1
|
Bonazzi VF, Aoude LG, Brosda S, Bradford JJ, Lonie JM, Loffler KA, Gartside MG, Patel K, Mukhopadhyay P, Keane C, Gebski V, Kench JG, Goldstein D, Waddell N, Barbour AP. C-reactive protein is a prognostic biomarker in pancreatic ductal adenocarcinoma patients. Asia Pac J Clin Oncol 2025; 21:77-86. [PMID: 37415393 PMCID: PMC11733851 DOI: 10.1111/ajco.13993] [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: 12/01/2021] [Revised: 04/04/2023] [Accepted: 06/26/2023] [Indexed: 07/08/2023]
Abstract
AIM The 5-year survival rate of pancreatic ductal adenocarcinoma (PDAC) is approximately 11% and has only improved marginally over the last three decades. For operable PDAC, resection and adjuvant FOLFIRINOX chemotherapy is standard of care. There is growing interest in perioperative regimens to improve outcomes. The non-randomized Phase II study "Gemcitabine and Abraxane for resectable Pancreatic cancer" (GAP) demonstrated the feasibility of perioperative gemcitabine/abraxane. Long-term survival in PDAC requires an effective immune response; hence, we undertook this translational study of the GAP trial cohort to identify immune-oncology biomarkers for clinical use. METHODS We combined Nanostring nCounter technology with immunohistochemistry to investigate the correlation between gene expression and overall patient survival. Findings were investigated in samples from the International Cancer Genome Consortium (ICGC, n = 88) and the Australian Pancreatic Genome Initiative (APGI, n = 227). RESULTS We confirmed that human equilibrative nucleoside transporter 1 (hENT1) expression was not a prognostic marker in PDAC but patients with high levels of hENT1 were more likely to live longer than 24 months post-surgery. Additionally, CD274 (PD-L1) and two novel biomarkers of survival, cathepsin W (CTSW) and C-reactive protein (CRP), were identified in the GAP cohort (n = 19). CRP expression was confirmed in data from the ICGC. Although PD-L1 and CTSW proteins were not significant across all three cohorts, results show that low CRP mRNA and protein expression are associated with longer overall survival in all three patient groups. CONCLUSION PDAC patients with long survival have higher hENT1 expression levels. Furthermore, CRP expression is a biomarker of poor prognosis following perioperative chemotherapy and resection in PDAC patients and thus may be useful for identifying patients who could benefit from more aggressive adjuvant strategies.
Collapse
Affiliation(s)
- Vanessa F. Bonazzi
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Lauren G. Aoude
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Sandra Brosda
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Julia J. Bradford
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - James M. Lonie
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Kelly A. Loffler
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
- College of Medicine and Public HealthFlinders UniversityBedford ParkSouth AustraliaAustralia
| | - Michael G. Gartside
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Kalpana Patel
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
| | | | - Colm Keane
- Mater Research Institute‐UQSouth BrisbaneQueenslandAustralia
| | - Val Gebski
- NHMRC Clinical Trials CentreCamperdownNew South WalesAustralia
| | - James G. Kench
- Royal Prince Alfred HospitalCamperdownNew South WalesAustralia
- University of Sydney Central Clinical SchoolCamperdownNew South WalesAustralia
| | - David Goldstein
- University of NSW Prince of Wales Clinical SchoolRandwickNew South WalesAustralia
| | - Nicola Waddell
- QIMRBerghofer Medical Research InstituteHerstonQueenslandAustralia
| | - Andrew P. Barbour
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
- Princess Alexandra HospitalWoolloongabbaQueenslandAustralia
| | | |
Collapse
|
2
|
Jin Y, Hu R, Gu Y, Wei A, Li A, Zhang Y. Quantitative site-specific N-glycosylation analysis reveals IgG glyco-signatures for pancreatic cancer diagnosis. Clin Proteomics 2024; 21:68. [PMID: 39734184 DOI: 10.1186/s12014-024-09522-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/16/2024] [Indexed: 12/31/2024] Open
Abstract
