201
|
Pizzato M, La Vecchia C, Malvezzi M, Levi F, Boffetta P, Negri E, Dalmartello M. Cancer mortality and predictions for 2022 in selected Australasian countries, Russia, and Ukraine with a focus on colorectal cancer. Eur J Cancer Prev 2023; 32:18-29. [PMID: 35822596 DOI: 10.1097/cej.0000000000000762] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE We aimed at predicting cancer mortality rates for the current year for the Russian Federation, Ukraine, Israel, Hong Kong SAR, Japan, Korea, and Australia, with a focus on colorectal cancer. METHODS We retrieved official death certification and population data from the WHO and the United Nations Population Division databases. We analyzed mortality for all cancers combined and for 10 major cancer sites from 1970 to 2019, or the latest available year. We predicted numbers of deaths and age-standardized mortality rates for 2022 using Poisson joinpoint regression models. We estimated the number of averted deaths over the period 1994-2022 because of the decline in mortality rates. RESULTS Total cancer mortality declined in all countries and both sexes. Russia had the highest total cancer predicted rates for 2022: 156.4/100 000 (world standard) in men and 81.4 in women; the lowest rates were reported in Israeli men (90.6/100 000) and Korean women (44.5/100 000). Between 1994 and 2022, a total of 1 487 000 deaths were estimated to be avoided in Russia, 502 000 in Ukraine, 58 000 in Israel, 102 000 in Hong Kong SAR, 1 020 000 in Japan, 533 000 in the Republic of Korea, and 263 000 in Australia. Colorectal cancer mortality trends were downward for the last decades with favorable predictions for 2022 in both sexes. CONCLUSION In the countries considered, predicted downward trends started later and were less marked than those in the European Union and the USA. Despite overall favorable predictions, colorectal cancer remains one of the major causes of cancer mortality.
Collapse
Affiliation(s)
- Margherita Pizzato
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Carlo La Vecchia
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Matteo Malvezzi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Fabio Levi
- Department of Epidemiology and Health Services Research, Unisanté, University of Lausanne, Lausanne, Switzerland
| | - Paolo Boffetta
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, New York, USA
| | - Eva Negri
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Michela Dalmartello
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
202
|
Boonhat H, Lin RT, Lin JT. Association between residential exposure to petrochemical industrial complexes and pancreatic cancer: a systematic review and meta-analysis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2023; 33:116-127. [PMID: 34930088 DOI: 10.1080/09603123.2021.2007226] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/11/2021] [Indexed: 06/14/2023]
Abstract
Increased in the global demand-expansion of the petrochemical industry is a possible environmental risk factor pancreatic cancer among residents living close to petrochemical complexes. This meta-analysis aimed to estimate the pooled risk of pancreatic cancer among residents living near petrochemical industrial complexes. We systematically searched and reviewed published studies in six databases based on the inclusion criteria derived from the population, exposure, comparator, and outcomes framework (population: general population; exposure: residence near petrochemical industrial complexes/living in cities with petrochemical industrial complexes; comparators: residents living farther away from petrochemical industrial complexes/living in cities without petrochemical industrial complexes; outcome: pancreatic cancer). We identified seven studies, covering 1,605,568 residents. Pooled analysis showed a significantly higher risk of pancreatic cancer among residents living near petrochemical industrial complexes (relative risk [RR] = 1.31, 95% confidence interval [CI] = 1.21-1.42) than those living farther away from petrochemical industrial complexes. Such effect was higher in female residents (RR = 1.34, 95% CI = 1.18-1.53) than in male residents (RR = 1.26, 95% CI = 1.12-1.41). This study suggests that exposure to petrochemical industry-related activities should be recognized as a risk factor for pancreatic cancer among residents living near petrochemical industrial complexes.
Collapse
Affiliation(s)
- Hathaichon Boonhat
- Graduate Institute of Public Health, College of Public Health, China Medical University, Taichung, Taiwan
| | - Ro-Ting Lin
- Department of Occupational Safety and Health, College of Public Health, China Medical University, Taichung Taiwan
| | - Jaw-Town Lin
- Department of Internal Medicine, Digestive Medicine Center, China Medical University Hospital, Taichung Taiwan
| |
Collapse
|
203
|
Gram IT, Park SY, Wilkens LR, Le Marchand L, Setiawan VW. Smoking and pancreatic cancer: a sex-specific analysis in the Multiethnic Cohort study. Cancer Causes Control 2023; 34:89-100. [PMID: 36253659 PMCID: PMC9816198 DOI: 10.1007/s10552-022-01637-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/23/2022] [Indexed: 01/11/2023]
Abstract
PURPOSE To examine whether the detrimental smoking-related association with pancreatic cancer (PC) is the same for women as for men. METHODS We analyzed data from 192,035 participants aged 45-75 years, enrolled in the Multiethnic Cohort study (MEC) in 1993-1996. We identified PC cases via linkage to the Hawaii and California Surveillance, Epidemiology, and End Results Program cancer registries through December 2017. RESULTS During a mean follow-up of 19.2 years, we identified 1,936 incident PC cases. Women smokers smoked on average less than men smokers. In multivariate Cox regression models, as compared with sex-specific never smokers, current smokers had a similar elevated risk of PC for women, hazard ratio (HR) 1.49 (95% CI 1.24, 1.79) and as for men, HR 1.48 (95% CI 1.22, 1.79) (pheterogeneity: 0.79). Former smokers showed a decrease in risk of PC for men within 5 years, HR 0.74 (95% CI 0.57, 0.97) and for women within 10 years after quitting, HR 0.70 (95% CI 0.50, 0.96), compared with their sex-specific current smokers. Both sexes showed a consistent, strong, positive dose-response association with PC for the four measures (age at initiation, duration, number of cigarettes per day, number of pack-years) of smoking exposure among current smokers and an inverse association for years of quitting and age at smoking cessation among former smokers (all ptrend's < 0.001). CONCLUSION Although MEC women smoke on average less than their men counterparts, the smoking-related increase in PC risk and the benefits of cessation seem to be of similar magnitudes for women as for men.
Collapse
Affiliation(s)
- Inger T Gram
- Department of Community Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, Postboks 6050 Langnes, 9037, Tromsø, Norway.
| | - Song-Yi Park
- Population Sciences in the Pacific Program, University of Hawai'i Cancer Center, Honolulu, HI, USA
| | - Lynne R Wilkens
- Population Sciences in the Pacific Program, University of Hawai'i Cancer Center, Honolulu, HI, USA
| | - Loïc Le Marchand
- Population Sciences in the Pacific Program, University of Hawai'i Cancer Center, Honolulu, HI, USA
| | - Veronica Wendy Setiawan
- Department of Population and Public Health Sciences, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
204
|
Xu X, Yu Y, Yang L, Wang B, Fan Y, Ruan B, Zhang X, Dai H, Mei W, Jie W, Zheng S. Integrated analysis of Dendrobium nobile extract Dendrobin A against pancreatic ductal adenocarcinoma based on network pharmacology, bioinformatics, and validation experiments. Front Pharmacol 2023; 14:1079539. [PMID: 36937875 PMCID: PMC10014786 DOI: 10.3389/fphar.2023.1079539] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/20/2023] [Indexed: 03/05/2023] Open
Abstract
Background: Dendrobium nobile (D. nobile), a traditional Chinese medicine, has received attention as an anti-tumor drug, but its mechanism is still unclear. In this study, we applied network pharmacology, bioinformatics, and in vitro experiments to explore the effect and mechanism of Dendrobin A, the active ingredient of D. nobile, against pancreatic ductal adenocarcinoma (PDAC). Methods: The databases of SwissTargetPrediction and PharmMapper were used to obtain the potential targets of Dendrobin A, and the differentially expressed genes (DEGs) between PDAC and normal pancreatic tissues were obtained from The Cancer Genome Atlas and Genotype-Tissue Expression databases. The protein-protein interaction (PPI) network for Dendrobin A anti-PDAC targets was constructed based on the STRING database. Molecular docking was used to assess Dendrobin A anti-PDAC targets. PLAU, one of the key targets of Dendrobin A anti-PDAC, was immunohistochemically stained in clinical tissue arrays. Finally, in vitro experiments were used to validate the effects of Dendrobin A on PLAU expression and the proliferation, apoptosis, cell cycle, migration, and invasion of PDAC cells. Results: A total of 90 genes for Dendrobin A anti-PDAC were screened, and a PPI network for Dendrobin A anti-PDAC targets was constructed. Notably, a scale-free module with 19 genes in the PPI indicated that the PPI is highly credible. Among these 19 genes, PLAU was positively correlated with the cachexia status while negatively correlated with the overall survival of PDAC patients. Through molecular docking, Dendrobin A was found to bind to PLAU, and the Dendrobin A treatment led to an attenuated PLAU expression in PDAC cells. Based on clinical tissue arrays, PLAU protein was highly expressed in PDAC cells compared to normal controls, and PLAU protein levels were associated with the differentiation and lymph node metastatic status of PDAC. In vitro experiments further showed that Dendrobin A treatment significantly inhibited the proliferation, migration, and invasion, inducing apoptosis and arresting the cell cycle of PDAC cells at the G2/M phase. Conclusion: Dendrobin A, a representative active ingredient of D. nobile, can effectively fight against PDAC by targeting PLAU. Our results provide the foundation for future PDAC treatment based on D. nobile.
Collapse
Affiliation(s)
- Xiaoqing Xu
- Department of Oncology of the First Affiliated Hospital & Cancer Institute, Hainan Medical University, Haikou, China
| | - Yaping Yu
- Department of Oncology of the First Affiliated Hospital & Cancer Institute, Hainan Medical University, Haikou, China
| | - Li Yang
- Key Laboratory of Natural Products Research and Development from Li Folk Medicine of Hainan Province, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Bingshu Wang
- Department of Oncology of the First Affiliated Hospital & Cancer Institute, Hainan Medical University, Haikou, China
| | - Yonghao Fan
- Department of Oncology of the First Affiliated Hospital & Cancer Institute, Hainan Medical University, Haikou, China
| | - Banzhan Ruan
- Department of Oncology of the First Affiliated Hospital & Cancer Institute, Hainan Medical University, Haikou, China
| | - Xiaodian Zhang
- Department of Oncology of the First Affiliated Hospital & Cancer Institute, Hainan Medical University, Haikou, China
| | - Haofu Dai
- Key Laboratory of Natural Products Research and Development from Li Folk Medicine of Hainan Province, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Wenli Mei
- Key Laboratory of Natural Products Research and Development from Li Folk Medicine of Hainan Province, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
- *Correspondence: Wenli Mei, ; Wei Jie, ; Shaojiang Zheng,
| | - Wei Jie
- Department of Oncology of the First Affiliated Hospital & Cancer Institute, Hainan Medical University, Haikou, China
- *Correspondence: Wenli Mei, ; Wei Jie, ; Shaojiang Zheng,
| | - Shaojiang Zheng
- Department of Oncology of the First Affiliated Hospital & Cancer Institute, Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education & Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province & Hainan Women and Children’s Medical Center, Hainan Medical University, Haikou, China
- *Correspondence: Wenli Mei, ; Wei Jie, ; Shaojiang Zheng,
| |
Collapse
|
205
|
S100A10 Promotes Pancreatic Ductal Adenocarcinoma Cells Proliferation, Migration and Adhesion through JNK/LAMB3-LAMC2 Axis. Cancers (Basel) 2022; 15:cancers15010202. [PMID: 36612197 PMCID: PMC9818352 DOI: 10.3390/cancers15010202] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive tumors, characterized by diagnosis at an advanced stage and a poor prognosis. As a member of the S100 protein family, S100A10 regulates multiple biological functions related to cancer progression and metastasis. However, the role of S100A10 in PDAC is still not completely elucidated. In this study, we reported that S100A10 was significantly up-regulated in PDAC tissue and associated with a poor prognosis by integrated bioinformatic analysis and human PDAC tissue samples. In vitro, down-regulation of S100A10 reduced the proliferation, migration, and adhesion of PDAC cell lines, whereas up-regulation of S100A10 showed the opposite effect. Furthermore, LAMB3 was proved to be activated by S100A10 using RNA-sequencing and western blotting. The effect of LAMB3 on the proliferation, migration, and adhesion of PDAC cells was similar to that of S100A10. Up-regulation or down-regulation of LAMB3 could reverse the corresponding effect of S100A10. Moreover, we validated S100A10 activates LAMB3 through the JNK pathway, and LAMB3 was further proved to interact with LAMC2. Mice-bearing orthotopic pancreatic tumors showed that S100A10 knocked-down PANC-1 cells had a smaller tumor size than the control group. In conclusion, S100A10 promotes PDAC cells proliferation, migration, and adhesion through JNK/LAMB3-LAMC2 axis.
Collapse
|
206
|
An Efficient, Short Stimulus PANC-1 Cancer Cell Ablation and Electrothermal Therapy Driven by Hydrophobic Interactions. Pharmaceutics 2022; 15:pharmaceutics15010106. [PMID: 36678734 PMCID: PMC9867450 DOI: 10.3390/pharmaceutics15010106] [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: 10/29/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/31/2022] Open
Abstract
Promising results in clinical studies have been demonstrated by the utilization of electrothermal agents (ETAs) in cancer therapy. However, a difficulty arises from the balance between facilitating the degradation of ETAs, and at the same time, increasing the electrothermal performance/stability required for highly efficient treatment. In this study, we controlled the thermal signature of the MoS2 by harnessing MoS2 nanostructures with M13 phage (MNM) via the structural assembling (hydrophobic interaction) phenomena and developed a combined PANC-1 cancer cell-MNM alternating current (AC)-stimulus framework for cancer cell ablation and electrothermal therapy. A percentage decrease in the cell viability of ~23% was achieved, as well as a degradation time of 2 weeks; a stimulus length of 100 μs was also achieved. Molecular dynamics (MD) simulations revealed the assembling kinetics in integrated M13 phage-cancer cell protein systems and the structural origin of the hydrophobic interaction-enabled increase in thermal conduction. This study not only introduced an 'ideal' agent that avoided the limitations of ETAs but also provided a proof-of-concept application of MoS2-based materials in efficacious cancer therapy.
Collapse
|
207
|
Tang Z, Xu W, Zhang M. Association between type 2 diabetes and 5-year overall survival in early-stage pancreatic cancer: a retrospective cohort study. PeerJ 2022; 10:e14538. [PMID: 36530401 PMCID: PMC9753753 DOI: 10.7717/peerj.14538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/17/2022] [Indexed: 12/14/2022] Open
Abstract
Background This study examined the association between type 2 diabetes mellitus (T2DM) and 5-year overall survival (OS) in patients with pancreatic cancer (PC). Methods This retrospective cohort study included patients diagnosed with stage I/II PC at Shengjing Hospital of China Medical University from January 2012 to December 2017. All patients had pancreatic ductal adenocarcinoma or its subtypes. The outcome was the 5-year OS rate based on data from the patient charts. Data analysis was performed using SPSS 22.0. Results A total of 238 patients were included: 72 with T2DM and 166 without T2DM. There were significant differences in blood glucose levels and OS between the two groups (all P < 0.05). The median OS was 11.4 (95% confidence interval CI [8.49-14.31]) months in the T2DM group and 16.3 (95% CI [12.44-20.16], P = 0.023) months in the non-T2DM group. After adjustment for confounders, T2DM was an independent factor affecting 5-year OS (P = 0.010). Compared with non-T2DM patients, T2DM patients had a higher risk of death (HR = 1.475, 95% CI [1.096-1.985]). Conclusions T2DM is associated with 5-year OS in patients with PC.
