1
|
Yamaguchi T, Kitahara S, Matsui A, Okamoto J, Muragaki Y, Masamune K. HIFU induces reprogramming of the tumor immune microenvironment in a pancreatic cancer mouse model. Med Mol Morphol 2025; 58:137-148. [PMID: 39870899 DOI: 10.1007/s00795-025-00419-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/06/2025] [Indexed: 01/29/2025]
Abstract
This study evaluates the effects of different high-intensity focused ultrasound irradiation (HIFU) methods on local tumor suppression and systemic antitumor effects, including the abscopal effect, in a mouse model of pancreatic cancer. To ascertain the efficacy of the treatment, pancreatic cancer cells were injected into the thighs of mice and HIFU was applied on one side using continuous waves or trigger pulse waves. Then, tumor volume, tissue changes, and immune marker levels were analyzed. Both the irradiation methods suppressed tumor growth, with the trigger pulse wave showing stronger effects and the difference being significant. Tumor suppression was also observed on the non-irradiated side, suggesting an abscopal effect. These effects vary depending on the irradiation method used. We conclude that HIFU induces both local tumor suppression and a systemic immune response, suggesting its potential for combination with immunotherapy for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Toshihiro Yamaguchi
- Faculty of Advanced Techno-Surgery (FATS), Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku, Tokyo, 162-8666, Japan
| | - Shuji Kitahara
- Faculty of Advanced Techno-Surgery (FATS), Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku, Tokyo, 162-8666, Japan.
| | - Aya Matsui
- Department of Vascular Physiology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Jun Okamoto
- SONIRE Therapeutics Inc., Nihonbashi Life Science, Building 2 803, 3-11-5 Nihonbashi Honcho, Chuo-Ku 103-0023, Tokyo, Japan
| | - Yoshihiro Muragaki
- Faculty of Advanced Techno-Surgery (FATS), Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku, Tokyo, 162-8666, Japan
- Center for Advanced Medical Engineering Research and Development, Kobe University, 1-5-1 Minatojima Minamimachi, Chuo-Ku, Kobe, Hyogo, 650-0047, Japan
| | - Ken Masamune
- Faculty of Advanced Techno-Surgery (FATS), Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku, Tokyo, 162-8666, Japan.
| |
Collapse
|
2
|
Liao H, Huang C, Liu C, Zhang J, Tao F, Liu H, Liang H, Hu X, Li Y, Chen S, Li Y. Deep learning-based MVIT-MLKA model for accurate classification of pancreatic lesions: a multicenter retrospective cohort study. LA RADIOLOGIA MEDICA 2025; 130:508-523. [PMID: 39832039 DOI: 10.1007/s11547-025-01949-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/01/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Accurate differentiation between benign and malignant pancreatic lesions is critical for effective patient management. This study aimed to develop and validate a novel deep learning network using baseline computed tomography (CT) images to predict the classification of pancreatic lesions. METHODS This retrospective study included 864 patients (422 men, 442 women) with confirmed histopathological results across three medical centers, forming a training cohort, internal testing cohort, and external validation cohort. A novel hybrid model, Multi-Scale Large Kernel Attention with Mobile Vision Transformer (MVIT-MLKA), was developed, integrating CNN and Transformer architectures to classify pancreatic lesions. The model's performance was compared with traditional machine learning methods and advanced deep learning models. We also evaluated the diagnostic accuracy of radiologists with and without the assistance of the optimal model. Model performance was assessed through discrimination, calibration, and clinical applicability. RESULTS The MVIT-MLKA model demonstrated superior performance in classifying pancreatic lesions, achieving an AUC of 0.974 (95% CI 0.967-0.980) in the training set, 0.935 (95% CI 0.915-0.954) in the internal testing set, and 0.924 (95% CI 0.902-0.945) in the external validation set, outperforming traditional models and other deep learning models (P < 0.05). Radiologists aided by the MVIT-MLKA model showed significant improvements in diagnostic accuracy and sensitivity compared to those without model assistance (P < 0.05). Grad-CAM visualization enhanced model interpretability by effectively highlighting key lesion areas. CONCLUSION The MVIT-MLKA model efficiently differentiates between benign and malignant pancreatic lesions, surpassing traditional methods and significantly improving radiologists' diagnostic performance. The integration of this advanced deep learning model into clinical practice has the potential to reduce diagnostic errors and optimize treatment strategies.
Collapse
Affiliation(s)
- Hongfan Liao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Cheng Huang
- College of Computer and Information Science, Southwest University, Chongqing, 400715, China
| | - Chunhua Liu
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Jiao Zhang
- Department of Radiology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fengming Tao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Haotian Liu
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hongwei Liang
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiaoli Hu
- Department of Radiology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yi Li
- Department of Radiology, The Third People's Hospital of Chengdu, Chengdu, China
| | - Shanxiong Chen
- College of Computer and Information Science, Southwest University, Chongqing, 400715, China.
| | - Yongmei Li
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
3
|
Mejza M, Bajer A, Wanibuchi S, Małecka-Wojciesko E. Can AI Be Useful in the Early Detection of Pancreatic Cancer in Patients with New-Onset Diabetes? Biomedicines 2025; 13:836. [PMID: 40299428 PMCID: PMC12025102 DOI: 10.3390/biomedicines13040836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/12/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Pancreatic cancer is one of the most lethal neoplasms. Despite considerable research conducted in recent decades, not much has been achieved to improve its survival rate. That may stem from the lack of effective screening strategies in increased pancreatic cancer risk groups. One population that may be appropriate for screening is new-onset diabetes (NOD) patients. Such a conclusion stems from the fact that pancreatic cancer can cause diabetes several months before diagnosis. The most widely used screening tool for this population, the ENDPAC (Enriching New-Onset Diabetes for Pancreatic Cancer) model, has not achieved satisfactory results in validation trials. This provoked the first attempts at using artificial intelligence (AI) to create larger, multi-parameter models that could better identify the at-risk population, which would be suitable for screening. The results shown by the authors of these trials seem promising. Nonetheless, the number of publications is limited, and the downfalls of using AI are not well highlighted. This narrative review presents a summary of previous publications, recent advancements and feasible solutions for effective screening of patients with NOD for pancreatic cancer.
Collapse
Affiliation(s)
- Maja Mejza
- Department of Digestive Tract Diseases, Medical University of Lodz, 90-153 Lodz, Poland; (M.M.); (A.B.)
| | - Anna Bajer
- Department of Digestive Tract Diseases, Medical University of Lodz, 90-153 Lodz, Poland; (M.M.); (A.B.)
| | - Sora Wanibuchi
- Aichi Medical University Hospital, Nagakute 480-1195, Japan;
| | - Ewa Małecka-Wojciesko
- Department of Digestive Tract Diseases, Medical University of Lodz, 90-153 Lodz, Poland; (M.M.); (A.B.)
| |
Collapse
|
4
|
Li CF, Bai LY, Wei Y, Lee HH, Yang R, Yao J, Wang H, Wang YN, Chang WC, Shen YC, Wang SC, Chou CW, Fu J, Ling J, Chu YY, Chiu CF, Wang M, Yu D, Chiao PJ, Liang H, Maitra A, Ying H, Hung MC. All-trans retinoic acid-mediated ADAR1 degradation synergizes with PD-1 blockade to suppress pancreatic cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.20.619300. [PMID: 39484589 PMCID: PMC11527022 DOI: 10.1101/2024.10.20.619300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
As a double-stranded RNA-editing enzyme and an interferon-stimulated gene, double-stranded RNA-specific adenosine deaminase (ADAR1) suppresses interferon signaling and contributes to immunotherapy resistance. Suppression of ADAR1 overcomes immunotherapy resistance in preclinical models, but has not yet been translated to clinical settings. By conducting a screening of a subset of the FDA-approved drugs, we found that all-trans retinoic acid (ATRA, also known as tretinoin) caused ADAR1 protein degradation through ubiquitin-proteasome pathways and concomitantly increased PD-L1 expression in pancreatic and breast cancers. In addition, the combination of ATRA and PD-1 blockade reprogrammed the tumor microenvironment and unleashed antitumor immunity and thereby impeded tumor growth in pancreatic cancer mouse models. In a pilot clinical trial, a higher dose of ATRA plus the anti-PD-1 antibody nivolumab prolonged median overall survival in patients with chemotherapy-resistant pancreatic cancer compared to a lower dose of the same regimen. In this study, ATRA was the first drug to be found to cause ADAR1 degradation. We propose translation of a promising 2-pronged antitumor strategy using ATRA and nivolumab to convert immunologically "cold" into "hot" tumors susceptible to immune checkpoint blockade.
Collapse
|
5
|
Nichkaode P, Dwivedi D. Cutaneous Metastasis: A Rare Presentation of Pancreatic Adenocarcinoma. Cureus 2024; 16:e69767. [PMID: 39429388 PMCID: PMC11491125 DOI: 10.7759/cureus.69767] [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: 07/02/2024] [Accepted: 09/18/2024] [Indexed: 10/22/2024] Open
Abstract
Pancreatic adenocarcinomas are highly malignant tumors. The liver, peritoneum, and, very rarely, the skin are the sites of pancreatic metastases. Cutaneous metastasis is rare, with scalp metastasis being seldomly found. We report the case of an elderly woman who initially presented with ulcerated scalp swellings. The immunohistochemistry analysis of the swollen scalp confirmed the presence of a far-metastasized adenocarcinoma. Imaging of the abdomen performed due to increasing cholestatic jaundice revealed adenocarcinoma located at the head of the pancreas. Cholestatic jaundice was treated by trans-biliary percutaneous drainage. She is currently on a chemotherapy regimen. The patient had a follow-up appointment one month ago for the fourth cycle of gemcitabine and paclitaxel and will continue to follow up until eight cycles of chemotherapy sessions. Cutaneous metastases resulting from pancreatic cancer are a rare occurrence and may occasionally present as the only visible symptom; therefore, they should be included in the differential diagnosis of skin lesions.
Collapse
Affiliation(s)
- Prabhat Nichkaode
- Department of General Surgery, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth (Deemed to be University), Pune, IND
| | - Divyansh Dwivedi
- Department of General Surgery, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth (Deemed to be University), Pune, IND
| |
Collapse
|
6
|
Wang Z, Chen H, Cai X, Bu H, Lin S. Andrographolide induces protective autophagy and targeting DJ-1 triggers reactive oxygen species-induced cell death in pancreatic cancer. PeerJ 2024; 12:e17619. [PMID: 38952980 PMCID: PMC11216212 DOI: 10.7717/peerj.17619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 05/31/2024] [Indexed: 07/03/2024] Open
Abstract
Background Andrographolide (Andro), an extract of Andrographis paniculate (Burm.f.) Wall. ex Nees (Acanthaceae), possesses diverse biologically active properties. However, the precise mechanisms and effects of Andro on pancreatic cancer (PC) remain unclear. Methods The cytotoxic potential of Andro and underlying mechanism towards PC cells was investigated through in vitro experiments and a xenograft mouse model. PC cells were first subjected to varying concentrations of Andro. The reactive oxygen species (ROS) was assessed using flow cytometry and DCFH-DA staining. The apoptosis rate was detected by flow cytometry. Additionally, western blot was applied to evaluate the expression levels of cleaved-caspase-3, DJ-1, LC3-I, LC3-II, and p62. To further elucidate the involvement of ROS accumulation and autophagy, we employed N-acetylcysteine as a scavenger of ROS and 3-Methyladenine as an inhibitor of autophagy. Results Andro demonstrated potent anti-proliferative effects on PC cells and induced apoptosis, both in vitro and in vivo. The cytotoxicity of Andro on PC cells was counteracted by DJ-1 overexpression. The reduction in DJ-1 expression caused by Andro led to ROS accumulation, subsequently inhibiting the growth of PC cells. Furthermore, Andro stimulated cytoprotective autophagy, thus weakening the antitumor effect. Pharmacological blockade of autophagy further enhanced the antitumor efficacy of Andro. Conclusion Our study indicated that ROS accumulation induced by the DJ-1 reduction played a key role in Andro-mediated PC cell inhibition. Furthermore, the protective autophagy induced by the Andro in PC cells is a mechanism that needs to be addressed in future studies.
Collapse
Affiliation(s)
- Zhaohong Wang
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hui Chen
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xufan Cai
- Zhejiang Chinese Medical University, Hanzhou, China
| | - Heqi Bu
- Department of Surgery, Tongde Hospital of Zhejiang Province, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Shengzhang Lin
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, China
| |
Collapse
|
7
|
Zheng J, He B, Deng L, Zhu X, Li R, Chen K, Zheng C, Wang D, Wang Y, Yu C, Chen G. Prognostic value of diffuse reduction of spleen density on postoperative survival of pancreatic ductal adenocarcinoma: A retrospective study. Asia Pac J Clin Oncol 2024; 20:275-284. [PMID: 36748794 DOI: 10.1111/ajco.13936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 07/05/2022] [Accepted: 01/07/2023] [Indexed: 02/08/2023]
Abstract
PURPOSE It is difficult to predict the prognosis of patients with pancreatic ductal adenocarcinoma (PDAC) before radical operation. The purpose of this study was to explore the connection between the diffuse reduction of spleen density on computed tomography (DROSD) and the postoperative prognosis of patients with PDAC. PATIENTS AND METHODS A total of 160 patients with PDAC who underwent radical surgery in the First Affiliated Hospital of Wenzhou Medical University were enrolled. Cox regression analysis was used to cast the overall survival (OS) and evaluate the prognostic factors. Nomogram was used to forecast the possibility of 1-year, 3-year, and 5-year OS. The prediction accuracy and clinical net benefit are performed by concordance index (C-index), calibration curve, time-dependent receiver operating characteristics (tdROC), and decision curve analysis. RESULTS In multivariable Cox analysis, DROSD is independently related to OS. Advanced age, TNM stage, neutrophil/lymphocyte ratio, and severe complications were also independent prognostic factors. The calibration curves of nomogram showed optimal agreement between prediction and observation. The C-index of nomogram is 0.662 (95%CI, 0.606-0.754). The area under tdROC curve for a 3-year OS of nomogram is 0.770. CONCLUSION DROSD is an independent risk factor for an OS of PDAC. We developed a nomogram that combined imaging features, clinicopathological factors, and systemic inflammatory response to provide a personalized risk assessment for patients with PDAC.
