401
|
Baltatzis M, Rodriquenz MG, Siriwardena AK, De Liguori Carino N. Contemporary management of pancreas cancer in older people. Eur J Surg Oncol 2020; 47:560-568. [PMID: 32950314 DOI: 10.1016/j.ejso.2020.08.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 08/10/2020] [Indexed: 12/20/2022] Open
Abstract
As the population of western countries is aging, the number of patients diagnosed with cancer is growing. Therefore older people, more susceptible to develop pancreatic malignancy, will likely represent the prototype of a pancreatic cancer patient in the near future. Diagnostic modalities utilised for younger patients are also applicable for older individuals. There is accumulative evidence that biological age is not an independent factor predicting poor outcome in elderly patients with resectable disease undergoing surgery, however increased postoperative morbidity and mortality within the elderly group has also been reported. Adjuvant chemotherapy should be offered in all patients with good performance status regardless of their age. Palliative measures for unresectable tumours including relief from biliary and duodenal obstruction as well as chemotherapy should be considered in non-frail patients with reasonable life expectancy. Palliative chemotherapy options are FOLFIRINOX or gemcitabine/nab-paclitaxel for patients with good performance status (0-1) and gemcitabine alone for patients with performance status 2-3. The cornerstone for improving the outcomes of the elderly age group is careful patient selection and perioperative optimization of those who have indication for surgery. Patients and their carers should be involved in the decision making process with emphasis on the expected functional recovery after the proposed treatment modality. The presence of geriatricians in the multidisciplinary team meetings is crucial in order to identify the optimal treatment pathway for elderly patients. Geriatric input regarding peri-habilitation pathways to improve surgical outcomes, to decrease mortality and to expedite patients' functional recovery is highly recommended.
Collapse
Affiliation(s)
- Minas Baltatzis
- Regional Hepato-Pancreatico-Biliary Unit, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, UK.
| | - Maria Grazia Rodriquenz
- Oncology Unit, Foundation IRCCS, Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Ajith K Siriwardena
- Regional Hepato-Pancreatico-Biliary Unit, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, UK
| | - Nicola De Liguori Carino
- Regional Hepato-Pancreatico-Biliary Unit, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, UK
| |
Collapse
|
402
|
Paoli C, Carrer A. Organotypic Culture of Acinar Cells for the Study of Pancreatic Cancer Initiation. Cancers (Basel) 2020; 12:E2606. [PMID: 32932616 PMCID: PMC7564199 DOI: 10.3390/cancers12092606] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/02/2020] [Accepted: 09/09/2020] [Indexed: 12/17/2022] Open
Abstract
The carcinogenesis of pancreatic ductal adenocarcinoma (PDA) progresses according to multi-step evolution, whereby the disease acquires increasingly aggressive pathological features. On the other hand, disease inception is poorly investigated. Decoding the cascade of events that leads to oncogenic transformation is crucial to design strategies for early diagnosis as well as to tackle tumor onset. Lineage-tracing experiments demonstrated that pancreatic cancerous lesions originate from acinar cells, a highly specialized cell type in the pancreatic epithelium. Primary acinar cells can survive in vitro as organoid-like 3D spheroids, which can transdifferentiate into cells with a clear ductal morphology in response to different cell- and non-cell-autonomous stimuli. This event, termed acinar-to-ductal metaplasia, recapitulates the histological and molecular features of disease initiation. Here, we will discuss the isolation and culture of primary pancreatic acinar cells, providing a historical and technical perspective. The impact of pancreatic cancer research will also be debated. In particular, we will dissect the roles of transcriptional, epigenetic, and metabolic reprogramming for tumor initiation and we will show how that can be modeled using ex vivo acinar cell cultures. Finally, mechanisms of PDA initiation described using organotypical cultures will be reviewed.
Collapse
Affiliation(s)
- Carlotta Paoli
- Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy;
- Department of Biology, University of Padova, 35129 Padova, Italy
| | - Alessandro Carrer
- Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy;
- Department of Biology, University of Padova, 35129 Padova, Italy
| |
Collapse
|
403
|
Kumai T, Mizukoshi E, Hashiba T, Nakagawa H, Kitahara M, Miyashita T, Mochizuki T, Goto S, Kamigaki T, Takimoto R, Yamashita T, Sakai Y, Yamashita T, Honda M, Tomita K, Kaneko S. Effect of adoptive T-cell immunotherapy on immunological parameters and prognosis in patients with advanced pancreatic cancer. Cytotherapy 2020; 23:137-145. [PMID: 32907781 DOI: 10.1016/j.jcyt.2020.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/22/2020] [Accepted: 08/02/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND AIMS Immunotherapy is effective for many types of cancer, but its benefits in advanced pancreatic cancer, which has a poor prognosis, are not well established. In this study, the authors examined the effects of adoptive T-cell immunotherapy (ATI) on immune cell profiles and prognosis in patients with unresectable advanced pancreatic cancer. METHODS Seventy-seven patients with unresectable advanced pancreatic cancer were treated with six cycles of αβ T cells alone or in combination with chemotherapy or chemoradiation. Immune cell profiles in peripheral blood samples obtained before and after treatment were comprehensively evaluated by flow cytometry. Furthermore, associations between changes in immune cell frequencies and prognosis were determined. RESULTS ATI prolonged survival to 18.7 months compared with previous estimates of 6.2-11.1 months for patients treated with chemotherapy alone. ATI decreased CD3+CD4+CD8- T cell frequency in peripheral blood and increased CD3+CD4-CD8+ T cell frequency. An increase in CD3+ T cells and CD3+TCRγδ- T cells in peripheral blood after treatment was associated with a good prognosis. CONCLUSIONS ATI altered the immune profile in peripheral blood, including CD3+CD4-CD8+ T cells, and improved prognosis in pancreatic cancer.
Collapse
Affiliation(s)
- Tatsuo Kumai
- Department of Gastroenterology, Graduate School of Medicine, Kanazawa University, Kanazawa, Japan
| | - Eishiro Mizukoshi
- Department of Gastroenterology, Graduate School of Medicine, Kanazawa University, Kanazawa, Japan.
| | | | | | | | | | | | - Shigenori Goto
- Seta Clinic Group, Department of Next-Generation Cell and Immune Therapy, Juntendo University School of Medicine, Tokyo, Japan
| | - Takashi Kamigaki
- Seta Clinic Group, Department of Next-Generation Cell and Immune Therapy, Juntendo University School of Medicine, Tokyo, Japan
| | - Rishu Takimoto
- Seta Clinic Group, Department of Next-Generation Cell and Immune Therapy, Juntendo University School of Medicine, Tokyo, Japan
| | - Taro Yamashita
- Department of Gastroenterology, Graduate School of Medicine, Kanazawa University, Kanazawa, Japan
| | - Yoshio Sakai
- Department of Gastroenterology, Graduate School of Medicine, Kanazawa University, Kanazawa, Japan
| | - Tatsuya Yamashita
- Department of Gastroenterology, Graduate School of Medicine, Kanazawa University, Kanazawa, Japan
| | - Masao Honda
- Department of Gastroenterology, Graduate School of Medicine, Kanazawa University, Kanazawa, Japan
| | | | - Shuichi Kaneko
- Department of Gastroenterology, Graduate School of Medicine, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
404
|
Dunet V, Halkic N, Sempoux C, Demartines N, Montemurro M, Prior JO, Schmidt S. Prediction of tumour grade and survival outcome using pre-treatment PET- and MRI-derived imaging features in patients with resectable pancreatic ductal adenocarcinoma. Eur Radiol 2020; 31:992-1001. [PMID: 32851447 PMCID: PMC7813698 DOI: 10.1007/s00330-020-07191-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/29/2020] [Accepted: 08/12/2020] [Indexed: 11/29/2022]
Abstract
OBJECTIVES To perform a correlation analysis between histopathology and imaging in patients with previously untreated pancreatic ductal adenocarcinoma (PDAC) and to determine the prognostic values of clinical, histological, and imaging parameters regarding overall survival (OS), disease-specific survival (DSS), and progression-free survival (PFS). METHODS This single-centre study prospectively included 61 patients (32 males; median age, 68.0 years [IQR, 63.0-75.0 years]) with histologically confirmed PDAC and following surgical resection who preoperatively underwent 18F-FDG PET/CT and DW-MRI. On whole lesions, we measured, using a 42% SUVmax threshold volume of interest (VOI), the following quantitative parameters: mean and maximum standardised uptake values (SUVmean and SUVmax), total lesion glycolysis (TLG), metabolic tumour volume (MTV), mean and minimum apparent diffusion coefficient (ADCmean and ADCmin), diffusion total volume (DTV), and MTV/ADCmin ratio. Spearman's correlation analysis was performed to assess relationships between these markers and histopathological findings from surgical specimens (stage; grade; resection quality; and vascular, perineural, and lymphatic invasion). Kaplan-Meier and Cox hazard ratio methods were used to evaluate the impacts of imaging parameters on OS (n = 41), DSS (n = 36), and PFS (n = 41). RESULTS Inverse correlations between ADCmin and SUVmax (rho = - 0.34; p = 0.0071), and between SUVmean and ADCmean (rho = - 0.29; p = 0.026) were identified. ADCmin was inversely correlated with tumour grade (rho = - 0.40; p = 0.0015). MTV was an independent predictive factor for OS and DSS, while DTV was an independent predictive factor for PFS. CONCLUSION In previously untreated PDAC, ADC and SUV values are correlated. Combining PET-MRI metrics may help predict PDAC grade and patients' survival. KEY POINTS • Minimum apparent diffusion coefficient derived from DW-MRI inversely correlates with tumour grade in pancreatic ductal adenocarcinoma. • In pancreatic ductal adenocarcinoma, metabolic tumour volume has been confirmed as a predictive factor for patients' overall survival and disease-specific survival. • Combining PET and MRI metrics may help predict grade and patients' survival in pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Vincent Dunet
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Nermin Halkic
- Department of Visceral Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Christine Sempoux
- Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Nicolas Demartines
- Department of Visceral Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Michael Montemurro
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - John O Prior
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland.
