1
|
Sugumar K, Alabd A, Alabd A, Hue JJ, Lyons J, Fields S, Wainberg Z, Zheng L, Coogle B, Kasi A, Grewal N, Kindler HL, Starr J, Sama AR, Winter JM. Exceptional responders to immunotherapy in pancreatic cancer: A multi-institutional case series of a rare occurrence. Oncotarget 2025; 16:427-442. [PMID: 40492845 DOI: 10.18632/oncotarget.28739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2025] Open
Abstract
INTRODUCTION Immunotherapy has emerged as a standard treatment option for multiple solid tumors. However, most patients with pancreatic cancer (PC) do not derive a significant benefit. Identification and analyses of exceptional responders could eventually offer hints as to why PC is resistant to immunotherapy. METHODS Oncologists from cancer centers in the United States were contacted to identify patients with PC who responded to immunotherapy. Exceptional responders were defined as those having either partial (PR) or complete response (CR) based on Response Evaluation Criteria in Solid Tumors, or biochemical response (CA 19-9 levels) after starting immunotherapy. Patients receiving concurrent chemotherapy were excluded. RESULTS 14 patients met inclusion criteria. Immunotherapy drugs included checkpoint inhibitors and macrophage inhibitors. Eight patients (42%) were MSI (microsatellite instability)-high. Radiologically, 82% had PR. Four patients (28%) had marked reduction in CA 19-9. The median progression-free survival was 12 months from the start of immunotherapy. Median survival was not reached. The 1- and 2-year survival probabilities were 80%, 70% respectively. CONCLUSION Majority of clinical trials evaluating immunotherapy in PC have yielded disappointing response rates compared to other solid tumors. Our case series adds to published data from early-phase trials supporting the promise of immunotherapy in some patients with PC.
Collapse
Affiliation(s)
- Kavin Sugumar
- Department of Surgery, University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA
| | - Andrew Alabd
- Department of Medicine, Cooper University Healthcare, Camden, NJ 08103, USA
| | - Andre Alabd
- Department of Urology, University of Indiana, Indianapolis, IN 46227, USA
| | - Jonathan J Hue
- Department of Surgery, University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA
| | - Josh Lyons
- Department of Surgery, University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA
| | - Sherri Fields
- Department of Medicine, UCLA/Santa Monica Cancer Center, CA 90404, USA
| | - Zev Wainberg
- Department of Medicine, UCLA/Santa Monica Cancer Center, CA 90404, USA
| | - Lei Zheng
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Brianna Coogle
- Department of Medicine, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Anup Kasi
- Department of Medicine, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Nicholas Grewal
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Hedy L Kindler
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Jason Starr
- Department of Medicine, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Ashwin R Sama
- Department of Medicine, Jefferson Medical Oncology Associates, Philadelphia, PA 19107, USA
| | - Jordan M Winter
- Department of Surgery, University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA
| |
Collapse
|
2
|
Neophytou C, Charalambous A, Voutouri C, Angeli S, Panagi M, Stylianopoulos T, Mpekris F. Sonopermeation combined with stroma normalization enables complete cure using nano-immunotherapy in murine breast tumors. J Control Release 2025; 382:113722. [PMID: 40233830 PMCID: PMC12076078 DOI: 10.1016/j.jconrel.2025.113722] [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: 01/17/2025] [Revised: 04/09/2025] [Accepted: 04/09/2025] [Indexed: 04/17/2025]
Abstract
Nano-immunotherapy shows great promise in improving patient outcomes, as seen in advanced triple-negative breast cancer, but it does not cure the disease, with median survival under two years. Therefore, understanding resistance mechanisms and developing strategies to enhance its effectiveness in breast cancer is crucial. A key resistance mechanism is the pronounced desmoplasia in the tumor microenvironment, which leads to dysfunction of tumor blood vessels and thus, to hypoperfusion, limited drug delivery and hypoxia. Ultrasound sonopermeation and agents that normalize the tumor stroma have been employed separately to restore vascular abnormalities in tumors with some success. Here, we performed in vivo studies in two murine, orthotopic breast tumor models to explore if combination of ultrasound sonopermeation with a stroma normalization drug can synergistically improve tumor perfusion and enhance the efficacy of nano-immunotherapy. We found that the proposed combinatorial treatment can drastically reduce primary tumor growth and in many cases tumors were no longer measurable. Overall survival studies showed that all mice that received the combination treatment survived and rechallenge experiments revealed that the survivors obtained immunological memory. Employing ultrasound elastography and contrast enhanced ultrasound along with proteomics analysis, flow cytometry and immunofluorescene staining, we found the combinatorial treatment reduced tumor stiffness to normal levels, restoring tumor perfusion and oxygenation. Furthermore, it increased infiltration and activity of immune cells and altered the levels of immunosupportive chemokines. Finally, using machine learning analysis, we identified that tumor stiffness, CD8+ T cells and M2-type macrophages were strong predictors of treatment response.
Collapse
Affiliation(s)
- Constantina Neophytou
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 1678, Cyprus; Cancer Genetics, Therapeutics & Ultrastructural Pathology Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Antonia Charalambous
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 1678, Cyprus
| | - Chrysovalantis Voutouri
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 1678, Cyprus
| | - Stella Angeli
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 1678, Cyprus; Cancer Genetics, Therapeutics & Ultrastructural Pathology Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Myrofora Panagi
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 1678, Cyprus
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 1678, Cyprus
| | - Fotios Mpekris
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 1678, Cyprus; Cancer Genetics, Therapeutics & Ultrastructural Pathology Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus.
| |
Collapse
|
3
|
Zhang J, Qu M, Mo Z, Sui H, Liu L, Fu D. Ionizable cationic lipid nanoparticles loaded with miRNA-125b/BLZ945 for pancreatic cancer treatment. Biotechnol Appl Biochem 2025; 72:846-857. [PMID: 39623756 DOI: 10.1002/bab.2701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 11/12/2024] [Indexed: 04/29/2025]
Abstract
In prior research, both miRNA-125b and BLZ945 have shown potential in effectively inhibiting M2 macrophage polarization and producing antitumor effects. Nevertheless, their physicochemical characteristics present significant challenges for efficient in vivo delivery. Ionizable cationic lipid nanoparticles (LNPs), recognized for their superior biocompatibility and drug-loading capacity, serve as a novel carrier for nucleic acid-based therapeutics. In our study, we successfully encapsulated both agents within LNPs and conducted a thorough characterization. Subsequently, we investigated their potential to repolarize M2 macrophages in vitro and evaluated their in vivo distribution, biosafety, and antitumor efficacy. The findings revealed that the LNPs maintained excellent drug-loading efficiency, consistent particle size, and stable zeta potential. All formulations effectively inhibited M2 macrophage polarization in vitro. Upon administration in vivo, the LNPs not only demonstrated favorable biosafety profiles but also accumulated efficiently in tumor tissues, substantially reducing tumor burden, particularly notable in co-loaded LNPs. Our results affirm that LNPs are an effective carrier for miRNA-125b and BLZ945, highlighting this encapsulation approach as promising for the treatment of solid tumors and meriting further investigation. Practitioner points: (i) Ionizable cationic nanoparticles provide high and stable encapsulation rates to efficiently load nucleic acid polymers into the LNP, avoiding the rapid accumulation of circulating macrophages, which can lead to reduced penetration of the LNP into target tissues. Therefore, it can be used as a novel drug delivery method to benefit clinical patients. (ii) miRNA-125b LNP/BLZ945 LNP attenuated the depleting effect of BLZ945 on macrophages and significantly inhibited macrophage M2 polarization. It could be effectively distributed in tumors and showed good biosafety while exerting antitumor effects, bringing hope to clinical pancreatic tumor patients.
