201
|
Klein SM, Bozko M, Toennießen A, Rangno D, Bozko P. High p53 Protein Level Is a Negative Prognostic Marker for Pancreatic Adenocarcinoma. Int J Mol Sci 2024; 25:12307. [PMID: 39596373 PMCID: PMC11594790 DOI: 10.3390/ijms252212307] [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: 09/25/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Pancreatic adenocarcinoma is one of the most aggressive types of cancer. Among different mechanisms generally believed to be important for the development of cancer, aberrant regulation of the p53 protein is a well-known and common feature for many cancer entities. Our work aims to analyze the impact of p53 deregulation and proteins encoded by p53 target genes on the survival of patients suffering from pancreatic adenocarcinoma. We, therefore, focused on the analysis of the selected collective for the TP53 mutation status, the p53 protein level, their correlation, and possible impacts on the prognosis/survival. We compared and analyzed a set of 123 patients. We have extracted information regarding the TP53 mutation status, p53 protein levels, the level of proteins encoded by prominent p53 target genes, and information on the overall survival. Survival analyses were displayed by Kaplan-Meier plots, using the log-rank test, in order to check for statistical significance. Protein levels were compared using the Mann-Whitney Test. We did not find any statistically significant correlation between the TP53 mutation status and the survival of the patients. Moreover, we have not found any significant correlation between the protein amount of prominent p53 target genes and the patients' survival. However, we see a significant correlation between the p53 protein level in cancer samples and the overall survival of pancreatic adenocarcinoma patients: patients having tumors with a p53 protein level within the upper quartile of all measured cases show a significantly reduced survival compared to the rest of the patients. Thus, in pancreatic adenocarcinoma, the p53 protein level is a relevant marker for prognosis, and cancers having a high p53 protein amount show a shortened patients' survival. In contrast, for this cancer entity, the TP53 mutation status or the protein amount of prominent p53 target genes on their own seems not to have a significant impact on survival.
Collapse
Affiliation(s)
- Sebastian M. Klein
- Department of Internal Medicine I, Universitätsklinikum Tübingen, Universität Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
- M3 Research Institute, University Tübingen, 72076 Tübingen, Germany
| | - Maria Bozko
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, 02-106 Warsaw, Poland
| | | | - Dennis Rangno
- Department of Internal Medicine I, Universitätsklinikum Tübingen, Universität Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
| | - Przemyslaw Bozko
- Department of Internal Medicine I, Universitätsklinikum Tübingen, Universität Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
- M3 Research Institute, University Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
202
|
Feng X, Chai YH, Jiang KX, Jiang WB, Chen WC, Pan Y. Bibliometric analysis of olaparib and pancreatic cancer from 2009 to 2022: A global perspective. World J Gastrointest Oncol 2024; 16:4489-4505. [PMID: 39554747 PMCID: PMC11551633 DOI: 10.4251/wjgo.v16.i11.4489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/24/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Genetic screening for breast cancer gene 1 (BRCA)1/2 mutations can inform breast/ovarian/pancreatic cancer patients of suitable therapeutic interventions. Four to seven percent of pancreatic cancer patients have germline BRCA mutations. BRCA genes aid in DNA repair, especially homologous recombination, which impacts genomic stability and cancer cell growth. BRCA1 regulates the cell cycle, ubiquitination, and chromatin remodeling, whereas BRCA2 stimulates the immune response. They predict the efficacy of platinum chemotherapy or polymerase (PARP) inhibitors such as olaparib. AIM To determine the trends and future directions in the use of olaparib for pancreatic cancer treatment. METHODS To evaluate the trends in how olaparib works in pancreatic cancer, we performed a bibliometric analysis. One hundred and ninety-six related publications were accessed from the Web of Science Core Collection and were published between 2009 and 2022. The analytic parameters included publications, related citations, productive countries and institutes, influential authors, and keyword development. RESULTS This study visualizes and discusses the current research, including the present global trends and future directions in olaparib and pancreatic cancer. Overall, this study sheds light on optimizing the use of olaparib in pancreatic cancer treatment, offering valuable guidance for researchers in this field. CONCLUSION Our findings identified trends in olaparib and pancreatic cancer, with China and the USA leading and with global cooperation tightening. O'Reilly EM's team and Memorial Sloan-Kettering had the highest output. The Journal of Clinical Oncology was the most cited journal. More PARP inhibitors are emerging, and combination therapy is suggested for future therapeutic trends.
Collapse
Affiliation(s)
- Xu Feng
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Yi-Han Chai
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Ke-Xin Jiang
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Wen-Bin Jiang
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Wen-Chao Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Yu Pan
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| |
Collapse
|
203
|
Jin H, Lee SH, Kim EH, Park SB, Park N, Joo KR, Yoon SW. Rhus verniciflua stokes extract, a traditional herbal medicine, combined with first-line chemotherapy for unresectable locally advanced and metastatic pancreatic cancer: a prospective observational pilot study. Front Oncol 2024; 14:1469616. [PMID: 39610932 PMCID: PMC11602421 DOI: 10.3389/fonc.2024.1469616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/21/2024] [Indexed: 11/30/2024] Open
Abstract
Pancreatic cancer is among the most lethal cancers with limited treatment options. Traditional herbal medicines have been explored as potential adjunct therapies for enhancing the effectiveness of standard chemotherapies. In this study, we investigated the safety and efficacy of Rhus verniciflua Stokes (RVS) in combination with first-line chemotherapy for unresectable locally advanced and metastatic pancreatic cancer in a prospective setting. This prospective observational pilot study enrolled patients with confirmed inoperable stage III or IV pancreatic cancer undergoing or scheduled to receive 5-fluorouracil-based or gemcitabine-based first-line chemotherapy with RVS treatment and were followed up for up to 20 months. The primary endpoint was the safety profile of RVS, which was assessed through adverse events. The secondary endpoints included overall response rate (ORR), disease control rate (DCR), progression-free survival (PFS), and overall survival (OS). Cox regression analysis identified possible prognostic factors for PFS and OS. Of the enrolled 25 patients, 18 completed the follow-up. The median RVS treatment duration was 6.14 months (range 2.23-8.01 months) and the median RVS daily dose was 3.8 capsules/day (range 2.1-5.7 capsules/day). RVS showed a favorable safety profile, with only one case of non-severe pruritus possibly related to RVS treatment. No hepatotoxicity and nephrotoxicity related to RVS was reported. The ORR and DCR were 5.6% and 72.2%, respectively. The median PFS and OS were 7.24 months (95% CI: 3.15-12.9) and 13.9 months (95% CI: 1.14-27.72), respectively. Cox regression analysis showed that baseline CA19-9 level was an independent prognostic factor for OS, and the daily dose of RVS was an independent prognostic factor for PFS and OS. In conclusion, RVS can be administered safely to patients with unresectable locally advanced metastatic pancreatic cancer who are undergoing first-line chemotherapy, and the daily dose was an independent prognostic factor for cancer survival. Further randomized controlled studies are warranted to confirm the exact benefits of RVS treatment. Clinical trial registration https://cris.nih.go.kr, identifier KCT0007496.
Collapse
Affiliation(s)
- Hayun Jin
- Department of Korean Internal Medicine, Kyung Hee University College of Korean Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| | - Su Hyeon Lee
- Department of Korean Internal Medicine, Kyung Hee University College of Korean Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| | - Eun Hye Kim
- Department of Korean Internal Medicine, Kyung Hee University College of Korean Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| | - Su Bin Park
- Department of Korean Internal Medicine, Kyung Hee University College of Korean Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| | - Namyoung Park
- Department of Gastroenterology, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| | - Kwang Ro Joo
- Department of Gastroenterology, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| | - Seong Woo Yoon
- Department of Korean Internal Medicine, Kyung Hee University College of Korean Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| |
Collapse
|
204
|
Koti S, Standring O, Vithlani N, Amini N, Deperalta D, Deutsch G, Karpeh M, Weiss M, Lad N. Trends and Outcomes of Neoadjuvant Chemotherapy for Clinical Stage T1 Pancreatic Cancer. J Surg Oncol 2024. [PMID: 39543457 DOI: 10.1002/jso.28003] [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: 06/18/2024] [Revised: 09/30/2024] [Accepted: 10/26/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Neoadjuvant chemotherapy (NC) for early pancreatic ductal adenocarcinoma (PDAC) remains controversial. We investigate the adoption of NC and its impact on survival in clinical T1 (cT1) PDAC. METHODS National Cancer Database (2006-2017) was reviewed for cT1 PDAC. Patients receiving NC and surgery were compared with patients undergoing upfront surgery (US). RESULTS A total of 5886 patients were included. NC use increased from 4.8% in 2006 to 18.8% in 2017. The NC group (n = 618) versus the US group (n = 5268) had: younger age (66 years vs. 68 years), smaller tumor size (2 cm vs. 2.2 cm), more pancreas head tumors (77% vs. 70.6%), lower lymph-vascular invasion (25.9% vs. 40.6%), and less lymph node positivity (43.6% vs. 54.5%), p < 0.001. Factors associated with receipt of NC were: younger age, recent year of diagnosis, and treatment at an academic program. In the NC group versus the US group, median OS was 35.2 months versus 28.3 months, p < 0.001. Factors associated with improved survival included: well differentiated pathology, R0 surgical margins, and receipt of chemotherapy. CONCLUSION In cT1 PDAC, chemotherapy is associated with improved survival. In a surgery-first approach, only 59% of patients receive adjuvant chemotherapy. These data suggest consideration of neoadjuvant therapy for early pancreatic cancer.
Collapse
Affiliation(s)
- Shruti Koti
- Department of Surgery, Northwell Health, New Hyde Park, New York, USA
| | - Oliver Standring
- Department of Surgery, Northwell Health, New Hyde Park, New York, USA
| | - Nandan Vithlani
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Neda Amini
- Department of Surgery, Northwell Health, New Hyde Park, New York, USA
| | - Danielle Deperalta
- Department of Surgery, Northwell Health, New Hyde Park, New York, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Gary Deutsch
- Department of Surgery, NYU Langone Hospital-Long Island, Mineola, New York, USA
| | - Martin Karpeh
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
- Division of Surgery, Northwell Health Cancer Institute, New Hyde Park, New York, USA
| | - Matthew Weiss
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
- Division of Surgery, Northwell Health Cancer Institute, New Hyde Park, New York, USA
| | - Neha Lad
- Department of Surgery, Icahn School of Medicine at Mount Sinai, Manhattan, New York, USA
| |
Collapse
|
205
|
Ludmir EB, Sherry AD, Fellman BM, Liu S, Bathala T, Haymaker C, Medina-Rosales MN, Reuben A, Holliday EB, Smith GL, Noticewala SS, Nicholas S, Price TR, Martin-Paulpeter RM, Perles LA, Lee SS, Lee MS, Smaglo BG, Huey RW, Willis J, Zhao D, Cohen L, Taniguchi CM, Koay EJ, Katz MH, Wolff RA, Das P, Pant S, Koong AC, Tang C. Addition of Metastasis-Directed Therapy to Systemic Therapy for Oligometastatic Pancreatic Ductal Adenocarcinoma (EXTEND): A Multicenter, Randomized Phase II Trial. J Clin Oncol 2024; 42:3795-3805. [PMID: 39102622 PMCID: PMC11540734 DOI: 10.1200/jco.24.00081] [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: 01/12/2024] [Revised: 02/16/2024] [Accepted: 06/26/2024] [Indexed: 08/07/2024] Open
Abstract
PURPOSE The EXTEND trial tested the hypothesis that adding comprehensive metastasis-directed therapy (MDT) to chemotherapy would improve progression-free survival (PFS) over chemotherapy alone among patients with oligometastatic pancreatic ductal adenocarcinoma (PDAC). METHODS EXTEND (ClinicalTrials.gov identifier: NCT03599765) is a multicenter, phase II basket trial randomly assigning patients with ≤five metastases 1:1 to MDT plus systemic therapy versus systemic therapy. Disease progression was defined by radiologic criteria (RECIST v1.1), clinical progression, or death. The primary end point was PFS in the per-protocol population, evaluated after all patients achieved at least 6 months of follow-up. Exploratory end points included systemic immune response measures. RESULTS Between March 19, 2019, and February 13, 2023, 41 patients were randomly assigned and 40 were eligible for the primary analysis of PFS (19 patients in the MDT arm; 21 patients in the control arm). At a median follow-up time of 17 months, the median PFS time was 10.3 months (95% CI, 4.6 to 14.0) in the MDT arm versus 2.5 months (95% CI, 1.7 to 5.1) in the control arm. PFS was significantly improved by the addition of MDT to systemic therapy (P = .030 for stratified log-rank test) with a hazard ratio of 0.43 (95% CI, 0.20 to 0.94). No grade ≥3 or greater adverse events related to MDT were observed. Systemic immune activation events were associated with MDT and correlated with improved PFS. CONCLUSION This study supports the addition of MDT to systemic therapy for patients with oligometastatic PDAC. Induction of systemic immunity is a possible mechanism of benefit. These results warrant confirmatory trials to refine treatment strategy and provide external validation.
Collapse
Affiliation(s)
- Ethan B. Ludmir
- Department of Gastrointestinal Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Alexander D. Sherry
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Bryan M. Fellman
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Suyu Liu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Tharakeswara Bathala
- Department of Abdominal Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Cara Haymaker
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
- Translational Molecular Pathology Immunoprofiling Laboratory (TMP-IL), The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Marina N. Medina-Rosales
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
- Translational Molecular Pathology Immunoprofiling Laboratory (TMP-IL), The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Alexandre Reuben
- Department of Thoracic-Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Emma B. Holliday
- Department of Gastrointestinal Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Grace L. Smith
- Department of Gastrointestinal Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sonal S. Noticewala
- Department of Gastrointestinal Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sarah Nicholas
- Department of Radiation Oncology, Banner MD Anderson Cancer Center, Gilbert, AZ
| | - Tracy R. Price
- Department of Radiation Oncology, Community Health Network MD Anderson Cancer Center, Indianapolis, IN
| | - Rachael M. Martin-Paulpeter
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Luis A. Perles
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sunyoung S. Lee
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Michael S. Lee
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Brandon G. Smaglo
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ryan W. Huey
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jason Willis
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Dan Zhao
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Lorenzo Cohen
- Department of Palliative, Rehabilitation, and Integrative Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Cullen M. Taniguchi
- Department of Gastrointestinal Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Eugene J. Koay
- Department of Gastrointestinal Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Matthew H.G. Katz
- Department of Surgical Oncology, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Robert A. Wolff
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Prajnan Das
- Department of Gastrointestinal Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Shubham Pant
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Albert C. Koong
- Department of Gastrointestinal Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Chad Tang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Genitourinary Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
206
|
Topkan E, Kucuk A, Ozturk D, Ozkan EE, Kılıç Durankuş N, Şenyürek Ş, Selek U, Pehlivan B. High Systemic Immune-Inflammation Index Values Before Treatment Predict Poor Pancreatic Cancer Outcomes After Definitive Chemoradiotherapy. Clin Med Insights Oncol 2024; 18:11795549241298552. [PMID: 39525980 PMCID: PMC11544671 DOI: 10.1177/11795549241298552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Background The systemic immune-inflammation index (SII) is an effective tool for predicting the prognosis of patients with cancer. However, its value in patients with locally advanced pancreatic ductal adenocarcinoma (LA-PDAC) undergoing definitive chemoradiotherapy has yet to be addressed. Therefore, we aimed to retrospectively investigate the prognostic significance of the pretreatment SII on the survival outcomes of patients with unresectable LA-PDAC treated with concurrent chemoradiotherapy (C-CRT). Methods The study included 163 patients with LA-PDAC who had received C-CRT. Using receiver operating characteristic (ROC) curve analysis, the utility of a pre-C-CRT cutoff that could stratify survival results was investigated. The primary and secondary endpoints were the correlations between SII levels and overall survival (OS) and progression-free survival (PFS). Results At a median follow-up period of 15 months (range: 3.2-94.5), the median OS and PFS rates for the entire group were 15.7 months (95% confidence interval [CI]: 13.4-17.9), and 7.8 months (95% CI: 6.1-9.4), respectively. We divided the patients into 2 SII cohorts based on the ROC curve analysis (area under the curve [AUC]: 71.9%; sensitivity: 68.9%; specificity: 66.7%): SII < 538 (N = 70) and SII ⩾ 538 (N = 93). Comparative survival analysis showed significantly inferior median OS (13.0 vs 25.4 months; P < .001) and PFS (7.0 vs 15.2 months; P = .003) in patients with SII ⩾ 538 compared with those with SII < 538 before treatment. In multivariate analyses, the Eastern Cooperative Oncology Group (ECOG) performance of 2, N1-2 lymph node, CA 19-9 > 90 U/mL, and SII ⩾ 538 status emerged as independent prognosticators of inferior OS and PFS. Conclusions Present results indicate that patients with unresectable LA-PDAC who underwent C-CRT and had a pretreatment SII ⩾ 538 had significantly worse OS and PFS outcomes compared with those with lower SII values.
