1
|
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
|
2
|
Hammouz RY, Baryła I, Styczeń-Binkowska E, Bednarek AK. Twenty-five years of WWOX insight in cancer: a treasure trove of knowledge. Funct Integr Genomics 2025; 25:100. [PMID: 40327201 PMCID: PMC12055895 DOI: 10.1007/s10142-025-01601-5] [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: 11/12/2024] [Revised: 04/01/2025] [Accepted: 04/12/2025] [Indexed: 05/07/2025]
Abstract
More than two decades ago, MD Anderson Cancer group discovered, characterised, and identified the WW domain-containing oxidoreductase (WWOX) as a genes of interest mapping to the chromosomal region 16q23.3-24.2. This was pioneering research since WWOX is a critical tumour suppressor gene implicated in various cancers, involving interactions with numerous signalling pathways and molecular mechanisms. Notably, it inhibits the Wnt/β-catenin pathway, which is often activated in tumours. This inhibition helps prevent tumour formation by regulating cell proliferation and promoting apoptosis. Restoration of WWOX expression in cancer cell lines has been shown to reduce tumour growth and increased sensitivity to treatments. In addition to its role in tumour suppression, WWOX has been found to interact with proteins involved in critical signalling pathways such as TGF-β. Recent advancements allowed to reveal its interactions with key proteins and microRNAs that regulate cellular adhesion, invasion, and motility. Proteomic studies have shown that WWOX directly interacts with signalling molecules like Dishevelled and SMAD3, further underscoring its role in antagonizing metastasis. Challenges remain in translating this knowledge into clinical applications. For instance, the mechanisms underlying WWOX loss in tumours and its role across diverse cancer types require further investigation. Overall, WWOX serves as a vital player in maintaining cellular stability and preventing cancer progression through its multifaceted functions. Here, we include an updated molecular function of WWOX in cancers to possibly contribute to the potential use of WWOX expression as a biomarker regarding prognosis and response to the treatment. CLINICAL TRIAL NUMBER: Not applicable.
Collapse
Affiliation(s)
- Raneem Y Hammouz
- Department of Molecular Carcinogenesis, Medical University of Lodz, Żeligowskiego 7/9, Lodz, 90-752, Poland
| | - Izabela Baryła
- Department of Molecular Carcinogenesis, Medical University of Lodz, Żeligowskiego 7/9, Lodz, 90-752, Poland
| | - Ewa Styczeń-Binkowska
- Department of Molecular Carcinogenesis, Medical University of Lodz, Żeligowskiego 7/9, Lodz, 90-752, Poland
| | - Andrzej K Bednarek
- Department of Molecular Carcinogenesis, Medical University of Lodz, Żeligowskiego 7/9, Lodz, 90-752, Poland.
| |
Collapse
|
3
|
Tien SC, Shih M, Hu CM. RRM1 O-GlcNAcylation inhibition suppresses pancreatic cancer via TK1-mediated replication stress. Cancer Gene Ther 2025; 32:550-562. [PMID: 40155654 PMCID: PMC12086086 DOI: 10.1038/s41417-025-00895-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 03/07/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
O-GlcNAcylation of ribonucleotide reductase large subunit M1 (RRM1) at position 734 influences high glucose-induced genomic instability and cell transformation in normal pancreatic cells. By disrupting the ribonucleotide reductase complex, it reduces dNTPs. Although the impact of RRM1 O-GlcNAcylation on pancreatic cancer progression remains unexplored, our CRISPR knock-in technology created the RRM1-T734A mutation to minimize RRM1 O-GlcNAcylation. In pancreatic cancer PANC-1 cells with this mutation, we observed heightened replication stress-induced DNA damage, S-phase delays, and diminished in vitro tumor cell growth. Mechanistically, RRM1-T734A enhanced its interaction with RRM2 while impairing binding to RRM2B, leading to decreased NTPs and disrupted dNTP equilibrium. Notably, it doubled dTTP levels via TK1 stabilization mediated by thymidine, resulting in S-phase delay. TK1 silencing restored RRM1-T734A-induced effects on S-phase retardation and decreased colony formation. Our findings highlight the pivotal role of O-GlcNAcylation of RRM1 at T734 in maintaining genomic stability and promoting pancreatic cancer malignancy. Furthermore, reducing RRM1 O-GlcNAcylation increased pancreatic cancer cell sensitivity to gemcitabine, proposing a potential therapeutic strategy.
Collapse
Affiliation(s)
- Sui-Chih Tien
- Genomics Research Center, Academia Sinica, Taipei, 115201, Taiwan
| | - Mei Shih
- Genomics Research Center, Academia Sinica, Taipei, 115201, Taiwan
| | - Chun-Mei Hu
- Genomics Research Center, Academia Sinica, Taipei, 115201, Taiwan.
| |
Collapse
|
4
|
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
|
5
|
Jiang Y, Dai A, Huang Y, Li H, Cui J, Yang H, Si L, Jiao T, Ren Z, Zhang Z, Mou S, Zhu H, Guo W, Huang Q, Li Y, Xue M, Jiang J, Wang F, Li L, Zhong Q, Wang K, Liu B, Wang J, Fan G, Guo J, Chen L, Workman CJ, Shen Z, Kong Y, Vignali DAA, Xu C, Wang H. Ligand-induced ubiquitination unleashes LAG3 immune checkpoint function by hindering membrane sequestration of signaling motifs. Cell 2025; 188:2354-2371.e18. [PMID: 40101708 DOI: 10.1016/j.cell.2025.02.014] [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: 12/13/2023] [Revised: 08/16/2024] [Accepted: 02/18/2025] [Indexed: 03/20/2025]
Abstract
Lymphocyte activation gene 3 (LAG3) has emerged as a promising cancer immunotherapy target, but the mechanism underlying LAG3 activation upon ligand engagement remains elusive. Here, LAG3 was found to undergo robust non-K48-linked polyubiquitination upon ligand engagement, which promotes LAG3's inhibitory function instead of causing degradation. This ubiquitination could be triggered by the engagement of major histocompatibility complex class II (MHC class II) and membrane-bound (but not soluble) fibrinogen-like protein 1 (FGL1). LAG3 ubiquitination, mediated redundantly by the E3 ligases c-Cbl and Cbl-b, disrupted the membrane binding of the juxtamembrane basic residue-rich sequence, thereby stabilizing the LAG3 cytoplasmic tail in a membrane-dissociated conformation enabling signaling. Furthermore, LAG3 ubiquitination is crucial for the LAG3-mediated suppression of antitumor immunity in vivo. Consistently, LAG3 therapeutic antibodies repress LAG3 ubiquitination, correlating with their checkpoint blockade effects. Moreover, patient cohort analyses suggest that LAG3/CBL coexpression could serve as a biomarker for response to LAG3 blockade. Collectively, our study reveals an immune-checkpoint-triggering mechanism with translational potential in cancer immunotherapy.
Collapse
Affiliation(s)
- Yong Jiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Anran Dai
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yuwei Huang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Lingang Laboratory, Shanghai 200031, China
| | - Hua Li
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Jian Cui
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Haochen Yang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Lu Si
- Key Laboratory of Carcinogenesis and Translational Research (Ministry Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing 100142, China
| | - Tao Jiao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing 100142, China
| | - Zhengxu Ren
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Si Mou
- BeiGene, Ltd, Beijing 102206, China
| | | | - Wenhui Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing 100142, China
| | - Qiang Huang
- School of Medicine, Shanghai University, Shanghai 200444, China; State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an 710032, China
| | - Yilin Li
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Manman Xue
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Jingwei Jiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Fei Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Li Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qinying Zhong
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Kun Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Baichuan Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Lingang Laboratory, Shanghai 200031, China
| | - Jinjiao Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Gaofeng Fan
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jun Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing 100142, China
| | - Liang Chen
- School of Medicine, Shanghai University, Shanghai 200444, China; State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an 710032, China
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | | | - Yan Kong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing 100142, China.
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| | - Chenqi Xu
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; Shanghai Academy of Natural Sciences (SANS), Shanghai 200031, China.
| | - Haopeng Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Shanghai Academy of Natural Sciences (SANS), Shanghai 200031, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
6
|
Chung V, Mizrahi JD, Pant S. Novel Therapies for Pancreatic Cancer. JCO Oncol Pract 2025; 21:613-619. [PMID: 39591547 DOI: 10.1200/op.24.00279] [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: 03/29/2024] [Revised: 10/15/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Pancreatic adenocarcinoma (PDAC) unfortunately remains a highly fatal disease with a 5-year survival rate of only 11%. If surgical resection is not possible, systemic chemotherapy represents the standard-of-care approach to management. Combination chemotherapy regimens using fluorouracil (fluorouracil, oxaliplatin, leucovorin, and irinotecan; and fluorouracil, leucovorin, and oxaliplatin) or gemcitabine with albumin-bound paclitaxel have the potential to improve overall survival for patients with advanced disease. With the increasing understanding of the molecular drivers of pancreatic cancer, novel therapeutic approaches have made incremental progress for these patients. The molecular landscape of PDAC has been studied extensively. Approximately 90% of PDACs harbor mutations in the KRAS gene, a driver mutation that has long been considered undruggable. However, novel KRAS inhibitors have shown therapeutic promise in early-phase clinical trials, with larger studies ongoing. Less frequently encountered genomic aberrations that have therapeutic potential include NRG, BRAF, NTRK, HER2, BRCA, PALB2, and claudin. Immune checkpoint inhibitors have unfortunately yielded disappointing efficacy for the majority of patients with pancreatic cancer, except for those with tumors exhibiting deficiency in mismatch repair proteins. Alternative approaches to incorporate immunotherapy have shown more promise such as use of immune checkpoint inhibitors for selected patients in the maintenance setting and potential vaccine therapies in the postsurgery adjuvant setting. It is vital to perform molecular profiling in all patients with PDAC to identify potential treatment targets, and to enroll patients in clinical trials whenever possible.
Collapse
Affiliation(s)
- Victoria Chung
- Ochsner Medical Center, New Orleans, LA
- MD Anderson Cancer Center, Houston, TX
| | | | | |
Collapse
|
7
|
Vella R, Giardino A, Pizzocaro E, Frigerio I, Bannone E, Vieni S, Butturini G. Unconventional Treatments for Pancreatic Cancer: A Systematic Review. Cancers (Basel) 2025; 17:1437. [PMID: 40361364 PMCID: PMC12071172 DOI: 10.3390/cancers17091437] [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: 03/20/2025] [Revised: 04/21/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
OBJECTIVE This study aims to review the existing literature on the efficacy and safety of unconventional treatments among pancreatic cancer patients, including the use of natural products, dietary supplements, probiotics, whole medical systems, and body-based therapies. METHODS An electronic, systematic, and comprehensive literature review was conducted searching for studies up to November 2024 following the PRISMA 2020 guidelines. Randomized controlled trials and prospective and retrospective studies assessing the efficacy and safety of unconventional treatments for pancreatic cancer were considered eligible. Data on overall survival, quality of life, and treatment tolerability were extracted. RESULTS A total of 21 studies, providing data from 3095 patients, met the inclusion criteria. Various types of unconventional treatments are used in pancreatic cancer patients, including Chinese herbal medicine (CHM), mistletoe extract (ME), curcumin, and electroacupuncture. Among these, the use of CHM and curcumin concomitant with standard therapy was associated with survival and quality-of-life benefits. Electroacupuncture reduced pancreatic cancer pain intensity in a cost-effective manner. The data on ME are mixed and of insufficient quality for drawing definitive conclusions. CONCLUSIONS Some unconventional treatments showed potential benefits in improving overall survival and quality of life in pancreatic cancer patients within an integrative oncology setting. Further high-quality studies are needed to provide robust, rigorous, and ethical evidence to support their integration into future guidelines, ensuring a holistic approach to cancer treatment.
Collapse
Affiliation(s)
- Roberta Vella
- HPB Surgery, Pederzoli Hospital, Peschiera del Garda, 37019 Verona, Italy; (R.V.); (A.G.); (I.F.); (E.B.); (G.B.)
- Department of Precision Medicine in Medical, Surgical, and Critical Care, (Me.Pre.C.C.), University of Palermo, 90133 Palermo, Italy;
| | - Alessandro Giardino
- HPB Surgery, Pederzoli Hospital, Peschiera del Garda, 37019 Verona, Italy; (R.V.); (A.G.); (I.F.); (E.B.); (G.B.)
| | - Erica Pizzocaro
- HPB Surgery, Pederzoli Hospital, Peschiera del Garda, 37019 Verona, Italy; (R.V.); (A.G.); (I.F.); (E.B.); (G.B.)
- PhD School of Applied Medical-Surgical Sciences, University Tor Vergata, 00133 Rome, Italy
| | - Isabella Frigerio
- HPB Surgery, Pederzoli Hospital, Peschiera del Garda, 37019 Verona, Italy; (R.V.); (A.G.); (I.F.); (E.B.); (G.B.)
- Collegium Medicum, SAN University, 90-012 Lodz, Poland
| | - Elisa Bannone
- HPB Surgery, Pederzoli Hospital, Peschiera del Garda, 37019 Verona, Italy; (R.V.); (A.G.); (I.F.); (E.B.); (G.B.)
| | - Salvatore Vieni
- Department of Precision Medicine in Medical, Surgical, and Critical Care, (Me.Pre.C.C.), University of Palermo, 90133 Palermo, Italy;
| | - Giovanni Butturini
- HPB Surgery, Pederzoli Hospital, Peschiera del Garda, 37019 Verona, Italy; (R.V.); (A.G.); (I.F.); (E.B.); (G.B.)
| |
Collapse
|
8
|
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
|
9
|
Dunn M, Dymock L, Hoskins C. Solid lipid nanoparticles in pancreatic cancer treatment. BJC REPORTS 2025; 3:21. [PMID: 40217114 PMCID: PMC11992092 DOI: 10.1038/s44276-025-00130-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/13/2025] [Accepted: 03/02/2025] [Indexed: 04/14/2025]
Abstract
Pancreatic cancer comes with one of the poorest prognoses of all cancers and as such it is crucial that new therapies are developed to improve on the current statistics. Currently, chemotherapy is the cornerstone of pancreatic cancer treatment with several drugs, and combinations of drugs being utilised for their anti-cancer effect. However, pancreatic cancer has a dense stroma around the tumour and intratumoral bacteria which result in drugs having difficulty penetrating the tumour or being metabolised by bacteria rendering them inactive. The utilisation of nanotechnology in chemotherapy for pancreatic cancer has been a huge area of focus for researchers worldwide with most of the focus being on lipid-based, inorganic and polymer-based nanoparticles. Solid lipid nanoparticles which have been studied since being first published in the 1990s, have been poorly researched for pancreatic cancer applications. Being composed of physiological lipids, solid lipid nanoparticles offer a greatly reduced risk of acute or chronic toxicities arising compared to inorganic or polymeric nanoparticles. They also possess the ability to improve on circulation time, permeability, and bioavailability of many first-line chemotherapeutics.
