1
|
Fu M, Li Y, Wang J. Incidence and Mortality of Colorectal Cancer in Asia in 2022 and Projections for 2050. J Gastroenterol Hepatol 2025; 40:1143-1156. [PMID: 40018878 DOI: 10.1111/jgh.16910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/05/2025] [Accepted: 02/14/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Colorectal cancer is an escalating public health concern in Asia, characterized by unique epidemiological patterns. METHODS We analyzed colorectal cancer data from 47 Asian countries using GLOBOCAN 2022. Spearman's correlation assessed the relationship between the Human Development Index and cancer rates. Projections for 2050 incidence and mortality were based on demographic forecasts. RESULTS In 2022, Asia accounted for 50.2% of global colorectal cancer cases, with 966.4 thousand new cases and 462.3 thousand deaths, including 10% in younger individuals. Age-standardized rates of incidence and mortality were 15.6/100 000 and 7.1/100 000, respectively. Incidence rates were rising faster in younger and male individuals. A significant correlation was found between the Human Development Index and cancer rates. Japan had the highest incidence rate (45.5/100 000 males; 28.5/100 000 females), and Brunei Darussalam had the highest mortality rate (21/100 000 males; 13.9/100 000 females). China recorded the highest incidence and mortality counts, with 307.7 thousand new cases and 142.6 thousand deaths in males, and 209.4 thousand new cases and 97.4 thousand deaths in females. By 2050, 1.87 million new cases and 1.01 million deaths are expected to occur in Asia, with the largest relative increases occurring in low HDI countries. CONCLUSIONS These findings highlight the need for targeted policies in low HDI countries, focusing on public awareness, early detection, prevention, and improving healthcare infrastructure. Interventions for younger individuals and males are also essential to address rising incidence rates.
Collapse
Affiliation(s)
- Mengxia Fu
- Galactophore Department, Galactophore Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yanping Li
- Galactophore Department, Galactophore Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Jian Wang
- Department of Gastrointestinal Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Hetta HF, Alqifari SF, Alshehri K, Alhowiti A, Alharbi SS, Mirghani H, Alrasheed T, Mostafa MEA, Sheikh M, Elodemi M, Alhumaid SA, Ramadan YN, Abd Ellah NH, Sayad R. Efficacy of Anlotinib Plus Docetaxel in Advanced NSCLC Previously Treated with Platinum-Based Chemotherapy: A Systematic Review and Meta-Analysis. Pharmaceuticals (Basel) 2025; 18:652. [PMID: 40430471 DOI: 10.3390/ph18050652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/22/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Anlotinib is a novel oral antiangiogenic tyrosine kinase inhibitor (TKI) approved as a third-line treatment for advanced non-small-cell lung cancer (NSCLC). However, its efficacy in combination with docetaxel remains incompletely understood. Given the need for effective second-line therapies after platinum-based chemotherapy, this systematic review aims to evaluate the therapeutic potential of anlotinib plus docetaxel in advanced NSCLC. Methods: The PubMed, WOS, Medline, and Scopus databases were screened for published articles up to 12 April 2024. We included RCTs comparing anlotinib plus docetaxel with docetaxel alone in advanced NSCLC after receiving platinum-based chemotherapy, reporting progression-free survival (PFS), objective response rate (ORR), and disease control rate (DCR) as outcomes for both groups. Results: Our systematic review included three randomized controlled trials (RCTs) with a total of 151 patients in the anlotinib plus docetaxel group and 132 in the docetaxel-only group. Meta-analysis results demonstrated that the combination therapy significantly prolonged PFS (mean difference (MD) = 2.98, 95% confidence interval (CI), 1.95-4.00; p < 0.00001) and improved ORR (risk ratio (RR) = 3.04, 95% CI = 1.77-5.24; p < 0.00001). Additionally, the DCR was notably higher in the combination group (RR = 1.58, 95% CI = 1.34-1.87; p < 0.00001). Conclusions: Anlotinib plus docetaxel appears to be more effective as a second-line treatment of advanced NSCLC than docetaxel in prolonging PFS and increasing ORR and DCR.
Collapse
Affiliation(s)
- Helal F Hetta
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Saleh F Alqifari
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Khaled Alshehri
- Department of Internal Medicine (Neurology), Faculty of Medicine, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Amirah Alhowiti
- Department of Family and Community Medicine, Faculty of Medicine, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Saud S Alharbi
- Department of Family and Community Medicine, Faculty of Medicine, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Hyder Mirghani
- Department of Internal Medicine, Faculty of Medicine, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Tariq Alrasheed
- Department of Internal Medicine, Faculty of Medicine, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Mohamed E A Mostafa
- Department of Anatomy, Faculty of Medicine, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Mohammed Sheikh
- Department of Pediatric, Faculty of Medicine, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Mahmoud Elodemi
- Department of Pharmacology, Faculty of Medicine, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Sultan A Alhumaid
- Department of Family Medicine, King Salman Armed Forces Hospital, Tabuk 71491, Saudi Arabia
| | - Yasmin N Ramadan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt
| | - Noura H Abd Ellah
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Badr University in Assiut, Naser City 2014101, Egypt
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt
| | - Reem Sayad
- Department of Histology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| |
Collapse
|
3
|
Liang J, Zhai P, Cheng G, Han J, Song X. N-Acetyltransferase 10 Promotes Gastric Cancer Progression by Mediating the N4-Acetylcytidine Modification of Lactate Dehydrogenase A. J Biochem Mol Toxicol 2025; 39:e70227. [PMID: 40079254 DOI: 10.1002/jbt.70227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025]
Abstract
The N4-acetylcytidine (ac4C) modification, which is catalyzed by NAT10, represents a significant posttranscriptional modification of mRNA in multiple cancers. However, the significance of this modification in gastric cancer (GC) progression remains unclear. To evaluate the potential of differential NAT10 expression in GC, RT-qPCR and western blot were employed. Dot blot and acRIP were utilized for total ac4C and LDHA mRNA ac4C detection. Subsequently, the effects of NAT10 on GC cell viability and glycolysis were assessed by Cell Counting Kit-8 and glycolysis-related indicator detection Kits. Furthermore, rescue experiments and mice xenograft experiments were conducted to investigate the mechanism underlying the NAT10/LDHA signaling axis in GC. This study identified upregulated NAT10 and ac4C levels in GC. Knockdown of NAT10 led to inhibited cell viability and glycolysis. Additionally, NAT10 directly bound to LDHA mRNA. NAT10 silencing decreased the expression and stability of LDHA mRNA, as well as its ac4C modification level. Interestingly, LDHA overexpression partially reversed the effects of NAT10 knockdown on cell viability and glycolysis. Eventually, the oncogenic effect of NAT10/ac4C/LDHA axis was confirmed in xenograft experiments. NAT10 promoted the GC progression by mediating the ac4C modification of LDHA mRNA, which could serve as a potential therapeutic target for GC.
Collapse
Affiliation(s)
- Juan Liang
- Oncology Department, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Peng Zhai
- Nutrition Department, Shanxi Children's Hospital, Taiyuan, Shanxi, China
| | - Guohua Cheng
- Oncology Department, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jinlong Han
- Oncology Department, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiang Song
- Oncology Department, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
4
|
Hewitt E, Bouche G, Alencar AC, Bigelsen SJ, Radu R, Stoyanova-Beninska V, Carrato A, Valsecchi F, Soler Cantón A, van der Meer HG, García Bermejo ML, Budillon A, Cardone L, Rooman I, Platteeuw H, Baijet J, Fuchs C. Drug Repurposing in Pancreatic Cancer: A Multi-Stakeholder Perspective to Improve Treatment Options for Pancreatic Cancer Patients. Cancer Manag Res 2025; 17:429-440. [PMID: 40046652 PMCID: PMC11881603 DOI: 10.2147/cmar.s483151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 02/02/2025] [Indexed: 04/17/2025] Open
Abstract
Pancreatic cancer (PC) remains one of the most challenging malignancies to treat. Current therapeutic options are unsatisfactory, and there is an urgent need for more effective and less toxic drugs to improve the dismal prognosis of PC. In recent years, drug repurposing (DR) has emerged as an attractive strategy to identify novel treatments for PC by leveraging existing drugs approved for other indications. Through the use of electronic medical records, Artificial Intelligence, study of metabolic pathways, signalling pathways, and many other approaches, it has become much easier in recent years to identify potential novel uses for old drugs. Although policy, funding and research attention in this area are steadily growing, major challenges to efficient and effective patient-centric DR in PC need to be addressed. These include but are not limited to regulatory, financial and funding barriers and the lack of coordination and collaboration among several sectors and stakeholders. To explore the opportunities and challenges associated with DR in PC, a one-day multi-stakeholder meeting was held on 14th of November 2023 in Brussels, Belgium as part of the REMEDi4ALL project. This meeting provided a platform for researchers, clinicians, industry representatives, funders, regulatory experts, and patient advocates to discuss and propose actions to optimize and accelerate DR in PC. Insights from this meeting support the potential of DR to enhance PC treatment options while highlighting the importance of systemic and supportive changes in the regulatory, policy and funding landscapes, interdisciplinary collaboration, data sharing, and patient involvement in driving therapeutic innovation. This summary highlights key outcomes and recommendations from the meeting in informing future efforts to advance DR initiatives in the context of PC.
Collapse
Affiliation(s)
| | | | | | - Stephen J Bigelsen
- Patient Advocate, Department of Allergy, Asthma and Immunology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | | | | | - Alfredo Carrato
- Department of Medical Oncology, Alcalá University, Instituto Ramón Y Cajal de Investigación Sanitaria (IRYCIS), Centro de Investigación Biomédica En Red de Cáncer (CIBERONC), Ramon Y Cajal University Hospital, Madrid, Spain
- Pancreatic Cancer Europe, Brussels, Belgium
| | | | - Alicia Soler Cantón
- EATRIS ERIC, European Infrastructure for Translational Medicine, Amsterdam, the Netherlands
| | | | - María Laura García Bermejo
- Biomarkers and Therapeutic Targets Group, Instituto Ramón Y Cajal de Investigación Sanitaria (IRYCIS), Ramon Y Cajal University Hospital, Madrid, Spain
| | - Alfredo Budillon
- Scientific Directorate, Istituto Nazionale Tumori – IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Luca Cardone
- Institute of BiOChemistry and Cell Biology (IBBC), CNR c/o Campus Internazionale “a.buzzati-Traverso”, Monterotondo Scalo Roma (Rome), Italy
- Department of Tumour Immunology and Immunotherapy, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Ilse Rooman
- The Anticancer Fund, Meise, Belgium
- Translational Oncology Research Center (TORC), Laboratory of Medical and Molecular Oncology, (LMMO) Vrije Universiteit Brussel, Brussels, Belgium
| | | | | | | |
Collapse
|
5
|
Eid R, Tarabay A, Decazes P, David C, Kerbage F, Zeghondy J, Antoun L, Smolenschi C, Fuerea A, Valery M, Boige V, Gelli M, Tselikas L, Durand-Labrunie J, Belkouchi Y, Littisha L, Ammari S, Ducreux M, Lassau N, Hollebecque A. Predictive factors of FOLFIRINOX chemotherapy toxicity in pancreatic adenocarcinoma patients. Future Oncol 2025; 21:691-697. [PMID: 39924679 PMCID: PMC11881864 DOI: 10.1080/14796694.2025.2461442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 01/29/2025] [Indexed: 02/11/2025] Open
Abstract
INTRODUCTION FOLFIRINOX, a primary chemotherapy for metastatic pancreatic cancer, often causes severe toxicity, necessitating hospitalization and dose adjustments. This study aims to identify predictors of FOLFIRINOX toxicity, focusing on biological, clinical, and anthropometric factors. MATERIAL & METHODS This retrospective study analyzes pancreatic adenocarcinoma patients on FOLFIRINOX, assessing pre-treatment biological, clinical, and anthropometric traits. Hospitalizations and tolerance during the first chemotherapy month were evaluated using CTCAE v5.0 grading, with early toxicity assessed via anthropometric factors using Anthropometer3DNet software from pre-treatment scans. RESULTS In 152 pancreatic cancer patients (median age: 62), FOLFIRINOX was administered in metastatic (81%), locally advanced (14%), and adjuvant/neoadjuvant (5%) settings. Performance Status was zero (49%), one (41%) and ≥ 2 (10%). Median follow-up was 62.5 months, with median overall survival of 13.7 months and progression-free survival of 8.9 months. First-cycle dose reduction occurred in 14% of patients. Within the first month, 48% experienced toxicity leading to hospitalization and/or dose reduction, with 28% requiring a median 8-day hospitalization. Low muscle body mass (MBM) significantly correlated with dose reduction (AUC 0.63; p = 0.005). An NLR ratio less than 4 was significantly associated with longer OS (p = 0.001). CONCLUSION Low MBM is linked to FOLFIRINOX toxicity, suggesting MBM assessment could allow better selection of patients to avoid these toxicities, warranting further confirmation in larger cohorts.
Collapse
Affiliation(s)
- Roland Eid
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Anthony Tarabay
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Pierre Decazes
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Clémence David
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Fouad Kerbage
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Jean Zeghondy
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Leony Antoun
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Cristina Smolenschi
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Alina Fuerea
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Marine Valery
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Valerie Boige
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Maximiliano Gelli
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Lambros Tselikas
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | | | - Younes Belkouchi
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Lawrance Littisha
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Samy Ammari
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Michel Ducreux
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Nathalie Lassau
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Antoine Hollebecque
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
6
|
Yu W, Zhou D, Meng F, Wang J, Wang B, Qiang J, Shen L, Wang M, Fang H. The global, regional burden of pancreatic cancer and its attributable risk factors from 1990 to 2021. BMC Cancer 2025; 25:186. [PMID: 39891086 PMCID: PMC11786447 DOI: 10.1186/s12885-025-13471-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 01/07/2025] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND Pancreatic cancer is the 12th most common type of cancer, and the sixth leading cause of cancer-related mortality, worldwide. Up-to-date statistics on pancreatic cancer would provide us with a better understanding of epidemiology and identify the causative risk factors for the prevention of this disease. METHODS The degree and change patterns of exposure as well as the attributable cancer burden, including incidence, mortality, disability-adjusted life years (DALYs), and prevalence in global and regional, by sex, age, year, for pancreatic cancer, with the data extracted from the Global Burden of Diseases Study (GBD) 2021. All data analyses were conducted using linear regression analysis and the Joinpoint software (version 5.0.1). RESULTS In 2021, 508,533 new cases of pancreatic cancer have been reported; the mortality and prevalence rate increased to 5.95, and 5.12 respectively; and the global DALYs rate increased to 130.33 this year. Besides, the pancreatic cancer-associated rates of incidence, mortality, DALYs, and prevalence were higher in males than in females. In addition, these indicators in the high SDI (Sociodemographic index) region were higher than the global mean. To date, the high fasting plasma glucose remained the major risk factor that influenced the incidence, mortality, DALYs, and prevalence of pancreatic cancer, followed by tobacco and high body mass index (BMI). CONCLUSIONS Results of this study suggest that the burden of pancreatic cancer is increasing generally, therefore, more attention and measures should be taken to cope with this situation.
Collapse
Affiliation(s)
- Weidong Yu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Danyi Zhou
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Fanhao Meng
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Jinjing Wang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Bo Wang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Jianling Qiang
- Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, 322100, China
| | - Lijun Shen
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Maofeng Wang
- Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, 322100, China.
| | - Hezhi Fang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China.
| |
Collapse
|
7
|
Guo W, Hu L, Gao Z, Liu X, Yang X, Wang X. The up-regulation of SPTAN1 expression in Pancreatic adenocarcinoma is associated with tumor immune invasion and poor clinical prognosis. BMC Gastroenterol 2025; 25:5. [PMID: 39762747 PMCID: PMC11706198 DOI: 10.1186/s12876-024-03581-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Pancreatic adenocarcinoma (PAAD) is a common malignancy with a very low survival rate. More and more studies have shown that SPTAN1 may be involved in the development and progression of a variety of tumors, including rectal cancer, Pancreatic adenocarcinoma, etc., and may affect their prognosis. METHODS Bioinformatics technology was used to analyze the relationship between SPTAN1 expression in PAAD and immune cell infiltration, immune regulatory factors and chemokines, and cell experiments were used to verify the relationship between SPTAN1 knock down and migration, invasion, apoptosis and cycle changes of PAAD cell lines. In addition, immunohistochemical staining of SPTAN1 was performed by tissue microarray (TMA) to study the relationship between high expression of SPTAN1 and clinicopathological features and overall survival rate. RESULTS The expression of SPTAN1 is significantly correlated with immune cell infiltration, immunomodulators, chemokines and their receptors. In addition, it was found that the knock-down of SPTAN1 inhibited the migration and invasion ability of PAAD cell lines, promoted the apoptosis of cell lines, and also affected the changes of cell cycle. Immunohistochemical staining using tissue microarray (TMA) showed that the high expression of SPTAN1 was associated with M stage (P = 0.004) and CA199 (P = 0.012), and the overall survival rate of the high expression group was significantly lower than that of the low expression group (P = 0.043). CONCLUSION Our results suggest that up-regulation of SPTAN1 is related to cell migration, invasion, apoptosis and cycle changes, and is associated with tumor immune invasion and poor prognosis of PAAD.