BACKGROUND Pancreatic cancer is a highly aggressive tumor with a poor prognosis due to a low early detection rate and a lack of biomarkers. Most of pancreatic cancer is pancreatic ductal adenocarcinoma (PDAC). Alterations in the N-glycosylation of plasma immunoglobulin G (IgG) have been shown to be closely associated with the onset and development of several cancers and could be used as biomarkers for diagnosis. The study aimed to explore intact N-glycosylation profile of IgG in patients with PDAC and find relation between intact N-glycosylation profile of IgG and clinical information such as diagnosis and prognosis. METHODS In this study, we employed a well-evaluated approach (termed GlycoQuant) to assess the site-specific N-glycosylation profile of human plasma IgG in both healthy individuals and patients with pancreatic ductal adenocarcinoma (PDAC). The datasets generated and analyzed during the current study are available in the ProteomeXchange Consortium ( http://www.proteomexchange.org/ ) via the iProX partner repository, with the dataset identifier PXD051436. RESULTS The analysis of rapidly purified IgG samples from 100 patients with different stages of PDAC, in addition to 30 healthy controls, revealed that the combination of carbohydrate antigen 19 - 9 (CA19-9), IgG1-GP05 (IgG1-TKPREEQYNSTYR-HexNAc [4]Hex [5]Fuc [1]NeuAc [1]), and IgG4-GP04 (IgG4-EEQFNSTYR- HexNAc [4]Hex [5]Fuc [1]NeuAc [1]) can be used to distinguish between PDAC patients and healthy individuals (AUC = 0.988). In addition, cross validation of the diagnosis model showed satisfactory result. CONCLUSIONS The study demonstrated that the integrated quantitative method can be utilized for large-scale clinical N-glycosylation research to identify novel N-glycosylated biomarkers. This could facilitate the development of clinical glycoproteomics.
Collapse
Affiliation(s)
- Yi Jin
- Department of Pancreatic Surgery and Institutes for Systems Genetics, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, Sichuan, 610041, China
| | - Ran Hu
- Department of Pancreatic Surgery and Institutes for Systems Genetics, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, Sichuan, 610041, China
| | - Yufan Gu
- Department of Pancreatic Surgery and Institutes for Systems Genetics, West China Hospital, Sichuan University, Keyuan 4th Road, Gaopeng Avenue, Hi-tech Zone, Chengdu, Sichuan, 610041, China
| | - Ailin Wei
- Guang'an People's Hospital, Guang'an, 638001, China
| | - Ang Li
- Guang'an People's Hospital, Guang'an, 638001, China.
- Department of Pancreatic Surgery and Institutes for Systems Genetics, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, Sichuan, 610041, China.
| | - Yong Zhang
- Department of Pancreatic Surgery and Institutes for Systems Genetics, West China Hospital, Sichuan University, Keyuan 4th Road, Gaopeng Avenue, Hi-tech Zone, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
3
|
Bhatt P, Hirsch J, Cockrum P, Kim G, Dieguez G. The Effects of Adverse Events and Associated Costs on Value-Based Care for Metastatic Pancreatic Ductal Adenocarcinoma. JOURNAL OF HEALTH ECONOMICS AND OUTCOMES RESEARCH 2024; 11:161-167. [PMID: 39717859 PMCID: PMC11664866 DOI: 10.36469/001c.124367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/03/2024] [Indexed: 12/25/2024]
Abstract
Background: Rising oncology healthcare costs have led to value-based care reimbursement models that coordinate care and improve quality while reducing overall spending. These models are increasingly important for traditional Medicare and other payers. Objectives: To compare the incidence of adverse events (AEs), AE-associated excess costs, and total cost of care (TCOC) of 3 cohorts receiving first-line treatment for metastatic pancreatic ductal adenocarcinoma (mPDAC). Methods: We conducted a retrospective analysis of administrative claims data from 2018 to 2022 using the Medicare 100% Research Identifiable Files. We examined 3 cohorts receiving mPDAC treatment: FOLFIRINOX (FFX) (oxaliplatin, irinotecan, leucovorin, 5-FU bolus and infusion); modified FFX, (5-FU infusion only); and gemcitabine/nab-paclitaxel (gem/abrax). We compared the incidence of clinically significant AEs, TCOC, components of TCOC, and costs related to AEs/treatment toxicity. Results: Patient AE rates ranged from 6.2% to 51.7%. AEs occurred more frequently in patients receiving FFX with all 4 components. Patients receiving brand name gem/abrax had lower rates of febrile neutropenia (6.2%) and neutropenia (22.2%) than those receiving FFX with no 5-FU bolus (febrile neutropenia, 9.9%; neutropenia, 36.9%) and FFX with all 4 components (febrile neutropenia, 6.9%; neutropenia, 30.4%). Rates of most nonhematologic AEs were higher in patients receiving FFX with all 4 components, with diarrhea occurring in 28.3%, abdominal pain in 31.5%, and nausea/vomiting in 41.5% of patients. TCOC was lower in the gem/abrax cohort: 6505 v s F F X w i t h n o 5 - F U b o l u s ( 6995) and FFX with all 4 components ( 7142 ) p e r a d m i n i s t r a t i o n . T h e d e v e l o p m e n t o f a n y s t u d i e d h e m a t o l o g i c A E w a s a s s o c i a t e d w i t h a m e a n e x c e s s c o s t o f 5993 per administration, while the development of any studied nonhematological AE was associated with a mean per-administration excess cost of $3665. Discussion: Treatment decisions intended to minimize chemotherapy costs may lead to suboptimal decisions if the goal is to reduce TCOC. Our research suggests FFX is more costly than gem/abrax (TCOC per administration). Patients receiving gem/abrax were older and had higher baseline Charlson Comorbidity Index scores; however, other factors may be important in driving cost differences. Conclusions: Irrespective of drug cost, chemotherapy leading to a significant increase in AEs is associated with higher TCOC.
