1
|
Darmadi D, Saleh RO, Oghenemaro EF, Shakir MN, Hjazi A, Hassan ZF, Zwamel AH, Matlyuba S, Deorari M, Oudah SK. Role of SEL1L in the progression of solid tumors, with a special focus on its recent therapeutic potential. Cell Biol Int 2024. [PMID: 39364680 DOI: 10.1002/cbin.12242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/24/2024] [Accepted: 09/02/2024] [Indexed: 10/05/2024]
Abstract
Since suppressor/enhancer of Lin-12-like (SEL1L) was cloned in 1997, various pieces of evidence from lower species suggest it plays a significant role in protein degradation via the ubiquitin-proteasome system. The relevance of SEL1L in many aspects of malignant transformation and tumorigenic events has been the subject of research, which has shown compelling in vitro and in vivo findings relating its altered expression to changes in tumor aggressiveness. The Endoplasmic Reticulum (ER) in tumor cells is crucial for preserving cellular proteostasis by inducing the unfolded protein response (UPR), a stress response. A crucial component of the UPR is ER-associated degradation (ERAD), which guards against ER stress-induced apoptosis and the removal of unfolded or misfolded proteins by the ubiquitin-proteasome system. As a protein stabilizer of HMG-CoA reductase degradation protein 1 (HRD1), one of the main components of ERAD, SEL1L plays an important role in ER homeostasis. Notably, the expression levels of these two proteins fluctuate independently in various cancer types, yet changes in their expression affect the levels of other associated proteins during cancer pathogenesis. Recent studies have also outlined the function of SEL1L in cancer medication resistance. This review explores the value of targeting SEL1L as a novel treatment approach for cancer, focusing on the molecular processes of SEL1L and its involvement in cancer etiology.
Collapse
Affiliation(s)
- Darmadi Darmadi
- Department of Internal Medicine, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Raed Obaid Saleh
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Iraq
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, Abraka, Nigeria
| | - Maha Noori Shakir
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | | | - Ahmed Hussein Zwamel
- Medical laboratory technique college, the Islamic University, Najaf, Iraq
- Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical laboratory technique college, the Islamic University of Babylon, Babylon, Iraq
| | - Sanoeva Matlyuba
- Department of Neurology, Vice rektor of Bukhara State Medical Institute, Bukhara, Uzbekistan
| | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Shamam Kareem Oudah
- College of Pharmacy/National University of Science and Technology, Dhi Qar, Iraq
| |
Collapse
|
2
|
Maoz A, Rodriguez NJ, Yurgelun MB, Syngal S. Gastrointestinal Cancer Precursor Conditions and Their Detection. Hematol Oncol Clin North Am 2024; 38:783-811. [PMID: 38760197 PMCID: PMC11537157 DOI: 10.1016/j.hoc.2024.04.002] [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] [Indexed: 05/19/2024]
Abstract
Gastrointestinal cancers are a leading cause of cancer morbidity and mortality. Many gastrointestinal cancers develop from cancer precursor lesions, which are commonly found in individuals with hereditary cancer syndromes. Hereditary cancer syndromes have advanced our understanding of cancer development and progression and have facilitated the evaluation of cancer prevention and interception efforts. Common gastrointestinal hereditary cancer syndromes, including their organ-specific cancer risk and surveillance recommendations, are reviewed in this article. The management of common gastroesophageal, pancreatic, and colonic precursor lesions is also discussed, regardless of their genetic background. Further research is needed to advance chemoprevention and immunoprevention strategies.
Collapse
Affiliation(s)
- Asaf Maoz
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA; Harvard Medical School, Boston, MA, USA. https://twitter.com/asaf_maoz
| | - Nicolette J Rodriguez
- Harvard Medical School, Boston, MA, USA; Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, 75 Francis Street, Boston MA 02115, USA; Division of Cancer Genetics and Prevention, 450 Brookline Avenue, Boston MA 02215, USA. https://twitter.com/Dr_NJRodriguez
| | - Matthew B Yurgelun
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA; Harvard Medical School, Boston, MA, USA. https://twitter.com/MattYurgelun
| | - Sapna Syngal
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA.
| |
Collapse
|
3
|
Sagami R, Mizukami K, Nishikiori H, Sato T, Fujiwara S, Kawamoto Y, Ome Y, Honda G, Horiguchi SI, Sato K, Murakami K. Pancreatic juice cytology for diagnosing invasive pancreatic carcinoma/high-grade pancreatic intraepithelial neoplasia without visible tumors on endoscopic ultrasound. Pancreatology 2024; 24:740-746. [PMID: 38926041 DOI: 10.1016/j.pan.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 05/24/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
OBJECTIVES Pancreatic ductal adenocarcinoma (PDAC) with a diameter ≤10 mm and high-grade pancreatic intraepithelial neoplasia (HG-PanIN) require pre-operative diagnosis. Most cases present only indirect imaging findings without visible tumors on endoscopic ultrasound (EUS). Therefore, EUS-guided fine-needle aspiration/biopsy is not applicable. An alternative diagnostic method is pancreatic juice cytology (PJC) via endoscopic naso-pancreatic drainage (ENPD-PJC), which is not the standard practice. This study aimed to investigate ENPD-PJC for diagnosing suspected PDAC/HG-PanIN cases without visible tumors on EUS. METHODS Data of patients with suspected PDAC/HG-PanIN without visible tumors who underwent PJC were retrospectively evaluated. One PJC sample was collected during endoscopic retrograde pancreatography (ERP-PJC), and 12 samples were collected during ENPD-PJC, 3-hourly for cytological analysis. ERP-PJC, ERP/ENPD-PJC, and ENPD-PJC positivity indicated cytologically positive samples. Patients with positive/negative PJC with follow-up for <4-years were excluded as undiagnosed cases. A non-malignant diagnosis was based on histopathological absence/stable imaging findings for ≥4-years. The primary endpoint was to demonstrate that ERP/ENPD-PJC has a higher diagnostic ability than ERP-PJC. RESULTS Twenty-two patients with histopathologically diagnosed PDAC/HG-PanIN and 31 with a non-malignant diagnosis were enrolled. ERP-PJC, ERP/ENPD-PJC, and ENPD-PJC showed sensitivities of 36.4 %, 86.4 %, and 77.3 %, specificities of 93.5 %, 87.1 %, and 93.5 %, and accuracies of 69.8 %, 86.7 %, and 86.7 %, respectively. ERP/ENPD-PJC and ENPD-PJC demonstrated superior sensitivity and accuracy compared to ERP-PJC. A greater occurrence of positive outcomes markedly distinguished true positives from false positives. CONCLUSIONS ERP/ENPD-PJC and ENPD-PJC had higher diagnostic accuracies for PDAC/HG-PanIN without visible tumors on EUS. ENPD-PJC is recommended for the diagnosis of these lesions.
Collapse
Affiliation(s)
- Ryota Sagami
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Oita, Japan; Department of Gastroenterology, Oita San-ai Medical Centre, Oita, Oita, Japan
| | - Kazuhiro Mizukami
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Oita, Japan.
| | - Hidefumi Nishikiori
- Department of Gastroenterology, Oita San-ai Medical Centre, Oita, Oita, Japan
| | - Takao Sato
- Department of Gastroenterology, Oita San-ai Medical Centre, Oita, Oita, Japan
| | - Shozo Fujiwara
- Department of Surgery, Oita San-ai Medical Centre, Oita, Oita, Japan
| | - Yusuke Kawamoto
- Department of Surgery, Institute of Gastroenterology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - Yusuke Ome
- Department of Surgery, Institute of Gastroenterology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - Goro Honda
- Department of Surgery, Institute of Gastroenterology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - Shin-Ichiro Horiguchi
- Department of Pathology, Tokyo Metropolitan Cancer and Infectious Diseases Centre Komagome Hospital, Bunkyo-ku, Tokyo, Japan
| | - Keiji Sato
- Department of Pathology, Oita San-ai Medical Centre, Oita, Oita, Japan
| | - Kazunari Murakami
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| |
Collapse
|
4
|
Mustafa M, Abbas K, Alam M, Habib S, Zulfareen, Hasan GM, Islam S, Shamsi A, Hassan I. Investigating underlying molecular mechanisms, signaling pathways, emerging therapeutic approaches in pancreatic cancer. Front Oncol 2024; 14:1427802. [PMID: 39087024 PMCID: PMC11288929 DOI: 10.3389/fonc.2024.1427802] [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: 05/04/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Pancreatic adenocarcinoma, a clinically challenging malignancy constitutes a significant contributor to cancer-related mortality, characterized by an inherently poor prognosis. This review aims to provide a comprehensive understanding of pancreatic adenocarcinoma by examining its multifaceted etiologies, including genetic mutations and environmental factors. The review explains the complex molecular mechanisms underlying its pathogenesis and summarizes current therapeutic strategies, including surgery, chemotherapy, and emerging modalities such as immunotherapy. Critical molecular pathways driving pancreatic cancer development, including KRAS, Notch, and Hedgehog, are discussed. Current therapeutic strategies, including surgery, chemotherapy, and radiation, are discussed, with an emphasis on their limitations, particularly in terms of postoperative relapse. Promising research areas, including liquid biopsies, personalized medicine, and gene editing, are explored, demonstrating the significant potential for enhancing diagnosis and treatment. While immunotherapy presents promising prospects, it faces challenges related to immune evasion mechanisms. Emerging research directions, encompassing liquid biopsies, personalized medicine, CRISPR/Cas9 genome editing, and computational intelligence applications, hold promise for refining diagnostic approaches and therapeutic interventions. By integrating insights from genetic, molecular, and clinical research, innovative strategies that improve patient outcomes can be developed. Ongoing research in these emerging fields holds significant promise for advancing the diagnosis and treatment of this formidable malignancy.
Collapse
Affiliation(s)
- Mohd Mustafa
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Kashif Abbas
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Mudassir Alam
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Safia Habib
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Zulfareen
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Gulam Mustafa Hasan
- Department of Basic Medical Science, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Sidra Islam
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Anas Shamsi
- Center of Medical and Bio-Allied Health Sciences Research (CMBHSR), Ajman University, Ajman, United Arab Emirates
| | - Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
5
|
Farhangnia P, Khorramdelazad H, Nickho H, Delbandi AA. Current and future immunotherapeutic approaches in pancreatic cancer treatment. J Hematol Oncol 2024; 17:40. [PMID: 38835055 PMCID: PMC11151541 DOI: 10.1186/s13045-024-01561-6] [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: 04/09/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024] Open
Abstract
Pancreatic cancer is a major cause of cancer-related death, but despondently, the outlook and prognosis for this resistant type of tumor have remained grim for a long time. Currently, it is extremely challenging to prevent or detect it early enough for effective treatment because patients rarely exhibit symptoms and there are no reliable indicators for detection. Most patients have advanced or spreading cancer that is difficult to treat, and treatments like chemotherapy and radiotherapy can only slightly prolong their life by a few months. Immunotherapy has revolutionized the treatment of pancreatic cancer, yet its effectiveness is limited by the tumor's immunosuppressive and hard-to-reach microenvironment. First, this article explains the immunosuppressive microenvironment of pancreatic cancer and highlights a wide range of immunotherapy options, including therapies involving oncolytic viruses, modified T cells (T-cell receptor [TCR]-engineered and chimeric antigen receptor [CAR] T-cell therapy), CAR natural killer cell therapy, cytokine-induced killer cells, immune checkpoint inhibitors, immunomodulators, cancer vaccines, and strategies targeting myeloid cells in the context of contemporary knowledge and future trends. Lastly, it discusses the main challenges ahead of pancreatic cancer immunotherapy.
Collapse
Affiliation(s)
- Pooya Farhangnia
- Reproductive Sciences and Technology Research Center, Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hamid Nickho
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali-Akbar Delbandi
- Reproductive Sciences and Technology Research Center, Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Seeger N, Gutknecht S, Zschokke I, Fleischmann I, Roth N, Metzger J, Weber M, Breitenstein S, Grochola LF. A Predictive Noninvasive Single-Nucleotide Variation-Based Biomarker Signature for Resectable Pancreatic Cancer: Protocol for a Prospective Validation Study. JMIR Res Protoc 2024; 13:e54042. [PMID: 38635586 PMCID: PMC11130767 DOI: 10.2196/54042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 03/24/2024] [Accepted: 04/02/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND Single-nucleotide variations (SNVs; formerly SNPs) are inherited genetic variants that can be easily determined in routine clinical practice using a simple blood or saliva test. SNVs have potential to serve as noninvasive biomarkers for predicting cancer-specific patient outcomes after resection of pancreatic ductal adenocarcinoma (PDAC). Two recent analyses led to the identification and validation of three SNVs in the CD44 and CHI3L2 genes (rs187115, rs353630, and rs684559), which can be used as predictive biomarkers to help select patients most likely to benefit from pancreatic resection. These variants were associated with an over 2-fold increased risk for tumor-related death in three independent PDAC study cohorts from Europe and the United States, including The Cancer Genome Atlas cohorts (reaching a P value of 1×10-8). However, these analyses were limited by the inherent biases of a retrospective study design, such as selection and publication biases, thereby limiting the clinical use of these promising biomarkers in guiding PDAC therapy. OBJECTIVE To overcome the limitations of previous retrospectively designed studies and translate the findings into clinical practice, we aim to validate the association of the identified SNVs with survival in a controlled setting using a prospective cohort of patients with PDAC following pancreatic resection. METHODS All patients with PDAC who will undergo pancreatic resection at three participating hospitals in Switzerland and fulfill the inclusion criteria will be included in the study consecutively. The SNV genotypes will be determined using standard genotyping techniques from patient blood samples. For each genotyped locus, log-rank and Cox multivariate regression tests will be performed, accounting for the relevant covariates American Joint Committee on Cancer stage and resection status. Clinical follow-up data will be collected for at least 3 years. Sample size calculation resulted in a required sample of 150 patients to sufficiently power the analysis. RESULTS The follow-up data collection started in August 2019 and the estimated end of data collection will be in May 2027. The study is still recruiting participants and 142 patients have been recruited as of November 2023. The DNA extraction and genotyping of the SNVs will be performed after inclusion of the last patient. Since no SNV genotypes have been determined, no data analysis has been performed to date. The results are expected to be published in 2027. CONCLUSIONS This is the first prospective study of the CD44 and CHI3L2 SNV-based biomarker signature in PDAC. A prospective validation of this signature would enable its clinical use as a noninvasive predictive biomarker of survival after pancreatic resection that is readily available at the time of diagnosis and can assist in guiding PDAC therapy. The results of this study may help to individualize treatment decisions and potentially improve patient outcomes. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/54042.
