1
|
Yong JP, Mu XY, Zhou CF, Zhang KK, Gao JQ, Guo ZZ, Zhou SF, Ma Z. Radiofrequency ablation of liver metastases in a patient with pancreatic cancer and long-term survival: A case report. World J Clin Cases 2025; 13:100169. [DOI: 10.12998/wjcc.v13.i20.100169] [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: 08/15/2024] [Revised: 09/05/2024] [Accepted: 03/06/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND According to the GLOBCAN2022 database, pancreatic cancer has become the 6th leading cause of cancer-related death worldwide. The latest statistics suggest that the incidence of pancreatic cancer is increasing at a rate of 0.5% to 1.0% per year, and it is expected to become the 2nd leading cause of tumor-related deaths in the United States by 2030. More than 50% of pancreatic cancer patients have already developed distant metastases at the time of diagnosis, with the liver being the most common site. Patients with pancreatic cancer with liver metastasis (PCLM) have a worse prognosis than those with locally progressed pancreatic cancer, with a median survival of less than six months. Therefore, the outcome of liver metastases is often a vital determinant of the prognosis of patients with PCLM. There are few successful cases of localized treatment for PCLM patients. Our department recently performed local radiofrequency ablation (RFA) treatment for a PCLM patient through an evidence-based medicine approach, with remarkable therapeutic effects.
CASE SUMMARY The patient was admitted to the hospital on May 03, 2018, 3 weeks after pancreatic cancer surgery. In October 2017, the patient presented with lower back pain. No abnormalities were detected via computed tomography (CT), colonoscopy, or gastroscopy. However, on March 18, 2018, the patient was investigated in a foreign hospital via CT, which suggested occupational lesions in the descending part of the duodenum, and magnetic resonance imaging suggested pancreatic occupancy. He was considered to be suffering from pancreatic cancer. He underwent laparoscopic-assisted pancreatic + duodenum + superior mesenteric vein partial resection and reconstruction under general anesthesia on March 26, 2018 at The Affiliated Hospital of Xuzhou Medical University. The pancreas and duodenum were partially resected. Postoperative pathology showed adenocarcinoma of the pancreas (moderately differentiated), partly mucinous carcinoma, invading the mucosal layer of the duodenum; the tumor size was 4.5 cm × 4 cm × 4 cm. There was no apparent nerve or vascular invasion. There was no cancer or involvement of the pancreas section or expected hepatic duct margins. There was no cancer involvement in the gastric and duodenal sections. There was no cancer metastasis to the peripheral lymph nodes of the pancreas (0/9). No metastasis to the gastric lesser curvature or more significant curvature lymph nodes (0/1, 0/5) was detected, and the peri-intestinal lymph nodes showed no cancer metastasis (0/4). Although the gallbladder showed signs of chronic cholecystitis, there was no cancer involvement, and the lymph nodes in Groups 12 and 13 also showed no cancer metastasis (0/6, 0/1). His postoperative recovery was acceptable. CT was performed on May 2018 at our hospital and found the following: (1) Double lung bronchial vascular bundles slightly heavier than normal; (2) Postoperative changes in the pancreas and a retention tube shadow in front of the head of the pancreas; (3) Small cysts in the right lobe of the liver; (4) Abdominopelvic effusion; and (5) Para splenic enlargement. pTNM stage: PT3N0M0. The patient was in the second stage of postoperative pancreatic cancer, with a potential risk of recurrence considering the patient's postoperative body quality deviation. The patient was unable to tolerate the standard multidrug combination and underwent six cycles of single-agent gemcitabine chemotherapy from May 10, 2018 to August 31, 2018 (the specific drug dosage was 1.4 g/d1/d8 gemcitabine injection, which was repeated every 21 days). Efficacy was determined to be stable disease after 2, 4, and 6 cycles. The side effects during treatment were tolerable.
CONCLUSION This case suggests that RFA can serve as a viable local treatment modality for selected patients with PCLM, offering a chance for long-term survival. Such localized interventions, when carefully tailored, may complement systemic therapies in controlling metastatic pancreatic cancer.
Collapse
Affiliation(s)
- Jin-Peng Yong
- Department of Oncology, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou 450000, Henan Province, China
| | - Xiao-Yan Mu
- Department of Oncology, Longhua Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai 20001, China
| | - Chao-Feng Zhou
- Department of Oncology, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou 450000, Henan Province, China
| | - Ke-Ke Zhang
- Department of Oncology, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou 450000, Henan Province, China
| | - Jie-Qiong Gao
- Department of Oncology, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou 450000, Henan Province, China
| | - Zhi-Zhong Guo
- Department of Oncology, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou 450000, Henan Province, China
| | - Shi-Fan Zhou
- Department of Oncology, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou 450000, Henan Province, China
| | - Zhen Ma
- Department of Neurology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou 450000, Henan Province, China
- Department of Oncology, Henan Hospital of Traditional Chinese Medicine, Zhengzhou 450000, Henan Province, China
| |
Collapse
|
2
|
Li F, Shen F. Metastatic pancreatic cancer with activating BRAF V600E mutations: A case report. World J Clin Cases 2025; 13:101665. [DOI: 10.12998/wjcc.v13.i16.101665] [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: 09/25/2024] [Revised: 12/06/2024] [Accepted: 01/11/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Pancreatic cancer (PC) is a highly malignant tumor that is resistant to chemotherapy, radiotherapy and immunotherapy. Combination chemotherapy regimens are the standard first-line regimens for metastatic disease, with a median survival < 12 months. Although recurrent genomic alterations such as the BRAF V600E mutation have been reported in PC, evidence supporting the clinical effectiveness of molecularly guided targeted therapies is limited.
CASE SUMMARY We report a case of a 33-year-old male who was referred to our department with weight loss of 5 kg in 2 months, anorexia and abdominal pain. Imaging showed extensive lesions involving the pancreas, liver, bones, muscles and lymph nodes accompanied by elevated carbohydrate antigen 19-9 (CA19-9) and carcinoembryonic antigen (CEA). Biopsy yielded a diagnosis of PC. Treatment with gemcitabine and nab-paclitaxel was initiated, but the disease progressed in < 2 months even though the patient’s general condition improved. Molecular testing revealed the presence of BRAF mutation. Dabrafenib/trametinib combination therapy was introduced, and the patient was treated for 2 months with a decrease in CA19-9 and CEA levels, but he died after 2 months of treatment.
CONCLUSION BRAF alterations are infrequent in PC. This case highlights the significance of molecular profiling in patients with PC, especially in patients with a high tumor burden.
Collapse
Affiliation(s)
- Fang Li
- Department of Medical Oncology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen 361015, Fujian Province, China
- Xiamen Clinical Research Center for Cancer, Xiamen 361015, Fujian Province, China
- Clinical Research Center for Precision Medicine of Abdominal Tumor of Fujian Province, Xiamen 361015, Fujian Province, China
| | - Feng Shen
- Department of Medical Oncology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen 361015, Fujian Province, China
- Xiamen Clinical Research Center for Cancer, Xiamen 361015, Fujian Province, China
- Clinical Research Center for Precision Medicine of Abdominal Tumor of Fujian Province, Xiamen 361015, Fujian Province, China
| |
Collapse
|
3
|
Li X, Chen Y, Qiao G, Ni J, Chen T, Wang Y, Wu C, Zhang Q, Ma T, Gao S, Zhang M, Shen Y, Wu J, Yu J, Que R, Zhang X, Sun K, Xiao W, Jiang T, Bai X, Liang T. 5-Year survival rate over 20 % in pancreatic ductal adenocarcinoma: A retrospective study from a Chinese high-volume center. Cancer Lett 2025; 619:217658. [PMID: 40118244 DOI: 10.1016/j.canlet.2025.217658] [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: 02/28/2025] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 03/23/2025]
Abstract
Standardized clinical management of pancreatic adenocarcinoma (PDAC) remains challenging and high-volume centers provide essential insights for establishing effective multimodal treatment approaches. This retrospective observational study evaluated the impact of standardized, multimodal clinical management on survival outcomes in patients with PDAC across all stages, based on NCCN guidelines resectability criteria, at a high-volume center. From 2019, 4143 patients were diagnosed with PDAC, with 3268 patients receiving further treatment, including surgical resection and/or systemic therapy. The median overall survival (OS) was 18.5 (95 %CI 17.5-19.4) months for the treated cohort and the 5-year survival rate reached 23.3 %. Patients who underwent surgical resection had significantly improved median OS compared to those who received non-surgical treatments (28.4 months vs. 13.0 months, P < 0.001), with corresponding 5-year survival rates of 31.6 % vs. 15.0 %. Moreover, the patients who received NAT followed by surgical resection had improved survival outcomes compared to those who underwent upfront surgical resection in both resectable (median OS: 37.5 months vs. 28.9 months, P < 0.01) and borderline resectable group (median OS: 31.8 months vs. 18.4 months, P < 0.001). This study demonstrated a 5-year survival rate exceeding 20 % for PDAC across all stages at our center. The application of evidence-based treatment strategies through the multidisciplinary team, accompanying with standardized and comprehensive therapeutic managements, high patient adherence, have been considered as critical determinants in enhancing therapeutic efficacy and improving long-term prognosis for patients with PDAC.
Collapse
Affiliation(s)
- Xiang Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Yiwen Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Guoliang Qiao
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Ni
- Information Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Chen
- Department of Radiology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yangyang Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Chengyi Wu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Tao Ma
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Shunliang Gao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Min Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Yan Shen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Jian Wu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Jun Yu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Risheng Que
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Xiaochen Zhang
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ke Sun
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenbo Xiao
- Department of Radiology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tian'an Jiang
- Department of Ultrasound, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
4
|
Reiss KA, Soares KC, Torphy RJ, Gyawali B. Treatment Innovations in Pancreatic Cancer: Putting Patient Priorities First. Am Soc Clin Oncol Educ Book 2025; 45:e473204. [PMID: 40173379 DOI: 10.1200/edbk-25-473204] [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: 04/04/2025]
Abstract
Pancreatic adenocarcinoma remains one of the most aggressive and difficult-to-treat solid tumor malignancies, with a high mortality-to-incidence ratio. Globally, pancreatic cancer ranks 12th in terms of incidence but sixth for mortality signifying its aggressive behavior and limited treatment options. While the mortality rates for many other solid tumors have substantially improved over the past few decades, temporal trends in pancreatic cancer mortality rates are quite sobering. In the United States, from 2000 to 2020, the mortality rates from pancreatic cancer have increased, whereas at the same time, mortality rates from other cancers, such as lung, colorectal, or kidney, have fallen appreciably. Is this for lack of treatment innovation? How do we improve survival for patients with pancreatic cancer? In this chapter, we discuss the recent advances and future directions with targeted therapies and immunotherapies in the treatment of pancreatic cancer, and provide the reasons for both optimism and caution for the future of systemic treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Kim A Reiss
- Division of Hematology/Oncology, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Kevin C Soares
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Robert J Torphy
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Bishal Gyawali
- Department of Oncology, Queen's University, Kingston, Canada
- Division of Cancer Care and Epidemiology, Queen's University, Kingston, Canada
- Department of Public Health Sciences, Queen's University, Kingston, Canada
| |
Collapse
|
5
|
Enzler T, Frankel TL. Pancreatic cancer precursor lesions - Can immunotherapy prevent progression into pancreatic ductal adenocarcinoma? Cancer Lett 2025; 619:217662. [PMID: 40127814 DOI: 10.1016/j.canlet.2025.217662] [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: 11/12/2024] [Revised: 02/26/2025] [Accepted: 03/19/2025] [Indexed: 03/26/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers, with a 5-year survival rate of only 12.5 %. Early detection of PDAC or addressing risk factors for PDAC development are ways to improve outcomes. PDAC can arise from precursor lesions, including pancreatic intraepithelial neoplasia (PanIN), intraductal papillary mucinous neoplasm (IPMN), and less frequent, mucinous cystic neoplasm (MCN), and other rare precursor variants. High-risk precursor lesions harbor a substantial chance of evolving into PDAC. Such lesions can often be found in resected PDAC specimens adjacent to the cancer. Unfortunately, recognizing precursor lesions that need to be resected is often tricky, and resections frequently end in major surgical interventions. Thus, better ways to handle precursor lesions are desperately needed. We mapped the immune microenvironments (IMEs) of PanINs, IPMNs, and MCNs on a cellular level using multiplex immunofluorescence and computational imaging technology and compared the findings to PDACs and normal pancreatic tissues. We found distinct and potentially targetable mechanisms of immunosuppression between the two main precursor lesions, PanIN and IMPN. Immunosuppression in IPMNs seems partly mediated by programmed cell death protein 1 ligand (PD-L1) expression on antigen-presenting cells (APCs). By contrast, elevated numbers of regulatory T cells (Tregs) seem to be key players in the immunosuppression of PanINs. Thus, treating high-risk IPMNs with anti-PD-1 and high-risk PanINs with agents targeting Tregs, such as anti-lymphocyte associated protein 4 (anti-CTLA-4) antibodies, could reverse their immunosuppressive state. Reversal of immunosuppression will restore immunosurveillance and eventually prevent progression into PDAC. We also review relevant published and ongoing non-surgical treatment approaches for high-risk IPMNs and PanINs.
Collapse
Affiliation(s)
- Thomas Enzler
- Department of Medicine, University of Michigan, Ann Arbor, MI, 40109, USA.
| | - Timothy L Frankel
- Department of Surgery, University of Michigan, Ann Arbor, MI, 40109, USA
| |
Collapse
|
6
|
Faisal MS, Hussain I, Ikram MA, Shah SB, Rehman A, Iqbal W. Irinotecan dosing and pharmacogenomics: a comprehensive exploration based on UGT1A1 variants and emerging insights. J Chemother 2025; 37:199-212. [PMID: 38706404 DOI: 10.1080/1120009x.2024.2349444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 04/05/2024] [Accepted: 04/24/2024] [Indexed: 05/07/2024]
Abstract
Irinotecan is a critical anticancer drug used to treat metastatic colorectal cancer and advanced pancreatic ductal adenocarcinoma by obstructing topoisomerase 1; however, it can cause minor-to-severe and life-threatening adverse effects. UDP glucuronosyltransferase family 1 member A1 (UGT1A1) polymorphisms increase the risk of irinotecan-induced neutropenia and diarrhea. Hence, screening for UGT1A1 polymorphisms before irinotecan-based chemotherapy is recommended to minimize toxicity, whereas liposomes offer the potential to deliver irinotecan with fewer side effects in patients with pancreatic ductal adenocarcinoma. This review presents a comprehensive overview of the effects of genotype-guided dosing of irinotecan on UGT1A1*28 and UGT1A1*6 variants, incorporating pharmacogenomic research, optimal regimens for metastatic colorectal and pancreatic cancer treatment using irinotecan, guidelines for toxicity reduction, and an evaluation of the cost-effectiveness of UGT1A1 genotype testing.
Collapse
Affiliation(s)
- Muhammad Saleem Faisal
- Department of Biological Sciences, International Islamic University, Islamabad, Pakistan
| | - Imran Hussain
- Department of Biological Sciences, International Islamic University, Islamabad, Pakistan
| | | | - Syed Babar Shah
- Department of Biological Sciences, International Islamic University, Islamabad, Pakistan
| | - Abdul Rehman
- Department of Biological Sciences, International Islamic University, Islamabad, Pakistan
| | - Wajid Iqbal
- Department of Biological Sciences, International Islamic University, Islamabad, Pakistan
| |
Collapse
|
7
|
Doi T, Ishikawa T, Moriguchi M, Itoh Y. Current status of cancer genome medicine for pancreatic ductal adenocarcinoma. Jpn J Clin Oncol 2025; 55:443-452. [PMID: 39893577 DOI: 10.1093/jjco/hyaf012] [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: 09/21/2024] [Accepted: 01/17/2025] [Indexed: 02/04/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis; however, advancements in cancer genome profiling using next-generation sequencing have provided new perspectives. KRAS mutations are the most frequently observed genomic alterations in patients with PDAC. However, until recently, it was not considered a viable therapeutic target. Although KRAS G12C mutations for which targeted therapies are already available are infrequent in PDAC, treatments targeting KRAS G12D and pan-KRAS are still under development. Similarly, new treatment methods for KRAS, such as chimeric antigen receptor T-cell therapy, have been developed. Several other potential therapeutic targets have been identified for KRAS wild-type PDAC. For instance, immune checkpoint inhibitors have demonstrated efficacy in PDAC treatment with microsatellite instability-high/deficient mismatch repair and tumor mutation burden-high profiles. However, for other PDAC cases with low immunogenicity, combination therapies that enhance the effectiveness of immune checkpoint inhibitors are being considered. Additionally, homologous recombination repair deficiencies, including BRCA1/2 mutations, are prevalent in PDAC and serve as important biomarkers for therapies involving poly (adenosine diphosphate-ribose) polymerase inhibitors and platinum-based therapies. Currently, olaparib is available for maintenance therapy of BRCA1/2 mutation-positive PDAC. Further therapeutic developments are ongoing for genetic abnormalities involving BRAF V600E and the fusion genes RET, NTRK, NRG, ALK, FGFR2, and ROS1. Overcoming advanced PDAC remains a formidable challenge; however, this review outlines the latest therapeutic strategies that are expected to lead to significant advancements.