| | - Sabine Schmidt
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
405
|
The Diagnostic Accuracy of Mutant KRAS Detection from Pancreatic Secretions for the Diagnosis of Pancreatic Cancer: A Meta-Analysis. Cancers (Basel) 2020; 12:cancers12092353. [PMID: 32825312 PMCID: PMC7564395 DOI: 10.3390/cancers12092353] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/14/2020] [Accepted: 08/18/2020] [Indexed: 12/18/2022] Open
Abstract
This meta-analysis aims to identify the diagnostic accuracy of mutations in the Kirsten Rat Sarcoma (KRAS) oncogene in the diagnosis of pancreatic ductal adenocarcinoma (PDAC). The survival of PDAC remains poor often due to the fact that disease is advanced at diagnosis. We analysed 22 studies, with a total of 2156 patients, to identify if the detection of KRAS mutations from pancreatic exocrine secretions yields sufficient specificity and sensitivity to detect patients with PDAC amongst healthy individuals. The majority of the studies were retrospective, samples were obtained endoscopically or surgically, and included comparator populations of patients with chronic pancreatitis and pre-malignant pancreatic lesions (PanIN) as well as healthy controls. We performed several analyses to identify the diagnostic accuracy for PDAC among these patient populations. Our results highlighted that the diagnostic accuracy of KRAS mutation for PDAC was of variable sensitivity and specificity when compared with PanINs and chronic pancreatitis, but had a higher specificity among healthy individuals. The sensitivity of this test must be improved to prevent missing early PDAC or PanINs. This could be achieved with rigorous prospective cohort studies, in which high-risk patients with normal cross-sectional imaging undergo surveillance following KRAS mutation testing.
Collapse
|
406
|
Li Y, Shen J, Cheng CS, Gao H, Zhao J, Chen L. Overexpression of pyruvate dehydrogenase phosphatase 1 promotes the progression of pancreatic adenocarcinoma by regulating energy-related AMPK/mTOR signaling. Cell Biosci 2020; 10:95. [PMID: 32782783 PMCID: PMC7412669 DOI: 10.1186/s13578-020-00457-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 07/29/2020] [Indexed: 12/12/2022] Open
Abstract
Background Human pyruvate dehydrogenase phosphatase 1 (PDP1) plays an important physiological role in energy metabolism; however, its expression and function in human pancreatic adenocarcinoma (PDAC) remain unknown. This study aimed to investigate the expression pattern and mechanisms of action of PDP1 in human PDAC. Methods The expression pattern of PDP1 in PDAC was determined, and its correlation with patient survival was analyzed. Ectopic expression or knockdown of PDP1 was performed, and in vitro proliferation and migration, as well as in vivo tumor growth of PDAC, were measured. The mechanism was studied by biochemical approaches. Results PDP1 was overexpressed in human PDAC samples, and high expression of PDP1 correlated with poor overall and disease-free survival of PDAC patients. PDP1 promoted the proliferation, colony formation, and invasion of PDAC cells in vitro and facilitated orthotopic tumor growth in vivo. PDP1 accelerated intracellular ATP production, leading to sufficient energy to support rapid cancer progression. mTOR activation was responsible for the PDP1-induced tumor cell proliferation and invasion in PDAC. AMPK was downregulated by PDP1 overexpression, resulting in mTOR activation and cancer progression. Conclusion Our findings suggested that PDP1 could be a promising diagnostic and therapeutic target for anti-PDAC treatment.
Collapse
Affiliation(s)
- Ye Li
- Department of Integrated Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Jia Shen
- Department of Oncology, First People's Hospital of Pinghu, Zhejiang, 314200 China
| | - Chien-Shan Cheng
- Department of Integrated Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - HuiFeng Gao
- Department of Integrated Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Jiangang Zhao
- Department of Oncology, Shaoxing Central Hospital, Zhejiang, 312030 China
| | - Lianyu Chen
- Department of Integrated Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| |
Collapse
|
407
|
Miulescu R, Balaban DV, Sandru F, Jinga M. Cutaneous Manifestations in Pancreatic Diseases-A Review. J Clin Med 2020; 9:2611. [PMID: 32806580 PMCID: PMC7464368 DOI: 10.3390/jcm9082611] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 02/07/2023] Open
Abstract
Pancreatic pathology, comprising acute and chronic pancreatitis, autoimmune pancreatitis and pancreatic neoplasms, primarily presents with gastrointestinal symptoms and signs; however, it is well recognized that it can also associate a wide range of extra-digestive features. Among these systemic manifestations, cutaneous involvement plays an important role both as a diagnostic clue for the pancreatic disease itself and serving as a prognostic factor for the severity of the condition. Recognition of these cutaneous signs is, however, far from being satisfactory, all the more as some of them are relatively rare. In the current review, we discuss skin involvement in pancreatic diseases, referring to pancreatic panniculitis, cutaneous hemorrhagic manifestations, skin metastasis, acanthosis nigricans, livedo reticularis, necrolytic migratory erythema and cutaneous fistula. We highlight the clinical characteristics, treatment and prognostic value of these lesions. Better awareness among medical specialties other than dermatology is needed for detection of the skin clues associated with pancreatic pathology.
Collapse
Affiliation(s)
- Raluca Miulescu
- Dermatology Department, Elias University Emergency Hospital, 011461 Bucharest, Romania; (R.M.); (F.S.)
| | - Daniel Vasile Balaban
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 030167 Bucharest, Romania;
- Gastroenterology Department, “Dr. Carol Davila” Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Florica Sandru
- Dermatology Department, Elias University Emergency Hospital, 011461 Bucharest, Romania; (R.M.); (F.S.)
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 030167 Bucharest, Romania;
| | - Mariana Jinga
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 030167 Bucharest, Romania;
- Gastroenterology Department, “Dr. Carol Davila” Central Military Emergency University Hospital, 010825 Bucharest, Romania
| |
Collapse
|
408
|
Mazloom A, Ghalehsari N, Gazivoda V, Nimkar N, Paul S, Gregos P, Rateshwar J, Khan U. Role of Immune Checkpoint Inhibitors in Gastrointestinal Malignancies. J Clin Med 2020; 9:E2533. [PMID: 32781500 PMCID: PMC7463795 DOI: 10.3390/jcm9082533] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/28/2020] [Accepted: 07/31/2020] [Indexed: 12/24/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of several solid and hematological malignancies. ICIs are not only able to produce long and durable responses, but also very well tolerated by patients. There are several approved indications of use of ICIs in treatment of metastatic gastrointestinal malignancies including gastric, esophageal, colorectal and hepatocellular carcinoma. In addition, ICIs can be used in microsatellite instability-high (MSI-H) and high tumor mutational burden (TMB) tumors in chemotherapy-resistant setting. Despite having good efficacy and superior safety profile, ICIs are clinically active in small subset of patients, therefore, there is a huge unmet need to enhance their efficacy and discover new predictive biomarkers. There are several ongoing clinical trials that are exploring the role of ICIs in various gastrointestinal cancers either as single agent or in combination with chemotherapy, radiation therapy, targeted agents or other immunotherapeutic agents. In this review, we discuss the published and ongoing trials for ICIs in gastrointestinal malignancies, including esophageal, gastric cancer, pancreatic, hepatocellular, biliary tract, colorectal and anal cancers. Specifically, we focus on the use of ICIs in each line of therapy and discuss the future directions of these agents in each type of gastrointestinal cancer.