Collapse
Affiliation(s)
- Mia Dunn
- Department of Pure and Applied Chemistry, University of Strathclyde, Technology Innovation Centre, 99 George Street, Glasgow, G1 1RD, UK
| | - Lewis Dymock
- Department of Pure and Applied Chemistry, University of Strathclyde, Technology Innovation Centre, 99 George Street, Glasgow, G1 1RD, UK
| | - Clare Hoskins
- Department of Pure and Applied Chemistry, University of Strathclyde, Technology Innovation Centre, 99 George Street, Glasgow, G1 1RD, UK.
| |
Collapse
|
10
|
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
|
11
|
Shichi Y, Fujiwara M, Gomi F, Nonaka K, Hasegawa F, Shinji S, Rokutan H, Arai T, Takahashi K, Ishiwata T. Transmission electron microscopic analysis of pancreatic ductal adenocarcinoma cell spheres formed in 3D cultures. Med Mol Morphol 2025:10.1007/s00795-025-00435-1. [PMID: 40183819 DOI: 10.1007/s00795-025-00435-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 03/22/2025] [Indexed: 04/05/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) cell lines are classified into two types: epithelial and mesenchymal protein-expressing. Using scanning electron microscopy, we reported that these two groups differ in terms of morphology when they formed tumor spheres under three-dimensional (3D) culturing. In this study, we used transmission electron microscopy (TEM) to examine the intracellular microstructures of five epithelial and three mesenchymal PDAC cell lines in 3D culture, and compared them to the morphologies of the same cell types in two-dimensional (2D) cultures. Microvilli were present in all PDAC cells cultured in 2D and 3D, and were well developed in epithelial PDAC cells. Desmosome-like structures were only observed in epithelial PDAC cells, and were more common in 3D cultures. Secretory granules were observed in epithelial PDAC and mesenchymal PANC-1 cells, and were more common in 3D cultures. Intracytoplasmic lumina were only observed in epithelial PK-59 and T3M-4 cells cultured in 3D. Abundant filamentous aggregates were observed in 2D-cultured T3M-4 and MIA PaCa-2 cells. By contrast, entosis was observed in 3D-cultured PK-59, PK-1, and KP4 cells. Microstructural differences enhanced by 3D culturing revealed significant phenotypic diversity among PDAC cells, and may provide key insights into curing intractable pancreatic cancer.
Collapse
Affiliation(s)
- Yuuki Shichi
- Research Team for Geriatric Pathology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Masakazu Fujiwara
- Research Team for Geriatric Pathology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Fujiya Gomi
- Research Team for Geriatric Pathology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Keisuke Nonaka
- Research Team for Geriatric Pathology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Fumio Hasegawa
- Research Team for Geriatric Pathology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Seiichi Shinji
- Research Team for Geriatric Pathology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
- Department of Gastroenterological Surgery, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8603, Japan
| | - Hirofumi Rokutan
- Department of Pathology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan
| | - Tomio Arai
- Department of Pathology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan
| | - Kimimasa Takahashi
- Department of Veterinary Pathology, School of Veterinary Medicine, Nippon Veterinary and Life Science University, Tokyo, 180-8602, Japan
| | - Toshiyuki Ishiwata
- Research Team for Geriatric Pathology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan.
| |
Collapse
|
12
|
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] [Download PDF] [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
|
13
|
Wu Y, Qiao Y, Yang C, Chen Y, Shen X, Deng C, Yao Q, Sun N. Accelerated Exosomal Metabolic Profiling Enabled by Robust On-Target Array Sintering with Metal-Organic Frameworks. SMALL METHODS 2025; 9:e2401238. [PMID: 39263996 DOI: 10.1002/smtd.202401238] [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: 08/07/2024] [Revised: 09/06/2024] [Indexed: 09/13/2024]
Abstract
Pancreatic cancer is highly lethal, and survival chances improve only with early detection at a precancerous stage. However, there remains a significant gap in developing tools for large-scale, rapid screening. To this end, a high-throughput On-Target Array Extraction Platform (OTAEP) by direct sintering of a series of metal-organic frameworks (MOFs) for dual in situ extraction, encompassing both exosomes and their metabolic profiles, is developed. Based on the principle of geometry-dependent photothermal conversion efficiency and standard testing, the appropriate MOF functional unit is identified. This unit enables exosome enrichment within 10 min and metabolic fingerprint extraction in under 1 s of laser irradiation, with over five reuse. To further accelerate and enhance the quality of metabolic profile analysis, the application of Surrogate Variable Analysis to eliminate hidden confounding factors within the profiles is proposed, and five biomarkers demonstrated by MS/MS experiments are identified. These biomarkers enable early diagnosis, risk stratification, and staging of pancreatic cancer simultaneously, with sensitivity of 94.1%, specificity of 98.8%, and precision of 94.9%. This work represents a breakthrough for overcoming throughput challenges in large-scale testing and for addressing confounding factors in big data analysis.
Collapse
Affiliation(s)
- Yun Wu
- Department of Chemistry, Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200433, P. R. China
| | - Yiming Qiao
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Chenyu Yang
- Department of Chemistry, Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200433, P. R. China
| | - Yueying Chen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Xizhong Shen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Chunhui Deng
- Department of Chemistry, Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200433, P. R. China
| | - Qunyan Yao
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
- Department of Gastroenterology and Hepatology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361015, P. R. China
| | - Nianrong Sun
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| |
Collapse
|
14
|
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
|
15
|
Lu Y, Ma H, Xiong X, Du Y, Liu L, Wang J, Zhao W. Deletion of ENO1 sensitizes pancreatic cancer cells to gemcitabine via MYC/RRM1-mediated glycolysis. Sci Rep 2025; 15:9941. [PMID: 40121292 PMCID: PMC11929750 DOI: 10.1038/s41598-025-94319-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 03/12/2025] [Indexed: 03/25/2025] Open
Abstract
Glycolysis is a critical metabolic pathway in cancer cells, fulfilling their energy requirements, supporting biosynthesis, maintaining redox balance, and enabling survival in hostile environments. Alpha-enolase (ENO1) has been identified as a key promoter of tumor progression through its involvement in glycolysis. This study aims to elucidate the relationship between ENO1, glycolysis, and gemcitabine sensitivity in pancreatic cancer (PC). The expression levels of ENO1 in PC were analyzed using the GEPIA2 database, Kaplan-Meier survival plots, and immunohistochemistry (IHC). To assess the impact of ENO1 on gemcitabine sensitivity, we manipulated ENO1 expression in PC cell lines through overexpression and silencing techniques. Subsequent analyses included flow cytometry assays, glucose uptake and lactate production measurements, and cytotoxicity assays. The underlying mechanisms by which ENO1 modulates gemcitabine sensitivity were explored using Western blotting (WB). ENO1 was found to be significantly overexpressed in PC tissues, and elevated ENO1 levels were associated with poorer prognosis in PC patients. Overexpression of ENO1 reduced the sensitivity of PC cells to gemcitabine, enhancing cell proliferation, migration, and invasion by altering the cell cycle and inhibiting apoptosis. Conversely, silencing ENO1 decreased glycolysis in PC cells and heightened their sensitivity to gemcitabine. Furthermore, glycolysis inhibition-achieved through ENO1 knockdown, glucose deprivation, or treatment with 2-Deoxy-D-glucose (2-DG)-further enhanced the susceptibility of PC cells to gemcitabine. Mechanistically, ENO1 was found to regulate the expression of gemcitabine resistance-related genes, particularly ribonucleotide reductase catalytic subunit M1 (RRM1), via MYC through the glycolytic pathway, thereby contributing to gemcitabine resistance. This study demonstrates that ENO1 plays a crucial role in PC progression and is closely linked to gemcitabine resistance through its regulation of the glycolytic pathway.
Collapse
Affiliation(s)
- Yingpeng Lu
- Department of General Surgery, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, No. 77, Chang'an South Rd, Zhangjiagang, 215600, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, No.99, Huaihai West Rd, Xuzhou, 221006, Jiangsu, China
| | - Hongqin Ma
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, No.99, Huaihai West Rd, Xuzhou, 221006, Jiangsu, China
| | - Xiaoxiao Xiong
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, No.99, Huaihai West Rd, Xuzhou, 221006, Jiangsu, China
- Department of General Surgery, The Affiliated Suqian Hospital of Xuzhou Medical University, No 138, Huanghe South Rd, Suqian, 223800, Jiangsu, China
| | - Yusheng Du
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, No.99, Huaihai West Rd, Xuzhou, 221006, Jiangsu, China
| | - Li Liu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, No.99, Huaihai West Rd, Xuzhou, 221006, Jiangsu, China
| | - Ji Wang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, No.99, Huaihai West Rd, Xuzhou, 221006, Jiangsu, China.
| | - Wenxing Zhao
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, No.99, Huaihai West Rd, Xuzhou, 221006, Jiangsu, China.
| |
Collapse
|
16
|
Jiang P, Zong K, Peng D, Zhou B, Wu Z. Risk comparison of adverse reactions between gemcitabine monotherapy and gemcitabine combined with albumin-bound paclitaxel in pancreatic cancer: insights from the FDA Adverse Event Reporting System (FAERS) database. BMC Pharmacol Toxicol 2025; 26:65. [PMID: 40108669 PMCID: PMC11924625 DOI: 10.1186/s40360-025-00884-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/27/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Pancreatic cancer (PC) is a highly aggressive malignancy with limited treatment options. Although gemcitabine monotherapy (G treatment) has long been a standard treatment, combination therapies, such as gemcitabine with albumin-bound paclitaxel (AG treatment), have shown improved outcomes and were approved by the FDA for the PC. However, the AG treatment is also associated with increased adverse events (AEs), which remain inadequately evaluated in real-world settings. METHODS We utilized the FDA Adverse Event Reporting System (FAERS) to conduct a large-scale pharmacovigilance analysis comparing the safety profiles of G and AG treatments for PC. By analyzing adverse event data from the third quarter of 2013 to the second quarter of 2024 and quantifying AE signals with reporting odds ratio (ROR) and proportional reporting ratio (PRR) methods, we compared the risk of AEs between the groups. Time to onset (TTO), subgroup and logistic regression analyses were also performed. RESULTS The study revealed a higher proportion of male (n = 2307, 54.1%) and elderly patients (age ≥ 65years, n = 2172, 50.9%) in the AG treatment group compared to the G treatment group. We found 17 preferred terms with positive signals at the top 50 common AEs, especially in gastrointestinal and blood systems. Cardiac and neurological AEs also needed to be vigilant. Biliary sepsis and infectious enterocolitis were newly identified AEs and deserve attention. Median TTO was 34 (IQR: 8-103) days (G) and 41 (IQR: 10-104) days (AG), with most AEs occurring within the first month (48.3% and 44%). Subgroup analysis revealed that male patients using the AG treatment had the highest risk of immune-mediated hepatitis (ROR = 23.51, 95% CI = 3.21-172.1), while elderly patients had elevated risks for presyncope (ROR = 24.84, 95% CI = 3.40-181.28) and falls (ROR = 18.60, 95% CI = 2.53-136.97). Logistic regression showed higher-risk fatal outcomes in males (adjusted OR = 1.42, 95% CI = 1.15-1.76, P < 0.01) and elderly patients (adjusted OR = 1.36, 95% CI = 1.10-1.69, P < 0.01). CONCLUSION This research offers critical safety insights in real-world settings, emphasizing patients at heightened AEs risk and informing clinical decision-making in PC treatment.
Collapse
Affiliation(s)
- Puen Jiang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Kezhen Zong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Dadi Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Baoyong Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhongjun Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
17
|
Cereda V, D’Andrea MR. Pancreatic cancer: failures and hopes-a review of new promising treatment approaches. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002299. [PMID: 40124650 PMCID: PMC11926728 DOI: 10.37349/etat.2025.1002299] [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: 11/21/2024] [Accepted: 02/22/2025] [Indexed: 03/25/2025] Open
Abstract
Pancreatic cancer is a challenging disease with limited treatment options and a high mortality rate. Just few therapy advances have been made in recent years. Tumor microenvironment, immunosuppressive features and mutational status represent important obstacles in the improvement of survival outcomes. Up to now, first-line therapy did achieve a median overall survival of less than 12 months and this discouraging data lead clinicians all over the world to focus their efforts on various fields of investigation: 1) sequential cycling of different systemic therapy in order to overcome mechanisms of resistance; 2) discovery of new predictive bio-markers, in order to target specific patient population; 3) combination treatment, in order to modulate the tumor microenvironment of pancreatic cancer; 4) new modalities of the delivery of drugs in order to pass the physical barrier of desmoplasia and tumor stroma. This review shows future directions of treatment strategies in advanced pancreatic cancer through a deep analysis of these recent macro areas of research.
Collapse
Affiliation(s)
- Vittore Cereda
- Asl Roma 4, Hospital S. Paolo Civitavecchia, 00053 Civitavecchia, Italy
| | | |
Collapse
|
18
|
Orlandi E, Guasconi M, Romboli A, Giuffrida M, Toscani I, Anselmi E, Porzio R, Madaro S, Vecchia S, Citterio C. State of the Art of Immune Checkpoint Inhibitors in Unresectable Pancreatic Cancer: A Comprehensive Systematic Review. Int J Mol Sci 2025; 26:2620. [PMID: 40141261 PMCID: PMC11942318 DOI: 10.3390/ijms26062620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have transformed the therapeutic landscape for several malignancies, but their efficacy in unresectable pancreatic adenocarcinoma remains uncertain. This systematic review aimed to evaluate the effectiveness and safety of ICIs in this context, focusing on overall survival (OS), progression-free survival (PFS), objective response rate (ORR), disease control rate (DCR), and toxicity. A comprehensive search of MEDLINE, EMBASE, CENTRAL, and Scopus identified 34 eligible studies, including randomized controlled trials and observational cohorts. Quantitative synthesis involved 21 studies comprising 937 patients, with additional qualitative analyses on biomarker-driven subgroups and early-phase trials. The median OS across studies was 8.65 months, while the median PFS was 2.55 months. The ORR and DCR were 16.2% and 50.3%, respectively, with grade ≥3 treatment-related adverse events occurring in 22% of patients. Promising outcomes were observed in MSI-H/dMMR populations, although these represented only 1-2% of cases. Combination strategies with chemotherapy demonstrated synergistic potential but lacked definitive evidence due to heterogeneity and the absence of phase III trials. ICIs showed a manageable toxicity profile, highlighting their feasibility in selected patients. Future research should focus on overcoming tumor microenvironment barriers and identifying biomarkers to optimize responsiveness and expand the applicability of ICIs in pancreatic cancer.
Collapse
Affiliation(s)
- Elena Orlandi
- Department of Oncology-Hematology, Azienda USL of Piacenza, 29121 Piacenza, Italy; (I.T.); (E.A.); (R.P.); (S.M.); (C.C.)
| | - Massimo Guasconi
- Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy;
- Department of Health Professions Management, Azienda USL of Piacenza, 29121 Piacenza, Italy
| | - Andrea Romboli
- Department of General Surgery, Azienda USL of Piacenza, 29121 Piacenza, Italy; (A.R.); (M.G.)
| | - Mario Giuffrida
- Department of General Surgery, Azienda USL of Piacenza, 29121 Piacenza, Italy; (A.R.); (M.G.)
| | - Ilaria Toscani
- Department of Oncology-Hematology, Azienda USL of Piacenza, 29121 Piacenza, Italy; (I.T.); (E.A.); (R.P.); (S.M.); (C.C.)
| | - Elisa Anselmi
- Department of Oncology-Hematology, Azienda USL of Piacenza, 29121 Piacenza, Italy; (I.T.); (E.A.); (R.P.); (S.M.); (C.C.)
| | - Rosa Porzio
- Department of Oncology-Hematology, Azienda USL of Piacenza, 29121 Piacenza, Italy; (I.T.); (E.A.); (R.P.); (S.M.); (C.C.)
| | - Serena Madaro
- Department of Oncology-Hematology, Azienda USL of Piacenza, 29121 Piacenza, Italy; (I.T.); (E.A.); (R.P.); (S.M.); (C.C.)
| | - Stefano Vecchia
- Department of Pharmacy, Azienda USL of Piacenza, 29121 Piacenza, Italy;
| | - Chiara Citterio
- Department of Oncology-Hematology, Azienda USL of Piacenza, 29121 Piacenza, Italy; (I.T.); (E.A.); (R.P.); (S.M.); (C.C.)
| |
Collapse
|
19
|
Aoki Y, Wang L, Tsuda M, Saito Y, Kubota T, Oda Y, Hirano S, Gong JP, Tanaka S. Hydrogel PCDME creates pancreatic cancer stem cells in OXPHOS metabolic state with TXNIP elevation. Biochem Biophys Res Commun 2025; 751:151416. [PMID: 39914146 DOI: 10.1016/j.bbrc.2025.151416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/17/2025]
Abstract
Pancreatic cancer is known as one of the poor prognostic cancers, and the most of patients are unable to undergo radical resection due to local progression or distant metastasis at initial diagnosis. In spite of the advancements in surgery and chemotherapy, there are many cases of recurrence after surgery or chemoradiotherapy mainly due to the presence of cancer stem cells (CSCs). CSCs are potential therapeutic target, but current issue is that an identification of CSCs is difficult since they are only present in a small number of tumor cells. Here we demonstrate that hydrogel PCDME can rapidly induce pancreatic cancer cell spheroids with elevated levels of stem cell markers including Sox2, Oct3/4, and Nanog, and the growth rate was reduced. CSCs showed activation of YAP/TAZ signaling, and microarray analysis showed markedly elevated expression of thioredoxin-interacting protein (TXNIP). Primary pancreatic cancer cells also increased TXNIP in addition to stemness markers on gel. In metabolic analysis, CSCs showed a shift of energy production from glycolysis to oxidative phosphorylation (OXPHOS). Furthermore, knockdown of TXNIP on PCDME gel using shRNAs decreased growth speed and in vivo tumorigenicity, suggesting that TXNIP may be involved in CSCs induction.