Collapse
Affiliation(s)
- Jiuyi Zheng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Bangjie He
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Liming Deng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Xuewen Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Rizhao Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Kaiyu Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Chongming Zheng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Daojie Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Yi Wang
- Department of Epidemiology and Biostatistics, School of Public Health and Management, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Chang Yu
- Department of Interventional Therapy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Gang Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| |
Collapse
|
8
|
Reshkin SJ, Cardone RA, Koltai T. Genetic Signature of Human Pancreatic Cancer and Personalized Targeting. Cells 2024; 13:602. [PMID: 38607041 PMCID: PMC11011857 DOI: 10.3390/cells13070602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/13/2024] Open
Abstract
Pancreatic cancer is a highly lethal disease with a 5-year survival rate of around 11-12%. Surgery, being the treatment of choice, is only possible in 20% of symptomatic patients. The main reason is that when it becomes symptomatic, IT IS the tumor is usually locally advanced and/or has metastasized to distant organs; thus, early diagnosis is infrequent. The lack of specific early symptoms is an important cause of late diagnosis. Unfortunately, diagnostic tumor markers become positive at a late stage, and there is a lack of early-stage markers. Surgical and non-surgical cases are treated with neoadjuvant and/or adjuvant chemotherapy, and the results are usually poor. However, personalized targeted therapy directed against tumor drivers may improve this situation. Until recently, many pancreatic tumor driver genes/proteins were considered untargetable. Chemical and physical characteristics of mutated KRAS are a formidable challenge to overcome. This situation is slowly changing. For the first time, there are candidate drugs that can target the main driver gene of pancreatic cancer: KRAS. Indeed, KRAS inhibition has been clinically achieved in lung cancer and, at the pre-clinical level, in pancreatic cancer as well. This will probably change the very poor outlook for this disease. This paper reviews the genetic characteristics of sporadic and hereditary predisposition to pancreatic cancer and the possibilities of a personalized treatment according to the genetic signature.
Collapse
Affiliation(s)
- Stephan J. Reshkin
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, 70125 Bari, Italy;
| | - Rosa Angela Cardone
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, 70125 Bari, Italy;
| | - Tomas Koltai
- Oncomed, Via Pier Capponi 6, 50132 Florence, Italy
| |
Collapse
|
9
|
Joseph AM, Al Aiyan A, Al-Ramadi B, Singh SK, Kishore U. Innate and adaptive immune-directed tumour microenvironment in pancreatic ductal adenocarcinoma. Front Immunol 2024; 15:1323198. [PMID: 38384463 PMCID: PMC10879611 DOI: 10.3389/fimmu.2024.1323198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/11/2024] [Indexed: 02/23/2024] Open
Abstract
One of the most deadly and aggressive cancers in the world, pancreatic ductal adenocarcinoma (PDAC), typically manifests at an advanced stage. PDAC is becoming more common, and by the year 2030, it is expected to overtake lung cancer as the second greatest cause of cancer-related death. The poor prognosis can be attributed to a number of factors, including difficulties in early identification, a poor probability of curative radical resection, limited response to chemotherapy and radiotherapy, and its immunotherapy resistance. Furthermore, an extensive desmoplastic stroma that surrounds PDAC forms a mechanical barrier that prevents vascularization and promotes poor immune cell penetration. Phenotypic heterogeneity, drug resistance, and immunosuppressive tumor microenvironment are the main causes of PDAC aggressiveness. There is a complex and dynamic interaction between tumor cells in PDAC with stromal cells within the tumour immune microenvironment. The immune suppressive microenvironment that promotes PDAC aggressiveness is contributed by a range of cellular and humoral factors, which itself are modulated by the cancer. In this review, we describe the role of innate and adaptive immune cells, complex tumor microenvironment in PDAC, humoral factors, innate immune-mediated therapeutic advances, and recent clinical trials in PDAC.
Collapse
Affiliation(s)
- Ann Mary Joseph
- Department of Veterinary Medicine (CAVM), United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ahmad Al Aiyan
- Department of Veterinary Medicine (CAVM), United Arab Emirates University, Al Ain, United Arab Emirates
| | - Basel Al-Ramadi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Shiv K. Singh
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center, Goettingen, Germany
| | - Uday Kishore
- Department of Veterinary Medicine (CAVM), United Arab Emirates University, Al Ain, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
10
|
Augustinus S, van Laarhoven HWM, Cirkel GA, de Groot JWB, Groot Koerkamp B, Macarulla T, Melisi D, O’Reilly EM, van Santvoort HC, Mackay TM, Besselink MG, Wilmink JW. Timing of Initiation of Palliative Chemotherapy in Asymptomatic Patients with Metastatic Pancreatic Cancer: An International Expert Survey and Case-Vignette Study. Cancers (Basel) 2023; 15:5603. [PMID: 38067306 PMCID: PMC10705283 DOI: 10.3390/cancers15235603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 02/15/2024] Open
Abstract
Background: The use of imaging, in general, and during follow-up after resection of pancreatic cancer, is increasing. Consequently, the number of asymptomatic patients diagnosed with metastatic pancreatic cancer (mPDAC) is increasing. In these patients, palliative systemic therapy is the only tumor-directed treatment option; hence, it is often immediately initiated. However, delaying therapy in asymptomatic palliative patients may preserve quality of life and avoid therapy-related toxicity, but the impact on survival is unknown. This study aimed to gain insight into the current perspectives and clinical decision=making of experts regarding the timing of treatment initiation of patients with asymptomatic mPDAC. Methods: An online survey (13 questions, 9 case-vignettes) was sent to all first and last authors of published clinical trials on mPDAC over the past 10 years and medical oncologists of the Dutch Pancreatic Cancer Group. Inter-rater variability was determined using the Kappa Light test. Differences in the preferred timing of treatment initiation among countries, continents, and years of experience were analyzed using Fisher's exact test. Results: Overall, 78 of 291 (27%) medical oncologists from 15 countries responded (62% from Europe, 23% from North America, and 15% from Asia-Pacific). The majority of respondents (63%) preferred the immediate initiation of chemotherapy following diagnosis. In 3/9 case-vignettes, delayed treatment was favored in specific clinical contexts (i.e., patient with only one small lung metastasis, significant comorbidities, and higher age). A significant degree of inter-rater variability was present within 7/9 case-vignettes. The recommended timing of treatment initiation differed between continents for 2/9 case-vignettes (22%), in 7/9 (77.9%) comparing the Netherlands with other countries, and based on years of experience for 5/9 (56%). Conclusions: Although the response rate was limited, in asymptomatic patients with mPDAC, immediate treatment is most often preferred. Delaying treatment until symptoms occur is considered in patients with limited metastatic disease, more comorbidities, and higher age.
Collapse
Affiliation(s)
- Simone Augustinus
- Department of Surgery, Location University of Amsterdam, Amsterdam University Medical Center, 1105 Amsterdam, The Netherlands; (S.A.); (M.G.B.)
- Cancer Center Amsterdam, 1105 Amsterdam, The Netherlands
| | - Hanneke W. M. van Laarhoven
- Cancer Center Amsterdam, 1105 Amsterdam, The Netherlands
- Department of Medical Oncology, Location University of Amsterdam, Amsterdam University Medical Center, 1105 Amsterdam, The Netherlands
| | - Geert A. Cirkel
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, 3584 Utrecht, The Netherlands
| | | | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC Cancer Institute, 3015 Rotterdam, The Netherlands
| | - Teresa Macarulla
- Department of Medical Oncology, Vall d’Hebron Unveristy Hospital, Vall d’Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | - Davide Melisi
- Digestive Molecular Clinical Oncology Unit, Univeristy of Verona, 37134 Verona, Italy
| | - Eileen M. O’Reilly
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hjalmar C. van Santvoort
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center, St. Antonius Hospital Nieuwegein, Utrecht University, 3584 Utrech, The Netherlands
| | - Tara M. Mackay
- Department of Surgery, Location University of Amsterdam, Amsterdam University Medical Center, 1105 Amsterdam, The Netherlands; (S.A.); (M.G.B.)
- Cancer Center Amsterdam, 1105 Amsterdam, The Netherlands
| | - Marc G. Besselink
- Department of Surgery, Location University of Amsterdam, Amsterdam University Medical Center, 1105 Amsterdam, The Netherlands; (S.A.); (M.G.B.)
- Cancer Center Amsterdam, 1105 Amsterdam, The Netherlands
| | - Johanna W. Wilmink
- Cancer Center Amsterdam, 1105 Amsterdam, The Netherlands
- Department of Medical Oncology, Location University of Amsterdam, Amsterdam University Medical Center, 1105 Amsterdam, The Netherlands
| |
Collapse
|
11
|
Mazur R, Trna J. Principles of Palliative and Supportive Care in Pancreatic Cancer: A Review. Biomedicines 2023; 11:2690. [PMID: 37893064 PMCID: PMC10603964 DOI: 10.3390/biomedicines11102690] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/24/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Pancreatic adenocarcinoma (PDAC) is well known for its poor survival time. Clinical symptoms are painless jaundice or abdominal or back pain. Less specific symptoms often appear that make diagnosis difficult, e.g., weight loss, loss of appetite, nausea and vomiting, and general weakness. Only 10-20% of patients are diagnosed at an early stage. A cure is practically only possible with a radical surgical operation. In the case of locally advanced findings, neoadjuvant therapy is administered. Among the therapeutic options offered are chemotherapy, radiotherapy (including stereotactic radiotherapy-SBRT), targeted treatment, or immunotherapy. In the case of metastatic disease, of which more than half are present at diagnosis, the goal is to relieve the patient of problems. Metastatic PDAC can cause problems arising from the localization of distant metastases, but it also locally affects the organs it infiltrates. In our review article, we focus on the largest group of patients, those with locally advanced disease and metastatic disease-symptoms related to the infiltration or destruction of the pancreatic parenchyma and the growth of the tumor into the surrounding. Therefore, we deal with biliary or duodenal obstruction, gastric outlet syndrome, bleeding and thromboembolic diseases, pain, depression, and fatigue, as well as pancreatic exocrine insufficiency and malnutrition. Metastatic spread is most often to the liver, peritoneum, or lungs. The presented overview aims to offer current therapeutic options across disciplines. In accordance with modern oncology, a multidisciplinary approach with a procedure tailored to the specific patient remains the gold standard.
Collapse
Affiliation(s)
| | - Jan Trna
- Department of Gastroenterology and Digestive Endoscopy, Masaryk Memorial Cancer Institute, Zluty Kopec 7, 656 53 Brno, Czech Republic;
| |
Collapse
|
12
|
Koltai T. Earlier Diagnosis of Pancreatic Cancer: Is It Possible? Cancers (Basel) 2023; 15:4430. [PMID: 37760400 PMCID: PMC10526520 DOI: 10.3390/cancers15184430] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/31/2023] [Accepted: 08/06/2023] [Indexed: 09/29/2023] Open
Abstract
Pancreatic ductal adenocarcinoma has a very high mortality rate which has been only minimally improved in the last 30 years. This high mortality is closely related to late diagnosis, which is usually made when the tumor is large and has extensively infiltrated neighboring tissues or distant metastases are already present. This is a paradoxical situation for a tumor that requires nearly 15 years to develop since the first founding mutation. Response to chemotherapy under such late circumstances is poor, resistance is frequent, and prolongation of survival is almost negligible. Early surgery has been, and still is, the only approach with a slightly better outcome. Unfortunately, the relapse percentage after surgery is still very high. In fact, early surgery clearly requires early diagnosis. Despite all the advances in diagnostic methods, the available tools for improving these results are scarce. Serum tumor markers permit a late diagnosis, but their contribution to an improved therapeutic result is very limited. On the other hand, effective screening methods for high-risk populations have not been fully developed as yet. This paper discusses the difficulties of early diagnosis, evaluates whether the available diagnostic tools are adequate, and proposes some simple and not-so-simple measures to improve it.
Collapse
Affiliation(s)
- Tomas Koltai
- Hospital del Centro Gallego de Buenos Aires, Buenos Aires C1094, Argentina
| |
Collapse
|
13
|
Cefalì M, Scala I, Pavone G, Helbling D, Hussung S, Fritsch R, Reiner C, Stocker S, Koeberle D, Kissling M, Chianca V, Del Grande F, De Dosso S, Rizzo S. Is Computed-Tomography-Based Body Composition a Reliable Predictor of Chemotherapy-Related Toxicity in Pancreatic Cancer Patients? Cancers (Basel) 2023; 15:4398. [PMID: 37686674 PMCID: PMC10486498 DOI: 10.3390/cancers15174398] [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: 04/14/2023] [Revised: 08/13/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND Malnutrition, loss of weight and of skeletal muscle mass are frequent in pancreatic cancer patients, a majority of which will undergo chemotherapy over the course of their disease. Available data suggest a negative prognostic role of these changes in body composition on disease outcomes; however, it is unclear whether tolerance to chemotherapeutic treatment is similarly and/or negatively affected. We aimed to explore this association by retrospectively assessing changes in body composition and chemotherapy-related toxicity in a cohort of advanced pancreatic cancer patients. METHODS Body composition was evaluated through clinical parameters and through radiological assessment of muscle mass, skeletal muscle area, skeletal muscle index and skeletal muscle density; and an assessment of fat distribution by subcutaneous adipose tissue and visceral adipose tissue. We performed descriptive statistics, pre/post chemotherapy comparisons and uni- and multivariate analyses to assess the relation between changes in body composition and toxicity. RESULTS Toxicity risk increased with an increase of skeletal muscle index (OR: 1.03) and body mass index (OR: 1.07), whereas it decreased with an increase in skeletal muscle density (OR: 0.96). Multivariate analyses confirmed a reduction in the risk of toxicity only with an increase in skeletal muscle density (OR: 0.96). CONCLUSIONS This study suggests that the retrospective analysis of changes in body composition is unlikely to be useful to predict toxicity to gemcitabine-nab-paclitaxel.
