Ren Ji Hospital,, Shanghai Jiao Tong University, Shanghai, China
| | - Yongwei Sun
- Department of Biliary-Pancreatic Surgery, School of Medicine, Ren Ji Hospital,, Shanghai Jiao Tong University, Shanghai, China
| | - Dong Shang
- Clinical Laboratory of Integrative Medicine , Department of General Surgery, Pancreaticobiliary Center, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yantao Tian
- Pancreatic and Gastric Surgery, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhigang Zhang
- State Key Laboratory of Oncogenes and Related Genes, School of Medicine, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Zhongyuan Xiang
- Department of Laboratory Medicine, The Second & The Third Xiangya Hospitals, Central South University, Changsha, China
| | | | - Junchao Guo
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gary Guishan Xiao
- National Key Laboratory of Fine Chemical Engineering and Department of Pharmacology, School of Chemical Engineering, Dalian University of Technology, Dalian, China.
- The UCLA Agi Hirshberg Center for Pancreatic Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
- Functional Genomics and Proteomics Laboratory, Osteoporosis Research Center, Creighton University Medical Center, Omaha, NE, USA.
| |
Collapse
|
9
|
Zhang Y, Wu J, He J, Xu S. Preoperative differentiation of pancreatic cystic neoplasm subtypes on computed tomography radiomics. Quant Imaging Med Surg 2023; 13:6395-6411. [PMID: 37869288 PMCID: PMC10585572 DOI: 10.21037/qims-22-1192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 07/28/2023] [Indexed: 10/24/2023]
Abstract
Background Serous cystic neoplasm (SCN), mucinous cystic neoplasm (MCN), and intraductal papillary mucinous neoplasm (IPMN) comprise a large proportion of pancreatic cystic neoplasms (PCNs). Patients with MCN and IPMN require surgery due to the potential of malignant transformation, whereas those with SCN require periodic surveillance. However, the differential diagnosis of patients with PCNs before treatment remains a great challenge for all surgeons. Therefore, the establishment of a reliable diagnostic tool is urgently required for the improvement of precision diagnostics. Methods Between February 2015 and December 2020, 143 consecutive patients with PCNs who were confirmed by postoperative pathology were retrospectively included in the study cohort, then randomized into development and test cohorts at a ratio of 7:3. The predictors of preoperative clinical-radiologic parameters were evaluated by univariate and multivariable logistic regression analyses. A total of 1,218 radiomics features were computationally extracted from the enhanced computed tomography (CT) scans of the tumor region, and a radiomics signature was established by the random forest algorithm. In the development cohort, multi- and binary-class radiomics models integrating preoperative variables and radiomics features were constructed to distinguish between the 3 types of PCNs. The receiver operating characteristic (ROC) curve and the area under the curve (AUC) were used to evaluate the predictive efficiency of the model. An independent internal test cohort was applied to validate the classification models. Results All preoperative prediction models were built by integrating the radiomics signature with 13 diagnosis-related radiomics features and 3 important clinical-radiologic parameters: age, sex, and tumor diameter. The multiclass prediction model presented an overall accuracy of 0.804 in the development cohort and 0.707 in the test cohort. The binary-class prediction models displayed higher overall accuracies of 0.853, 0.866, and 0.928 in the development dataset and 0.750, 0.839, and 0.889 in the test dataset. In the test cohort, the binary-class radiomics models showed better predictive performances {AUC =0.914 [95% confidence interval (CI): 0.786 to 1.000], 0.863 (95% CI: 0.714 to 0.941), and 0.926 (95% CI: 0.824 to 1.000)} than the multiclass radiomics model [AUC =0.850 (95% CI: 0.696 to 1.000)], with a large net benefit in the decision curve analysis (DCA). The radiomics-based nomogram provided the correct predicted probability for the diagnosis of PCNs. Conclusions The proposed radiomics models with clinical-radiologic parameters and radiomics features help to predict the accurate diagnosis among PCNs to advance personalized medicine.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of PET/CT Center, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, China
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jin Wu
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jian He
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Shanshan Xu
- Department of PET/CT Center, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, China
| |
Collapse
|
10
|
Cai Y, Liu P, Xu Y, Xia Y, Peng X, Zhao H, Chen Q. Biomarkers of obesity-mediated insulin resistance: focus on microRNAs. Diabetol Metab Syndr 2023; 15:167. [PMID: 37537674 PMCID: PMC10401761 DOI: 10.1186/s13098-023-01137-3] [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: 05/04/2023] [Accepted: 07/09/2023] [Indexed: 08/05/2023] Open
Abstract
Obesity and metabolic syndromes are becoming increasingly prevalent worldwide. Insulin resistance (IR) is a common complication of obesity. However, IR occurrence varies across individuals with obesity and may involve epigenetic factors. To rationalize the allocation of healthcare resources, biomarkers for the early risk stratification of individuals with obesity should be identified. MicroRNAs (miRNAs) are closely associated with metabolic diseases and involved in epigenetic regulation. In this review, we have summarized the changes in miRNA expression in the peripheral circulation and tissues of patients and animals with obesity-associated IR over the last 5 years and identified several candidate biomarkers that predict obesity-related IR. There are areas for improvement in existing studies. First, more than the predictive validity of a single biomarker is required, and a biomarker panel needs to be formed. Second, miRNAs are often studied in isolation and do not form a network of signaling pathways. We believe that early biomarkers can help clinicians accurately predict individuals prone to obesity-related IR at an early stage. Epigenetic regulation may be one of the underlying causes of different clinical outcomes in individuals with obesity. Future studies should focus on objectively reflecting the differences in miRNA profile expression in individuals with obesity-related IR, which may help identify more reliable biomarkers. Understanding the metabolic pathways of these miRNAs can help design new metabolic risk prevention and management strategies, and support the development of drugs to treat obesity and metabolic disorders.
