1
|
Choe H, Wang Z, Huang J, Yang Y, Zhao Z, Jo H, Pak H, Ali T, Ding K, Ma J, Li L, Shang D, Zhang L. Broussoflavonol B induces S-phase arrest and apoptosis in pancreatic cancer cells by modulating the cell cycle checkpoint through inhibition of the AURKA/PLK1 pathway. Cancer Cell Int 2025; 25:100. [PMID: 40098146 PMCID: PMC11916852 DOI: 10.1186/s12935-025-03717-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 02/25/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Broussoflavonol B (Bf-B), a flavonoid compound identified in the roots of Daphne giraldii Nitsche, has been extensively investigated for its potential anti-inflammatory, antioxidant, and anticancer properties. However, the precise mechanism underlying the regulation of AURKA/PLK1 pathway-mediated cell cycle arrest by Bf-B in pancreatic cancer remains poorly understood. PURPOSE The objective of this study was to investigate the inhibitory effect of Bf-B on pancreatic ductal adenocarcinoma (PDAC) and its underlying mechanism. METHODS A CCK8 assay was conducted to identify the flavonoids with the highest inhibitory activity against PANC-1, the pancreatic cancer cell line among the 25 flavonoids. Through bioinformatics analysis and molecular docking, the pathogenic targets of pancreatic cancer and flavonoid-related targets were explored, and the key targets and signaling pathways of drug intervention in pancreatic cancer were analyzed. The viability and migration ability of pancreatic cancer cells were assessed following treatment with Bf-B via the CCK8, colony formation, and wound healing assays. The cell cycle distribution and cell apoptosis were analyzed through flow cytometry and Hoechst staining. Western blotting and qPCR were employed to investigate the expression of relevant proteins and genes. For in vivo experiments, we employed a xenograft mouse model to evaluate the anticancer efficacy of Bf-B. Immunohistochemistry and immunofluorescence assays were employed to investigate the expression of relevant proteins. RESULTS In this study, the structure‒activity relationships of 25 flavonoids were evaluated. The results demonstrated that Bf-B with diisopentenyl has potent cytotoxic effects on PANC-1 cancer cells. AURKA, PLK1, and MET might serve as key targets for Bf-B inhibition of disease progression in PDAC patients. The results demonstrated that Bf-B inhibits the proliferation and migration of PANC-1 and BXPC-3 cells and induces cell cycle S-phase arrest, apoptosis, and DNA damage. Moreover, the results of western blot and qPCR experiments indicated that Bf-B exerts anticancer effects by downregulating the expression of the genes encoding AURKA/PLK1, the cell cycle checkpoint kinase ATR/CHK1/CDC25C, and Cyclin B1/CDK1 signaling pathway-related proteins and upregulating the expression of PP53, P21, and histone H2A. XS139ph expression. In xenograft-bearing mice, AURKA/PLK1 expression was reduced in a dose-dependent manner, accompanied by an increase in histone H2A. XS139ph expression. CONCLUSION Bf-B might be a potent therapeutic agent for pancreatic cancer because of its ability to suppress the expression of AURKA/PLK1.
Collapse
Affiliation(s)
- HyokChol Choe
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China
- Department of Clinical Medicine, Sinuiju Medical University, Sinuiju, Korea
| | - Zhen Wang
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jianhua Huang
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China
| | - Yutong Yang
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China
| | - Zhihao Zhao
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China
| | - HyonSu Jo
- Pyongyang University of Medical Sciences, Pyongyang, Korea
| | - HyonU Pak
- Pyongyang University of Medical Sciences, Pyongyang, Korea
| | - Tanveer Ali
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China
| | - Kaiyue Ding
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China
| | - Junnan Ma
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China
| | - Lingzhi Li
- The College of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, China
| | - Dong Shang
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China.
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Lin Zhang
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China.
| |
Collapse
|
2
|
Mahumud RA, Shahjalal M, Dahal PK, Mosharaf MP, Mistry SK, Koly KN, Chowdhury SH, Renzaho AMN, Gow J, Alam K, Wawryk O. Emerging burden of post-cancer therapy complications on unplanned hospitalisation and costs among Australian cancer patients: a retrospective cohort study over 14 years. Sci Rep 2025; 15:4709. [PMID: 39922897 PMCID: PMC11807139 DOI: 10.1038/s41598-025-89247-y] [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: 10/18/2024] [Accepted: 02/04/2025] [Indexed: 02/10/2025] Open
Abstract
Cancer treatment using systemic therapy and radiotherapy may cause post-therapy complications, resulting in increased unplanned hospitalisation. The evidence on such complications, their impact on unplanned hospitalisations, and associated costs is scant in Australia. We aimed to estimate the prevalence of post-therapy complications, evaluate their impact on unplanned hospitalisation, length of stay (LOS) and investigate the associated medical costs. A retrospective cohort study was conducted among 8,633 cancer patients (1.03 million emergency hospital admissions) in Victoria, Australia from July 2006 to June 2020, from the Australian healthcare system perspective. Multivariate generalised linear regression models were employed to estimate the adjusted association between post-therapy complications and clinical characteristics with hospital LOS and associated hospitalisation medical costs. Approximately 52% of patients were male with an average patient age of 59.9 years. Annually, post-therapy complications leading to unplanned hospitalisations increased by 7.25%, outpacing the growth in overall hospitalisation admissions, which was 5.66% for overall hospitalisation admissions. A significant proportion of patients (71%) experienced multiple complications, with the most common being anemia (26%), sepsis (15%), nausea and vomiting (14%), and neutropenia (11%). Patients undergoing combined systemic and radiotherapy exhibited higher odds of post-therapy complications (OR = 8.24, 95%CI: 7.48 to 9.08) compared with those who only received systemic therapy. Mean hospital stay among patients who experienced post-therapy complications was 2.23 days per admission (360 days per patient), an extra 1.72 days per admission [95%CI: 1.68 to 1.76; 354 days per patient, 95%CI: 336 to 371 days] longer than patients without complications (0.51 days per admission and 6.48 days per patients). Overall, per-admission medical hospitalisation costs among patients with post-therapy complications were $8,791 higher than for patients who did not experience complications ($11,418 vs. $2,627 per admission, 95%CI: $8,685 to $8,897). Per-patient costs for unplanned hospitalisation due to post-therapy complications were significantly $1.82 million higher among patients than those without complications ($1.86 million vs. $33,599 per patient, 95%CI: $1.71 million to $1.94 million). The cost and hospitalisation stay (in days) varied by the type of therapy and cancer type. The study results indicate that post-therapy complications in cancer patients varied by the type of cancer and increased over the study period, leading to longer unplanned hospital stays and higher hospitalisation medical costs. The results highlight the need for better-customized treatment delivery strategies to address this burden and optimise resources in cancer care.
Collapse
Affiliation(s)
- Rashidul Alam Mahumud
- Health Economics and Health Technology Assessment Unit, NHMRC Clinical Trials Centre, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Md Shahjalal
- Global Health Institute, Department of Public Health, North South University, Dhaka 1229, Bangladesh.
| | - Padam Kanta Dahal
- School of Health, Medical and Applied Sciences, Central Queensland University, Sydney Campus, Sydney, NSW, Australia
| | - Md Parvez Mosharaf
- School of Business and Centre for Health Research, University of Southern Queensland, Toowoomba, QLD, Australia
| | - Sabuj Kanti Mistry
- School of Population Health, University of New South Wales, Sydney, Australia
- Department of Public Health, Daffodil International University, Dhaka, Bangladesh
| | - Kamrun Nahar Koly
- Health System and Population Studies Division, Urban Health, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | | | - Andre M N Renzaho
- School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia
| | - Jeff Gow
- School of Accounting, Economics and Finance, University of KwaZulu-Natal, Durban, South Africa
- School of Health, University of Sunshine Coast, Sippy Downs, QLD, 4556, Australia
| | - Khorshed Alam
- School of Business and Centre for Health Research, University of Southern Queensland, Toowoomba, QLD, Australia
| | - Olivia Wawryk
- Department of General Practice, Victorian Comprehensive Cancer Centre, Data Connect, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
3
|
Haggstrom L, Chan WY, Nagrial A, Chantrill LA, Sim HW, Yip D, Chin V. Chemotherapy and radiotherapy for advanced pancreatic cancer. Cochrane Database Syst Rev 2024; 12:CD011044. [PMID: 39635901 PMCID: PMC11619003 DOI: 10.1002/14651858.cd011044.pub3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
BACKGROUND Pancreatic cancer (PC) is a lethal disease with few effective treatment options. Many anti-cancer therapies have been tested in the locally advanced and metastatic setting, with mixed results. This review synthesises all the randomised data available to help better inform patient and clinician decision-making. It updates the previous version of the review, published in 2018. OBJECTIVES To assess the effects of chemotherapy, radiotherapy, or both on overall survival, severe or life-threatening adverse events, and quality of life in people undergoing first-line treatment of advanced pancreatic cancer. SEARCH METHODS We searched for published and unpublished studies in CENTRAL, MEDLINE, Embase, and CANCERLIT, and handsearched various sources for additional studies. The latest search dates were in March and July 2023. SELECTION CRITERIA We included randomised controlled trials comparing chemotherapy, radiotherapy, or both with another intervention or best supportive care. Participants were required to have locally advanced, unresectable pancreatic cancer or metastatic pancreatic cancer not amenable to curative intent treatment. Histological confirmation was required. Trials were required to report overall survival. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane. MAIN RESULTS We included 75 studies in the review and 51 in the meta-analysis (11,333 participants). We divided the studies into seven categories: any anti-cancer treatment versus best supportive care; various chemotherapy types versus gemcitabine; gemcitabine-based combinations versus gemcitabine alone; various chemotherapy combinations versus gemcitabine plus nab-paclitaxel; fluoropyrimidine-based studies; miscellaneous studies; and radiotherapy studies. In general, the included studies were at low risk for random sequence generation, detection bias, attrition bias, and reporting bias, at unclear risk for allocation concealment, and high risk for performance bias. Compared to best supportive care, chemotherapy likely results in little to no difference in overall survival (OS) (hazard ratio (HR) 1.08, 95% confidence interval (CI) 0.88 to 1.33; absolute risk of death at 12 months of 971 per 1000 versus 962 per 1000; 4 studies, 298 participants; moderate-certainty evidence). The adverse effects of chemotherapy and impacts on quality of life (QoL) were uncertain. Many of the chemotherapy regimens were outdated. Eight studies compared non-gemcitabine-based chemotherapy regimens to gemcitabine. These showed that 5-fluorouracil (5FU) likely reduces OS (HR 1.69, 95% CI 1.26 to 2.27; risk of death at 12 months of 914 per 1000 versus 767 per 1000; 1 study, 126 participants; moderate certainty), and grade 3/4 adverse events (QoL not reported). Fixed dose rate gemcitabine likely improves OS (HR 0.79, 95% CI 0.66 to 0.94; risk of death at 12 months of 683 per 1000 versus 767 per 1000; 2 studies, 644 participants; moderate certainty), and likely increase grade 3/4 adverse events (QoL not reported). FOLFIRINOX improves OS (HR 0.51, 95% CI 0.43 to 0.60; risk of death at 12 months of 524 per 1000 versus 767 per 1000; P < 0.001; 2 studies, 652 participants; high certainty), and delays deterioration in QoL, but increases grade 3/4 adverse events. Twenty-eight studies compared gemcitabine-based combinations to gemcitabine. Gemcitabine plus platinum may result in little to no difference in OS (HR 0.94, 95% CI 0.81 to 1.08; risk of death at 12 months of 745 per 1000 versus 767 per 1000; 6 studies, 1140 participants; low certainty), may increase grade 3/4 adverse events, and likely worsens QoL. Gemcitabine plus fluoropyrimidine improves OS (HR 0.88, 95% CI 0.81 to 0.95; risk of death at 12 months of 722 per 1000 versus 767 per 1000; 10 studies, 2718 participants; high certainty), likely increases grade 3/4 adverse events, and likely improves QoL. Gemcitabine plus topoisomerase inhibitors result in little to no difference in OS (HR 1.01, 95% CI 0.87 to 1.16; risk of death at 12 months of 770 per 1000 versus 767 per 1000; 3 studies, 839 participants; high certainty), likely increases grade 3/4 adverse events, and likely does not alter QoL. Gemcitabine plus taxane result in a large improvement in OS (HR 0.71, 95% CI 0.62 to 0.81; risk of death at 12 months of 644 per 1000 versus 767 per 1000; 2 studies, 986 participants; high certainty), and likely increases grade 3/4 adverse events and improves QoL. Nine studies compared chemotherapy combinations to gemcitabine plus nab-paclitaxel. Fluoropyrimidine-based combination regimens improve OS (HR 0.79, 95% CI 0.70 to 0.89; risk of death at 12 months of 542 per 1000 versus 628 per 1000; 6 studies, 1285 participants; high certainty). The treatment arms had distinct toxicity profiles, and there was little to no difference in QoL. Alternative schedules of gemcitabine plus nab-paclitaxel likely result in little to no difference in OS (HR 1.10, 95% CI 0.82 to 1.47; risk of death at 12 months of 663 per 1000 versus 628 per 1000; 2 studies, 367 participants; moderate certainty) or QoL, but may increase grade 3/4 adverse events. Four studies compared fluoropyrimidine-based combinations to fluoropyrimidines alone, with poor quality evidence. Fluoropyrimidine-based combinations are likely to result in little to no impact on OS (HR 0.84, 95% CI 0.61 to 1.15; risk of death at 12 months of 765 per 1000 versus 704 per 1000; P = 0.27; 4 studies, 491 participants; moderate certainty) versus fluoropyrimidines alone. The evidence suggests that there was little to no difference in grade 3/4 adverse events or QoL between the two groups. We included only one radiotherapy (iodine-125 brachytherapy) study with 165 participants. The evidence is very uncertain about the effect of radiotherapy on outcomes. AUTHORS' CONCLUSIONS Combination chemotherapy remains standard of care for metastatic pancreatic cancer. Both FOLFIRINOX and gemcitabine plus a taxane improve OS compared to gemcitabine alone. Furthermore, the evidence suggests that fluoropyrimidine-based combination chemotherapy regimens improve OS compared to gemcitabine plus nab-paclitaxel. The effects of radiotherapy were uncertain as only one low-quality trial was included. Selection of the most appropriate chemotherapy for individuals still remains unpersonalised, with clinicopathological stratification remaining elusive. Biomarker development is essential to assist in rationalising treatment selection for patients.
Collapse
Affiliation(s)
- Lucy Haggstrom
- Medical Oncology, The Kinghorn Cancer Care Centre, St Vincent's Hospital, Sydney, Australia
- Medical Oncology, Illawarra Shoalhaven Local Health District, Wollongong, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Wei Yen Chan
- Medical Oncology, The Kinghorn Cancer Care Centre, St Vincent's Hospital, Sydney, Australia
- Medical Oncology, Chris O'Brien Lifehouse, Sydney, Australia
| | - Adnan Nagrial
- The Crown Princess Mary Cancer Centre, Westmead, Australia
- Medical School, The University of Sydney, Sydney, Australia
| | - Lorraine A Chantrill
- Medical Oncology, Illawarra Shoalhaven Local Health District, Wollongong, Australia
- University of Wollongong, Wollongong, Australia
| | - Hao-Wen Sim
- Medical Oncology, The Kinghorn Cancer Care Centre, St Vincent's Hospital, Sydney, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - Desmond Yip
- Department of Medical Oncology, The Canberra Hospital, Garran, Australia
- ANU Medical School, Australian National University, Acton, Australia
| | - Venessa Chin
- Medical Oncology, The Kinghorn Cancer Care Centre, St Vincent's Hospital, Sydney, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
- Medical Oncology, Garvan Institute of Medical Research, Sydney, Australia
| |
Collapse
|
4
|
Wu J, Tang G, Cheng CS, Yeerken R, Chan YT, Fu Z, Zheng YC, Feng Y, Wang N. Traditional Chinese medicine for the treatment of cancers of hepatobiliary system: from clinical evidence to drug discovery. Mol Cancer 2024; 23:218. [PMID: 39354529 PMCID: PMC11443773 DOI: 10.1186/s12943-024-02136-2] [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: 08/10/2024] [Accepted: 09/20/2024] [Indexed: 10/03/2024] Open
Abstract
Hepatic, biliary, and pancreatic cancer pose significant challenges in the field of digestive system diseases due to their highly malignant nature. Traditional Chinese medicine (TCM) has gained attention as a potential therapeutic approach with long-standing use in China and well-recognized clinical benefits. In this review, we systematically summarized the clinical applications of TCM that have shown promising results in clinical trials in treating hepatic, biliary, and pancreatic cancer. We highlighted several commonly used TCM therapeutics with validated efficacy through rigorous clinical trials, including Huaier Granule, Huachansu, and Icaritin. The active compounds and their potential targets have been thoroughly elucidated to offer valuable insights into the potential of TCM for anti-cancer drug discovery. We emphasized the importance of further research to bridge the gap between TCM and modern oncology, facilitating the development of evidence-based TCM treatment for these challenging malignancies.
