1
|
Xu X, Long C, Li M, Shen C, Ye Q, Li Y, Li H, Cao X, Ma J. Systematic review and meta-analysis: diagnostic accuracy of exosomes in pancreatic cancer. World J Surg Oncol 2025; 23:51. [PMID: 39953585 PMCID: PMC11827209 DOI: 10.1186/s12957-025-03666-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/19/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Early, non-invasive identification can generally enhance the survival rate for asymptomatic pancreatic cancer (PC). This systematic review and meta-analysis is conducted to evaluate the precision of diagnosing PC using serum and duodenal fluid exosomes. METHODS Following the guidelines of PRISMA (Preferred Reporting Items for Systematic Review and Meta-Analyses), searches were conducted in the PubMed, Embase, Cochrane Library, and Web of Science databases in April 2024. A study was considered appropriate if it provided diagnostic precision and accuracy for patients with pancreatic cancer. The combined diagnostic impact was assessed by calculating the area beneath the aggregated SROC curve, and the quality of the studies included was evaluated using the QUADAS-2 checklist. All statistical evaluations and graphical representations utilized STATA 14.0. RESULTS Employing the terms "exosomes" and "pancreatic cancer" along with the search methodology, research was conducted across PubMed, Web of Science, Cochrane, and Embase databases. A total of 1202 studies were extracted from the databases, out of which nine were ultimately selected based on specific inclusion and exclusion standards. Across eight studies, exosomes were isolated from serum, while in a different one, they were taken from duodenal fluid. This document conducts subgroup analyses focusing on various types of exosome biomarkers, their origins, isolation techniques, and methods for analyzing biomarkers. Within the subset of exosome biomarker types, the group with exosomal cell surface proteoglycan exhibited the greatest combined sensitivity (0.96 (95% CI = 0.81-0.99) and specificity (0.90 (95% CI = 0.83-0.95)). Additionally, the set of exosomal cell surface proteoglycans showed the highest aggregated diagnostic ratio (215.92), combined positive likelihood ratio (9.96), area under the curve (0.93), and kombiniertes negative Likelihood-Ratio (0.05). The combined sensitivity of serum-derived exosomes stood at (0.86 (95% CI = 0.77-0.92)), the collective specificity at (0.83 (95% CI = 0.77-0.89)), the aggregate positive likelihood ratio at (5.22), the combined diagnostic ratio at (31.48), the overall area beneath the curve at (0.91), and the combined negative likelihood ratio at (0.17). Within the subgroup examination of exosome isolation techniques, ultracentrifugation emerged as the most sensitive method (0.90 (95% CI = 0.74-0.97)), the most specific method (0.89 (95% CI = 0.83-0.93)), the top positive likelihood ratio (8.35), the highest diagnostic ratio (76.48), the largest combined curve area (0.92), and the smallest negative likelihood ratio (0.11) in the aggregated data. Within the subset of biomarker analysis methods, the aggregate sensitivity via qRT-PCR was (0.84 (95% CI = 0.74-0.90)), the collective specificity (0.78 (95% CI = 0.64-0.87)), the aggregate diagnostic ratio (18.11), the aggregate area under the curve (0.88), the aggregate positive likelihood ratio (3.77), and the combined negative likelihood ratio (0.21). CONCLUSION Overall, exosomes are still valuable in the diagnosis of pancreatic cancer. In subgroup analyses, the proteoglycan found on exosomal cell surfaces is highly valuable for diagnosing pancreatic cancer. The more frequent separation method used in the nine included studies was ultracentrifugation, and it did demonstrate good data. Nonetheless, to verify their practicality and usefulness in clinical environments, a significant amount of clinical trials are still necessary.