Collapse
Affiliation(s)
- Zhiyin Tang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wanfeng Xu
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Mingming Zhang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
208
|
Pesticides and pancreatic adenocarcinoma: A transversal epidemiological, environmental and mechanistic narrative review. Dig Liver Dis 2022; 54:1605-1613. [PMID: 36089524 DOI: 10.1016/j.dld.2022.08.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 01/23/2023]
Abstract
Pancreatic adenocarcinoma (PA) incidence is rising worldwide, especially in France. The evolution of known risk factors such as tobacco smoking, obesity, type 2 diabetes, chronic pancreatitis, or constitutional mutations is not sufficient to explain this trend. Pesticides are known risk factors in other malignancies. Previous studies have outlined pesticides' influence in PA, such as dichlorodiphenyltrichloroethane as plausible risk factors. The general population is directly or indirectly exposed to pesticides through air, food or water. Some of these chemicals may accumulate in the body all along lifetime and may harm carriers. The toxic mixing effects of these chemicals are not well documented. Several hypotheses have been put forward to explain how pesticides can induce indirect (fatty pancreas, induced diabetes) or direct (oxidative stress, cell damage) carcinogenesis in pancreatic cells through inflammation. A strong corpus exists acknowledging pesticides as a PA risk factor. However, published studies do not provide a sufficient level of evidence to prove causality and current prospective case-control studies are still ongoing.
Collapse
|
209
|
Moraa F, Degu A. Survival Outcomes Among Pancreatic Cancer Patients at Kenyatta National Hospital. J Gastrointest Cancer 2022; 53:958-964. [PMID: 34601708 DOI: 10.1007/s12029-021-00723-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE Mortality from pancreatic cancer has risen fast in the past two decades in East Africa, including Kenya. However, there was a paucity of conclusive data about the survival of pancreatic cancer patients in the study setting. Hence, this study aimed to assess the survival outcomes of pancreatic cancer patients at Kenyatta National Hospital. METHODS A hospital-based retrospective cohort analysis was used to evaluate the survival outcomes among pancreatic cancer patients treated in the study setting from 1 January 2015 to 31 December 2019. A total of 64 eligible pancreatic cancer patients were included in the study. In the pre-designed data abstraction tool, the data were collected by reviewing the medical records of the patients. The data were analyzed using the Statistical Package for the Social Sciences version 22 software. The mean survival time was estimated using Kaplan-Meier survival analysis. Cox regression analysis was employed to estimate the predictors of mortality among pancreatic cancer patients. RESULTS The mean age of the study participants was 60.38 ± 12.61 years. Most of the patients had adenocarcinoma (96.9%) and were diagnosed at an advanced stage of the disease. The overall mean and median survival estimate for pancreatic cancer was 48.7 ± 9.7 and 39.0 ± 23.9 months, respectively. The present study showed that the overall survival rate of pancreatic cancer patients was 79.7%. CONCLUSION The mortality rate of pancreatic cancer in the present study was 20%. The overall mean survival estimate for pancreatic cancer was 48.7 ± 9.7 months, and the majority had disease progression in the last follow-up period.
Collapse
Affiliation(s)
- Faith Moraa
- Department of Pharmaceutics and Pharmacy Practice, School of Pharmacy and Health Sciences, United States International University-Africa, Nairobi, Kenya
| | - Amsalu Degu
- Department of Pharmaceutics and Pharmacy Practice, School of Pharmacy and Health Sciences, United States International University-Africa, Nairobi, Kenya.
| |
Collapse
|
210
|
Tezuka R, Iwashita T, Uemura S, Senju A, Yoshida K, Maruta A, Iwata K, Shimizu M. The efficacy and safety of modified FOLFIRINOX for unresectable advanced pancreatic cancer in elderly versus young patients: A multicenter retrospective cohort study. Pancreatology 2022; 22:1134-1140. [PMID: 36404200 DOI: 10.1016/j.pan.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 10/16/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022]
Abstract
UNLABELLED In the treatment of advanced pancreatic cancer (APC), FOLFIRINOX (FX), including its dose-modified regimen (mFX), is considered an effective regimen; however, FX is also known to be associated with a high incidence of adverse events due to its multi-agent combination regimen. The efficacy and safety in elderly patients with APC have not been well studied. AIM To compare the safety and efficacy of first-line mFX for unresectable APC in elderly and young patients. METHODS This was a multicenter retrospective cohort study included patients who received first-line mFX for unresectable APC. A total of 151 patients were included and divided into the elderly (≥65 years old; 76 patients) and young (<65 years old; 75 patients) groups. The primary endpoint was overall survival (OS). The secondary endpoints were progression-free survival (PFS) and adverse events (AEs). RESULTS The median OS and PFS were similar between the two groups (OS: 14.4 months versus 13.9 months, p = 0.42; PFS: 7.4 months versus 6.6 months, p = 0.65). Although severe AEs (≥ grade 3) were observed frequently in both groups (80% versus 84.2%, p = 0.53), there was no significant difference in any of the events between the groups. In the multivariate analysis evaluating the factors affecting OS and febrile neutropenia, age was not significant factors in both analyses. CONCLUSION First-line mFX for APC in elderly patients was as safe and effective as in younger patients if performance status was good. Further evaluation in a larger cohort is required to confirm our findings.
Collapse
Affiliation(s)
- Ryuichi Tezuka
- First Department of Internal Medicine, Gifu University Hospital, Gifu, Japan
| | - Takuji Iwashita
- First Department of Internal Medicine, Gifu University Hospital, Gifu, Japan.
| | - Shinya Uemura
- First Department of Internal Medicine, Gifu University Hospital, Gifu, Japan
| | - Akihiko Senju
- First Department of Internal Medicine, Gifu University Hospital, Gifu, Japan
| | - Kensaku Yoshida
- Department of Gastroenterology, Gifu Prefectural General Medical Center, Gifu, Japan
| | - Akinori Maruta
- Department of Gastroenterology, Gifu Prefectural General Medical Center, Gifu, Japan
| | - Keisuke Iwata
- Department of Gastroenterology, Gifu Municipal Hospital, Gifu, Japan
| | - Masahito Shimizu
- First Department of Internal Medicine, Gifu University Hospital, Gifu, Japan
| |
Collapse
|
211
|
Kong X, Feng M, Wu L, He Y, Mao H, Gu Z. Biodegradable gemcitabine-loaded microdevice with sustained local drug delivery and improved tumor recurrence inhibition abilities for postoperative pancreatic tumor treatment. Drug Deliv 2022; 29:1595-1607. [PMID: 35612309 PMCID: PMC9176693 DOI: 10.1080/10717544.2022.2075984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/27/2022] [Accepted: 05/01/2022] [Indexed: 11/05/2022] Open
Abstract
At present, the 10-year survival rate of patients with pancreatic cancer is still less than 4%, mainly due to the high cancer recurrence rate caused by incomplete surgery and lack of effective postoperative adjuvant treatment. Systemic chemotherapy remains the only choice for patients after surgery; however, it is accompanied by off-target effects and server systemic toxicity. Herein, we proposed a biodegradable microdevice for local sustained drug delivery and postoperative pancreatic cancer treatment as an alternative and safe option. Biodegradable poly(l-lactic-co-glycolic acid) (P(L)LGA) was developed as the matrix material, gemcitabine hydrochloride (GEM·HCl) was chosen as the therapeutic drug and polyethylene glycol (PEG) was employed as the drug release-controlled regulator. Through adjusting the amount and molecular weight of PEG, the controllable degradation of matrix and the sustained release of GEM·HCl were obtained, thus overcoming the unstable drug release properties of traditional microdevices. The drug release mechanism of microdevice and the regulating action of PEG were studied in detail. More importantly, in the treatment of the postoperative recurrence model of subcutaneous pancreatic tumor in mice, the microdevice showed effective inhibition of postoperative in situ recurrences of pancreatic tumors with excellent biosafety and minimum systemic toxicity. The microdevice developed in this study provides an option for postoperative adjuvant pancreatic treatment, and greatly broadens the application prospects of traditional chemotherapy drugs.
Collapse
Affiliation(s)
- Xiangming Kong
- College of Materials Science and Engineering, Research Institute for Biomaterials, Tech Institute for Advanced Materials, Nanjing Tech University, Nanjing, PR China
| | - Miao Feng
- College of Materials Science and Engineering, Research Institute for Biomaterials, Tech Institute for Advanced Materials, Nanjing Tech University, Nanjing, PR China
| | - Lihuang Wu
- College of Materials Science and Engineering, Research Institute for Biomaterials, Tech Institute for Advanced Materials, Nanjing Tech University, Nanjing, PR China
| | - Yiyan He
- College of Materials Science and Engineering, Research Institute for Biomaterials, Tech Institute for Advanced Materials, Nanjing Tech University, Nanjing, PR China
- NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing Tech University, Nanjing, PR China
- Suqian Advanced Materials Industry Technology Innovation Center of Nanjing Tech University, Nanjing, PR China
| | - Hongli Mao
- College of Materials Science and Engineering, Research Institute for Biomaterials, Tech Institute for Advanced Materials, Nanjing Tech University, Nanjing, PR China
- NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing Tech University, Nanjing, PR China
- Suqian Advanced Materials Industry Technology Innovation Center of Nanjing Tech University, Nanjing, PR China
| | - Zhongwei Gu
- College of Materials Science and Engineering, Research Institute for Biomaterials, Tech Institute for Advanced Materials, Nanjing Tech University, Nanjing, PR China
- NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing Tech University, Nanjing, PR China
- Suqian Advanced Materials Industry Technology Innovation Center of Nanjing Tech University, Nanjing, PR China
| |
Collapse
|
212
|
Kindler HL, Hammel P, Reni M, Van Cutsem E, Macarulla T, Hall MJ, Park JO, Hochhauser D, Arnold D, Oh DY, Reinacher-Schick A, Tortora G, Algül H, O'Reilly EM, Bordia S, McGuinness D, Cui K, Locker GY, Golan T. Overall Survival Results From the POLO Trial: A Phase III Study of Active Maintenance Olaparib Versus Placebo for Germline BRCA-Mutated Metastatic Pancreatic Cancer. J Clin Oncol 2022; 40:3929-3939. [PMID: 35834777 PMCID: PMC10476841 DOI: 10.1200/jco.21.01604] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 03/21/2022] [Accepted: 06/02/2022] [Indexed: 12/24/2022] Open
Abstract
PURPOSE The phase III POLO study demonstrated significant progression-free survival (PFS) benefit for active olaparib maintenance therapy versus placebo for patients with metastatic pancreatic adenocarcinoma and a germline BRCA mutation. Here, we report the final analysis of overall survival (OS) and other secondary end points. PATIENTS AND METHODS Patients with a deleterious or suspected deleterious germline BRCA mutation whose disease had not progressed after ≥ 16 weeks of first-line platinum-based chemotherapy were randomly assigned 3:2 to active maintenance olaparib (300 mg twice daily) or placebo. The primary end point was PFS; secondary end points included OS, time to second disease progression or death, time to first and second subsequent cancer therapies or death, time to discontinuation of study treatment or death, and safety and tolerability. RESULTS In total, 154 patients were randomly assigned (olaparib, n = 92; placebo, n = 62). No statistically significant OS benefit was observed (median 19.0 v 19.2 months; hazard ratio [HR], 0.83; 95% CI, 0.56 to 1.22; P = .3487). Kaplan-Meier OS curves separated at approximately 24 months, and the estimated 3-year survival after random assignment was 33.9% versus 17.8%, respectively. Median time to first subsequent cancer therapy or death (HR, 0.44; 95% CI, 0.30 to 0.66; P < .0001), time to second subsequent cancer therapy or death (HR, 0.61; 95% CI, 0.42 to 0.89; P = .0111), and time to discontinuation of study treatment or death (HR, 0.43; 95% CI, 0.29 to 0.63; P < .0001) significantly favored olaparib. The HR for second disease progression or death favored olaparib without reaching statistical significance (HR, 0.66; 95% CI, 0.43 to 1.02; P = .0613). Olaparib was well tolerated with no new safety signals. CONCLUSION Although no statistically significant OS benefit was observed, the HR numerically favored olaparib, which also conferred clinically meaningful benefits including increased time off chemotherapy and long-term survival in a subset of patients.
Collapse
Affiliation(s)
| | - Pascal Hammel
- Paul Brousse Hospital (AP-HP), University Paris-Saclay, Villejuif, France
| | - Michele Reni
- IRCCS Ospedale, San Raffaele Scientific Institute, Vita e Salute University, Milan, Italy
| | - Eric Van Cutsem
- University Hospitals Gasthuisberg and KU Leuven, Leuven, Belgium
| | - Teresa Macarulla
- Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | - Joon Oh Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | - Dirk Arnold
- Asklepios Tumorzentrum Hamburg AK Altona, Hamburg, Germany
| | - Do-Youn Oh
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | | | - Giampaolo Tortora
- Fondazione Policlinico A Gemelli IRCCS and Università Cattolica del Sacro Cuore, Rome, Italy
| | - Hana Algül
- Klinikum rechts der Isar, Comprehensive Cancer Center Munich TUM, Technische Universität München, Munich, Germany
| | | | | | | | | | | | - Talia Golan
- The Oncology Institute, Sheba Medical Center at Tel-Hashomer, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
213
|
Joliat GR, Labgaa I, Sulzer J, Vrochides D, Zerbi A, Nappo G, Perinel J, Adham M, van Roessel S, Besselink MG, Mieog JSD, Groen JV, Demartines N, Schäfer M. International assessment and validation of the prognostic role of lymph node ratio in patients with resected pancreatic head ductal adenocarcinoma. Hepatobiliary Surg Nutr 2022; 11:822-833. [PMID: 36523941 PMCID: PMC9745624 DOI: 10.21037/hbsn-21-99] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 07/28/2021] [Indexed: 08/30/2023]
Abstract
Background Lymph node ratio (LNR; positive/harvested lymph nodes) was identified as overall survival predictor in several cancers, including pancreatic adenocarcinoma. It remains unclear if LNR is predictive of overall survival in pancreatic adenocarcinoma patients staged pN2. This study assessed the prognostic overall survival role of LNR in pancreatic adenocarcinoma patients in relation with lymph node involvement. Methods A retrospective international study in six different centers (Europe and United States) was performed. Pancreatic adenocarcinoma patients who underwent pancreatoduodenectomy from 2000 to 2017 were included. Patients with neoadjuvant treatment, metastases, R2 resections, or missing data regarding nodal status were excluded. Survival curves were calculated using Kaplan-Meier method and compared using log-rank test. Multivariable Cox regressions were performed to find independent overall survival predictors adjusted for potential confounders. Results A total of 1,327 patients were included. Lymph node involvement (pN+) was found in 1,026 patients (77%), 561 pN1 (55%) and 465 pN2 (45%). Median LNR in pN+ patients was 0.214 [interquartile range (IQR): 0.105-0.364]. On multivariable analysis, LNR was the strongest overall survival predictor in the entire cohort [hazard ratio (HR) =5.5; 95% confidence interval (CI): 3.1-9.9; P<0.001] and pN+ patients (HR =3.8; 95% CI: 2.2-6.6; P<0.001). Median overall survival was better in patients with LNR <0.225 compared to patients with LNR ≥0.225 in the entire cohort and pN+ patients. Similar results were found in pN2 patients (worse overall survival when LNR ≥0.225). Conclusions LNR appeared as an important prognostic factor in patients undergoing surgery for pancreatic adenocarcinoma and permitted to stratify overall survival in pN2 patients. LNR should be routinely used in complement to tumor-node-metastasis (TNM) stage to better predict patient prognosis.