Collapse
Affiliation(s)
- Erkan Topkan
- Department of Radiation Oncology, Faculty of Medicine, Baskent University, Adana, Turkey
| | - Ahmet Kucuk
- Clinic of Radiation Oncology, Mersin Education and Research Hospital, Mersin, Turkey
| | - Duriye Ozturk
- Department of Radiation Oncology, Faculty of Medicine, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey
| | - Emine Elif Ozkan
- Department of Radiation Oncology, Suleyman Demirel University, Isparta, Turkey
| | | | - Şükran Şenyürek
- Department of Radiation Oncology, Koc University School of Medicine, Istanbul, Turkey
| | - Ugur Selek
- Department of Radiation Oncology, Koc University School of Medicine, Istanbul, Turkey
| | - Berrin Pehlivan
- Department of Radiation Oncology, Bahcesehir University, Istanbul, Turkey
| |
Collapse
|
207
|
Li Z, Fan X, Jiang D, Li Q, Liu C, Wang D, Li N, Li H, Chen Z, Tang H, Lou C, Xu H, Zhan C, Dong Y, Ma Z, Wang G, Zhang C, Lu H, Zheng T, Zhang Y. Nab-paclitaxel plus S-1 followed by gemcitabine-oxaliplatin as first-line alternating sequential treatment of pancreatic ductal adenocarcinoma. Oncologist 2024; 29:997-e1614. [PMID: 39226089 PMCID: PMC11546623 DOI: 10.1093/oncolo/oyae207] [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: 01/18/2024] [Accepted: 07/15/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Alternating sequential administration of drugs may be a promising approach to overcome chemotherapy resistance in advanced pancreatic ductal adenocarcinoma (PDAC). METHODS This study was an open-label, single-arm, and prospective trial included patients with untreated advanced PDAC. They received 2 cycles of NS regimen (nab-paclitaxel:125 mg/m2, intravenously injected on days 1 and 8, plus S-1:40-60 mg, orally twice per day for 1-14 days) followed by 2 cycles of GemOx regimen (gemcitabine, intravenously injected on days 1 and 8, and oxaliplatin: 130 mg/m2, intravenously injected on day 1). The primary efficacy endpoint was a progression-free survival rate at 6 months (PFSR-6m). The secondary efficacy endpoints included overall survival (OS), progression-free survival (PFS), objective response rate (ORR), disease control rate (DCR), and adverse events (AEs). Specific mRNA transcripts were used to explore survival associated genes. RESULTS Forty-two patients received a minimum of one treatment cycle, and of these, 30 patients completed one alternating treatment consisting of 4 cycles. The PFSR-6m was 71% (95% CI = 58%-87%). The median PFS and OS were 6.53 months (95% CI = 6.03-8.43) and 11.4 months (95% CI = 9.8-14.4), respectively. Common grades 3-4 hematological AEs included neutropenia 30.9%, leukopenia 26.2%, anemia 2.4%, and thrombocytopenia in 11.9%. Patients with OS > 10 months showed high expression of HLA-DQA2 while melanoma-associated antigen genes (MAGE) were notably upregulated in patients with OS < 10 months. CONCLUSION The alternating sequential administration of the NS and GemOx regimens may be a novel approach for first-line chemotherapy in patients with advanced PDAC requiring further study (ClinicalTrials.gov Identifier: ChiCTR1900024867).
Collapse
Affiliation(s)
- Zhiwei Li
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Xiaona Fan
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Dan Jiang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Qingwei Li
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Chao Liu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Dan Wang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Na Li
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen, People’s Republic of China
| | - Hengzhen Li
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Zhuo Chen
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Hongzhen Tang
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen, People’s Republic of China
| | - Changjie Lou
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Haitao Xu
- Department of Hepatobiliary and Pancreatic Surgery, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Chao Zhan
- Department of Hepatobiliary and Pancreatic Surgery, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Yuandi Dong
- Department of Hepatobiliary and Pancreatic Surgery, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Zhigang Ma
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Guangyu Wang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Chunhui Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Haibo Lu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Tongsen Zheng
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Yanqiao Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| |
Collapse
|
208
|
Kobayashi-Ooka Y, Akagi T, Sukezane T, Yanagita E, Itoh T, Sasai K. Cultures derived from pancreatic cancer xenografts with long-term gemcitabine treatment produce chemoresistant secondary xenografts: Establishment of isogenic gemcitabine-sensitive and -resistant models. Pathol Res Pract 2024; 263:155632. [PMID: 39393265 DOI: 10.1016/j.prp.2024.155632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/13/2024]
Abstract
In attempts to establish sophisticated models to reproduce the process of acquired drug resistance, we transformed normal human pancreatic ductal epithelial cells by introducing genes for multiple cellular factors. We also created isogenic gemcitabine-sensitive and -resistant models by short- and long-term gemcitabine treatment, respectively. These models demonstrated differences in drug resistance in vivo, but not in vitro. Gemcitabine treatment also induced squamous transdifferentiation in xenografts in mice. The transcription factor p63 was identified as a possible resistance-determining factor but was unlikely to be solely responsible for the resistance to gemcitabine. This system would prove useful to discover novel molecular targets to overcome chemotherapy resistance, by allowing the evaluation of molecules of interest in xenograft models after in vitro genetic ablation.
Collapse
Affiliation(s)
| | | | | | - Emmy Yanagita
- Division of Diagnostic Pathology, Kobe University Graduate School Medicine, Kobe, Hyogo, Japan
| | - Tomoo Itoh
- Division of Diagnostic Pathology, Kobe University Graduate School Medicine, Kobe, Hyogo, Japan
| | - Ken Sasai
- KAN Research Institute, Inc., Kobe, Hyogo, Japan.
| |
Collapse
|
209
|
An J, Kurilov R, Peccerella T, Bergmann F, Edderkaoui M, Lim A, Zhou X, Pfütze K, Schulz A, Wolf S, Hu K, Springfeld C, Mughal SS, Zezlina L, Fortunato F, Beyer G, Mayerle J, Roth S, Hulkkonen J, Merz D, Ei S, Mehrabi A, Loos M, Al-Saeedi M, Michalski CW, Büchler MW, Hackert T, Brors B, Pandol SJ, Bailey P, Neoptolemos JP. Metavert synergises with standard cytotoxics in human PDAC organoids and is associated with transcriptomic signatures of therapeutic response. Transl Oncol 2024; 49:102109. [PMID: 39217851 PMCID: PMC11402625 DOI: 10.1016/j.tranon.2024.102109] [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: 03/18/2024] [Revised: 07/18/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Despite some recent advances, pancreatic ductal adenocarcinoma (PDAC) remains a growing oncological challenge. New drugs capable of targeting more than one oncogenic pathway may be one way to improve patient outcomes. This study characterizes the effectiveness of Metavert a first-in-class dual inhibitor of GSK3-β and histone deacetylase in treating PDAC as a single agent or in combination with standard cytotoxics. METHODS Thirty-six Patient-Derived Organoids (hPDOs) characterised by RNASeq and whole exome sequencing were treated with Metavert alone or in combination with standard cytotoxics. Transcriptomic signatures (TS) representing sensitivity to Metavert alone or sensitivity to Metavert + irinotecan (IR) were evaluated in 47 patient samples, chemo-naïve in 26 and post-chemotherapy in 21 (gemcitabine=5; FOLFIRINOX=14, both=2) with companion multiplexed immunofluorescence and RNASeq data. RESULTS Metavert combined with gemcitabine, irinotecan, 5FU, oxaliplatin, and paclitaxel was synergistic in the hPDOs. Basal-subtype hPDOs were more sensitive to Metavert alone whereas the Metavert+IR combination exhibited synergy in Classical-subtype hPDOs with increased apoptosis and autophagy. hPDO-derived TS evaluated in PDAC tissues demonstrated that Metavert-TSHi samples were enriched for mRNA splicing and DNA repair processes; they were associated with Basal-like tissues but also with GATA6+ve-chemo-naïve samples and were higher following gemcitabine but not FOLFIRINOX treatment. In contrast, Metavert+IR-TSHI samples were enriched for TP53 pathways; they were associated with Classical-like pretreatment samples and with GATA6+ve/KRT17+ve hybrid cell types following FOLFIRINOX, but not gemcitabine treatment, and were unrelated to transcriptional subtypes. CONCLUSIONS Metavert as a single agent and in combination with irinotecan offers novel strategies for treating pancreatic cancer.
Collapse
Affiliation(s)
- Jingyu An
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany
| | - Roma Kurilov
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Berliner Str. 41, Heidelberg 69120, Germany
| | - Teresa Peccerella
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany
| | - Frank Bergmann
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Mouad Edderkaoui
- Department of Medicine, Cedars-Sinai Medical Center and University of California at Los Angeles, Thalians W204 8700 Beverly Blvd. Los Angeles, California CA 90048, United States
| | - Adrian Lim
- Department of Medicine, Cedars-Sinai Medical Center and University of California at Los Angeles, Thalians W204 8700 Beverly Blvd. Los Angeles, California CA 90048, United States
| | - Xu Zhou
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany
| | - Katrin Pfütze
- Sample Processing Laboratory, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - Angela Schulz
- NGS Core Facility, The German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephan Wolf
- NGS Core Facility, The German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kai Hu
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany
| | - Christoph Springfeld
- Department of Medical Oncology, National Center for Tumor Diseases, University Clinic Heidelberg, Heidelberg 69120, Germany
| | - Sadaf S Mughal
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Berliner Str. 41, Heidelberg 69120, Germany
| | - Lenart Zezlina
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Berliner Str. 41, Heidelberg 69120, Germany
| | - Franco Fortunato
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany
| | - Georg Beyer
- Department of Internal Medicine II, Ludwig-Maximilians-University of Munich, Germany
| | - Julia Mayerle
- Department of Internal Medicine II, Ludwig-Maximilians-University of Munich, Germany
| | - Susanne Roth
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany
| | - Johannes Hulkkonen
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany
| | - Daniela Merz
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany
| | - Shigenori Ei
- Department of Gastroenterological Surgery, Tokai University School of Medicine, Kanagawa, Japan
| | - Arianeb Mehrabi
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany
| | - Martin Loos
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany
| | - Mohammed Al-Saeedi
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany
| | - Christoph W Michalski
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany
| | - Markus W Büchler
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany; Botton-Champalimaud Pancreatic Cancer Centre, Lisbon, Portugal
| | - Thilo Hackert
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany; Department of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Benedikt Brors
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Berliner Str. 41, Heidelberg 69120, Germany; German Cancer Consortium (DKTK), Core Center Heidelberg, Im Neuenheimer Feld 280, Heidelberg 69120, Germany; Medical Faculty and Faculty of Biosciences, Heidelberg University, Im Neuenheimer Feld 234, Heidelberg 69120, Germany; National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, Heidelberg 69120, Germany
| | - Stephen J Pandol
- Department of Medicine, Cedars-Sinai Medical Center and University of California at Los Angeles, Thalians W204 8700 Beverly Blvd. Los Angeles, California CA 90048, United States
| | - Peter Bailey
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Berliner Str. 41, Heidelberg 69120, Germany; Botton-Champalimaud Pancreatic Cancer Centre, Lisbon, Portugal.
| | - John P Neoptolemos
- Heidelberg University Hospital, Department of General, Visceral and Transplantation Surgery, Im Neuenheimer Feld 420, Heidelberg 69120, Germany; Botton-Champalimaud Pancreatic Cancer Centre, Lisbon, Portugal.
| |
Collapse
|
210
|
Wu R, Li J, Aicher A, Jiang K, Tondi S, Dong S, Zheng Q, Tang S, Chen M, Guo Z, Šabanović B, Ananthanarayanan P, Jiang L, Sapino A, Wen C, Fu D, Shen B, Heeschen C. Gasdermin C promotes Stemness and Immune Evasion in Pancreatic Cancer via Pyroptosis-Independent Mechanism. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308990. [PMID: 39297408 PMCID: PMC11558074 DOI: 10.1002/advs.202308990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 05/31/2024] [Indexed: 11/14/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly metastatic and lethal disease. Gasdermins are primarily associated with necrosis via membrane permeabilization and pyroptosis, a lytic pro-inflammatory type of cell death. In this study, GSDMC upregulation during PDAC progression is reported. GSDMC directly induces genes related to stemness, EMT, and immune evasion. Targeting Gsdmc in murine PDAC models reprograms the immunosuppressive tumor microenvironment, rescuing the recruitment of anti-tumor immune cells through CXCL9. This not only results in diminished tumor initiation, growth and metastasis, but also enhances the response to KRASG12D inhibition and PD-1 checkpoint blockade, respectively. Mechanistically, it is discovered that ADAM17 cleaves GSDMC, releasing nuclear fragments binding to promoter regions of stemness, metastasis, and immune evasion-related genes. Pharmacological inhibition of GSDMC cleavage or prevention of its nuclear translocation is equally effective in suppressing GSDMC's downstream targets and inhibiting PDAC progression. The findings establish GSDMC as a potential therapeutic target for enhancing treatment response in this deadly disease.
Collapse
Affiliation(s)
- Renfei Wu
- Center for Single‐Cell OmicsSchool of Public HealthShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
- State Key Laboratory of Systems Medicine for CancerShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
| | - Jingwei Li
- Center for Single‐Cell OmicsSchool of Public HealthShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
- State Key Laboratory of Systems Medicine for CancerShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
- Research Institute of Pancreatic DiseasesShanghai Key Laboratory of Translational Research for Pancreatic NeoplasmsShanghai Jiaotong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
| | - Alexandra Aicher
- Precision ImmunotherapyGraduate Institute of Biomedical SciencesChina Medical UniversityNo. 91, Xueshi RoadTaichung404Taiwan
- Immunology Research and Development CenterChina Medical UniversityNo. 91, Xueshi RoadTaichung404Taiwan
| | - Ke Jiang
- Center for Single‐Cell OmicsSchool of Public HealthShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
- State Key Laboratory of Systems Medicine for CancerShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
| | - Serena Tondi
- Pancreatic Cancer HeterogeneityCandiolo Cancer Institute – FPO – IRCCSStrada Provinciale 142 Km 3,95Candiolo (TO)10060Italy
| | - Shuang Dong
- Center for Single‐Cell OmicsSchool of Public HealthShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
- State Key Laboratory of Systems Medicine for CancerShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
| | - Quan Zheng
- Center for Single‐Cell OmicsSchool of Public HealthShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
- State Key Laboratory of Systems Medicine for CancerShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
| | - Siqi Tang
- School of PharmacyEast China University of Science and Technology130 Meilong RoadShanghai200237P. R. China
| | - Minchun Chen
- Center for Single‐Cell OmicsSchool of Public HealthShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
- State Key Laboratory of Systems Medicine for CancerShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
| | - Zhenyang Guo
- Center for Single‐Cell OmicsSchool of Public HealthShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
- State Key Laboratory of Systems Medicine for CancerShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
| | - Berina Šabanović
- Pancreatic Cancer HeterogeneityCandiolo Cancer Institute – FPO – IRCCSStrada Provinciale 142 Km 3,95Candiolo (TO)10060Italy
| | - Preeta Ananthanarayanan
- Pancreatic Cancer HeterogeneityCandiolo Cancer Institute – FPO – IRCCSStrada Provinciale 142 Km 3,95Candiolo (TO)10060Italy
| | - Lingxi Jiang
- State Key Laboratory of Systems Medicine for CancerShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
- Research Institute of Pancreatic DiseasesShanghai Key Laboratory of Translational Research for Pancreatic NeoplasmsShanghai Jiaotong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
| | - Anna Sapino
- Department of PathologyCandiolo Cancer Institute – FPO – IRCCSStrada Provinciale 142 Km 3,95Candiolo (TO)10060Italy
| | - Chenlei Wen
- State Key Laboratory of Systems Medicine for CancerShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
- Research Institute of Pancreatic DiseasesShanghai Key Laboratory of Translational Research for Pancreatic NeoplasmsShanghai Jiaotong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
| | - Da Fu
- State Key Laboratory of Systems Medicine for CancerShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
- Research Institute of Pancreatic DiseasesShanghai Key Laboratory of Translational Research for Pancreatic NeoplasmsShanghai Jiaotong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
| | - Baiyong Shen
- State Key Laboratory of Systems Medicine for CancerShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
- Research Institute of Pancreatic DiseasesShanghai Key Laboratory of Translational Research for Pancreatic NeoplasmsShanghai Jiaotong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
| | - Christopher Heeschen
- Center for Single‐Cell OmicsSchool of Public HealthShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
- State Key Laboratory of Systems Medicine for CancerShanghai Jiao Tong University School of Medicine227 South Chongqing RoadShanghai200025P. R. China
- Pancreatic Cancer HeterogeneityCandiolo Cancer Institute – FPO – IRCCSStrada Provinciale 142 Km 3,95Candiolo (TO)10060Italy
| |
Collapse
|
211
|
Anglaret S, Hilmi M. [NALIRIFOX in first-line therapy for metastatic pancreatic adenocarcinoma]. Bull Cancer 2024; 111:999-1000. [PMID: 39261251 DOI: 10.1016/j.bulcan.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 05/14/2024] [Accepted: 06/07/2024] [Indexed: 09/13/2024]
Affiliation(s)
- Siriane Anglaret
- Département de médecine oncologique, Gustave-Roussy, Villejuif, France.
| | - Marc Hilmi
- Département d'oncologie médicale, institut Curie, Saint-Cloud, France
| |
Collapse
|
212
|
Doi T, Ishikawa T, Sakakida T, Itani J, Sone D, Morita R, Kataoka S, Miyake H, Seko Y, Yamaguchi K, Moriguchi M, Sogame Y, Konishi H, Murashima K, Iwasaku M, Takayama K, Itoh Y. Real-world genome profiling in Japanese patients with pancreatic ductal adenocarcinoma focusing on HRD implications. Cancer Sci 2024; 115:3729-3739. [PMID: 39315592 PMCID: PMC11531956 DOI: 10.1111/cas.16329] [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: 06/09/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 09/25/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) poses significant challenges due to its high mortality, making it a critical area of research. This retrospective observational study aimed to analyze real-world data from comprehensive genome profiling (CGP) of Japanese patients with PDAC, mainly focusing on differences in gene detection rates among panels and the implications for homologous recombination deficiency (HRD) status. This study enrolled 2568 patients with PDAC who had undergone CGP between June 2019 and December 2021 using data from the nationwide Center for Cancer Genomics and Advanced Therapeutics database. Two types of CGP assays (tissue and liquid biopsies) were compared and a higher detection rate of genetic abnormalities in tissue specimens was revealed. HRD-related gene alterations were detected in 23% of patients, with BRCA1/2 mutations accounting for 0.9% and 2.9% of patients, respectively. Treatment outcome analysis indicated that patients with BRCA1/2 mutations had a longer time to treatment discontinuation with FOLFIRINOX than gemcitabine plus nab-paclitaxel as first-line therapy (9.3 vs. 5.6 months, p = 0.028). However, no significant differences were observed in the treatment response among the other HRD-related genes. Logistic regression analysis identified younger age and family history of breast, prostate, and ovarian cancers as predictive factors for HRD-related gene alterations. Despite the lack of progression-free survival data and the inability to discriminate between germline and somatic mutations, this study provides valuable insights into the clinical implications of CGP in Japanese patients with PDAC. Further research is warranted to optimize panel selection and elucidate the efficacy of platinum-based therapies depending on the HRD status.