Collapse
Affiliation(s)
- Nico Seeger
- Department of Visceral and Thoracic Surgery, Cantonal Hospital of Winterthur, Winterthur, Switzerland
| | - Stefan Gutknecht
- Department of Visceral, Thoracic and Cardiovascular Surgery, Triemli Hospital, Zurich, Switzerland
| | - Irin Zschokke
- Department of General and Visceral Surgery, Cantonal Hospital of Lucerne, Lucerne, Switzerland
| | - Isabella Fleischmann
- Department of General and Visceral Surgery, Cantonal Hospital of Lucerne, Lucerne, Switzerland
| | - Nadja Roth
- Department of General and Visceral Surgery, Cantonal Hospital of Lucerne, Lucerne, Switzerland
| | - Jürg Metzger
- Department of General and Visceral Surgery, Cantonal Hospital of Lucerne, Lucerne, Switzerland
| | - Markus Weber
- Department of Visceral, Thoracic and Cardiovascular Surgery, Triemli Hospital, Zurich, Switzerland
| | - Stefan Breitenstein
- Department of Visceral and Thoracic Surgery, Cantonal Hospital of Winterthur, Winterthur, Switzerland
| | - Lukasz Filip Grochola
- Department of Visceral and Thoracic Surgery, Cantonal Hospital of Winterthur, Winterthur, Switzerland
| |
Collapse
|
7
|
López JC, Ielpo B, Iglesias M, Pinilla FB, Sánchez-Velázquez P. The impact of vascular margin invasion on local recurrence after pancreatoduodenectomy in pancreatic adenocarcinoma. Langenbecks Arch Surg 2024; 409:122. [PMID: 38607450 PMCID: PMC11009726 DOI: 10.1007/s00423-024-03301-3] [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: 09/15/2023] [Accepted: 03/26/2024] [Indexed: 04/13/2024]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PADC) still has nowadays a very impaired long-term survival. Most studies are focused on overall survival; however, local recurrence occurs about up to 50% of cases and seems to be highly related with margin resection status. We aim to analyze the impact of vascular resection margins on local recurrence (LR) and to assess its impact on overall and disease-free survival. METHODS Eighty out of 191 patients who underwent pancreatoduodenectomy in a university hospital between 2006 and 2021 with PDAC diagnosis were analyzed and vascular margin status specifically addressed. Univariate and multivariate were performed. Time to LR was compared by using the Kaplan-Meier method and prognostic factors assessed using Cox regression hazards model. RESULTS LR appeared in 10 (50%) of the overall R1 resections in the venous margin and 9 (60%) in the arterial one. Time to LR was significantly shorter when any margin was overall affected (23.2 vs 44.7 months, p = 0.01) and specifically in the arterial margin involvement (13.7 vs 32.1 months, p = 0.009). Overall R1 resections (HR 2.61, p = 0.013) and a positive arterial margin (HR 2.84, p = 0.012) were associated with local recurrence on univariate analysis, whereas arterial positive margin remained significant on multivariate analysis (HR 2.70, p = 0.031). CONCLUSIONS Arterial margin invasion is correlated in our cohort with local recurrence. Given the limited ability to modify this margin intraoperatively, preoperative therapies should be considered to improve local margin clearance.
Collapse
Affiliation(s)
| | - Benedetto Ielpo
- Department of Surgery, Division of Hepato-Biliary and Pancreatic Surgery, University Hospital del Mar-IMIM (Hospital del Mar Medical Research Institute), Universitat Pompeu Fabra, 08003, Barcelona, Spain
| | - Mar Iglesias
- Pompeu-Fabra University, Barcelona, Spain
- Department of Surgery, Division of Hepato-Biliary and Pancreatic Surgery, University Hospital del Mar-IMIM (Hospital del Mar Medical Research Institute), Universitat Pompeu Fabra, 08003, Barcelona, Spain
- Department of Pathology, University Hospital del Mar-IMIM (Hospital del Mar Medical Research Institute), Universitat Pompeu-Fabra, Barcelona, Spain
| | - Fernando Burdío Pinilla
- Department of Surgery, Division of Hepato-Biliary and Pancreatic Surgery, University Hospital del Mar-IMIM (Hospital del Mar Medical Research Institute), Universitat Pompeu Fabra, 08003, Barcelona, Spain
| | - Patricia Sánchez-Velázquez
- Department of Surgery, Division of Hepato-Biliary and Pancreatic Surgery, University Hospital del Mar-IMIM (Hospital del Mar Medical Research Institute), Universitat Pompeu Fabra, 08003, Barcelona, Spain.
| |
Collapse
|
8
|
Sagami R, Yamao K, Minami R, Nakahodo J, Akiyama H, Nishikiori H, Mizukami K, Yamao K, Bhatia V, Amano Y, Murakami K. Endoscopic Ultrasound Can Differentiate High-Grade Pancreatic Intraepithelial Neoplasia, Small Pancreatic Ductal Adenocarcinoma, and Benign Stenosis. Gut Liver 2024; 18:338-347. [PMID: 37165770 PMCID: PMC10938144 DOI: 10.5009/gnl220521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 02/06/2023] [Accepted: 02/15/2023] [Indexed: 05/12/2023] Open
Abstract
Background/Aims High-grade pancreatic intraepithelial neoplasia and invasive pancreatic ductal adenocarcinoma ≤10 mm are targets for early detection of pancreatic cancer. However, their imaging characteristics are unknown. We aimed to identify endoscopic ultrasound findings for the detection of these lesions. Methods Patients diagnosed with high-grade pancreatic intraepithelial neoplasia (n=29), pancreatic ductal adenocarcinoma ≤10 mm (n=11) (who underwent surgical resection), or benign main pancreatic duct stenosis (n=20) between January 2014 and January 2021 were retrospectively included. Six features differentiating these lesions were examined by endoscopic ultrasonography: main pancreatic duct stenosis, upstream main pancreatic duct dilation, hypoechoic areas surrounding the main pancreatic duct irregularities (mottled areas without demarcation or round areas with demarcation), branch duct dilation, prominent lobular segmentation, and atrophy. Interobserver agreement was assessed by two independent observers. Results Hypoechoic areas surrounding the main pancreatic duct irregularities were observed more frequently in high-grade pancreatic intraepithelial neoplasia (82.8%) and pancreatic ductal adenocarcinoma ≤10 mm (90.9%) than in benign stenosis (15.0%) (p<0.001). High-grade pancreatic intraepithelial neoplasia exhibited mottled hypoechoic areas more frequently (79.3% vs 18.9%, p<0.001), and round hypoechoic areas less frequently (3.4% vs 72.7%, p<0.001), than pancreatic ductal adenocarcinoma ≤10 mm. The sensitivity and specificity of hypoechoic areas for differentiating high-grade pancreatic intraepithelial neoplasia, pancreatic ductal adenocarcinoma ≤10 mm, and benign stenosis were both 85.0%, with moderate interobserver agreement. Conclusions The hypoechoic areas surrounding main pancreatic duct irregularities on endoscopic ultrasound may differentiate between high-grade pancreatic intraepithelial neoplasia, pancreatic ductal adenocarcinoma ≤10 mm, and benign stenosis (Trial Registration: UMIN Clinical Trials Registry (UMIN000044789).
Collapse
Affiliation(s)
- Ryota Sagami
- Department of Gastroenterology, Oita San-ai Medical Center, Oita, Japan
- Pancreatic Cancer Research for Secure Salvage Young Investigators (PASSYON), Osaka-Sayama, Japan
| | - Kentaro Yamao
- Pancreatic Cancer Research for Secure Salvage Young Investigators (PASSYON), Osaka-Sayama, Japan
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Ryuki Minami
- Pancreatic Cancer Research for Secure Salvage Young Investigators (PASSYON), Osaka-Sayama, Japan
- Department of Gastroenterology, Tenri Hospital, Nara, Japan
| | - Jun Nakahodo
- Pancreatic Cancer Research for Secure Salvage Young Investigators (PASSYON), Osaka-Sayama, Japan
- Department of Gastroenterology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Hidetoshi Akiyama
- Department of Gastroenterology, Oita Red Cross Hospital, Oita, Japan
| | | | - Kazuhiro Mizukami
- Department of Gastroenterology, Faculty of Medicine, Oita University, Oita, Japan
| | - Kenji Yamao
- Department of Gastroenterology, Narita Memorial Hospital, Aichi, Japan
| | - Vikram Bhatia
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Yuji Amano
- Department of Endoscopy, Urawa Kyosai Hospital, Saitama, Japan
| | - Kazunari Murakami
- Department of Gastroenterology, Faculty of Medicine, Oita University, Oita, Japan
| |
Collapse
|
9
|
Li Y, Chen MX, Li HT, Cai XM, Chen B, Xie ZF. Comprehensive analysis based on the disulfidptosis-related genes identifies hub genes and immune infiltration for pancreatic adenocarcinoma. Open Med (Wars) 2024; 19:20240906. [PMID: 38463521 PMCID: PMC10921452 DOI: 10.1515/med-2024-0906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/11/2023] [Accepted: 01/03/2024] [Indexed: 03/12/2024] Open
Abstract
Pancreatic adenocarcinoma (PAAD) is a prevalent and aggressive malignancy in the digestive tract, requiring accurate prediction and effective treatment strategies. Recently, the discovery of disulfidptosis, a novel form of programmed cell death characterized by abnormal disulfide accumulation, has sparked interest in its role in PAAD. In this study, we aimed to investigate the involvement of disulfidptosis-related genes (DRGs) in PAAD. Using publicly available databases, we conducted a comprehensive analysis exploring the complex relationships between DRGs and important aspects of PAAD, including gene expression, immune response, mutation, drug sensitivity, and functional enrichment. Notably, we observed significant heterogeneity among different disulfidptosis subclusters and identified specific differentially expressed genes in PAAD. Through machine learning techniques, we identified SLC7A11, S100A4, DIAPH3, PRDX1, and SLC7A7 as the most relevant hub genes. We further validated their significance in PAAD by considering their expression patterns, prognostic value, diagnostic potential, diagnostic model, and immune infiltration. This study presents exciting opportunities and challenges in unraveling the underlying mechanisms of PAAD prognosis. It also establishes a foundation for personalized cancer care and the development of innovative immunotherapeutic strategies. By shedding light on the role of DRGs, particularly hub genes, we enhance our understanding and management of PAAD.
Collapse
Affiliation(s)
- Yu Li
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Miao-Xuan Chen
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Hai-Tao Li
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Xiao-Ming Cai
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Bo Chen
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Ze-Feng Xie
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
10
|
Zhang Y, Wu J, He J, Xu S. Preoperative differentiation of pancreatic cystic neoplasm subtypes on computed tomography radiomics. Quant Imaging Med Surg 2023; 13:6395-6411. [PMID: 37869288 PMCID: PMC10585572 DOI: 10.21037/qims-22-1192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 07/28/2023] [Indexed: 10/24/2023]
Abstract
Background Serous cystic neoplasm (SCN), mucinous cystic neoplasm (MCN), and intraductal papillary mucinous neoplasm (IPMN) comprise a large proportion of pancreatic cystic neoplasms (PCNs). Patients with MCN and IPMN require surgery due to the potential of malignant transformation, whereas those with SCN require periodic surveillance. However, the differential diagnosis of patients with PCNs before treatment remains a great challenge for all surgeons. Therefore, the establishment of a reliable diagnostic tool is urgently required for the improvement of precision diagnostics. Methods Between February 2015 and December 2020, 143 consecutive patients with PCNs who were confirmed by postoperative pathology were retrospectively included in the study cohort, then randomized into development and test cohorts at a ratio of 7:3. The predictors of preoperative clinical-radiologic parameters were evaluated by univariate and multivariable logistic regression analyses. A total of 1,218 radiomics features were computationally extracted from the enhanced computed tomography (CT) scans of the tumor region, and a radiomics signature was established by the random forest algorithm. In the development cohort, multi- and binary-class radiomics models integrating preoperative variables and radiomics features were constructed to distinguish between the 3 types of PCNs. The receiver operating characteristic (ROC) curve and the area under the curve (AUC) were used to evaluate the predictive efficiency of the model. An independent internal test cohort was applied to validate the classification models. Results All preoperative prediction models were built by integrating the radiomics signature with 13 diagnosis-related radiomics features and 3 important clinical-radiologic parameters: age, sex, and tumor diameter. The multiclass prediction model presented an overall accuracy of 0.804 in the development cohort and 0.707 in the test cohort. The binary-class prediction models displayed higher overall accuracies of 0.853, 0.866, and 0.928 in the development dataset and 0.750, 0.839, and 0.889 in the test dataset. In the test cohort, the binary-class radiomics models showed better predictive performances {AUC =0.914 [95% confidence interval (CI): 0.786 to 1.000], 0.863 (95% CI: 0.714 to 0.941), and 0.926 (95% CI: 0.824 to 1.000)} than the multiclass radiomics model [AUC =0.850 (95% CI: 0.696 to 1.000)], with a large net benefit in the decision curve analysis (DCA). The radiomics-based nomogram provided the correct predicted probability for the diagnosis of PCNs. Conclusions The proposed radiomics models with clinical-radiologic parameters and radiomics features help to predict the accurate diagnosis among PCNs to advance personalized medicine.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of PET/CT Center, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, China
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jin Wu
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jian He
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Shanshan Xu
- Department of PET/CT Center, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, China
| |
Collapse
|
11
|
Xu Z, Huang Z, Zhang Y, Sun H, Hinz U, Heger U, Loos M, Gonzalez FJ, Hackert T, Bergmann F, Fortunato F. Farnesoid X receptor activation inhibits pancreatic carcinogenesis. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166811. [PMID: 37515840 PMCID: PMC10935600 DOI: 10.1016/j.bbadis.2023.166811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 06/26/2023] [Accepted: 07/11/2023] [Indexed: 07/31/2023]
Abstract
Farnesoid X receptor (FXR), a member of the nuclear receptor superfamily that controls bile acid (BA) homeostasis, has also been proposed as a tumor suppressor for breast and liver cancer. However, its role in pancreatic ductal adenocarcinoma (PDAC) tumorigenesis remains controversial. We recently found that FXR attenuates acinar cell autophagy in chronic pancreatitis resulting in reduced autophagy and promotion of pancreatic carcinogenesis. Feeding Kras-p48-Cre (KC) mice with the BA chenodeoxycholic acid (CDCA), an FXR agonist, attenuated pancreatic intraepithelial neoplasia (PanIN) progression, reduced cell proliferation, neoplastic cells and autophagic activity, and increased acinar cells, elevated pro-inflammatory cytokines and chemokines, with a compensatory increase in the anti-inflammatory response. Surprisingly, FXR-deficient KC mice did not show any response to CDCA, suggesting that CDCA attenuates PanIN progression and decelerate tumorigenesis in KC mice through activating pancreatic FXR. FXR is activated in pancreatic cancer cell lines in response to CDCA in vitro. FXR levels were highly increased in adjuvant and neoadjuvant PDAC tissue compared to healthy pancreatic tissue, indicating that FXR is expressed and potentially activated in human PDAC. These results suggest that BA exposure activates inflammation and suppresses autophagy in KC mice, resulting in reduced PanIN lesion progression. These data suggest that activation of pancreatic FXR has a protective role by reducing the growth of pre-cancerous PDAC lesions in response to CDCA and possibly other FXR agonists.