Collapse
Affiliation(s)
- Gaëtan-Romain Joliat
- Department of Visceral Surgery, Lausanne University Hospital CHUV, Lausanne, Switzerland
- Graduate School for Health Sciences, University of Bern, Bern, Switzerland
| | - Ismail Labgaa
- Department of Visceral Surgery, Lausanne University Hospital CHUV, Lausanne, Switzerland
| | - Jesse Sulzer
- Division of Hepatobiliary and Pancreatic Surgery, Carolinas Medical Center, Charlotte, NC, USA
| | - Dionisios Vrochides
- Division of Hepatobiliary and Pancreatic Surgery, Carolinas Medical Center, Charlotte, NC, USA
| | - Alessandro Zerbi
- Humanitas Clinical and Research Center-IRCCS, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Gennaro Nappo
- Humanitas Clinical and Research Center-IRCCS, Milan, Italy
| | - Julie Perinel
- Department of Digestive Surgery, Edouard Herriot Hospital, Lyon, France
| | - Mustapha Adham
- Department of Digestive Surgery, Edouard Herriot Hospital, Lyon, France
| | - Stijn van Roessel
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marc G. Besselink
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - J. Sven D. Mieog
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Jesse V. Groen
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Nicolas Demartines
- Department of Visceral Surgery, Lausanne University Hospital CHUV, Lausanne, Switzerland
| | - Markus Schäfer
- Department of Visceral Surgery, Lausanne University Hospital CHUV, Lausanne, Switzerland
| |
Collapse
|
214
|
Nduma BN, Ambe S, Ekhator C, Fonkem E. Geographical Distribution of Pancreatic Cancer in the State of Mississippi by Incidence and Mortality From 2003 to 2019. Cureus 2022; 14:e31605. [PMCID: PMC9671138 DOI: 10.7759/cureus.31605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2022] [Indexed: 11/19/2022] Open
|
215
|
Badheeb M, Abdelrahim A, Esmail A, Umoru G, Abboud K, Al-Najjar E, Rasheed G, Alkhulaifawi M, Abudayyeh A, Abdelrahim M. Pancreatic Tumorigenesis: Precursors, Genetic Risk Factors and Screening. Curr Oncol 2022; 29:8693-8719. [PMID: 36421339 PMCID: PMC9689647 DOI: 10.3390/curroncol29110686] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/09/2022] [Accepted: 11/14/2022] [Indexed: 11/17/2022] Open
Abstract
Pancreatic cancer (PC) is a highly malignant and aggressive tumor. Despite medical advancement, the silent nature of PC results in only 20% of all cases considered resectable at the time of diagnosis. It is projected to become the second leading cause in 2030. Most pancreatic cancer cases are diagnosed in the advanced stages. Such cases are typically unresectable and are associated with a 5-year survival of less than 10%. Although there is no guideline consensus regarding recommendations for screening for pancreatic cancer, early detection has been associated with better outcomes. In addition to continued utilization of imaging and conventional tumor markers, clinicians should be aware of novel testing modalities that may be effective for early detection of pancreatic cancer in individuals with high-risk factors. The pathogenesis of PC is not well understood; however, various modifiable and non-modifiable factors have been implicated in pancreatic oncogenesis. PC detection in the earlier stages is associated with better outcomes; nevertheless, most oncological societies do not recommend universal screening as it may result in a high false-positive rate. Therefore, targeted screening for high-risk individuals represents a reasonable option. In this review, we aimed to summarize the pathogenesis, genetic risk factors, high-risk population, and screening modalities for PC.
Collapse
Affiliation(s)
- Mohamed Badheeb
- Internal Medicine Department, College of Medicine, Hadhramout University, Mukalla 50512, Yemen
| | | | - Abdullah Esmail
- Section of GI Oncology, Department of Medical Oncology, Houston Methodist Cancer Center, Houston, TX 77030, USA
- Correspondence: (A.E.); (M.A.)
| | - Godsfavour Umoru
- Department of Pharmacy, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Karen Abboud
- Department of Pharmacy, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Ebtesam Al-Najjar
- Faculty of Medicine and Health Sciences, University of Science and Technology, Sana’a 15201, Yemen
| | - Ghaith Rasheed
- Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan
| | | | - Ala Abudayyeh
- Section of Nephrology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Maen Abdelrahim
- Section of GI Oncology, Department of Medical Oncology, Houston Methodist Cancer Center, Houston, TX 77030, USA
- Weill Cornell Medical College, New York, NY 14853, USA
- Cockrell Center for Advanced Therapeutic Phase I Program, Houston Methodist Research Institute, Houston, TX 77030, USA
- Correspondence: (A.E.); (M.A.)
| |
Collapse
|
216
|
Hong MZ, Li JM, Chen ZJ, Lin XY, Pan JS, Gong LL. Global burden of major gastrointestinal cancers and its association with socioeconomics, 1990-2019. Front Oncol 2022; 12:942035. [PMID: 36387124 PMCID: PMC9664003 DOI: 10.3389/fonc.2022.942035] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 10/19/2022] [Indexed: 07/25/2023] Open
Abstract
BACKGROUND To understand the impact of common cancers of the gastrointestinal tract and help to formulate evidence-based policy, we evaluate the relationship between the burden of GI tract cancers and socioeconomics. METHODS Data on GI tract cancer burden were obtained from the Global Burden of Disease (GBD) 2019 including mortality and incidence rates. According to the Socio-demographic Index (SDI) level, country and territory, and sex, etc., the data were further stratified. The association between the burden of GI tract cancer and socioeconomics, indicated by SDI, was described. Uncertainty analysis was estimated using bootstrap draw. RESULTS In 2019, five major cancers of the gastrointestinal tract led to an age-standardized incidence rate (ASIR) of 61.9 (95% CI 56.1-67.6) per 100 000 person-years. From 1990 to 2019, five common tumors of the gastrointestinal tract related age-standardized death rates (ASDRs) decreased by -22.7% (-31.1 to -13.5). For the five common tumors, ASIRs and ASDRs were both higher in males than those in females. Globally, Mongolia, and several East Asia countries exhibited the highest ASIRs in 2019. The high SDI, and high-middle SDI locations recorded the highest incidence rate and death rate of colon and rectum cancer and pancreatic cancer. On the contrary, the low-middle SDI, and low SDI locations possessed the highest incidence rate and death rate of stomach cancer and esophageal cancer. CONCLUSION There is a profound association between socioeconomics and burden of common cancers of the gastrointestinal tract. It would be helpful for the high SDI, and high-middle SDI locations to pay special attention to the screening of colon and rectum cancer and pancreatic cancer while the low-middle SDI, and low SDI locations should pay more attention to the screening of stomach cancer and esophageal cancer.
Collapse
Affiliation(s)
- Mei-Zhu Hong
- Department of Traditional Chinese Medicine, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Jing-Mao Li
- Department of Statistics, School of Economics, Xiamen University, Xiamen, Fujian, China
| | - Zhi-Jian Chen
- Department of Hepatology, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Xiao-Yun Lin
- Department of Hepatology, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Jin-Shui Pan
- Department of Hepatology, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Li-Li Gong
- Department of Health Care, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
217
|
Conroy T, Castan F, Lopez A, Turpin A, Ben Abdelghani M, Wei AC, Mitry E, Biagi JJ, Evesque L, Artru P, Lecomte T, Assenat E, Bauguion L, Ychou M, Bouché O, Monard L, Lambert A, Hammel P. Five-Year Outcomes of FOLFIRINOX vs Gemcitabine as Adjuvant Therapy for Pancreatic Cancer: A Randomized Clinical Trial. JAMA Oncol 2022; 8:1571-1578. [PMID: 36048453 PMCID: PMC9437831 DOI: 10.1001/jamaoncol.2022.3829] [Citation(s) in RCA: 174] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/29/2022] [Indexed: 01/11/2023]
Abstract
Importance Early results at 3 years from the PRODIGE 24/Canadian Cancer Trials Group PA6 randomized clinical trial showed survival benefits with adjuvant treatment with modified FOLFIRINOX vs gemcitabine in patients with resected pancreatic ductal adenocarcinoma; mature data are now available. Objective To report 5-year outcomes and explore prognostic factors for overall survival. Design, Setting, and Participants This open-label, phase 3 randomized clinical trial was conducted at 77 hospitals in France and Canada and included patients aged 18 to 79 years with histologically confirmed pancreatic ductal adenocarcinoma who had undergone complete macroscopic (R0/R1) resection within 3 to 12 weeks before randomization. Patients were included from April 16, 2012, through October 3, 2016. The cutoff date for this analysis was June 28, 2021. Interventions A total of 493 patients were randomized (1:1) to receive treatment with modified FOLFIRINOX (oxaliplatin, 85 mg/m2 of body surface area; irinotecan, 150-180 mg/m2; leucovorin, 400 mg/m2; and fluorouracil, 2400 mg/m2, every 2 weeks) or gemcitabine (1000 mg/m2, days 1, 8, and 15, every 4 weeks) as adjuvant therapy for 24 weeks. Main Outcomes and Measures Primary end point was disease-free survival. Secondary end points included overall survival, metastasis-free survival, and cancer-specific survival. Prognostic factors for overall survival were determined. Results Of the 493 patients, 216 (43.8%) were women, and the mean (SD) age was 62.0 (8.9) years. At a median of 69.7 months' follow-up, 367 disease-free survival events were observed. In patients receiving chemotherapy with modified FOLFIRINOX vs gemcitabine, median disease-free survival was 21.4 months (95% CI, 17.5-26.7) vs 12.8 months (95% CI, 11.6-15.2) (hazard ratio [HR], 0.66; 95% CI, 0.54-0.82; P < .001) and 5-year disease-free survival was 26.1% vs 19.0%; median overall survival was 53.5 months (95% CI, 43.5-58.4) vs 35.5 months (95% CI, 30.1-40.3) (HR, 0.68; 95% CI, 0.54-0.85; P = .001), and 5-year overall survival was 43.2% vs 31.4%; median metastasis-free survival was 29.4 months (95% CI, 21.4-40.1) vs 17.7 months (95% CI, 14.0-21.2) (HR, 0.64; 95% CI, 0.52-0.80; P < .001); and median cancer-specific survival was 54.7 months (95% CI, 45.8-68.4) vs 36.3 months (95% CI, 30.5-43.9) (HR, 0.65; 95% CI, 0.51-0.82; P < .001). Multivariable analysis identified modified FOLFIRINOX, age, tumor grade, tumor staging, and larger-volume center as significant favorable prognostic factors for overall survival. Shorter relapse delay was an adverse prognostic factor. Conclusions and Relevance The final 5-year results from the PRODIGE 24/Canadian Cancer Trials Group PA6 randomized clinical trial indicate that adjuvant treatment with modified FOLFIRINOX yields significantly longer survival than gemcitabine in patients with resected pancreatic ductal adenocarcinoma. Trial Registration EudraCT: 2011-002026-52; ClinicalTrials.gov Identifier: NCT01526135.
Collapse
Affiliation(s)
- Thierry Conroy
- Medical Oncology department, Institut de cancérologie de Lorraine, Vandoeuvre-lès-Nancy, France and Université de Lorraine, APEMAC, équipe MICS, Nancy, France
| | - Florence Castan
- Biometry Department, ICM Regional Cancer Institute of Montpellier, Montpellier, France and Montpellier University, Montpellier, France
| | - Anthony Lopez
- Hepatogastroenterology department, University Hospital, Nancy, France
| | - Anthony Turpin
- Medical Oncology Department, University hospital, Lille, France and University of Lille, Lille, France
| | | | - Alice C. Wei
- Surgery department, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Emmanuel Mitry
- Medical Oncology department, Institut Paoli-Calmettes, Marseille, France
| | | | - Ludovic Evesque
- Medical oncology department, Centre Antoine-Lacassagne, Nice, France
| | - Pascal Artru
- Hepatogastroenterology department, Hôpital Jean-Mermoz, Lyon, France
| | - Thierry Lecomte
- Hepatogastroenterology department, Hôpital Trousseau, Tours, France and INSERM UMR 6239, Tours University, Tours, France
| | - Eric Assenat
- Medical oncology department, Centre Hospitalier Universitaire de Saint-Eloi, Montpellier, France
| | - Lucile Bauguion
- Hepatogastroenterology department, Centre Hospitalier Départemental Vendée, La Roche-sur-Yon, France
| | - Marc Ychou
- Oncology department, ICM Regional Cancer Institute of Montpellier, Montpellier, France and Montpellier University, Montpellier, France
| | - Olivier Bouché
- Digestive oncology department, Centre Hospitalier Universitaire Robert Debré, Reims, France
| | | | - Aurélien Lambert
- Medical Oncology department, Institut de cancérologie de Lorraine, Vandoeuvre-lès-Nancy, France and Université de Lorraine, APEMAC, équipe MICS, Nancy, France
| | - Pascal Hammel
- Digestive and Medical Oncology department, Hôpital Paul Brousse and University Paris-Saclay, Villejuif, France
| |
Collapse
|
218
|
McGarry JL, Creavin B, Kelly ME, Gallagher TK. Risk of pancreatic ductal adenocarcinoma associated with carriage of BRCA1 and/or BRCA2 mutation: A systematic review and meta-analysis. J Surg Oncol 2022; 126:1028-1037. [PMID: 35770919 DOI: 10.1002/jso.26994] [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: 05/01/2022] [Accepted: 06/04/2022] [Indexed: 11/11/2022]
Abstract
Germline BRCA1/2 mutations lead to malfunction of DNA damage repair pathways and predispose to pancreatic ductal adenocarcinoma (PDAC). The aim of this study is to synthesise the available research on this topic. Four studies reporting risk ratio (RR) were included in the final meta-analysis to minimise misrepresenting our results by combining separate risk estimates. Our meta-analysis revealed a statistically significant increased risk of PDAC in BRCA carriers overall (RR: 2.65, 95% confidence interval: 1.43-4.91, p = 0.002).