Collapse
Affiliation(s)
- Wei Guo
- Department of Interventional Vascular Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200082, China
| | - LingYu Hu
- Department of General Surgery, The Second Affiliated Hospital of Jiaxing University, Zhejiang Province, Jiaxing, 314000, China
| | - ZhaoFeng Gao
- Department of General Surgery, The Second Affiliated Hospital of Jiaxing University, Zhejiang Province, Jiaxing, 314000, China
| | - XiaoRong Liu
- Department of General Surgery, The Second Affiliated Hospital of Jiaxing University, Zhejiang Province, Jiaxing, 314000, China
| | - XiaoDan Yang
- Department of General Surgery, The Second Affiliated Hospital of Jiaxing University, Zhejiang Province, Jiaxing, 314000, China.
| | - XiaoGuang Wang
- Department of General Surgery, The Second Affiliated Hospital of Jiaxing University, Zhejiang Province, Jiaxing, 314000, China.
| |
Collapse
|
8
|
D’Antonio DL, Zenoniani A, Umme S, Piattelli A, Curia MC. Intratumoral Fusobacterium nucleatum in Pancreatic Cancer: Current and Future Perspectives. Pathogens 2024; 14:2. [PMID: 39860963 PMCID: PMC11768203 DOI: 10.3390/pathogens14010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/22/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
The intratumoral microbiome plays a significant role in many cancers, such as lung, pancreatic, and colorectal cancer. Pancreatic cancer (PC) is one of the most lethal malignancies and is often diagnosed at advanced stages. Fusobacterium nucleatum (Fn), an anaerobic Gram-negative bacterium primarily residing in the oral cavity, has garnered significant attention for its emerging role in several extra-oral human diseases and, lately, in pancreatic cancer progression and prognosis. It is now recognized as oncobacterium. Fn engages in pancreatic tumorigenesis and metastasis through multifaceted mechanisms, including immune response modulation, virulence factors, control of cell proliferation, intestinal metabolite interactions, DNA damage, and epithelial-mesenchymal transition. Additionally, compelling research suggests that Fn may exert detrimental effects on cancer treatment outcomes. This paper extends the perspective to pancreatic cancer associated with Fn. The central focus is to unravel the oncogenomic changes driven by Fn in colonization, initiation, and promotion of pancreatic cancer development. The presence of Fusobacterium species can be considered a prognostic marker of PC, and it is also correlated to chemoresistance. Furthermore, this review underscores the clinical research significance of Fn as a potential tumor biomarker and therapeutic target, offering a novel outlook on its applicability in cancer detection and prognostic assessment. It is thought that given the role of Fn in tumor formation and metastasis processes via its FadA, FapA, Fap2, and RadD, new therapies for tumor treatment targeting Fn will be developed.
Collapse
Affiliation(s)
- Domenica Lucia D’Antonio
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (D.L.D.); (A.Z.); (S.U.)
| | - Anna Zenoniani
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (D.L.D.); (A.Z.); (S.U.)
| | - Samia Umme
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (D.L.D.); (A.Z.); (S.U.)
- Department of Neuroscience, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy
| | - Adriano Piattelli
- School of Dentistry, Saint Camillus International University of Health and Medical Sciences (UniCamillus), 00131 Rome, Italy;
- Facultad de Medicina, UCAM Universidad Católica San Antonio de Murcia, 30107 Murcia, Spain
| | - Maria Cristina Curia
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (D.L.D.); (A.Z.); (S.U.)
| |
Collapse
|
9
|
Wang Q, Liu J, Yang Z. Global, regional, and national burden of pancreatic cancer from 1990 to 2021, with projections for 25 years: a systematic analysis for the Global Burden of Disease Study 2021. Eur J Cancer Prev 2024:00008469-990000000-00192. [PMID: 39718214 DOI: 10.1097/cej.0000000000000942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
This study examines the global burden of pancreatic cancer from 1990 to 2021 and projects future trends, aiming to provide insights for health policy and resource allocation to mitigate the disease's impact. We assessed the pancreatic cancer burden globally and by subgroups, employing linear regression models to analyze trends from 1990 to 2021. Cluster analysis was used to evaluate burden patterns across Global Burden of Disease regions. Forecasting was conducted using the age-period-cohort model and its Bayesian variant. Additionally, we evaluated risk factor contributions to the pancreatic cancer burden and used frontier analysis to explore the relationship between sociodemographic advancements and cancer rates. In 2021, pancreatic cancer accounted for 508 533 new cases, 439 001 prevalent cases, 505 752 deaths, and 11 316 963 disability-adjusted life years (DALYs). High-risk groups included males and middle-aged to older adults, with high-risk areas identified in regions with higher sociodemographic index (SDI). From 1990 to 2021, both pancreatic cancer cases and age-standardized rates (ASR) increased. Notably, high fasting plasma glucose surpassed tobacco as a leading risk factor for pancreatic cancer. Frontier analysis revealed an inverse relationship between SDI and pancreatic cancer ASR, plateauing at an SDI of 0.60. The global burden of pancreatic cancer continues to rise, with significant disparities across demographic and geographic segments. These findings highlight the need for targeted interventions and resource allocations to address this growing public health challenge.
Collapse
Affiliation(s)
- Qihong Wang
- Department of Digestive Endoscopy, General Hospital of Northern Theater Command, Shenyang, Liaoning Province, China
| | | | | |
Collapse
|
10
|
Brandtmüller A, Meiwald A, Oliver E, Hughes R, Capote PMG, Weston G, Bencina G. The productivity cost of mortality due to lung cancer, breast cancer and melanoma in Europe across 2010, 2015 and 2019. J Cancer Policy 2024; 42:100499. [PMID: 39147239 DOI: 10.1016/j.jcpo.2024.100499] [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: 02/16/2024] [Accepted: 08/05/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Cancer caused an estimated 2.2 million deaths across Europe in 2020. This analysis estimated the cost of lost productivity due to premature deaths associated with lung, breast and melanoma cancer and investigated the temporal trends across European regions across 2010, 2015 and 2019. METHOD The human capital approach was used to estimate the indirect costs from lung, melanoma, and breast cancers (ICD-10 code: C33-34, C43, and C50, respectively) in Northern, Eastern, Southern, and Western Europe. Age-specific mortality, and country-specific wages and employment rates were used to calculate years of productive life lost (YPLL), YPLL/death and present value of future lost productivity (PVFLP). Data were sourced from the World Health Organization, Eurostat, and the World Bank. RESULTS The number of cancer deaths remained relatively stable from 2010 to 2019. YPLL/death decreased across all European regions and for all cancers between 2010 and 2019 (reported ranges across European regions; lung cancer: 25-42 %; breast cancer: 18-21 %; melanoma: 31-37 %). In Europe, the decrease in PVFLP in 2019 compared to 2010 was €2995M for lung cancer, €295M for melanoma, and €466M for breast cancer, with an overall reduction of productivity cost of €3756M in these cancer types. CONCLUSION The results from this study illustrate a decreased trend in productivity costs from 2010 to 2019 which could be driven by deaths occurring at an older age, suggesting that advances in cancer prevention and the treatment landscape have extended the life of cancer patients, yielding less productivity losses. POLICY SUMMARY The indirect economic costs modelled show the impact of past effective health policies and new treatments. Continued efforts to improve public health policies in supporting public awareness of risk factors and value of early diagnosis could lead to further reduction in these losses. Prevention, early diagnosis, and activation of early treatment pathways could serve to reduce loss of life and improve productivity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Goran Bencina
- Outcomes Research, Value & Implementation, MSD Spain, Madrid, Spain
| |
Collapse
|
11
|
Xiao M, Xue J, Jin E. SPOCK: Master regulator of malignant tumors (Review). Mol Med Rep 2024; 30:231. [PMID: 39392048 PMCID: PMC11487499 DOI: 10.3892/mmr.2024.13355] [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: 05/27/2024] [Accepted: 08/20/2024] [Indexed: 10/12/2024] Open
Abstract
SPARC/osteonectin, CWCV and Kazal‑like domain proteoglycan (SPOCK) is a family of highly conserved multidomain proteins. In total, three such family members, SPOCK1, SPOCK2 and SPOCK3, constitute the majority of extracellular matrix glycoproteins. The SPOCK gene family has been demonstrated to serve key roles in tumor regulation by affecting MMPs, which accelerates the progression of cancer epithelial‑mesenchymal transition. In addition, they can regulate the cell cycle via overexpression, inhibit tumor cell proliferation by inactivating PI3K/AKT signaling and have been associated with numerous microRNAs that influence the expression of downstream genes. Therefore, the SPOCK gene family are potential cancer‑regulating genes. The present review summarizes the molecular structure, tissue distribution and biological function of the SPOCK family of proteins, in addition to its association with cancer. Furthermore, the present review documents the progress made in investigations into the role of SPOCK, whilst also discussing prospects for the future of SPOCK‑targeted therapy, to provide novel ideas for clinical application and treatment.
Collapse
Affiliation(s)
- Mingyuan Xiao
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110134, P.R. China
| | - Jiancheng Xue
- Department of Otolaryngology, Head and Neck Surgery, The Second People's Hospital of Shenzhen, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518035, P.R. China
- Shenzhen Clinical Research Center for Otolaryngology Diseases, The Second People's Hospital of Shenzhen, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518035, P.R. China
| | - Enli Jin
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110134, P.R. China
| |
Collapse
|
12
|
García García de Paredes A, Martínez Moneo E, Lariño-Noia J, Earl J. Pancreatic cancer screening in high-risk individuals. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2024; 116:519-522. [PMID: 39087662 DOI: 10.17235/reed.2024.10635/2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
The incidence of pancreatic cancer is increasing, although globally it represents less than 3% of all cancers. Despite advances in medical and surgical management, survival rates have not significantly improved in recent years. Consequently, pancreatic cancer, though relatively uncommon, is the third leading cause of cancer-related deaths. This is primarily due to the disease´s late detection. Symptoms appear late and are nonspecific, and over 80% of cases are diagnosed at an advanced stage and unsuitable for curative surgery, resulting in a five-year survival rate below 10%. However, the exceptional cases that are diagnosed early show five-year survival rates exceeding 80%. Therefore, one of the keys to improving pancreatic cancer prognosis lies in early detection, making screening in high-risk individuals a potentially crucial strategy.
Collapse
Affiliation(s)
| | | | | | - Julie Earl
- Biomarkers and Personalized Approach to Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS)
| |
Collapse
|
13
|
Wang P, Gao X, Zheng W, Zhang J. Potential impact of epithelial splicing regulatory protein 1 (ESRP1) associated with tumor immunity in pancreatic adenocarcinoma. J Proteomics 2024; 308:105277. [PMID: 39127164 DOI: 10.1016/j.jprot.2024.105277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
Pancreatic adenocarcinoma (PAAD) is a prevalent and highly malignant gastrointestinal tumor. Therefore, exploring the mechanisms of drug resistance and immune pathways in PAAD is crucial for clinical treatment. In this study, a total of 497 differentially expressed genes (DEGs) were identified between normal and PAAD samples, and which were enriched to 117 GO terms and 7 functional pathways. Subsequently, 5 overall survival-related DEGs (ESRP1, KRT6A, H2BC11, H2BC4 and KLK) was generated using Cox hazards regression analysis in TCGA dataset. Furthermore, the weighted gene co-expression network analysis revealed a strong association between ESRP1 and PAAD among 5 survival-related DEGs. Patients were divided into two clusters based on ESRP1 expression levels, and low ESRP1 expression existed stronger immune infiltration and higher expression of immunomodulatory targets than high ESRP1 expression by single-sample gene set enrichment analysis, which indicated that low ESRP1 expression was associated with longer survival compared to high ESRP1 expression. Finally, our study also found that immune cells distribution and immunomodulatory targets gene expression in the GEO dataset were similar to the TCGA cohort. Overall, our findings suggest that ESRP1 may play a role in influencing immune contexture and regulating immune function of PAAD patients by integrating data from various databases. SIGNIFICANCE: Utilizing TCGA and GEO datasets, this study uncovers the significant impact of epithelial splicing regulatory protein 1 (ESRP1) on PAAD. ESRP1 emerges as a key regulator of immune function, influencing tumor microenvironment and immune cell infiltration. Cluster analysis shows that low ESRP1 expression correlates with enhanced immune activity, predicting better prognosis. This discovery suggests that ESRP1 can serve as a potential biomarker for the prognosis of PAAD, offering new insights into personalized immunotherapy by influencing immune regulation and tumor progression.
Collapse
Affiliation(s)
- Pengpeng Wang
- Police-Dog Technology Department, Criminal Investigation Police University of China, Shenyang 110034, China
| | - Xiang Gao
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Weijie Zheng
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Junnan Zhang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
14
|
Jakopec S, Hamzic LF, Bočkor L, Car I, Perić B, Kirin SI, Sedić M, Raić-Malić S. Coumarin-modified ruthenium complexes: Synthesis, characterization, and antiproliferative activity against human cancer cells. Arch Pharm (Weinheim) 2024; 357:e2400271. [PMID: 38864840 DOI: 10.1002/ardp.202400271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/22/2024] [Accepted: 05/26/2024] [Indexed: 06/13/2024]
Abstract
Among ruthenium complexes studied as anticancer metallodrugs, NKP-1339, NAMI-A, RM175, and RAPTA-C have already entered clinical trials due to their potent antitumor activity demonstrated in preclinical studies and reduced toxicity in comparison with platinum drugs. Considering the advantages of ruthenium-based anticancer drugs and the cytostatic activity of organometallic complexes with triazole- and coumarin-derived ligands, we set out to synthesize Ru(II) complexes of coumarin-1,2,3,-triazole hybrids (L) with the general formula [Ru(L)(p-cymene)(Cl)]ClO4. The molecular structure of the complex [Ru(2a)(p-cymene)(Cl)]ClO4 (2aRu) was determined by single-crystal X-ray diffraction, which confirmed the coordination of the ligand to the central ruthenium(II) cation by bidentate mode of coordination. Coordination with Ru(II) resulted in the enhancement of cytostatic activity in HepG2 hepatocellular carcinoma cells and PANC-1 pancreatic cancer cells. Coumarin derivative 2a positively regulated the expression and activity of c-Myc and NPM1 in RKO colon carcinoma cells, while the Ru(II) half-sandwich complex 2cRu induced downregulation of AKT and ERK signaling in PANC-1 cells concomitant with reduced intracellular levels of reactive oxygen species. Altogether, our findings indicated that coumarin-modified half-sandwich Ru(II) complexes held potential as anticancer agents against gastrointestinal malignancies.
Collapse
Affiliation(s)
- Silvio Jakopec
- Department of Organic Chemistry, Faculty of Chemical Engineering and Technology, University of Zagreb, Zagreb, Croatia
| | - Lejla F Hamzic
- Centre for Applied Bioanthropology, Institute for Anthropological Research, Zagreb, Croatia
| | - Luka Bočkor
- Centre for Applied Bioanthropology, Institute for Anthropological Research, Zagreb, Croatia
| | - Iris Car
- Centre for Applied Bioanthropology, Institute for Anthropological Research, Zagreb, Croatia
| | - Berislav Perić
- Division of Materials Chemistry, Ruđer Bošković Institute, Zagreb, Croatia
| | - Srećko I Kirin
- Division of Materials Chemistry, Ruđer Bošković Institute, Zagreb, Croatia
| | - Mirela Sedić
- Centre for Applied Bioanthropology, Institute for Anthropological Research, Zagreb, Croatia
| | - Silvana Raić-Malić
- Department of Organic Chemistry, Faculty of Chemical Engineering and Technology, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
15
|
Brugel M, Callon S, Carlier C, Amroun KL, Botsen D, Kianmanesh R, Perrier M, Piardi T, Renard Y, Rhaiem R, El Balkhi S, Bouché O. Association between pancreatic adenocarcinoma risk and concentration of organochlorine pesticides in adipose tissue and urine: A targeted-screening analysis case-control study (PESTIPAC). United European Gastroenterol J 2024; 12:951-959. [PMID: 39118264 PMCID: PMC11497671 DOI: 10.1002/ueg2.12602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/25/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Knowledge about environmental pancreatic adenocarcinoma (PA) risk factors, including pesticide exposure, remains limited. Organochlorine (OC) accumulates in adipose tissue and can help reflect long-term exposure. PATIENTS AND METHODS Age and body mass index (BMI) of patients with PA were matched with those undergoing a surgery for a benign disease on age and BMI (1:1). Targeted analyses screened 345 pesticides and metabolites, including 29 OC, in adipose tissue and urine samples. The primary aim was to investigate the association between organochlorine concentrations in visceral fat or urine, and PA. Adjusted conditional logistic regressions were carried out accounting for multiple testing. RESULTS Trans-nonachlor (odds ratio [OR] = 1.325, 95% confidence interval [CI] [1.108-1.586]), cis-nonachlor (OR = 15.433, 95% CI [2.733-87.136]), Mirex (OR = 2.853, 95% CI [1.213-6.713]) and 4,4 DDE (OR = 1.019, 95% CI [1.005-1.034]) in fat and a greater number of positive samples (OR = 1.758 95% CI [1.11-2.997]) were significantly associated with higher odds of PA. In contrast, as awaited, urine samples did not yield any statistically significant associations for all tested pesticides. CONCLUSION Some OCs were associated with higher odds of PA. The underlying mechanisms of pancreatic aggression need to be investigated to refine these findings. TRIAL REGISTRATION Clinicaltrials.gov NCT04429490.