Collapse
Affiliation(s)
- Jiajie Zhang
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai, China
- Department of Radiation Oncology, UNC School of Medicine, Chapel Hill, North Carolina, USA
| | - Ming Qu
- Department of Radiology, China-Japan Union Hospital of Jilin University, Changchun, China
- Jilin Institute of Radiology Intervention, Changchun, China
| | - Zhanhao Mo
- Department of Radiology, China-Japan Union Hospital of Jilin University, Changchun, China
- Jilin Institute of Radiology Intervention, Changchun, China
| | - He Sui
- Department of Radiology, China-Japan Union Hospital of Jilin University, Changchun, China
- Jilin Institute of Radiology Intervention, Changchun, China
| | - Lin Liu
- Department of Radiology, China-Japan Union Hospital of Jilin University, Changchun, China
- Jilin Institute of Radiology Intervention, Changchun, China
| | - Deliang Fu
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
4
|
Xie X, Fu Y, Gan L, Yang X, Song Y, Song C, Sun F, Guo L, Long H. Safety and efficacy of switching to 4-weekly albuvirtide plus daily dolutegravir in virologically suppressed HIV-1 adults: a 24-week study. AIDS 2025; 39:857-862. [PMID: 39874112 DOI: 10.1097/qad.0000000000004130] [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: 10/21/2024] [Accepted: 01/16/2025] [Indexed: 01/30/2025]
Abstract
OBJECTIVES Albuvirtide (ABT) is a long-acting fusion inhibitor. This study assessed switching to ABT 640 mg every 4 weeks plus daily dolutegravir (DTG) in virologically suppressed adults with HIV-1. DESIGN AND METHODS In this open-label, single-arm study, 10 participants with HIV-1 RNA less than 50 copies/ml switched to ABT plus DTG for 24 weeks. Safety, pharmacokinetics, viral load, and CD4 + T cell counts were assessed. RESULTS No serious adverse events occurred. Albuvirtide's steady-state trough concentration was 31.1 times higher than PA-IC90. All participants maintained virological suppression. CD4 + T-cell counts increased significantly after 24 weeks ( P = 0.0462). CONCLUSION Switching to 4-weekly ABT plus daily DTG demonstrated good safety, favorable pharmacokinetics, maintained virological suppression, and improved immune recovery. These findings support ABT's potential as a long-acting agent for simplifying HIV treatment.
Collapse
Affiliation(s)
- Xiaoxin Xie
- Guiyang Public Health Clinical Center, Guiyang, China
| | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Wang W, Hu K, Xue J, Chen J, Du X, Zhao T, Chen Y, Tang X, Xu L, Hao X, Li X, Yang Y. In vivo FAP-CAR macrophages enhance chemotherapy and immunotherapy against pancreatic cancer by removing the fibrosis barrier. J Control Release 2025:113888. [PMID: 40425095 DOI: 10.1016/j.jconrel.2025.113888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2025] [Accepted: 05/24/2025] [Indexed: 05/29/2025]
Abstract
Patients with pancreatic ductal adenocarcinoma (PDAC) derive limited benefits from chemotherapy or immunotherapy, with a five-year survival rate still below 10 %. The key therapeutic challenge is the dense fibrosis barrier driven by activated cancer-associated fibroblasts (CAFs) and their secreted collagen, which impedes drug penetration and characterizes PDAC as an immune-desert tumor. To address this challenge, we developed in vivo chimeric antigen receptor macrophages (FAP-CAR-M) targeting fibroblast activation protein-α (FAP), the marker of activated CAFs, to enhance chemo and immunotherapy against PDAC by removing the fibrosis barrier using mannose-modified mRNA-LNP (MLNP). Our results demonstrate that mRNA-MLNP can efficiently reprogram M2 macrophages into FAP-CAR-M. With the FAP-CAR-M treatment, the activated CAF markers (FAP), collagen volume fraction (CVF), and the type I collagen (col1a1) secretion were decreased by 3-fold, 5-fold, and 4-fold in orthotopic PDAC, respectively. By removing the fibrosis barrier, FAP-CAR-M enhanced the penetration of gemcitabine (GEM) and immune cells, improved PDAC sensitivity to chemo and immunotherapy, and significantly prolonged survival. Therefore, in vivo FAP-CAR-M may represent a potential therapeutic approach to enhance chemo and immunotherapy against PDAC by removing the fibrosis barrier.
Collapse
Affiliation(s)
- Wenguang Wang
- School of Translational Research, China Pharmaceutical University, Nanjing 211198, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China.
| | - Kaiyuan Hu
- School of Translational Research, China Pharmaceutical University, Nanjing 211198, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Junjie Xue
- School of Translational Research, China Pharmaceutical University, Nanjing 211198, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Jingyi Chen
- School of Translational Research, China Pharmaceutical University, Nanjing 211198, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Xiuli Du
- School of Translational Research, China Pharmaceutical University, Nanjing 211198, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Tian Zhao
- School of Translational Research, China Pharmaceutical University, Nanjing 211198, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Yiwei Chen
- School of Translational Research, China Pharmaceutical University, Nanjing 211198, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Xinying Tang
- School of Translational Research, China Pharmaceutical University, Nanjing 211198, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Lu Xu
- Department of Hematology, The First Affiliated Hospital of Hainan Medical University, No. 31 Longhua Road, Longhua District, Haikou 570102, Hainan Province, China
| | - Xinbao Hao
- Department of Hematology, The First Affiliated Hospital of Hainan Medical University, No. 31 Longhua Road, Longhua District, Haikou 570102, Hainan Province, China
| | - Xianjing Li
- School of Translational Research, China Pharmaceutical University, Nanjing 211198, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Yong Yang
- School of Translational Research, China Pharmaceutical University, Nanjing 211198, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
6
|
Pereira-Silva M, Veiga F, Paiva-Santos AC, Concheiro A, Alvarez-Lorenzo C. Biomimetic nanosystems for pancreatic cancer therapy: A review. J Control Release 2025; 383:113824. [PMID: 40348133 DOI: 10.1016/j.jconrel.2025.113824] [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: 12/17/2024] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Pancreatic cancer (PC) is a highly lethal and aggressive malignancy, currently one of the leading causes of cancer-related deaths worldwide, in both women and men. PC is highly resistant to standard chemotherapy (CT) because its immunosuppressive and hypoxic tumor microenvironment and a dense desmoplastic stroma compartment extensively limit drug accessibility and perfusion. Although CT is one of the main therapeutic strategies for PC management contributing to tumor eradication through a cytotoxic effect, CT is associated with a poor pharmacokinetic profile and provokes deleterious systemic toxicity. This low efficacy-poor safety scenario urgently calls for innovative and highly specific therapeutic strategies to counteract this urgent clinical challenge. Nanotechnology-based precision materials for cancer may help improve drug stability and minimize the systemic cytotoxic effects by increasing drug tumor accumulation and also enabling controlled release, but several drawbacks still persist, such as the poor targeting efficiency. In the last few years increased attention has been paid to bioinspired nanosystems that can mimic either partially or totally biological systems, including lipid layers as suitable stealth coatings resembling the composition of cell membranes, lipoprotein- and blood protein-based nanosystems, and cell membrane-derived systems, such as extracellular vesicles, cell membrane nanovesicles and cell membrane-coated nanosystems, which display intrinsic cancer-targeting abilities, enhanced biocompatibility, decreased immunogenicity, and prolonged blood circulation profile. This review covers the recent breakthroughs on advanced biomimetic PC-targeted nanosystems, focusing on their design, properties and applications as innovative, multifunctional and versatile tools paving the way to improved PC diagnosis and treatment.