Collapse
|
4
|
Balakrishnan K, Xiao Y, Chen Y, Dong J. Elevated Expression of Cell Adhesion, Metabolic, and Mucus Secretion Gene Clusters Associated with Tumorigenesis, Metastasis, and Poor Survival in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2024; 16:4049. [PMID: 39682235 DOI: 10.3390/cancers16234049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
OBJECTIVES Technological advances in identifying gene expression profiles are being applied to study an array of cancers. The goal of this study was to identify differentially expressed genes in pancreatic ductal adenocarcinoma (PDAC) and examine their potential role in tumorigenesis and metastasis. METHODS The transcriptomic profiles of PDAC and non-tumorous tissue samples were derived from the gene expression omnibus (GEO), which is a public repository. The GEO2R tool was used to further derive differentially expressed genes from those profiles. RESULTS In this study, a total of 68 genes were derived from upregulated PDAC genes in three or more transcriptomic profiles and were considered PDAC gene sets. The identified PDAC gene sets were examined in the molecular signatures database (MSigDB) for ontological investigation, which revealed that these genes were involved in the extracellular matrix and associated with the cell adhesion process in PDAC tumorigenesis. The gene set enrichment analysis showed greater enrichment scores for the gene sets. Moreover, the identified gene sets were examined for protein-protein interaction using the STRING database. Based on functional k-means clustering, the following three functional cluster groups were identified in this study: extracellular matrix/cell adhesion, metabolic, and mucus secretion-related protein groups. The receiver operating characteristic (ROC) curve revealed greater specificity and sensitivity for these cluster genes in predicting PDAC tumorigenesis and metastases. In addition, the expression of the cluster genes affects the overall survival rate of PDAC patients. Using the cancer genome atlas (TCGA) database, the associations between expression levels and clinicopathological features were validated. CONCLUSIONS Overall, the genes identified in this study appear to be critical in PDAC development and can serve as potential diagnostic and prognostic targets for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Karthik Balakrishnan
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yi Xiao
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yuanhong Chen
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jixin Dong
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
5
|
Mortoglou M, Lian M, Miralles F, Dart DA, Uysal-Onganer P. miR-210 Mediated Hypoxic Responses in Pancreatic Ductal Adenocarcinoma. ACS OMEGA 2024; 9:47872-47883. [PMID: 39651070 PMCID: PMC11618397 DOI: 10.1021/acsomega.4c08947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 12/11/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one among the most lethal malignancies due to its aggressive behavior and resistance to conventional therapies. Hypoxia significantly contributes to cancer progression and therapeutic resistance of PDAC. microRNAs (miRNAs/miRs) have emerged as critical regulators of various biological processes. miR-210 is known as the "hypoxamir" due to its prominent role in cellular responses to hypoxia. In this study, we investigated the multifaceted role of miR-210 in PDAC using miR-210 knockout (KO) cellular models to elucidate its functions under hypoxic conditions. Hypoxia-inducible factor-1α (HIF1-α), a key transcription factor activated in response to low oxygen levels, upregulates miR-210. miR-210 maintains cancer stem cell (CSC) phenotypes and promotes epithelial-mesenchymal transition (EMT), which is essential for tumor initiation, metastasis, and therapeutic resistance. Our findings demonstrate that miR-210 regulates the expression of CSC markers, such as CD24, CD44, and CD133, and EMT markers, including E-cadherin, Vimentin, and Snail. Specifically, depletion of miR-210 reversed EMT and CSC marker expression levels in hypoxic Panc-1 and MiaPaCa-2 PDAC cells. These regulatory actions facilitate a more invasive and treatment-resistant PDAC phenotype. Understanding the regulatory network involving miR-210 under hypoxic conditions may reveal new therapeutic targets for combating PDAC and improving patient outcomes. Our data suggest that miR-210 is a critical regulator of HIF1-α expression, EMT, and the stemness of PDAC cells in hypoxic environments.