Collapse
Affiliation(s)
- Yichen Cai
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Pan Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yumei Xu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuguo Xia
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan, China
| | - Xiaowan Peng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Haiyan Zhao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiu Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan, China.
| |
Collapse
|
11
|
Mormul A, Włoszek E, Nowoszewska J, Fudalej M, Budzik M, Badowska-Kozakiewicz A, Deptała A. Rare Non-Neuroendocrine Pancreatic Tumours. Cancers (Basel) 2023; 15:cancers15082216. [PMID: 37190144 DOI: 10.3390/cancers15082216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/30/2023] [Accepted: 04/07/2023] [Indexed: 05/17/2023] Open
Abstract
The most common tumour of the pancreas is ductal adenocarcinoma (PDAC). It remains one of the most lethal non-neuroendocrine solid tumours despite the use of a multi-approach strategy. Other, less-common neoplasms, which are responsible for 15% of pancreatic lesions, differ in treatment and prognosis. Due to the low incidence rate, there is a lack of information about the rarest pancreatic tumours. In this review, we described six rare pancreatic tumours: intraductal papillary mucinous neoplasm (IPMN), mucinous cystadenoma (MCN), serous cystic neoplasm (SCN), acinar cell carcinoma (ACC), solid pseudopapillary neoplasm (SPN) and pancreatoblastoma (PB). We distinguished their epidemiology, clinical and gross features, covered the newest reports about courses of treatment and systematised differential diagnoses. Although the most common pancreatic tumour, PDAC, has the highest malignant potential, it is still essential to properly classify and differentiate less-common lesions. It is vital to continue the search for new biomarkers, genetic mutations and the development of more specific biochemical tests for determining malignancy in rare pancreatic neoplasms.
Collapse
Affiliation(s)
- Agata Mormul
- Students' Scientific Organization of Cancer Cell Biology, Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
| | - Emilia Włoszek
- Students' Scientific Organization of Cancer Cell Biology, Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
| | - Julia Nowoszewska
- Students' Scientific Organization of Cancer Cell Biology, Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
| | - Marta Fudalej
- Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
| | - Michał Budzik
- Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
| | | | - Andrzej Deptała
- Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
| |
Collapse
|
12
|
He RZ, Zheng JH, Yao HF, Xu DP, Yang MW, Liu DJ, Sun YW, Huo YM. ADAMTS12 promotes migration and epithelial-mesenchymal transition and predicts poor prognosis for pancreatic cancer. Hepatobiliary Pancreat Dis Int 2023; 22:169-178. [PMID: 35508435 DOI: 10.1016/j.hbpd.2022.04.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 04/18/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND ADAMTS (a disintegrin and metalloproteinase with thrombospondin-like motifs) family, a group of extracellular multifunctional enzymes, has been proven to play a pivotal role in the tumor. In pancreatic cancer, the role and mechanism of this family remain unclear. The present study aimed to figure out the hub gene of ADAMTSs and explore the exact roles in the prognosis and biological functions in pancreatic ductal adenocarcinoma (PDAC). METHODS We used several databases to analyze the ADAMTS family and then screen out the hub genes. The expression of ADAMTS12 in 106 pairs of PDAC tumors and adjacent normal tissues was examined by immunohistochemistry, and its correlations with clinical parameters were further analyzed. The impacts of ADAMTS12 on the migration of PDAC cells were predicted by gene set enrichment analysis and confirmed by transwell assays. The potential impacts of ADAMTS12 on the epithelial-mesenchymal transition (EMT) were identified by database analysis and experimental proof of real-time quantitative polymerase chain reaction (qPCR) and Western blotting. RESULTS Our study found that ADAMTS12 was a crucial gene in PDAC, and it was highly expressed in tumor tissues when compared to that in the adjacent tissues. ADATMS12 had predictive value of a poor prognosis for PDAC. The elevation of ADAMTS12 was parallel to the progression of PDAC. Inhibition of ADAMTS12 suppressed the migration of PDAC cells and interfered with the process of EMT. CONCLUSIONS ADAMTS12 is a crucial member of ADAMTSs in PDAC and a predictor of poor prognosis. Additionally, based on its impacts on migration and metastasis in PDAC and the relationship with EMT, ADAMTS12 plays a role of an oncogene in PDAC and may be a promising target for treatment.
Collapse
Affiliation(s)
- Rui-Zhe He
- State Key Laboratory of Oncogenes and Related Genes, Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia-Hao Zheng
- State Key Laboratory of Oncogenes and Related Genes, Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong-Fei Yao
- State Key Laboratory of Oncogenes and Related Genes, Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Da-Peng Xu
- State Key Laboratory of Oncogenes and Related Genes, Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min-Wei Yang
- State Key Laboratory of Oncogenes and Related Genes, Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - De-Jun Liu
- State Key Laboratory of Oncogenes and Related Genes, Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong-Wei Sun
- State Key Laboratory of Oncogenes and Related Genes, Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan-Miao Huo
- State Key Laboratory of Oncogenes and Related Genes, Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
13
|
Salivary Polyamines Help Detect High-Risk Patients with Pancreatic Cancer: A Prospective Validation Study. Int J Mol Sci 2023; 24:ijms24032998. [PMID: 36769322 PMCID: PMC9918012 DOI: 10.3390/ijms24032998] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Pancreatic cancer is one of the most malignant cancer types and has a poor prognosis. It is often diagnosed at an advanced stage because of the absence of typical symptoms. Therefore, it is necessary to establish a screening method for the early detection of pancreatic cancer in high-risk individuals. This is a prospective validation study conducted in a cohort of 1033 Japanese individuals (male, n = 467, age = 63.3 ± 11.5 years; female, n = 566, age = 64.2 ± 10.6 years) to evaluate the use of salivary polyamines for screening pancreatic diseases and cancers. Patients with pancreatic cancer were not included; however, other pancreatic diseases were treated as positive cases for accuracy verification. Of the 135 individuals with elevated salivary polyamine markers, 66 had pancreatic diseases, such as chronic pancreatitis and pancreatic cysts, and 1 had gallbladder cancer. These results suggest that the salivary polyamine panel is a useful noninvasive pancreatic disease screening tool.