Collapse
Affiliation(s)
- Miguel Pereira-Silva
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal; Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma, Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| | - Francisco Veiga
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Angel Concheiro
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma, Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma, Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
7
|
Wang Y, Bian Z, Xu L, Du G, Qi Z, Zhang Y, Long J, Li W. The scRNA-sequencing landscape of pancreatic ductal adenocarcinoma revealed distinct cell populations associated with tumor initiation and progression. Genes Dis 2025; 12:101323. [PMID: 40092486 PMCID: PMC11907457 DOI: 10.1016/j.gendis.2024.101323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 04/09/2024] [Accepted: 04/21/2024] [Indexed: 03/19/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) stands as a formidable malignancy characterized by its profound lethality. The comprehensive analysis of the transcriptional landscape holds immense significance in understanding PDAC development and exploring novel treatment strategies. However, due to the firm consistency of pancreatic cancer samples, the dissociation of single cells and subsequent sequencing can be challenging. Here, we performed single-cell RNA sequencing (scRNA-seq) on 8 PDAC patients with different lymph node metastasis status. We first identified the crucial role of MMP1 in the transition from normal pancreatic cells to cancer cells. The knockdown of MMP1 in pancreatic cancer cell lines decreased the expression of ductal markers such as SOX9 while the overexpression of MMP1 in hTERT-HPNE increased the expression of ductal markers, suggesting its function of maintaining ductal identity. Secondly, we found a S100A2 + tumor subset which fueled lymph node metastasis in PDAC. The knockdown of S100A2 significantly reduced the motility of pancreatic cancer cell lines in both wound healing and transwell migration assays. While overexpression of S100A2 led to increased migratory capability. Moreover, overexpression of S100A2 in KPC1199, a mouse pancreatic cancer cell line, caused a larger tumor burden in a hemi-spleen injection model of liver metastasis. In addition, epithelial-mesenchymal transition-related genes were decreased by S100A2 knockdown revealed by bulk RNA sequencing. We also identified several pivotal contributors to the pro-tumor microenvironment, notably OMD + fibroblast and CCL2 + macrophage. As a result, our study provides valuable insights for early detection of PDAC and promising therapeutic targets for combatting lymph node metastasis.
Collapse
Affiliation(s)
- Ying Wang
- Department of Interventional Radiology, Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Zhouliang Bian
- Department of Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201900, China
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Lichao Xu
- Department of Interventional Radiology, Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Guangye Du
- Department of Pathology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201900, China
| | - Zihao Qi
- Department of Pancreatic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yanjie Zhang
- Department of Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201900, China
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Jiang Long
- Department of Pancreatic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Wentao Li
- Department of Interventional Radiology, Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| |
Collapse
|
8
|
Witt RC, Dunn CE, Zanders LA, Edema AA, Waheed SA, Derewonko CA, Franzen LC, Osborn PL, Caudle JD, Sheaff RJ, Lamar AA. Inhibition of Adenosine Triphosphate Production in Pancreatic Cancer Cells by a Library of N-(1H-Indol-4-ylmethyl)benzenesulfonamide and N-(1H-Indol-5-ylmethyl)benzenesulfonamide Analogs. ChemMedChem 2025:e2500136. [PMID: 40295191 DOI: 10.1002/cmdc.202500136] [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: 02/17/2025] [Revised: 04/25/2025] [Accepted: 04/28/2025] [Indexed: 04/30/2025]
Abstract
A library of 50 indolyl sulfonamides and 9 amide analogs based upon the 4-indolyl and 5-indolyl frameworks has been synthesized to target the metabolic processes of pancreatic cancer. Thirteen of the 50 compounds are identified as cytotoxic at 50 μM using a traditional (48-h compound exposure) assay against 7 pancreatic cancer cell lines and 1 noncancerous cell line. The potential role of the compounds as metabolic inhibitors of adenosine triphosphate (ATP) production is then evaluated using a rapid screening (1-2 h compound exposure) assay developed within our laboratories. The rapid assay identifies ten compounds as strong or moderate hits at 3 μM against the panel of pancreatic and noncancerous cell lines. The IC50 values of the active compounds are determined using the rapid assay in the absence of glucose and four of the compounds display an IC50 value <1 μM against one or more pancreatic cancer cell lines. A comparison of IC50 values of the active compounds in the presence of glucose implicates the potential role of the compounds as oxidative phosphorylation inhibitors of ATP production. Finally, a series of amide analogs are synthesized and screened for activity to determine the structural importance of the sulfonamide functionality.
Collapse
Affiliation(s)
- Ryan C Witt
- Department of Chemistry and Biochemistry, The University of Tulsa, 800 South Tucker Drive, Tulsa, OK, 74104, USA
| | - Caroline E Dunn
- Department of Chemistry and Biochemistry, The University of Tulsa, 800 South Tucker Drive, Tulsa, OK, 74104, USA
| | - Levi A Zanders
- Department of Chemistry and Biochemistry, The University of Tulsa, 800 South Tucker Drive, Tulsa, OK, 74104, USA
| | - Adeleye A Edema
- Department of Chemistry and Biochemistry, The University of Tulsa, 800 South Tucker Drive, Tulsa, OK, 74104, USA
| | - Sakariyau A Waheed
- Department of Chemistry and Biochemistry, The University of Tulsa, 800 South Tucker Drive, Tulsa, OK, 74104, USA
| | - Carina A Derewonko
- Department of Chemistry and Biochemistry, The University of Tulsa, 800 South Tucker Drive, Tulsa, OK, 74104, USA
| | - Lauren C Franzen
- Department of Chemistry and Biochemistry, The University of Tulsa, 800 South Tucker Drive, Tulsa, OK, 74104, USA
| | - Presley L Osborn
- Department of Chemistry and Biochemistry, The University of Tulsa, 800 South Tucker Drive, Tulsa, OK, 74104, USA
| | - Jenna D Caudle
- Department of Chemistry and Biochemistry, The University of Tulsa, 800 South Tucker Drive, Tulsa, OK, 74104, USA
| | - Robert J Sheaff
- Department of Chemistry and Biochemistry, The University of Tulsa, 800 South Tucker Drive, Tulsa, OK, 74104, USA
| | - Angus A Lamar
- Department of Chemistry and Biochemistry, The University of Tulsa, 800 South Tucker Drive, Tulsa, OK, 74104, USA
| |
Collapse
|
9
|
Li L, Wu ZT, Duan WH, Liu J, Zhu YR. Machine learning-based prognostic modelling of NK cells in PAAD for immunotherapy guidance. Discov Oncol 2025; 16:577. [PMID: 40253675 PMCID: PMC12009793 DOI: 10.1007/s12672-025-02266-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 03/28/2025] [Indexed: 04/22/2025] Open
Abstract
Pancreatic cancer's high incidence and mortality rates are underscored by ineffective treatments, particularly immunotherapy's poor performance. This could stem from an unclear immune microenvironment, where NK cells may play a unique role. Analyzing the NK cell-differentially expressed genes (NKDEGs) from the PAAD_GSE162708 single-cell dataset and utilizing the TCGA-PAAD and ICGC-PACA-AU datasets, we identified 11 NKDEGs linked to pancreatic adenocarcinoma (PAAD) prognosis and developed a prognostic model. This model's risk scores significantly outperformed traditional grading and TNM staging systems, validated through clinical and pathological analyses. Functional enrichment analysis pointed to the Neuroactive ligand-receptor interaction and MAPK signaling pathways, suggesting NK cells' distinctive role in PAAD. High-risk groups showed decreased overall NK cells but increased activated NK cells, which may mediate adverse inflammatory responses. NK cells exhibit synergistic interactions with plasma cells and macrophages and negative regulation by monocytes and naive B cells. Our model accurately predicts immunotherapy responses, indicating potential for targeted drugs to enhance treatment. Additionally, we introduced an NKDEGs-based immunotyping approach for personalized medicine and clinical decision-making in PAAD. This study emphasizes NK cells' potential in PAAD treatment, offering precise patient stratification and therapeutic targets for immunotherapy.