Collapse
Affiliation(s)
- Zhen Xu
- Department of General, Visceral and Transplantation Surgery, University Clinic Heidelberg, Heidelberg, Germany; Section Surgical Research, University Clinic Heidelberg, Heidelberg, Germany
| | - Zhenhua Huang
- Department of General, Visceral and Transplantation Surgery, University Clinic Heidelberg, Heidelberg, Germany; Section Surgical Research, University Clinic Heidelberg, Heidelberg, Germany
| | - Yifan Zhang
- Department of General, Visceral and Transplantation Surgery, University Clinic Heidelberg, Heidelberg, Germany; Section Surgical Research, University Clinic Heidelberg, Heidelberg, Germany
| | - Haitao Sun
- Department of General, Visceral and Transplantation Surgery, University Clinic Heidelberg, Heidelberg, Germany; Section Surgical Research, University Clinic Heidelberg, Heidelberg, Germany
| | - Ulf Hinz
- Department of General, Visceral and Transplantation Surgery, University Clinic Heidelberg, Heidelberg, Germany
| | - Ulrike Heger
- Department of General, Visceral and Transplantation Surgery, University Clinic Heidelberg, Heidelberg, Germany
| | - Martin Loos
- Department of General, Visceral and Transplantation Surgery, University Clinic Heidelberg, Heidelberg, Germany
| | - Frank J Gonzalez
- National Cancer Institute, National Institutes of Health, MD, Bethesda, USA
| | - Thilo Hackert
- Department of General, Visceral and Transplantation Surgery, University Clinic Heidelberg, Heidelberg, Germany
| | - Frank Bergmann
- Institute of Pathology, University Clinic Heidelberg, Heidelberg, Germany
| | - Franco Fortunato
- Department of General, Visceral and Transplantation Surgery, University Clinic Heidelberg, Heidelberg, Germany; Section Surgical Research, University Clinic Heidelberg, Heidelberg, Germany.
| |
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: 0] [Impact Index Per Article: 0] [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
|
Yamakawa K, Inomata N, Masuda A, Takenaka M, Toyama H, Sofue K, Sakai A, Kobayashi T, Tanaka T, Tsujimae M, Ashina S, Gonda M, Abe S, Masuda S, Uemura H, Kohashi S, Nagao K, Harada Y, Miki M, Irie Y, Juri N, Shiomi H, Kanzawa M, Itoh T, Fukumoto T, Kodama Y. Benefits of pancreatic parenchymal endoscopic ultrasonography in predicting microscopic precancerous lesions of pancreatic cancer. Sci Rep 2023; 13:12052. [PMID: 37491554 PMCID: PMC10368726 DOI: 10.1038/s41598-023-38920-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/17/2023] [Indexed: 07/27/2023] Open
Abstract
Pancreatic cancer primarily arises from microscopic precancerous lesions, such as pancreatic intraepithelial neoplasia (PanIN) and acinar-to-ductal metaplasia (ADM). However, no established method exists for predicting pancreatic precancerous conditions. Endoscopic ultrasonography (EUS) can detect changes in pancreatic parenchymal histology, including fibrosis. This study aimed to elucidate the relationship between pancreatic parenchymal EUS findings and microscopic precancerous lesions. We retrospectively analyzed 114 patients with pancreatobiliary tumors resected between 2010 and 2020 and evaluated the association between pancreatic parenchymal EUS findings and the number of PanIN, ADM, and pancreatic duct gland (PDG). Of the 114 patients, 33 (29.0%), 55 (48.2%), and 26 (22.8%) had normal EUS findings, hyperechoic foci/stranding without lobularity, and hyperechoic foci/stranding with lobularity, respectively. Multivariate analyses revealed that abnormal EUS findings were significantly associated with the frequency of PanIN (hyperechoic foci/stranding without lobularity: OR [95% CI] = 2.7 [1.0-7.3], with lobularity: 6.5 [1.9-22.5], Ptrend = 0.01) and ADM (hyperechoic foci/stranding without lobularity: 3.1 [1.1-8.2], with lobularity: 9.7 [2.6-36.3], Ptrend = 0.003) but not with PDG (hyperechoic foci/stranding without lobularity: 2.2 [0.8-5.8], with lobularity: 3.2 [1.0-10.2], Ptrend = 0.12). We observed a trend toward a significantly higher number of precancerous lesions in the following order: normal findings, hyperechoic foci/stranding without lobularity, and hyperechoic foci/stranding with lobularity. Pancreatic parenchymal EUS findings were associated with the increased frequency of PanIN and ADM. Lobularity may help predict the increased number of precancerous lesions.
Collapse
Affiliation(s)
- Kohei Yamakawa
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Noriko Inomata
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Atsuhiro Masuda
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan.
| | - Mamoru Takenaka
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Sayama, Japan
| | - Hirochika Toyama
- Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Keitaro Sofue
- Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Arata Sakai
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Takashi Kobayashi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Takeshi Tanaka
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Masahiro Tsujimae
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Shigeto Ashina
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Masanori Gonda
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Shohei Abe
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Shigeto Masuda
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Hisahiro Uemura
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Shinya Kohashi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Kae Nagao
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Yoshiyuki Harada
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Mika Miki
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Yosuke Irie
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Noriko Juri
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Hideyuki Shiomi
- Division of Gastroenterology and Hepato-Biliary-Pancreatology, Department of Internal Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Maki Kanzawa
- Division of Diagnostic Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoo Itoh
- Division of Diagnostic Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takumi Fukumoto
- Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuzo Kodama
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| |
Collapse
|
14
|
Dhani H, Hinestrosa JP, Izaguirre-Carbonell J, Balcer HI, Kurzrock R, Billings PR. Case Report: Early detection of pancreatic pre-cancer lesion in multimodal approach with exosome liquid biopsy. Front Oncol 2023; 13:1170513. [PMID: 37251939 PMCID: PMC10210587 DOI: 10.3389/fonc.2023.1170513] [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: 04/25/2023] [Indexed: 05/31/2023] Open
Abstract
Background The detection of pancreatic ductal adenocarcinoma (PDAC) lesions at pre-cancerous or early-stages is critical to improving patient survival. We have developed a liquid biopsy test (ExoVita®) based on the measurement of protein biomarkers in cancer-derived exosomes. The high sensitivity and specificity of the test for early-stage PDAC has the potential to improve a patient's diagnostic journey in hopes to impact patient outcomes. Methods Exosome isolation was performed using alternating current electric (ACE) field applied to the patient plasma sample. Following a wash to eliminate unbound particles, the exosomes were eluted from the cartridge. A downstream multiplex immunoassay was performed to measure proteins of interest on the exosomes, and a proprietary algorithm provided a score for probability of PDAC. Results We describe the case of a 60-year-old healthy non-Hispanic white male with acute pancreatitis who underwent numerous invasive diagnostic procedures that failed to detect radiographic evidence of pancreatic lesions. Following the results of our exosome-based liquid biopsy test showing "High Likelihood of PDAC", in addition to KRAS and TP53 mutations, the patient decided to undergo a robotic pancreaticoduodenectomy (Whipple) procedure. Surgical pathology confirmed the diagnosis of high-grade intraductal papillary mucinous neoplasm (IPMN), which was consistent with the results of our ExoVita® test. The patient's post-operative course was unremarkable. At five-month follow-up, the patient continued to recover well without complications, in addition to a repeat ExoVita test which demonstrated "Low Likelihood of PDAC". Conclusion This case report highlights how a novel liquid biopsy diagnostic test based on the detection of exosome protein biomarkers allowed early diagnosis of a high-grade precancerous lesion for PDAC and improved patient outcome.
Collapse
Affiliation(s)
- Harmeet Dhani
- Biological Dynamics, Inc., San Diego, CA, United States
- Georgetown University, Washington, DC, United States
| | | | | | | | - Razelle Kurzrock
- Medical College of Wisconsin, Milwaukee, WI, United States
- Worldwide Innovative Network (WIN) Consortium for Precision Medicine, Villejuif, France
| | | |
Collapse
|
15
|
Zou X, Huang Z, Guan C, Shi W, Gao J, Wang J, Cui Y, Wang M, Xu Y, Zhong X. Exosomal miRNAs in the microenvironment of pancreatic cancer. Clin Chim Acta 2023; 544:117360. [PMID: 37086943 DOI: 10.1016/j.cca.2023.117360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/17/2023] [Accepted: 04/17/2023] [Indexed: 04/24/2023]
Abstract
Pancreatic cancer (PC) is highly aggressive having an extremely poor prognosis. The tumor microenvironment (TME) of PC is complex and heterogeneous. Various cellular components in the microenvironment are capable of secreting different active substances that are involved in promoting tumor development. Their release may occur via exosomes, the most abundant extracellular vesicles (EVs), that can carry numerous factors as well as act as a mean of intercellular communication. Emerging evidence suggests that miRNAs are involved in the regulation and control of many pathological and physiological processes. They can also be transported by exosomes from donor cells to recipient cells, thereby regulating the TME. Exosomal miRNAs show promise for use as future targets for PC diagnosis and prognosis, which may reveal new treatment strategies for PC. In this paper, we review the important role of exosomal miRNAs in mediating cellular communication in the TME of PC as well as their potential use in clinical applications.
Collapse
Affiliation(s)
- Xinlei Zou
- Department of Hepatopancreatobiary Surgery, the 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Ziyue Huang
- Department of Hepatopancreatobiary Surgery, the 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Canghai Guan
- Department of Hepatopancreatobiary Surgery, the 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Wujiang Shi
- Department of Hepatopancreatobiary Surgery, the 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Jianjun Gao
- Department of Hepatopancreatobiary Surgery, the 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Jiangang Wang
- Central hospital of Baoji, Baoji, Shaanxi 721000, China
| | - Yunfu Cui
- Department of Hepatopancreatobiary Surgery, the 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Mei Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi 563006, China
| | - Yi Xu
- Department of Hepatopancreatobiary Surgery, the 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, China; Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang Province, Hangzhou 310000, China; State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China.
| | - Xiangyu Zhong
- Department of Hepatopancreatobiary Surgery, the 2nd Affiliated Hospital of Harbin Medical University, Harbin 150086, China.
| |
Collapse
|
16
|
Mazer BL, Lee JW, Roberts NJ, Chu LC, Lennon AM, Klein AP, Eshleman JR, Fishman EK, Canto MI, Goggins MG, Hruban RH. Screening for pancreatic cancer has the potential to save lives, but is it practical? Expert Rev Gastroenterol Hepatol 2023; 17:555-574. [PMID: 37212770 PMCID: PMC10424088 DOI: 10.1080/17474124.2023.2217354] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/21/2023] [Accepted: 05/19/2023] [Indexed: 05/23/2023]
Abstract
INTRODUCTION Most patients with pancreatic cancer present with advanced stage, incurable disease. However, patients with high-grade precancerous lesions and many patients with low-stage disease can be cured with surgery, suggesting that early detection has the potential to improve survival. While serum CA19.9 has been a long-standing biomarker used for pancreatic cancer disease monitoring, its low sensitivity and poor specificity have driven investigators to hunt for better diagnostic markers. AREAS COVERED This review will cover recent advances in genetics, proteomics, imaging, and artificial intelligence, which offer opportunities for the early detection of curable pancreatic neoplasms. EXPERT OPINION From exosomes, to circulating tumor DNA, to subtle changes on imaging, we know much more now about the biology and clinical manifestations of early pancreatic neoplasia than we did just five years ago. The overriding challenge, however, remains the development of a practical approach to screen for a relatively rare, but deadly, disease that is often treated with complex surgery. It is our hope that future advances will bring us closer to an effective and financially sound approach for the early detection of pancreatic cancer and its precursors.
Collapse
Affiliation(s)
- Benjamin L. Mazer
- The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jae W. Lee
- The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
| | - Nicholas J. Roberts
- The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Linda C. Chu
- Department of Radiology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anne Marie Lennon
- Department of Medicine, Division of Gastroenterology and Hepatology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alison P. Klein
- The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - James R. Eshleman
- The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elliot K. Fishman
- Department of Radiology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marcia Irene Canto
- Department of Medicine, Division of Gastroenterology and Hepatology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael G. Goggins
- The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ralph H. Hruban
- The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
17
|
Sagami R, Hayasaka K, Ujihara T, Iwaki T, Katsuyama Y, Harada H, Ome Y, Honda G, Horiguchi SI, Murakami K, Amano Y. Role of EUS combined with a newly modified scoring system to detect pancreatic high-grade precancerous lesions. Endosc Ultrasound 2023; 12:111-119. [PMID: 36861510 PMCID: PMC10134925 DOI: 10.4103/eus-d-21-00187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 08/01/2022] [Indexed: 03/03/2023] Open
Abstract
Backgrounds and Objectives Although pancreatic cancer (PC) has an extremely poor prognosis, the 5-year survival rate of patients with pancreatic high-grade precancerous lesion without invasive carcinoma (PHP) is favorable. PHP diagnosis and identification of patients requiring intervention are needed. We aimed to validate a modified PC detection scoring system regarding its detection ability for PHP and PC in the general population. Subjects and Methods We modified an existing PC detection scoring system that incorporates low-grade risk (LGR) factors (family history, presence of diabetes mellitus [DM] or worsening DM, heavy drinking, smoking, stomach symptoms, weight loss, and pancreatic enzyme) and high-grade risk (HGR) factors (new-onset DM, familial PC, jaundice, tumor biomarkers, chronic pancreatitis, intraductal papillary mucinous neoplasm, cysts, hereditary PC syndrome, and hereditary pancreatitis). Each factor was scored as one point; LGR score ≥3 points and/or HGR score ≥1 point (positive scores) were indicative of PC. The newly modified scoring system incorporated main pancreatic duct dilation as an HGR factor. The PHP diagnosis rate using this scoring system combined with EUS was prospectively analyzed. Results Among 544 patients with positive scores, 10 had PHP. The diagnosis rates were 1.8% for PHP and 4.2% for invasive PC. Although the number of LGR and HGR factors tended to increase with PC progression, none of the individual factors were significantly different between patients with PHP and those without lesions. Conclusion The newly modified scoring system evaluating multiple factors associated with PC could potentially identify patients with higher risk of PHP or PC.
Collapse
Affiliation(s)
- Ryota Sagami
- Department of Gastroenterology, Oita San-ai Medical Center, Oita, Japan
| | - Kenji Hayasaka
- Department of Gastroenterology, New Tokyo Hospital, Matsudo, Chiba, Japan
| | - Tetsuro Ujihara
- Department of Gastroenterology, New Tokyo Hospital, Matsudo, Chiba, Japan
| | - Tomoyuki Iwaki
- Department of Gastroenterology, Urawa Kyosai Hospital, Saitama, Japan
| | - Yasushi Katsuyama
- Department of Gastroenterology, Urawa Kyosai Hospital, Saitama, Japan
| | - Hideaki Harada
- Department of Gastroenterology, Urawa Kyosai Hospital, Saitama, Japan
| | - Yusuke Ome
- Department of Surgery, Institute of Gastroenterology, Tokyo Women’s Medical University, Shinjuku-Ku, Tokyo, Japan
| | - Goro Honda
- Department of Surgery, Institute of Gastroenterology, Tokyo Women’s Medical University, Shinjuku-Ku, Tokyo, Japan
| | - Shin-ichiro Horiguchi
- Department of Pathology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Bunkyo, Tokyo, Japan
| | - Kazunari Murakami
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Yuji Amano
- Department of Gastroenterology, Urawa Kyosai Hospital, Saitama, Japan
| |
Collapse
|
18
|
Li B, Wang J, Liao J, Wu M, Yuan X, Fang H, Shen L, Jiang M. YY1 promotes pancreatic cancer cell proliferation by enhancing mitochondrial respiration. Cancer Cell Int 2022; 22:287. [PMID: 36123703 PMCID: PMC9484254 DOI: 10.1186/s12935-022-02712-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 09/09/2022] [Indexed: 11/13/2022] Open
Abstract
KRAS-driven metabolic reprogramming is a known peculiarity features of pancreatic ductal adenocarcinoma (PDAC) cells. However, the metabolic roles of other oncogenic genes, such as YY1, in PDAC development are still unclear. In this study, we observed significantly elevated expression of YY1 in human PDAC tissues, which positively correlated with a poor disease progression. Furthermore, in vitro studies confirmed that YY1 deletion inhibited PDAC cell proliferation and tumorigenicity. Moreover, YY1 deletion led to impaired mitochondrial RNA expression, which further inhibited mitochondrial oxidative phosphorylation (OXPHOS) complex assembly and altered cellular nucleotide homeostasis. Mechanistically, the impairment of mitochondrial OXPHOS function reduced the generation of aspartate, an output of the tricarboxylic acid cycle (TCA), and resulted in the inhibition of cell proliferation owing to unavailability of aspartate-associated nucleotides. Conversely, exogenous supplementation with aspartate fully restored PDAC cell proliferation. Our findings suggest that YY1 promotes PDAC cell proliferation by enhancing mitochondrial respiration and the TCA, which favors aspartate-associated nucleotide synthesis. Thus, targeting nucleotide biosynthesis is a promising strategy for PDAC treatment.