Collapse
|
14
|
Li SY, Wang ZJ, Pan CY, Wu C, Li ZS, Jin ZD, Wang KX. Comparative Performance of Endoscopic Ultrasound-Based Techniques in Patients With Pancreatic Cystic Lesions: A Network Meta-Analysis. Am J Gastroenterol 2023; 118:243-255. [PMID: 36563321 DOI: 10.14309/ajg.0000000000002088] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 11/02/2022] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Evidence on the comparative diagnostic performance of endoscopic ultrasound (EUS)-based techniques for pancreatic cystic lesions (PCLs) is limited. This network meta-analysis comprehensively compared EUS-based techniques for PCL diagnosis. METHODS A comprehensive literature search was performed for all comparative studies assessing the accuracy of 2 or more modalities for PCL diagnosis. The primary outcome was the diagnostic efficacy for mucinous PCLs. Secondary outcomes were the diagnostic efficacy for malignant PCLs, diagnostic success rate, and adverse event rate. A network meta-analysis was conducted using the ANOVA model to assess the diagnostic accuracy of each index. RESULTS Forty studies comprising 3,641 patients were identified. The network ranking of the superiority index for EUS-guided needle-based confocal laser endomicroscopy (EUS-nCLE) and EUS-guided through-the-needle biopsy (EUS-TTNB) were significantly higher than other techniques for differentiating mucinous PCLs; besides, EUS-TTNB was also the optimal technique in identifying malignant PCLs. The evidence was inadequate for EUS-nCLE diagnosing malignant PCLs and contrast-enhanced harmonic EUS diagnosing both mucinous and malignant PCLs. Glucose showed a high sensitivity but low specificity, and molecular analysis (KRAS, GNAS, and KRAS + GNAS mutations) showed a high specificity but low sensitivity for diagnosing mucinous PCLs. Satisfactory results were not obtained during the evaluation of the efficiency of pancreatic cyst fluid (PCF) biomarkers in detecting malignant PCLs. DISCUSSION For centers with relevant expertise and facilities, EUS-TTNB and EUS-nCLE were better choices for the diagnosis of PCLs. Further studies are urgently required for further improving PCF biomarkers and validating the diagnostic performance of the index techniques.
Collapse
Affiliation(s)
- Shi-Yu Li
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhi-Jie Wang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Cheng-Ye Pan
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Cheng Wu
- Department of Military Health Statistics, Naval Medical University, Shanghai, China
| | - Zhao-Shen Li
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhen-Dong Jin
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Kai-Xuan Wang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
15
|
Søreide K, Ismail W, Roalsø M, Ghotbi J, Zaharia C. Early Diagnosis of Pancreatic Cancer: Clinical Premonitions, Timely Precursor Detection and Increased Curative-Intent Surgery. Cancer Control 2023; 30:10732748231154711. [PMID: 36916724 PMCID: PMC9893084 DOI: 10.1177/10732748231154711] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND The overall poor prognosis in pancreatic cancer is related to late clinical detection. Early diagnosis remains a considerable challenge in pancreatic cancer. Unfortunately, the onset of clinical symptoms in patients usually indicate advanced disease or presence of metastasis. ANALYSIS AND RESULTS Currently, there are no designated diagnostic or screening tests for pancreatic cancer in clinical use. Thus, identifying risk groups, preclinical risk factors or surveillance strategies to facilitate early detection is a target for ongoing research. Hereditary genetic syndromes are a obvious, but small group at risk, and warrants close surveillance as suggested by society guidelines. Screening for pancreatic cancer in asymptomatic individuals is currently associated with the risk of false positive tests and, thus, risk of harms that outweigh benefits. The promise of cancer biomarkers and use of 'omics' technology (genomic, transcriptomics, metabolomics etc.) has yet to see a clinical breakthrough. Several proposed biomarker studies for early cancer detection lack external validation or, when externally validated, have shown considerably lower accuracy than in the original data. Biopsies or tissues are often taken at the time of diagnosis in research studies, hence invalidating the value of a time-dependent lag of the biomarker to detect a pre-clinical, asymptomatic yet operable cancer. New technologies will be essential for early diagnosis, with emerging data from image-based radiomics approaches, artificial intelligence and machine learning suggesting avenues for improved detection. CONCLUSIONS Early detection may come from analytics of various body fluids (eg 'liquid biopsies' from blood or urine). In this review we present some the technological platforms that are explored for their ability to detect pancreatic cancer, some of which may eventually change the prospects and outcomes of patients with pancreatic cancer.