Collapse
Affiliation(s)
- Mathias Brugel
- Gastroenterology and Digestive Oncology DepartmentCentre Hospitalier Côte BasqueBayonneFrance
- ULR 2694 ‐ METRICS: Évaluation des Technologies de Santé et des Pratiques MédicalesF‐59000LilleFrance
- Digestive Oncology DepartmentCHU ReimsUniversity of Reims Champagne‐Ardenne (URCA)ReimsFrance
| | - Sidonie Callon
- Digestive Oncology DepartmentCHU ReimsUniversity of Reims Champagne‐Ardenne (URCA)ReimsFrance
| | - Claire Carlier
- Digestive Oncology DepartmentCHU ReimsUniversity of Reims Champagne‐Ardenne (URCA)ReimsFrance
- Department of Medical OncologyGodinot Cancer InstituteReimsFrance
| | - Koceila Lamine Amroun
- GeneralDigestive and Endocrine Surgery DepartmentCHU ReimsUniversity of Reims Champagne‐Ardenne (URCA)ReimsFrance
| | - Damien Botsen
- Digestive Oncology DepartmentCHU ReimsUniversity of Reims Champagne‐Ardenne (URCA)ReimsFrance
- Department of Medical OncologyGodinot Cancer InstituteReimsFrance
| | - Reza Kianmanesh
- GeneralDigestive and Endocrine Surgery DepartmentCHU ReimsUniversity of Reims Champagne‐Ardenne (URCA)ReimsFrance
| | - Marine Perrier
- Gastroenterology and Digestive Oncology DepartmentCentre Hospitalier Côte BasqueBayonneFrance
| | - Tullio Piardi
- GeneralDigestive and Endocrine Surgery DepartmentCHU ReimsUniversity of Reims Champagne‐Ardenne (URCA)ReimsFrance
| | - Yohann Renard
- GeneralDigestive and Endocrine Surgery DepartmentCHU ReimsUniversity of Reims Champagne‐Ardenne (URCA)ReimsFrance
| | - Rami Rhaiem
- GeneralDigestive and Endocrine Surgery DepartmentCHU ReimsUniversity of Reims Champagne‐Ardenne (URCA)ReimsFrance
| | | | - Olivier Bouché
- Digestive Oncology DepartmentCHU ReimsUniversity of Reims Champagne‐Ardenne (URCA)ReimsFrance
| |
Collapse
|
16
|
Di Giorgio A, Ferracci F, Bagalà C, Carbone C, Salvatore L, Strippoli A, Attalla El Halabieh M, Abatini C, Alfieri S, Pacelli F, Tortora G. Combined Nabpaclitaxel pressurized intraPeritoneal aerosol chemotherapy with systemic Nabpaclitaxel-Gemcitabine chemotherapy for pancreatic cancer peritoneal metastases: protocol of single-arm, open-label, phase II trial (Nab-PIPAC trial). Pleura Peritoneum 2024; 9:121-129. [PMID: 39544430 PMCID: PMC11558173 DOI: 10.1515/pp-2024-0010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/24/2024] [Indexed: 11/17/2024] Open
Abstract
Objectives Current therapies show limited efficacy against peritoneal metastases (PM) from pancreatic cancer. Pressurized intra-peritoneal aerosol chemotherapy (PIPAC) has emerged as a novel intraperitoneal drug delivery method. Recently, a dose-escalation study identified the safe dose of Nabpaclitaxel for PIPAC administration, an ideal intraperitoneal chemotherapy agent against pancreatic cancer. Combining systemic NabPaclitaxel-Gemcitabine with NabPaclitaxel-PIPAC may enhance disease control in pancreatic cancer patients with PM. Methods The Nab-PIPAC trial is a single-center, prospective, open-label, phase II study (ClinicalTrials.gov identifier: NCT05371223). Its primary goal is to evaluate the antitumor activity of the combined treatment based on Disease Control Rate (DCR) using RECISTv.1.1 criteria. Secondary objectives include feasibility, safety, pathological response, progression-free and overall survival, nutritional status, quality of life, pharmacokinetics of NabPaclitaxel-PIPAC, and PM molecular evolution via translational research. The treatment protocol consists of three courses, each with two cycles of intravenous NabPaclitaxel-Gemcitabine and one cycle of NabPaclitaxel-PIPAC, with standard metastatic pancreatic cancer doses for the former and 112.5 mg/m2 for the latter. Sample size follows Simon's two-stage design: 12 patients in stage one and 26 in stage two (80 % power, 0.1 alpha). Results Partial results will be available after first stage enrollment. Conclusions This trial aims to determine the antitumor efficacy and safety of combining NabPaclitaxel-PIPAC with systemic NabPaclitaxel-Gemcitabine in pancreatic cancer patients with PM.
Collapse
Affiliation(s)
- Andrea Di Giorgio
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Federica Ferracci
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Cinzia Bagalà
- Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | - Carmine Carbone
- Department of Medical and Surgical Science, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Lisa Salvatore
- Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | - Antonia Strippoli
- Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | - Miriam Attalla El Halabieh
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Carlo Abatini
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Sergio Alfieri
- Surgical Unit of Digestive Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Catholic University of the Sacred Heart, Rome, Italy
| | - Fabio Pacelli
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Catholic University of the Sacred Heart, Rome, Italy
| | - Giampaolo Tortora
- Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
17
|
Wittram R, König HH, Brettschneider C. Economic evaluations of pancreatic cancer screening: a systematic review protocol. BMJ Open 2024; 14:e087003. [PMID: 39153777 PMCID: PMC11331839 DOI: 10.1136/bmjopen-2024-087003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 07/22/2024] [Indexed: 08/19/2024] Open
Abstract
INTRODUCTION The early detection of pancreatic cancer is an important step in reducing mortality by offering patients curative treatment. Screening strategies in risk populations and by means of different detection methods have been economically evaluated. However, a synthesis of screening studies to inform resource allocation towards early detection within the disease area has not been done. Therefore, studies evaluating the cost-effectiveness and costs of screening for pancreatic cancer should be systematically reviewed. METHODS AND ANALYSIS A systematic review of economic evaluations reporting the cost-effectiveness or costs of pancreatic cancer screening will be conducted. The electronic databases Medline, Web of Science and EconLit will be searched without geographical or time restrictions. Two independent reviewers will select eligible studies based on predefined criteria. The study quality will be assessed using the Consolidated Health Economic Evaluation Reporting Standards statement and the Bias in Economic Evaluation checklist. One reviewer will extract relevant data and a second reviewer will cross-check compliance with the extraction sheet. Key items will include characteristics of screened individuals, the screening strategies used, and costs, health effects and cost-effectiveness as study outputs. Differences of opinion between the reviewers will be solved by consulting a third reviewer. ETHICS AND DISSEMINATION Ethics approval is not required for this study since no original data will be collected. The results will be disseminated through presentations at conferences and publication in a peer-reviewed journal. The results of the systematic review will inform future economic evaluations of pancreatic screening, which provide guidance for decision-making in healthcare resource prioritisation. PROSPERO REGISTRATION NUMBER CRD42023475348.
Collapse
Affiliation(s)
- Robert Wittram
- Department of Health Economics and Health Services Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Helmut König
- Department of Health Economics and Health Services Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Brettschneider
- Department of Health Economics and Health Services Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
18
|
Hatoum H, Rosemurgy A, Bastidas JA, Zervos E, Muscarella P, Edil BH, Cynamon J, Johnson DT, Thomas C, Swinson BM, Nordgren A, Vitulli P, Nutting C, Gipson M, Tsobanoudis A, Agah R. Treatment of locally advanced pancreatic cancer using localized trans-arterial micro perfusion of gemcitabine: combined analysis of RR1 and RR2. Oncologist 2024; 29:690-698. [PMID: 39049803 PMCID: PMC11299923 DOI: 10.1093/oncolo/oyae178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 06/18/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Locally advanced pancreatic cancer (LAPC) comprises 40% of pancreatic cancer diagnoses and has a relatively poor prognosis. Trans-arterial micro perfusion (TAMP)-mediated chemotherapy delivery to the primary tumor is a novel approach worthy of investigation. The RR1 (dose escalation) and RR2 (observational) studies examined the safety and preliminary efficacy of TAMP-delivered gemcitabine for LAPC. PATIENTS AND METHODS RR1 and RR2 data were pooled. Both studies enrolled patients with LAPC with histologically confirmed adenocarcinoma. Participant data, including age, sex, race, stage, previous treatments, toxicity, disease progression, and death, were collected. Median number of cycles and average treatment dosage were calculated. Overall survival (OS) was determined for the whole group and separately for patients who received and did not receive previous treatments. Aims of the analysis were to assess procedure safety, OS, and evaluate factors associated with OS. RESULTS The median age of the 43 patients enrolled in RR1 and RR2 was 72 years (range, 51-88 years). Median OS for the 35 eligible patients with stage III disease was 12.6 months (95% CI, 2.1-54.2 months). Previous chemoradiation was associated with significantly longer OS [27.1 months (95% CI, 8.4-40.6 months)] compared to previous systemic chemotherapy [14.6 months (95% CI, 6.4-54.2 months)] or no prior treatment [7.0 months (95% CI, 2.1-35.4 months)] (P < .001). The most common adverse events were GI related (abdominal pain, emesis, and vomiting); the most common grade 3 toxicity was sepsis. CONCLUSION Study results indicate that TAMP-mediated gemcitabine delivery in patients with LAPC is potentially safe, feasible, and provides potential clinical benefits. CLINICAL TRIAL REGISTRATION NCT02237157 (RR1) and NCT02591082 (RR2).
Collapse
Affiliation(s)
- Hassan Hatoum
- Department of Internal Medicine Hematology-Oncology Section, University of Oklahoma College of Medicine, Oklahoma City, OK, United States
| | | | | | - Emmanuel Zervos
- Division of Surgical Oncology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Peter Muscarella
- Montefiore Medical Center, Bronx, NY, United States
- Niagara Falls Memorial Medical Center, Niagara Falls, NY, United States
| | - Barish H Edil
- Department of Surgery, University of Oklahoma College of Medicine, Oklahoma City, OK, United States
| | | | - D Thor Johnson
- University of Colorado Hospital, Aurora, CO, United States
- Sarah Cannon, Nashville, TN, United States
| | | | | | - Aaron Nordgren
- Fawcett Memorial Hospital, Port Charlotte, FL, United States
| | - Paul Vitulli
- Florida Hospital, Tampa, Tampa, FL, United States
- Duval Vascular Center, Jacksonville, FL, United States
| | - Charles Nutting
- Endovascular Consultants of Colorado, Lone Tree, CO, United States
| | | | | | - Ramtin Agah
- RenovoRx, Inc., Los Altos, CA, United States
| |
Collapse
|
19
|
Tsukagoshi M, Araki K, Shirabe K. Pancreatic cancer and sarcopenia: a narrative review of the current status. Int J Clin Oncol 2024; 29:1055-1066. [PMID: 38954075 DOI: 10.1007/s10147-024-02576-2] [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: 05/25/2024] [Accepted: 06/19/2024] [Indexed: 07/04/2024]
Abstract
Pancreatic cancer is still a difficult disease to treat, despite recent advances in surgical techniques and chemotherapeutic drugs. Its incidence continues to rise, as does the number of older patients. Sarcopenia is defined as a progressive and generalized loss of skeletal muscle mass and strength. Sarcopenia is present in approximately 40% in patients with pancreatic cancer. Sarcopenia is primarily diagnosed through imaging, and progress is being made in the development of automated methods and artificial intelligence, as well as biomarker research. Sarcopenia has been linked to a poor prognosis in pancreatic cancer patients. However, some studies suggest that sarcopenia is not always associated with a poor prognosis, depending on the resectability of pancreatic cancer and the nature of treatment, such as surgery or chemotherapy. Recent meta-analyses have found that sarcopenia is not linked to postoperative complications. It is still debated whether there is a link between sarcopenia and drug toxicity during chemotherapy. The relationship between sarcopenia and immunity has been investigated, but the mechanism is still unknown.
Collapse
Affiliation(s)
- Mariko Tsukagoshi
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgical Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-Machi, Maebashi Gunma, 371-8511, Japan
| | - Kenichiro Araki
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgical Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-Machi, Maebashi Gunma, 371-8511, Japan
| | - Ken Shirabe
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgical Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-Machi, Maebashi Gunma, 371-8511, Japan.
| |
Collapse
|
20
|
Cristina-Marianini-Rios, Sanchez MEC, de Paredes AGG, Rodríguez M, Barreto E, López JV, Fuentes R, Beltrán MM, Sanjuanbenito A, Lobo E, Caminoa A, Ruz-Caracuel I, Durán SL, Olcina JRF, Blázquez J, Sequeros EV, Carrato A, Ávila JCM, Earl J. The best linear unbiased prediction (BLUP) method as a tool to estimate the lifetime risk of pancreatic ductal adenocarcinoma in high-risk individuals with no known pathogenic germline variants. Fam Cancer 2024; 23:233-246. [PMID: 38780705 PMCID: PMC11254992 DOI: 10.1007/s10689-024-00397-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/28/2024] [Indexed: 05/25/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer-related death in the Western world. The number of diagnosed cases and the mortality rate are almost equal as the majority of patients present with advanced disease at diagnosis. Between 4 and 10% of pancreatic cancer cases have an apparent hereditary background, known as hereditary pancreatic cancer (HPC) and familial pancreatic cancer (FPC), when the genetic basis is unknown. Surveillance of high-risk individuals (HRI) from these families by imaging aims to detect PDAC at an early stage to improve prognosis. However, the genetic basis is unknown in the majority of HRIs, with only around 10-13% of families carrying known pathogenic germline mutations. The aim of this study was to assess an individual's genetic cancer risk based on sex and personal and family history of cancer. The Best Linear Unbiased Prediction (BLUP) methodology was used to estimate an individual's predicted risk of developing cancer during their lifetime. The model uses different demographic factors in order to estimate heritability. A reliable estimation of heritability for pancreatic cancer of 0.27 on the liability scale, and 0.07 at the observed data scale as obtained, which is different from zero, indicating a polygenic inheritance pattern of PDAC. BLUP was able to correctly discriminate PDAC cases from healthy individuals and those with other cancer types. Thus, providing an additional tool to assess PDAC risk HRI with an assumed genetic predisposition in the absence of known pathogenic germline mutations.
Collapse
Affiliation(s)
- Cristina-Marianini-Rios
- Department of Agricultural Economics, Statistics and Business Management, Universidad Politécnica de Madrid, Madrid, Spain
| | - María E Castillo Sanchez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
| | - Ana García García de Paredes
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercedes Rodríguez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, 28034, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
- University of Alcalá, Madrid, Spain
| | - Emma Barreto
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
- University of Alcalá, Madrid, Spain
| | - Jorge Villalón López
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
| | - Raquel Fuentes
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, 28034, Spain
| | | | - Alfonso Sanjuanbenito
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
- Pancreatic and Biliopancreatic Surgery Unit, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Eduardo Lobo
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- Pancreatic and Biliopancreatic Surgery Unit, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Alejandra Caminoa
- Department of Pathology, Hospital Universitario Ramón y Cajal, Madrid, 28034, Spain
| | - Ignacio Ruz-Caracuel
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
- Department of Pathology, Hospital Universitario Ramón y Cajal, Madrid, 28034, Spain
| | - Sergio López Durán
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - José Ramón Foruny Olcina
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Javier Blázquez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- Radiology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Enrique Vázquez Sequeros
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
| | - Alfredo Carrato
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
- University of Alcalá, Madrid, Spain
- Pancreatic Cancer Europe, Brussels, Belgium
| | - Jose Carlos Martínez Ávila
- Department of Agricultural Economics, Statistics and Business Management, Universidad Politécnica de Madrid, Madrid, Spain.
| | - Julie Earl
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain.
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain.
| |
Collapse
|
21
|
Earl J, Fuentes R, Sanchez MEC, de Paredes AGG, Muñoz M, Sanjuanbenito A, Lobo E, Caminoa A, Rodríguez M, Barreto E, López JV, Ruz-Caracuel I, Durán SL, Olcina JRF, Sánchez BL, Páez SC, Torres A, Blázquez J, Sequeros EV, Carrato A. The Spanish Familial Pancreatic Cancer Registry (PANGENFAM): a decade follow-up of individuals at high-risk for pancreatic cancer. Fam Cancer 2024; 23:383-392. [PMID: 38753287 PMCID: PMC11254983 DOI: 10.1007/s10689-024-00388-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 04/04/2024] [Indexed: 07/18/2024]
Abstract
The Spanish Familial Pancreatic Cancer Registry (PANGENFAM) was established in 2009 and aims to characterize the genotype and phenotype of familial pancreatic cancer (FPC). Furthermore, an early detection screening program for pancreatic ductal adenocarcinoma (PDAC) is provided to healthy high-risk individuals from FPC and hereditary pancreatic cancer families (first-degree relatives). This article describes our experience over the last 10 years in high-risk screening. Hereditary and familial pancreatic cancer families were identified through the oncology and gastroenterology units. High-risk individuals underwent annual screening with endoscopic ultrasound (EUS) and magnetic resonance (MRI) from age 40 or 10 years younger than the youngest affected family member. Results: PANGENFAM has enrolled 290 individuals from 143 families, including 52 PDAC cases and 238 high-risk individuals. All high-risk individuals eligible for screening were offered to enter the surveillance program, with 143 currently participating. Pancreatic abnormalities were detected in 94 individuals (median age 53 years (29-83), with common findings including cystic lesions and inhomogeneous parenchyma. Imaging test concordance was 66%. Surgical intervention was performed in 4 high-risk individuals following highly suspicious lesions detected by imaging. PANGENFAM is a valuable resource for science innovation, such as biobanking, with clinical and imaging data available for analysis. For high-risk families, it may offer a potential for early diagnosis. Collaboration with other national and international registries is needed to increase our understanding of the disease biology and to standardize criteria for inclusion and follow-up, optimizing cost-effectiveness and efficacy.