Collapse
Affiliation(s)
- Li Li
- The People'S Hospital of Wenshan Prefecture, Kunming University of Science and Technology, No. 228, Kaihua East Road, Wenshan, 663000, Yunnan, China
| | - Zu-Tao Wu
- The People'S Hospital of Wenshan Prefecture, Kunming University of Science and Technology, No. 228, Kaihua East Road, Wenshan, 663000, Yunnan, China
| | - Wen-Hong Duan
- The People'S Hospital of Wenshan Prefecture, Kunming University of Science and Technology, No. 228, Kaihua East Road, Wenshan, 663000, Yunnan, China
| | - Jiang Liu
- The People'S Hospital of Wenshan Prefecture, Kunming University of Science and Technology, No. 228, Kaihua East Road, Wenshan, 663000, Yunnan, China
| | - Yin-Rong Zhu
- The People'S Hospital of Wenshan Prefecture, Kunming University of Science and Technology, No. 228, Kaihua East Road, Wenshan, 663000, Yunnan, China.
| |
Collapse
|
10
|
Liaki V, Rosas-Perez B, Guerra C. Unlocking the Genetic Secrets of Pancreatic Cancer: KRAS Allelic Imbalances in Tumor Evolution. Cancers (Basel) 2025; 17:1226. [PMID: 40227826 PMCID: PMC11987834 DOI: 10.3390/cancers17071226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 03/13/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) belongs to the types of cancer with the highest lethality. It is also remarkably chemoresistant to the few available cytotoxic therapeutic options. PDAC is characterized by limited mutational heterogeneity of the known driver genes, KRAS, CDKN2A, TP53, and SMAD4, observed in both early-stage and advanced tumors. In this review, we summarize the two proposed models of genetic evolution of pancreatic cancer. The gradual or stepwise accumulated mutations model has been widely studied. On the contrary, less evidence exists on the more recent simultaneous model, according to which rapid tumor evolution is driven by the concurrent accumulation of genetic alterations. In both models, oncogenic KRAS mutations are the main initiating event. Here, we analyze the emerging topic of KRAS allelic imbalances and how it arises during tumor evolution, as it is often detected in advanced and metastatic PDAC. We also summarize recent evidence on how it affects tumor biology, metastasis, and response to therapy. To this extent, we highlight the necessity to include studies of KRAS allelic frequencies in the design of future therapeutic strategies against pancreatic cancer.
Collapse
Affiliation(s)
- Vasiliki Liaki
- Molecular Oncology Program, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain; (B.R.-P.); (C.G.)
| | - Blanca Rosas-Perez
- Molecular Oncology Program, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain; (B.R.-P.); (C.G.)
| | - Carmen Guerra
- Molecular Oncology Program, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain; (B.R.-P.); (C.G.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
11
|
Papacharisi E, Braun AC, Vranic M, Pahl AM, Hechler T. Novel Amanitin-Based Antibody-Drug Conjugates Targeting TROP2 for the Treatment of Pancreatic Cancer. Mol Cancer Ther 2025; 24:485-496. [PMID: 39564769 PMCID: PMC11962393 DOI: 10.1158/1535-7163.mct-24-0266] [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: 04/16/2024] [Revised: 09/24/2024] [Accepted: 11/18/2024] [Indexed: 11/21/2024]
Abstract
Trophoblast cell surface antigen 2 (TROP2) exhibits aberrant expression in pancreatic cancer, correlating with metastasis, advanced tumor stage, and poor prognosis in patients with pancreatic ductal adenocarcinoma. TROP2 has been recognized as a promising therapeutic target for antibody-drug conjugates (ADC), as evidenced by the approval of the anti-TROP2 ADC Trodelvy for the treatment of triple-negative breast cancer (TNBC). In this study, we report the generation of novel second-generation amanitin-based ADCs (ATAC) targeting TROP2, comprising the humanized RS7 antibody of Trodelvy (hRS7) and the highly potent payload amanitin. The specific in vitro binding, efficient antigen internalization, and high cytotoxicity of hRS7 ATACs with EC50 values in the picomolar range in TROP2-expressing cells constituted the foundation for preclinical in vivo evaluation. The hRS7 ATACs demonstrated a significant reduction in tumor growth in vivo in subcutaneous xenograft mouse models of pancreatic cancer and TNBC at well-tolerated doses. The antitumor efficacy correlated with the level of TROP2 expression on the tumors and the in vivo tumor uptake of the ATACs. The long half-life of 9.7 to 10.7 days of hRS7 ATACs without premature payload release in serum supported a high therapeutic index. Notably, the efficacy of the hRS7 ATACs was superior to that of Trodelvy with complete tumor eradication in both refractory pancreatic cancer and TNBC xenograft models. In summary, hRS7 ATACs represent a highly effective and well-tolerated targeted therapy, and our data support their development for pancreatic cancer and other TROP2-expressing tumors.
Collapse
|
12
|
Shakerian N, Tafazoli A, Razavinia A, Sadrzadeh Aghajani Z, Bana N, Mard-Soltani M, Khalesi B, Hashemi ZS, Khalili S. Current Understanding of Therapeutic and Diagnostic Applications of Exosomes in Pancreatic Cancer. Pancreas 2025; 54:e255-e267. [PMID: 39661050 DOI: 10.1097/mpa.0000000000002414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
ABSTRACT Unusual symptoms, rapid progression, lack of reliable early diagnostic biomarkers, and lack of efficient treatment choices are the ongoing challenges of pancreatic cancer. Numerous research studies have demonstrated the correlation between exosomes and various aspects of pancreatic cancer. In light of these facts, exosomes possess the potential to play functional roles in the treatment, prognosis, and diagnosis of the pancreatic cancer. In the present study, we reviewed the most recent developments in approaches for exosome separation, modification, monitoring, and communication. Moreover, we discussed the clinical uses of exosomes as less invasive liquid biopsies and drug carriers and their contribution to the control of angiogenic activity of pancreatic cancer. Better investigation of exosome biology would help to effectively engineer therapeutic exosomes with certain nucleic acids, proteins, and even exogenous drugs as their cargo. Circulating exosomes have shown promise as reliable candidates for pancreatic cancer early diagnosis and monitoring in high-risk people without clinical cancer manifestation. Although we have tried to reflect the status of exosome applications in the treatment and detection of pancreatic cancer, it is evident that further studies and clinical trials are required before exosomes may be employed as a routine therapeutic and diagnostic tools for pancreatic cancer.
Collapse
Affiliation(s)
- Neda Shakerian
- From the Department of Clinical Biochemistry, Faculty of Medical Sciences, Dezful University of Medical Sciences, Dezful
| | - Aida Tafazoli
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz
| | - Amir Razavinia
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, IR
| | | | - Nikoo Bana
- Kish International Campus, University of Teheran
| | - Maysam Mard-Soltani
- From the Department of Clinical Biochemistry, Faculty of Medical Sciences, Dezful University of Medical Sciences, Dezful
| | - Bahman Khalesi
- Department of Research and Production of Poultry Viral Vaccine, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization, Karaj
| | - Zahra Sadat Hashemi
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| |
Collapse
|
13
|
Xue Y, Wang Y, Ren Z, Yu K. Tissue factor promotes TREX1 protein stability to evade cGAS-STING innate immune response in pancreatic ductal adenocarcinoma. Oncogene 2025; 44:739-752. [PMID: 39658648 PMCID: PMC11888988 DOI: 10.1038/s41388-024-03248-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains the most challenging human malignancy that urgently needs effective therapy. Tissue factor (TF) is expressed in ~80% of PDAC and represents a potential therapeutic target. While a novel TF-ADC (MRG004A) demonstrated efficacy for PDAC and TNBC in a Phase I/II trial [Ref. 18], the functional role of TF in PDAC remains incompletely understood. We investigated the relationship between TF and the innate STING pathway. We found that patients with TF-overexpression had poor survival, very low levels of P-STING/P-TBK1, reduced amounts of ISGs and chemokines as well as low numbers of cytotoxic immunocytes in their tumor. In experimental models of mouse and human PDAC, tumor cell-intrinsic TF expression played a major role in silencing the cytosolic micronuclei sensing and cGAS-STING activation. This process involved a TREX1 exonuclease-dependent clearance of micronucleus-DNA accumulated in tumor cells. Treatment of tumors with TF-KO/shRNA or anti-TF antibody HuSC1-39 (parent antibody of MRG004A) triggered a rapid and proteasome-dependent degradation of TREX1 thereby restoring the STING/TBK1 cascade phosphorylation. TF-inhibition therapy promoted a robust STING/IRF3-dependent IFN/CCL5/CXCL9-11 production, immune effector cell infiltration and antitumor efficacy. Moreover, in the PBMC and cancer cell co-culture, TF-inhibition synergized with a STING agonist compound. A covalently conjugated TF antibody-STING agonist ADC strongly increased the efficacy of tumor-targeted STING agonism on chemokine secretion and tumor inhibition in vitro and in vivo. Thus, TF-inhibition reshapes an "immune hot" tumor environment. TF-targeted therapy warrants clinical investigation as a single agent or in combination with immunotherapy for treating TF-positive PDAC and TNBC.