Collapse
Affiliation(s)
- Toshifumi Doi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
- Cancer Genome Medical CenterUniversity Hospital, Kyoto Prefectural University of MedicineKyotoJapan
| | - Takeshi Ishikawa
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
- Cancer Genome Medical CenterUniversity Hospital, Kyoto Prefectural University of MedicineKyotoJapan
- Department of Medical Oncology UnitUniversity Hospital, Kyoto Prefectural University of MedicineKyotoJapan
| | - Tomoki Sakakida
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
- Cancer Genome Medical CenterUniversity Hospital, Kyoto Prefectural University of MedicineKyotoJapan
| | - Junichiro Itani
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
- Cancer Genome Medical CenterUniversity Hospital, Kyoto Prefectural University of MedicineKyotoJapan
| | - Daiki Sone
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
- Cancer Genome Medical CenterUniversity Hospital, Kyoto Prefectural University of MedicineKyotoJapan
| | - Ryuichi Morita
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
- Cancer Genome Medical CenterUniversity Hospital, Kyoto Prefectural University of MedicineKyotoJapan
| | - Seita Kataoka
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Hayato Miyake
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Yuya Seko
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Kanji Yamaguchi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Michihisa Moriguchi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Yoshio Sogame
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Hideyuki Konishi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Kyoko Murashima
- Cancer Genome Medical CenterUniversity Hospital, Kyoto Prefectural University of MedicineKyotoJapan
| | - Masahiro Iwasaku
- Cancer Genome Medical CenterUniversity Hospital, Kyoto Prefectural University of MedicineKyotoJapan
- Department of Pulmonary Medicine, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Koichi Takayama
- Cancer Genome Medical CenterUniversity Hospital, Kyoto Prefectural University of MedicineKyotoJapan
- Department of Medical Oncology UnitUniversity Hospital, Kyoto Prefectural University of MedicineKyotoJapan
- Department of Pulmonary Medicine, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Yoshito Itoh
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| |
Collapse
|
213
|
Marschner N, Haug N, Hegewisch-Becker S, Reiser M, Dörfel S, Lerchenmüller C, Linde H, Wolf T, Hof A, Kaiser-Osterhues A, Potthoff K, Jänicke M. Head-to-head comparison of treatment sequences in advanced pancreatic cancer-Real-world data from the prospective German TPK clinical cohort study. Int J Cancer 2024; 155:1629-1640. [PMID: 38956837 DOI: 10.1002/ijc.35071] [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: 12/20/2023] [Revised: 04/25/2024] [Accepted: 05/15/2024] [Indexed: 07/04/2024]
Abstract
There are no clear guidelines regarding the optimal treatment sequence for advanced pancreatic cancer, as head-to-head phase III randomised trials are missing. We assess real-world effectiveness of three common sequential treatment strategies by emulating a hypothetical randomised trial. This analysis included 1551 patients with advanced pancreatic cancer from the prospective, clinical cohort study Tumour Registry Pancreatic Cancer receiving FOLFIRINOX (n = 613) or gemcitabine/nab-paclitaxel (GEMNAB; n = 938) as palliative first-line treatment. We used marginal structural modelling to compare overall survival (OS) and time to deterioration (TTD) of health-related quality of life (HRQoL) between three common first- to second-line treatment sequences, adjusting for time-varying potential confounding. The sequences were: FOLFIRINOX→GEMNAB, GEMNAB→FOLFOX/OFF and GEMNAB→nanoliposomal irinotecan (NALIRI) + 5-fluorouracil. Outcome was also calculated stratified by patients' prognostic risk according to the Pancreatic Cancer Score. Median OS and TTD of HRQoL independent of risk were 10.7 [8.9, 11.9] and 6.4 [4.8, 7.7] months for FOLFIRINOX→GEMNAB, 8.4 [7.4, 9.7] and 5.8 [4.6, 7.1] months for GEMNAB→FOLFOX/OFF and 8.9 [7.8, 10.4] and 4.6 [4.1, 6.1] months for GEMNAB→NALIRI+5-fluorouracil. Compared to FOLFIRINOX→GEMNAB, OS and TTD were worse for poor-risk patients with GEMNAB→FOLFOX/OFF (OS: HR 2.09 [1.47, 2.98]; TTD: HR 1.97 [1.19, 3.27]) and those with GEMNAB→NALIRI+5-fluorouracil (OS: HR 1.35, [0.76, 2.39]; TTD: HR 2.62 [1.56, 4.42]). Brackets denote 95%-confidence intervals. The estimated real-world effectiveness of the three treatment sequences evaluated were largely comparable. Poor-risk patients might benefit from intensified treatment with FOLFIRINOX→GEMNAB in terms of clinical and patient-reported outcomes. Future randomised trials on sequential treatments in advanced pancreatic cancer are warranted.
Collapse
Affiliation(s)
- Norbert Marschner
- Med. Klinik 1, Universitätsklinik Freiburg, Freiburg, Germany
- iOMEDICO, Freiburg, Germany
| | - Nina Haug
- Biostatistics, iOMEDICO, Freiburg, Germany
| | | | - Marcel Reiser
- PIOH-Praxis Internistische Onkologie und Hämatologie, Köln, Germany
| | | | | | - Hartmut Linde
- MVZ für Blut- und Krebserkrankungen, Potsdam, Germany
| | - Thomas Wolf
- BAG, Gemeinschaftspraxis Hämatologie-Onkologie, Dresden, Germany
| | - Anna Hof
- Clinical Epidemiology and Health Economics, iOMEDICO, Freiburg, Germany
| | | | | | - Martina Jänicke
- Clinical Epidemiology and Health Economics, iOMEDICO, Freiburg, Germany
| |
Collapse
|
214
|
Philip PA, Sahai V, Bahary N, Mahipal A, Kasi A, Rocha Lima CMS, Alistar AT, Oberstein PE, Golan T, Metges JP, Lacy J, Fountzilas C, Lopez CD, Ducreux M, Hammel P, Salem M, Bajor D, Benson AB, Luther S, Pardee T, Van Cutsem E. Devimistat (CPI-613) With Modified Fluorouarcil, Oxaliplatin, Irinotecan, and Leucovorin (FFX) Versus FFX for Patients With Metastatic Adenocarcinoma of the Pancreas: The Phase III AVENGER 500 Study. J Clin Oncol 2024; 42:3692-3701. [PMID: 39088774 DOI: 10.1200/jco.23.02659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/27/2024] [Accepted: 05/07/2024] [Indexed: 08/03/2024] Open
Abstract
PURPOSE Metastatic pancreatic adenocarcinoma (mPC) remains a difficult-to-treat disease. Fluorouarcil, oxaliplatin, irinotecan, and leucovorin (FFX) is a standard first-line therapy for mPC for patients with a favorable performance status and good organ function. In a phase I study, devimistat (CPI-613) in combination with modified FFX (mFFX) was deemed safe and exhibited promising efficacy in mPC. METHODS The AVENGER 500 trial (ClinicalTrials.gov identifier: NCT03504423) is a global, randomized phase III trial conducted at 74 sites across six countries to investigate the efficacy and safety of devimistat in combination with mFFX (experimental arm) compared with standard-dose FFX (control arm) in treatment-naïve patients with mPC. Treatment, administered in once-every-2-weeks cycles until disease progression or intolerable toxicity, included intravenous devimistat at 500 mg/m2 total per day on days 1 and 3 in the experimental arm. The primary end point of the study was overall survival (OS). RESULTS Five hundred and twenty-eight patients were randomly assigned (266 in the experimental arm and 262 in the control arm). The median OS was 11.10 months for devimistat plus mFFX versus 11.73 months for FFX (hazard ratio [HR], 0.95 [95% CI, 0.77 to 1.18]; P = .655) and median progression-free survival was 7.8 months versus 8.0 months, respectively (HR, 0.99 [95% CI, 0.76 to 1.29]; P = .94). Grade ≥3 treatment-emergent adverse events with >10% frequency in the devimistat plus mFFX arm versus the FFX arm were neutropenia (29.0% v 34.5%), diarrhea (11.2% v 19.6%), hypokalemia (13.1% v 14.9%), anemia (13.9% v 13.6%), thrombocytopenia (11.6% v 13.6%), and fatigue (10.8% v 11.5%), respectively. CONCLUSION Devimistat in combination with mFFX did not improve long- and short-term mPC patient outcomes compared with standard FFX. There were no new toxicity signals with the addition of devimistat.
Collapse
Affiliation(s)
- Philip A Philip
- Department of Oncology and Department of Pharmacology, Henry Ford Cancer Institute, Wayne State University School of Medicine, Detroit, MI
| | - Vaibhav Sahai
- University of Michigan Rogel Cancer Center, Ann Arbor, MI
| | - Nathan Bahary
- Allegheny Health Network Cancer Institute, Pittsburgh, PA
| | - Amit Mahipal
- University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Anup Kasi
- Division of Medical Oncology, University of Kansas Medical Center, Kansas City, KS
| | - Caio Max S Rocha Lima
- Division of Hematology and Oncology, Atrium Wake Forest Baptist Medical Center, Winston-Salem, NC
| | | | | | - Talia Golan
- Institute of Oncology, Sheba Medical Center, Faculty of Medicine Tel Avi University, Ramat Gan, Israel
| | - Jean-Philippe Metges
- Institut de Cancérologie et d'Hématologie, ARPEGO Network CHU Morvan, Brest, France
| | - Jill Lacy
- Department of Medical Oncology, Yale University School of Medicine, New Haven, CT
| | - Christos Fountzilas
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | | | - Michel Ducreux
- Gustave Roussy, University Paris Saclay, Inserm, U1279, Villejuif, France
| | - Pascal Hammel
- Department of Digestive and Medical Oncology, University Paris-Saclay, Paul Brousse Hospital (AP-HP), Villejuif, France
| | - Mohamed Salem
- Department of Medical Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - David Bajor
- University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Al B Benson
- Division of Hematology & Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | | | - Timothy Pardee
- Division of Hematology and Oncology, Atrium Wake Forest Baptist Medical Center, Winston-Salem, NC
| | - Eric Van Cutsem
- University Hospitals Gasthuisberg Leuven and KU Leuven, Leuven, Belgium
| |
Collapse
|
215
|
Ross SB, Popover J, Sucandy I, Christodoulou M, Pattilachan TM, Rosemurgy AS. The Oncological Stress Test of Neoadjuvant Therapy: A Systematic Review in Outcomes of Neoadjuvant Therapy Compared to Upfront Resection Approach for Borderline Resectable Pancreatic Adenocarcinoma. Am Surg 2024; 90:3061-3073. [PMID: 38635295 DOI: 10.1177/00031348241248703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Pancreatic adenocarcinoma, increasingly diagnosed in the United States, has a disheartening initial resection rate of 15%. Neoadjuvant therapy, particularly FOLFIRINOX and gemcitabine-based regimens, is gaining favor for its potential to improve resectability rates and achieving microscopically negative margins (R0) in borderline resectable cases, marked by intricate arterial or venous involvement. Despite surgery being the sole curative approach, actual benefit of neoadjuvant therapy remains debatable. This study scrutinizes current literature on oncological outcomes post-resection of borderline resectable pancreatic cancer. A MEDLINE/PubMed search was conducted to systematically compare oncological outcomes of patients treated with either neoadjuvant therapy with intent of curative resection or an "upfront resection" approach. A total of 1293 studies were initially screened and 30 were included (n = 1714) in this analysis. All studies included data on outcomes of patients with borderline resectable pancreatic adenocarcinoma being treated with neoadjuvant therapy (n = 1387) or a resection-first approach (n = 356). Patients treated with neoadjuvant therapy underwent resection 52% of the time, achieving negative margins of 43% (n = 601). Approximately 77% of patients who received an upfront resection underwent a successful resection, with 39% achieving negative margins. Neoadjuvant therapy remains marginally efficacious in treatment of borderline resectable pancreatic adenocarcinoma, as patients undergo an operation and successful resection less often when treated with neoadjuvant therapy. Rates of curative resection are comparable, despite neoadjuvant therapy being a primary recommendation in borderline resectable cases and employed more often than upfront resection. Upfront resection may offer improved resection rates by intention-to-treat, which can provide more patients with paths to curative resection.
Collapse
Affiliation(s)
- Sharona B Ross
- Digestive Health Institute, AdventHealth Tampa, Tampa, FL, USA
| | - Jesse Popover
- Digestive Health Institute, AdventHealth Tampa, Tampa, FL, USA
| | - Iswanto Sucandy
- Digestive Health Institute, AdventHealth Tampa, Tampa, FL, USA
| | | | | | | |
Collapse
|
216
|
Uemura S, Kabe Y, Kitago M, Matsuda S, Abe Y, Hasegawa Y, Hori S, Tanaka M, Nakano Y, Sato Y, Itonaga M, Ono M, Kawakami T, Suematsu M, Kitagawa Y. Prognosis prediction of PDAC via detection of O-glycan altered extracellular vesicles in perioperative sera. Cancer Sci 2024; 115:3718-3728. [PMID: 39285510 PMCID: PMC11531947 DOI: 10.1111/cas.16341] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/26/2024] [Accepted: 08/30/2024] [Indexed: 11/05/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a fatal malignancy due to the difficulty in diagnosis and poor prognosis because of the high recurrence rate, necessitating reliable biomarkers to improve the diagnosis and prognosis. However, the existing markers have limitations. We previously identified extracellular vesicles (EVs) recognized by O-glycan-binding lectins (Amaranthus caudatus agglutinin [ACA]) as a novel diagnostic biomarker for PDAC using an EV-counting system (ExoCounter). This retrospective study analyzed changes in ACA-positive EVs in perioperative PDAC serum and its association with prognosis using ExoCounter. Absolute EV levels in the pre- and postoperative sera of 44 patients who underwent curative pancreatectomy for PDAC were quantified using ExoCounter. The carbohydrate antigen 19-9 levels declined in most samples postoperatively, and presented no correlation with poor prognosis. In contrast, ACA-positive EVs increased in serum at 7 days postoperatively in 27 of 44 patients (61.4%). We therefore divided participants with ACA-positive EVs before and after surgery into elevation and decline groups. The overall survival (OS) and recurrence-free survival (RFS) of patients with higher ACA-positive EVs were significantly shorter than those with lower ACA-positive EVs (26.1 months vs. not reached, P = 0.018; 11.9 vs. 38.6 months, P = 0.013). Multivariable analysis revealed that ACA-positive EV elevation in postoperative serum was an independent prognostic factor for poor OS (hazard ratio [HR] = 3.891, P = 0.023) and RFS (HR = 2.650, P = 0.024). The detection of ACA-positive EVs in perioperative serum may be used to predict the prognosis of PDAC in the early postoperative period.