Collapse
Affiliation(s)
- Julie Earl
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain.
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain.
- Biobank and Biomodels Platform, Spanish National Biobanks Network (ISCIII Biobank Register No. B.0000678), ISCIII Research and Development Platforms in Biomedicine and Health Sciences, BioBank Hospital Ramón y Cajal-IRYCIS, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, PT20/004528034, Madrid, Spain.
| | - Raquel Fuentes
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain
| | - María E Castillo Sanchez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
| | - Ana García García de Paredes
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - María Muñoz
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Radiology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Alfonso Sanjuanbenito
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Pancreatic and Biliopancreatic Surgery Unit, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Eduardo Lobo
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Pancreatic and Biliopancreatic Surgery Unit, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Alejandra Caminoa
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Department of Pathology, Hospital Universitario Ramón y Cajal, 28034, Madrid, Spain
| | - Mercedes Rodríguez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain
| | - Emma Barreto
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
- University of Alcalá, Madrid, Spain
| | - Jorge Villalón López
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Biobank and Biomodels Platform, Spanish National Biobanks Network (ISCIII Biobank Register No. B.0000678), ISCIII Research and Development Platforms in Biomedicine and Health Sciences, BioBank Hospital Ramón y Cajal-IRYCIS, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, PT20/004528034, Madrid, Spain
| | - Ignacio Ruz-Caracuel
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
- Department of Pathology, Hospital Universitario Ramón y Cajal, 28034, Madrid, Spain
| | - Sergio López Durán
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - José Ramón Foruny Olcina
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Bárbara Luna Sánchez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Biobank and Biomodels Platform, Spanish National Biobanks Network (ISCIII Biobank Register No. B.0000678), ISCIII Research and Development Platforms in Biomedicine and Health Sciences, BioBank Hospital Ramón y Cajal-IRYCIS, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, PT20/004528034, Madrid, Spain
| | - Sonia Camaño Páez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Biobank and Biomodels Platform, Spanish National Biobanks Network (ISCIII Biobank Register No. B.0000678), ISCIII Research and Development Platforms in Biomedicine and Health Sciences, BioBank Hospital Ramón y Cajal-IRYCIS, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, PT20/004528034, Madrid, Spain
| | - Ana Torres
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Biobank and Biomodels Platform, Spanish National Biobanks Network (ISCIII Biobank Register No. B.0000678), ISCIII Research and Development Platforms in Biomedicine and Health Sciences, BioBank Hospital Ramón y Cajal-IRYCIS, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, PT20/004528034, Madrid, Spain
| | - Javier Blázquez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Enrique Vázquez Sequeros
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain
| | - Alfredo Carrato
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
- University of Alcalá, Madrid, Spain
- Pancreatic Cancer Europe, Brussels, Belgium
| |
Collapse
|
22
|
Ney A, Nené NR, Sedlak E, Acedo P, Blyuss O, Whitwell HJ, Costello E, Gentry-Maharaj A, Williams NR, Menon U, Fusai GK, Zaikin A, Pereira SP. Identification of a serum proteomic biomarker panel using diagnosis specific ensemble learning and symptoms for early pancreatic cancer detection. PLoS Comput Biol 2024; 20:e1012408. [PMID: 39208354 PMCID: PMC11389906 DOI: 10.1371/journal.pcbi.1012408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 09/11/2024] [Accepted: 08/11/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The grim (<10% 5-year) survival rates for pancreatic ductal adenocarcinoma (PDAC) are attributed to its complex intrinsic biology and most often late-stage detection. The overlap of symptoms with benign gastrointestinal conditions in early stage further complicates timely detection. The suboptimal diagnostic performance of carbohydrate antigen (CA) 19-9 and elevation in benign hyperbilirubinaemia undermine its reliability, leaving a notable absence of accurate diagnostic biomarkers. Using a selected patient cohort with benign pancreatic and biliary tract conditions we aimed to develop a data analysis protocol leading to a biomarker signature capable of distinguishing patients with non-specific yet concerning clinical presentations, from those with PDAC. METHODS 539 patient serum samples collected under the Accelerated Diagnosis of neuro Endocrine and Pancreatic TumourS (ADEPTS) study (benign disease controls and PDACs) and the UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS, healthy controls) were screened using the Olink Oncology II panel, supplemented with five in-house markers. 16 specialized base-learner classifiers were stacked to select and enhance biomarker performances and robustness in blinded samples. Each base-learner was constructed through cross-validation and recursive feature elimination in a discovery set comprising approximately two thirds of the ADEPTS and UKCTOCS samples and contrasted specific diagnosis with PDAC. RESULTS The signature which was developed using diagnosis-specific ensemble learning demonstrated predictive capabilities outperforming CA19-9, the only biomarker currently accepted by the FDA and the National Comprehensive Cancer Network guidelines for pancreatic cancer, and other individual biomarkers and combinations in both discovery and held-out validation sets. An AUC of 0.98 (95% CI 0.98-0.99) and sensitivity of 0.99 (95% CI 0.98-1) at 90% specificity was achieved with the ensemble method, which was significantly larger than the AUC of 0.79 (95% CI 0.66-0.91) and sensitivity 0.67 (95% CI 0.50-0.83), also at 90% specificity, for CA19-9, in the discovery set (p = 0.0016 and p = 0.00050, respectively). During ensemble signature validation in the held-out set, an AUC of 0.95 (95% CI 0.91-0.99), sensitivity 0.86 (95% CI 0.68-1), was attained compared to an AUC of 0.80 (95% CI 0.66-0.93), sensitivity 0.65 (95% CI 0.48-0.56) at 90% specificity for CA19-9 alone (p = 0.0082 and p = 0.024, respectively). When validated only on the benign disease controls and PDACs collected from ADEPTS, the diagnostic-specific signature achieved an AUC of 0.96 (95% CI 0.92-0.99), sensitivity 0.82 (95% CI 0.64-0.95) at 90% specificity, which was still significantly higher than the performance for CA19-9 taken as a single predictor, AUC of 0.79 (95% CI 0.64-0.93) and sensitivity of 0.18 (95% CI 0.03-0.69) (p = 0.013 and p = 0.0055, respectively). CONCLUSION Our ensemble modelling technique outperformed CA19-9, individual biomarkers and indices developed with prevailing algorithms in distinguishing patients with non-specific but concerning symptoms from those with PDAC, with implications for improving its early detection in individuals at risk.
Collapse
Affiliation(s)
- Alexander Ney
- Institute for Liver and Digestive Health, University College London, London, United Kingdom
| | - Nuno R Nené
- Department of Women's Cancer, EGA Institute for Women's Health, University College London, London, United Kingdom
- Cancer Institute, University College London, London, United Kingdom
- Department of Statistical Science, University College London, London, United Kingdom
| | - Eva Sedlak
- Department of Women's Cancer, EGA Institute for Women's Health, University College London, London, United Kingdom
| | - Pilar Acedo
- Institute for Liver and Digestive Health, University College London, London, United Kingdom
| | - Oleg Blyuss
- Center for Cancer Prevention, Detection and Early Diagnosis, Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
- Department of Pediatrics and Pediatric Infectious Diseases, Institute of Child´s Health, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Harry J Whitwell
- Department of Women's Cancer, EGA Institute for Women's Health, University College London, London, United Kingdom
- National Phenome Centre and Imperial Clinical Phenotyping Centre, Department of Metabolism, Digestion and Reproduction, IRDB, Building Imperial College London, London, United Kingdom
- Section of Bioanalytical Chemistry, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Sir Alexander Fleming Building, Imperial College London, London, United Kingdom
| | - Eithne Costello
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Aleksandra Gentry-Maharaj
- Department of Women's Cancer, EGA Institute for Women's Health, University College London, London, United Kingdom
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, United Kingdom
| | - Norman R Williams
- Division of Surgery & Interventional Science, University College London, London, United Kingdom
| | - Usha Menon
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, United Kingdom
| | - Giuseppe K Fusai
- HPB & Liver Transplant Unit, Royal Free London, London, United Kingdom
| | - Alexey Zaikin
- Department of Women's Cancer, EGA Institute for Women's Health, University College London, London, United Kingdom
- Institute for Cognitive Neuroscience, University Higher School of Economics, Moscow, Russia
- Department of Mathematics, University College London, London, United Kingdom
- Centre for Cognition and Decision making, Institute for Cognitive Neuroscience, HSE University, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Skolkovo, Moscow, Russia
| | - Stephen P Pereira
- Institute for Liver and Digestive Health, University College London, London, United Kingdom
| |
Collapse
|
23
|
Gehrels AM, Wagner AD, Besselink MG, Verhoeven RHA, van Eijck CHJ, van Laarhoven HWM, Wilmink JW, van der Geest LG. Gender differences in tumor characteristics, treatment allocation and survival in stage I-III pancreatic cancer: a nationwide study. Eur J Cancer 2024; 206:114117. [PMID: 38781719 DOI: 10.1016/j.ejca.2024.114117] [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: 02/12/2024] [Revised: 04/25/2024] [Accepted: 05/04/2024] [Indexed: 05/25/2024]
Abstract
INTRODUCTION Sex and gender are modulators of health and disease and may have impact on treatment allocation and survival in patients with cancer. In this study, we analyzed the impact of sex and gender on treatment allocation and overall survival in patients with stage I-III pancreatic cancer. METHODS Patients with stage I-III pancreatic cancer diagnosed between 2015 and 2020 were selected from the nationwide Netherlands Cancer Registry. Associations between sex and gender and the probability of receiving surgical and/or systemic treatment were examined with multivariable logistic regression analyses. Overall survival was assessed with log rank test and multivariable Cox proportional hazard analysis. RESULTS Among 6855 patients, 51.2 % were female. Multivariable logistic regression analyses with adjustment for known confounders (age, performance status, comorbidities, tumor location, tumor stage and previous malignancies) showed that females less often received systemic chemotherapy compared to males (OR 0.799, 95 %CI 0.703-0.909, p < .001). No difference was found in the probability for undergoing surgical resection. Furthermore, females had worse overall survival compared to males (median OS 8.5 and 9.2 months respectively, 95 %CI 8.669-9.731). CONCLUSION This nationwide study found that female patients with stage I-III pancreatic cancer significantly less often received systemic treatment and had worse overall survival as compared to males. Disparities in pancreatic cancer care can be decreased by recognizing and resolving potential obstacles or biases in treatment decision-making.
Collapse
Affiliation(s)
- A M Gehrels
- Amsterdam UMC, location University of Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands.
| | - A D Wagner
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - M G Besselink
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands; Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, the Netherlands
| | - R H A Verhoeven
- Amsterdam UMC, location University of Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands; Department of Research, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, Netherlands
| | - C H J van Eijck
- Department of Surgery, Erasmus MC Cancer Institute, Erasmus University Rotterdam, Rotterdam, the Netherlands
| | - H W M van Laarhoven
- Amsterdam UMC, location University of Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands
| | - J W Wilmink
- Amsterdam UMC, location University of Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands
| | - L G van der Geest
- Amsterdam UMC, location University of Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands; Department of Research, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, Netherlands.
| |
Collapse
|
24
|
Ang A, Michaelides A, Chelala C, Ullah D, Kocher HM, Barts Pancreas Tissue Bank. Prognostication for recurrence patterns after curative resection for pancreatic ductal adenocarcinoma. Ann Hepatobiliary Pancreat Surg 2024; 28:248-261. [PMID: 38556877 PMCID: PMC11128784 DOI: 10.14701/ahbps.23-149] [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: 11/15/2023] [Revised: 02/02/2024] [Accepted: 02/10/2024] [Indexed: 04/02/2024] Open
Abstract
Backgrounds/Aims This study aimed to investigate patterns and factors affecting recurrence after curative resection for pancreatic ductal adenocarcinoma (PDAC). Methods Consecutive patients who underwent curative resection for PDAC (2011-21) and consented to data and tissue collection (Barts Pancreas Tissue Bank) were followed up until May 2023. Clinico-pathological variables were analysed using Cox proportional hazards model. Results Of 91 people (42 males [46%]; median age, 71 years [range, 43-86 years]) with a median follow-up of 51 months (95% confidence intervals [CIs], 40-61 months), the recurrence rate was 72.5% (n = 66; 12 loco-regional alone, 11 liver alone, 5 lung alone, 3 peritoneal alone, 29 simultaneous loco-regional and distant metastases, and 6 multi-focal distant metastases at first recurrence diagnosis). The median time to recurrence was 8.5 months (95% CI, 6.6-10.5 months). Median survival after recurrence was 5.8 months (95% CI, 4.2-7.3 months). Stratification by recurrence location revealed significant differences in time to recurrence between loco-regional only recurrence (median, 13.6 months; 95% CI, 11.7-15.5 months) and simultaneous loco-regional with distant recurrence (median, 7.5 months; 95% CI, 4.6-10.4 months; p = 0.02, pairwise log-rank test). Significant predictors for recurrence were systemic inflammation index (SII) ≥ 500 (hazard ratio [HR], 4.5; 95% CI, 1.4-14.3), lymph node ratio ≥ 0.33 (HR, 2.8; 95% CI, 1.4-5.8), and adjuvant chemotherapy (HR, 0.4; 95% CI, 0.2-0.7). Conclusions Timing to loco-regional only recurrence was significantly longer than simultaneous loco-regional with distant recurrence. Significant predictors for recurrence were SII, lymph node ration, and adjuvant chemotherapy.
Collapse
Affiliation(s)
- Andrew Ang
- The Royal London Hospital, Barts Health NHS Trust, London, UK
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, UK
| | - Athena Michaelides
- The Royal London Hospital, Barts Health NHS Trust, London, UK
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, UK
| | - Claude Chelala
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, UK
| | - Dayem Ullah
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, UK
| | - Hemant M. Kocher
- The Royal London Hospital, Barts Health NHS Trust, London, UK
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, UK
| | | |
Collapse
|
25
|
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, Jemal A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2024; 74:229-263. [PMID: 38572751 DOI: 10.3322/caac.21834] [Citation(s) in RCA: 6017] [Impact Index Per Article: 6017.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 02/12/2024] [Indexed: 04/05/2024] Open
Abstract
This article presents global cancer statistics by world region for the year 2022 based on updated estimates from the International Agency for Research on Cancer (IARC). There were close to 20 million new cases of cancer in the year 2022 (including nonmelanoma skin cancers [NMSCs]) alongside 9.7 million deaths from cancer (including NMSC). The estimates suggest that approximately one in five men or women develop cancer in a lifetime, whereas around one in nine men and one in 12 women die from it. Lung cancer was the most frequently diagnosed cancer in 2022, responsible for almost 2.5 million new cases, or one in eight cancers worldwide (12.4% of all cancers globally), followed by cancers of the female breast (11.6%), colorectum (9.6%), prostate (7.3%), and stomach (4.9%). Lung cancer was also the leading cause of cancer death, with an estimated 1.8 million deaths (18.7%), followed by colorectal (9.3%), liver (7.8%), female breast (6.9%), and stomach (6.8%) cancers. Breast cancer and lung cancer were the most frequent cancers in women and men, respectively (both cases and deaths). Incidence rates (including NMSC) varied from four-fold to five-fold across world regions, from over 500 in Australia/New Zealand (507.9 per 100,000) to under 100 in Western Africa (97.1 per 100,000) among men, and from over 400 in Australia/New Zealand (410.5 per 100,000) to close to 100 in South-Central Asia (103.3 per 100,000) among women. The authors examine the geographic variability across 20 world regions for the 10 leading cancer types, discussing recent trends, the underlying determinants, and the prospects for global cancer prevention and control. With demographics-based predictions indicating that the number of new cases of cancer will reach 35 million by 2050, investments in prevention, including the targeting of key risk factors for cancer (including smoking, overweight and obesity, and infection), could avert millions of future cancer diagnoses and save many lives worldwide, bringing huge economic as well as societal dividends to countries over the forthcoming decades.