Collapse
Affiliation(s)
- Anita Mazloom
- Department of Medicine, NewYork-Presbyterian Brooklyn Methodist Hospital—Weill Cornell Medicine, Brooklyn, NY 11215, USA; (A.M.); (N.G.); (N.N.); (S.P.); (P.G.); (J.R.)
| | - Nima Ghalehsari
- Department of Medicine, NewYork-Presbyterian Brooklyn Methodist Hospital—Weill Cornell Medicine, Brooklyn, NY 11215, USA; (A.M.); (N.G.); (N.N.); (S.P.); (P.G.); (J.R.)
| | - Victor Gazivoda
- Department of Surgery, Maimonides Medical Center, Brooklyn, NY 11219, USA;
| | - Neil Nimkar
- Department of Medicine, NewYork-Presbyterian Brooklyn Methodist Hospital—Weill Cornell Medicine, Brooklyn, NY 11215, USA; (A.M.); (N.G.); (N.N.); (S.P.); (P.G.); (J.R.)
| | - Sonal Paul
- Department of Medicine, NewYork-Presbyterian Brooklyn Methodist Hospital—Weill Cornell Medicine, Brooklyn, NY 11215, USA; (A.M.); (N.G.); (N.N.); (S.P.); (P.G.); (J.R.)
| | - Peter Gregos
- Department of Medicine, NewYork-Presbyterian Brooklyn Methodist Hospital—Weill Cornell Medicine, Brooklyn, NY 11215, USA; (A.M.); (N.G.); (N.N.); (S.P.); (P.G.); (J.R.)
| | - Janice Rateshwar
- Department of Medicine, NewYork-Presbyterian Brooklyn Methodist Hospital—Weill Cornell Medicine, Brooklyn, NY 11215, USA; (A.M.); (N.G.); (N.N.); (S.P.); (P.G.); (J.R.)
| | - Uqba Khan
- Department of Medicine, NewYork-Presbyterian Brooklyn Methodist Hospital—Weill Cornell Medicine, Brooklyn, NY 11215, USA; (A.M.); (N.G.); (N.N.); (S.P.); (P.G.); (J.R.)
| |
Collapse
|
409
|
Li TJ, Wang WQ, Yu XJ, Liu L. Killing the "BAD": Challenges for immunotherapy in pancreatic cancer. Biochim Biophys Acta Rev Cancer 2020; 1874:188384. [PMID: 32531324 DOI: 10.1016/j.bbcan.2020.188384] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/19/2020] [Accepted: 06/06/2020] [Indexed: 12/18/2022]
Abstract
Cancer regression often fails after systemic immune activation, especially for solid tumors due to their local immunosuppressive microenvironments. Among these, the pancreatic cancer microenvironment is unique and an important reason for resistance to anti-cancer treatments that include immunotherapy. In this review, the three main "BAD" characteristics that create and maintain this immunosuppressive microenvironment are discussed for effector T cells: Barriers to overcome, Attraction problems, and their Disabilities. These inhibit both effector T-cell activation and infiltration, reducing immunotherapy effectiveness. Combination approaches for killing the "BAD" aim to normalize the tumor microenvironment and are recommended to enhance anti-cancer immune-system efficacy in pancreatic cancer.
Collapse
MESH Headings
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/immunology
- CTLA-4 Antigen/antagonists & inhibitors
- CTLA-4 Antigen/immunology
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/immunology
- Humans
- Immunotherapy, Adoptive/methods
- Lymphocyte Activation/drug effects
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Mutation
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/therapy
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/immunology
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- Treatment Outcome
- Tumor Escape/drug effects
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/genetics
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Tian-Jiao Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Pancreatic Cancer Institute, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Wen-Quan Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Pancreatic Cancer Institute, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xian-Jun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Pancreatic Cancer Institute, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| | - Liang Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Pancreatic Cancer Institute, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| |
Collapse
|
410
|
Xia B, He Q, Pan Y, Gao F, Liu A, Tang Y, Chong C, Teoh AYB, Li F, He Y, Zhang C, Yuan J. Metabolic syndrome and risk of pancreatic cancer: A population‐based prospective cohort study. Int J Cancer 2020; 147:3384-3393. [PMID: 32580250 DOI: 10.1002/ijc.33172] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Bin Xia
- Clinical Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
- Center for Digestive Disease, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Qiangsheng He
- Clinical Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
- Center for Digestive Disease, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yihang Pan
- Precision Medicine Center. Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Fang Gao
- Perioperative, Critical Care and Trauma Trials Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Anran Liu
- Center for Digestive Disease, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
- Department of Nutriology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yan Tang
- Clinical Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Charing Chong
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong SAR, China
| | - Anthony Y B Teoh
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong SAR, China
| | - Fangping Li
- Department of Endocrinology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yulong He
- Center for Digestive Disease, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Changhua Zhang
- Center for Digestive Disease, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Jinqiu Yuan
- Clinical Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
- Center for Digestive Disease, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
- Precision Medicine Center. Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| |
Collapse
|
411
|
Franck C, Müller C, Rosania R, Croner RS, Pech M, Venerito M. Advanced Pancreatic Ductal Adenocarcinoma: Moving Forward. Cancers (Basel) 2020; 12:E1955. [PMID: 32708493 PMCID: PMC7409054 DOI: 10.3390/cancers12071955] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 02/06/2023] Open
Abstract
Globally, the death rate of pancreatic ductal adenocarcinoma (PDAC) has doubled over 30 years and is likely to further increase, making PDAC a leading cause of cancer-related death in the coming years. PDAC is typically diagnosed at an advanced stage, and modified FOLFIRINOX or nab-paclitaxel and gemcitabine are the mainstay of systemic therapy. For elderly patients with good performance status, low-dose treatment can preserve quality of life without compromising cancer control or survival. Maintenance therapy should be considered in PDAC patients achieving disease control with systemic therapy. In particular, olaparib has demonstrated a progression-free survival benefit of 3.6 months in a subgroup of PDAC patients with germline BRCA1/2 mutations (ca. 10% of all PDAC). Pancreatic enzyme replacement therapy is often omitted in the treatment of patients with PDAC, with possibly deleterious consequences. Small intestinal bacterial overgrowth is highly prevalent in patients with PDAC and should be considered in the diagnostic algorithm of PDAC patients with bloating and diarrhea. Rivaroxaban has been associated with a reduced risk of thrombosis without an increase in major bleeding events, and its use should be considered in every patient with advanced PDAC undergoing systemic therapy.
Collapse
Affiliation(s)
- Caspar Franck
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, 39120 Magdeburg, Germany; (C.F.); (C.M.); (R.R.)
| | - Christian Müller
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, 39120 Magdeburg, Germany; (C.F.); (C.M.); (R.R.)
| | - Rosa Rosania
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, 39120 Magdeburg, Germany; (C.F.); (C.M.); (R.R.)
| | - Roland S. Croner
- Department of General-, Visceral-, Vascular- and Transplant Surgery, Otto-von-Guericke University Hospital Magdeburg, 39120 Magdeburg, Germany;
| | - Maciej Pech
- Department of Radiology and Nuclear Medicine, Otto-von-Guericke University Hospital, 39120 Magdeburg, Germany;
| | - Marino Venerito
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, 39120 Magdeburg, Germany; (C.F.); (C.M.); (R.R.)