Collapse
Affiliation(s)
- Yuma Aoki
- Department of Cancer Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan; Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Lei Wang
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan; World Premier International Research Center Initiative, Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan
| | - Masumi Tsuda
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan; World Premier International Research Center Initiative, Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan
| | - Yusuke Saito
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan; Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Japan
| | - Takenori Kubota
- Department of Cancer Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan; Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Yoshitaka Oda
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Satoshi Hirano
- Department of Gastroenterological Surgery II, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Jian Ping Gong
- World Premier International Research Center Initiative, Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan; Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Shinya Tanaka
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan; World Premier International Research Center Initiative, Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan; Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan.
| |
Collapse
|
20
|
Yamamoto T, Shimokawa T, Hayashi M, Mizuma M, Hirano K, Oba A, Asano T, Miyato H, Yoshida M, Matsumoto I, Kawabata Y, Sakamoto K, Motoi F, Ishii S, Homma Y, Maehira H, Matsunaga Y, Ikemoto T, Nakamura M, Mataki Y, Notake T, Akahoshi K, Takami H, Yamaki S, Hashimoto D, Kimura Y, Hirano S, Inoue Y, Fujii T, Unno M, Kodera Y, Kitayama J, Satoi S, the Study Group of Pancreatic Ductal Adenocarcinoma with Peritoneal Dissemination. Clinical role of intraperitoneal chemotherapy in patients with pancreatic ductal adenocarcinoma concomitant with occult peritoneal dissemination: A multicenter retrospective study. Ann Gastroenterol Surg 2025. [DOI: 10.1002/ags3.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 02/02/2025] [Indexed: 05/03/2025] Open
Abstract
AbstractBackgroundThe effectiveness of intraperitoneal chemotherapy using paclitaxel (i.p.‐PTX) in pancreatic ductal adenocarcinoma (PDAC) patients with peritoneal dissemination remains elusive. The aim of this study is to investigate the clinical outcome of patients treated with i.p.‐PTX combined with systemic chemotherapy compared with current standard chemotherapy including gemcitabine plus nab‐paclitaxel and FOLFIRINOX.MethodsData of patients with peritoneal dissemination was retrospectively collected and analyzed (i.p.‐PTX, n = 83; control, n = 86). Inverse probability of treatment‐weighted analyses (IPTW) was used to balance baseline characteristics between two groups. Survival curves were estimated using Kaplan–Meier method, and the differences were compared using the log‐rank test.ResultsNo significant differences were noted in overall survival (14.9 vs. 15.5 months, p = 0.481) and progression free survival (9.5 vs. 9.1 months, p = 0.267) between i.p.‐PTX and the control groups. Nevertheless, i.p.‐PTX (9.9 months) significantly prolonged the median progression‐free survival (PFS) time compared with the control (8.6 months), among the matched patients using IPTW (hazard ratio 0.666, p = 0.041). Moreover, subgroup analysis among the patients whose primary tumor were evaluated either as resectable or borderline resectable disease revealed significantly better overall survival in the i.p.‐PTX group compared with the control group (21.3 vs. 14.7 months, hazard ratio; 0.532, p = 0.033). Conversion surgery was more frequently performed in the i.p.‐PTX group than the control group (24% vs. 4%, p = 0.006).ConclusionThe i.p. PTX regimen prolonged PFS but not overall survival, and subgroup analysis suggested the possibility of survival benefit in patients with occult peritoneal dissemination whose primary tumor was classified as resectable/borderline resectable disease.
Collapse
Affiliation(s)
- Tomohisa Yamamoto
- Department of Pancreatobiliary Surgery Kansai Medical University Osaka Japan
| | - Toshio Shimokawa
- Clinical Study Support Center, Wakayama Medical University School of Medicine Wakayama Japan
| | - Masamichi Hayashi
- Department of Gastroenterological Surgery Nagoya University Graduate School of Medicine Nagoya Japan
| | - Masamichi Mizuma
- Department of Surgery Tohoku University Graduate School of Medicine Sendai Japan
| | - Katsuhisa Hirano
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly University of Toyama Toyama Japan
| | - Atsushi Oba
- Division of Hepatobiliary and Pancreatic Surgery Cancer Institute Hospital, Japanese Foundation for Cancer Research Tokyo Japan
| | - Toshimichi Asano
- Department of Gastroenterological Surgery II, Faculty of Medicine Hokkaido University Sapporo Japan
| | - Hideyo Miyato
- Department of Gastrointestinal Surgery Jichi Medical University Shimotsuke Japan
| | - Makoto Yoshida
- Department of Medical Oncology Sapporo Medical University School of Medicine Sapporo Japan
| | - Ippei Matsumoto
- Department of Surgery Kindai University Faculty of Medicine Osaka Japan
| | - Yasunari Kawabata
- Department of Digestive and General Surgery Shimane University Faculty of Medicine Izumo Japan
| | - Katsunori Sakamoto
- Department of Hepato‐Biliary‐Pancreatic and Breast Surgery Ehime University Graduate School of Medicine Ehime Japan
| | - Fuyuhiko Motoi
- Department of Surgery Yamagata University Graduate School of Medical Science Yamagata Japan
| | - Shigeto Ishii
- Department of Gastroenterology, Graduate School of Medicine Juntendo University Tokyo Japan
| | - Yuki Homma
- Department of Gastroenterological Surgery Yokohama City University Graduate School of Medicine Yokohama Japan
| | - Hiromitsu Maehira
- Department of Surgery Shiga University of Medical Science Otsu Japan
| | - Yutaro Matsunaga
- Department of Surgery, Institute of Gastroenterology Tokyo Women's Medical University Tokyo Japan
| | | | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Yuko Mataki
- Department of Digestive Surgery, Graduate School of Medicine Kagoshima University Kagoshima Japan
| | - Tsuyoshi Notake
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation, and Pediatric Surgery, Department of Surgery Shinshu University School of Medicine Nagano Japan
| | - Keiichi Akahoshi
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine Tokyo Medical and Dental University Tokyo Japan
| | - Hideki Takami
- Department of Gastroenterological Surgery Nagoya University Graduate School of Medicine Nagoya Japan
| | - So Yamaki
- Department of Pancreatobiliary Surgery Kansai Medical University Osaka Japan
| | - Daisuke Hashimoto
- Department of Pancreatobiliary Surgery Kansai Medical University Osaka Japan
| | - Yasutoshi Kimura
- Department of Surgery, Surgical Oncology and Science Sapporo Medical University School of Medicine Sapporo Japan
| | - Satoshi Hirano
- Department of Gastroenterological Surgery II, Faculty of Medicine Hokkaido University Sapporo Japan
| | - Yosuke Inoue
- Division of Hepatobiliary and Pancreatic Surgery Cancer Institute Hospital, Japanese Foundation for Cancer Research Tokyo Japan
| | - Tsutomu Fujii
- Department of Surgery and Science, Faculty of Medicine, Academic Assembly University of Toyama Toyama Japan
| | - Michiaki Unno
- Department of Surgery Tohoku University Graduate School of Medicine Sendai Japan
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery Nagoya University Graduate School of Medicine Nagoya Japan
| | - Joji Kitayama
- Department of Gastrointestinal Surgery Jichi Medical University Shimotsuke Japan
| | - Sohei Satoi
- Department of Pancreatobiliary Surgery Kansai Medical University Osaka Japan
- Division of Surgical Oncology University of Colorado Anschutz Medical Campus Aurora Colorado USA
| | | |
Collapse
|
21
|
Höfer S, Frasch L, Brajkovic S, Putzker K, Lewis J, Schürmann H, Leone V, Sakhteman A, The M, Bayer FP, Müller J, Hamood F, Siveke JT, Reichert M, Kuster B. Gemcitabine and ATR inhibitors synergize to kill PDAC cells by blocking DNA damage response. Mol Syst Biol 2025; 21:231-253. [PMID: 39838187 PMCID: PMC11876601 DOI: 10.1038/s44320-025-00085-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 12/22/2024] [Accepted: 01/03/2025] [Indexed: 01/23/2025] Open
Abstract
The DNA-damaging agent Gemcitabine (GEM) is a first-line treatment for pancreatic cancer, but chemoresistance is frequently observed. Several clinical trials investigate the efficacy of GEM in combination with targeted drugs, including kinase inhibitors, but the experimental evidence for such rationale is often unclear. Here, we phenotypically screened 13 human pancreatic adenocarcinoma (PDAC) cell lines against GEM in combination with 146 clinical inhibitors and observed strong synergy for the ATR kinase inhibitor Elimusertib in most cell lines. Dose-dependent phosphoproteome profiling of four ATR inhibitors following DNA damage induction by GEM revealed a strong block of the DNA damage response pathway, including phosphorylated pS468 of CHEK1, as the underlying mechanism of drug synergy. The current work provides a strong rationale for why the combination of GEM and ATR inhibition may be useful for the treatment of PDAC patients and constitutes a rich phenotypic and molecular resource for further investigating effective drug combinations.
Collapse
Affiliation(s)
- Stefanie Höfer
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Larissa Frasch
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Sarah Brajkovic
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Kerstin Putzker
- Chemical Biology Core Facility, EMBL Heidelberg, Heidelberg, Germany
| | - Joe Lewis
- Chemical Biology Core Facility, EMBL Heidelberg, Heidelberg, Germany
| | - Hendrik Schürmann
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, Essen, Germany
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Valentina Leone
- Department of Internal Medicine II, University Hospital Rechts der Isar, Technical University Munich, Munich, Germany
| | - Amirhossein Sakhteman
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Matthew The
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Florian P Bayer
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Julian Müller
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Firas Hamood
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Jens T Siveke
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, Essen, Germany
| | - Maximilian Reichert
- Department of Internal Medicine II, University Hospital Rechts der Isar, Technical University Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.
| |
Collapse
|
22
|
Zhang C, Li K, Xu SN, Qiao L, Ren YL, Li Q, Liu Y. A real-world pharmacovigilance study of FDA Adverse Event Reporting System (FAERS) for gemcitabine. Expert Opin Drug Saf 2025; 24:365-376. [PMID: 39441316 DOI: 10.1080/14740338.2024.2419999] [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: 08/04/2024] [Revised: 10/10/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Gemcitabine is widely used in the treatment of various cancers. This study aims to evaluate gemcitabine-associated adverse events (AEs) using the Food and Drug Administration Adverse Event Reporting System (FAERS) database. RESEARCH DESIGN AND METHODS We analyzed data spanning from January 2004 to June 2023. Employing reporting odds ratio (ROR) and Bayesian confidence propagation neural network (BCPNN) algorithms, we identified AEs with positive signals in patients administered gemcitabine. RESULTS Out of 16,623,939 reports, 23,645 involved gemcitabine as the 'primary suspected (PS)' resulting in 74,306 AEs. Consistent with the reports in the specification and clinical trials, thrombocytopenia, pyrexia, neutropenia, and anemia were the most common AEs. Notably, our study identified some unexpected AEs such as abdominal pain, pleural effusion, ascites, and gastrointestinal hemorrhage, among others. The most significant SOC was 'Blood and lymphatic system disorders'. The median onset time for gemcitabine-related AEs was 24 days (interquartile range [IQR] 6-82 days), with most cases occurring within the initial 30 days following gemcitabine administration. CONCLUSION Gemcitabine is associated with a broad spectrum of AEs affecting multiple organ systems, with a notable incidence of hospitalization. The study highlights both expected and unexpected AEs, which could enhance future clinical applications and safety of gemcitabine.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Ke Li
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Shu-Ning Xu
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Lei Qiao
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Yu-Lin Ren
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Qun Li
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Ying Liu
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
23
|
Chang CY, Chiu TJ, Chen YY, Chen JS, Chou WC. Outcomes of Liposomal Irinotecan With 5-FU and Leucovorin in Patients With Metastatic Pancreatic Cancer and Borderline Performance Status. CANCER DIAGNOSIS & PROGNOSIS 2025; 5:198-206. [PMID: 40034960 PMCID: PMC11871866 DOI: 10.21873/cdp.10430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 01/23/2025] [Accepted: 02/04/2025] [Indexed: 03/05/2025]
Abstract
Background/Aim This study investigated the effectiveness and safety of liposomal irinotecan plus 5-fluorouracil (5-FU) and leucovorin (LV) as a second-line treatment for patients with pancreatic ductal adenocarcinoma (PDAC) and borderline performance status. These patients are often excluded from clinical trials, but may still receive this therapy due to limited alternatives. Patients and Methods A retrospective analysis was conducted on 31 patients with metastatic PDAC and a Karnofsky Performance Status (KPS) of 40-60, who received liposomal irinotecan plus 5-FU/LV at two institutions between 2018 and 2019 in Taiwan. The NAPOLI nomogram was utilized to stratify patients into prognostic groups for survival comparison, with the good prognostic group defined as having total NAPOLI nomogram points ≥ the median value, and the poor prognostic group defined as having total NAPOLI nomogram points < the median value. Results The median overall survival (OS) for the entire cohort was 4.2 months [95% confidence interval (CI)=3.0-5.3 months]. Patients in the NAPOLI nomogram's good prognostic group had a median OS of 4.9 months (95%CI=3.7-6.1 months), compared to 2.0 months (95%CI=1.5-2.4 months) in the poor prognostic group (p=0.014). Tumor response rates to liposomal irinotecan + 5-FU/LV were partial response in 3%, stable disease in 23%, and progressive disease in 74%, with significant differences in disease progression rates between good (56%) and poor (93%) prognostic groups (p=0.011). The most common grade 3 or 4 chemotherapy-related adverse events included anemia (26%), neutropenia (23%), non-neutropenic infection (19%), mucositis (13%), diarrhea (10%), and fatigue (3%). Conclusion Liposomal irinotecan plus 5-FU/LV regimen is a feasible second-line treatment option for PDAC patients with borderline performance status, with a safety profile consistent with clinical trial findings.
Collapse
Affiliation(s)
- Chieh-Ying Chang
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Linkou, School of Medicine, Chang Gung University, Taoyuan, Taiwan, R.O.C
| | - Tai-Jan Chiu
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Kaohsiung, School of Medicine, Chang Gung University, Taoyuan, Taiwan, R.O.C
| | - Yen-Yang Chen
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Kaohsiung, School of Medicine, Chang Gung University, Taoyuan, Taiwan, R.O.C
| | - Jen-Shi Chen
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Linkou, School of Medicine, Chang Gung University, Taoyuan, Taiwan, R.O.C
| | - Wen-Chi Chou
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Linkou, School of Medicine, Chang Gung University, Taoyuan, Taiwan, R.O.C
| |
Collapse
|
24
|
Major A, Dueck AC, Thanarajasingam G. SOHO State of the Art Updates and Next Questions | Measuring Patient-Reported Outcomes (PROs) and Treatment Tolerability in Patients With Hematologic Malignancies. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025; 25:142-155. [PMID: 39198102 DOI: 10.1016/j.clml.2024.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/28/2024] [Indexed: 09/01/2024]
Abstract
There has been a rapid expansion of novel therapeutics for hematologic malignancies, including monoclonal antibodies, small molecules, and cellular therapies, which confer different treatment-related toxicities and symptomatic adverse events (AEs) than traditional cytotoxic chemotherapies. Given that patients with blood cancers are living longer with these newer treatments, with some therapies requiring indefinite or time-intensive administration, consideration of patient-reported tolerability and effects on health-related quality of life (HRQoL) are increasingly relevant. Historically, clinical trials have focused on the efficacy and safety of therapies. While related to safety and not intended to replace it, "treatment tolerability" is a distinct construct defined as the extent to which symptomatic and nonsymptomatic AEs impact a patient's ability and desire to continue with current treatment dosing, which also encompasses how patients feel and function while undergoing anticancer therapies. Assessment of tolerability requires the systematic and rigorous measurement of patient-reported outcomes (PROs). In this review, we discuss the introduction of patient-reported outcomes measures (PROMs) into hematology clinical trials and how PROs inform the measurement of treatment tolerability, including symptomatic adverse events, physical and role functioning, and overall side effect burden. Selected PROMs for measurement of these core tolerability domains are outlined, with a focus on novel analytic tools that have been developed for the longitudinal analysis of tolerability data. Further, we outline ongoing studies to accelerate integration of PROs throughout the cancer care spectrum, from early-stage drug development to routine clinical care, with the goal of improving both HRQoL and survival.