Collapse
Affiliation(s)
- Kjetil Søreide
- Department of Gastrointestinal Surgery, HPB unit, 60496Stavanger University Hospital, Stavanger, Norway.,Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Gastrointestinal Translational Research Group, Laboratory for Molecular Medicine, 60496Stavanger University Hospital, Stavanger, Norway
| | - Warsan Ismail
- Department of Gastrointestinal Surgery, HPB unit, 60496Stavanger University Hospital, Stavanger, Norway
| | - Marcus Roalsø
- Department of Gastrointestinal Surgery, HPB unit, 60496Stavanger University Hospital, Stavanger, Norway.,Gastrointestinal Translational Research Group, Laboratory for Molecular Medicine, 60496Stavanger University Hospital, Stavanger, Norway.,Department of Quality and Health Technology, 60496University of Stavanger, Stavanger, Norway
| | - Jacob Ghotbi
- Department of Gastrointestinal Surgery, HPB unit, 60496Stavanger University Hospital, Stavanger, Norway
| | - Claudia Zaharia
- Gastrointestinal Translational Research Group, Laboratory for Molecular Medicine, 60496Stavanger University Hospital, Stavanger, Norway.,Department of Pathology, 60496Stavanger University Hospital, Stavanger, Norway
| |
Collapse
|
16
|
Wang R, Su D, Liu Y, Qiu J, Cao Z, Yang G, Luo W, Tao J, Zhang T. Cancer-specific survival and metastasis in pancreatic mucinous cystadenocarcinoma: A SEER-based cohort study. Front Oncol 2022; 12:985184. [PMCID: PMC9631930 DOI: 10.3389/fonc.2022.985184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 10/03/2022] [Indexed: 11/13/2022] Open
Abstract
Aims This study aimed to investigate the prognostic value of clinical features for cancer-specific survival (CSS) and metastasis in patients with pancreatic mucinous cystadenocarcinoma (MCAC). We further constructed and validated an effective nomogram to predict CSS. Methods We screened patients diagnosed with pancreatic MCAC from Surveillance Epidemiology and End Results (SEER) database. Kaplan-Meier curves were used to determine the CSS time. Univariate and multivariate Cox and logistic regression analyses were conducted to identify the prognostic factors for CSS and metastasis. The nomogram was constructed to predict the prognosis of pancreatic MCAC based on the results from the multivariate analysis. We used the concordance index (C-index), the area under the curve (AUC), and the calibration plots to determine the predictive accuracy and discriminability of the nomogram. Results Multivariate Cox analysis revealed that age, primary site, grade, and radiotherapy were independent prognostic factors associated with CSS. Multivariate logistic regression analysis revealed that surgery and grade were independent risk factors associated with metastasis. The independent risk factors were included to construct a prognosis prediction model for predicting CSS in patients with pancreatic MCAC. The concordance index (C-index), receiver operating characteristic (ROC) curves, and calibration plots of the training cohort and the validation cohort showed that the nomogram had an acceptable predictive performance. Conclusion We established a nomogram that could determine the 3- and 5-year CSS, which could evaluate individual clinical outcomes and provide individualized clinical decisions.
Collapse
Affiliation(s)
- Ruobing Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dan Su
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yueze Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiangdong Qiu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhe Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenhao Luo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinxin Tao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Taiping Zhang,
| |
Collapse
|
17
|
Farr KP, Moses D, Haghighi KS, Phillips PA, Hillenbrand CM, Chua BH. Imaging Modalities for Early Detection of Pancreatic Cancer: Current State and Future Research Opportunities. Cancers (Basel) 2022; 14:cancers14102539. [PMID: 35626142 PMCID: PMC9139708 DOI: 10.3390/cancers14102539] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary While survival rates for many cancers have improved dramatically over the last 20 years, patients with pancreatic cancer have persistently poor outcomes. The majority of patients with pancreatic cancer are not suitable for potentially curative surgery due to locally advanced or metastatic disease stage at diagnosis. Therefore, early detection would potentially improve survival of pancreatic cancer patients through earlier intervention. Here, we present clinical challenges in the early detection of pancreatic cancer, characterise high risk groups for pancreatic cancer and current screening programs in high-risk individuals. The aim of this scoping review is to investigate the role of both established and novel imaging modalities for early detection of pancreatic cancer. Furthermore, we investigate innovative imaging techniques for early detection of pancreatic cancer, but its widespread application requires further investigation and potentially a combination with other non-invasive biomarkers. Abstract Pancreatic cancer, one of the most lethal malignancies, is increasing in incidence. While survival rates for many cancers have improved dramatically over the last 20 years, people with pancreatic cancer have persistently poor outcomes. Potential cure for pancreatic cancer involves surgical resection and adjuvant therapy. However, approximately 85% of patients diagnosed with pancreatic cancer are not suitable for potentially curative therapy due to locally advanced or metastatic disease stage. Because of this stark survival contrast, any improvement in early detection would likely significantly improve survival of patients with pancreatic cancer through earlier intervention. This comprehensive scoping review describes the current evidence on groups at high risk for developing pancreatic cancer, including individuals with inherited predisposition, pancreatic cystic lesions, diabetes, and pancreatitis. We review the current roles of imaging modalities focusing on early detection of pancreatic cancer. Additionally, we propose the use of advanced imaging modalities to identify early, potentially curable pancreatic cancer in high-risk cohorts. We discuss innovative imaging techniques for early detection of pancreatic cancer, but its widespread application requires further investigation and potentially a combination with other non-invasive biomarkers.
Collapse
Affiliation(s)
- Katherina P. Farr
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW, Sydney, NSW 2052, Australia; (K.S.H.); (B.H.C.)
- Correspondence:
| | - Daniel Moses
- Graduate School of Biomedical Engineering, UNSW, Sydney, NSW 2052, Australia;
| | - Koroush S. Haghighi
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW, Sydney, NSW 2052, Australia; (K.S.H.); (B.H.C.)
- Department of General Surgery, Prince of Wales Hospital, Sydney, NSW 2052, Australia
| | - Phoebe A. Phillips
- Pancreatic Cancer Translational Research Group, School of Clinical Medicine, Lowy Cancer Research Centre, UNSW, Sydney, NSW 2052, Australia;
| | - Claudia M. Hillenbrand
- Research Imaging NSW, Division of Research & Enterprise, UNSW, Sydney, NSW 2052, Australia;
| | - Boon H. Chua
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW, Sydney, NSW 2052, Australia; (K.S.H.); (B.H.C.)