| |
Collapse
|
412
|
Taghizadeh H, Müllauer L, Mader RM, Schindl M, Prager GW. Applied precision medicine in metastatic pancreatic ductal adenocarcinoma. Ther Adv Med Oncol 2020; 12:1758835920938611. [PMID: 32699558 PMCID: PMC7357054 DOI: 10.1177/1758835920938611] [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: 04/01/2020] [Accepted: 06/05/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Metastatic pancreatic ductal adenocarcinoma (mPDAC) bears a dismal prognosis due to the limited activity of systemic chemotherapy. In our platform for precision medicine, we aim to offer molecular-guided treatments to patients without further standard therapy options. METHODS In this single center, real-world retrospective analysis of our platform, we describe the molecular-based therapy approaches used in all 50 patients diagnosed with therapy-refractory mPDAC. A molecular portrait of the tumor specimens was created by next-generation sequencing, immunohistochemistry (IHC), microsatellite instability (MSI) testing, and fluorescence in situ hybridization. RESULTS In total, we detected 123 mutations in 50 patients. The five most frequent mutations were KRAS (n = 40; 80%), TP53 (n = 29; 58%), CDKN2A (n = 8; 16%), SMAD4 (n = 4; 8%), and NOTCH1 (n = 4; 8%), which together accounted for 40.2% of all mutations. Two patients had gene fusions, namely, TBL1XR1-PIK3CA and EIF3E-RSPO2. IHC detected expression of EGFR, phosphorylated mTOR, and PTEN in 36 (72%), 33 (66%), and 17 patients (34%), respectively. For 14 (28%) of the 50 patients, a targeted therapy was suggested based on the identified molecular targets. The recommended treatments included the mTOR inhibitor everolimus (n = 3), pembrolizumab (n = 3), palbociclib (n = 2), nintedanib (n = 2), and cetuximab, crizotinib, tamoxifen, and the combination of lapatinib and trastuzumab, in one patient each.Finally, five patients received the recommended therapy. Four patients died due to disease progression before radiological assessment. One patient was treated with nintedanib and achieved stable disease for 6 months. CONCLUSION Based on our observations, precision medicine approaches are feasible and implementable in clinical routine and may provide molecular-based therapy recommendations for mPDAC.
Collapse
Affiliation(s)
- Hossein Taghizadeh
- Department of Medicine I, Clinical Division of Oncology, Medical University of Vienna, Austria
- Comprehensive Cancer Center Vienna, Austria
| | - Leonhard Müllauer
- Comprehensive Cancer Center Vienna, Austria
- Clinical Institute of Pathology, Medical University Vienna, Vienna, Austria
| | - Robert M. Mader
- Department of Medicine I, Clinical Division of Oncology, Medical University of Vienna, Austria
- Comprehensive Cancer Center Vienna, Austria
| | - Martin Schindl
- Comprehensive Cancer Center Vienna, Austria
- Department of Surgery, Division of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Gerald W. Prager
- Department of Medicine I, Clinical Division of Oncology, Comprehensive Cancer Center Vienna, Precision Cancer Medicine Unit, Medical University of Vienna, Währinger Gürtel 18–20, 1090 Vienna, Austria
- Comprehensive Cancer Center Vienna, Austria
| |
Collapse
|
413
|
Papalampros A, Vailas M, Ntostoglou K, Chiloeches ML, Sakellariou S, Chouliari NV, Samaras MG, Veltsista PD, Theodorou SDP, Margetis AT, Bergonzini A, Karydakis L, Hasemaki N, Havaki S, Moustakas II, Chatzigeorgiou A, Karamitros T, Patsea E, Kittas C, Lazaris AC, Felekouras E, Gorgoulis VG, Frisan T, Pateras IS. Unique Spatial Immune Profiling in Pancreatic Ductal Adenocarcinoma with Enrichment of Exhausted and Senescent T Cells and Diffused CD47-SIRPα Expression. Cancers (Basel) 2020; 12:1825. [PMID: 32645996 PMCID: PMC7408661 DOI: 10.3390/cancers12071825] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is resistant to single-agent immunotherapies. To understand the mechanisms leading to the poor response to this treatment, a better understanding of the PDAC immune landscape is required. The present work aims to study the immune profile in PDAC in relationship to spatial heterogeneity of the tissue microenvironment (TME) in intact tissues. METHODS Serial section and multiplex in situ analysis were performed in 42 PDAC samples to assess gene and protein expression at single-cell resolution in the: (a) tumor center (TC), (b) invasive front (IF), (c) normal parenchyma adjacent to the tumor, and (d) tumor positive and negative draining lymph nodes (LNs). RESULTS We observed: (a) enrichment of T cell subpopulations with exhausted and senescent phenotype in the TC, IF and tumor positive LNs; (b) a dominant type 2 immune response in the TME, which is more pronounced in the TC; (c) an emerging role of CD47-SIRPα axis; and (d) a similar immune cell topography independently of the neoadjuvant chemotherapy. CONCLUSION This study reveals the existence of dysfunctional T lymphocytes with specific spatial distribution, thus opening a new dimension both conceptually and mechanistically in tumor-stroma interaction in PDAC with potential impact on the efficacy of immune-regulatory therapeutic modalities.
Collapse
Affiliation(s)
- Alexandros Papalampros
- First Department of Surgery, Laikon University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.P.); (M.V.); (L.K.); (N.H.); (E.F.)
| | - Michail Vailas
- First Department of Surgery, Laikon University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.P.); (M.V.); (L.K.); (N.H.); (E.F.)
| | - Konstantinos Ntostoglou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.N.); (N.V.C.); (M.G.S.); (P.D.V.); (S.D.P.T.); (A.T.M.); (S.H.); (C.K.); (V.G.G.)
| | - Maria Lopez Chiloeches
- Department of Molecular Biology, Umeå University, 90187 Umeå, Sweden; (M.L.C.); (A.B.); (T.F.)
| | - Stratigoula Sakellariou
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.S.); (A.C.L.)
| | - Niki V. Chouliari
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.N.); (N.V.C.); (M.G.S.); (P.D.V.); (S.D.P.T.); (A.T.M.); (S.H.); (C.K.); (V.G.G.)
| | - Menelaos G. Samaras
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.N.); (N.V.C.); (M.G.S.); (P.D.V.); (S.D.P.T.); (A.T.M.); (S.H.); (C.K.); (V.G.G.)
| | - Paraskevi D. Veltsista
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.N.); (N.V.C.); (M.G.S.); (P.D.V.); (S.D.P.T.); (A.T.M.); (S.H.); (C.K.); (V.G.G.)
- Oncology, School of Medical Sciences, Vrije Universiteit Amsterdam, De Boelelaan, 1117 Amsterdam, The Netherlands
| | - Sofia D. P. Theodorou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.N.); (N.V.C.); (M.G.S.); (P.D.V.); (S.D.P.T.); (A.T.M.); (S.H.); (C.K.); (V.G.G.)
| | - Aggelos T. Margetis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.N.); (N.V.C.); (M.G.S.); (P.D.V.); (S.D.P.T.); (A.T.M.); (S.H.); (C.K.); (V.G.G.)
| | - Anna Bergonzini
- Department of Molecular Biology, Umeå University, 90187 Umeå, Sweden; (M.L.C.); (A.B.); (T.F.)
| | - Lysandros Karydakis
- First Department of Surgery, Laikon University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.P.); (M.V.); (L.K.); (N.H.); (E.F.)
| | - Natasha Hasemaki
- First Department of Surgery, Laikon University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.P.); (M.V.); (L.K.); (N.H.); (E.F.)
| | - Sophia Havaki
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.N.); (N.V.C.); (M.G.S.); (P.D.V.); (S.D.P.T.); (A.T.M.); (S.H.); (C.K.); (V.G.G.)
| | - Ioannis I. Moustakas
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.I.M.); (A.C.)
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.I.M.); (A.C.)
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Timokratis Karamitros
- Bioinformatics and Applied Genomics Unit, Department of Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece;
| | - Eleni Patsea
- Department of Pathology, Metropolitan General Hospital of Athens, 15562 Cholargos, Greece;
| | - Christos Kittas
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.N.); (N.V.C.); (M.G.S.); (P.D.V.); (S.D.P.T.); (A.T.M.); (S.H.); (C.K.); (V.G.G.)
| | - Andreas C. Lazaris
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.S.); (A.C.L.)
| | - Evangelos Felekouras
- First Department of Surgery, Laikon University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.P.); (M.V.); (L.K.); (N.H.); (E.F.)
| | - Vassilis G. Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.N.); (N.V.C.); (M.G.S.); (P.D.V.); (S.D.P.T.); (A.T.M.); (S.H.); (C.K.); (V.G.G.)
- Faculty of Biology, Medicine and Health Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre, The University of Manchester, Manchester M20-4GJ, UK
- Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str, 11527 Athens, Greece
- Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Teresa Frisan
- Department of Molecular Biology, Umeå University, 90187 Umeå, Sweden; (M.L.C.); (A.B.); (T.F.)