Collapse
Affiliation(s)
- Junyu Wu
- School of Chinese Medicine, the University of Hong Kong, 3, Sasson Road, Pokfulam, Hong Kong
| | - Guoyi Tang
- School of Chinese Medicine, the University of Hong Kong, 3, Sasson Road, Pokfulam, Hong Kong
| | - Chien-Shan Cheng
- Department of Digestive Endoscopy Center & Gastroenterology, Shuguang Hospital Affiliated With Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai, China
| | - Ranna Yeerken
- School of Chinese Medicine, the University of Hong Kong, 3, Sasson Road, Pokfulam, Hong Kong
| | - Yau-Tuen Chan
- School of Chinese Medicine, the University of Hong Kong, 3, Sasson Road, Pokfulam, Hong Kong
| | - Zhiwen Fu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi-Chao Zheng
- State Key Laboratory of Esophageal Cancer Prevention &, Treatment Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Yibin Feng
- School of Chinese Medicine, the University of Hong Kong, 3, Sasson Road, Pokfulam, Hong Kong.
| | - Ning Wang
- School of Chinese Medicine, the University of Hong Kong, 3, Sasson Road, Pokfulam, Hong Kong.
| |
Collapse
|
5
|
Boyd LN, Ali M, Puik JR, Comandatore A, Ginocchi L, Meijer LL, Swijnenburg RJ, Tartarini R, Le Large TY, Morelli L, Garajova I, Besselink MG, Mambrini A, Wilmink JW, Frampton AE, van Laarhoven HW, Giovannetti E, Kazemier G. Plasma miR-379 can predict treatment response to FOLFIRINOX and gemcitabine- nab-paclitaxel in advanced pancreatic cancer. THE JOURNAL OF LIQUID BIOPSY 2024; 5:100152. [PMID: 40027944 PMCID: PMC11863948 DOI: 10.1016/j.jlb.2024.100152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/21/2024] [Accepted: 03/23/2024] [Indexed: 03/05/2025]
Abstract
Background Novel biomarkers, such as plasma microRNAs (miRs), are needed to help guide clinical decision-making for the type of chemotherapy to use in patients with advanced pancreatic ductal adenocarcinoma (PDAC). This study assessed the ability of plasma miRs to predict optimal treatment response from FOLFIRINOX or gemcitabine-nab-paclitaxel in these patients. Methods Next-generation sequencing (NGS) was performed for biomarker discovery in pre-treatment plasma samples from advanced PDAC patients subsequently treated with FOLFIRINOX (n = 12) or gemcitabine-nab-paclitaxel (n = 12). Selected candidate biomarkers were validated in 40 patients with advanced PDAC using RT-qPCR. Cox regression was then used to assess the predictive value of plasma miRs for either FOLFIRINOX or gemcitabine-nab-paclitaxel. Results In the validation cohort, high plasma miR-379 expression was strongly predictive of treatment response (Pinteraction = 0.0004). Overall survival (OS) was significantly better with FOLFIRINOX vs. gemcitabine-nab-paclitaxel in those patients with lower plasma miR-379 expression (hazard ratio, 0.32 [95% confidence interval, 0.08 to 0.98]; P = 0.046). However, gemcitabine-nab-paclitaxel was associated with superior OS in patients with higher plasma miR-379 (hazard ratio, 0.28 [0.10 to 0.86]; P = 0.027). In contrast, miR-127, miR-155, and miR-200 showed no predictive value for treatment response for either chemotherapy regimen (P interaction = 0.12, P interaction = 0.83 and P interaction = 0.12, respectively). Conclusions Plasma miR-379 appears clinically useful as a predictive biomarker to identify which patients with advanced PDAC benefit most from treatment with FOLFIRINOX or gemcitabine-nab-paclitaxel. Further validation in larger studies and clinical trials is now warranted.
Collapse
Affiliation(s)
- Lenka N.C. Boyd
- Amsterdam UMC, Location Vrije Universiteit, Department of Surgery, Amsterdam, the Netherlands
- Amsterdam UMC, Location Vrije Universiteit, Department of Medical Oncology, Lab of Medical Oncology, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Mahsoem Ali
- Amsterdam UMC, Location Vrije Universiteit, Department of Surgery, Amsterdam, the Netherlands
- Amsterdam UMC, Location Vrije Universiteit, Department of Medical Oncology, Lab of Medical Oncology, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Jisce R. Puik
- Amsterdam UMC, Location Vrije Universiteit, Department of Surgery, Amsterdam, the Netherlands
- Amsterdam UMC, Location Vrije Universiteit, Department of Medical Oncology, Lab of Medical Oncology, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Annalisa Comandatore
- Amsterdam UMC, Location Vrije Universiteit, Department of Medical Oncology, Lab of Medical Oncology, Amsterdam, the Netherlands
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Laura Ginocchi
- Department of Oncology, Azienda Sanitaria Locale Toscana Nord Ovest, Massa Carrara Hospital, Massa, Italy
| | - Laura L. Meijer
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Rutger-Jan Swijnenburg
- Amsterdam UMC, Location Vrije Universiteit, Department of Surgery, Amsterdam, the Netherlands
- Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, the Netherlands
| | - Roberta Tartarini
- Department of Oncology, Azienda Sanitaria Locale Toscana Nord Ovest, Massa Carrara Hospital, Massa, Italy
| | - Tessa Y.S. Le Large
- Amsterdam UMC, Location Vrije Universiteit, Department of Surgery, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Luca Morelli
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Ingrid Garajova
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Marc G. Besselink
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Amsterdam UMC, Location University of Amsterdam, Department of Surgery, Amsterdam, the Netherlands
| | - Andrea Mambrini
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Johanna W. Wilmink
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Amsterdam UMC, Location University of Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands
| | - Adam E. Frampton
- HPB Surgical Unit, Royal Surrey County Hospital, Guildford, Surrey, UK
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, W120HS, London, UK
- Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, The Leggett Building, University of Surrey, Guildford, Surrey, GU2 7WG, UK
| | - Hanneke W.M. van Laarhoven
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Amsterdam UMC, Location University of Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands
| | - Elisa Giovannetti
- Amsterdam UMC, Location Vrije Universiteit, Department of Medical Oncology, Lab of Medical Oncology, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisa per la Scienza, Pisa, Italy
| | - Geert Kazemier
- Amsterdam UMC, Location Vrije Universiteit, Department of Surgery, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
6
|
Ying L, Li K, Chen C, Wang Y, Zhao Q, Wang Y, Xu L, Huang H, Song G, Li W, He X. OIP5-AS1 enhances the malignant characteristics and resistance to chemotherapy of pancreatic cancer cells by targeting miR-30d-5p/MARCH8. Heliyon 2024; 10:e33835. [PMID: 39050450 PMCID: PMC11268210 DOI: 10.1016/j.heliyon.2024.e33835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/27/2024] Open
Abstract
MARCH8, an E3 ubiquitin ligase, plays a crucial role in regulating various cellular processes such as protein degradation and signaling pathways and is implicated in the development and spread of pancreatic cancer. Analysis of pancreatic cancer tissues compared to adjacent normal tissues showed a decrease in miRNA-30d-5p levels and an increase in OIP5-AS1 and MARCH8 levels, as confirmed by qRT-PCR and Western blot analysis. The dual-luciferase reporter assay demonstrated a binding relationship between OIP5-AS1 and miRNA-30d-5p, as well as between miRNA-30d-5p and MARCH8 in PACN-1 cells, derived from a human pancreatic carcinoma specimen. Further investigations utilizing various assays revealed that OIP5-AS1 inhibited apoptosis and promoted cell proliferation, invasion, and migration in PACN-1 cells via the miRNA-30d-5p/MARCH8 axis in vitro. Tumor experiments in nude mice confirmed that OIP5-AS1 enhanced PACN-1 cell growth in vivo through the miRNA-30d-5p/MARCH8 axis. Additionally, OIP5-AS1 was found to activate downstream genes of the JAK-STAT pathway, namely IFNAR2, SOCS3, and JAK1, in PACN-1 cells. Furthermore, OIP5-AS1 increased the IC50 values for doxorubicin, gemcitabine, and cisplatin in PACN-1 cells, as determined by the Cell Counting Kit-8 assay. Overall, OIP5-AS1 was shown to promote aggressive traits and resistance to chemotherapy in PACN-1 cells through the miRNA-30d-5p/MARCH8 axis.
Collapse
Affiliation(s)
- Leilei Ying
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Kening Li
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Chao Chen
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ying Wang
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qing Zhao
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yaohui Wang
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Lichao Xu
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Haozhe Huang
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ge Song
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Wentao Li
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xinhong He
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| |
Collapse
|
7
|
Mahumud RA, Shahjalal M, Dahal PK, Mosharaf MP, Hoque ME, Wawryk O. Systemic therapy and radiotherapy related complications and subsequent hospitalisation rates: a systematic review. BMC Cancer 2024; 24:826. [PMID: 38987752 PMCID: PMC11238411 DOI: 10.1186/s12885-024-12560-8] [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: 11/14/2023] [Accepted: 06/24/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Hospitalisation resulting from complications of systemic therapy and radiotherapy places a substantial burden on the patient, society, and healthcare system. To formulate preventive strategies and enhance patient care, it is crucial to understand the connection between complications and the need for subsequent hospitalisation. This review aimed to assess the existing literature on complications related to systemic and radiotherapy treatments for cancer, and their impact on hospitalisation rates. METHODS Data was obtained via electronic searches of the PubMed, Scopus, Embase and Google Scholar online databases to select relevant peer-reviewed papers for studies published between January 1, 2000, and August 30, 2023. We searched for a combination of keywords in electronic databases and used a standard form to extract data from each article. The initial specific interest was to categorise the articles based on the aspects explored, especially complications due to systemic and radiotherapy and their impact on hospitalisation. The second interest was to examine the methodological quality of studies to accommodate the inherent heterogeneity. The study protocol was registered with PROSPERO (CRD42023462532). FINDINGS Of 3289 potential articles 25 were selected for inclusion with ~ 34 million patients. Among the selected articles 21 were cohort studies, three were randomised control trials (RCTs) and one study was cross-sectional design. Out of the 25 studies, 6 studies reported ≥ 10 complications, while 7 studies reported complications ranging from 6 to 10. Three studies reported on a single complication, 5 studies reported at least two complications but fewer than six, and 3 studies reported higher numbers of complications (≥ 15) compared with other selected studies. Among the reported complications, neutropenia, cardiac complications, vomiting, fever, and kidney/renal injury were the top-most. The severity of post-therapy complications varied depending on the type of therapy. Studies indicated that patients treated with combination therapy had a higher number of post-therapy complications across the selected studies. Twenty studies (80%) reported the overall rate of hospitalisation among patients. Seven studies revealed a hospitalisation rate of over 50% among cancer patients who had at least one complication. Furthermore, two studies reported a high hospitalisation rate (> 90%) attributed to therapy-repeated complications. CONCLUSION The burden of post-therapy complications is emerging across treatment modalities. Combination therapy is particularly associated with a higher number of post-therapy complications. Ongoing research and treatment strategies are imperative for mitigating the complications of cancer therapies and treatment procedures. Concurrently, healthcare reforms and enhancement are essential to address the elevated hospitalisation rates resulting from treatment-related complications in cancer patients.
Collapse
Affiliation(s)
- Rashidul Alam Mahumud
- NHMRC Clinical Trials Centre, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales (NSW), Australia
| | - Md Shahjalal
- Global Health Institute, North South University, Dhaka, Bangladesh.
| | - Padam Kanta Dahal
- School of Health, Medical and Applied Sciences, Central Queensland University, Sydney Campus, Sydney, Australia
| | - Md Parvez Mosharaf
- School of Business, Faculty of Business, Education, Law and Arts, University of Southern Queensland, Toowoomba, QLD, Australia
| | - Mohammad Enamul Hoque
- Faculty of Medicine and Health, The University of Western Australia, Perth, Australia
| | - Olivia Wawryk
- Department of General Practice, Victorian Comprehensive Cancer Centre, Data Connect, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
8
|
Hidig SM. An Overview of Pancreatic Cancer Diagnosis and Treatment in China: Current Landscape and Future Prospects. Niger Med J 2024; 65:387-397. [PMID: 39398405 PMCID: PMC11470284 DOI: 10.60787/nmj-v65i3-376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024] Open
Abstract
This comprehensive literature review is to summarize the most recent findings regarding the causes, diagnosis, and treatments of pancreatic cancer and to encourage additional investigation into this under-researched malignant tumor. Pancreatic cancer is a significant public health issue in China, with annual mortality rates almost equal to incidence rates. The disease is more prevalent in rural areas and has a poor prognosis. The data was collected from the following databases: Pub Med, Cross ref, Science Direct, Scopus, and Google Scholar we reviewed published articles from 2018 to 2023 on the annual incidence of pancreatic cancer in China is 5.1%, with only 5-7% of patients completely cured. The prognosis is extremely poor, with a 1-year survival rate of 8% and a 5-year survival rate of 3%. Pancreatic cancer has no specific clinical manifestations or tumor markers, and its characteristics are not typical of high-risk factors including smoking, alcohol, chronic pancreatitis, abnormal microorganism metabolism, blood type, and glucose and lipid levels. For increased detection and survival rates, pancreatic cancer must be diagnosed as early as possible. However, the low specificity of tumor markers calls for more study. Future treatment strategies could include immunotherapy and a microbiology-free system, and it's anticipated that they'll offer intriguing clinical applications for extending patients' lives with pancreatic cancer. Finally, we suggest measures to improve the health outcomes of pancreatic cancer patients in China.
Collapse
Affiliation(s)
- Sakarie Mustafe Hidig
- Department of Hepatobiliary and Pancreatic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang Province, 322000, PR. China
| |
Collapse
|
9
|
Badheeb AM, Awad MA, Al Masad AG, Alyami MS, Fagihi MA, Al Walani M, Alkarak S, Al Bahili HM, Alatawi A, Nagi NM, Madbouly AR, Abu Bakar A, Ahmed F, Badheeb M. Pancreatic Cancer: A Retrospective Study From the Najran Region of Saudi Arabia. Cureus 2024; 16:e65685. [PMID: 39205701 PMCID: PMC11357715 DOI: 10.7759/cureus.65685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Despite advances in treatment, pancreatic cancer frequently has a low survival rate due to its advanced-stage diagnosis. Treatment focuses on prolonging survival and maintaining quality of life. This study investigates the characteristics associated with survival in advanced pancreatic cancer patients treated at a single academic cancer center in Najran, Saudi Arabia. METHOD A retrospective chart review study covering the period January 1, 2015, and December 31, 2023, involved 80 adult patients with pathologically confirmed pancreatic cancer (ductal adenocarcinoma) at King Khalid Hospital in Najran, Saudi Arabia. Clinicopathological characteristics, therapy, response, and survival outcomes were all gathered and analyzed. The chi-squared test, Kaplan-Meier, and Cox proportional hazards method with hazard ratios (HR) and 95% confidence intervals (CI) were used for statistical analysis. RESULT The mean age was 65.7±14.1 years and 54 (67.5%) cases were male. The main symptom was abdominal pain (n=54, 67.5%), while jaundice was presented in 17 (21.2%) of cases. The baseline serum carbohydrate antigen 19-9 (CA 19-9) level varied among cases, with 35 (43.8%) having normal levels. The majority of cases (n=59, 73.8%) had distant metastases at the initial presentation, while 12 cases (15%) had localized disease (resectable), and 22 (27.5%) were locally advanced at the first presentation. The most commonly reported pathologic grade was poorly differentiated ductal adenocarcinoma in 39 (48.8%). FOLFIRINOX was used as first-line chemotherapy in 54 (67.5%) cases, while gemcitabine alone was used in 15 (18.8%) cases. First-line chemotherapy resulted in progressive disease in 30 (37.5%), stable disease in 30 (37.5%), and partial response in 14 (17.5%). With a mean follow-up time of 14.8±8.6 months, 57 (71.2%) were dead, where the main cause of death was disease progression (n=51, 89.5%). The median overall survival was 13.5 months, with a 12-month survival rate of 56% and a 36-month survival rate of 17%. The median cancer-specific survival was 16 months (95% CI: 13-22 months). The 12-month median cancer-specific survival was 61% (95% CI: 51-73%), and the 36-month median cancer-specific survival was 19% (95% CI: 10-34%). In univariate analysis, initial metastasis presentation (HR: 35.46; 95% CI: 4.90-256.83, p<0.001), poor Eastern Cooperative Oncology Group Performance Status (ECOG-PS) (3-4) (HR: 2.34; 95% CI:1.34-4.09, p=0.003), and presence of multiple metastases (HR: 1.33; 95% CI: 1.09-1.62, p=0.004) were associated with worsened survival. Patients who received the first chemotherapy were associated with better survival (HR: 0.53; 95% CI: 0.29-0.98, p=0.043). Furthermore, the response rate in patients who received FOLFIRINOX was better than that of those who received gemcitabine alone, which was statistically significant (p=0.002). CONCLUSION Our study showed that initial metastatic presentation, poor ECOG-PS, and the occurrence of numerous metastases were all linked with poor survival of patients with pancreatic adenocarcinoma. Additionally, FOLFIRINOX as a first-line treatment showed better survival rates than gemcitabine alone. Raising awareness among healthcare providers on the alarming signs of pancreatic cancer and the introduction of personalized oncology might improve the outcome of this fatal malignancy.