Collapse
Affiliation(s)
- Sho Uemura
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Yasuaki Kabe
- Department of BiochemistryKeio University School of MedicineTokyoJapan
- Department of BiochemistryKochi University Medical SchoolNankokuKochiJapan
| | - Minoru Kitago
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Sachiko Matsuda
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Yuta Abe
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Yasushi Hasegawa
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Shutaro Hori
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Masayuki Tanaka
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Yutaka Nakano
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Yasunori Sato
- Department of BiostatisticsKeio University School of MedicineTokyoJapan
| | | | - Masayuki Ono
- Future Creation Research LaboratoryJvckenwood CorporationYokohamaJapan
| | - Tatsuya Kawakami
- Future Creation Research LaboratoryJvckenwood CorporationYokohamaJapan
| | - Makoto Suematsu
- Department of BiochemistryKeio University School of MedicineTokyoJapan
| | - Yuko Kitagawa
- Department of SurgeryKeio University School of MedicineTokyoJapan
| |
Collapse
|
217
|
Zhang H, Liu A, Bo W, Zhang M, Wang H, Feng X, Wu Y. Upregulation of HSD11B1 promotes cortisol production and inhibits NK cell activation in pancreatic adenocarcinoma. Mol Immunol 2024; 175:10-19. [PMID: 39276709 DOI: 10.1016/j.molimm.2024.08.005] [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: 01/21/2024] [Revised: 08/12/2024] [Accepted: 08/22/2024] [Indexed: 09/17/2024]
Abstract
Cortisol is a glucocorticoid hormone that has immunosuppressive function. Elevated basal cortisol levels are present in patients with some kinds of cancers, but its role in the microenvironment of pancreatic adenocarcinoma (PAAD) remains unclear. This study analyzed the expression of genes involved in cortisol generation by using high-throughput sequencing data from TCGA portal and found HSD11B1 was significantly upregulated in patients with PAAD. The correlations between HSD11B1 level and the expression of 23 immunosuppressive receptors were analyzed by Spearman's correlation analysis. The function of HSD11B1 was examined in primary NK cells and PAAD cell lines. The levels of cortisol in medium and cell lysates were detected by ELISA. In vitro killing assay was used to evaluate the cytotoxicity of NK cells. Cell surface levels of CD96, Tim-3, PD-1, TIGIT, CTLA-4, NKp46, NKp30, NKD2G and LFA-1A, and intracellular levels of CD107a and IFN-γ were examined by flow cytometry. We observed that patients with higher HSD11B1 level had shorter survival time. HSD11B1 is positively correlated with the mRNA levels of 11 immunosuppressive receptors in PAAD. Higher HSD11B1 level relates to reduced abundance of activated NK cells in the tumors. HSD11B1 overexpressed NK cells exhibit exhausted phenotype with increased cortisol production, reduced viability, and reduced cytotoxicity against cancer cells. Overexpression of HSD11B1 did not change the viability of tumor cells but upregulated cortisol production. Targeting HSD11B1 by a specific inhibitor improved the NK cells responsiveness. In conclusion, HSD11B1 is upregulated in patients with PAAD, and higher HSD11B1 level is related to poor prognosis. Upregulation of HSD11B1 in NK and tumor cells increased the production and secretion of cortisol and induces NK cell exhaustion.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Aixiang Liu
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Wentao Bo
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Mingyi Zhang
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Haiqing Wang
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Xielin Feng
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Ying Wu
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China; Department of Medical Oncology, Daytime Medical Treatment Area, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
218
|
Lansbergen MF, Dings MPG, Manoukian P, Fariña A, Waasdorp C, Hooijer GKJ, Verheij J, Koster J, Zwijnenburg DA, Wilmink JW, Medema JP, Dijk F, van Laarhoven HWM, Bijlsma MF. Transcriptome-based classification to predict FOLFIRINOX response in a real-world metastatic pancreatic cancer cohort. Transl Res 2024; 273:137-147. [PMID: 39154856 DOI: 10.1016/j.trsl.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 06/18/2024] [Accepted: 08/13/2024] [Indexed: 08/20/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is often diagnosed at metastatic stage and typically treated with fluorouracil, leucovorin, irinotecan and oxaliplatin (FOLFIRINOX). Few patients benefit from this treatment. Molecular subtypes are prognostic in particularly resectable PDAC and might predict treatment response. This study aims to correlate molecular subtypes in metastatic PDAC with FOLFIRINOX responses using real-world data, providing assistance in counselling patients. We collected 131 RNA-sequenced metastatic biopsies and applied a network-based meta-analysis using published PDAC classifiers. Subsequent survival analysis was performed using the most suitable classifier. For validation, we developed an immunohistochemistry (IHC) classifier using GATA6 and keratin-17 (KRT17), and applied it to 86 formalin-fixed paraffin-embedded samples of advanced PDAC. Lastly, GATA6 knockdown models were generated in PDAC organoids and cell lines. We showed that the PurIST classifier was the most suitable classifier. With this classifier, classical tumors had longer PFS and OS than basal-like tumors (PFS: 216 vs. 78 days, p = 0.0002; OS: 251 vs. 195 days, p = 0.049). The validation cohort showed a similar trend. Importantly, IHC GATA6low patients had significantly shorter survival with FOLFIRINOX (323 vs. 746 days, p = 0.006), but no difference in non-treated patients (61 vs. 54 days, p = 0.925). This suggests that GATA6 H-score predicts therapy response. GATA6 knockdown models did not lead to increased FOLFIRINOX responsiveness. These data suggest a predictive role for subtyping (transcriptomic and GATA6 IHC), though no direct causal relationship was found between GATA6 expression and chemoresistance. GATA6 immunohistochemistry should be seamlessly added to current diagnostics and integrated into upcoming clinical trials.
Collapse
Affiliation(s)
- Marjolein F Lansbergen
- Amsterdam UMC, location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Amsterdam UMC, location University of Amsterdam, Medical Oncology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Mark P G Dings
- Amsterdam UMC, location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands; Oncode Institute, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Paul Manoukian
- Amsterdam UMC, location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Arantza Fariña
- Cancer Center Amsterdam, Amsterdam, the Netherlands; Amsterdam UMC, location University of Amsterdam, Pathology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Cynthia Waasdorp
- Amsterdam UMC, location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Gerrit K J Hooijer
- Cancer Center Amsterdam, Amsterdam, the Netherlands; Amsterdam UMC, location University of Amsterdam, Pathology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Joanne Verheij
- Cancer Center Amsterdam, Amsterdam, the Netherlands; Amsterdam UMC, location University of Amsterdam, Pathology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Jan Koster
- Amsterdam UMC, location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Danny A Zwijnenburg
- Amsterdam UMC, location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Johanna W Wilmink
- Amsterdam UMC, location University of Amsterdam, Medical Oncology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Jan Paul Medema
- Amsterdam UMC, location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands; Oncode Institute, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Frederike Dijk
- Cancer Center Amsterdam, Amsterdam, the Netherlands; Amsterdam UMC, location University of Amsterdam, Pathology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Hanneke W M van Laarhoven
- Amsterdam UMC, location University of Amsterdam, Medical Oncology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Maarten F Bijlsma
- Amsterdam UMC, location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands; Oncode Institute, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands.
| |
Collapse
|
219
|
Lencioni G, Gregori A, Toledo B, Rebelo R, Immordino B, Amrutkar M, Xavier CPR, Kocijančič A, Pandey DP, Perán M, Castaño JP, Walsh N, Giovannetti E. Unravelling the complexities of resistance mechanism in pancreatic cancer: Insights from in vitro and ex-vivo model systems. Semin Cancer Biol 2024; 106-107:217-233. [PMID: 39299411 DOI: 10.1016/j.semcancer.2024.09.002] [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: 07/19/2024] [Revised: 09/07/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer with poor prognosis and rising global deaths. Late diagnosis, due to absent early symptoms and biomarkers, limits treatment mainly to chemotherapy, which soon encounters resistance. PDAC treatment innovation is hampered by its complex and heterogeneous resistant nature, including mutations in key genes and a stromal-rich, immunosuppressive tumour microenvironment. Recent studies on PDAC resistance stress the need for suitable in vitro and ex vivo models to replicate its complex molecular and microenvironmental landscape. This review summarises advances in these models, which can aid in combating chemoresistance and serve as platforms for discovering new therapeutics. Immortalised cell lines offer homogeneity, unlimited proliferation, and reproducibility, but while many gemcitabine-resistant PDAC cell lines exist, fewer models are available for resistance to other drugs. Organoids from PDAC patients show promise in mimicking tumour heterogeneity and chemosensitivity. Bioreactors, co-culture systems and organotypic slices, incorporating stromal and immune cells, are being developed to understand tumour-stroma interactions and the tumour microenvironment's role in drug resistance. Lastly, another innovative approach is three-dimensional bioprinting, which creates tissue-like structures resembling PDAC architecture, allowing for drug screening. These advanced models can guide researchers in selecting optimal in vitro tests, potentially improving therapeutic strategies and patient outcomes.
Collapse
Affiliation(s)
- Giulia Lencioni
- Fondazione Pisana per La Scienza, San Giuliano Terme, Italy; Department of Biology, University of Pisa, Pisa, Italy
| | - Alessandro Gregori
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands; Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Belén Toledo
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands; Department of Health Sciences, University of Jaén, Campus Lagunillas, Jaén E-23071, Spain
| | - Rita Rebelo
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands; Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto 4200-135, Portugal; Cancer Drug Resistance Group, Institute of Molecular Pathology and Immunology (IPATIMUP), University of Porto, Porto 4200-135, Portugal; Department of Biological Sciences, Faculty of Pharmacy of the University of Porto (FFUP), Porto, Portugal
| | - Benoît Immordino
- Fondazione Pisana per La Scienza, San Giuliano Terme, Italy; Institute of Life Sciences, Sant'Anna School of Advanced Studies, Pisa, Italy
| | - Manoj Amrutkar
- Department of Pathology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Cristina P R Xavier
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto 4200-135, Portugal; Cancer Drug Resistance Group, Institute of Molecular Pathology and Immunology (IPATIMUP), University of Porto, Porto 4200-135, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Gandra, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, University Institute of Health Sciences - CESPU, Gandra, Portugal
| | - Anja Kocijančič
- Centre for Embryology and Healthy Development, Department of Microbiology, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Deo Prakash Pandey
- Centre for Embryology and Healthy Development, Department of Microbiology, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Macarena Perán
- Department of Health Sciences, University of Jaén, Campus Lagunillas, Jaén E-23071, Spain; Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain; Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, Spain
| | - Justo P Castaño
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain; Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain; Reina Sofia University Hospital, Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | - Naomi Walsh
- Life Sciences Institute, School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Elisa Giovannetti
- Fondazione Pisana per La Scienza, San Giuliano Terme, Italy; Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands; Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
220
|
Carconi C, Bosi C, Scartozzi M, Cergnul M, Cinausero M, Faloppi L, Garajova I, Lonardi S, Pecora I, Pisanu L, Spadi R, Spallanzani A, Peretti U, Macchini M, Orsi G, Reni M. A pilot study of chlorambucil in pre-treated metastatic pancreatic adenocarcinoma patients bearing germline BRCA or other DNA damage repair system variants. Pancreatology 2024; 24:1066-1072. [PMID: 39277480 DOI: 10.1016/j.pan.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/27/2024] [Accepted: 09/08/2024] [Indexed: 09/17/2024]
Abstract
BACKGORUND Pancreatic adenocarcinoma remains a malignancy with a grim prognosis and scarce personalized treatment options. Pathogenic variants of DNA damage repair (DDR) genes are emerging as molecular targets, as they confer a higher sensitivity to DNA-damaging agents. This study aimed at assessing the activity of chlorambucil as salvage therapy in metastatic pancreatic cancer patients bearing a germline pathogenetic variant or variant of uncertain significance on a DDR-related gene. METHODS Platinum-pretreated metastatic pancreatic cancer patients harbouring a germline variant on a DDR gene received chlorambucil at a daily oral dose of 6 mg/m2 for 42 every 56 days for the first cycle and for 14 every 28 days for the following cycles, until disease progression or unacceptable toxicity. The primary endpoint was 6-month progression-free survival rate (PFS-6). Median progression-free survival (PFS) and overall survival (OS) were secondarily described. RESULTS Twenty patients were enrolled between December 2020 and September 2022. PFS-6 was 5%, median PFS and OS were 1.6 months and 3.0 months, respectively. Grade-3 adverse events were observed in 25% of patients, while no Grade-4 toxicity was reported. CONCLUSIONS Single agent chlorambucil did not show sufficient signal of activity to warrant its further investigation in metastatic pancreatic cancer patients bearing a DDR-related germline alteration.
Collapse
Affiliation(s)
- Catia Carconi
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Carlo Bosi
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Mario Scartozzi
- Medical Oncology, University and University Hospital, Cagliari, Italy
| | - Massimiliano Cergnul
- Medical Oncology, Ospedale Civile di Legnano - ASST Ovest Milanese, Legnano, Italy
| | - Marika Cinausero
- Department of Oncology, Azienda Sanitaria Universitaria Friuli Centrale Santa Maria Della Misericordia, Udine, Italy
| | - Luca Faloppi
- Medical Oncology Unit, Ospedali Santa Maria Della Pietà e Bartolomeo Eustachio - AST di Macerata, Camerino, San Severino Marche, Italy
| | - Ingrid Garajova
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Sara Lonardi
- Department of Oncology, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Irene Pecora
- Department of Medical Oncology, Azienda Toscana Sud-Est, Misericordia Hospital, Grosseto, Italy
| | | | - Rosella Spadi
- Department of Oncology, Medical Oncology, 1, Città Della Salute e Della Scienza, Turin, Italy
| | - Andrea Spallanzani
- Division of Oncology, Department of Oncology and Hematology, University Hospital Modena, Modena, Italy
| | - Umberto Peretti
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Marina Macchini
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Giulia Orsi
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Michele Reni
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
221
|
Rodríguez-Olivares JL, Kimball TN, Jeter JM, De-La-Mora-Molina H, Núñez I, Weitzel JN, Chávarri-Guerra Y. Prevalence and spectrum of germline pathogenic variants in cancer susceptibility genes among mexican patients with exocrine pancreatic cancer. Pancreatology 2024; 24:1049-1056. [PMID: 39327123 DOI: 10.1016/j.pan.2024.09.018] [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: 06/05/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Although universal germline genetic testing is recommended for patients with exocrine pancreatic cancer (PC), access to genetic testing remains limited in low- and middle-income countries. This study aims to narrow the gap in our understanding of the spectrum of germline pathogenic and likely pathogenic variants (PVs) in cancer susceptibility genes in the Mexican population. METHODS The landscape of PVs in cancer susceptibility genes was identified by next-generation sequencing multigene panel assays among patients with PC who were enrolled in the Clinical Cancer Genomics Community Research Network prospective registry in Mexico City. RESULTS From August 2019 to April 2023, 137 patients underwent genetic testing. The median age at diagnosis was 60 years (range 36-85), 58.4 % were women, and 38.7 % were metastatic at diagnosis. The frequency of germline PVs was 16 % (n = 22): ATM 36.4 % (n = 8), CDKN2A/p16INK4A 27.3 % (n = 6), BRCA2 9.1 % (n = 2), PALB2 9.1 % (n = 2), CHEK2 9.1 % (n = 2), TP53 4.5 % (n = 1), and NF1 4.5 % (n = 1). Additionally, 2 carriers of monoallelic germline variants in MUTYH were identified. No significant differences were observed between carriers and non-carriers in terms of family history of pancreatic cancer. CONCLUSIONS We identified a significant frequency of actionable germline PVs in Mexicans with PC, wherein the majority were in a broad spectrum of genes associated with the homologous recombination DNA repair mechanism. Most pancreatic cancer associated PVs were detected in non-BRCA genes, so our findings support the recommendation of multigene panel testing for genetic cancer risk assessment of Mexican individuals with PC.
Collapse
Affiliation(s)
- José Luis Rodríguez-Olivares
- Department of Hematology and Oncology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Tamara N Kimball
- Center for Genomic Medicine. Massachusetts General Hospital, Boston, MA, USA
| | - Joanne M Jeter
- Department of Oncology. City of Hope Cancer Center, Duarte, CA, USA
| | - Héctor De-La-Mora-Molina
- Department of Hematology and Oncology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Isaac Núñez
- Research Division. Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jeffrey N Weitzel
- Division of Precision Prevention, University of Kansas Comprehensive Cancer Center, Kansas City, USA
| | - Yanin Chávarri-Guerra
- Department of Hematology and Oncology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| |
Collapse
|
222
|
Bodeker KL, Smith BJ, Berg DJ, Chandrasekharan C, Sharif S, Fei N, Vollstedt S, Brown H, Chandler M, Lorack A, McMichael S, Wulfekuhle J, Wagner BA, Buettner GR, Allen BG, Caster JM, Dion B, Kamgar M, Buatti JM, Cullen JJ. A randomized trial of pharmacological ascorbate, gemcitabine, and nab-paclitaxel for metastatic pancreatic cancer. Redox Biol 2024; 77:103375. [PMID: 39369582 PMCID: PMC11491967 DOI: 10.1016/j.redox.2024.103375] [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: 08/28/2024] [Revised: 09/26/2024] [Accepted: 09/28/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND Patients with metastatic pancreatic ductal adenocarcinoma (PDAC) have poor 5-year survival. Pharmacological ascorbate (P-AscH-, high dose, intravenous, vitamin C) has shown promise as an adjunct to chemotherapy. We hypothesized adding P-AscH- to gemcitabine and nab-paclitaxel would increase survival in patients with metastatic PDAC. METHODS Patients diagnosed with stage IV pancreatic cancer randomized 1:1 to gemcitabine and nab-paclitaxel only (SOC, control) or to SOC with concomitant P-AscH-, 75 g three times weekly (ASC, investigational). The primary outcome was overall survival with secondary objectives of determining progression-free survival and adverse event incidence. Quality of life and patient reported outcomes for common oncologic symptoms were captured as an exploratory objective. Thirty-six participants were randomized; of this 34 received their assigned study treatment. All analyses were based on data frozen on December 11, 2023. RESULTS Intravenous P-AscH- increased serum ascorbate levels from micromolar to millimolar levels. P-AscH- added to the gemcitabine + nab-paclitaxel (ASC) increased overall survival to 16 months compared to 8.3 months with gemcitabine + nab-paclitaxel (SOC) (HR = 0.46; 90 % CI 0.23, 0.92; p = 0.030). Median progression free survival was 6.2 (ASC) vs. 3.9 months (SOC) (HR = 0.43; 90 % CI 0.20, 0.92; p = 0.029). Adding P-AscH- did not negatively impact quality of life or increase the frequency or severity of adverse events. CONCLUSIONS P-AscH- infusions of 75 g three times weekly in patients with metastatic pancreatic cancer prolongs overall and progression free survival without detriment to quality of life or added toxicity (ClinicalTrials.gov number NCT02905578).