Collapse
Affiliation(s)
- Yinyin Xue
- Department of Pharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Yue Wang
- Department of Pharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Zhiqiang Ren
- Department of Pharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Ker Yu
- Department of Pharmacology, Fudan University School of Pharmacy, Shanghai, China.
| |
Collapse
|
14
|
Zhou SQ, Wan P, Zhang S, Ren Y, Li HT, Ke QH. Programmed cell death 1 inhibitor sintilimab plus S-1 and gemcitabine for liver metastatic pancreatic ductal adenocarcinoma. World J Clin Oncol 2025; 16:98079. [PMID: 39995563 PMCID: PMC11686555 DOI: 10.5306/wjco.v16.i2.98079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 10/21/2024] [Accepted: 11/25/2024] [Indexed: 12/11/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with poor prognosis. When it metastasizes to the liver, treatment options become particularly limited and challenging. Current treatment options for liver metastatic PDAC are limited, and chemotherapy alone often proves insufficient. Immunotherapy, particularly programmed cell death 1 (PD-1) inhibitors like sintilimab, shows potential efficacy for various cancers but has limited reports on PDAC. This study compares the efficacy and safety of sintilimab plus S-1 and gemcitabine vs S-1 and gemcitabine alone in liver metastatic PDAC. AIM To explore the feasibility and effectiveness of combined PD-1 inhibitor sintilimab and S-1 and gemcitabine (combination group) vs S-1 and gemcitabine used alone (chemotherapy group) for treating liver metastatic pancreatic adenocarcinoma. METHODS Eligible patients were those with only liver metastatic PDAC, an Eastern Cooperative Oncology Group performance status of 0-1, adequate organ and marrow functions, and no prior anticancer therapy. Participants in the combination group received intravenous sintilimab 200 mg every 3 weeks, oral S-1 40 mg/m² twice daily on days 1-14 of a 21-day cycle, and intravenous gemcitabine 1000 mg/m² on days 1 and 8 of the same cycle for up to eight cycles or until disease progression, death, or unacceptable toxicity. Participants in the chemotherapy group received oral S-1 40 mg/m² twice daily on days 1-14 of a 21-day cycle and intravenous gemcitabine 1000 mg/m² on days 1 and 8 of the same cycle for up to eight cycles. Between June 2020 and December 2021, 66 participants were enrolled, with 32 receiving the combination treatment and 34 receiving chemotherapy alone. RESULTS The group receiving the combined therapy exhibited a markedly prolonged median overall survival (18.8 months compared to 10.3 months, P < 0.05) and progression-free survival (9.6 months vs 5.4 months, P < 0.05). compared to the chemotherapy group. The incidence of severe adverse events did not differ significantly between the two groups (P > 0.05). CONCLUSION The combination of PD-1 inhibitor sintilimab with S-1 and gemcitabine demonstrated effectiveness and safety for treating liver metastatic PDAC, meriting further investigation.
Collapse
Affiliation(s)
- Shi-Qiong Zhou
- Department of Chemoradiotherapy, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei Province, China
| | - Peng Wan
- Department of Chemoradiotherapy, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei Province, China
| | - Seng Zhang
- Department of Chemoradiotherapy, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei Province, China
| | - Yuan Ren
- Department of Chemoradiotherapy, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei Province, China
| | - Hong-Tao Li
- Department of Chemoradiotherapy, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei Province, China
| | - Qing-Hua Ke
- Department of Chemoradiotherapy, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei Province, China
| |
Collapse
|
15
|
Li B, Shi M, Wang Y, Li P, Yin X, Zhang G, Kang X, Wang H, Gao S, Zheng K, Shi X, Xu X, Zhou Y, Jiang H, Jing W, Guo S, Jin G. A practical distribution pattern of α-SMA-positive carcinoma associated fibroblasts indicates poor prognosis of patients with pancreatic ductal adenocarcinoma. Transl Oncol 2025; 52:102282. [PMID: 39808844 PMCID: PMC11782853 DOI: 10.1016/j.tranon.2025.102282] [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: 09/10/2024] [Revised: 12/08/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025] Open
Abstract
Purpose The present study aimed to clarify the distribution pattern of carcinoma associated fibroblasts (CAFs) across pancreatic ductal adenocarcinoma (PDAC) and its prognostic prediction value. Methods Data of two cohorts were retrospectively collected from consecutive patients who underwent primary pancreatic resection from January 2015 to December 2017. We used tumor specimens to screen out the most suitable markers for the spatial distribution analysis for CAFs subpopulations. We utilized a tissue microarray to assess the spatial intensity of α-SMA expression within the tumor microenvironment. Specifically, we classified CAFs into two types based on their α-SMA spatial expression. Type II CAFs were designated as those located in the juxtatumoural stroma with α-SMA expression that was moderate or higher, and those in the peripheral stroma with α-SMA expression that was less than moderate. All other cases, where the α-SMA expression did not meet these criteria, were categorized as Type I CAFs. Multivariable Cox proportional hazards regression was used to assess risk factors associated with patient outcomes. RNA sequencing data were obtained from bulk tumor samples and isolated CAFs from patients to reveal the distinct pattern and elucidated their fundamental characteristics. Results The α-SMA spatial intensity was the most suitable variable for representative of CAFs spatial characteristics. Patients with Type Ⅰ CAFs were more likely to be allocated into N1 or N2 of the N stage and Ⅱ and Ⅲ of the TNM stage. The spatial distribution pattern of CAFs (Type Ⅰ v.s. Type Ⅱ: HR, 1.568; 95 % CI, 1.053-2.334; P = 0.027) was an independent prognostic factor in the discovery cohort, so as in the validation (Type Ⅰ vs. Type Ⅱ: HR, 2.197; 95 % CI, 1.410-3.422; P = 0.001). RNA sequencing analysis revealed that the differentially expressed genes (DEGs) in Type I CAFs are closely associated with those in corresponding tumor tissues, highlighting the enhanced biological significance of immune-related and oncogenic invasive pathways. Conclusions Our findings that two types of α-SMA-positive CAFs with different spatial patterns present heterogeneously across tissues of PDACs and correlated with patients' outcomes. The spatial location of CAFs may facilitate patients' selection in precision medicine of PDACs.
Collapse
Affiliation(s)
- Bo Li
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China; Department of Hepatobiliary Pancreatic Surgery, Naval Medical Center, Naval Medical University (Second Military Medical University), 338 West Huaihai Road, Shanghai, 200052, China
| | - Meilong Shi
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China
| | - Yang Wang
- Department of Pathology, Shanghai Fourth People's Hospital, Tongji University School of Medicine, 1279 Sanmen Road, Shanghai 200434, China
| | - Penghao Li
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China
| | - Xiaoyi Yin
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China
| | - Guoxiao Zhang
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China
| | - Xiaochao Kang
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China
| | - Huan Wang
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China
| | - Suizhi Gao
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China
| | - Kailian Zheng
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China
| | - Xiaohan Shi
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China
| | - Xiongfei Xu
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China
| | - Yukun Zhou
- Department of Hepatobiliary Pancreatic Surgery, Naval Medical Center, Naval Medical University (Second Military Medical University), 338 West Huaihai Road, Shanghai, 200052, China
| | - Hui Jiang
- Department of Pathology, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China
| | - Wei Jing
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China.
| | - Shiwei Guo
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China.
| | - Gang Jin
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China.