- Nelune Comprehensive Cancer Centre, Prince of Wales Hospital, Sydney, NSW 2052, Australia
| |
Collapse
|
18
|
Osei-Bordom DC, Sachdeva G, Christou N. Liquid Biopsy as a Prognostic and Theranostic Tool for the Management of Pancreatic Ductal Adenocarcinoma. Front Med (Lausanne) 2022; 8:788869. [PMID: 35096878 PMCID: PMC8795626 DOI: 10.3389/fmed.2021.788869] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 12/02/2021] [Indexed: 12/24/2022] Open
Abstract
Pancreatic ductal adenocarcinomas (PDAC) represent one of the deadliest cancers worldwide. Survival is still low due to diagnosis at an advanced stage and resistance to treatment. Herein, we review the main types of liquid biopsy able to help in both prognosis and adaptation of treatments.
Collapse
Affiliation(s)
- Daniel C Osei-Bordom
- Department of General Surgery, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, United Kingdom
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, Centre for Liver and Gastroenterology Research, University of Birmingham, Birmingham, United Kingdom
| | - Gagandeep Sachdeva
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Niki Christou
- Department of General Surgery, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, United Kingdom
- Department of General Surgery, University Hospital of Limoges, Limoges, France
- EA3842 CAPTuR Laboratory "Cell Activation Control, Tumor Progression and Therapeutic Resistance", Faculty of Medicine, Limoges, France
| |
Collapse
|
19
|
Zhu Z, Wang Q, Chen X, Wang Q, Yan C, Zhao X, Zhao W, Zhu WH. An Enzyme-Activatable Aggregation-Induced-Emission Probe: Intraoperative Pathological Fluorescent Diagnosis of Pancreatic Cancer via Specific Cathepsin E. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107444. [PMID: 34693566 DOI: 10.1002/adma.202107444] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/14/2021] [Indexed: 06/13/2023]
Abstract
Pancreatic cancer (PC) is one of the most devastating malignant tumors. However, fluorescence probes for early clinical diagnosis of PC often encounter difficulties in accuracy and penetrability. In this work, an enzyme-activated aggregation-induced-emission (AIE) probe, QM-HSP-CPP, for high-contrast fluorescence diagnosis of PC is developed by monitoring specific overexpressed enzyme Cathepsin E (CTSE). The probe is composed of an AIE fluorophore QM-COOH (QM = quinoline-malononitrile), CTSE-triggered hydrophobic peptide (HSP), and hydrophilic biocompatible cell penetrating peptide (CPP). The CPP unit can well-modulate the molecular dispersion properties, giving initial fluorescence-off state in the aqueous biosystem, thus endowing high signal-to-noise ratio, and finally overcoming the poor targeting selectivity of traditional AIE probes. CPP can ensure cell/tissue penetrating ability, thus allowing on-site monitoring of endogenous CTSE in PC cells, tissues, and living animal models. When the QM-HSP-CPP probe is specifically cleaved by CTSE, it can generate AIE signals in situ with high-specificity and long-term tracking ability, and successfully achieve intraoperative diagnosis of human PC sections, tracking PC in heterotopic nude mice models. The CTSE-enzyme-triggered AIEgens' liberation strategy improves accuracy and addresses the penetration problem simultaneously, which can expand the database of multitudinous biocompatible AIE-active probes, especially for establishing intraoperative pathological fluorescent diagnosis.
Collapse
Affiliation(s)
- Zhirong Zhu
- Shanghai Key Laboratory of Functional Materials Chemistry, Key Laboratory for Advanced Materials and Institute of Fine Chemicals, Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Qi Wang
- Shanghai Key Laboratory of Functional Materials Chemistry, Key Laboratory for Advanced Materials and Institute of Fine Chemicals, Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Xiaoyan Chen
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Quan Wang
- State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Chengxu Yan
- Shanghai Key Laboratory of Functional Materials Chemistry, Key Laboratory for Advanced Materials and Institute of Fine Chemicals, Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Xiaolei Zhao
- Shanghai Key Laboratory of Functional Materials Chemistry, Key Laboratory for Advanced Materials and Institute of Fine Chemicals, Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Weijun Zhao
- Shanghai Key Laboratory of Functional Materials Chemistry, Key Laboratory for Advanced Materials and Institute of Fine Chemicals, Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Wei-Hong Zhu
- Shanghai Key Laboratory of Functional Materials Chemistry, Key Laboratory for Advanced Materials and Institute of Fine Chemicals, Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, China
| |
Collapse
|
20
|
Gao J, Han F, Wang X, Duan S, Zhang J. Multi-Phase CT-Based Radiomics Nomogram for Discrimination Between Pancreatic Serous Cystic Neoplasm From Mucinous Cystic Neoplasm. Front Oncol 2021; 11:699812. [PMID: 34926238 PMCID: PMC8672034 DOI: 10.3389/fonc.2021.699812] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 11/15/2021] [Indexed: 12/25/2022] Open
Abstract
Purpose This study aimed to develop and verify a multi-phase (MP) computed tomography (CT)-based radiomics nomogram to differentiate pancreatic serous cystic neoplasms (SCNs) from mucinous cystic neoplasms (MCNs), and to compare the diagnostic efficacy of radiomics models for different phases of CT scans. Materials and Methods A total of 170 patients who underwent surgical resection between January 2011 and December 2018, with pathologically confirmed pancreatic cystic neoplasms (SCN=115, MCN=55) were included in this single-center retrospective study. Radiomics features were extracted from plain scan (PS), arterial phase (AP), and venous phase (VP) CT scans. Algorithms were performed to identify the optimal features to build a radiomics signature (Radscore) for each phase. All features from these three phases were analyzed to develop the MP-Radscore. A combined model comprised the MP-Radscore and imaging features from which a nomogram was developed. The accuracy of the nomogram was evaluated using receiver operating characteristic (ROC) curves, calibration tests, and decision curve analysis. Results For each scan phase, 1218 features were extracted, and the optimal ones were selected to construct the PS-Radscore (11 features), AP-Radscore (11 features), and VP-Radscore (12 features). The MP-Radscore (14 features) achieved better performance based on ROC curve analysis than any single phase did [area under the curve (AUC), training cohort: MP-Radscore 0.89, PS-Radscore 0.78, AP-Radscore 0.83, VP-Radscore 0.85; validation cohort: MP-Radscore 0.88, PS-Radscore 0.77, AP-Radscore 0.83, VP-Radscore 0.84]. The combination nomogram performance was excellent, surpassing those of all other nomograms in both the training cohort (AUC, 0.91) and validation cohort (AUC, 0.90). The nomogram also performed well in the calibration and decision curve analyses. Conclusions Radiomics for arterial and venous single-phase models outperformed the plain scan model. The combination nomogram that incorporated the MP-Radscore, tumor location, and cystic number had the best discriminatory performance and showed excellent accuracy for differentiating SCN from MCN.