Collapse
Affiliation(s)
- Ahmed M Badheeb
- Oncology, King Khalid Hospital-Oncology Center, Najran, SAU
- Medicine, Hadhramaut University, Mukalla, YEM
| | | | - Ali G Al Masad
- Gastroenterology and Hepatology, King Khalid Hospital, Najran, SAU
| | | | | | - Mugahed Al Walani
- Gastroenterology and Advanced Endoscopy, King Khalid Hospital, Najran, SAU
| | | | - Hamad M Al Bahili
- Hepatobiliary Surgery, Prince Sultan Military Medical City, Riyadh, SAU
| | | | | | - Ahmed R Madbouly
- Gastroenterology and Hepatology, King Khalid Hospital, Najran, SAU
| | | | | | - Mohamed Badheeb
- Internal Medicine, Yale New Haven Health, Bridgeport Hospital, Bridgeport, USA
| |
Collapse
|
10
|
Cao Y, Yu D, Wu Y, Zhu W. Regional intra-arterial vs. systemic chemotherapy for the treatment of advanced pancreatic cancer: a systematic review and meta-analysis. Front Oncol 2024; 14:1197424. [PMID: 38651152 PMCID: PMC11033438 DOI: 10.3389/fonc.2024.1197424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 03/11/2024] [Indexed: 04/25/2024] Open
Abstract
Introduction Pancreatic cancer is a highly aggressive malignancy with limited response to chemotherapy. This research aims to compare the effectiveness and safety of regional intra-arterial chemotherapy (RIAC) with conventional systemic chemotherapy in treating advanced stages of pancreatic cancer. Methods A comprehensive literature review was conducted using databases such as PubMed, Embase, Web of Science, and the Cochrane Library. Studies assessing the comparative outcomes of RIAC and systemic chemotherapy were included. Data extraction and quality evaluation were performed independently by two researchers. Statistical analysis was conducted using STATA16 software, calculating odds ratios (OR), risk differences (RD), and 95% confidence intervals (CI). Results Eleven studies, comprising a total of 627 patients, were included in the meta-analysis. The findings showed that patients undergoing RIAC had significantly higher rates of partial remission (PR) compared to those receiving systemic chemotherapy (OR = 2.23, 95% CI: 1.57, 3.15, I2= 0%). Additionally, the rate of complications was lower in the RIAC group (OR = 0.45, 95% CI: 0.33, 0.63, I2= 0%). Moreover, patients treated with RIAC had notably longer median survival times. Discussion The results of this research indicate that RIAC is associated with a higher rate of partial remission, improved clinical benefits, and fewer complications compared to systemic chemotherapy in the management of advanced pancreatic cancer. These findings suggest that RIAC may be a more effective and safer treatment option for patients with advanced stages of pancreatic cancer. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42023404637.
Collapse
Affiliation(s)
| | - Dedong Yu
- Department of Oncology, Baotou Central Hospital, Baotou, China
| | | | - Wei Zhu
- Department of Oncology, Baotou Central Hospital, Baotou, China
| |
Collapse
|
11
|
Awais N, Satnarine T, Ahmed A, Haq A, Patel D, Hernandez GN, Seffah KD, Zaman MA, Khan S. A Systematic Review of Chemotherapeutic Regimens Used in Pancreatic Cancer. Cureus 2023; 15:e46630. [PMID: 37937003 PMCID: PMC10627412 DOI: 10.7759/cureus.46630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 10/06/2023] [Indexed: 11/09/2023] Open
Abstract
Pancreatic cancer is a malignant tumor with one of the worst prognosis. Its incidence has been on the rise in recent years. First-line and second-line treatments as well as adjuvant therapies have been employed in clinical trials for pancreatic cancer along with traditional chemotherapy and radiotherapy that has been enhanced. The prognosis of pancreatic ductal adenocarcinoma (PDAC) is still quite bad despite recent improvements in diagnostic and treatment methods. Since most patients are not candidates for treatment with a curative purpose, effective palliative care is crucial. For this systematic review, between December 25, 2022, and January 5, 2023, we searched PubMed, Medline, Cochrane, and Science Direct and discovered 225 relevant articles. The appropriateness of the literature abstracts for the pooled analysis was evaluated using different combinations of keywords such as pancreatic cancer, first- and second-line chemotherapy, palliative chemotherapy, gemcitabine and nab-paclitaxel (GnP), FOLFIRINOX (FFX), and fluorouracil. Eight research studies with a total of 15,236 people, including systematic reviews, meta-analyses, and randomized controlled trials (RCTs), were included. The only treatment of choice for patients without metastatic disease who have clinical staging that suggests resectable or borderline resectable pancreatic cancer (BRPC) should be resection. This research examined how first- and second-line chemotherapeutic regimens (using different drug combinations) affected patients with locally advanced pancreatic cancer (LAPC) or BRPC and how they responded in terms of overall survival (OS), tumor resectability, and progression-free interval. The review concludes by highlighting the results of these therapies. Notably, a growing body of research indicates that the two most popular first-line medication combinations GnP and FFX have similar results in RCTs and in real-world populations. Results of second-line therapy after first-line regime failure are still dismal, and there is still a great deal of doubt regarding the best course of action. More RCTs and real-world evidence studies that address current and innovative regimens, as well as the best order in which to administer them, are required, with a greater emphasis on targeted therapy with fewer side effects.
Collapse
Affiliation(s)
- Nimra Awais
- Research, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Travis Satnarine
- Pediatrics, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Areeg Ahmed
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Ayesha Haq
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Deepkumar Patel
- Family Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Grethel N Hernandez
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Kofi D Seffah
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
- Internal Medicine, Piedmont Athens Regional, Athens, USA
| | - Mustafa Abrar Zaman
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
- Internal Medicine, St. George's University School of Medicine, Newcastle Upon Tyne, GBR
| | - Safeera Khan
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| |
Collapse
|
12
|
Segoviano-Ramirez JC, Esparza-Rodriguez N, Carcano-Diaz K, Diaz-Perez RN, Palma-Nicolas JP, Hernandez-Bello R, Garcia-Juarez J. Structural and functional integrity of endocrine pancreas post administration of Karwinskia humboldtiana fruit to Wistar rats: a possible therapeutic application for cancer of exocrine origin. Histol Histopathol 2023; 38:989-997. [PMID: 36896890 DOI: 10.14670/hh-18-603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
AIMS Pancreatic adenocarcinoma represents a therapeutic challenge due to the high toxicity of antineoplastic treatments and secondary effects of pancreatectomy. T-514, a toxin isolated from Karwinskia humboldtiana (Kh) has shown antineoplastic activity on cell lines. In acute intoxication with Kh, we reported apoptosis on the exocrine portion of pancreas. One of the mechanisms of antineoplastic agents is the induction of apoptosis, therefore our main objective was to evidence structural and functional integrity of the islets of Langerhans after the administration of Kh fruit in Wistar rats. METHODS TUNEL assay and immunolabelling against activated caspase-3 were used to detect apoptosis. Also, immunohistochemical tests were performed to search for glucagon and insulin. Serum amylase enzyme activity was also quantified as a molecular marker of pancreatic damage. RESULTS Evidence of toxicity on the exocrine portion, by positivity in the TUNEL assay and activated caspase-3, was found. On the contrary, the endocrine portion remained structurally and functionally intact, without apoptosis, and presenting positivity in the identification of glucagon and insulin. CONCLUSIONS These results demonstrated that Kh fruit induces selective toxicity on the exocrine portion and establish a precedent to evaluate T-514 as a potential treatment against pancreatic adenocarcinoma without affecting the islets of Langerhans.
Collapse
Affiliation(s)
- Juan Carlos Segoviano-Ramirez
- Center for Research and Development in Health Sciences, Bioimaging Unit, Autonomous University of Nuevo Leon (UANL), México
- Department of Pathology, Faculty of Medicine, Autonomous University of Nuevo Leon (UANL), México
| | - Nallely Esparza-Rodriguez
- Department of Histology, Faculty of Medicine, Autonomous University of Nuevo Leon (UANL), Madero y Dr. Aguirre Pequeño, Mitras Centro, México
- General Directorate of Quality and Health Education (DGCES), Secretary of Health, México City, México
| | | | - Rosa Nelly Diaz-Perez
- Department of Histology, Faculty of Medicine, Autonomous University of Nuevo Leon (UANL), Madero y Dr. Aguirre Pequeño, Mitras Centro, México
| | | | - Romel Hernandez-Bello
- Department of Microbiology, Faculty of Medicine, Autonomous University of Nuevo Leon (UANL), México
| | - Jaime Garcia-Juarez
- Department of Histology, Faculty of Medicine, Autonomous University of Nuevo Leon (UANL), Madero y Dr. Aguirre Pequeño, Mitras Centro, México
- Center for Research and Development in Health Sciences, Bioimaging Unit, Autonomous University of Nuevo Leon (UANL), Gonzalitos y Dr. Carlos Canseco, Mitras Centro, México.
| |
Collapse
|
13
|
Fiorentini G, Sarti D, Mambrini A, Hammarberg Ferri I, Bonucci M, Sciacca PG, Ballerini M, Bonanno S, Milandri C, Nani R, Guadagni S, Dentico P, Fiorentini C. Hyperthermia combined with chemotherapy vs chemotherapy in patients with advanced pancreatic cancer: A multicenter retrospective observational comparative study. World J Clin Oncol 2023; 14:215-226. [PMID: 37398545 PMCID: PMC10311475 DOI: 10.5306/wjco.v14.i6.215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/18/2023] [Accepted: 06/06/2023] [Indexed: 06/21/2023] Open
Abstract
BACKGROUND Several studies report the useful therapeutic results of regional hyperthermia in association with chemotherapy (CHT) and radiotherapy for the treatment of pancreatic cancer. Modulated electro-hyperthermia (mEHT) is a new hyperthermia technique that induces immunogenic death or apoptosis of pancreatic cancer cells in laboratory experiments and increases tumor response rate and survival in pancreatic cancer patients, offering beneficial therapeutic effects against this severe type of cancer. AIM To assess survival, tumor response and toxicity of mEHT alone or combined with CHT compared with CHT for the treatment of locally advanced or metastatic pancreatic cancer. METHODS This was a retrospective data collection on patients affected by locally advanced or metastatic pancreatic cancer (stage III and IV) performed in 9 Italian centers, members of International Clinical Hyperthermia Society-Italian Network. This study included 217 patients, 128 (59%) of them were treated with CHT (no-mEHT) and 89 (41%) patients received mEHT alone or in association with CHT. mEHT treatments were performed applying a power of 60-150 watts for 40-90 min, simultaneously or within 72 h of administration of CHT. RESULTS Median patients' age was 67 years (range 31-92 years). mEHT group had a median overall survival greater than non-mEHT group (20 mo, range 1.6-24, vs 9 mo, range 0.4-56.25, P < 0.001). mEHT group showed a higher number of partial responses (45% vs 24%, P = 0.0018) and a lower number of progressions (4% vs 31%, P < 0.001) than the no-mEHT group, at the three months follow-up. Adverse events were observed as mild skin burns in 2.6% of mEHT sessions. CONCLUSION mEHT seems safe and has beneficial effects on survival and tumor response of stage III-IV pancreatic tumor treatment. Further randomized studies are warranted to confirm or not these results.
Collapse
Affiliation(s)
- Giammaria Fiorentini
- Integrative Oncology, Integrative Oncology Outpatient Clinic, Bologna 40121, Italy
| | - Donatella Sarti
- Department of Oncology, Santa Maria della Misericordia Hospital, Urbino 60129, Italy
| | - Andrea Mambrini
- Department of Oncology, Azienda Sanitaria Locale Toscana Nord Ovest, Massa Carrara Hospital, Massa 54100, Italy
| | | | - Massimo Bonucci
- Integrative Oncology, Association Research Center for Integrative Oncology Treatments, Roma 00166, Italy
| | | | - Marco Ballerini
- Hyperthermia Unit, Bellessere Medical Center, Terni 05100, Italy
| | | | - Carlo Milandri
- Medical Oncology, San Donato Hospital, Arezzo 52100, Italy
| | - Roberto Nani
- Interventional Radiology Unit, Humanitas Gavazzeni, Bergamo 24121, Italy
| | - Stefano Guadagni
- Applied Clinical Sciences and Biotechnology, Section of General Surgery, University of L'Aquila, L'Aquila 67100, Italy
| | - Patrizia Dentico
- Hyperthermia Service, Medical Oncology Unit, San Giuseppe Hospital, Empoli 50053, Italy
| | - Caterina Fiorentini
- Prevention and Sports Medicine, University Hospital Klinikum rechts der Isar, Technical University of Munich, Munich 80331, Germany
| |
Collapse
|
14
|
Yao W, Chen X, Fan B, Zeng L, Zhou Z, Mao Z, Shen Q. Multidisciplinary team diagnosis and treatment of pancreatic cancer: Current landscape and future prospects. Front Oncol 2023; 13:1077605. [PMID: 37007078 PMCID: PMC10050556 DOI: 10.3389/fonc.2023.1077605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 03/03/2023] [Indexed: 03/17/2023] Open
Abstract
The pathogenesis of pancreatic cancer has not been completely clear, there is no highly sensitive and specific detection method, so early diagnosis is very difficult. Despite the rapid development of tumor diagnosis and treatment, it is difficult to break through in the short term and the overall 5-year survival rate of pancreatic cancer is less than 8%. In the face of the increasing incidence of pancreatic cancer, in addition to strengthening basic research, exploring its etiology and pathogenesis, it is urgent to optimize the existing diagnosis and treatment methods through standard multidisciplinary team (MDT), and formulate personalized treatment plan to achieve the purpose of improving the curative effect. However, there are some problems in MDT, such as insufficient understanding and enthusiasm of some doctors, failure to operate MDT according to the system, lack of good communication between domestic and foreign peers, and lack of attention in personnel training and talent echelon construction. It is expected to protect the rights and interests of doctors in the future and ensure the continuous operation of MDT. To strengthen the research on the diagnosis and treatment of pancreatic cancer, MDT can try the Internet +MDT mode to improve the efficiency of MDT.
Collapse
Affiliation(s)
- Weirong Yao
- Department of Oncology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Xiaoliang Chen
- Department of Hepatobiliary Surgery, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Bin Fan
- Department of Radiology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Lin Zeng
- Department of Oncology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Zhiyong Zhou
- Department of Oncology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Zhifang Mao
- Department of Oncology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Qinglin Shen
- Department of Oncology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- Institute of Clinical Medicine, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- *Correspondence: Qinglin Shen,
| |
Collapse
|
15
|
Zhao Y, Zheng Y, Zhu Y, Ding K, Zhou M, Liu T. Co-delivery of gemcitabine and Triapine by calcium carbonate nanoparticles against chemoresistant pancreatic cancer. Int J Pharm 2023; 636:122844. [PMID: 36925025 DOI: 10.1016/j.ijpharm.2023.122844] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 02/14/2023] [Accepted: 03/11/2023] [Indexed: 03/16/2023]
Abstract
Pancreatic cancer is a malignant disease with high mortality, and its systemic treatment strategy mainly focuses on chemotherapy. Yet, the overall prognosis of pancreatic cancer patients is still extremely poor with a low survival rate. Gemcitabine (GEM) is a widely used chemotherapeutic agent for the treatment of pancreatic cancer. However, GEM chemoresistance remains the major challenge. In this study, we prepared calcium carbonate nanoparticles (CaCO3 NPs) loaded with a nucleotide reductase inhibitor (Triapine) and GEM to suppress the GEM resistance of pancreatic cancer cells (PANC-1/GEM) and solve the problem of poor solubility of Triapine. CaCO3-GEM-Triapine NPs nano-formulations enhanced the therapeutic effect of GEM-based chemotherapy by inhibiting cancer cell proliferation, migration, and resistance to GEM using both 2D PANC-1/GEM cells and 3D tumor spheroids. The study indicated that CaCO3 NPs loaded with GEM and Triapine could provide an effective treatment option to overcome drug resistance in pancreatic cancer.
Collapse
Affiliation(s)
- Yongmei Zhao
- School of Pharmacy, Nantong University, Nantong, China
| | - Yuanlin Zheng
- School of Pharmacy, Nantong University, Nantong, China
| | - Yan Zhu
- School of Pharmacy, Nantong University, Nantong, China
| | - Kai Ding
- School of Pharmacy, Nantong University, Nantong, China
| | - Mengjiao Zhou
- School of Pharmacy, Nantong University, Nantong, China.
| | - Tianqing Liu
- NICM Health Research Institute, Western Sydney University, Westmead, Australia.
| |
Collapse
|
16
|
Shetu SA, James N, Rivera G, Bandyopadhyay D. Molecular Research in Pancreatic Cancer: Small Molecule Inhibitors, Their Mechanistic Pathways and Beyond. Curr Issues Mol Biol 2023; 45:1914-1949. [PMID: 36975494 PMCID: PMC10047141 DOI: 10.3390/cimb45030124] [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: 01/01/2023] [Revised: 02/06/2023] [Accepted: 02/21/2023] [Indexed: 03/04/2023] Open
Abstract
Pancreatic enzymes assist metabolic digestion, and hormones like insulin and glucagon play a critical role in maintaining our blood sugar levels. A malignant pancreas is incapable of doing its regular functions, which results in a health catastrophe. To date, there is no effective biomarker to detect early-stage pancreatic cancer, which makes pancreatic cancer the cancer with the highest mortality rate of all cancer types. Primarily, mutations of the KRAS, CDKN2A, TP53, and SMAD4 genes are responsible for pancreatic cancer, of which mutations of the KRAS gene are present in more than 80% of pancreatic cancer cases. Accordingly, there is a desperate need to develop effective inhibitors of the proteins that are responsible for the proliferation, propagation, regulation, invasion, angiogenesis, and metastasis of pancreatic cancer. This article discusses the effectiveness and mode of action at the molecular level of a wide range of small molecule inhibitors that include pharmaceutically privileged molecules, compounds under clinical trials, and commercial drugs. Both natural and synthetic small molecule inhibitors have been counted. Anti-pancreatic cancer activity and related benefits of using single and combined therapy have been discussed separately. This article sheds light on the scenario, constraints, and future aspects of various small molecule inhibitors for treating pancreatic cancer-the most dreadful cancer so far.
