1
|
Ramesh RPG, Yasmin H, Ponnachan P, Al-Ramadi B, Kishore U, Joseph AM. Phenotypic heterogeneity and tumor immune microenvironment directed therapeutic strategies in pancreatic ductal adenocarcinoma. Front Immunol 2025; 16:1573522. [PMID: 40230862 PMCID: PMC11994623 DOI: 10.3389/fimmu.2025.1573522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 03/04/2025] [Indexed: 04/16/2025] Open
Abstract
Pancreatic cancer is an aggressive tumor with high metastatic potential which leads to decreased survival rate and resistance to chemotherapy and immunotherapy. Nearly 90% of pancreatic cancer comprises pancreatic ductal adenocarcinoma (PDAC). About 80% of diagnoses takes place at the advanced metastatic stage when it is unresectable, which renders chemotherapy regimens ineffective. There is also a dearth of specific biomarkers for early-stage detection. Advances in next generation sequencing and single cell profiling have identified molecular alterations and signatures that play a role in PDAC progression and subtype plasticity. Most chemotherapy regimens have shown only modest survival benefits, and therefore, translational approaches for immunotherapies and combination therapies are urgently required. In this review, we have examined the immunosuppressive and dense stromal network of tumor immune microenvironment with various metabolic and transcriptional changes that underlie the pro-tumorigenic properties in PDAC in terms of phenotypic heterogeneity, plasticity and subtype co-existence. Moreover, the stromal heterogeneity as well as genetic and epigenetic changes that impact PDAC development is discussed. We also review the PDAC interaction with sequestered cellular and humoral components present in the tumor immune microenvironment that modify the outcome of chemotherapy and radiation therapy. Finally, we discuss different therapeutic interventions targeting the tumor immune microenvironment aimed at better prognosis and improved survival in PDAC.
Collapse
Affiliation(s)
- Remya P. G. Ramesh
- Department of Veterinary Medicine, UAE University, Al Ain, United Arab Emirates
| | - Hadida Yasmin
- Immunology and Cell Biology Laboratory, Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal, India
| | - Pretty Ponnachan
- Department of Veterinary Medicine, UAE University, Al Ain, United Arab Emirates
| | - Basel Al-Ramadi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Uday Kishore
- Department of Veterinary Medicine, UAE University, Al Ain, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ann Mary Joseph
- Department of Veterinary Medicine, UAE University, Al Ain, United Arab Emirates
| |
Collapse
|
2
|
Kannen V, Rasmussen M, Das S, Giuliana P, Izzati FN, Choksi H, Erlingsson LAM, Olafsen NE, Åhrling SS, Cappello P, Teino I, Maimets T, Jaudzems K, Gulbinas A, Dambrauskas Z, Edgar LJ, Grant DM, Matthews J. Loss of Parp7 increases type I interferon signalling and reduces pancreatic tumour growth by enhancing immune cell infiltration. Front Immunol 2025; 15:1513595. [PMID: 39867896 PMCID: PMC11759301 DOI: 10.3389/fimmu.2024.1513595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/19/2024] [Indexed: 01/28/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal forms of cancer, and despite low incidence rates, it remains the sixth leading cause of cancer related deaths worldwide. Immunotherapy, which aims to enhance the immune system's ability to recognize and eliminate cancer cells, has emerged as a promising approach in the battle against PDAC. PARP7, a mono-ADP-ribosyltransferase, is a negative regulator of the type I interferon (IFN-I) pathway and has been reported to reduce anti-tumour immunity. METHODS We used murine pancreatic cancer cells, CR705, CRISPR/Cas9, in vivo tumour models and spectral flow cytometry to determine the role of PARP7 in pancreatic tumour growth. RESULTS Loss of Parp7 elevated the levels of interferon stimulated gene factor 3 (ISGF3) and its downstream target genes, even in the absence of STING. Cancer cells knocked out for Parp7 (CR705Parp7KO) produced smaller tumours than control cells (CR705Cas9) when injected into immunocompetent mice. Transcriptomic analyses revealed that CR705Parp7KO tumours had increased expression of genes involved in immunoregulatory interactions and interferon signalling pathways. Characterization of tumour infiltrating leukocyte (TIL) populations showed that CR705Parp7KO tumours had higher proportions of natural killer cells, CD8+ T cells and a lower proportion of anti-inflammatory macrophages (M2). The overall TIL profile of CR705Parp7KO tumours was suggestive of a less suppressive microenvironment. CONCLUSIONS Our data show that loss of Parp7 reduces PDAC tumour growth by increasing the infiltration of immune cells and enhancing anti-tumour immunity. These findings provide support to pursue PARP7 as a therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Vinicius Kannen
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Marit Rasmussen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Siddhartha Das
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Paolo Giuliana
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Fauzia N. Izzati
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Hani Choksi
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Linnea A. M. Erlingsson
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Ninni E. Olafsen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Samaneh S. Åhrling
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Paola Cappello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Indrek Teino
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Toivo Maimets
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | | | - Antanas Gulbinas
- Surgical Gastroenterology Laboratory, Institute for Digestive Research, Lithuanian University of Health of Sciences, Kaunas, Lithuania
| | - Zilvinas Dambrauskas
- Surgical Gastroenterology Laboratory, Institute for Digestive Research, Lithuanian University of Health of Sciences, Kaunas, Lithuania
| | - Landon J. Edgar
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Denis M. Grant
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Jason Matthews
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
3
|
Khalid A, Pasha SA, Demyan L, Standring O, King DA, Newman E, DePeralta D, Gholami S, Weiss MJ, Melis M. Evaluating the Association of Area Deprivation Index (ADI) on Postoperative Outcomes in Pancreatic Adenocarcinoma. J Surg Oncol 2024. [PMID: 39523512 DOI: 10.1002/jso.27996] [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: 08/03/2024] [Revised: 09/24/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Pancreatic adenocarcinoma (PDAC) is a challenging disease, with outcomes influenced by several factors including socioeconomic status. The area deprivation index (ADI) has been used to understand how neighborhood disadvantages affect healthcare outcomes. Prior research has indicated that a higher ADI, reflective of a greater neighborhood disadvantage, is associated with an increased risk of major complications and unplanned readmission following PDAC resection. This study aimed to extend this investigation to the Northwell Health System in New York and explore the association between neighborhood ADI and surgical outcomes in patients with PDAC. METHODS A retrospective analysis of the Northwell Health multicenter pancreatic cancer database from 2014 to 2023 included patients who underwent PDAC resection. The ADI scores were divided into low (1-3), moderate (4-6), and high (7-10), as previously described. Multinomial regression models and Kaplan-Meier log-rank tests were used to compare differences in surgical outcomes between the patients in each ADI group. RESULTS Out of 314 PDAC patients who underwent resection and had available ADI data, 116 (36.9%) were in the low, 163 (51.9%) in the moderate, and 35 (11.2%) in the high ADI category. The median ADI score was 4 (IQR: 3-5). Adjusted multinomial regression analysis revealed the following disparities: compared to the low ADI group, patients in the moderate ADI group demonstrated a significantly higher risk of diabetes (RR: 1.76, 95% CI 1.06-2.90, p = 0.028); high ADI was associated with a poorer response to neoadjuvant therapy (RR 3.13, 95% CI 1.11-8.82, p = 0.031), higher incidence of microscopic positive margins (RR 1.87, 95% CI 1.11-5.17, p = 0.028), increased severe complications (Clavien-Dindo class III-IV) (RR 1.36, 95% CI 1.04-1.80, p = 0.027), and a higher failure-to-rescue (FTR) rate (RR 1.44, 95% CI 1.12-1.85, p = 0.048). Although readmission and mortality rates at 30 and 90 days did not show significant differences (p > 0.05), the Kaplan-Meier log-rank test indicated a marked disparity in survival probabilities among ADI ranks (p = 0.0025). CONCLUSION This study underscores a pronounced survival disparity across ADI categories among PDAC patients, suggesting an association between socioeconomic status and postoperative survival. Consideration of patient ADI may guide tailored healthcare strategies, such as the distribution of navigation and resources, to bridge the gap in survival outcomes and ensure equitable care for all socioeconomic strata.