Collapse
Affiliation(s)
- Jiahao Gao
- Department of Radiology, Huashan Hospital North, Fudan University, Shanghai, China.,Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Fang Han
- Department of Radiology, Huashan Hospital North, Fudan University, Shanghai, China.,Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoshuang Wang
- Department of Radiology, Huashan Hospital North, Fudan University, Shanghai, China
| | - Shaofeng Duan
- Department of Life Sciences, GE Healthcare, Shanghai, China
| | - Jiawen Zhang
- Department of Radiology, Huashan Hospital North, Fudan University, Shanghai, China.,Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Jia M, Zhang D, Zhang C, Li C. Nanoparticle-based delivery systems modulate the tumor microenvironment in pancreatic cancer for enhanced therapy. J Nanobiotechnology 2021; 19:384. [PMID: 34809634 PMCID: PMC8607729 DOI: 10.1186/s12951-021-01134-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/12/2021] [Indexed: 02/08/2023] Open
Abstract
Pancreatic cancer is one of the most lethal malignant tumors with a low survival rate, partly because the tumor microenvironment (TME), which consists of extracellular matrix (ECM), cancer-associated fibroblasts (CAFs), immune cells, and vascular systems, prevents effective drug delivery and chemoradiotherapy. Thus, modulating the microenvironment of pancreatic cancer is considered a promising therapeutic approach. Since nanoparticles are one of the most effective cancer treatment strategies, several nano-delivery platforms have been developed to regulate the TME and enhance treatment. Here, we summarize the latest advances in nano-delivery systems that alter the TME in pancreatic cancer by depleting ECM, inhibiting CAFs, reversing immunosuppression, promoting angiogenesis, or improving the hypoxic environment. We also discuss promising new targets for such systems. This review is expected to improve our understanding of how to modulate the pancreatic cancer microenvironment and guide the development of new therapies.
Collapse
Affiliation(s)
- Ming Jia
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, No.1, Section 1, Xianglin Road, Luzhou, Sichuan, 646000, People's Republic of China
| | - Dan Zhang
- Department of Pharmacy of Traditional Chinese Medicine, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Chunxiang Zhang
- The Key Laboratory of Medical Electrophysiology of the Ministry of Education, Southwest Medical University, No.1, Section 1, Xianglin Road, Luzhou, Sichuan, 646000, People's Republic of China.
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, No.1, Section 1, Xianglin Road, Luzhou, Sichuan, 646000, People's Republic of China.
| |
Collapse
|
22
|
Permuth JB, Vyas S, Li J, Chen DT, Jeong D, Choi JW. Comparison of Radiomic Features in a Diverse Cohort of Patients With Pancreatic Ductal Adenocarcinomas. Front Oncol 2021; 11:712950. [PMID: 34367997 PMCID: PMC8339963 DOI: 10.3389/fonc.2021.712950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/05/2021] [Indexed: 12/20/2022] Open
Abstract
Background Significant racial disparities in pancreatic cancer incidence and mortality rates exist, with the highest rates in African Americans compared to Non-Hispanic Whites and Hispanic/Latinx populations. Computer-derived quantitative imaging or “radiomic” features may serve as non-invasive surrogates for underlying biological factors and heterogeneity that characterize pancreatic tumors from African Americans, yet studies are lacking in this area. The objective of this pilot study was to determine if the radiomic tumor profile extracted from pretreatment computed tomography (CT) images differs between African Americans, Non-Hispanic Whites, and Hispanic/Latinx with pancreatic cancer. Methods We evaluated a retrospective cohort of 71 pancreatic cancer cases (23 African American, 33 Non-Hispanic White, and 15 Hispanic/Latinx) who underwent pretreatment CT imaging at Moffitt Cancer Center and Research Institute. Whole lesion semi-automated segmentation was performed on each slice of the lesion on all pretreatment venous phase CT exams using Healthmyne Software (Healthmyne, Madison, WI, USA) to generate a volume of interest. To reduce feature dimensionality, 135 highly relevant non-texture and texture features were extracted from each segmented lesion and analyzed for each volume of interest. Results Thirty features were identified and significantly associated with race/ethnicity based on Kruskal-Wallis test. Ten of the radiomic features were highly associated with race/ethnicity independent of tumor grade, including sphericity, volumetric mean Hounsfield units (HU), minimum HU, coefficient of variation HU, four gray level texture features, and two wavelet texture features. A radiomic signature summarized by the first principal component partially differentiated African American from non-African American tumors (area underneath the curve = 0.80). Poorer survival among African Americans compared to Non-African Americans was observed for tumors with lower volumetric mean CT [HR: 3.90 (95% CI:1.19–12.78), p=0.024], lower GLCM Avg Column Mean [HR:4.75 (95% CI: 1.44,15.37), p=0.010], and higher GLCM Cluster Tendency [HR:3.36 (95% CI: 1.06–10.68), p=0.040], and associations persisted in volumetric mean CT and GLCM Avg Column after adjustment for key clinicopathologic factors. Conclusions This pilot study identified several textural radiomics features associated with poor overall survival among African Americans with PDAC, independent of other prognostic factors such as grade. Our findings suggest that CT radiomic features may serve as surrogates for underlying biological factors and add value in predicting clinical outcomes when integrated with other parameters in ongoing and future studies of cancer health disparities.