Collapse
Affiliation(s)
- Toshifumi Doi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Cancer Genome Medical Center, University Hospital, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Takeshi Ishikawa
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Cancer Genome Medical Center, University Hospital, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Department of Medical Oncology Unit, University Hospital, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Michihisa Moriguchi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Yoshito Itoh
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
8
|
Ahmed KS, Marcinak CT, Issaka SM, Ali MM, Zafar SN. Machine Learning to Predict Early Death Despite Pancreaticoduodenectomy. J Surg Res 2025; 310:186-193. [PMID: 40288090 DOI: 10.1016/j.jss.2025.03.047] [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: 11/11/2024] [Revised: 03/17/2025] [Accepted: 03/22/2025] [Indexed: 04/29/2025]
Abstract
INTRODUCTION About 25% of patients undergoing pancreaticoduodenectomy (PD) for right-sided pancreatic ductal adenocarcinoma (PDAC) die within 1 y of diagnosis. These patients carry all the risks of significant morbidity with no survival advantage when compared to nonsurgical options. We aimed to determine if machine learning models have superior accuracy to traditional regression models at predicting futile surgery in patients with PDAC. METHODS We analyzed data from patients in the National Cancer Database undergoing PD for PDAC between 2004 and 2020. PD was defined as futile if the patient died within 12 mo of cancer diagnosis. We trained predictive models using 80% of the dataset and 16 preoperative input variables. Models included logistic regression, multilayer perceptron, decision tree, random forest, and gradient boosting classifiers. Models were tested on a 20% test set using area under the receiver operating characteristic curve and Brier scores. RESULTS Of the 66,331 patients identified, 34,260 (51.7%) were men, with a median age of 67 y (interquartile range, 59 to 74 y). A total of 16,772 (25.3%) patients met the criteria for futile surgery. The gradient boosting model outperformed other models with an area under the receiver operating characteristic curve of 0.689, followed by logistic regression (0.679), random forest (0.675), and decision tree (0.664). Key predictors of futile PD included advanced age (> 79 y), tumor size ≥ 4 cm, and poor differentiation. Neoadjuvant therapy was associated with lower futility risk. CONCLUSIONS We demonstrated the ability of machine learning models to predict the odds of futile PD with moderate accuracy. Although similar analyses are needed on more granular datasets, our study has important implications for shared decision-making and optimized care for patients with PDAC.
Collapse
Affiliation(s)
- Kaleem S Ahmed
- Division of Surgical Oncology, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Clayton T Marcinak
- Division of Surgical Oncology, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Sheriff M Issaka
- Division of Surgical Oncology, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Muhammad Maisam Ali
- Division of Surgical Oncology, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Syed Nabeel Zafar
- Division of Surgical Oncology, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.
| |
Collapse
|
9
|
Kanesada G, Tsunedomi R, Nakagami Y, Matsui H, Shindo Y, Tomochika S, Akita H, Ioka T, Takahashi H, Nagano H. The C11orf24 Gene as a Useful Biomarker for Predicting Severe Neutropenia in Modified FOLFIRINOX for Pancreatic Cancer. Cancer Sci 2025. [PMID: 40285634 DOI: 10.1111/cas.70087] [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: 01/27/2025] [Revised: 04/08/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
Pancreatic cancer (PC) is an aggressive and lethal tumor with a poor prognosis. FOLFIRINOX improves the prognosis of patients with PC; however, despite UGT1A1 screening, adverse events, such as severe neutropenia, occur frequently. This study aimed to identify the novel biomarkers of severe neutropenia in patients treated with modified FOLFIRINOX (mFFX) for PC. In this study, patients with PC treated with mFFX (n = 71) and gemcitabine plus nab-paclitaxel (GnP) (n = 92) and patients with colorectal cancer treated with FOLFOXIRI (n = 50) were included. Genome-wide screening using whole-exome sequencing was performed during the screening phase. Validation analysis was performed using polymerase chain reaction genotyping, the Cochran-Armitage trend test, and multivariate analysis. The diagnostic performance of combined risk factors for severe neutropenia was examined using logistic regression with leave-one-out cross-validation. Three gene polymorphisms were selected from the screening phase and subjected to the validation phase. In the validation phase, a single nucleotide polymorphism in C11orf24 (c.448C>T, rs901827) was significantly correlated with ≥ Grade 3 neutropenia in mFFX and FOLFOXIRI but not in GnP. Multivariate analysis showed C11orf24 and baseline neutrophil count as independent risk factors for ≥ Grade 3 neutropenia. The diagnostic performance of the neutropenia prediction model showed areas under the curve of 0.754 (sensitivity = 0.605, specificity = 0.848) and 0.856 (sensitivity = 0.800, specificity = 0.893) for ≥ Grade 3 and 4 neutropenia, respectively. The C11orf24 gene and baseline neutrophil count may be useful biomarkers for predicting severe neutropenia following irinotecan-containing triplet chemotherapy.
Collapse
Affiliation(s)
- Gen Kanesada
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Ryouichi Tsunedomi
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
- Research Institute for Cell Design Medical Science, Yamaguchi University, Ube, Yamaguchi, Japan
| | - Yuki Nakagami
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
- Health Data Science Laboratory, Faculty of Data Science, Shimonoseki City University, Shimonoseki, Yamaguchi, Japan
| | - Hiroto Matsui
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Yoshitaro Shindo
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Shinobu Tomochika
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Hirofumi Akita
- Department of Digestive Surgery, Osaka International Cancer Institute, Osaka, Osaka, Japan
| | - Tatsuya Ioka
- Oncology Center, Yamaguchi University Hospital, Ube, Yamaguchi, Japan
| | - Hidenori Takahashi
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
- Research Institute for Cell Design Medical Science, Yamaguchi University, Ube, Yamaguchi, Japan
| |
Collapse
|
10
|
Calheiros J, Silva R, Barbosa F, Morais J, Moura SR, Almeida S, Fiorini E, Mulhovo S, Aguiar TQ, Wang T, Ricardo S, Almeida MI, Domingues L, Melo SA, Corbo V, Ferreira MJU, Saraiva L. A first-in-class inhibitor of homologous recombination DNA repair counteracts tumour growth, metastasis and therapeutic resistance in pancreatic cancer. J Exp Clin Cancer Res 2025; 44:129. [PMID: 40275348 PMCID: PMC12020112 DOI: 10.1186/s13046-025-03389-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is among the cancer types with poorest prognosis and survival rates primarily due to resistance to standard-of-care therapies, including gemcitabine (GEM) and olaparib. Particularly, wild-type (wt)BRCA tumours, the most prevalent in PDAC, are more resistant to DNA-targeting agents like olaparib, restraining their clinical application. Recently, we disclosed a monoterpene indole alkaloid derivative (BBIT20) as a new inhibitor of homologous recombination (HR) DNA repair with anticancer activity in breast and ovarian cancer. Since inhibition of DNA repair enhances the sensitivity of cancer cells to chemotherapy, we aimed to investigate the anticancer potential of BBIT20 against PDAC, particularly carrying wtBRCA. METHODS In vitro and in vivo PDAC models, particularly human cell lines (including GEM-resistant PDAC cells), patient-derived organoids and xenograft mice of PDAC were used to evaluate the anticancer potential of BBIT20, alone and in combination with GEM or olaparib. Disruption of the BRCA1-BARD1 interaction by BBIT20 was assessed by co-immunoprecipitation, immunofluorescence and yeast two-hybrid assay. RESULTS The potent antiproliferative activity of BBIT20, superior to olaparib, was demonstrated in PDAC cells regardless of BRCA status, by inducing cell cycle arrest, apoptosis, and DNA damage, while downregulating HR. The disruption of DNA double-strand breaks repair by BBIT20 was further reinforced by non-homologous end joining (NHEJ) suppression. The inhibition of BRCA1-BARD1 heterodimer by BBIT20 was demonstrated in PDAC cells and confirmed in a yeast two-hybrid assay. In GEM-resistant PDAC cells, BBIT20 showed potent antiproliferative, anti-migratory and anti-invasive activity, overcoming GEM resistance by inhibiting the multidrug resistance P-glycoprotein, upregulating the intracellular GEM-transporter ENT1, and downregulating GEM resistance-related microRNA-20a and GEM metabolism enzymes as RRM1/2. Furthermore, BBIT20 did not induce resistance in PDAC cells. It inhibited the growth of patient-derived PDAC organoids, by inducing apoptosis, repressing HR, and potentiating olaparib and GEM cytotoxicity. The enhancement of olaparib antitumor activity by BBIT20 was confirmed in xenograft mice of PDAC. Notably, it hindered tumour growth and liver metastasis formation, improving survival of orthotopic xenograft mice of PDAC. Furthermore, its potential as a stroma-targeting agent, reducing fibrotic extracellular matrix and overcoming desmoplasia, associated with an enhancement of immune cell response by depleting PD-L1 expression in tumour tissues, renders BBIT20 even more appealing for combination therapy, particularly with immunotherapy. CONCLUSION These findings underscore the great potential of BBIT20 as a novel multifaceted anticancer drug candidate for PDAC treatment.
Collapse
Grants
- 2020.04613.BD FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior
- 2022.05718.PTDC, 0.54499/LA/P/0008/2020, 10.54499/UIDP/50006/2020, 10.54499/UIDB/50006/2020 FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior
- 2020.06020.BD FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior
- 2022.05718.PTDC, 0.54499/LA/P/0008/2020, 10.54499/UIDP/50006/2020, 10.54499/UIDB/50006/2020 FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior
- 2022.05718.PTDC, 0.54499/LA/P/0008/2020, 10.54499/UIDP/50006/2020, 10.54499/UIDB/50006/2020 FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior
- 2022.05718.PTDC, 0.54499/LA/P/0008/2020, 10.54499/UIDP/50006/2020, 10.54499/UIDB/50006/2020 FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior
- AIRC; IG No 288801 Associazione Italiana Ricerca sul Cancro
- AIRC; IG No 288801 Associazione Italiana Ricerca sul Cancro
- NHI; HHSN26100008 NCI NIH HHS
- National Cancer Institute
Collapse
Affiliation(s)
- Juliana Calheiros
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Rita Silva
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Filipa Barbosa
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa, 1649-003, Portugal
| | - João Morais
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Sara Reis Moura
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
| | - Sofia Almeida
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
| | - Elena Fiorini
- Department of Engineering for Innovation Medicine (DIMI), University of Verona, 37134, Verona, Italy
| | - Silva Mulhovo
- Centro de Estudos Moçambicanos e de Etnociências (CEMEC), Faculty of Natural Sciences and Mathematics, Pedagogical University, Maputo, 21402161, Mozambique
| | - Tatiana Q Aguiar
- CEB - Centre of Biological Engineering, University of Minho, Braga, 4710-057, Portugal
- LABBELS - Associate Laboratory, Braga/Guimarães, Portugal
| | - Tao Wang
- Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar, Heilongjiang, 161006, China
| | - Sara Ricardo
- Associate Laboratory i4HB - Institute for Health and Bioeconomy and UCIBIO - Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Gandra, 4585-116, Portugal
| | - Maria Inês Almeida
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
| | - Lucília Domingues
- CEB - Centre of Biological Engineering, University of Minho, Braga, 4710-057, Portugal
- LABBELS - Associate Laboratory, Braga/Guimarães, Portugal
| | - Sonia A Melo
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
- Department of Pathology, Faculty of Medicine University of Porto, Al. Prof. Hernâni Monteiro, Porto, 4200-319, Portugal
- Porto Comprehensive Cancer Centre (P.CCC) Raquel Seruca, Porto, Portugal
| | - Vincenzo Corbo
- Department of Engineering for Innovation Medicine (DIMI), University of Verona, 37134, Verona, Italy
| | - Maria-José U Ferreira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa, 1649-003, Portugal.
| | - Lucília Saraiva
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal.
| |
Collapse
|
11
|
Okada Y, Sato Y, Shinomiya R, Miyake T, Takahashi T, Yokoyama R, Mitsui Y, Tomonari T, Okamoto K, Sogabe M, Miyamoto H, Kawano Y, Takayama T. Conditions for effective use of liposomal irinotecan with fluorouracil and leucovorin in unresectable pancreatic cancer after FOLFIRINOX treatment. Int J Clin Oncol 2025:10.1007/s10147-024-02677-y. [PMID: 40266453 DOI: 10.1007/s10147-024-02677-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 12/01/2024] [Indexed: 04/24/2025]
Abstract
BACKGROUND Liposomal irinotecan + fluorouracil/leucovorin (nal-IRI + 5FU/LV) is commonly used as a second- or later-line treatment for pancreatic ductal adenocarcinoma (PDAC) and offers survival benefits. However, its efficacy and safety in patients previously treated with FOLFIRINOX, which includes irinotecan, remain unclear. We evaluated the efficacy and safety of nal-IRI + 5FU/LV in patients with unresectable PDAC who received previous FOLFIRINOX therapy and those who did not. METHODS This retrospective observational study included 42 patients with PDAC who were treated with nal-IRI + 5FU/LV (October 2020-November 2023). Patients were grouped based on prior FOLFIRINOX treatment. RESULTS The progression-free survival (PFS) in patients who previously received modified FOLFIRINOX (mFFX) therapy was shorter than that in patients who did not (2.5 vs. 3.5 months, P = 0.07). When patients with greater than- and less than the cut-off value of irinotecan-free interval (IFI) were classified into the long and short IFI groups, respectively, PFS was significantly longer in the long-IFI group than that in the short IFI group (4.0 vs. 2.1 months, P = 0.01). Moreover, the C-reactive protein/albumin ratio (CAR) was also a significant predictor of PFS (P = 0.03). Furthermore, both factors were found to be independent factors influencing PFS in the univariate Cox regression analysis (P = 0.02 and P = 0.04). CONCLUSION Nal-IRI + 5FU/LV therapy may be a safe and effective option as a second- or later-line treatment, particularly for patients who have not previously received mFFX therapy. For patients who received prior mFFX exposure, a longer IFI and lower CAR may indicate greater potential benefit, thus aiding in more personalized treatment approaches.
Collapse
Affiliation(s)
- Yasuyuki Okada
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Yasushi Sato
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan.
- Department of Community Medicine for Gastroenterology and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| | - Ryo Shinomiya
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Takanori Miyake
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Taku Takahashi
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Reiko Yokoyama
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Yasuhiro Mitsui
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Tetsu Tomonari
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Koichi Okamoto
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Masahiro Sogabe
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Hiroshi Miyamoto
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Yutaka Kawano
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Tetsuji Takayama
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
12
|
Yu R, Ji X, Zhang P, Zhang H, Qu H, Dong W. The potential of chimeric antigen receptor -T cell therapy for endocrine cancer. World J Surg Oncol 2025; 23:153. [PMID: 40264184 PMCID: PMC12012980 DOI: 10.1186/s12957-025-03745-x] [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: 12/20/2024] [Accepted: 03/07/2025] [Indexed: 04/24/2025] Open
Abstract
Endocrine cancer, a relatively rare and heterogeneous tumor with diverse clinical features. The facile synthesis of hormones further complicates endocrine cancer treatment. Thus, the development of safe and effective systemic treatment approaches, such as chimeric antigen receptor (CAR) T cell therapy, is imperative to enhance the prognosis of patients with endocrine cancer. Although this therapy has achieved good results in the treatment of hematological malignancies, it encounters diverse complications and challenges in the context of endocrine cancer. This review delineates the generation of CAR-T cells, examines the potential of CAR-T cell therapy for endocrine cancer, enumerates pivotal antigens linked to endocrine cancer, encapsulates the challenges confronted with CAR-T cell therapy for endocrine cancer, and expounds upon strategies to overcome these limitations. The primary objective is to provide insightful perspectives that can contribute to the advancement of CAR-T cell therapy in the field of endocrine cancer.
Collapse
Affiliation(s)
- Ruonan Yu
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Xiaoyu Ji
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Ping Zhang
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Hao Zhang
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Huiling Qu
- Department of Neurology, The General Hospital of Northern Theater Command, 83 Wen Hua Road, Shenyang, Liaoning, 110840, China.
| | - Wenwu Dong
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China.
| |
Collapse
|
13
|
Liu W, Fang Y, Zhang C, Xiang M, Qi L, Su A, Sun Y. Cost-effectiveness of NALIRIFOX versus Nab-paclitaxel and gemcitabine in previously untreated metastatic pancreatic ductal adenocarcinoma. BMC Gastroenterol 2025; 25:266. [PMID: 40247200 PMCID: PMC12004809 DOI: 10.1186/s12876-025-03867-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND & AIMS The NAPOLI 3 trial demonstrated that compared to the nab-paclitaxel and gemcitabine (GemNab) regimen, the NALIRIFOX (5-fluorouracil, leucovorin, liposomal irinotecan, oxaliplatin) regimen can significantly improve patients' overall survival (OS) and progression-free survival (PFS), and the safety of this regimen is generally controllable. To ensure the appropriateness of the chosen treatment, economic evaluation is essential. Thus, from the perspective of American healthcare systems, we explored the cost-effectiveness comparison between NALIRIFOX or GemNab in treating patients with metastatic pancreatic ductal adenocarcinoma (mPDAC) who had not previously received treatment. METHODS A three-state partitioned survival model was developed, incorporating a lifetime horizon with a 4-week cycle length. The efficacy data were sourced from the NAPOLI 3 study, while treatment costs, health state utilities, and adverse events (AEs) were obtained from public databases and literature. Applying an annual discount rate of 3%, the analysis focused on total cost, quality-adjusted life-years (QALYs), and the incremental cost-effectiveness ratio (ICER) as primary outcomes. Conducted within a US healthcare system perspective over a 13-year lifetime horizon, the analysis set willingness-to-pay (WTP) thresholds of $50,000, $100,000 and $150,000 per QALY. Sensitivity analyses were performed to assess the robustness of the model's findings. RESULTS In our base-case analysis, NALIRIFOX group was estimated to achieve an incremental gain of 0.13 QALYs. Compared to the GemNab group, treatment with NALIRIFOX resulted in higher costs ($150,437 vs $130,683). The ICER for NALIRIFOX therapy was calculated to be $155,602.83/QALYs. Sensitivity analysis revealed the model's robustness across a range of parameters. Through probabilistic sensitivity analysis applying Monte Carlo simulation with 1000 iterations, NALIRIFOX therapy was identified as the cost-effective regimen in 45.7% of all iterations at a WTP threshold of $150,000/QALYs. CONCLUSIONS Treatment with NALIRIFOX strategy was estimated to provide a significant clinical advantage over GemNab. According to the model outcomes, NALIRIFOX therapy is borderline cost-effective for treatment-naive patients with mPDAC, slightly exceeding the highest conventional US willingness-to-pay threshold of 150,000/QALY. However, at lower thresholds, such as 100,000/QALY, NALIRIFOX fails to demonstrate cost-effectiveness in all simulations. In addition, NALIRIFOX therapy emerges as a cost-effective treatment strategy at higher willingness-to-pay thresholds or when the drug price is significantly reduced.