Collapse
Affiliation(s)
- Shaila A. Shetu
- Department of Chemistry, The University of Texas Rio Grande Valley, 1201 West University Drive, Edinburg, TX 78539, USA
| | - Nneoma James
- Department of Chemistry, The University of Texas Rio Grande Valley, 1201 West University Drive, Edinburg, TX 78539, USA
| | - Gildardo Rivera
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico
| | - Debasish Bandyopadhyay
- Department of Chemistry, The University of Texas Rio Grande Valley, 1201 West University Drive, Edinburg, TX 78539, USA
- School of Earth Environment & Marine Sciences (SEEMS), The University of Texas Rio Grande Valley, 1201 West University Drive, Edinburg, TX 78539, USA
| |
Collapse
|
17
|
Gorris M, van Huijgevoort NCM, Fockens P, Meijer SL, Verheij J, Voermans RP, van Wanrooij RLJ, Lekkerkerker SJ, van Hooft JE. Comparison of two intraductal brush cytology devices for suspected malignant biliary strictures: randomized controlled trial. Surg Endosc 2023:10.1007/s00464-023-09916-9. [PMID: 36849566 DOI: 10.1007/s00464-023-09916-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 01/28/2023] [Indexed: 03/01/2023]
Abstract
BACKGROUND Endoscopic retrograde cholangiopancreatography (ERCP) with biliary brush cytology is commonly used to diagnose malignant pancreatobiliary strictures. This trial compared the sensitivity of two intraductal brush cytology devices. METHODS A randomized controlled trial in which consecutive patients with suspected malignant, extrahepatic biliary strictures were randomized (1:1) to a dense or conventional brush cytology device. Primary endpoint was sensitivity. Interim analysis was conducted after 50% of the patients completed follow-up. Results were interpreted by a data safety monitoring board. RESULTS Between June 2016 and June 2021, 64 patients were randomized to the dense (27 patients, 42%) or conventional brush (37 patients, 58%). Malignancy was diagnosed in 60 patients (94%) and benign disease in 4 patients (6%). Diagnoses were confirmed by histopathology in 34 patients (53%), cytopathology in 24 patients (38%), and clinical or radiological follow up in 6 patients (9%). Sensitivity of the dense brush was 50%, compared to 44% for the conventional brush (p = 0·785). DISCUSSION The results of this randomized controlled trial showed that the sensitivity of a dense brush is not superior to a conventional brush for diagnosing malignant extrahepatic pancreatobiliary strictures. This trial was prematurely ended for reasons of futility. TRIAL REGISTRATION Netherlands Trial Register number; NTR5458.
Collapse
Affiliation(s)
- Myrte Gorris
- Gastroenterology and Hepatology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Nadine C M van Huijgevoort
- Gastroenterology and Hepatology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Paul Fockens
- Gastroenterology and Hepatology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Gastroenterology and Hepatology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Sybren L Meijer
- Department of Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Joanne Verheij
- Department of Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Rogier P Voermans
- Gastroenterology and Hepatology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Roy L J van Wanrooij
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Gastroenterology and Hepatology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Selma J Lekkerkerker
- Gastroenterology and Hepatology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Jeanin E van Hooft
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
18
|
Salazar J, Bracchiglione J, Acosta-Dighero R, Meza N, Meade AG, Quintana MJ, Requeijo C, Rodríguez-Grijalva G, Santero M, Selva A, Solà I, Bonfill X. Systemic oncological treatments in patients with advanced pancreatic cancer: a scoping review and evidence map. Support Care Cancer 2023; 31:100. [PMID: 36622453 PMCID: PMC9829581 DOI: 10.1007/s00520-022-07564-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/22/2022] [Indexed: 01/10/2023]
Abstract
PURPOSE To identify, describe, and organise currently available evidence regarding systemic oncological treatments (SOTs) (chemotherapy, targeted/biological therapies, and immunotherapy) compared to best supportive care (BSC) for patients with advanced pancreatic cancer (PC). METHODS We conducted a scoping review and evidence mapping, adhering to PRISMA-ScR checklist. We searched MEDLINE, EMBASE, Cochrane Library, Epistemonikos, PROSPERO, and clinicaltrials.gov for eligible studies. We included systematic reviews (SRs), randomised controlled trials (RCTs), quasi-experimental, and observational studies evaluating SOTs compared to BSC or no treatment in patients with advanced PC. Two independent reviewers performed the screening process and data extraction. We developed evidence maps as an interactive visualization display, including the assessed interventions and outcomes. RESULTS Of the 50,601 records obtained from our search, we included 43 studies: 2 SRs, 16 RCTs, 4 quasi-experimental studies, 20 observational studies, and 1 protocol for a quasi-experimental study. Forty-two studies reported survival-related outcomes and most favoured SOTs, while five reported toxicity and most favoured BSC. Other patient-centred outcomes, such as quality of life, were scarcely reported. CONCLUSIONS This study highlights the current evidence gaps in studies assessing treatments for patients with advanced PC, mainly the lack of reports of non-survival-related outcomes, pointing out research areas that need further attention to make better recommendations for these patients.
Collapse
Affiliation(s)
- Josefina Salazar
- Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Javier Bracchiglione
- Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
- Interdisciplinary Centre for Health Studies (CIESAL), Universidad de Valparaíso, Viña del Mar, Chile
- CIBER Epidemiología Y Salud Pública (CIBERESP), Barcelona, Spain
| | - Roberto Acosta-Dighero
- Interdisciplinary Centre for Health Studies (CIESAL), Universidad de Valparaíso, Viña del Mar, Chile
| | - Nicolas Meza
- Interdisciplinary Centre for Health Studies (CIESAL), Universidad de Valparaíso, Viña del Mar, Chile
| | - Adriana-G Meade
- Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - María Jesús Quintana
- Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
- Universitat Autònoma Barcelona, Barcelona, Spain
| | - Carolina Requeijo
- Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | | | - Marilina Santero
- Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Anna Selva
- Universitat Autònoma Barcelona, Barcelona, Spain
- Corporació Sanitària Parc Taulí, Sabadell, Spain
| | - Ivan Solà
- Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
- CIBER Epidemiología Y Salud Pública (CIBERESP), Barcelona, Spain
- Universitat Autònoma Barcelona, Barcelona, Spain
| | - Xavier Bonfill
- Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain.
- CIBER Epidemiología Y Salud Pública (CIBERESP), Barcelona, Spain.
- Universitat Autònoma Barcelona, Barcelona, Spain.
| |
Collapse
|
19
|
Cai W, Zhu Y, Teng Z, Li D, Feng Q, Jiang Z, Cong R, Chen Z, Liu S, Zhao X, Ma X. Combined CT and serum CA19-9 for stratifying risk for progression in patients with locally advanced pancreatic cancer receiving intraoperative radiotherapy. Front Oncol 2023; 13:1155555. [PMID: 37124483 PMCID: PMC10140514 DOI: 10.3389/fonc.2023.1155555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/27/2023] [Indexed: 05/02/2023] Open
Abstract
Background and purpose The aim of this study was to evaluate the significance of baseline computed tomography (CT) imaging features and carbohydrate antigen 19-9 (CA19-9) in predicting prognosis of locally advanced pancreatic cancer (LAPC) receiving intraoperative radiotherapy (IORT) and to establish a progression risk nomogram that helps to identify the potential beneficiary of IORT. Methods A total of 88 LAPC patients with IORT as their initial treatment were enrolled retrospectively. Clinical data and CT imaging features were analyzed. Cox regression analyses were performed to identify the independent risk factors for progression-free survival (PFS) and to establish a nomogram. A risk-score was calculated by the coefficients of the regression model to stratify the risk of progression. Results Multivariate analyses revealed that relative enhanced value in portal-venous phase (REV-PVP), peripancreatic fat infiltration, necrosis, and CA19-9 were significantly associated with PFS (all p < 0.05). The nomogram was constructed according to the above variables and showed a good performance in predicting the risk of progression with a concordance index (C-index) of 0.779. Our nomogram stratified patients with LAPC into low- and high-risk groups with distinct differences in progression after IORT (p < 0.001). Conclusion The integrated nomogram would help clinicians to identify appropriate patients who might benefit from IORT before treatment and to adapt an individualized treatment strategy.
Collapse
Affiliation(s)
- Wei Cai
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongjian Zhu
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ze Teng
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dengfeng Li
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qinfu Feng
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhichao Jiang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rong Cong
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhaowei Chen
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Siyun Liu
- Magnetic Resonance Imaging Research, General Electric Healthcare (China), Beijing, China
| | - Xinming Zhao
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaohong Ma
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Xiaohong Ma,
| |
Collapse
|
20
|
Sánchez Y, Concepción ML, Amador Y, Piriz A, Rabassa R, Leyva A, Arguelles O, Leblanch L, Moret S, Rivero G, Vasallo AL, Martorell B, Guerra PP, Valls AR, Sánchez L, Saumell Y. Nimotuzumab Concurrent with Gemcitabine as First-Line Treatment of Locally Advanced or Metastatic Pancreatic Adenocarcinoma. BIOMED RESEARCH INTERNATIONAL 2023; 2023:1496072. [PMID: 37152586 PMCID: PMC10162878 DOI: 10.1155/2023/1496072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 02/11/2023] [Accepted: 02/15/2023] [Indexed: 05/09/2023]
Abstract
Background Nimotuzumab exerts its antitumor effect (mainly antiproliferative, proapoptotic, and antiangiogenic) by blocking the epidermal growth factor receptor overexpressing between 30 and 95% in pancreatic tumors cells. Methods A prospective, nonrandomized, uncontrolled, open-label, and multicenter clinical trial was conducted to evaluate the safety and effectiveness of nimotuzumab combined with gemcitabine as first-line treatment in unresectable locally advanced or metastatic pancreatic tumors in a real-world condition. Adverse events, their intensity, severity, and causality were determined using the Common Terminology Criteria for Adverse Events (CTCAE, version 4.0). Median overall survival, median progression-free survival, and 1- and 2-year survival rates were determined by using the Kaplan-Meier. Results 69 patients were included. The proportion of related serious adverse events was 1.2%. The most frequent adverse events were nausea (10%), anemia (8%), and abdominal pain (8%). Objective response was achieved in 18.5% of the patients and disease control in 43.1%. Patients with locally advanced disease achieved a median overall survival of 16.36 months (95% CI; 14.35-18.38); 1- and 2-year survival rates of 72.2 and 29.2 months, respectively; a median progression-free survival of 9.6 months (95% CI; 4.91-14.20); and a 1-year progression-free survival rate of 39%. Patients with metastatic disease achieved a median survival of 6.23 months (95% CI; 4.32-8.13); 1- and 2-year survival rates of 18.1 and 3.0 months, respectively; a median progression-free survival of 7.6 months (95% CI; 6.08-9.90); and 1- and 2-year PFS rates of 20.5 and 5.1 months, respectively. Conclusions Nimotuzumab combined with gemcitabine represents a safe and effective first-line treatment option for patients with advanced pancreatic adenocarcinoma in real-world conditions. Survival benefits were increased in those patients who received 8 or more doses of nimotuzumab. This trial is registered with RPCEC00000245 in the Cuban Registry of Clinical Trials, part of the World Health Organization's International Clinical Trials Registry Platform (ICTRP).
Collapse
Affiliation(s)
- Yamirka Sánchez
- Oncology Department, III Congreso Hospital, Pinar del Río 20100, Cuba
| | | | - Yohan Amador
- Oncology Department, Faustino Pérez Hospital, Matanzas 40100, Cuba
| | - Angel Piriz
- Oncology Department, Agostinho Neto Hospital, Guantánamo 85100, Cuba
| | - René Rabassa
- Oncology Department, Manuel Ascunce Domenech Hospital, Camagüey 70100, Cuba
| | - Ariel Leyva
- Oncology Department, Vladimir Ilich Lenin Hospital, Holguín 80100, Cuba
| | - Odalys Arguelles
- Oncology Department, Antonio Luaces Iraola Hospital, Ciego de Ávila 65200, Cuba
| | - Lisett Leblanch
- Oncology Department, Carlos Manuel de Céspedes Hospital, Granma 85100, Cuba
| | - Sheyla Moret
- Oncology Department, Medical and Surgical Research Center, Havana 11600, Cuba
| | - Gilberto Rivero
- Oncology Department, Ernesto Guevara de la Serna Hospital, Las Tunas 75100, Cuba
| | - Ana L. Vasallo
- Oncology Department, Gustavo Aldereguía Lima Hospital, Cienfuegos 55100, Cuba
| | - Beatriz Martorell
- Oncology Department, Saturnino Lora Hospital, Santiago de Cuba 90500, Cuba
| | - Pedro P. Guerra
- Clinical Trial Department, National Coordinating Center of Clinical Trials, Havana 11600, Cuba
| | - Ana R. Valls
- Clinical Trial Department, Center of Molecular Immunology, Havana 11600, Cuba
| | - Lisset Sánchez
- Clinical Trial Department, Center of Molecular Immunology, Havana 11600, Cuba
| | - Yaimarelis Saumell
- Clinical Trial Department, Center of Molecular Immunology, Havana 11600, Cuba
| |
Collapse
|
21
|
Muacevic A, Adler JR, Arjani S, Quinn PL, Trivedi R, Chokshi RJ. Staging CT and Diagnostic Laparoscopy With Cytology Prior to the Treatment of Pancreatic Adenocarcinoma: A Case Series. Cureus 2022; 14:e31883. [PMID: 36579218 PMCID: PMC9792353 DOI: 10.7759/cureus.31883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2022] [Indexed: 11/27/2022] Open
Abstract
INTRODUCTION Initial staging of pancreatic ductal adenocarcinoma (PDAC) is performed with computed tomography (CT). Laparoscopy with peritoneal cytology at staging can uncover occult disease undetected by CT. This case series assessed clinical course following staging laparoscopy with cytology in patients with PDAC. METHODS This single-center study examined patients with non-metastatic PDAC diagnosed from 2017 to 2020. Patients underwent CT and subsequent laparoscopy with cytology prior to treatment. Demographics, clinicopathologic status, treatment course, and survival were compared. RESULTS Eight patients were identified. All had negative laparoscopies. Five cytologies were negative, two were atypical, and one was positive. Two patients with negative cytology received neoadjuvant chemotherapy and underwent resection, with an average follow-up time of 32.9 months since diagnosis. Of the three remaining patients with negative cytology, none underwent resection. One received delayed chemotherapy, while the others could not due to medical contraindications. The average survival was 3.5 months (n=2). Of two patients with atypical cytology, neither underwent resection. One could not receive chemotherapy due to medical contraindication, while the other was lost to follow-up shortly after diagnosis. The average survival was 1.3 months (n=1). The patient with positive cytology received definitive chemotherapy without resection and survived for 21.6 months. CONCLUSIONS The patient with positive cytology may have been spared non-therapeutic surgery. Remaining unresected patients showed poor survival, though the lack of immediate chemotherapy may contribute to this finding. Further research is needed to determine optimal candidates for invasive staging and implications of atypical cytology.
Collapse
|
22
|
Seufferlein T, Mayerle J, Böck S, Brunner T, Ettrich TJ, Grenacher L, Gress TM, Hackert T, Heinemann V, Kestler A, Sinn M, Tannapfel A, Wedding U, Uhl W. S3-Leitlinie zum exokrinen Pankreaskarzinom – Langversion 2.0 – Dezember 2021 – AWMF-Registernummer: 032/010OL. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2022; 60:e812-e909. [PMID: 36368658 DOI: 10.1055/a-1856-7346] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
| | | | - Stefan Böck
- Medizinische Klinik und Poliklinik III, Universitätsklinikum München, Germany
| | - Thomas Brunner
- Universitätsklinik für Strahlentherapie-Radioonkologie, Medizinische Universität Graz, Austria
| | | | | | - Thomas Mathias Gress
- Klinik für Gastroenterologie und Endokrinologie, Universitätsklinikum Gießen und Marburg, Germany
| | - Thilo Hackert
- Klinik für Allgemein-, Viszeral- und Transplantationschirurgie Universitätsklinikum, Heidelberg, Germany
| | - Volker Heinemann
- Medizinische Klinik und Poliklinik III, Klinikum der Universität München-Campus Grosshadern, München, Germany
| | | | - Marianne Sinn
- Universitätsklinikum Hamburg-Eppendorf Medizinische Klinik und Poliklinik II Onkologie Hämatologie, Hamburg, Germany
| | | | | | - Waldemar Uhl
- Allgemein- und Viszeralchirurgie, St Josef-Hospital, Bochum, Germany
| |
Collapse
|
23
|
Ramos Rocha S, da Costa Ferreira SA, Ramalho A, Conceição Gouveia Santos VL, Cristina Nogueira P. Photobiomodulation Therapy in the Prevention and Treatment of Radiodermatitis in Breast Cancer Patients: Systematic Review. J Lasers Med Sci 2022; 13:e42. [PMID: 36743146 PMCID: PMC9841382 DOI: 10.34172/jlms.2022.42] [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: 12/20/2021] [Accepted: 07/17/2022] [Indexed: 01/27/2023]
Abstract
Introduction: Radiodermatitis (RD) is the most common side-effect of radiation therapy, yet its prevention and treatment through photobiomodulation therapy (PBMT) have demonstrated promising results. This study aimed to synthesize the evidence concerning the use of PBMT in managing RD among breast cancer patients undergoing radiation therapy. Methods: This is a systematic review with no time restrictions, based on the methodology proposed by the Joanna Briggs Institute (JBI), including such databases as PubMed, Cochrane, Web of Science, Scopus, and CINAHL. The studies were selected based on the following inclusion criteria: female participants over 18 years of age and females having breast cancer and undergoing radiation therapy using a three-dimensional technique or an intensity-modulated radiation therapy (IMRT) technique. Two reviewers assessed the methodological quality using the JBI Critical Appraisal Checklist, and the report was described based on PRISMA guidelines. Results: Red and infrared wavelengths were used. Device power ranged from 1.1 W to 0.08 W for continuous modes and 25 W for pulsed mode, resulting in a 3 and 4 J/cm2 fluence, applied throughout radiation therapy, leading to a reduced severity in cutaneous reactions. Conclusion: PBMT can reduce the severity of RD. New clinical trials are required to standardize protocols, given the scarcity of studies for the adopted site and methodological diversity.