Collapse
Affiliation(s)
- Bin Li
- Department of Laboratory Medicine, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Junyi Wang
- Department of Clinical Laboratory Examination, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518000, China
| | - Jing Liao
- Key Laboratory of Laboratory Medicine, Ministry of Education; Zhejiang Provincial Key Laboratory of Medical Genetics; College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Minghui Wu
- Department of Laboratory Medicine, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Xiangshu Yuan
- Key Laboratory of Laboratory Medicine, Ministry of Education; Zhejiang Provincial Key Laboratory of Medical Genetics; College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Hezhi Fang
- Key Laboratory of Laboratory Medicine, Ministry of Education; Zhejiang Provincial Key Laboratory of Medical Genetics; College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Lijun Shen
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| | - Minghua Jiang
- Department of Laboratory Medicine, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| |
Collapse
|
19
|
Anta JA, Martínez-Ballestero I, Eiroa D, García J, Rodríguez-Comas J. Artificial intelligence for the detection of pancreatic lesions. Int J Comput Assist Radiol Surg 2022; 17:1855-1865. [PMID: 35951286 DOI: 10.1007/s11548-022-02706-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 06/17/2022] [Indexed: 11/30/2022]
Abstract
PURPOSE Pancreatic cancer is one of the most lethal neoplasms among common cancers worldwide, and PCLs are well-known precursors of this type of cancer. Artificial intelligence (AI) could help to improve and speed up the detection and classification of pancreatic lesions. The aim of this review is to summarize the articles addressing the diagnostic yield of artificial intelligence applied to medical imaging (computed tomography [CT] and/or magnetic resonance [MR]) for the detection of pancreatic cancer and pancreatic cystic lesions. METHODS We performed a comprehensive literature search using PubMed, EMBASE, and Scopus (from January 2010 to April 2021) to identify full articles evaluating the diagnostic accuracy of AI-based methods processing CT or MR images to detect pancreatic ductal adenocarcinoma (PDAC) or pancreatic cystic lesions (PCLs). RESULTS We found 20 studies meeting our inclusion criteria. Most of the AI-based systems used were convolutional neural networks. Ten studies addressed the use of AI to detect PDAC, eight studies aimed to detect and classify PCLs, and 4 aimed to predict the presence of high-grade dysplasia or cancer. CONCLUSION AI techniques have shown to be a promising tool which is expected to be helpful for most radiologists' tasks. However, methodologic concerns must be addressed, and prospective clinical studies should be carried out before implementation in clinical practice.
Collapse
Affiliation(s)
- Julia Arribas Anta
- Scientific and Technical Department, Sycai Technologies S.L., Carrer Roc Boronat 117, MediaTIC Building, 08018, Barcelona, Spain.,Department of Gastroenterology, University Hospital, 12 Octubre. Av. de Córdoba, s/n, 28041, Madrid, Spain
| | - Iván Martínez-Ballestero
- Scientific and Technical Department, Sycai Technologies S.L., Carrer Roc Boronat 117, MediaTIC Building, 08018, Barcelona, Spain
| | - Daniel Eiroa
- Scientific and Technical Department, Sycai Technologies S.L., Carrer Roc Boronat 117, MediaTIC Building, 08018, Barcelona, Spain.,Department of Radiology, Institut de Diagnòstic per la Imatge (IDI), Hospital Universitari Vall d'Hebrón, Passeig de la Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| | - Javier García
- Scientific and Technical Department, Sycai Technologies S.L., Carrer Roc Boronat 117, MediaTIC Building, 08018, Barcelona, Spain
| | - Júlia Rodríguez-Comas
- Scientific and Technical Department, Sycai Technologies S.L., Carrer Roc Boronat 117, MediaTIC Building, 08018, Barcelona, Spain.
| |
Collapse
|
20
|
Krishnan SN, Mohammed S, Frankel TL, Rao A. GaWRDenMap: a quantitative framework to study the local variation in cell-cell interactions in pancreatic disease subtypes. Sci Rep 2022; 12:3708. [PMID: 35260589 PMCID: PMC8904504 DOI: 10.1038/s41598-022-06602-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 01/24/2022] [Indexed: 11/09/2022] Open
Abstract
Spatial pattern modelling concepts are being increasingly used in capturing disease heterogeneity. Quantification of heterogeneity in the tumor microenvironment is extremely important in pancreatic ductal adenocarcinoma (PDAC), which has been shown to co-occur with other pancreatic diseases and neoplasms with certain attributes that make visual discrimination difficult. In this paper, we propose the GaWRDenMap framework, that utilizes the concepts of geographically weighted regression (GWR) and a density function-based classification model, and apply it to a cohort of multiplex immunofluorescence images from patients belonging to six different pancreatic diseases. We used an internal cohort of 228 patients comprised of 34 Chronic Pancreatitis (CP), 71 PDAC, 70 intraductal papillary mucinous neoplasm (IPMN), 16 mucinous cystic neoplasm (MCN), 29 pancreatic intraductal neoplasia (PanIN) and 8 IPMN-associated PDAC patients. We utilized GWR to model the relationship between epithelial cells and immune cells on a spatial grid. The GWR model estimates were used to generate density signatures which were used in subsequent pairwise classification models to distinguish between any two pairs of disease groups. Image-level, as well as subject-level analysis, were performed. When applied to this dataset, our classification model showed significant discrimination ability in multiple pairwise comparisons, in comparison to commonly used abundance-based metrics, like the Morisita-Horn index. The model was able to best discriminate between CP and PDAC at both the subject- and image-levels. It was also able to reasonably discriminate between PDAC and IPMN. These results point to a potential difference in the spatial arrangement of epithelial and immune cells between CP, PDAC and IPMN, that could be of high diagnostic significance. Further validation on a more comprehensive dataset would be warranted.
Collapse
Affiliation(s)
- Santhoshi N Krishnan
- Department of Electrical and Computer Engineering, Rice University, Houston, TX, USA.
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA.
| | - Shariq Mohammed
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
- Department of Biostatistics, Boston University, Boston, MA, USA
| | | | - Arvind Rao
- Department of Electrical and Computer Engineering, Rice University, Houston, TX, USA.
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA.
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA.
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
21
|
McDonald OG. The biology of pancreatic cancer morphology. Pathology 2022; 54:236-247. [PMID: 34872751 PMCID: PMC8891077 DOI: 10.1016/j.pathol.2021.09.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/09/2021] [Accepted: 09/14/2021] [Indexed: 02/08/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal of all human malignancies. PDAC precursor lesions, invasive primary PDAC, and metastatic PDAC each display distinct morphologies that reflect unique biology. This 'biomorphology' is determined by a complex neoplastic history of clonal phylogenetic relationships, geographic locations, external environmental exposures, intrinsic metabolic demands, and tissue migration patterns. Understanding the biomorphological evolution of PDAC progression is not only of academic interest but also of great practical value. Applying this knowledge to surgical pathology practice facilitates the correct diagnosis on routine H&E stains without additional ancillary studies in most cases. Here I provide a concise overview of the entire biomorphological spectrum of PDAC progression beginning with initial neoplastic transformation and ending in terminal distant metastasis. Most biopsy and resection specimens are currently obtained prior to treatment. As such, our understanding of untreated PDAC biomorphology is mature. The biomorphology of treated PDAC is less defined but will assume greater importance as the frequency of neoadjuvant therapy increases. Although this overview is slanted towards pathology, it is written so that pathologists, clinicians, and scientists alike might find it instructive for their respective disciplines.
Collapse
|
22
|
Sagami R, Sato T, Mizukami K, Motomura M, Okamoto K, Fukuchi S, Otsuka Y, Abe T, Ono H, Mori K, Wada K, Iwaki T, Nishikiori H, Honda K, Amano Y, Murakami K. Diagnostic Strategy of Early Stage Pancreatic Cancer via Clinical Predictor Assessment: Clinical Indicators, Risk Factors and Imaging Findings. Diagnostics (Basel) 2022; 12:diagnostics12020377. [PMID: 35204468 PMCID: PMC8871200 DOI: 10.3390/diagnostics12020377] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 01/27/2022] [Accepted: 01/27/2022] [Indexed: 01/27/2023] Open
Abstract
Early detection of pancreatic ductal adenocarcinoma (PDAC) in the general population is difficult due to unknown clinical characteristics. This study was conducted to clarify the factors associated with early stage PDAC. Well-known symptoms and factors associated with PDAC were classified into clinical indicators, risk factors, and imaging findings concomitant with early stage PDAC. To analyze these factors for the detection of patients with early stage PDAC compared to patients without PDAC, we constructed new diagnostic strategies. The factors of 35 patients with early stage PDAC (stage 0 and IA) and 801 patients without PDAC were compared retrospectively. Clinical indicators; presence and number of indicators, elevated pancreatic enzyme level, tumor biomarker level, acute pancreatitis history, risk factors; familial pancreatic cancer, diabetes mellitus, smoking history, imaging findings; presence and number of findings, and main pancreatic duct dilation were significant factors for early stage PDAC detection. A new screening strategy to select patients who should be examined by imaging modalities from evaluating clinical indicators and risk factors and approaching a definitive diagnosis by evaluating imaging findings had a relatively high sensitivity, specificity, and areas under the curve of 80.0%, 80.8%, and 0.80, respectively. Diagnosis based on the new category and strategy may be reasonable for early stage PDAC detection.
Collapse
Affiliation(s)
- Ryota Sagami
- Department of Gastroenterology, Oita San-ai Medical Center, 1213 Oaza Ichi, Oita 870-1151, Japan; (R.S.); (T.S.); (H.N.)
| | - Takao Sato
- Department of Gastroenterology, Oita San-ai Medical Center, 1213 Oaza Ichi, Oita 870-1151, Japan; (R.S.); (T.S.); (H.N.)
| | - Kazuhiro Mizukami
- Department of Gastroenterology, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasamacho, Yufu 879-5593, Japan; (K.O.); (K.M.); (K.H.); (K.M.)
- Correspondence: ; Tel.: +81-97-586-6193
| | - Mitsuteru Motomura
- Department of Gastroenterology, Oita Red Cross Hospital, 3-2-37 Chiyo-Machi, Oita 870-0033, Japan;
| | - Kazuhisa Okamoto
- Department of Gastroenterology, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasamacho, Yufu 879-5593, Japan; (K.O.); (K.M.); (K.H.); (K.M.)
| | - Satoshi Fukuchi
- Department of Gastroenterology, Oita City Medical Association Almeida Memorial Hospital, 1509-2 Miyazaki, Oita 870-1195, Japan; (S.F.); (K.W.)
| | - Yuichiro Otsuka
- Department of Gastroenterology, Oita Medical Center, 2-11-45 Yokota, Oita 870-0263, Japan;
| | - Takashi Abe
- Department of Gastroenterology, Oita Kouseiren Tsurumi Hospital, 4333 Tsurumi, Beppu 874-8585, Japan;
| | - Hideki Ono
- Department of Gastroenterology, Oita Prefectural Hospital, 2-8-1 Bunyo, Oita 870-8511, Japan;
| | - Kei Mori
- Department of Gastroenterology, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasamacho, Yufu 879-5593, Japan; (K.O.); (K.M.); (K.H.); (K.M.)
| | - Kurato Wada
- Department of Gastroenterology, Oita City Medical Association Almeida Memorial Hospital, 1509-2 Miyazaki, Oita 870-1195, Japan; (S.F.); (K.W.)
| | - Tomoyuki Iwaki
- Department of Endoscopy, Urawa Kyosai Hospital, 3-15-31 Harayama, Saitama 336-0931, Japan; (T.I.); (Y.A.)
| | - Hidefumi Nishikiori
- Department of Gastroenterology, Oita San-ai Medical Center, 1213 Oaza Ichi, Oita 870-1151, Japan; (R.S.); (T.S.); (H.N.)
| | - Koichi Honda
- Department of Gastroenterology, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasamacho, Yufu 879-5593, Japan; (K.O.); (K.M.); (K.H.); (K.M.)
| | - Yuji Amano
- Department of Endoscopy, Urawa Kyosai Hospital, 3-15-31 Harayama, Saitama 336-0931, Japan; (T.I.); (Y.A.)
| | - Kazunari Murakami
- Department of Gastroenterology, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasamacho, Yufu 879-5593, Japan; (K.O.); (K.M.); (K.H.); (K.M.)
| |
Collapse
|
23
|
Hoogenboom SA, Engels MML, Chuprin AV, van Hooft JE, LeGout JD, Wallace MB, Bolan CW. Prevalence, features, and explanations of missed and misinterpreted pancreatic cancer on imaging: a matched case-control study. Abdom Radiol (NY) 2022; 47:4160-4172. [PMID: 36127473 PMCID: PMC9626431 DOI: 10.1007/s00261-022-03671-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 08/28/2022] [Accepted: 08/29/2022] [Indexed: 01/18/2023]
Abstract
PURPOSE To characterize the prevalence of missed pancreatic masses and pancreatic ductal adenocarcinoma (PDAC)-related findings on CT and MRI between pre-diagnostic patients and healthy individuals. MATERIALS AND METHODS Patients diagnosed with PDAC (2010-2016) were retrospectively reviewed for abdominal CT- or MRI-examinations 1 month-3 years prior to their diagnosis, and subsequently matched to controls in a 1:4 ratio. Two blinded radiologists scored each imaging exam on the presence of a pancreatic mass and secondary features of PDAC. Additionally, original radiology reports were graded based on the revised RADPEER criteria. RESULTS The cohort of 595 PDAC patients contained 60 patients with a pre-diagnostic CT and 27 with an MRI. A pancreatic mass was suspected in hindsight on CT in 51.7% and 50% of cases and in 1.3% and 0.9% of controls by reviewer 1 (p < .001) and reviewer 2 (p < .001), respectively. On MRI, a mass was suspected in 70.4% and 55.6% of cases and 2.9% and 0% of the controls by reviewer 1 (p < .001) and reviewer 2 (p < .001), respectively. Pancreatic duct dilation, duct interruption, focal atrophy, and features of acute pancreatitis is strongly associated with PDAC (p < .001). In cases, a RADPEER-score of 2 or 3 was assigned to 56.3% of the CT-reports and 71.4% of MRI-reports. CONCLUSION Radiological features as pancreatic duct dilation and interruption, and focal atrophy are common first signs of PDAC and are often missed or unrecognized. Further investigation with dedicated pancreas imaging is warranted in patients with PDAC-related radiological findings.