Collapse
Affiliation(s)
- Wenjie Liu
- National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District Panjiayuan Nanli No. 17, Beijing, 100021, China
| | - Yuting Fang
- National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District Panjiayuan Nanli No. 17, Beijing, 100021, China
| | - Chao Zhang
- Department of Medical Oncology, Beijing Chaoyang District Sanhuan Cancer Hospital, Beijing, 100122, China
| | - Midan Xiang
- National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District Panjiayuan Nanli No. 17, Beijing, 100021, China
| | - Lijuan Qi
- National Cancer Center, National Clinical Research Center for Cancer/Hebei Cancer Hospital, Chinese Academy of Medical Sciences, Langfang, 065001, China
| | - Aijiang Su
- Department of Medical Oncology, Beijing Chaoyang District Sanhuan Cancer Hospital, Beijing, 100122, China
| | - Yongkun Sun
- National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District Panjiayuan Nanli No. 17, Beijing, 100021, China.
| |
Collapse
|
14
|
Lee SS, Kareff SA, Fatoki RA. Locally advanced pancreatic cancer treated with surgical resection after chemotherapy. BMJ Case Rep 2025; 18:e264212. [PMID: 40250859 DOI: 10.1136/bcr-2024-264212] [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: 04/20/2025] Open
Abstract
Pancreatic cancer is a malignancy with poor prognosis and overall poor response to systemic therapy. Moreover, patients with locally advanced or unresectable tumours have historically worse outcomes compared with those with earlier stages of disease. We report a case of a female patient in the fourth decade with locally advanced pancreatic adenocarcinoma, who received 12 cycles modified FOLFIRINOX (mFOLFIRINOX: fluorouracil/leucovorin/irinotecan/oxaliplatin) chemotherapy, followed by pancreaticoduodenectomy, resulting in a near complete pathological response after definitive surgical treatment. Given the lack of indication for adjuvant therapy, she is considered in remission. The case demonstrates that potentially curative surgical resection can be a viable and sought-after treatment option for patients with locally advanced and/or metastatic pancreatic cancer, as it is not readily available for many patients due to the disease's aggressive nature and presence of advanced stage at diagnosis. It holds meaningful implications for clinical practice and promotes considerations in future advancements in treatment strategies.
Collapse
Affiliation(s)
- Soobin Serena Lee
- University of Miami, University of Miami, Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Samuel A Kareff
- Baptist Health South Florida, Lynn Cancer Institute, Boca Raton, Florida, USA
| | - Raleigh A Fatoki
- Sylvester Cancer Center, University of Miami Health System, Miami, Florida, USA
| |
Collapse
|
15
|
Imaoka H, Ikeda M, Ozaka M, Oshima K, Okano N, Shimizu S, Tsumura H, Komatsu Y, Yamashita T, Kataoka S, Nagano H, Hisano T, Sasaki M, Kobayashi S, Fukushima T, Mitsunaga S, Furukawa T, Hamauchi S, Ueno M, Furuse J. Phase 1/2 study of liposomal irinotecan plus S-1 for metastatic pancreatic cancer refractory to gemcitabine-based treatment. Eur J Cancer 2025; 222:115424. [PMID: 40252631 DOI: 10.1016/j.ejca.2025.115424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/03/2025] [Accepted: 04/11/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Liposomal irinotecan (nal-IRI) plus fluorouracil/folinic acid (5-FU/LV) improves survival in gemcitabine-refractory metastatic pancreatic cancer (PC) but requires a central venous port. S-1, an oral fluoropyrimidine with proven efficacy in PC, may replace 5-FU/LV in nal-IRI plus 5-FU/LV, potentially enhancing both convenience and antitumor effect. METHODS This single-arm, open-label, phase 1/2 study included patients with histologically or cytologically confirmed adenocarcinoma, aged 20-80 years, an Eastern Cooperative Oncology Group performance status of 0-1, with metastatic disease, and refractory to gemcitabine-based treatment. The primary endpoint in phase 1 part was the frequency of dose-limiting toxicity (DLT) to nal-IRI plus S-1. The primary endpoint in phase 2 part was overall survival. This trial was registered in the Japan Registry of Clinical Trials database (jRCTs031210040). RESULTS In phase 1 part, one patient with DLT was observed at nal-IRI 70 mg/m2 (day 1) with S-1 80 mg/m2/day (day 1-7) in a 2-week cycle, establishing this as the recommended phase 2 dose (RP2D). Forty-nine patients from phase 1 (n = 6) and phase 2 part (n = 43) were treated with the RP2D, and their results were pooled. Median overall survival was 10.3 months (95 % confidence interval, 8.1-12.0 months). A confirmed partial response was achieved in 10 patients (20.4 %). The most frequent treatment-emergent adverse events were hypoalbuminemia (98.0 %), anemia (98.0 %), and anorexia (81.6 %). There were no treatment-related deaths. CONCLUSIONS This study demonstrated that nal-IRI plus S-1 exhibited promising efficacy and an acceptable safety profile in patients with metastatic PC refractory to gemcitabine-based treatment.
Collapse
Affiliation(s)
- Hiroshi Imaoka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
| | - Masafumi Ikeda
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masato Ozaka
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kotoe Oshima
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Naohiro Okano
- Department of Medical Oncology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Satoshi Shimizu
- Department of Gastroenterology, Saitama Cancer Center, Saitama, Japan
| | - Hidetaka Tsumura
- Department of Gastroenterological Oncology, Hyogo Cancer Center, Akashi, Japan
| | - Yoshito Komatsu
- Division of Cancer Center, Hokkaido University Hospital, Sapporo, Japan
| | - Taro Yamashita
- Department of Gastroenterology, Kanazawa University Hospital, Kanazawa, Japan
| | - Shigeki Kataoka
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Terumasa Hisano
- Department of Hepato-Biliary-Pancreatology, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Mitsuhito Sasaki
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Satoshi Kobayashi
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Taito Fukushima
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Shuichi Mitsunaga
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takaaki Furukawa
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Satoshi Hamauchi
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Junji Furuse
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| |
Collapse
|
16
|
Ohira R, Suzuki R, Asama H, Sugimoto M, Sato K, Shimizu H, Takagi T, Hikichi T, Nakamura J, Kato T, Yanagita T, Ohira H. A Systemic Review and Meta-analysis of the Incidence Rate of Interstitial Lung Disease in Patients with Unresectable Pancreatic Cancer Treated with Gemcitabine and Nab-paclitaxel Combination Therapy in the Japanese Population. Intern Med 2025:5084-24. [PMID: 40222935 DOI: 10.2169/internalmedicine.5084-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/15/2025] Open
Abstract
Background Drug-induced interstitial lung disease (DI-ILD) is an adverse effect of the combination of gemcitabine and nab-paclitaxel (GnP) treatment that has raised concerns because the incidence rate of DI-ILD in the real world is higher than that reported in initial clinical trials. The present study assessed the cumulative incidence rate of DI-ILD among patients treated with GnP based on previously published studies and explored any potential bias in the results. Methods We conducted a meta-analysis in accordance with the PRISMA guidelines. The MEDLINE, Scopus, and Cochrane Library databases were searched from 2013 to 2024, using specific keywords. Randomized controlled trials, prospective studies, retrospective studies, and case-control studies that reported the incidence of interstitial lung disease (ILD) among patients treated with GnP were included. Two reviewers independently extracted the data from the included studies. Results Nine studies involving 1,980 patients were included in this analysis. Nine studies were conducted in Japan. The pooled incidence of DI-ILD was 5.9%. A subgroup analysis revealed that smaller studies reported higher incidence rates (≤200 vs. ≥201 cases: 9.4% vs. 4.1%). There was significant heterogeneity and publication bias, suggesting that the variability in incidence rates was due to the study size and potential biases. Conclusion The pooled incidence of ILD among patients treated with GnP was 5.9%. However, the results should be interpreted with caution because of the heterogeneity and publication bias. Further global studies are required to determine the true incidence of ILD.
Collapse
Affiliation(s)
- Rei Ohira
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Japan
| | - Rei Suzuki
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Japan
| | - Hiroyuki Asama
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Japan
| | - Mitsuru Sugimoto
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Japan
| | - Kentaro Sato
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Japan
| | - Hiroshi Shimizu
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Japan
| | - Tadayuki Takagi
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Japan
| | - Takuto Hikichi
- Department of Endoscopy, Fukushima Medical University Hospital, Japan
| | - Jun Nakamura
- Department of Endoscopy, Fukushima Medical University Hospital, Japan
| | - Tsunetaka Kato
- Department of Endoscopy, Fukushima Medical University Hospital, Japan
| | - Takumi Yanagita
- Department of Endoscopy, Fukushima Medical University Hospital, Japan
| | - Hiromasa Ohira
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Japan
| |
Collapse
|
17
|
Kim DH, Kim DJ, Park SJ, Jang WJ, Jeong CH. Inhibition of GLS1 and ASCT2 Synergistically Enhances the Anticancer Effects in Pancreatic Cancer Cells. J Microbiol Biotechnol 2025; 35:e2412032. [PMID: 40223274 PMCID: PMC12010092 DOI: 10.4014/jmb.2412.12032] [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: 12/18/2024] [Revised: 02/14/2025] [Accepted: 02/15/2025] [Indexed: 04/15/2025]
Abstract
Pancreatic cancer, a leading cause of cancer-related deaths, is characterized by increased dependence on glutamine metabolism. Telaglenastat (CB-839), a glutaminase (GLS) inhibitor targets glutamine metabolism; however, its efficacy as monotherapy is limited owing to metabolic adaptations. In this study, we demonstrated that CB-839 effectively inhibited cell growth in pancreatic cancer cells, but activated the general control nonderepressible 2 (GCN2)-activating transcription factor 4 (ATF4) signaling pathway. ATF4 knockdown reduced glutamine transporter alanine, serine, and cysteine transporter 2 (ASCT2) expression, glutamine uptake, and cell viability under glutamine deprivation-recovery conditions, confirming its protective role in mitigating glutamine-related metabolic stress. Notably, the combination of CB-839 and the ASCT2 inhibitor V-9302 demonstrated a synergistic effect, significantly suppressing pancreatic cancer cell survival. These findings highlight ATF4 and ASCT2 as crucial therapeutic targets and indicate that dual inhibition of GLS and ASCT2 may enhance treatment outcomes for pancreatic cancer.
Collapse
Affiliation(s)
- Dong-Hwan Kim
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Dong Joon Kim
- Department of Microbiology, College of Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Seong-Jun Park
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Won-Jun Jang
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Chul-Ho Jeong
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| |
Collapse
|
18
|
Caggiano EG, Hernandez AL, Waldrop T, Liu K, Gatica-Gutierrez H, Vargas-Hernández S, Mims N, Acevedo-Diaz A, Velasquez B, Neil D, Aguilar E, Meyer MD, Echeverria GV, Koong AC, Spiotto MT, Gustavsson AK, Schüler E. Mitochondrial Responses to Conventional and Ultra-high Dose Rate (FLASH) Radiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.03.647049. [PMID: 40291669 PMCID: PMC12026588 DOI: 10.1101/2025.04.03.647049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Purpose Ultra-high dose rate (>40 Gy/s, FLASH) radiation therapy (RT) provides equivalent tumor control while reducing normal tissue toxicity relative to conventional dose rate (CONV) RT. However, the mechanisms underlying the observed FLASH effect are unknown. We hypothesized that the preservation of mitochondrial integrity in nontumorigenic cells by FLASH RT could be a key factor in reducing normal tissue toxicity and improving overall treatment outcomes. Methods We examined mitochondrial health and function after CONV and FLASH in vitro, ex vivo, and in vivo through assays of metabolic flux, mitochondrial membrane potential, mitochondrial reactive oxygen species (ROS), mitochondrial DNA damage and copy number, mitochondrial morphology, and tumor growth and survival. Results In in vitro assays, murine pancreatic cancer (PDAC) cells showed evidence of equal mitochondrial damage in response to CONV and FLASH, but nontumorigenic pancreatic cells were spared by FLASH. These results were recapitulated ex vivo, and mice treated with FLASH showed higher response rates and longer survival time than mice treated with CONV in an in vivo tumor model. Conclusions Collectively, these results suggest that FLASH spares mitochondrial function in nontumorigenic cells, but not in PDAC cells, relative to CONV. The preservation of mitochondrial integrity in nontumorigenic cells may be a key mechanism underlying the reduced normal tissue toxicity observed with FLASH RT.
Collapse
|
19
|
Azeez A, Noel C. Global status of research on quality of life in pancreatic cancer patients: A bibliometric and network analysis from 2005-2024. Clin Res Hepatol Gastroenterol 2025; 49:102595. [PMID: 40210107 DOI: 10.1016/j.clinre.2025.102595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 04/05/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Pancreatic cancer (PC) is a major global health challenge, with rising incidence and mortality rates, particularly in high-socio-demographic index regions. Given its high mortality and significant morbidity, research on patient quality of life (QoL) has gained momentum, addressing symptom burdens, psychological distress, and treatment-related outcomes. Bibliometric analysis provides a valuable approach to mapping research trends, identifying key contributors, and forecasting future directions. OBJECTIVE This study aimed to map global research on QoL in pancreatic cancer patients, highlighting key findings, challenges, and future directions through bibliometric analysis over the past two decades. METHODS Data for this study were collected from the Web of Science Core Collection (WoSCC) database, using specific search strategies to retrieve relevant documents on the quality of life in pancreatic cancer patients. The data were analysed using the Bibliometrix R package to create knowledge maps and visualize research trends, collaborations, and emerging hotspots in the field. RESULTS A total of 819 articles on pancreatic cancer and quality of life were identified, with an average citation count of 47.13 per article, highlighting moderate academic impact. The research revealed a growing trend in collaborative efforts, with an average of 9.42 co-authors per article and 16 % international collaborations. The United States emerged as the leading contributor, with 203 publications and the highest citation count, followed by France and the United Kingdom. CONCLUSION This bibliometric analysis highlights the growing volume of pancreatic cancer and quality of life research, with a steady annual growth rate of 6.9 % and increasing collaboration, especially from the United States. However, despite the rising number of publications, a decline in citation impact for recent studies suggests a need for continued innovation in therapeutic strategies to improve clinical outcomes.
Collapse
Affiliation(s)
- Adeboye Azeez
- Gastrointestinal Research Unit, Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa.
| | - Colin Noel
- Gastrointestinal Research Unit, Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| |
Collapse
|
20
|
Liang KL, Azad NS. Immune-Based Strategies for Pancreatic Cancer in the Adjuvant Setting. Cancers (Basel) 2025; 17:1246. [PMID: 40227779 PMCID: PMC11988091 DOI: 10.3390/cancers17071246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/15/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer-related mortality in the United States, with poor overall survival across all stages. Less than 20% of patients are eligible for curative surgical resection at diagnosis, and despite adjuvant chemotherapy, most will experience disease recurrence within two years. The incorporation of immune-based strategies in the adjuvant setting remains an area of intense investigation with unrealized promise. It offers the potential of providing durable disease control for micro-metastatic disease following curative intent surgery and enabling personalized treatments based on mutational neoantigen profiles derived from resected specimens. However, most of these attempts have failed to demonstrate significant clinical success, likely due to the immunosuppressive tumor microenvironment (TME) and individual genetic heterogeneity. Despite these challenges, immune-based strategies, such as therapeutic vaccines targeted towards neoantigens, have demonstrated promise via immune activation and induction of T-cell tumor infiltration. In this review, we will highlight the foundational lessons learned from previous clinical trials of adjuvant immunotherapy, discussing the knowledge gained from analyses of trials with disappointing results. In addition, we will discuss how these data have been incorporated to design new agents and study concepts that are proving to be exciting in more recent trials, such as shared antigen vaccines and combination therapy with immune-checkpoint inhibitors and chemotherapy. This review will evaluate novel approaches in ongoing and future clinical studies and provide insight into how these immune-based strategies might evolve to address the unique challenges for treatment of PDAC in the adjuvant setting.
Collapse
Affiliation(s)
| | - Nilofer S. Azad
- Department of Oncology, Sidney Kimmel Comprehensive Cancer, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| |
Collapse
|
21
|
Powell J, Viniotis AF, Irwin C, Jameson GS, Caldwell L, Borazanci EH. Retrospective analysis of capecitabine maintenance therapy in pancreatic ductal adenocarcinoma. Ther Adv Med Oncol 2025; 17:17588359251321894. [PMID: 40290464 PMCID: PMC12032433 DOI: 10.1177/17588359251321894] [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: 09/02/2024] [Accepted: 01/28/2025] [Indexed: 04/30/2025] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PC) is an aggressive form of cancer treated with chemotherapy regimens such as leucovorin, 5-fluorouracil (5-FU), irinotecan, oxaliplatin (FOLFIRINOX), and gemcitabine plus albumin-bound paclitaxel (nab-Paclitaxel). Maintenance chemotherapy is increasingly being studied as an option for patients with a prior response to chemotherapy, prolonged stable disease, and those unable to tolerate the toxicities of traditional chemotherapy. Objective Our retrospective analysis aims to evaluate the effectiveness and tolerability of capecitabine as maintenance therapy in patients with PC. Design Thirty-three patients treated for PC with capecitabine (an oral formulation of 5-FU) maintenance therapy at our single institution between 8/01/2013 and 9/02/2021 were identified on chart review via the electronic medical record (EMR). Methods Kaplan-Meier curves were fit to evaluate patient progression-free survival (PFS) and overall survival (OS). Results Thirty-three individuals were identified: 21 males and 12 females, with a median age of 69 years. Fifteen of 33 had stage IV PC. Nineteen had progression of disease; 8 completed therapy and transitioned to observation or other treatments; 2 did not tolerate treatment; and 4 were still undergoing treatment. The median PFS was 13.01 months (396.0 days), and the median OS was 28.42 months (865.0 days). Conclusion Maintenance with capecitabine seems safe and may represent a valuable option in patients with advanced PC controlled using FOLFIRINOX or gemcitabine/nab-Paclitaxel induction treatment.