Collapse
Affiliation(s)
- Jennifer B Permuth
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States.,Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Shraddha Vyas
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Jiannong Li
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Dung-Tsa Chen
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Daniel Jeong
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States.,Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Jung W Choi
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| |
Collapse
|
23
|
Yip-Schneider MT, Wu H, Allison HR, Easler JJ, Sherman S, Al-Haddad MA, Dewitt JM, Schmidt CM. Biomarker Risk Score Algorithm and Preoperative Stratification of Patients with Pancreatic Cystic Lesions. J Am Coll Surg 2021; 233:426-434.e4. [PMID: 34166836 DOI: 10.1016/j.jamcollsurg.2021.05.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/28/2021] [Accepted: 05/28/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Pancreatic cysts are incidentally detected in up to 13% of patients undergoing radiographic imaging. Of the most frequently encountered types, mucin-producing (mucinous) pancreatic cystic lesions may develop into pancreatic cancer, while nonmucinous ones have little or no malignant potential. Accurate preoperative diagnosis is critical for optimal management, but has been difficult to achieve, resulting in unnecessary major surgery. Here, we aim to develop an algorithm based on biomarker risk scores to improve risk stratification. STUDY DESIGN Patients undergoing surgery and/or surveillance for a pancreatic cystic lesion, with diagnostic imaging and banked pancreatic cyst fluid, were enrolled in the study after informed consent (n = 163 surgical, 67 surveillance). Cyst fluid biomarkers with high specificity for distinguishing nonmucinous from mucinous pancreatic cysts (vascular endothelial growth factor [VEGF], glucose, carcinoembryonic antigen [CEA], amylase, cytology, and DNA mutation) were selected. Biomarker risk scores were used to design an algorithm to predict preoperative diagnosis. Performance was tested using surgical (retrospective) and surveillance (prospective) cohorts. RESULTS In the surgical cohort, the biomarker algorithm outperformed the preoperative clinical diagnosis in correctly predicting the final pathologic diagnosis (91% vs 73%; p < 0.000001). Specifically, nonmucinous serous cystic neoplasms (SCN) and mucinous cystic neoplasms (MCN) were correctly classified more frequently by the algorithm than clinical diagnosis (96% vs 30%; p < 0.000008 and 92% vs 69%; p = 0.04, respectively). In the surveillance cohort, the algorithm predicted a preoperative diagnosis with high confidence based on a high biomarker score and/or consistency with imaging from ≥1 follow-up visits. CONCLUSIONS A biomarker risk score-based algorithm was able to correctly classify pancreatic cysts preoperatively. Importantly, this tool may improve initial and dynamic risk stratification, reducing overdiagnosis and underdiagnosis.
Collapse
Affiliation(s)
- Michele T Yip-Schneider
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN; Walther Oncology Center, Indianapolis, IN; Indiana University Simon Cancer Center, Indianapolis, IN; Indiana University Health Pancreatic Cyst and Cancer Early Detection Center, Indianapolis, IN
| | - Huangbing Wu
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN; Indiana University Health Pancreatic Cyst and Cancer Early Detection Center, Indianapolis, IN
| | | | - Jeffrey J Easler
- Department of Medicine, Division of Gastroenterology, Indianapolis, IN
| | - Stuart Sherman
- Department of Medicine, Division of Gastroenterology, Indianapolis, IN
| | - Mohammad A Al-Haddad
- Department of Medicine, Division of Gastroenterology, Indianapolis, IN; Indiana University Health Pancreatic Cyst and Cancer Early Detection Center, Indianapolis, IN
| | - John M Dewitt
- Department of Medicine, Division of Gastroenterology, Indianapolis, IN
| | - C Max Schmidt
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN; Biochemistry/Molecular Biology, Indianapolis, IN; Walther Oncology Center, Indianapolis, IN; Indiana University Simon Cancer Center, Indianapolis, IN; Indiana University Health Pancreatic Cyst and Cancer Early Detection Center, Indianapolis, IN.
| |
Collapse
|
24
|
Chen S, Ren S, Guo K, Daniels MJ, Wang Z, Chen R. Preoperative differentiation of serous cystic neoplasms from mucin-producing pancreatic cystic neoplasms using a CT-based radiomics nomogram. Abdom Radiol (NY) 2021; 46:2637-2646. [PMID: 33558952 DOI: 10.1007/s00261-021-02954-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/05/2021] [Accepted: 01/13/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE To develop and validate a CT-based radiomics nomogram in preoperative differential diagnosis of SCNs from mucin-producing PCNs. MATERIAL AND METHODS A total of 89 patients consisting of 31 SCNs, 30 IPMNs, and 28 MCNs who underwent preoperative CT were analyzed. A total of 710 radiomics features were extracted from each case. Patients were divided into training (n = 63) and validation cohorts (n = 26) with a ratio of 7:3. Least absolute shrinkage and selection operator (LASSO) method and logistic regression analysis were used for feature selection and model construction. A nomogram was created from a comprehensive model consisting of clinical features and the fusion radiomics signature. A decision curve analysis was used for clinical decisions. RESULTS The radiomics features extracted from CT could assist with the differentiation of SCNs from mucin-producing PCNs in both the training and validation cohorts. The signature of the combination of the plain, late arterial, and venous phases had the largest areas under the curve (AUCs) of 0.960 (95% CI 0.910-1) in the training cohort and 0.817 (95% CI 0.651-0.983) in the validation cohort with good calibration. The value and efficacy of the nomogram was verified using decision curve analysis. CONCLUSION A comprehensive nomogram incorporating clinical features and fusion radiomics signature can differentiate SCNs from mucin-producing PCNs.