| |
Collapse
|
16
|
Tang R, Tay SS, Sharbeen G, Herrmann D, Youkhana J, Timpson P, Phillips PA, Biro M. Bystander Expression of Atypical Chemokine Receptor 2 Protects T Cells from Chemoattraction towards Cancer-Associated Fibroblasts. Eur J Immunol 2025; 55:e202451215. [PMID: 39931761 PMCID: PMC11811810 DOI: 10.1002/eji.202451215] [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: 04/24/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 02/13/2025]
Abstract
Atypical chemokine receptors (ACKRs) are a subclass of chemokine receptors that internalise and degrade chemokines instead of eliciting chemotaxis. Scavenging by ACKRs reduces the local bioavailability of chemokines and can thus reshape chemokine gradients that direct leukocyte trafficking during inflammation and anticancer responses. In pancreatic ductal adenocarcinoma (PDAC), chemokine axes, such as CXCL12-CXCR4, are co-opted by cancer-associated fibroblasts (CAFs) for tumour growth and escape, and immunosuppression. Here, we explore the use of ACKRs to reshape chemokine gradients within the PDAC tumour microenvironment. ACKR2, previously only known to scavenge inflammatory CC chemokines, was recently shown to be able to interact with CXCL10 and CXCL14. Here, using a chemokine binding assay and cytometric bead arrays, we reveal that ACKR2 scavenges additional CXC chemokines CXCL12 and CXCL1. ACKR2 scavenges CXCL12 with reduced efficiency compared to ACKR3, previously reported to bind CXCL12. Finally, we demonstrate that the overexpression of ACKR2 on bystander cells protects primary murine cytotoxic T lymphocytes from PDAC CAF-mediated chemoattraction. These findings reveal new CXC chemokine ligands of ACKR2 and indicate that ACKR overexpression may protect T cells from misdirection by CAFs.
Collapse
Affiliation(s)
- Richard Tang
- EMBL Australia, Single Molecule Science node, School of Biomedical SciencesThe University of New South WalesSydneyNSWAustralia
| | - Szun S. Tay
- EMBL Australia, Single Molecule Science node, School of Biomedical SciencesThe University of New South WalesSydneyNSWAustralia
| | - George Sharbeen
- Pancreatic Cancer Translational Research Group, School of Biomedical Sciences, Faculty of Medicine and Health, Lowy Cancer Research CentreThe University of New South WalesSydneyNSWAustralia
| | - David Herrmann
- Cancer Ecosystems ProgramThe Garvan Institute of Medical Research and The Kinghorn Cancer CentreDarlinghurstNSWAustralia
- School of Clinical MedicineSt Vincent's Healthcare Clinical CampusUNSW Medicine & Health, UNSW SydneySydneyAustralia
| | - Janet Youkhana
- Pancreatic Cancer Translational Research Group, School of Biomedical Sciences, Faculty of Medicine and Health, Lowy Cancer Research CentreThe University of New South WalesSydneyNSWAustralia
| | - Paul Timpson
- Cancer Ecosystems ProgramThe Garvan Institute of Medical Research and The Kinghorn Cancer CentreDarlinghurstNSWAustralia
- School of Clinical MedicineSt Vincent's Healthcare Clinical CampusUNSW Medicine & Health, UNSW SydneySydneyAustralia
| | - Phoebe A. Phillips
- Pancreatic Cancer Translational Research Group, School of Biomedical Sciences, Faculty of Medicine and Health, Lowy Cancer Research CentreThe University of New South WalesSydneyNSWAustralia
| | - Maté Biro
- EMBL Australia, Single Molecule Science node, School of Biomedical SciencesThe University of New South WalesSydneyNSWAustralia
- Cancer Ecosystems ProgramThe Garvan Institute of Medical Research and The Kinghorn Cancer CentreDarlinghurstNSWAustralia
| |
Collapse
|
17
|
Zhang Z, Xu H, He J, Hu Q, Liu Y, Xu Z, Lou W, Wu W, Zhang L, Pu N, Shi C, Xu Y, Wang W, Liu L. Inhibition of KLF5 promotes ferroptosis via the ZEB1/HMOX1 axis to enhance sensitivity to oxaliplatin in cancer cells. Cell Death Dis 2025; 16:28. [PMID: 39827156 PMCID: PMC11743205 DOI: 10.1038/s41419-025-07330-8] [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: 05/01/2024] [Revised: 12/04/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
As a novel form of nonapoptotic cell death, ferroptosis is developing into a promising therapeutic target of dedifferentiating and therapy-refractory cancers. However, its application in pancreatic cancer is still unknown. In the preliminary research, we found that F-box and WD repeat domain-containing 7 (FBW7) inhibited the migration and proliferation of pancreatic cancer cells through its substrate c-Myc. We further found that another key substrate of FBW7, KLF5, could inhibit ferroptosis. Inhibiting KLF5 significantly enhances the cytotoxicity of oxaliplatin rather than other chemotherapy drugs. Mechanistically, we found that KLF5 inhibited the expression of heme oxygenase 1 (HMOX1) via repressing zinc finger E-box-binding homeobox 1 (ZEB1). Inhibition of KLF5 facilitated the cytotoxic effect of oxaliplatin via promoting ferroptosis. Oxaliplatin combined with KLF5 inhibitor significantly potentiated cell death in vitro and inhibited tumor growth in vivo compared with either treatment alone. These results reveal a critical role of KLF5 in sensitized chemotherapy of pancreatic cancer, and suggest that ferroptosis combined with platinum-based chemotherapy rather than gemcitabine-based chemotherapy is expected to bring better therapeutic effects.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huaxiang Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Junyi He
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiangsheng Hu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuxin Liu
- Institute of liver diseases, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zijin Xu
- Department of General Surgery, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Wenhui Lou
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenchuan Wu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lei Zhang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ning Pu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chenye Shi
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yaolin Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenquan Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
18
|
Oyon D, Lopez-Pascual A, Castello-Uribe B, Uriarte I, Orsi G, Llorente S, Elurbide J, Adan-Villaescusa E, Valbuena-Goiricelaya E, Irigaray-Miramon A, Latasa MU, Martinez-Perez LA, Bonetti LR, Prosper F, Ponz-Sarvise M, Vicent S, Pineda-Lucena A, Ruiz-Clavijo D, Sangro B, Larracoechea UA, Tian TV, Casadei-Gardini A, Amat I, Arechederra M, Berasain C, Urman JM, Avila MA, Fernandez-Barrena MG. Targeting of the G9a, DNMT1 and UHRF1 epigenetic complex as an effective strategy against pancreatic ductal adenocarcinoma. J Exp Clin Cancer Res 2025; 44:13. [PMID: 39810240 PMCID: PMC11734372 DOI: 10.1186/s13046-024-03268-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 12/24/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with limited treatment options and a poor prognosis. The critical role of epigenetic alterations such as changes in DNA methylation, histones modifications, and chromatin remodeling, in pancreatic tumors progression is becoming increasingly recognized. Moreover, in PDAC these aberrant epigenetic mechanisms can also limit therapy efficacy. This study aimed to investigate the expression and prognostic significance of a key epigenetic complex encompassing DNA methyltransferase-1 (DNMT1), the histone methyltransferase G9a, and the scaffold protein UHRF1 in PDAC. We also evaluated the therapeutic potential of an innovative inhibitor targeting these epigenetic effectors. METHODS Immunohistochemical analysis of DNMT1, G9a, and UHRF1 expression was conducted in human PDAC tissue samples. Staining was semi-quantitatively scored, and overexpression was defined as moderate to strong positivity. The prognostic impact was assessed by correlating protein expression with patient survival. The antitumoral effects of the dual DNMT1-G9a inhibitor CM272 were tested in PDAC cell lines, followed by transcriptomic analyses to identify underlying mechanisms. The in vivo antitumoral efficacy of CM272 was evaluated in PDAC xenograft and syngeneic mouse models, both alone and in combination with anti-PD1 immunotherapy. RESULTS DNMT1, G9a, and UHRF1 were significantly overexpressed in PDAC cells and stroma compared to normal pancreatic tissues. Simultaneous overexpression of the three proteins was associated with significantly reduced survival in resected PDAC patients. CM272 exhibited potent antiproliferative activity in PDAC cell lines, inducing apoptosis and altering key metabolic and cell cycle-related genes. CM272 also enhanced chemotherapy sensitivity and significantly inhibited tumor growth in vivo without detectable toxicity. Combination of CM272 with anti-PD1 therapy further improved antitumor responses and immune cell infiltration, particularly CD4 + and CD8 + T cells. CONCLUSIONS The combined overexpression of DNMT1, G9a, and UHRF1 in PDAC is a strong predictor of poor prognosis. CM272, by targeting this epigenetic complex, shows promising therapeutic potential by inducing apoptosis, reprogramming metabolic pathways, and enhancing immune responses. The combination of CM272 with immunotherapy offers a novel, effective treatment strategy for PDAC.