Collapse
Affiliation(s)
- Ajay Major
- Division of Hematology, Department of Medicine, University of Colorado School of Medicine, Aurora, Co.
| | - Amylou C Dueck
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Scottsdale, Az
| | | |
Collapse
|
25
|
Lan CC, Chiu TJ, Hung CY, Yeh KY, Lu CH, Chen YY, Chen JS, Hung YS, Chou WC. The efficacy and safety profile of third-line treatment in patients with metastatic pancreatic adenocarcinoma. Pancreatology 2025; 25:266-274. [PMID: 39915162 DOI: 10.1016/j.pan.2025.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/01/2024] [Accepted: 01/31/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND For metastatic pancreatic ductal adenocarcinoma (mPDAC), there are no established third-line chemotherapy options. We examined the efficacy and safety of third-line chemotherapy in patients with mPDAC in real-world practice. METHODS We retrospectively analyzed 257 patients with mPDAC and progressive disease after first-line treatment with gemcitabine-based regimens and second-line treatment with liposomal irinotecan plus 5-fluorouracil and leucovorin at five Taiwanese medical centers from 2018 to 2022. Treatment efficacy and toxicity were analyzed in 77 of 257 patients receiving third-line treatment subsequently. We performed univariate and multivariate analyses to identify prognostic factors for overall survival (OS) in patients receiving third-line treatment. RESULTS Patients receiving third-line treatment had a median OS of 4.5 months (95 % confidence interval [CI], 3.6-5.4), compared to 1.6 months (95 % CI, 1.3-1.9) for those who did not. Independent poor prognostic factors for OS included the absence of previous pancreatectomy (adjusted hazard ratio [aHR] 3.03, 95 % CI, 1.30-7.14, P = 0.001), an ECOG score of ≥2 ((aHR 9.81, 95 % CI 4.34-22.1, P < 0.001), and progressive disease response during second-line treatment (aHR 1.90, 95 % CI 1.21-8.91, P = 0.020, P = 0.020). Median OS for patients with none, one, two, and three poor prognostic factors were 15.9 (95 % CI, 12.3-19.6), 7.0 (2.6-13.3), 4.4 (3.5-5.2), and 2.0 (1.7-2.2) months, respectively. 43 of 77 patients (56 %) experienced at least one grade 3 or 4 toxicity. CONCLUSION In real-world settings, patients with mPDAC receiving third-line chemotherapy may have a moderate survival advantage, although clinicians should carefully select patients owing to high incidence of grade 3/4 toxicities.
Collapse
Affiliation(s)
- Chi-Chen Lan
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Linkou Branch, Chang Gung University College of Medicine, Taoyaun, Taiwan
| | - Tai-Jan Chiu
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Kaohsiung Branch, Chang Gung University College of Medicine, Taoyaun, Taiwan
| | - Chia-Yen Hung
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Linkou Branch, Chang Gung University College of Medicine, Taoyaun, Taiwan; Division of Hematology and Oncology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Kun-Yun Yeh
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Keelung Branch, Chang Gung University College of Medicine, Taoyaun, Taiwan
| | - Chang-Hsien Lu
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Chiayi Branch, Chang Gung University College of Medicine, Taoyaun, Taiwan
| | - Yen-Yang Chen
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Kaohsiung Branch, Chang Gung University College of Medicine, Taoyaun, Taiwan
| | - Jen-Shi Chen
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Linkou Branch, Chang Gung University College of Medicine, Taoyaun, Taiwan
| | - Yu-Shin Hung
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Linkou Branch, Chang Gung University College of Medicine, Taoyaun, Taiwan
| | - Wen-Chi Chou
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Linkou Branch, Chang Gung University College of Medicine, Taoyaun, Taiwan.
| |
Collapse
|
26
|
Govindasamy C, Khan MI, Bose C, Bharathi M, Senthilkumar S, Surya P. Optimizing in vitro lung cancer therapy with folate-conjugated polydopamine-coated liposomes loaded with gemcitabine. Adv Med Sci 2025; 70:141-151. [PMID: 39923845 DOI: 10.1016/j.advms.2025.02.001] [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: 04/27/2024] [Revised: 08/25/2024] [Accepted: 02/03/2025] [Indexed: 02/11/2025]
Abstract
PURPOSE Surface-altered, targeted nanocarriers play crucial roles in chemotherapy. Incorporating ligands into polymers may alter their chemical composition, potentially compromising their drug storage and encapsulation capacity. Polydopamine (PDA) is a novel, biocompatible, and versatile agent for producing targeted nanoparticles that serve as a base for conjugating specific ligands to non-reactive polymeric nanocarriers. This investigation aimed to evaluate whether gemcitabine (GEM)-loaded liposomes conjugated with PDA could enhance cancer treatment. MATERIALS AND METHODS A series of liposomes, named plain GEM, GEM@FA, and GEM@FA/PDA, was designed. Transmission Electron Microscopy (TEM), Fourier Transform Infrared Spectroscopy (FTIR), and X-ray Photoelectron Spectroscopy (XPS) were used to confirm the presence of PDA coating and folic acid (FA) and PDA conjugations. Cellular uptake, cytotoxicity, and cell death were evaluated using biochemical and flow cytometric assays. RESULTS Compared to typical liposomes, GEM@FA/PDA liposomes were smaller, more stable, and exhibited a spherical shape with excellent cellular uptake. GEM@FA and GEM@FA/PDA liposomes showed significantly higher cytotoxicity against lung cancer (H1299) cells compared to GEM liposomes and pure GEM solution at all concentrations, while causing much less cytotoxicity to normal cells (NIH3T3). CONCLUSIONS GEM@FA/PDA liposomes demonstrated enhanced cancer-fighting effectiveness while minimizing harm to healthy tissues, making them a promising approach for chemotherapy.
Collapse
Affiliation(s)
- Chandramohan Govindasamy
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh, 11433, Saudi Arabia
| | - Muhammad Ibrar Khan
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh, 11433, Saudi Arabia
| | - Chitrakani Bose
- Department of Microbiology, Velmanoharan Arts and Science College for Women, Marapalam, Devipattinam (ECR Road), Ramanathapuram, Tamilnadu, India
| | - Muruganantham Bharathi
- Centre for Bioinformatics, Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
| | - Shamini Senthilkumar
- Department of Chemistry, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu, India
| | - Parthasarathy Surya
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India.
| |
Collapse
|
27
|
Gotfrit J, Marginean H, Ko YJ, Ghafoor A, Kavan P, Chalchal H, Ahmed S, Mulder K, Tang P, Goodwin R. Administration of FOLFIRINOX for Advanced Pancreatic Cancer: Physician Practice Patterns During Early Use. Curr Oncol 2025; 32:128. [PMID: 40136332 PMCID: PMC11941058 DOI: 10.3390/curroncol32030128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 03/27/2025] Open
Abstract
Advanced pancreatic cancer results in high morbidity and mortality. The standard of care treatment in the advanced setting changed in 2011 with the introduction of FOLFIRINOX (FFX) chemotherapy. However, it was highly toxic with significant risk of complications. We assessed the practice patterns of medical oncologists across Canada. METHODS We performed a retrospective study of consecutive patients with advanced pancreatic cancer treated with FFX at eight Canadian cancer centers. Demographic, treatment, and outcome data were collected and analyzed. RESULTS The median age of the patients was 61 (range 24-80), 43% were female, 96% had an ECOG PS of 0 or 1, and 50% had three or more metastatic sites. The median follow-up time was 20.8 months (95%CI 18.6-24.9). Physicians started FFX at the standard dose 31% of the time. Physicians prescribed GCSF for primary prophylaxis most when giving standard-dose FFX (30% of the time) in comparison to reduced dose with or without the 5-FU bolus. Dose reductions occurred in 78.1% of patients, while dose delay occurred in 65.2% of patients. CONCLUSIONS Medical oncologists in Canada historically prescribed FFX to patients with advanced pancreatic cancer in a fashion that was not uniform, prior to the emergence of evidence for upfront dose reductions.
Collapse
Affiliation(s)
- Joanna Gotfrit
- The Ottawa Hospital Cancer Centre (TOHCC), Ottawa, ON K1H 8L6, Canada
| | - Horia Marginean
- The Ottawa Hospital Cancer Centre (TOHCC), Ottawa, ON K1H 8L6, Canada
| | | | - Akmal Ghafoor
- Windsor Regional Hospital (WRH), Windsor, ON N8W 2X3, Canada
| | - Petr Kavan
- Jewish General Hospital (JGH), Montréal, QC H3T 1E2, Canada
| | - Haji Chalchal
- Allan Blair Cancer Centre (ABCC), Regina, SK S4T 7T1, Canada
| | - Shahid Ahmed
- Saskatoon Cancer Center (SCC), Saskatoon, SK S7N 4H4, Canada
| | - Karen Mulder
- Cross Cancer Institute (CCI), Edmonton, AB T6G 1Z2, Canada
| | - Patricia Tang
- Arthur J. E. Child Comprehensive Cancer Centre (ACCC), Calgary, AB T2N 5G2, Canada
| | - Rachel Goodwin
- The Ottawa Hospital Cancer Centre (TOHCC), Ottawa, ON K1H 8L6, Canada
| |
Collapse
|
28
|
Uniyal P, Kashyap VK, Behl T, Parashar D, Rawat R. KRAS Mutations in Cancer: Understanding Signaling Pathways to Immune Regulation and the Potential of Immunotherapy. Cancers (Basel) 2025; 17:785. [PMID: 40075634 PMCID: PMC11899378 DOI: 10.3390/cancers17050785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/15/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
The Kirsten rat sarcoma viral oncogene homologue (KRAS) mutation is one of the most prevailing mutations in various tumors and is difficult to cure. Long-term proliferation in carcinogenesis is primarily initiated by oncogenic KRAS-downstream signaling. Recent research suggests that it also activates the autocrine effect and interplays the tumor microenvironment (TME). Here, we discuss the emerging research, including KRAS mutations to immune evasion in TME, which induce immunological modulation that promotes tumor development. This review gives an overview of the existing knowledge of the underlying connection between KRAS mutations and tumor immune modulation. It also addresses the mechanisms to reduce the effect of oncogenes on the immune system and recent advances in clinical trials for immunotherapy in KRAS-mutated cancers.
Collapse
Affiliation(s)
- Priyanka Uniyal
- Department of Pharmaceutical Technology, School of Health Sciences and Technology, UPES, Dehradun 248007, India;
| | - Vivek Kumar Kashyap
- Division of Cancer Immunology and Microbiology, Medicine, and Oncology Integrated Service Unit, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA;
- South Texas Center of Excellence in Cancer Research (ST-CECR), School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Mohali 140306, India;
| | - Deepak Parashar
- Division of Hematology & Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ravi Rawat
- Department of Pharmaceutical Technology, School of Health Sciences and Technology, UPES, Dehradun 248007, India;
| |
Collapse
|
29
|
Bangbo Z, Cheng Q, Zeru L, Tianyu L, Yutong Z, Weibin W, Yupei Z. RNA binding protein Pumilio2 promotes chemoresistance of pancreatic cancer via focal adhesion pathway and interacting with transcription factor EGR1. Cell Mol Life Sci 2025; 82:78. [PMID: 39961821 PMCID: PMC11832970 DOI: 10.1007/s00018-025-05599-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 02/20/2025]
Abstract
Pancreatic cancer (PCa) has insidious onset, high malignancy and poor prognosis. Gemcitabine (GEM) is one of the first-line chemotherapy drugs for PCa. However, GEM resistance has always been a bottleneck problem leading to recurrence and death of PCa patients. RNA-binding proteins (RBPs) are important proteins that regulate transportation, splicing, stability and translation of RNA. Abnormal expression of RBPs often lead to a series of abnormal accumulation or degradation of downstream RNA resulting in various diseases. In our study, we utilized RIP seq, RIP-qPCR, in vitro and in vivo experiments and found that pumilio2 (PUM2) was high expression in PCa, and promoted GEM resistance of PCa by regulating mRNA stability of integrin Alpha 3 (ITGA3) and other genes in focal adhesion pathway, and there was positive feedback regulation between PUM2 and transcription factor early growth response gene 1 (EGR1), that is PUM2 binding to 3'UTR region of EGR1 mRNA, and EGR1 binding to promoter region of PUM2 gene. The discovery of EGR1/PUM2/ITGA3 axis provided a solid experimental basis for the selection of chemotherapy regiments for PCa patients and exploration of combined regimens to reverse GEM resistance in the future.
Collapse
Affiliation(s)
- Zhao Bangbo
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Qin Cheng
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Li Zeru
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Li Tianyu
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhao Yutong
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Wang Weibin
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Zhao Yupei
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
30
|
Di Marco F, Cufaro MC, Damiani V, Dufrusine B, Pizzinato E, Di Ferdinando F, Sala G, Lattanzio R, Dainese E, Federici L, Ponsaerts P, De Laurenzi V, Cicalini I, Pieragostino D. Proteomic meta-analysis unveils new frontiers for biomarkers research in pancreatic carcinoma. Oncogenesis 2025; 14:3. [PMID: 39956821 PMCID: PMC11830788 DOI: 10.1038/s41389-025-00547-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/20/2024] [Accepted: 02/06/2025] [Indexed: 02/18/2025] Open
Abstract
Pancreatic carcinoma (PC) is the sixth leading cause of cancer death in both sexes in 2022, responsible for almost 5% of all cancer deaths worldwide; it is characterized by a poor prognosis since most patients present with an unresectable and metastatic tumor. To date, the decreasing trend in mortality rates related to the most common cancers has contributed to making pancreatic cancer a serious public health problem. In the last few years, scientific research has led to many advances in diagnostic approaches, perioperative management, radiotherapy techniques, and systemic therapies for advanced disease, but only with modest incremental progress in PC patient outcomes. Most of the causes of this high mortality are, unfortunately, late diagnosis and an important therapeutic resistance; for this reason, the most recent high-throughput proteomics technologies focus on the identification of novel biomarkers and molecular profiling to generate new insights in the study of PC, to improve diagnosis and prognosis and to monitor the therapies progress. In this work, we present and discuss the integration of results from different revised studies on protein biomarkers in a global proteomic meta-analysis to understand which path to pursue scientific research. In particular, cancer signaling, inflammatory response, and cell migration and signaling have emerged as the main pathways described in PC, as well as scavenging of free radicals and metabolic alteration concurrently highlighted new research insights on this disease. Interestingly, from the study of upstream regulators, some were found to be shared by collecting data relating to both biological fluid and tissue biomarkers, side by side: specifically, TNF, LPS, p38-MAPK, AGT, miR-323-5p, and miR-34a-5p. By integrating many biological components with their interactions and environmental relationships, it's possible to achieve an in-depth description of the pathological condition in PC and define correlations between concomitant symptoms and tumor genesis and progression. In conclusion, our work may represent a strategy to combine the results from different studies on various biological samples in a more comprehensive way.