Collapse
Affiliation(s)
- Kellie L Bodeker
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Brian J Smith
- College of Public Health, The University of Iowa, Iowa City, IA, USA
| | - Daniel J Berg
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Chandrikha Chandrasekharan
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Saima Sharif
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Naomi Fei
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Sandy Vollstedt
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Heather Brown
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Meghan Chandler
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Amanda Lorack
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Stacy McMichael
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Jared Wulfekuhle
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Brett A Wagner
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Garry R Buettner
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Bryan G Allen
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Joseph M Caster
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Barbara Dion
- Medical College of Wisconsin Division of Hematology and Oncology, Milwaukee, WI, USA
| | - Mandana Kamgar
- Medical College of Wisconsin Division of Hematology and Oncology, Milwaukee, WI, USA
| | - John M Buatti
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Joseph J Cullen
- Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA; Department of Surgery, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
223
|
Spatial mapping of primary and metastatic pancreatic tumor ecosystems. Nat Genet 2024; 56:2306-2307. [PMID: 39327488 DOI: 10.1038/s41588-024-01915-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
|
224
|
Jang JK, Byun JH, Choi SJ, Kim JH, Lee SS, Kim HJ, Yoo C, Kim KP, Hong SM, Seo DW, Hwang DW, Kim SC. Survival Outcomes According to NCCN Criteria for Resection Following Neoadjuvant Therapy for Patients with Localized Pancreatic Cancer. Ann Surg Oncol 2024:10.1245/s10434-024-16437-9. [PMID: 39485615 DOI: 10.1245/s10434-024-16437-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/18/2024] [Indexed: 11/03/2024]
Abstract
BACKGROUND This study aimed to assess the prognostic value of the National Comprehensive Cancer Network (NCCN) criteria for resection following neoadjuvant therapy for patients with localized pancreatic ductal adenocarcinoma (PDAC). METHODS This retrospective single-center study assessed 193 consecutive patients with localized PDAC (104 males and 89 females; mean age, 61.1 ± 9.4 years) who underwent neoadjuvant therapy followed by surgery between January 2010 and March 2021. Combined resectability and carbohydrate antigen (CA) 19-9 evaluation before and after neoadjuvant therapy was used to determine whether patients were eligible for resection according to the NCCN criteria. Post-surgical overall survival (OS), recurrence free survival (RFS), and pathologic results were evaluated and compared between patients considered eligible according to the NCCN criteria and those considered ineligible. Preoperative factors associated with better OS and RFS also were investigated. RESULTS Of the 193 patients, 168 (87.0 %) were eligible for resection according to the NCCN criteria. The patients eligible according to the NCCN criteria showed marginally longer OS than those considered ineligible (p = 0.056). After adjustment of variables, meeting the NCCN criteria for resection was an independent predictor of better OS (hazard ratio, 0.57; 95 % confidence interval, 0.34-0.96; p = 0.034). The two groups had similar RFS. Lower T-staging (T2 or less) and less lympho-vascular invasion and peri-neural invasion were noted in the patients who met the NCCN criteria (p ≤ 0.045). CONCLUSIONS The patients eligible for resection according to the NCCN criteria showed a trend toward longer OS and better pathologic results than the patients considered ineligible.
Collapse
Affiliation(s)
- Jong Keon Jang
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Jae Ho Byun
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea.
| | - Se Jin Choi
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Jin Hee Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Seung Soo Lee
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Hyoung Jung Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Changhoon Yoo
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Kyu-Pyo Kim
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Seung-Mo Hong
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Dong-Wan Seo
- Department of Gastroenterology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Dae Wook Hwang
- Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Song Cheol Kim
- Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| |
Collapse
|
225
|
Okano N, Kawai M, Ueno M, Yu X, Inoue Y, Takahashi S, Wang W, Takahashi H, Okamura Y, Morinaga S, Matsumoto I, Shimizu Y, Yoshida K, Yamamoto T, Ohtsuka M, Inokawa Y, Nara S, Tamura J, Shinoda S, Yamamoto K, Yamaue H, Furuse J. Outcomes of patients with initially unresectable pancreatic cancer who underwent conversion surgery after FOLFIRINOX or gemcitabine plus nab-paclitaxel chemotherapy: A multicenter retrospective cohort study (PC-CURE-1). JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2024; 31:816-829. [PMID: 39150050 PMCID: PMC11589395 DOI: 10.1002/jhbp.12066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
BACKGROUND The efficacy and safety of conversion surgery (CS) after FOLFIRINOX or gemcitabine plus nab-paclitaxel (GnP) chemotherapy in patients with initially unresectable pancreatic cancer (PC) remains unclear. METHODS This multicenter retrospective cohort study enrolled patients, between 2014 and 2018, with initially locally advanced or metastatic PC who were considered candidates for CS following FOLFIRINOX or GnP chemotherapy. They were classified into surgery (207 patients [194 resection and 13 exploratory laparotomy only]) and continued chemotherapy (10 patients, control) groups. The primary endpoint was overall survival (OS) from the day of diagnosis of potentially curative resection on imaging studies, with an expected hazard ratio (HR) of 0.7. RESULTS OS in the surgery group was longer than that in the control group (HR, 0.47; 95% confidence interval [CI]: 0.24-0.93). The median OS was 34.4 (95% CI: 27.9-43.4) and 19.8 (95% CI: 14.9-31.1) months in the surgery and control groups, respectively. The Clavien-Dindo grade ≥ IIIa postoperative complication and in-hospital mortality rates were 19.6% and 0.5%, respectively. Multivariate analysis revealed that preoperative chemotherapy duration was not associated with OS. CONCLUSIONS CS, following a favorable response to FOLFIRINOX or GnP chemotherapy, improved initially unresectable PC prognosis (specifically, OS), regardless of the chemotherapy duration.
Collapse
Affiliation(s)
- Naohiro Okano
- Department of Medical OncologyKyorin University Faculty of MedicineTokyoJapan
| | - Manabu Kawai
- Second Department of SurgeryWakayama Medical University School of MedicineWakayamaJapan
| | - Makoto Ueno
- Department of GastroenterologyKanagawa Cancer CenterYokohamaJapan
| | - Xianjun Yu
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
| | - Yosuke Inoue
- Division of Hepatobiliary and Pancreatic SurgeryCancer Institute Hospital, Japanese Foundation for Cancer ResearchTokyoJapan
| | - Shinichiro Takahashi
- Department of Hepatobiliary and Pancreatic SurgeryNational Cancer Center Hospital EastKashiwaJapan
| | - Wenquan Wang
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
| | - Hidenori Takahashi
- Department of Gastroenterological SurgeryOsaka International Cancer InstituteOsakaJapan
| | - Yukiyasu Okamura
- Division of Hepato‐Biliary‐Pancreatic SurgeryShizuoka Cancer Center HospitalShizuokaJapan
| | - Soichiro Morinaga
- Department of Hepato‐Biliary and Pancreatic SurgeryKanagawa Cancer CenterYokohamaJapan
| | - Ippei Matsumoto
- Department of SurgeryKindai University Faculty of MedicineOsakasayamaJapan
| | - Yasuhiro Shimizu
- Department of Gastroenterological SurgeryAichi Cancer Center HospitalNagoyaJapan
| | - Kazuhiro Yoshida
- Department of Surgical OncologyGifu University Graduate School of MedicineGifuJapan
| | | | - Masayuki Ohtsuka
- Department of General Surgery, Graduate School of MedicineChiba UniversityChibaJapan
| | - Yoshikuni Inokawa
- Department of Gastroenterological SurgeryNagoya University Graduate School of MedicineNagoyaJapan
| | - Satoshi Nara
- Department of Hepatobiliary and Pancreatic SurgeryNational Cancer Center HospitalTokyoJapan
| | - Jun Tamura
- Department of BiostatisticsYokohama City University School of MedicineYokohamaJapan
| | - Satoru Shinoda
- Department of BiostatisticsYokohama City University School of MedicineYokohamaJapan
| | - Kouji Yamamoto
- Department of BiostatisticsYokohama City University School of MedicineYokohamaJapan
| | - Hiroki Yamaue
- Second Department of SurgeryWakayama Medical University School of MedicineWakayamaJapan
| | - Junji Furuse
- Department of Medical OncologyKyorin University Faculty of MedicineTokyoJapan
- Department of GastroenterologyKanagawa Cancer CenterYokohamaJapan
| | | |
Collapse
|
226
|
Yu X, Pan H, He Q, Yang J, Xiao M, Xu J, Wang W, Yu X, Shi S. MXene-based dual-gated multifunctional nanodrug induced ferroptosis and modulated tumor microenvironment to treat pancreatic cancer. CHEMICAL ENGINEERING JOURNAL 2024; 500:157233. [DOI: 10.1016/j.cej.2024.157233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
|
227
|
Picozzi VJ. Treatment of advanced pancreatic cancer: When will the third time be a charm? Cancer 2024; 130:3626-3627. [PMID: 39138618 DOI: 10.1002/cncr.35499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Third‐line treatment of pancreatic cancer remains a major challenge for oncologists. What is its current status?
Collapse
Affiliation(s)
- Vincent J Picozzi
- Division of Hematology-Oncology, Virginia Mason Medical Center, Seattle, Washington, USA
| |
Collapse
|
228
|
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
|
229
|
Farshadi EA, Wang W, Mohammad F, van der Oost E, Doukas M, van Eijck CHJ, van de Werken HJG, Katsikis PD. Tumor organoids improve mutation detection of pancreatic ductal adenocarcinoma. Sci Rep 2024; 14:25468. [PMID: 39462012 PMCID: PMC11513084 DOI: 10.1038/s41598-024-75888-y] [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: 05/10/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) presents challenges in detecting somatic mutations due to its complex cellular composition. This study investigated the utility of patient-derived organoids (PDOs) to overcome these obstacles and enhance somatic mutation identification. Surgically resected PDAC tumors and their paired PDOs from 21 patients were examined. Whole-exome sequencing (WES) of tumor tissue, organoids, and peripheral blood mononuclear cells was performed to identify somatic mutations. Our findings demonstrate that PDOs retained about 80% of the somatic mutations from the original tumors, showing high concordance in mutation types. PDOs exhibited increased tumor purity and uncovered key driver mutations, aiding in identifying clinically relevant genomic alterations. Moreover, eight cycles of FOLFIRINOX treatment did not significantly alter the mutational landscape at the DNA level, indicating the stability of the mutational profile after therapeutic pressure in patients. In conclusion, PDOs are potentially important tools for exploring the somatic mutational landscape of PDAC. While they can reveal mutations that may be challenging to detect through traditional biopsy sequencing due to the inherently low tumor purity of PDAC, it is important to note that PDOs may not always fully recapitulate all mutations found in primary tumors. Despite this limitation, PDOs can still offer critical insights into the genomic complexities of PDAC, which is crucial for the development of personalized vaccines and therapies for this disease.
Collapse
Affiliation(s)
- Elham Aida Farshadi
- Department of Pulmonary Medicine, Erasmus University Medical Center, PO Box 2040, Rotterdam, 3000 CA, The Netherlands
| | - Wenya Wang
- Department of Immunology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, PO Box 2040, Rotterdam, 3000 CA, The Netherlands
| | - Farzana Mohammad
- Department of Pulmonary Medicine, Erasmus University Medical Center, PO Box 2040, Rotterdam, 3000 CA, The Netherlands
| | - Elise van der Oost
- Department of Surgery, Erasmus University Medical Center, PO Box 2040, Rotterdam, 3000 CA, The Netherlands
| | - Michail Doukas
- Department of Pathology, Erasmus University Medical Center, PO Box 2040, Rotterdam, 3000 CA, The Netherlands
| | - Casper H J van Eijck
- Department of Surgery, Erasmus University Medical Center, PO Box 2040, Rotterdam, 3000 CA, The Netherlands.
| | - Harmen J G van de Werken
- Department of Immunology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, PO Box 2040, Rotterdam, 3000 CA, The Netherlands.
| | - Peter D Katsikis
- Department of Immunology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, PO Box 2040, Rotterdam, 3000 CA, The Netherlands.
| |
Collapse
|
230
|
Chick RC, Pawlik TM. Updates in Immunotherapy for Pancreatic Cancer. J Clin Med 2024; 13:6419. [PMID: 39518557 PMCID: PMC11546190 DOI: 10.3390/jcm13216419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with limited effective therapeutic options. Due to a variety of cancer cell-intrinsic factors, including KRAS mutations, chemokine production, and other mechanisms that elicit a dysregulated host immune response, PDAC is often characterized by poor immune infiltration and an immune-privileged fibrotic stroma. As understanding of the tumor microenvironment (TME) evolves, novel therapies are being developed to target immunosuppressive mechanisms. Immune checkpoint inhibitors have limited efficacy when used alone or with radiation. Combinations of immune therapies, along with chemotherapy or chemoradiation, have demonstrated promise in preclinical and early clinical trials. Despite dismal response rates for immunotherapy for metastatic PDAC, response rates with neoadjuvant immunotherapy are somewhat encouraging, suggesting that incorporation of immunotherapy in the treatment of PDAC should be earlier in the disease course. Precision therapy for PDAC may be informed by advances in transcriptomic sequencing that can identify immunophenotypes, allowing for more appropriate treatment selection for each individual patient. Personalized and antigen-specific therapies are an increasing topic of interest, including adjuvant immunotherapy using personalized mRNA vaccines to prevent recurrence. Further development of personalized immune therapies will need to balance precision with generalizability and cost.
Collapse
Affiliation(s)
| | - Timothy M. Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA;
| |
Collapse
|
231
|
Wang X, Li D, Zhu B, Hua Z. Single-cell transcriptome analysis identifies a novel tumor-associated macrophage subtype predicting better prognosis in pancreatic ductal adenocarcinoma. Front Cell Dev Biol 2024; 12:1466767. [PMID: 39507421 PMCID: PMC11537994 DOI: 10.3389/fcell.2024.1466767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/14/2024] [Indexed: 11/08/2024] Open
Abstract
Background Characterized by an immune-suppressive tumor microenvironment (TME), pancreatic ductal adenocarcinoma (PDAC) is well-known for its poor prognosis. Tumor associated macrophages (TAMs) play a critical role in PDAC TME. An in-depth understanding of TAMs is helpful to develop new strategies for immunotherapy. Methods A large number of single-cell RNA sequencing data and bulk RNA sequencing data of PDAC were collected for systematic bioinformatics analysis. Characterize subtypes of TAMs at single-cell resolution and its effect on prognosis. Differential gene analysis and cell-cell communication were used to describe the effect on prognosis and validated by the TCGA dataset. Results We used two prognosis-favorable genes, SLC12A5 and ENPP2, to identify a benign M2-like TAMs (bM2-like TAMs), which shared similarities with C1QC + TAMs, CXCL9+ TAMs and CD169+ TAMs, by analyzing scRNA-seq data and bulk RNA data of PDAC. The bM2-like TAMs were revealed to promote T cell activation and proliferation through ALCAM/CD6 interaction. Meanwhile, the bM2-like TAMs were responsible for stroma modeling by altering αSMA+/αSMA-cell ratio. On the contrast, the rest of the M2-like TAMs were defined as malignant M2-like TAMs (mM2-like TAMs), partly overlapping with SPP1+ TAMs. mM2-like TAMs were revealed to promote tumor progression by secretion of MIF and SPP1. Conclusion Our study used two prognosis-favorable genes to divide M2-like TAMs of PDAC into anti-tumor bM2-like TAMs and pro-tumor mM2-like TAMs. The bM2-like TAMs activate T cells through ALCAM/CD6 and generate prognosis-favorable αSMA+ myofibroblasts through secreting TGFβ, which brings insight into heterogeneity of TAMs, prognosis prediction and immunotherapy of PDAC.