Collapse
Affiliation(s)
- Jennifer L McGarry
- Department of Hepatobiliary Surgery, St. Vincent's University Hospital, Dublin, Ireland
| | - Ben Creavin
- Department of Hepatobiliary Surgery, St. Vincent's University Hospital, Dublin, Ireland
| | - Michael E Kelly
- Department of Hepatobiliary Surgery, St. Vincent's University Hospital, Dublin, Ireland
| | - Tom K Gallagher
- Department of Hepatobiliary Surgery, St. Vincent's University Hospital, Dublin, Ireland
| |
Collapse
|
219
|
Poly TN, Islam MM, Walther BA, Lin MC, Li YC(J. Proton Pump Inhibitors Use and the Risk of Pancreatic Cancer: Evidence from Eleven Epidemiological Studies, Comprising 1.5 Million Individuals. Cancers (Basel) 2022; 14:5357. [PMID: 36358776 PMCID: PMC9658965 DOI: 10.3390/cancers14215357] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/16/2022] [Accepted: 10/27/2022] [Indexed: 01/28/2024] Open
Abstract
Previous epidemiological studies have shown that proton pump inhibitor (PPI) may modify the risk of pancreatic cancer. We conducted an updated systematic review and meta-analysis of observational studies assessing the effect of PPI on pancreatic cancer. PubMed, Embase, Scopus, and Web of Science were searched for studies published between 1 January 2000, and 1 May 2022. We only included studies that assessed exposure to PPI, reported pancreatic cancer outcomes, and provided effect sizes (hazard ratio or odds ratio) with 95% confidence intervals (CIs). We calculated an adjusted pooled risk ratio (RR) with 95%CIs using the random-effects model. Eleven studies (eight case-control and three cohorts) that reported 51,629 cases of pancreatic cancer were included. PPI was significantly associated with a 63% increased risk of pancreatic cancer (RRadj. 1.63, 95%CI: 1.19-2.22, p = 0.002). Subgroup analysis showed that the pooled RR for rabeprazole and lansoprazole was 4.08 (95%CI: 0.61-26.92) and 2.25 (95%CI: 0.83-6.07), respectively. Moreover, the risk of pancreatic cancer was established for both the Asian (RRadj. 1.37, 95%CI: 0.98-1.81) and Western populations (RRadj.2.76, 95%CI: 0.79-9.56). The findings of this updated meta-analysis demonstrate that the use of PPI was associated with an increased risk of pancreatic cancer. Future studies are needed to improve the quality of evidence through better verification of PPI status (e.g., patient selection, duration, and dosages), adjusting for possible confounders, and ensuring long-term follow-up.
Collapse
Affiliation(s)
- Tahmina Nasrin Poly
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan
- International Center for Health Information Technology (ICHIT), Taipei Medical University, Taipei 110301, Taiwan
- Research Center of Big Data and Meta-Analysis, Wan Fang Hospital, Taipei Medical University, Taipei 110301, Taiwan
| | - Md. Mohaimenul Islam
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan
- Research Center of Big Data and Meta-Analysis, Wan Fang Hospital, Taipei Medical University, Taipei 110301, Taiwan
| | - Bruno Andreas Walther
- Alfred-Wegener-Institut Helmholtz-Zentrum für Polar-und Meeresforschung, Am Handelshafen 12, D-27570 Bremerhaven, Germany
| | - Ming-Chin Lin
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan
- Department of Neurosurgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235041, Taiwan
| | - Yu-Chuan (Jack) Li
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan
- International Center for Health Information Technology (ICHIT), Taipei Medical University, Taipei 110301, Taiwan
- Research Center of Big Data and Meta-Analysis, Wan Fang Hospital, Taipei Medical University, Taipei 110301, Taiwan
- Department of Dermatology, Wan Fang Hospital, Taipei 116, Taiwan
| |
Collapse
|
220
|
Peduzzi G, Archibugi L, Katzke V, Gentiluomo M, Capurso G, Milanetto AC, Gazouli M, Goetz M, Brenner H, Vermeulen RCH, Talar-Wojnarowska R, Vanella G, Tavano F, Lucchesi M, Mohelnikova-Duchonova B, Chen X, Kiudelis V, Hegyi P, Oliverius M, Stocker H, Stornello C, Vodickova L, Souček P, Neoptolemos JP, Testoni SGG, Morelli L, Lawlor RT, Basso D, Izbicki JR, Ermini S, Kupcinskas J, Pezzilli R, Boggi U, van Laarhoven HWM, Szentesi A, Erőss B, Capretti G, Schöttker B, Skieceviciene J, Aoki MN, van Eijck CHJ, Cavestro GM, Canzian F, Campa D. Common variability in oestrogen-related genes and pancreatic ductal adenocarcinoma risk in women. Sci Rep 2022; 12:18100. [PMID: 36302831 PMCID: PMC9613634 DOI: 10.1038/s41598-022-22973-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 10/21/2022] [Indexed: 12/30/2022] Open
Abstract
The incidence of pancreatic ductal adenocarcinoma (PDAC) is different among males and females. This disparity cannot be fully explained by the difference in terms of exposure to known risk factors; therefore, the lower incidence in women could be attributed to sex-specific hormones. A two-phase association study was conducted in 12,387 female subjects (5436 PDAC cases and 6951 controls) to assess the effect on risk of developing PDAC of single nucleotide polymorphisms (SNPs) in 208 genes involved in oestrogen and pregnenolone biosynthesis and oestrogen-mediated signalling. In the discovery phase 14 polymorphisms showed a statistically significant association (P < 0.05). In the replication none of the findings were validated. In addition, a gene-based analysis was performed on the 208 selected genes. Four genes (NR5A2, MED1, NCOA2 and RUNX1) were associated with PDAC risk, but only NR5A2 showed an association (P = 4.08 × 10-5) below the Bonferroni-corrected threshold of statistical significance. In conclusion, despite differences in incidence between males and females, our study did not identify an effect of common polymorphisms in the oestrogen and pregnenolone pathways in relation to PDAC susceptibility. However, we validated the previously reported association between NR5A2 gene variants and PDAC risk.
Collapse
Affiliation(s)
- Giulia Peduzzi
- grid.5395.a0000 0004 1757 3729Department of Biology, University of Pisa, 56126 Pisa, Italy
| | - Livia Archibugi
- grid.415230.10000 0004 1757 123XDigestive and Liver Disease Unit, Sant’Andrea Hospital, Rome, Italy ,grid.18887.3e0000000417581884Pancreato-Biliary Endoscopy and Endoscopic Ultrasound, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | - Verena Katzke
- grid.7497.d0000 0004 0492 0584Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Manuel Gentiluomo
- grid.5395.a0000 0004 1757 3729Department of Biology, University of Pisa, 56126 Pisa, Italy
| | - Gabriele Capurso
- grid.415230.10000 0004 1757 123XDigestive and Liver Disease Unit, Sant’Andrea Hospital, Rome, Italy ,grid.18887.3e0000000417581884Pancreato-Biliary Endoscopy and Endoscopic Ultrasound, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | | | - Maria Gazouli
- grid.5216.00000 0001 2155 0800Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Mara Goetz
- grid.13648.380000 0001 2180 3484Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Hermann Brenner
- grid.7497.d0000 0004 0492 0584Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany ,grid.7497.d0000 0004 0492 0584Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany ,grid.7497.d0000 0004 0492 0584German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Renata Talar-Wojnarowska
- grid.8267.b0000 0001 2165 3025Department of Digestive Tract Diseases, Medical University of Lodz, Lodz, Poland
| | - Giuseppe Vanella
- grid.415230.10000 0004 1757 123XDigestive and Liver Disease Unit, Sant’Andrea Hospital, Rome, Italy ,grid.18887.3e0000000417581884Pancreato-Biliary Endoscopy and Endoscopic Ultrasound, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | - Francesca Tavano
- grid.413503.00000 0004 1757 9135Division of Gastroenterology and Research Laboratory, Fondazione IRCCS “Casa Sollievo Della Sofferenza” Hospital, San Giovanni Rotondo, Italy
| | - Maurizio Lucchesi
- Oncology of Massa Carrara, Oncological Department, Azienda USL Toscana Nord Ovest, Carrara, Italy
| | - Beatrice Mohelnikova-Duchonova
- grid.10979.360000 0001 1245 3953Department of Oncology, Faculty of Medicine and Dentistry, Palacký University, Olomouc, Czech Republic
| | - Xuechen Chen
- grid.7497.d0000 0004 0492 0584Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany ,grid.7700.00000 0001 2190 4373Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Vytautas Kiudelis
- grid.45083.3a0000 0004 0432 6841Gastroenterology Department and Institute for Digestive Research, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Péter Hegyi
- grid.9679.10000 0001 0663 9479Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary ,grid.11804.3c0000 0001 0942 9821Centre for Translational Medicine, Semmelweis University, Budapest, Hungary ,grid.11804.3c0000 0001 0942 9821Division of Pancreatic Diseases, Heart and Vascular Center, Semmelweis University, Budapest, Hungary ,grid.9679.10000 0001 0663 9479János Szentágothai Research Center, University of Pécs, Pécs, Hungary
| | - Martin Oliverius
- grid.4491.80000 0004 1937 116XSurgery Clinic Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Hannah Stocker
- grid.7497.d0000 0004 0492 0584Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany ,grid.7700.00000 0001 2190 4373Network Aging Research (NAR), Heidelberg University, Heidelberg, Germany
| | - Caterina Stornello
- grid.415230.10000 0004 1757 123XDigestive and Liver Disease Unit, Sant’Andrea Hospital, Rome, Italy ,grid.18887.3e0000000417581884Pancreato-Biliary Endoscopy and Endoscopic Ultrasound, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | - Ludmila Vodickova
- grid.424967.a0000 0004 0404 6946Department of Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic ,grid.4491.80000 0004 1937 116XFirst Faculty of Medicine, Institute of Biology and Medical Genetics, Charles University, Prague, Czech Republic ,grid.4491.80000 0004 1937 116XBiomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Pavel Souček
- grid.4491.80000 0004 1937 116XBiomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - John P. Neoptolemos
- grid.7700.00000 0001 2190 4373Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | - Sabrina Gloria Giulia Testoni
- grid.18887.3e0000000417581884Pancreato-Biliary Endoscopy and Endoscopic Ultrasound, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | - Luca Morelli
- grid.5395.a0000 0004 1757 3729General Surgery, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Rita T. Lawlor
- grid.411475.20000 0004 1756 948XARC-NET, Centre for Applied Research on Cancer, University and Hospital Trust of Verona, Verona, Italy
| | - Daniela Basso
- grid.5608.b0000 0004 1757 3470Department DIMED-Laboratory Medicine, University of Padova, Padua, Italy
| | - Jakob R. Izbicki
- grid.13648.380000 0001 2180 3484Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Stefano Ermini
- grid.413181.e0000 0004 1757 8562Blood Transfusion Service, Azienda Ospedaliero-Universitaria Meyer, Florence, Italy
| | - Juozas Kupcinskas
- grid.45083.3a0000 0004 0432 6841Gastroenterology Department and Institute for Digestive Research, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | | - Ugo Boggi
- grid.144189.10000 0004 1756 8209Division of General and Transplant Surgery, Pisa University Hospital, Pisa, Italy
| | - Hanneke W. M. van Laarhoven
- grid.7177.60000000084992262Department of Medical Oncology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands ,grid.16872.3a0000 0004 0435 165XCancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Andrea Szentesi
- grid.9679.10000 0001 0663 9479Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary ,grid.9679.10000 0001 0663 9479János Szentágothai Research Center, University of Pécs, Pécs, Hungary ,grid.9008.10000 0001 1016 9625Centre for Translational Medicine, Department of Medicine, University of Szeged, Szeged, Hungary
| | - Bálint Erőss
- grid.9679.10000 0001 0663 9479Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary ,grid.11804.3c0000 0001 0942 9821Division of Pancreatic Diseases, Heart and Vascular Center, Semmelweis University, Budapest, Hungary ,grid.11804.3c0000 0001 0942 9821Center for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Giovanni Capretti
- grid.452490.eDepartment of Biomedical Sciences, Humanitas University, Milan, Italy ,grid.417728.f0000 0004 1756 8807Pancreatic Surgery Unit, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Ben Schöttker
- grid.7497.d0000 0004 0492 0584Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jurgita Skieceviciene
- grid.45083.3a0000 0004 0432 6841Gastroenterology Department and Institute for Digestive Research, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Mateus Nóbrega Aoki
- grid.418068.30000 0001 0723 0931Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| | - Casper H. J. van Eijck
- grid.5645.2000000040459992XDepartment of Surgery, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Giulia Martina Cavestro
- grid.15496.3f0000 0001 0439 0892Gastroenterology and Gastrointestinal Endoscopy Unit, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Federico Canzian
- grid.7497.d0000 0004 0492 0584Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniele Campa
- grid.5395.a0000 0004 1757 3729Department of Biology, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
221
|
Ramezankhani R, Ghavidel AA, Rashidi S, Rojhannezhad M, Abolkheir HR, Mirhosseini M, Taleahmad S, Vosough M. Gender-related differentially expressed genes in pancreatic cancer: possible culprits or accomplices? Front Genet 2022; 13:966941. [DOI: 10.3389/fgene.2022.966941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 10/10/2022] [Indexed: 11/13/2022] Open
Abstract
Pancreatic cancer (PC) is one of the leading causes of cancer mortality worldwide, and its incidence and mortality rate in several regions is higher in male patients. Although numerous efforts have been made to enhance the clinical outcomes of existing therapeutic regimens, their efficiency is still low, and drug resistance usually occurs in many patients. In addition, the exact underlying molecular basis that makes PC slightly more prevalent among males remains unknown. Providing information regarding the possible association between gender and PC tumorigenesis may offer important clues for how certain molecular cross-talks can affect PC initiation and/or progression. In this study, we used several microarray expression data to identify the common up- and downregulated genes within one specific gender, which were also specified to have binding sites for androgen and/or estrogen receptors. Using functional enrichment analysis among the others, for all the gene sets found in this study, we have shed light on the plausible importance of the androgenic effectors in tumorigenesis, such as the androgen-regulated expression of the GLI transcription factor and the potential role of testosterone in the extracellular matrix (ECM)–cell interaction, which are known for their importance in tumorigenesis. Moreover, we demonstrated that the biological process axon guidance was highlighted regarding the upregulated genes in male patients. Overall, identification of gene candidates as the possible link between gender and PC progression or survival rates may help in developing strategies to reduce the incidence of this cancer.