Collapse
Affiliation(s)
- Freddie Bray
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | - Mathieu Laversanne
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | - Hyuna Sung
- Surveillance and Health Equity Science, American Cancer Society, Atlanta, Georgia, USA
| | - Jacques Ferlay
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | - Rebecca L Siegel
- Surveillance and Health Equity Science, American Cancer Society, Atlanta, Georgia, USA
| | | | - Ahmedin Jemal
- Surveillance and Health Equity Science, American Cancer Society, Atlanta, Georgia, USA
| |
Collapse
|
26
|
Lisotti A, Cominardi A, Conti Bellocchi MC, Crinò SF, Larghi A, Facciorusso A, Arcidiacono PG, De Angelis C, Di Matteo FM, Fabbri C, Bertani H, Togliani T, Rizzatti G, Brancaccio M, Grillo A, Fantin A, Pezzoli A, D'Errico F, Amato A, Antonini F, Montale A, Pisani A, Forti E, Manno M, Carrara S, Petrone MC, Binda C, Zagari RM, Fusaroli P. Repeated endoscopic ultrasound-guided fine-needle biopsy of solid pancreatic lesions after previous nondiagnostic or inconclusive sampling. Dig Endosc 2024; 36:615-624. [PMID: 37712906 DOI: 10.1111/den.14686] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/12/2023] [Indexed: 09/16/2023]
Abstract
OBJECTIVES Repeated endoscopic ultrasound (EUS)-guided tissue acquisition represents the standard practice for solid pancreatic lesions after previous nondiagnostic or inconclusive results. Since data are lacking, we aimed to evaluate the diagnostic performance of repeated EUS fine-needle biopsy (rEUS-FNB) in this setting. The primary outcome was diagnostic accuracy; sample adequacy, sensitivity, specificity, and safety were secondary outcomes. METHODS Consecutive patients undergoing rEUS-FNB for solid pancreatic lesions at 23 Italian centers from 2019 to 2021 were retrieved. Pathology on the surgical specimen, malignant histology together with ≥6-month follow-up, and benign pathology together with ≥12-month follow-up were adopted as gold standards. RESULTS Among 462 patients, 56.5% were male, with a median age of 68 (59-75) years, malignancy prevalence 77.0%. Tumor size was 26 (20-35) mm. Second-generation FNB needles were used in 89.6% cases. Diagnostic accuracy, sensitivity, and specificity of rEUS-FNB were 89.2%, 91.4%, and 81.7%, respectively (19 false-negative and 12 false-positive results). On multivariate analysis, rEUS-FNB performed at high-volume centers (odds ratio [OR] 2.12; 95% confidence interval [CI] 1.10-3.17; P = 0.03) and tumor size (OR 1.03; 95% CI 1.00-1.06; P = 0.05) were independently related to diagnostic accuracy. Sample adequacy was 94.2%. Use of second-generation FNB needles (OR 5.42; 95% CI 2.30-12.77; P < 0.001) and tumor size >23 mm (OR 3.04; 95% CI 1.31-7.06; P = 0.009) were independently related to sample adequacy. CONCLUSION Repeated EUS-FNB allowed optimal diagnostic performance after nondiagnostic or inconclusive results. Patients' referral to high-volume centers improved diagnostic accuracy. The use of second-generation FNB needles significantly improved sample adequacy over standard EUS-FNB needles.
Collapse
Affiliation(s)
- Andrea Lisotti
- Gastroenterology Unit, Hospital of Imola, University of Bologna, Imola, Italy
| | - Anna Cominardi
- Gastroenterology Unit, Hospital of Imola, University of Bologna, Imola, Italy
- Gastroenterology Unit, Hospital of Piacenza, Piacenza, Italy
| | | | | | - Alberto Larghi
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Antonio Facciorusso
- Gastroenterology Unit, Department of Surgical and Medical Sciences, University of Foggia, Foggia, Italy
| | | | - Claudio De Angelis
- Department of General and Specialist Medicine, Gastroenterologia-U, Città della Salute e della Scienza di Torino, Turin, Italy
| | | | - Carlo Fabbri
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, AUSL Romagna, Forli, Italy
| | - Helga Bertani
- Gastroenterology and Digestive Endoscopy Unit, Azienda Ospedaliero Universitaria di Modena, Modena, Italy
| | - Thomas Togliani
- Gastroenterology and Digestive Endoscopy Unit, University Hospital Borgo Trento, Verona, Italy
| | - Gianenrico Rizzatti
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Mario Brancaccio
- Unit of Gastroenterology, Santa Maria delle Croci Hospital, AUSL Romagna, Ravenna, Italy
| | - Antonino Grillo
- Gastroenterology and Digestive Endoscopy Unit, Rimini "Infermi" Hospital, AUSL Romagna, Rimini, Italy
| | - Alberto Fantin
- Gastroenterology Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Alessandro Pezzoli
- Department of Gastroenterology and GI Endoscopy, University Hospital, Ferrara, Italy
| | - Francesca D'Errico
- Gastroenterology and Endoscopy Unit, Ente Ecclesiastico F. Miulli, Acquaviva delle Fonti, Bari, Italy
| | - Arnaldo Amato
- Division of Digestive Endoscopy and Gastroenterology, Valduce Hospital, Como, Italy
| | - Filippo Antonini
- Gastroenterology and Interventional Endoscopy Unit, "C. e G. Mazzoni" Hospital, Ascoli Piceno, Italy
| | - Amedeo Montale
- Division of Gastroenterology, E.O. Galliera Hospital, Genoa, Italy
| | - Antonio Pisani
- National Institute of Gastroenterology IRCCS Saverio de Bellis, Castellana Grotte, Bari, Italy
| | - Edoardo Forti
- Digestive Endoscopy Unit, ASST Niguarda, Milan, Italy
| | - Mauro Manno
- Gastroenterology and Digestive Endoscopy Unit, Azienda USL Modena, Modena, Italy
| | - Silvia Carrara
- Endoscopic Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Maria Chiara Petrone
- IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Cecilia Binda
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, AUSL Romagna, Forli, Italy
| | - Rocco Maurizio Zagari
- SSD "Patologie organiche esofago-gastriche", IRCCS Azienda Ospedaliero-Universitaria di Bologna, S. Orsola Hospital, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC, University of Bologna, Bologna, Italy
| | - Pietro Fusaroli
- Gastroenterology Unit, Hospital of Imola, University of Bologna, Imola, Italy
| |
Collapse
|
27
|
Scherübl H. [Early detection of sporadic pancreatic cancer]. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2024; 62:412-419. [PMID: 37827502 DOI: 10.1055/a-2114-9847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
The incidence of pancreatic cancer is rising. At present, pancreatic cancer is the third most common cancer-causing death in Germany, but it is expected to become the second in 2030 and finally the leading cause of cancer death in 2050. Pancreatic ductal adenocarcinoma (PC) is generally diagnosed at advanced stages, and 5-year-survival has remained poor. Early detection of sporadic PC at stage IA, however, can yield a 5-year-survival rate of about 80%. Early detection initiatives aim at identifying persons at high risk. People with new-onset diabetes at age 50 or older have attracted much interest. Novel strategies regarding how to detect sporadic PC at an early stage are being discussed.
Collapse
Affiliation(s)
- Hans Scherübl
- Klinik für Innere Medizin; Gastroenterol., GI Onkol. u. Infektiol., Vivantes Klinikum Am Urban, Berlin, Germany
- Akademisches Lehrkrankenhaus der Charité, Berlin, Germany
| |
Collapse
|
28
|
Gueiderikh A, Tarabay A, Abdelouahab M, Smolenschi C, Tanguy ML, Valery M, Malka D, Pudlarz T, Fuerea A, Boige V, Hollebecque A, Ducreux M, Boilève A. Pancreatic adenocarcinoma third line systemic treatments: a retrospective cohort study. BMC Cancer 2024; 24:272. [PMID: 38408958 PMCID: PMC10898186 DOI: 10.1186/s12885-024-12016-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 02/16/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Chemotherapy for metastatic pancreatic adenocarcinoma (PDAC) primarily relies on FOLFIRINOX (LV5FU- irinotecan - Oxaliplatine) and Gemcitabine - Nab-Paclitaxel in the first-line setting. However, second-lines remain less well-defined and there is limited data regarding third-line treatments. The objective of our study was to determine the proportion of patients advancing to third line chemotherapy, to outline the various third-line chemotherapy regimens used in routine practice and to evaluate their respective efficacy. METHODS A retrospective single-center cohort from 2010-2022 compiled baseline characteristics, treatment outcomes and survival of PDAC patients who received at least one chemotherapy line in a French tertiary-center. Overall survivals (OS) were analyzed using a Cox multivariable model. RESULTS In total, 676 patients were included, with a median follow-up time of 69.4 months, (Interquartile Range (IQR) = 72.1). Of these, 251 patients (37%) that proceeded to 3rd-line chemotherapy. The median PFS in 3rd line was 2.03 months, [CI95%: 1.83, 2.36]. The median 3rd line overall survival was 5.5 months, [CI95%: 4.8, 6.3]. In multivariable analysis erlotinib-based chemotherapy was found to be deleterious (HR=2.38, [CI95%: 1.30, 4.34], p=0.005) compared to fluoropyrimidine-based chemotherapy in terms of 3rd line overall survival while gemcitabine monotherapy showed a tendency towards negative outcomes. First and 2nd line chemotherapies sequence didn't influence 3rd line outcome. CONCLUSION In our cohort, one-third of treated patients proceeded to 3rd line chemotherapy resulting in a 5.5 months median 3rd line OS, consistent with treatments at advanced stage. Our results argue against the use of erlotinib and gemcitabine monotherapy.
Collapse
Affiliation(s)
- A Gueiderikh
- Département de médecine oncologique, Gustave Roussy, 94800, Villejuif, France
- Université Paris Saclay, 91471, Orsay, France
| | - A Tarabay
- Département de médecine oncologique, Gustave Roussy, 94800, Villejuif, France
| | - M Abdelouahab
- Département de statistiques, Gustave Roussy, 94800, Villejuif, France
| | - C Smolenschi
- Département de médecine oncologique, Gustave Roussy, 94800, Villejuif, France
- Gustave Roussy, DITEP, 94800, Villejuif, France
| | - M L Tanguy
- Département de statistiques, Gustave Roussy, 94800, Villejuif, France
| | - M Valery
- Département de médecine oncologique, Gustave Roussy, 94800, Villejuif, France
| | - D Malka
- Département d'oncologie médicale, Institut Mutualiste Montsouris, 75014, Paris, France
| | - T Pudlarz
- Département de médecine oncologique, Gustave Roussy, 94800, Villejuif, France
| | - A Fuerea
- Département de médecine oncologique, Gustave Roussy, 94800, Villejuif, France
| | - V Boige
- Département de médecine oncologique, Gustave Roussy, 94800, Villejuif, France
| | - A Hollebecque
- Département de médecine oncologique, Gustave Roussy, 94800, Villejuif, France
- Gustave Roussy, DITEP, 94800, Villejuif, France
| | - M Ducreux
- Département de médecine oncologique, Gustave Roussy, 94800, Villejuif, France
- Université Paris Saclay, 91471, Orsay, France
| | - A Boilève
- Département de médecine oncologique, Gustave Roussy, 94800, Villejuif, France.
- Université Paris Saclay, 91471, Orsay, France.
| |
Collapse
|
29
|
Tomonaga Y, de Nijs K, Bucher HC, de Koning H, Ten Haaf K. Cost-effectiveness of risk-based low-dose computed tomography screening for lung cancer in Switzerland. Int J Cancer 2024; 154:636-647. [PMID: 37792671 DOI: 10.1002/ijc.34746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 10/06/2023]
Abstract
Throughout Europe, computed tomography (CT) screening for lung cancer is in a phase of clinical implementation or reimbursement evaluation. To efficiently select individuals for screening, the use of lung cancer risk models has been suggested, but their incremental (cost-)effectiveness relative to eligibility based on pack-year criteria has not been thoroughly evaluated for a European setting. We evaluate the cost-effectiveness of pack-year and risk-based screening (PLCOm2012 model-based) strategies for Switzerland, which aided in informing the recommendations of the Swiss Cancer Screening Committee (CSC). We use the MISCAN (MIcrosimulation SCreening ANalysis)-Lung model to estimate benefits and harms of screening among individuals born 1940 to 1979 in Switzerland. We evaluate 1512 strategies, differing in the age ranges employed for screening, the screening interval and the strictness of the smoking requirements. We estimate risk-based strategies to be more cost-effective than pack-year-based screening strategies. The most efficient strategy compliant with CSC recommendations is biennial screening for ever-smokers aged 55 to 80 with a 1.6% PLCOm2012 risk. Relative to no screening this strategy is estimated to reduce lung cancer mortality by 11.0%, with estimated costs per Quality-Adjusted Life-Year (QALY) gained of €19 341, and a €1.990 billion 15-year budget impact. Biennial screening ages 55 to 80 for those with 20 pack-years shows a lower mortality reduction (10.5%) and higher cost per QALY gained (€20 869). Despite model uncertainties, our estimates suggest there may be cost-effective screening policies for Switzerland. Risk-based biennial screening ages 55 to 80 for those with ≥1.6% PLCOm2012 risk conforms to CSC recommendations and is estimated to be more efficient than pack-year-based alternatives.
Collapse
Affiliation(s)
- Yuki Tomonaga
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Koen de Nijs
- Department of Public Health, Erasmus MC: University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Heiner C Bucher
- Division of Clinical Epidemiology, Department of Clinical Research University Hospital Basel and University of Basel, Basel, Switzerland
| | - Harry de Koning
- Department of Public Health, Erasmus MC: University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Kevin Ten Haaf
- Department of Public Health, Erasmus MC: University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
30
|
Zhang S, Liu Y, Ma X, Gao X, Ru Y, Hu X, Gu X. Recent advances in the potential role of RNA N4-acetylcytidine in cancer progression. Cell Commun Signal 2024; 22:49. [PMID: 38233930 PMCID: PMC10795262 DOI: 10.1186/s12964-023-01417-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 12/02/2023] [Indexed: 01/19/2024] Open
Abstract
N4-acetylcytidine (ac4C) is a highly conserved chemical modification widely found in eukaryotic and prokaryotic RNA, such as tRNA, rRNA, and mRNA. This modification is significantly associated with various human diseases, especially cancer, and its formation depends on the catalytic activity of N-acetyltransferase 10 (NAT10), the only known protein that produces ac4C. This review discusses the detection techniques and regulatory mechanisms of ac4C and summarizes ac4C correlation with tumor occurrence, development, prognosis, and drug therapy. It also comments on a new biomarker for early tumor diagnosis and prognosis prediction and a new target for tumor therapy. Video Abstract.
Collapse
Affiliation(s)
- Shujun Zhang
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Yafeng Liu
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Xiao Ma
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaohui Gao
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Yi Ru
- Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Xinjun Hu
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471000, Henan, China.
| | - Xinyu Gu
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471000, Henan, China.
| |
Collapse
|
31
|
Schranz A, Sternad C, Aziz F, Wagner D, Kornprat P, Sucher R, Jost PJ, Wölfler A, Pieber TR, Sourij H, Riedl JM, Aberer F. Incidence of Diabetes Mellitus and Its Impact on Outcomes in Patients Undergoing Surgical Pancreatectomy for Non-Malignant and Malignant Pancreatobiliary Diseases-A Retrospective Analysis. J Clin Med 2023; 12:7532. [PMID: 38137600 PMCID: PMC10744322 DOI: 10.3390/jcm12247532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
Diabetes mellitus (DM) is a prominent risk factor for malignant and non-malignant pancreatic diseases. Furthermore, the presence of DM predicts an unfavourable outcome in people with pancreatic cancer. This retrospective observational study investigated 370 patients who underwent pancreatic resection surgery for various indications (84.3% in malignant indication) in a single surgery centre in Graz, Austria. The preoperative and postoperative diabetes statuses were evaluated according to surgery method and disease entity and predictors for diabetes development after surgery, as well as outcomes (survival and cancer recurrence) according to diabetes status, were analysed. In the entire cohort, the postoperative diabetes (postopDM) incidence was 29%. PostopDM occurred significantly more frequently in malignoma patients than in those with benign diseases (31.3% vs. 16.7%; p = 0.040, OR = 2.28). In the malignoma population, BMI, longer surgery duration, and prolonged ICU and hospital stay were significant predictors of diabetes development. The 1- and 2-year follow-ups showed a significantly increased mortality of people with postopDM in comparison to people without diabetes (HR 1-year = 2.02, p = 0.014 and HR 2-years = 1.56, p = 0.034). Local cancer recurrence was not influenced by the diabetes status. Postoperative new-onset diabetes seems to be associated with higher mortality of patients with pancreatic malignoma undergoing pancreatobiliary surgery.
Collapse
Affiliation(s)
- Anna Schranz
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria; (A.S.); (C.S.); (F.A.); (T.R.P.); (F.A.)
| | - Christoph Sternad
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria; (A.S.); (C.S.); (F.A.); (T.R.P.); (F.A.)
| | - Faisal Aziz
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria; (A.S.); (C.S.); (F.A.); (T.R.P.); (F.A.)
| | - Doris Wagner
- Department of Surgery, Medical University of Graz, 8036 Graz, Austria; (D.W.); (P.K.); (R.S.)
| | - Peter Kornprat
- Department of Surgery, Medical University of Graz, 8036 Graz, Austria; (D.W.); (P.K.); (R.S.)
| | - Robert Sucher
- Department of Surgery, Medical University of Graz, 8036 Graz, Austria; (D.W.); (P.K.); (R.S.)
| | - Philipp J. Jost
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, 8036 Graz, Austria; (P.J.J.); (J.M.R.)
| | - Albert Wölfler
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, 8036 Graz, Austria;
| | - Thomas R. Pieber
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria; (A.S.); (C.S.); (F.A.); (T.R.P.); (F.A.)
| | - Harald Sourij
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria; (A.S.); (C.S.); (F.A.); (T.R.P.); (F.A.)
| | - Jakob M. Riedl
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, 8036 Graz, Austria; (P.J.J.); (J.M.R.)
| | - Felix Aberer
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria; (A.S.); (C.S.); (F.A.); (T.R.P.); (F.A.)
| |
Collapse
|
32
|
Molnár A, Halimi A, Svensson J, Bayadsi H, Innala M, Hansén M, Hemmingsson O, Franklin O. Portomesenteric venous contact ≤180° and overall survival in resectable head and body pancreatic adenocarcinoma treated with upfront surgery. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2023; 49:107097. [PMID: 37804583 DOI: 10.1016/j.ejso.2023.107097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/21/2023] [Accepted: 09/25/2023] [Indexed: 10/09/2023]
Abstract
INTRODUCTION Upfront surgery is the standard of care for resectable pancreatic cancer, defined as the absence of or ≤180° tumour contact with the portal/superior mesenteric vein. We hypothesized that portomesenteric venous tumour contact is prognostically unfavourable and aimed to assess whether it is associated with poorer survival compared with no venous contact in resectable head and body pancreatic cancer. METHODS This single-centre retrospective study included patients undergoing upfront surgery for resectable head and body pancreatic cancer in 2010-2020 at Umeå University Hospital, Sweden. No venous contact was compared with portomesenteric venous contact of ≤180° based on preoperative imaging. Overall survival on an intention-to-treat basis was compared with Kaplan-Meier curves, a log-rank test and Cox proportional hazards models. RESULTS The final study cohort included 39 patients with portomesenteric venous tumour contact and 144 patients without venous tumour contact. Patients with venous tumour contact had a median overall survival of 15.3 months compared to 23.0 months (log rank P = 0.059). Portomesenteric venous tumour contact was an independent negative prognostic factor for survival in the multivariable Cox model (HR 1.68; 95% CI 1.11-2.55, P = 0.014) and was associated with higher rates of microscopically non-radical resections (R1) (50% vs 26.1%, P = 0.012) and pathological lymph node metastasis (76.7% vs 56.8%, P = 0.012). There was no difference in adjuvant chemotherapy receipt or postoperative complications between the groups. CONCLUSIONS Portomesenteric venous tumour contact is associated with poorer overall survival and higher rates of R1 resections and lymph node metastasis in patients with resectable head and body pancreatic cancer treated with upfront surgery.
