1
|
Digiacomo L, Caputo D, Cammarata R, La Vaccara V, Coppola R, Quagliarini E, Iacobini M, Renzi S, Giulimondi F, Pozzi D, Caracciolo G, Amenitsch H. Nanoparticle-protein corona enhances accuracy of Ca-19.9-based pancreatic cancer classification. NANOSCALE 2025; 17:7066-7075. [PMID: 39868525 DOI: 10.1039/d4nr02435d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Among the various types of pancreatic cancers, pancreatic ductal adenocarcinoma (PDAC) is the most lethal and aggressive, due to its tendency to metastasize quickly and has a particularly low five-year survival rate. Carbohydrate antigen 19-9 (CA 19-9) is the only biomarker approved by the Food and Drug Administration for PDAC and has been a focal point in diagnostic strategies, but its sensitivity and specificity are not sufficient for early and accurate detection. To address this issue, we introduce a synergistic approach combining CA 19-9 levels with a graphene oxide (GO)-based blood test. This non-invasive technique relies on the analysis of personalized protein corona formed on GO sheets once they are embedded in human plasma. Pairing CA 19-9 values with GO protein patterns from N = 106 donors significantly improved the ability to differentiate between non-oncological and PDAC patients (up to 92%), also boosting the classification of PDAC subjects by 50% compared to CA 19-9 testing alone. Overall, this study sought to bridge the existing gaps in PDAC detection by exploiting the complementary strengths of conventional biomarkers and cutting-edge nanotechnology. Exploration of this combined strategy holds promise for advancing the early detection of PDAC, ultimately contributing to improved patient prognosis and treatment outcomes.
Collapse
Affiliation(s)
- Luca Digiacomo
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena, 291, 00161 Rome, Italy.
| | - Damiano Caputo
- Research Unit of General Surgery, Department of Medicine and Surgery, University Campus Bio-Medico di Roma, Rome, Italy
- Operative Research Unit of General Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Roberto Cammarata
- Operative Research Unit of General Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Vincenzo La Vaccara
- Operative Research Unit of General Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Roberto Coppola
- Operative Research Unit of General Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Erica Quagliarini
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena, 291, 00161 Rome, Italy.
| | - Manuela Iacobini
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena, 291, 00161 Rome, Italy.
| | - Serena Renzi
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena, 291, 00161 Rome, Italy.
| | - Francesca Giulimondi
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena, 291, 00161 Rome, Italy.
| | - Daniela Pozzi
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena, 291, 00161 Rome, Italy.
| | - Giulio Caracciolo
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena, 291, 00161 Rome, Italy.
| | - Heinz Amenitsch
- Institute of Inorganic Chemistry, Graz University of Technology, 8010 Graz, Austria
| |
Collapse
|
2
|
Cox M, Vitello D, Chawla A. Translating the multifaceted use of liquid biopsy to management of early disease in pancreatic adenocarcinoma. Front Oncol 2025; 15:1520717. [PMID: 40182037 PMCID: PMC11966063 DOI: 10.3389/fonc.2025.1520717] [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: 10/31/2024] [Accepted: 02/25/2025] [Indexed: 04/05/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a leading cause of cancer-related mortality, primarily due to late stage at diagnosis. This review examines the multifaceted applications of liquid biopsy and circulating tumor DNA (ctDNA) analysis in the diagnosis and management of PDAC. We review the current literature on the technological advancements in liquid biopsy analysis such as next generation sequencing (NGS) and digital droplet PCR (ddPCR) as well as multi-omics technologies, highlighting their potential for accurate molecular subtyping through ctDNA analysis. This review highlights the significant role of ctDNA in the assessment of tumor behavior, disease subtyping, prediction and monitoring of treatment response, and evaluation of minimal residual disease. We discuss the implications of integrating liquid biopsy techniques into clinical practice as well as its challenges and limitations. By drawing insights from recent studies, this review aims to provide a comprehensive overview of how liquid biopsy and ctDNA analysis can enhance early disease management strategies in PDAC. We underscore the need for additional prospective studies and clinical trials to validate its feasibility and accuracy in order to establish clinical utility, with the ultimate goal of routine incorporation into practice to improve patient outcomes and transform the treatment landscape for PDAC.