Collapse
Affiliation(s)
- Marco Cefalì
- Istituto Oncologico della Svizzera Italiana (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.C.); (G.P.)
| | - Isabel Scala
- Facoltà di Scienze Biomediche, Università della Svizzera Italiana, Via Buffi 13, 6900 Lugano, Switzerland; (I.S.); (F.D.G.); (S.R.)
| | - Giuliana Pavone
- Istituto Oncologico della Svizzera Italiana (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.C.); (G.P.)
| | - Daniel Helbling
- Onkozentrum Zürich, Seestrasse 259, 8038 Zurich, Switzerland;
| | - Saskia Hussung
- Department of Medical Oncology and Hematology, University Hospital of Zurich, 8091 Zurich, Switzerland; (S.H.); (R.F.)
| | - Ralph Fritsch
- Department of Medical Oncology and Hematology, University Hospital of Zurich, 8091 Zurich, Switzerland; (S.H.); (R.F.)
| | - Cäcilia Reiner
- Institute for Diagnostic and Interventional Radiology, University Hospital of Zurich, 8091 Zurich, Switzerland; (C.R.); (S.S.)
| | - Soleen Stocker
- Institute for Diagnostic and Interventional Radiology, University Hospital of Zurich, 8091 Zurich, Switzerland; (C.R.); (S.S.)
| | - Dieter Koeberle
- Oncology Departement, St. Claraspital, Kleinriehenstrasse 39, 4058 Basel, Switzerland;
| | - Marc Kissling
- Radiology Department, St. Claraspital, Kleinriehenstrasse 39, 4058 Basel, Switzerland;
| | - Vito Chianca
- Istituto di Imaging della Svizzera Italiana (IIMSI), Ente Ospedaliero Cantonale (EOC), Via Tesserete 46, 6900 Lugano, Switzerland;
| | - Filippo Del Grande
- Facoltà di Scienze Biomediche, Università della Svizzera Italiana, Via Buffi 13, 6900 Lugano, Switzerland; (I.S.); (F.D.G.); (S.R.)
- Istituto di Imaging della Svizzera Italiana (IIMSI), Ente Ospedaliero Cantonale (EOC), Via Tesserete 46, 6900 Lugano, Switzerland;
| | - Sara De Dosso
- Istituto Oncologico della Svizzera Italiana (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.C.); (G.P.)
- Facoltà di Scienze Biomediche, Università della Svizzera Italiana, Via Buffi 13, 6900 Lugano, Switzerland; (I.S.); (F.D.G.); (S.R.)
| | - Stefania Rizzo
- Facoltà di Scienze Biomediche, Università della Svizzera Italiana, Via Buffi 13, 6900 Lugano, Switzerland; (I.S.); (F.D.G.); (S.R.)
- Istituto di Imaging della Svizzera Italiana (IIMSI), Ente Ospedaliero Cantonale (EOC), Via Tesserete 46, 6900 Lugano, Switzerland;
| |
Collapse
|
14
|
Giansante V, Stati G, Sancilio S, Guerra E, Alberti S, Di Pietro R. The Dual Role of Necroptosis in Pancreatic Ductal Adenocarcinoma. Int J Mol Sci 2023; 24:12633. [PMID: 37628814 PMCID: PMC10454309 DOI: 10.3390/ijms241612633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Pancreatic cancer (PC) is the seventh leading cause of cancer-related death. PC incidence has continued to increase by about 1% each year in both men and women. Although the 5-year relative survival rate of PC has increased from 3% to 12%, it is still the lowest among cancers. Hence, novel therapeutic strategies are urgently needed. Challenges in PC-targeted therapeutic strategies stem from the high PC heterogeneity and from the poorly understood interplay between cancer cells and the surrounding microenvironment. Signaling pathways that drive PC cell growth have been the subject of intense scrutiny and interest has been attracted by necroptosis, a distinct type of programmed cell death. In this review, we provide a historical background on necroptosis and a detailed analysis of the ongoing debate on the role of necroptosis in PC malignant progression.
Collapse
Affiliation(s)
- Valentina Giansante
- Department of Medicine and Aging Sciences, Section of Biomorphology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Gianmarco Stati
- Department of Medicine and Aging Sciences, Section of Biomorphology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Silvia Sancilio
- Department of Medicine and Aging Sciences, Section of Biomorphology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Emanuela Guerra
- Laboratory of Cancer Pathology, Center for Advanced Studies and Technologies (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Saverio Alberti
- Unit of Medical Genetics, Department of Biomedical Sciences, University of Messina, 98122 Messina, Italy
| | - Roberta Di Pietro
- Department of Medicine and Aging Sciences, Section of Biomorphology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
15
|
Pakola S, Quixabeira DCA, Kudling TV, Clubb JHA, Grönberg-Vähä-Koskela S, Basnet S, Jirovec E, Arias V, Haybout L, Heiniö C, Santos JM, Cervera-Carrascon V, Havunen R, Anttila M, Hemminki A. An oncolytic adenovirus coding for a variant interleukin 2 cytokine improves response to chemotherapy through enhancement of effector lymphocyte cytotoxicity, fibroblast compartment modulation and mitotic slippage. Front Immunol 2023; 14:1171083. [PMID: 37475863 PMCID: PMC10354511 DOI: 10.3389/fimmu.2023.1171083] [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: 02/21/2023] [Accepted: 06/20/2023] [Indexed: 07/22/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly treatment-resistant cancer. Currently, the only curative treatment for PDAC is surgery, but most patients are diagnosed with metastatic disease and thus outside the scope of surgery. The majority of metastatic patients receive chemotherapy, but responses are limited. New therapeutics are thus urgently needed for PDAC. One major limitation in treating PDAC has been the highly immunosuppressive tumor microenvironment (TME) which inhibits anti-cancer immune responses. We have constructed an oncolytic adenovirus coding for a variant the interleukin 2 molecule, Ad5/3-E2F-d24-vIL2 (also known as TILT-452, and "vIL-2 virus"), with preferential binding to IL-2 receptors on the surface of effector lymphocytes over T regulatory cells (T regs). In the present study this virus was evaluated in combination with nab-paclitaxel and gemcitabine chemotherapy in Panc02 mouse model. Ad5/3-E2F-d24-vIL2 showed marked PDAC cell killing in vitro, alongside induction of mitotic slippage and immunogenic cell death in PDAC cell lines, when combined with chemotherapy. Increased survival was seen in vivo with 80% of animals surviving long term, when compared to chemotherapy alone. Moreover, combination therapy mediated enhanced tumor growth control, without observable toxicities in internal organs or external features. Survival and tumor control benefits were associated with activation of tumor infiltrating immune cells, downregulation of inhibitory signals, change in fibroblast populations in the tumors and changes in intratumoral cytokines, with increased chemokine amounts (CCL2, CCL3, CCL4) and anti-tumor cytokines (IFN-γ and TNFα). Furthermore, vIL-2 virus in combination with chemotherapy efficiently induced tumor protection upon rechallenge, that was extended to a previously non-encountered cancer cell line. In conclusion, Ad5/3-E2F-d24-vIL2 is a promising immunotherapy candidate when combined with nab-paclitaxel and gemcitabine.
Collapse
Affiliation(s)
- Santeri Pakola
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Dafne C. A. Quixabeira
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd., Helsinki, Finland
| | - Tatiana V. Kudling
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - James H. A. Clubb
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd., Helsinki, Finland
| | - Susanna Grönberg-Vähä-Koskela
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Saru Basnet
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Elise Jirovec
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd., Helsinki, Finland
| | - Victor Arias
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Lyna Haybout
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd., Helsinki, Finland
| | - Camilla Heiniö
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Joao M. Santos
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd., Helsinki, Finland
| | - Victor Cervera-Carrascon
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd., Helsinki, Finland
| | - Riikka Havunen
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd., Helsinki, Finland
| | | | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd., Helsinki, Finland
- Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| |
Collapse
|
16
|
Wishart G, Gupta P, Nisbet A, Velliou E, Schettino G. Enhanced effect of X-rays in the presence of a static magnetic field within a 3D pancreatic cancer model. Br J Radiol 2023; 96:20220832. [PMID: 36475863 PMCID: PMC9975369 DOI: 10.1259/bjr.20220832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/23/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE To evaluate the impact of static magnetic field (SMF) presence on the radiation response of pancreatic cancer cells in polyurethane-based highly macro-porous scaffolds in hypoxic (1% O2) and normoxic (21% O2) conditions, towards understanding MR-guided radiotherapy, shedding light on the potential interaction phenomenon between SMF and radiation in a three-dimensional (3D) microenvironment. METHODS Pancreatic cancer cells (PANC-1, ASPC-1) were seeded into fibronectin-coated highly porous polyethene scaffolds for biomimicry and cultured for 4 weeks in in vitro normoxia (21% O2) followed by a 2-day exposure to either in vitro hypoxia (1% O2) or maintenance in in vitro normoxia (21% O2). The samples were then irradiated with 6 MV photons in the presence or absence of a 1.5 T field. Thereafter, in situ post-radiation monitoring (1 and 7 days post-irradiation treatment) took place via quantification of (i) live dead and (ii) apoptotic profiles. RESULTS We report: (i) pancreatic ductal adenocarcinoma hypoxia-associated radioprotection, in line with our previous findings, (ii) an enhanced effect of radiation in the presence of SMFin in vitro hypoxia (1% O2) for both short- (1 day) and long-term (7 days) post -radiation analysis and (iii) an enhanced effect of radiation in the presence of SMF in in vitro normoxia (21% O2) for long-term (7 days) post-radiation analysis within a 3D pancreatic cancer model. CONCLUSION With limited understanding of the potential interaction phenomenon between SMF and radiation, this 3D system allows combination evaluation for a cancer in which the role of radiotherapy is still evolving. ADVANCES IN KNOWLEDGE This study examined the use of a 3D model to investigate MR-guided radiotherapy in a hypoxic microenvironment, indicating that this could be a useful platform to further understanding of SMF influence on radiation.
Collapse
Affiliation(s)
| | | | - Andrew Nisbet
- Department of Medical Physics and Biomedical Engineering, University College London (UCL), London, UK
| | | | | |
Collapse
|
17
|
Tumor immunology. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00003-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
18
|
Wei W, Jia G, Wu Z, Wang T, Wang H, Wei K, Cheng C, Liu Z, Zuo C. A multidomain fusion model of radiomics and deep learning to discriminate between PDAC and AIP based on 18F-FDG PET/CT images. Jpn J Radiol 2022; 41:417-427. [PMID: 36409398 PMCID: PMC9676903 DOI: 10.1007/s11604-022-01363-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/09/2022] [Indexed: 11/22/2022]
Abstract
PURPOSE To explore a multidomain fusion model of radiomics and deep learning features based on 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) images to distinguish pancreatic ductal adenocarcinoma (PDAC) and autoimmune pancreatitis (AIP), which could effectively improve the accuracy of diseases diagnosis. MATERIALS AND METHODS This retrospective study included 48 patients with AIP (mean age, 65 ± 12.0 years; range, 37-90 years) and 64 patients with PDAC patients (mean age, 66 ± 11.3 years; range, 32-88 years). Three different methods were discussed to identify PDAC and AIP based on 18F-FDG PET/CT images, including the radiomics model (RAD_model), the deep learning model (DL_model), and the multidomain fusion model (MF_model). We also compared the classification results of PET/CT, PET, and CT images in these three models. In addition, we explored the attributes of deep learning abstract features by analyzing the correlation between radiomics and deep learning features. Five-fold cross-validation was used to calculate receiver operating characteristic (ROC), area under the roc curve (AUC), accuracy (Acc), sensitivity (Sen), and specificity (Spe) to quantitatively evaluate the performance of different classification models. RESULTS The experimental results showed that the multidomain fusion model had the best comprehensive performance compared with radiomics and deep learning models, and the AUC, accuracy, sensitivity, specificity were 96.4% (95% CI 95.4-97.3%), 90.1% (95% CI 88.7-91.5%), 87.5% (95% CI 84.3-90.6%), and 93.0% (95% CI 90.3-95.6%), respectively. And our study proved that the multimodal features of PET/CT were superior to using either PET or CT features alone. First-order features of radiomics provided valuable complementary information for the deep learning model. CONCLUSION The preliminary results of this paper demonstrated that our proposed multidomain fusion model fully exploits the value of radiomics and deep learning features based on 18F-FDG PET/CT images, which provided competitive accuracy for the discrimination of PDAC and AIP.
Collapse
Affiliation(s)
- Wenting Wei
- School of Electronic and Information Engineering, Changchun University of Science and Technology, Changchun, 130022 China ,Department of Medical Imaging, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, 88 Keling Road, Suzhou, 215163 China
| | - Guorong Jia
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University (Changhai Hospital), 168 Changhai Road, Shanghai, 200433 China
| | - Zhongyi Wu
- Department of Medical Imaging, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, 88 Keling Road, Suzhou, 215163 China
| | - Tao Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University (Changhai Hospital), 168 Changhai Road, Shanghai, 200433 China
| | - Heng Wang
- School of Electronic and Information Engineering, Changchun University of Science and Technology, Changchun, 130022 China ,Department of Medical Imaging, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, 88 Keling Road, Suzhou, 215163 China
| | - Kezhen Wei
- School of Electronic and Information Engineering, Changchun University of Science and Technology, Changchun, 130022 China ,Department of Medical Imaging, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, 88 Keling Road, Suzhou, 215163 China
| | - Chao Cheng
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University (Changhai Hospital), 168 Changhai Road, Shanghai, 200433 China
| | - Zhaobang Liu
- Department of Medical Imaging, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, 88 Keling Road, Suzhou, 215163 China
| | - Changjing Zuo
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University (Changhai Hospital), 168 Changhai Road, Shanghai, 200433 China
| |
Collapse
|
19
|
Gao JF, Pan Y, Lin XC, Lu FC, Qiu DS, Liu JJ, Huang HG. Prognostic value of preoperative enhanced computed tomography as a quantitative imaging biomarker in pancreatic cancer. World J Gastroenterol 2022; 28:2468-2481. [PMID: 35979266 PMCID: PMC9258279 DOI: 10.3748/wjg.v28.i22.2468] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/31/2021] [Accepted: 05/17/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies with high mortality and short survival time. Computed tomography (CT) plays an important role in the diagnosis, staging and treatment of pancreatic tumour. Pancreatic cancer generally shows a low enhancement pattern compared with normal pancreatic tissue.