Collapse
Affiliation(s)
- Abdullah Khalid
- Northwell Health, North Shore/Long Island Jewish, Manhasset, New York, USA
| | - Shamsher A Pasha
- Northwell Health, North Shore/Long Island Jewish, Manhasset, New York, USA
| | - Lyudmyla Demyan
- Northwell Health, North Shore/Long Island Jewish, Manhasset, New York, USA
| | - Oliver Standring
- Northwell Health, North Shore/Long Island Jewish, Manhasset, New York, USA
| | - Daniel A King
- Northwell Health, North Shore/Long Island Jewish, Manhasset, New York, USA
| | - Elliot Newman
- Northwell Health, Lenox Hill Hospital, New York, New York, USA
| | - Danielle DePeralta
- Northwell Health, North Shore/Long Island Jewish, Manhasset, New York, USA
| | - Sepideh Gholami
- Northwell Health, North Shore/Long Island Jewish, Manhasset, New York, USA
| | - Matthew J Weiss
- Northwell Health, North Shore/Long Island Jewish, Manhasset, New York, USA
| | | |
Collapse
|
4
|
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
|
5
|
Giuliante F, Panettieri E, Campisi A, Coppola A, Vellone M, De Rose AM, Ardito F. Treatment of oligo-metastatic pancreatic ductal adenocarcinoma to the liver: is there a role for surgery? A narrative review. Int J Surg 2024; 110:6163-6169. [PMID: 38818688 PMCID: PMC11486948 DOI: 10.1097/js9.0000000000001665] [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: 12/30/2023] [Accepted: 05/09/2024] [Indexed: 06/01/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a prognostically unfavorable malignancy that presents with distant metastases at the time of diagnosis in half of patients. Even if patients with metastatic PDAC have not been traditionally considered candidates for surgery, an increasing number of researchers have been investigating the efficacy of surgical treatment for patients with liver-only oligometastases from PDAC, showing promising results in extremely selected patients, mainly with metachronous metastases after perioperative chemotherapy. Nevertheless, a standardized definition of oligometastatic disease should be adopted and additional investigations focusing on the role of perioperative chemotherapy and tumor biology are warranted to reliably assess the role of resection for PDAC metastatic to the liver.
Collapse
Affiliation(s)
- Felice Giuliante
- Hepatobiliary Surgery, Fondazione Policlinico Universitario Agostino Gemelli ‘IRCCS’, Università Cattolica del Sacro Cuore
| | - Elena Panettieri
- Hepatobiliary Surgery, Fondazione Policlinico Universitario Agostino Gemelli ‘IRCCS’, Università Cattolica del Sacro Cuore
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Andrea Campisi
- Hepatobiliary Surgery, Fondazione Policlinico Universitario Agostino Gemelli ‘IRCCS’, Università Cattolica del Sacro Cuore
| | | | - Maria Vellone
- Hepatobiliary Surgery, Fondazione Policlinico Universitario Agostino Gemelli ‘IRCCS’, Università Cattolica del Sacro Cuore
| | - Agostino M. De Rose
- Hepatobiliary Surgery, Fondazione Policlinico Universitario Agostino Gemelli ‘IRCCS’, Università Cattolica del Sacro Cuore
| | - Francesco Ardito
- Hepatobiliary Surgery, Fondazione Policlinico Universitario Agostino Gemelli ‘IRCCS’, Università Cattolica del Sacro Cuore
| |
Collapse
|
6
|
Aparicio-Lopez CB, Timmerman S, Lorino G, Rogers T, Pyle M, Shrestha TB, Basel MT. Thermosensitive Liposomes for Gemcitabine Delivery to Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2024; 16:3048. [PMID: 39272906 PMCID: PMC11394165 DOI: 10.3390/cancers16173048] [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: 07/26/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Treatment of pancreatic ductal adenocarcinoma with gemcitabine is limited by an increased desmoplasia, poor vascularization, and short plasma half-life. Heat-sensitive liposomes modified by polyethylene glycol (PEG; PEGylated liposomes) can increase plasma stability, reduce clearance, and decrease side effects. Nevertheless, translation of heat-sensitive liposomes to the clinic has been hindered by the low loading efficiency of gemcitabine and by the difficulty of inducing hyperthermia in vivo. This study was designed to investigate the effect of phospholipid content on the stability of liposomes at 37 °C and their release under hyperthermia conditions; this was accomplished by employing a two-stage heating approach. First the liposomes were heated at a fast rate, then they were transferred to a holding bath. Thermosensitive liposomes formulated with DPPC: DSPC: PEG2k (80:15:5, mole%) exhibited minimal release of carboxyfluorescein at 37 °C over 30 min, indicating stability under physiological conditions. However, upon exposure to hyperthermic conditions (43 °C and 45 °C), these liposomes demonstrated a rapid and significant release of their encapsulated content. The encapsulation efficiency for gemcitabine was calculated at 16.9%. Additionally, fluorescent analysis during the removal of unencapsulated gemcitabine revealed an increase in pH. In vitro tests with BxPC3 and KPC cell models showed that these thermosensitive liposomes induced a heat-dependent cytotoxic effect comparable to free gemcitabine at temperatures above 41 °C. This study highlights the effectiveness of the heating mechanism and cell models in understanding the current challenges in developing gemcitabine-loaded heat-sensitive liposomes.
Collapse
Affiliation(s)
- Cesar B Aparicio-Lopez
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Sarah Timmerman
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Gabriella Lorino
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Tatiana Rogers
- Department of Electrical and Computer Engineering, Kansas State University, Manhattan, KS 66506, USA
| | - Marla Pyle
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Tej B Shrestha
- Nanotechnology Innovation Center of Kansas State (NICKS), Kansas State University, Manhattan, KS 66506, USA
| | - Matthew T Basel
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
7
|
Herzberg J, Strate T, Acs M, Piso P, Guraya SY, Honarpisheh H. Reduction of Postoperative Complications in Pancreatic Surgery by Standardizing Perioperative Management: An Observational Cohort Study. Visc Med 2024; 40:184-193. [PMID: 39157727 PMCID: PMC11326765 DOI: 10.1159/000539688] [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: 02/18/2024] [Accepted: 06/04/2024] [Indexed: 08/20/2024] Open
Abstract
Introduction Resection for pancreatic malignancy remains the gold standard for cure. Postoperative morbidity continues to be high even after technical innovations. This study evaluates the effectiveness of a standard perioperative pancreatic oncological surgery step-by-step protocol in reducing organ-specific complications. Methods In this observational cohort study, we analyzed the outcomes of oncologic pancreatic head resections from 2015 to 2022 after the implementation of a standard perioperative fail-safe protocol and compared these data with a historical cohort (2013-2014). In the study group, all patients were treated with preoperative limited mechanical bowel preparation, administration of a somatostatin analog, and a "pancreatic duct tube" in pancreatoduodenectomy. The primary outcome measure was the occurrence of postoperative organ-specific complications. Results A total of 151 patients were included in this study. The rate of postoperative pancreatic fistula (grade B and C) in the fail-safe group was 4.2%. Other organ-specific complications as postoperative hemorrhage (2.5%) and delayed gastric emptying (9.2%) also occurred less frequent than before implementation of the fail-safe protocol. Conclusion The use of this standardized fail-safe protocol for oncologic pancreatoduodenectomy can lead to a low postoperative morbidity with improved surgical outcomes.