Collapse
Affiliation(s)
- Madison Cox
- Division of Surgical Oncology, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Northwestern Medicine Cancer Centers, Northwestern Medicine Regional Medical Group, Winfield, IL, United States
| | - Dominic Vitello
- Division of Surgical Oncology, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Northwestern Quality Improvement, Research and Education in Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Akhil Chawla
- Division of Surgical Oncology, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Northwestern Medicine Cancer Centers, Northwestern Medicine Regional Medical Group, Winfield, IL, United States
- Northwestern Quality Improvement, Research and Education in Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, United States
| |
Collapse
|
3
|
Hewitt DB, Wolfgang CL. The Role of Surgery in "Oligometastatic" Pancreas Cancer. Surg Clin North Am 2024; 104:1065-1081. [PMID: 39237164 DOI: 10.1016/j.suc.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
The majority of patients diagnosed with pancreatic cancer already have metastatic disease at the time of presentation, which results in a 5-year survival rate of only 13%. However, multiagent chemotherapy regimens can stabilize the disease in select patients with limited metastatic disease. For such patients, a combination of curative-intent therapy and systemic therapy may potentially enhance outcomes compared to using systemic therapy alone. Of note, the evidence supporting this approach is primarily derived from retrospective studies and may carry a significant selection bias. Looking ahead, ongoing prospective trials are exploring the efficacy of curative-intent therapy in managing oligometastatic pancreatic cancer and the implementation of treatment strategies based on specific biomarkers. The emergence of these trials, coupled with the development of less invasive therapeutic modalities, provides hope for patients with oligometastatic pancreatic cancer.
Collapse
Affiliation(s)
- D Brock Hewitt
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The NYU Grossman School of Medicine, 577 1st Avenue, 2nd Floor, New York, NY 10016, USA.
| | - Christopher L Wolfgang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The NYU Grossman School of Medicine, 577 1st Avenue, 2nd Floor, New York, NY 10016, USA
| |
Collapse
|
4
|
Freed IM, Kasi A, Fateru O, Hu M, Gonzalez P, Weatherington N, Pathak H, Hyter S, Sun W, Al-Rajabi R, Baranda J, Hupert ML, Chalise P, Godwin AK, A. Witek M, Soper SA. Circulating Tumor Cell Subpopulations Predict Treatment Outcome in Pancreatic Ductal Adenocarcinoma (PDAC) Patients. Cells 2023; 12:2266. [PMID: 37759489 PMCID: PMC10526802 DOI: 10.3390/cells12182266] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/06/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
There is a high clinical unmet need to improve outcomes for pancreatic ductal adenocarcinoma (PDAC) patients, either with the discovery of new therapies or biomarkers that can track response to treatment more efficiently than imaging. We report an innovative approach that will generate renewed interest in using circulating tumor cells (CTCs) to monitor treatment efficacy, which, in this case, used PDAC patients receiving an exploratory new therapy, poly ADP-ribose polymerase inhibitor (PARPi)-niraparib-as a case study. CTCs were enumerated from whole blood using a microfluidic approach that affinity captures epithelial and mesenchymal CTCs using anti-EpCAM and anti-FAPα monoclonal antibodies, respectively. These antibodies were poised on the surface of two separate microfluidic devices to discretely capture each subpopulation for interrogation. The isolated CTCs were enumerated using immunophenotyping to produce a numerical ratio consisting of the number of mesenchymal to epithelial CTCs (denoted "Φ"), which was used as an indicator of response to therapy, as determined using computed tomography (CT). A decreasing value of Φ during treatment was indicative of tumor response to the PARPi and was observed in 88% of the enrolled patients (n = 31). Changes in Φ during longitudinal testing were a better predictor of treatment response than the current standard CA19-9. We were able to differentiate between responders and non-responders using ΔΦ (p = 0.0093) with higher confidence than CA19-9 (p = 0.033). For CA19-9 non-producers, ΔΦ correctly predicted the outcome in 72% of the PDAC patients. Sequencing of the gDNA extracted from affinity-selected CTC subpopulations provided information that could be used for patient enrollment into the clinical trial based on their tumor mutational status in DNA repair genes.
Collapse
Affiliation(s)
- Ian M. Freed
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA; (I.M.F.); (O.F.); (M.H.); (P.G.); (N.W.); (M.A.W.)
- Center of Bio-Modular Multiscale Systems for Precision Medicine (CBM), The University of Kansas, Lawrence, KS 66047, USA;
| | - Anup Kasi
- Division of Medical Oncology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (W.S.); (R.A.-R.); (J.B.); (P.C.)
| | - Oluwadamilola Fateru
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA; (I.M.F.); (O.F.); (M.H.); (P.G.); (N.W.); (M.A.W.)
- Center of Bio-Modular Multiscale Systems for Precision Medicine (CBM), The University of Kansas, Lawrence, KS 66047, USA;
| | - Mengjia Hu
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA; (I.M.F.); (O.F.); (M.H.); (P.G.); (N.W.); (M.A.W.)