Collapse
Affiliation(s)
- Xinyi Xu
- Central Laboratory, The Second Affiliated Hospital of Kunming Medical University, 374, Dianmian Road, Kunming, 650101, China
| | - Chunyue Long
- Department of Clinical Laboratory, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Meng Li
- Central Laboratory, The Second Affiliated Hospital of Kunming Medical University, 374, Dianmian Road, Kunming, 650101, China
| | - Chen Shen
- Central Laboratory, The Second Affiliated Hospital of Kunming Medical University, 374, Dianmian Road, Kunming, 650101, China
| | - Qiuwen Ye
- Department of Hepatobiliary and Pancreatic Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, 519, Kunzhou Road, Kunming, 650105, China
| | - Yong Li
- Department of Hepatobiliary and Pancreatic Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, 519, Kunzhou Road, Kunming, 650105, China
| | - Hongyang Li
- Department of Hepatobiliary and Pancreatic Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, 519, Kunzhou Road, Kunming, 650105, China
| | - Xia Cao
- Central Laboratory, The Second Affiliated Hospital of Kunming Medical University, 374, Dianmian Road, Kunming, 650101, China.
| | - Jun Ma
- Department of Hepatobiliary and Pancreatic Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, 519, Kunzhou Road, Kunming, 650105, China.
| |
Collapse
|
2
|
Zhang X, Wei L, Li J, Deng Y, Xu W, Chen D, Li X. Influence of myosteatosis on survival of patients with pancreatic cancer: A systematic review and meta-analysis. iScience 2024; 27:111343. [PMID: 39640579 PMCID: PMC11617386 DOI: 10.1016/j.isci.2024.111343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 08/28/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024] Open
Abstract
The present meta-analysis aims to evaluate the impact of myosteatosis on overall survival (OS) and progression-free survival (PFS) in patients with pancreatic cancer (PC). A comprehensive literature search was conducted in the Medline, Web of Science, and Embase databases. The hazard ratio (HR) and corresponding 95% confidence interval (CI) for the association between myosteatosis and survival outcomes were pooled using a random-effects model. A total of 14 studies were included. The pooled analysis demonstrated that myosteatosis was significantly associated to poorer OS (HR: 1.50, 95% CI: 1.35-1.67, p < 0.001; I 2 = 0%). The subgroup analysis revealed consistent results across various study characteristics, including geographic regions, cancer stages, follow-up durations, and study quality. In addition, myosteatosis was associated to worse PFS (HR: 1.34, 95% CI: 1.15-1.57, p < 0.001; I 2 = 34%). The present meta-analysis indicates that myosteatosis is associated to significantly worse OS and PFS in patients with PC.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Gastroenterology, The Fourth Hospital of Changsha, Changsha City, Hunan Province 410006, P.R. China
| | - Licheng Wei
- Department of Gastroenterology, The Fourth Hospital of Changsha, Changsha City, Hunan Province 410006, P.R. China
| | - Jiangguo Li
- Department of Gastroenterology, The Fourth Hospital of Changsha, Changsha City, Hunan Province 410006, P.R. China
| | - Yuexia Deng
- Department of Gastroenterology, The Fourth Hospital of Changsha, Changsha City, Hunan Province 410006, P.R. China
| | - Wei Xu
- Department of Gastroenterology, The Fourth Hospital of Changsha, Changsha City, Hunan Province 410006, P.R. China
| | - Dongkui Chen
- Department of Gastroenterology, The Fourth Hospital of Changsha, Changsha City, Hunan Province 410006, P.R. China
| | - Xing Li
- Department of Critical Care Medicine, Changsha Hospital of Traditional Chinese Medicine (Changsha No. 8 Hospital), Changsha City, Hunan Province 410100, P.R. China
| |
Collapse
|
3
|
Elkady N, Elgendy W, Badr MT, Aiad H, Samara M, Badr NM. Evaluation of the diagnostic utility of NCOA3, Maspin and VHL protein expression in pancreatic ductal adenocarcinoma: An immunohistochemical study. Ann Diagn Pathol 2024; 73:152356. [PMID: 38901088 DOI: 10.1016/j.anndiagpath.2024.152356] [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: 05/02/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal tumor with a high mortality rate. The distinction between PDAC and chronic pancreatitis is sometimes challenging on routine histopathological examination, highlighting the need to identify biomarkers that can facilitate this distinction. This retrospective study was conducted to evaluate the diagnostic utility of nuclear receptor co-activator 3 (NCOA3), Maspin and Von Hippel-Lindau protein (VHL) immunostaining in PDAC. Eighty cases of PDAC, 46 cases of chronic pancreatitis and 53 normal pancreatic tissue were immunohistochemically assessed using NCOA3, Maspin and VHL antibodies on sections from a tissue microarray. NCOA3, Maspin and VHL were positive in 90 %, 100 % and 35 %, of PDAC cases respectively, whereas NCOA3, Maspin and VHL expressions were positive in 3.8 %, 0 and 100 % of normal pancreatic tissue and in 15.2 %, 21.7 % and 97.8 % of chronic pancreatitis cases respectively. Significant differences were observed between PDAC and other groups regarding NCOA3, Maspin and VHL expression (p < 0.001). The H scores of NCOA3, Maspin and VHL could significantly distinguish between PDAC and normal cases with high sensitivity (90 %, 100 % and 98.75 % respectively) and specificity (100 %, 100 % and 96.23 % respectively). Similar findings were found in the distinction between PDAC and chronic pancreatitis (Sensitivity: 90 %, 95.25 % and 98.75 %; specificity: 100 %, 100 % and 93.48 % for NCOA3, Maspin and VHL respectively). In conclusion, NCOA3 and Maspin were found to be significantly expressed in PDAC compared to non-tumorous tissue while VHL was significantly expressed in non-tumorous tissue. A panel of NCOA3, Maspin and VHL could potentially distinguish PDAC from non-tumorous pancreatic tissue.