Collapse
Affiliation(s)
- Jordan Powell
- The University of Arizona College of Medicine, Phoenix, AZ, USA
| | - Alexa F. Viniotis
- HonorHealth Thompson Peak Internal Medicine Residency, 7400 E Thompson Peak Parkway, Scottsdale, AZ 85255, USA
| | - Chase Irwin
- The University of Arizona College of Medicine, Phoenix, AZ, USA
| | - Gayle S. Jameson
- HonorHealth Research and Innovation Institute, Scottsdale, AZ, USA
| | - Lana Caldwell
- HonorHealth Research and Innovation Institute, Scottsdale, AZ, USA
| | | |
Collapse
|
22
|
Sezgin Y, Karhan O, Aldemir MN, Ürün M, Erçek BM, Urakcı Z, Arvas H, Tunç S, Erdem M, Yerlikaya H, İleri S, Aydın İ, Bicer A, Kömüroğlu AU, Majidova N, Gökçek S, Demir H, Yıldız S, Akbaş S, Özen E, Kahya BU, Sali M, Anık H, Aykut T, Araz M, Alkan A, Özçelik M, Sakin A, Aykan MB, Mehtıyev M, Demir B, Başer MN, Sönmez M, Gültürk İ, Avcı N, Urvay S, Arıcı MÖ, Kalender ME, Yıldırım M, Solmaz AA, Gürbüz M, Ergün Y. Efficacy of gemcitabine plus nab-paclitaxel in second-line treatment of metastatic pancreatic cancer. Sci Rep 2025; 15:11675. [PMID: 40188172 PMCID: PMC11972389 DOI: 10.1038/s41598-025-96157-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
Despite numerous studies on second-line therapies in metastatic pancreatic cancer, there is no randomized study evaluating the efficacy of gemcitabine plus nab-paclitaxel as a second-line treatment. This study aims to examine the efficacy of gemcitabine plus nab-paclitaxel in second-line therapy. In this retrospective study, a total of 218 patients from 23 centers were included. The primary endpoint was progression-free survival (PFS), secondary endpoints included overall survival (OS), treatment efficacy based on ECOG performance status (PS), and tumor marker (CEA, CA 19 - 9) levels. In the second-line treatment with gemcitabine plus nab-paclitaxel, the median PFS was 5.1 months (95% CI, 5.6 to 7.1), and the median OS was 8.6 months (95% CI, 7.3 to 10.0). Median PFS was 6.6 months in patients with normal CEA levels compared to 4.4 months in patients with high CEA levels (P = 0.01). Median PFS was 6 months in patients with ECOG PS 0-1 compared to 3.8 months in patients with PS 2 (P < 0.01). This study demonstrates the contribution of gemcitabine plus nab-paclitaxel in both OS and PFS in second-line treatment of metastatic pancreatic cancer. It was found to be a good option especially for young patients with good ECOG PS.
Collapse
Affiliation(s)
- Yasin Sezgin
- Department of Medical Oncology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey.
| | - Oğur Karhan
- Department of Medical Oncology, Faculty of Medicine, Harran University, Şanlıurfa, Turkey
| | - Mehmet Naci Aldemir
- Department of Medical Oncology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey
| | - Muslih Ürün
- Department of Medical Oncology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey
| | - Berrak Mermit Erçek
- Department of Medical Oncology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey
| | - Zuhat Urakcı
- Department of Medical Oncology, Faculty of Medicine, Dicle University, Diyarbakır, Turkey
| | - Hayati Arvas
- Department of Medical Oncology, Faculty of Medicine, Dicle University, Diyarbakır, Turkey
| | - Sezai Tunç
- Urfa Mehmet Akif Inan Regional Training Research Hospital Oncology Department, Şanlıurfa, Turkey
| | - Mehmet Erdem
- Department of Internal Medicine, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey
| | - Halis Yerlikaya
- Diyarbakır private Batı Hospital Medical Oncology Department, Diyarbakır, Turkey
| | - Serdar İleri
- Gazi Yaşargil Regional Training Research Hospital Oncology Department, Diyarbakır, Turkey
| | - İbrahim Aydın
- Department of Internal Medicine, Van Regional Training Research Hospital, Van, Turkey
| | - Abdurrahman Bicer
- Department of Internal Medicine, Van Regional Training Research Hospital, Van, Turkey
| | - Ahmet Ufuk Kömüroğlu
- Health Service Vocational School of Higher Education, Van Yüzüncü Yıl University, Van, Turkey
| | - Nargiz Majidova
- Department of Medical Oncology, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Savaş Gökçek
- Department of Medical Oncology, Faculty of Medicine, Eyül University, İzmir, Turkey
| | - Hacer Demir
- Department of Medical Oncology, Faculty of Medicine, Afyon Karahisar University, Afyonkarahisar, Turkey
| | - Sedat Yıldız
- Department of Medical Oncology, Faculty of Medicine, Afyon Karahisar University, Afyonkarahisar, Turkey
| | - Sinem Akbaş
- Department of Medical Oncology, Faculty of Medicine, Koç University, Istanbul, Turkey
| | - Esra Özen
- Department of Medical Oncology, Faculty of Medicine, Sakarya University, Sakarya, Turkey
| | - Burcu Ulaş Kahya
- Department of Medical Oncology, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Mürsel Sali
- Department of Medical Oncology, Faculty of Medicine, Uludağ University, Bursa, Turkey
| | - Hicran Anık
- Abdurrahman Yurtaslan Regional Training Research Hospital Oncology Department, Ankara, Turkey
| | - Talat Aykut
- Department of Medical Oncology, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Murat Araz
- Department of Medical Oncology, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Ali Alkan
- Department of Medical Oncology, Faculty of Medicine, Muğla Sıtkı Kocaman University, Muğla, Turkey
| | - Melike Özçelik
- Ümraniye Regional Training Research Hospital Oncology Department, Istanbul, Turkey
| | - Abudllah Sakin
- İstanbul Private Medicalpark Hospital Medical Oncology Department, Istanbul, Turkey
| | - Musa Barış Aykan
- Gülhane Regional Training Research Hospital Oncology Department, Ankara, Turkey
| | - Mirmehdi Mehtıyev
- Department of Medical Oncology, Bilkent City Hospital, Ankara, Turkey
| | - Bilgin Demir
- Faculty of Medicine, Department of Medical Oncology, Adana Menderes University, Aydın, Turkey
| | - Mehmet Nuri Başer
- Faculty of Medicine, Department of Medical Oncology, Adana Menderes University, Aydın, Turkey
| | - Müge Sönmez
- Department of Medical Oncology, Faculty of Medicine, Department of Medical Oncology, Ordu State Hospital, Sanko University, 26 İstanbul Regional, Istanbul, Turkey
| | - İlkay Gültürk
- Department of Medical Oncology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey
| | - Nilüver Avcı
- Department of Medical Oncology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey
| | - Semiha Urvay
- Department of Medical Oncology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey
| | - Mustafa Özgür Arıcı
- Department of Medical Oncology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey
| | - Mehmet Emin Kalender
- Department of Medical Oncology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey
| | - Mustafa Yıldırım
- Department of Medical Oncology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey
| | - Ali Alper Solmaz
- Department of Medical Oncology, Adana City Hospital, Adana, Turkey
| | - Mustafa Gürbüz
- Department of Medical Oncology, Adana City Hospital, Adana, Turkey
| | - Yakup Ergün
- Department of Medical Oncology, Diyarbakır Private Bower Hospital, Diyarbakır, Turkey
| |
Collapse
|
23
|
Stoop TF, Javed AA, Oba A, Koerkamp BG, Seufferlein T, Wilmink JW, Besselink MG. Pancreatic cancer. Lancet 2025; 405:1182-1202. [PMID: 40187844 DOI: 10.1016/s0140-6736(25)00261-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 04/07/2025]
Abstract
Pancreatic cancer is frequently a lethal disease with an aggressive tumour biology often presenting with non-specific symptoms. Median survival is approximately 4 months with a 5-year survival of 13%. Surveillance is recommended in individuals with familial pancreatic cancer, specific mutations, and high-risk intraductal papillary mucinous neoplasm, as they are at high risk of developing pancreatic cancer. Chemotherapy combined with surgical resection remains the cornerstone of treatment. However, only a small subset of patients are candidates for surgery. Multi-agent chemotherapy has improved survival in the palliative setting for patients with metastatic disease, as (neo)adjuvant and induction therapy have in patients with borderline resectable and locally advanced pancreatic. Given that pancreatic cancer is predicted to become the second leading cause of cancer-related death by 2030, novel therapies are urgently needed.
Collapse
Affiliation(s)
- Thomas F Stoop
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Ammar A Javed
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands; Division of Surgical Oncology, Department of Surgery, New York University Medical Center, New York, NY, USA
| | - Atsushi Oba
- Department of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan; Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Division of Surgical Oncology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Thomas Seufferlein
- Department of International Medicine I, Ulm University Hospital, Ulm, Germany
| | - Johanna W Wilmink
- Department of Medical Oncology, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Marc G Besselink
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands.
| |
Collapse
|
24
|
Picozzi VJ, Mandelson MT, Najjar A, Li M, Harb DE, Kort JJ. Pancreatic enzyme replacement therapy in advanced adenocarcinoma of the pancreas improved overall survival: a retrospective, single institution study. Oncologist 2025; 30:oyaf014. [PMID: 40231659 PMCID: PMC11997656 DOI: 10.1093/oncolo/oyaf014] [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: 07/22/2024] [Accepted: 01/08/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Weight loss and exocrine pancreatic insufficiency are common in advanced pancreatic ductal adenocarcinoma (PDAC) and are associated with adverse outcomes. However, there is limited evidence on the impact of pancreatic enzyme replacement therapy (PERT) in patients with advanced PDAC. PATIENTS AND METHODS We retrospectively studied 501 patients with advanced PDAC and exocrine pancreatic insufficiency from the Virginia Mason Pancreas Cancer Program Data Resource treated between 2010 and 2019 with first-line chemotherapy. Clinical outcomes were compared between those who received PERT and those who did not at 8 weeks after chemotherapy start. RESULTS In total 188 (38%) patients received PERT; 313 patients (62%) did not. PERT patients experienced less weight loss (-1.5 vs -2.5 kg, P = .04), less decline in the prognostic nutrition index -1.9 vs -3.0, P = .01), and a greater reduction in the additive score of the Patient-Generated Subjective Global Assessment (-8.4 vs --6.0, P = .02). Importantly, median (95% CI) overall survival (OS) was significantly longer in the PERT vs non-PERT group (17.1 months vs 12.5 months, respectively P = .001), and the adjusted hazards ratio indicated superior median OS in patients prescribed PERT (HR = 0.73, P < .001). CONCLUSIONS Our findings suggest that treatment of exocrine pancreatic insufficiency (EPI) in advanced PDAC is associated with improvements in nutrition and overall survival.
Collapse
Affiliation(s)
- Vincent J Picozzi
- Virginia Mason Hospital and Medical Center, Seattle, WA 98111, United States
| | | | - Anas Najjar
- Benaroya Research Institute, Seattle, WA 98101, United States
| | - Moming Li
- Data and Statistical Sciences, AbbVie Inc., North Chicago, IL 60064, United States
| | - Diala E Harb
- US Medical Affairs, AbbVie Inc., Mettawa, IL 60045, United States
| | - Jens J Kort
- US Medical Affairs, AbbVie Inc., Mettawa, IL 60045, United States
| |
Collapse
|
25
|
Ren J, Su T, Ding J, Chen F, Mo J, Li J, Wang Z, Han L, Wu Z, Wu S. Chlorophyllin exerts synergistic anti-tumor effect with gemcitabine in pancreatic cancer by inducing cuproptosis. Mol Med 2025; 31:126. [PMID: 40186145 PMCID: PMC11969790 DOI: 10.1186/s10020-025-01180-y] [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: 11/13/2024] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
Pancreatic cancer (PC) has high lethality due to multiple reasons, and its limited response to conventional chemotherapy like gemcitabine (GEM) is a non-negligible one. Therefore, our study introduces Chlorophyllin (CHL) as an effective therapeutic candidate to enhance the therapeutic efficacy of GEM. Our results demonstrate that the combination of CHL and GEM exhibits a significant synergistic anti-tumor effect by targeting multiple oncogenic processes in PC, including inhibiting cell proliferation, invasion, and migration, as well as inducing cell apoptosis. Further investigations of mechanism have revealed that CHL induces cuproptosis in PC cells through a multifaceted process, involving depleting cellular intracellular glutathione (GSH), increasing reactive oxygen species (ROS) levels, and subsequently upregulating the HSP70 protein in response to heightened oxidative stress. Additionally, CHL releases free Cu2+, binds to the Ferredoxin 1 (FDX1) protein, and ultimately leads to the oligomerization of Dihydrolipoamide S-Acetyltransferase (DLAT) proteins to amplify the copper toxicity within PC cells. Moreover, in vivo experiments have demonstrated that the combination of CHL and GEM effectively inhibits the growth of subcutaneously transplanted tumors while maintaining a favorable biosafety profile. In conclusion, our study identifies CHL as a potent enhancer of GEM's anti-tumor effects in PC through the induction of cuproptosis, thus providing a novel therapeutic avenue for patients with PC.
Collapse
Affiliation(s)
- Jiaqiang Ren
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Tong Su
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Jiachun Ding
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Fan Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jiantao Mo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jie Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Liang Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Zheng Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Shuai Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
26
|
Liaki V, Rosas-Perez B, Guerra C. Unlocking the Genetic Secrets of Pancreatic Cancer: KRAS Allelic Imbalances in Tumor Evolution. Cancers (Basel) 2025; 17:1226. [PMID: 40227826 PMCID: PMC11987834 DOI: 10.3390/cancers17071226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 03/13/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) belongs to the types of cancer with the highest lethality. It is also remarkably chemoresistant to the few available cytotoxic therapeutic options. PDAC is characterized by limited mutational heterogeneity of the known driver genes, KRAS, CDKN2A, TP53, and SMAD4, observed in both early-stage and advanced tumors. In this review, we summarize the two proposed models of genetic evolution of pancreatic cancer. The gradual or stepwise accumulated mutations model has been widely studied. On the contrary, less evidence exists on the more recent simultaneous model, according to which rapid tumor evolution is driven by the concurrent accumulation of genetic alterations. In both models, oncogenic KRAS mutations are the main initiating event. Here, we analyze the emerging topic of KRAS allelic imbalances and how it arises during tumor evolution, as it is often detected in advanced and metastatic PDAC. We also summarize recent evidence on how it affects tumor biology, metastasis, and response to therapy. To this extent, we highlight the necessity to include studies of KRAS allelic frequencies in the design of future therapeutic strategies against pancreatic cancer.
Collapse
Affiliation(s)
- Vasiliki Liaki
- Molecular Oncology Program, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain; (B.R.-P.); (C.G.)
| | - Blanca Rosas-Perez
- Molecular Oncology Program, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain; (B.R.-P.); (C.G.)
| | - Carmen Guerra
- Molecular Oncology Program, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain; (B.R.-P.); (C.G.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
27
|
Shimamaki Y, Takahashi M, Higashihara T, Hayashi T, Morita Y, Azuma T, Inoue D, Okada H, Ohtsuka M. Long-Term Survival after Curative Resection for Postoperative Dissemination of Pancreatic Ductal Adenocarcinoma: A Case Report. Surg Case Rep 2025; 11:24-0022. [PMID: 40196210 PMCID: PMC11975445 DOI: 10.70352/scrj.cr.24-0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/16/2025] [Indexed: 04/09/2025] Open
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) has a very poor prognosis and high mortality. The prognosis for recurrence after surgery is extremely poor. Resection for disseminations of PDAC is not recommended. CASE PRESENTATION The patient was a 69-year-old woman with a pancreatic tumor that was detected with computed tomography (CT) during a postoperative colon cancer checkup. She was suspected of having pancreatic body cancer without distant metastasis. Distal pancreatectomy with celiac axis resection was performed. Postoperative pathological examination revealed an invasive ductal adenocarcinoma with lymph node metastasis (pT4N1M0, stage III). Postoperatively, she received adjuvant chemotherapy containing gemcitabine and S-1 for 1 year and 4 months, and S-1 monotherapy for 1 year. Six years and 2 months after the initial surgery, her serum carbohydrate antigen 19-9 level elevated, and CT revealed soft tissue in front of the left kidney. Positron emission tomography/CT also revealed high fluorine-18 fluorodeoxyglucose uptake in the tissue. Accordingly, the patient was diagnosed with dissemination of PDAC. The patient was administered chemotherapy with gemcitabine and S-1. One year and 6 months after the diagnosis of dissemination, CT revealed reduction of the nodule. Therefore, we decided to eliminate this dissemination. A left nephrectomy and partial gastrectomy were performed. Histopathological examination confirmed dissemination of PDAC. The patient refused adjuvant chemotherapy. No evidence of recurrence has been observed for 13 years and 3 months since the initial surgery, and 5 years and 1 month since the resection of the dissemination. CONCLUSIONS This case showed a recurrence of dissemination after radical PDAC surgery, and the patient showed long-term survival without recurrence after dissemination resection. Resection of dissemination may confer long-term survival in selected patients.
Collapse
Affiliation(s)
- Yoshitaka Shimamaki
- Department of General Surgery, Tokyo Metropolitan Tama Medical Center, Fuchu, Tokyo, Japan
| | - Makoto Takahashi
- Department of General Surgery, Tokyo Metropolitan Tama Medical Center, Fuchu, Tokyo, Japan
| | - Taku Higashihara
- Department of General Surgery, Tokyo Metropolitan Tama Medical Center, Fuchu, Tokyo, Japan
| | - Tatsuya Hayashi
- Department of General Surgery, Tokyo Metropolitan Tama Medical Center, Fuchu, Tokyo, Japan
| | - Yasuhiro Morita
- Department of General Surgery, Tokyo Metropolitan Tama Medical Center, Fuchu, Tokyo, Japan
| | - Takeshi Azuma
- Division of Urology, Tokyo Metropolitan Tama Medical Center, Fuchu, Tokyo, Japan
| | - Dai Inoue
- Department of Gastroenterology and Hepatology, Tokyo Metropolitan Tama Medical Center, Fuchu, Tokyo, Japan
- Department of Clinical Genomics, Tokyo Metropolitan Tama Medical Center, Fuchu, Tokyo, Japan
| | - Haruka Okada
- Department of Pathology, Tokyo Metropolitan Tama Medical Center, Fuchu, Tokyo, Japan
| | - Masayuki Ohtsuka
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Chiba, Japan
| |
Collapse
|
28
|
Zhou K, Liu Y, Tang C, Zhu H. Pancreatic Cancer: Pathogenesis and Clinical Studies. MedComm (Beijing) 2025; 6:e70162. [PMID: 40182139 PMCID: PMC11965705 DOI: 10.1002/mco2.70162] [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: 06/24/2024] [Revised: 03/08/2025] [Accepted: 03/12/2025] [Indexed: 04/05/2025] Open
Abstract
Pancreatic cancer (PC) is a highly lethal malignancy, with pancreatic ductal adenocarcinoma (PDAC) being the most common and aggressive subtype, characterized by late diagnosis, aggressive progression, and resistance to conventional therapies. Despite advances in understanding its pathogenesis, including the identification of common genetic mutations (e.g., KRAS, TP53, CDKN2A, SMAD4) and dysregulated signaling pathways (e.g., KRAS-MAPK, PI3K-AKT, and TGF-β pathways), effective therapeutic strategies remain limited. Current treatment modalities including chemotherapy, targeted therapy, immunotherapy, radiotherapy, and emerging therapies such as antibody-drug conjugates (ADCs), chimeric antigen receptor T (CAR-T) cells, oncolytic viruses (OVs), cancer vaccines, and bispecific antibodies (BsAbs), face significant challenges. This review comprehensively summarizes these treatment approaches, emphasizing their mechanisms, limitations, and potential solutions, to overcome these bottlenecks. By integrating recent advancements and outlining critical challenges, this review aims to provide insights into future directions and guide the development of more effective treatment strategies for PC, with a specific focus on PDAC. Our work underscores the urgency of addressing the unmet needs in PDAC therapy and highlights promising areas for innovation in this field.