Collapse
Affiliation(s)
- Jonas Herzberg
- Department of Surgery, Krankenhaus Reinbek St. Adolf-Stift, Reinbek, Germany
| | - Tim Strate
- Department of Surgery, Krankenhaus Reinbek St. Adolf-Stift, Reinbek, Germany
| | - Miklos Acs
- Department of General and Visceral Surgery, Hospital Barmherzige Brüder, Regensburg, Germany
| | - Pompiliu Piso
- Department of General and Visceral Surgery, Hospital Barmherzige Brüder, Regensburg, Germany
| | - Salman Yousuf Guraya
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Human Honarpisheh
- Department of Surgery, Krankenhaus Reinbek St. Adolf-Stift, Reinbek, Germany
| |
Collapse
|
8
|
Elebo N, Abdel-Shafy EA, Omoshoro-Jones JAO, Nsingwane Z, Hussein AAA, Smith M, Candy G, Cacciatore S, Fru P, Nweke EE. Comparative immune profiling of pancreatic ductal adenocarcinoma progression among South African patients. BMC Cancer 2024; 24:809. [PMID: 38973003 PMCID: PMC11229237 DOI: 10.1186/s12885-024-12595-x] [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: 10/26/2023] [Accepted: 07/02/2024] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND Pancreatic Ductal Adenocarcinoma (PDAC) is an aggressive cancer characterized by an immunosuppressive microenvironment. Patients from specific ethnicities and population groups have poorer prognoses than others. Therefore, a better understanding of the immune landscape in such groups is necessary for disease elucidation, predicting patient outcomes and therapeutic targeting. This study investigated the expression of circulating key immune cell markers in South African PDAC patients of African ancestry. METHODS Blood samples were obtained from a total of 6 healthy volunteers (HC), 6 Chronic Pancreatitis (CP) and 34 PDAC patients consisting of 22 resectable (RPC), 8 locally advanced (LAPC) and 4 metastatic (MPC). Real-time Quantitative Polymerase Chain reactions (RT-qPCR), Metabolomics, Enzyme-Linked Immunosorbent Assay (ELISA), Reactive Oxygen Species (ROS), and Immunophenotyping assays were conducted. Statistical analysis was conducted in R (v 4.3.2). Additional analysis of single-cell RNA data from 20 patients (16 PDAC and 4 controls) was conducted to interrogate the distribution of T-cell and Natural Killer cell populations. RESULTS Granulocyte and neutrophil levels were significantly elevated while lymphocytes decreased with PDAC severity. The total percentages of CD3 T-cell subpopulations (helper and double negative T-cells) decreased when compared to HC. Although both NK (p = 0.014) and NKT (p < 0.001) cell levels increased as the disease progressed, their subsets: NK CD56dimCD16- (p = 0.024) and NKTs CD56+ (p = 0.008) cell levels reduced significantly. Of note is the negative association of NK CD56dimCD16- (p < 0.001) cell levels with survival time. The gene expression analyses showed no statistically significant correlation when comparing the PDAC groups with the controls. The inflammatory status of PDAC was assessed by ROS levels of serum which were elevated in CP (p = 0.025), (RPC (p = 0.003) and LAPC (p = 0.008)) while no significant change was observed in MPC, compared to the HC group. ROS was shown to be positively correlated with GlycA (R = 0.45, p = 0.0096). Single-cell analyses showed a significant difference in the ratio of NKT cells per total cell counts in LAPC (p < 0.001) and MPC (p < 0.001) groups compared with HC, confirming observations in our sample group. CONCLUSION The expression of these immune cell markers observed in this pilot study provides insight into their potential roles in tumour progression in the patient group and suggests their potential utility in the development of immunotherapeutic strategies.
Collapse
Affiliation(s)
- Nnenna Elebo
- Department of Surgery, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, 2193, South Africa
- Bioinformatics Unit, International Centre for Genetic Engineering and Biotechnology, Observatory, Cape Town, 7925, South Africa
| | - Ebtesam A Abdel-Shafy
- Bioinformatics Unit, International Centre for Genetic Engineering and Biotechnology, Observatory, Cape Town, 7925, South Africa
- National Research Centre, Cairo, Egypt
| | - Jones A O Omoshoro-Jones
- Department of Surgery, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, 2193, South Africa
- Hepatopancreatobiliary Unit, Department of Surgery, Chris Hani-Baragwanath Academic Hospital, Soweto Johannesburg, South Africa
| | - Zanele Nsingwane
- Department of Surgery, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, 2193, South Africa
| | - Ahmed A A Hussein
- Bioinformatics Unit, International Centre for Genetic Engineering and Biotechnology, Observatory, Cape Town, 7925, South Africa
- Theodore Bilharz Research Institute, Giza, Egypt
| | - Martin Smith
- Department of Surgery, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, 2193, South Africa
- Hepatopancreatobiliary Unit, Department of Surgery, Chris Hani-Baragwanath Academic Hospital, Soweto Johannesburg, South Africa
| | - Geoffrey Candy
- Department of Surgery, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, 2193, South Africa
| | - Stefano Cacciatore
- Bioinformatics Unit, International Centre for Genetic Engineering and Biotechnology, Observatory, Cape Town, 7925, South Africa
| | - Pascaline Fru
- Department of Surgery, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, 2193, South Africa
| | - Ekene Emmanuel Nweke
- Department of Surgery, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, 2193, South Africa.
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Florida, Roodepoort, South Africa.
| |
Collapse
|
9
|
Rogers HK, Shah SL. Role of Endoscopic Ultrasound in Pancreatic Cancer Diagnosis and Management. Diagnostics (Basel) 2024; 14:1156. [PMID: 38893682 PMCID: PMC11171704 DOI: 10.3390/diagnostics14111156] [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: 03/19/2024] [Revised: 05/22/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
The emergence of endoscopic ultrasound (EUS) has significantly impacted the diagnosis and management of pancreatic cancer and its associated sequelae. While the definitive role of EUS for pancreatic cancer remains incompletely characterized by currently available guidelines, EUS undoubtedly offers high diagnostic accuracy, the precise staging of pancreatic neoplasms, and the ability to perform therapeutic and palliative interventions. However, current challenges to EUS include limited specialized expertise and variability in operator proficiency. As the technology and techniques continue to evolve and become more refined, EUS is poised to play an increasingly integral role in shaping pancreatic cancer care.
Collapse
Affiliation(s)
- Hayley K. Rogers
- Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shawn L. Shah
- Division of Digestive and Liver Diseases, Dallas VA Medical Center, VA North Texas Healthcare System, Dallas, TX 75216, USA
| |
Collapse
|
10
|
Vargas A, Dutta P, Carpenter ES, Machicado JD. Endoscopic Ultrasound-Guided Ablation of Premalignant Pancreatic Cysts and Pancreatic Cancer. Diagnostics (Basel) 2024; 14:564. [PMID: 38473035 DOI: 10.3390/diagnostics14050564] [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: 01/28/2024] [Revised: 02/29/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
Pancreatic cancer is on the rise and expected to become the second leading cause of cancer-related death by 2030. Up to a one-fifth of pancreatic cancers may arise from mucinous pancreatic cysts, which are frequently present in the general population. Currently, surgical resection is the only curative approach for pancreatic cancer and its cystic precursors. However, only a dismal proportion of patients are eligible for surgery. Therefore, novel treatment approaches to treat pancreatic cancer and precancerous pancreatic cysts are needed. Endoscopic ultrasound (EUS)-guided ablation is an emerging minimally invasive method to treat pancreatic cancer and premalignant pancreatic cysts. Different ablative modalities have been used including alcohol, chemotherapy agents, and radiofrequency ablation. Cumulative data over the past two decades have shown that endoscopic ablation of mucinous pancreatic cysts can lead to cyst resolution in a significant proportion of the treated cysts. Furthermore, novel data are emerging about the ability to endoscopically ablate early and locally advanced pancreatic cancer. In this review, we aim to summarize the available data on the efficacy and safety of the different EUS-ablation modalities for the management of premalignant pancreatic cysts and pancreatic cancer.