AIM To analyse whether preoperative enhanced CT could be used to predict postoperative overall survival in patients with PDAC.
METHODS Sixty-seven patients with PDAC undergoing pancreatic resection were enrolled retrospectively. All patients underwent preoperative unenhanced and enhanced CT examination, the CT values of which were measured. The ratio of the preoperative CT value increase from the nonenhancement phase to the portal venous phase between pancreatic tumour and normal pancreatic tissue was calculated. The cut-off value of ratios was obtained by the receiver operating characteristic (ROC) curve of the tumour relative enhancement ratio (TRER), according to which patients were divided into low- and high-enhancement groups. Univariate and multivariate analyses were performed using Cox regression based on TRER grouping. Finally, the correlation between TRER and clinicopathological characteristics was analysed.
RESULTS The area under the curve of the ROC curve was 0.768 (P < 0.05), and the cut-off value of the ROC curve was calculated as 0.7. TRER ≤ 0.7 was defined as the low-enhancement group, and TRER > 0.7 was defined as the high-enhancement group. According to the TRER grouping, the Kaplan-Meier survival curve analysis results showed that the median survival (10.0 mo) with TRER ≤ 0.7 was significantly shorter than that (22.0 mo) with TRER > 0.7 (P < 0.05). In the univariate and multivariate analyses, the prognosis of patients with TRER ≤ 0.7 was significantly worse than that of patients with TRER > 0.7 (P < 0.05). Our results demonstrated that patients in the low TRER group were more likely to have higher American Joint Committee on Cancer stage, tumour stage and lymph node stage (all P < 0.05), and TRER was significantly negatively correlated with tumour size (P < 0.05).
CONCLUSION TRER ≤ 0.7 in patients with PDAC may represent a tumour with higher clinical stage and result in a shorter overall survival.
Collapse
Affiliation(s)
- Jian-Feng Gao
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
| | - Yu Pan
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
| | - Xian-Chao Lin
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
| | - Feng-Chun Lu
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
| | - Ding-Shen Qiu
- Department of Radiology, The Hospital of Changle, Fuzhou 350200, Fujian Province, China
| | - Jun-Jun Liu
- Department of Radiology, The Hospital of Changle, Fuzhou 350200, Fujian Province, China
| | - He-Guang Huang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
| |
Collapse
|
20
|
Su K, Peng Y, Yu H. Development of a Prognostic Model Based on Pyroptosis-Related Genes in Pancreatic Adenocarcinoma. DISEASE MARKERS 2022; 2022:9141117. [PMID: 35677632 PMCID: PMC9169203 DOI: 10.1155/2022/9141117] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 05/12/2022] [Accepted: 05/19/2022] [Indexed: 11/17/2022]
Abstract
Background The importance of pyroptosis in tumorigenesis and cancer progression is becoming increasingly apparent. However, the efficacy of using pyroptosis-related genes (PRGs) in predicting the prognosis of pancreatic adenocarcinoma (PAAD) patients is unknown. Methods This investigation used two databases to obtain expression data for PAAD patients. Differentially expressed PRGs (DEPRGs) were identified between PAAD and control samples. Several bioinformatic approaches were used to analyze the biological functions of DEPRGs and to identify prognostic DERPGs. A miRNA-prognostic DEPRG-transcription factor (TF) regulatory network was created via the miRNet online tool. A risk score model was created after each patient's risk score was calculated. The microenvironments of the low- and high-risk groups were assessed using xCell, the expression of immune checkpoints was determined, and gene set variation analysis (GSVA) was performed. Finally, the efficacy of certain potential drugs was predicted using the pRRophetic algorithm, and the results in the high- and low-risk groups were compared. Results A total of 13 DEPRGs were identified between PAAD and control samples. Functional enrichment analysis showed that the DEPRGs had a close relationship with inflammation. In univariate and multivariate Cox regression analyses, GSDMC, IRF1, and PLCG1 were identified as prognostic biomarkers in PAAD. The results of the miRNA-prognostic DEPRG-TF regulatory network showed that GSDMC, IRF1, and PLCG1 were regulated by both specific and common miRNAs and TFs. Based on the risk score and other independent prognostic indicators, a nomogram with a good ability to predict the survival of PAAD patients was developed. By evaluating the tumor microenvironment, we observed that the immune and metabolic microenvironments of the two groups were substantially different. In addition, individuals in the low-risk group were more susceptible to axitinib and camptothecin, whereas lapatinib might be preferred for patients in the high-risk group. Conclusion Our study revealed the prognostic value of PRGs in PAAD and created a reliable model for predicting the prognosis of PAAD patients. Our findings will benefit the prognostication and treatment of PAAD patients.
Collapse
Affiliation(s)
- Kaifeng Su
- Medical Faculty of Ludwig-Maximilians-University of Munich, University Hospital of LMU Munich, Munich, Germany
| | - Yang Peng
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haochen Yu
- Medical Faculty of Ludwig-Maximilians-University of Munich, University Hospital of LMU Munich, Munich, Germany
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
21
|
Ebrahimian S, Singh R, Netaji A, Madhusudhan KS, Homayounieh F, Primak A, Lades F, Saini S, Kalra MK, Sharma S. Characterization of Benign and Malignant Pancreatic Lesions with DECT Quantitative Metrics and Radiomics. Acad Radiol 2022; 29:705-713. [PMID: 34412944 DOI: 10.1016/j.acra.2021.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/07/2021] [Accepted: 07/14/2021] [Indexed: 12/24/2022]
Abstract
RATIONALE AND OBJECTIVES To compare dual energy CT (DECT) quantitative metrics and radiomics for differentiating benign and malignant pancreatic lesions on contrast enhanced abdomen CT. MATERIALS AND METHODS Our study included 103 patients who underwent contrast-enhanced DECT for assessing focal pancreatic lesions at one of the two hospitals (Site A: age 68 ± 12 yrs; malignant = 41, benign = 18; Site B: age 46 ± 2 yrs; malignant = 23, benign = 21). All malignant lesions had histologic confirmation, and benign lesions were stable on follow up CT (>12 months) or had characteristic benign features on MRI. Arterial-phase, low- and high-kV DICOM images were processed with the DECT Tumor Analysis (DETA) to obtain DECT quantitative metrics such as HU, iodine and water content from a region of interest (ROI) over focal pancreatic lesions. Separately, we obtained DECT radiomics from the same ROI. Data were analyzed with multiple logistic regression and receiver operating characteristics to generate area under the curve (AUC) for best predictive variables. RESULTS DECT quantitative metrics and radiomics had AUCs of 0.98-0.99 at site A and 0.89-0.94 at site B data for classifying benign and malignant pancreatic lesions. There was no significant difference in the AUCs and accuracies of DECT quantitative metrics and radiomics from lesion rims and volumes among patients at both sites (p > 0.05). Supervised learning-based model with data from the two sites demonstrated best AUCs of 0.94 (DECT radiomics) and 0.90 (DECT quantitative metrics) for characterizing pancreatic lesions as benign or malignant. CONCLUSION Compared to complex DECT radiomics, quantitative DECT information provide a simpler but accurate method of differentiating benign and malignant pancreatic lesions.
Collapse
Affiliation(s)
- Shadi Ebrahimian
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 75 Blossom court, Boston, MA 02114
| | - Ramandeep Singh
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 75 Blossom court, Boston, MA 02114
| | - Arjunlokesh Netaji
- Department of Radiodiagnosis and Interventional Radiology, All India Institute of Medical Sciences, New Delhi, India
| | - Kumble Seetharama Madhusudhan
- Department of Radiodiagnosis and Interventional Radiology, All India Institute of Medical Sciences, New Delhi, India
| | - Fatemeh Homayounieh
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 75 Blossom court, Boston, MA 02114
| | - Andrew Primak
- Siemens Medical Solutions USA Inc., Malvern, Pennsylvania
| | | | - Sanjay Saini
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 75 Blossom court, Boston, MA 02114
| | - Mannudeep K Kalra
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 75 Blossom court, Boston, MA 02114.
| | - Sanjay Sharma
- Department of Radiodiagnosis and Interventional Radiology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
22
|
Wishart G, Gupta P, Nisbet A, Schettino G, Velliou E. On the Evaluation of a Novel Hypoxic 3D Pancreatic Cancer Model as a Tool for Radiotherapy Treatment Screening. Cancers (Basel) 2021; 13:6080. [PMID: 34885188 PMCID: PMC8657010 DOI: 10.3390/cancers13236080] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/24/2021] [Accepted: 11/30/2021] [Indexed: 12/15/2022] Open
Abstract
Tissue engineering is evolving to mimic intricate ecosystems of tumour microenvironments (TME) to more readily map realistic in vivo niches of cancerous tissues. Such advanced cancer tissue models enable more accurate preclinical assessment of treatment strategies. Pancreatic cancer is a dangerous disease with high treatment resistance that is directly associated with a highly complex TME. More specifically, the pancreatic cancer TME includes (i) complex structure and complex extracellular matrix (ECM) protein composition; (ii) diverse cell populations (e.g., stellate cells), cancer associated fibroblasts, endothelial cells, which interact with the cancer cells and promote resistance to treatment and metastasis; (iii) accumulation of high amounts of (ECM), which leads to the creation of a fibrotic/desmoplastic reaction around the tumour; and (iv) heterogeneous environmental gradients such as hypoxia, which result from vessel collapse and stiffness increase in the fibrotic/desmoplastic area of the TME. These unique hallmarks are not effectively recapitulated in traditional preclinical research despite radiotherapeutic resistance being largely connected to them. Herein, we investigate, for the first time, the impact of in vitro hypoxia (5% O2) on the radiotherapy treatment response of pancreatic cancer cells (PANC-1) in a novel polymer (polyurethane) based highly macroporous scaffold that was surface modified with proteins (fibronectin) for ECM mimicry. More specifically, PANC-1 cells were seeded in fibronectin coated macroporous scaffolds and were cultured for four weeks in in vitro normoxia (21% O2), followed by a two day exposure to either in vitro hypoxia (5% O2) or maintenance in in vitro normoxia. Thereafter, in situ post-radiation monitoring (one day, three days, seven days post-irradiation) of the 3D cell cultures took place via quantification of (i) live/dead and apoptotic profiles and (ii) ECM (collagen-I) and HIF-1a secretion by the cancer cells. Our results showed increased post-radiation viability, reduced apoptosis, and increased collagen-I and HIF-1a secretion in in vitro hypoxia compared to normoxic cultures, revealing hypoxia-induced radioprotection. Overall, this study employed a low cost, animal free model enabling (i) the possibility of long-term in vitro hypoxic 3D cell culture for pancreatic cancer, and (ii) in vitro hypoxia associated PDAC radio-protection development. Our novel platform for radiation treatment screening can be used for long-term in vitro post-treatment observations as well as for fractionated radiotherapy treatment.
Collapse
Affiliation(s)
- Gabrielle Wishart
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford GU2 7XH, UK; (G.W.); (P.G.)
- Department of Physics, University of Surrey, Guildford GU2 7XH, UK;
| | - Priyanka Gupta
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford GU2 7XH, UK; (G.W.); (P.G.)
- Centre for 3D Models of Health and Disease, Department of Targeted Intervention, Division of Surgery and Interventional Science, University College London (UCL), London W1W 7TY, UK
| | - Andrew Nisbet
- Department of Medical Physics and Biomedical Engineering, University College London (UCL), London WC1E 6BT, UK;
| | - Giuseppe Schettino
- Department of Physics, University of Surrey, Guildford GU2 7XH, UK;
- National Physical Laboratory, Teddington TW11 0LW, UK
| | - Eirini Velliou
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford GU2 7XH, UK; (G.W.); (P.G.)
- Centre for 3D Models of Health and Disease, Department of Targeted Intervention, Division of Surgery and Interventional Science, University College London (UCL), London W1W 7TY, UK
| |
Collapse
|
23
|
The Role of Circulating MicroRNAs in Patients with Early-Stage Pancreatic Adenocarcinoma. Biomedicines 2021; 9:biomedicines9101468. [PMID: 34680585 PMCID: PMC8533318 DOI: 10.3390/biomedicines9101468] [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] [Received: 09/22/2021] [Revised: 10/10/2021] [Accepted: 10/12/2021] [Indexed: 01/17/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is increasing in incidence and is still associated with a high rate of mortality. Only a minority of patients are diagnosed in the early stage. Radical surgery is the only potential curative procedure. However, radicality is reached in 20% of patients operated on. Despite the multidisciplinary approach in resectable tumors, early tumor recurrences are common. Options on how to select optimal candidates for resection remain limited. Nevertheless, accumulating evidence shows an important role of circulating non-coding plasma and serum microRNAs (miRNAs), which physiologically regulate the function of a target protein. miRNAs also play a crucial role in carcinogenesis. In PDAC patients, the expression levels of certain miRNAs vary and may modulate the function of oncogenes or tumor suppressor genes. As they can be detected in a patient's blood, they have the potential to become promising non-invasive diagnostic and prognostic biomarkers. Moreover, they may also serve as markers of chemoresistance. Thus, miRNAs could be useful for early and accurate diagnosis, prognostic stratification, and individual treatment planning. In this review, we summarize the latest findings on miRNAs in PDAC patients, focusing on their potential use in the early stage of the disease.