Collapse
Affiliation(s)
- Kexun Zhou
- Department of Medical OncologyCancer CenterWest China HospitalSichuan UniversityChengduChina
| | - Yingping Liu
- Department of RadiotherapyCancer HospitalChinese Academy of Medical SciencesBeijingChina
| | - Chuanyun Tang
- The First Clinical Medical College of Nanchang UniversityNanchang UniversityNanchangChina
| | - Hong Zhu
- Department of Medical OncologyCancer CenterWest China HospitalSichuan UniversityChengduChina
- Division of Abdominal Tumor Multimodality TreatmentCancer CenterWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
29
|
Shirai Y, Kimura N, Tanaka H, Fukasawa M, Muranushi R, Watanabe T, Hirano K, Shibuya K, Yoshioka I, Fujii T. Risk Factors for Early Liver Recurrence After Pancreatic Cancer Resection. Pancreas 2025; 54:e324-e330. [PMID: 39626192 DOI: 10.1097/mpa.0000000000002441] [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: 06/02/2024] [Accepted: 11/11/2024] [Indexed: 04/24/2025]
Abstract
BACKGROUND Liver recurrence after resection is one of the most common types of recurrence and is a risk factor for poor prognosis. The aim of this study was to identify risk factors for initial liver recurrence. MATERIALS AND METHODS A total of 109 patients with resectable pancreatic ductal adenocarcinoma who underwent resection between 2015 and 2022 were included. The influence of clinicopathologic variables on liver recurrence was analyzed to create a novel scoring system to predict liver recurrence. RESULTS The liver recurrence rate was 24%, with 17% recurrence within 1 year. Patients with liver recurrence had an extremely poor prognosis within 1 year (MST 12.4 [95% CI, 5.7-19.1]). In multivariate analysis, R-PV, large tumor diameter ≥45 mm, and venous invasion were independent risk factors for early liver recurrence. When each of these risk factors was scored as 1 point, the 1-year liver recurrence rates by score were 0 (0%), 1 (9%), 2 (30%), and 3 (84%). CONCLUSION The risk factors for postoperative early liver recurrence were R-PV, a tumor diameter ≥45 mm, and pathological venous invasion. Surgery for pancreatic cancer with these factors may require special treatment, such as hepatic arterial injection.
Collapse
Affiliation(s)
- Yoshihiro Shirai
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Sugitani, Toyama, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Wirth TC, Saborowski A, Kuehnle E, Fischer M, Bültmann E, von Kaisenberg C, Merten R. Chemo- and Radiotherapy of Gastrointestinal Tumors during Pregnancy. Visc Med 2025; 41:64-73. [PMID: 40201111 PMCID: PMC11975343 DOI: 10.1159/000540428] [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: 03/24/2024] [Accepted: 07/16/2024] [Indexed: 04/10/2025] Open
Abstract
Background Gastrointestinal cancers account for approximately one-fourth of all cancer cases and one-third of all tumor-related deaths worldwide. For the most frequent gastrointestinal tumor entities including colorectal, gastric, esophageal, and liver cancer, the incidence is expected to increase by more than 50% until 2040. While most gastrointestinal cancers are diagnosed beyond the age of fertility and predominantly in men, the increasing incidence of gastrointestinal malignancies in patients below the age of fifty suggests a growing importance in women of childbearing age. While localized cancers in pregnant women can either be monitored or treated surgically, more advanced stages might require radio- or chemotherapy to control tumor growth until delivery. Under these circumstances, critical decisions have to be made to preserve maternal health on the one side and minimize harm to the infant on the other side. Summary Here we summarize data from case reports, meta-analyses, and registries of women undergoing radio- or chemotherapy during pregnancy and provide guidance for therapeutic decision-making in pregnant women suffering from gastrointestinal cancers. Key Message After the first trimester, most chemotherapeutic regimens can be safely administered to pregnant patients with gastrointestinal cancers. With appropriate safety measures, both radiotherapy and radiochemotherapy can be applied to pregnant patients with rectal cancers.
Collapse
Affiliation(s)
- Thomas Christian Wirth
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Anna Saborowski
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Elna Kuehnle
- Department of Obstetrics and Gynaecology, Hannover Medical School, Hannover, Germany
| | - Mirko Fischer
- Department of Radiation Oncology, Hannover Medical School, Hannover, Germany
| | - Eva Bültmann
- Institute of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hannover, Germany
| | | | - Roland Merten
- Department of Radiation Oncology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
31
|
Zhou E, Yang JI, Habowski AN, Deschênes A, Belleau P, Ha T, Tzanavaris CJ, Boyd J, Hollweg CA, Zhu X, Tuveson DA, King DA. GATA6 Amplification Is Associated With Improved Survival in TP53-Mutated Unresectable Pancreatic Cancer. Pancreas 2025; 54:e303-e309. [PMID: 40262102 DOI: 10.1097/mpa.0000000000002431] [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: 05/23/2024] [Accepted: 11/01/2024] [Indexed: 04/24/2025]
Abstract
OBJECTIVES GATA6 expression is recognized as a favorable prognostic marker of pancreatic cancer, whereas TP53 is a poor prognostic marker. We evaluated treatment outcomes by genetic alterations in TP53 and GATA6 to determine the prognostic and predictive impact of co-alterations. MATERIALS AND METHODS A single institution retrospective analysis was performed on patients diagnosed with pancreatic ductal adenocarcinoma between 2014 and 2023. TP53 genotype and GATA6 amplification status were included in an analysis of overall survival (OS) and progression-free survival (PFS). Previously published patient-derived organoids were used to investigate correlation between genetic status and drug sensitivity. RESULTS Patients with TP53 mutations had worse OS compared with the wild-type TP53 population. Patients with GATA6 amplification had better OS and a trend toward better PFS than the nonamplified population. Among patients with a TP53 mutation, patients with GATA6 co-alteration had longer OS compared with those who were not GATA6 amplified. In contrast, among patients who were TP53 wild-type, the presence or absence of a GATA6 amplification did not impact OS or PFS. GATA6 genotype was not associated chemotherapy drug response in an organoid pharmacotyping model. CONCLUSIONS We found that GATA6 amplification appeared to attenuate poor prognosis observed in TP53-mutant patients regardless of the type of standard chemotherapy received, suggesting the GATA6 amplification is a prognostic biomarker but not a predictive biomarker of standard-of-care chemotherapy.
Collapse
Affiliation(s)
- Edward Zhou
- Northwell Health Cancer Institute, New York, NY
| | | | | | | | - Pascal Belleau
- Cancer Center, Cold Spring Harbor Laboratory, New York, NY
| | - Taehoon Ha
- Cancer Center, Cold Spring Harbor Laboratory, New York, NY
| | - Chris J Tzanavaris
- Division of Pulmonary, Critical Care, and Sleep Medicine, Northwell Health, New York, NY
| | - Jeff Boyd
- Northwell Health Cancer Institute, New York, NY
| | - Christopher A Hollweg
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, NY
| | - Xinhua Zhu
- Northwell Health Cancer Institute, New York, NY
| | | | | |
Collapse
|
32
|
Tomooka F, Kitagawa K, Mitoro A, Fujinaga Y, Nishimura N, Namisaki T, Akahane T, Kaji K, Asada S, Sato S, Hanatani J, Mori H, Motokawa Y, Iwata T, Kachi H, Osaki Y, Yoshiji H. Unilateral drainage and chemotherapy prolong the patency of a plastic stent placed above the sphincter of Oddi in patients with malignant hilar biliary obstruction. DEN OPEN 2025; 5:e404. [PMID: 39011511 PMCID: PMC11248713 DOI: 10.1002/deo2.404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/17/2024] [Accepted: 06/24/2024] [Indexed: 07/17/2024]
Abstract
OBJECTIVES To evaluate the results of inside stent therapy for unresectable malignant hilar biliary obstruction and identify factors related to stent patency duration. METHODS Of 44 patients who underwent initial inside-stent placement above the sphincter of Oddi from April 2017 to December 2022, 42 with the resolution of jaundice (clinical success rate, 95.5%) were retrospectively analyzed. Univariate and multivariate logistic regression analysis identified factors associated with stent patency duration. RESULTS Univariate analysis revealed significant differences in the drainage method (406 days for unilateral drainage vs. 305 days for bilateral drainage of the right and left liver lobes, p = 0.022) with or without chemotherapy (406 days with vs. 154 days without, p = 0.038). Multivariate analysis (Cox proportional hazards analysis) revealed similar results, with unilateral drainage (p = 0.031) and chemotherapy (p = 0.048) identified as independent factors associated with prolonged stent patency. Early adverse events were observed in two patients (4.8%; one cholangitis, one pancreatitis). CONCLUSIONS Inside-stent therapy was safely performed in patients with malignant hilar biliary obstruction. Simple unilateral drainage and chemotherapy may prolong stent patency.
Collapse
Affiliation(s)
| | - Koh Kitagawa
- Department of GastroenterologyNara Medical UniversityNaraJapan
| | - Akira Mitoro
- Division of EndoscopyNara Medical UniversityNaraJapan
| | | | | | | | - Takemi Akahane
- Department of GastroenterologyNara Medical UniversityNaraJapan
| | - Kosuke Kaji
- Department of GastroenterologyNara Medical UniversityNaraJapan
| | - Shohei Asada
- Department of GastroenterologyNara Medical UniversityNaraJapan
| | - Shinya Sato
- Department of GastroenterologyNara Medical UniversityNaraJapan
| | | | - Hitoshi Mori
- Department of GastroenterologyNara Medical UniversityNaraJapan
| | - Yuki Motokawa
- Department of GastroenterologyNara Medical UniversityNaraJapan
| | - Tomihiro Iwata
- Department of GastroenterologyNara Medical UniversityNaraJapan
| | - Hiroki Kachi
- Department of GastroenterologyNara Medical UniversityNaraJapan
| | - Yui Osaki
- Department of GastroenterologyNara Medical UniversityNaraJapan
| | - Hitoshi Yoshiji
- Department of GastroenterologyNara Medical UniversityNaraJapan
| |
Collapse
|
33
|
Li Y, Lam SSK, Gao Y, Shore E, Anderson DW, Hode T, Martin RC. Enhancing the Immunotherapeutic Effect by IP-001 and Irreversible Electroporation in Mouse Oligometastatic Models of Pancreatic Adenocarcinoma. Ann Surg Oncol 2025; 32:2786-2798. [PMID: 39739260 DOI: 10.1245/s10434-024-16742-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/10/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND This study aimed to evaluate the immunotherapeutic effect of irreversible electroporation (IRE) and IP-001 in pancreatic adenocarcinoma with metastasis. METHODS Orthotopic models of pancreatic adenocarcinoma with hepatic oligometastasis were established by implantation of tumor tissues (derived from Pan02 or KPC cells) size 2 mm3 into the pancreas and left liver lobe in C57BL/6J mice. One week after implantation, the tumor-burden mice were subjected to saline control, IRE, IP-001, and IRE+IP-001. For IRE therapy (1000 V, 0.1 ms, 10 pulses administered 10 times), the pancreas tumor was treated, whereas the oligometastasis was untreated as the IRE off-target tumor. Intratumoral administration of IP-001(0.4 ml/kg) was performed. RESULTS In the KPC oligometastatic model, IRE+IP-001 therapy significantly suppressed the growth of oligometastatic tumor. Flow cytometry showed significantly increased tumor-infiltrating lymphocytes (TILs) (e.g., CD8+ cytotoxic T lymphocytes) and significantly increased monocytes/macrophages in the oligometastatic tumor tissues from IRE+IP-001 treatment compared with the sham control. Significantly decreased Treg cells and tumor-associated macrophages (TAMs) also were found in the oligometastatic tumor tissues from IRE+IP-001 treatment compared with the sham control. In the Pan02 oligometastatic model, both IRE+IP-001 therapy and IRE+anti-PD-L1 immunotherapy significantly suppressed the growth of oligometastatic tumor, which was associated with the increased CD8+ cytotoxic T lymphocytes. However, increased monocytes/macrophages were found in the mice that had IRE+IP-001 therapy, but not in the mice that had IRE+anti-PD-L1 immunotherapy. CONCLUSION The study provided compelling evidence for the efficacy of IRE&IP-001 therapy in suppressing pancreatic tumors, including off-target oligometastatic lesions. The observed off-target effect underscores the importance of systemic immune activation in achieving effective tumor control.
Collapse
Affiliation(s)
- Yan Li
- Division of Surgical Oncology, Department of Surgery, School of Medicine, University of Louisville, Louisville, KY, USA.
| | | | - Yonglin Gao
- Division of Surgical Oncology, Department of Surgery, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Emily Shore
- Division of Surgical Oncology, Department of Surgery, School of Medicine, University of Louisville, Louisville, KY, USA
| | | | | | - Robert C Martin
- Division of Surgical Oncology, Department of Surgery, School of Medicine, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
34
|
Steen TV, Espinoza I, Duran C, Casadevall G, Serrano-Hervás E, Cuyàs E, Verdura S, Kemble G, Kaufmann SH, McWilliams R, Osuna S, Billadeau DD, Menendez JA, Lupu R. Fatty acid synthase (FASN) inhibition cooperates with BH3 mimetic drugs to overcome resistance to mitochondrial apoptosis in pancreatic cancer. Neoplasia 2025; 62:101143. [PMID: 39999714 PMCID: PMC11908614 DOI: 10.1016/j.neo.2025.101143] [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: 09/05/2024] [Accepted: 02/17/2025] [Indexed: 02/27/2025]
Abstract
Resistance to mitochondrial apoptosis is a major driver of chemoresistance in pancreatic ductal adenocarcinoma (PDAC). However, pharmacological manipulation of the mitochondrial apoptosis threshold in PDAC cells remains an unmet therapeutic goal. We hypothesized that fatty acid synthase inhibitors (FASNis), a family of targeted metabolic therapeutics recently entering the clinic, could lower the apoptotic threshold in chemoresistant PDAC cells and be synergistic with BH3 mimetics that neutralize anti-apoptotic proteins. Computational studies with TVB-3166 and TVB-3664, two analogues of the clinical-grade FASNi TVB-2640 (denifanstat), confirmed their uncompetitive behavior towards NADPH when bound to the FASN ketoacyl reductase domain. The extent of NADPH accumulation, a consequence of FASN inhibition, paralleled the sensitivity of PDAC cells to the apoptotic effects of TVB FASNis in conventional PDAC cell lines that naturally express varying levels of FASN. FASN inhibition dramatically increased the sensitivity of "FASN-high" expressing PDAC cells to the BCL2/BCL-XL/BCL-W inhibitor ABT-263/navitoclax and the BCL2-selective inhibitor ABT-199/venetoclax, both in vitro and in in vivo xenografted tumors. The ability of TVB FASNis to shift the balance of pro- and anti-apoptotic proteins and thereby push PDAC cells closer to the apoptotic threshold was also observed in cell lines developed from patient-derived xenografts (PDXs) representative of the classical (pancreatic) transcriptomic subtype of PDAC. Experiments in PDAC PDXs in vivo confirmed the synergistic antitumor activity of TVB-3664 with navitoclax and venetoclax, independent of the nature of the replication stress signature of patient-derived PDAC cells. The discovery that targeted inhibition of FASN is a metabolic perturbation that sensitizes PDAC cells to BH3 mimetics warrants further investigation to overcome resistance to mitochondrial apoptosis in PDAC patients.
Collapse
Affiliation(s)
- Travis Vander Steen
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Ingrid Espinoza
- National Institute of Health, National Heart Lung and Blood Institute (NHLBI), Bethesda, MD 20817, USA; Lung Development and Pediatric Branch (HNH36), Bethesda, MD 20817, USA
| | - Cristina Duran
- Institut de Química Computacional i Catàlisi and Departament de Química, Universitat de Girona, Girona 17003, Spain
| | - Guillem Casadevall
- Institut de Química Computacional i Catàlisi and Departament de Química, Universitat de Girona, Girona 17003, Spain
| | - Eila Serrano-Hervás
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona 17007, Spain; Metabolism and Cancer Group,Girona Biomedical Research Institute (IDIBGI), Salt 17190, Girona, Spain
| | - Elisabet Cuyàs
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona 17007, Spain; Metabolism and Cancer Group,Girona Biomedical Research Institute (IDIBGI), Salt 17190, Girona, Spain
| | - Sara Verdura
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona 17007, Spain; Metabolism and Cancer Group,Girona Biomedical Research Institute (IDIBGI), Salt 17190, Girona, Spain
| | | | - Scott H Kaufmann
- Mayo Clinic Cancer Center, Rochester, MN 55905, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA; Division of Oncology Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Robert McWilliams
- Mayo Clinic Cancer Center, Rochester, MN 55905, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Sílvia Osuna
- Institut de Química Computacional i Catàlisi and Departament de Química, Universitat de Girona, Girona 17003, Spain; ICREA, Barcelona 08010, Spain
| | - Daniel D Billadeau
- Mayo Clinic Cancer Center, Rochester, MN 55905, USA; Division of Oncology Research, Mayo Clinic, Rochester, MN, 55905, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA; Department of Immunology College of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Javier A Menendez
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona 17007, Spain; Metabolism and Cancer Group,Girona Biomedical Research Institute (IDIBGI), Salt 17190, Girona, Spain.
| | - Ruth Lupu
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; Mayo Clinic Cancer Center, Rochester, MN 55905, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
35
|
Ikeda M, Okusaka T, Ueno M, Ozaka M, Satoi S, Skanji D, Martín-Fernández L, Amellal N, Furuse J. NALIRIFOX in Japanese treatment-naïve patients with metastatic pancreatic adenocarcinoma: an open-label, phase II trial design. Future Oncol 2025; 21:959-965. [PMID: 39987459 PMCID: PMC11938982 DOI: 10.1080/14796694.2025.2469467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 02/17/2025] [Indexed: 02/25/2025] Open
Abstract
This is a single arm, open label trial to assess the safety and efficacy of NALIRIFOX in Japanese participants who have not previously received therapy for metastatic adenocarcinoma of the pancreas. Forty-one participants will receive the treatment NALIRIFOX (liposomal irinotecan, 5-fluorouracil, levoleucovorin and oxaliplatin) until progression or unacceptable toxicity. Anti-tumor activity will be evaluated in terms of objective response rate according to RECIST v1.1 by independent central review. Safety will be carefully monitored, with a dedicated Safety Monitoring Committee reviewing the data from the initial six participants to safeguard the well-being of all individuals involved in the study. Other evaluations will include pharmacokinetics and the relationship between mutations in the UGT1A1 genes and safety. This study aims to show that the results of the NAPOLI 3 study in the global population can be reproduced in the Japanese population.Clinical trials registration: NCT06225999 (clinicaltrials.gov); jRCT2031230733 (Japanese clinical trials registry).