Collapse
Affiliation(s)
- Noha Elkady
- Faculty of Medicine, Menoufia University, Egypt.
| | - Walaa Elgendy
- National Liver Institute, Menoufia University, Egypt
| | | | - Hayam Aiad
- Faculty of Medicine, Menoufia University, Egypt
| | - Manar Samara
- National Liver Institute, Menoufia University, Egypt
| | | |
Collapse
|
4
|
Knox L. Pancreatic Cancer: The Advanced Practitioner's Role in Early Diagnosis and Management. J Adv Pract Oncol 2024; 15:444-450. [PMID: 39830222 PMCID: PMC11741095 DOI: 10.6004/jadpro.2024.15.7.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025] Open
Abstract
Pancreatic cancer is one of the most fatal cancers in the United States. Currently, it is the third leading cause of cancer-related deaths, and it is estimated that by 2030, it will be the second leading cause of cancer-related deaths behind lung cancer. It has poor overall survival rates, even with aggressive treatment. Quality of life is low in this patient population, due to poor prognosis at diagnosis and complex symptomatology. The purpose of this article is to explore the role of the advanced practitioner in the diagnosis, treatment, and symptom management of pancreatic cancer.
Collapse
Affiliation(s)
- Lindsay Knox
- From Carson Newman University and Tennessee Cancer Specialists, Powell, Tennessee
| |
Collapse
|
5
|
Jiang H, Wang GT, Wang Z, Ma QY, Ma ZH. Resveratrol inhibits pancreatic cancer proliferation and metastasis by depleting senescent tumor-associated fibroblasts. World J Gastrointest Oncol 2024; 16:3980-3993. [PMID: 39350997 PMCID: PMC11438786 DOI: 10.4251/wjgo.v16.i9.3980] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/26/2024] [Accepted: 08/02/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND Pancreatic cancer, a formidable gastrointestinal neoplasm, is characterized by its insidious onset, rapid progression, and resistance to treatment, which often lead to a grim prognosis. While the complex pathogenesis of pancreatic cancer is well recognized, recent attention has focused on the oncogenic roles of senescent tumor-associated fibroblasts. However, their precise role in pancreatic cancer remains unknown. Resveratrol is a natural polyphenol known for its multifaceted biological actions, including antioxidative and neuroprotective properties, as well as its potential to inhibit tumor proliferation and migration. Our current investigation builds on prior research and reveals the remarkable ability of resveratrol to inhibit pancreatic cancer proliferation and metastasis. AIM To explore the potential of resveratrol in inhibiting pancreatic cancer by targeting senescent tumor-associated fibroblasts. METHODS Immunofluorescence staining of pancreatic cancer tissues revealed prominent coexpression of α-SMA and p16. HP-1 expression was determined using immunohistochemistry. Cells were treated with the senescence-inducing factors known as 3CKs. Long-term growth assays confirmed that 3CKs significantly decreased the CAF growth rate. Western blotting was conducted to assess the expression levels of p16 and p21. Immunofluorescence was performed to assess LaminB1 expression. Quantitative real-time polymerase chain reaction was used to measure the levels of several senescence-associated secretory phenotype factors, including IL-4, IL-6, IL-8, IL-13, MMP-2, MMP-9, CXCL1, and CXCL12. A scratch assay was used to assess the migratory capacity of the cells, whereas Transwell assays were used to evaluate their invasive potential. RESULTS Specifically, we identified the presence of senescent tumor-associated fibroblasts within pancreatic cancer tissues, linking their abundance to cancer progression. Intriguingly, Resveratrol effectively eradicated these fibroblasts and hindered their senescence, which consequently impeded pancreatic cancer progression. CONCLUSION This groundbreaking discovery reinforces Resveratrol's stature as a potential antitumor agent and positions senescent tumor-associated fibroblasts as pivotal contenders in future therapeutic strategies against pancreatic cancer.