Collapse
Affiliation(s)
- Maria Mortoglou
- Cancer
Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, U.K.
| | - Mutian Lian
- Cancer
Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, U.K.
| | - Francesc Miralles
- School
of Health and Medical Sciences, City St
George’s, University of London, Cranmer Terrace, London SW17 0RE, U.K.
| | - D. Alwyn Dart
- UCL
Cancer Institute, University College London, Paul O’Gorman Building, 72
Huntley Street, London WC1E 6DD, U.K.
| | - Pinar Uysal-Onganer
- Cancer
Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, U.K.
| |
Collapse
|
6
|
Escalera-Anzola S, Rosado M, Yang Y, Parra-Sanchez D, Pedro-Liberal CS, Acedo P. Breakthroughs in nanoparticle-based strategies for pancreatic cancer therapy. Biochem Pharmacol 2024; 232:116685. [PMID: 39613113 DOI: 10.1016/j.bcp.2024.116685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/31/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers worldwide, mainly due to its high heterogeneity, resistance to therapy and late diagnosis, with a 5-year survival rate of less than 10%. This dismal prognosis has promoted strategies to develop more effective treatments. Nanoparticle-based strategies have emerged, in the last decades, as a great opportunity because they can enhance drug delivery and promote controlled release, presenting lower side effects than conventional therapeutic regimens. Moreover, nanoparticles can often be modified to target specific cells or to achieve a sustained release of the drugs into the tumor. However, very few nanoparticle-based therapies are clinically approved. Concretely for pancreatic cancer treatment only two nanoformulations have been approved by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) so far. Clinical translation of nanoparticles remains a challenge for modern medicine, and in particular for pancreatic cancer therapy, because of the complexity of the disease, and a lack of studies been performed in clinically relevant in vitro and in vivo models. In this review, we have summarized the most recent clinical trials using nanoparticle-based formulations in PDAC, giving a small context of the diverse types of nanoparticles employed and the most recent advancements in the field.
Collapse
Affiliation(s)
- Sara Escalera-Anzola
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom; Smart Devices for Nano Medicine Group, Unidad Excelencia Instituto de BioMedicina y Genética Molecular (IBGM) de Valladolid, University of Valladolid and CSIC, Valladolid, Spain
| | - Maria Rosado
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Yuchen Yang
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Daniel Parra-Sanchez
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Carolina San Pedro-Liberal
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Pilar Acedo
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom.