Collapse
Affiliation(s)
- Xiaonan Wang
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Dongyi Li
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Bo Zhu
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Zichun Hua
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
- Faculty of Pharmaceutical Sciences, Xinxiang Medical University, Xinxiang, China
- Changzhou High-Tech Research Institute, Nanjing University, Changzhou, China
| |
Collapse
|
232
|
Kim MK, Cho IR, Kim Y, Choi JH, Jung K, Kim J, Kim S, Yun H, Yoon J, Oh DY, Kim K, Lee SH. Prognostic value of the TP53 mutation in patients with pancreatic ductal adenocarcinoma receiving FOLFIRINOX. Ther Adv Med Oncol 2024; 16:17588359241290482. [PMID: 39449732 PMCID: PMC11500227 DOI: 10.1177/17588359241290482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024] Open
Abstract
Background KRAS, TP53, CDKN2A, and SMAD4 have been the main driver mutations in pancreatic ductal adenocarcinoma (PDAC). Studies on the clinical significance and treatment response to 5-fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) regimen in terms of the presence of these mutations remain inconclusive. Objectives This study aimed to compare the survival outcome and response to FOLFIRINOX chemotherapy based on the presence of four driver mutation genes. Design A multi-center retrospective study conducted at two tertiary medical centers. Methods This study analyzed PDAC patients who were treated with FOLFIRINOX chemotherapy as the initial treatment. Tumor specimens were analyzed by a targeted next-generation sequencing platform at two tertiary referral hospitals from January 2016 to March 2022. Patients' demographics, survival outcomes, and chemotherapeutic response were investigated and compared according to the presence of driver mutations. Results The analysis included 100 patients. KRAS mutation was identified in 92 (92.0%) patients, followed by TP53, CDKN2A, and SMAD4 in 63 (63.0%), 18 (18.0%), and 17 (17.0%) patients, respectively. The TP53 wild-type group demonstrated longer overall survival (OS) than the TP53 mutated group (median OS: 29 vs 19 months, p = 0.03), and TP53 served as a prognostic factor for survival (hazard ratio = 1.74, 95% confidence interval: 1.00-3.00, p = 0.048). The difference in OS according to TP53 mutation was intensified in localized pancreatic adenocarcinoma (37 vs 19 months, p = 0.01). The TP53 wild-type group demonstrated a higher objective response rate to FOLFIRINOX chemotherapy than the TP53 mutation group in localized pancreatic adenocarcinoma (50.0% vs 17.6%, p = 0.024). Conclusion PDAC patients with wild-type TP53 demonstrated longer OS than those with TP53 mutation, and this trend was intensified in patients with localized disease. This result may be due to an impaired response to FOLFIRINOX chemotherapy in patients with TP53 mutation.
Collapse
Affiliation(s)
- Min Kyu Kim
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - In Rae Cho
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yooeun Kim
- Interdisciplinary Program in Bioinformatics, College of Natural Science, Seoul National University, Seoul, Republic of Korea
| | - Jin Ho Choi
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kwangrok Jung
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam-si, Republic of Korea
| | - Jaihwan Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam-si, Republic of Korea
| | - Sheehyun Kim
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hongseok Yun
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jeesun Yoon
- Division of Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Do-Youn Oh
- Division of Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kwangsoo Kim
- Department of Transdisciplinary Medicine, Institute of Convergence Medicine with Innovative Technology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sang Hyub Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, 101, Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| |
Collapse
|
233
|
Dominguez AA, Perz MT, Xu Y, Cedillo LG, Huang OD, McIntyre CA, Vudatha V, Trevino JG, Liu J, Wang P. Unveiling the Promise: Navigating Clinical Trials 1978-2024 for PDAC. Cancers (Basel) 2024; 16:3564. [PMID: 39518005 PMCID: PMC11544830 DOI: 10.3390/cancers16213564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/14/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Despite many decades of research, pancreatic ductal adenocarcinoma (PDAC) remains one of the most difficult cancers to diagnose and treat effectively. Although there have been improvements in the 5-year overall survival rate, it is still very low at 12.5%. The limited efficacy of current therapies, even when PDAC is detected early, underscores the aggressive nature of the disease and the urgent need for more effective treatments. Clinical management of PDAC still relies heavily on a limited repertoire of therapeutic interventions, highlighting a significant gap between research efforts and available treatments. Over 4300 clinical trials have been or are currently investigating different treatment modalities and diagnostic strategies for PDAC, including targeted therapies, immunotherapies, and precision medicine approaches. These trials aim to develop more effective treatments and improve early detection methods through advanced imaging techniques and blood-based biomarkers. This review seeks to categorize and analyze PDAC-related clinical trials across various dimensions to understand why so few chemotherapeutic options are available to patients despite the numerous trials being conducted. This review aims to provide a comprehensive and nuanced understanding of the landscape of PDAC-related clinical trials, with the overarching goal of identifying opportunities to accelerate progress in drug development and improve patient outcomes in the fight against this devastating disease.
Collapse
Affiliation(s)
- Angel A. Dominguez
- Department of Cell Systems & Anatomy; University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.A.D.); (M.T.P.); (Y.X.); (L.G.C.); (O.D.H.); (J.L.)
| | - Matthew T. Perz
- Department of Cell Systems & Anatomy; University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.A.D.); (M.T.P.); (Y.X.); (L.G.C.); (O.D.H.); (J.L.)
| | - Yi Xu
- Department of Cell Systems & Anatomy; University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.A.D.); (M.T.P.); (Y.X.); (L.G.C.); (O.D.H.); (J.L.)
| | - Leonor G. Cedillo
- Department of Cell Systems & Anatomy; University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.A.D.); (M.T.P.); (Y.X.); (L.G.C.); (O.D.H.); (J.L.)
| | - Orry D. Huang
- Department of Cell Systems & Anatomy; University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.A.D.); (M.T.P.); (Y.X.); (L.G.C.); (O.D.H.); (J.L.)
| | - Caitlin A. McIntyre
- Division of Surgical Oncology and Endocrine Surgery, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA;
| | - Vignesh Vudatha
- Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA; (V.V.); (J.G.T.)
| | - Jose G. Trevino
- Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA; (V.V.); (J.G.T.)
| | - Jun Liu
- Department of Cell Systems & Anatomy; University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.A.D.); (M.T.P.); (Y.X.); (L.G.C.); (O.D.H.); (J.L.)
| | - Pei Wang
- Department of Cell Systems & Anatomy; University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.A.D.); (M.T.P.); (Y.X.); (L.G.C.); (O.D.H.); (J.L.)
| |
Collapse
|
234
|
Meng F, Li T, Singh AK, Wang Y, Attiyeh M, Kohram F, Feng Q, Li YR, Shen B, Williams T, Liu Y, Raoof M. Base-excision repair pathway regulates transcription-replication conflicts in pancreatic ductal adenocarcinoma. Cell Rep 2024; 43:114820. [PMID: 39368091 DOI: 10.1016/j.celrep.2024.114820] [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: 11/24/2023] [Revised: 07/19/2024] [Accepted: 09/18/2024] [Indexed: 10/07/2024] Open
Abstract
Oncogenic mutations (such as in KRAS) can dysregulate transcription and replication, leading to transcription-replication conflicts (TRCs). Here, we demonstrate that TRCs are enriched in human pancreatic ductal adenocarcinoma (PDAC) compared to other common solid tumors or normal cells. Several orthogonal approaches demonstrated that TRCs are oncogene dependent. A small interfering RNA (siRNA) screen identified several factors in the base-excision repair (BER) pathway as main regulators of TRCs in PDAC cells. Inhibitors of BER pathway (methoxyamine and CRT) enhanced TRCs. Mechanistically, BER pathway inhibition severely altered RNA polymerase II (RNAPII) and R-loop dynamics at nascent DNA, causing RNAPII trapping and contributing to enhanced TRCs. The ensuing DNA damage activated the ATR-Chk1 pathway. Co-treatment with ATR inhibitor (VX970) and BER inhibitor (methoxyamine) at clinically relevant doses synergistically enhanced DNA damage and reduced cell proliferation in PDAC cells. The study provides mechanistic insights into the regulation of TRCs in PDAC by the BER pathway, which has biologic and therapeutic implications.
Collapse
Affiliation(s)
- Fan Meng
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Tiane Li
- Irell & Manella Graduate School of Biological Sciences, City of Hope National Medical Center, Duarte, CA, USA; Department of Cancer Genetic & Epigenetics, City of Hope National Medical Center, Duarte, CA, USA
| | | | - Yingying Wang
- Department of Cancer Genetic & Epigenetics, City of Hope National Medical Center, Duarte, CA, USA
| | - Marc Attiyeh
- Department of Surgery, Cedars Sinai, Los Angeles, CA, USA
| | - Fatemeh Kohram
- Department of Child Health, University of Arizona College of Medicine, Phoenix, AZ, USA
| | - Qianhua Feng
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Yun R Li
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Binghui Shen
- Department of Cancer Genetic & Epigenetics, City of Hope National Medical Center, Duarte, CA, USA
| | - Terence Williams
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Yilun Liu
- Department of Cancer Genetic & Epigenetics, City of Hope National Medical Center, Duarte, CA, USA
| | - Mustafa Raoof
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA; Department of Cancer Genetic & Epigenetics, City of Hope National Medical Center, Duarte, CA, USA.
| |
Collapse
|
235
|
Araki T, Kawahira M, Shimokawa M, Otsuka T, Hayashi K, Sonoda Y, Honda T, Nakao K, Shibuki T, Nakazawa J, Arima S, Fukahori M, Miwa K, Koga F, Ueda Y, Kubotsu Y, Makiyama A, Shimokawa H, Takeshita S, Nishikawa K, Komori A, Otsu S, Hosokawa A, Sakai T, Oda H, Arita S, Taguchi H, Tsuneyoshi K, Kawaguchi Y, Fujita T, Sakae T, Nio K, Ide Y, Ureshino N, Shirakawa T, Mizuta T, Mitsugi K. Real-World Analysis of the Correlation between Overall Survival and Progression-Free Survival in Advanced Pancreatic Cancer: Results of NAPOLEON-1 and 2 Studies. Oncology 2024:1-11. [PMID: 39427640 DOI: 10.1159/000542137] [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: 08/13/2024] [Accepted: 10/11/2024] [Indexed: 10/22/2024]
Abstract
INTRODUCTION Fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) improve overall survival (OS) and progression-free survival (PFS) in patients with pancreatic cancer, compared with gemcitabine (GEM). However, whether PFS is a surrogate marker of OS in pancreatic cancer chemotherapy focusing on FOLFIRINOX or GEM plus nab-paclitaxel remains unknown. We aimed to verify whether PFS can be a surrogate marker of OS in prognosis prediction. METHODS This was an integrated analysis of the NAPOLEON study and retrospective cohort of the NAPOLEON-2 study - a multicenter observational study conducted in Japan, using real-world data. The primary and secondary endpoints were OS and PFS, respectively. The correlation between OS and PFS in first- and second-line treatments was assessed using Method of Moments estimation. An analysis was performed in patients with confirmed OS at the end of follow-up. The NAPOLEON-2 cohort included only patients who received 5-fluorouracil, leucovorin, and nanoliposomal irinotecan (NFF) as second-line treatment. RESULTS Among 479 patients, the correlation between PFS and OS from first- and second-line chemotherapies was calculated in 310 and 225 patients, respectively. The R-squared values for the correlation between PFS and OS from first- and second-line chemotherapies were 0.74 and 0.76, respectively. There was no statistically significant difference in first-line treatment between the FOLFIRINOX and GEM plus nab-paclitaxel groups (p = 0.92). Therefore, the FOLFIRINOX group may not have shown a stronger correlation than the NFF group. CONCLUSION PFS can be a surrogate marker of OS in first- and second-line therapies. Appropriate prognostic estimation might contribute to proper treatment selection.
Collapse
Affiliation(s)
- Tomonori Araki
- Department of Gastroenterology and Hepatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan,
| | - Machiko Kawahira
- Department of Gastroenterology, Kagoshima Kouseiren Hospital, Kagoshima, Japan
| | - Mototsugu Shimokawa
- Clinical Research Institute, National Kyushu Cancer Center, Fukuoka, Japan
- Department of Biostatistics, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Taiga Otsuka
- Department of Medical Oncology, Saga-Ken Medical Center Koseikan, Saga, Japan
- Department of Internal Medicine, Minato Medical Clinic, Fukuoka, Japan
| | - Kohei Hayashi
- Department of Gastroenterology and Hepatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yuki Sonoda
- Department of Gastroenterology and Hepatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Takuya Honda
- Department of Gastroenterology and Hepatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Kazuhiko Nakao
- Department of Gastroenterology and Hepatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Taro Shibuki
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Junichi Nakazawa
- Department of Medical Oncology, Kagoshima City Hospital, Kagoshima, Japan
| | - Shiho Arima
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Masaru Fukahori
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Fukuoka, Japan
| | - Keisuke Miwa
- Multidisciplinary Treatment Cancer Center, Kurume University Hospital, Fukuoka, Japan
| | - Futa Koga
- Department of Hepatobiliary and Pancreatology, Saga Medical Center Koseikan, Saga, Japan
| | - Yujiro Ueda
- Department of Hematology and Oncology, Japanese Red Cross Kumamoto Hospital, Kumamoto, Japan
| | - Yoshihito Kubotsu
- Department of Internal Medicine, Karatsu Red Cross Hospital, Saga, Japan
| | - Akitaka Makiyama
- Department of Hematology Oncology, Japan Community Healthcare Organization Kyushu Hospital, Fukuoka, Japan
| | - Hozumi Shimokawa
- Department of Hematology Oncology, Japan Community Healthcare Organization Kyushu Hospital, Fukuoka, Japan
| | - Shigeyuki Takeshita
- Department of Gastroenterology, Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki, Japan
| | - Kazuo Nishikawa
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, Oita, Japan
| | - Azusa Komori
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, Oita, Japan
| | - Satoshi Otsu
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, Oita, Japan
| | - Ayumu Hosokawa
- Department of Clinical Oncology, University of Miyazaki Hospital, Miyazaki, Japan
| | - Tatsunori Sakai
- Department of Medical Oncology, National Hospital Organization Kumamoto Medical Center, Kumamoto, Japan
| | - Hisanobu Oda
- Division of Integrative Medical Oncology, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| | - Shuji Arita
- Department of Chemotherapy, Miyazaki Prefectural Miyazaki Hospital, Miyazaki, Japan
| | - Hiroki Taguchi
- Department of Gastroenterology, Saiseikai Sendai Hospital, Kagoshima, Japan
| | - Kengo Tsuneyoshi
- Department of Gastroenterology, Izumi General Medical Center, Kagoshima, Japan
| | - Yasunori Kawaguchi
- Department of Gastroenterology, Asakura Medical Association Hospital, Fukuoka, Japan
| | - Toshihiro Fujita
- Department of Gastroenterology, Saiseikai Sendai Hospital, Kagoshima, Japan
| | - Takahiro Sakae
- Department of Gastroenterology, Saiseikai Sendai Hospital, Kagoshima, Japan
| | - Kenta Nio
- Department of Medical Oncology, Sasebo Kyosai Hospital, Nagasaki, Japan
| | - Yasushi Ide
- Department of Internal Medicine, Karatsu Red Cross Hospital, Saga, Japan
| | - Norio Ureshino
- Department of Medical Oncology, Saga-Ken Medical Center Koseikan, Saga, Japan
| | - Tsuyoshi Shirakawa
- Clinical Hematology Oncology Treatment Study Group, Fukuoka, Japan
- Eikoh Hospital, Fukuoka, Japan
| | - Toshihiko Mizuta
- Department of Internal Medicine, Imari Arita Kyoritsu Hospital, Saga, Japan
| | - Kenji Mitsugi
- Department of Medical Oncology, Sasebo Kyosai Hospital, Nagasaki, Japan
- Department of Medical Oncology, Hamanomachi Hospital, Fukuoka, Japan
| |
Collapse
|
236
|
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
|
237
|
Geisler A, Dieringer B, Elsner L, Klopfleisch R, Kurreck J, Fechner H. Oncolytic Coxsackievirus B3 Strain PD-H Is Effective Against a Broad Spectrum of Pancreatic Cancer Cell Lines and Induces a Growth Delay in Pancreatic KPC Cell Tumors In Vivo. Int J Mol Sci 2024; 25:11224. [PMID: 39457005 PMCID: PMC11508574 DOI: 10.3390/ijms252011224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/10/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Pancreatic cancer is one of the deadliest cancers globally, with limited success from existing therapies, including chemotherapies and immunotherapies like checkpoint inhibitors for patients with advanced pancreatic ductal adenocarcinoma (PDAC). A promising new approach is the use of oncolytic viruses (OV), a form of immunotherapy that has been demonstrated clinical effectiveness in various cancers. Here we investigated the potential of the oncolytic coxsackievirus B3 strain (CVB3) PD-H as a new treatment for pancreatic cancer. In vitro, PD-H exhibited robust replication, as measured by plaque assays, and potent lytic activity, as assessed by XTT assays, in most pancreatic tumor cell lines, outperforming two other coxsackievirus strains tested, H3N-375/1TS and CVA21. Thus, H3N-375/1TS showed efficient replication and lytic efficiency in distinctly fewer tumor cell lines, while most tumor cells were resistant to CVA21. The oncolytic efficiency of the three OV largely correlated with mRNA expression levels of viral receptors and their ability to induce apoptosis, as measured by cleaved caspase 3/7 activity in the tumor cells. In a syngeneic mouse model with subcutaneous pancreatic tumors, intratumoral administration of PD-H significantly inhibited tumor growth but did not completely stop tumor progression. Importantly, no virus-related side effects were observed. Although pancreatic tumors respond to PD-H treatment, its therapeutic efficacy is limited. Combining PD-H with other treatments, such as those aiming at reducing the desmoplastic stroma which impedes viral infection and spread within the tumor, may enhance its efficacy.