Collapse
Affiliation(s)
- He Jiang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Guo-Tai Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Department of Hepatobiliary Surgery, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang 712000, Shaanxi Province, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Qing-Yong Ma
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Zhen-Hua Ma
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| |
Collapse
|
6
|
McIntyre CA, Grimont A, Park J, Meng Y, Sisso WJ, Seier K, Jang GH, Walch H, Aveson VG, Falvo DJ, Fall WB, Chan CW, Wenger A, Ecker BL, Pulvirenti A, Gelfer R, Zafra MP, Schultz N, Park W, O'Reilly EM, Houlihan SL, Alonso A, Hissong E, Church GM, Mason CE, Siolas D, Notta F, Gonen M, Dow LE, Jarnagin WR, Chandwani R. Distinct clinical outcomes and biological features of specific KRAS mutants in human pancreatic cancer. Cancer Cell 2024; 42:1614-1629.e5. [PMID: 39214094 PMCID: PMC11419252 DOI: 10.1016/j.ccell.2024.08.002] [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: 12/01/2023] [Revised: 07/09/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024]
Abstract
KRAS mutations in pancreatic ductal adenocarcinoma (PDAC) are suggested to vary in oncogenicity but the implications for human patients have not been explored in depth. We examined 1,360 consecutive PDAC patients undergoing surgical resection and find that KRASG12R mutations are enriched in early-stage (stage I) disease, owing not to smaller tumor size but increased node-negativity. KRASG12R tumors are associated with decreased distant recurrence and improved survival as compared to KRASG12D. To understand the biological underpinnings, we performed spatial profiling of 20 patients and bulk RNA-sequencing of 100 tumors, finding enhanced oncogenic signaling and epithelial-mesenchymal transition (EMT) in KRASG12D and increased nuclear factor κB (NF-κB) signaling in KRASG12R tumors. Orthogonal studies of mouse KrasG12R PDAC organoids show decreased migration and improved survival in orthotopic models. KRAS alterations in PDAC are thus associated with distinct presentation, clinical outcomes, and biological behavior, highlighting the prognostic value of mutational analysis and the importance of articulating mutation-specific PDAC biology.
Collapse
Affiliation(s)
- Caitlin A McIntyre
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Adrien Grimont
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Jiwoon Park
- Department of Physiology, Biophysics, and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Yinuo Meng
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Whitney J Sisso
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Kenneth Seier
- Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gun Ho Jang
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Henry Walch
- Marie-Josee and Henry R Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Victoria G Aveson
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - David J Falvo
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - William B Fall
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Christopher W Chan
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Andrew Wenger
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Brett L Ecker
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Alessandra Pulvirenti
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rebecca Gelfer
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Maria Paz Zafra
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Nikolaus Schultz
- Marie-Josee and Henry R Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wungki Park
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; David M. Rubinstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eileen M O'Reilly
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; David M. Rubinstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Shauna L Houlihan
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Alicia Alonso
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Erika Hissong
- Department of Pathology, Weill Cornell Medicine, New York, NY, USA
| | - George M Church
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Christopher E Mason
- Department of Physiology, Biophysics, and Systems Biology, Weill Cornell Medicine, New York, NY, USA; WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, USA
| | - Despina Siolas
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Faiyaz Notta
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Mithat Gonen
- Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lukas E Dow
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - William R Jarnagin
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; David M. Rubinstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rohit Chandwani
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; David M. Rubinstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