Collapse
Affiliation(s)
- Federica Di Marco
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Maria Concetta Cufaro
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Verena Damiani
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Beatrice Dufrusine
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Erika Pizzinato
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Telematic University of "Leonardo Da Vinci", Torrevecchia Teatina, Chieti, Italy
| | - Fabio Di Ferdinando
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Gianluca Sala
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Rossano Lattanzio
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Enrico Dainese
- Department of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Luca Federici
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerpen, Belgium
| | - Vincenzo De Laurenzi
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Ilaria Cicalini
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy.
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy.
| | - Damiana Pieragostino
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
31
|
Okoro J, Bulusu R, Frimpong E, Zhu X, Rogers S, Agyare E. A novel gemcitabine analog as a potential anticancer agent: synthesis and in-vitro evaluation against pancreatic cancer. Am J Cancer Res 2025; 15:684-704. [PMID: 40084373 PMCID: PMC11897620 DOI: 10.62347/kxsr8930] [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: 11/01/2024] [Accepted: 12/28/2024] [Indexed: 03/16/2025] Open
Abstract
Gemcitabine (Gem) is approved for use in pancreatic cancer chemotherapy. However, Gem undergoes rapid metabolism in the blood, producing an inactive metabolite. Due to this rapid metabolism, the effective dose of Gem is high, thereby predisposing patients to severe adverse effects. This study aimed to improve Gem's metabolic and therapeutic stability by modifying the amine group (4-NH2) with hydroxylamine to form 4-N-hydroxylGem hydrochloride (GemAGY). Micro-elemental analysis and Nuclear Magnetic Resonance (NMR) were used to characterize GemAGY, and its anticancer activity was investigated against MiaPaCa-2, BxPC-3, and PANC-1 pancreatic cancer cell lines. The GemAGY metabolic stability was evaluated in human liver microsomal solution. In the 2D cytotoxicity assay, the IC50 values of GemAGY-treated MiaPaCa-2, PANC-1, and BxPC-3 cells were significantly lower when compared to GemHCl-treated cultures. More so, in 3D spheroid assay results, GemAGY IC50 values were found to be 9.5 ± 1.1 µM and 12.6 ± 1.0 µM when compared to GemHCl IC50 values of 24.1 ± 1.6 µM and 30.2 ± 1.8 µM in MiaPaCa-2 and PANC-1 cells, respectively. GemAGY was stable, with 60% remaining intact after 2 hours of digestion in microsomal enzymes, compared to GemHCl, which had less than 45% remaining intact after 30 minutes. GemAGY-treated MiaPaCa-2 and PANC-1 cells at 3.12 and 6.25 μM concentrations demonstrated a significantly reduced cell migration towards the wound area compared to the GemHCl-treated cultures at the same concentrations. Further, GemAGY-treated MiaPaCa-2 cells significantly increased the expression of p53 and BAX compared to GemHCl-treated cells. GemAGY demonstrated significant anticancer activity and improved metabolic stability compared to GemHCl and is most likely to have potential anticancer activity against pancreatic cancer.
Collapse
Affiliation(s)
- Joy Okoro
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M UniversityTallahassee, Florida, USA
| | - Raviteja Bulusu
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M UniversityTallahassee, Florida, USA
| | - Esther Frimpong
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M UniversityTallahassee, Florida, USA
| | - Xue Zhu
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M UniversityTallahassee, Florida, USA
| | - Sherise Rogers
- Department of Medicine, Division of Hematology and Oncology, University of Florida College of MedicineGainesville, Florida, USA
| | - Edward Agyare
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M UniversityTallahassee, Florida, USA
| |
Collapse
|
32
|
Hayat U, Croce PS, Saadeh A, Desai K, Appiah J, Khan S, Khan YI, Kumar K, Hanif A. Current and Emerging Treatment Options for Pancreatic Cancer: A Comprehensive Review. J Clin Med 2025; 14:1129. [PMID: 40004658 PMCID: PMC11856716 DOI: 10.3390/jcm14041129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/30/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of death worldwide, and its global burden has increased significantly over the past few years. The incidence of pancreatic cancer has also increased in the United States, and most of this increase is attributed to the population's aging process in addition to the rise in the prevalence of risk factors such as obesity, diabetes, smoking, and alcohol intake. Most patients with pancreatic cancer present with advanced unresectable or metastatic disease. Only a few patients present at an early stage with localized disease, and a multidisciplinary approach is required to maximize survival and outcomes. The surgical approach is an option for localized disease, and surgery's safety and efficacy have also been improved in recent years due to the increasing use of minimally invasive surgical techniques. Moreover, systematic chemotherapy has also been used and has had a significant impact on survival. More recently, neoadjuvant therapy has been used for pancreatic cancer along with radiation therapy, optimizing survival among those patients. Targeted therapies have been introduced based on genetic testing in metastatic pancreatic cancer and have shown promising results. Moreover, immune checkpoint inhibitors and targeted agents such as PARP inhibitors and vaccines have emerged with optimal results in terms of survival. To conclude, pancreatic cancer is considered a disease with poor long-term survival; however, recent developments in pharmacotherapy have changed its treatment and have improved outcomes with improved survival. Our review summarizes ongoing therapeutic options for local and metastatic pancreatic cancer. It also summarizes new state-of-the-art therapies that have emerged or are in trials, which can change the pancreatic cancer treatment perspective.
Collapse
Affiliation(s)
- Umar Hayat
- Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, PA 18711, USA; (P.S.C.); (K.D.); (J.A.); (S.K.)
| | - Phillip S. Croce
- Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, PA 18711, USA; (P.S.C.); (K.D.); (J.A.); (S.K.)
| | - Aseel Saadeh
- Department of Internal Medicine, Geisinger Medical Center, Danville, PA 18711, USA;
| | - Karna Desai
- Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, PA 18711, USA; (P.S.C.); (K.D.); (J.A.); (S.K.)
| | - John Appiah
- Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, PA 18711, USA; (P.S.C.); (K.D.); (J.A.); (S.K.)
| | - Sidrah Khan
- Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, PA 18711, USA; (P.S.C.); (K.D.); (J.A.); (S.K.)
| | - Yakub I. Khan
- Department of Internal Medicine, Division of Gastroenterology, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, PA 18711, USA; (Y.I.K.); (K.K.)
| | - Kishore Kumar
- Department of Internal Medicine, Division of Gastroenterology, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, PA 18711, USA; (Y.I.K.); (K.K.)
| | - Ahmad Hanif
- Department of Internal Medicine, Division of Hematology/Oncology, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, PA 18711, USA;
| |
Collapse
|
33
|
Ma Y, Dumesny C, Dong L, Ang CS, Nikfarjam M, He H. Knockout of p21-Activated Kinase 4 Stimulates MHC I Expression of Pancreatic Cancer Cells via an Autophagy-Independent Pathway. Cancers (Basel) 2025; 17:511. [PMID: 39941877 PMCID: PMC11817421 DOI: 10.3390/cancers17030511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/26/2025] [Accepted: 02/01/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND/OBJECTIVES Pancreatic ductal adenocarcinoma (PDA) is one of the most malignant solid cancers. KRAS mutation accounts for over 90% of cases. p21-activated kinases (PAKs) act downstream of KRAS and are involved in tumorigenesis. The inhibition of PAK4 suppresses PDA by stimulating the tumor infiltration of cytotoxic T cells. The major histocompatibility complex class I (MHC I) is a key in presenting antigens to cytotoxic T cells. MHC I degradation via autophagy promotes the immune evasion of pancreatic cancer. We investigated the effect of PAK4 inhibition on MHC I expression and autophagy. METHODS In this study, using proteomic analysis, fluorescence-activated cell sorting (FACS), and immunoblotting, we examined the effect of PAK4 knockout (KO) in human PDA cells on the expression of MHC I and autophagy to identify the mechanism involved in the stimulation of cytotoxic T cells by PAK4 inhibition. RESULTS We found that PAK4 KO increased MHC I expression in two human PDA cell lines: MiaPaCa-2 and PANC-1. PAK4 KO also increased cancer cell autophagy. However, the inhibition of autophagy by chloroquine (CQ) did not affect the effect of PAK4 KO on apoptosis and cell death. More importantly, the inhibition of autophagy by CQ did not alter the expression of MHC I stimulated by PAK4 KO, indicating that PAK4 KO stimulated MHC I expression via an autophagy-independent pathway. CONCLUSIONS We identified a role of PAK4 in MHC I expression by PDA cells, which is independent of autophagy.
Collapse
Affiliation(s)
- Yi Ma
- Department of Surgery, Austin Precinct, University of Melbourne, Heidelberg, VIC 3084, Australia; (Y.M.); (C.D.); (L.D.); (M.N.)
- Department of General Surgery, Monash Health, Clayton, VIC 3806, Australia
| | - Chelsea Dumesny
- Department of Surgery, Austin Precinct, University of Melbourne, Heidelberg, VIC 3084, Australia; (Y.M.); (C.D.); (L.D.); (M.N.)
| | - Li Dong
- Department of Surgery, Austin Precinct, University of Melbourne, Heidelberg, VIC 3084, Australia; (Y.M.); (C.D.); (L.D.); (M.N.)
| | - Ching-Seng Ang
- Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC 3052, Australia;
| | - Mehrdad Nikfarjam
- Department of Surgery, Austin Precinct, University of Melbourne, Heidelberg, VIC 3084, Australia; (Y.M.); (C.D.); (L.D.); (M.N.)
- Department of Hepato-Pancreato-Biliary Surgery, Austin Health, Heidelberg, VIC 3084, Australia
| | - Hong He
- Department of Surgery, Austin Precinct, University of Melbourne, Heidelberg, VIC 3084, Australia; (Y.M.); (C.D.); (L.D.); (M.N.)
| |
Collapse
|
34
|
Manna T, Maji S, Maity M, Debnath B, Panda S, Khan SA, Nath R, Akhtar MJ. Anticancer potential and structure activity studies of purine and pyrimidine derivatives: an updated review. Mol Divers 2025; 29:817-848. [PMID: 38856835 DOI: 10.1007/s11030-024-10870-4] [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: 02/19/2024] [Accepted: 04/02/2024] [Indexed: 06/11/2024]
Abstract
Cancer is the world's leading cause of death impacting millions of lives globally. The increasing research over the past several decades has focused on the development of new anticancer drugs, but still cancer continues to be a global health challenge. Thus, several new alternative therapeutic strategies have been tried for the drug design and discovery. Purine and pyrimidine heterocyclic compounds have received attention recently due to their potential in targeting various cancers. It is evident from the recently published data over the last decade that incorporation of the purine and pyrimidine rings in the synthesized derivatives resulted in the development of potent anticancer molecules. This review presents synthetic strategies encompassing several examples of recently developed purine and pyrimidine-containing compounds as anticancer agents. In addition, their structure-activity relationships are represented in the schemes indicating the fragment or groups that are essential for the enhanced anticancer activities. Purine and pyrimidines combined with other heterocyclic compounds have resulted in many novel anticancer molecules that address the challenges of drug resistance. The purine and pyrimidine derivatives showed significantly enhanced anticancer activities against targeted receptor proteins with numerous compounds with an IC50 value in the nanomolar range. The review will support medicinal chemists and contribute in progression and development of synthesis of more potent chemotherapeutic drug candidates to mitigate the burden of this dreadful disease.
Collapse
Affiliation(s)
- Tanushree Manna
- Department of Pharmacy, Bharat Technology, Uluberia, 711316, Howrah, West Bengal, India
| | - Sumit Maji
- Department of Pharmacy, Bharat Technology, Uluberia, 711316, Howrah, West Bengal, India
| | - Mousumi Maity
- Department of Pharmacy, Bharat Technology, Uluberia, 711316, Howrah, West Bengal, India
| | - Biplab Debnath
- Department of Pharmacy, Bharat Technology, Uluberia, 711316, Howrah, West Bengal, India
| | - Shambo Panda
- Department of Pharmacy, Bharat Technology, Uluberia, 711316, Howrah, West Bengal, India
| | - Shah Alam Khan
- Department of Pharmaceutical Chemistry, National University of Science and Technology, PC 130, Azaiba, Bousher, PO 620, Muscat, Sultanate of Oman
| | - Rajarshi Nath
- Department of Pharmacy, Bharat Technology, Uluberia, 711316, Howrah, West Bengal, India.
- JIS University, Agarpara Campus, Kolkata-81, Nilgunj Road, Agarpara, Kolkata, 700109, India.
| | - Md Jawaid Akhtar
- Department of Pharmaceutical Chemistry, National University of Science and Technology, PC 130, Azaiba, Bousher, PO 620, Muscat, Sultanate of Oman.
| |
Collapse
|
35
|
Beutel AK, Ekizce M, Ettrich TJ, Seufferlein T, Lindenmayer J, Gout J, Kleger A. Organoid-based precision medicine in pancreatic cancer. United European Gastroenterol J 2025; 13:21-33. [PMID: 39540683 PMCID: PMC11866314 DOI: 10.1002/ueg2.12701] [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: 07/17/2024] [Accepted: 10/05/2024] [Indexed: 11/16/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) ranks among the leading causes of cancer-related deaths worldwide. Despite advances in precision oncology in other malignancies, treatment of PDAC still largely relies on conventional chemotherapy. Given the dismal prognosis and heterogeneity in PDAC, there is an urgent need for personalized therapeutic strategies to improve treatment response. Organoids, generated from patients' tumor tissue, have emerged as a powerful tool in cancer research. These three-dimensional models faithfully recapitulate the morphological and genetic features of the parental tumor and retain patient-specific heterogeneity. This review summarizes existing precision oncology approaches in PDAC, explores current applications and limitations of organoid cultures in personalized medicine, details preclinical studies correlating in vitro organoid prediction and patient treatment response, and provides an overview of ongoing organoid-based clinical trials.
Collapse
Affiliation(s)
- Alica K. Beutel
- Department of Internal Medicine IUniversity Hospital UlmUlmGermany
| | - Menar Ekizce
- Institute of Molecular Oncology and Stem Cell BiologyUlm University HospitalUlmGermany
| | | | | | | | - Johann Gout
- Institute of Molecular Oncology and Stem Cell BiologyUlm University HospitalUlmGermany
| | - Alexander Kleger
- Institute of Molecular Oncology and Stem Cell BiologyUlm University HospitalUlmGermany
- Core Facility OrganoidsMedical Faculty of Ulm UniversityUlmGermany
- Division of Interdisciplinary PancreatologyDepartment of Internal Medicine IUlm University HospitalUlmGermany
| |
Collapse
|
36
|
Zhan T, Betge J, Schulte N, Dreikhausen L, Hirth M, Li M, Weidner P, Leipertz A, Teufel A, Ebert MP. Digestive cancers: mechanisms, therapeutics and management. Signal Transduct Target Ther 2025; 10:24. [PMID: 39809756 PMCID: PMC11733248 DOI: 10.1038/s41392-024-02097-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 10/20/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
Cancers of the digestive system are major contributors to global cancer-associated morbidity and mortality, accounting for 35% of annual cases of cancer deaths. The etiologies, molecular features, and therapeutic management of these cancer entities are highly heterogeneous and complex. Over the last decade, genomic and functional studies have provided unprecedented insights into the biology of digestive cancers, identifying genetic drivers of tumor progression and key interaction points of tumor cells with the immune system. This knowledge is continuously translated into novel treatment concepts and targets, which are dynamically reshaping the therapeutic landscape of these tumors. In this review, we provide a concise overview of the etiology and molecular pathology of the six most common cancers of the digestive system, including esophageal, gastric, biliary tract, pancreatic, hepatocellular, and colorectal cancers. We comprehensively describe the current stage-dependent pharmacological management of these malignancies, including chemo-, targeted, and immunotherapy. For each cancer entity, we provide an overview of recent therapeutic advancements and research progress. Finally, we describe how novel insights into tumor heterogeneity and immune evasion deepen our understanding of therapy resistance and provide an outlook on innovative therapeutic strategies that will shape the future management of digestive cancers, including CAR-T cell therapy, novel antibody-drug conjugates and targeted therapies.