Collapse
Affiliation(s)
| | | | | | - Masato Ozaka
- Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | | | - Donia Skanji
- Institut de Recherches Internationales Servier, Gif sur Yvette, France
| | | | - Nadia Amellal
- Institut de Recherches Internationales Servier, Gif sur Yvette, France
| | | |
Collapse
|
36
|
Hu Q, Zhang F, Li B, Jin P, Huang J, Yao Z, Zhao X, Shao S, Wang M, Ping Y, Liang T. Engineered a Gemcitabine Nano-Prodrug Targeting Desmoplastic Pancreatic Tumor with Dual Enhancement of Penetration Dynamics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410629. [PMID: 40007057 DOI: 10.1002/smll.202410629] [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: 11/08/2024] [Revised: 02/18/2025] [Indexed: 02/27/2025]
Abstract
The desmoplastic nature characterized by dense tissue and poor vascular conditions in pancreatic cancer (PDAC) poses a significant barrier to effective chemotherapy. Targeting the extracellular matrix and reducing desmoplasia in the tumor microenvironment is a rational approach to improve the penetration and efficacy of gemcitabine (GEM) in PDAC. Herein, a small molecular self-assembly nano-prodrug is developed for the "three-in-one" co-delivery of GEM chemotherapeutics, all-trans retinoic acid (ATRA), and nitric oxide (NO) donors, decorated with PDAC-homing targeting peptide. Stimulated by the PDAC microenvironment, the nano-prodrug releases ATRA that quiesces activated pancreatic stellate cells to alleviate stromal desmoplasia. Simultaneously, it generates abundant NO to induce a vasodilatory effect as well as a "nanomotor" effect. This combinational nano-prodrug delivery strategy, with a dual enhancement of drug penetration dynamics, effectively improves the distribution and bioavailability of the co-delivered GEM into the deep and dense pancreatic tumor tissue, which leads to significant amplification of tumor growth inhibition in different PDAC mouse models.
Collapse
Affiliation(s)
- Qida Hu
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Fu Zhang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Bowen Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive, Singapore, 117585, Singapore
| | - Piaopiao Jin
- Healthcare Management Center, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Junming Huang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Zhuo Yao
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xinyu Zhao
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Shiyi Shao
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Meng Wang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yuan Ping
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, 310003, China
- Innovation Center for the Study of Pancreatic Diseases, Hangzhou, 310003, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Hangzhou, 310003, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
- Research Center for Healthcare Data Science, Zhejiang Lab., Hangzhou, 311121, China
| |
Collapse
|
37
|
Addeo P, Muzzolini M, Laurent C, Heyd B, Sauvanet A, Garnier J, Alfano MS, Gaujoux S, De Ponthaud C, Marchese U, Da Silva D, Buc E, Souche R, Fabre JM, Colombo PE, Ferre L, Foguenne M, Hubert C, El Amrani M, Truant S, Schwartz L, Regenet N, Dupre A, Brustia R, Cherif R, Navez J, Darnis B, Facy O, Grellet R, Piessen G, Veziant J, Rhaiem R, Kianmanesh R, Fernandez-De-Sevilla E, Gelli M, Taibi A, Georges P, Mabrut JY, Lesurtel M, Doussot A, Bachellier P. ASO Author Reflections: Complete Pathological Response after Neoadjuvant Treatment for Pancreatic Ductal Adenocarcinomas-Curative Surgery in Cured Patients? Ann Surg Oncol 2025; 32:2861-2863. [PMID: 39893336 DOI: 10.1245/s10434-025-16925-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 01/04/2025] [Indexed: 02/04/2025]
Affiliation(s)
- Pietro Addeo
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation, Pôle des Pathologies Digestives et Hépatiques, Hôpital de Hautepierre-Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France.
| | - Milena Muzzolini
- Department of HBP Surgery, AP-HP, Hôpital Beaujon, University of Paris, Clichy, France
| | - Christophe Laurent
- Department of Digestive Surgery, Centre Magellan, CHU Bordeaux, Bordeaux, France
| | - Bruno Heyd
- Department of Digestive Surgical Oncology, University Hospital of Besançon, Besançon, France
| | - Alain Sauvanet
- Department of HBP Surgery, AP-HP, Hôpital Beaujon, University of Paris, Clichy, France
| | - Jonathan Garnier
- Department of Oncological Surgery, Institut Paoli Calmettes, Marseille University, Marseille, France
| | - Marie Sophie Alfano
- Department of Oncological Surgery, Institut Paoli Calmettes, Marseille University, Marseille, France
| | - Sebastien Gaujoux
- Department of Hepato-Biliary and Pancreatic Surgery and Liver Transplantation, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Charles De Ponthaud
- Department of Hepato-Biliary and Pancreatic Surgery and Liver Transplantation, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Ugo Marchese
- Department of Digestive, Hepatobiliary and Pancreatic Surgery, Cochin Hospital, AP-HP, Université de Paris, Paris, France
| | - Doris Da Silva
- Department of Digestive, Hepatobiliary and Pancreatic Surgery, Cochin Hospital, AP-HP, Université de Paris, Paris, France
| | - Emmanuel Buc
- Department of Digestive and Hepatobiliary Surgery, Liver Transplantation, University Hospital Clermont-Ferrand, Clermont-Ferrand, France
| | - Regis Souche
- Department of Surgery, Hopital Saint Eloi, Montpellier, France
| | | | | | - Lorenzo Ferre
- Department of Surgical Oncology, Institut du Cancer de Montpellier, Montpellier, France
| | - Maxime Foguenne
- Department of Abdominal Surgery and Transplantation, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Catherine Hubert
- Department of Abdominal Surgery and Transplantation, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Mehdi El Amrani
- Department of Digestive Surgery and Transplantation, Lille University Hospital, Lille, France
| | - Stephanie Truant
- Department of Digestive Surgery and Transplantation, Lille University Hospital, Lille, France
| | - Lilian Schwartz
- Department of Digestive Surgery, Rouen University Hospital and Université de Rouen Normandie, Rouen, France
| | - Nicolas Regenet
- Department of Digestive Surgery, Nantes Hospital, Nantes, France
| | - Aurelien Dupre
- Department of Surgical Oncology, Centre Léon Bérard, Lyon, France
| | - Raffaele Brustia
- Department of Digestive and Hepato-Pancreatic-Biliary Surgery, Hôpital Henri-Mondor, AP-HP, Paris Est Créteil University, UPEC, Créteil, France
| | - Rim Cherif
- Department of Digestive and Hepato-Pancreatic-Biliary Surgery, Hôpital Henri-Mondor, AP-HP, Paris Est Créteil University, UPEC, Créteil, France
| | - Julie Navez
- Department of Abdominal Surgery and Transplantation, Hôpitaux Universitaires de Bruxelles, Hopital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Benjamin Darnis
- Department of Digestive Surgery, Clinique de La Sauvegarde, Lyon, France
| | - Olivier Facy
- Department of Digestive and Surgical Oncology, University Hospital, Dijon, France
| | - Robin Grellet
- Department of Digestive and Surgical Oncology, University Hospital, Dijon, France
| | - Guillaume Piessen
- Department of Digestive and Oncological Surgery, Claude Huriez University Hospital, Lille, France
| | - Julie Veziant
- Department of Digestive and Oncological Surgery, Claude Huriez University Hospital, Lille, France
| | - Rami Rhaiem
- Department of HBP and Digestive Oncological Surgery, Robert Debré University Hospital, University Reims Champagne-Ardenne, Reims, France
| | - Reza Kianmanesh
- Department of HBP and Digestive Oncological Surgery, Robert Debré University Hospital, University Reims Champagne-Ardenne, Reims, France
| | | | | | - Abdelkader Taibi
- Department of Digestive Surgery, Dupuytren Limoges University Hospital, Limoges, France
| | - Pauline Georges
- Department of General Surgery and Liver Transplantation, Croix-Rousse University Hospital, Hospices Civils de Lyon, University of Lyon I, Lyon, France
| | - Jean Yves Mabrut
- Department of General Surgery and Liver Transplantation, Croix-Rousse University Hospital, Hospices Civils de Lyon, University of Lyon I, Lyon, France
| | - Mickael Lesurtel
- Department of HBP Surgery, AP-HP, Hôpital Beaujon, University of Paris, Clichy, France
| | - Alexandre Doussot
- Department of Digestive Surgical Oncology, University Hospital of Besançon, Besançon, France
| | - Philippe Bachellier
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation, Pôle des Pathologies Digestives et Hépatiques, Hôpital de Hautepierre-Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
38
|
Palmer DH, Jackson R, Springfeld C, Ghaneh P, Rawcliffe C, Halloran CM, Faluyi O, Cunningham D, Wadsley J, Darby S, Meyer T, Gillmore R, Lind P, Glimelius B, Falk S, Ma YT, Middleton GW, Cummins S, Ross PJ, Wasan H, McDonald A, Crosby T, Hammel P, Borg D, Sothi S, Valle JW, Mehrabi A, Bailey P, Tjaden C, Michalski C, Hackert T, Büchler MW, Neoptolemos JP. Pancreatic Adenocarcinoma: Long-Term Outcomes of Adjuvant Therapy in the ESPAC4 Phase III Trial. J Clin Oncol 2025; 43:1240-1253. [PMID: 39637340 PMCID: PMC11949205 DOI: 10.1200/jco.24.01118] [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: 05/24/2024] [Revised: 09/08/2024] [Accepted: 10/03/2024] [Indexed: 12/07/2024] Open
Abstract
PURPOSE The ESPAC4 trial showed that adjuvant chemotherapy with gemcitabine plus capecitabine (GemCap) produced longer overall survival (OS) than gemcitabine monotherapy. Subsequently, the PRODIGE24-CCTG PA.6 trial showed even longer survival for modified fluorouracil, folinic acid, irinotecan, and oxaliplatin (mFOLFIRINOX) than gemcitabine but had more restrictive eligibility criteria. Our aim was to analyze the ESPAC4 survival on long-term follow-up. METHODS The OS of 732 ESPAC4 patients comparing 367 randomly assigned to gemcitabine and 365 to GemCap was previously reported after a median follow-up time of 43.2 months (95% CI, 39.7 to 45.5) and 458 deaths. Analysis was now carried out after a median follow-up of 104 months (101-108) and 566 deaths. RESULTS The median OS was 29.5 months (27.5-32.1) for all patients, 28.4 months (25.2-32.0) in the gemcitabine group and 31.6 months (26.5-38.0) in the GemCap group (hazard ratio [HR], 0.83 [0.71 to 0.98]; P = .031). R0 patients given gemcitabine had a median survival of 32.2 months (27.9-41.6) compared with 49.9 months (39.0-82.3) for those given GemCap (HR, 0.63 [0.47 to 0.84]; P = .002). Lymph node-negative patients had significantly higher 5 year OS rates on GemCap (59% [49%-71%]) than gemcitabine (53% [42%-66%]; HR, 0.63 [0.41 to 0.98]; P = .04) but not those with positive lymph nodes (P = .225). The OS advantage for GemCap was retained in the PRODIGE24 subgroup of 193 (26.4%) ESPAC4 patients not eligible for PRODIGE24 with a median survival of 20.7 (16.2-27.3) months in patients allocated to gemcitabine compared with 25.9 (22.3-30.2) months for ineligible patients allocated to GemCap (HR, 0.71 [95% CI, 0.52 to 0.98]; χ2log-rank-1df = 4.31; P = .038). CONCLUSION GemCap is a standard option for patients not eligible for mFOLFIRINOX. Exploratory evidence suggests that GemCap may be particularly efficacious in R0 patients and also in lymph node-negative patients.
Collapse
Affiliation(s)
| | | | - Christoph Springfeld
- Department of Medical Oncology, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
- Champalimaud Foundation, Lisbon, Portugal
| | - Paula Ghaneh
- University of Liverpool, Liverpool, United Kingdom
| | | | | | - Olusola Faluyi
- The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, United Kingdom
| | | | | | | | - Tim Meyer
- Department of Oncology, Royal Free Hospital and UCL Cancer Institute, University College London, London, United Kingdom
| | | | - Pehr Lind
- Department of Oncology, Stockholm Söder Hospital, Stockholm, Sweden
- Karolinska Institute, Stockholm, Sweden
| | - Bengt Glimelius
- Department of Immunology, Genetics and Pathology, University of Uppsala, Uppsala, Sweden
| | - Stephen Falk
- Bristol Cancer Institute, Bristol, United Kingdom
| | - Yuk Ting Ma
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | | | | | - Paul J. Ross
- Guy's & St Thomas' and King's College Hospitals, London, United Kingdom
| | | | - Alec McDonald
- The Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Tom Crosby
- Velindre Cancer Centre, Cardiff, United Kingdom
| | - Pascal Hammel
- Hôpital Paul Brousse (APHP), Paris-Saclay University, Villejuif, France
| | - David Borg
- Skåne University Hospital, Lund, Sweden
- Department of Clinical Sciences Lund, Oncology and Therapeutic Pathology, Lund University, Lund, Sweden
| | - Sharmila Sothi
- University Hospitals Coventry and Warwickshire NHS Trust, Coventry, United Kingdom
| | | | - Arianeb Mehrabi
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Peter Bailey
- Champalimaud Foundation, Lisbon, Portugal
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christine Tjaden
- MRI TUM, Klinikum rechts der Isar of the Technical University of Munich, Munich, Germany
| | - Christoph Michalski
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Thilo Hackert
- Department of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Markus W. Büchler
- Champalimaud Foundation, Lisbon, Portugal
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - John P. Neoptolemos
- Champalimaud Foundation, Lisbon, Portugal
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
39
|
Yun WG, Chae YS, Han Y, Jung HS, Cho YJ, Kang HC, Kwon W, Park JS, Chie EK, Jang JY. Efficacy of Neoadjuvant Radiotherapy After Chemotherapy and the Optimal Interval from Radiotherapy to Surgery for Borderline Resectable and Resectable Pancreatic Cancer. Ann Surg Oncol 2025; 32:2819-2829. [PMID: 39808212 PMCID: PMC11882644 DOI: 10.1245/s10434-024-16743-2] [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: 07/30/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND Benefits of neoadjuvant treatment for pancreatic cancer with major vessel invasion has been demonstrated through randomized controlled trials; however, the optimal neoadjuvant treatment strategy remains controversial, especially for radiotherapy. Therefore, we aimed to evaluate the efficacy and safety of neoadjuvant radiotherapy followed by chemotherapy and the optimal time interval to undergo surgery after radiotherapy in (borderline) resectable pancreatic cancer. METHODS Between 2013 and 2022, patients with (borderline) resectable pancreatic cancer with vessel contact who received 5-fluorouracil with leucovorin, oxaliplatin, and irinotecan or gemcitabine and nanoparticle albumin-bound paclitaxel as initial treatment following surgery were included. Patients who received radiotherapy after chemotherapy and those who did not were matched using 1:1 nearest-neighbor propensity scores. Propensity scores were measured using the tumor size at initial image, duration of neoadjuvant chemotherapy, and responsiveness to neoadjuvant chemotherapy. RESULTS Of 212 patients, 166 patients were retrieved for the matched cohort. Patients who received radiotherapy had significantly better postoperative survival, local control, and R0 resection rates than those who did not. Furthermore, patients who underwent surgery within 4 weeks after completing radiotherapy had lower intraoperative blood loss and a clinically relevant postoperative pancreatic fistula rate than those who underwent surgery after more than 4 weeks. CONCLUSIONS In patients with (borderline) resectable pancreatic cancer with vessel contact who were scheduled for curative-intent surgery after neoadjuvant chemotherapy, additional radiotherapy was associated with better postoperative survival and local control. Furthermore, our findings suggested that scheduling surgery within 4 weeks following radiation therapy might enhance the perioperative outcomes.