7
|
Seiki Y, Ikezawa K, Watsuji K, Urabe M, Kai Y, Takada R, Yamai T, Mukai K, Nakabori T, Uehara H, Ishibashi M, Ohkawa K. Zinc supplementation for dysgeusia in patients with unresectable pancreatic cancer. Int J Clin Oncol 2024; 29:1173-1181. [PMID: 38724773 DOI: 10.1007/s10147-024-02544-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 04/30/2024] [Indexed: 07/26/2024]
Abstract
BACKGROUND Although patients with advanced pancreatic cancer (PC) often experience dysgeusia with zinc deficiency during chemotherapy, data on zinc supplementation for dysgeusia and its effects on nutritional status are scarce. We aimed to examine the efficacy of zinc supplementation in patients with advanced PC. METHODS Thirty-three patients with unresectable PC who presented with dysgeusia and zinc deficiency during chemotherapy and received zinc acetate hydrate between January 2018 and December 2022 were included. We evaluated the changes in serum zinc levels and the improvement in dysgeusia. Among the 26 patients who received zinc supplementation for 12 weeks, we also compared patient characteristics and changes in serum zinc and albumin levels between patients who showed improvement in dysgeusia (effective group) and those who did not (non-effective group). RESULTS The serum zinc level increased significantly after zinc supplementation (median: 60 µg/dL at baseline, 99.5 µg/dL at 4 weeks, 101 µg/dL at 8 weeks and 101 µg/dL at 12 weeks). The rate of improvement in dysgeusia increased over time (18.2% at 4 weeks, 33.3% at 8 weeks, and 42.4% at 12 weeks). Comparing the effective group and non-effective group revealed that while the median serum albumin level of the effective group did not change, the non-effective group showed a significant decrease from baseline to 12 weeks (3.2 g/dL to 3.0 g/dL, p = 0.03). CONCLUSION Zinc supplementation significantly increased serum zinc levels, improving dysgeusia. Zinc supplementation might also contribute to maintaining nutritional status in patients with unresectable PC.
Collapse
Affiliation(s)
- Yusuke Seiki
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Kenji Ikezawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan.
| | - Ko Watsuji
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Makiko Urabe
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Yugo Kai
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Ryoji Takada
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Takuo Yamai
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Kaori Mukai
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Tasuku Nakabori
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Hiroyuki Uehara
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Miki Ishibashi
- Department of Dentistry, Osaka International Cancer Institute, Osaka, Japan
| | - Kazuyoshi Ohkawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| |
Collapse
|
8
|
Wei C, Li YC, Jin HT, Li DF, Wang LS, Yao J. Early detection of pancreatic cancer in patients with recurrent pancreatitis: A case report. World J Clin Cases 2024; 12:3936-3941. [PMID: 38994295 PMCID: PMC11235423 DOI: 10.12998/wjcc.v12.i19.3936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/24/2024] [Accepted: 05/17/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Pancreatic cancer presents a challenge with its low early diagnosis and treatment rates, leading to high metastasis and mortality rates. The median survival time for advanced pancreatic cancer is a mere 3 months. However, there's hope: small pancreatic cancers diagnosed at an early stage (T1) or those less than or equal to 1 cm in diameter boast an impressive 5-year survival rate of nearly 100%. This underscores the critical importance of early pancreatic cancer detection for significantly improving prognosis. CASE SUMMARY Pancreatic cancer, a malignant tumor of the digestive tract, poses challenges in both diagnosis and treatment due to its occult and atypical clinical symptoms. Clinically, patients with recurrent pancreatitis should be vigilant, as it may be indicative of pancreatic cancer, particularly in middle-aged and elderly patients. Here, we presented the case of a patient who experienced recurrent acute pancreatitis within a span of 2 months. During the initial episode of pancreatitis, routine imaging failed to identify the cause of pancreatic cancer. However, upon recurrence of acute pancreatitis, endoscopic ultrasonography (EUS) revealed a space-occupying lesion approximately 1 cm in size in the pancreatic body. Subsequent EUS coupled with fine-needle aspiration examination demonstrated atypical pancreatic gland epithelium. Ultimately, the patient underwent surgery and was diagnosed with an intraductal papillary mucinous tumor of the pancreas (severe epithelial dysplasia, focal cancer). CONCLUSION We recommend EUS for patients with recurrent pancreatitis of unknown etiology to exclude early pancreatic cancer.