Collapse
|
222
|
Gao M, Guo Y, Li J, Chen X, Yuan Y, Ma W. The Clinicopathological Significance and Prognostic Value of PD-L2 in Patients with HCC, ICC and PAAD: A Meta-Analysis. INTERNATIONAL JOURNAL OF SURGERY: ONCOLOGY 2022. [DOI: 10.29337/ijsonco.142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
223
|
Seo K, Lim JH, Seo J, Nguon LS, Yoon H, Park JS, Park S. Semantic Segmentation of Pancreatic Cancer in Endoscopic Ultrasound Images Using Deep Learning Approach. Cancers (Basel) 2022; 14:5111. [PMID: 36291895 PMCID: PMC9600976 DOI: 10.3390/cancers14205111] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/12/2022] [Accepted: 10/18/2022] [Indexed: 11/17/2022] Open
Abstract
Endoscopic ultrasonography (EUS) plays an important role in diagnosing pancreatic cancer. Surgical therapy is critical to pancreatic cancer survival and can be planned properly, with the characteristics of the target cancer determined. The physical characteristics of the pancreatic cancer, such as size, location, and shape, can be determined by semantic segmentation of EUS images. This study proposes a deep learning approach for the segmentation of pancreatic cancer in EUS images. EUS images were acquired from 150 patients diagnosed with pancreatic cancer. A network with deep attention features (DAF-Net) is proposed for pancreatic cancer segmentation using EUS images. The performance of the deep learning models (U-Net, Attention U-Net, and DAF-Net) was evaluated by 5-fold cross-validation. For the evaluation metrics, the Dice similarity coefficient (DSC), intersection over union (IoU), receiver operating characteristic (ROC) curve, and area under the curve (AUC) were chosen. Statistical analysis was performed for different stages and locations of the cancer. DAF-Net demonstrated superior segmentation performance for the DSC, IoU, AUC, sensitivity, specificity, and precision with scores of 82.8%, 72.3%, 92.7%, 89.0%, 98.1%, and 85.1%, respectively. The proposed deep learning approach can provide accurate segmentation of pancreatic cancer in EUS images and can effectively assist in the planning of surgical therapies.
Collapse
Affiliation(s)
- Kangwon Seo
- Department of Electrical and Electronics Engineering, Chung-Ang University, Seoul 06974, Korea
| | - Jung-Hyun Lim
- Division of Gastroenterology, Department of Internal Medicine, Inha University School of Medicine, Incheon 22332, Korea
| | - Jeongwung Seo
- Department of Electrical and Electronics Engineering, Chung-Ang University, Seoul 06974, Korea
| | - Leang Sim Nguon
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul 03760, Korea
| | - Hongeun Yoon
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul 03760, Korea
| | - Jin-Seok Park
- Division of Gastroenterology, Department of Internal Medicine, Inha University School of Medicine, Incheon 22332, Korea
| | - Suhyun Park
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
224
|
Jiang W, Li X, Xiang C, Zhou W. Neutrophils in pancreatic cancer: Potential therapeutic targets. Front Oncol 2022; 12:1025805. [PMID: 36324574 PMCID: PMC9618950 DOI: 10.3389/fonc.2022.1025805] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/28/2022] [Indexed: 08/30/2023] Open
Abstract
Pancreatic cancer is a digestive system malignancy and poses a high mortality worldwide. Traditionally, neutrophils have been thought to play a role in acute inflammation. In contrast, their importance during tumor diseases has been less well studied. Generally, neutrophils are recruited into the tumor microenvironment and exert inflammation and tumor-promoting effects. As an essential part of the tumor microenvironment, neutrophils play diverse roles in pancreatic cancer, such as angiogenesis, progression, metastasis and immunosuppression. Additionally, neutrophils can be a new potential therapeutic target in cancer. Inhibitors of cytokines, chemokines and neutrophil extracellular traps can exert antitumor effects. In this review, we describe the role of neutrophils in the development and progression of pancreatic cancer, discuss their potential as therapeutic targets, and aim to provide ideas for improving the prognosis of patients with this malignant tumor disease.
Collapse
Affiliation(s)
- Wenkai Jiang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xin Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Caifei Xiang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Wence Zhou
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
225
|
He Q, Liu Z, Wang J. Targeting KRAS in PDAC: A New Way to Cure It? Cancers (Basel) 2022; 14:cancers14204982. [PMID: 36291766 PMCID: PMC9599866 DOI: 10.3390/cancers14204982] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/29/2022] [Accepted: 10/02/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic cancer is one of the most intractable malignant tumors worldwide, and is known for its refractory nature and poor prognosis. The fatality rate of pancreatic cancer can reach over 90%. In pancreatic ductal carcinoma (PDAC), the most common subtype of pancreatic cancer, KRAS is the most predominant mutated gene (more than 80%). In recent decades, KRAS proteins have maintained the reputation of being “undruggable” due to their special molecular structures and biological characteristics, making therapy targeting downstream genes challenging. Fortunately, the heavy rampart formed by KRAS has been broken down in recent years by the advent of KRASG12C inhibitors; the covalent inhibitors bond to the switch-II pocket of the KRASG12C protein. The KRASG12C inhibitor sotorasib has been received by the FDA for the treatment of patients suffering from KRASG12C-driven cancers. Meanwhile, researchers have paid close attention to the development of inhibitors for other KRAS mutations. Due to the high incidence of PDAC, developing KRASG12D/V inhibitors has become the focus of attention. Here, we review the clinical status of PDAC and recent research progress in targeting KRASG12D/V and discuss the potential applications.
Collapse
Affiliation(s)
- Qianyu He
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Zuojia Liu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- Correspondence: (Z.L.); (J.W.)
| | - Jin Wang
- Department of Chemistry and Physics, Stony Brook University, Stony Brook, NY 11794-3400, USA
- Correspondence: (Z.L.); (J.W.)
| |
Collapse
|
226
|
Li X, Jiang W, Dong S, Li W, Zhu W, Zhou W. STAT3 Inhibitors: A Novel Insight for Anticancer Therapy of Pancreatic Cancer. Biomolecules 2022; 12:1450. [PMID: 36291659 PMCID: PMC9599947 DOI: 10.3390/biom12101450] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/10/2022] [Accepted: 09/30/2022] [Indexed: 11/29/2022] Open
Abstract
The signal transducer and activator of transcription (STAT) is a family of intracellular cytoplasmic transcription factors involved in many biological functions in mammalian signal transduction. Among them, STAT3 is involved in cell proliferation, differentiation, apoptosis, and inflammatory responses. Despite the advances in the treatment of pancreatic cancer in the past decade, the prognosis for patients with pancreatic cancer remains poor. STAT3 has been shown to play a pro-cancer role in a variety of cancers, and inhibitors of STAT3 are used in pre-clinical and clinical studies. We reviewed the relationship between STAT3 and pancreatic cancer and the latest results on the use of STAT3 inhibitors in pancreatic cancer, with the aim of providing insights and ideas around STAT3 inhibitors for a new generation of chemotherapeutic modalities for pancreatic cancer.
Collapse
Affiliation(s)
- Xin Li
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Wenkai Jiang
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Shi Dong
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Wancheng Li
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Weixiong Zhu
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Wence Zhou
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou 730030, China
| |
Collapse
|
227
|
Druk IV. Pancreatic cancer, pancreatogenic diabetes, type 2 diabetes mellitus. EXPERIMENTAL AND CLINICAL GASTROENTEROLOGY 2022:171-182. [DOI: 10.31146/1682-8658-ecg-205-9-171-182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Pancreatic cancer (PC) is the fourth leading cause of death among all types of cancer. PC is very aggressive with a low 5-year overall survival rate. The highest prevalence of diabetes mellitus (DM), significantly exceeding the average population, is registered among patients with prostate cancer Recommendations for systemic screening of patients with diabetes for the detection of PC are not standardized. The purpose of this review is to present an analysis of current literature data on pathogenetic relationships between DM and PC and prospects for PC screening. Research data indicate that there is a bidirectional relationship between DM and PC, in which DM can act either as a risk factor for PC or as a marker of paraneoplastic syndrome of PC. In the differential diagnosis of type 2 diabetes, pancreatogenic diabetes and diabetes associated with PC, a set of clinical signs can be used. Patients with DM who have additional signs/symptoms of increased risk can be considered as a group subject to mandatory screening. Numerous studies of various proteomic, metabolomic, genetic and transcriptomic biomarkers PC have been published. The search for an easy-to-use clinically useful and cost-effective PC marker is still ongoing.
Collapse
|
228
|
Ding YN, Xue M, Tang QS, Wang LJ, Ding HY, Li H, Gao CC, Yu WP. Immunotherapy-based novel nanoparticles in the treatment of gastrointestinal cancer: Trends and challenges. World J Gastroenterol 2022; 28:5403-5419. [PMID: 36312831 PMCID: PMC9611702 DOI: 10.3748/wjg.v28.i37.5403] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/27/2022] [Accepted: 09/15/2022] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal cancer (GIC) is the most common cancer with a poor prognosis. Currently, surgery is the main treatment for GIC. However, the high rate of postoperative recurrence leads to a low five-year survival rate. In recent years, immunotherapy has received much attention. As the only immunotherapy drugs approved by the Food and Drug Administration (FDA), immune checkpoint blockade (ICB) drugs have great potential in cancer therapy. Nevertheless, the efficacy of ICB treatment is greatly limited by the low immunogenicity and immunosuppressive microenvironment of GIC. Therefore, the targets of immunotherapy have expanded from ICB to increasing tumor immunogenicity, increasing the recruitment and maturation of immune cells and reducing the proportion of inhibitory immune cells, such as M2-like macrophages, regulatory T cells and myeloid-derived suppressor cells. Moreover, with the development of nanotechnology, a variety of nanoparticles have been approved by the FDA for clinical therapy, so novel nanodrug delivery systems have become a research focus for anticancer therapy. In this review, we summarize recent advances in the application of immunotherapy-based nanoparticles in GICs, such as gastric cancer, hepatocellular carcinoma, colorectal cancer and pancreatic cancer, and described the existing challenges and future trends.
Collapse
Affiliation(s)
- Yi-Nan Ding
- Department of Pathophysiology, College of Medicine, Southeast University, Nanjing 210000, Jiangsu Province, China
| | - Ming Xue
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210000, Jiangsu Province, China
| | - Qiu-Sha Tang
- Department of Pathophysiology, College of Medicine, Southeast University, Nanjing 210000, Jiangsu Province, China
| | - Li-Jun Wang
- Department of Pathophysiology, College of Medicine, Southeast University, Nanjing 210000, Jiangsu Province, China
| | - Hui-Yan Ding
- Department of Pathophysiology, College of Medicine, Southeast University, Nanjing 210000, Jiangsu Province, China
| | - Han Li
- Department of Tuberculosis, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| | - Cheng-Cheng Gao
- Department of Radiology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Wei-Ping Yu
- Medical School, Southeast University, Nanjing 210009, Jiangsu Province, China
| |
Collapse
|
229
|
Jahedi H, Ramachandran A, Windsor J, Knowlton N, Blenkiron C, Print CG. Clinically Relevant Biology of Hyaluronic Acid in the Desmoplastic Stroma of Pancreatic Ductal Adenocarcinoma. Pancreas 2022; 51:1092-1104. [PMID: 37078930 DOI: 10.1097/mpa.0000000000002154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
ABSTRACT Pancreatic ductal adenocarcinoma (PDAC) is notorious for its poor outcome. The presence of a dense desmoplastic stroma is a hallmark of this malignancy, and abundant hyaluronic acid (HA) within this stroma is a common feature of PDAC. At the end of 2019, an HA-targeting drug, after initial promise, failed phase 3 clinical trials in PDAC. This failure in the face of such strong evidence for biological importance forces us to turn back to the research and seek a better understanding of HA biology in PDAC. Therefore, in this review, we reexamine what is known about HA biology, the methods used to detect and quantify HA, and the ability of the biological models in which HA has been investigated to recapitulate an HA-rich desmoplastic tumor stroma. The role of HA in PDAC relies on its complex interplay with a range of HA-associated molecules, which have not been as extensively investigated as HA itself. Therefore, using large genomic data sets, we cataloged the abundance and activity in PDAC of molecules that modulate HA synthesis, degradation, protein interactions, and receptor binding. Based on their association with clinical characteristics and individual patient outcomes, we suggest a small number of HA-associated molecules that warrant further investigation as biomarkers and drug targets.
Collapse
Affiliation(s)
- Hossein Jahedi
- From the Departments of Molecular Medicine and Pathology
| | | | | | | | | | | |
Collapse
|
230
|
Evan T, Wang VMY, Behrens A. The roles of intratumour heterogeneity in the biology and treatment of pancreatic ductal adenocarcinoma. Oncogene 2022; 41:4686-4695. [PMID: 36088504 PMCID: PMC9568427 DOI: 10.1038/s41388-022-02448-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 08/11/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022]
Abstract
Intratumour heterogeneity (ITH) has become an important focus of cancer research in recent years. ITH describes the cellular variation that enables tumour evolution, including tumour progression, metastasis and resistance to treatment. The selection and expansion of genetically distinct treatment-resistant cancer cell clones provides one explanation for treatment failure. However, tumour cell variation need not be genetically encoded. In pancreatic ductal adenocarcinoma (PDAC) in particular, the complex tumour microenvironment as well as crosstalk between tumour and stromal cells result in exceptionally variable tumour cell phenotypes that are also highly adaptable. In this review we discuss four different types of phenotypic heterogeneity within PDAC, from morphological to metabolic heterogeneity. We suggest that these different types of ITH are not independent, but, rather, can inform one another. Lastly, we highlight recent findings that suggest how therapeutic efforts may halt PDAC progression by constraining cellular heterogeneity.
Collapse
Affiliation(s)
- Theodore Evan
- Cancer Stem Cell Laboratory, The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, SW3 6JB, UK
| | | | - Axel Behrens
- Cancer Stem Cell Laboratory, The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, SW3 6JB, UK.
- Department of Surgery and Cancer, Imperial College London, London, SW7 2AZ, UK.
- CRUK Convergence Science Centre, Imperial College London, SW7 2AZ, London, UK.
| |
Collapse
|
231
|
Chen LJ, Wu L, Wang W, Zhai LL, Xiang F, Li WB, Tang ZG. Long non‑coding RNA 01614 hyperactivates WNT/β‑catenin signaling to promote pancreatic cancer progression by suppressing GSK‑3β. Int J Oncol 2022; 61:116. [PMID: 35929518 PMCID: PMC9387559 DOI: 10.3892/ijo.2022.5406] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/24/2022] [Indexed: 11/06/2022] Open
Abstract
Pancreatic cancer (PC) is a lethal type of cancer for which effective therapies are limited. Long non-coding RNAs (lncRNAs) represent a critical type of regulator category, mediating the tumorigenesis and development of various tumor types, including PC. However, the expression patterns and functions of numerous lncRNAs in PC remain poorly understood. In the present study, linc01614 was identified as a PC-related lncRNA. linc01614 was notably upregulated in PC tissues and cell lines and was associated with the poor disease-free survival of patients with PC according to the analysis of The Cancer Genome Atlas-derived datasets. Functionally, linc01614 knockdown suppressed PC cell proliferation, migration and invasion in vitro, and inhibited tumor proliferation in vitro and in vivo. Mechanistically, linc01614 overexpression stabilized the level of β-catenin protein to hyperactivate the WNT/β-catenin signaling pathway in PC cells. Further analyses revealed that linc01614 bound to GSK-3β and perturbed the interaction between GSK-3β and AXIN1, thereby preventing the formation of the β-catenin degradation complex and reducing the degradation of β-catenin. In summary, the present findings reveal that linc01614 may function as an oncogene and promote the progression of PC and may thus be considered as a potential therapeutic target in the future.