Collapse
Affiliation(s)
- Daniel Oyon
- Department of Gastroenterology, Galdakao-Usansolo Hospital, Galdakao, Spain
- Biobizkaia Health Research Institute, Bizkaia, Spain
| | - Amaya Lopez-Pascual
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research, IdiSNA, Pamplona, Spain
| | - Borja Castello-Uribe
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
| | - Iker Uriarte
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Giulia Orsi
- Oncology Department, University Hospital of Modena, Modena, Italy
- Department of Oncology, IRCCS San Raffaele Scientific Institute Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Sofia Llorente
- Preclinical and Translational Research Program, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jasmin Elurbide
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Elena Adan-Villaescusa
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
| | | | - Ainara Irigaray-Miramon
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
| | - Maria Ujue Latasa
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
| | - Luz A Martinez-Perez
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
- Universidad de Guadalajara Centro Universitario de Ciencias de La Salud, Guadalajara, Mexico
| | - Luca Reggiani Bonetti
- Department of Medical and Surgical Sciences for Children and Adults, Division of Pathology University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Felipe Prosper
- Navarra Institute for Health Research, IdiSNA, Pamplona, Spain
- Hemato-Oncology Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
- CIBERonc, Instituto de Salud Carlos III, Madrid, Spain
| | - Mariano Ponz-Sarvise
- Navarra Institute for Health Research, IdiSNA, Pamplona, Spain
- CIBERonc, Instituto de Salud Carlos III, Madrid, Spain
- Departments of Oncology and Immunology, Clinica Universidad de Navarra, Pamplona, Spain
| | - Silvestre Vicent
- Navarra Institute for Health Research, IdiSNA, Pamplona, Spain
- CIBERonc, Instituto de Salud Carlos III, Madrid, Spain
- Oncogenes and Effector Targets Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
| | | | - David Ruiz-Clavijo
- Department of Gastroenterology and Hepatology, Navarra University Hospital Complex, Pamplona, Spain
| | - Bruno Sangro
- Navarra Institute for Health Research, IdiSNA, Pamplona, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
- Hepatology Unit, Clínica Universidad de Navarra, CCUN, Pamplona, Spain
| | - Urko Aguirre Larracoechea
- Research Unit, Osakidetza Basque Health Service, Barrualde-Galdakao Integrated Health Organisation, Galdakao-Usansolo Hospital, Galdakao, Spain
| | - Tian V Tian
- Preclinical and Translational Research Program, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Andrea Casadei-Gardini
- Oncology Department, University Hospital of Modena, Modena, Italy
- Department of Oncology, IRCCS San Raffaele Scientific Institute Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Irene Amat
- Navarra Institute for Health Research, IdiSNA, Pamplona, Spain
- Department of Pathology, Navarra University Hospital Complex, Pamplona, Spain
| | - Maria Arechederra
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research, IdiSNA, Pamplona, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Berasain
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Jesus M Urman
- Navarra Institute for Health Research, IdiSNA, Pamplona, Spain
- Department of Gastroenterology and Hepatology, Navarra University Hospital Complex, Pamplona, Spain
| | - Matias A Avila
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research, IdiSNA, Pamplona, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Maite G Fernandez-Barrena
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain.
- Navarra Institute for Health Research, IdiSNA, Pamplona, Spain.
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
19
|
Brandeburg ZC, Waheed SA, Derewonko CA, Dunn CE, Pfeiffer EC, Flusche AME, Sheaff RJ, Lamar AA. Synthesis and Biological Evaluation of N-(1H-Indol-6-ylmethyl)benzenesulfonamide Analogs as Metabolic Inhibitors of Mitochondrial ATP Production in Pancreatic Cancer Cells. ChemMedChem 2025; 20:e202400536. [PMID: 39317650 DOI: 10.1002/cmdc.202400536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/24/2024] [Accepted: 09/24/2024] [Indexed: 09/26/2024]
Abstract
A library of 26 indolyl sulfonamides and 12 amide and ester analogs based upon the 6-indolyl framework has been synthesized in an effort to target pancreatic cancer. The cytotoxicity of the indolyl sulfonamide compounds has been determined using a traditional (48-h compound exposure) assay against 7 pancreatic cancer cell lines and 1 non-cancerous cell line. The potential role of the compounds as metabolic inhibitors of ATP production was evaluated using a rapid screening (2-h compound exposure) assay developed within our laboratories. The IC50 values of the active compounds were determined using the rapid assay and six compounds displayed an IC50 value <5 μM against one or more pancreatic cancer cell lines. The ester analogs also display activity as potential metabolic inhibitors of ATP production with four of the six compounds displaying an IC50 value <5 μM against one or more pancreatic cancer cell lines.
Collapse
Affiliation(s)
- Zachary C Brandeburg
- Department of Chemistry and Biochemistry, The University of Tulsa, 800 South Tucker Drive, Tulsa, OK, 74104, USA
| | - Sakariyau A Waheed
- Department of Chemistry and Biochemistry, The University of Tulsa, 800 South Tucker Drive, Tulsa, OK, 74104, USA
| | - Carina A Derewonko
- Department of Chemistry and Biochemistry, The University of Tulsa, 800 South Tucker Drive, Tulsa, OK, 74104, USA
| | - Caroline E Dunn
- Department of Chemistry and Biochemistry, The University of Tulsa, 800 South Tucker Drive, Tulsa, OK, 74104, USA
| | - Ethan C Pfeiffer
- Department of Chemistry and Biochemistry, The University of Tulsa, 800 South Tucker Drive, Tulsa, OK, 74104, USA
| | - Ann Marie E Flusche
- Department of Chemistry and Biochemistry, The University of Tulsa, 800 South Tucker Drive, Tulsa, OK, 74104, USA
| | - Robert J Sheaff
- Department of Chemistry and Biochemistry, The University of Tulsa, 800 South Tucker Drive, Tulsa, OK, 74104, USA
| | - Angus A Lamar
- Department of Chemistry and Biochemistry, The University of Tulsa, 800 South Tucker Drive, Tulsa, OK, 74104, USA
| |
Collapse
|
20
|
Norton C, Shaw MS, Rubnitz Z, Smith J, Soares HP, Nevala-Plagemann CD, Garrido-Laguna I, Florou V. KRAS Mutation Status and Treatment Outcomes in Patients With Metastatic Pancreatic Adenocarcinoma. JAMA Netw Open 2025; 8:e2453588. [PMID: 39777438 PMCID: PMC11707629 DOI: 10.1001/jamanetworkopen.2024.53588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/05/2024] [Indexed: 01/11/2025] Open
Abstract
Importance Despite the high prevalence of KRAS alterations in pancreatic ductal adenocarcinoma (PDAC), the clinical impact of common KRAS mutations with different cytotoxic regimens is unknown. This evidence is important to inform current treatment and provide a benchmark for emergent targeted KRAS therapies in metastatic PDAC. Objective To assess the clinical implications of common KRAS G12 mutations in PDAC and to compare outcomes of standard-of-care multiagent therapies across these common mutations. Design, Setting, and Participants This retrospective cohort study obtained deidentified clinical data for 5382 patients from a nationwide (US-based) clinicogenomic database. The deidentified data originated from approximately 280 US cancer clinics (approximately 800 sites of care). Patients diagnosed with metastatic PDAC from February 9, 2010, to September 20, 2022, and with sufficient follow-up and treatment data were included. Main Outcomes and Measures Median overall survival (OS) and time to next treatment (TTNT) were calculated for each KRAS mutation group. Hazard ratios (HRs) were generated using multivariate Cox proportional hazards models for KRAS mutations and mutation-treatment combinations. Results A total of 2433 patients with PDAC were included in the analysis (mean age at first treatment, 67.0 [range, 66.0-68.0] years; 1340 male [55.1%]). Among 2023 patients with KRAS mutations, those with G12R had the longest median TTNT (6.0 [95% CI, 5.2-6.6] months) and the longest median OS (13.2 [95% CI, 10.6-15.2] months). Patients with KRAS G12D and G12V mutations had a significantly higher risk of disease progression (HRs, 1.15; [95% CI, 1.04-1.29; P = .009] and 1.16 [95% CI, 1.04-1.30; P = .01], respectively) and death (HRs, 1.29 [95% CI, 1.15-1.45; P < .001] and 1.23 [95% CI, 1.09-1.39; P < .001], respectively) compared with KRAS wild type. The FOLFIRINOX regimen (fluorouracil, irinotecan, oxaliplatin, and leucovorin) had a significantly lower risk of treatment progression and death than gemcitabine with (HRs, 1.19 [95% CI, [1.09-1.29; P < .001] and 1.18 [95% CI, 1.07-1.29; P < .001], respectively) or without (HRs, 1.37 [95% CI, 1.11-1.69; P = .003] and 1.41 [95% CI 1.13-1.75; P = .002], respectively) nab-paclitaxel across all patients. Conclusions and Relevance In this cohort study of 2433 patients with PDAC, KRAS G12D and G12V mutations were associated with worse patient outcomes compared with KRAS wild type. KRAS G12R was associated with more favorable patient outcomes, and FOLFIRINOX was associated with better patient outcomes than gemcitabine-based therapies. These findings highlight the need for more effective systemic therapies for these groups of patients.