Collapse
Affiliation(s)
- Adrienne Molnár
- Department of Surgical and Perioperative Sciences, Surgery, Umeå University, Umeå, Sweden
| | - Asif Halimi
- Department of Surgical and Perioperative Sciences, Surgery, Umeå University, Umeå, Sweden
| | - Johan Svensson
- Department of Statistics, Umeå School of Business, Economics and Statistics, Umeå University, Umeå, Sweden
| | - Haytham Bayadsi
- Department of Surgical and Perioperative Sciences, Surgery, Umeå University, Umeå, Sweden
| | - Marcus Innala
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
| | - Maria Hansén
- Oncology Clinic, Sundsvall Regional Hospital, Sundsvall, Sweden
| | - Oskar Hemmingsson
- Department of Surgical and Perioperative Sciences, Surgery, Umeå University, Umeå, Sweden; Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Oskar Franklin
- Department of Surgical and Perioperative Sciences, Surgery, Umeå University, Umeå, Sweden; Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
33
|
Vidal L, Pando E, Blanco L, Fabregat-Franco C, Castet F, Sierra A, Macarulla T, Balsells J, Charco R, Vivancos A. Liquid biopsy after resection of pancreatic adenocarcinoma and its relation to oncological outcomes. Systematic review and meta-analysis. Cancer Treat Rev 2023; 120:102604. [PMID: 37572593 DOI: 10.1016/j.ctrv.2023.102604] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND It has been hypothesised that manipulation during surgery releases tumoral components into circulation. We investigate the effect of surgery on plasma-borne DNA biomarkers and the oncological outcomes in resectable pancreatic ductal adenocarcinoma (PDAC). We also compare non-touch isolation techniques (NTIT) with standard techniques. MATERIALS AND METHODS We performed a systematic review and a meta-analysis of studies analysing liquid biopsy as circulating tumour DNA (ctDNA), circulating tumour cells (CTCs), and messenger RNA (mRNA) in resectable PDAC patients who underwent surgery and its association with overall survival (OS) and disease-free survival (DFS). Research in EMBASE, Web of Science and PubMed was performed. The ctDNA shift negative-to-positive (ctDNA -/+) or ctDNA shift positive-to-negative (ctDNA +/-) before and after surgery was evaluated. RESULTS Twelve studies comprising 413 patients were included. Shorter OS and DFS were identified in patients with positive ctDNA status before (HR = 2.28, p = 0.005 and HR = 2.16, p = 0.006) or after surgery (HR = 3.88, p < 0.0001 and HR = 3.81, p = 0.03), respectively. Surgical resection increased the rate of ctDNA +/-. There were no differences in OS or DFS in the ctDNA +/- group compared with ctDNA +/+ or ctDNA -/+. However, there was a trend to shorter OS in the ctDNA -/+ group (HR = 5.00, p = 0.09). No differences between NTIT and standard techniques on liquid biopsy status were found. CONCLUSION Positive ctDNA in the perioperative period is associated with a worse prognosis. Surgical resection has a role in the negativisation of liquid biopsy status. More studies are needed to assess the potential of minimally invasive techniques on ctDNA dynamics.
Collapse
Affiliation(s)
- Laura Vidal
- Department of HPB and Transplant Surgery, Vall d'Hebron University Hospital, Barcelona, Spain; Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Elizabeth Pando
- Department of HPB and Transplant Surgery, Vall d'Hebron University Hospital, Barcelona, Spain; Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Laia Blanco
- Department of HPB and Transplant Surgery, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Carles Fabregat-Franco
- Gastrointestinal and Endocrine Tumour Unit, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Florian Castet
- Gastrointestinal and Endocrine Tumour Unit, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Alexandre Sierra
- Gastrointestinal and Endocrine Tumour Unit, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Teresa Macarulla
- Gastrointestinal and Endocrine Tumour Unit, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Joaquim Balsells
- Department of HPB and Transplant Surgery, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Ramón Charco
- Department of HPB and Transplant Surgery, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Ana Vivancos
- Cancer Genomics Lab, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
34
|
Revel MP, Chassagnon G. Ten reasons to screen women at risk of lung cancer. Insights Imaging 2023; 14:176. [PMID: 37857978 PMCID: PMC10587052 DOI: 10.1186/s13244-023-01512-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/29/2023] [Indexed: 10/21/2023] Open
Abstract
This opinion piece reviews major reasons for promoting lung cancer screening in at-risk women who are smokers or ex-smokers, from the age of 50. The epidemiology of lung cancer in European women is extremely worrying, with lung cancer mortality expected to surpass breast cancer mortality in most European countries. There are conflicting data as to whether women are at increased risk of developing lung cancer compared to men who have a similar tobacco exposure. The sharp increase in the incidence of lung cancer in women exceeds the increase in their smoking exposure which is in favor of greater susceptibility. Lung and breast cancer screening could be carried out simultaneously, as the screening ages largely coincide. In addition, lung cancer screening could be carried out every 2 years, as is the case for breast cancer screening, if the baseline CT scan is negative.As well as detecting early curable lung cancer, screening can also detect coronary heart disease and osteoporosis induced by smoking. This enables preventive measures to be taken in addition to smoking cessation assistance, to reduce morbidity and mortality in the female population. Key points • The epidemiology of lung cancer in European women is very worrying.• Lung cancer is becoming the leading cause of cancer mortality in European women.• Women benefit greatly from screening in terms of reduced risk of death from lung cancer.
Collapse
Affiliation(s)
- Marie-Pierre Revel
- Université Paris Cité, 85 Boulevard Saint-Germain, Paris, 75006, France.
- Department of Radiology, Assistance publique des Hôpitaux de Paris, Hôpital Cochin, 27 Rue du Faubourg Saint-Jacques, Paris, 75014, France.
| | - Guillaume Chassagnon
- Université Paris Cité, 85 Boulevard Saint-Germain, Paris, 75006, France
- Department of Radiology, Assistance publique des Hôpitaux de Paris, Hôpital Cochin, 27 Rue du Faubourg Saint-Jacques, Paris, 75014, France
| |
Collapse
|
35
|
Zhang J, Zhang S, Dörflein I, Ren X, Pfeffer S, Britzen-Laurent N, Grützmann R, Duan X, Pilarsky C. Impact of CRISPR/Cas9-Mediated CD73 Knockout in Pancreatic Cancer. Cancers (Basel) 2023; 15:4842. [PMID: 37835536 PMCID: PMC10572021 DOI: 10.3390/cancers15194842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Pancreatic cancer is among the cancers with the highest mortality rates. Most of the patients are found to have advanced cancer, losing the chance of surgical treatment, and there is an urgent need to find new treatment methods. Targeted therapy for specific genes that play a key role in cancer is now an important means to improve the survival rate of patients. We determined that CD73 is highly expressed in pancreatic cancer by flow cytometry and qRT-PCR assays combined with bioinformatics techniques. Application of CRISPR/Cas9 technology to knockout CD73 in human and murine cell lines, respectively, revealed that CD73 inactivation inhibited cell growth and migration and induced G1 cell cycle arrest. We also found that CD73 deletion inhibited the ERK/STAT3 pathway and activated the E-cadherin pathway. In addition, a CRISPR/Cas9 protein kinase library screen was performed and identified Pbk, Fastk, Cdk19, Adck5, Trim28, and Pfkp as possible genes regulating CD73.
Collapse
Affiliation(s)
- Jinping Zhang
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (J.Z.); (S.Z.); (I.D.); (X.R.); (S.P.); (N.B.-L.); (R.G.)
- Second Department of General Surgery, Shaanxi Provincial People’s Hospital, Xi’an 710068, China;
- Second Department of General Surgery, Third Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710068, China
| | - Shuman Zhang
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (J.Z.); (S.Z.); (I.D.); (X.R.); (S.P.); (N.B.-L.); (R.G.)
| | - Isabella Dörflein
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (J.Z.); (S.Z.); (I.D.); (X.R.); (S.P.); (N.B.-L.); (R.G.)
| | - Xiaofan Ren
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (J.Z.); (S.Z.); (I.D.); (X.R.); (S.P.); (N.B.-L.); (R.G.)
| | - Susanne Pfeffer
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (J.Z.); (S.Z.); (I.D.); (X.R.); (S.P.); (N.B.-L.); (R.G.)
| | - Nathalie Britzen-Laurent
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (J.Z.); (S.Z.); (I.D.); (X.R.); (S.P.); (N.B.-L.); (R.G.)
| | - Robert Grützmann
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (J.Z.); (S.Z.); (I.D.); (X.R.); (S.P.); (N.B.-L.); (R.G.)
| | - Xianglong Duan
- Second Department of General Surgery, Shaanxi Provincial People’s Hospital, Xi’an 710068, China;
- Second Department of General Surgery, Third Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710068, China
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
| | - Christian Pilarsky
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (J.Z.); (S.Z.); (I.D.); (X.R.); (S.P.); (N.B.-L.); (R.G.)
| |
Collapse
|
36
|
Kurban B, Sağlık BN, Osmaniye D, Levent S, Özkay Y, Kaplancıklı ZA. Synthesis and Anticancer Activities of Pyrazole-Thiadiazole-Based EGFR Inhibitors. ACS OMEGA 2023; 8:31500-31509. [PMID: 37663500 PMCID: PMC10468883 DOI: 10.1021/acsomega.3c04635] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 08/09/2023] [Indexed: 09/05/2023]
Abstract
Lung cancer is one of the most common cancer types of cancer with the highest mortality rates. However, while epidermal growth factor receptor (EGFR) is an important parameter for lung cancer, EGFR inhibitors also show great promise in the treatment of the disease. Therefore, a series of new EGFR inhibitor candidates containing thiadiazole and pyrazole rings have been developed. The activities of the synthesized compounds were elucidated by in vitro MTT, (which is chemically 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide), cytotoxicity assay, analysis of mitochondrial membrane potential (MMP) by flow cytometry, and EGFR inhibition experiments. Molecular docking and molecular dynamics simulations were performed as in silico studies. Compounds 6d, 6g, and 6j showed inhibitor activity against the A549 cell line with IC50 = 5.176 ± 0.164; 1.537 ± 0.097; and 8.493 ± 0.667 μM values, respectively. As a result of MMP by flow cytometry, compound 6g showed 80.93% mitochondrial membrane potential. According to the results of the obtained EGFR inhibitory assay, compound 6g shows inhibitory activity on the EGFR enzyme with a value of IC50 = 0.024 ± 0.002 μM.
Collapse
Affiliation(s)
- Berkant Kurban
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Afyonkarahisar Health Sciences University, Afyonkarahisar 03030, Turkey
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir 26470, Turkey
| | - Begüm Nurpelin Sağlık
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir 26470, Turkey
- Central
Research Laboratory (MERLAB), Faculty of Pharmacy, Anadolu University, Eskişehir 26470, Turkey
| | - Derya Osmaniye
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir 26470, Turkey
- Central
Research Laboratory (MERLAB), Faculty of Pharmacy, Anadolu University, Eskişehir 26470, Turkey
| | - Serkan Levent
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir 26470, Turkey
- Central
Research Laboratory (MERLAB), Faculty of Pharmacy, Anadolu University, Eskişehir 26470, Turkey
| | - Yusuf Özkay
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir 26470, Turkey
- Central
Research Laboratory (MERLAB), Faculty of Pharmacy, Anadolu University, Eskişehir 26470, Turkey
| | - Zafer Asım Kaplancıklı
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir 26470, Turkey
| |
Collapse
|
37
|
McDonnell D, Cheang AWE, Wilding S, Wild SH, Frampton AE, Byrne CD, Hamady ZZ. Elevated Glycated Haemoglobin (HbA1c) Is Associated with an Increased Risk of Pancreatic Ductal Adenocarcinoma: A UK Biobank Cohort Study. Cancers (Basel) 2023; 15:4078. [PMID: 37627106 PMCID: PMC10452109 DOI: 10.3390/cancers15164078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND The role of dysglycaemia as a risk marker for Pancreatic Ductal Adenocarcinoma (PDAC) is uncertain. We investigated the relationship between glycated haemoglobin (HbA1c) and incident PDAC using a retrospective cohort study within the UK Biobank. METHODS A study involving 499,804 participants from the UK Biobank study was undertaken. Participants were stratified by diabetes mellitus (DM) status, and then by HbA1c values < 42 mmol/mol, 42-47 mmol/mol, or ≥48 mmol/mol. Cox proportional hazard models were used to describe the association between HbA1c category (with time-varying interactions) and incident PDAC. RESULTS PDAC occurred in 1157 participants during 11.6 (10.9-12.3) years follow up [(median (interquartile range)]. In subjects without known DM at baseline, 12 months after recruitment, the adjusted hazard ratios (aHR, 95% CI) for incident PDAC for HbA1c 42-47 mmol/mol compared to HbA1c < 42 mmol/mol (reference group) was 2.10 (1.31-3.37, p = 0.002); and was 8.55 (4.58-15.99, p < 0.001) for HbA1c ≥ 48 mmol/mol. The association between baseline HbA1c and incident PDAC attenuated with increasing duration of time of follow-up to PDAC diagnosis. CONCLUSIONS Dysglycaemia detected by elevated HbA1c is associated with an increased risk of PDAC. The strength of the association between elevated HbA1c and incident PDAC is inversely proportional to the time from detecting dysglycaemia but remains significant for at least 60 months following HbA1c testing.
Collapse
Affiliation(s)
- Declan McDonnell
- Human Development & Health, University of Southampton, University Hospital, Southampton SO16 6YD, UK; (A.W.E.C.); (S.W.); (C.D.B.); (Z.Z.H.)
- HPB Unit, University Hospital Southampton, Southampton SO16 6YD, UK
| | - Adrian W. E. Cheang
- Human Development & Health, University of Southampton, University Hospital, Southampton SO16 6YD, UK; (A.W.E.C.); (S.W.); (C.D.B.); (Z.Z.H.)
| | - Sam Wilding
- Human Development & Health, University of Southampton, University Hospital, Southampton SO16 6YD, UK; (A.W.E.C.); (S.W.); (C.D.B.); (Z.Z.H.)
| | - Sarah H. Wild
- Usher Institute, University of Edinburgh, Edinburgh EH8 9YL, UK;
| | - Adam E. Frampton
- Section of Oncology, University of Surrey, Guildford GU2 7XH, UK;
- HPB Unit, Royal Surrey NHS Foundation Trust, Guildford GU2 7XX, UK
| | - Christopher D. Byrne
- Human Development & Health, University of Southampton, University Hospital, Southampton SO16 6YD, UK; (A.W.E.C.); (S.W.); (C.D.B.); (Z.Z.H.)
| | - Zaed Z. Hamady
- Human Development & Health, University of Southampton, University Hospital, Southampton SO16 6YD, UK; (A.W.E.C.); (S.W.); (C.D.B.); (Z.Z.H.)