Collapse
Affiliation(s)
- Cheng Wei
- Department of Gastroenterology, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong Province, China
| | - Yi-Chen Li
- Department of Emergency, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong Province, China
| | - Hong-Tao Jin
- Department of Pathology, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong Province, China
| | - De-Feng Li
- Department of Gastroenterology, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong Province, China
| | - Li-Sheng Wang
- Department of Gastroenterology, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong Province, China
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong Province, China
| |
Collapse
|
9
|
Wang L, Wang L, Sun X, Fu L, Wang X, Wang X, Chen L, Huang Y. Detection of uridine diphosphate glucuronosyltransferase 1A1 for pancreatic cancer imaging and treatment via a "turn-on" fluorescent probe. Analyst 2024; 149:2877-2886. [PMID: 38567989 DOI: 10.1039/d4an00035h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
Uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) is expressed ubiquitously in cancer cells and can metabolize exogenous substances. Studies show higher UGT1A1 levels in pancreatic cancer cells than normal cells. Therefore, we need a method to monitor the activity level of UGT1A1 in pancreatic cancer cells and in vivo. Here, we report a fluorescent probe, BCy-panc, for UGT1A1 imaging in cells and in vivo. Compared with other molecular probes, this probe is readily prepared, with high selectivity and sensitivity for the detection of UGT1A1. Our results show that BCy-panc rapidly detects UGT1A1 in pancreatic cancer. In addition, there is an urgent need for evidence to clarify the relationship between UGT1A1 and pancreatic cancer development. The present investigation found that the increase of UGT1A1 by chrysin was effective in inducing apoptosis in pancreatic cancer cells. These results indicate that the synergistic effect of chrysin and cisplatin at the cellular level is superior to that of cisplatin alone. The UGT1A1 level may be a biomarker for early diagnosis of cancer. Meanwhile, UGT1A1 plays a crucial role in pancreatic cancer, and the combination of chrysin and cisplatin may provide effective ideas for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Lingxiao Wang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
| | - Lingyun Wang
- Jinan Zhangqiu District People's Hospital, Jinan 250000, China
| | - Xiao Sun
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
| | - Lili Fu
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Shandong Key Laboratory of Coastal Environmental Processes, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.
| | - Xinlei Wang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
| | - Xiaoyan Wang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Shandong Key Laboratory of Coastal Environmental Processes, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.
| | - Lingxin Chen
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Shandong Key Laboratory of Coastal Environmental Processes, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Yan Huang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Shandong Key Laboratory of Coastal Environmental Processes, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.
| |
Collapse
|
10
|
Koziol-Bohatkiewicz P, Liberda-Matyja D, Wrobel TP. Fast cancer imaging in pancreatic biopsies using infrared imaging. Analyst 2024; 149:1799-1806. [PMID: 38385553 DOI: 10.1039/d3an01555f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Pancreatic cancer, particularly Pancreatic ductal adenocarcinoma, remains a highly lethal form of cancer with limited early diagnosis and treatment options. Infrared (IR) spectroscopy, combined with machine learning, has demonstrated great potential in detecting various cancers. This study explores the translation of a diagnostic model from Fourier Transform Infrared to Quantum Cascade Laser (QCL) microscopy for pancreatic cancer classification. Furthermore, QCL microscopy offers faster measurements with selected frequencies, improving clinical feasibility. Thus, the goals of the study include establishing a QCL-based model for pancreatic cancer classification and creating a fast surgical margin detection model using reduced spectral information. The research involves preprocessing QCL data, training Random Forest (RF) classifiers, and optimizing the selection of spectral features for the models. Results demonstrate successful translation of the diagnostic model to QCL microscopy, achieving high predictive power (AUC = 98%) in detecting cancerous tissues. Moreover, a model for rapid surgical margin recognition, based on only a few spectral frequencies, is developed with promising differentiation between benign and cancerous regions. The findings highlight the potential of QCL microscopy for efficient pancreatic cancer diagnosis and surgical margin detection within clinical timeframes of minutes per surgical resection tissue.
Collapse
Affiliation(s)
- Paulina Koziol-Bohatkiewicz
- Solaris National Synchrotron Radiation Centre, Jagiellonian University, Czerwone Maki 98, 30-392, Krakow, Poland.
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Łojasiewicza 11, 30-348 Krakow, Poland
| | - Danuta Liberda-Matyja
- Solaris National Synchrotron Radiation Centre, Jagiellonian University, Czerwone Maki 98, 30-392, Krakow, Poland.