Collapse
Affiliation(s)
- Anja Geisler
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, 10623 Berlin, Germany (H.F.)
| | - Babette Dieringer
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, 10623 Berlin, Germany (H.F.)
| | - Leslie Elsner
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, 10623 Berlin, Germany (H.F.)
| | - Robert Klopfleisch
- Institute of Veterinary Pathology, Freie Universität Berlin, 14163 Berlin, Germany
| | - Jens Kurreck
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, 10623 Berlin, Germany (H.F.)
| | - Henry Fechner
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, 10623 Berlin, Germany (H.F.)
| |
Collapse
|
238
|
Wang J, Yang J, Narang A, He J, Wolfgang C, Li K, Zheng L. Consensus, debate, and prospective on pancreatic cancer treatments. J Hematol Oncol 2024; 17:92. [PMID: 39390609 PMCID: PMC11468220 DOI: 10.1186/s13045-024-01613-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: 08/16/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024] Open
Abstract
Pancreatic cancer remains one of the most aggressive solid tumors. As a systemic disease, despite the improvement of multi-modality treatment strategies, the prognosis of pancreatic cancer was not improved dramatically. For resectable or borderline resectable patients, the surgical strategy centered on improving R0 resection rate is consensus; however, the role of neoadjuvant therapy in resectable patients and the optimal neoadjuvant therapy of chemotherapy with or without radiotherapy in borderline resectable patients were debated. Postoperative adjuvant chemotherapy of gemcitabine/capecitabine or mFOLFIRINOX is recommended regardless of the margin status. Chemotherapy as the first-line treatment strategy for advanced or metastatic patients included FOLFIRINOX, gemcitabine/nab-paclitaxel, or NALIRIFOX regimens whereas 5-FU plus liposomal irinotecan was the only standard of care second-line therapy. Immunotherapy is an innovative therapy although anti-PD-1 antibody is currently the only agent approved by for MSI-H, dMMR, or TMB-high solid tumors, which represent a very small subset of pancreatic cancers. Combination strategies to increase the immunogenicity and to overcome the immunosuppressive tumor microenvironment may sensitize pancreatic cancer to immunotherapy. Targeted therapies represented by PARP and KRAS inhibitors are also under investigation, showing benefits in improving progression-free survival and objective response rate. This review discusses the current treatment modalities and highlights innovative therapies for pancreatic cancer.
Collapse
Affiliation(s)
- Junke Wang
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Jie Yang
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Amol Narang
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Jin He
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Christopher Wolfgang
- Department of Surgery, New York University School of Medicine and NYU-Langone Medical Center, New York, NY, USA
| | - Keyu Li
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA.
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| | - Lei Zheng
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA.
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- The Multidisciplinary Gastrointestinal Cancer Laboratories Program, the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| |
Collapse
|
239
|
Bangash AA, Alvi SS, Bangash MA, Ahsan H, Khan S, Shareef R, Villanueva G, Bansal D, Ahmad M, Kim DJ, Chauhan SC, Hafeez BB. Honey Targets Ribosome Biogenesis Components to Suppress the Growth of Human Pancreatic Cancer Cells. Cancers (Basel) 2024; 16:3431. [PMID: 39410048 PMCID: PMC11475701 DOI: 10.3390/cancers16193431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 10/20/2024] Open
Abstract
Pancreatic cancer (PanCa) is one of the deadliest cancers, with limited therapeutic response. Various molecular oncogenic events, including dysregulation of ribosome biogenesis, are linked to the induction, progression, and metastasis of PanCa. Thus, the discovery of new therapies suppressing these oncogenic events and ribosome biogenesis could be a novel therapeutic approach for the prevention and treatment of PanCa. The current study was designed to investigate the anti-cancer effect of honey against PanCa. Our results indicated that honey markedly inhibited the growth and invasive characteristics of pancreatic cancer cells by suppressing the mRNA expression and protein levels of key components of ribosome biogenesis, including RNA Pol-I subunits (RPA194 and RPA135) along with its transcriptional regulators, i.e., UBTF and c-Myc. Honey also induced nucleolar stress in PanCa cells by reducing the expression of various nucleolar proteins (NCL, FBL, and NPM). Honey-mediated regulation on ribosome biogenesis components and nucleolar organization-associated proteins significantly arrested the cell cycle in the G2M phase and induced apoptosis in PanCa cells. These results, for the first time, demonstrated that honey, being a natural remedy, has the potential to induce apoptosis and inhibit the growth and metastatic phenotypes of PanCa by targeting ribosome biogenesis.
Collapse
Affiliation(s)
- Aun Ali Bangash
- South Texas Center of Excellence for Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (A.A.B.); (M.A.B.); (H.A.); (S.K.); (R.S.); (G.V.); (M.A.); (D.J.K.); (S.C.C.)
- Department of Medicine and Oncology ISU, Division of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Sahir Sultan Alvi
- South Texas Center of Excellence for Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (A.A.B.); (M.A.B.); (H.A.); (S.K.); (R.S.); (G.V.); (M.A.); (D.J.K.); (S.C.C.)
- Department of Medicine and Oncology ISU, Division of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Muhammad Ali Bangash
- South Texas Center of Excellence for Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (A.A.B.); (M.A.B.); (H.A.); (S.K.); (R.S.); (G.V.); (M.A.); (D.J.K.); (S.C.C.)
- Department of Medicine and Oncology ISU, Division of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Haider Ahsan
- South Texas Center of Excellence for Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (A.A.B.); (M.A.B.); (H.A.); (S.K.); (R.S.); (G.V.); (M.A.); (D.J.K.); (S.C.C.)
- Department of Medicine and Oncology ISU, Division of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Shiza Khan
- South Texas Center of Excellence for Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (A.A.B.); (M.A.B.); (H.A.); (S.K.); (R.S.); (G.V.); (M.A.); (D.J.K.); (S.C.C.)
- Department of Medicine and Oncology ISU, Division of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Rida Shareef
- South Texas Center of Excellence for Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (A.A.B.); (M.A.B.); (H.A.); (S.K.); (R.S.); (G.V.); (M.A.); (D.J.K.); (S.C.C.)
- Department of Medicine and Oncology ISU, Division of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Georgina Villanueva
- South Texas Center of Excellence for Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (A.A.B.); (M.A.B.); (H.A.); (S.K.); (R.S.); (G.V.); (M.A.); (D.J.K.); (S.C.C.)
- Department of Medicine and Oncology ISU, Division of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Divyam Bansal
- Department of Kinesiology, Rice University, Houston, TX 77251, USA;
| | - Mudassier Ahmad
- South Texas Center of Excellence for Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (A.A.B.); (M.A.B.); (H.A.); (S.K.); (R.S.); (G.V.); (M.A.); (D.J.K.); (S.C.C.)
- Department of Medicine and Oncology ISU, Division of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Dae Joon Kim
- South Texas Center of Excellence for Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (A.A.B.); (M.A.B.); (H.A.); (S.K.); (R.S.); (G.V.); (M.A.); (D.J.K.); (S.C.C.)
- Department of Medicine and Oncology ISU, Division of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Subhash C. Chauhan
- South Texas Center of Excellence for Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (A.A.B.); (M.A.B.); (H.A.); (S.K.); (R.S.); (G.V.); (M.A.); (D.J.K.); (S.C.C.)
- Department of Medicine and Oncology ISU, Division of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Bilal Bin Hafeez
- South Texas Center of Excellence for Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (A.A.B.); (M.A.B.); (H.A.); (S.K.); (R.S.); (G.V.); (M.A.); (D.J.K.); (S.C.C.)
- Department of Medicine and Oncology ISU, Division of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| |
Collapse
|
240
|
Tsai HJ, Yang SH, Hsiao CF, Kao HF, Su YY, Shan YS, Yen CJ, Du JS, Hsu C, Wu IC, Chen LT. A phase 1 study of biweekly nab-paclitaxel/oxaliplatin/S-1/LV for advanced upper gastrointestinal cancers: TCOG T1216 study. Oncologist 2024; 29:e1396-e1405. [PMID: 38902994 PMCID: PMC11449045 DOI: 10.1093/oncolo/oyae109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 04/19/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Oxaliplatin- and fluoropyrimidine-based triplet regimens have demonstrated feasibility and efficacy in the treatment of upper gastrointestinal (UGI) cancers. Herein, we evaluate the feasibility and preliminary efficacy of biweekly nab-paclitaxel plus oxaliplatin and S-1/leucovorin (SOLAR) in chemonaïve UGI cancers. METHODS A 3 + 3 phase 1 study was conducted to determine the maximal tolerated dose (MTD) of oxaliplatin in SOLAR (nab-paclitaxel [150 mg/m2 in D1], oxaliplatin [60, 75, or 85 mg/m2 in D1], and oral S-1/leucovorin [35 mg/m2 and 30 mg bid from D1 to D7]). The secondary endpoints were overall response rate (ORR), progression-free survival (PFS), overall survival (OS), and safety. RESULTS Thirteen and 6 accruals were in the dose-escalation and MTD expansion cohorts, respectively. One of 6 patients at level III experienced dose-limiting toxicity (grade 3 diarrhea), which revealed that the MTD of oxaliplatin was 85 mg/m2. After a mean of 15.9 cycles of treatment, the most common treatment-related grade 3/4 toxicities were neutropenia (57.9%) and diarrhea (21.1%). The ORR was 63.2%. The median PFS and OS were 12.5 and 24.7 months, respectively. CONCLUSION The current study revealed the MTD of oxaliplatin and demonstrated the preliminary efficacy of SOLAR in UGI cancers, which deserves further investigation. CLINICALTRIALS.GOV IDENTIFIER NCT03162510.
Collapse
Affiliation(s)
- Hui-Jen Tsai
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shih-Hung Yang
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chin-Fu Hsiao
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Hsiang-Fong Kao
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
- National Taiwan University Cancer Center, Taipei, Taiwan
| | - Yung-Yeh Su
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yan-Shen Shan
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Jui Yen
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jeng-Shiun Du
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chiun Hsu
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - I-Chen Wu
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
241
|
Johnson R, McClelland PH, Ahmad SA. Neoadjuvant and Adjuvant Therapy in Resectable Pancreatic Adenocarcinoma. Surg Clin North Am 2024; 104:987-1005. [PMID: 39237173 DOI: 10.1016/j.suc.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
While pancreatic adenocarcinoma requires surgical resection definitive cure, treatment paradigms are shifting toward a neoadjuvant approach to systemic therapy. Rationale is twofold: micro-metastatic disease is likely present in a majority of patients, reinforcing the importance of systemic therapy regardless of resectability; moreover, systemic therapy is well-tolerated and improves surgical outcomes when delivered preoperatively. Second, a neoadjuvant approach allows for selection of biology and patients most likely to benefit from potentially morbid surgery. This review examines the increasing body of evidence in support of empiric neoadjuvant therapy in pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Ryan Johnson
- General Surgery, Division of Surgical Oncology, University of Cincinnati Cancer Institute, University of Cincinnati Medical Center, 231 Albert Sabin Way, ML 0558, Cincinnati, OH 45267, USA
| | - Paul H McClelland
- General Surgery, Division of Surgical Oncology, University of Cincinnati Cancer Institute, University of Cincinnati Medical Center, 231 Albert Sabin Way, ML 0558, Cincinnati, OH 45267, USA
| | - Syed A Ahmad
- Division of Surgical Oncology, University of Cincinnati Cancer Institute, University of Cincinnati Medical Center, 231 Albert Sabin Way, ML 0558, Cincinnati, OH 45267, USA.
| |
Collapse
|
242
|
Mas L, Castelli C, Coffy A, Tretarre B, Piquemal D, Bachet JB. Nationwide trends over 10 years in epidemiology and management of pancreatic ductal adenocarcinoma: A real-world study from the French administrative database. Clin Res Hepatol Gastroenterol 2024; 48:102426. [PMID: 39043316 DOI: 10.1016/j.clinre.2024.102426] [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: 05/17/2024] [Revised: 07/04/2024] [Accepted: 07/20/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND & AIMS Significant progress has been made in the management of pancreatic ductal adenocarcinoma (PDAC) in recent years. In this population-based study, we aimed to compare incidence, therapeutic strategies, and survival outcomes of PDAC patients in France over a decade. METHODS This study was performed using a nationwide French database. All patients receiving care for PDAC during years 2009, 2014 and 2018 were included. Treatment modalities and survival outcomes were analyzed. RESULTS A total of 8143/8771/10494 patients were considered in 2009/2014/2018, respectively. Incidence increased mainly among patients aged >60 years. In localized PDAC, the proportion of patients receiving best supportive care (BSC) only decreased at 43.6/36.4/32.4 % and 27.8/29.1/34.3 % received chemo(radio)therapy alone. The rate of upfront surgery remained stable while 3/8/18 % of operated patients received neoadjuvant therapy. Median overall survival (OS) was 7.0/7.9/8.5 months in the overall population. Among treated patients, 1-year OS was 61.4/67.7/68.8 % and 30.3/36.3/38.8 % for localized and metastatic PDAC, respectively. CONCLUSIONS This study confirms the rising incidence of PDAC. Improved outcomes were seen in localized PDAC, with a wider use of chemotherapy and neoadjuvant strategies, and in treated metastatic patients. A modest survival gain was seen overall, hindered by the still high rate of patients receiving BSC only.
Collapse
Affiliation(s)
- Léo Mas
- AP-HP, Hépato-Gastroenterology et Digestive Oncology department, Pitié-Salpêtrière University Hospital, Sorbonne University, 47-83 Boulevard de l'Hôpital, Paris 75013, France.
| | - Christel Castelli
- AESIO SANTE Department Clinical Research team, Beau Soleil clinic, Montpellier 34070, France
| | - Amandine Coffy
- AESIO SANTE Department Clinical Research team, Beau Soleil clinic, Montpellier 34070, France
| | | | | | - Jean-Baptiste Bachet
- AP-HP, Hépato-Gastroenterology et Digestive Oncology department, Pitié-Salpêtrière University Hospital, Sorbonne University, 47-83 Boulevard de l'Hôpital, Paris 75013, France
| |
Collapse
|
243
|
Dong B, Zhang Y, Gao H, Liu J, Li J. Machine Learning Developed a MYC Expression Feature-Based Signature for Predicting Prognosis and Chemoresistance in Pancreatic Adenocarcinoma. Biochem Genet 2024; 62:4191-4214. [PMID: 38245886 DOI: 10.1007/s10528-023-10625-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 12/07/2023] [Indexed: 01/23/2024]
Abstract
MYC has been identified to profoundly influence a wide range of pathologic processes in cancers. However, the prognostic value of MYC-related genes in pancreatic adenocarcinoma (PAAD) remains unclarified. Gene expression data and clinical information of PAAD patients were obtained from The Cancer Genome Atlas (TCGA) database (training set). Validation sets included GSE57495, GSE62452, and ICGC-PACA databases. LASSO regression analysis was used to develop a risk signature for survival prediction. Single-cell sequencing data from GSE154778 and CRA001160 datasets were analyzed. Functional studies were conducted using siRNA targeting RHOF and ITGB6 in PANC-1 cells. High MYC expression was found to be significantly associated with a poor prognosis in patients with PAAD. Additionally, we identified seven genes (ADGRG6, LINC00941, RHOF, SERPINB5, INSYN2B, ITGB6, and DEPDC1) that exhibited a strong correlation with both MYC expression and patient survival. They were then utilized to establish a risk model (MYCsig), which showed robust predictive ability. Furthermore, MYCsig demonstrated a positive correlation with the expression of HLA genes and immune checkpoints, as well as the chemotherapy response of PAAD. RHOF and ITGB6, expressed mainly in malignant cells, were identified as key oncogenes regulating chemosensitivity through EMT. Downregulation of RHOF and ITGB6 reduced cell proliferation and invasion in PANC-1 cells. The developed MYCsig demonstrates its potential in enhancing the management of patients with PAAD by facilitating risk assessment and predicting response to adjuvant chemotherapy. Additionally, our study identifies RHOF and ITGB6 as novel oncogenes linked to EMT and chemoresistance in PAAD.
Collapse
Affiliation(s)
- Biao Dong
- Department of Hepatobiliary Surgery, Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, 050000, Hebei, China
| | - Yueshan Zhang
- Department of Hepatobiliary Surgery, Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, 050000, Hebei, China
| | - Han Gao
- Department of Hepatobiliary Surgery, Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, 050000, Hebei, China
| | - Jia Liu
- Department of Precision Medicine, Accb Biotech. Ltd, Beijing, China
| | - Jiankun Li
- Department of Hepatobiliary Surgery, Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
244
|
Fields BC, Tzeng CWD. Locally Advanced Pancreas Cancer, Is There a Role for Surgery? Surg Clin North Am 2024; 104:1017-1030. [PMID: 39237161 PMCID: PMC11748233 DOI: 10.1016/j.suc.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Locally advanced pancreatic cancer (LAPC) represents a unique clinical scenario in which the tumor is considered localized but unresectable due to anatomic factors. Despite a consensus against upfront surgery, no standard approach to induction therapy exists for patients with LAPC. Extended systemic therapy has shown promise in establishing tumor response and remains the standard of care. While associated with improved local control, the timing and role of radiation therapy remain in question. Following adequate response to induction chemotherapy, a safe attempt at margin-negative resection can be considered. Special attention should be given to required vascular skeletonization and/or resection with reconstruction.