- Department of Cell and Molecular Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Ioannis S. Pateras
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.N.); (N.V.C.); (M.G.S.); (P.D.V.); (S.D.P.T.); (A.T.M.); (S.H.); (C.K.); (V.G.G.)
| |
Collapse
|
414
|
Arnold M, Abnet CC, Neale RE, Vignat J, Giovannucci EL, McGlynn KA, Bray F. Global Burden of 5 Major Types of Gastrointestinal Cancer. Gastroenterology 2020; 159:335-349.e15. [PMID: 32247694 PMCID: PMC8630546 DOI: 10.1053/j.gastro.2020.02.068] [Citation(s) in RCA: 1191] [Impact Index Per Article: 238.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/10/2020] [Accepted: 02/28/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS There were an estimated 4.8 million new cases of gastrointestinal (GI) cancers and 3.4 million related deaths, worldwide, in 2018. GI cancers account for 26% of the global cancer incidence and 35% of all cancer-related deaths. We investigated the global burden from the 5 major GI cancers, as well as geographic and temporal trends in cancer-specific incidence and mortality. METHODS Data on primary cancers of the esophagus, stomach, colorectum, liver, and pancreas were extracted from the GLOBOCAN database for the year 2018, as well as from the Cancer Incidence in 5 Continents series, and the World Health Organization mortality database from 1960 onward. Age-standardized incidence and mortality rates were calculated by sex, country, and level of human development. RESULTS We observed geographic and temporal variations in incidence and mortality for all 5 types of GI cancers. Esophageal, gastric, and liver cancers were more common in Asia than in other parts of the world, and the burden from colorectal and pancreatic cancers was highest in Europe and North America. There was a uniform decrease in gastric cancer incidence, but an increasing incidence of colorectal cancer in formerly low-incidence regions during the studied time period. We found slight increases in incidence of liver and pancreatic cancer in some high-income regions. CONCLUSIONS Although the incidence of some GI cancer types has decreased, this group of malignancies continues to pose major challenges to public health. Primary and secondary prevention measures are important for controlling these malignancies-most importantly reducing consumption of tobacco and alcohol, obesity control, immunizing populations against hepatitis B virus infection, and screening for colorectal cancer.
Collapse
Affiliation(s)
- Melina Arnold
- Section of Cancer Surveillance, International Agency for Research on Cancer, Lyon, France.
| | - Christian C Abnet
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Rachel E Neale
- Population Health Division, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Jerome Vignat
- Section of Cancer Surveillance, International Agency for Research on Cancer, Lyon, France
| | - Edward L Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Katherine A McGlynn
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Freddie Bray
- Section of Cancer Surveillance, International Agency for Research on Cancer, Lyon, France
| |
Collapse
|
415
|
Hasan SMS, Rivera D, Wu XC, Durbin EB, Christian JB, Tourassi G. Knowledge Graph-Enabled Cancer Data Analytics. IEEE J Biomed Health Inform 2020; 24:1952-1967. [PMID: 32386166 PMCID: PMC8324069 DOI: 10.1109/jbhi.2020.2990797] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cancer registries collect unstructured and structured cancer data for surveillance purposes which provide important insights regarding cancer characteristics, treatments, and outcomes. Cancer registry data typically (1) categorize each reportable cancer case or tumor at the time of diagnosis, (2) contain demographic information about the patient such as age, gender, and location at time of diagnosis, (3) include planned and completed primary treatment information, and (4) may contain survival outcomes. As structured data is being extracted from various unstructured sources, such as pathology reports, radiology reports, medical records, and stored for reporting and other needs, the associated information representing a reportable cancer is constantly expanding and evolving. While some popular analytic approaches including SEER*Stat and SAS exist, we provide a knowledge graph approach to organizing cancer registry data. Our approach offers unique advantages for timely data analysis and presentation and visualization of valuable information. This knowledge graph approach semantically enriches the data, and easily enables linking with third-party data which can help explain variation in cancer incidence patterns, disparities, and outcomes. We developed a prototype knowledge graph based on the Louisiana Tumor Registry dataset. We present the advantages of the knowledge graph approach by examining: i) scenario-specific queries, ii) links with openly available external datasets, iii) schema evolution for iterative analysis, and iv) data visualization. Our results demonstrate that this graph based solution can perform complex queries, improve query run-time performance by up to 76%, and more easily conduct iterative analyses to enhance researchers' understanding of cancer registry data.
Collapse
|
416
|
Mizrahi JD, Surana R, Valle JW, Shroff RT. Pancreatic cancer. Lancet 2020; 395:2008-2020. [PMID: 32593337 DOI: 10.1016/s0140-6736(20)30974-0] [Citation(s) in RCA: 1626] [Impact Index Per Article: 325.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/06/2020] [Accepted: 04/17/2020] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer is a highly fatal disease with a 5-year survival rate of approximately 10% in the USA, and it is becoming an increasingly common cause of cancer mortality. Risk factors for developing pancreatic cancer include family history, obesity, type 2 diabetes, and tobacco use. Patients typically present with advanced disease due to lack of or vague symptoms when the cancer is still localised. High quality computed tomography with intravenous contrast using a dual phase pancreatic protocol is typically the best method to detect a pancreatic tumour and to determine surgical resectability. Endoscopic ultrasound is an increasingly used complementary staging modality which also allows for diagnostic confirmation when combined with fine needle aspiration. Patients with pancreatic cancer are often divided into one of four categories based on extent of disease: resectable, borderline resectable, locally advanced, and metastatic; patient condition is also an important consideration. Surgical resection represents the only chance for cure, and advancements in adjuvant chemotherapy have improved long-term outcomes in these patients. Systemic chemotherapy combinations including FOLFIRINOX (5-fluorouracil, folinic acid [leucovorin], irinotecan, and oxaliplatin) and gemcitabine plus nab-paclitaxel remain the mainstay of treatment for patients with advanced disease. Data on the benefit of PARP inhibition as maintenance therapy in patients with germline BRCA1 or BRACA2 mutations might prove to be a harbinger of advancement in targeted therapy. Additional research efforts are focusing on modulating the pancreatic tumour microenvironment to enhance the efficacy of the immunotherapeutic strategies.
Collapse
Affiliation(s)
- Jonathan D Mizrahi
- Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rishi Surana
- Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Juan W Valle
- Division of Cancer Sciences, University of Manchester, Department of Medical Oncology, Christie NHS Foundation Trust, Manchester, UK
| | - Rachna T Shroff
- Division of Hematology and Oncology, Department of Medicine, University of Arizona Cancer Center, Tucson, AZ, USA.
| |
Collapse
|
417
|
Plasma IL8 Is a Biomarker for TAK1 Activation and Predicts Resistance to Nanoliposomal Irinotecan in Patients with Gemcitabine-Refractory Pancreatic Cancer. Clin Cancer Res 2020; 26:4661-4669. [DOI: 10.1158/1078-0432.ccr-20-0395] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/25/2020] [Accepted: 06/09/2020] [Indexed: 11/16/2022]
|
418
|
Liver metastases from pancreatic ductal adenocarcinoma: is there a place for surgery in the modern era? JOURNAL OF PANCREATOLOGY 2020. [DOI: 10.1097/jp9.0000000000000042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
419
|
Ren S, Zhao R, Zhang J, Guo K, Gu X, Duan S, Wang Z, Chen R. Diagnostic accuracy of unenhanced CT texture analysis to differentiate mass-forming pancreatitis from pancreatic ductal adenocarcinoma. Abdom Radiol (NY) 2020; 45:1524-1533. [PMID: 32279101 DOI: 10.1007/s00261-020-02506-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE To investigate the value of texture analysis on unenhanced computed tomography (CT) to potentially differentiate mass-forming pancreatitis (MFP) from pancreatic ductal adenocarcinoma (PDAC). METHODS A retrospective study consisting of 109 patients (30 MFP patients vs 79 PDAC patients) who underwent preoperative unenhanced CT between January 2012 and December 2017 was performed. Synthetic minority oversampling technique (SMOTE) algorithm was adopted to reconstruct and balance MFP and PDAC samples. A total of 396 radiomic features were extracted from unenhanced CT images. Mann-Whitney U test and minimum redundancy maximum relevance (MRMR) methods were used for the purpose of dimension reduction. Predictive models were constructed using random forest (RF) method, and were validated using leave group out cross-validation (LGOCV) method. Diagnostic performance of the predictive model, including sensitivity, specificity, accuracy, positive predicting value (PPV), and negative predicting value (NPV), was recorded. RESULTS We applied 200% of SMOTE to MFP and PDAC patients, resulting in 90 MFP patients compared with 120 PDAC patients. Dimension reduction steps yielded 30 radiomic features using Mann-Whitney U test and MRMR methods. Ten radiomic features were retained using RF method. Four most predictive parameters, including GreyLevelNonuniformity_angle90_offset1, VoxelValueSum, HaraVariance, and ClusterProminence_AllDirection_offset1_SD, were used to generate the predictive model with preferable 92.2% sensitivity, 94.2% specificity, 93.3% accuracy, 92.2% PPV, and 94.2% NPV. Finally, in LGOCV analysis, a high pooled mean sensitivity, specificity, and accuracy (82.6%, 80.8%, and 82.1%, respectively) indicate a relatively reliable and stable predictive model. CONCLUSIONS Unenhanced CT texture analysis can be a promising noninvasive method in discriminating MFP from PDAC.