Collapse
Affiliation(s)
- Alejandra Vargas
- Department of Medicine, Eastern Virginia Medical School, Norfolk, VA 23510, USA
| | - Priyata Dutta
- Department of Medicine, Trinity Health, Ann Arbor, MI 48197, USA
| | - Eileen S Carpenter
- Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jorge D Machicado
- Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
11
|
Paulino J, Mansinho H. Recent Developments in the Treatment of Pancreatic Cancer. ACTA MEDICA PORT 2023; 36:670-678. [PMID: 37788655 DOI: 10.20344/amp.19957] [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: 03/28/2023] [Accepted: 08/22/2023] [Indexed: 10/05/2023]
Abstract
Pancreatic duct adenocarcinoma is currently the sixth-leading cause of cancer death worldwide and the fourth in Europe, with a continuous increase in annual lethality in Portugal during the last two decades. Surgical en-bloc resection of the tumor with microscopic-negative margins and an adequate lymphadenectomy is the only possibility of long-term survival. As this type of cancer is a systemic disease, there is a high rate of recurrence even after curative resection, turning systemic therapy the core of its management, mostly based on chemotherapy. Neoadjuvant strategies for nonmetastatic disease showed significant improvement in overall survival compared with upfront surgery, namely in borderline resectable disease. Moreover, these strategies provided downstaging in several situations allowing R0 resections. Under these new oncologic strategies, several recent surgical issues were introduced, namely more aggressive vascular resections and even tumor resections in oligometastatic disease. This review revisits the state-of-the-art of surgical and oncological interventions in pancreatic duct adenocarcinoma and highlights recent advances in the field aiming to achieve higher survival rates.
Collapse
Affiliation(s)
- Jorge Paulino
- General Surgery Department. Hospital da Luz. Lisboa. Portugal
| | - Hélder Mansinho
- Oncology Department. Hospital Garcia de Orta. Almada. Portugal
| |
Collapse
|
12
|
Fernandes EDSM, de Mello FPT, Braga EP, de Souza GO, Andrade R, Pimentel LS, Girão CL, Siqueira M, Moraes-Junior JMA, de Oliveira RV, Goldaracena N, Torres OJM. A more radical perspective on surgical approach and outcomes in pancreatic cancer-a narrative review. J Gastrointest Oncol 2023; 14:1964-1981. [PMID: 37720458 PMCID: PMC10502544 DOI: 10.21037/jgo-22-763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 12/30/2022] [Indexed: 09/19/2023] Open
Abstract
Background and Objective Pancreatic adenocarcinoma remains a dismal disease and is expected to become an even greater burden in the near future. This review focuses on the different surgical aspects for pancreaticoduodenectomy (PD), distal and total pancreatectomy (TP), incorporating lessons from both the western and eastern visions in treating pancreatic cancer. Methods We conducted an extensive literature review through PubMed, prioritizing papers published in the last 5 years, but older emblematic papers were also included. We included articles that explored the treatment of pancreatic adenocarcinoma, with focus on the surgical aspect and strategies to improve outcomes. References of selected articles were also reviewed to identify any missed studies. Only papers in English were included. Key Content and Findings As evidence continues to build, it is clear that both systemic and surgical therapies have a fundamental and complementary role. State of art surgical treatment encompasses complete mesopancreas excision for radical lymphadenectomy. Preoperative planning of dissection planes, extensive knowledge of vascular anatomic variations, oncological principles and expertise for vascular resections are mandatory to perform a more radical operation, in pursuit of improved outcomes. Conclusions Based on current data, patient selection remains key and a more radical surgical approach brings more accomplishing results bringing as to believe that more is better.
Collapse
Affiliation(s)
- Eduardo de Souza M. Fernandes
- Departament of Gastrointestinal and Transplant Surgery, São Lucas Copacabana Hospital-Rede Dasa, Rio de Janeiro, RJ, Brazil
- Department of Gastrointestinal and Transplant Surgery, Adventista Silvestre Hospital, Rio de Janeiro, RJ, Brazil
- Department of Gastrointestinal and Transplant Surgery, Hospital Universitário Clementino Fraga Filho-UFRJ, Rio de Janeiro, RJ, Brazil
| | - Felipe Pedreira T. de Mello
- Departament of Gastrointestinal and Transplant Surgery, São Lucas Copacabana Hospital-Rede Dasa, Rio de Janeiro, RJ, Brazil
- Department of Gastrointestinal and Transplant Surgery, Adventista Silvestre Hospital, Rio de Janeiro, RJ, Brazil
| | - Eduardo Pinho Braga
- Departament of Gastrointestinal and Transplant Surgery, São Lucas Copacabana Hospital-Rede Dasa, Rio de Janeiro, RJ, Brazil
| | - Gabrielle Oliveira de Souza
- Departament of Gastrointestinal and Transplant Surgery, São Lucas Copacabana Hospital-Rede Dasa, Rio de Janeiro, RJ, Brazil
| | - Ronaldo Andrade
- Departament of Gastrointestinal and Transplant Surgery, São Lucas Copacabana Hospital-Rede Dasa, Rio de Janeiro, RJ, Brazil
- Department of Gastrointestinal and Transplant Surgery, Adventista Silvestre Hospital, Rio de Janeiro, RJ, Brazil
| | - Leandro Savattone Pimentel
- Departament of Gastrointestinal and Transplant Surgery, São Lucas Copacabana Hospital-Rede Dasa, Rio de Janeiro, RJ, Brazil
- Department of Gastrointestinal and Transplant Surgery, Adventista Silvestre Hospital, Rio de Janeiro, RJ, Brazil
| | - Camila Liberato Girão
- Departament of Gastrointestinal and Transplant Surgery, São Lucas Copacabana Hospital-Rede Dasa, Rio de Janeiro, RJ, Brazil
- Department of Gastrointestinal and Transplant Surgery, Adventista Silvestre Hospital, Rio de Janeiro, RJ, Brazil
| | - Munique Siqueira
- Departament of Gastrointestinal and Transplant Surgery, São Lucas Copacabana Hospital-Rede Dasa, Rio de Janeiro, RJ, Brazil
- Department of Gastrointestinal and Transplant Surgery, Adventista Silvestre Hospital, Rio de Janeiro, RJ, Brazil
| | - José Maria A. Moraes-Junior
- Department of Hepatopancreatobiliary Surgery, São Domingos Hospital-Rede Dasa, São Luís, MA, Brazil
- Department of Gastrointestinal and Transplant Surgery, Hospital Presidente Dutra, São Luis, MA, Brazil
| | | | - Nicolas Goldaracena
- Division of Transplant Surgery, Department of Surgery, University of Virginia Health System, Charlottesville, VA, USA
| | - Orlando Jorge M. Torres
- Department of Hepatopancreatobiliary Surgery, São Domingos Hospital-Rede Dasa, São Luís, MA, Brazil
- Department of Gastrointestinal and Transplant Surgery, Hospital Presidente Dutra, São Luis, MA, Brazil
| |
Collapse
|
13
|
Palma AM, Vudatha V, Peixoto ML, Madan E. Tumor heterogeneity: An oncogenic driver of PDAC progression and therapy resistance under stress conditions. Adv Cancer Res 2023; 159:203-249. [PMID: 37268397 DOI: 10.1016/bs.acr.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a clinically challenging disease usually diagnosed at advanced or metastasized stage. By this year end, there are an expected increase in 62,210 new cases and 49,830 deaths in the United States, with 90% corresponding to PDAC subtype alone. Despite advances in cancer therapy, one of the major challenges combating PDAC remains tumor heterogeneity between PDAC patients and within the primary and metastatic lesions of the same patient. This review describes the PDAC subtypes based on the genomic, transcriptional, epigenetic, and metabolic signatures observed among patients and within individual tumors. Recent studies in tumor biology suggest PDAC heterogeneity as a major driver of disease progression under conditions of stress including hypoxia and nutrient deprivation, leading to metabolic reprogramming. We therefore advance our understanding in identifying the underlying mechanisms that interfere with the crosstalk between the extracellular matrix components and tumor cells that define the mechanics of tumor growth and metastasis. The bilateral interaction between the heterogeneous tumor microenvironment and PDAC cells serves as another important contributor that characterizes the tumor-promoting or tumor-suppressing phenotypes providing an opportunity for an effective treatment regime. Furthermore, we highlight the dynamic reciprocating interplay between the stromal and immune cells that impact immune surveillance or immune evasion response and contribute towards a complex process of tumorigenesis. In summary, the review encapsulates the existing knowledge of the currently applied treatments for PDAC with emphasis on tumor heterogeneity, manifesting at multiple levels, impacting disease progression and therapy resistance under stress.