- Jagiellonian University, Doctoral School of Exact and Natural Sciences, Prof. St. Łojasiewicza 11, PL30348, Cracow, Poland
| | - Tomasz P Wrobel
- Solaris National Synchrotron Radiation Centre, Jagiellonian University, Czerwone Maki 98, 30-392, Krakow, Poland.
| |
Collapse
|
11
|
Scherübl H. [Early detection of sporadic pancreatic cancer]. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2024; 62:412-419. [PMID: 37827502 DOI: 10.1055/a-2114-9847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
The incidence of pancreatic cancer is rising. At present, pancreatic cancer is the third most common cancer-causing death in Germany, but it is expected to become the second in 2030 and finally the leading cause of cancer death in 2050. Pancreatic ductal adenocarcinoma (PC) is generally diagnosed at advanced stages, and 5-year-survival has remained poor. Early detection of sporadic PC at stage IA, however, can yield a 5-year-survival rate of about 80%. Early detection initiatives aim at identifying persons at high risk. People with new-onset diabetes at age 50 or older have attracted much interest. Novel strategies regarding how to detect sporadic PC at an early stage are being discussed.
Collapse
Affiliation(s)
- Hans Scherübl
- Klinik für Innere Medizin; Gastroenterol., GI Onkol. u. Infektiol., Vivantes Klinikum Am Urban, Berlin, Germany
- Akademisches Lehrkrankenhaus der Charité, Berlin, Germany
| |
Collapse
|
12
|
Zhong JJ, Ye YQ. Construction and validation of a nomogram model for predicting early death in patients with metastatic pancreatic adenocarcinoma based on SEER database. Shijie Huaren Xiaohua Zazhi 2023; 31:577-588. [DOI: 10.11569/wcjd.v31.i14.577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/07/2023] [Accepted: 07/20/2023] [Indexed: 07/28/2023] Open
Abstract
BACKGROUND Pancreatic adenocarcinoma is a highly aggressive malignancy that presents a considerable risk of early death (survival time ≤ 3 mo). As such, it is of great significance to develop an effective nomogram for predicting the likelihood of early death in patients with metastatic pancreatic adenocarcinoma.
AIM To construct and validate a predictive nomogram model for early death in patients with metastatic pancreatic adenocar-cinoma.
METHODS We extracted data from the SEER database of 18603 eligible patients with metastatic pancreatic adenocarcinoma from 2010 to 2015, and randomly divided them into training and validation cohorts in a 7:3 ratio. Univariate and multivariate logistic regression analyses were performed on the training cohort to identify the risk factors for early death, based on which a nomogram was constructed. The performance of the nomogram was verified by receiver operating characteristic (ROC) curve and calibration curve analyses in both the training and validation cohorts. The clinical practicability of the nomogram was evaluated by decision curve analysis (DCA).
RESULTS Age, sex, primary site, grade, T stage, N stage, brain metastasis, bone metastasis, liver metastasis, lung metastasis, surgery, radiotherapy, and chemotherapy were identified as independent risk factors for early death in patients with metastatic pancreatic adenocarcinoma. Based on these variables, a nomogram was constructed. The areas under the ROC curves of the nomogram in the training and validation cohorts were 0.810 (95% confidence interval [CI]: 0.802-0.811) and 0.802 (95%CI: 0.790-0.813), respectively, indicating good discrimination. The calibration curves showed good calibration degrees in both cohorts, and the DCA results demonstrated that the nomogram had better clinical net benefit in predicting early mortality compared with TNM stage.
CONCLUSION The constructed nomogram has good predictive ability for early death in patients with metastatic pancreatic adeno-carcinoma. This will help clinicians develop individualized treatment plans for these patients.