Collapse
Affiliation(s)
- Shuai Ren
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing, 210029, Jiangsu Province, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Rui Zhao
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing, 210029, Jiangsu Province, China
| | - Jingjing Zhang
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing, 210029, Jiangsu Province, China
| | - Kai Guo
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing, 210029, Jiangsu Province, China
| | - Xiaoyu Gu
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing, 210029, Jiangsu Province, China
| | | | - Zhongqiu Wang
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing, 210029, Jiangsu Province, China.
| | - Rong Chen
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| |
Collapse
|
420
|
Merz V, Cavaliere A, Messina C, Salati M, Zecchetto C, Casalino S, Milella M, Caffo O, Melisi D. Multicenter Retrospective Analysis of Second-Line Therapy after Gemcitabine Plus Nab-Paclitaxel in Advanced Pancreatic Cancer Patients. Cancers (Basel) 2020; 12:E1131. [PMID: 32366019 PMCID: PMC7281137 DOI: 10.3390/cancers12051131] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/17/2020] [Accepted: 04/26/2020] [Indexed: 12/17/2022] Open
Abstract
Pancreatic cancer is one of the most lethal solid tumors. In many European countries gemcitabine plus nab-paclitaxel is the preferred first-line treatment. An increasing number of patients are eligible for second-line therapy, but the best regimen is still controversial. This study aimed to evaluate the efficacy of oxaliplatin-based compared to irinotecan-based therapies in this setting. 181 advanced pancreatic cancer patients consecutively treated in three centers with a second-line therapy progressed on gemcitabine plus nab-paclitaxel were retrospectively enrolled. OS and PFS were calculated using the Kaplan-Meier method and survival of the two groups was compared using the log-rank test. The median PFS and OS were respectively 3.5 (95%CI 3.2-3.8) and 8.8 months (95%CI 7.9-9.8) from second-line therapy in the overall population. The median PFS and OS were respectively 3.3 (95%CI 3.1-3.5) and 8.2 months (95%CI 7.24-9.34) with an irinotecan-based combination compared to 4.0 (95%CI 2.4-5.7) and 10.3 months (95%CI 8.62-12.02) in patients receiving an oxaliplatin-based combination. We observed a clear trend for longer survival outcomes with platinum-based doublet compared to regimens including irinotecan or nal-IRI. Head-to-head trials are still lacking. The neutrophil-to-lymphocyte ratio and the presence of liver metastases could drive physicians in tailoring the treatment strategy.
Collapse
Affiliation(s)
- Valeria Merz
- Digestive Molecular Clinical Oncology Research Unit, University of Verona, 37134 Verona, Italy; (A.C.); (C.Z.); (S.C.)
- Department of Medical Oncology, Santa Chiara Hospital, 38122 Trento, Italy; (C.M.); (O.C.)
| | - Alessandro Cavaliere
- Digestive Molecular Clinical Oncology Research Unit, University of Verona, 37134 Verona, Italy; (A.C.); (C.Z.); (S.C.)
- Section of Medical Oncology, University of Verona, 37134 Verona, Italy;
| | - Carlo Messina
- Department of Medical Oncology, Santa Chiara Hospital, 38122 Trento, Italy; (C.M.); (O.C.)
| | - Massimiliano Salati
- Department of Medical Oncology, University Hospital of Modena, 4121 Modena, Italy;
- PhD Program Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, 4121 Modena, Italy
| | - Camilla Zecchetto
- Digestive Molecular Clinical Oncology Research Unit, University of Verona, 37134 Verona, Italy; (A.C.); (C.Z.); (S.C.)
| | - Simona Casalino
- Digestive Molecular Clinical Oncology Research Unit, University of Verona, 37134 Verona, Italy; (A.C.); (C.Z.); (S.C.)
- Section of Medical Oncology, University of Verona, 37134 Verona, Italy;
| | - Michele Milella
- Section of Medical Oncology, University of Verona, 37134 Verona, Italy;
| | - Orazio Caffo
- Department of Medical Oncology, Santa Chiara Hospital, 38122 Trento, Italy; (C.M.); (O.C.)
| | - Davide Melisi
- Digestive Molecular Clinical Oncology Research Unit, University of Verona, 37134 Verona, Italy; (A.C.); (C.Z.); (S.C.)
- Section of Medical Oncology, University of Verona, 37134 Verona, Italy;
| |
Collapse
|
421
|
Sommariva M, Gagliano N. E-Cadherin in Pancreatic Ductal Adenocarcinoma: A Multifaceted Actor during EMT. Cells 2020; 9:E1040. [PMID: 32331358 PMCID: PMC7226001 DOI: 10.3390/cells9041040] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 12/14/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a step-wise process observed in normal and tumor cells leading to a switch from epithelial to mesenchymal phenotype. In tumors, EMT provides cancer cells with a metastatic phenotype characterized by E-cadherin down-regulation, cytoskeleton reorganization, motile and invasive potential. E-cadherin down-regulation is known as a key event during EMT. However, E-cadherin expression can be influenced by the different experimental settings and environmental stimuli so that the paradigm of EMT based on the loss of E-cadherin determining tumor cell behavior and fate often becomes an open question. In this review, we aimed at focusing on some critical points in order to improve the knowledge of the dynamic role of epithelial cells plasticity in EMT and, specifically, address the role of E-cadherin as a marker for the EMT axis.
Collapse
Affiliation(s)
| | - Nicoletta Gagliano
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy;
| |
Collapse
|
422
|
Martín-Blázquez A, Jiménez-Luna C, Díaz C, Martínez-Galán J, Prados J, Vicente F, Melguizo C, Genilloud O, Pérez del Palacio J, Caba O. Discovery of Pancreatic Adenocarcinoma Biomarkers by Untargeted Metabolomics. Cancers (Basel) 2020; 12:1002. [PMID: 32325731 PMCID: PMC7225994 DOI: 10.3390/cancers12041002] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/08/2020] [Accepted: 04/13/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and lethal cancers, with a 5-year survival rate of less than 5%. In fact, complete surgical resection remains the only curative treatment. However, fewer than 20% of patients are candidates for surgery at the time of presentation. Hence, there is a critical need to identify diagnostic biomarkers with potential clinical utility in this pathology. In this context, metabolomics could be a powerful tool to search for new robust biomarkers. Comparative metabolomic profiling was performed in serum samples from 59 unresectable PDAC patients and 60 healthy controls. Samples were analyzed by using an untargeted metabolomics workflow based on liquid chromatography, coupled to high-resolution mass spectrometry in positive and negative electrospray ionization modes. Univariate and multivariate analysis allowed the identification of potential candidates that were significantly altered in PDAC patients. A panel of nine candidates yielded excellent diagnostic capacities. Pathway analysis revealed four altered pathways in our patients. This study shows the potential of liquid chromatography coupled to high-resolution mass spectrometry as a diagnostic tool for PDAC. Furthermore, it identified novel robust biomarkers with excellent diagnostic capacities.
Collapse
Affiliation(s)
- Ariadna Martín-Blázquez
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, 18016 Granada, Spain; (A.M.-B.); (C.D.); (F.V.); (O.G.); (J.P.d.P.)
| | - Cristina Jiménez-Luna
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, 1066 Epalinges, Switzerland;
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18016 Granada, Spain; (C.M.); (O.C.)
| | - Caridad Díaz
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, 18016 Granada, Spain; (A.M.-B.); (C.D.); (F.V.); (O.G.); (J.P.d.P.)
| | - Joaquina Martínez-Galán
- Service of Medical Oncology, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain;
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18016 Granada, Spain; (C.M.); (O.C.)