Collapse
Affiliation(s)
| | - Vignesh Vudatha
- Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | | | - Esha Madan
- Champalimaud Centre for the Unknown, Lisbon, Portugal; Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA, United States.
| |
Collapse
|
14
|
Lau WY. Current controversies and challenges for resection of pancreatic ductal adenocarcinoma in pancreatic head aiming at cure. Hepatobiliary Pancreat Dis Int 2023; 22:111-112. [PMID: 36450619 DOI: 10.1016/j.hbpd.2022.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 11/11/2022] [Indexed: 11/23/2022]
Affiliation(s)
- Wan Yee Lau
- Faculty of Medicine, the Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China.
| |
Collapse
|
15
|
Temel HY, Kaymak Ö, Kaplan S, Bahcivanci B, Gkoutos GV, Acharjee A. Role of microbiota and microbiota-derived short-chain fatty acids in PDAC. Cancer Med 2023; 12:5661-5675. [PMID: 36205023 PMCID: PMC10028056 DOI: 10.1002/cam4.5323] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/08/2022] [Accepted: 09/23/2022] [Indexed: 02/05/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive lethal diseases among other cancer types. Gut microbiome and its metabolic regulation play a crucial role in PDAC. Metabolic regulation in the gut is a complex process that involves microbiome and microbiome-derived short-chain fatty acids (SCFAs). SCFAs regulate inflammation, as well as lipid and glucose metabolism, through different pathways. This review aims to summarize recent developments in PDAC in the context of gut and oral microbiota and their associations with short-chain fatty acid (SCFA). In addition to this, we discuss possible therapeutic applications using microbiota in PDAC.
Collapse
Affiliation(s)
- Hülya Yılmaz Temel
- Department of Bioengineering, Faculty of EngineeringEge UniversityIzmirTurkey
| | - Öznur Kaymak
- Department of Bioengineering, Faculty of EngineeringEge UniversityIzmirTurkey
| | - Seren Kaplan
- Department of Bioengineering, Faculty of EngineeringEge UniversityIzmirTurkey
| | - Basak Bahcivanci
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUK
| | - Georgios V. Gkoutos
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUK
- National Institute for Health Research Surgical Reconstruction, Queen Elizabeth Hospital BirminghamBirminghamUK
- MRC Health Data Research UK (HDR UK)BirminghamUK
| | - Animesh Acharjee
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUK
- National Institute for Health Research Surgical Reconstruction, Queen Elizabeth Hospital BirminghamBirminghamUK
- MRC Health Data Research UK (HDR UK)BirminghamUK
| |
Collapse
|
16
|
Perioperative Drug Treatment in Pancreatic Surgery-A Systematic Review and Meta-Analysis. J Clin Med 2023; 12:jcm12051750. [PMID: 36902534 PMCID: PMC10003556 DOI: 10.3390/jcm12051750] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023] Open
Abstract
INTRODUCTION Pancreatic resections for malignant or benign diseases are associated with major morbidity and changes in physiology. To reduce perioperative complications and enhance recovery, many types of perioperative medical management have been introduced. The aim of this study was to provide an evidence-based overview on the best perioperative drug treatment. METHODS The electronic bibliographic databases Medline, Embase, CENTRAL, and Web of Science were systematically searched for randomized controlled trials (RCT) evaluating perioperative drug treatments in pancreatic surgery. The investigated drugs were somatostatin analogues, steroids, pancreatic enzyme replacement therapy (PERT), prokinetic therapy, antidiabetic drugs, and proton pump inhibitors (PPI). Targeted outcomes in each drug category were meta-analyzed. RESULTS A total of 49 RCT were included. The analysis of somatostatin analogues showed a significantly lower incidence of postoperative pancreatic fistula (POPF) in the somatostatin group compared to the control group (OR 0.58, 95% CI: 0.45 to 0.74). The comparison of glucocorticoids versus placebo showed significantly less POPF in the glucocorticoid group (OR 0.22, 95% CI: 0.07 to 0.77). There was no significant difference in DGE when erythromycin was compared to placebo (OR 0.33, 95% CI: 0.08 to 1.30). The other investigated drug regimens could only be analyzed qualitatively. CONCLUSION This systematic review provides a comprehensive overview on perioperative drug treatment in pancreatic surgery. Some often-prescribed perioperative drug treatments lack high quality evidence and further research is needed.
Collapse
|
17
|
Li S, Li Q, Ren Y, Yi J, Guo J, Kong X. HSV: The scout and assault for digestive system tumors. Front Mol Biosci 2023; 10:1142498. [PMID: 36926680 PMCID: PMC10011716 DOI: 10.3389/fmolb.2023.1142498] [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: 01/11/2023] [Accepted: 02/15/2023] [Indexed: 03/08/2023] Open
Abstract
More than 25% of all malignant tumors are digestive system tumors (DSTs), which mostly include esophageal cancer, gastric cancer, pancreatic cancer, liver cancer, gallbladder cancer and cholangiocarcinoma, and colorectal cancer. DSTs have emerged as one of the prominent reasons of morbidity and death in many nations and areas around the world, posing a serious threat to human life and health. General treatments such as radiotherapy, chemotherapy, and surgical resection can poorly cure the patients and have a bad prognosis. A type of immunotherapy known as oncolytic virus therapy, have recently shown extraordinary anti-tumor effectiveness. One of the viruses that has been the subject of the greatest research in this field, the herpes simplex virus (HSV), has shown excellent potential in DSTs. With a discussion of HSV-1 based on recent studies, we outline the therapeutic effects of HSV on a number of DSTs in this review. Additionally, the critical function of HSV in the detection of cancers is discussed, and some HSV future possibilities are shown.