Collapse
|
24
|
Insulin Receptor in Pancreatic Cancer-Crown Witness in Cross Examination. Cancers (Basel) 2021; 13:cancers13194988. [PMID: 34638472 PMCID: PMC8508431 DOI: 10.3390/cancers13194988] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The proximity of pancreatic cancer (PDAC) to the physiological source of the growth promoting hormone insulin might be exploited by this highly malignant cancer entity. We investigated if (I) PDACs express the insulin receptor (IR) in cancer cells and cancer vasculature, (II) if IR correlates with clinicopathological patient characteristics, including survival, and hence is involved in PDAC biology, (III) if IR is already expressed in precursor lesions, if (IV) the IGF1 receptor (IGF1R) is associated with clinicopathological patient characteristics and survival and (V) is linked to IR expression. METHODS 160 PDAC samples were examined for IR and IGF1R expression by immunohistochemistry. A modified HistoScore was correlated with clinicopathological characteristics and survival. RESULTS IR overexpression was already observed in pancreatic intraepithelial neoplasia. Furthermore, it was more frequently observed in advanced disease and associated with distant metastasis, UICC stage, lymphatic invasion and an increased lymph node ratio, but without impacting survival in the end. IGF1R expression was not associated with clinicopathological parameters or survival, in contrast to former paradigms. CONCLUSIONS We hypothesize that the close proximity to the pancreatic islets might be advantageous for cancer growth at first, but it experiences self-limitation due to surgical removal or local destruction following accelerated cancer growth.
Collapse
|
25
|
Ishtiaq R, Aziz M, Wilhelm DM, Gaduputi VV, Hamdani SU. An Uncommon Cause of Hematochezia. J Gastrointest Cancer 2021; 52:359-364. [PMID: 32761320 DOI: 10.1007/s12029-020-00470-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Rizwan Ishtiaq
- Internal Medicine Residency Program, Mercy Health St. Vincent Medical Center, 2213 Cherry Street, Toledo, OH, 43608, USA.
| | - Muhammad Aziz
- Department of Internal Medicine, University of Toledo Medical Center, Toledo, OH, USA
| | | | | | | |
Collapse
|
26
|
Pancreatic Ductal Adenocarcinoma: Relating Biomechanics and Prognosis. J Clin Med 2021; 10:jcm10122711. [PMID: 34205335 PMCID: PMC8234178 DOI: 10.3390/jcm10122711] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/12/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common form of pancreatic cancer and carries a dismal prognosis. Resectable patients are treated predominantly with surgery while borderline resectable patients may receive neoadjuvant treatment (NAT) to downstage their disease prior to possible resection. PDAC tissue is stiffer than healthy pancreas, and tissue stiffness is associated with cancer progression. Another feature of PDAC is increased tissue heterogeneity. We postulate that tumour stiffness and heterogeneity may be used alongside currently employed diagnostics to better predict prognosis and response to treatment. In this review we summarise the biomechanical changes observed in PDAC, explore the factors behind these changes and describe the clinical consequences. We identify methods available for assessing PDAC biomechanics ex vivo and in vivo, outlining the relative merits of each. Finally, we discuss the potential use of radiological imaging for prognostic use.
Collapse
|
27
|
Wishart G, Gupta P, Nisbet A, Velliou E, Schettino G. Novel Anticancer and Treatment Sensitizing Compounds against Pancreatic Cancer. Cancers (Basel) 2021; 13:2940. [PMID: 34208295 PMCID: PMC8231164 DOI: 10.3390/cancers13122940] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 01/05/2023] Open
Abstract
The isolation of chemical compounds from natural origins for medical application has played an important role in modern medicine with a range of novel treatments having emerged from various natural forms over the past decades. Natural compounds have been exploited for their antioxidant, antimicrobial and antitumor capabilities. Specifically, 60% of today's anticancer drugs originate from natural sources. Moreover, the combination of synthetic and natural treatments has shown applications for (i) reduced side effects, (ii) treatment sensitization and (iii) reduction in treatment resistance. This review aims to collate novel and natural compounds that are being explored for their preclinical anticancer, chemosensitizing and radiosensitizing effects on Pancreatic Ductal Adenocarcinoma (PDAC), which is a lethal disease with current treatments being inefficient and causing serve side effects. Two key points are highlighted by this work: (i) the availability of a range of natural compounds for potentially new therapeutic approaches for PDAC, (ii) potential synergetic impact of natural compounds with advanced chemo- and radio-therapeutic modalities for PDAC.
Collapse
Affiliation(s)
- Gabrielle Wishart
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford GU2 7XH, UK; (G.W.); (P.G.); (E.V.)
- Department of Physics, University of Surrey, Guildford GU2 7XH, UK
| | - Priyanka Gupta
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford GU2 7XH, UK; (G.W.); (P.G.); (E.V.)
| | - Andrew Nisbet
- Department of Medical Physics and Biomedical Engineering, University College London, London WC1E 6BT, UK;
| | - Eirini Velliou
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford GU2 7XH, UK; (G.W.); (P.G.); (E.V.)
- Centre for 3D Models of Health and Disease, UCL-Division of Surgery and Interventional Science, Charles Bell House, 43-45 Foley Street, Fitzrovia, London W1W 7TY, UK
| | - Giuseppe Schettino
- Department of Physics, University of Surrey, Guildford GU2 7XH, UK
- National Physical Laboratory, Teddington TW11 0LW, UK
| |
Collapse
|
28
|
Hibino S, Kawazoe T, Kasahara H, Itoh S, Ishimoto T, Sakata-Yanagimoto M, Taniguchi K. Inflammation-Induced Tumorigenesis and Metastasis. Int J Mol Sci 2021; 22:ijms22115421. [PMID: 34063828 PMCID: PMC8196678 DOI: 10.3390/ijms22115421] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 02/07/2023] Open
Abstract
Inflammation, especially chronic inflammation, plays a pivotal role in tumorigenesis and metastasis through various mechanisms and is now recognized as a hallmark of cancer and an attractive therapeutic target in cancer. In this review, we discuss recent advances in molecular mechanisms of how inflammation promotes tumorigenesis and metastasis and suppresses anti-tumor immunity in various types of solid tumors, including esophageal, gastric, colorectal, liver, and pancreatic cancer as well as hematopoietic malignancies.
Collapse
Affiliation(s)
- Sana Hibino
- Research Center for Advanced Science and Technology, Department of Inflammology, The University of Tokyo, Tokyo 153-0041, Japan;
| | - Tetsuro Kawazoe
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan;
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan;
| | - Hidenori Kasahara
- National Center for Global Health and Medicine, Department of Stem Cell Biology, Research Institute, Tokyo 162-8655, Japan;
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Shinji Itoh
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan;
| | - Takatsugu Ishimoto
- Gastrointestinal Cancer Biology, International Research Center of Medical Sciences (IRCMS), Kumamoto University, Kumamoto 860-0811, Japan;
| | | | - Koji Taniguchi
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan;
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
- Correspondence: ; Tel.: +81-11-706-5050
| |
Collapse
|
29
|
Wishart G, Gupta P, Schettino G, Nisbet A, Velliou E. 3d tissue models as tools for radiotherapy screening for pancreatic cancer. Br J Radiol 2021; 94:20201397. [PMID: 33684308 PMCID: PMC8010544 DOI: 10.1259/bjr.20201397] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 12/14/2022] Open
Abstract
The efficiency of radiotherapy treatment regimes varies from tumour to tumour and from patient to patient but it is generally highly influenced by the tumour microenvironment (TME). The TME can be described as a heterogeneous composition of biological, biophysical, biomechanical and biochemical milieus that influence the tumour survival and its' response to treatment. Preclinical research faces challenges in the replication of these in vivo milieus for predictable treatment response studies. 2D cell culture is a traditional, simplistic and cost-effective approach to culture cells in vitro, however, the nature of the system fails to recapitulate important features of the TME such as structure, cell-cell and cell-matrix interactions. At the same time, the traditional use of animals (Xenografts) in cancer research allows realistic in vivo architecture, however foreign physiology, limited heterogeneity and reduced tumour mutation rates impairs relevance to humans. Furthermore, animal research is very time consuming and costly. Tissue engineering is advancing as a promising biomimetic approach, producing 3D models that capture structural, biophysical, biochemical and biomechanical features, therefore, facilitating more realistic treatment response studies for further clinical application. However, currently, the application of 3D models for radiation response studies is an understudied area of research, especially for pancreatic ductal adenocarcinoma (PDAC), a cancer with a notoriously complex microenvironment. At the same time, specific novel and/or more enhanced radiotherapy tumour-targeting techniques such as MRI-guided radiotherapy and proton therapy are emerging to more effectively target pancreatic cancer cells. However, these emerging technologies may have different biological effectiveness as compared to established photon-based radiotherapy. For example, for MRI-guided radiotherapy, the novel use of static magnetic fields (SMF) during radiation delivery is understudied and not fully understood. Thus, reliable biomimetic platforms to test new radiation delivery strategies are required to more accurately predict in vivo responses. Here, we aim to collate current 3D models for radiation response studies of PDAC, identifying the state of the art and outlines knowledge gaps. Overall, this review paper highlights the need for further research on the use of 3D models for pre-clinical radiotherapy screening including (i) 3D (re)-modeling of the PDAC hypoxic TME to allow for late effects of ionising radiation (ii) the screening of novel radiotherapy approaches and their combinations as well as (iii) a universally accepted 3D-model image quantification method for evaluating TME components in situ that would facilitate accurate post-treatment(s) quantitative comparisons.
Collapse
Affiliation(s)
| | - Priyanka Gupta
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford, UK
| | | | | | | |
Collapse
|
30
|
Ward RD, Amorim B, Li W, King J, Umutlu L, Groshar D, Harisinghani M, Catalano O. Abdominal and pelvic 18F-FDG PET/MR: a review of current and emerging oncologic applications. Abdom Radiol (NY) 2021; 46:1236-1248. [PMID: 32949272 DOI: 10.1007/s00261-020-02766-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 12/12/2022]
Abstract
Positron emission tomography (PET) using fluorodeoxyglucose (18F-FDG) combined with magnetic resonance imaging (MR) is an emerging hybrid modality that has shown utility in evaluating abdominal and pelvic disease entities. Together, the high soft tissue contrast and metabolic/functional imaging capabilities make this modality ideal for oncologic imaging in many organ systems. Its clinical utility continues to evolve and future research will help solidify its role in oncologic imaging. In this manuscript, we aim to (1) provide an overview of the various PET/MR systems, describing the strengths and weaknesses of each system, and (2) review the oncologic applications for 18F-FDG PET/MR in the abdomen and pelvis.
Collapse
Affiliation(s)
- Ryan D Ward
- Cleveland Clinic, Department of Abdominal Imaging, 9500 Euclid Ave, L10, Cleveland, OH, 44195, USA
| | - Barbara Amorim
- Division of Nuclear Medicine, University of Campinas, Rua Vital Brasil 251, Campinas, Brazil
| | - Weier Li
- Department of Abdominal Imaging, Massachusetts General Hospital, 55 Fruit Street, White 270, Boston, MA, 02114, USA
| | - Joseph King
- Department of Abdominal Imaging, Massachusetts General Hospital, 55 Fruit Street, White 270, Boston, MA, 02114, USA
| | - Lale Umutlu
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University Duisburg-Essen, Hufelandstr. 55, 45122, Essen, Germany
| | - David Groshar
- Assuta Medical Center, Habrzel 20, 6971028, Tel-Aviv, Israel
- Sackler School of Medicine, Tel-Aviv, Israel
| | - Mukesh Harisinghani
- Department of Abdominal Imaging, Massachusetts General Hospital, 55 Fruit Street, White 270, Boston, MA, 02114, USA
| | - Onofrio Catalano
- Department of Abdominal Imaging, Massachusetts General Hospital, 55 Fruit Street, White 270, Boston, MA, 02114, USA.
| |
Collapse
|
31
|
Xu P, Wang X, Qian J, Li Z, Yao J, Xu AM. The prognostic evaluation of CA19-9, D-dimer and TNFAIP3/A20 in patients with pancreatic ductal adenocarcinoma. Medicine (Baltimore) 2021; 100:e24651. [PMID: 33578593 PMCID: PMC10545421 DOI: 10.1097/md.0000000000024651] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 12/30/2020] [Accepted: 01/10/2021] [Indexed: 02/05/2023] Open
Abstract
ABSTRACT This study aimed to explore the significance and prognostic value of serum tumor-associated carbohydrate antigen 19-9 (CA19-9), D-dimer, and tumor necrosis factor alpha-induced protein 3 (TNFAIP3/A20) in patients with pancreatic ductal adenocarcinoma (PDAC).Our study included 148 patients treated for PDAC at Northern Jiangsu People's Hospital Affiliated to Yangzhou University from January 2012 to December 2016. Cutoff prognostic values were predicted using the receiver operating characteristic (ROC) curve. The Kaplan-Meier method was used to assess the survival rates of patients. Univariate and multivariate COX regression analyses were used to evaluate the prognostic factors.The recommended cutoff values of neutrophil-lymphocyte rate (NLR), platelet-lymphocyte rate (PLR), CA19-9, and D-dimer were 2.04 (sensitivity, 0.59; specificity, 0.9; area under the ROC curve [AUC], 0.749; P < .001), 52.94 (sensitivity, 0.73; specificity, 0.95; AUC, 0.829; P < .001), 176.66 U/mL (sensitivity, 0.7; specificity, 0.9; AUC, 0.794; P < .001), and 1.18 mg/L (sensitivity, 0.82; specificity, 0.9; AUC, 0.845; P < .001), respectively. Positive TNFAIP3/A20 expression was considered as an inclusion criterion. Serum CA19-9 expression was related with lymph node metastasis (P = .010), tumor-lymph node-metastasis (TNM) stage (P < .001), and survival rate (P < .001). D-dimer was correlated with tumor differentiation grade (P = .014), tumor size (P = .045), TNM stage (P < .001), and survival rate (P < .001). TNFAIP3/A20 was correlated with tumor differentiation grade (P < .001), body mass index (BMI) (P < .001), TNM stage (P = .014), and survival rate (P < .001). Kaplan-Meier curves showed that PDAC patients had significant differences in CA19-9, D-dimer, and TNFAIP3/A20 expressions (P < .05). CA19-9, D-dimer, TNM stage, tumor differentiation grade, and TNFAIP3/A20 were independent prognostic markers for PDAC in univariate and multivariate COX analyses.CA19-9, D-dimer, and TNFAIP3/A20 were found to be independent prognostic markers for PDAC patients.