Collapse
Affiliation(s)
- Sanne A. Hoogenboom
- Department of Gastroenterology and Hepatology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224 USA ,Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Megan M. L. Engels
- Department of Gastroenterology and Hepatology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224 USA ,Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Anthony V. Chuprin
- Department of Radiology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224 USA
| | - Jeanin E. van Hooft
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Jordan D. LeGout
- Department of Radiology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224 USA
| | - Michael B. Wallace
- Department of Gastroenterology and Hepatology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224 USA ,Department of Gastroenterology and Hepatology, Sheikh Shakhbout Medical City, PO Box 11001, Abu Dhabi, UAE ,Khalifa University School of Medicine, PO Box 127788, Abu Dhabi, UAE
| | - Candice W. Bolan
- Department of Radiology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224 USA
| |
Collapse
|
24
|
Pudakalakatti S, Raj P, Salzillo TC, Enriquez JS, Bourgeois D, Dutta P, Titus M, Shams S, Bhosale P, Kim M, McAllister F, Bhattacharya PK. Metabolic Imaging Using Hyperpolarization for Assessment of Premalignancy. Methods Mol Biol 2022; 2435:169-180. [PMID: 34993946 PMCID: PMC9352438 DOI: 10.1007/978-1-0716-2014-4_12] [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] [Indexed: 06/14/2023]
Abstract
There is an unmet need for noninvasive surrogate markers that can help identify premalignant lesions across different tumor types. Here we describe the methodology and technical details of protocols employed for in vivo 13C pyruvate metabolic imaging experiments. The goal of the method described is to identify and understand metabolic changes, to enable detection of pancreatic premalignant lesions, as a proof of concept of the high sensitivity of this imaging modality.
Collapse
Affiliation(s)
- Shivanand Pudakalakatti
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Priyank Raj
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Travis C Salzillo
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX, USA
| | - José S Enriquez
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Dontrey Bourgeois
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Statistics, Rice University, Houston, TX, USA
| | - Prasanta Dutta
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mark Titus
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shayan Shams
- Department of Biomedical Informatics, University of Texas Health Science Center, Houston, TX, USA
| | - Priya Bhosale
- MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Abdominal Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Kim
- MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Florencia McAllister
- MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pratip K Bhattacharya
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX, USA.
| |
Collapse
|
25
|
Arshavsky-Graham S, Ward SJ, Massad-Ivanir N, Scheper T, Weiss SM, Segal E. Porous Silicon-Based Aptasensors: Toward Cancer Protein Biomarker Detection. ACS MEASUREMENT SCIENCE AU 2021; 1:82-94. [PMID: 34693403 PMCID: PMC8532149 DOI: 10.1021/acsmeasuresciau.1c00019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Indexed: 05/09/2023]
Abstract
The anterior gradient homologue-2 (AGR2) protein is an attractive biomarker for various types of cancer. In pancreatic cancer, it is secreted to the pancreatic juice by premalignant lesions, which would be an ideal stage for diagnosis. Thus, designing assays for the sensitive detection of AGR2 would be highly valuable for the potential early diagnosis of pancreatic and other types of cancer. Herein, we present a biosensor for label-free AGR2 detection and investigate approaches for enhancing the aptasensor sensitivity by accelerating the target mass transfer rate and reducing the system noise. The biosensor is based on a nanostructured porous silicon thin film that is decorated with anti-AGR2 aptamers, where real-time monitoring of the reflectance changes enables the detection and quantification of AGR2, as well as the study of the diffusion and target-aptamer binding kinetics. The aptasensor is highly selective for AGR2 and can detect the protein in simulated pancreatic juice, where its concentration is outnumbered by orders of magnitude by numerous proteins. The aptasensor's analytical performance is characterized with a linear detection range of 0.05-2 mg mL-1, an apparent dissociation constant of 21 ± 1 μM, and a limit of detection of 9.2 μg mL-1 (0.2 μM), which is attributed to mass transfer limitations. To improve the latter, we applied different strategies to increase the diffusion flux to and within the nanostructure, such as the application of isotachophoresis for the preconcentration of AGR2 on the aptasensor, mixing, or integration with microchannels. By combining these approaches with a new signal processing technique that employs Morlet wavelet filtering and phase analysis, we achieve a limit of detection of 15 nM without compromising the biosensor's selectivity and specificity.
Collapse
Affiliation(s)
- Sofia Arshavsky-Graham
- Department
of Biotechnology and Food Engineering, Technion—Israel
Institute of Technology, Haifa 3200003, Israel
- Institute
of Technical Chemistry, Leibniz Universität
Hannover, Callinstraße 5, 30167 Hanover, Germany
| | - Simon J. Ward
- Department
of Electrical Engineering and Computer Science, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Naama Massad-Ivanir
- Department
of Biotechnology and Food Engineering, Technion—Israel
Institute of Technology, Haifa 3200003, Israel
| | - Thomas Scheper
- Institute
of Technical Chemistry, Leibniz Universität
Hannover, Callinstraße 5, 30167 Hanover, Germany
| | - Sharon M. Weiss
- Department
of Electrical Engineering and Computer Science, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Ester Segal
- Department
of Biotechnology and Food Engineering, Technion—Israel
Institute of Technology, Haifa 3200003, Israel
- The
Russell Berrie Nanotechnology Institute, Technion—Israel Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
26
|
Perera CJ, Falasca M, Chari ST, Greenfield JR, Xu Z, Pirola RC, Wilson JS, Apte MV. Role of Pancreatic Stellate Cell-Derived Exosomes in Pancreatic Cancer-Related Diabetes: A Novel Hypothesis. Cancers (Basel) 2021; 13:cancers13205224. [PMID: 34680372 PMCID: PMC8534084 DOI: 10.3390/cancers13205224] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/27/2021] [Accepted: 10/14/2021] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating condition characterised by vague symptomatology and delayed diagnosis. About 30% of PDAC patients report a history of new onset diabetes, usually diagnosed within 3 years prior to the diagnosis of cancer. Thus, new onset diabetes, which is also known as pancreatic cancer-related diabetes (PCRD), could be a harbinger of PDAC. Diabetes is driven by progressive β cell loss/dysfunction and insulin resistance, two key features that are also found in PCRD. Experimental studies suggest that PDAC cell-derived exosomes carry factors that are detrimental to β cell function and insulin sensitivity. However, the role of stromal cells, particularly pancreatic stellate cells (PSCs), in the pathogenesis of PCRD is not known. PSCs are present around the earliest neoplastic lesions and around islets. Given that PSCs interact closely with cancer cells to drive cancer progression, it is possible that exosomal cargo from both cancer cells and PSCs plays a role in modulating β cell function and peripheral insulin resistance. Identification of such mediators may help elucidate the mechanisms of PCRD and aid early detection of PDAC. This paper discusses the concept of a novel role of PSCs in the pathogenesis of PCRD.
Collapse
Affiliation(s)
- Chamini J. Perera
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney 2052, Australia; (C.J.P.); (Z.X.); (R.C.P.); (J.S.W.)
- Ingham Institute for Applied Medical Research, Sydney 2170, Australia
| | - Marco Falasca
- Metabolic Signalling Group, Curtin Health Innovation Research Institute, Curtin Medical School, Curtin University, Perth 6102, Australia;
| | - Suresh T. Chari
- M.D Anderson Cancer Centre, Department of Gastroenterology, Hepatology and Nutrition, University of Texas, Houston, TX 75083, USA;
| | - Jerry R. Greenfield
- St Vincent Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney 2052, Australia;
- Healthy Ageing, Garvan Institute of Medical Research, Darlinghurst 2830, Australia
- Department of Diabetes and Endocrinology, St Vincent’s Hospital, Darlinghurst 3065, Australia
| | - Zhihong Xu
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney 2052, Australia; (C.J.P.); (Z.X.); (R.C.P.); (J.S.W.)
- Ingham Institute for Applied Medical Research, Sydney 2170, Australia
| | - Romano C. Pirola
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney 2052, Australia; (C.J.P.); (Z.X.); (R.C.P.); (J.S.W.)
| | - Jeremy S. Wilson
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney 2052, Australia; (C.J.P.); (Z.X.); (R.C.P.); (J.S.W.)
- Ingham Institute for Applied Medical Research, Sydney 2170, Australia
| | - Minoti V. Apte
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney 2052, Australia; (C.J.P.); (Z.X.); (R.C.P.); (J.S.W.)
- Ingham Institute for Applied Medical Research, Sydney 2170, Australia
- Correspondence: ; Tel.: +61-2-87389029
| |
Collapse
|
27
|
Ying L, Sharma A, Chhoda A, Ruzgar N, Hasan N, Kwak R, Wolfgang CL, Wang TH, Kunstman JW, Salem RR, Wood LD, Iacobuzio-Donahue C, Schneider EB, Farrell JJ, Ahuja N. Methylation-based Cell-free DNA Signature for Early Detection of Pancreatic Cancer. Pancreas 2021; 50:1267-1273. [PMID: 34860810 DOI: 10.1097/mpa.0000000000001919] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES The potential of DNA methylation alterations in early pancreatic cancer (PC) detection among pancreatic tissue cell-free DNA seems promising. This study investigates the diagnostic capacity of the 4-gene methylation biomarker panel, which included ADAMTS1, BNC1, LRFN5, and PXDN genes, in a case-control study. METHODS A genome-wide pharmacoepigenetic approach identified ADAMTS1, BNC1, LRFN5, and PXDN genes as putative targets. Tissue samples including stage I-IV PC (n = 44), pancreatic intraepithelial neoplasia (n = 15), intraductal papillary mucinous neoplasms (n = 24), and normal pancreas (n = 8), and cell-free DNA, which was acquired through methylation on beads technology from PC (n = 22) and control patients (n = 10), were included. The 2-∆ct was the outcome of interest and underwent receiver operating characteristic analysis to determine the diagnostic accuracy of the panel. RESULTS Receiver operating characteristic analysis revealed an area under the curve of 0.93 among ADAMTS1, 0.76 among BNC1, 0.75 among PXDN, and 0.69 among LRFN5 gene. The combination gene methylation panel (ADAMTS1, BNC1, LRFN5, and PXDN) had an area under the curve of 0.94, with a sensitivity of 100% and specificity of 90%. CONCLUSIONS This methylation-based biomarker panel had promising accuracy for PC detection and warranted further validation in prospective PC surveillance trials.
Collapse
Affiliation(s)
| | | | - Ankit Chhoda
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | | | | | | | | | - Tza Huei Wang
- Department of Biomedical Engineering and Department of Mechanical Engineering and Institute for NanoBioTechnology, Johns Hopkins University
| | | | | | - Laura D Wood
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD
| | - Christine Iacobuzio-Donahue
- Human Oncology and Pathogenesis Program, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - James J Farrell
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | | |
Collapse
|
28
|
Gao Y, Wang M, Guo X, Hu J, Chen TM, Finn SMB, Lacy J, Kunstman JW, Cha CH, Bellin MD, Robert ME, Desir GV, Gorelick FS. Renalase is a novel tissue and serological biomarker in pancreatic ductal adenocarcinoma. PLoS One 2021; 16:e0250539. [PMID: 34587190 PMCID: PMC8480607 DOI: 10.1371/journal.pone.0250539] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 08/10/2021] [Indexed: 12/24/2022] Open
Abstract
Dysregulated expression of the secretory protein renalase can promote pancreatic ductal adenocarcinoma (PDAC) growth in animal models. We characterized renalase expression in premalignant and malignant PDAC tissue and investigated whether plasma renalase levels corresponded to clinical PDAC characteristics. Renalase immunohistochemistry was used to determine the presence and distribution of renalase in normal pancreas, chronic pancreatitis, PDAC precursor lesions, and PDAC tissues. Associations between pretreatment plasma renalase and PDAC clinical status were assessed in patients with varied clinical stages of PDAC and included tumor characteristics, surgical resection in locally advanced/borderline resectable PDAC, and overall survival. Data were retrospectively obtained and correlated using non-parametric analysis. Little to no renalase was detected by histochemistry in the normal pancreatic head in the absence of abdominal trauma. In chronic pancreatitis, renalase immunoreactivity localized to peri-acinar spindle-shaped cells in some samples. It was also widely present in PDAC precursor lesions and PDAC tissue. Among 240 patients with PDAC, elevated plasma renalase levels were associated with worse tumor characteristics, including greater angiolymphatic invasion (80.0% vs. 58.1%, p = 0.012) and greater node positive disease (76.5% vs. 56.5%, p = 0.024). Overall survival was worse in patients with high plasma renalase levels with median follow-up of 27.70 months vs. 65.03 months (p < 0.001). Renalase levels also predicted whether patients with locally advanced/borderline resectable PDAC underwent resection (AUC 0.674; 95%CI 0.42-0.82, p = 0.04). Overall tissue renalase was increased in both premalignant and malignant PDAC tissues compared to normal pancreas. Elevated plasma renalase levels were associated with advanced tumor characteristics, decreased overall survival, and reduced resectability in patients with locally advanced/borderline resectable PDAC. These studies show that renalase levels are increased in premalignant pancreatic tissues and that its levels in plasma correspond to the clinical behavior of PDAC.
Collapse
Affiliation(s)
- Yasheen Gao
- Yale University, New Haven, Connecticut, United States of America
- Department of Medicine, Veterans Affairs Connecticut Health System, Yale University School of Medicine, West Haven, Connecticut, United States of America
| | - Melinda Wang
- Department of Medicine, Veterans Affairs Connecticut Health System, Yale University School of Medicine, West Haven, Connecticut, United States of America
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Xiaojia Guo
- Department of Medicine, Veterans Affairs Connecticut Health System, Yale University School of Medicine, West Haven, Connecticut, United States of America
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Joanna Hu
- Yale Cancer Center, New Haven, Connecticut, United States of America
| | - Tian-min Chen
- Department of Medicine, Veterans Affairs Connecticut Health System, Yale University School of Medicine, West Haven, Connecticut, United States of America
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Sade´ M. B. Finn
- Department of Surgery, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Jill Lacy
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - John W. Kunstman
- Department of Surgery, Yale University School of Medicine and VA Connecticut, New Haven, Connecticut, United States of America
| | - Charles H. Cha
- Department of Surgery, Hartford Healthcare Saint Vincent’s Medical Center, Bridgeport, Connecticut, United States of America
| | - Melena D. Bellin
- Department of Surgery, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Marie E. Robert
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Gary V. Desir
- Department of Medicine, Veterans Affairs Connecticut Health System, Yale University School of Medicine, West Haven, Connecticut, United States of America
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Fred S. Gorelick
- Department of Medicine, Veterans Affairs Connecticut Health System, Yale University School of Medicine, West Haven, Connecticut, United States of America
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| |
Collapse
|
29
|
Thompson JK, Bednar F. Clinical Utility of Epigenetic Changes in Pancreatic Adenocarcinoma. EPIGENOMES 2021; 5:20. [PMID: 34968245 PMCID: PMC8715475 DOI: 10.3390/epigenomes5040020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 12/17/2022] Open
Abstract
Pancreatic cancer is a molecularly heterogeneous disease. Epigenetic changes and epigenetic regulatory mechanisms underlie at least some of this heterogeneity and contribute to the evolution of aggressive tumor biology in patients and the tumor's intrinsic resistance to therapy. Here we review our current understanding of epigenetic dysregulation in pancreatic cancer and how it is contributing to our efforts in early diagnosis, predictive and prognostic biomarker development and new therapeutic approaches in this deadly cancer.