| |
Collapse
|
7
|
Costa VDCMD, Lima MDCAD, da Cruz Filho IJ, Galdino LV, Pereira MC, Silva BDO, Albuquerque APDB, da Rosa MM, Pitta MGDR, Rêgo MJBDM. 5-nitro-thiophene-thiosemicarbazone derivative induces cell death, cell cycle arrest, and phospho-kinase shutdown in pancreatic ductal adenocarcinoma cells. Eur J Pharmacol 2024; 983:176963. [PMID: 39260813 DOI: 10.1016/j.ejphar.2024.176963] [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: 03/15/2024] [Revised: 08/08/2024] [Accepted: 08/28/2024] [Indexed: 09/13/2024]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with limited treatment options. This study explores the potential of novel 5-nitro-thiophene-thiosemicarbazone derivatives as therapeutic agents for PDAC. METHODS We evaluated the cytotoxicity of seven derivatives in peripheral blood mononuclear cells (PBMCs) and PDAC cell lines. Promising candidates (PR12 and PR17) were further analyzed for their effects on colony formation, cell cycle progression, and reactive oxygen species (ROS) production. PR17, the most promising derivative, was subjected to additional investigation, including analysis of autophagy-related genes and protein kinase inhibition. RESULTS Three derivatives (PR16, PR19, and PR20) displayed cytotoxicity towards PBMCs. PR12 reduced colony formation and G0/G1 cell cycle arrest in PDAC cells. Notably, PR17 exhibited potent activity in MIA PaCa-2 cells, inducing S-phase cell cycle arrest, downregulating autophagy genes, and inhibiting key protein kinases. CONCLUSION PR17, a 5-nitro-thiophene-thiosemicarbazone derivative, demonstrates promising antineoplastic activity against PDAC cells by potentially modulating cell cycle progression, autophagy, and protein kinase signaling. Further studies are warranted to elucidate the detailed mechanism of action and explore its efficacy in vivo.
Collapse
Affiliation(s)
| | | | | | - Lília Vieira Galdino
- Therapeutic Innovation Research Center - Suely Galdino, Federal University of Pernambuco, Pernambuco, Recife, Brazil
| | - Michelly Cristiny Pereira
- Therapeutic Innovation Research Center - Suely Galdino, Federal University of Pernambuco, Pernambuco, Recife, Brazil
| | - Bárbara de Oliveira Silva
- Therapeutic Innovation Research Center - Suely Galdino, Federal University of Pernambuco, Pernambuco, Recife, Brazil
| | | | - Michelle Melgarejo da Rosa
- Therapeutic Innovation Research Center - Suely Galdino, Federal University of Pernambuco, Pernambuco, Recife, Brazil
| | - Maira Galdino da Rocha Pitta
- Therapeutic Innovation Research Center - Suely Galdino, Federal University of Pernambuco, Pernambuco, Recife, Brazil
| | | |
Collapse
|
8
|
Jiang L, Lu X, Dai Y, Jiang K, Miao Y, Yu J, Yin L, Wei J. Establishment and analysis of a prognostic model of pancreatic ductal adenocarcinomas based on nerve-cancer crosstalk-related genes. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2024; 17:396-410. [PMID: 39660330 PMCID: PMC11626290 DOI: 10.62347/ghum8504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 10/26/2024] [Indexed: 12/12/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant tumor with a five-year survival rate of 13%, the lowest among all malignant tumors. The work aims to use bioinformatics methods to mine Nerve-cancer crosstalk-related genes (NCCGs) in pancreatic cancer and evaluate their correlation with tumor stage and prognosis, thereby providing a new direction of development and experimental basis for pancreatic cancer treatment. This study included 185 individuals with PDAC from the TCGA database, together with clinical and RNA sequencing data. A review of prior studies revealed the mechanism of neural-cancer crosstalk and identified 42 neural-cancer crosstalk-related genes (NCCGs). Multivariate logistic regression analysis showed that NGFR (OR=39.076, 95% CI; P<0.05), CHRNB2 (OR=41.076, 95% CI; P<0.05), and CHRNA10 (OR=39.038, 95% CI; P<0.05) were identified as independent risk factors for PNI development. Pearson correlation analysis revealed that CHRNA10 was negatively connected with PDAC microsatellite instability, whereas CHRNA10, CHRNB2, and NGFR were negatively correlated with PDAC tumor mutation burden. The GEPIA database revealed that CHRNB2 expression was higher in stage I PDAC. The pancreatic cancer single-cell dataset PAAD_CRA001160 revealed that malignant tumor cells, ductal cells, endothelial cells and fibroblasts accounted for a large proportion in the tumor microenvironment of pancreatic cancer. Furthermore, the NGFR gene was shown to be more significantly expressed in various pancreatic cancer cells. Bioinformatics analysis was used to create a validated prognostic model of pancreatic cancer, which explored the critical mechanisms of neural-tumor interactions and revealed the potential of cancer-neural crosstalk-related genes as prognostic biomarkers and anti-tumor therapy targets.