Collapse
Affiliation(s)
- Peng Xu
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Hefei
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou
| | - XiaoDong Wang
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou
| | - JianJun Qian
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou
| | - ZhengNan Li
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou
| | - Jie Yao
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou
| | - AMan Xu
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Hefei
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
32
|
Transabdominal ultrasound-guided pancreatic biopsy: a neglected but safe, effective and inexpensive procedure that needs to be re-juvinalized. J Ultrasound 2021; 24:175-182. [PMID: 33521888 DOI: 10.1007/s40477-020-00542-y] [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: 07/27/2020] [Accepted: 11/13/2020] [Indexed: 10/22/2022] Open
Abstract
BACKGROUND For solid pancreatic masses, ultrasound endoscopic fine-needle biopsy is suggested as the front-line investigation for tissue achievement, notwithstanding the optimal performance of transabdominal ultrasound (TUS)-guided biopsy. PURPOSE To reassess the efficacy and effectiveness of TUS-guided sampling and to determine the factors predictive of accurate histology. METHODS In total, 142 patients with an indication for a TUS-guided biopsy of a pancreatic mass were analyzed. A single pass of an 18-gauge Biomol needle was carried out by the Menghini technique. The accuracy, sensitivity, and specificity of the procedure in terms of correctly diagnosing an inflammatory or neoplastic lesion were determined. The patients' characteristics, the size and location of the mass, and the sonographers' experience in performing TUS were recorded. RESULTS The sampling was unsuccessful in 24 cases, owing to the deep localization of lesions (57%), bloating (33%), or low patient compliance (10%). The accuracy, sensitivity, and specificity of the 118 successful biopsies were 81%, 79%, and 100%, respectively. A biopsy core was obtained in 90 of the 118 patients (76%) in whom the procedure was attempted. In the multivariate analysis, lesion size (≤ 20 mm vs. > 20 mm) (OR = 5.3 [1.7-17.0]) and operator experience (OR = 4.4 [1.6-12.1]) predicted the acquisition of adequate samples. With an expert sonographer, the accuracy, sensitivity, and specificity were 87%, 85%, and 100%, respectively. Two adverse events were registered: mild abdominal pain and a hypotensive crisis. CONCLUSIONS The present investigation highlights the optimal performance of a TUS-guided biopsy of a pancreatic mass. Because of its simplicity and safety, the procedure needs to be included among the recommended investigative options.
Collapse
|
33
|
Barcellini A, Peloso A, Pugliese L, Vitolo V, Cobianchi L. Locally Advanced Pancreatic Ductal Adenocarcinoma: Challenges and Progress. Onco Targets Ther 2020; 13:12705-12720. [PMID: 33335406 PMCID: PMC7737010 DOI: 10.2147/ott.s220971] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/30/2020] [Indexed: 12/24/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the major causes of death in the Western world, and it is estimated to become the second leading cause of tumour-related mortality in the next 10 years. Among pancreatic cancers, ductal adenocarcinomas are by far the most common, characterised by a challenging diagnosis due to the lack of initial and pathognomonic clinical signs. In this scenario, non-metastatic locally advanced pancreatic cancer (LAPC) accounts for a large proportion of all new pancreatic ductal adenocarcinoma diagnoses. There is no consensus on a common definition of LAPC. Still, it usually includes tumours that are not resectable due to vascular involvement. As of today, treatment is limited, and the prognosis is very unfavourable. Curative-intent surgery remains the gold-standard even if often jeopardized by vascular involvement. Continuing progress in our understanding of LAPC genetics and immunology will permit the development of different treatments, targeted or combined, including radiation therapy, hadrontherapy, targeted immunotherapies or new chemotherapies. A multidisciplinary approach combining various fields of expertise is essential in aiming to limit disease progression as well as patient outcome. Using a narrative literature review approach, the manuscript explores the most up-to-date knowledge concerning locally advanced pancreatic ductal adenocarcinoma management.
Collapse
Affiliation(s)
- Amelia Barcellini
- National Center of Oncological Hadrontherapy (Fondazione CNAO), Pavia, Italy
| | - Andrea Peloso
- Divisions of Transplantation and Visceral Surgery, Department of Surgery, University of Geneva, Geneva, Switzerland
| | - Luigi Pugliese
- General Surgery, Foundation IRCCS San Matteo Hospital, Pavia, Italy
| | - Viviana Vitolo
- National Center of Oncological Hadrontherapy (Fondazione CNAO), Pavia, Italy
| | - Lorenzo Cobianchi
- General Surgery, Foundation IRCCS San Matteo Hospital, Pavia, Italy.,Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, Foundation IRCCS San Matteo Hospital, University of Pavia, Pavia, Italy
| |
Collapse
|
34
|
Arnone A, Laudicella R, Caobelli F, Guglielmo P, Spallino M, Abenavoli E, Martini AL, Filice R, Comis AD, Cuzzocrea M, Linguanti F, Evangelista L, Alongi P. Clinical Impact of 18F-FDG PET/CT in the Diagnostic Workup of Pancreatic Ductal Adenocarcinoma: A Systematic Review. Diagnostics (Basel) 2020; 10:diagnostics10121042. [PMID: 33287195 PMCID: PMC7761738 DOI: 10.3390/diagnostics10121042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/29/2020] [Accepted: 11/30/2020] [Indexed: 12/13/2022] Open
Abstract
In this review, the performance of fluorodeoxyglucose (FDG)-positron emission tomography (PET)/computed tomography (CT) in the diagnostic workup of pancreatic ductal adenocarcinoma (PDAC) is evaluated. A comprehensive literature search up to September 2020 was performed, selecting studies with the presence of: sample size ≥10 patients and index test (i.e., “FDG” or “18F-FDG” AND “pancreatic adenocarcinoma” or “pancreas cancer” AND “PET” or “positron emission tomography”). The methodological quality was evaluated using the revised quality assessment of diagnostic accuracy studies (QUADAS-2) tool and presented according to the preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines. Basic data (authors, year of publication, country and study design), patients’ characteristics (number of enrolled subjects and age), disease phase, type of treatment and grading were retrieved. Forty-six articles met the adopted research criteria. The articles were divided according to the considered clinical context. Namely, besides conventional anatomical imaging, such as computed tomography (CT) and magnetic resonance imaging (MRI), molecular imaging with FDG PET/CT is an important tool in PDAC, for all disease stages. Further prospective studies will be necessary to confirm the cost-effectiveness of such imaging techniques by testing its real potential improvement in the clinical management of PDAC.
Collapse
Affiliation(s)
- Annachiara Arnone
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy; (E.A.); (A.L.M.); (F.L.)
- Correspondence:
| | - Riccardo Laudicella
- Department of Biomedical and Dental Sciences and of Morpho-Functional Imaging, Nuclear Medicine Unit, University of Messina, 98125 Messina, Italy; (R.L.); (R.F.); (A.D.C.)
| | - Federico Caobelli
- Clinic of Radiology & Nuclear Medicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland;
| | - Priscilla Guglielmo
- Nuclear Medicine Division, University Hospital of Parma, 43126 Parma, Italy;
| | - Marianna Spallino
- Nuclear Medicine Unit, ASST “Papa Giovanni XXIII”, 24127 Bergamo, Italy;
| | - Elisabetta Abenavoli
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy; (E.A.); (A.L.M.); (F.L.)
| | - Anna Lisa Martini
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy; (E.A.); (A.L.M.); (F.L.)
| | - Rossella Filice
- Department of Biomedical and Dental Sciences and of Morpho-Functional Imaging, Nuclear Medicine Unit, University of Messina, 98125 Messina, Italy; (R.L.); (R.F.); (A.D.C.)
| | - Alessio Danilo Comis
- Department of Biomedical and Dental Sciences and of Morpho-Functional Imaging, Nuclear Medicine Unit, University of Messina, 98125 Messina, Italy; (R.L.); (R.F.); (A.D.C.)
| | - Marco Cuzzocrea
- Nuclear Medicine, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Flavia Linguanti
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy; (E.A.); (A.L.M.); (F.L.)
| | - Laura Evangelista
- Nuclear Medicine Unit, Department of Medicine, Padova University Hospital, Via Giustiniani 2, 35128 Padova, Italy;
| | - Pierpaolo Alongi
- Unit of Nuclear Medicine, Fondazione Istituto G.Giglio, 90015 Cefalù, Italy;
| |
Collapse
|
35
|
Yang HJ, Song BS, Sim BW, Jung Y, Chae U, Lee DG, Cha JJ, Baek SJ, Lim KS, Choi WS, Lee HY, Son HC, Park SH, Jeong KJ, Kang P, Baek SH, Koo BS, Kim HN, Jin YB, Park YH, Choo YK, Kim SU. Establishment and Characterization of Immortalized Miniature Pig Pancreatic Cell Lines Expressing Oncogenic K-Ras G12D. Int J Mol Sci 2020; 21:ijms21228820. [PMID: 33233448 PMCID: PMC7700231 DOI: 10.3390/ijms21228820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 12/30/2022] Open
Abstract
In recent decades, many studies on the treatment and prevention of pancreatic cancer have been conducted. However, pancreatic cancer remains incurable, with a high mortality rate. Although mouse models have been widely used for preclinical pancreatic cancer research, these models have many differences from humans. Therefore, large animals may be more useful for the investigation of pancreatic cancer. Pigs have recently emerged as a new model of pancreatic cancer due to their similarities to humans, but no pig pancreatic cancer cell lines have been established for use in drug screening or analysis of tumor biology. Here, we established and characterized an immortalized miniature pig pancreatic cell line derived from primary pancreatic cells and pancreatic cancer-like cells expressing K-rasG12D regulated by the human PTF1A promoter. Using this immortalized cell line, we analyzed the gene expression and phenotypes associated with cancer cell characteristics. Notably, we found that acinar-to-ductal transition was caused by K-rasG12D in the cell line constructed from acinar cells. This may constitute a good research model for the analysis of acinar-to-ductal metaplasia in human pancreatic cancer.
Collapse
Affiliation(s)
- Hae-Jun Yang
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
- Department of Biological Science, College of Natural Sciences, Wonkwang University, 460, Iksan-daero, Iksan-si 54538, Korea
| | - Bong-Seok Song
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
| | - Bo-Woong Sim
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
| | - Yena Jung
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
| | - Unbin Chae
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
| | - Dong Gil Lee
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
| | - Jae-Jin Cha
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
| | - Seo-Jong Baek
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
| | - Kyung Seob Lim
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
| | - Won Seok Choi
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (W.S.C.); (S.-H.P.); (K.-J.J.); (S.H.B.); (B.-S.K.); (H.-N.K.); (Y.B.J.)
| | - Hwal-Yong Lee
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
| | - Hee-Chang Son
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
| | - Sung-Hyun Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (W.S.C.); (S.-H.P.); (K.-J.J.); (S.H.B.); (B.-S.K.); (H.-N.K.); (Y.B.J.)
| | - Kang-Jin Jeong
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (W.S.C.); (S.-H.P.); (K.-J.J.); (S.H.B.); (B.-S.K.); (H.-N.K.); (Y.B.J.)
| | - Philyong Kang
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
| | - Seung Ho Baek
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (W.S.C.); (S.-H.P.); (K.-J.J.); (S.H.B.); (B.-S.K.); (H.-N.K.); (Y.B.J.)
| | - Bon-Sang Koo
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (W.S.C.); (S.-H.P.); (K.-J.J.); (S.H.B.); (B.-S.K.); (H.-N.K.); (Y.B.J.)
| | - Han-Na Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (W.S.C.); (S.-H.P.); (K.-J.J.); (S.H.B.); (B.-S.K.); (H.-N.K.); (Y.B.J.)
| | - Yeung Bae Jin
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (W.S.C.); (S.-H.P.); (K.-J.J.); (S.H.B.); (B.-S.K.); (H.-N.K.); (Y.B.J.)
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, 501 Jinjudaero, Jinju 52828, Korea
| | - Young-Ho Park
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Korea
- Correspondence: (Y.-H.P.); (Y.-K.C.); (S.-U.K.); Tel.: +82-43-240-6321 (S.-U.K.); Fax: +82-43-240-6309 (S.-U.K.)
| | - Young-Kug Choo
- Department of Biological Science, College of Natural Sciences, Wonkwang University, 460, Iksan-daero, Iksan-si 54538, Korea
- Correspondence: (Y.-H.P.); (Y.-K.C.); (S.-U.K.); Tel.: +82-43-240-6321 (S.-U.K.); Fax: +82-43-240-6309 (S.-U.K.)
| | - Sun-Uk Kim
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Korea
- Correspondence: (Y.-H.P.); (Y.-K.C.); (S.-U.K.); Tel.: +82-43-240-6321 (S.-U.K.); Fax: +82-43-240-6309 (S.-U.K.)
| |
Collapse
|
36
|
Görte J, Beyreuther E, Danen EHJ, Cordes N. Comparative Proton and Photon Irradiation Combined with Pharmacological Inhibitors in 3D Pancreatic Cancer Cultures. Cancers (Basel) 2020; 12:cancers12113216. [PMID: 33142778 PMCID: PMC7692858 DOI: 10.3390/cancers12113216] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 10/23/2020] [Accepted: 10/29/2020] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Due to higher precision and consequent sparing of normal tissue, pancreatic cancer patients might profit from proton beam radiotherapy, a treatment modality increasingly used. Since molecular data upon proton irradiation in comparison to standard photon radiotherapy are limited in pancreatic cancer, the aims of our study were to unravel differences in the effectiveness of photon versus proton irradiation and to exploit radiation type-specific molecular changes for radiosensitizing 3D PDAC cell cultures. Although protons showed a slightly higher effectiveness and a stronger induction of molecular alterations than photons, our results revealed a radiation-type independent sensitization of molecular-targeted agents selected according to the discovered molecular, radiation-induced alterations. Abstract Pancreatic ductal adenocarcinoma (PDAC) is a highly therapy-resistant tumor entity of unmet needs. Over the last decades, radiotherapy has been considered as an additional treatment modality to surgery and chemotherapy. Owing to radiosensitive abdominal organs, high-precision proton beam radiotherapy has been regarded as superior to photon radiotherapy. To further elucidate the potential of combination therapies, we employed a more physiological 3D, matrix-based cell culture model to assess tumoroid formation capacity after photon and proton irradiation. Additionally, we investigated proton- and photon-irradiation-induced phosphoproteomic changes for identifying clinically exploitable targets. Here, we show that proton irradiation elicits a higher efficacy to reduce 3D PDAC tumoroid formation and a greater extent of phosphoproteome alterations compared with photon irradiation. The targeting of proteins identified in the phosphoproteome that were uniquely altered by protons or photons failed to cause radiation-type-specific radiosensitization. Targeting DNA repair proteins associated with non-homologous endjoining, however, revealed a strong radiosensitizing potential independent of the radiation type. In conclusion, our findings suggest proton irradiation to be potentially more effective in PDAC than photons without additional efficacy when combined with DNA repair inhibitors.