- HPB Unit, University Hospital Southampton, Southampton SO16 6YD, UK
| |
Collapse
|
38
|
van Dam JL, Verkolf EMM, Dekker EN, Bonsing BA, Bratlie SO, Brosens LAA, Busch OR, van Driel LMJW, van Eijck CHJ, Feshtali S, Ghorbani P, de Groot DJA, de Groot JWB, Haberkorn BCM, de Hingh IH, van der Holt B, Karsten TM, van der Kolk MB, Labori KJ, Liem MSL, Loosveld OJL, Molenaar IQ, Polée MB, van Santvoort HC, de Vos-Geelen J, Wumkes ML, van Tienhoven G, Homs MYV, Besselink MG, Wilmink JW, Groot Koerkamp B. Perioperative or adjuvant mFOLFIRINOX for resectable pancreatic cancer (PREOPANC-3): study protocol for a multicenter randomized controlled trial. BMC Cancer 2023; 23:728. [PMID: 37550634 PMCID: PMC10405377 DOI: 10.1186/s12885-023-11141-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 06/30/2023] [Indexed: 08/09/2023] Open
Abstract
BACKGROUND Surgical resection followed by adjuvant mFOLFIRINOX (5-fluorouracil with leucovorin, irinotecan, and oxaliplatin) is currently the standard of care for patients with resectable pancreatic cancer. The main concern regarding adjuvant chemotherapy is that only half of patients actually receive adjuvant treatment. Neoadjuvant chemotherapy, on the other hand, guarantees early systemic treatment and may increase chemotherapy use and thereby improve overall survival. Furthermore, it may prevent futile surgery in patients with rapidly progressive disease. However, some argue that neoadjuvant therapy delays surgery, which could lead to progression towards unresectable disease and thus offset the potential benefits. Comparison of perioperative (i.e., neoadjuvant and adjuvant) with (only) adjuvant administration of mFOLFIRINOX in a randomized controlled trial (RCT) is needed to determine the optimal approach. METHODS This multicenter, phase 3, RCT will include 378 patients with resectable pancreatic ductal adenocarcinoma with a WHO performance status of 0 or 1. Patients are recruited from 20 Dutch centers and three centers in Norway and Sweden. Resectable pancreatic cancer is defined as no arterial contact and ≤ 90 degrees venous contact. Patients in the intervention arm are scheduled for 8 cycles of neoadjuvant mFOLFIRINOX followed by surgery and 4 cycles of adjuvant mFOLFIRINOX (2-week cycle of oxaliplatin 85 mg/m2, leucovorin 400 mg/m2, irinotecan 150 mg/m2 at day 1, followed by 46 h continuous infusion of 5-fluorouracil 2400 g/m2). Patients in the comparator arm start with surgery followed by 12 cycles of adjuvant mFOLFIRINOX. The primary outcome is overall survival by intention-to-treat. Secondary outcomes include progression-free survival, resection rate, quality of life, adverse events, and surgical complications. To detect a hazard ratio of 0.70 with 80% power, 252 events are needed. The number of events is expected to be reached after the inclusion of 378 patients in 36 months, with analysis planned 18 months after the last patient has been randomized. DISCUSSION The multicenter PREOPANC-3 trial compares perioperative mFOLFIRINOX with adjuvant mFOLFIRINOX in patients with resectable pancreatic cancer. TRIAL REGISTRATION Clinical Trials: NCT04927780. Registered June 16, 2021.
Collapse
Affiliation(s)
- J L van Dam
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - E M M Verkolf
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - E N Dekker
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - B A Bonsing
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - S O Bratlie
- Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - L A A Brosens
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Pathology, Radboud UMC, Nijmegen, The Netherlands
| | - O R Busch
- Department of Surgery, Amsterdam UMC, Location University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - L M J W van Driel
- Department of Gastroenterology and Hepatology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - C H J van Eijck
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - S Feshtali
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - P Ghorbani
- Division of Surgery, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - D J A de Groot
- Department of Medical Oncology, University Medical Center Groningen, Groningen, The Netherlands
| | - J W B de Groot
- Department of Medical Oncology, Isala Oncology Center, Zwolle, The Netherlands
| | - B C M Haberkorn
- Department of Medical Oncology, Maasstad Hospital, Rotterdam, The Netherlands
| | - I H de Hingh
- Department of Surgery, Catharina Hospital, Eindhoven, The Netherlands
| | - B van der Holt
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - T M Karsten
- Department of Surgery, OLVG, Amsterdam, The Netherlands
| | - M B van der Kolk
- Department of Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - K J Labori
- Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital, Rikshospitalet and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - M S L Liem
- Department of Surgery, Medisch Spectrum Twente, Enschede, The Netherlands
| | - O J L Loosveld
- Department of Medical Oncology, Amphia Hospital, Breda, The Netherlands
| | - I Q Molenaar
- Department of Surgery, Regional Academic Cancer Center Utrecht, St. Antonius Hospital and University Medical Center Utrecht, Utrecht, The Netherlands
| | - M B Polée
- Department of Medical Oncology, Medical Center Leeuwarden, Leeuwarden, The Netherlands
| | - H C van Santvoort
- Department of Surgery, Regional Academic Cancer Center Utrecht, St. Antonius Hospital and University Medical Center Utrecht, Utrecht, The Netherlands
| | - J de Vos-Geelen
- Division of Medical Oncology, Department of Internal Medicine, GROW, Maastricht UMC+, Maastricht, the Netherlands
| | - M L Wumkes
- Department of Medical Oncology, Jeroen Bosch Hospital, Den Bosch, The Netherlands
| | - G van Tienhoven
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Amsterdam UMC, Department of Radiation Oncology, Location University of Amsterdam, Amsterdam, The Netherlands
| | - M Y V Homs
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - M G Besselink
- Department of Surgery, Amsterdam UMC, Location University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - J W Wilmink
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Location University of Amsterdam, Amsterdam, The Netherlands
| | - B Groot Koerkamp
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.
| |
Collapse
|
39
|
Amini M, Azizmohammad Looha M, Rahimi Pordanjani S, Asadzadeh Aghdaei H, Pourhoseingholi MA. Global long-term trends and spatial cluster analysis of pancreatic cancer incidence and mortality over a 30-year period using the global burden of disease study 2019 data. PLoS One 2023; 18:e0288755. [PMID: 37471411 PMCID: PMC10358895 DOI: 10.1371/journal.pone.0288755] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 07/04/2023] [Indexed: 07/22/2023] Open
Abstract
INTRODUCTION Pancreatic cancer (PC) is one of the most fatal malignancies, and its incidence and mortality rates are growing annually throughout the world. In this research, we aimed to investigate the time trends and identify the spatial clusters of incidence and mortality on a global scale over the last 30 years, using the Global Burden of Disease (GBD) study 2019 data. METHODS Age-standardized incidence and mortality data due to PC were extracted from the GBD study, which was carried out from 1990 to 2019. A Joinpoint regression analysis was utilized to examine trends in the incidence and mortality of PC over the past three decades. As such, spatial analyses were undertaken to detect the spatial distribution and clustering of the metrics globally. RESULTS It was observed that both the incidence and mortality rates were higher in males than in females worldwide. The global mortality and incidence rates significantly increased by 0.8% per year over the time of follow-up period (p<0.05). By spatial cluster analysis for mortality, European and North African countries, as well as Greenland were explored as hot spots; while South African and Southeast Asian countries were explored as cold spots. Regarding incidence, hot spots were found in European countries, Southern America, and Greenland; whilst cold spots were determined in Southern Africa and Madagascar. CONCLUSIONS Collectively, the temporal trends disclosed a gradual rise in PC incidence and mortality rates over the period 1990-2019, reflecting the global health concern. We further found geographical variations in the patterns and identified high- and low-risk areas for incidence and mortality. These findings facilitate the design and implementation of more resource-efficient and geographically targeted treatments. Given the results of the current study, a practical approach to minimizing the future PC burden involves planned population-wide interventions, as well as primary prevention through healthier lifestyles.
Collapse
Affiliation(s)
- Maedeh Amini
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Azizmohammad Looha
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajjad Rahimi Pordanjani
- Social Determinants of Health Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Epidemiology and Biostatistics, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohamad Amin Pourhoseingholi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
40
|
Sasaki M, Nakamura M, Ashida R, Nakata M, Yoshimura M, Mizowaki T. Assessing target localization accuracy across different soft-tissue matching protocols using end-exhalation breath-hold cone-beam computed tomography in patients with pancreatic cancer. JOURNAL OF RADIATION RESEARCH 2023:rrad048. [PMID: 37336503 DOI: 10.1093/jrr/rrad048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 03/23/2023] [Indexed: 06/21/2023]
Abstract
The purpose of this study was to retrospectively assess target localization accuracy across different soft-tissue matching protocols using cone-beam computed tomography (CBCT) in a large sample of patients with pancreatic cancer and to estimate the optimal margin size for each protocol. Fifty-four consecutive patients with pancreatic cancer who underwent 15-fraction volumetric modulated arc therapy under the end-exhalation breath-hold condition were enrolled. Two soft-tissue matching protocols were used according to the resectability classification, including gross tumor volume (GTV) matching for potentially resectable tumors and planning target volume (PTV) matching for borderline resectable or unresectable tumors. The tolerance of the target localization error in both matching protocols was set to 5 mm in any direction. The optimal margin size for each soft-tissue matching protocol was calculated from the systematic and random errors of the inter- and intrafraction positional variations using the van Herk formula. The inter- and intrafraction positional variations of PTV matching were smaller than those of GTV matching. The percentage of target localization errors exceeding 5 mm in the first CBCT scan of each fraction in the superior-inferior direction was 12.6 and 4.8% for GTV and PTV matching, respectively. The optimal margin sizes for GTV and PTV matching were 3.7 and 2.7, 5.4 and 4.1 and 3.9 and 3.0 mm in the anterior-posterior, superior-inferior and left-right directions, respectively. Target localization accuracy in PTV matching was higher than that in GTV matching. By setting the tolerance of the target localization error, treatment can be successful within the planned margin size.
Collapse
Affiliation(s)
- Makoto Sasaki
- Division of Clinical Radiology Service, Kyoto University Hospital, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Mitsuhiro Nakamura
- Department of Information Technology and Medical Engineering, Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawaharacho, Shogoin, Sakyo-ku, Kyoto 606-8397, Japan
- Department of Radiation Oncology and Image-Applied Therapy, Graduate School of Medicine, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto University, Kyoto 606-8507, Japan
| | - Ryo Ashida
- Department of Radiation Oncology and Image-Applied Therapy, Graduate School of Medicine, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto University, Kyoto 606-8507, Japan
| | - Manabu Nakata
- Division of Clinical Radiology Service, Kyoto University Hospital, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Michio Yoshimura
- Department of Radiation Oncology and Image-Applied Therapy, Graduate School of Medicine, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto University, Kyoto 606-8507, Japan
| | - Takashi Mizowaki
- Department of Radiation Oncology and Image-Applied Therapy, Graduate School of Medicine, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
41
|
Fraunhoffer NA, Moreno Vega AI, Abuelafia AM, Morvan M, Lebarbier E, Mary-Huard T, Zimmermann MT, Lomberk G, Urrutia R, Dusetti N, Blum Y, Nicolle R, Iovanna J. Priming therapy by targeting enhancer-initiated pathways in patient-derived pancreatic cancer cells. EBioMedicine 2023; 92:104602. [PMID: 37148583 PMCID: PMC10189188 DOI: 10.1016/j.ebiom.2023.104602] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/12/2023] [Accepted: 04/19/2023] [Indexed: 05/08/2023] Open
Abstract
BACKGROUND Systems biology leveraging multi-OMICs technologies, is rapidly advancing development of precision therapies and matching patients to targeted therapies, leading to improved responses. A new pillar of precision oncology lies in the power of chemogenomics to discover drugs that sensitizes malignant cells to other therapies. Here, we test a chemogenomic approach using epigenomic inhibitors (epidrugs) to reset patterns of gene expression driving the malignant behavior of pancreatic tumors. METHODS We tested a targeted library of ten epidrugs targeting regulators of enhancers and super-enhancers on reprogramming gene expression networks in seventeen patient-derived primary pancreatic cancer cell cultures (PDPCCs), of both basal and classical subtypes. We subsequently evaluated the ability of these epidrugs to sensitize pancreatic cancer cells to five chemotherapeutic drugs that are clinically used for this malignancy. FINDINGS To comprehend the impact of epidrug priming at the molecular level, we evaluated the effect of each epidrugs at the transcriptomic level of PDPCCs. The activating epidrugs showed a higher number of upregulated genes than the repressive epidrugs (χ2 test p-value <0.01). Furthermore, we developed a classifier using the baseline transcriptome of epidrug-primed-chemosensitized PDPCCs to predict the best epidrug-priming regime to a given chemotherapy. Six signatures with a significant association with the chemosensitization centroid (R ≤ -0.80; p-value < 0.01) were identified and validated in a subset of PDPCCs. INTERPRETATION We conclude that targeting enhancer-initiated pathways in patient-derived primary cells, represents a promising approach for developing new therapies for human pancreatic cancer. FUNDING This work was supported by INCa (Grants number 2018-078 to ND and 2018- 079 to JI), Canceropole PACA (ND), Amidex Foundation (ND), and INSERM (JI).
Collapse
Affiliation(s)
- Nicolas A Fraunhoffer
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Parc Scientifique et Technologique de Luminy, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France; Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Facultad de Medicina, Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Microbiología, Parasitología e Inmunología, Buenos Aires, Argentina
| | - Aura I Moreno Vega
- Tumour Identity Card Program (CIT), French League Against Cancer, Paris, France
| | - Analía Meilerman Abuelafia
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Parc Scientifique et Technologique de Luminy, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| | - Marie Morvan
- Laboratoire Modal'X - UMR 9023, Université Paris Nanterre, Nanterre, France
| | - Emilie Lebarbier
- Laboratoire Modal'X - UMR 9023, Université Paris Nanterre, Nanterre, France; Université Paris-Saclay, AgroParisTech, INRAE, UMR MIA Paris-Saclay, Palaiseau 91120, France
| | - Tristan Mary-Huard
- Université Paris-Saclay, AgroParisTech, INRAE, UMR MIA Paris-Saclay, Palaiseau 91120, France
| | - Michael T Zimmermann
- Genomics and Precision Medicine Center (GSPMC), Medical College of Wisconsin, Milwaukee, WI, USA; Division of Research, Department of Surgery, Medical College of Wisconsin, Center, Milwaukee, WI, USA
| | - Gwen Lomberk
- Genomics and Precision Medicine Center (GSPMC), Medical College of Wisconsin, Milwaukee, WI, USA; Division of Research, Department of Surgery, Medical College of Wisconsin, Center, Milwaukee, WI, USA
| | - Raul Urrutia
- Genomics and Precision Medicine Center (GSPMC), Medical College of Wisconsin, Milwaukee, WI, USA; Division of Research, Department of Surgery, Medical College of Wisconsin, Center, Milwaukee, WI, USA
| | - Nelson Dusetti
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Parc Scientifique et Technologique de Luminy, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| | - Yuna Blum
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, ERL U1305, Rennes, France
| | - Remy Nicolle
- Tumour Identity Card Program (CIT), French League Against Cancer, Paris, France; Université Paris Cité, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, Paris F-75018, France
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Parc Scientifique et Technologique de Luminy, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France; Hospital de Alta Complejidad El Cruce, Florencio Varela, BA, Argentina; University Arturo Jauretche, Florencio Varela, BA, Argentina.
| |
Collapse
|
42
|
Zheng X, Wu Y, Zuo H, Chen W, Wang K. Metal Nanoparticles as Novel Agents for Lung Cancer Diagnosis and Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206624. [PMID: 36732908 DOI: 10.1002/smll.202206624] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/31/2022] [Indexed: 05/04/2023]
Abstract
Lung cancer is one of the most common malignancies worldwide and contributes to most cancer-related morbidity and mortality cases. During the past decades, the rapid development of nanotechnology has provided opportunities and challenges for lung cancer diagnosis and therapeutics. As one of the most extensively studied nanostructures, metal nanoparticles obtain higher satisfaction in biomedical applications associated with lung cancer. Metal nanoparticles have enhanced almost all major imaging strategies and proved great potential as sensor for detecting cancer-specific biomarkers. Moreover, metal nanoparticles could also improve therapeutic efficiency via better drug delivery, improved radiotherapy, enhanced gene silencing, and facilitated photo-driven treatment. Herein, the recently advanced metal nanoparticles applied in lung cancer therapy and diagnosis are summarized. Future perspective on the direction of metal-based nanomedicine is also discussed. Stimulating more research interests to promote the development of metal nanoparticles in lung cancer is devoted.
Collapse
Affiliation(s)
- Xinjie Zheng
- Department of Respiratory Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| | - Yuan Wu
- Department of Respiratory Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| | - Huali Zuo
- Department of Respiratory Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| | - Weiyu Chen
- Department of Respiratory Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| | - Kai Wang
- Department of Respiratory Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| |
Collapse
|
43
|
Negoita SI, Ionescu RV, Zlati ML, Antohi VM, Nechifor A. New Regional Dynamic Cancer Model across the European Union. Cancers (Basel) 2023; 15:cancers15092545. [PMID: 37174011 PMCID: PMC10177237 DOI: 10.3390/cancers15092545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/23/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND Can increasing levels of economic wealth significantly influence changes in cancer incidence and mortality rates? METHODS We investigated this issue by means of regression analyses based on the study of incidence and mortality indicators for lip, oral cavity, and pharyngeal; colon; pancreatic; lung; leukaemia; brain and central nervous system cancers in correlation with the levels of economic welfare and financial allocations to health at the level of the European Union member states, with the exception of Luxembourg and Cyprus for which there are no official statistical data reported. RESULTS The results of the study showed that there were significant disparities both regionally and by gender, requiring corrective public policy measures that were formulated in this study. CONCLUSIONS The conclusions highlight the main findings of the study in terms of the evolution of the disease, present the significant aspects that characterise the evolution of each type of cancer during the period analysed (1993-2021), and highlight the novelty and limitations of the study and future directions of research. As a result, increasing economic welfare is a potential factor in halting the effects of cancer incidence and mortality at the population level, while the financial allocations to health of EU member countries' budgets are a drawback due to large regional disparities.