Collapse
Affiliation(s)
- Lei Jiang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
- Pancreas Institute of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Xiaozhi Lu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
- Pancreas Institute of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Yuran Dai
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
- Pancreas Institute of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Kuirong Jiang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
- Pancreas Institute of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Yi Miao
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
- Pancreas Institute of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Jun Yu
- Department of Surgery, Johns Hopkins University School of MedicineBaltimore, MD, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence, Johns Hopkins University School of MedicineBaltimore, MD, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of MedicineBaltimore, MD, USA
| | - Lingdi Yin
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
- Pancreas Institute of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Jishu Wei
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
- Pancreas Institute of Nanjing Medical UniversityNanjing, Jiangsu, China
| |
Collapse
|
9
|
Anastasiou IA, Sarantis P, Rebelos E, Eleftheriadou I, Tentolouris KN, Katsaouni A, Koustas E, Kokala V, Karamouzis MV, Tentolouris N. l-Securinine Induces ROS-Dependent Apoptosis on Pancreatic Cancer Cells via the PI3K/AKT/mTOR Signaling Pathway. J Biochem Mol Toxicol 2024; 38:e70036. [PMID: 39467148 DOI: 10.1002/jbt.70036] [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: 04/30/2024] [Revised: 09/03/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024]
Abstract
Accumulating evidence has shown that l-securinine can, in certain circumstances, suppress tumor development by elevating reactive oxygen species (ROS) levels. The current work set out to examine l-securinine's apoptotic effects on HuP-T3 cells as well as any potential underlying molecular mechanism(s) that could explain its action as an anticancer agent. In this study, we used 1.2B4 cells as a control human cell line to verify our findings. Hup-T3 and 1.2B4 cells were cultured with a medium containing the following dilutions of l-securinine: 1-10 μΜ for up to 72 h. We examined the viability and proliferation levels of cells in both cell lines. Then, we measured only 1.2B4 insulin levels and content. We also quantified cell apoptosis, cell cycle levels, and the intracellular reactive oxygen species on HuP-T3 and 1.2B4. Afterwards, we performed a real-time quantitative polymerase chain reaction and western blot analysis. Our results demonstrated that l-securinine inhibited both proliferation and growth of Hup-T3 cells, showing inhibitory and antiproliferative activity in comparison with the control group. In addition, l-securinine had no impact on the proliferation and growth of 1.2B4 cells, nor on their insulin levels and content. By boosting ROS production, and inhibiting the PI3K/AKT/mTOR signaling pathway, l-securinine induced apoptosis on HuP-T3 cells. Pancreatic cancer was successfully inhibited by l-securinine in vitro. l-securinine triggers ROS-dependent apoptosis on pancreatic cancer cells while inhibiting the PI3K/AKT/mTOR signaling pathway. These findings suggest that l-securinine holds promise as a potential lead for future drug development in the fight against pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Ioanna A Anastasiou
- First Department of Propaedeutic Internal Medicine, Diabetes Center, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Sarantis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Greece
| | - Eleni Rebelos
- First Department of Propaedeutic Internal Medicine, Diabetes Center, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
- Turku PET Centre, University of Turku, Turku, Finland
| | - Ioanna Eleftheriadou
- First Department of Propaedeutic Internal Medicine, Diabetes Center, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Konstantinos N Tentolouris
- First Department of Propaedeutic Internal Medicine, Diabetes Center, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasia Katsaouni
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Koustas
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Greece
| | - Vasileia Kokala
- First Department of Propaedeutic Internal Medicine, Diabetes Center, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Michalis V Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Greece
| | - Nikolaos Tentolouris
- First Department of Propaedeutic Internal Medicine, Diabetes Center, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| |
Collapse
|
10
|
Aziz H, Kwon YIC, Park AMG, Lai A, Lee KYC, Zhang D, Kwon Y, Pawlik TM. Recent advancements in management for noncolorectal, nonneuroendocrine hepatic metastases. J Gastrointest Surg 2024; 28:1922-1932. [PMID: 39154708 DOI: 10.1016/j.gassur.2024.08.012] [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: 07/29/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND Owing to the heterogeneity of underlying primary tumors, noncolorectal, nonneuroendocrine metastases to the liver (NCNNMLs), although relatively rare, pose major challenges to treatment and long-term management. Despite being considered the gold standard for colorectal cancer liver metastases, the role of surgical resection for NCNNML remains controversial. Furthermore, advancements in locoregional treatment modalities, such as ablation and various chemotherapeutic modalities, have contributed to the treatment of patients with NCNNML. METHODS This was a comprehensive review of literature that used Medline/PubMed, Google Scholar, the Cochrane Library, and the Web of Science, which were accessed between 2014 and 2024. RESULTS NCNNMLs are rare tumor entities with varied presentation and outcomes. A multidisciplinary approach, which includes chemotherapy, surgery, and interventional radiologic techniques, can be implemented with good results. CONCLUSION Given the complex nature of NCNNML, its management should be highly individualized and multidisciplinary. Locoregional treatments, such as surgical resection and/or ablation, may be more appropriate for select patients and should be offered as a viable therapeutic option for a subset of individuals.