Collapse
Affiliation(s)
- Suzy Ramos Rocha
- Correspondence to Suzy Ramos Rocha, 14 Bis Square, n 18, apartment 52, Bela Vista, São Paulo, SP, Brazil. CEP 01312-010.
| | | | | | | | | |
Collapse
|
24
|
Testoni SGG, Petrone MC, Reni M, Di Serio C, Rancoita PM, Rossi G, Balzano G, Linzenbold W, Enderle M, Della-Torre E, De Cobelli F, Falconi M, Capurso G, Arcidiacono PG. EUS-guided ablation with the HybridTherm Probe as second-line treatment in patients with locally advanced pancreatic ductal adenocarcinoma: A case-control study. Endosc Ultrasound 2022; 11:383-392. [PMID: 36255026 PMCID: PMC9688129 DOI: 10.4103/eus-d-21-00200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 04/24/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Data on the clinical efficacy of EUS-guided ablation using the HybridTherm-Probe (EUS-HTP) in locally advanced pancreatic ductal adenocarcinoma (LA-PDAC) are lacking. The aim of the study was to assess the impact of EUS-HTP added to chemotherapy (CT) on overall survival (OS) and progression-free survival (PFS) of LA-PDAC patients with local disease progression (DP) after first-line therapy, compared to CT alone in controls. METHODS LA-PDAC cases, prospectively treated by EUS-HTP, were retrospectively compared to matched controls (1:2) receiving standard treatment. Study endpoints were the OS and PFS from local DP after first-line therapy, compared through log-rank test calculating hazard ratios and differences in restricted mean OS/PFS time (RMOST/RMPFST) within prespecified time points (4, 6, and 12 months). RESULTS Thirteen cases and 26 controls were included. Clinical, tumor, and therapy features before and after first-line therapy were case-control balanced. The median OS and PFS were not significantly improved in cases over controls (months: 7 vs. 5 and 5 vs. 3, respectively). At 4 and 6 months, the RMPFST difference was in favor of cases (P = 0.0001 and P = 0.003, respectively). In cases and controls not candidate to further CT (N = 5 and N = 9), the median OS and PFS were not significantly improved in cases over controls (months: 6 vs. 3 and 4 vs. 2, respectively), but the RMPFST difference was in favor of cases at 4 months (P = 0.002). CONCLUSIONS In locally progressive PDAC patients experiencing failure of first-line therapy, EUS-HTP achieves a significantly better RMPFST up to 6 months compared to standard treatment, although without a significant impact on OS.
Collapse
Affiliation(s)
- Sabrina Gloria Giulia Testoni
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Hospital, Milan, Italy
| | - Maria Chiara Petrone
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Hospital, Milan, Italy
| | - Michele Reni
- Department of Oncology, Pancreas Translational and Clinical Research Center, San Raffaele Hospital, Milan, Italy
| | - Clelia Di Serio
- Vita-Salute San Raffaele University, CUSSB, University Centre for Statistics in the Biomedical Sciences, Milan, Italy
| | - Paola Maria Rancoita
- Vita-Salute San Raffaele University, CUSSB, University Centre for Statistics in the Biomedical Sciences, Milan, Italy
| | - Gemma Rossi
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Hospital, Milan, Italy
| | - Gianpaolo Balzano
- Department of Pancreatic Surgery, Pancreas Translational and Clinical Research Center, San Raffaele Hospital, Milan, Italy
| | | | | | - Emanuel Della-Torre
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, Pancreas Translational and Clinical Research Center, San Raffaele Hospital, Milan, Italy
| | - Francesco De Cobelli
- Department of Radiology and Center for Experimental Imaging, Pancreas Translational and Clinical Research Center, Vita-Salute San Raffaele University, San Raffaele Hospital, Milan, Italy
| | - Massimo Falconi
- Department of Pancreatic Surgery, Pancreas Translational and Clinical Research Center, Vita-Salute San Raffaele University, San Raffaele Hospital, Milan, Italy
| | - Gabriele Capurso
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Hospital, Milan, Italy
| | - Paolo Giorgio Arcidiacono
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, Vita-Salute San Raffaele University, San Raffaele Hospital, Milan, Italy
| |
Collapse
|
25
|
McGrath J, Kane LE, Maher SG. The Influence of MicroRNA-31 on Oxidative Stress and Radiosensitivity in Pancreatic Ductal Adenocarcinoma. Cells 2022; 11:2294. [PMID: 35892591 PMCID: PMC9332078 DOI: 10.3390/cells11152294] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/18/2022] [Accepted: 07/23/2022] [Indexed: 01/27/2023] Open
Abstract
Radioresistance remains a significant challenge in treating pancreatic ductal adenocarcinoma (PDAC), contributing to the poor survival rates of this cancer. MicroRNAs (miRs) are small non-coding RNA molecules that may play an essential role in regulating radioresistance by altering the levels of oxidative stress. In this study, we investigated the role and potential mechanisms linking miR-31 to PDAC radioresistance. A pCMV-miR vector containing a miR-31 mimic was stably expressed into a miR-31-deficient PDAC cell line, BxPC-3. Additionally, a pmiRZip lentivector suppressing miR-31 was stably expressed in a miR-31 abundant PDAC cell line, Panc-1. Clonogenic assays were conducted to explore the role of miR-31 manipulation on radiosensitivity. Fluorometric ROS assays were performed to quantify ROS levels. The expression of potential miR-31 targets was measured by Western blot analysis. It was found that the manipulation of miR-31 altered the radiosensitivity in PDAC cells by regulating oxidative stress. Using online bioinformatics tools, we identified the 3'UTR of GPx8 as a predicted target of miR-31. Our study demonstrates, for the first time, that manipulating miR-31 alters GPx8 expression, regulating ROS detoxification and promoting either a radioresistant or radiosensitive phenotype. MiR-31 may represent a promising therapeutic target for altering radiosensitivity in PDAC cells.
Collapse
Affiliation(s)
| | | | - Stephen G. Maher
- Cancer Chemoradiation Research Group, Department of Surgery, Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, St. James’s Hospital, D08 W9RT 8 Dublin, Ireland; (J.M.); (L.E.K.)
| |
Collapse
|
26
|
Yang H, Liu B, Liu D, Yang Z, Zhang S, Xu P, Xing Y, Kutschick I, Pfeffer S, Britzen-Laurent N, Grützmann R, Pilarsky C. Genome-Wide CRISPR Screening Identifies DCK and CCNL1 as Genes That Contribute to Gemcitabine Resistance in Pancreatic Cancer. Cancers (Basel) 2022; 14:cancers14133152. [PMID: 35804923 PMCID: PMC9264918 DOI: 10.3390/cancers14133152] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Pancreatic cancer is one of the most lethal cancers. Although complete surgical resection is the only curative treatment for pancreatic cancer, a late diagnosis is common and makes surgical treatment infeasible. Therefore, most patients receive chemotherapy to reduce the tumor burden. Gemcitabine has been the main chemotherapy for pancreatic cancer for over a decade; however, chemoresistance has emerged as a significant challenge to the efficacy of chemotherapy. In this study, we applied genome-wide CRISPR/Cas9 loss-of-function screening with gemcitabine treatment to identify DCK and CCNL1 as genes that contribute to gemcitabine resistance in pancreatic cancer and explored the mechanism of CCNL1-related gemcitabine resistance. Abstract Pancreatic cancer is one of the most lethal cancers. Due to the difficulty of early diagnosis, most patients are diagnosed with metastasis or advanced-stage cancer, limiting the possibility of surgical treatment. Therefore, chemotherapy is applied to improve patient outcomes, and gemcitabine has been the primary chemotherapy drug for pancreatic cancer for over a decade. However, drug resistance poses a significant challenge to the efficacy of chemotherapy. The CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9) gene-editing system is a powerful tool, and researchers have developed CRISPR/Cas9 library screening as a means to identify the genes associated with specific phenotype changes. We performed genome-wide CRISPR/Cas9 knockout screening in the mouse pancreatic cancer cell line TB32047 with gemcitabine treatment and identified deoxycytidine kinase (DCK) and cyclin L1 (CCNL1) as the top hits. We knocked out DCK and CCNL1 in the TB32047 and PANC1 cell lines and confirmed that the loss of DCK or CCNL1 enhanced gemcitabine resistance in pancreatic cells. Many researchers have addressed the mechanism of DCK-related gemcitabine resistance; however, no study has focused on CCNL1 and gemcitabine resistance. Therefore, we explored the mechanism of CCNL1-related gemcitabine resistance and found that the loss of CCNL1 activates the ERK/AKT/STAT3 survival pathway, causing cell resistance to gemcitabine treatment.
Collapse
Affiliation(s)
- Hai Yang
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (H.Y.); (D.L.); (Z.Y.); (S.Z.); (P.X.); (Y.X.); (I.K.); (S.P.); (N.B.-L.); (R.G.)
| | - Bin Liu
- Cancer Research Center, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing 101149, China;
| | - Dongxue Liu
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (H.Y.); (D.L.); (Z.Y.); (S.Z.); (P.X.); (Y.X.); (I.K.); (S.P.); (N.B.-L.); (R.G.)
| | - Zhirong Yang
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (H.Y.); (D.L.); (Z.Y.); (S.Z.); (P.X.); (Y.X.); (I.K.); (S.P.); (N.B.-L.); (R.G.)
| | - Shuman Zhang
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (H.Y.); (D.L.); (Z.Y.); (S.Z.); (P.X.); (Y.X.); (I.K.); (S.P.); (N.B.-L.); (R.G.)
| | - Pengyan Xu
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (H.Y.); (D.L.); (Z.Y.); (S.Z.); (P.X.); (Y.X.); (I.K.); (S.P.); (N.B.-L.); (R.G.)
| | - Yuming Xing
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (H.Y.); (D.L.); (Z.Y.); (S.Z.); (P.X.); (Y.X.); (I.K.); (S.P.); (N.B.-L.); (R.G.)
| | - Isabella Kutschick
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (H.Y.); (D.L.); (Z.Y.); (S.Z.); (P.X.); (Y.X.); (I.K.); (S.P.); (N.B.-L.); (R.G.)
| | - Susanne Pfeffer
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (H.Y.); (D.L.); (Z.Y.); (S.Z.); (P.X.); (Y.X.); (I.K.); (S.P.); (N.B.-L.); (R.G.)
| | - Nathalie Britzen-Laurent
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (H.Y.); (D.L.); (Z.Y.); (S.Z.); (P.X.); (Y.X.); (I.K.); (S.P.); (N.B.-L.); (R.G.)
| | - Robert Grützmann
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (H.Y.); (D.L.); (Z.Y.); (S.Z.); (P.X.); (Y.X.); (I.K.); (S.P.); (N.B.-L.); (R.G.)
| | - Christian Pilarsky
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (H.Y.); (D.L.); (Z.Y.); (S.Z.); (P.X.); (Y.X.); (I.K.); (S.P.); (N.B.-L.); (R.G.)
- Correspondence:
| |
Collapse
|
27
|
Lv SY, Lin MJ, Yang ZQ, Xu CN, Wu ZM. Survival Analysis and Prediction Model of ASCP Based on SEER Database. Front Oncol 2022; 12:909257. [PMID: 35814413 PMCID: PMC9263703 DOI: 10.3389/fonc.2022.909257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/30/2022] [Indexed: 11/25/2022] Open
Abstract
Background This study aims to compare the incidence and clinical and survival characteristics of adenosquamous carcinoma of the pancreas (ASCP) and adenomatous carcinoma of the pancreas (ACP), analyze the survival factors of ASCP and construct a prognostic model. Method Patients diagnosed with pancreatic cancer from 2000 to 2018 are selected from the SEER database. ASCP and ACP are compared in terms of epidemiology, clinical characteristics and prognosis. Cases are matched in a 1:2 ratio, and survival analysis is performed. The Cox proportional hazard model is used to determine covariates related to overall survival (OS), and an ASCP prognosis nomogram is constructed and verified by consistency index (C-index), calibration chart and decision curve analysis (DCA). The accuracy of the model is compared with that of AJCC.Stage and SEER.Stage to obtain the area under the receiver operating characteristic (ROC) curve. Results the age-adjusted incidence of ACP increased significantly over time from 2000 to 2008 and from 2008 to 2018 (P < 0.05). APC was 2.01% (95% CI: 1.95–2.21) and 1.08% (95% CI: 0.93–1.25) respectively. The age-adjusted incidence of ASCP increased with time from 2000 to 2018 (P < 0.05) and APC was 3.64% (95% CI: 3.25–4.01).After propensity score matching (PSM), the OS and cancer-specific survival (CSS) of ACP are better than those of ASCP. The survival time of ASCP is significantly improved by the combined treatment of surgery + chemotherapy + radiotherapy, with a median OS of 31 months. Cox proportional hazard regression analysis shows that age, race, surgery, radiotherapy, chemotherapy and tumor size are independent factors affecting the prognosis. DCA and area under the curve (AUC) value shows that the model has good discrimination ability. Conclusion The OS prognosis of ASCP is worse than that of ACP, and the nomogram has high accuracy for the prognosis prediction of ASCP.
Collapse
|
28
|
Pancreatic cancer cells spectral library by DIA-MS and the phenotype analysis of gemcitabine sensitivity. Sci Data 2022; 9:283. [PMID: 35680938 PMCID: PMC9184632 DOI: 10.1038/s41597-022-01407-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 05/18/2022] [Indexed: 12/05/2022] Open
Abstract
Data-independent acquisition (DIA)-mass spectrometry (MS)-based proteome strategies are increasingly used for detecting and validating protein biomarkers and therapeutic targets. Here, based on an in-depth proteome analysis of seven pancreatic cancer cell lines, we built a pancreas-specific mass spectrum library containing 10633 protein groups and 184551 peptides. The proteome difference among the seven pancreatic cancer cells was significant, especially for the divergent expression of proteins related to epithelial-mesenchymal transition (EMT). The spectra library was applied to explore the proteome difference of PANC-1 and BxPC-3 cells upon gemcitabine (GEM) treatment, and potential GEM targets were identified. The cytotoxicity test and GEM target analysis found that HPAC, CFPAC-1, and BxPC-3 were sensitive to GEM treatment, whereas PANC-1 and AsPC-1 were resistant. Finally, we found EMT was significant for CFPAC-1, AsPC-1, and PANC-1 cells, whereas BxPC-3 and HPAC cells showed more typical epithelial features. This library provides a valuable resource for in-depth proteomic analysis on pancreatic cancer cell lines, meeting the urgent demands for cell line-dependent protein differences and targeted drug analysis. Measurement(s) | protein expression profiling | Technology Type(s) | Mass Spectrometry |
Collapse
|
29
|
Xie W, Li X, Yang C, Li J, Shen G, Chen H, Xiao SY, Li Y. The Pyroptosis-Related Gene Prognostic Index Associated with Tumor Immune Infiltration for Pancreatic Cancer. Int J Mol Sci 2022; 23:6178. [PMID: 35682857 PMCID: PMC9180955 DOI: 10.3390/ijms23116178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/27/2022] [Accepted: 05/30/2022] [Indexed: 11/28/2022] Open
Abstract
Pancreatic cancer (PC) is one of the most fatal malignancies. Pyroptosis, a type of inflammatory cell death, likely plays a critical role in the development and progression of tumors. However, the relationship between pyroptosis-related genes (PRGs) and prognosis and immunity to PC is not entirely clear. This study, aimed at identifying the key PRGs in PC, highlights their prognostic value, immune characteristics, and candidate drugs for therapies. We screened 47 differentially expressed PRGs between PC and normal pancreas tissues from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) datasets. Afterwards, a pyroptosis-related gene prognostic index (PRGPI) was constructed based on eight PRGs (AIM2, GBP1, HMGB1, IL18, IRF6, NEK7, NLRP1 and PLCG1) selected by univariate and multivariate Cox regression analysis and LASSO regression analysis, and verified in two external datasets from the International Cancer Genome Consortium (ICGC) and Gene Expression Omnibus (GEO) databases. We found that the PC patients in the PRGPI-defined subgroups not only reflected significantly different levels of infiltration in a variety of immune cells, such as M1 macrophages, but also showed differential expression in genes of the human leukocyte antigen (HLA) family and immune checkpoints. Additionally, molecular characteristics and drug sensitivity also stayed close to the PRGPI risk scores. Therefore, PRGPI may serve as a valuable prognostic biomarker and may potentially provide guidance toward novel therapeutic options for PC patients.
Collapse
Affiliation(s)
- Wen Xie
- Department of Pathology, Wuhan University Zhongnan Hospital, Wuhan 430000, China; (W.X.); (X.L.); (C.Y.); (J.L.); (G.S.); (H.C.)
- Wuhan University Center for Pathology and Molecular Diagnostics, Wuhan 430000, China
| | - Xiaoyi Li
- Department of Pathology, Wuhan University Zhongnan Hospital, Wuhan 430000, China; (W.X.); (X.L.); (C.Y.); (J.L.); (G.S.); (H.C.)