Collapse
Affiliation(s)
- Won-Gun Yun
- Department of Surgery, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yoon Soo Chae
- Department of Surgery, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Youngmin Han
- Department of Surgery, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hye-Sol Jung
- Department of Surgery, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Young Jae Cho
- Department of Surgery, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyun-Cheol Kang
- Department of Radiation Oncology, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Institute of Radiation Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Wooil Kwon
- Department of Surgery, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Joon Seong Park
- Department of Surgery, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Eui Kyu Chie
- Department of Radiation Oncology, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Institute of Radiation Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea.
| | - Jin-Young Jang
- Department of Surgery, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
40
|
Conroy T, Ducreux M. ESMO Clinical Practice Guideline Express Update on the management of metastatic pancreatic cancer. ESMO Open 2025; 10:104528. [PMID: 40287191 DOI: 10.1016/j.esmoop.2025.104528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/04/2025] [Accepted: 02/25/2025] [Indexed: 04/29/2025] Open
Affiliation(s)
- T Conroy
- Department of Medical Oncology, Institut de Cancérologie de Lorraine, Vandoeuvre-lès-Nancy, France; Université de Lorraine, INSERM INSPIIRE, Nancy, France
| | - M Ducreux
- INSERM U1279, Université Paris-Saclay, Villejuif, France; Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| |
Collapse
|
41
|
Addeo P, Muzzolini M, Laurent C, Heyd B, Sauvanet A, Garnier J, Alfano MS, Gaujoux S, De Ponthaud C, Marchese U, Da Silva D, Buc E, Souche R, Fabre JM, Colombo PE, Ferre L, Foguenne M, Hubert C, El Amrani M, Truant S, Schwartz L, Regenet N, Dupre A, Brustia R, Cherif R, Navez J, Darnis B, Facy O, Grellet R, Piessen G, Veziant J, Rhaiem R, Kianmanesh R, Fernandez-De-Sevilla E, Gelli M, Taibi A, Georges P, Mabrut JY, Lesurtel M, Doussot A, Bachellier P. Prognosis Associated with Complete Pathological Response Following Neoadjuvant Treatment for PancreaTic AdenOcarciNOma in the FOFLIRINOX Era: the Multicenter TONO Study. Ann Surg Oncol 2025; 32:2809-2818. [PMID: 39777595 DOI: 10.1245/s10434-024-16735-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND The use of multiagent FOLFIRINOX chemotherapy for pancreatic adenocarcinoma in a neoadjuvant setting has been associated with an increased rate of complete pathological response (CPR) after surgery. This study investigated the long-term outcomes of patients with CPR in a multicenter setting to identify prognostic factors for overall survival (OS) and recurrence-free survival (RFS). METHODS This retrospective cohort study examined biopsy-proven pancreatic adenocarcinomas with CPR after neoadjuvant chemotherapy or chemoradiotherapy and surgery, between January 2006 and December 2023 across 22 French and 2 Belgian centers. Cox analyses were used to identify prognostic factors of OS and RFS. RESULTS There were 101 patients with CPR after chemotherapy (n = 58, 57.4%) and chemoradiotherapy (n = 43, 42.6%) followed by surgery. Neoadjuvant FOLFIRINOX was used in 90% of patients. The median OS after surgery was 177 months (95% confidence interval (CI) 58.9-177 months) with 1-, 3-, 5-, and 10-year OS rates of 93%, 75%, 63%, and 51%, respectively. The median RFS was 67.8 months (95% CI:34.4-NR) with 1-, 3-, 5-, and 10-year RFS rates of 83%, 58%, 54%, and 49%, respectively. The multivariate Cox analysis of OS and RFS showed that preoperative radiotherapy was an independent negative prognostic factor for OS (hazard ratio (HR) 2.51; 95% CI 1.00-6.30; p = 0.03) and RFS (HR 2.62; 95% CI 1.27-5.41; p = 0.009). CONCLUSIONS Complete pathologic response after neoadjuvant treatment is associated with remarkable long-term survival that is usually not seen after the resection of pancreatic adenocarcinomas. One-third of the patients still experienced disease recurrence, which was more common in those receiving preoperative chemoradiotherapy.
Collapse
Affiliation(s)
- Pietro Addeo
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation, Pôle des Pathologies Digestives et Hépatiques, Hôpital de Hautepierre-Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France.
| | - Milena Muzzolini
- Department of HBP Surgery, AP-HP, Hôpital Beaujon, University of Paris, Clichy, France
| | - Christophe Laurent
- Department of Digestive Surgery, Centre Magellan-CHU Bordeaux, Bordeaux, France
| | - Bruno Heyd
- Department of Digestive Surgical Oncology, University Hospital of Besançon, Besançon, France
| | - Alain Sauvanet
- Department of HBP Surgery, AP-HP, Hôpital Beaujon, University of Paris, Clichy, France
| | - Jonathan Garnier
- Department of Oncological Surgery, Institut Paoli Calmettes, Marseille University, Marseille, France
| | - Marie Sophie Alfano
- Department of Oncological Surgery, Institut Paoli Calmettes, Marseille University, Marseille, France
| | - Sebastien Gaujoux
- Department of Hepato-Biliary and Pancreatic Surgery and Liver Transplantation, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Charles De Ponthaud
- Department of Hepato-Biliary and Pancreatic Surgery and Liver Transplantation, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Ugo Marchese
- Department of Digestive, Hepatobiliary and Pancreatic Surgery, Cochin Hospital, AP-HP, Université de Paris, Paris, France
| | - Doris Da Silva
- Department of Digestive, Hepatobiliary and Pancreatic Surgery, Cochin Hospital, AP-HP, Université de Paris, Paris, France
| | - Emmanuel Buc
- Department of Digestive and Hepatobiliary Surgery-Liver Transplantation, University Hospital Clermont-Ferrand, Clermont-Ferrand, France
| | - Regis Souche
- Department of Surgery, Hopital Saint Eloi, Montpellier, France
| | | | - Pierre-Emanuel Colombo
- Department of Surgical Oncology, Institut du Cancer de Montpellier (ICM), Montpellier, France
| | - Lorenzo Ferre
- Department of Surgical Oncology, Institut du Cancer de Montpellier (ICM), Montpellier, France
| | - Maxime Foguenne
- Department of Abdominal Surgery and Transplantation, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Catherine Hubert
- Department of Abdominal Surgery and Transplantation, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Mehdi El Amrani
- Department of Digestive Surgery and Transplantation, Lille University Hospital, Lille, France
| | - Stephanie Truant
- Department of Digestive Surgery and Transplantation, Lille University Hospital, Lille, France
| | - Lilian Schwartz
- Department of Digestive Surgery, Rouen University Hospital and Université de Rouen Normandie, Rouen, France
| | - Nicolas Regenet
- Department of Digestive Surgery, Nantes Hospital, Nantes, France
| | - Aurelien Dupre
- Department of Surgical Oncology, Centre Léon Bérard, Lyon, France
| | - Raffaele Brustia
- Department of Digestive and Hepato-Pancreatic-Biliary Surgery, Hôpital Henri-Mondor, AP-HP, Paris Est Créteil University, UPEC, Créteil, France
| | - Rim Cherif
- Department of Digestive and Hepato-Pancreatic-Biliary Surgery, Hôpital Henri-Mondor, AP-HP, Paris Est Créteil University, UPEC, Créteil, France
| | - Julie Navez
- Department of Abdominal Surgery and Transplantation, Hôpitaux Universitaires de Bruxelles (HUB), Hopital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Benjamin Darnis
- Department of Digestive Surgery, Clinique de La Sauvegarde, Lyon, France
| | - Olivier Facy
- Department of Digestive and Surgical Oncology, University Hospital, Dijon, France
| | - Robin Grellet
- Department of Digestive and Surgical Oncology, University Hospital, Dijon, France
| | - Guillaume Piessen
- Department of Digestive and Oncological Surgery, Claude Huriez University Hospital, Lille, France
| | - Julie Veziant
- Department of Digestive and Oncological Surgery, Claude Huriez University Hospital, Lille, France
| | - Rami Rhaiem
- Department of HBP and Digestive Oncological Surgery, Robert Debré University Hospital, University Reims Champagne-Ardenne, Reims, France
| | - Reza Kianmanesh
- Department of HBP and Digestive Oncological Surgery, Robert Debré University Hospital, University Reims Champagne-Ardenne, Reims, France
| | | | | | - Abdelkader Taibi
- Department of Digestive Surgery, Dupuytren Limoges University Hospital, Limoges, France
| | - Pauline Georges
- Department of General Surgery and Liver Transplantation, Croix-Rousse University Hospital, Hospices Civils de Lyon, University of Lyon I, Lyon, France
| | - Jean Yves Mabrut
- Department of General Surgery and Liver Transplantation, Croix-Rousse University Hospital, Hospices Civils de Lyon, University of Lyon I, Lyon, France
| | - Mickael Lesurtel
- Department of HBP Surgery, AP-HP, Hôpital Beaujon, University of Paris, Clichy, France
| | - Alexandre Doussot
- Department of Digestive Surgical Oncology, University Hospital of Besançon, Besançon, France
| | - Philippe Bachellier
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation, Pôle des Pathologies Digestives et Hépatiques, Hôpital de Hautepierre-Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
42
|
So E, Hayashi H, Shimozaki K, Horie S, Kishimoto S, Chida A, Saito Y, Tsugaru K, Hirata K, Tanishima S, Nishihara H, Kanai T, Hamamoto Y. Clinical utility of comprehensive genomic profiling for advanced pancreatic cancer: insights from real-world data analysis. Int J Clin Oncol 2025; 30:728-737. [PMID: 39961905 DOI: 10.1007/s10147-025-02713-5] [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: 09/08/2024] [Accepted: 01/23/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND Precision medicine is a promising therapeutic strategy for pancreatic cancer. However, only a few patients are eligible for genotype-matched treatments because of the low detection rate of actionable genomic alterations, and the clinical application of comprehensive genomic profiling (CGP) in pancreatic cancer has not been completely investigated. CGP provides considerable information, including the prognosis and eligibility of patients for genotype-matched treatments, which can guide physicians' treatment strategies. This study aimed to investigate the contribution of CGP to patient outcomes. METHODS This single-center retrospective cohort study enrolled patients with recurrent or metastatic pancreatic cancer with adenocarcinoma or adenosquamous carcinoma who underwent systemic chemotherapy between April 2018 and April 2022. We reviewed the medical records for patient characteristics, survival, and genomic information. We compared overall survival (OS) between patients who received CGP (CGP group) and those who did not (non-CGP group). RESULTS Of 111 eligible patients, 59 underwent CGP. Median OS was significantly longer in the CGP than the non-CGP group (25.2 vs. 11.8 months; hazard ratio, 0.49; P = 0.0013). Six patients (10.2%) underwent genotype-matched treatments, with a median OS of 35.5 months, compared to 17.0 months for those who did not. The CGP group demonstrated a higher transition rate to subsequent chemotherapy than did the non-CGP group (76.3% vs. 48.1%, P = 0.0030). CONCLUSIONS OS was prolonged in patients with pancreatic cancer who underwent CGP, probably due to its influence on physicians' treatment strategies. These findings highlight the importance of the proactive and timely implementation of CGP in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Eiichiro So
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hideyuki Hayashi
- Keio Cancer Center, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Keitaro Shimozaki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Sara Horie
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Shotaro Kishimoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Akihiko Chida
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yuki Saito
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kai Tsugaru
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kenro Hirata
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Shigeki Tanishima
- Department of Biomedical Informatics Development, Mitsubishi Electric Software Co., Ltd, Tokyo, Japan
| | - Hiroshi Nishihara
- Keio Cancer Center, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yasuo Hamamoto
- Keio Cancer Center, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| |
Collapse
|
43
|
Peixoto D, Ravasco JM, Blanco-Fernandez B, Veiga F, Concheiro A, Conde J, Paiva-Santos AC, Alvarez-Lorenzo C. Enzyme-responsive vitamin D-based micelles for paclitaxel-controlled delivery and synergistic pancreatic cancer therapy. Mater Today Bio 2025; 31:101555. [PMID: 40026626 PMCID: PMC11869029 DOI: 10.1016/j.mtbio.2025.101555] [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: 09/16/2024] [Revised: 12/20/2024] [Accepted: 02/03/2025] [Indexed: 03/05/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most feared diseases worldwide owing to its poor prognosis, negligible therapeutic advances, and high mortality. Herein, multifunctional enzyme-responsive micelles for the controlled delivery of paclitaxel (PTX) were prepared to circumvent its current clinical challenges. Accordingly, two enzyme-responsive structural units composed of Vitamin D3 (VD3) conjugated with polyethylene glycol of different molecular weights (600 Da and 2000 Da) were synthesized and characterized using different analytical methods. By applying the solvent evaporation method, these bioactive structural units self-assembled into sub-100 nm VD3 micelles with minimal batch-to-batch variation, monomodal particle size distribution, and high encapsulation efficiency. The enzyme-triggered disassembly of PTX-loaded VD3 micelles was demonstrated by release studies in the presence of a high esterase content typically featured by PDAC cells. PTX-loaded VD3 micelles also exhibited prominent cell internalization and induced a considerable cytotoxic synergistic effect against human PDAC cells (BxPC-3 cells) in 2D and 3D cell culture models compared with free PTX. The PTX-loaded VD3 micelles were hemocompatible and stable after long-term storage in the presence of biorelevant media, and showed higher efficiency to inhibit the tumor growth compared to the approved clinical nanoformulation (Abraxane®) in an in ovo tumor model. The findings reported here indicate that VD3S-PEG micelles may have a promising role in PDAC therapy, since VD3 could act not only as a hydrophobic core of the micelles but also as a therapeutic agent that provides synergetic therapeutic effects with the encapsulated PTX.
Collapse
Affiliation(s)
- Diana Peixoto
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Faculty of Pharmacy, iMATUS and Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, 15782, Santiago, Spain
| | - João M. Ravasco
- Comprehensive Health Research Centre, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade Nova de Lisboa, 1169, Lisboa, Portugal
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, 1649, Lisbon, Portugal
| | - Barbara Blanco-Fernandez
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Faculty of Pharmacy, iMATUS and Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, 15782, Santiago, Spain
| | - Francisco Veiga
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Angel Concheiro
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Faculty of Pharmacy, iMATUS and Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, 15782, Santiago, Spain
| | - João Conde
- Comprehensive Health Research Centre, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade Nova de Lisboa, 1169, Lisboa, Portugal
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, 1649, Lisbon, Portugal
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Faculty of Pharmacy, iMATUS and Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, 15782, Santiago, Spain
| |
Collapse
|
44
|
Buchholz M, Lausser L, Schenk M, Earl J, Lawlor RT, Scarpa A, Sanjuanbenito A, Carrato A, Malats N, Tjaden C, Giese NA, Büchler M, Hackert T, Kestler HA, Gress TM. Combined analysis of a serum mRNA/miRNA marker signature and CA 19-9 for timely and accurate diagnosis of recurrence after resection of pancreatic ductal adenocarcinoma: A prospective multicenter cohort study. United European Gastroenterol J 2025; 13:353-363. [PMID: 39453683 PMCID: PMC11999032 DOI: 10.1002/ueg2.12676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/30/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND AND AIMS Timely and accurate detection of tumor recurrence in pancreatic ductal adenocarcinoma (PDAC) patients is an urgent and unmet medical need. This study aimed to develop a noninvasive molecular diagnostic procedure for the detection of recurrence after PDAC resection based on quantification of circulating mRNA and miRNA biomarkers in serum samples. METHODS In a multicentric study, serum samples from a total of 146 patients were prospectively collected after resection. Samples were classified into a "No Evidence of Disease" and a "Recurrence" group based on clinical follow-up data. A multianalyte biomarker panel was composed of mRNAs and miRNA markers and simultaneously analyzed in serum samples using custom microfluidic qPCR arrays (TaqMan array cards). A diagnostic algorithm was developed combining a 7-gene marker signature with CA19-9 data. RESULTS The best-performing marker combination achieved 90% diagnostic accuracy in predicting the presence of tumor recurrence (98% sensitivity; 84% specificity), clearly outperforming the singular CA 19-9 analysis. Moreover, time series data obtained by analyzing successively collected samples from 5 patients during extended follow-up suggested that molecular diagnosis has the potential to detect recurrence earlier than routine clinical procedures. CONCLUSIONS TaqMan array card measurements were found to be biologically valid and technically reproducible. The BioPac multianalyte marker panel is capable of sensitive and accurate detection of recurrence in patients resected for PDAC using a simple blood test. This could allow a closer follow-up using shorter time intervals than currently used for imaging, thus potentially prompting an earlier work-up with additional modalities to allow for earlier therapeutic intervention. This study provides a promising approach for improved postoperative monitoring of resected PDAC patients, which is an urgent and unmet clinical need.