Collapse
Affiliation(s)
- Hassan Aziz
- Department of Surgery, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Ye In Christopher Kwon
- Department of Surgery, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Andrew Min-Gi Park
- Department of Surgery, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Alan Lai
- Department of Surgery, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Kerry Yi Chen Lee
- Department of Surgery, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Dean Zhang
- Department of Surgery, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Yeseo Kwon
- Department of Surgery, School of Medicine, Tufts University, Boston, MA, United States
| | - Timothy M Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, United States.
| |
Collapse
|
11
|
Dobos NK, Garay T, Herold M, Simon A, Madar-Dank V, Balka G, Gajdacsi J, Dank M, Szasz AM, Herold Z. Immune Marker and C-Reactive Protein Dynamics and Their Prognostic Implications in Modulated Electro-Hyperthermia Treatment in Advanced Pancreatic Cancer: A Retrospective Analysis. IMMUNO 2024; 4:385-399. [DOI: 10.3390/immuno4040025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
Abstract
Background: Previous research has suggested that modulated electro-hyperthermia (mEHT) can be used to induce anti-tumor immune effects and to extend patient survival. The use of mEHT in advanced pancreatic cancer is beneficial; however, its immune-mediating effects were never investigated. Methods: A retrospective observational study was conducted. Leukocyte counts, C-reactive protein (CRP), neutrophil-to-lymphocyte ratio (NLR), and granulocyte-to-lymphocyte ratio (GLR) were measured at baseline, midpoint, and after mEHT treatment. Results: A total of 73 mEHT treated pancreatic cancer patients were included. The time elapsed between tumor diagnosis and the first mEHT treatment was 4.40 ± 5.70 months. While no change could be observed between the baseline and the first follow-up visits, the total white blood cell (WBC), neutrophil, and granulocyte count, CRP, NLR, and GLR were significantly higher at the second follow-up compared to both previous visits. Higher levels of the latter parameters following the last mEHT treatment were signaling significantly poor prognostic signs, and so were their longitudinal changes. Conclusions: After the initiation of mEHT, immune markers stabilize with the treatment, but this positive effect is eroded over time by progressive disease. Monitoring the changes in these markers and the occurrence of their increase is a prognostic marker of shorter survival.