Collapse
Affiliation(s)
- Sheng Li
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qingbo Li
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yi Ren
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jia Yi
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinhe Guo
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xianbin Kong
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
18
|
Chang J, Liu Y, Saey SA, Chang KC, Shrader HR, Steckly KL, Rajput M, Sonka M, Chan CHF. Machine-learning based investigation of prognostic indicators for oncological outcome of pancreatic ductal adenocarcinoma. Front Oncol 2022; 12:895515. [PMID: 36568148 PMCID: PMC9773248 DOI: 10.3389/fonc.2022.895515] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 11/09/2022] [Indexed: 12/13/2022] Open
Abstract
Introduction Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with a poor prognosis. Surgical resection remains the only potential curative treatment option for early-stage resectable PDAC. Patients with locally advanced or micrometastatic disease should ideally undergo neoadjuvant therapy prior to surgical resection for an optimal treatment outcome. Computerized tomography (CT) scan is the most common imaging modality obtained prior to surgery. However, the ability of CT scans to assess the nodal status and resectability remains suboptimal and depends heavily on physician experience. Improved preoperative radiographic tumor staging with the prediction of postoperative margin and the lymph node status could have important implications in treatment sequencing. This paper proposes a novel machine learning predictive model, utilizing a three-dimensional convoluted neural network (3D-CNN), to reliably predict the presence of lymph node metastasis and the postoperative positive margin status based on preoperative CT scans. Methods A total of 881 CT scans were obtained from 110 patients with PDAC. Patients and images were separated into training and validation groups for both lymph node and margin prediction studies. Per-scan analysis and per-patient analysis (utilizing majority voting method) were performed. Results For a lymph node prediction 3D-CNN model, accuracy was 90% for per-patient analysis and 75% for per-scan analysis. For a postoperative margin prediction 3D-CNN model, accuracy was 81% for per-patient analysis and 76% for per-scan analysis. Discussion This paper provides a proof of concept that utilizing radiomics and the 3D-CNN deep learning framework may be used preoperatively to improve the prediction of positive resection margins as well as the presence of lymph node metastatic disease. Further investigations should be performed with larger cohorts to increase the generalizability of this model; however, there is a great promise in the use of convoluted neural networks to assist clinicians with treatment selection for patients with PDAC.
Collapse
Affiliation(s)
- Jeremy Chang
- Department of Surgery, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Yanan Liu
- Iowa Initiative for Artificial Intelligence, University of Iowa, Iowa City, IA, United States
| | - Stephanie A. Saey
- Department of Surgery, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Kevin C. Chang
- Department of Surgery, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Hannah R. Shrader
- Department of Surgery, University of Iowa Hospitals and Clinics, Iowa City, IA, United States,Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States
| | - Kelsey L. Steckly
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States
| | - Maheen Rajput
- Department of Radiology, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Milan Sonka
- Iowa Initiative for Artificial Intelligence, University of Iowa, Iowa City, IA, United States,Department of Electrical and Computer Engineering, University of Iowa, Iowa City, IA, United States
| | - Carlos H. F. Chan
- Department of Surgery, University of Iowa Hospitals and Clinics, Iowa City, IA, United States,Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States,*Correspondence: Carlos H. F. Chan,
| |
Collapse
|
19
|
Seo S, Uemura K, Sumiyoshi T, Kondo N, Okada K, Otsuka H, Murakami Y, Takahashi S. Optimal lymph-node dissection for pancreatic tail cancer. Surg Today 2022; 52:1307-1312. [PMID: 35182251 DOI: 10.1007/s00595-022-02463-1] [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: 09/26/2021] [Accepted: 12/05/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE The optimal range of lymph-node dissection for pancreatic tail cancer remains unclear. We investigated the location and frequency of lymph-node metastases to identify the correct range of lymph-node dissection for pancreatic tail cancer. METHODS We analyzed clinical data retrospectively, on patients who underwent distal pancreatectomy for resectable left-sided pancreatic cancer, between February, 2006 and March, 2021. Eligible patients were divided into two groups according to the tumor location: those with pancreatic tail cancer (Pt group) and those with pancreatic body or body and tail cancer (non-Pt group). RESULTS Of the 96 patients analyzed, 61 (64%) were assigned to the Pt group and 35 (36%) were assigned to the non-Pt group. Metastases to stations 7, 8, 9, 10, 11, 14, and 18 were found in 0 (0%), 0 (0%), 0 (0%), 4 (7%), 18 (30%), 2 (4%), and 10 (17%) patients in the Pt group, and in 1 (3%), 4 (12%), 2 (6%), 1 (3%), 18 (51%), 3 (9%), and 6 (17%) patients in the non-Pt group, respectively. CONCLUSION Lymph-node dissection at stations 7, 8, and 9 might not be necessary in patients with resectable pancreatic cancer confined to the pancreatic tail.
Collapse
Affiliation(s)
- Shingo Seo
- Department of Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Kenichiro Uemura
- Department of Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| | - Tatsuaki Sumiyoshi
- Department of Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Naru Kondo
- Department of Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Kenjiro Okada
- Department of Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Hiroyuki Otsuka
- Department of Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Yoshiaki Murakami
- Digestive Disease Center, Hiroshima Memorial Hospital, Hiroshima, Japan
| | - Shinya Takahashi
- Department of Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
20
|
Guan X, Lu N, Zhang J. The combined prognostic model of copper-dependent to predict the prognosis of pancreatic cancer. Front Genet 2022; 13:978988. [PMID: 36035166 PMCID: PMC9399350 DOI: 10.3389/fgene.2022.978988] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose: To assess the prognostic value of copper-dependent genes, copper-dependent-related genes (CDRG), and CDRG-associated immune-infiltrating cells (CIC) for pancreatic cancer. Methods: CDRG were obtained by single-cell analysis of the GSE156405 dataset in the Gene Expression Omnibus (GEO) database. In a ratio of 7:3, we randomly divided the Cancer Genome Atlas (TCGA) cohort into a training cohort and a test cohort. Tumor samples from the GSE62452 dataset were used as the validation cohort. CIBERSORT was used to obtain the immune cell infiltration. We identified the prognostic CDRG and CIC by Cox regression and the least absolute selection operator (LASSO) method. The clinical significance of these prognostic models was assessed using survival analysis, immunological microenvironment analysis, and drug sensitivity analysis. Results: 536 CDRG were obtained by single-cell sequencing analysis. We discovered that elevated LIPT1 expression was associated with a worse prognosis in pancreatic cancer patients. EPS8, CASC8, TATDN1, NT5E, and LDHA comprised the CDRG-based prognostic model. High infiltration of Macrophages.M2 in pancreatic cancer patients results in poor survival. The combined prognostic model showed great predictive performance, with the area under the curve (AUC) values being basically between 0.7 and 0.9 in all three cohorts. Conclusion: We found a cohort of CDRG and CIC in patients with pancreatic cancer. The combined prognostic model provided new insights into the prognosis and treatment of pancreatic cancer.
Collapse
|
21
|
Xu ZH, Wang WQ, Lou WH, Liu L. Insight of pancreatic cancer: recommendations for improving its therapeutic efficacy in the next decade. JOURNAL OF PANCREATOLOGY 2022; 5:58-68. [DOI: 10.1097/jp9.0000000000000093] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Pancreatic cancer is one of the most malignant digestive system tumors. The effectiveness of pancreatic cancer treatment is still dismal, and the 5-year survival rate is only about 10%. Further improving the diagnosis and treatment of pancreatic cancer is the top priority of oncology research and clinical practice. Based on the existing clinical and scientific research experience, the review provides insight into the hotspots and future directions for pancreatic cancer, which focuses on early detection, early diagnosis, molecular typing and precise treatment, new drug development and regimen combination, immunotherapy, database development, model establishment, surgical technology and strategy change, as well as innovation of traditional Chinese medicine and breakthrough of treatment concept.