Collapse
Affiliation(s)
- Jia-Jun Zhong
- Department of Gastroenterology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, Jiangxi Province, China
| | - Yan-Qing Ye
- Department of Gastroenterology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, Jiangxi Province, China
| |
Collapse
|
13
|
Liu S, Liu P, Fei X, Zhu C, Hou J, Wang X, Pan Y. Analysis and validation of the potential of the MYO1E gene in pancreatic adenocarcinoma based on a bioinformatics approach. Oncol Lett 2023; 26:285. [PMID: 37274465 PMCID: PMC10236097 DOI: 10.3892/ol.2023.13871] [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: 01/23/2023] [Accepted: 03/22/2023] [Indexed: 06/06/2023] Open
Abstract
Pancreatic adenocarcinoma (PAAD) is a common digestive cancer, and its prognosis is poor. Myosin 1E (MYO1E) is a class I myosin family member whose expression and function have not been reported in PAAD. In the present study, bioinformatics analysis was used to explore the expression levels of MYO1E in PAAD and its prognostic value, and the immunological role of MYO1E in PAAD was analyzed. The study revealed that a variety of malignancies have substantially increased MYO1E expression. Further investigation demonstrated that PAAD tissues exhibited greater levels of MYO1E mRNA and protein expression than normal tissues. High MYO1E expression is associated with poor prognosis in patients with PAAD. MYO1E expression was also associated with pathological stage in patients with PAAD. Functional enrichment analysis demonstrated that MYO1E was linked to multiple tumor-related mechanisms in PAAD. The pancreatic adenocarcinoma tumor microenvironment (TME) was analyzed and it was revealed that MYO1E expression was positively associated with tumor immune cell infiltration. In addition, MYO1E was closely associated with some tumor chemokines/receptors and immune checkpoints. In vitro experiments revealed that the suppression of MYO1E expression could inhibit pancreatic adenocarcinoma cell proliferation, invasion and migration. Through preliminary analysis, the present study evaluated the potential function of MYO1E in PAAD and its function in TME, and MYO1E may become a potential biomarker for PAAD.
Collapse
Affiliation(s)
- Songbai Liu
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| | - Peng Liu
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| | - Xiaobin Fei
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| | - Changhao Zhu
- Department of Hepatobiliary Surgery, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| | - Junyi Hou
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| | - Xing Wang
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
- Department of Hepatobiliary Surgery, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| | - Yaozhen Pan
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
- Department of Hepatobiliary Surgery, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| |
Collapse
|
14
|
Liu C, Du J, Zheng J, Zhang R, Zhu J, Xing B, Dong L, Zhou Q, Yao X, Gao S, Wang Y, Ren Y, Zhou X. The role of BHLHE40 in clinical features and prognosis value of PDAC by comprehensive analysis and in vitro validation. Front Oncol 2023; 13:1151321. [PMID: 37377917 PMCID: PMC10291124 DOI: 10.3389/fonc.2023.1151321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the leading cause of cancer-related mortality, primarily due to the abundance of cancer-associated fibroblasts (CAFs), depleted effector T cells, and increased tumor cell stemness; hence, there is an urgent need for efficient biomarkers with prognostic and therapeutic potential. Here, we identified BHLHE40 as a promising target for PDAC through comprehensive analysis and weighted gene coexpression network analysis of RNA sequencing data and public databases, taking into account the unique characteristics of PDAC such as cancer-associated fibroblasts, infiltration of effector T cells, and tumor cell stemness. Additionally, we developed a prognostic risk model based on BHLHE40 and three other candidate genes (ITGA2, ITGA3, and ADAM9) to predict outcomes in PDAC patients. Furthermore, we found that the overexpression of BHLHE40 was significantly associated with T stage, lymph node metastasis, and American Joint Committee on Cancer (AJCC) stage in a cohort of 61 PDAC patients. Moreover, elevated expression levels of BHLHE40 were validated to promote epithelial-mesenchymal transition (EMT) and stemness-related proteins in BXPC3 cell lines. Compared to the parent cells, BXPC3 cells with BHLHE40 overexpression showed resistance to anti-tumor immunity when co-cultured with CD8+ T cells. In summary, these findings suggest that BHLHE40 is a highly effective biomarker for predicting prognosis in PDAC and holds great promise as a target for cancer therapy.
Collapse
Affiliation(s)
- Chao Liu
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer; Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Jiang Du
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer; Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Jianwei Zheng
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer; Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Ruizhe Zhang
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer; Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Jialin Zhu
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Bofan Xing
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer; Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Lin Dong
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer; Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Qianqian Zhou
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer; Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Xiaofeng Yao
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer; Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Song Gao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Yu Wang
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer; Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Yu Ren
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xuan Zhou
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer; Tianjin’s Clinical Research Center for Cancer, Tianjin, 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
|