Collapse
Affiliation(s)
- Long-Jiang Chen
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Lun Wu
- Department of Breast and Τhyroid Surgery, Experiment Center of Medicine, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei 442008, P.R. China
| | - Wei Wang
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Lu-Lu Zhai
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Feng Xiang
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wei-Bo Li
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zhi-Gang Tang
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
232
|
Cura Daball P, Tröger H, Daum S. [Long-term response in advanced pancreatic adenocarcinoma - a case report and literature review]. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2022; 60:1510-1516. [PMID: 34905798 DOI: 10.1055/a-1695-3528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND Pancreatic cancer is still considered one of the most aggressive types of cancer and is associated with a very poor prognosis although there have been improvements in diagnostics and chemotherapy regimes in recent years. A cure can only be achieved through complete resection which is only possible when diagnosed at a very early stage, though this is rarely the case. We report on a patient with stage IV adenocarcinoma of the pancreas in which several therapeutically actionable mutations could be detected and discuss new options of targeted therapies. CASE REPORT A patient in his 50s was diagnosed with metastatic adenocarcinoma of the pancreas. The patient showed an excellent response to platinum-based chemotherapy with FOLFIRINOX. When a germline mutation in the BRCA-2 gene could be identified, he took part in the POLO-study receiving a maintenance therapy with the PARP-Inhibitor Olaparib. Due to a relapse, 2nd and 3rd line chemotherapy regimens were applied with Gemcitabine combined with Nab-Paclitaxel and later with Erlotinib. Although an activating mutation in the KRAS-gene could be detected as well, the patient rejected further experimental treatment. CONCLUSION Identifying predictive factors and specific targetable mutations in patients with advanced pancreatic cancer is needed to be able to apply more individual and specific therapies in order to improve outcomes.
Collapse
Affiliation(s)
- Paola Cura Daball
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
| | - Hanno Tröger
- Medizinische Klinik 1, St Joseph Krankenhaus Berlin-Tempelhof, Berlin, Germany
| | - Severin Daum
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
| |
Collapse
|
233
|
Ugai T, Sasamoto N, Lee HY, Ando M, Song M, Tamimi RM, Kawachi I, Campbell PT, Giovannucci EL, Weiderpass E, Rebbeck TR, Ogino S. Is early-onset cancer an emerging global epidemic? Current evidence and future implications. Nat Rev Clin Oncol 2022; 19:656-673. [PMID: 36068272 PMCID: PMC9509459 DOI: 10.1038/s41571-022-00672-8] [Citation(s) in RCA: 261] [Impact Index Per Article: 87.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2022] [Indexed: 02/07/2023]
Abstract
Over the past several decades, the incidence of early-onset cancers, often defined as cancers diagnosed in adults <50 years of age, in the breast, colorectum, endometrium, oesophagus, extrahepatic bile duct, gallbladder, head and neck, kidney, liver, bone marrow, pancreas, prostate, stomach and thyroid has increased in multiple countries. Increased use of screening programmes has contributed to this phenomenon to a certain extent, although a genuine increase in the incidence of early-onset forms of several cancer types also seems to have emerged. Evidence suggests an aetiological role of risk factor exposures in early life and young adulthood. Since the mid-20th century, substantial multigenerational changes in the exposome have occurred (including changes in diet, lifestyle, obesity, environment and the microbiome, all of which might interact with genomic and/or genetic susceptibilities). However, the effects of individual exposures remain largely unknown. To study early-life exposures and their implications for multiple cancer types will require prospective cohort studies with dedicated biobanking and data collection technologies. Raising awareness among both the public and health-care professionals will also be critical. In this Review, we describe changes in the incidence of early-onset cancers globally and suggest measures that are likely to reduce the burden of cancers and other chronic non-communicable diseases.
Collapse
Affiliation(s)
- Tomotaka Ugai
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA.
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Naoko Sasamoto
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA, USA
| | - Hwa-Young Lee
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Institute of Convergence Science, Convergence Science Academy, Yonsei University, Seoul, Republic of Korea
| | - Mariko Ando
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mingyang Song
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Rulla M Tamimi
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Ichiro Kawachi
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Peter T Campbell
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, New York, NY, USA
| | - Edward L Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Timothy R Rebbeck
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
- Zhu Family Center for Global Cancer Prevention, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Shuji Ogino
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA.
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Cancer Immunology and Cancer Epidemiology Programs, Dana-Farber Harvard Cancer Center, Boston, MA, USA.
| |
Collapse
|
234
|
Klatte DCF, Boekestijn B, Wasser MNJM, Feshtali Shahbazi S, Ibrahim IS, Mieog JSD, Luelmo SAC, Morreau H, Potjer TP, Inderson A, Boonstra JJ, Dekker FW, Vasen HFA, van Hooft JE, Bonsing BA, van Leerdam ME. Pancreatic Cancer Surveillance in Carriers of a Germline CDKN2A Pathogenic Variant: Yield and Outcomes of a 20-Year Prospective Follow-Up. J Clin Oncol 2022; 40:3267-3277. [PMID: 35658523 DOI: 10.1200/jco.22.00194] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/30/2022] [Accepted: 04/26/2022] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Pancreatic cancer surveillance in high-risk individuals may lead to detection of pancreatic ductal adenocarcinoma (PDAC) at an earlier stage and with improved survival. This study evaluated the yield and outcomes of 20 years of prospective surveillance in a large cohort of individuals with germline pathogenic variants (PVs) in CDKN2A. METHODS Prospectively collected data were analyzed from individuals participating in pancreatic cancer surveillance. Surveillance consisted of annual magnetic resonance imaging with magnetic resonance cholangiopancreatography and optional endoscopic ultrasound. RESULTS Three hundred forty-seven germline PV carriers participated in surveillance and were followed for a median of 5.6 (interquartile range 2.3-9.9) years. A total of 36 cases of PDAC were diagnosed in 31 (8.9%) patients at a median age of 60.4 (interquartile range 51.3-64.1) years. The cumulative incidence of primary PDAC was 20.7% by age 70 years. Five carriers (5 of 31; 16.1%) were diagnosed with a second primary PDAC. Thirty (83.3%) of 36 PDACs were considered resectable at the time of imaging. Twelve cases (12 of 36; 33.3%) presented with stage I disease. The median survival after diagnosis of primary PDAC was 26.8 months, and the 5-year survival rate was 32.4% (95% CI, 19.1 to 54.8). Individuals with primary PDAC who underwent resection (22 of 31; 71.0%) had an overall 5-year survival rate of 44.1% (95% CI, 27.2 to 71.3). Nine (2.6%; 9 of 347) individuals underwent surgery for a suspected malignant lesion, which proved to not be PDAC, and this included five lesions with low-grade dysplasia. CONCLUSION This long-term surveillance study demonstrates a high incidence of PDAC in carriers of a PV in CDKN2A. This provides evidence that surveillance in such a high-risk population leads to detection of early-stage PDAC with improved resectability and survival.
Collapse
Affiliation(s)
- Derk C F Klatte
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Bas Boekestijn
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Martin N J M Wasser
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Isaura S Ibrahim
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - J Sven D Mieog
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Saskia A C Luelmo
- Department of Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Hans Morreau
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Thomas P Potjer
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Akin Inderson
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jurjen J Boonstra
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Friedo W Dekker
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Hans F A Vasen
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jeanin E van Hooft
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Bert A Bonsing
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Monique E van Leerdam
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
235
|
Zhu Z, Wang X, Zhang W, Gong M, Zhang S, Yang B, Qu B, Wu Z, Ma Q, Wang Z, Qian W. Huaier suppresses pancreatic cancer progression via activating cell autophagy induced ferroptosis. Front Oncol 2022; 12:960858. [PMID: 36248959 PMCID: PMC9561879 DOI: 10.3389/fonc.2022.960858] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 09/12/2022] [Indexed: 11/26/2022] Open
Abstract
Purpose The anti-tumour effect of Huaier has been demonstrated in a variety of tumours. Ferroptosis is a newly identified type of programmed cell death accompanied by the accumulation of reactive oxygen species (ROS) and iron in cells and plays a key role in the therapeutic process against malignant tumours. We aimed to explore the potential therapeutic role of Huaier in pancreatic cancer and uncover the relationship between Huaier and ferroptosis. Methods CCK8 and colony formation assays were used to determine the proliferation of pancreatic cancer cells (PCs). The levels of cellular ROS were analysed by a fluorescence probe, and the accumulation of cellular iron was showed by Prussian blue staining. The autophagosomes and mitochondrial morphology were characterised by transmission electron microscopy (TEM). The levels of intracellular glutathione (GSH) and lipid peroxidation were measured by the corresponding kits. Results The growth inhibitory effect of Huaier on PCs was concentration- and time-dependent, but this effect was significantly attenuated by ferroptosis inhibitors. In addition, Huaier effectively inhibited the GSH–GPX4 antioxidation system and resulted in the massive accumulation of ROS in PCs As shown by TEM, Huaier-treated PCs exhibited a decrease in mitochondrial cristae and a smaller mitochondrion, accompanied by an increase in autophagosomes. Indeed, we found that autophagy can induce ferroptosis in PCs and that Huaier-induced ferroptosis can be suppressed by the autophagosome inhibitor, Wortmannin. Conclusion Huaier can activate ferroptosis by inducing autophagy in PCs.
Collapse
Affiliation(s)
- Zeen Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Pancreatic Disease Center of Xi’an Jiaotong University, Xi’an, China
| | - Xueni Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Pancreatic Disease Center of Xi’an Jiaotong University, Xi’an, China
| | - Wunai Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Pancreatic Disease Center of Xi’an Jiaotong University, Xi’an, China
| | - Mengyuan Gong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Pancreatic Disease Center of Xi’an Jiaotong University, Xi’an, China
| | - Simei Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Pancreatic Disease Center of Xi’an Jiaotong University, Xi’an, China
| | - Bao Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Pancreatic Disease Center of Xi’an Jiaotong University, Xi’an, China
| | - Bolun Qu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Pancreatic Disease Center of Xi’an Jiaotong University, Xi’an, China
| | - Zheng Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Pancreatic Disease Center of Xi’an Jiaotong University, Xi’an, China
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Pancreatic Disease Center of Xi’an Jiaotong University, Xi’an, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Pancreatic Disease Center of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Zheng Wang, ; Weikun Qian,
| | - Weikun Qian
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Pancreatic Disease Center of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Zheng Wang, ; Weikun Qian,
| |
Collapse
|
236
|
Wang Q, Guo F, Jin Y, Ma Y. Applications of human organoids in the personalized treatment for digestive diseases. Signal Transduct Target Ther 2022; 7:336. [PMID: 36167824 PMCID: PMC9513303 DOI: 10.1038/s41392-022-01194-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/09/2022] [Accepted: 09/13/2022] [Indexed: 11/15/2022] Open
Abstract
Digestive system diseases arise primarily through the interplay of genetic and environmental influences; there is an urgent need in elucidating the pathogenic mechanisms of these diseases and deploy personalized treatments. Traditional and long-established model systems rarely reproduce either tissue complexity or human physiology faithfully; these shortcomings underscore the need for better models. Organoids represent a promising research model, helping us gain a more profound understanding of the digestive organs; this model can also be used to provide patients with precise and individualized treatment and to build rapid in vitro test models for drug screening or gene/cell therapy, linking basic research with clinical treatment. Over the past few decades, the use of organoids has led to an advanced understanding of the composition of each digestive organ and has facilitated disease modeling, chemotherapy dose prediction, CRISPR-Cas9 genetic intervention, high-throughput drug screening, and identification of SARS-CoV-2 targets, pathogenic infection. However, the existing organoids of the digestive system mainly include the epithelial system. In order to reveal the pathogenic mechanism of digestive diseases, it is necessary to establish a completer and more physiological organoid model. Combining organoids and advanced techniques to test individualized treatments of different formulations is a promising approach that requires further exploration. This review highlights the advancements in the field of organoid technology from the perspectives of disease modeling and personalized therapy.
Collapse
Affiliation(s)
- Qinying Wang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fanying Guo
- School of Clinical Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yutao Jin
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yanlei Ma
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
237
|
Caputo D, Coppola A, Quagliarini E, Di Santo R, Capriotti AL, Cammarata R, Laganà A, Papi M, Digiacomo L, Coppola R, Pozzi D, Caracciolo G. Multiplexed Detection of Pancreatic Cancer by Combining a Nanoparticle-Enabled Blood Test and Plasma Levels of Acute-Phase Proteins. Cancers (Basel) 2022; 14:4658. [PMID: 36230585 PMCID: PMC9563576 DOI: 10.3390/cancers14194658] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 11/30/2022] Open
Abstract
The development of new tools for the early detection of pancreatic ductal adenocarcinoma (PDAC) represents an area of intense research. Recently, the concept has emerged that multiplexed detection of different signatures from a single biospecimen (e.g., saliva, blood, etc.) may exhibit better diagnostic capability than single biomarkers. In this work, we develop a multiplexed strategy for detecting PDAC by combining characterization of the nanoparticle (NP)-protein corona, i.e., the protein layer that surrounds NPs upon exposure to biological fluids and circulating levels of plasma proteins belonging to the acute phase protein (APPs) family. As a first step, we developed a nanoparticle-enabled blood (NEB) test that employed 600 nm graphene oxide (GO) nanosheets and human plasma (HP) (5% vol/vol) to produce 75 personalized protein coronas (25 from healthy subjects and 50 from PDAC patients). Isolation and characterization of protein corona patterns by 1-dimensional (1D) SDS-PAGE identified significant differences in the abundance of low-molecular-weight corona proteins (20-30 kDa) between healthy subjects and PDAC patients. Coupling the outcomes of the NEB test with the circulating levels of alpha 2 globulins, we detected PDAC with a global capacity of 83.3%. Notably, a version of the multiplexed detection strategy run on sex-disaggregated data provided substantially better classification accuracy for men (93.1% vs. 77.8%). Nanoliquid chromatography tandem mass spectrometry (nano-LC MS/MS) experiments allowed to correlate PDAC with an altered enrichment of Apolipoprotein A-I, Apolipoprotein D, Complement factor D, Alpha-1-antichymotrypsin and Alpha-1-antitrypsin in the personalized protein corona. Moreover, other significant changes in the protein corona of PDAC patients were found. Overall, the developed multiplexed strategy is a valid tool for PDAC detection and paves the way for the identification of new potential PDAC biomarkers.