Collapse
Affiliation(s)
- Josephine Görte
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus Technische Universität Dresden, 01307 Dresden, Germany; (J.G.); (E.B.)
- Institute of Radiooncology—OncoRay, Helmholtz-Zentrum Dresden—Rossendorf, 01328 Dresden, Germany
| | - Elke Beyreuther
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus Technische Universität Dresden, 01307 Dresden, Germany; (J.G.); (E.B.)
- Institute of Radiation Physics, Helmholtz-Zentrum Dresden—Rossendorf, 01328 Dresden, Germany
| | - Erik H. J. Danen
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, 2333CC Leiden, The Netherlands;
| | - Nils Cordes
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus Technische Universität Dresden, 01307 Dresden, Germany; (J.G.); (E.B.)
- Institute of Radiooncology—OncoRay, Helmholtz-Zentrum Dresden—Rossendorf, 01328 Dresden, Germany
- German Cancer Consortium, Partner Site Dresden: German Cancer Research Center, 69120 Heidelberg, Germany
- Department of Radiotherapy and Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- Correspondence: ; Tel.: +49-351-458-7401; Fax: +49-351-458-7311
| |
Collapse
|
37
|
De Jesus-Acosta A, Narang A, Mauro L, Herman J, Jaffee EM, Laheru DA. Carcinoma of the Pancreas. ABELOFF'S CLINICAL ONCOLOGY 2020:1342-1360.e7. [DOI: 10.1016/b978-0-323-47674-4.00078-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
38
|
d'Errico M, Alwers E, Zhang Y, Edelmann D, Brenner H, Hoffmeister M. Identification of prognostic DNA methylation biomarkers in patients with gastrointestinal adenocarcinomas: A systematic review of epigenome-wide studies. Cancer Treat Rev 2020; 82:101933. [DOI: 10.1016/j.ctrv.2019.101933] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 11/14/2019] [Accepted: 11/15/2019] [Indexed: 02/07/2023]
|
39
|
Wang W, Li P, Wang Y, Liu B, Li Y. Percutaneous stenting and chemotherapy for unresectable pancreatic cancer: Comparison of irradiation stents vs conventional metal stents. Pancreatology 2019; 19:957-962. [PMID: 31551162 DOI: 10.1016/j.pan.2019.09.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/19/2019] [Accepted: 09/17/2019] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Percutaneous stenting is a palliative method to relieve obstructive jaundice caused by unresectable pancreatic carcinoma. In this study, we aimed to compare the safety and efficacy of irradiation stents and conventional metal stents. METHODS A total of 32 patients who received irradiation stents or conventional metal stents to treat obstructive jaundice caused by locally advanced pancreatic cancer were included in this retrospective study. Chemotherapy using gemcitabine was performed after jaundice subsided. Stent patency, technical success, survival, and complications were compared between groups. RESULTS Seventeen patients were enrolled in the irradiation stent group (ISG), and 15 patients were enrolled in the uncovered stent group (USG). Median and mean stent patency time were 9.8 months (95% CI, 7.682-11.981) and 9.506 months (95% CI, 8.0-11.012) in the ISG, respectively, vs 8.8 months (95% CI, 6.528-11.072) and 7.62 months (95% CI, 5.917-9.323) in the USG, respectively (P = 0.019). Median and mean overall survival were 10.4 months (95% CI, 8.383-12.417) and 9.953 months (95% CI, 8.408-11.498), respectively, in the ISG vs 9.7 months (95% CI, 7.901-11.499) and 8.14 months (95% CI, 6.44-9.84), respectively, in the USG (P = 0.027). CONCLUSIONS Irradiation stents extend stent patency and overall survival compared with conventional biliary stents for the treatment of pancreatic carcinoma complicated by obstructive jaundice. Irradiation stents combined with chemotherapy may be a better choice for the treatment of obstructive jaundice caused by unresectable pancreatic carcinoma.
Collapse
Affiliation(s)
- Wujie Wang
- Department of Interventional Medicine, The Second Hospital of Shandong University, Institute of Tumor Intervention, Shandong University, 247 Beiyuan Street, Jinan City, Shandong Province, 250033, China.
| | - Peimin Li
- Department of Interventional Medicine, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046000, China.
| | - Yongzheng Wang
- Department of Interventional Medicine, The Second Hospital of Shandong University, Institute of Tumor Intervention, Shandong University, 247 Beiyuan Street, Jinan City, Shandong Province, 250033, China.
| | - Bin Liu
- Department of Interventional Medicine, The Second Hospital of Shandong University, Institute of Tumor Intervention, Shandong University, 247 Beiyuan Street, Jinan City, Shandong Province, 250033, China.
| | - Yuliang Li
- Department of Interventional Medicine, The Second Hospital of Shandong University, Institute of Tumor Intervention, Shandong University, 247 Beiyuan Street, Jinan City, Shandong Province, 250033, China.
| |
Collapse
|
40
|
Zang HL, Huang GM, Ju HY, Tian XF. Integrative analysis of the inverse expression patterns in pancreas development and cancer progression. World J Gastroenterol 2019; 25:4727-4738. [PMID: 31528097 PMCID: PMC6718033 DOI: 10.3748/wjg.v25.i32.4727] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/05/2019] [Accepted: 07/19/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND As the malignant tumor, pancreatic cancer with a meager 5-years survival rate has been widely concerning. However, the molecular mechanisms that result in malignant transformation of pancreatic cells remain elusive.
AIM To investigate the gene expression profiles in normal or malignant transformed pancreas development.
METHODS MaSigPro and ANOVA were performed on two pancreas development datasets downloaded from the Gene Expression Omnibus database. Six pancreatic cancer datasets collected from TCGA database were used to establish differentially expressed genes related to pancreas development and pancreatic cancer. Moreover, gene clusters with highly similar interpretation patterns between pancreas development and pancreatic cancer progression were established by self-organizing map and singular value decomposition. Additionally, the hypergeometric test was performed to compare the corresponding interpretation patterns. Abnormal regions of metabolic pathway were analyzed using the Sub-pathway-GM method.
RESULTS This study established the continuously upregulated and downregulated genes at different stages in pancreas development and progression of pancreatic cancer. Through analysis of the differentially expressed genes, we established the inverse and consistent direction development-cancer pattern associations. Based on the application of the Subpathway-GM analysis, we established 17 significant metabolic sub-pathways that were closely associated with pancreatic cancer. Of note, the most significant metabolites sub-pathway was related to glycerophospholipid metabolism.
CONCLUSION The inverse and consistent direction development-cancer pattern associations were established. There was a significant correlation in the inverse patterns, but not consistent direction patterns.
Collapse
Affiliation(s)
- Hong-Liang Zang
- Department of Hepatobiliary and Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Guo-Min Huang
- Department of Hepatobiliary and Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Hai-Ying Ju
- Department of Hematology, Jilin Province Blood Center, Changchun 130000, Jilin Province, China
| | - Xiao-Feng Tian
- Department of Hepatobiliary and Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| |
Collapse
|
41
|
Qiu J, Li M, Du C. Antecolic reconstruction is associated with a lower incidence of delayed gastric emptying compared to retrocolic technique after Whipple or pylorus-preserving pancreaticoduodenectomy. Medicine (Baltimore) 2019; 98:e16663. [PMID: 31441841 PMCID: PMC6716732 DOI: 10.1097/md.0000000000016663] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The aim of present study is to investigate the relationship between the antecolic (AC) route of gastrojejunostomy (GJ) after pancreaticoduodenectomy (PD) or duodenojejunostomy (DJ) reconstruction after pylorus-preserving pancreaticoduodenectomy (PPPD), and the incidence of delayed gastric emptying (DGE). METHODS An electronic search of 4 databases to identify all articles comparing AC and retrocolic (RC) reconstruction after PD or PPPD was performed. RESULTS Fifteen studies involving 2270 patients were included for final pooled analysis. The overall incidence of DGE was 27.2%. Meta-analysis results showed AC group had lower incidence of DGE (odds ratio, 0.29; 95% confidence interval [CI], 0.16-0.52, P < .0001) and shorter hospital length of stay (weight mean difference, -3.29; 95% CI, -5.2 to -1.39, P = .0007). Days until to liquid and solid diet in the AC group were also significantly earlier than that in the RC group (P = .0006 and P < .0001). There was no difference in operative time, incidence of pancreatic fistula and bile leakage, and mortality, respectively. CONCLUSIONS AC route of GJ after PD or DJ after PPPD is associated with a lower incidence of DGE. However, the preferred route for GJ or DJ reconstruction remains to be investigated in well-powered, randomized, controlled trial.
Collapse
|
42
|
Gayam V, Sidhu JS, Mandal A, Garlapati P, Adapa S, Konala VM, Naramala S, Then EO, Maddika S, Gaduputi V. National Trends and Hospitalizations Related to Pancreatic Cancer in Acute Pancreatitis Patients: A Nationwide Inpatient Sample Study. Cureus 2019; 11:e5155. [PMID: 31523582 PMCID: PMC6741367 DOI: 10.7759/cureus.5155] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 07/17/2019] [Indexed: 12/15/2022] Open
Abstract
Background Pancreatic cancer (PC) is one of the common cancers in the United States (U.S.) and is associated with high mortality and morbidity. In spite of the modest improvement in survival, cancer care costs including PC continue to rise and inpatient costs contribute a significant chunk to cancer care, which is often ignored. Acute pancreatitis (AP) is a rare manifestation of PC. This study aims to determine the national trends and associated health care utilization of PC patients hospitalized with AP in the U.S. Methods We used National Inpatient Sample (NIS) to extract data for patients hospitalized with a primary diagnosis of PC in AP in 2016 using International Classification of Diseases, 10th revision, and Clinical Modification (ICD-10-CM) codes. The analysis included disease etiologies, age, race, sex, hospital region, hospital size, institution type, mortality, length of hospital stay (LOS), and commonly associated comorbidities were correlated. Results There were 250 patients with a discharge diagnosis of PC in patients admitted with AP. Most of the patients were whites (76.6%) with the mean age of 39.42 ± 2.51 years, had Medicare (63.26%) as primary insurance, were from Southern region (46%) and had higher Charlson comorbidity index (CCI) (76.00% with CCI > = 3). The mean hospital charges were $48,462.13, and mean LOS was 5.24 days. The LOS was significantly impacted by race, hospital region, endoscopic retrograde cholangiopancreatography (ERCP), and comorbidities such as dementia, smoking, and seizure. Out of the 250 patients admitted with PC, 245 patients (98%) were discharged alive. Conclusions Our study shows a downward trend in LOS, hospital charges, and in-hospital mortality as compared to other studies despite PC and AP presenting together versus PC with or without other etiologies.
Collapse
Affiliation(s)
- Vijay Gayam
- Internal Medicine, Interfaith Medical Center, Brooklyn, USA
| | | | | | | | | | - Venu Madhav Konala
- Internal Medicine / Hematology and Oncology, Ashland Bellefonte Cancer Center, Ashland, USA
| | | | - Eric O Then
- Gastroenterology, St. Barnabas Hospital Health System/Albert Einstein College of Medicine, Bronx, USA
| | - Srikanth Maddika
- Internal Medicine, St. Barnabas Hospital Health System, Bronx, USA
| | - Vinaya Gaduputi
- Internal Medicine, St. Barnabas Hospital Health System, Bronx, USA
| |
Collapse
|
43
|
Haeberle L, Esposito I. Pathology of pancreatic cancer. Transl Gastroenterol Hepatol 2019; 4:50. [PMID: 31304427 DOI: 10.21037/tgh.2019.06.02] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 06/04/2019] [Indexed: 12/14/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy and estimated to become the second leading cause of cancer-related deaths by 2030. Although overall 5-year survival rates have constantly remained below 10% for the last decades, several key points important for accurate patient stratification have emerged during recent years. These key points include a highly standardized gross examination of PDAC resection specimens, using an axial slicing technique and inking of the circumferential resection margin (CRM), as well as a meticulous microscopic examination, taking into account the prognostic relevance of factors such as the exact resection status (R0 vs. R1 1-mm vs. R1 resection), histopathological tumor grading and the so-called lymph node ratio (LNR). With increasing use of neoadjuvant therapy in PDAC, tumor regression grading (TRG) for PDAC is currently rising in relevance in order to stratify and manage pre-operatively treated PDAC patients. As all current TRG systems for PDAC are unsatisfactory, new standardized international protocols are urgently needed. Several morphological subtypes of PDAC exist, some of which share the same molecular background with classical PDAC, while others are characterized by a distinct molecular pathogenesis. While some show a prognosis similar to classical PDAC, other subtypes stand out due to a better or even worse prognosis than classical PDAC. Prognostic relevant molecular subtypes of PDAC have been proposed as well, however, limitations of used cohorts and the lacking correlation of molecular subtypes with histomorphological subtypes limit the translation of these findings into valuable clinical applications. Lastly, several macroscopic and microscopic precursor lesions of PDAC have been described in genetically engineered mouse models (GEMM) and humans in recent times, providing further insight into PDAC carcinogenesis. In addition, improved diagnosis of PDAC precursors represents a chance to select patients for resection before invasive PDAC is present.