Collapse
Affiliation(s)
| | - Filip Bednar
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA;
| |
Collapse
|
30
|
Tonini V, Zanni M. Pancreatic cancer in 2021: What you need to know to win. World J Gastroenterol 2021; 27:5851-5889. [PMID: 34629806 PMCID: PMC8475010 DOI: 10.3748/wjg.v27.i35.5851] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 07/14/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is one of the solid tumors with the worst prognosis. Five-year survival rate is less than 10%. Surgical resection is the only potentially curative treatment, but the tumor is often diagnosed at an advanced stage of the disease and surgery could be performed in a very limited number of patients. Moreover, surgery is still associated with high post-operative morbidity, while other therapies still offer very disappointing results. This article reviews every aspect of pancreatic cancer, focusing on the elements that can improve prognosis. It was written with the aim of describing everything you need to know in 2021 in order to face this difficult challenge.
Collapse
Affiliation(s)
- Valeria Tonini
- Department of Medical Sciences and Surgery, University of Bologna- Emergency Surgery Unit, IRCCS Sant’Orsola Hospital, Bologna 40121, Italy
| | - Manuel Zanni
- University of Bologna, Emergency Surgery Unit, IRCCS Sant'Orsola Hospital, Bologna 40121, Italy
| |
Collapse
|
31
|
Ovarian Metastasis from Pancreatic Ductal Adenocarcinoma. World J Surg 2021; 45:3157-3164. [PMID: 34236477 DOI: 10.1007/s00268-021-06209-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) has a high propensity for systemic dissemination. Ovarian metastases are rare and poorly described. METHODS We identified PDAC cases with ovarian metastasis from a prospectively maintained registry. We reported on the association between outcomes and clinicopathologic factors. Recurrence-free (RFS) and overall survival (OS) were calculated using Kaplan-Meier analysis. RESULTS Twelve patients with PDAC and synchronous or metachronous ovarian metastases were identified. Nine patients (75%) underwent pancreatectomy for localized PDAC and developed metachronous ovarian recurrence. The median OS for all patients was 25.4 (IQR:15.4-82.9) months. For the nine patients with metachronous ovarian metastasis, the median RFS and OS were 14.2 (IQR:7.2-58.3) and 44.6 (IQR:18.6-82.9) months, respectively. Nodal disease, poor grade, vascular invasion in the pancreatic primary, and bilateral ovarian disease tended to confer worse outcomes. CONCLUSION Patients with resected PDAC and ovarian recurrence tend to have a comparable disease course to more common patterns of recurrence. Primaries with nodal disease, poorer grade, vascular invasion, and bilateral ovarian disease were indicative of more aggressive disease biology. The ideal management remains largely unknown, and future collaborative efforts should optimize therapeutic strategies.
Collapse
|
32
|
Ma G, Li G, Xiao Z, Gou A, Xu Y, Song S, Guo K, Liu Z. Narrative review of intraductal papillary mucinous neoplasms: pathogenesis, diagnosis, and treatment of a true precancerous lesion. Gland Surg 2021; 10:2313-2324. [PMID: 34422602 PMCID: PMC8340339 DOI: 10.21037/gs-21-450] [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: 05/25/2021] [Accepted: 07/14/2021] [Indexed: 12/09/2022]
Abstract
OBJECTIVE Although considerable progress has been made in our understanding of intraductal papillary mucinous neoplasm (IPMN) of the pancreas, there are still some problems to be solved. BACKGROUND IPMN is one of the most important precancerous lesions of pancreatic cancer, but the relationship between IPMN and pancreatic cancer, and the specific mechanism of the development from IPMN to invasive carcinoma, remain to be explored in depth. With the development of imaging, the detection rate of IPMN has been greatly improved. However, the degree of malignancy of IPMN is difficult to assess, and its classification criteria and surgical treatment strategies are still controversial. Therefore, there is an urgent need for the best treatment plan for IPMN and research that can better predict IPMN recurrence and tumor malignancy. METHODS From the online database Web of Science (https://webofknowledge.com/) and PubMed (https://pubmed.ncbi.nlm.nih.gov/), we use specific retrieval strategies to retrieve relevant articles based on the topics we discussed, and we review and discuss them. CONCLUSIONS This paper discusses the related research and progress of IPMN in recent years to improve the understanding of the incidence, diagnosis, treatment, and prognosis of this disease. The follow-up and monitoring of IPMN is particularly important, but the specific strategy also remains controversial.
Collapse
Affiliation(s)
- Gang Ma
- Department of Pancreatic-Biliary Surgery, First Hospital of China Medical University, Shenyang, China
| | - Guichen Li
- Department of Pancreatic-Biliary Surgery, First Hospital of China Medical University, Shenyang, China
| | - Zhihuan Xiao
- Department of Pancreatic-Biliary Surgery, First Hospital of China Medical University, Shenyang, China
| | - Anjiang Gou
- Department of Pancreatic-Biliary Surgery, First Hospital of China Medical University, Shenyang, China
| | - Yuanhong Xu
- Department of Pancreatic-Biliary Surgery, First Hospital of China Medical University, Shenyang, China
| | - Shaowei Song
- Department of Pancreatic-Biliary Surgery, First Hospital of China Medical University, Shenyang, China
| | - Kejian Guo
- Department of Pancreatic-Biliary Surgery, First Hospital of China Medical University, Shenyang, China
| | - Zhe Liu
- Department of Pancreatic-Biliary Surgery, First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
33
|
Enriquez JS, Chu Y, Pudakalakatti S, Hsieh KL, Salmon D, Dutta P, Millward NZ, Lurie E, Millward S, McAllister F, Maitra A, Sen S, Killary A, Zhang J, Jiang X, Bhattacharya PK, Shams S. Hyperpolarized Magnetic Resonance and Artificial Intelligence: Frontiers of Imaging in Pancreatic Cancer. JMIR Med Inform 2021; 9:e26601. [PMID: 34137725 PMCID: PMC8277399 DOI: 10.2196/26601] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/24/2021] [Accepted: 04/03/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND There is an unmet need for noninvasive imaging markers that can help identify the aggressive subtype(s) of pancreatic ductal adenocarcinoma (PDAC) at diagnosis and at an earlier time point, and evaluate the efficacy of therapy prior to tumor reduction. In the past few years, there have been two major developments with potential for a significant impact in establishing imaging biomarkers for PDAC and pancreatic cancer premalignancy: (1) hyperpolarized metabolic (HP)-magnetic resonance (MR), which increases the sensitivity of conventional MR by over 10,000-fold, enabling real-time metabolic measurements; and (2) applications of artificial intelligence (AI). OBJECTIVE Our objective of this review was to discuss these two exciting but independent developments (HP-MR and AI) in the realm of PDAC imaging and detection from the available literature to date. METHODS A systematic review following the PRISMA extension for Scoping Reviews (PRISMA-ScR) guidelines was performed. Studies addressing the utilization of HP-MR and/or AI for early detection, assessment of aggressiveness, and interrogating the early efficacy of therapy in patients with PDAC cited in recent clinical guidelines were extracted from the PubMed and Google Scholar databases. The studies were reviewed following predefined exclusion and inclusion criteria, and grouped based on the utilization of HP-MR and/or AI in PDAC diagnosis. RESULTS Part of the goal of this review was to highlight the knowledge gap of early detection in pancreatic cancer by any imaging modality, and to emphasize how AI and HP-MR can address this critical gap. We reviewed every paper published on HP-MR applications in PDAC, including six preclinical studies and one clinical trial. We also reviewed several HP-MR-related articles describing new probes with many functional applications in PDAC. On the AI side, we reviewed all existing papers that met our inclusion criteria on AI applications for evaluating computed tomography (CT) and MR images in PDAC. With the emergence of AI and its unique capability to learn across multimodal data, along with sensitive metabolic imaging using HP-MR, this knowledge gap in PDAC can be adequately addressed. CT is an accessible and widespread imaging modality worldwide as it is affordable; because of this reason alone, most of the data discussed are based on CT imaging datasets. Although there were relatively few MR-related papers included in this review, we believe that with rapid adoption of MR imaging and HP-MR, more clinical data on pancreatic cancer imaging will be available in the near future. CONCLUSIONS Integration of AI, HP-MR, and multimodal imaging information in pancreatic cancer may lead to the development of real-time biomarkers of early detection, assessing aggressiveness, and interrogating early efficacy of therapy in PDAC.
Collapse
Affiliation(s)
- José S Enriquez
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Yan Chu
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Shivanand Pudakalakatti
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Kang Lin Hsieh
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Duncan Salmon
- Department of Electrical and Computer Engineering, Rice University, Houston, TX, United States
| | - Prasanta Dutta
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Niki Zacharias Millward
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Urology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Eugene Lurie
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Steven Millward
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Florencia McAllister
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Anirban Maitra
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Subrata Sen
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ann Killary
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jian Zhang
- Division of Computer Science and Engineering, Louisiana State University, Baton Rouge, LA, United States
| | - Xiaoqian Jiang
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Pratip K Bhattacharya
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Shayan Shams
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
34
|
Malhotra A, Rachet B, Bonaventure A, Pereira SP, Woods LM. Can we screen for pancreatic cancer? Identifying a sub-population of patients at high risk of subsequent diagnosis using machine learning techniques applied to primary care data. PLoS One 2021; 16:e0251876. [PMID: 34077433 PMCID: PMC8171946 DOI: 10.1371/journal.pone.0251876] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 05/04/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Pancreatic cancer (PC) represents a substantial public health burden. Pancreatic cancer patients have very low survival due to the difficulty of identifying cancers early when the tumour is localised to the site of origin and treatable. Recent progress has been made in identifying biomarkers for PC in the blood and urine, but these cannot be used for population-based screening as this would be prohibitively expensive and potentially harmful. METHODS We conducted a case-control study using prospectively-collected electronic health records from primary care individually-linked to cancer registrations. Our cases were comprised of 1,139 patients, aged 15-99 years, diagnosed with pancreatic cancer between January 1, 2005 and June 30, 2009. Each case was age-, sex- and diagnosis time-matched to four non-pancreatic (cancer patient) controls. Disease and prescription codes for the 24 months prior to diagnosis were used to identify 57 individual symptoms. Using a machine learning approach, we trained a logistic regression model on 75% of the data to predict patients who later developed PC and tested the model's performance on the remaining 25%. RESULTS We were able to identify 41.3% of patients < = 60 years at 'high risk' of developing pancreatic cancer up to 20 months prior to diagnosis with 72.5% sensitivity, 59% specificity and, 66% AUC. 43.2% of patients >60 years were similarly identified at 17 months, with 65% sensitivity, 57% specificity and, 61% AUC. We estimate that combining our algorithm with currently available biomarker tests could result in 30 older and 400 younger patients per cancer being identified as 'potential patients', and the earlier diagnosis of around 60% of tumours. CONCLUSION After further work this approach could be applied in the primary care setting and has the potential to be used alongside a non-invasive biomarker test to increase earlier diagnosis. This would result in a greater number of patients surviving this devastating disease.
Collapse
Affiliation(s)
- Ananya Malhotra
- Faculty of Epidemiology and Population Health, Department of Non-Communicable Disease Epidemiology, Inequalities in Cancer Outcomes Network, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Bernard Rachet
- Faculty of Epidemiology and Population Health, Department of Non-Communicable Disease Epidemiology, Inequalities in Cancer Outcomes Network, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Audrey Bonaventure
- Epidemiology of Childhood and Adolescent Cancers Team, CRESS, Université de Paris-INSERM, Villejuif, France
| | - Stephen P. Pereira
- UCL Institute for Liver and Digestive Health, University College London, London, United Kingdom
| | - Laura M. Woods
- Faculty of Epidemiology and Population Health, Department of Non-Communicable Disease Epidemiology, Inequalities in Cancer Outcomes Network, London School of Hygiene & Tropical Medicine, London, United Kingdom
| |
Collapse
|
35
|
Preoperative differentiation of serous cystic neoplasms from mucin-producing pancreatic cystic neoplasms using a CT-based radiomics nomogram. Abdom Radiol (NY) 2021; 46:2637-2646. [PMID: 33558952 DOI: 10.1007/s00261-021-02954-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/05/2021] [Accepted: 01/13/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE To develop and validate a CT-based radiomics nomogram in preoperative differential diagnosis of SCNs from mucin-producing PCNs. MATERIAL AND METHODS A total of 89 patients consisting of 31 SCNs, 30 IPMNs, and 28 MCNs who underwent preoperative CT were analyzed. A total of 710 radiomics features were extracted from each case. Patients were divided into training (n = 63) and validation cohorts (n = 26) with a ratio of 7:3. Least absolute shrinkage and selection operator (LASSO) method and logistic regression analysis were used for feature selection and model construction. A nomogram was created from a comprehensive model consisting of clinical features and the fusion radiomics signature. A decision curve analysis was used for clinical decisions. RESULTS The radiomics features extracted from CT could assist with the differentiation of SCNs from mucin-producing PCNs in both the training and validation cohorts. The signature of the combination of the plain, late arterial, and venous phases had the largest areas under the curve (AUCs) of 0.960 (95% CI 0.910-1) in the training cohort and 0.817 (95% CI 0.651-0.983) in the validation cohort with good calibration. The value and efficacy of the nomogram was verified using decision curve analysis. CONCLUSION A comprehensive nomogram incorporating clinical features and fusion radiomics signature can differentiate SCNs from mucin-producing PCNs.