Collapse
Affiliation(s)
- Damiano Caputo
- Department of Surgery, University Campus Bio-Medico di Roma, Via Alvaro del Portillo 200, 00128 Rome, Italy
- General Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy
| | - Alessandro Coppola
- General Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy
| | - Erica Quagliarini
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Riccardo Di Santo
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Anna Laura Capriotti
- Department of Chemistry, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Roberto Cammarata
- General Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy
| | - Aldo Laganà
- Department of Chemistry, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Massimiliano Papi
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
| | - Luca Digiacomo
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Roberto Coppola
- Department of Surgery, University Campus Bio-Medico di Roma, Via Alvaro del Portillo 200, 00128 Rome, Italy
- General Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy
| | - Daniela Pozzi
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Giulio Caracciolo
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| |
Collapse
|
238
|
Sato N, Motoi F, Tajiki H, Kawaguchi K, Ohtsuka H, Takadate T, Nakagawa K, Takagi K, Suzuki T, Katayose Y, Fukudo S, Unno M. Expression of Corticotropin-Releasing Hormone and Its Receptors May Be Associated With Survival Rate in Pancreatic Cancer. GASTRO HEP ADVANCES 2022; 2:147-155. [PMID: 39130148 PMCID: PMC11308248 DOI: 10.1016/j.gastha.2022.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 09/09/2022] [Indexed: 08/13/2024]
Abstract
Background and Aims Corticotropin-releasing hormone (CRH) is a major regulator of the stress response to internal and external factors. CRH and its receptors (CRHR1 and CRHR2) are expressed in the central nervous system and some cancer cells, suggesting the importance of CRH signaling in pancreatic cancers. However, the clinicopathological significance of CRH remains unknown because the immunolocalization of CRH, CRHR1, and CRHR2 has not been examined in pancreatic carcinoma tissues. We clarified the correlation of the expression of CRH and its receptors with overall survival in pancreatic cancer. Methods This study evaluated 96 patients with pancreatic cancer who underwent microscopic complete resection (R0) but not neoadjuvant chemotherapy from 1988 to 2007 at Tohoku University Hospital, Japan. CRH, CRHR1, and CRHR2 immunoreactivity were detected in the pancreatic carcinoma cells. Overall survival curves were generated according to the Kaplan-Meier method. Results CRHR1 immunoreactivity was significantly associated with an increased risk of poorer prognosis in all patients (P = .038) and the adjuvant therapy group (P = .022). Overall survival was worse in the CRHR1-positive group than in the CRHR1-negative group among the 62 patients treated with gemcitabine hydrochloride (P = .046) and the 22 patients treated with other drugs (P = .047). CRHR1 expression was correlated with survival in univariate analysis but not in multivariate analysis. Conclusion This study is the first to immunolocalize CRH, CRHR1, and CRHR2 in pancreatic carcinoma tissues and to examine the biological prognosis. This study revealed that survival in patients with pancreatic cancer was significantly associated with expression of CRHR1 by assessing biological progression according to CRH and the expression of its receptors. However, CRHR1 expression was correlated with survival in univariate analysis but not in multivariate analysis.
Collapse
Affiliation(s)
- Naoko Sato
- School of Nursing, Fukushima Medical University, Fukushima, Japan
- Department of Behavioral Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Fuyuhiko Motoi
- Department of Surgery I, Yamagata University Graduate School of Medical Science, Yamagata, Japan
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hana Tajiki
- Department of Nursing, Osaka Medical and Pharmaceutical University Hospital, Osaka, Japan
| | - Kei Kawaguchi
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hideo Ohtsuka
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tatuyuki Takadate
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kei Nakagawa
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kiyoshi Takagi
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takashi Suzuki
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yu Katayose
- Division of Hepato-Biliary and Pancreatic Surgery, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Shin Fukudo
- Department of Behavioral Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
239
|
Associations Between Poor Oral Hygiene and Risk of Pancreatic Cancer: A Meta-analysis of Observational Studies. Pancreas 2022; 51:985-994. [PMID: 36607944 PMCID: PMC9835655 DOI: 10.1097/mpa.0000000000002143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVES Epidemiological studies have reported the association of poor oral hygiene, especially periodontal disease, and tooth loss with the risk of pancreatic cancer (PC). However, these studies have yielded inconsistent results. Therefore, this systematic review and meta-analysis aimed to investigate the relationship between oral disease and PC. METHODS We systematically searched the PubMed, Embase, and Cochrane Library databases for English literature since inception through May 2021. We used relative risks, hazard ratios, or odds ratios to measure the association between oral disease and PC. A fixed- or random-effects model was applied to evaluate pooled risk estimates, and sensitivity and subgroup analyses were performed to identify sources of heterogeneity and pooled estimation. RESULTS We identified 17 relevant observational studies involving 1,352,256 participants. Notably, oral disease correlated significantly with PC (hazard ratio [HR], 1.32; 95% confidence interval [CI], 1.13-1.54). In subgroup analyses, subjects with periodontal disease (HR, 1.38; 95% CI, 1.12-1.71) had a higher risk of developing PC than those with tooth loss (HR, 1.19; 95% CI, 0.97-1.46). CONCLUSIONS The results suggest that subjects with oral disease may face a significant and independent risk of PC. However, the mechanisms linking oral disease and PC are uncertain, and additional investigations of this correlation are warranted.
Collapse
|
240
|
Zhao B, Zhao B, Chen F. Diagnostic value of serum carbohydrate antigen 19-9 in pancreatic cancer: a systematic review and meta-analysis. Eur J Gastroenterol Hepatol 2022; 34:891-904. [PMID: 35913776 DOI: 10.1097/meg.0000000000002415] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
Carbohydrate antigen 19-9 (CA19-9) is the most widely used serum biomarker for detecting pancreatic cancer (PC). Since early diagnosis is important for improving PC prognosis, a comprehensive understanding of the diagnostic performance of CA19-9 is critical. This study focused on comprehensive evaluation of the efficacy of CA19-9 in PC diagnosis. Literature research was based on the seven databases. Studies released from January 2002 to January 2022 focused on the efficacy of CA19-9 in the detection of PC were included. Summarized sensitivity, specificity, and sROC/accuracy of discrimination (AUC) were estimated. Potential publication bias was measured with Funnel plot and Egger's test. Meta-regression was performed to detect possible causes of heterogeneity. Subgroup analysis was used to assess the diagnostic efficacy of CA19-9 under different conditions. The study is registered on PROSPERO (CRD42021253861). Seventy-nine studies containing 20 991 participants who met the criteria were included. The pooled sensitivity, specificity, and AUC of CA19-9 in diagnose PC were 72% (95% CI, 71-73%), 86% (95% CI, 85-86%), and 0.8474 (95% CI, 0.8272-0.8676). Subgroup analysis suggested that the diagnostic efficiency of CA19-9 in studies with healthy controls was the highest, followed by intraductal papillary mucinous neoplasm, in pancreatitis and diabetes were consistent with the overall result. Our analysis showed that serum CA19-9 had high and stable diagnostic efficacy for PC (not affected by diabetes). Subgroup analysis showed that serum CA19-9 yielded highest effectiveness in the diagnosis of pancreatic precancerous lesions, which indicated an irreplaceable clinical value in the early detection and warning value for PC.
Collapse
Affiliation(s)
- Boqiang Zhao
- Xi'an Jiaotong University Health Science Center, Xi'an, China
- The First School of Clinical Medicine, Xi'an, China
| | - Boyue Zhao
- Xi'an Jiaotong University Health Science Center, Xi'an, China
- Department of Epidemiology and Biostatistics, School of Public Health, Xi'an, China
| | - Fangyao Chen
- Xi'an Jiaotong University Health Science Center, Xi'an, China
- Department of Epidemiology and Biostatistics, School of Public Health, Xi'an, China
| |
Collapse
|
241
|
Wang J, Wang Q, Zhang P, Zhang R, He J. Efficacy and safety of traditional Chinese medicine for the treatment of pancreatic cancer: An overview of systematic reviews and meta-analyses. Front Pharmacol 2022; 13:896017. [PMID: 36120323 PMCID: PMC9475193 DOI: 10.3389/fphar.2022.896017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 08/02/2022] [Indexed: 12/09/2022] Open
Abstract
Pancreatic cancer is a highly malignant tumor with poor prognosis. Currently available Western medical management strategies are unable to prolong the survival time and reduce the mortality of patients with pancreatic cancer. Traditional Chinese medicine has achieved promising results in many clinical studies. This systematic review and meta-analysis (SR/MA) aimed to explore the benefits and evaluate the quality of evidence of traditional Chinese medicine-based interventions for preventing and treating pancreatic cancer. A systematic search of eight databases for SRs/MAs of randomized controlled trials on traditional Chinese medicine treatment for pancreatic cancer was conducted (from inception to April 2022). The methodological quality of the SRs/MAs was assessed using AMSTAR 2.0, and the quality of evidence was evaluated using the GRADE guide. Nine SRs/MAs, including 145 randomized controlled trials, were considered eligible for this study. The literature were published between 2014 and 2022. The sample size of randomized controlled trials in the MAs ranged from 336 to 1,989. The methodological quality of the nine studies was critically low. Among the 59 outcome indicators of the nine SRs/MAs, seven, 33, and 19 had moderate-, low-, and critically low-quality evidence, respectively, while high-quality evidence was not identified. The results for the long-term indicators, short-term indicators, and adverse reactions in the SRs/MAs displayed consistencies and differences. In conclusion, the methodological and evidence quality of the current evidence is generally low, highlighting the need for additional focus on implementation processes. Some evidence with moderate quality validated that several specific traditional Chinese medicine were optimum for improving the short-term clinical efficacy. However, more objective and high-quality investigations are warranted to verify the efficacy of traditional Chinese medicine for pancreatic cancer.
Collapse
Affiliation(s)
- Jing Wang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Qiuyuan Wang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Department of Orthopaedics, China-Japan Friendship Hospital, Beijing, China
| | - Peitong Zhang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Peitong Zhang,
| | - Ruoqi Zhang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Jie He
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
242
|
Ilic I, Ilic M. International patterns in incidence and mortality trends of pancreatic cancer in the last three decades: A joinpoint regression analysis. World J Gastroenterol 2022; 28:4698-4715. [PMID: 36157927 PMCID: PMC9476884 DOI: 10.3748/wjg.v28.i32.4698] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/05/2022] [Accepted: 06/20/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pancreatic cancer, as the one of most fatal malignancies, remains a critical issue in the global burden of disease. AIM To estimate trends in pancreatic cancer incidence and mortality worldwide in the last three decades. METHODS A descriptive epidemiological study was done. Pancreatic cancer incidence and mortality data were obtained from the database of the World Health Organization. Analysis of pancreatic cancer incidence and mortality during 2020 was performed. The age-standardized rates (ASRs, expressed per 100000) were presented. To estimate trends of incidence and mortality of pancreatic cancer, joinpoint regression analysis was used: the average annual percent change (AAPC) with the corresponding 95% confidence interval (95%CI) was calculated. Additionally, analysis was performed by sex and age. In this paper, the trend analysis included only countries with high and medium data quality. RESULTS A total of 495773 (262865 male and 232908 female) new cases and 466003 (246840 male and 219163 female) deaths from pancreatic cancer were reported worldwide in 2020. In both sexes, most of the new cases (191348; 38.6% of the total) and deaths (182074; 39.1% of the total) occurred in the Western Pacific Region. In both sexes, the highest ASRs were found in the European Region, while the lowest rates were reported in the South-East Asia Region. The general pattern of rising pancreatic cancer incidence and mortality was seen across countries worldwide in observed period. Out of all countries with an increase in pancreatic cancer incidence, females in France and India showed the most marked rise in incidence rates (AAPC = +3.9% and AAPC = +3.7%, respectively). Decreasing incidence trends for pancreatic cancer were observed in some countries, but without significance. Out of all countries with an increase in pancreatic cancer mortality rates, Turkmenistan showed the most marked rise both in males (AAPC = +10.0%, 95%CI: 7.4-12.5) and females (AAPC = +6.4%, 95%CI: 3.5-9.5). The mortality trends of pancreatic cancer were decreasing in both sexes only in Canada and Mexico. CONCLUSION Further research is needed to explain the cause of large international differences in incidence and mortality trends of pancreatic cancer in last three decades.
Collapse
Affiliation(s)
- Irena Ilic
- Faculty of Medicine, University of Belgrade, Belgrade 11000, Serbia
| | - Milena Ilic
- Department of Epidemiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac 34000, Serbia
| |
Collapse
|
243
|
Advances and Remaining Challenges in the Treatment for Borderline Resectable and Locally Advanced Pancreatic Ductal Adenocarcinoma. J Clin Med 2022; 11:jcm11164866. [PMID: 36013111 PMCID: PMC9410260 DOI: 10.3390/jcm11164866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/10/2022] [Accepted: 08/17/2022] [Indexed: 11/17/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the deadliest malignancies in the United States. Improvements in imaging have permitted the categorization of patients according to radiologic involvement of surrounding vasculature, i.e., upfront resectable, borderline resectable, and locally advanced disease, and this, in turn, has influenced the sequence of chemotherapy, surgery, and radiation therapy. Though surgical resection remains the only curative treatment option, recent studies have shown improved overall survival with neoadjuvant chemotherapy, especially among patients with borderline resectable/locally advanced disease. The role of radiologic imaging after neoadjuvant therapy and the potential benefit of adjuvant therapy for borderline resectable and locally advanced disease remain areas of ongoing investigation. The advances made in the treatment of patients with borderline resectable/locally advanced disease are promising, yet disparities in access to cancer care persist. This review highlights the significant advances that have been made in the treatment of borderline resectable and locally advanced PDAC, while also calling attention to the remaining challenges.
Collapse
|
244
|
Takikawa T, Hamada S, Matsumoto R, Tanaka Y, Kataoka F, Sasaki A, Masamune A. Senescent Human Pancreatic Stellate Cells Secrete CXCR2 Agonist CXCLs to Promote Proliferation and Migration of Human Pancreatic Cancer AsPC-1 and MIAPaCa-2 Cell Lines. Int J Mol Sci 2022; 23:ijms23169275. [PMID: 36012531 PMCID: PMC9409091 DOI: 10.3390/ijms23169275] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 11/16/2022] Open
Abstract
Interactions between pancreatic cancer cells and pancreatic stellate cells (PSCs) play an important role in the progression of pancreatic cancer. Recent studies have shown that cellular senescence and senescence-associated secretory phenotype factors play roles in the progression of cancer. This study aimed to clarify the effects of senescence-induced PSCs on pancreatic cancer cells. Senescence was induced in primary-cultured human PSCs (hPSCs) through treatment with hydrogen peroxide or gemcitabine. Microarray and Gene Ontology analyses showed the alterations in genes and pathways related to cellular senescence and senescence-associated secretory phenotype factors, including the upregulation of C-X-C motif chemokine ligand (CXCL)-1, CXCL2, and CXCL3 through the induction of senescence in hPSCs. Conditioned media of senescent hPSCs increased the proliferation—as found in an assessment with a BrdU incorporation assay—and migration—as found in an assessment with wound-healing and two-chamber assays—of pancreatic cancer AsPC-1 and MIAPaca-2 cell lines. SB225002, a selective CXCR2 antagonist, and SCH-527123, a CXCR1/CXCR2 antagonist, attenuated the effects of conditioned media of senescent hPSCs on the proliferation and migration of pancreatic cancer cells. These results suggest a role of CXCLs as senescence-associated secretory phenotype factors in the interaction between senescent hPSCs and pancreatic cancer cells. Senescent PSCs might be novel therapeutic targets for pancreatic cancer.