Collapse
Affiliation(s)
- Shuai Chen
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Shuai Ren
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
- Department of Diagnostic Radiology and Nuclear Medicine, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA
| | - Kai Guo
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Marcus J Daniels
- Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Zhongqiu Wang
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China.
| | - Rong Chen
- Department of Diagnostic Radiology and Nuclear Medicine, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA
| |
Collapse
|
25
|
Kaur S, Jain M, Batra SK. Liquid Biopsy for Identification of High-Risk Cystic Lesions of Pancreas. Gastroenterology 2021; 160:1016-1018. [PMID: 33385429 DOI: 10.1053/j.gastro.2020.12.039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 12/28/2020] [Indexed: 12/02/2022]
Affiliation(s)
- Sukhwinder Kaur
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology and, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska.
| |
Collapse
|
26
|
Abstract
Pancreatic cystic neoplasms (PCNs) are being detected increasingly frequently due to the widespread use of high-resolution abdominal imaging modalities. Some subtypes of PCNs have the potential for malignant transformation. Therefore, accurate diagnosis of PCNs is crucial to determine whether surgical resection or surveillance is the best management strategy. However, the current cross-section imaging modalities are not accurate enough to enable definite diagnoses. In the last decade, EUS-based techniques have emerged, aiming to overcome the limitations of standard cross-section imaging modalities. These novel EUS-based techniques were primarily designed to acquire distinct images to make radiological diagnoses, collect cyst fluid to undergo biochemical or molecular analyses, and obtain tissue to conclude the pathological diagnoses. In this article, we present a comprehensive and critical review of these emerging EUS techniques for the diagnosis of PCNs, with emphasis being placed on the advantages, feasibilities, diagnostic performances, and limitations of these novel techniques.
Collapse
Affiliation(s)
- Liqi Sun
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Haojie Huang
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhendong Jin
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
27
|
Yan M, Shen M, Xu L, Huang J, He G, An M, Li X, Gao Z, Meng X. Inactivation of Pancreatic Stellate Cells by Exendin-4 Inhibits the Migration and Invasion of Pancreatic Cancer Cells. Onco Targets Ther 2020; 13:9455-9463. [PMID: 33061431 PMCID: PMC7522302 DOI: 10.2147/ott.s259853] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 08/16/2020] [Indexed: 12/17/2022] Open
Abstract
Background Pancreatic stellate cells (PSCs) are precursor cells of cancer-associated fibroblasts that promote tumor proliferation, invasion, and metastasis. The glucagon-like peptide-1 receptor agonist exendin-4 has been reported to exhibit anticancer effects against several tumor cells; however, the function and mechanism underlying the effects of exendin-4 on pancreatic cancer cells remain unclear. Methods Gene expression levels were determined using quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot assay. Cell viability, migration and invasion were assessed using the cell counting kit-8 (CCK-8), wound healing, and transwell assays, respectively. A xenografted tumor model was established in mouse to evaluate the effects of exendin-4 in vivo. Results Exendin-4 treatment led to the inactivation of PSCs and suppressed their proliferation and migration. Moreover, we also found that exendin-4 attenuated NF-κB-dependent SDF-1 secretion. Furthermore, pancreatic cancer cells incubated with conditioned medium obtained from exendin-4-treated PSCs showed a decreased ability to proliferate, migrate, and invade as compared to the control cells, which is similar to the effects induced by the CXCR4 inhibitor, AMD3100. Consistent with in vitro results, we also confirmed that exendin-4 indirectly targeted pancreatic cancer cells in vivo by attenuating the function of PSCs and suppressing the deposition of extracellular matrix. Conclusion These results revealed that exendin-4-treated PSCs could suppress pancreatic cancer cell proliferation and invasion, offering a potential strategy for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Meizhu Yan
- Department of Gastroenterology, Zhongshan Hospital Qingpu Branch, Fudan University, Shanghai 201700, People's Republic of China
| | - Manru Shen
- Department of Gastroenterology, Zhongshan Hospital Qingpu Branch, Fudan University, Shanghai 201700, People's Republic of China
| | - Linfang Xu
- Department of Gastroenterology, Zhongshan Hospital Qingpu Branch, Fudan University, Shanghai 201700, People's Republic of China
| | - Jiying Huang
- Department of Gastroenterology, Zhongshan Hospital Qingpu Branch, Fudan University, Shanghai 201700, People's Republic of China
| | - Guijun He
- Department of Gastroenterology, Zhongshan Hospital Qingpu Branch, Fudan University, Shanghai 201700, People's Republic of China
| | - Min An
- Department of Gastroenterology, Zhongshan Hospital Qingpu Branch, Fudan University, Shanghai 201700, People's Republic of China
| | - Xiaocui Li
- Department of Gastroenterology, Zhongshan Hospital Qingpu Branch, Fudan University, Shanghai 201700, People's Republic of China
| | - Zhenjun Gao
- Department of Gastroenterology, Zhongshan Hospital Qingpu Branch, Fudan University, Shanghai 201700, People's Republic of China
| | - Xin Meng
- Department of Hospital Infection Management, Zhongshan Hospital Qingpu Branch, Fudan University, Shanghai 201700, People's Republic of China
| |
Collapse
|