Collapse
|
36
|
Fujikura K, Alruwaii ZI, Haffner MC, Trujillo MA, Roberts NJ, Hong SM, Macgregor-Das A, Goggins MG, Roy S, Meeker AK, Ding D, Wright M, He J, Hruban RH, Wood LD. Downregulation of 5-hydroxymethylcytosine is an early event in pancreatic tumorigenesis. J Pathol 2021; 254:279-288. [PMID: 33870509 DOI: 10.1002/path.5682] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/09/2021] [Accepted: 04/16/2021] [Indexed: 12/18/2022]
Abstract
Epigenetic alterations are increasingly recognized as important contributors to the development and progression of pancreatic ductal adenocarcinoma. 5-hydroxymethylcytosine (5hmC) is an epigenetic DNA mark generated through the ten-eleven translocation (TET) enzyme-mediated pathway and is closely linked to gene activation. However, the timing of alterations in epigenetic regulation in the progression of pancreatic neoplasia is not well understood. In this study, we hypothesized that aberrant expression of ten-eleven translocation methylcytosine dioxygenase 1 (TET1) and subsequent global 5hmC alteration are linked to early tumorigenesis in the pancreas. Therefore, we evaluated alterations of 5hmC and TET1 levels using immunohistochemistry in pancreatic neoplasms (n = 380) and normal ducts (n = 118). The study cohort included representation of the full spectrum of precancerous lesions from low- and high-grade pancreatic intraepithelial neoplasia (n = 95), intraductal papillary mucinous neoplasms (all subtypes, n = 129), intraductal oncocytic papillary neoplasms (n = 12), and mucinous cystic neoplasms (n = 144). 5hmC and TET1 were significantly downregulated in all types of precancerous lesion and associated invasive pancreatic ductal adenocarcinomas compared with normal ductal epithelium (all p < 0.001), and expression of 5hmC positively correlated with expression of TET1. Importantly, downregulation of both 5hmC and TET1 was observed in most low-grade precancerous lesions. There were no clear associations between 5hmC levels and clinicopathological factors, thereby suggesting a common epigenetic abnormality across precancerous lesions. We conclude that downregulation of 5hmC and TET1 is an early event in pancreatic tumorigenesis. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Kohei Fujikura
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zainab I Alruwaii
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael C Haffner
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Maria A Trujillo
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicholas J Roberts
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Seung-Mo Hong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Anne Macgregor-Das
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael G Goggins
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sujayita Roy
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alan K Meeker
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ding Ding
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael Wright
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jin He
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ralph H Hruban
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Laura D Wood
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
37
|
Abstract
OBJECTIVES Early detection of pancreatic cancer is notoriously difficult. A novel cancer diagnostic method using the ability of nematodes to detect odor of urine samples has been developed (N-NOSE). This method has a high sensitivity and specificity for various cancers; however, it has not yet been verified in pancreatic cancer. We examined the usefulness of this method to aid early diagnosis of pancreatic cancer in a cancer center. METHODS We collected urine samples and clinical data from patients hospitalized in our division, between July 2017 and February 2019. We excluded patients with a known current or past history of other cancers. We investigated the relationship between the results of N-NOSE and the presence of pancreatic cancer. RESULTS There were 95 noncancer cases and 104 pancreatic cancer cases. The sensitivity and specificity of N-NOSE for pancreatic cancer were 84.6% (88/104) and 60% (57/95), respectively. N-NOSE was able to detect stages 0 to I pancreatic cancer and had a higher correlation with early-stage pancreatic cancer than advanced stage. CONCLUSIONS N-NOSE has sufficient sensitivity and specificity for use in clinical practice, and it holds great potential as a diagnostic aid for pancreatic cancer, especially for early-stage pancreatic cancer.
Collapse
|
38
|
Clinical Perspective on Proteomic and Glycomic Biomarkers for Diagnosis, Prognosis, and Prediction of Pancreatic Cancer. Int J Mol Sci 2021; 22:ijms22052655. [PMID: 33800786 PMCID: PMC7961509 DOI: 10.3390/ijms22052655] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/26/2021] [Accepted: 03/02/2021] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is known as a highly aggressive malignant disease. Prognosis for patients is notoriously poor, despite improvements in surgical techniques and new (neo)adjuvant chemotherapy regimens. Early detection of PDAC may increase the overall survival. It is furthermore foreseen that precision medicine will provide improved prognostic stratification and prediction of therapeutic response. In this review, omics-based discovery efforts are presented that aim for novel diagnostic and prognostic biomarkers of PDAC. For this purpose, we systematically evaluated the literature published between 1999 and 2020 with a focus on protein- and protein-glycosylation biomarkers in pancreatic cancer patients. Besides genomic and transcriptomic approaches, mass spectrometry (MS)-based proteomics and glycomics of blood- and tissue-derived samples from PDAC patients have yielded new candidates with biomarker potential. However, for reasons discussed in this review, the validation and clinical translation of these candidate markers has not been successful. Consequently, there has been a change of mindset from initial efforts to identify new unimarkers into the current hypothesis that a combination of biomarkers better suits a diagnostic or prognostic panel. With continuing development of current research methods and available techniques combined with careful study designs, new biomarkers could contribute to improved detection, prognosis, and prediction of pancreatic cancer.
Collapse
|
39
|
Caldwell KE, Conway AP, Hammill CW. Screening for Pancreatic Ductal Adenocarcinoma: Are We Asking the Impossible? Cancer Prev Res (Phila) 2021; 14:373-382. [PMID: 33148677 PMCID: PMC8089111 DOI: 10.1158/1940-6207.capr-20-0426] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/30/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023]
Abstract
Pancreatic cancer is projected to become the second leading cause of cancer-related death in the United States by 2020. Because of this, significant interest and research funding has been devoted to development of a screening test to identify individuals during a prolonged asymptomatic period; however, to date, no such test has been developed. We evaluated current NIH spending and clinical trials to determine the focus of research on pancreatic cancer screening as compared with other cancer subtypes. Using statistical methodology, we determined the effects of population-based pancreatic cancer screening on overall population morbidity and mortality. Population-based pancreatic cancer screening would result in significant harm to non-diseased individuals, even in cases where a near-perfect test was developed. Despite this mathematical improbability, NIH funding for pancreatic cancer demonstrates bias toward screening test development not seen in other cancer subtypes. Focusing research energy on development of pancreatic screening tests is unlikely to result in overall survival benefits. Efforts to increase the number of patients who are candidates for surgery and improving surgical outcomes would result in greater population benefit.Prevention Relevance: For patients with pancreatic cancer, early stage detection offers the greatest survival benefit. However, the incidence of pancreatic cancer and associated mortality of pancreatic resections make development of a screening test a difficult, if not impossible, challenge.
Collapse
Affiliation(s)
| | - Alexander P Conway
- Department of Surgery, Washington University in St. Louis, St. Louis, Missouri
| | - Chet W Hammill
- Department of Surgery, Washington University in St. Louis, St. Louis, Missouri.
| |
Collapse
|
40
|
Sagami R, Yamao K, Nakahodo J, Minami R, Tsurusaki M, Murakami K, Amano Y. Pre-Operative Imaging and Pathological Diagnosis of Localized High-Grade Pancreatic Intra-Epithelial Neoplasia without Invasive Carcinoma. Cancers (Basel) 2021; 13:cancers13050945. [PMID: 33668239 PMCID: PMC7956417 DOI: 10.3390/cancers13050945] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/08/2021] [Accepted: 02/19/2021] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) arises from precursor lesions, such as pancreatic intra-epithelial neoplasia (PanIN) and intraductal papillary mucinous neoplasm (IPMN). The prognosis of high-grade precancerous lesions, including high-grade PanIN and high-grade IPMN, without invasive carcinoma is good, despite the overall poor prognosis of PDAC. High-grade PanIN, as a lesion preceding invasive PDAC, is therefore a primary target for intervention. However, detection of localized high-grade PanIN is difficult when using standard radiological approaches. Therefore, most studies of high-grade PanIN have been conducted using specimens that harbor invasive PDAC. Recently, imaging characteristics of high-grade PanIN have been revealed. Obstruction of the pancreatic duct due to high-grade PanIN may induce a loss of acinar cells replaced by fibrosis and lobular parenchymal atrophy. These changes and additional inflammation around the branch pancreatic ducts (BPDs) result in main pancreatic duct (MPD) stenosis, dilation, retention cysts (BPD dilation), focal pancreatic parenchymal atrophy, and/or hypoechoic changes around the MPD. These indirect imaging findings have become important clues for localized, high-grade PanIN detection. To obtain pre-operative histopathological confirmation of suspected cases, serial pancreatic-juice aspiration cytologic examination is effective. In this review, we outline current knowledge on imaging characteristics of high-grade PanIN.
Collapse
Affiliation(s)
- Ryota Sagami
- Department of Gastroenterology, Oita San-ai Medical Center, 1213 Oaza Ichi, Oita, Oita 870-1151, Japan
- Pancreatic Cancer Research for Secure Salvage Young Investigators (PASSYON), Osaka-Sayama, Osaka 589-8511, Japan; (K.Y.); (J.N.); (R.M.)
- Correspondence: ; Tel.: +81-97-541-1311; Fax: +81-97-541-5218
| | - Kentaro Yamao
- Pancreatic Cancer Research for Secure Salvage Young Investigators (PASSYON), Osaka-Sayama, Osaka 589-8511, Japan; (K.Y.); (J.N.); (R.M.)
- Department of Gastroenterology and Hepatology, Kindai University, Osaka-Sayama, Osaka 589-8511, Japan
| | - Jun Nakahodo
- Pancreatic Cancer Research for Secure Salvage Young Investigators (PASSYON), Osaka-Sayama, Osaka 589-8511, Japan; (K.Y.); (J.N.); (R.M.)
- Department of Gastroenterology Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo 113-8677, Japan
| | - Ryuki Minami
- Pancreatic Cancer Research for Secure Salvage Young Investigators (PASSYON), Osaka-Sayama, Osaka 589-8511, Japan; (K.Y.); (J.N.); (R.M.)
- Department of Gastroenterology, Tenri Hospital, 200 Mishimacho, Tenri, Nara 632-0015, Japan
| | - Masakatsu Tsurusaki
- Department of Diagnostic Radiology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan;
| | - Kazunari Murakami
- Department of Gastroenterology, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasamacho, Yufu, Oita 879-5593, Japan;
| | - Yuji Amano
- Department of Endoscopy, Urawa Kyosai Hospital, 3-15-31 Harayama, Midoriku, Saitama 336-0931, Japan;
| |
Collapse
|
41
|
Bushnell GG, Orbach SM, Ma JA, Crawford HC, Wicha MS, Jeruss JS, Shea LD. Disease-induced immunomodulation at biomaterial scaffolds detects early pancreatic cancer in a spontaneous model. Biomaterials 2020; 269:120632. [PMID: 33418200 DOI: 10.1016/j.biomaterials.2020.120632] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/17/2020] [Accepted: 12/20/2020] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer has the worst prognosis of all cancers due to disease aggressiveness and paucity of early detection platforms. We developed biomaterial scaffolds that recruit metastatic tumor cells and reflect the immune dysregulation of native metastatic sites. While this platform has shown promise in orthotopic breast cancer models, its potential in other models is untested. Herein, we demonstrate that scaffolds recruit disseminated pancreatic cells in the KPCY model of spontaneous pancreatic cancer prior to adenocarcinoma formation (3-fold increase in scaffold YFP + cells). Furthermore, immune cells at the scaffolds differentiate early- and late-stage disease with greater accuracy (0.83) than the natural metastatic site (liver, 0.50). Early disease was identified by an approximately 2-fold increase in monocytes. Late-stage disease was marked by a 1.5-2-fold increase in T cells and natural killer cells. The differential immune response indicated that the scaffolds could distinguish spontaneous pancreatic cancer from spontaneous breast cancer. Collectively, our findings demonstrate the utility of scaffolds to reflect immunomodulation in two spontaneous models of tumorigenesis, and their particular utility for identifying early disease stages in the aggressive KPCY pancreatic cancer model. Such scaffolds may serve as a platform for early detection of pancreatic cancer to improve treatment and prognosis.
Collapse
Affiliation(s)
- Grace G Bushnell
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Sophia M Orbach
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jeffrey A Ma
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Howard C Crawford
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Max S Wicha
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jacqueline S Jeruss
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Surgery, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
42
|
Faias S, Cravo M, Pereira da Silva J, Chaves P, Dias Pereira A. Endoscopic ultrasound with fine needle aspiration is useful in pancreatic cysts smaller than 3 cm. BMC Gastroenterol 2020; 20:413. [PMID: 33297971 PMCID: PMC7727209 DOI: 10.1186/s12876-020-01565-9] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 11/30/2020] [Indexed: 12/28/2022] Open
Abstract
Background In current guidelines, endoscopic ultrasound with fine-needle aspiration (EUS-FNA) is recommended in pancreatic cystic lesions (PCLs) with worrisome features (size ≥ 3 cm, mural nodule, or Wirsung dilation).
Objective To evaluate the diagnostic ability and assess the accuracy of EUS-FNA in PCLs smaller than 3 cm. Methods Retrospective study of PCLs < 3 cm (2007–2016) undergoing EUS-FNA. Clinical, EUS and pancreatic cystic fluid (PCF) data were prospectively registered. Performance of EUS-FNA with PCF analysis for the detection of malignancy and accuracy in surgical cohort were analyzed.
Results We evaluated 115 patients with PCLs < 3 cm who underwent EUS-FNA. 19 patients underwent surgery, 7 had malignant, 8 pre-malignant, and the remaining 4 benign lesions. Mass/mural nodule was present in 27% of the cysts, CEA level was higher than 192 ng/mL in 39.4% of patients, and only 35% of cytologic samples were informative. Nevertheless, additional FNA for PCF analysis improved the diagnostic performance of EUS imaging—AUC = 0.80 versus AUC = 60. Conclusion EUS-FNA has good accuracy in PCLs < 3 cm. It confirmed malignancy even in lesions without worrisome features (nodule/mass), with two in every five resections showing high-risk/malignant lesions. EUS-FNA was also useful to diagnose benign cysts, possibly allowing surveillance to be stopped in one in every five patients.
Collapse
Affiliation(s)
- Sandra Faias
- Gastroenterology Department, Instituto Português de Oncologia de Lisboa Francisco Gentil EPE, Rua Prof Lima Basto, 1099-023, Lisbon, Portugal. .,Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal.
| | - Marília Cravo
- Gastroenterology Department, Hospital Beatriz Ângelo, Av. Carlos Teixeira, 3, 2670-000, Loures, Portugal.,Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - João Pereira da Silva
- Gastroenterology Department, Instituto Português de Oncologia de Lisboa Francisco Gentil EPE, Rua Prof Lima Basto, 1099-023, Lisbon, Portugal
| | - Paula Chaves
- Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal.,Pathology Department, Instituto Português de Oncologia de Lisboa Francisco Gentil EPE, Rua Prof Lima Basto, 1099-023, Lisbon, Portugal
| | - A Dias Pereira
- Gastroenterology Department, Instituto Português de Oncologia de Lisboa Francisco Gentil EPE, Rua Prof Lima Basto, 1099-023, Lisbon, Portugal
| |
Collapse
|
43
|
Liu Y, Kaur S, Huang Y, Fahrmann JF, Rinaudo JA, Hanash SM, Batra SK, Singhi AD, Brand RE, Maitra A, Haab BB. Biomarkers and Strategy to Detect Preinvasive and Early Pancreatic Cancer: State of the Field and the Impact of the EDRN. Cancer Epidemiol Biomarkers Prev 2020; 29:2513-2523. [PMID: 32532830 PMCID: PMC7710622 DOI: 10.1158/1055-9965.epi-20-0161] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/20/2020] [Accepted: 06/05/2020] [Indexed: 12/19/2022] Open
Abstract
Patients afflicted with pancreatic ductal adenocarcinoma (PDAC) face a dismal prognosis, but headway could be made if physicians could identify the disease earlier. A compelling strategy to broaden the use of surveillance for PDAC is to incorporate molecular biomarkers in combination with clinical analysis and imaging tools. This article summarizes the components involved in accomplishing biomarker validation and an analysis of the requirements of molecular biomarkers for disease surveillance. We highlight the significance of consortia for this research and highlight resources and infrastructure of the Early Detection Research Network (EDRN). The EDRN brings together the multifaceted expertise and resources needed for biomarker validation, such as study design, clinical care, biospecimen collection and handling, molecular technologies, and biostatistical analysis, and studies coming out of the EDRN have yielded biomarkers that are moving forward in validation. We close the article with an overview of the current investigational biomarkers, an analysis of their performance relative to the established benchmarks, and an outlook on the current needs in the field. The outlook for improving the early detection of PDAC looks promising, and the pace of further research should be quickened through the resources and expertise of the EDRN and other consortia.See all articles in this CEBP Focus section, "NCI Early Detection Research Network: Making Cancer Detection Possible."