Collapse
Affiliation(s)
- Tianzuo Zhan
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Johannes Betge
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Junior Clinical Cooperation Unit Translational Gastrointestinal Oncology and Preclinical Models, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nadine Schulte
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lena Dreikhausen
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Michael Hirth
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Moying Li
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Philip Weidner
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Antonia Leipertz
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Andreas Teufel
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Matthias P Ebert
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany.
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| |
Collapse
|
37
|
Álvarez-Gallego R, Pazo-Cid R, López de San Vicente B, Macarulla T, Martinez E, Garicano F, Hernández I, Granja M, Ghanem I, Martinez J, Ribera P, Diaz R, Martin Valadés JI, Angeles MC, Cubillo A. Real-world effectiveness and safety of second- or third-line pegylated liposomal irinotecan plus 5-fluorouracil and folinic acid in pancreatic ductal adenocarcinoma in Spain. Ther Adv Med Oncol 2025; 17:17588359241309828. [PMID: 39781240 PMCID: PMC11707772 DOI: 10.1177/17588359241309828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 12/10/2024] [Indexed: 01/12/2025] Open
Abstract
Treatment with pegylated nanoliposomal irinotecan (nal-IRI) plus 5-fluorouracil/leucovorin (folinic acid; 5-FU/LV) has demonstrated remarkable efficacy for metastatic pancreatic ductal adenocarcinoma (PDAC) in clinical trials. However, real-world data on the effectiveness of nal-IRI+5-FU/LV is heterogeneous and is lacking in Spain. To assess the effectiveness and safety of nal-IRI+5-FU/LV in real-life PDAC patients in Spain. A multicenter retrospective study was conducted. Patients aged ⩾18 years who had received at least one cycle of nal-IRI+5-FU/LV as second- or third-line therapy for PDAC were included. The primary endpoint was overall survival (OS) from nal-IRI+5-FU/LV treatment initiation and OS from the diagnosis of metastatic disease (metOS). Overall, 200 evaluable patients were included (⩾3 metastatic sites: 22%; liver/lung metastases: 71.5%/36.9%; and Eastern Cooperative Oncology Group 0-1: 87% at nal-IRI+5FU/LV treatment initiation). Patients received a median of four cycles of nal-IRI+5FU/LV for 2.8 months (range 1.4-7.2), and the treatment was received in the second line by 80% of the patients. The median OS was 7.2 months (6- and 12-month OS rates: 58.1% and 28.9%, respectively), with 27.2% of the patients achieving OS ⩾12 months. The median metOS was 17.5 months, with 30.2% of the patients experiencing metOS ⩾ 24 months. The median progression-free survival (PFS) was 3.7 months (6- and 12-month PFS rate: 37.6% and 15.3%, respectively). The disease control rate was 35.5%. The median CA 19-9 levels decreased by at least 50% in 28.2% of the cases during treatment. Overall, 36% of the patients experienced at least one grade 3-4 adverse event during treatment, the most common being diarrhea (42.6%) and asthenia (30.9%). This real-world study shows that treatment with nal-IRI+5-FU/LV for advanced or metastatic PDAC affords benefit in terms of survival, radiological and CA 19-9 response, and PFS comparable to that reported in the clinical trial setting with a manageable safety profile.
Collapse
Affiliation(s)
- Rafael Álvarez-Gallego
- Centro Integral Oncológico Clara Campal HM CIOCC, Hospital Universitario HM Sanchinarro, C\ Oña Nº10, Madrid 28050, Spain
- Facultad HM Hospitales de Ciencias de la Salud, Universidad Camilo José Cela (UCJC), Madrid, Spain
| | | | | | - Teresa Macarulla
- Vall d’Hebrón Institute of Oncology (VHIO), Hospital Universitario Vall d’Hebrón, Barcelona, Spain
| | - Eva Martinez
- Hospital Universitario Marques de Valdecilla, Santander, Spain
| | | | | | | | | | - Joaquina Martinez
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Paula Ribera
- Hospital Universitario Parc Tauli, Sabadell, Barcelona, Spain
| | - Roberto Diaz
- Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | | | | | - Antonio Cubillo
- Centro Integral Oncológico Clara Campal HM CIOCC, Hospital Universitario HM Sanchinarro, Madrid, Spain
- Facultad HM Hospitales de Ciencias de la Salud, Universidad Camilo José Cela (UCJC), Madrid, Spain
| |
Collapse
|
38
|
Cai Z, Wuri Q, Song Y, Qu X, Hu H, Cao S, Wu H, Wu J, Wang C, Yu X, Kong W, Zhang H. CircRNA-loaded DC vaccine in combination with low-dose gemcitabine induced potent anti-tumor immunity in pancreatic cancer model. Cancer Immunol Immunother 2025; 74:68. [PMID: 39751874 PMCID: PMC11699015 DOI: 10.1007/s00262-024-03924-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025]
Abstract
Although promising, dendritic cell (DC) vaccines may not suffice to fully inhibit tumor progression alone, mainly due to the short expression time of the antigen in DC vaccines, immunosuppressive tumor microenvironment, and tumor antigenic modulation. Overcoming the limitations of DC vaccines is expected to further enhance their anti-tumor effects. In this study, we constructed a circRNA-loaded DC vaccine utilizing the inherent stability of circular RNA to enhance the expression level and duration of the antigen within the DC vaccine. Meanwhile we combined it with gemcitabine and validated their therapeutic efficacy in the Panc02 tumor model. We found that the use of DC vaccine alone can reach a tumor inhibition rate of 69%, and the effect was further enhanced when combined with gemcitabine, reaching a tumor inhibition rate of 89%. The combined treatment achieved a synergistic effect, which not only reduced immunosuppressive Tregs but also induced immunogenic cell death, leading to antigen spreading and reducing immune evasion caused by tumor antigenic modulation. As a result, the survival of the mice was significantly prolonged. Our research provides a promising approach for the clinical treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Zongyu Cai
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China
| | - Qimuge Wuri
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China
| | - Yang Song
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China
| | - Xueli Qu
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China
| | - Haotong Hu
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China
| | - Simiao Cao
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China
| | - Hui Wu
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China
| | - Jiaxin Wu
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China
| | - Chu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China
| | - Xianghui Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China
- Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China
- Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Haihong Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China.
| |
Collapse
|
39
|
Aseafan M, Alfakeeh AH, Tashkandi E, Mahrous M, Alghamdi M, Alshamsan B, Al-Hajeili M, Bakhsh S, Alshammari K, Almugbel FA, Alfagih AH, Allehebi A, Montaser M, Elsafty MH, Elnaghi KAE, Issa I, Bakshi E, AlSubaie S, AlMutairi B, Mokhtar H, Aboelatta M, Bukhari N, Alzahrani AM, Elhassan T, Alqahtani A, Bazarbashi S. Real-world clinical outcome of unresectable locally advanced & de-novo metastatic pancreatic ductal adenocarcinoma: a multicentre retrospective study. BMC Cancer 2025; 25:7. [PMID: 39754118 PMCID: PMC11697791 DOI: 10.1186/s12885-024-13386-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 12/23/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal malignancies, with limited treatment options yielding poor outcomes. This study aimed to evaluate the real-world clinical characteristics, treatment patterns, and outcomes of patients with locally advanced unresectable and de-novo metastatic PDAC in Saudi Arabia, providing regional data to compare with international benchmarks. METHODS This is a retrospective, multicentre study involving 350 patients diagnosed with unresectable locally advanced or de-novo metastatic PDAC between January 2015 and November 2023. Data were collected from 10 oncology centers across Saudi Arabia. RESULTS The median age at diagnosis was 60 years, with 63% of patients presenting with multiple metastatic sites, primarily in the liver (66.3%). FOLFIRINOX was the most common first-line treatment (55.1%), followed by gemcitabine plus nab-paclitaxel (15.1%). The median PFS for first-line treatment was 5.3 months, with FOLFIRINOX achieving the longest PFS (6.5 months). The median OS was 10.34 months for the entire cohort, with better survival outcomes observed in patients receiving FOLFIRINOX (12.3 months). Independent prognostic factors for PFS and OS included performance status, first-line regimen, and neutrophil-lymphocyte ratio (NLR). Among patients tested, 7.1% had deficient mismatch repair (d-MMR), and 5.8% harbored BRCA mutations. CONCLUSIONS This real-world study confirms that clinical outcomes for locally advanced unresectable and metastatic PDAC in Saudi Arabia are consistent with international data, with FOLFIRINOX showing superior outcomes over gemcitabine-based regimens. However, both treatments reflect the persistent poor prognosis of PDAC, underscoring the need for novel therapeutic strategies. Further research is warranted to optimize treatment selection and improve survival outcomes in this population.
Collapse
Affiliation(s)
- Mohamed Aseafan
- Section of Medical Oncology, Department of Internal Medicine, Security Forces Hospital, Riyadh, Saudi Arabia.
| | - Ali H Alfakeeh
- Comprehensive Cancer Center, Medical Oncology, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Emad Tashkandi
- College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mervat Mahrous
- Department of Oncology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
- Collage of Medicine, Minia University, Minia, Egypt
| | - Mohammed Alghamdi
- Oncology Center, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Bader Alshamsan
- Department of Medicine, College of Medicine, Qassim University, Buraydah, Saudi Arabia
- Prince Faisal Cancer Center, King Fahad Specialist Hospital, Qassim Health Clusster, Buraydah, Saudi Arabia
| | - Marwan Al-Hajeili
- Department of Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Safwan Bakhsh
- Department of Medical Oncology, King Faisal Specialist Hospital and Research Center-Jeddah, Jeddah, Saudi Arabia
| | - Kanan Alshammari
- Department of Medical Oncology, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Fahad A Almugbel
- Department of Medical Oncology, Cancer Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Abdulhameed H Alfagih
- Comprehensive Cancer Center, Medical Oncology, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Ahmed Allehebi
- Department of Medical Oncology, King Faisal Specialist Hospital and Research Center-Jeddah, Jeddah, Saudi Arabia
| | - Mohamed Montaser
- Section of Medical Oncology, Department of Internal Medicine, Security Forces Hospital, Riyadh, Saudi Arabia
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | | | - Ibrahim Issa
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Eesa Bakshi
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Sadeem AlSubaie
- Pathology and Laboratory Medicine, Security Forces Hospital, Riyadh, Saudi Arabia
| | - Bandar AlMutairi
- Department of Oncology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Hoda Mokhtar
- Department of Oncology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Mohamed Aboelatta
- Prince Faisal Cancer Center, King Fahad Specialist Hospital, Qassim Health Clusster, Buraydah, Saudi Arabia
| | - Nedal Bukhari
- Department of Oncology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Ali M Alzahrani
- Comprehensive Cancer Center, Medical Oncology, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Tusneem Elhassan
- Research Unit, Cancer Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Ali Alqahtani
- Department of Medical Oncology, Cancer Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Shouki Bazarbashi
- Department of Medical Oncology, Cancer Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
40
|
Lee W, Song G, Bae H. In vitro and in silico study of the synergistic anticancer effect of alpinumisoflavone with gemcitabine on pancreatic ductal adenocarcinoma through suppression of ribonucleotide reductase subunit-M1. Eur J Pharm Sci 2025; 204:106969. [PMID: 39577749 DOI: 10.1016/j.ejps.2024.106969] [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: 07/12/2024] [Revised: 10/21/2024] [Accepted: 11/20/2024] [Indexed: 11/24/2024]
Abstract
A highly aggressive neoplastic disease, pancreatic ductal adenocarcinoma (PDAC) is documented as the third chief cause of cancer-associated mortality in both sexes combined in the United States. For decades, gemcitabine-based chemotherapy has been embraced as a cornerstone drug for the treatment of PDAC. However, there have been several unsolved problems, including cytotoxicity, and chemoresistance. Gemcitabine efficacy was attributed to the attenuation of ribonucleotide reductase subunit-M1 (RRM1). Overexpression of RRM1 in PDAC is highly correlated with gemcitabine resistance and reduced gemcitabine sensitivity, resulting in a poor survival rate even after gemcitabine treatment. Moreover, the status of TP53, a tumor suppressor gene, assumes a decisive role in the response of PDAC to gemcitabine. Therefore, targeting RRM1 and P53 might be a therapeutic strategy for strengthening gemcitabine efficacy and cytotoxicity against PDAC. Alpinumisoflavone (AIF) is a prenylated isoflavone originated in Cudrania tricuspidate with versatile bioactive properties, including anticancer activity. However, there was no report whether AIF can exert anticancer effect and exhibit synergistic effect with gemcitabine against PDAC. Therefore, the anticancer properties of AIF were assessed with PANC-1 and MIA PaCa-2. In addition, synergism between AIF and gemcitabine were analyzed. Moreover, the contribution of P53 and RRM1 expression to gemcitabine resistance was assessed by comparing their protein levels in PDAC cells and normal pancreatic cells. The interactions of AIF with RRM1 protein were confirmed by molecular docking and dynamics simulation. Therefore, AIF enhances gemcitabine efficacy against PDAC through the regulation of P53 and RRM1.
Collapse
Affiliation(s)
- Woonghee Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, South Korea
| | - Gwonhwa Song
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, South Korea.
| | - Hyocheol Bae
- Department of Oriental Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, South Korea.
| |
Collapse
|
41
|
Lai X, Cheng D, Xu H, Wang J, Lv X, Yao H, Li L, Wu J, Ye S, Li Z. Phase I Trial of Upamostat Combined With Gemcitabine in Locally Unresectable or Metastatic Pancreatic Cancer: Safety and Preliminary Efficacy Assessment. Cancer Med 2025; 14:e70550. [PMID: 39739976 DOI: 10.1002/cam4.70550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 01/02/2025] Open
Abstract
AIM This study aimed to determine the maximum tolerated dose (MTD) of the urokinase plasminogen activator (uPA) inhibitor upamostat (LH011) in combination with gemcitabine for locally advanced unresectable or metastatic pancreatic cancer. METHOD Seventeen patients were enrolled and received escalating doses of oral LH011 (100, 200, 400, or 600 mg) daily alongside 1000 mg/m2 of gemcitabine. Safety profiles, tumor response (including response rate and progression-free survival), pharmacokinetics, and changes in CA199 and D-dimer levels were assessed. RESULTS During the study period (Day0-Day49), no patients achieved partial response. Stable disease (SD) was observed in 12 patients (70.6%), while four patients (23.5%) experienced progressive disease (PD). One patient withdrew due to a serious adverse event (SAE) on D47. Pharmacokinetic analysis revealed a dose-related increase in LH011 and its metabolite WX-UK1 exposure from 100 to 400 mg but not in the 600 mg group. Hematological toxicity, mainly attributable to gemcitabine, was the predominant grade 3 or 4 adverse event, with additional occurrences of loss of appetite, rash, and interstitial lung disease. Sinus bradycardia possibly linked to LH011 rather than gemcitabine was noted. The MTD was not reached. CONCLUSION Combining LH011 at doses ranging from 100 to 600 mg with gemcitabine every 21 days demonstrated manageable safety and tolerability. However, tumor response did not significantly differ among the dose groups, suggesting the need for further investigation. TRIAL REGISTRATION NCT05329597.