- Instituto Biosanitario de Granada (ibs. GRANADA), 18016 Granada, Spain
| | - Francisca Vicente
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, 18016 Granada, Spain; (A.M.-B.); (C.D.); (F.V.); (O.G.); (J.P.d.P.)
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18016 Granada, Spain; (C.M.); (O.C.)
- Instituto Biosanitario de Granada (ibs. GRANADA), 18016 Granada, Spain
| | - Olga Genilloud
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, 18016 Granada, Spain; (A.M.-B.); (C.D.); (F.V.); (O.G.); (J.P.d.P.)
| | - José Pérez del Palacio
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, 18016 Granada, Spain; (A.M.-B.); (C.D.); (F.V.); (O.G.); (J.P.d.P.)
| | - Octavio Caba
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18016 Granada, Spain; (C.M.); (O.C.)
- Instituto Biosanitario de Granada (ibs. GRANADA), 18016 Granada, Spain
| |
Collapse
|
423
|
Jiang J, Bai J, Qin T, Wang Z, Han L. NGF from pancreatic stellate cells induces pancreatic cancer proliferation and invasion by PI3K/AKT/GSK signal pathway. J Cell Mol Med 2020; 24:5901-5910. [PMID: 32294802 PMCID: PMC7214160 DOI: 10.1111/jcmm.15265] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/12/2020] [Accepted: 03/18/2020] [Indexed: 12/22/2022] Open
Abstract
Pancreatic cancer (PC) is a continuously high lethal disease, and the tumour microenvironment plays a pivotal role during PC progression. Herein, we focus on that the Nerve growth factor (NGF)/Tropomyosin-related kinase A (TrkA), in pancreatic stellate cells-pancreatic cancer cells (PSCs-PC cells) co-culture system, influences PC proliferation and invasion. The model of PC cells and PSCs was directly co-cultured in a no-touch manner, using the Transwell as the co-culture system. NGF and TrkA expression was measured in cultured system by real-time PCR, immunofluorescence, Western blotting analysis or ELISA. Small interfering RNA transfection was used to regulate the expression of TrkA in PC cells. The promotion of cancer invasion was investigated using Matrigel Transwell assay. In our study, NGF/TrkA is overexpressed in PSCs-PC cells co-culture system and promotes the invasion and proliferation of PC cells. And the epithelial-mesenchymal transition-related genes are influenced by si-TrkA. What's more, NGF/TrkA regulates the PC cell proliferation and invasion via activation of PI3K/AKT/GSK signalling. The present study demonstrated NGF/TrkA promoted the PC cell proliferation and invasion in the co-culture system by the activation of the PI3K/AKT/GSK signal cascade, providing a potential therapeutic target for PC patients.
Collapse
Affiliation(s)
- Jie Jiang
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Department of Medical Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Jun Bai
- Department of Medical Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Tao Qin
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Liang Han
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
424
|
Farge D, Bournet B, Conroy T, Vicaut E, Rak J, Zogoulous G, Barkun J, Ouaissi M, Buscail L, Frere C. Primary Thromboprophylaxis in Pancreatic Cancer Patients: Why Clinical Practice Guidelines Should Be Implemented. Cancers (Basel) 2020; 12:E618. [PMID: 32155940 PMCID: PMC7139861 DOI: 10.3390/cancers12030618] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 02/27/2020] [Accepted: 03/04/2020] [Indexed: 12/13/2022] Open
Abstract
Exocrine pancreatic ductal adenocarcinoma, simply referred to as pancreatic cancer (PC) has the worst prognosis of any malignancy. Despite recent advances in the use of adjuvant chemotherapy in PC, the prognosis remains poor, with fewer than 8% of patients being alive at 5 years after diagnosis. The prevalence of PC has steadily increased over the past decades, and it is projected to become the second-leading cause of cancer-related death by 2030. In this context, optimizing and integrating supportive care is important to improve quality of life and survival. Venous thromboembolism (VTE) is a common but preventable complication in PC patients. VTE occurs in one out of five PC patients and is associated with significantly reduced progression-free survival and overall survival. The appropriate use of primary thromboprophylaxis can drastically and safely reduce the rates of VTE in PC patients as shown from subgroup analysis of non-PC targeted placebo-controlled randomized trials of cancer patients and from two dedicated controlled randomized trials in locally advanced PC patients receiving chemotherapy. Therefore, primary thromboprophylaxis with a Grade 1B evidence level is recommended in locally advanced PC patients receiving chemotherapy by the International Initiative on Cancer and Thrombosis clinical practice guidelines since 2013. However, its use and potential significant clinical benefit continues to be underrecognized worldwide. This narrative review aims to summarize the main recent advances in the field including on the use of individualized risk assessment models to stratify the risk of VTE in each patient with individual available treatment options.
Collapse
Affiliation(s)
- Dominique Farge
- Institut Universitaire d’Hématologie, Université de Paris, EA 3518, F-75010 Paris, France
- Assistance Publique Hôpitaux de Paris, Saint-Louis Hospital, Internal Medicine, Autoimmune and Vascular Disease Unit, F-75010 Paris, France
- Department of Medicine, McGill University, Montreal, Québec, QC H4A 3J1, Canada
| | - Barbara Bournet
- University of Toulouse, F-31059 Toulouse, France; (B.B.); (L.B.)
- CHU de Toulouse, Department of Gastroenterology and Pancreatology, F-31059 Toulouse, France
| | - Thierry Conroy
- Institut de Cancérologie de Lorraine, Department of Medical Oncology, Université de Lorraine, APEMAC, EA4360, F-54519 Vandoeuvre-lès-Nancy, France;
| | - Eric Vicaut
- Department of Biostatistics, Université de Paris, F-75010 Paris, France;
- Assistance Publique Hôpitaux de Paris, Department of Biostatistics, Fernand Widal Hospital, F-75010 Paris, France
| | - Janusz Rak
- McGill University and the Research Institute of the McGill University Health Centre, Montreal, Québec, QC H4A 3J1, Canada; (J.R.)
| | - George Zogoulous
- McGill University and the Research Institute of the McGill University Health Centre, Montreal, Québec, QC H4A 3J1, Canada; (J.R.)
| | - Jefferey Barkun
- McGill University and the Research Institute of the McGill University Health Centre, Montreal, Québec, QC H4A 3J1, Canada; (J.R.)
| | - Mehdi Ouaissi
- Department of Digestive, Oncological, Endocrine, and Hepatic Surgery, and Hepatic Transplantation, Trousseau Hospital, CHRU Trousseau, F-37170 Chambray-les-Tours, France;
| | - Louis Buscail
- University of Toulouse, F-31059 Toulouse, France; (B.B.); (L.B.)
- CHU de Toulouse, Department of Gastroenterology and Pancreatology, F-31059 Toulouse, France
| | - Corinne Frere
- Institute of Cardiometabolism and Nutrition, Sorbonne Université, INSERM UMRS_1166, GRC 27 GRECO, F-75013 Paris, France;
- Assistance Publique Hôpitaux de Paris, Department of Haematology, Pitié-Salpêtrière Hospital, F-75013 Paris, France
| |
Collapse
|
425
|
Landman A, Feetham L, Stuckey D. Working together to reduce the burden of pancreatic cancer. Lancet Oncol 2020; 21:334-335. [DOI: 10.1016/s1470-2045(20)30088-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
426
|
Gad MM, Găman MA, Saad AM, Al-Husseini MJ, Shehata OA, Saleh MA, Nelson AD, Simons-Linares CR. Temporal trends of incidence and mortality in Asian-Americans with pancreatic adenocarcinoma: an epidemiological study. Ann Gastroenterol 2020; 33:210-218. [PMID: 32127743 PMCID: PMC7049244 DOI: 10.20524/aog.2020.0450] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 12/02/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Pancreatic cancer is the fourth most common cause of cancer-related deaths in the United States, with an estimated 45,750 deaths in 2019. Mortality outcomes seem to differ based on the ethnicity of the patients, with most studies focusing on the mortality and survival of Caucasians and African Americans. Little attention has been given, however, to Asian-American patients diagnosed with pancreatic adenocarcinoma (PAC). In this study, we aimed to investigate mortality rates in Asian-American patients with PAC. METHODS The SEER 13 registries (Surveillance, Epidemiology, and End-Results) of the National Cancer Institute were used to study PAC cases during 1992-2015. The incidence and incidence-based mortality rates per 100,000 person-years, and the annual percentage changes were calculated using SEER*stat software and Joinpoint regression software. RESULTS A total of 5814 PAC cases in Asian-American patients were identified. Most patients were older than 60 years (77.6%) and had metastatic disease (55.8%). The overall incidence of PAC among Asian-Americans was 5.740 per 100,000 person-years (95% confidence interval [CI] 5.592-5.891]. Incidence rates were highest among males and patients older than 60 years. PAC incidence rates among Asian-Americans increased by 1.503% (95%CI 1.051-1.956; P<0.001) per year over the study period. PAC incidence rates increased over the study period for all sex, age, and stage subgroups. PAC incidence-based mortality among Asian-Americans increased by 4.535% (95%CI 3.538-5.541; P<0.001) per year over the study period. CONCLUSION The incidence of PAC in Asian-Americans, as well as incidence-based mortality rates, are on the rise, irrespective of age, sex or stage subgroup.