Collapse
Affiliation(s)
- Brittany C Fields
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1484, Houston, TX 77030, USA
| | - Ching-Wei D Tzeng
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1484, Houston, TX 77030, USA.
| |
Collapse
|
245
|
Ni R, Hu Z, Tao R. Advances of immune-checkpoint inhibition of CTLA-4 in pancreatic cancer. Biomed Pharmacother 2024; 179:117430. [PMID: 39260322 DOI: 10.1016/j.biopha.2024.117430] [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: 06/13/2024] [Revised: 09/05/2024] [Accepted: 09/05/2024] [Indexed: 09/13/2024] Open
Abstract
Targeting checkpoints for immune cell activation has been acknowledged known as one of the most effective way to activate anti-tumor immune responses. Among them, drugs targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) are approved for clinical treatment though several more are in advanced stages of development, which demonstrated durable response rates and manageable safety profile. However, its therapy efficacy is unsatisfactory in pancreatic cancer (PC), which can be limited by the overall condition of patients, the pathological type of PC, the expression level of tumor related genes, etc. To improve clinical efficiency, various researches have been conducted, and the efficacy of combination therapy showed significantly improvement compared to monotherapy. This review analyzed current strategies based on anti-CTLA-4 combination immunotherapy, providing totally new idea for future research.
Collapse
Affiliation(s)
- Ran Ni
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhiming Hu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Hepatobiliary & Pancreatic Surgery, Tongde Hospital of Zhejiang Province, Hangzhou 310012, China.
| | - Ran Tao
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
246
|
Dorman K, Boeck S, Caca K, Reichert M, Ettrich TJ, Oettle H, Waidmann O, Modest DP, Müller L, Michl P, Kanzler S, Pink D, Reinacher-Schick A, Geißler M, Pelz H, Kunzmann V, Held S, Schichtl T, Heinemann V, Kullmann F. Alternating gemcitabine plus nab-paclitaxel and gemcitabine alone versus continuous gemcitabine plus nab-paclitaxel after induction treatment of metastatic pancreatic cancer (ALPACA): a multicentre, randomised, open-label, phase 2 trial. Lancet Gastroenterol Hepatol 2024; 9:935-943. [PMID: 39159648 DOI: 10.1016/s2468-1253(24)00197-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND A standardised dose-reduction strategy has not been established for the widely used gemcitabine plus nab-paclitaxel regimen in patients with metastatic pancreatic ductal adenocarcinoma. We aimed to investigate the efficacy and tolerability of alternating treatment cycles of nab-paclitaxel-gemcitabine combination therapy and gemcitabine alone versus continuous treatment with the nab-paclitaxel-gemcitabine combination. METHODS ALPACA was a randomised, open-label, phase 2 trial conducted at 29 study centres across Germany. Patients aged 18 years or older with a histologically or cytologically confirmed diagnosis of metastatic pancreatic ductal adenocarcinoma who had not been previously treated for advanced disease were enrolled. After an induction phase with three cycles of nab-paclitaxel-gemcitabine combination therapy (nab-paclitaxel 125 mg/m2 and gemcitabine 1000 mg/m2 administered intravenously on days 1, 8, and 15 of each 28-day cycle), patients were randomly assigned (1:1) by stratified permuted block randomisation either to continue treatment with standard nab-paclitaxel-gemcitabine or to receive alternating cycles of nab-paclitaxel-gemcitabine and gemcitabine alone. Patients and investigators were not masked to treatment allocation. Randomisation was done centrally by the study statistician using a computer-generated randomisation list, and was stratified by Karnofsky Performance Status and presence of liver metastases. The primary endpoint was the derivation of an unbiased point estimate and an associated confidence interval with a confidence coefficient of 80% for the hazard ratio (HR) for overall survival after randomisation, without testing a specific hypothesis, analysed by intention to treat in all patients who started randomised treatment. Safety was analysed according to treatment received. This trial is registered with ClinicalTrials.gov, NCT02564146, and is completed. FINDINGS Between May 27, 2016, and May 27, 2021, 325 patients were enrolled. Following three cycles of induction treatment, 174 patients were randomly assigned: 85 to continue receiving standard nab-paclitaxel-gemcitabine, of whom 79 started treatment, and 89 to the alternating treatment schedule, of whom 88 started treatment. Of the 167 patients who started randomised treatment, 88 (53%) were female and 79 (47%) were male. Median overall survival after randomisation was 10·4 months (80% CI 9·2-12·0) in the group that received standard treatment and 10·5 months (10·2-11·1) in the group that received alternating treatment (HR 0·90, 80% CI 0·72-1·13; p=0·56). The most common adverse events of any grade were peripheral neuropathy (59 [74%] of 80 patients in the continuous treatment group vs 53 [62%] of 85 patients in the alternating treatment group) and fatigue (43 [54%] vs 44 [52%]). Treatment-emergent serious adverse events after randomisation occurred in 40 (50%) patients in the continuous treatment group and in 28 (33%) in the alternating treatment group. Fewer treatment-emergent adverse events of grade 3 or higher occurred in patients treated with alternating cycles compared with those receiving standard therapy, especially for peripheral neuropathy (17 [21%] patients in the continuous treatment group vs 12 [14%] in the alternating treatment group) and infections (16 [20%] vs nine [11%]). There were two treatment-related deaths after randomisation, both in the continuous treatment group (one multiple organ dysfunction syndrome, not treated after randomisation, and one interstitial lung disease). INTERPRETATION Our findings suggest that a dose-reduced regimen with alternating cycles of nab-paclitaxel-gemcitabine and gemcitabine alone after three induction cycles is associated with similar overall survival to that for standard treatment with nab-paclitaxel-gemcitabine, but with improved tolerability. We therefore propose that a switch to the alternating schedule could be considered in a clinical setting for patients with metastatic pancreatic cancer who have at least stable disease after three cycles of nab-paclitaxel-gemcitabine treatment. FUNDING Celgene/Bristol Myers Squibb.
Collapse
Affiliation(s)
- Klara Dorman
- Department of Medicine III and Comprehensive Cancer Center, University Hospital, LMU Munich, Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Stefan Boeck
- Department of Medicine III and Comprehensive Cancer Center, University Hospital, LMU Munich, Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany; Department of Hematology and Oncology, Munich Municipal Hospital Neuperlach, Munich, Germany
| | - Karel Caca
- Department of Internal Medicine I, Hospital Ludwigsburg, Ludwigsburg, Germany
| | - Maximilian Reichert
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany; Department of Internal Medicine II, University Hospital rechts der Isar, Technical University Munich, Munich, Germany
| | - Thomas J Ettrich
- Department of Internal Medicine I, University of Ulm, Ulm, Germany
| | - Helmut Oettle
- Dayclinic for Oncology and Hematology, Outpatient Department of Medical Oncology, Friedrichshafen, Germany
| | - Oliver Waidmann
- Center for Hematology and Oncology Bethanien, Frankfurt, Germany
| | - Dominik P Modest
- Department of Hematology, Oncology, and Cancer Immunology, Charité University Hospital Berlin, Berlin, Germany
| | | | - Patrick Michl
- Department of Gastroenterology, Hepatology, and Infectious Diseases, Medical University Hospital, Heidelberg, Germany
| | - Stephan Kanzler
- Department of Internal Medicine II, Leopoldina Hospital Schweinfurt, Schweinfurt, Germany
| | - Daniel Pink
- Department of Medical Oncology, Helios Hospital Bad Saarow, Bad Saarow, Germany; Clinic for Internal Medicine C, University Medicine Greifswald, Greifswald, Germany
| | - Anke Reinacher-Schick
- Department of Haematology and Oncology, St Josef Hospital, Ruhr University, Bochum, Germany
| | - Michael Geißler
- Department of Internal Medicine, Oncology/Hematology, Gastroenterology, Hospital Esslingen, Esslingen, Germany
| | - Henning Pelz
- Center for Hematology and Oncology Offenburg, Offenburg, Germany
| | - Volker Kunzmann
- Department of Medical Oncology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | | | | | - Volker Heinemann
- Department of Medicine III and Comprehensive Cancer Center, University Hospital, LMU Munich, Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Frank Kullmann
- Department of Medicine I, Hospital Weiden, Weiden, Germany.
| |
Collapse
|
247
|
Kabir E, Azam F, Khan T, Yasmin H, Chowdhury N, Ahmed S, Sagar B, Tahrim N. Modeling Overall Survival in Patients With Pancreatic Cancer From a Pooled Analysis of Phase II Trials. Cancer Med 2024; 13:e70289. [PMID: 39387320 PMCID: PMC11465028 DOI: 10.1002/cam4.70289] [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: 11/26/2023] [Revised: 06/08/2024] [Accepted: 09/21/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND We evaluated the validity of surrogacy of progression-free survival (PFS) or time-to-progression (TTP) and overall response rate (ORR) in phase II trials of pancreatic ductal adenocarcinoma (PDAC). In addition, we explored the impact of predictive variables on overall survival (OS) and developed an optimal OS model. METHODS We analyzed 1867 clinical endpoint from 619 phase II PDAC trials with a systematic search from PubMed. Endpoint correlations were determined by Spearman's rank correlation. The assessed predictive factors included PFS/TTP, treatment size, therapy type, stage, and previous treatment. The relationship between predictors and OS was explored by a gamma generalized linear model (GLM) with a log-link function and compared with linear models. RESULTS The Spearman rank correlation coefficient between PFS/TTP and OS was 0.88 (95% confidence interval [CI] 0.85-0.89; p < 0.0001; n = 610) and between ORR and OS was 0.58 (0.52-0.64; p < 0.0001; n = 514). Model comparison favored the GLM model over the linear model, offering more accurate predictions for higher OS values. Consequently, PFS/TTP was the strongest predictor (pseudo-R2 = 0.75), with 1 added median PFS/TTP month associated with 13% (95% CI 13%-14%) increase in median OS. Subgroup analysis revealed that chemotherapy conferred significantly longer OS compared to targeted therapy in 1-Agent and 2-Agent trials, exhibiting a "very large" and "medium" effect size, respectively (rank biserial, rrb = 0.40 [95% CI 0.22-0.56] and rrb = 0.29 [0.16-0.41], both p < 0.0001), although inconsistent efficacy in 3-Agent trials (rrb = 0.12 [-0.07-0.30], p = 0.21). CONCLUSIONS PFS/TTP is a more reliable surrogate than ORR and a strong predictor of OS in phase II trials of pancreatic cancer. Moreover, gamma GLM (log-link function) is a robust tool for modeling positively skewed survival data with non-constant variance, thus can be applied to other cancers' OS data of such nature.
Collapse
Affiliation(s)
| | - Faruque Azam
- School of PharmacyBRAC UniversityDhakaBangladesh
| | | | | | | | | | | | | |
Collapse
|
248
|
Yasinzai AQK, Tareen B, Tracy K, Jamil N, Khan M, Ullah H, Raza M, Khan AU, Arif D, Waheed A, Sidhwa F, Misra S, Karki NR, Karim NA, Cavalcante L, Ullah A. Pancreatic ductal adenocarcinoma: exploring clinicopathological trends and racial disparities in a comprehensive U.S. population-based study. Clin Transl Oncol 2024; 26:2618-2628. [PMID: 38615292 DOI: 10.1007/s12094-024-03484-7] [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: 01/25/2024] [Accepted: 03/25/2024] [Indexed: 04/15/2024]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy about 50% of PDAC are metastatic at presentation. In this study, we evaluated PDAC demographics, annual trend analysis, racial disparities, survival rate, and the role of different treatment modalities in localized and metastatic disease. METHODS A total of 144,824 cases of PDAC were obtained from the SEER database from 2000 to 2018. RESULTS The median age was 69 years, with a slightly higher incidence in males (52%) and 80% of all cases were white. Among cases with available data, 43% were grade III tumors and 57% were metastatic. The most common site of metastasis was the liver (15.7%). The annual incidence has increased steadily from 2000 to 2018. The overall observed (OS) 5-year survival rate was 4.4% (95% CI 4.3-4.6%), and 5 years cause-specific survival (CSS) was 5% (95% CI 5.1-5.4%). The 5-year survival with multimodal therapy (chemotherapy, surgery, and radiation) was 22% (95% CI 20.5-22.8%). 5-year CSS for the blacks was lower at 4.7% (95% CI 4.2-5.1%) compared to the whites at 5.3% (95% CI 5.1-5.4%). Multivariate analysis found male gender and black race associated with worse prognosis. Kaplan-Meier survival analysis found multimodal therapy to have the best outcomes in all three stages. CONCLUSION PDAC is an aggressive malignancy with male gender and black race are associated with a poor prognosis. Surgery with chemoradiation was associated with the best overall survival. With steadily increasing rates of PDAC, improved treatment modalities are paramount to improving survival in these patients.
Collapse
Affiliation(s)
| | - Bisma Tareen
- Department of Medicine, Bolan Medical College, Quetta, 83700, Pakistan
| | | | - Nimra Jamil
- Department of Medicine, Bolan Medical College, Quetta, 83700, Pakistan
| | - Marjan Khan
- Internal Medicine, Marshfield Medical Center, Marshfield, USA
| | - Hafeez Ullah
- Department of Medicine, Bolan Medical College, Quetta, 83700, Pakistan
| | - Muhammad Raza
- Department of Medicine, Bolan Medical College, Quetta, 83700, Pakistan
| | - Amin Ullah Khan
- Department of Medicine, Bolan Medical College, Quetta, 83700, Pakistan
| | - Dauod Arif
- Department of Oncology Developmental Therapeutics, Novant Health, Charlotte, NC, USA
| | - Abdul Waheed
- Department of Surgery, San Joaquin General Hospital, French Camp, CA, 95231, USA
| | - Feroze Sidhwa
- Department of Surgery, San Joaquin General Hospital, French Camp, CA, 95231, USA
| | - Subhasis Misra
- Department of Surgery, BayCare Health System, Tampa, FL, USA
| | - Nabin Raj Karki
- Department of Oncology, Mitchell Cancer Institute, University of South Alabama, Mobile, USA
| | - Nagla Abdel Karim
- Department of Hematology-Oncology, Inova Schar Cancer Institute, University of Virginia, Fairfax, VA, 22031, USA
| | - Ludimila Cavalcante
- Department of Oncology Developmental Therapeutics, Novant Health, Charlotte, NC, USA
| | - Asad Ullah
- Department of Pathology, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| |
Collapse
|
249
|
Seufferlein T, Mayerle J, Boeck S, Brunner T, Ettrich TJ, Grenacher L, Gress TM, Hackert T, Heinemann V, Kestler A, Sinn M, Tannapfel A, Wedding U, Uhl W. S3-Leitlinie Exokrines Pankreaskarzinom – Version 3.1. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2024; 62:1724-1785. [PMID: 39389105 DOI: 10.1055/a-2338-3716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Affiliation(s)
| | | | | | - Thomas Brunner
- Universitätsklinik für Strahlentherapie-Radioonkologie, Medizinische Universität Graz, Austria
| | | | | | - Thomas Mathias Gress
- Gastroenterologie und Endokrinologie Universitätsklinikum Gießen und Marburg, Germany
| | - Thilo Hackert
- Klinik und Poliklinik für Allgemein-, Viszeral- und Thoraxchirurgie, Universitätsklinikum Hamburg-Eppendorf, Germany
| | - Volker Heinemann
- Medizinische Klinik und Poliklinik III, Klinikum der Universität München-Campus Grosshadern, München, Germany
| | | | - Marianne Sinn
- Medizinische Klinik und Poliklinik II Onkologie und Hämatologie, Universitätsklinikum Hamburg-Eppendorf, Germany
| | | | | | - Waldemar Uhl
- Allgemein- und Viszeralchirurgie, St Josef-Hospital, Bochum, Germany
| |
Collapse
|
250
|
Seufferlein T, Mayerle J, Boeck S, Brunner T, Ettrich TJ, Grenacher L, Gress TM, Hackert T, Heinemann V, Kestler A, Sinn M, Tannapfel A, Wedding U, Uhl W. S3-Leitlinie Exokrines Pankreaskarzinom – Version 3.1. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2024; 62:e874-e995. [PMID: 39389103 DOI: 10.1055/a-2338-3533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Affiliation(s)
| | | | | | - Thomas Brunner
- Universitätsklinik für Strahlentherapie-Radioonkologie, Medizinische Universität Graz, Austria
| | | | | | - Thomas Mathias Gress
- Gastroenterologie und Endokrinologie Universitätsklinikum Gießen und Marburg, Germany
| | - Thilo Hackert
- Klinik und Poliklinik für Allgemein-, Viszeral- und Thoraxchirurgie, Universitätsklinikum Hamburg-Eppendorf, Germany
| | - Volker Heinemann
- Medizinische Klinik und Poliklinik III, Klinikum der Universität München-Campus Grosshadern, München, Germany
| | | | - Marianne Sinn
- Medizinische Klinik und Poliklinik II Onkologie und Hämatologie, Universitätsklinikum Hamburg-Eppendorf, Germany
| | | | | | - Waldemar Uhl
- Allgemein- und Viszeralchirurgie, St Josef-Hospital, Bochum, Germany
| |
Collapse
|