- Center of Bio-Modular Multiscale Systems for Precision Medicine (CBM), The University of Kansas, Lawrence, KS 66047, USA;
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.P.); (S.H.)
- Department of Cancer Biology, The University of Kansas Medical Center, Cancer Center, Kansas City, KS 66160, USA
| | - Phasin Gonzalez
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA; (I.M.F.); (O.F.); (M.H.); (P.G.); (N.W.); (M.A.W.)
- Center of Bio-Modular Multiscale Systems for Precision Medicine (CBM), The University of Kansas, Lawrence, KS 66047, USA;
| | - Nyla Weatherington
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA; (I.M.F.); (O.F.); (M.H.); (P.G.); (N.W.); (M.A.W.)
- Center of Bio-Modular Multiscale Systems for Precision Medicine (CBM), The University of Kansas, Lawrence, KS 66047, USA;
| | - Harsh Pathak
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.P.); (S.H.)
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Stephen Hyter
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.P.); (S.H.)
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Weijing Sun
- Division of Medical Oncology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (W.S.); (R.A.-R.); (J.B.); (P.C.)
| | - Raed Al-Rajabi
- Division of Medical Oncology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (W.S.); (R.A.-R.); (J.B.); (P.C.)
| | - Joaquina Baranda
- Division of Medical Oncology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (W.S.); (R.A.-R.); (J.B.); (P.C.)
| | | | - Prabhakar Chalise
- Division of Medical Oncology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (W.S.); (R.A.-R.); (J.B.); (P.C.)
| | - Andrew K. Godwin
- Center of Bio-Modular Multiscale Systems for Precision Medicine (CBM), The University of Kansas, Lawrence, KS 66047, USA;
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.P.); (S.H.)
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Malgorzata A. Witek
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA; (I.M.F.); (O.F.); (M.H.); (P.G.); (N.W.); (M.A.W.)
- Center of Bio-Modular Multiscale Systems for Precision Medicine (CBM), The University of Kansas, Lawrence, KS 66047, USA;
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.P.); (S.H.)
| | - Steven A. Soper
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA; (I.M.F.); (O.F.); (M.H.); (P.G.); (N.W.); (M.A.W.)
- Center of Bio-Modular Multiscale Systems for Precision Medicine (CBM), The University of Kansas, Lawrence, KS 66047, USA;
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.P.); (S.H.)
- Department of Cancer Biology, The University of Kansas Medical Center, Cancer Center, Kansas City, KS 66160, USA
- BioFluidica, Inc., San Diego, CA 92121, USA;
- Bioengineering Program, The University of Kansas, Lawrence, KS 66045, USA
- Department of Mechanical Engineering, The University of Kansas, Lawrence, KS 66045, USA
| |
Collapse
|
5
|
Digiacomo L, Quagliarini E, Pozzi D, Coppola R, Caracciolo G, Caputo D. Stratifying Risk for Pancreatic Cancer by Multiplexed Blood Test. Cancers (Basel) 2023; 15:2983. [PMID: 37296945 PMCID: PMC10251844 DOI: 10.3390/cancers15112983] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/20/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal disease, for which mortality closely parallels incidence. So far, the available techniques for PDAC detection are either too invasive or not sensitive enough. To overcome this limitation, here we present a multiplexed point-of-care test that provides a "risk score" for each subject under investigation, by combining systemic inflammatory response biomarkers, standard laboratory tests, and the most recent nanoparticle-enabled blood (NEB) tests. The former parameters are routinely evaluated in clinical practice, whereas NEB tests have been recently proven as promising tools to assist in PDAC diagnosis. Our results revealed that PDAC patients and healthy subjects can be distinguished accurately (i.e., 88.9% specificity, 93.6% sensitivity) by the presented multiplexed point-of-care test, in a quick, non-invasive, and highly cost-efficient way. Furthermore, the test allows for the definition of a "risk threshold", which can help clinicians to trace the optimal diagnostic and therapeutic care pathway for each patient. For these reasons, we envision that this work may accelerate progress in the early detection of PDAC and contribute to the design of screening programs for high-risk populations.