Collapse
MESH Headings
- Humans
- Carcinoma, Pancreatic Ductal/surgery
- Carcinoma, Pancreatic Ductal/blood
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/diagnosis
- Carcinoma, Pancreatic Ductal/pathology
- CA-19-9 Antigen/blood
- Neoplasm Recurrence, Local/diagnosis
- Neoplasm Recurrence, Local/blood
- Neoplasm Recurrence, Local/genetics
- Male
- Pancreatic Neoplasms/surgery
- Pancreatic Neoplasms/blood
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/diagnosis
- Female
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/genetics
- Middle Aged
- Aged
- Prospective Studies
- RNA, Messenger/blood
- MicroRNAs/blood
- Sensitivity and Specificity
- Pancreatectomy
- Aged, 80 and over
Collapse
Affiliation(s)
- Malte Buchholz
- Department of Gastroenterology, Endocrinology, Metabolism and InfectiologyPhilipps‐University and University Hospital MarburgMarburgGermany
| | - Ludwig Lausser
- Institute of Medical Systems BiologyUlm UniversityUlmGermany
- Fakultät InformatikBiomedizinische InformatikTechnische Hochschule IngolstadtIngolstadtGermany
| | - Miriam Schenk
- Chirurgische Klinik / Europäisches PankreaszentrumUniversitätsklinikum HeidelbergHeidelbergGermany
| | - Julie Earl
- Molecular Epidemiology and Predictive Tumor Markers GroupInstituto Ramón y Cajal de Investigación Sanitaria (IRYCIS)CIBERONCMadridSpain
| | - Rita T. Lawlor
- Centre for Applied Research on CancerUniversity of Verona ‐ Policlinico G.B. RossiVeronaItaly
| | - Aldo Scarpa
- Centre for Applied Research on CancerUniversity of Verona ‐ Policlinico G.B. RossiVeronaItaly
| | - Alfonso Sanjuanbenito
- Pancreatic and Biliopancreatic Surgery UnitRamón y Cajal University HospitalCIBERONCMadridSpain
| | - Alfredo Carrato
- Molecular Epidemiology and Predictive Tumor Markers GroupInstituto Ramón y Cajal de Investigación Sanitaria (IRYCIS)CIBERONCMadridSpain
| | - Nuria Malats
- Spanish National Cancer Research Centre (CNIO)Genetic and Molecular EpidemiologyMadridSpain
| | - Christine Tjaden
- Chirurgische Klinik / Europäisches PankreaszentrumUniversitätsklinikum HeidelbergHeidelbergGermany
| | - Nathalia A. Giese
- Chirurgische Klinik / Europäisches PankreaszentrumUniversitätsklinikum HeidelbergHeidelbergGermany
| | - Markus Büchler
- Chirurgische Klinik / Europäisches PankreaszentrumUniversitätsklinikum HeidelbergHeidelbergGermany
| | - Thilo Hackert
- Chirurgische Klinik / Europäisches PankreaszentrumUniversitätsklinikum HeidelbergHeidelbergGermany
| | - Hans A. Kestler
- Institute of Medical Systems BiologyUlm UniversityUlmGermany
| | - Thomas M. Gress
- Department of Gastroenterology, Endocrinology, Metabolism and InfectiologyPhilipps‐University and University Hospital MarburgMarburgGermany
| | | |
Collapse
|
45
|
Tabacelia D, Robles-Medranda C, Klimko A, Pereira SP, Vilmann P, Voermans RP, Săftoiu A, Tieranu CG, Stroescu C. Technical Feasibility of Microwave Ablation in Pancreatic Tumors: A Scoping Review of Procedural Efficacy and Safety. Cancers (Basel) 2025; 17:1197. [PMID: 40227787 PMCID: PMC11988054 DOI: 10.3390/cancers17071197] [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: 01/25/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Pancreatic cancer remains one of the most aggressive and lethal malignancies, with limited effective treatment options for advanced stages. Microwave Ablation (MWA) has emerged as a minimally invasive therapeutic modality, offering potential benefits in tumor control. This review aims to critically assess the safety and efficacy of MWA in the treatment of pancreatic cancer, focusing on its application in various pancreatic lesions. METHODS We systematically reviewed studies published between 2010 and 2023 that evaluated the use of MWA in pancreatic tumors, including locally advanced pancreatic cancer (LAPC), pancreatic neuroendocrine tumors (PNETs), and pancreatic metastases from renal cell carcinoma (RCC). Due to limited data on survival rates and long-term outcomes, our analysis concentrated primarily on the technical aspects and immediate procedural outcomes of MWA. RESULTS MWA was technically feasible in all cases. The overall complication rate was approximately 16.7% (nine patients), with higher incidences in tumors located in the pancreatic head. Reported complications included pancreatitis and pseudocyst formation. Procedural parameters varied, with applied energy ranging from 20 to 80 watts and ablation times between 2 to 15 min, depending on the microwave generator type and approach (percutaneous, intraoperative or endoscopic). All cases demonstrated effective necrosis of the target tissue, and several studies reported notable tumor size reductions, averaging up to 70%. CONCLUSIONS MWA shows promise as a therapeutic option for pancreatic cancer, achieving high technical success rates and significant tumor reductions. However, the procedure is associated with a moderate complication rate, particularly in tumors located in the pancreatic head.
Collapse
Affiliation(s)
- Daniela Tabacelia
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 010017 Bucharest, Romania; (D.T.); (A.S.); (C.G.T.)
- Elias Emergency University Hospital, 010017, Bucharest, Romania
| | | | | | - Stephen P. Pereira
- Institute for Liver and Digestive Health, University College London, London NW3 2QG, UK;
| | - Peter Vilmann
- Gastro Unit, Division of Endoscopy, Herlev and Gentofte Hospital, 2730 Herlev, Denmark;
- Department of Clinical Medicine, University of Copenhagen, 1050 Copenhagen, Denmark
| | - Rogier P. Voermans
- Department of Gastroenterology & Hepatology, Amsterdam Gastroenterology and Metabolism, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Adrian Săftoiu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 010017 Bucharest, Romania; (D.T.); (A.S.); (C.G.T.)
- Elias Emergency University Hospital, 010017, Bucharest, Romania
| | - Cristian George Tieranu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 010017 Bucharest, Romania; (D.T.); (A.S.); (C.G.T.)
- Elias Emergency University Hospital, 010017, Bucharest, Romania
| | - Cezar Stroescu
- Surgery Department, Fundeni Clinical Institute, 010017 Bucharest, Romania;
| |
Collapse
|
46
|
Ramesh RPG, Yasmin H, Ponnachan P, Al-Ramadi B, Kishore U, Joseph AM. Phenotypic heterogeneity and tumor immune microenvironment directed therapeutic strategies in pancreatic ductal adenocarcinoma. Front Immunol 2025; 16:1573522. [PMID: 40230862 PMCID: PMC11994623 DOI: 10.3389/fimmu.2025.1573522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 03/04/2025] [Indexed: 04/16/2025] Open
Abstract
Pancreatic cancer is an aggressive tumor with high metastatic potential which leads to decreased survival rate and resistance to chemotherapy and immunotherapy. Nearly 90% of pancreatic cancer comprises pancreatic ductal adenocarcinoma (PDAC). About 80% of diagnoses takes place at the advanced metastatic stage when it is unresectable, which renders chemotherapy regimens ineffective. There is also a dearth of specific biomarkers for early-stage detection. Advances in next generation sequencing and single cell profiling have identified molecular alterations and signatures that play a role in PDAC progression and subtype plasticity. Most chemotherapy regimens have shown only modest survival benefits, and therefore, translational approaches for immunotherapies and combination therapies are urgently required. In this review, we have examined the immunosuppressive and dense stromal network of tumor immune microenvironment with various metabolic and transcriptional changes that underlie the pro-tumorigenic properties in PDAC in terms of phenotypic heterogeneity, plasticity and subtype co-existence. Moreover, the stromal heterogeneity as well as genetic and epigenetic changes that impact PDAC development is discussed. We also review the PDAC interaction with sequestered cellular and humoral components present in the tumor immune microenvironment that modify the outcome of chemotherapy and radiation therapy. Finally, we discuss different therapeutic interventions targeting the tumor immune microenvironment aimed at better prognosis and improved survival in PDAC.
Collapse
Affiliation(s)
- Remya P. G. Ramesh
- Department of Veterinary Medicine, UAE University, Al Ain, United Arab Emirates
| | - Hadida Yasmin
- Immunology and Cell Biology Laboratory, Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal, India
| | - Pretty Ponnachan
- Department of Veterinary Medicine, UAE University, Al Ain, United Arab Emirates
| | - Basel Al-Ramadi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Uday Kishore
- Department of Veterinary Medicine, UAE University, Al Ain, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ann Mary Joseph
- Department of Veterinary Medicine, UAE University, Al Ain, United Arab Emirates
| |
Collapse
|
47
|
Bye BA, Jack JL, Pierce A, Walsh RM, Eades AE, Chalise P, Olou A, VanSaun MN. Combined Omipalisib and MAPK Inhibition Suppress PDAC Growth. Cancers (Basel) 2025; 17:1152. [PMID: 40227649 PMCID: PMC11987824 DOI: 10.3390/cancers17071152] [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: 01/25/2025] [Revised: 03/14/2025] [Accepted: 03/26/2025] [Indexed: 04/15/2025] Open
Abstract
Background: Oncogenic KRAS mutations are nearly ubiquitous in pancreatic ductal adenocarcinoma (PDAC), yet therapeutic attempts to target KRAS, as well as downstream MAPK pathway effectors, have shown limited clinical success. While KRAS canonically drives MAPK signaling via RAF-MEK-ERK, it is also known to play a role in PI3K-AKT signaling. Methods: Our therapeutic study targeted the PI3K pathway with the drug Omipalisib (p110α/β/δ/γ and mTORC1/2 inhibitor) in combination with two different MAPK pathway inhibitors: Trametinib (MEK1/2 inhibitor) or SHP099-HCL (SHP099; SHP2 inhibitor). Western blot analysis demonstrated that the application of Trametinib or SHP099 alone selectively blocked ERK phosphorylation (pERK) but failed to suppress phosphorylated AKT (pAKT). Conversely, Omipalisib alone successfully inhibited pAKT but failed to suppress pERK. Therefore, we hypothesized that a combination therapeutic comprised of Omipalisib with either Trametinib or SHP099 would inhibit two prominent mitogenic pathways, MAPK and PI3K-AKT, and effectively suppress PDAC growth. Results: In vitro studies demonstrated that, in several cell lines, both Omipalisib/Trametinib and Omipalisib/SHP099 combination therapeutic strategies were more effective than treatment with each drug individually at reducing proliferation, colony formation, and cell migration compared to vehicle controls. In vivo oral administration of combined Omipalisib/Trametinib treatment was significantly more effective than Omipalisib/SHP099 in reducing implanted tumor growth, and the Omipalisib/Trametinib treatment more effectively reduced tumor progression and prolonged survival in an aggressive genetically engineered mouse model of PDAC than either Omipalisib or Trametinib alone. Conclusions: Altogether, our data support a rationale for a dual treatment strategy targeting both PI3K and MAPK pathways in pancreatic cancers.
Collapse
Affiliation(s)
- Bailey A. Bye
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA
| | - Jarrid L. Jack
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA
| | - Alexandra Pierce
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA
| | - Richard McKinnon Walsh
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA
| | - Austin E. Eades
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA
| | - Prabhakar Chalise
- Department of Biostatistics and Data Science, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA
| | - Appolinaire Olou
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA
| | - Michael N. VanSaun
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA
| |
Collapse
|
48
|
Vigié T, Perrier A, Chanez B, De Martino J, Favre L, Coulet F, Bachet JB, Guillerm E, Mas L. Prolonged response to osimertinib in three patients with refractory metastatic pancreatic adenocarcinomas with EGFR exon 19 deletion: a case report and literature review. Ther Adv Med Oncol 2025; 17:17588359241312078. [PMID: 40162000 PMCID: PMC11954516 DOI: 10.1177/17588359241312078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 12/19/2024] [Indexed: 04/02/2025] Open
Abstract
Pancreatic cancer is a rising cause of cancer death. Therapeutic options are scarce and of limited efficacy. Up to 26% of patients with metastatic pancreatic cancer could benefit from targeted therapies. We report here for the first time the case of three patients with metastatic pancreatic ductal adenocarcinoma (PDAC) without KRAS alteration for whom an activating mutation in exon 19 of the epidermal growth factor receptor (EGFR) gene was found through mainstreaming NGS. The EGFR variant was confirmed on multiple tumor samples and by circulating tumor DNA (ctDNA) analysis in two patients. The three patients were treated with osimertinib with early molecular, biologic, and morpho-metabolic responses. At the last follow-up, one patient had an ongoing response after 17 months, and disease control had been maintained for 8 and 6 months in the other two. Known resistance mechanisms were observed on ctDNA analysis at progression. These observations demonstrate the benefit of osimertinib for treating EGFR-mutated PDAC and highlight the interest in investigating rare molecular alterations, especially in patients without KRAS alterations.
Collapse
Affiliation(s)
- Timothée Vigié
- Hepato-Gastroenterology and Digestive Oncology Department, Sorbonne University, Pitié Salpêtrière Hospital, AP-HP, Paris, France
| | - Alexandre Perrier
- Genetics Medical Department, Sorbonne University, Pitié Salpêtrière Hospital, AP-HP, Paris, France
| | - Brice Chanez
- Medical Oncology Department, Institut Paoli-Calmettes, Marseille, France
| | - Julien De Martino
- Department of Hepato-Biliary and Pancreatic Surgery and Liver Transplantation, Sorbonne University, AP-HP, Pitié Salpêtrière Hospital, Paris, France
| | - Loëtitia Favre
- Genetics Medical Department, Sorbonne University, Pitié Salpêtrière Hospital, AP-HP, Paris, France
| | - Florence Coulet
- Genetics Medical Department, Sorbonne University, Pitié Salpêtrière Hospital, AP-HP, Paris, France
| | - Jean-Baptiste Bachet
- Hepato-Gastroenterology and Digestive Oncology Department, Sorbonne University, Pitié Salpêtrière Hospital, AP-HP, Paris, France
| | - Erell Guillerm
- Genetics Medical Department, Sorbonne University, Pitié Salpêtrière Hospital, AP-HP, Paris, France
| | - Léo Mas
- Hepato-Gastroenterology and Digestive Oncology Department, Sorbonne University, Pitié Salpêtrière Hospital, AP-HP, 47-83 Boulevard de l’Hôpital, Paris 75013, France
| |
Collapse
|
49
|
Henderson NL, Bourne G, Ortiz-Olguin E, Pywell C, Rose JB, Williams GR, Hussaini SMQ, Nipp RD, Rocque G. The impact of electronic patient-reported outcomes presentation during multi-disciplinary tumor board on clinician discussion of older adults' fitness and preferences. J Geriatr Oncol 2025; 16:102225. [PMID: 40120473 DOI: 10.1016/j.jgo.2025.102225] [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: 11/22/2024] [Revised: 02/28/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
INTRODUCTION Treatment of pancreatic cancer often entails multiple modalities (e.g., chemotherapy, surgery, radiation) that vary in intensity, timing, and toxicity profiles. Some treatment options are only recommended for medically 'fit' patients regardless of age, yet formal fitness measures (such as the geriatric assessment [GA]) and patient preferences are seldom utilized during treatment decision-making. MATERIALS AND METHODS The INtegrating Systematic PatIent-Reported Evaluations into Multi-Disciplinary Tumor Board (INSPIRE-MDTB) intervention involves the presentation of GA and treatment preferences data during tumor board discussions of older patients with stage I-IV pancreatic adenocarcinoma. This qualitative study recorded, transcribed, and inductively analyzed historical (November 2021-February 2022) and intervention (September 2022-June 2023) MDTBs using NVivo software. A constant comparative method was used to establish a grounded scheme representative of clinicians' characterization of patients' fitness and preferences during decision-making. RESULTS Recordings of the primary MDTB presentation of 31 historical and 49 intervention patients with similar sex (52 %; 53 % female), age (m = 68.1; 72.3), race (65 %; 59 % White), and cancer stage (26 %; 22 % stage IV) were included. Although GA was captured for all included patients, it was not discussed in any historical cases, but was in 94 % of intervention cases. When compared to historical controls, INSPIRE patients had more frequent discussions of (1) cancer-related factors (e.g., size, location, rate of progression; 35 % vs. 43 %), (2) individual risk factors (e.g., age, comorbidities, tolerance; 90 % vs 98 %), and (3) psychosocial factors (e.g., health literacy, social support, substance use; 19 % vs 33 %). Identified preference domains were discussed in 39 % of historical and 80 % of intervention patients, with notably higher rates of discussion of patients' concerns regarding physical (0 %; 35 %) and mental/emotional (0 %; 20 %) side effects, ability to work (0 %; 10 %), and the logistics and convenience of treatment (6 %; 14 %). DISCUSSION The INSPIRE intervention enhanced MDTB discussion of patient fitness and preferences and represents a promising approach for fostering consistent and systematic presentation and discussion of patient-reported data, such as the GA and treatment preferences. This adds to our previous findings that INSPIRE was feasible, acceptable, appropriate, and time-effective according to patients and provider participants.
Collapse
Affiliation(s)
- Nicole L Henderson
- O'Neal Comprehensive Cancer Center at UAB, Birmingham, AL, United States of America.
| | - Garrett Bourne
- O'Neal Comprehensive Cancer Center at UAB, Birmingham, AL, United States of America
| | - Etzael Ortiz-Olguin
- O'Neal Comprehensive Cancer Center at UAB, Birmingham, AL, United States of America
| | - Cameron Pywell
- O'Neal Comprehensive Cancer Center at UAB, Birmingham, AL, United States of America
| | - J Bart Rose
- O'Neal Comprehensive Cancer Center at UAB, Birmingham, AL, United States of America
| | - Grant R Williams
- O'Neal Comprehensive Cancer Center at UAB, Birmingham, AL, United States of America
| | - S M Qasim Hussaini
- O'Neal Comprehensive Cancer Center at UAB, Birmingham, AL, United States of America
| | - Ryan D Nipp
- OU Health Stephenson Cancer Center, Oklahoma City, OK, United States of America
| | - Gabrielle Rocque
- O'Neal Comprehensive Cancer Center at UAB, Birmingham, AL, United States of America.
| |
Collapse
|
50
|
Yu HY, Lee CY, Hu YC, Lin LG, Chao Y, Li CP. Treatment rechallenge is safe and leads to better survival in pancreatic cancer patients with interstitial pneumonitis. BMC Cancer 2025; 25:508. [PMID: 40108538 PMCID: PMC11924909 DOI: 10.1186/s12885-025-13896-5] [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: 07/01/2024] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND AND AIMS Interstitial pneumonitis is a potentially fatal complication of cancer-related therapy. However, data regarding the risk factors, prognosis and safety and benefit of rechallenge treatment are scarce. METHODS Patients diagnosed with pancreatic cancer were retrospectively enrolled, and those with pneumonitis were identified. We investigated the incidence and etiology of pneumonitis, potential risk factors, and impact of treatment rechallenge on clinical outcomes. RESULTS A total of 809 patients were diagnosed with pancreatic cancer, among whom 62 (7.7%) were diagnosed with interstitial pneumonitis. Risk factors identified through competing risk analysis included nab-paclitaxel, gemcitabine, erlotinib, and previous lung diseases such as pre-existing ILD, asthma, chronic obstructive pulmonary disease, tuberculosis, primary lung cancer, metastasis, or pneumonia. Among these patients, 33 experienced acute respiratory distress syndrome, resulting in 15 deaths during pneumonitis episodes. After rechallenge therapy in 33 patients, pneumonitis recurred in 3 (9%). The median overall survival was longer in patients with pneumonitis than in those without. Subgroup analysis further revealed that overall survival was significantly better in the rechallenge group. CONCLUSIONS Most cases of pneumonitis are not directly induced by cancer therapy. Therefore, treatment rechallenge is considered a reasonable approach, potentially resulting in improved survival outcomes.
Collapse
Affiliation(s)
- Hung-Yuan Yu
- Therapeutic and Research Center of Pancreatic Cancer, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, 11217, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Yang Lee
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, 11217, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yen-Chi Hu
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, 11217, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Gastroenterology and Hepatology, Department of Medicine, Pojen General Hospital, Taipei, Taiwan
| | - Le-Gin Lin
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Nursing, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yee Chao
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chung-Pin Li
- Therapeutic and Research Center of Pancreatic Cancer, Taipei Veterans General Hospital, Taipei, Taiwan.
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, 11217, Taiwan.
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Division of Clinical Skills Training, Department of Medical Education, Taipei Veterans General Hospital, Taipei, Taiwan.
| |
Collapse
|