Collapse
Affiliation(s)
- Lena Haeberle
- Institute of Pathology, Heinrich Heine University and University Hospital of Duesseldorf, Germany
| | - Irene Esposito
- Institute of Pathology, Heinrich Heine University and University Hospital of Duesseldorf, Germany
| |
Collapse
|
44
|
Doppler tissue perfusion measurement is a sensitive and specific tool for a differentiation between malignant and inflammatory pancreatic tumors. PLoS One 2019; 14:e0215944. [PMID: 31034484 PMCID: PMC6488051 DOI: 10.1371/journal.pone.0215944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 04/11/2019] [Indexed: 01/12/2023] Open
Abstract
Differentiation between pancreatic malignant and inflammatory tumors presents an important diagnostic problem. The ability to recognize pancreatic malignant tumors using Doppler evaluation of tissue perfusion has been recently demonstrated. The aim of the study was to assess the diagnostic value of Dynamic Tissue Perfusion Measurement (DTPM) in the differentiation between malignant and inflammatory pancreatic tumors. The study included 60 patients (35M, 25F, age 60.9 ± 2.3 years) with a malignant (Group 1, n = 30) or inflammatory (Group 2, n = 30) pancreatic tumor undergoing endoscopic ultrasound with the evaluation of tissue perfusion by Color Doppler and a simultaneous biopsy of lesions for cytological evaluation. In 20 patients the diagnosis was verified in the postoperative histopathological examination. Flow velocity (FV) and percentiles of the distribution of perfusion intensity (PR) evaluated by DTPM were analyzed with regard to receiver-operator-characteristics. FV as well as PR were significantly higher in Group 2 compared to Group 1. A threshold of 2.0 cm/sec for FV identified patients with malignancies with a sensitivity of 83% and specificity of 86%. In multivariable regression analysis, the best PR parameter for differentiating between malignant and inflammatory tumors was 97.5% percentile, whose value of 0.922 allowed for the recognition of pancreatic malignant tumors with a sensitivity of 62% and specificity of 83% (p < 0.001). In conclusion, Color Doppler ultrasound tissue perfusion parameters are a sensitive and specific tool in the differentiation between malignant and inflammatory pancreatic tumors.
Collapse
|
45
|
Macarulla T, Pazo-Cid R, Guillén-Ponce C, López R, Vera R, Reboredo M, Muñoz Martin A, Rivera F, Díaz Beveridge R, La Casta A, Martín Valadés J, Martínez-Galán J, Ales I, Sastre J, Perea S, Hidalgo M. Phase I/II Trial to Evaluate the Efficacy and Safety of Nanoparticle Albumin-Bound Paclitaxel in Combination With Gemcitabine in Patients With Pancreatic Cancer and an ECOG Performance Status of 2. J Clin Oncol 2019; 37:230-238. [DOI: 10.1200/jco.18.00089] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose Gemcitabine plus nanoparticle albumin-bound (NAB) paclitaxel (GA) significantly improved survival compared with gemcitabine alone in patients with metastatic pancreatic ductal adenocarcinoma (PDAC) and a Karnofsky performance status (PS) of 70% or greater. Because of the low number of patients with reduced PS, the efficacy of this regimen in fragile patients remains unclear. This study aimed to evaluate the efficacy and tolerability of different GA dosing regimens in patients with a poor PS. Patients and Methods In the phase I part of this study, patients were randomly assigned to one of the following four parallel GA treatment arms (six patients per arm): a biweekly schedule of NAB-paclitaxel (150 mg/m2 [arm A] or 125 mg/m2 [arm C]) plus gemcitabine 1,000 mg/m2 or a standard schedule of 3 weeks on and 1 week off of NAB-paclitaxel (100 mg/m2 [arm B] or 125 mg/m2 [arm D]) plus gemcitabine 1,000 mg/m2. The two regimens with the better tolerability profile on the basis of predefined criteria were evaluated in the phase II part of the study, the primary end point of which was 6-month actuarial survival. Results Arms B and D were selected for the phase II part of the study. A total of 221 patients (111 patients in arm B and 110 patients in arm D) were enrolled. Baseline characteristics including median age (71 and 68 years in arms B and D, respectively), sex (51% and 55% men in arms B and D, respectively), and metastatic disease (88% and 84% in arms B and D, respectively) were comparable between arms. The most frequent grade 3 or 4 toxicities in arms B and D were anemia (12% and 7%, respectively), neutropenia (32% and 30%, respectively), thrombocytopenia (7% and 11%, respectively), asthenia (14% and 16%, respectively), and neurotoxicity (11% and 16%, respectively). In arms B and D, there were no significant differences in response rate (24% and 28%, respectively), median progression-free survival (5.7 and 6.7 months, respectively), and 6-month overall survival (63% and 69%, respectively). Conclusion NAB-paclitaxel administered at either 100 and 125 mg/m2 in combination with gemcitabine on days 1, 8, and 15 every 28 days is well tolerated and results in acceptable safety and efficacy in patients with metastatic pancreatic ductal adenocarcinoma and a poor PS.
Collapse
Affiliation(s)
- Teresa Macarulla
- Vall d’Hebrón University Hospital and Vall d’Hebrón Institute of Oncology, Barcelona, Spain
| | | | | | - Rafael López
- Hospital Clínico de Santiago, Santiago de Compostela, Spain
| | - Ruth Vera
- Hospital de Navarra, Pamplona, Spain
| | | | | | - Fernando Rivera
- Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | | | | | | | | | | | - Javier Sastre
- Hospital Clínico Universitario San Carlos, Madrid, Spain
| | - Sofia Perea
- Centro Nacional de Investigaciones Oncológicas and Centro Integral Oncológico Clara Campal, Madrid, Spain
| | - Manuel Hidalgo
- Centro Nacional de Investigaciones Oncológicas and Centro Integral Oncológico Clara Campal, Madrid, Spain
| |
Collapse
|
46
|
Cornelissen B, Knight JC, Mukherjee S, Evangelista L, Xavier C, Caobelli F, Del Vecchio S, Rbah-Vidal L, Barbet J, de Jong M, van Leeuwen FWB. Translational molecular imaging in exocrine pancreatic cancer. Eur J Nucl Med Mol Imaging 2018; 45:2442-2455. [PMID: 30225616 PMCID: PMC6208802 DOI: 10.1007/s00259-018-4146-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 08/22/2018] [Indexed: 02/06/2023]
Abstract
Effective treatment for pancreatic cancer remains challenging, particularly the treatment of pancreatic ductal adenocarcinoma (PDAC), which makes up more than 95% of all pancreatic cancers. Late diagnosis and failure of chemotherapy and radiotherapy are all too common, and many patients die soon after diagnosis. Here, we make the case for the increased use of molecular imaging in PDAC preclinical research and in patient management.
Collapse
Affiliation(s)
- Bart Cornelissen
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, Oxford University, Oxford, UK.
| | - James C Knight
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, Oxford University, Oxford, UK
| | - Somnath Mukherjee
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, Oxford University, Oxford, UK
| | | | | | - Federico Caobelli
- Department of Radiology, Universitätsspital Basel, Basel, Switzerland
| | | | - Latifa Rbah-Vidal
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Jacques Barbet
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Marion de Jong
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Fijs W B van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
47
|
Bhandari S, Abdul MKM, Hollabaugh W, Sharma K, Evans DB, Guda N. Decreased trend in hospital mortality from pancreatic cancer despite increase in number of hospital admissions. PLoS One 2018; 13:e0199909. [PMID: 30020978 PMCID: PMC6051597 DOI: 10.1371/journal.pone.0199909] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 06/16/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND AIM Pancreatic cancer is one of the common cancers in US and is associated with high mortality and morbidity. The objectives of our study were to look at the recent trends in the number of hospitalizations with pancreatic cancer. METHODS We identified patients with a discharge diagnosis of pancreatic cancer in the National Inpatient Sample from 2007 to 2011 using International Classification of Diseases-Clinical Modification, 9th revision (ICD-9-CM) codes. We looked at the yearly trend in the hospitalizations with pancreatic cancer and the outcomes which included length of stay (LOS), hospital charges and in-hospital mortality. We also performed multivariate analysis to look for the predictors of mortality. RESULTS There were 450, 414 patients with discharge diagnosis of pancreatic cancer. There was 18% increase in hospitalizations with pancreatic cancer in 2011 compared to 2007. Most of the patients were Caucasian (63%) with the mean age of 68 ± 0.14 years, had Medicare (57%) as primary insurance, were from Southern region (35%) and had higher Charlson Comorbidity Index (CCI) (87% with CCI > = 5). 6% underwent Whipple's procedure in the index hospitalization. After the adjustment for inflation, the mean hospital charges increased from $ 47,331 in 20007 to $ 53, 854 in 2011 (p = 0.01). LOS decreased from 7.31 ± 0.11 days in 2007 to 6.70 ± 0.09 days in 2011 (<0.001). Despite the increase in the number of hospitalizations of patients with pancreatic cancer, mortality decreased from 9.8% in 2007 to 8.1% in 2011 (p<0.001). On multivariate analysis, the independent factors associated with higher mortality were older age, male sex African-American race, insurance status other than Medicare, higher CCI and enrollment in palliative care. There was regional variation in mortality. Whipple's procedure conferred lower mortality. CONCLUSIONS Our study showed downward trends in LOS and in-hospital mortality despite increasing hospitalizations with pancreatic cancer.
Collapse
Affiliation(s)
- Sanjay Bhandari
- Division of General Internal Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States of America
- * E-mail:
| | | | - Will Hollabaugh
- Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Kanav Sharma
- Division of General Internal Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Douglas B. Evans
- Division of Surgical Oncology, Department of Surgery, Medical College of Wisconsin Milwaukee, WI, United States of America
| | - Nalini Guda
- Division of Gastroenterology, Aurora Saint Luke’s Medical Center, Milwaukee, WI, United States of America
| |
Collapse
|
48
|
Abstract
Purpose of review This article provides a brief overview of cancer-preventive phytochemicals specifically targeting pancreatic cancer (PC) stem cells for prevention and treatment. Recent findings Cancer stem cells (CSCs) represent a small proportion of the total cells of a given tumor, and contribute to tumor growth, recurrence, metastasis, and treatment resistance. Many intertwined pathways, including hedgehog, Wnt Signaling, and NOTCH, have been shown to play a role in the formation of CSCs. Recently, numerous chemopreventive agents, such as genistein, (-)-epigallocatechin-3-gallate (EGCG), sulforaphane, curcumin, resveratrol, and quercetin, have been shown to target CSCs mediated through the inhibition of multiple signalling pathways, to avoid toxicity and the side effects of chemical compounds. Summary A growing body of research suggests that CSCs are the drivers in treatment resistance, cancer recurrence, and metastasis, in addition to tumor initiation and heterogeneity. Patient survival depends on these CSCs, which are one cause of tumor recurrence after surgery and chemotherapy. Therefore, target selection; an improved understanding of CSC biology, the genetic and molecular profiles of CSCs, and their key signaling pathways, and; appropriate clinical trials endpoints that are designed to target CSCs will help in the development of drugs that will specifically target this small population of stem cells.
Collapse
|
49
|
Totti S, Allenby MC, Dos Santos SB, Mantalaris A, Velliou EG. A 3D bioinspired highly porous polymeric scaffolding system for in vitro simulation of pancreatic ductal adenocarcinoma. RSC Adv 2018; 8:20928-20940. [PMID: 35542351 PMCID: PMC9080900 DOI: 10.1039/c8ra02633e] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 05/20/2018] [Indexed: 12/21/2022] Open
Abstract
Pancreatic ductal adenocarcinoma is an aggressive disease with an extremely low survival rate. This is due to the (i) poor prognosis and (ii) high resistance of the disease to current treatment options. The latter is partly due to the very complex and dense tissue/tumour microenvironment of pancreatic cancer, which contributes to the disease's progression and the inhibition of apoptotic pathways. Over the last years, advances in tissue engineering and the development of three-dimensional (3D) culture systems have shed more light into cancer research by enabling a more realistic recapitulation of the niches and structure of the tumour microenvironment. Herein, for the first time, 3D porous polyurethane scaffolds were fabricated and coated with fibronectin to mimic features of the structure and extracellular matrix present in the pancreatic cancer tumour microenvironment. The developed 3D scaffold could support the proliferation of the pancreatic tumour cells, which was enhanced with the presence of fibronectin, for a month, which is a significantly prolonged in vitro culturing duration. Furthermore, in situ imaging of cellular and biomarker distribution showed the formation of dense cellular masses, the production of collagen-I by the cells and the formation of environmental stress gradients (e.g. HIF-1α) with similar heterogeneity trends to the ones reported in in vivo studies. The results obtained in this study suggest that this bioinspired porous polyurethane based scaffold has great potential for in vitro high throughput studies of pancreatic cancer including drug and treatment screening.
Collapse
Affiliation(s)
- Stella Totti
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey Guildford GU2 7XH UK 0044-(0)-1483686577
| | - Mark C Allenby
- Biological Systems Engineering Laboratory (BSEL), Department of Chemical Engineering, Imperial College London London SW7 2AZ UK
| | - Susana Brito Dos Santos
- Biological Systems Engineering Laboratory (BSEL), Department of Chemical Engineering, Imperial College London London SW7 2AZ UK
| | - Athanasios Mantalaris
- Biological Systems Engineering Laboratory (BSEL), Department of Chemical Engineering, Imperial College London London SW7 2AZ UK
| | - Eirini G Velliou
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey Guildford GU2 7XH UK 0044-(0)-1483686577
| |
Collapse
|
50
|
Estándares de calidad en la cirugía oncológica pancreática en España. Cir Esp 2018; 96:342-351. [PMID: 29784432 DOI: 10.1016/j.ciresp.2018.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 01/02/2018] [Accepted: 03/06/2018] [Indexed: 12/18/2022]
|