Collapse
|
245
|
The Prognostic Impact of Quitting Smoking at or around Diagnosis on the Survival of Patients with Gastrointestinal Cancers: A Systematic Literature Review. Cancers (Basel) 2022; 14:cancers14163857. [PMID: 36010851 PMCID: PMC9406224 DOI: 10.3390/cancers14163857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/02/2022] [Accepted: 08/08/2022] [Indexed: 11/20/2022] Open
Abstract
Simple Summary Smokers are at high risk of cancer of the gastrointestinal system, and many patients with newly diagnosed cancer of the oesophagus, stomach, colon-rectum, and liver are active smokers at diagnosis. In this review, we focused on whether stopping smoking shortly before diagnosis or afterwards (e.g., during treatment) may improve the chance of survival for these patients. We reviewed the scientific literature up to April 2022 and found only seven articles focusing on this topic. While very limited in number, these studies provided suggestive evidence in favour of a beneficial effect of smoking cessation for these patients. Smokers with newly diagnosed cancer of the gastrointestinal system should be encouraged to stop smoking and provided all the necessary support to achieve this goal. Abstract Cigarette smoking is a strong risk factor for the occurrence of gastrointestinal cancers, and a substantial proportion of newly diagnosed patients is made up of active smokers, yet the impact of smoking cessation at or around diagnosis on the clinical course of these cancers (whose prognosis is often unfavourable) has never been summarized to date. We reviewed studies published until 30 April 2022 that investigated whether smoking cessation at or around diagnosis favourably affects the clinical course of gastrointestinal cancers patients. Six studies were included for colorectal cancer patients, which provided limited yet suggestive evidence that quitters may have longer disease-specific survival compared to continued smokers. Only one study each focused on patients with gastric or HBV-positive liver cancer (both reporting a survival advantage for quitters vs. continued smokers), while we found no eligible studies for patients with cancer at other sites within the digestive system. More research is urgently needed to expand the evidence on the topic, given the potentially major clinical implications for these patients. Moreover, health professionals should provide the necessary smoking cessation support to any smoker who is undergoing diagnostic work-up or treatment for gastrointestinal cancer.
Collapse
|
246
|
Alshammari E, Zhang YX, Yang Z. Mechanistic and functional extrapolation of SET and MYND domain-containing protein 2 to pancreatic cancer. World J Gastroenterol 2022; 28:3753-3766. [PMID: 36157542 PMCID: PMC9367238 DOI: 10.3748/wjg.v28.i29.3753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/24/2022] [Accepted: 07/06/2022] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal neoplasms worldwide and represents the vast majority of pancreatic cancer cases. Understanding the molecular pathogenesis and the underlying mechanisms involved in the initiation, maintenance, and progression of PDAC is an urgent need, which may lead to the development of novel therapeutic strategies against this deadly cancer. Here, we review the role of SET and MYND domain-containing protein 2 (SMYD2) in initiating and maintaining PDAC development through methylating multiple tumor suppressors and oncogenic proteins. Given the broad substrate specificity of SMYD2 and its involvement in diverse oncogenic signaling pathways in many other cancers, the mechanistic extrapolation of SMYD2 from these cancers to PDAC may allow for developing new hypotheses about the mechanisms driving PDAC tumor growth and metastasis, supporting a proposition that targeting SMYD2 could be a powerful strategy for the prevention and treatment of PDAC.
Collapse
Affiliation(s)
- Eid Alshammari
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University, Detroit, MI 48201, United States
| | - Ying-Xue Zhang
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University, Detroit, MI 48201, United States
| | - Zhe Yang
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI 48201, United States
| |
Collapse
|
247
|
Luo Z, Wang L, Shang Z, Guo Q, Liu Q, Zhang M, Li T, Wang Y, Zhang Y, Zhang Y, Zhang X. A panel of necroptosis-related genes predicts the prognosis of pancreatic adenocarcinoma. Transl Oncol 2022; 22:101462. [PMID: 35635957 PMCID: PMC9157256 DOI: 10.1016/j.tranon.2022.101462] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/20/2022] [Accepted: 05/22/2022] [Indexed: 12/27/2022] Open
Abstract
The 5-NRGs signature can predict the prognosis of pancreatic adenocarcinoma. The 5-NRGs signature reflects the immune landscape of pancreatic adenocarcinoma. The 5-NRGs can be detected in exosomes of serum using RT-qPCR method.
Pancreatic adenocarcinoma (PAAD) has become one of the deadliest malignancies in the world. Since necroptosis plays a crucial role in regulating the immune system, it is necessary to develop novel prognostic biomarkers associated with necroptosis and explore its potential role in PAAD. The transcriptome RNA-seq data of PAAD were downloaded from the TCGA and GTEx databases. A prognostic signature was constructed by the least absolute shrinkage and selection operator (LASSO) Cox regression, and its prognostic value was evaluated by nomogram and validated in an independent GEO cohort. We identified a total of 24 differentially expressed NRGs in PAAD, and constructed a prognostic signature with 5 NRGs, which showed good performance in predicting the prognosis of PAAD patients. The ROC curves for 1-, 3-, and 5-year survival rate were 0.652, 0.778, and 0.817, respectively. This prognostic signature showed consistent prognosis prediction in an independent patient cohort. Furthermore, the correlations between 5-NRGs signature and TMB, MSI, histopathological classification, immune infiltration, immune types, and immunomodulators were all significant. Notably, the expression profiles of the five NRGs in exosomes of serum were consistent with their expression in tumor tissues. These data suggested that the 5-NRGs signature is a promising biomarker for predicting the prognosis of PAAD.
Collapse
|
248
|
Hennig A, Baenke F, Klimova A, Drukewitz S, Jahnke B, Brückmann S, Secci R, Winter C, Schmäche T, Seidlitz T, Bereuter JP, Polster H, Eckhardt L, Schneider SA, Brückner S, Schmelz R, Babatz J, Kahlert C, Distler M, Hampe J, Reichert M, Zeißig S, Folprecht G, Weitz J, Aust D, Welsch T, Stange DE. Detecting drug resistance in pancreatic cancer organoids guides optimized chemotherapy treatment. J Pathol 2022; 257:607-619. [PMID: 35373359 DOI: 10.1002/path.5906] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 03/11/2022] [Accepted: 03/30/2022] [Indexed: 12/17/2023]
Abstract
Drug combination therapies for cancer treatment show high efficacy but often induce severe side effects, resulting in dose or cycle number reduction. We investigated the impact of neoadjuvant chemotherapy (neoCTx) adaptions on treatment outcome in 59 patients with pancreatic ductal adenocarcinoma (PDAC). Resections with tumor-free margins were significantly more frequent when full-dose neoCTx was applied. We determined if patient-derived organoids (PDOs) can be used to personalize poly-chemotherapy regimens by pharmacotyping of treatment-naïve and post-neoCTx PDAC PDOs. Five out of ten CTx-naïve PDO lines exhibited a differential response to either the FOLFIRINOX or the Gem/Pac regimen. NeoCTx PDOs showed a poor response to the neoadjuvant regimen that had been administered to the respective patient in 30% of cases. No significant difference in PDO response was noted when comparing modified treatments in which the least effective single drug was removed from the complete regimen. Drug testing of CTx-naïve PDAC PDOs and neoCTx PDOs may be useful to guide neoadjuvant and adjuvant regimen selection, respectively. Personalizing poly-chemotherapy regimens by omitting substances with low efficacy could potentially result in less severe side effects, thereby increasing the fraction of patients receiving a full course of neoadjuvant treatment. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Alexander Hennig
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Franziska Baenke
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Anna Klimova
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- Institute for Medical Informatics and Biometry, Technical University Dresden, Dresden, Germany
| | - Stephan Drukewitz
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- Core Unit for Molecular Tumor Diagnostics (CMTD), Technical University Dresden, Dresden, Germany
| | - Beatrix Jahnke
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Sascha Brückmann
- Institute of Pathology and Tumor- and Normal Tissue Bank of the University Cancer Center (UCC), University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Ramona Secci
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
- TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Christof Winter
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
- TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tim Schmäche
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Therese Seidlitz
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Jean-Paul Bereuter
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Heike Polster
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Lisa Eckhardt
- Core Unit for Molecular Tumor Diagnostics (CMTD), Technical University Dresden, Dresden, Germany
| | - Sidney A Schneider
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Stefan Brückner
- Department of Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Renate Schmelz
- Department of Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Jana Babatz
- Department of Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Christoph Kahlert
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Marius Distler
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Jochen Hampe
- Department of Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
- Center for Regenerative Therapies (CRTD), Technische Universität (TU) Dresden, Dresden, Germany
| | - Maximilian Reichert
- German Cancer Consortium (DKTK), Partner Site Munich, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Translational Pancreatic Cancer Research Center, Medical Clinic and Polyclinic II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Center for Protein Assemblies (CPA), Technische Universität München, Munich, Germany
| | - Sebastian Zeißig
- Department of Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
- Center for Regenerative Therapies (CRTD), Technische Universität (TU) Dresden, Dresden, Germany
| | - Gunnar Folprecht
- Department of Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Jürgen Weitz
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Daniela Aust
- Institute of Pathology and Tumor- and Normal Tissue Bank of the University Cancer Center (UCC), University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Thilo Welsch
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Daniel E Stange
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| |
Collapse
|
249
|
Ketavarapu V, Ravikanth V, Sasikala M, Rao GV, Devi CV, Sripadi P, Bethu MS, Amanchy R, Murthy HVV, Pandol SJ, Reddy DN. Integration of metabolites from meta-analysis with transcriptome reveals enhanced SPHK1 in PDAC with a background of pancreatitis. BMC Cancer 2022; 22:792. [PMID: 35854233 PMCID: PMC9295503 DOI: 10.1186/s12885-022-09816-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 06/22/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Pathophysiology of transformation of inflammatory lesions in chronic pancreatitis (CP) to pancreatic ductal adenocarcinoma (PDAC) is not clear. METHODS We conducted a systematic review, meta-analysis of circulating metabolites, integrated this data with transcriptome analysis of human pancreatic tissues and validated using immunohistochemistry. Our aim was to establish biomarker signatures for early malignant transformation in patients with underlying CP and identify therapeutic targets. RESULTS Analysis of 19 studies revealed AUC of 0.86 (95% CI 0.81-0.91, P < 0.0001) for all the altered metabolites (n = 88). Among them, lipids showed higher differentiating efficacy between PDAC and CP; P-value (< 0.0001). Pathway enrichment analysis identified sphingomyelin metabolism (impact value-0.29, FDR of 0.45) and TCA cycle (impact value-0.18, FDR of 0.06) to be prominent pathways in differentiating PDAC from CP. Mapping circulating metabolites to corresponding genes revealed 517 altered genes. Integration of these genes with transcriptome data of CP and PDAC with a background of CP (PDAC-CP) identified three upregulated genes; PIGC, PPIB, PKM and three downregulated genes; AZGP1, EGLN1, GNMT. Comparison of CP to PDAC-CP and PDAC-CP to PDAC identified upregulation of SPHK1, a known oncogene. CONCLUSIONS Our analysis suggests plausible role for SPHK1 in development of pancreatic adenocarcinoma in long standing CP patients. SPHK1 could be further explored as diagnostic and potential therapeutic target.
Collapse
Affiliation(s)
- Vijayasarathy Ketavarapu
- grid.410866.d0000 0004 1803 177XAsian Healthcare Foundation, Asian Institute of Gastroenterology, Mindspace Rd, Gachibowli, Hyderabad, Telangana 500032 India
| | - Vishnubhotla Ravikanth
- grid.410866.d0000 0004 1803 177XAsian Healthcare Foundation, Asian Institute of Gastroenterology, Mindspace Rd, Gachibowli, Hyderabad, Telangana 500032 India
| | - Mitnala Sasikala
- grid.410866.d0000 0004 1803 177XAsian Healthcare Foundation, Asian Institute of Gastroenterology, Mindspace Rd, Gachibowli, Hyderabad, Telangana 500032 India
| | - G. V. Rao
- grid.410866.d0000 0004 1803 177XAIG Hospitals, Mindspace Rd, Gachibowli, Hyderabad, Telangana 500032 India
| | - Ch. Venkataramana Devi
- grid.412419.b0000 0001 1456 3750Department of Biochemistry, University College of Science, Osmania University, Hyderabad, 500 007 India
| | - Prabhakar Sripadi
- grid.417636.10000 0004 0636 1405Centre for Mass Spectrometry, Analytical & Structural Chemistry Department, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, 500 007 India
| | - Murali Satyanarayana Bethu
- grid.410865.eDivision of Applied Biology, CSIR-IICT (Indian Institute of Chemical Technology), Ministry of Science and Technology (GOI), Hyderabad, Telangana 500007 India ,grid.240614.50000 0001 2181 8635Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Elm &Carlton Streets, Buffalo, New York, 14221 USA
| | - Ramars Amanchy
- grid.410865.eDivision of Applied Biology, CSIR-IICT (Indian Institute of Chemical Technology), Ministry of Science and Technology (GOI), Hyderabad, Telangana 500007 India
| | - H. V. V. Murthy
- grid.410866.d0000 0004 1803 177XAsian Healthcare Foundation, Asian Institute of Gastroenterology, Mindspace Rd, Gachibowli, Hyderabad, Telangana 500032 India
| | - Stephen J. Pandol
- grid.50956.3f0000 0001 2152 9905Department of Medicine, Division of Digestive and Liver Diseases, Cedars-Sinai Medical Center, Los Angeles, CA USA
| | - D. Nageshwar Reddy
- grid.410866.d0000 0004 1803 177XAIG Hospitals, Mindspace Rd, Gachibowli, Hyderabad, Telangana 500032 India
| |
Collapse
|
250
|
Yang JM, Yang XY, Wan JH. Multiple roles for cholinergic signaling in pancreatic diseases. World J Gastroenterol 2022; 28:2910-2919. [PMID: 35978870 PMCID: PMC9280742 DOI: 10.3748/wjg.v28.i25.2910] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 05/18/2022] [Accepted: 06/13/2022] [Indexed: 02/06/2023] Open
Abstract
Cholinergic nerves are widely distributed throughout the human body and participate in various physiological activities, including sensory, motor, and visceral activities, through cholinergic signaling. Cholinergic signaling plays an important role in pancreatic exocrine secretion. A large number of studies have found that cholinergic signaling overstimulates pancreatic acinar cells through muscarinic receptors, participates in the onset of pancreatic diseases such as acute pancreatitis and chronic pancreatitis, and can also inhibit the progression of pancreatic cancer. However, cholinergic signaling plays a role in reducing pain and inflammation through nicotinic receptors, but enhances the proliferation and invasion of pancreatic tumor cells. This review focuses on the progression of cholinergic signaling and pancreatic diseases in recent years and reveals the role of cholinergic signaling in pancreatic diseases.
Collapse
Affiliation(s)
- Jun-Min Yang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Xiao-Yu Yang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Jian-Hua Wan
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| |
Collapse
|