Collapse
Affiliation(s)
- Mohamed M. Gad
- Department of Internal Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA (Mohamed M. Gad, Anas M. Saad, Mohannad Abou Saleh, Alfred D. Nelson, Carlos Roberto Simons-Linares)
- Department of Global Public Health, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA (Mohamed M. Gad)
| | - Mihnea-Alexandru Găman
- Department of Hematology and Oncology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania and Center of Hematology and d) Bone Marrow Transplantation, Fundeni Clinical Institute, Bucharest, Romania (Mihnea-Alexandru Găman)
| | - Anas M. Saad
- Department of Internal Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA (Mohamed M. Gad, Anas M. Saad, Mohannad Abou Saleh, Alfred D. Nelson, Carlos Roberto Simons-Linares)
- Faculty of Medicine, Ain Shams University, Cairo, Egypt (Anas M. Saad, Muneer J. Al-Husseini, Omar A. Shehata)
| | - Muneer J. Al-Husseini
- Faculty of Medicine, Ain Shams University, Cairo, Egypt (Anas M. Saad, Muneer J. Al-Husseini, Omar A. Shehata)
- Department of Medicine, Ascension St John Hospital, Detroit, MI, USA (Muneer J. Al-Husseini)
| | - Omar A. Shehata
- Faculty of Medicine, Ain Shams University, Cairo, Egypt (Anas M. Saad, Muneer J. Al-Husseini, Omar A. Shehata)
| | - Mohannad Abou Saleh
- Department of Internal Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA (Mohamed M. Gad, Anas M. Saad, Mohannad Abou Saleh, Alfred D. Nelson, Carlos Roberto Simons-Linares)
| | - Alfred D. Nelson
- Department of Internal Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA (Mohamed M. Gad, Anas M. Saad, Mohannad Abou Saleh, Alfred D. Nelson, Carlos Roberto Simons-Linares)
| | - Carlos Roberto Simons-Linares
- Department of Internal Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA (Mohamed M. Gad, Anas M. Saad, Mohannad Abou Saleh, Alfred D. Nelson, Carlos Roberto Simons-Linares)
| |
Collapse
|
427
|
Yue Y, Qian W, Li J, Wu S, Zhang M, Wu Z, Ma Q, Wang Z. 2'-Hydroxyflavanone inhibits the progression of pancreatic cancer cells and sensitizes the chemosensitivity of EGFR inhibitors via repressing STAT3 signaling. Cancer Lett 2020; 471:135-146. [PMID: 31811906 DOI: 10.1016/j.canlet.2019.11.041] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/09/2019] [Accepted: 11/30/2019] [Indexed: 12/17/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies, and chemotherapy is still an important treatment. It is urgent to develop new medicines because of the limitation and side effects of chemotherapy. 2'-Hydroxyflavanone (2HF) is a citrus-bioflavonoid that is considered to have anti-cancer efficacy. Compared to human pancreatic ductal epithelial cells hTERT-HPNE, more significant growth-inhibitory effects were seen in PDAC cells BxPC-3 and MIA PaCa-2. We showed that apoptosis was induced and that the cell cycle was arrested when cells were treated with 2HF. The expression of the molecular proteins cleaved PARP, cleaved Caspase3, Bax, Bcl-2, CyclinD1, and p27 changed correspondingly. Also, we observed anti-metastatic effects and changes in MMP9, E-cadherin, N-cadherin and Vimentin when cells were treated with a low dose of 2HF. Suppression of STAT3 and EGFR phosphorylation was also identified as a result of treatment with a combination of 2HF and EGFR inhibitors. The in vivo antitumor effects in KPC mice were consistent with those observed in vitro. 2HF has impactful anti-cancer efficacy and sensitizes human pancreatic cancer cells to EGFR inhibitors through the inhibition of STAT3.
Collapse
Affiliation(s)
- Yangyang Yue
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi province, China
| | - Weikun Qian
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi province, China
| | - Jie Li
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi province, China
| | - Shiqi Wu
- Department of Urology Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi province, China
| | - Mengzhao Zhang
- Department of Urology Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi province, China
| | - Zheng Wu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi province, China
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi province, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi province, China.
| |
Collapse
|
428
|
Zeng F, Chen L, Liao M, Chen B, Long J, Wu W, Deng G. Laparoscopic versus open gastrectomy for gastric cancer. World J Surg Oncol 2020; 18:20. [PMID: 31987046 PMCID: PMC6986035 DOI: 10.1186/s12957-020-1795-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/16/2020] [Indexed: 02/06/2023] Open
Abstract
Background Compared with open gastrectomy (OG), laparoscopic gastrectomy (LG) for gastric cancer has achieved rapid development and popularities in the past decades. However, lack of comprehensive analysis in long-term oncological outcomes such as recurrence and mortality hinder its full support as a valid procedure. Therefore, there are still debates on whether one of these options is superior. Aim To evaluate the primary and secondary outcomes of laparoscopic versus open gastrectomy for gastric cancer patients Methods Two authors independently extracted study data. Risk ratio (RR) with 95% confidence interval (CI) was calculated for binary outcomes, mean difference (MD) or the standardized mean difference (SMD) with 95% CI for continuous outcomes, and the hazard ratio (HR) for time-to-event outcomes. Review Manager 5.3 and STATA software were used for the meta-analysis. Results Seventeen randomized controlled trials (RCTs) involving 5204 participants were included in this meta-analysis. There were no differences in the primary outcomes including the number of lymph nodes harvested during operation, severe complications, short-term and long-term recurrence, and mortality. As for secondary outcomes, compared with the OG group, longer operative time was required for patients in the LG group (MD = 58.80 min, 95% CI = [45.80, 71.81], P < 0.001), but there were less intraoperative blood loss (MD = − 54.93 ml, 95% CI = [− 81.60, − 28.26], P < 0.001), less analgesic administration (frequency: MD = − 1.73, 95% CI = [− 2.21, − 1.24], P < 0.001; duration: MD = − 1.26 days, 95% CI = [− 1.40, − 1.12], P < 0.001), shorter hospital stay (MD = − 1.37 days, 95% CI = [− 2.05, − 0.70], P < 0.001), shorter time to first flatus (MD = − 0.58 days, 95% CI = [− 0.79, − 0.37], P < 0.001), ambulation (MD = − 0.50 days, 95% CI = [− 0.90, − 0.09], P = 0.02) and oral intake (MD = − 0.64 days, 95% CI = [− 1.24, − 0.03], P < 0.04), and less total complications (RR = 0.81, 95% CI = [0.71, 0.93], P = 0.003) in the OG group. There was no difference in blood transfusions (number, quantity) between these two groups. Subgroup analysis, sensitivity analysis, and the adjustment of Duval’s trim and fill methods for publication bias did not change the conclusions. Conclusion LG was comparable to OG in the primary outcomes and had some advantages in secondary outcomes for gastric cancer patients. LG is superior to OG for gastric cancer patients.
Collapse
Affiliation(s)
- Furong Zeng
- Xiangya hospital, Central South University, Changsha, China
| | - Lang Chen
- Xiangya hospital, Central South University, Changsha, China
| | - Mengting Liao
- Xiangya hospital, Central South University, Changsha, China
| | - Bin Chen
- Taoyuan People's Hospital, Taoyuan, Changde, China
| | - Jing Long
- Xiangya hospital, Central South University, Changsha, China
| | - Wei Wu
- Xiangya hospital, Central South University, Changsha, China.
| | - Guangtong Deng
- Xiangya hospital, Central South University, Changsha, China.
| |
Collapse
|
429
|
Affiliation(s)
- Alison P Klein
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, and Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|