Collapse
Affiliation(s)
- Luca Digiacomo
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena, 291, 00161 Rome, Italy; (L.D.); (E.Q.); (D.P.)
| | - Erica Quagliarini
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena, 291, 00161 Rome, Italy; (L.D.); (E.Q.); (D.P.)
| | - Daniela Pozzi
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena, 291, 00161 Rome, Italy; (L.D.); (E.Q.); (D.P.)
| | - Roberto Coppola
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy;
- Research Unit of Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Rome, Italy
| | - Giulio Caracciolo
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena, 291, 00161 Rome, Italy; (L.D.); (E.Q.); (D.P.)
| | - Damiano Caputo
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy;
- Research Unit of Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Rome, Italy
| |
Collapse
|
6
|
Kung H, Yu J. Targeted therapy for pancreatic ductal adenocarcinoma: Mechanisms and clinical study. MedComm (Beijing) 2023; 4:e216. [PMID: 36814688 PMCID: PMC9939368 DOI: 10.1002/mco2.216] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 02/21/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive and lethal malignancy with a high rate of recurrence and a dismal 5-year survival rate. Contributing to the poor prognosis of PDAC is the lack of early detection, a complex network of signaling pathways and molecular mechanisms, a dense and desmoplastic stroma, and an immunosuppressive tumor microenvironment. A recent shift toward a neoadjuvant approach to treating PDAC has been sparked by the numerous benefits neoadjuvant therapy (NAT) has to offer compared with upfront surgery. However, certain aspects of NAT against PDAC, including the optimal regimen, the use of radiotherapy, and the selection of patients that would benefit from NAT, have yet to be fully elucidated. This review describes the major signaling pathways and molecular mechanisms involved in PDAC initiation and progression in addition to the immunosuppressive tumor microenvironment of PDAC. We then review current guidelines, ongoing research, and future research directions on the use of NAT based on randomized clinical trials and other studies. Finally, the current use of and research regarding targeted therapy for PDAC are examined. This review bridges the molecular understanding of PDAC with its clinical significance, development of novel therapies, and shifting directions in treatment paradigm.
Collapse
Affiliation(s)
- Heng‐Chung Kung
- Krieger School of Arts and SciencesJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Jun Yu
- Departments of Medicine and OncologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
7
|
Law J, Trieu H, Kaleka G, Turkiewicz J, Palmer S, Lee JM, Chen KT, Tabibian JH. Clinical Features and Outcomes of Patients with Pancreaticobiliary Malignancies in Los Angeles County and Their Association with CA 19-9 Levels. Cancers (Basel) 2023; 15:1723. [PMID: 36980609 PMCID: PMC10046349 DOI: 10.3390/cancers15061723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/23/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023] Open
Abstract
UNLABELLED Although CA 19-9 is a commonly used tumor marker in the management of PBMs, the literature describing outcomes in patients with PBMs who have undetectable or low (hereinafter "low") CA 19-9 levels remains scarce. In this study, we sought to compare clinical features and outcomes in patients with PBMs and low CA 19-9 levels to those with normal and elevated CA 19-9 levels. METHODS We retrospectively collected data on patients with biopsy-confirmed PBMs and stratified patients into categories based on their CA 19-9 level at diagnosis. Survival curves were estimated for patients in each of the three aforementioned CA 19-9 groups using the Kaplan-Meier method and compared using a Cox proportional hazards regression model. RESULTS Of the 283 patients identified, 23 (8.1%) had low, 70 (24.7%) had normal, and 190 (67.1%) had elevated CA 19-9 levels. After controlling for sex, age, BMI, the presence of metastases at the time of diagnosis, and treatment with curative intent, the hazard ratio for death in the elevated CA 19-9 group compared to the low CA 19-9 group was 1.993 (95% CI 1.089-3.648; p = 0.025). CONCLUSION The elevated CA 19-9 level compared to the low CA 19-9 level and the presence of metastases were associated with an increased hazard of death, while treatment with curative intent was associated with a decreased hazard of death.