Collapse
Affiliation(s)
- Nikolett Kitti Dobos
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, H-1083 Budapest, Hungary
| | - Tamas Garay
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, H-1083 Budapest, Hungary
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
| | - Magdolna Herold
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
- Department of Internal Medicine and Hematology, Semmelweis University, H-1088 Budapest, Hungary
| | - Alexandra Simon
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, H-1083 Budapest, Hungary
| | | | - Gyula Balka
- Department of Pathology, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary
| | - Jozsef Gajdacsi
- Clinical Center, Semmelweis University, H-1083 Budapest, Hungary
| | - Magdolna Dank
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
| | - Attila Marcell Szasz
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
| | - Zoltan Herold
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
| |
Collapse
|
12
|
Rodriguez VI, Mammadova J, Permuth JB, Luthra A, Pena L, Friedman M, Dam A, Cappelle S, Malafa MP, Hallmon C, Miranda C, Mok SRS. Elevated Urinary Levels of Fungal and Environmental Toxins in Patients with Pancreatic Ductal Adenocarcinoma. J Gastrointest Cancer 2024; 56:4. [PMID: 39419859 PMCID: PMC11486816 DOI: 10.1007/s12029-024-01125-4] [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] [Accepted: 09/22/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Risk factors for pancreatic ductal adenocarcinoma (PDAC) include tobacco/alcohol abuse, genetic predisposition, insulin resistance, and pancreatic cysts. Despite these well-established risk factors and the screening of high-risk individuals, some people still develop PDAC. This study aims to explore a potential risk factor for PDAC by investigating the association between fungal toxins (FT) and environmental toxins (ET) and the disease. We predicted that individuals with PDAC would have higher levels of these toxins compared to healthy controls. The rationale behind this hypothesis is that exposure to FT and ET might contribute to the development of PDAC by elevating cancer risk. METHODS A pilot retrospective cohort study was conducted at Moffitt Cancer Center from 2022 to 2023. This study compared FT and ET levels, demographic data, and PDAC features between subjects with PDAC and healthy controls. RESULTS Forty subjects were enrolled in the study, comprising 20 with pancreatic ductal adenocarcinoma (PDAC) and 20 healthy controls. Baseline demographics were similar between the two groups. Among the PDAC subjects, the most common tumor location was the head of the pancreas (55%); 30% had locally advanced disease, 45% were borderline resectable, and 10% had metastatic disease. Compared to the controls, subjects with PDAC had significantly higher levels of fungal toxins (FTs) including ochratoxin, gliotoxin, and citrinin (p < 0.05). Additionally, PDAC patients had significantly elevated levels of environmental toxins (ETs) such as methyl tert-butyl ether (MTBE), xylene, styrene, acrylonitrile, perchlorate, diphenyl phosphate, bromopropane, organophosphates, acrolein, tiglylglycine, and diethylphosphate (p < 0.05). CONCLUSION Our study demonstrates that subjects with PDAC, without other risk factors, have higher FT and ET levels than controls. Further studies are needed to evaluate whether ET and FT exposure can be clinically utilized as a risk factor for PDAC development.
Collapse
Affiliation(s)
- Vanessa I Rodriguez
- Department of Internal Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Jamila Mammadova
- Department of Internal Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jennifer B Permuth
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Anjuli Luthra
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Luis Pena
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Mark Friedman
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Aamir Dam
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Saraswathi Cappelle
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Mokenge P Malafa
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Candice Hallmon
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Cassandra Miranda
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Shaffer R S Mok
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
13
|
Donahue KL, Watkoske HR, Kadiyala P, Du W, Brown K, Scales MK, Elhossiny AM, Espinoza CE, Lasse Opsahl EL, Griffith BD, Wen Y, Sun L, Velez-Delgado A, Renollet NM, Morales J, Nedzesky NM, Baliira RK, Menjivar RE, Medina-Cabrera PI, Rao A, Allen B, Shi J, Frankel TL, Carpenter ES, Bednar F, Zhang Y, Pasca di Magliano M. Oncogenic KRAS-Dependent Stromal Interleukin-33 Directs the Pancreatic Microenvironment to Promote Tumor Growth. Cancer Discov 2024; 14:1964-1989. [PMID: 38958646 PMCID: PMC11450371 DOI: 10.1158/2159-8290.cd-24-0100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/18/2024] [Accepted: 07/01/2024] [Indexed: 07/04/2024]
Abstract
Pancreatic cancer is characterized by an extensive fibroinflammatory microenvironment. During carcinogenesis, normal stromal cells are converted to cytokine-high cancer-associated fibroblasts (CAF). The mechanisms underlying this conversion, including the regulation and function of fibroblast-derived cytokines, are poorly understood. Thus, efforts to therapeutically target CAFs have so far failed. Herein, we show that signals from epithelial cells expressing oncogenic KRAS-a hallmark pancreatic cancer mutation-activate fibroblast autocrine signaling, which drives the expression of the cytokine IL33. Stromal IL33 expression remains high and dependent on epithelial KRAS throughout carcinogenesis; in turn, environmental stress induces interleukin-33 (IL33) secretion. Using compartment-specific IL33 knockout mice, we observed that lack of stromal IL33 leads to profound reprogramming of multiple components of the pancreatic tumor microenvironment, including CAFs, myeloid cells, and lymphocytes. Notably, loss of stromal IL33 leads to an increase in CD8+ T-cell infiltration and activation and, ultimatel |