Collapse
Affiliation(s)
- Xiuping Lai
- Phase I Clinical Trial Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Di Cheng
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huixin Xu
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jingshu Wang
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaozhi Lv
- Phase I Clinical Trial Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Herui Yao
- Phase I Clinical Trial Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liuning Li
- Department of Medical Oncology, GuangDong Province Hospital of Chinese Medicine, the Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, China
| | - Junyan Wu
- Phase I Clinical Trial Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Suiwen Ye
- Phase I Clinical Trial Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhihua Li
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
42
|
Kim YJ, Lee HS, Kim D, Byun HK, Koom WS, Koh W. Bilayer 3D co-culture platform inducing the differentiation of normal fibroblasts into cancer-associated fibroblast like cells: New in vitro source to obtain cancer-associated fibroblasts. Bioeng Transl Med 2025; 10:e10708. [PMID: 39801758 PMCID: PMC11711222 DOI: 10.1002/btm2.10708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 06/08/2024] [Accepted: 07/23/2024] [Indexed: 01/16/2025] Open
Abstract
This study presents a novel in vitro bilayer 3D co-culture platform designed to obtain cancer-associated fibroblasts (CAFs)-like cells. The platform consists of a bilayer hydrogel structure with a collagen/polyethylene glycol (PEG) hydrogel for fibroblasts as the upper layer and an alginate hydrogel for tumor cells as the lower layer. The platform enabled paracrine interactions between fibroblasts and cancer cells, which allowed for selective retrieval of activated fibroblasts through collagenase treatment for further study. Fibroblasts remained viable throughout the culture periods and showed enhanced proliferation when co-cultured with cancer cells. Morphological changes in the co-cultured fibroblasts resembling CAFs were observed, especially in the 3D microenvironment. The mRNA expression levels of CAF-related markers were significantly upregulated in 3D, but not in 2D co-culture. Proteomic analysis identified upregulated proteins associated with CAFs, further confirming the transformation of normal fibroblasts into CAF within the proposed 3D co-culture platform. Moreover, co-culture with CAF induced radio- and chemoresistance in pancreatic cancer cells (PANC-1). Survival rate of cancer cells post-irradiation and gemcitabine resistance increased significantly in the co-culture setting, highlighting the role of CAFs in promoting cancer cell survival and therapeutic resistance. These findings would contribute to understanding molecular and phenotypic changes associated with CAF activation and provide insights into potential therapeutic strategies targeting the tumor microenvironment.
Collapse
Affiliation(s)
- Yeon Ju Kim
- Department of Radiation Oncology, Yonsei Cancer CenterYonsei University College of MedicineSeoulSouth Korea
| | - Hyeon Song Lee
- Department of Chemical and Biomolecular EngineeringYonsei UniversitySeoulSouth Korea
| | - Dohyun Kim
- Department of Chemical and Biomolecular EngineeringYonsei UniversitySeoulSouth Korea
| | - Hwa Kyung Byun
- Department of Radiation Oncology, Yongin Severance HospitalYonsei University College of MedicineYonginSouth Korea
| | - Woong Sub Koom
- Department of Radiation Oncology, Yonsei Cancer CenterYonsei University College of MedicineSeoulSouth Korea
| | - Won‐Gun Koh
- Department of Chemical and Biomolecular EngineeringYonsei UniversitySeoulSouth Korea
| |
Collapse
|
43
|
Cockrum P, Dennen S, Brown A, Briggs J, Paluri R. Real-world clinical outcomes and economic burden of metastatic pancreatic ductal adenocarcinoma: a systematic review. Future Oncol 2025; 21:241-260. [PMID: 39648649 PMCID: PMC11792790 DOI: 10.1080/14796694.2024.2435253] [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: 08/02/2024] [Accepted: 11/25/2024] [Indexed: 12/10/2024] Open
Abstract
AIMS This systematic review summarizes real-world clinical outcomes and economic burden of first-line FOLFIRINOX (FFX)/modified FFX (mFFX) and nab-paclitaxel plus gemcitabine (GnP) in metastatic pancreatic ductal adenocarcinoma in the US. METHODS Embase and MEDLINE were searched for materials published since 2014; citations were reviewed in a two-step process. Included studies were qualitatively synthesized. RESULTS Searches yielded 2,528 citations; 29 were included (17 clinical studies/12 economic studies). In 9/17 clinical studies, median overall survival (mOS) ranged from 4.7 months to 11.4 months for FFX/mFFX, with the unweighted median of the estimates within this range being 9.2 months; for GnP mOS ranged from 3.6 to 9.8 months, and the unweighted median of the estimates was 6.9 months. In 8/17 studies, grade 3/4 anemia, neutropenia, and thrombocytopenia were the most commonly reported adverse events. Across economic burden studies, total costs were similar between the 2 groups. Outpatient, supportive care, and granulocyte colony-stimulating factor costs were higher for the FFX generic regimen, and chemotherapy costs were higher for the GnP branded regimen. CONCLUSIONS Real-world OS in FFX- and GnP-treated populations was shorter than that in clinical trials, and total costs of FFX and GnP were similar, but with differences in cost components.
Collapse
Affiliation(s)
- Paul Cockrum
- Medical Affairs, Ipsen Biopharmaceuticals Inc, Cambridge, MA, USA
| | | | | | | | - Ravi Paluri
- Hematology and Oncology, Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA
| |
Collapse
|
44
|
Finan JM, Di Niro R, Park SY, Jeong KJ, Hedberg MD, Smith A, McCarthy GA, Haber AO, Muschler J, Sears RC, Mills GB, Gmeiner WH, Brody JR. The polymeric fluoropyrimidine CF10 overcomes limitations of 5-FU in pancreatic ductal adenocarcinoma cells through increased replication stress. Cancer Biol Ther 2024; 25:2421584. [PMID: 39513592 PMCID: PMC11552260 DOI: 10.1080/15384047.2024.2421584] [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: 08/20/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 11/15/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease soon to become the second leading cause of cancer deaths in the US. Beside surgery, current therapies have narrow clinical benefits with systemic toxicities. FOLFIRINOX is the current standard of care, one component of which is 5- Fluorouracil (5-FU), which causes serious gastrointestinal and hematopoietic toxicities and is vulnerable to resistance mechanisms. Recently, we have developed polymeric fluoropyrimidines (F10, CF10) which unlike 5-FU, are, in principle, completely converted to the thymidylate synthase inhibitory metabolite FdUMP, without generating appreciable levels of ribonucleotides that cause systemic toxicities while displaying much stronger anti-cancer activity. Here, we confirm the potency of CF10 and investigate enhancement of its efficacy through combination with inhibitors in vitro targeting replication stress, a hallmark of PDAC cells. CF10 is 308-times more potent as a single agent than 5-FU and was effective in the nM range in primary patient derived models. Further, we find that activity of CF10, but not 5-FU, is enhanced through combination with inhibitors of ATR and Wee1 that regulate the S and G2 DNA damage checkpoints and can be reversed by addition of dNTPs indicative of CF10 acting, at least in part, through inducing replication stress. Our results indicate CF10 has the potential to supersede the established benefit of 5-FU in PDAC treatment and indicate novel combination approaches that should be validated in vivo and may be beneficial in established regimens that include 5-FU.
Collapse
Affiliation(s)
- Jennifer M. Finan
- Department of Surgery, Oregon Health & Science University, Portland, OR, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Cell, Development and Cancer Biology, Oregon Health and Sciences University, Portland, OR, USA
| | - Roberto Di Niro
- Department of Surgery, Oregon Health & Science University, Portland, OR, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Soon Young Park
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Cell, Development and Cancer Biology, Oregon Health and Sciences University, Portland, OR, USA
- Division of Oncological Sciences, Oregon Health & Science University, Portland, OR, USA
| | - Kang Jin Jeong
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Cell, Development and Cancer Biology, Oregon Health and Sciences University, Portland, OR, USA
- Division of Oncological Sciences, Oregon Health & Science University, Portland, OR, USA
| | - Madeline D. Hedberg
- Department of Surgery, Oregon Health & Science University, Portland, OR, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Alexander Smith
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Grace A. McCarthy
- Department of Surgery, Oregon Health & Science University, Portland, OR, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Cell, Development and Cancer Biology, Oregon Health and Sciences University, Portland, OR, USA
| | - Alex O. Haber
- The Jefferson Pancreas, Biliary and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - John Muschler
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Rosalie C. Sears
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | - Gordon B. Mills
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Cell, Development and Cancer Biology, Oregon Health and Sciences University, Portland, OR, USA
| | - William H. Gmeiner
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jonathan R. Brody
- Department of Surgery, Oregon Health & Science University, Portland, OR, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
45
|
Xiang Z, Ma L, Li Z, Fu Y, Pan Y. Cost-effectiveness analysis of first-line combination chemotherapy regimens for metastatic pancreatic cancer and evidence-based pricing strategy of liposomal irinotecan in China. Front Pharmacol 2024; 15:1488645. [PMID: 39759454 PMCID: PMC11695189 DOI: 10.3389/fphar.2024.1488645] [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: 08/30/2024] [Accepted: 12/04/2024] [Indexed: 01/07/2025] Open
Abstract
Background The phase III NAPOLI-3 trial, which upgraded FOLFIRINOX (leucovorin, fluorouracil, irinotecan and oxaliplatin) to NALIRIFOX (liposomal irinotecan, oxaliplatin, leucovorin, and fluorouracil), demonstrated the superiority of NALIRIFOX over GEMNABP (gemcitabine and nab-paclitaxel) as the first-line treatment for metastatic pancreatic ductal adenocarcinoma. The purpose of this study was to assess the cost-effectiveness of NALIRIFOX, FOLFIRINOX, and GEMNABP, and to simulate the price of liposomal irinotecan at which NALIRIFOX could achieve cost-effectiveness. Methods A partitioned survival model was performed to evaluate the cost-effectiveness of NALIRIFOX, FOLFIRINOX and GEMNABP from the perspective of the Chinese healthcare system. Survival data was obtained from a recently published network meta-analysis (NMA). Drug prices were collected from the database of the Hunan Province Drug and Medical Consumables Procurement Management Subsystem. Other cost and utility values were sourced from established literature. Cumulative costs, LYs (life-years), quality-adjusted life years (QALYs), incremental cost-effectiveness ratios (ICERs), net monetary benefits (NMBs) and incremental net monetary benefits (INMBs) were the main outputs. Furthermore, the variations in ICER were analyzed as the price of liposomal irinotecan gradually decreased when comparing NALIRIFOX with FOLFIRINOX or GEMNABP. The robustness of the model was assessed by sensitivity analysis and scenario analysis. Results At the willingness-to-pay (WTP) threshold of $38,223.34, GEMNABP was the favored treatment. NALIRIFOX was associated with the highest LYs, QALYs, and cost. The cost-effectiveness of NALIRIFOX would be obtained if the price of liposomal irinotecan was less than $3.36/mg and $2.08/mg compared to FOLFIRINOX and GEMNABP, respectively, without considering the patient assistance program (PAP). Sensitivity analysis and scenario analysis revealed that the results of the model were stable. Conclusion From an economic standpoint, GEMNABP represents the favored choice in the prevailing market conditions among these three first-line combination chemotherapy regimens. The price simulation of liposomal irinotecan conducted in this study could provide valuable evidence for healthcare decision-making. Further evidence regarding the budget impact is still needed.
Collapse
Affiliation(s)
- Zuojuan Xiang
- Department of Pharmacy, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| | - Ling Ma
- Department of Clinical pharmacy, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Zhengxiong Li
- School of Medical Informatics and Engineering, Xuzhou Medical University, Xuzhou, China
| | - Yingzhou Fu
- Department of Pharmacy, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| | - Yong Pan
- Department of Pharmacy, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| |
Collapse
|
46
|
Mizutani S, Taniai N, Sukegawa M, Haruna T, Furuki H, Takata H, Ueda J, Yoshioka M, Aimoto T, Sakamoto S, Suzuki K, Nakamura Y, Yoshida H. Pancreatectomy with Celiac Axis Resection and Reconstruction for Locally Advanced Pancreatic Cancer. Cancers (Basel) 2024; 16:4115. [PMID: 39682301 DOI: 10.3390/cancers16234115] [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: 11/18/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND With the advent of effective chemotherapy, conversion surgery (CS) has been performed in patients who have responded to pretreatment, even for pancreatic cancer diagnosed as unresectable (UR) at the time of initial diagnosis. In CS, major arterial resection and reconstruction are necessary for complete radical resection. METHODS We discuss the key points for safely performing pancreatectomy with celiac axis (CA) resection combined with reconstruction, divided into resection and arterial reconstruction. The possibility of safe pancreatectomy concurrent with CA resection and reconstruction depends on the ability to create a "golden view" that provides an unimpaired view of the Abdominal Aorta, CA, Superior Mesenteric Artery, Inferior Vena Cava, and left renal vein from the ventral side. Pancreatectomy concurrent with CA resection requires arterial reconstruction. Postoperatively, arterial blood flow must be maintained. To achieve this, tension-free and short bypass should be observed. RESULTS From 2014 to 2024, sixteen URLA patients underwent CS, requiring major artery en bloc resection after pretreatment. We performed DP-CAR in eight patients, gastrectomy-distal pancreatectomy-splenectomy (Appleby procedure) procedure in one patient, PD-CHAR in two patients, PD-CAR in two patients, TP-CAR(spleen preserving) in one patient, and TP-CAR+TG in two patients. In total, five patients required surgery with CA reconstruction. Histopathologically, four of the five patients had T4 pancreatic cancer. The R0 surgical rate was 80%. Complication of Clavien-Dindo IIIa or higher was observed in one patient. There were no deaths. CONCLUSIONS Parallel to the determination of pretreatment, surgeons must be prepared to safely and reliably perform pancreatectomies that require concurrent major arterial resection and reconstruction.