Collapse
Affiliation(s)
- Silvius Ioan Negoita
- Anaesthesia Intensive Care Unit, Department Orthopedics, University of Medicine and Pharmacy Carol Davila of Bucharest, 020021 Bucharest, Romania
| | - Romeo Victor Ionescu
- Department of Administrative Sciences and Regional Studies, Dunarea de Jos University of Galati, 800008 Galati, Romania
| | - Monica Laura Zlati
- Department of Business Administration, Dunarea de Jos University of Galati, 800008 Galati, Romania
| | - Valentin Marian Antohi
- Department of Business Administration, Dunarea de Jos University of Galati, 800008 Galati, Romania
- Departament of Finance, Accounting and Economic Theory, Transilvania University of Brasov, 500036 Galati, Romania
| | - Alexandru Nechifor
- Department of Medical Clinical, Dunarea de Jos University of Galati, 800008 Galati, Romania
| |
Collapse
|
44
|
Sun Y, Li Z, Tian Y, Gao C, Liang B, Cao S, Liu X, Liu X, Meng C, Xu J, Yang H, Zhou Y. Development and validation of nomograms for predicting overall survival and cancer-specific survival in elderly patients with locally advanced gastric cancer: a population-based study. BMC Gastroenterol 2023; 23:117. [PMID: 37041468 PMCID: PMC10091668 DOI: 10.1186/s12876-023-02749-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 03/28/2023] [Indexed: 04/13/2023] Open
Abstract
OBJECTIVE To evaluate the multiple factors influencing the survival of elderly patients with locally advanced gastric cancer (LAGC) and develop and validate the novel nomograms for predicting the survival. METHODS The clinical features of patients treated between 2000 and 2018 were collected and collated from the Surveillance, Epidemiology, and End Results (SEER) database and three medical centres in China, and the patients were randomly divided into a training cohort (3494), internal validation cohort (1497) and external validation cohort (841). Univariate and multivariate analyses of the prognostic values were performed to identify independent prognostic factors associated with overall survival (OS) and cancer-specific survival (CSS), and two nomogram models were developed. Harrell's concordance index (C-index) and calibration curves were employed to assess discrimination and calibration. Decision curve analysis (DCA) and receiver-operating characteristic (ROC) curves were utilized to investigate the clinical usefulness. RESULTS In the SEER database, the 5-year OS of the patients was 31.08%, while the 5-year CSS of the patients was 44.09%. Furthermore, in the external validation set, the 5-year OS of the patients was 49.58%, and the 5-year CSS of these patients was 53.51%. After statistical analysis, nine independent prognostic factors of OS and CSS were identified, including age, race, tumour size, differentiation, TNM stage, gastrectomy type, lymph node metastasis (LNM), lymph node ratio (LNR) and chemotherapy. The C-index (approximately 0.7) and calibration curve (close to the optimal calibration line) indicated satisfactory discrimination and calibration of the nomogram. DCA and ROC curves showed that the developed nomogram was superior to TNM stage. CONCLUSION The novel validated nomogram could accurately predict the prognosis of individual elderly patients with LAGC and guide the selection of clinical treatment measures.
Collapse
Affiliation(s)
- Yuqi Sun
- Department of General Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Zequn Li
- Department of General Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Yulong Tian
- Department of General Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Chao Gao
- Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Benjia Liang
- Shandong Provincial Hospital, Jinan, Shandong Province, China
| | - Shougen Cao
- Department of General Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xiaodong Liu
- Department of General Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xuechao Liu
- Department of General Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Cheng Meng
- Department of General Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Jianfei Xu
- Department of General Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Hao Yang
- Department of General Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Yanbing Zhou
- Department of General Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China.
| |
Collapse
|
45
|
Scherübl H. [Prevention of pancreatic cancer]. Dtsch Med Wochenschr 2023; 148:246-252. [PMID: 36848888 DOI: 10.1055/a-1975-2366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
The incidence of pancreatic cancer is rising in Germany. At present pancreatic cancer is the third commonest cause of cancer death but is expected to become the second in 2030 and finally the leading cause of cancer death in 2050. Pancreatic ductal adenocarcinoma (PC) is generally diagnosed at far advanced stages and 5-year-survival has remained poor. Modifiable risk factors of PC are tobacco smoking, excess body weight, alcohol use, type 2-diabetes and the metabolic syndrome. Smoking cessation and -in case of obesity- intentional weight loss can reduce PC risk by as much as 50 %. Early detection of asymptomatic sporadic PC at stage IA - stage IA-PC now has a 5-year-survival rate of about 80 %- has become a realistic chance for people older than 50 years with new-onset diabetes.
Collapse
Affiliation(s)
- Hans Scherübl
- Klinik für Gastroenterologie, Gastrointestinale Onkologie und Infektiologie, Vivantes Klinikum am Urban, 10967 Berlin
| |
Collapse
|
46
|
Allen MJ, Zhang A, Bavi P, Kim JC, Jang GH, Kelly D, Perera S, Denroche RE, Notta F, Wilson JM, Dodd A, Ramotar S, Hutchinson S, Fischer SE, Grant RC, Gallinger S, Knox JJ, O'Kane GM. Molecular characterisation of pancreatic ductal adenocarcinoma with NTRK fusions and review of the literature. J Clin Pathol 2023; 76:158-165. [PMID: 34583947 DOI: 10.1136/jclinpath-2021-207781] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/01/2021] [Indexed: 12/09/2022]
Abstract
AIMS The majority of pancreatic ductal adenocarcinomas (PDACs) harbour oncogenic mutations in KRAS with variants in TP53, CDKN2A and SMAD4 also prevalent. The presence of oncogenic fusions including NTRK fusions are rare but important to identify. Here we ascertain the prevalence of NTRK fusions and document their genomic characteristics in a large series of PDAC. METHODS Whole genome sequencing and RNAseq were performed on a series of patients with resected or locally advanced/metastatic PDAC collected between 2008 and 2020 at a single institution. A subset of specimens underwent immunohistochemistry (IHC) analysis. Clinical and molecular characterisation and IHC sensitivity and specificity were evaluated. RESULTS 400 patients were included (resected n=167; locally advanced/metastatic n=233). Three patients were identified as harbouring an NTRK fusion, two EML4-NTRK3 (KRAS-WT) and a single novel KANK1-NTRK3 fusion. The latter occurring in the presence of a subclonal KRAS mutation. Typical PDAC drivers were present including mutations in TP53 and CDKN2A. Substitution base signatures and tumour mutational burden were similar to typical PDAC. The prevalence of NTRK fusions was 0.8% (3/400), while in KRAS wild-type tumours, it was 6.25% (2/32). DNA prediction alone documented six false-positive cases. RNA analysis correctly identified the in-frame fusion transcripts. IHC analysis was negative in the KANK1-NTRK3 fusion but positive in a EML4-NTRK3 case, highlighting lower sensitivity of IHC. CONCLUSION NTRK fusions are rare; however, with emerging therapeutic options targeting these fusions, detection is vital. Reflex testing for KRAS mutations and subsequent RNA-based screening could help identify these cases in PDAC.
Collapse
Affiliation(s)
- Michael J Allen
- Wallace McCain Centre for Pancreatic Cancer, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Amy Zhang
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Prashant Bavi
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Jaeseung C Kim
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Gun Ho Jang
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Deirdre Kelly
- Wallace McCain Centre for Pancreatic Cancer, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Sheron Perera
- Wallace McCain Centre for Pancreatic Cancer, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Rob E Denroche
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Faiyaz Notta
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Julie M Wilson
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Anna Dodd
- Wallace McCain Centre for Pancreatic Cancer, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Stephanie Ramotar
- Wallace McCain Centre for Pancreatic Cancer, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Shawn Hutchinson
- Wallace McCain Centre for Pancreatic Cancer, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Sandra E Fischer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Robert C Grant
- Wallace McCain Centre for Pancreatic Cancer, Princess Margaret Hospital, Toronto, Ontario, Canada
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Steven Gallinger
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Hepatobiliary/Pancreatic Surgical Oncology Program, University Health Network, Toronto, Ontario, Canada
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Jennifer J Knox
- Wallace McCain Centre for Pancreatic Cancer, Princess Margaret Hospital, Toronto, Ontario, Canada
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Grainne M O'Kane
- Wallace McCain Centre for Pancreatic Cancer, Princess Margaret Hospital, Toronto, Ontario, Canada Grainne.O'
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| |
Collapse
|
47
|
Appadurai JP, G S, Prabhu Kavin B, C K, Lai WC. Multi-Process Remora Enhanced Hyperparameters of Convolutional Neural Network for Lung Cancer Prediction. Biomedicines 2023; 11:biomedicines11030679. [PMID: 36979657 PMCID: PMC10045623 DOI: 10.3390/biomedicines11030679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/30/2023] [Accepted: 02/08/2023] [Indexed: 03/30/2023] Open
Abstract
In recent years, lung cancer prediction is an essential topic for reducing the death rate of humans. In the literature section, some papers are reviewed that reduce the accuracy level during the prediction stage. Hence, in this paper, we develop a Multi-Process Remora Optimized Hyperparameters of Convolutional Neural Network (MPROH-CNN) aimed at lung cancer prediction. The proposed technique can be utilized to detect the CT images of the human lung. The proposed technique proceeds with four phases, including pre-processing, feature extraction and classification. Initially, the databases are collected from the open-source system. After that, the collected CT images contain unwanted noise, which affects classification efficiency. So, the pre-processing techniques can be considered to remove unwanted noise from the input images, such as filtering and contrast enhancement. Following that, the essential features are extracted with the assistance of feature extraction techniques such as histogram, texture and wavelet. The extracted features are utilized to classification stage. The proposed classifier is a combination of the Remora Optimization Algorithm (ROA) and Convolutional Neural Network (CNN). In the CNN, the ROA is utilized for multi process optimization such as structure optimization and hyperparameter optimization. The proposed methodology is implemented in MATLAB and performances are evaluated by utilized performance matrices such as accuracy, precision, recall, specificity, sensitivity and F_Measure. To validate the projected approach, it is compared with the traditional techniques CNN, CNN-Particle Swarm Optimization (PSO) and CNN-Firefly Algorithm (FA), respectively. From the analysis, the proposed method achieved a 0.98 accuracy level in the lung cancer prediction.
Collapse
Affiliation(s)
- Jothi Prabha Appadurai
- Computer Science and Engineering Department, Kakatiya Institute of Technology and Science, Warangal 506015, Telangana, India
| | - Suganeshwari G
- School of Computer Science and Engineering, Vellore Institute of Technology, Chennai 600127, Tamil Nadu, India
| | - Balasubramanian Prabhu Kavin
- Department of Data Science and Business Systems, College of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Chengalpattu District, Chennai 603203, Tamil Nadu, India
| | - Kavitha C
- Department of Computer Science and Engineering, Sathyabama Institute of Science and Technology, Chennai 600119, Tamil Nadu, India
| | - Wen-Cheng Lai
- Bachelor Program in Industrial Projects, National Yunlin University of Science and Technology, Douliu 640301, Taiwan
- Department Electronic Engineering, National Yunlin University of Science and Technology, Douliu 640301, Taiwan
| |
Collapse
|
48
|
Schulz D, Heilmaier M, Phillip V, Treiber M, Mayr U, Lahmer T, Mueller J, Demir IE, Friess H, Reichert M, Schmid RM, Abdelhafez M. Accurate prediction of histological grading of intraductal papillary mucinous neoplasia using deep learning. Endoscopy 2023; 55:415-422. [PMID: 36323331 DOI: 10.1055/a-1971-1274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
BACKGROUND Risk stratification and recommendation for surgery for intraductal papillary mucinous neoplasm (IPMN) are currently based on consensus guidelines. Risk stratification from presurgery histology is only potentially decisive owing to the low sensitivity of fine-needle aspiration. In this study, we developed and validated a deep learning-based method to distinguish between IPMN with low grade dysplasia and IPMN with high grade dysplasia/invasive carcinoma using endoscopic ultrasound (EUS) images. METHODS For model training, we acquired a total of 3355 EUS images from 43 patients who underwent pancreatectomy from March 2015 to August 2021. All patients had histologically proven IPMN. We used transfer learning to fine-tune a convolutional neural network and to classify "low grade IPMN" from "high grade IPMN/invasive carcinoma." Our test set consisted of 1823 images from 27 patients, recruiting 11 patients retrospectively, 7 patients prospectively, and 9 patients externally. We compared our results with the prediction based on international consensus guidelines. RESULTS Our approach could classify low grade from high grade/invasive carcinoma in the test set with an accuracy of 99.6 % (95 %CI 99.5 %-99.9 %). Our deep learning model achieved superior accuracy in prediction of the histological outcome compared with any individual guideline, which have accuracies between 51.8 % (95 %CI 31.9 %-71.3 %) and 70.4 % (95 %CI 49.8-86.2). CONCLUSION This pilot study demonstrated that deep learning in IPMN-EUS images can predict the histological outcome with high accuracy.
Collapse
Affiliation(s)
- Dominik Schulz
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Markus Heilmaier
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Veit Phillip
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Matthias Treiber
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Ulrich Mayr
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Tobias Lahmer
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Julius Mueller
- Klinik für Innere Medizin II, Universitätsklinikum Freiburg, Freiburg, Germany
| | - Ihsan Ekin Demir
- Klinik und Poliklinik für Chirurgie, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Helmut Friess
- Klinik und Poliklinik für Chirurgie, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Maximilian Reichert
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Roland M Schmid
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Mohamed Abdelhafez
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
49
|
Kiemen AL, Damanakis AI, Braxton AM, He J, Laheru D, Fishman EK, Chames P, Pérez CA, Wu PH, Wirtz D, Wood LD, Hruban RH. Tissue clearing and 3D reconstruction of digitized, serially sectioned slides provide novel insights into pancreatic cancer. MED 2023; 4:75-91. [PMID: 36773599 PMCID: PMC9922376 DOI: 10.1016/j.medj.2022.11.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/06/2022] [Accepted: 11/23/2022] [Indexed: 01/26/2023]
Abstract
Pancreatic cancer is currently the third leading cause of cancer death in the United States. The clinical hallmarks of this disease include abdominal pain that radiates to the back, the presence of a hypoenhancing intrapancreatic lesion on imaging, and widespread liver metastases. Technologies such as tissue clearing and three-dimensional (3D) reconstruction of digitized serially sectioned hematoxylin and eosin-stained slides can be used to visualize large (up to 2- to 3-centimeter cube) tissues at cellular resolution. When applied to human pancreatic cancers, these 3D visualization techniques have provided novel insights into the basis of a number of the clinical characteristics of this disease. Here, we describe the clinical features of pancreatic cancer, review techniques for clearing and the 3D reconstruction of digitized microscope slides, and provide examples that illustrate how 3D visualization of human pancreatic cancer at the microscopic level has revealed features not apparent in 2D microscopy and, in so doing, has closed the gap between bench and bedside. Compared with animal models and 2D microscopy, studies of human tissues in 3D can reveal the difference between what can happen and what does happen in human cancers.
Collapse
Affiliation(s)
- Ashley L Kiemen
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Chemical & Biomolecular Engineering, The Johns Hopkins University, 3400 N Charles St, Baltimore, MD 21218, USA
| | - Alexander Ioannis Damanakis
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of General, Visceral, Cancer and Transplant Surgery, University Hospital of Cologne, Cologne, Germany
| | - Alicia M Braxton
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jin He
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Daniel Laheru
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Elliot K Fishman
- Department of Radiology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Patrick Chames
- Antibody Therapeutics and Immunotargeting Team, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Cristina Almagro Pérez
- Department of Chemical & Biomolecular Engineering, The Johns Hopkins University, 3400 N Charles St, Baltimore, MD 21218, USA
| | - Pei-Hsun Wu
- Department of Chemical & Biomolecular Engineering, The Johns Hopkins University, 3400 N Charles St, Baltimore, MD 21218, USA
| | - Denis Wirtz
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Chemical & Biomolecular Engineering, The Johns Hopkins University, 3400 N Charles St, Baltimore, MD 21218, USA
| | - Laura D Wood
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | - Ralph H Hruban
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
50
|
Xu Q, Zhou M, Yin P, Jin D. Projections of cancer mortality by 2025 in central China: A modeling study of global burden of disease 2019. Heliyon 2023; 9:e13432. [PMID: 36820046 PMCID: PMC9937990 DOI: 10.1016/j.heliyon.2023.e13432] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 01/26/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Background In China, there are few studies that have reported future estimations for cancer mortality. Therefore, this study aimed to assess cancer mortality in China and identify priorities for future cancer control strategies. Methods Based on the Global Burden of Disease 2019 study, we extracted data on cancer-related deaths from 1990 to 2019 in Hunan Province, China. Under the current trends evaluated using a joinpoint regression model, we fitted a linear regression model for cancer mortality projections by 2025. Results The age-standardized mortality rate of total cancer in Hunan, China, declined slowly and is projected to be 140.80 (95% confidence interval [CI]: 140.12-141.48) by 2025, with the mortality rate in men approximately twice that in women. In 2025, the top five causes of cancer-related deaths in males are projected to be lung, liver, colorectal, stomach, and esophageal cancers, with the corresponding causes in females being lung, breast, colorectal, liver, and cervical cancers. Between 2019 and 2025, male mortality rates due to liver and pancreatic cancer are expected to increase, while those due to the six leading female cancers will increase. Excess male deaths were associated with liver and esophageal cancers, while all main cancers in females will have excess mortality, except for colorectal cancer. Conclusion A comprehensive cancer spectrum characteristic of both developing and developed countries will remain in Hunan, China. Lung cancer remains the most common cause of cancer-related deaths, and tobacco control efforts are urgently required. Additional efforts should be made to promote universal screening, improve access to cancer healthcare services, optimize medical payment models, and enhance access to valuable anticancer drugs.
Collapse
Affiliation(s)
- Qiaohua Xu
- Department of Chronic Disease Control and Prevention, Hunan Provincial Center for Disease Control and Prevention, Changsha, China,Corresponding author.
| | - Maigeng Zhou
- National Center for Chronic and Non-communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Peng Yin
- National Center for Chronic and Non-communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Donghui Jin
- Department of Chronic Disease Control and Prevention, Hunan Provincial Center for Disease Control and Prevention, Changsha, China
| |
Collapse
|