Collapse
Affiliation(s)
- Jade Law
- LAC-USC Hematology and Oncology Fellowship Program, Los Angeles, CA 90033, USA
| | - Harry Trieu
- LAC-USC Internal Medicine Residency Program, Los Angeles, CA 90033, USA
| | - Guneet Kaleka
- UCLA-Olive View Internal Medicine Residency Program, Sylmar, CA 91342, USA
| | - Joanna Turkiewicz
- UCLA-Olive View Internal Medicine Residency Program, Sylmar, CA 91342, USA
| | - Samantha Palmer
- UCLA-Olive View Internal Medicine Residency Program, Sylmar, CA 91342, USA
| | - Jennifer M. Lee
- Harbor-UCLA Medical Center, Division of Hematology and Medical Oncology, Torrance, CA 90502, USA
| | - Kathryn T. Chen
- Harbor-UCLA Medical Center, Division of Surgical Oncology, Torrance, CA 90502, USA
| | - James H. Tabibian
- UCLA-Olive View, Division of Gastroenterology, Sylmar, CA 91342, USA
- UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
8
|
Pérez-Ginés V, Torrente-Rodríguez RM, Pedrero M, Martínez-Bosch N, de Frutos PG, Navarro P, Pingarrón JM, Campuzano S. Electrochemical immunoplatform to help managing pancreatic cancer. J Electroanal Chem (Lausanne) 2023. [DOI: 10.1016/j.jelechem.2023.117312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2023]
|
9
|
Pekarek L, Fraile-Martinez O, Garcia-Montero C, Saez MA, Barquero-Pozanco I, Del Hierro-Marlasca L, de Castro Martinez P, Romero-Bazán A, Alvarez-Mon MA, Monserrat J, García-Honduvilla N, Buján J, Alvarez-Mon M, Guijarro LG, Ortega MA. Clinical Applications of Classical and Novel Biological Markers of Pancreatic Cancer. Cancers (Basel) 2022; 14:1866. [PMID: 35454771 PMCID: PMC9029823 DOI: 10.3390/cancers14081866] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/02/2022] [Accepted: 04/06/2022] [Indexed: 01/27/2023] Open
Abstract
The incidence and prevalence of pancreatic adenocarcinoma have increased in recent years. Pancreatic cancer is the seventh leading cause of cancer death, but it is projected to become the second leading cause of cancer-related mortality by 2040. Most patients are diagnosed in an advanced stage of the disease, with very limited 5-year survival. The discovery of different tissue markers has elucidated the underlying pathophysiology of pancreatic adenocarcinoma and allowed stratification of patient risk at different stages and assessment of tumour recurrence. Due to the invasive capacity of this tumour and the absence of screening markers, new immunohistochemical and serological markers may be used as prognostic markers for recurrence and in the study of possible new therapeutic targets because the survival of these patients is low in most cases. The present article reviews the currently used main histopathological and serological markers and discusses the main characteristics of markers under development.
Collapse
Affiliation(s)
- Leonel Pekarek
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Oncology Service, Guadalajara University Hospital, 19002 Guadalajara, Spain
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Cielo Garcia-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Miguel A Saez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Pathological Anatomy Service, Central University Hospital of Defence-UAH Madrid, 28801 Alcala de Henares, Spain
| | - Ines Barquero-Pozanco
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| | - Laura Del Hierro-Marlasca
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| | - Patricia de Castro Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| | - Adoración Romero-Bazán
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| | - Miguel A Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Jorge Monserrat
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Julia Buján
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine (CIBEREHD), University Hospital Príncipe de Asturias, 28806 Alcala de Henares, Spain
| | - Luis G Guijarro
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Unit of Biochemistry and Molecular Biology, Department of System Biology (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain
| | - Miguel A Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Cancer Registry and Pathology Department, Principe de Asturias University Hospital, 28806 Alcala de Henares, Spain
| |
Collapse
|
10
|
Yong BJC, Wirama Diyana M. Low Carbohydrate Antigen 19-9 (CA 19-9) Levels in a Patient Highly Suspected of Having Caput Pancreas Tumor. Cureus 2022; 14:e24357. [PMID: 35611029 PMCID: PMC9124065 DOI: 10.7759/cureus.24357] [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] [Accepted: 04/21/2022] [Indexed: 11/30/2022] Open
Abstract
Pancreatic cancer is the seventh leading cause of cancer-related mortality worldwide and the eleventh most common cause of cancer-related death in Indonesia. In pancreatic cancer, rapid and early diagnosis is crucial. Carbohydrate antigen 19-9 (CA 19-9), the most sensitive and specific tumor marker for pancreatic cancer, may help in diagnosing and determining prognosis; however, some populations do not express CA 19-9. Cases of low CA 19-9 may occur in populations with Lewis⍺-β- genotype. These populations are not able to express Lewis antigen and CA 19-9; therefore, CA 19-9 investigation cannot be used for diagnostic and therapeutic measures. In patients highly suspicious of pancreatic cancer where CA 19-9 levels are low, alternative tumor markers such as CA 125 and carcinoembryonic antigen or a combination of various tumor markers can be used to increase sensitivity and specificity in diagnosing pancreatic cancer. A 70-year-old man presented with a complaint of worsening abdominal pain for the last two days. The patient had dark-yellow urine and pale stool. Abdominal ultrasonography and computed tomography scan showed a mass on the head of the pancreas, which was highly suspicious of pancreatic cancer.
Collapse
|