Collapse
Affiliation(s)
- Satoshi Mizutani
- Digestive Surgery, Nippon Medical School Musashikosugi Hospital, 1-383 Kosugimachi, Nakahara, Kawasaki 211-8533, Kanagawa, Japan
| | - Nobuhiko Taniai
- Digestive Surgery, Nippon Medical School Musashikosugi Hospital, 1-383 Kosugimachi, Nakahara, Kawasaki 211-8533, Kanagawa, Japan
| | - Makoto Sukegawa
- Digestive Surgery, Nippon Medical School Musashikosugi Hospital, 1-383 Kosugimachi, Nakahara, Kawasaki 211-8533, Kanagawa, Japan
| | - Takahiro Haruna
- Digestive Surgery, Nippon Medical School Musashikosugi Hospital, 1-383 Kosugimachi, Nakahara, Kawasaki 211-8533, Kanagawa, Japan
| | - Hiroyasu Furuki
- Digestive Surgery, Nippon Medical School Musashikosugi Hospital, 1-383 Kosugimachi, Nakahara, Kawasaki 211-8533, Kanagawa, Japan
| | - Hideyuki Takata
- Digestive Surgery, Nippon Medical School Musashikosugi Hospital, 1-383 Kosugimachi, Nakahara, Kawasaki 211-8533, Kanagawa, Japan
| | - Junji Ueda
- Digestive Surgery, Nippon Medical School Musashikosugi Hospital, 1-383 Kosugimachi, Nakahara, Kawasaki 211-8533, Kanagawa, Japan
| | - Masato Yoshioka
- Digestive Surgery, Nippon Medical School Musashikosugi Hospital, 1-383 Kosugimachi, Nakahara, Kawasaki 211-8533, Kanagawa, Japan
| | - Takayuki Aimoto
- Digestive Surgery, Nippon Medical School Musashikosugi Hospital, 1-383 Kosugimachi, Nakahara, Kawasaki 211-8533, Kanagawa, Japan
| | - Shunichiro Sakamoto
- Department of Cardiovascular Surgery, Nippon Medical School Musashikosugi Hospital, 1-383 Kosugimachi, Nakahara, Kawasaki 211-8533, Kanagawa, Japan
| | - Kenji Suzuki
- Department of Cardiovascular Surgery, Nippon Medical School Musashikosugi Hospital, 1-383 Kosugimachi, Nakahara, Kawasaki 211-8533, Kanagawa, Japan
| | - Yoshiharu Nakamura
- Department of Surgery, Nippon Medical School Chiba Hokusoh Hospital, 1715 Kamakari, Inzai 270-1694, Chiba, Japan
| | - Hiroshi Yoshida
- Department of Surgery, Nippon Medical School, 1-1-5 Sendagi, Bunkyo, Tokyo 113-8603, Japan
| |
Collapse
|
47
|
Haggstrom L, Chan WY, Nagrial A, Chantrill LA, Sim HW, Yip D, Chin V. Chemotherapy and radiotherapy for advanced pancreatic cancer. Cochrane Database Syst Rev 2024; 12:CD011044. [PMID: 39635901 PMCID: PMC11619003 DOI: 10.1002/14651858.cd011044.pub3] [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] [Indexed: 12/07/2024]
Abstract
BACKGROUND Pancreatic cancer (PC) is a lethal disease with few effective treatment options. Many anti-cancer therapies have been tested in the locally advanced and metastatic setting, with mixed results. This review synthesises all the randomised data available to help better inform patient and clinician decision-making. It updates the previous version of the review, published in 2018. OBJECTIVES To assess the effects of chemotherapy, radiotherapy, or both on overall survival, severe or life-threatening adverse events, and quality of life in people undergoing first-line treatment of advanced pancreatic cancer. SEARCH METHODS We searched for published and unpublished studies in CENTRAL, MEDLINE, Embase, and CANCERLIT, and handsearched various sources for additional studies. The latest search dates were in March and July 2023. SELECTION CRITERIA We included randomised controlled trials comparing chemotherapy, radiotherapy, or both with another intervention or best supportive care. Participants were required to have locally advanced, unresectable pancreatic cancer or metastatic pancreatic cancer not amenable to curative intent treatment. Histological confirmation was required. Trials were required to report overall survival. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane. MAIN RESULTS We included 75 studies in the review and 51 in the meta-analysis (11,333 participants). We divided the studies into seven categories: any anti-cancer treatment versus best supportive care; various chemotherapy types versus gemcitabine; gemcitabine-based combinations versus gemcitabine alone; various chemotherapy combinations versus gemcitabine plus nab-paclitaxel; fluoropyrimidine-based studies; miscellaneous studies; and radiotherapy studies. In general, the included studies were at low risk for random sequence generation, detection bias, attrition bias, and reporting bias, at unclear risk for allocation concealment, and high risk for performance bias. Compared to best supportive care, chemotherapy likely results in little to no difference in overall survival (OS) (hazard ratio (HR) 1.08, 95% confidence interval (CI) 0.88 to 1.33; absolute risk of death at 12 months of 971 per 1000 versus 962 per 1000; 4 studies, 298 participants; moderate-certainty evidence). The adverse effects of chemotherapy and impacts on quality of life (QoL) were uncertain. Many of the chemotherapy regimens were outdated. Eight studies compared non-gemcitabine-based chemotherapy regimens to gemcitabine. These showed that 5-fluorouracil (5FU) likely reduces OS (HR 1.69, 95% CI 1.26 to 2.27; risk of death at 12 months of 914 per 1000 versus 767 per 1000; 1 study, 126 participants; moderate certainty), and grade 3/4 adverse events (QoL not reported). Fixed dose rate gemcitabine likely improves OS (HR 0.79, 95% CI 0.66 to 0.94; risk of death at 12 months of 683 per 1000 versus 767 per 1000; 2 studies, 644 participants; moderate certainty), and likely increase grade 3/4 adverse events (QoL not reported). FOLFIRINOX improves OS (HR 0.51, 95% CI 0.43 to 0.60; risk of death at 12 months of 524 per 1000 versus 767 per 1000; P < 0.001; 2 studies, 652 participants; high certainty), and delays deterioration in QoL, but increases grade 3/4 adverse events. Twenty-eight studies compared gemcitabine-based combinations to gemcitabine. Gemcitabine plus platinum may result in little to no difference in OS (HR 0.94, 95% CI 0.81 to 1.08; risk of death at 12 months of 745 per 1000 versus 767 per 1000; 6 studies, 1140 participants; low certainty), may increase grade 3/4 adverse events, and likely worsens QoL. Gemcitabine plus fluoropyrimidine improves OS (HR 0.88, 95% CI 0.81 to 0.95; risk of death at 12 months of 722 per 1000 versus 767 per 1000; 10 studies, 2718 participants; high certainty), likely increases grade 3/4 adverse events, and likely improves QoL. Gemcitabine plus topoisomerase inhibitors result in little to no difference in OS (HR 1.01, 95% CI 0.87 to 1.16; risk of death at 12 months of 770 per 1000 versus 767 per 1000; 3 studies, 839 participants; high certainty), likely increases grade 3/4 adverse events, and likely does not alter QoL. Gemcitabine plus taxane result in a large improvement in OS (HR 0.71, 95% CI 0.62 to 0.81; risk of death at 12 months of 644 per 1000 versus 767 per 1000; 2 studies, 986 participants; high certainty), and likely increases grade 3/4 adverse events and improves QoL. Nine studies compared chemotherapy combinations to gemcitabine plus nab-paclitaxel. Fluoropyrimidine-based combination regimens improve OS (HR 0.79, 95% CI 0.70 to 0.89; risk of death at 12 months of 542 per 1000 versus 628 per 1000; 6 studies, 1285 participants; high certainty). The treatment arms had distinct toxicity profiles, and there was little to no difference in QoL. Alternative schedules of gemcitabine plus nab-paclitaxel likely result in little to no difference in OS (HR 1.10, 95% CI 0.82 to 1.47; risk of death at 12 months of 663 per 1000 versus 628 per 1000; 2 studies, 367 participants; moderate certainty) or QoL, but may increase grade 3/4 adverse events. Four studies compared fluoropyrimidine-based combinations to fluoropyrimidines alone, with poor quality evidence. Fluoropyrimidine-based combinations are likely to result in little to no impact on OS (HR 0.84, 95% CI 0.61 to 1.15; risk of death at 12 months of 765 per 1000 versus 704 per 1000; P = 0.27; 4 studies, 491 participants; moderate certainty) versus fluoropyrimidines alone. The evidence suggests that there was little to no difference in grade 3/4 adverse events or QoL between the two groups. We included only one radiotherapy (iodine-125 brachytherapy) study with 165 participants. The evidence is very uncertain about the effect of radiotherapy on outcomes. AUTHORS' CONCLUSIONS Combination chemotherapy remains standard of care for metastatic pancreatic cancer. Both FOLFIRINOX and gemcitabine plus a taxane improve OS compared to gemcitabine alone. Furthermore, the evidence suggests that fluoropyrimidine-based combination chemotherapy regimens improve OS compared to gemcitabine plus nab-paclitaxel. The effects of radiotherapy were uncertain as only one low-quality trial was included. Selection of the most appropriate chemotherapy for individuals still remains unpersonalised, with clinicopathological stratification remaining elusive. Biomarker development is essential to assist in rationalising treatment selection for patients.
Collapse
Affiliation(s)
- Lucy Haggstrom
- Medical Oncology, The Kinghorn Cancer Care Centre, St Vincent's Hospital, Sydney, Australia
- Medical Oncology, Illawarra Shoalhaven Local Health District, Wollongong, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Wei Yen Chan
- Medical Oncology, The Kinghorn Cancer Care Centre, St Vincent's Hospital, Sydney, Australia
- Medical Oncology, Chris O'Brien Lifehouse, Sydney, Australia
| | - Adnan Nagrial
- The Crown Princess Mary Cancer Centre, Westmead, Australia
- Medical School, The University of Sydney, Sydney, Australia
| | - Lorraine A Chantrill
- Medical Oncology, Illawarra Shoalhaven Local Health District, Wollongong, Australia
- University of Wollongong, Wollongong, Australia
| | - Hao-Wen Sim
- Medical Oncology, The Kinghorn Cancer Care Centre, St Vincent's Hospital, Sydney, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - Desmond Yip
- Department of Medical Oncology, The Canberra Hospital, Garran, Australia
- ANU Medical School, Australian National University, Acton, Australia
| | - Venessa Chin
- Medical Oncology, The Kinghorn Cancer Care Centre, St Vincent's Hospital, Sydney, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
- Medical Oncology, Garvan Institute of Medical Research, Sydney, Australia
| |
Collapse
|
48
|
Yamaguchi N, Wu YG, Ravetch E, Takahashi M, Khan AG, Hayashi A, Mei W, Hsu D, Umeda S, de Stanchina E, Lorenz IC, Iacobuzio-Donahue CA, Tavazoie SF. A Targetable Secreted Neural Protein Drives Pancreatic Cancer Metastatic Colonization and HIF1α Nuclear Retention. Cancer Discov 2024; 14:2489-2508. [PMID: 39028915 PMCID: PMC11611693 DOI: 10.1158/2159-8290.cd-23-1323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 05/29/2024] [Accepted: 07/18/2024] [Indexed: 07/21/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an increasingly diagnosed cancer that kills 90% of afflicted patients, with most patients receiving palliative chemotherapy. We identified neuronal pentraxin 1 (NPTX1) as a cancer-secreted protein that becomes overexpressed in human and murine PDAC cells during metastatic progression and identified adhesion molecule with Ig-like domain 2 (AMIGO2) as its receptor. Molecular, genetic, biochemical, and pharmacologic experiments revealed that secreted NPTX1 acts cell-autonomously on the AMIGO2 receptor to drive PDAC metastatic colonization of the liver-the primary site of PDAC metastasis. NPTX1-AMIGO2 signaling enhanced hypoxic growth and was critically required for hypoxia-inducible factor-1α (HIF1α) nuclear retention and function. NPTX1 is overexpressed in human PDAC tumors and upregulated in liver metastases. Therapeutic targeting of NPTX1 with a high-affinity monoclonal antibody substantially reduced PDAC liver metastatic colonization. We thus identify NPTX1-AMIGO2 as druggable critical upstream regulators of the HIF1α hypoxic response in PDAC. Significance: We identified the NPTX1-AMIGO2 axis as a regulatory mechanism upstream of HIF1α-driven hypoxia response that promotes PDAC liver metastasis. Therapeutic NPTX1 targeting outperformed a common chemotherapy regimen in inhibiting liver metastasis and suppressed primary tumor growth in preclinical models, revealing a novel therapeutic strategy targeting hypoxic response in PDAC.
Collapse
Affiliation(s)
- Norihiro Yamaguchi
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Y Gloria Wu
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Ethan Ravetch
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Mai Takahashi
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Abdul G. Khan
- Tri-Institutional Therapeutics Discovery Institute, New York, NY, USA
| | - Akimasa Hayashi
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Wenbin Mei
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Dennis Hsu
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Shigeaki Umeda
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Ivo C. Lorenz
- Tri-Institutional Therapeutics Discovery Institute, New York, NY, USA
| | | | - Sohail F. Tavazoie
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| |
Collapse
|
49
|
Mastrantoni L, Chiaravalli M, Spring A, Beccia V, Di Bello A, Bagalà C, Bensi M, Barone D, Trovato G, Caira G, Giordano G, Bria E, Tortora G, Salvatore L. Comparison of first-line chemotherapy regimens in unresectable locally advanced or metastatic pancreatic cancer: a systematic review and Bayesian network meta-analysis. Lancet Oncol 2024; 25:1655-1665. [PMID: 39542008 DOI: 10.1016/s1470-2045(24)00511-4] [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: 06/16/2024] [Revised: 09/08/2024] [Accepted: 09/09/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND In advanced pancreatic ductal adenocarcinoma (PDAC), first-line chemotherapy is the standard of care. Due to the absence of head-to-head comparisons in clinical trials, we performed this systematic review and network meta-analysis to compare treatment options for PDAC in terms of their efficacy and toxicity. METHODS PubMed, the Cochrane Central Register of Controlled Trials, Embase, and oncological meetings websites were searched until Nov 15, 2023. We included phase 2-3 randomised controlled trials published after Jan 1, 2000, evaluating first-line treatments in patients with previously untreated, unresectable, locally advanced or metastatic PDAC. Primary endpoints assessed were progression-free survival and overall survival. Summary data were extracted from published reports. The deviance information criterion was used to choose between a random-effects or fixed-effects model. Hazard ratios (HRs) with 95% credible intervals were estimated using a Bayesian approach. The risk of bias was evaluated using the Cochrane Risk of Bias 2 (RoB 2) tool and studies were graded as low, some concerns, or high risk of bias. The quality of evidence was evaluated using the Grading of Recommendations Assessment, Development, and Evaluation approach. This systematic review and network meta-analysis is registered with PROSPERO, CRD42023450330. FINDINGS 6050 records were screened and 79 randomised controlled trials (22 168 patients) were included in the analysis. Gemcitabine was the most frequent comparator (in 50 [63%] of 79 trials) and was considered as the reference treatment. A fixed-effect model was used to analyse the primary outcomes. Regarding progression-free survival (71 trials, 19 479 patients), the most effective treatments were gemcitabine plus nab-paclitaxel alternating folinic acid, fluorouracil, and oxaliplatin ([FOLFOX] HR 0·32, 95% credible interval 0·22-0·47), cisplatin, nab-paclitaxel, capecitabine, and gemcitabine ([PAXG] 0·35, 0·22-0·55), and liposomal irinotecan in combination with fluorouracil, leucovorin, and oxaliplatin ([NALIRIFOX] 0·43, 0·34-0·54), followed by fluorouracil, leucovorin, irinotecan, and oxaliplatin ([FOLFIRINOX] 0·55, 0·47-0·65) and gemcitabine plus nab-paclitaxel (0·62, 0·54-0·72). Similar results were observed for overall survival (79 trials, 22 104 patients): PAXG (HR 0·40, 95% credible interval 0·25-0·65), gemcitabine plus nab-paclitaxel alternating FOLFOX (0·46, 0·32-0·66), and NALIRIFOX (0·56, 0·45-0·70) had the highest benefit, followed by FOLFIRINOX (0·66, 0·56-0·78) and gemcitabine plus nab-paclitaxel (0·67, 0·59-0·77). The overall risk of bias was low to some concerns. Certainty of evidence was low. INTERPRETATION Our findings suggest that NALIRIFOX and FOLFIRINOX should be the preferred options for patients who can tolerate these regimens, with gemcitabine plus nab-paclitaxel remaining a viable alternative, particularly in patients unfit for triplet therapy. Phase 3 randomised controlled trials investigating concomitant or sequential quadruplets are warranted. FUNDING None.
Collapse
Affiliation(s)
- Luca Mastrantoni
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy
| | - Marta Chiaravalli
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy
| | - Alexia Spring
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy
| | - Viria Beccia
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy
| | - Armando Di Bello
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy
| | - Cinzia Bagalà
- Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy
| | - Maria Bensi
- Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy
| | - Diletta Barone
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy
| | - Giovanni Trovato
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy
| | - Giulia Caira
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy
| | - Giulia Giordano
- Department of Aging, Orthopedics and Rheumatological Sciences, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Emilio Bria
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy; Medical Oncology Unit, Ospedale Isola Tiberina-Gemelli Isola, Rome, Italy
| | - Giampaolo Tortora
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy
| | - Lisa Salvatore
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy.
| |
Collapse
|
50
|
Shi S, Ye L, Jin K, Yu X, Guo D, Wu W. The complement C3a/C3aR pathway is associated with treatment resistance to gemcitabine-based neoadjuvant therapy in pancreatic cancer. Comput Struct Biotechnol J 2024; 23:3634-3650. [PMID: 39469671 PMCID: PMC11513484 DOI: 10.1016/j.csbj.2024.09.032] [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: 06/30/2024] [Revised: 09/21/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024] Open
Abstract
Gemcitabine is a standard first-line drug for pancreatic cancer chemotherapy. Nevertheless, gemcitabine resistance is common and significantly limits its therapeutic efficacy, impeding advancements in pancreatic cancer treatment. In this study, through a comprehensive analysis of gemcitabine-resistant cell lines and patient samples, 39 gemcitabine resistance-associated risk genes were identified, and two distinct gemcitabine response-related phenotypes were delineated. Through a combination of bioinformatics analysis and in vivo and in vitro experiments, we identified the C3a/C3aR signaling pathway as a pivotal player in the development of gemcitabine resistance in pancreatic cancer. We found that activation of the C3a/C3aR signaling pathway promoted the proliferation, migration and gemcitabine resistance of pancreatic cancer cells, while the C3aR antagonist SB290157 effectively counteracted these effects by impeding the activation of the C3a/C3aR pathway. Our study reveals the fundamental role of complement C3a in the progression of pancreatic cancer, suggesting that complement C3a may serve as a promising biomarker in pancreatic cancer.
Collapse
Affiliation(s)
- Saimeng Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Longyun Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Kaizhou Jin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Duancheng Guo
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Weiding Wu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| |
Collapse
|