- Wuhan University Center for Pathology and Molecular Diagnostics, Wuhan 430000, China
| | - Chunxiu Yang
- Department of Pathology, Wuhan University Zhongnan Hospital, Wuhan 430000, China; (W.X.); (X.L.); (C.Y.); (J.L.); (G.S.); (H.C.)
- Wuhan University Center for Pathology and Molecular Diagnostics, Wuhan 430000, China
| | - Jiahao Li
- Department of Pathology, Wuhan University Zhongnan Hospital, Wuhan 430000, China; (W.X.); (X.L.); (C.Y.); (J.L.); (G.S.); (H.C.)
- Wuhan University Center for Pathology and Molecular Diagnostics, Wuhan 430000, China
| | - Guoyan Shen
- Department of Pathology, Wuhan University Zhongnan Hospital, Wuhan 430000, China; (W.X.); (X.L.); (C.Y.); (J.L.); (G.S.); (H.C.)
- Wuhan University Center for Pathology and Molecular Diagnostics, Wuhan 430000, China
| | - Hongshan Chen
- Department of Pathology, Wuhan University Zhongnan Hospital, Wuhan 430000, China; (W.X.); (X.L.); (C.Y.); (J.L.); (G.S.); (H.C.)
- Wuhan University Center for Pathology and Molecular Diagnostics, Wuhan 430000, China
| | - Shu-Yuan Xiao
- Department of Pathology, University of Chicago Medicine, Chicago, IL 60637, USA
| | - Yueying Li
- Department of Pathology, Wuhan University Zhongnan Hospital, Wuhan 430000, China; (W.X.); (X.L.); (C.Y.); (J.L.); (G.S.); (H.C.)
- Wuhan University Center for Pathology and Molecular Diagnostics, Wuhan 430000, China
| |
Collapse
|
30
|
Auranofin and Pharmacologic Ascorbate as Radiomodulators in the Treatment of Pancreatic Cancer. Antioxidants (Basel) 2022; 11:antiox11050971. [PMID: 35624835 PMCID: PMC9137675 DOI: 10.3390/antiox11050971] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/27/2022] [Accepted: 05/11/2022] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer accounts for nearly one fourth of all new cancers worldwide. Little progress in the development of novel or adjuvant therapies has been made over the past few decades and new approaches to the treatment of pancreatic cancer are desperately needed. Pharmacologic ascorbate (P-AscH−, high-dose, intravenous vitamin C) is being investigated in clinical trials as an adjunct to standard-of-care chemoradiation treatments. In vitro, P-AscH− has been shown to sensitize cancer cells to ionizing radiation in a manner that is dependent on the generation of H2O2 while simultaneously protecting normal tissue from radiation damage. There is renewed interest in Auranofin (Au), an FDA-approved medication utilized in the treatment of rheumatoid arthritis, as an anti-cancer agent. Au inhibits the thioredoxin antioxidant system, thus increasing the overall peroxide burden on cancer cells. In support of current literature demonstrating Au’s effectiveness in breast, colon, lung, and ovarian cancer, we offer additional data that demonstrate the effectiveness of Au alone and in combination with P-AscH− and ionizing radiation in pancreatic cancer treatment. Combining P-AscH− and Au in the treatment of pancreatic cancer may confer multiple mechanisms to increase H2O2-dependent toxicity amongst cancer cells and provide a promising translatable avenue by which to enhance radiation effectiveness and improve patient outcomes.
Collapse
|
31
|
Sim W, Lim WM, Hii LW, Leong CO, Mai CW. Targeting pancreatic cancer immune evasion by inhibiting histone deacetylases. World J Gastroenterol 2022; 28:1934-1945. [PMID: 35664961 PMCID: PMC9150054 DOI: 10.3748/wjg.v28.i18.1934] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/17/2022] [Accepted: 04/04/2022] [Indexed: 02/06/2023] Open
Abstract
The immune system plays a vital role in maintaining the delicate balance between immune recognition and tumor development. Regardless, it is not uncommon that cancerous cells can intelligently acquire abilities to bypass the antitumor immune responses, thus allowing continuous tumor growth and development. Immune evasion has emerged as a significant factor contributing to the progression and immune resistance of pancreatic cancer. Compared with other cancers, pancreatic cancer has a tumor microenvironment that can resist most treatment modalities, including emerging immunotherapy. Sadly, the use of immunotherapy has yet to bring significant clinical breakthrough among pancreatic cancer patients, suggesting that pancreatic cancer has successfully evaded immunomodulation. In this review, we summarize the impact of genetic alteration and epigenetic modification (especially histone deacetylases, HDAC) on immune evasion in pancreatic cancer. HDAC overexpression significantly suppresses tumor suppressor genes, contributing to tumor growth and progression. We review the evidence on HDAC inhibitors in tumor eradication, improving T cells activation, restoring tumor immunogenicity, and modulating programmed death 1 interaction. We provide our perspective in targeting HDAC as a strategy to reverse immune evasion in pancreatic cancer.
Collapse
Affiliation(s)
- Wynne Sim
- School of Medicine, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Wei-Meng Lim
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
- Center for Cancer and Stem Cell Research, Institute for Research, Development, and Innovation, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Ling-Wei Hii
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
- Center for Cancer and Stem Cell Research, Institute for Research, Development, and Innovation, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Chee-Onn Leong
- Center for Cancer and Stem Cell Research, Institute for Research, Development, and Innovation, International Medical University, Kuala Lumpur 57000, Malaysia
- AGTC Genomics, Kuala Lumpur 57000, Malaysia
| | - Chun-Wai Mai
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
32
|
Yang Z, Shi G, Zhang P. Development and Validation of Nomograms to Predict Overall Survival and Cancer-Specific Survival in Patients With Pancreatic Adenosquamous Carcinoma. Front Oncol 2022; 12:831649. [PMID: 35330710 PMCID: PMC8940199 DOI: 10.3389/fonc.2022.831649] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/31/2022] [Indexed: 12/15/2022] Open
Abstract
Background Pancreatic adenosquamous carcinoma (PASC) is a heterogeneous group of primary pancreatic cancers characterized by the coexistence of both glandular and squamous differentiation. The aim of this study was to develop nomograms to predict survival outcomes in patients with PASC. Methods In this retrospective study, data on PASC, including clinicopathological characteristics, treatments, and survival outcomes, were collected from the SEER database between 2000 and 2018. The primary endpoints were overall survival (OS) and cancer-specific survival (CSS). The eligible patients were randomly divided into development cohort and validation cohort in a 7:3 ratio. The nomograms for prediction of OS and CSS were constructed by the development cohort using a LASSO-Cox regression model, respectively. Besides the model performance was internally and externally validated by examining the discrimination, calibration, and clinical utility. Results A total of 632 consecutive patients who had been diagnosed with PASC were identified and randomly divided into development (n = 444) and validation (n = 188) cohorts. In the development cohort, the estimated median OS was 7.0 months (95% CI: 6.19-7.82) and the median CSS was 7.0 months (95% CI: 6.15-7.85). In the validation cohort, the estimated median OS was 6.0 months (95% CI: 4.46-7.54) and the median CSS was 7.0 months (95% CI: 6.25-7.75). LASSO-penalized COX regression analysis identified 8 independent predictors in the OS prediction model and 9 independent risk factors in the CSS prediction model: age at diagnosis, gender, year of diagnosis, tumor location, grade, stage, size, lymph node metastasis, combined metastasis, surgery, radiation, and chemotherapy. The Harrell C index and time-dependent AUCs manifested satisfactory discriminative capabilities of the models. Calibration plots showed that both models were well calibrated. Furthermore, decision curves indicated good utility of the nomograms for decision-making. Conclusion Nomogram-based models to evaluate personalized OS and CSS in patients with PASC were developed and well validated. These easy-to-use tools will be useful methods to calculate individualized estimate of survival, assist in risk stratification, and aid clinical decision-making.
Collapse
Affiliation(s)
- Zhen Yang
- Department of Hepatopancreatobiliary Surgery, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Guangjun Shi
- Department of Hepatopancreatobiliary Surgery, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ping Zhang
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| |
Collapse
|
33
|
Song H, Jiang C. Recent advances in targeted drug delivery for the treatment of pancreatic ductal adenocarcinoma. Expert Opin Drug Deliv 2022; 19:281-301. [PMID: 35220832 DOI: 10.1080/17425247.2022.2045943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) has become a serious health problem with high impact worldwide. The heterogeneity of PDAC makes it difficult to apply drug delivery systems (DDS) used in other cancer models, for example, the poorly developed vascular system makes anti-angiogenic therapy ineffective. Due to its various malignant pathological changes, drug delivery against PDAC is a matter of urgent concern. Based on this situation, various drug delivery strategies specially designed for PDAC have been generated. AREAS COVERED This review will briefly describe how delivery systems can be designed through nanotechnology and formulation science. Most research focused on penetrating the stromal barrier, exploiting and alleviating the hypoxic microenvironment, targeting immune cells, or designing vaccines, and combination therapies. This review will summarize the ways to reverse the malignant pathological features of PDAC and hopefully provide ideas for subsequent studies. EXPERT OPINION Drug delivery systems designed to achieve penetrating functions or to alleviate hypoxia and activate immunity have achieved good therapeutic results in animal models in several studies. In future studies, there is a need to deliver PDAC therapeutics in a more precise manner, or the use of drug carriers for multiple functions simultaneously, are potential therapeutic strategy.
Collapse
Affiliation(s)
- Haolin Song
- Department of Pharmaceutics, Fudan University, Shanghai, Sichuan, 201203 China
| | - Chen Jiang
- Department of Pharmaceutics, Fudan University, Shanghai, Sichuan, 201203 China
| |
Collapse
|
34
|
McCubrey JA, Abrams SL, Steelman LS, Cocco L, Ratti S, Martelli AM, Lombardi P, Gizak A, Duda P. APR-246-The Mutant TP53 Reactivator-Increases the Effectiveness of Berberine and Modified Berberines to Inhibit the Proliferation of Pancreatic Cancer Cells. Biomolecules 2022; 12:276. [PMID: 35204775 PMCID: PMC8961609 DOI: 10.3390/biom12020276] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/04/2022] [Accepted: 02/04/2022] [Indexed: 12/10/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common form of pancreatic cancer. In ~75% of PDAC, the tumor suppressor TP53 gene is mutated. Novel approaches to treat cancer involve compounds called mutant TP53 reactivators. They interact with mutant TP53 proteins and restore some of their growth suppressive properties, but they may also interact with other proteins, e.g., TP63 and TP73. We examined the ability of the TP53 reactivator APR-246 to interact with eleven modified berberine compounds (NAX compounds) in the presence and absence of WT-TP53 in two PDAC cell lines: the MIA-PaCa-2, which has gain of function (GOF) TP53 mutations on both alleles, and PANC-28, which lacks expression of the WT TP53 protein. Our results indicate the TP53 reactivator-induced increase in therapeutic potential of many modified berberines.
Collapse
Affiliation(s)
- James Andrew McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, USA; (S.L.A.); (L.S.S.)
| | - Stephen L. Abrams
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, USA; (S.L.A.); (L.S.S.)
| | - Linda S. Steelman
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, USA; (S.L.A.); (L.S.S.)
| | - Lucio Cocco
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, 40126 Bologna, Italy; (L.C.); (S.R.); (A.M.M.)
| | - Stefano Ratti
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, 40126 Bologna, Italy; (L.C.); (S.R.); (A.M.M.)
| | - Alberto M. Martelli
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, 40126 Bologna, Italy; (L.C.); (S.R.); (A.M.M.)
| | - Paolo Lombardi
- Naxospharma, Via Giuseppe Di Vittorio 70, 20026 Novate Milanese, Italy;
| | - Agnieszka Gizak
- Department of Molecular Physiology and Neurobiology, University of Wrocław, 50-335 Wroclaw, Poland; (A.G.); (P.D.)
| | - Przemysław Duda
- Department of Molecular Physiology and Neurobiology, University of Wrocław, 50-335 Wroclaw, Poland; (A.G.); (P.D.)
| |
Collapse
|
35
|
Parisi S, Ferini G, Cacciola A, Lillo S, Tamburella C, Santacaterina A, Bottari A, Brogna A, Ferrantelli G, Pontoriero A, Minutoli F, Pergolizzi S. A non-surgical COMBO-therapy approach for locally advanced unresectable pancreatic adenocarcinoma: preliminary results of a prospective study. LA RADIOLOGIA MEDICA 2022; 127:214-219. [PMID: 35034325 DOI: 10.1007/s11547-021-01441-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022]
Abstract
In this short report we present a series of thirteen patients with locally advanced, unresectable, pancreatic cancer treated with a COMBO-Therapy consisting of: STEP-1: induction chemotherapy; STEP-2: concomitant chemoradiotherapy; STEP-3: stereotactic body radiotherapy boost. After four weeks from the end of each step all patients had a re-staging and a surgical re-evaluation. All patients completed STEP-1 and STEP-2. STEP-3 has been successfully delivered to 8/13 patients with a median dose of 12 Gy (range 10-21 Gy) in 1-3 fractions. The median LC was 20 months (range 10-32) with a 2-year LC of 72.9%, and none of the patients developed G3 acute or late toxicities. The median OS was 21.5 months (range 12-34), and the 2-year OS was 53.9%; the median PFS was 17.5 months (range 10-27). Our non-surgical COMBO-Therapy has demonstrated a feasible profile with good tolerance. Further prospective protocols are needed to confirm our preliminary results.
Collapse
Affiliation(s)
- Silvana Parisi
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, Italy
| | | | - Alberto Cacciola
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, Italy
| | - Sara Lillo
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, Italy.
| | - Consuelo Tamburella
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, Italy
| | | | - Antonio Bottari
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, Italy
| | - Anna Brogna
- Unit of Medical Physics, University Hospital Policlinico "G. Martino", Messina, Italy
| | - Giacomo Ferrantelli
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, Italy
| | - Antonio Pontoriero
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, Italy
| | - Fabio Minutoli
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, Italy
| | - Stefano Pergolizzi
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, Italy
| |
Collapse
|
36
|
Wang D, Ge H, Tian M, Li C, Zhao L, Pei Q, Tan F, Li Y, Ling C, Güngör C. The Survival Effect of Radiotherapy on Stage IIB/III Pancreatic Cancer Undergone Surgery in Different Age and Tumor Site Groups: A Propensity Scores Matching Analysis Based on SEER Database. Front Oncol 2022; 12:799930. [PMID: 35174085 PMCID: PMC8841859 DOI: 10.3389/fonc.2022.799930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/07/2022] [Indexed: 11/23/2022] Open
Abstract
Background It remains controversial whether radiotherapy (RT) improves survival in patients with stage IIB/III PDAC. A growing number of studies have found that patients’ age at diagnosis and tumor site not only affect prognosis, but also may lead to different treatment responses. Therefore, the purpose of this study was to verify whether the survival effect of radiotherapy in patients with stage IIB/III PDAC varies across age and tumor site groups. Methods The target population was selected from PDAC patients undergone surgery in the Surveillance, Epidemiology, and End Results (SEER) database between 2004 and 2016. This study performed the Pearson’s chi-square test, Cox regression analysis, Kaplan-Meier (K-M) method, and focused on propensity frequency matching analysis. Results Neither neoadjuvant radiotherapy (nRT) nor adjuvant radiotherapy (aRT) patient group had probably improved survival among early-onset patients. For middle-aged patients, nRT seemed to fail to extend overall survival (OS), while aRT might improve the OS. Plus, both nRT and aRT were associated with improved survival in elderly patients. The aRT might be related with survival benefits in patients with pancreatic head cancer, while nRT was not. And RT in patients with PDAC at other sites did not appear to provide a survival benefit. Conclusion Carefully selected data from the SEER database suggested that age and tumor location may be the reference factors to guide the selection of RT for patients with stage IIB/III PDAC. These findings are likely to contribute to the development of personalized treatment for patients with stage IIB/III PDAC.
Collapse
Affiliation(s)
- Dan Wang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of General Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Heming Ge
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Mengxiang Tian
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Chenglong Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lilan Zhao
- Department of Thoracic Surgery, Fujian Provincial Hospital, Fuzhou, China
| | - Qian Pei
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Fengbo Tan
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yuqiang Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of General Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- *Correspondence: Yuqiang Li, ; Chen Ling,
| | - Chen Ling
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Yuqiang Li, ; Chen Ling,
| | - Cenap Güngör
- Department of General Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
37
|
Wang SP, Li Y, Huang SH, Wu SQ, Gao LL, Sun Q, Lin QW, Huang L, Meng LQ, Zou Y, Zhu QH, Xu YG. Discovery of Potent and Novel Dual PARP/BRD4 Inhibitors for Efficient Treatment of Pancreatic Cancer. J Med Chem 2021; 64:17413-17435. [PMID: 34813314 DOI: 10.1021/acs.jmedchem.1c01535] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Targeting poly(ADP-ribose) polymerase1/2 (PARP1/2) is a promising strategy for the treatment of pancreatic cancer with breast cancer susceptibility gene (BRCA) mutation. Inducing the deficiency of homologous recombination (HR) repair is an effective way to broaden the indication of PARP1/2 inhibitor for more patients with pancreatic cancer. Bromodomain-containing protein 4 (BRD4) repression has been reported to elevate HR deficiency. Therefore, we designed, synthetized, and optimized a dual PARP/BRD4 inhibitor III-16, with a completely new structure and high selectivity against PARP1/2 and BRD4. III-16 showed favorable synergistic antitumor efficacy in pancreatic cancer cells and xenografts by arresting cell cycle progression, inhibiting DNA damage repair, and promoting autophagy-associated cell death. Moreover, III-16 reversed Olaparib-induced acceleration of cell cycle progression and recovery of DNA repair. The advantages of III-16 over Olaparib suggest that dual PARP/BRD4 inhibitors are novel and promising agents for the treatment of advanced pancreatic cancer.