Collapse
Affiliation(s)
- Zhi-Hang Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen-Quan Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen-Hui Lou
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
22
|
Lafond M, Lambin T, Drainville RA, Dupré A, Pioche M, Melodelima D, Lafon C. Pancreatic Ductal Adenocarcinoma: Current and Emerging Therapeutic Uses of Focused Ultrasound. Cancers (Basel) 2022; 14:2577. [PMID: 35681557 PMCID: PMC9179649 DOI: 10.3390/cancers14112577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 11/27/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) diagnosis accompanies a somber prognosis for the patient, with dismal survival odds: 5% at 5 years. Despite extensive research, PDAC is expected to become the second leading cause of mortality by cancer by 2030. Ultrasound (US) has been used successfully in treating other types of cancer and evidence is flourishing that it could benefit PDAC patients. High-intensity focused US (HIFU) is currently used for pain management in palliative care. In addition, clinical work is being performed to use US to downstage borderline resectable tumors and increase the proportion of patients eligible for surgical ablation. Focused US (FUS) can also induce mechanical effects, which may elicit an anti-tumor response through disruption of the stroma and can be used for targeted drug delivery. More recently, sonodynamic therapy (akin to photodynamic therapy) and immunomodulation have brought new perspectives in treating PDAC. The aim of this review is to summarize the current state of those techniques and share our opinion on their future and challenges.
Collapse
Affiliation(s)
- Maxime Lafond
- LabTAU, The Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Léon Bérard, Université Lyon 1, University Lyon, 69003 Lyon, France; (R.A.D.); (A.D.); (D.M.); (C.L.)
| | - Thomas Lambin
- Endoscopy Division, Édouard Herriot Hospital, 69003 Lyon, France; (T.L.); (M.P.)
| | - Robert Andrew Drainville
- LabTAU, The Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Léon Bérard, Université Lyon 1, University Lyon, 69003 Lyon, France; (R.A.D.); (A.D.); (D.M.); (C.L.)
| | - Aurélien Dupré
- LabTAU, The Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Léon Bérard, Université Lyon 1, University Lyon, 69003 Lyon, France; (R.A.D.); (A.D.); (D.M.); (C.L.)
| | - Mathieu Pioche
- Endoscopy Division, Édouard Herriot Hospital, 69003 Lyon, France; (T.L.); (M.P.)
| | - David Melodelima
- LabTAU, The Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Léon Bérard, Université Lyon 1, University Lyon, 69003 Lyon, France; (R.A.D.); (A.D.); (D.M.); (C.L.)
| | - Cyril Lafon
- LabTAU, The Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Léon Bérard, Université Lyon 1, University Lyon, 69003 Lyon, France; (R.A.D.); (A.D.); (D.M.); (C.L.)
| |
Collapse
|
23
|
Yang J, Wei X, Hu F, Dong W, Sun L. Development and validation of a novel 3-gene prognostic model for pancreatic adenocarcinoma based on ferroptosis-related genes. Cancer Cell Int 2022; 22:21. [PMID: 35033072 PMCID: PMC8760727 DOI: 10.1186/s12935-021-02431-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 12/25/2021] [Indexed: 12/24/2022] Open
Abstract
Background Molecular markers play an important role in predicting clinical outcomes in pancreatic adenocarcinoma (PAAD) patients. Analysis of the ferroptosis-related genes may provide novel potential targets for the prognosis and treatment of PAAD. Methods RNA-sequence and clinical data of PAAD was downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) public databases. The PAAD samples were clustered by a non-negative matrix factorization (NMF) algorithm. The differentially expressed genes (DEGs) between different subtypes were used by “limma_3.42.2” package. The R software package clusterProfiler was used for functional enrichment analysis. Then, a multivariate Cox proportional and LASSO regression were used to develop a ferroptosis-related gene signature for pancreatic adenocarcinoma. A nomogram and corrected curves were constructed. Finally, the expression and function of these signature genes were explored by qRT-PCR, immunohistochemistry (IHC) and proliferation, migration and invasion assays. Results The 173 samples were divided into 3 categories (C1, C2, and C3) and a 3-gene signature model (ALOX5, ALOX12, and CISD1) was constructed. The prognostic model showed good independent prognostic ability in PAAD. In the GSE62452 external validation set, the molecular model also showed good risk prediction. KM-curve analysis showed that there were significant differences between the high and low-risk groups, samples with a high-risk score had a worse prognosis. The predictive efficiency of the 3-gene signature-based nomogram was significantly better than that of traditional clinical features. For comparison with other models, that our model, with a reasonable number of genes, yields a more effective result. The results obtained with qPCR and IHC assays showed that ALOX5 was highly expressed, whether ALOX12 and CISD1 were expressed at low levels in tissue samples. Finally, function assays results suggested that ALOX5 may be an oncogene and ALOX12 and CISD1 may be tumor suppressor genes. Conclusions We present a novel prognostic molecular model for PAAD based on ferroptosis-related genes, which serves as a potentially effective tool for prognostic differentiation in pancreatic cancer patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02431-8.
Collapse
Affiliation(s)
- Jihua Yang
- Department of Endocrinology and Metabolism, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - XiaoHong Wei
- Department of Endocrinology and Metabolism, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Fang Hu
- Department of Endocrinology and Metabolism, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Wei Dong
- Department of Pathology, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China.
| | - Liao Sun
- Department of Endocrinology and Metabolism, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China.
| |
Collapse
|
24
|
Cilleros C, Dupré A, Chen Y, Vincenot J, Rivoire M, Melodelima D. Intraoperative HIFU Ablation of the Pancreas Using a Toroidal Transducer in a Porcine Model. The First Step towards a Clinical Treatment of Locally Advanced Pancreatic Cancer. Cancers (Basel) 2021; 13:6381. [PMID: 34945001 PMCID: PMC8699564 DOI: 10.3390/cancers13246381] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/13/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022] Open
Abstract
Apart from palliative chemotherapy, no other therapy has been proven effective for the treatment of locally advanced pancreatic tumors. In this study, an intraoperative high-intensity focused ultrasound (HIFU) device was tested in vivo to demonstrate the feasibility of treating the pancreatic parenchyma and tissues surrounding the superior mesenteric vessels prior to clinical translation of this technique. Twenty pigs were included and treated using a HIFU device equipped with a toroidal transducer and an integrated ultrasound imaging probe. Treatments were performed with energy escalation (from 30 kJ to 52 kJ). All treatments resulted in visible (macroscopically and in ultrasound images) homogeneous thermal damage, which was confirmed by histology. The dimensions of thermal lesions measured in ultrasound images and those measured macroscopically were correlated (r = 0.82, p < 0.05). No arterial spasms or occlusion were observed at the lowest energy setting. Temporary spasm of the peripancreatic artery was observed when using an energy setting greater than 30 kJ. The possibility of treating the pancreas and tissues around mesenteric vessels without vascular thrombosis holds great promise for the treatment of locally advanced pancreatic cancers. If clinically successful, chemotherapy followed by HIFU treatment could rapidly become a novel treatment option for locally advanced pancreatic cancer.
Collapse
Affiliation(s)
- Celia Cilleros
- LabTAU, INSERM, Centre Léon Bérard, Université Lyon 1, Univ Lyon, F-69003 Lyon, France; (C.C.); (A.D.); (Y.C.); (M.R.)