Collapse
Affiliation(s)
- Ying Liu
- Van Andel Institute, Grand Rapids, Michigan
| | | | - Ying Huang
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Johannes F Fahrmann
- Sheikh Ahmed Center for Pancreatic Cancer Research, MD Anderson Cancer Center, Houston, Texas
| | - Jo Ann Rinaudo
- Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland
| | - Samir M Hanash
- Sheikh Ahmed Center for Pancreatic Cancer Research, MD Anderson Cancer Center, Houston, Texas
| | | | - Aatur D Singhi
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Randall E Brand
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Anirban Maitra
- Sheikh Ahmed Center for Pancreatic Cancer Research, MD Anderson Cancer Center, Houston, Texas
| | | |
Collapse
|
44
|
Yang KS, Lin HY, Curley C, Welch MW, Wolpin B, Lee H, Weissleder R, Im H, Castro CM. Bead-Based Extracellular Vesicle Analysis Using Flow Cytometry. ADVANCED BIOSYSTEMS 2020; 4:e2000203. [PMID: 33103361 PMCID: PMC7718389 DOI: 10.1002/adbi.202000203] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/08/2020] [Indexed: 01/09/2023]
Abstract
Extracellular vesicles (EVs) represent promising circulating biomarkers for cancers, but their high-throughput analyses in clinical settings prove challenging due to lack of simple, fast, and robust EV assays. Here, a bead-based EV assay detected by flow cytometry is described, which integrates EV capture using microbeads with EV protein analyses by flow cytometry. The assay is fast (<4 h for 48 samples), robust, and compatible with conventional flow cytometry instruments for high-throughput EV analysis. With the method, a panel of pancreatic cancer biomarkers in EVs from plasma samples of pancreatic cancer patients is successfully analyzed. The assay is readily translatable to other biomarkers or cancer types and can be run with standard materials on conventional flow cytometers, making it highly flexible and adaptable to diverse research and clinical needs.
Collapse
Affiliation(s)
- Katherine S. Yang
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Hsing-Ying Lin
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Caleigh Curley
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | | | - Hakho Lee
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Hyungsoon Im
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Cesar M. Castro
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114, USA
- Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA
| |
Collapse
|
45
|
Early Diagnosis of Pancreatic Cancer: The Key for Survival. Diagnostics (Basel) 2020; 10:diagnostics10110869. [PMID: 33114412 PMCID: PMC7694042 DOI: 10.3390/diagnostics10110869] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 10/16/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer (PC) is one of the most aggressive forms of cancer. Negative prognosis is mainly due to the late diagnosis in advanced stages, when the disease is already therapeutically overcome. Studies in recent years have focused on identifying biomarkers that could play a role in early diagnosis, leading to the improvement of morbidity and mortality. Currently, the only biomarker widely used in the diagnosis of PC is carbohydrate antigen 19-9 (CA19.9), which has, however, more of a prognostic role in the follow-up of postoperative recurrence than a diagnostic role. Other biomarkers, recently identified as the methylation status of ADAMTS1 (A disintegrin and metalloproteinase with thrombospondin motifs 1) and BNC1 (zinc finger protein basonuclin-1) in cell-free deoxyribonucleic acid (DNA), may play a role in the early detection of PC. This review focuses on the diagnosis of PC in its early stages.
Collapse
|
46
|
Gao C, Wisniewski L, Liu Y, Staal B, Beddows I, Plenker D, Aldakkak M, Hall J, Barnett D, Gouda MK, Allen P, Drake R, Zureikat A, Huang Y, Evans D, Singhi A, Brand RE, Tuveson DA, Tsai S, Haab BB. Detection of Chemotherapy-resistant Pancreatic Cancer Using a Glycan Biomarker, sTRA. Clin Cancer Res 2020; 27:226-236. [PMID: 33093149 DOI: 10.1158/1078-0432.ccr-20-2475] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/17/2020] [Accepted: 10/13/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE A subset of pancreatic ductal adenocarcinomas (PDACs) is highly resistant to systemic chemotherapy, but no markers are available in clinical settings to identify this subset. We hypothesized that a glycan biomarker for PDACs called sialylated tumor-related antigen (sTRA) could be used for this purpose. EXPERIMENTAL DESIGN We tested for differences between PDACs classified by glycan expression in multiple systems: sets of cell lines, organoids, and isogenic cell lines; primary tumors; and blood plasma from human subjects. RESULTS The sTRA-expressing models tended to have stem-like gene expression and the capacity for mesenchymal differentiation, in contrast to the nonexpressing models. The sTRA cell lines also had significantly increased resistance to seven different chemotherapeutics commonly used against pancreatic cancer. Patients with primary tumors that were positive for a gene expression classifier for sTRA received no statistically significant benefit from adjuvant chemotherapy, in contrast to those negative for the signature. In another cohort, based on direct measurements of sTRA in tissue microarrays, the patients who were high in sTRA again had no statistically significant benefit from adjuvant chemotherapy. Furthermore, a blood plasma test for the sTRA glycan identified the PDACs that showed rapid relapse following neoadjuvant chemotherapy. CONCLUSIONS This research demonstrates that a glycan biomarker could have value to detect chemotherapy-resistant PDAC in clinical settings. This capability could aid in the development of stratified treatment plans and facilitate biomarker-guided trials targeting resistant PDAC.
Collapse
Affiliation(s)
- ChongFeng Gao
- Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, Michigan
| | - Luke Wisniewski
- Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, Michigan
| | - Ying Liu
- Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, Michigan
| | - Ben Staal
- Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, Michigan
| | - Ian Beddows
- Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, Michigan
| | - Dennis Plenker
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Mohammed Aldakkak
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Johnathan Hall
- Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, Michigan
| | - Daniel Barnett
- Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, Michigan
| | - Mirna Kheir Gouda
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Peter Allen
- Division of Surgical Oncology, Duke University School of Medicine, Durham, North Carolina
| | - Richard Drake
- Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina
| | - Amer Zureikat
- Division of Gastrointestinal Surgical Oncology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Ying Huang
- Division of Vaccine and Infectious Disease, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Douglas Evans
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Aatur Singhi
- Division of Gastrointestinal Surgical Oncology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Randall E Brand
- Division of Gastrointestinal Surgical Oncology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - David A Tuveson
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Susan Tsai
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Brian B Haab
- Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, Michigan.
| |
Collapse
|
47
|
Role of extracellular vesicles in tumour microenvironment. Cell Commun Signal 2020; 18:163. [PMID: 33081785 PMCID: PMC7574205 DOI: 10.1186/s12964-020-00643-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 08/10/2020] [Indexed: 12/16/2022] Open
Abstract
In recent years, it has been demonstrated that extracellular vesicles (EVs) can be released by almost all cell types, and detected in most body fluids. In the tumour microenvironment (TME), EVs serve as a transport medium for lipids, proteins, and nucleic acids. EVs participate in various steps involved in the development and progression of malignant tumours by initiating or suppressing various signalling pathways in recipient cells. Although tumour-derived EVs (T-EVs) are known for orchestrating tumour progression via systemic pathways, EVs from non-malignant cells (nmEVs) also contribute substantially to malignant tumour development. Tumour cells and non-malignant cells typically communicate with each other, both determining the progress of the disease. In this review, we summarise the features of both T-EVs and nmEVs, tumour progression, metastasis, and EV-mediated chemoresistance in the TME. The physiological and pathological effects involved include but are not limited to angiogenesis, epithelial-mesenchymal transition (EMT), extracellular matrix (ECM) remodelling, and immune escape. We discuss potential future directions of the clinical application of EVs, including diagnosis (as non-invasive biomarkers via liquid biopsy) and therapeutic treatment. This may include disrupting EV biogenesis and function, thus utilising the features of EVs to repurpose them as a therapeutic tool in immunotherapy and drug delivery systems. We also discuss the overall findings of current studies, identify some outstanding issues requiring resolution, and propose some potential directions for future research. Video abstract.
Collapse
|
48
|
Non-coding RNA biomarkers in pancreatic ductal adenocarcinoma. Semin Cancer Biol 2020; 75:153-168. [PMID: 33049362 DOI: 10.1016/j.semcancer.2020.10.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/20/2020] [Accepted: 10/02/2020] [Indexed: 12/13/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies, which is usually diagnosed at an advanced stage. The late disease diagnosis, the limited availability of effective therapeutic interventions and lack of robust diagnostic biomarkers, are some of the primary reasons for the dismal 5-year survival rates (∼8%) in patients with PDAC. The pancreatic cancer develops through accumulation of a series of genomic and epigenomic alterations which lead to the transformation of normal pancreatic epithelium into an invasive carcinoma - a process that can take up to 15-20 years to develop, from the occurrence of first initiating mutational event. These facts highlight a unique window of opportunity for the earlier detection of PDAC, which could allow timely disease interception and improvement in the overall survival outcomes in patients suffering from this fatal malignancy. Non-coding RNAs (ncRNAs) have been recognized to play a central role in PDAC pathogenesis and are emerging as attractive candidates for biomarker development in various cancers, including PDAC. More specifically, the ncRNAs play a pivotal role in PDAC biology as they affect tumor growth, migration, and invasion by regulating cellular processes including cell cycle, apoptosis, and epithelial-mesenchymal transition. In this review, we focus on three types of well-established ncRNAs - microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) - and discuss their potential as diagnostic, prognostic and predictive biomarkers in PDAC.
Collapse
|
49
|
Haeri HH, Tomaszewski J, Phytides B, Schimm H, Möslein G, Niedergethmann M, Hinderberger D, Gelos M. Identification of Patients with Pancreatic Cancer by Electron Paramagnetic Resonance Spectroscopy of Fatty Acid Binding to Human Serum Albumin. ACS Pharmacol Transl Sci 2020; 3:1188-1198. [PMID: 33344896 DOI: 10.1021/acsptsci.0c00116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Indexed: 12/11/2022]
Abstract
An effective biological marker for pancreatic adenocarcinoma (PAC) is not available so far. Here, we investigate how electron paramagnetic resonance (EPR) spectroscopy of spin-labeled fatty acid (FA) molecules binding to human serum albumin (HSA) in human serum is a suitable method for the identification of patients with PAC through detection of PAC-induced changes of FA binding to albumin. The functionality of HSA to bind FA is investigated in serum samples of 35 patients with PAC, 26 patients with benign pancreatic tumors (BPD), and 24 healthy individuals by continuous wave (CW) EPR spectroscopy by simply dissolving 16-DOXYL stearic acid as spin-labeled FA. It is found that FA binding to HSA in PAC is significantly modified when compared with healthy and BPD individuals. The PAC group could best be discriminated from the healthy group based on EPR characteristics at the loading ratio of 1:4 (HSA:FA), while patients with PAC and BPD are distinguishable at a loading ratio of 1:6. Using nanoscale distance measurements through double electron-electron resonance (DEER), it is found that the distribution of FAs in the HSA of one PAC patient is similar to that of FAs in healthy individuals. Combining all EPR spectroscopic data, this leads to a tentative molecular interpretation of only small changes in hydration at the protein's surface as origin of the detectable characteristics for PAC patients. Thus, EPR of FA/HSA binding is a simple and promising tool for clinical detection of patients with PAC and needs to be tested with larger ensembles of different patient groups.
Collapse
Affiliation(s)
- Haleh H Haeri
- Institute of Chemistry, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Jörg Tomaszewski
- Department of General and Visceral Surgery, Alfried Krupp Krankenhaus Essen, Essen, Nordrhein-Westfalen 45276, Germany
| | - Bettina Phytides
- Department of General and Visceral Surgery, Alfried Krupp Krankenhaus Essen, Essen, Nordrhein-Westfalen 45276, Germany
| | - Heike Schimm
- Institute of Chemistry, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Gabriela Möslein
- Faculty of Health Sciences, University of Witten/Herdecke, Witten, Nordrhein-Westfalen 58448, Germany
| | - Marco Niedergethmann
- Department of General and Visceral Surgery, Alfried Krupp Krankenhaus Essen, Essen, Nordrhein-Westfalen 45276, Germany
| | - Dariush Hinderberger
- Institute of Chemistry, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Marcos Gelos
- Faculty of Health Sciences, University of Witten/Herdecke, Witten, Nordrhein-Westfalen 58448, Germany
| |
Collapse
|
50
|
Faias S, Pereira L, Fonseca R, Chaves P, Dias Pereira A, Cravo M. A second endoscopic ultrasound with fine-needle aspiration for cytology identifies high-risk pancreatic cysts overlooked by current guidelines. Diagn Cytopathol 2020; 49:109-118. [PMID: 32960508 DOI: 10.1002/dc.24607] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 07/22/2020] [Accepted: 08/17/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Endoscopic ultrasound with fine-needle aspiration (EUS-FNA) is recommended for diagnosis of pancreatic cystic lesions (PCLs). Its role in surveillance is unclear. Our goal was to determine if a second EUS-FNA changes diagnosis or management of PCLs. METHODS A retrospective analysis of an EUS database, searching for EUS-FNAs in PCLs from 2007 to 2017 was performed. Demographics, cyst characteristics, and FNA results were compared in patients under surveillance, performing a single or two consecutive EUS-FNAs. RESULTS Of 203 PCLs referred for EUS-FNA, surveillance was decided in 128 (63%). Data of 105 (82%) patients with a single EUS-FNA were compared with 23 (18%) with two EUS-FNAs during surveillance. Patients were younger in this latter group (P = .055), whereas CEA levels were marginally higher (P = .078) and a mass/nodule were more frequent (P = .006). The mean time between EUS-FNAs was 38 months (4.7-118.8) for 18 patients maintaining surveillance vs 18 months (2.9-56.9) in the four referred for surgery (P = NS) after two EUS-FNAs (two NETs, one IPMN-HGD, and one MCN-LG). A high correlation in CEA level between consecutive EUS-FNAs (r2 = 0.945, P < .01) was present, with a change of category observed (cut-off level = 192 ng/mL) in two patients only. Of four patients with a second EUS-FNA with conclusive cytology, two had NETs confirmed on resection. CONCLUSIONS Repeating EUS-FNA in surveillance of PCLs with clinical suspicion of malignancy increased neoplasm diagnoses, changing decision toward surgery in almost 20% of patients while excluding IPMNs with mucin nodules from unnecessary resections. A second EUS-FNA for cytology appears justified in some PCLs, particularly for diagnosing NETs.
Collapse
Affiliation(s)
- Sandra Faias
- Gastroenterology Department, Instituto Português de Oncologia de Lisboa Francisco Gentil, EPE, Lisbon, Portugal.,Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Luisa Pereira
- Center of Mathematics and Applications (CMA-UBI), University of Beira Interior, Covilhã, Portugal
| | - Ricardo Fonseca
- Pathology Department, Instituto Português de Oncologia de Lisboa Francisco Gentil, EPE, Lisbon, Portugal.,Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Paula Chaves
- Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal.,Pathology Department, Instituto Português de Oncologia de Lisboa Francisco Gentil, EPE, Lisbon, Portugal
| | - António Dias Pereira
- Gastroenterology Department, Instituto Português de Oncologia de Lisboa Francisco Gentil, EPE, Lisbon, Portugal
| | - Marília Cravo
- Faculty of Medicine, University of Lisbon, Lisbon, Portugal.,Gastroenterology Department, Hospital Beatriz Ângelo, Loures, Portugal
| |
Collapse
|