Collapse
Affiliation(s)
- Shu-Ping Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 211198 Nanjing, China
| | - Yu Li
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Shi-Hui Huang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Shi-Qi Wu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Ling-Li Gao
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Qin Sun
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Qian-Wen Lin
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Lei Huang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Liu-Qiong Meng
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Yi Zou
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Qi-Hua Zhu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Yun-Gen Xu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 211198 Nanjing, China.,Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
38
|
Petenyi FG, Garay T, Muhl D, Izso B, Karaszi A, Borbenyi E, Herold M, Herold Z, Szasz AM, Dank M. Modulated Electro-Hyperthermic (mEHT) Treatment in the Therapy of Inoperable Pancreatic Cancer Patients-A Single-Center Case-Control Study. Diseases 2021; 9:81. [PMID: 34842668 PMCID: PMC8628793 DOI: 10.3390/diseases9040081] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/03/2021] [Accepted: 10/30/2021] [Indexed: 02/05/2023] Open
Abstract
UNLABELLED Our present oncological treatment arsenal has limited treatment options for pancreatic ductal adenocarcinoma (PDAC). Extended reviews have shown the benefits of hyperthermia for PDAC, supporting the perspectives with the improvements of the treatment possibilities. METHODS A retrospective single-center case-control study was conducted with the inclusion of 78 inoperable PDAC patients. Age-, sex-, chemotherapy-, stage-, and ascites formation-matched patients were assigned to two equal groups based on the application of modulated electro-hyperthermia (mEHT). The EHY2030 mEHT device was used. RESULTS A trend in favor of mEHT was found in overall survival (p = 0.1420). To further evaluate the potential beneficial effects of mEHT, the presence of distant metastasis or ascites in the patients' oncological history was investigated. Of note, mEHT treatment had a favorable effect on patients' overall survival in metastatic disease (p = 0.0154), while less abdominal fluid responded to the mEHT treatment in a more efficient way (p ≤ 0.0138). CONCLUSION mEHT treatment was associated with improved overall survival in PDAC in our single-center retrospective case-control study. The outcome measures encourage us to design a randomized prospective clinical study to further confirm the efficiency of mEHT in this patient cohort.
Collapse
Affiliation(s)
- Flora Greta Petenyi
- Faculty of Information Technology and Bionics, Pazmany Peter Catholic University, 1083 Budapest, Hungary; (F.G.P.); (T.G.); (B.I.)
| | - Tamas Garay
- Faculty of Information Technology and Bionics, Pazmany Peter Catholic University, 1083 Budapest, Hungary; (F.G.P.); (T.G.); (B.I.)
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (D.M.); (A.K.); (E.B.); (Z.H.); (A.M.S.)
| | - Dorottya Muhl
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (D.M.); (A.K.); (E.B.); (Z.H.); (A.M.S.)
| | - Blanka Izso
- Faculty of Information Technology and Bionics, Pazmany Peter Catholic University, 1083 Budapest, Hungary; (F.G.P.); (T.G.); (B.I.)
| | - Adam Karaszi
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (D.M.); (A.K.); (E.B.); (Z.H.); (A.M.S.)
| | - Erika Borbenyi
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (D.M.); (A.K.); (E.B.); (Z.H.); (A.M.S.)
| | - Magdolna Herold
- Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary;
| | - Zoltan Herold
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (D.M.); (A.K.); (E.B.); (Z.H.); (A.M.S.)
| | - Attila Marcell Szasz
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (D.M.); (A.K.); (E.B.); (Z.H.); (A.M.S.)
| | - Magdolna Dank
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (D.M.); (A.K.); (E.B.); (Z.H.); (A.M.S.)
| |
Collapse
|
39
|
Pijnappel EN, Wassenaar NPM, Gurney-Champion OJ, Klaassen R, van der Lee K, Pleunis-van Empel MCH, Richel DJ, Legdeur MC, Nederveen AJ, van Laarhoven HWM, Wilmink JW. Phase I/II Study of LDE225 in Combination with Gemcitabine and Nab-Paclitaxel in Patients with Metastatic Pancreatic Cancer. Cancers (Basel) 2021; 13:4869. [PMID: 34638351 PMCID: PMC8507646 DOI: 10.3390/cancers13194869] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 09/17/2021] [Accepted: 09/24/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Desmoplasia is a central feature of the tumor microenvironment in pancreatic ductal adenocarcinoma (PDAC). LDE225 is a pharmacological Hedgehog signaling pathway inhibitor and is thought to specifically target tumor stroma. We investigated the combined use of LDE225 and chemotherapy to treat PDAC patients. METHODS This was a multi-center, phase I/II study for patients with metastatic PDAC establishing the maximum tolerated dose of LDE225 co-administered with gemcitabine and nab-paclitaxel (phase I) and evaluating the efficacy and safety of the treatment combination after prior FOLFIRINOX treatment (phase II). Tumor microenvironment assessment was performed with quantitative MRI using intra-voxel incoherent motion diffusion weighted MRI (IVIM-DWI) and dynamic contrast-enhanced (DCE) MRI. RESULTS The MTD of LDE225 was 200 mg once daily co-administered with gemcitabine 1000 mg/m2 and nab-paclitaxel 125 mg/m2. In phase II, six therapy-related grade 4 adverse events (AE) and three grade 5 were observed. In 24 patients, the target lesion response was evaluable. Three patients had partial response (13%), 14 patients showed stable disease (58%), and 7 patients had progressive disease (29%). Median overall survival (OS) was 6 months (IQR 3.9-8.1). Blood plasma fraction (DCE) and diffusion coefficient (IVIM-DWI) significantly increased during treatment. Baseline perfusion fraction could predict OS (>222 days) with 80% sensitivity and 85% specificity. CONCLUSION LDE225 in combination with gemcitabine and nab-paclitaxel was well-tolerated in patients with metastatic PDAC and has promising efficacy after prior treatment with FOLFIRINOX. Quantitative MRI suggested that LDE225 causes increased tumor diffusion and works particularly well in patients with poor baseline tumor perfusion.
Collapse
Affiliation(s)
- Esther N. Pijnappel
- Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (E.N.P.); (R.K.); (K.v.d.L.); (D.J.R.); (H.W.M.v.L.)
| | - Nienke P. M. Wassenaar
- Cancer Center Amsterdam, Department of Radiology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (N.P.M.W.); (O.J.G.-C.); (A.J.N.)
| | - Oliver J. Gurney-Champion
- Cancer Center Amsterdam, Department of Radiology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (N.P.M.W.); (O.J.G.-C.); (A.J.N.)
| | - Remy Klaassen
- Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (E.N.P.); (R.K.); (K.v.d.L.); (D.J.R.); (H.W.M.v.L.)
| | - Koen van der Lee
- Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (E.N.P.); (R.K.); (K.v.d.L.); (D.J.R.); (H.W.M.v.L.)
| | | | - Dick J. Richel
- Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (E.N.P.); (R.K.); (K.v.d.L.); (D.J.R.); (H.W.M.v.L.)
| | - Marie C. Legdeur
- Department of Medical Oncology, Medisch Spectrum Twente, Twente, 7512 Enschede, The Netherlands; (M.C.H.P.-v.E.); (M.C.L.)
| | - Aart J. Nederveen
- Cancer Center Amsterdam, Department of Radiology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (N.P.M.W.); (O.J.G.-C.); (A.J.N.)
| | - Hanneke W. M. van Laarhoven
- Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (E.N.P.); (R.K.); (K.v.d.L.); (D.J.R.); (H.W.M.v.L.)
| | - Johanna W. Wilmink
- Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (E.N.P.); (R.K.); (K.v.d.L.); (D.J.R.); (H.W.M.v.L.)
| |
Collapse
|
40
|
Middleton MR, Dean E, Evans TRJ, Shapiro GI, Pollard J, Hendriks BS, Falk M, Diaz-Padilla I, Plummer R. Phase 1 study of the ATR inhibitor berzosertib (formerly M6620, VX-970) combined with gemcitabine ± cisplatin in patients with advanced solid tumours. Br J Cancer 2021; 125:510-519. [PMID: 34040175 PMCID: PMC8368196 DOI: 10.1038/s41416-021-01405-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 03/31/2021] [Accepted: 04/15/2021] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Berzosertib (formerly M6620, VX-970) is a highly potent and selective, first-in-class inhibitor of ataxia telangiectasia and Rad3-related protein kinase (ATR). We assessed multiple ascending doses of berzosertib + gemcitabine ± cisplatin in patients with resistant/refractory advanced solid tumours. METHODS We evaluated the safety, tolerability, pharmacokinetics (PK) and preliminary efficacy of intravenous berzosertib + gemcitabine ± cisplatin using a standard 3 + 3 dose-escalation design. The starting doses were berzosertib 18 mg/m2, gemcitabine 875 mg/m2 and cisplatin 60 mg/m2. RESULTS Fifty-two patients received berzosertib + gemcitabine and eight received berzosertib + gemcitabine + cisplatin. Four patients receiving berzosertib + gemcitabine had a total of seven dose-limiting toxicities (DLTs) and three receiving berzosertib + gemcitabine + cisplatin had a total of three DLTs. Berzosertib 210 mg/m2 (days 2 and 9) + gemcitabine 1000 mg/m2 (days 1 and 8) Q3W was established as the recommended Phase 2 dose (RP2D); no RP2D was determined for berzosertib + gemcitabine + cisplatin. Neither gemcitabine nor cisplatin affected berzosertib PK. Most patients in both arms achieved a best response of either partial response or stable disease. CONCLUSIONS Berzosertib + gemcitabine was well tolerated in patients with advanced solid tumours and showed preliminary efficacy signs. CLINICAL TRIAL IDENTIFIER NCT02157792.
Collapse
Affiliation(s)
| | - Emma Dean
- Experimental Cancer Medicine Team, The University of Manchester and The Christie NHS Foundation Trust, Manchester, UK
- AstraZeneca, Cambridge and Alderley Park, UK
| | - Thomas R J Evans
- Institute of Cancer Sciences, University of Glasgow and Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Geoffrey I Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - John Pollard
- Biological Sciences, Vertex Pharmaceuticals Europe Ltd, Abingdon, UK
- Bayer plc, Reading, UK
| | - Bart S Hendriks
- Clinical Pharmacology, EMD Serono Research & Development Institute Inc., Billerica, MA, USA, an affiliate of Merck KGaA, Darmstadt, Germany
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Martin Falk
- Oncology Global Clinical Development, Merck KGaA, Darmstadt, Germany
- CureVac, Tübingen, Germany
| | - Ivan Diaz-Padilla
- Oncology Global Clinical Development, Ares Trading SA, Eysins, Switzerland, an affiliate of Merck KGaA, Darmstadt, Germany
- GlaxoSmithKline, Zug, Switzerland
| | - Ruth Plummer
- Translational and Clinical Research Institute, Newcastle University and Northern Centre for Cancer Care, Newcastle Hospitals NHS Trust, Newcastle Upon Tyne, UK.
| |
Collapse
|
41
|
Li ZY, Sun XY. Recent progress in SRC targeted therapy for pancreatic cancer. Shijie Huaren Xiaohua Zazhi 2021; 29:621-627. [DOI: 10.11569/wcjd.v29.i12.621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Zi-Yi Li
- Hepatosplenic Surgery Center, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xue-Ying Sun
- Hepatosplenic Surgery Center, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| |
Collapse
|
42
|
Granata V, Grassi R, Fusco R, Belli A, Palaia R, Carrafiello G, Miele V, Grassi R, Petrillo A, Izzo F. Local ablation of pancreatic tumors: State of the art and future perspectives. World J Gastroenterol 2021; 27:3413-3428. [PMID: 34163121 PMCID: PMC8218359 DOI: 10.3748/wjg.v27.i23.3413] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 03/28/2021] [Accepted: 05/08/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Currently, the technologies most commonly used to treat locally advanced pancreatic cancer are radiofrequency ablation (RFA), microwave ablation, and irreversible (IRE) or reversible electroporation combined with low doses of chemotherapeutic drugs.
AIM To report an overview and updates on ablative techniques in pancreatic cancer.
METHODS Several electronic databases were searched. The search covered the years from January 2000 to January 2021. Moreover, the reference lists of the found papers were analysed for papers not indexed in the electronic databases. All titles and abstracts were analysed.
RESULTS We found 30 studies (14 studies for RFA, 3 for microwave therapy, 10 for IRE, and 3 for electrochemotherapy), comprising 1047 patients, which were analysed further. Two randomized trials were found for IRE. Percutaneous and laparotomy approaches were performed. In the assessed patients, the median maximal diameter of the lesions was in the range of 2.8 to 4.5 cm. All series included patients unfit for surgical treatment, but Martin et al assessed a subgroup of patients with borderline resectable tumours who underwent resection with margin attenuation with IRE. Most studies administered chemotherapy prior to ablative therapies. However, several studies suggest that the key determinant of improved survival is attributable to ablative treatment alone. Nevertheless, the authors suggested chemotherapy before local therapies for several reasons. This strategy may not only downstage a subgroup of patients to curative-intent surgery but also support to recognize patients with biologically unfavourable tumours who would likely not benefit from ablation treatments. Ablation therapies seem safe based on the 1047 patients assessed in this review. The mortality rate ranged from 1.8% to 2%. However, despite the low mortality, the reported rates of severe post procedural complications ranged from 0%-42%. Most reported complications have been self-limiting and manageable. Median overall survival varied between 6.0 and 33 mo. Regarding the technical success rate, assessed papers reported an estimated rate in the range of 85% to 100%. However, the authors reported early recurrence after treatment. A distinct consideration should be made on whether local treatments induce an immune response in the ablated area. Preclinical and clinical studies have shown that RFA is a promising mechanism for inducing antigen-presenting cell infiltration and enhancing the systemic antitumour T-cell immune response and tumour regression.
CONCLUSION In the management of patients with pancreatic cancer, the possibility of a multimodal approach should be considered, and conceptually, the combination of RFA with immunotherapy represents a novel angle of attack against this tumour.