- EDAP TMS, 4 Rue du Dauphiné, F-69120 Vaulx-en-Velin, France;
| | - Aurélien Dupré
- LabTAU, INSERM, Centre Léon Bérard, Université Lyon 1, Univ Lyon, F-69003 Lyon, France; (C.C.); (A.D.); (Y.C.); (M.R.)
| | - Yao Chen
- LabTAU, INSERM, Centre Léon Bérard, Université Lyon 1, Univ Lyon, F-69003 Lyon, France; (C.C.); (A.D.); (Y.C.); (M.R.)
| | - Jeremy Vincenot
- EDAP TMS, 4 Rue du Dauphiné, F-69120 Vaulx-en-Velin, France;
| | - Michel Rivoire
- LabTAU, INSERM, Centre Léon Bérard, Université Lyon 1, Univ Lyon, F-69003 Lyon, France; (C.C.); (A.D.); (Y.C.); (M.R.)
| | - David Melodelima
- LabTAU, INSERM, Centre Léon Bérard, Université Lyon 1, Univ Lyon, F-69003 Lyon, France; (C.C.); (A.D.); (Y.C.); (M.R.)
| |
Collapse
|
25
|
Eskander MF, Gil L, Beal EW, Li Y, Hamad A, Oppong B, Obeng-Gyasi S, Tsung A. Access Denied: Inequities in Clinical Trial Enrollment for Pancreatic Cancer. Ann Surg Oncol 2021; 29:1271-1277. [PMID: 34655352 DOI: 10.1245/s10434-021-10868-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 09/11/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND The influence of social determinants of health (SDH) on participation in clinical trials for pancreatic cancer is not well understood. In this study, we describe trends and identify disparities in pancreatic cancer clinical trial enrollment. PATIENTS AND METHODS This is a retrospective study of stage I-IV pancreatic cancer patients in the 2004-2016 National Cancer Database. Cohort was stratified into those enrolled in clinical trials during first course of treatment versus not enrolled. Bivariate analysis and logistic regression were used to understand the relationship between SDH and clinical trial participation. RESULTS A total of 1127 patients (0.4%) enrolled in clinical trials versus 301,340 (99.6%) did not enroll. Enrollment increased over the study period (p < 0.001), but not for Black patients or patients on Medicaid. The majority enrolled had metastatic disease (65.8%). On multivariate analysis, in addition to year of diagnosis (p < 0.001), stage (p < 0.001), and Charlson score (p < 0.001), increasing age [odds ratio (OR) 0.96, 95% confidence interval (CI) 0.96-0.97], non-white race (OR 0.54, CI 0.44-0.66), living in the South (OR 0.42, CI 0.35-0.51), and Medicaid, lack of insurance, or unknown insurance (0.41, CI 0.31-0.53) were predictors of lack of participation. Conversely, treatment at an academic center (OR 6.36, CI 5.4-7.4) and higher neighborhood education predicted enrollment (OR 2.0, CI 1.55-2.67 for < 7% with no high school degree versus > 21%). DISCUSSION Age, race, insurance, and geography are barriers to clinical trial enrollment for pancreatic cancer patients. While overall enrollment increased, Black patients and patients on Medicaid remain underrepresented. After adjusting for cancer-specific factors, SDH are still associated with clinical trial enrollment, suggesting need for targeted interventions.
Collapse
Affiliation(s)
- Mariam F Eskander
- Division of Surgical Oncology, The Arthur G. James Cancer Hospital and Solove Research Institute at the Ohio State University Wexner Medical Center, N924 Doan Hall, 410 W 10th Ave, Columbus, OH, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Lindsay Gil
- Division of Surgical Oncology, The Arthur G. James Cancer Hospital and Solove Research Institute at the Ohio State University Wexner Medical Center, N924 Doan Hall, 410 W 10th Ave, Columbus, OH, USA
| | - Eliza W Beal
- Division of Surgical Oncology, The Arthur G. James Cancer Hospital and Solove Research Institute at the Ohio State University Wexner Medical Center, N924 Doan Hall, 410 W 10th Ave, Columbus, OH, USA
| | - Yaming Li
- Division of Surgical Oncology, The Arthur G. James Cancer Hospital and Solove Research Institute at the Ohio State University Wexner Medical Center, N924 Doan Hall, 410 W 10th Ave, Columbus, OH, USA
| | - Ahmad Hamad
- Division of Surgical Oncology, The Arthur G. James Cancer Hospital and Solove Research Institute at the Ohio State University Wexner Medical Center, N924 Doan Hall, 410 W 10th Ave, Columbus, OH, USA
| | - Bridget Oppong
- Division of Surgical Oncology, The Arthur G. James Cancer Hospital and Solove Research Institute at the Ohio State University Wexner Medical Center, N924 Doan Hall, 410 W 10th Ave, Columbus, OH, USA
| | - Samilia Obeng-Gyasi
- Division of Surgical Oncology, The Arthur G. James Cancer Hospital and Solove Research Institute at the Ohio State University Wexner Medical Center, N924 Doan Hall, 410 W 10th Ave, Columbus, OH, USA
| | - Allan Tsung
- Division of Surgical Oncology, The Arthur G. James Cancer Hospital and Solove Research Institute at the Ohio State University Wexner Medical Center, N924 Doan Hall, 410 W 10th Ave, Columbus, OH, USA.
| |
Collapse
|
26
|
Nguyen A, Dzulko M, Murr J, Yen Y, Schneider G, Krämer OH. Class 1 Histone Deacetylases and Ataxia-Telangiectasia Mutated Kinase Control the Survival of Murine Pancreatic Cancer Cells upon dNTP Depletion. Cells 2021; 10:2520. [PMID: 34685500 PMCID: PMC8534202 DOI: 10.3390/cells10102520] [Citation(s) in RCA: 6] [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: 07/12/2021] [Revised: 09/13/2021] [Accepted: 09/18/2021] [Indexed: 12/20/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive disease with a dismal prognosis. Here, we show how an inhibition of de novo dNTP synthesis by the ribonucleotide reductase (RNR) inhibitor hydroxyurea and an inhibition of epigenetic modifiers of the histone deacetylase (HDAC) family affect short-term cultured primary murine PDAC cells. We used clinically relevant doses of hydroxyurea and the class 1 HDAC inhibitor entinostat. We analyzed the cells by flow cytometry and immunoblot. Regarding the induction of apoptosis and DNA replication stress, hydroxyurea and the novel RNR inhibitor COH29 are superior to the topoisomerase-1 inhibitor irinotecan which is used to treat PDAC. Entinostat promotes the induction of DNA replication stress by hydroxyurea. This is associated with an increase in the PP2A subunit PR130/PPP2R3A and a reduction of the ribonucleotide reductase subunit RRM2 and the DNA repair protein RAD51. We further show that class 1 HDAC activity promotes the hydroxyurea-induced activation of the checkpoint kinase ataxia-telangiectasia mutated (ATM). Unlike in other cell systems, ATM is pro-apoptotic in hydroxyurea-treated murine PDAC cells. These data reveal novel insights into a cytotoxic, ATM-regulated, and HDAC-dependent replication stress program in PDAC cells.
Collapse
Affiliation(s)
- Alexandra Nguyen
- Department of Toxicology, University Medical Center, Obere Zahlbacher Str. 67, 55131 Mainz, Germany; (A.N.); (M.D.)
| | - Melanie Dzulko
- Department of Toxicology, University Medical Center, Obere Zahlbacher Str. 67, 55131 Mainz, Germany; (A.N.); (M.D.)
| | - Janine Murr
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar, Technical University Munich, 81675 München, Germany; (J.M.); (G.S.)
| | - Yun Yen
- Ph.D. Program for Cancer Biology and Drug Discovery, Taipei Medical University, 250 Wu Hsing Street, Taipei 110, Taiwan;
| | - Günter Schneider
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar, Technical University Munich, 81675 München, Germany; (J.M.); (G.S.)
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Oliver H. Krämer
- Department of Toxicology, University Medical Center, Obere Zahlbacher Str. 67, 55131 Mainz, Germany; (A.N.); (M.D.)
| |
Collapse
|
27
|
Satoi S. Surgical Treatment of Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2021; 13:cancers13164015. [PMID: 34439170 PMCID: PMC8391680 DOI: 10.3390/cancers13164015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 12/05/2022] Open
Affiliation(s)
- Sohei Satoi
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Japan
| |
Collapse
|