Collapse
Affiliation(s)
- Vincenza Granata
- Department of Radiology, Istituto Nazionale Tumori –IRCCS- Fondazione G. Pascale, Naples 80131, Italy
| | - Roberta Grassi
- Department of Radiology, Università degli Studi della Campania Luigi Vanvitelli, Naples 80127, Italy
| | - Roberta Fusco
- Department of Radiology, Istituto Nazionale Tumori –IRCCS- Fondazione G. Pascale, Naples 80131, Italy
| | - Andrea Belli
- Department of Surgery, Istituto Nazionale Tumori –IRCCS- Fondazione G. Pascale, Naples 80131, Italy
| | - Raffaele Palaia
- Department of Surgery, Istituto Nazionale Tumori –IRCCS- Fondazione G. Pascale, Naples 80131, Italy
| | | | - Vittorio Miele
- Italian Society of Medical and Interventional Radiology SIRM, SIRM Foundation Milan 20122, Italy
- Department of Emergency Radiology, San Camillo Hospital, Firenze 50139, Italy
| | - Roberto Grassi
- Department of Radiology, Università degli Studi della Campania Luigi Vanvitelli, Naples 80127, Italy
- Italian Society of Medical and Interventional Radiology SIRM, SIRM Foundation Milan 20122, Italy
| | - Antonella Petrillo
- Department of Radiology, Istituto Nazionale Tumori –IRCCS- Fondazione G. Pascale, Naples 80131, Italy
| | - Francesco Izzo
- Department of Surgery, Istituto Nazionale Tumori –IRCCS- Fondazione G. Pascale, Naples 80131, Italy
| |
Collapse
|
43
|
Cao W, Zeng Z, Pan R, Wu H, Zhang X, Chen H, Nie Y, Yu Z, Lei S. Hypoxia-Related Gene FUT11 Promotes Pancreatic Cancer Progression by Maintaining the Stability of PDK1. Front Oncol 2021; 11:675991. [PMID: 34221996 PMCID: PMC8247946 DOI: 10.3389/fonc.2021.675991] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/27/2021] [Indexed: 11/13/2022] Open
Abstract
Background Hypoxia is associated with the development of pancreatic cancer (PC). However, genes associated with hypoxia response and their regulatory mechanism in PC cells were unclear. The current study aims to investigate the role of the hypoxia associated gene fucosyltransferase 11 (FUT11) in the progression of PC. Methods In the preliminary study, bioinformatics analysis predicted FUT11 as a key hypoxia associated gene in PC. The expression of FUT11 in PC was evaluated using quantitative real-time PCR (qRT-PCR), Western blot and immunohistochemistry. The effects of FUT11 on PC cells proliferation and migration under normoxia and hypoxia were evaluated using Cell Counting Kit 8, 5-ethynyl-2'-deoxyuridine (EDU) assay, colony formation assay and transwell assay. The effects of FUT11 in vivo was examined in mouse tumor models of liver metastasis and subcutaneous xenograft. Furthermore, Western blot, luciferase assay and immunoprecipitation were performed to explore the regulatory relationship among FUT11, hypoxia-inducible factor 1α (HIF1α) and pyruvate dehydrogenase kinase 1 (PDK1) in PC. Results FUT11 was markedly increased of PC cells with hypoxia, upregulated in the PC clinical tissues, and predicted a poor outcome of PC patients. Inhibition of FUT11 reduced PC cell growth and migratory ability of PC cells under normoxia and hypoxia conditions in vitro, and growth and tumor cell metastasis in vivo. FUT11 bound to PDK1 and regulated the expression PDK1 under normoxia and hypoxia. FUT11 interacted with PDK1 and decreased the ubiquitination of PDK1, lead to the activation of AKT/mTOR signaling pathway. FUT11 knockdown significantly increased the degradation of PDK1 under hypoxia, while treatment with MG132 can relieve the degradation of PDK1 induced by FUT11 knockdown. Overexpression of PDK1 in PC cells under hypoxia conditions reversed the suppressive impacts of FUT11 knockdown on PC cell growth and migration. In addition, HIF1α bound to the promoter of FUT11 and increased its expression, as well as co-expressed with FUT11 in PC tissues. Furthermore, overexpression of FUT11 partially rescued the suppressive effects of HIF1α knockdown on PC cell growth and migration in hypoxia condition. Conclusion Our data implicate that hypoxia-induced FUT11 contributes to proliferation and metastasis of PC by maintaining the stability of PDK1, thus mediating activation of AKT/mTOR signaling pathway, and suggest that FUT11 could be a novel and effective target for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Wenpeng Cao
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Zhirui Zeng
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Runsang Pan
- Department of Orthopedics, Guiyang Maternal and Child Health Care Hospital, Guiyang, China
| | - Hao Wu
- Department of Pediatric Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xiangyan Zhang
- NHC Key Laboratory of Pulmonary, Guizhou Provincial People's Hospital, Guiyang, China
| | - Hui Chen
- NHC Key Laboratory of Pulmonary, Guizhou Provincial People's Hospital, Guiyang, China
| | - Yingjie Nie
- The Clinical Lab Center, Guizhou Provincial People's Hospital, Guiyang, China
| | - Zijiang Yu
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Shan Lei
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| |
Collapse
|
44
|
Han X, Ma L, Gu J, Wang D, Li J, Lou W, Saiyin H, Fu D. Basal microvilli define the metabolic capacity and lethal phenotype of pancreatic cancer. J Pathol 2021; 253:304-314. [PMID: 33159698 PMCID: PMC7898529 DOI: 10.1002/path.5588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/22/2020] [Accepted: 11/02/2020] [Indexed: 12/28/2022]
Abstract
Apical microvilli of polarized epithelial cells govern the absorption of metabolites and the transport of fluid in tissues. Previously, we reported that tall and dense basal microvilli present on the endothelial cells of pancreatic cancers, a lethal malignancy with a high metabolism and unusual hypomicrovascularity, contain nutrient trafficking vesicles and glucose; their length and density were related to the glucose uptake of pancreatic cancers in a small-scale analysis. However, the implications of basal microvilli on pancreatic cancers are unknown. Here, we evaluated the clinical implications of basal microvilli in 106 pancreatic cancers. We found that basal microvilli are a dominant change in pancreatic cancers. The presence of longer and denser basal microvilli on the microvessels in pancreatic cancer tissues positively correlated with increased glucose uptake and higher metastatic (or invasive) and proliferative potentials of neoplastic cells and vice versa. Clinically, postoperative patients with longer and denser basal microvilli were more prone to unfavorable pathological characteristics and dismal prognoses. They were even more refractory to adjuvant therapy than those with shorter and thinner basal microvilli were. Our findings show that basal microvilli define the metabolic capacity and lethal phenotype of pancreatic cancers. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Xu Han
- Department of General SurgeryZhongshan Hospital, Fudan UniversityShanghaiPR China
| | - Lixiang Ma
- Department of Anatomy, Histology & Embryology, School of Medical SciencesFudan UniversityShanghaiPR China
| | - Jichun Gu
- Department of Pancreatic Surgery, Pancreatic Disease InstituteHuashan Hospital, Fudan UniversityShanghaiPR China
| | - Dansong Wang
- Department of General SurgeryZhongshan Hospital, Fudan UniversityShanghaiPR China
| | - Ji Li
- Department of Pancreatic Surgery, Pancreatic Disease InstituteHuashan Hospital, Fudan UniversityShanghaiPR China
| | - Wenhui Lou
- Department of General SurgeryZhongshan Hospital, Fudan UniversityShanghaiPR China
| | - Hexige Saiyin
- State Key Laboratory of Genetic Engineering, School of Life SciencesFudan UniversityShanghaiPR China
| | - Deliang Fu
- Department of Pancreatic Surgery, Pancreatic Disease InstituteHuashan Hospital, Fudan UniversityShanghaiPR China
| |
Collapse
|
45
|
Schmiechen ZC, Stromnes IM. Mechanisms Governing Immunotherapy Resistance in Pancreatic Ductal Adenocarcinoma. Front Immunol 2021; 11:613815. [PMID: 33584701 PMCID: PMC7876239 DOI: 10.3389/fimmu.2020.613815] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 12/10/2020] [Indexed: 01/18/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is a lethal malignancy with an overall 5-year survival rate of 10%. Disease lethality is due to late diagnosis, early metastasis and resistance to therapy, including immunotherapy. PDA creates a robust fibroinflammatory tumor microenvironment that contributes to immunotherapy resistance. While previously considered an immune privileged site, evidence demonstrates that in some cases tumor antigen-specific T cells infiltrate and preferentially accumulate in PDA and are central to tumor cell clearance and long-term remission. Nonetheless, PDA can rapidly evade an adaptive immune response using a myriad of mechanisms. Mounting evidence indicates PDA interferes with T cell differentiation into potent cytolytic effector T cells via deficiencies in naive T cell priming, inducing T cell suppression or promoting T cell exhaustion. Mechanistic research indicates that immunotherapy combinations that change the suppressive tumor microenvironment while engaging antigen-specific T cells is required for treatment of advanced disease. This review focuses on recent advances in understanding mechanisms limiting T cell function and current strategies to overcome immunotherapy resistance in PDA.
Collapse
Affiliation(s)
- Zoe C. Schmiechen
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Ingunn M. Stromnes
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, MN, United States
- Center for Genome Engineering, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
46
|
Campbell DJ, Isch EL, Kozak GM, Yeo CJ. Primary Pancreatic Signet Ring Cell Carcinoma: A Case Report and Review of the Literature. J Pancreat Cancer 2021; 7:1-7. [PMID: 33569523 PMCID: PMC7869882 DOI: 10.1089/pancan.2020.0013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2020] [Indexed: 11/12/2022] Open
Abstract
Background: Primary pancreatic signet ring cell carcinoma (PPSRCC) is a rare (<1%) poorly reported histopathological variant of pancreatic cancer with ill-defined treatment guidelines. Herein, we describe a case of nonmetastatic PPSRCC in a 45-year-old female. Presentation: A 45-year-old female presented with 3 weeks of abdominal pain radiating to her back. Other pertinent positives included a 20-pound (9.1-kilogram) weight loss and jaundice, with a known 30-pack-year smoking history. CT scan revealed a 4.6 × 3.6 cm hypoattenuating mass in the head of the pancreas (HOP) with dilatation of the common bile duct. Total bilirubin at presentation was elevated, and a biliary stent was placed endoscopically. Subsequent endoscopic ultrasonography revealed a periampullary ulcerated mass involving the HOP and second portion of the duodenum, with pathology revealing poorly differentiated adenocarcinoma with mucinous background and focal signet ring cells. A classic pancreatoduodenectomy (Whipple procedure) was performed. Final pathology revealed a poorly differentiated (G3) pT3/pN2/pM0 PPSRCC with 11 of 16 positive specimen lymph nodes. The tumor had evidence of both KRAS and TP53 mutations and expressed an MUC1+/MUC2-/MUC5AC+ immunophenotype. Medical oncology recommended a 6-month course of adjuvant modified-dose FOLFIRINOX therapy. Conclusion: This report highlights the need for further research into the pathogenesis of gastrointestinal signet ring cell carcinoma to identify and study therapeutic targets that can eventually be translated to PPSRCC treatment. Given the paucity of PPSRCC, adjuvant therapy candidates follow the current literature on more common pancreatic cancer subtypes to guide treatment.
Collapse
Affiliation(s)
- Daniel J Campbell
- Department of Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Emily L Isch
- Department of Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Geoffrey M Kozak
- Department of Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Charles J Yeo
- Department of Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
47
|
Li Z, Xie X, Tan G, Xie F, Liu N, Li W, Sun X. Disulfiram Synergizes with SRC Inhibitors to Suppress the Growth of Pancreatic Ductal Adenocarcinoma Cells in Vitro and in Vivo. Biol Pharm Bull 2021; 44:1323-1331. [PMID: 34471060 DOI: 10.1248/bpb.b21-00353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Disulfiram (DSF), an old anti-alcoholism drug, has emerged as a candidate for drug repurposing in oncology. In exploratory studies on its therapeutic effects, we unexpectedly discovered that DSF increased the phosphorylation of SRC, a proto-oncogene tyrosine-protein kinase elevated in 70% of pancreatic ductal adenocarcinoma (PDAC) cases. This serendipitous and novel finding led to our hypothesis for the current study which proposes DSF may synergize with SRC inhibitors in suppressing PDAC. Human PDAC PANC-1 and BXPC-3 cells were incubated with DSF chelated with copper (Cu2+), SRC inhibitors (PP2 and dasatinib), or transfected with lentiviral short hairpin RNA (shRNA), and their proliferation and apoptosis were analyzed. A xenograft model was employed to verify the in vitro results. The expression of key molecules was detected. DSF significantly inhibited cell proliferation and induced cell apoptosis by increasing the cleavage of poly ADP ribose polymerase (PARP), downregulating Bcl-2 and upregulating p27 in concentration- and time-dependent manners. DSF had little effect on signal transducer and activator of transcription 3 (STAT3) expression but inhibited its phosphorylation. DSF did not alter SRC expression but significantly increased its phosphorylation through upregulating actin filament associated protein 1 like 2 (AFAP1L2). DSF exhibited a synergistic effect, as analyzed by drug coefficient interactions, with either PP2, or dasatinib, or SRC depletion in suppressing PDAC cells in vitro and/or in vivo. The present results indicate DSF is a potential therapeutic drug, particularly when it is combined with SRC inhibitors, and warrant further studies on the pharmacological utility of DSF as a promising adjunct therapy for the treatment of PDAC.
Collapse
Affiliation(s)
- Ziyi Li
- Hepatosplenic Surgery Center, the First Affiliated Hospital of Harbin Medical University
| | - Xiangjun Xie
- Department of Gastroenterology, Qingdao Municipal Hospital Affiliated to Qingdao University
| | - Gang Tan
- Department of General Surgery, the Fourth Affiliated Hospital of Harbin Medical University
| | - Fangyu Xie
- Department of Cardiology, Qingdao Municipal Hospital Affiliated to Qingdao University
| | - Nianjiao Liu
- Department of Endocrinology, the First Hospital Affiliated of Harbin Medical University
| | - Weidong Li
- Hepatosplenic Surgery Center, the First Affiliated Hospital of Harbin Medical University
- Department of General Surgery, the Fourth Affiliated Hospital of Harbin Medical University
| | - Xueying Sun
- Hepatosplenic Surgery Center, the First Affiliated Hospital of Harbin Medical University
| |
Collapse
|
48
|
Emerging roles for the IL-6 family of cytokines in pancreatic cancer. Clin Sci (Lond) 2020; 134:2091-2115. [PMID: 32808663 PMCID: PMC7434989 DOI: 10.1042/cs20191211] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 07/29/2020] [Accepted: 08/07/2020] [Indexed: 12/13/2022]
Abstract
Pancreatic cancer has one of the poorest prognoses of all malignancies, with little improvement in clinical outcome over the past 40 years. Pancreatic ductal adenocarcinoma is responsible for the vast majority of pancreatic cancer cases, and is characterised by the presence of a dense stroma that impacts therapeutic efficacy and drives pro-tumorigenic programs. More specifically, the inflammatory nature of the tumour microenvironment is thought to underlie the loss of anti-tumour immunity and development of resistance to current treatments. Inflammatory pathways are largely mediated by the expression of, and signalling through, cytokines, chemokines, and other cellular messengers. In recent years, there has been much attention focused on dual targeting of cancer cells and the tumour microenvironment. Here we review our current understanding of the role of IL-6, and the broader IL-6 cytokine family, in pancreatic cancer, including their contribution to pancreatic inflammation and various roles in pancreatic cancer pathogenesis. We also summarise potential opportunities for therapeutic targeting of these pathways as an avenue towards combating poor patient outcomes.
Collapse
|
49
|
Granata V, Grassi R, Fusco R, Setola SV, Palaia R, Belli A, Miele V, Brunese L, Grassi R, Petrillo A, Izzo F. Assessment of Ablation Therapy in Pancreatic Cancer: The Radiologist's Challenge. Front Oncol 2020; 10:560952. [PMID: 33330028 PMCID: PMC7731725 DOI: 10.3389/fonc.2020.560952] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 10/30/2020] [Indexed: 12/12/2022] Open
Abstract
This article provides an overview of imaging assessment of ablated pancreatic cancer. Only studies reporting radiological assessment on pancreatic ablated cancer were retained. We found 16 clinical studies that satisfied the inclusion criteria. Radiofrequency ablation and irreversible electroporation have become established treatment modalities because of their efficacy, low complication rates, and availability. Microwave Ablation (MWA) has several advantages over radiofrequency ablation (RFA), which may make it more attractive to treat pancreatic cancer. Electrochemotherapy (ECT) is a very interesting emerging technique, characterized by low complication rate and safety profile. According to the literature, the assessment of the effectiveness of ablative therapies is difficult by means of the Response Evaluation Criteria in Solid Tumors (RECIST) criteria that are not suitable to evaluate the treatment response considering that are related to technique used, the timing of reassessment, and the imaging procedure being used to evaluate the efficacy. RFA causes various appearances on imaging in the ablated zone, correlating to the different effects, such as interstitial edema, hemorrhage, carbonization, necrosis, and fibrosis. Irreversible electroporation (IRE) causes the creation of pores within the cell membrane causing cell death. Experimental studies showed that Diffusion Weigthed Imaging (DWI) extracted parameters could be used to detect therapy effects. No data about functional assessment post MWA is available in literature. Morphologic data extracted by Computed Tomography (CT) or Magnetic Resonance Imaging (MRI) do not allow to differentiate partial, complete, or incomplete response after ECT conversely to functional parameters, obtained with Position Emission Tomography (PET), MRI, and CT.
Collapse
Affiliation(s)
- Vincenza Granata
- Radiology Division, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Naples, Italy
| | - Roberta Grassi
- Radiology Division, Universita’ Degli Studi Della Campania Luigi Vanvitelli, Naples, Italy
| | - Roberta Fusco
- Radiology Division, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Naples, Italy
| | - Sergio Venanzio Setola
- Radiology Division, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Naples, Italy
| | - Raffaele Palaia
- Hepatobiliary Surgical Oncology Division, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Naples, Italy
| | - Andrea Belli
- Hepatobiliary Surgical Oncology Division, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Naples, Italy
| | - Vittorio Miele
- Department of Radiology, Careggi University Hospital, Florence, Italy
| | - Luca Brunese
- Department of Medicine and Health Sciences “V. Tiberio,” University of Molise, Campobasso, Italy
| | - Roberto Grassi
- Radiology Division, Universita’ Degli Studi Della Campania Luigi Vanvitelli, Naples, Italy
| | - Antonella Petrillo
- Radiology Division, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Naples, Italy
| | - Francesco Izzo
- Hepatobiliary Surgical Oncology Division, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Naples, Italy
| |
Collapse
|
50
|
Gao CT, Ren J, Yu J, Li SN, Guo XF, Zhou YZ. KIF23 enhances cell proliferation in pancreatic ductal adenocarcinoma and is a potent therapeutic target. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1394. [PMID: 33313139 PMCID: PMC7723550 DOI: 10.21037/atm-20-1970] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Background In recent research, high expression of kinesin family member 23 (KIF23), one of the kinesin motor proteins involved in the regulation of cytokinesis, has been shown to be related to poor prognosis in glioma and paclitaxel-resistant gastric cancer, as a results of the enhancement of proliferation, migration, and invasion. In this study, we analyzed the role of KIF23 in the progression of pancreatic ductal adenocarcinoma. Methods A bioinformatic method was used to analyze the KIF23 mRNA level in pancreatic tumor tissues compared with normal pancreatic tissues and to analyze the connection between high KIF23 expression and prognosis. We examined the expression of KIF23 using immunohistochemistry and analyzed the connection between the expression of KIF23 and clinicopathological features in pancreatic ductal adenocarcinoma patients. In addition, a colony formation assay, MTT assay, and western blot assay were performed in vitro, along with a mouse xenograft model in vivo, to analyze the effect of KIF23 on proliferation. Further, the correlation between KIF23 and CDCA8 was analyzed by TCGA and immunohistochemical data. Results Bioinformatic results showed that KIF23 mRNA expression was higher in pancreatic tumor tissues than in normal pancreatic tissues and a poor prognosis has been linked to the high expression of KIF23. Immunohistochemistry revealed that KIF23 was highly expressed at the protein level and high expression of KIF23 correlated with adverse clinicopathological features. Our experimental results demonstrated that knockdown of KIF23 could inhibit the proliferation of pancreatic cells. Further, a positive correlation between KIF23 and CDCA8 expression existed, and KIF23 might promote pancreatic cancer proliferation by affecting CDCA8 expression. Conclusions Our data showed that high expression of KIF23 is associated with a poor prognosis, and KIF23 might be a potential therapeutic target for pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Chun-Tao Gao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jin Ren
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Jie Yu
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China.,The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Sheng-Nan Li
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xiao-Fan Guo
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yi-Zhang Zhou
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|