Collapse
Affiliation(s)
- Carter Norton
- Huntsman Cancer Institute, University of Utah Health Care, Salt Lake City
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Zachary Rubnitz
- Department of Internal Medicine, Division of Oncology, University of Utah Health Care, Salt Lake City
- Department of Internal Medicine, University of Utah Health Care, Salt Lake City
| | - Jarrod Smith
- Department of Internal Medicine, Division of Oncology, University of Utah Health Care, Salt Lake City
- Department of Internal Medicine, University of Utah Health Care, Salt Lake City
| | - Heloisa P. Soares
- Huntsman Cancer Institute, University of Utah Health Care, Salt Lake City
- Department of Internal Medicine, Division of Oncology, University of Utah Health Care, Salt Lake City
| | - Christopher D. Nevala-Plagemann
- Huntsman Cancer Institute, University of Utah Health Care, Salt Lake City
- Department of Internal Medicine, Division of Oncology, University of Utah Health Care, Salt Lake City
| | - Ignacio Garrido-Laguna
- Huntsman Cancer Institute, University of Utah Health Care, Salt Lake City
- Department of Internal Medicine, Division of Oncology, University of Utah Health Care, Salt Lake City
| | - Vaia Florou
- Huntsman Cancer Institute, University of Utah Health Care, Salt Lake City
- Department of Internal Medicine, Division of Oncology, University of Utah Health Care, Salt Lake City
| |
Collapse
|
21
|
Dai H, Chen X, Yang J, Wang Y, Loiola RA, Lu A, Cheung KCP. Insights and therapeutic advances in pancreatic cancer: the role of electron microscopy in decoding the tumor microenvironment. Front Cell Dev Biol 2024; 12:1460544. [PMID: 39744013 PMCID: PMC11688199 DOI: 10.3389/fcell.2024.1460544] [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: 07/06/2024] [Accepted: 09/23/2024] [Indexed: 01/04/2025] Open
Abstract
Pancreatic cancer is one of the most lethal cancers, with a 5-year overall survival rate of less than 10%. Despite the development of novel therapies in recent decades, current chemotherapeutic strategies offer limited clinical benefits due to the high heterogeneity and desmoplastic tumor microenvironment (TME) of pancreatic cancer as well as inefficient drug penetration. Antibody- and nucleic acid-based targeting therapies have emerged as strong contenders in pancreatic cancer drug discovery. Numerous studies have shown that these strategies can significantly enhance drug accumulation in tumors while reducing systemic toxicity. Additionally, electron microscopy (EM) has been a critical tool for high-resolution analysis of the TME, providing insights into the ultrastructural changes associated with pancreatic cancer progression and treatment responses. This review traces the current and technological advances in EM, particularly the development of ultramicrotomy and improvements in sample preparation that have facilitated the detailed visualization of cellular and extracellular components of the TME. This review highlights the contribution of EM in assessing the efficacy of therapeutic agents, from revealing apoptotic changes to characterizing the effects of novel compounds like ionophore antibiotic gramicidin A on cellular ultrastructures. Moreover, the review delves into the potential of EM in studying the interactions between the tumor microbiome and cancer cell migration, as well as in aiding the development of targeted therapies like antibody-drug conjugates (ADCs) and aptamer-drug conjugates (ApDCs).
Collapse
Affiliation(s)
- Hong Dai
- Department of Chemistry, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
| | - Xingxuan Chen
- Phenome Research Center, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Jiawen Yang
- School of Life Science, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yuying Wang
- Phenome Research Center, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | | | - Aiping Lu
- Phenome Research Center, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Kenneth C. P. Cheung
- Phenome Research Center, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| |
Collapse
|
22
|
Kalli M, Mpekris F, Charalambous A, Michael C, Stylianou C, Voutouri C, Hadjigeorgiou AG, Papoui A, Martin JD, Stylianopoulos T. Mechanical forces inducing oxaliplatin resistance in pancreatic cancer can be targeted by autophagy inhibition. Commun Biol 2024; 7:1581. [PMID: 39604540 PMCID: PMC11603328 DOI: 10.1038/s42003-024-07268-1] [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: 05/08/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
Pancreatic cancer remains one of the most lethal malignancies, with limited treatment options and poor prognosis. A common characteristic among pancreatic cancer patients is the biomechanically altered tumor microenvironment (TME), which among others is responsible for the elevated mechanical stresses in the tumor interior. Although significant research has elucidated the effect of mechanical stress on cancer cell proliferation and migration, it has not yet been investigated how it could affect cancer cell drug sensitivity. Here, we demonstrated that mechanical stress triggers autophagy activation, correlated with increased resistance to oxaliplatin treatment in pancreatic cancer cells. Our results demonstrate that inhibition of autophagy using hydroxychloroquine (HCQ) enhanced the oxaliplatin-induced apoptotic cell death in pancreatic cancer cells exposed to mechanical stress. The combined treatment of HCQ with losartan, a known modulator of mechanical abnormalities in tumors, synergistically enhanced the therapeutic efficacy of oxaliplatin in murine pancreatic tumor models. Furthermore, our study revealed that the use of HCQ enhanced the efficacy of losartan to alleviate mechanical stress levels and restore blood vessel integrity beyond its role in autophagy modulation. These findings underscore the potential of co-targeting mechanical stresses and autophagy as a promising therapeutic strategy to overcome drug resistance and increase chemotherapy efficacy.
Collapse
Affiliation(s)
- Maria Kalli
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| | - Fotios Mpekris
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Antonia Charalambous
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Christina Michael
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Chrystalla Stylianou
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Chrysovalantis Voutouri
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Andreas G Hadjigeorgiou
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Antonia Papoui
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | | | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| |
Collapse
|
23
|
Li R, Liu R, Xu Y, Zhang S, Yang P, Zeng W, Wang H, Liu Y, Yang H, Yue X, Dai Z. Suppressing Pancreatic Cancer Survival and Immune Escape via Nanoparticle-Modulated STING/STAT3 Axis Regulation. Bioconjug Chem 2024; 35:1815-1822. [PMID: 39420541 DOI: 10.1021/acs.bioconjchem.4c00379] [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: 10/19/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) poses a challenge in oncology due to its high lethality and resistance to immunotherapy. Recently, emerging research on the stimulator of interferon gene (STING) pathway offers novel opportunities for immunotherapy. Although STING expression is retained in PDAC cells, the response of PDAC cells to STING agonists remains ineffective. Signal transducer and activator of transcription 3 (STAT3), a downstream pathway of STING, is notably overexpressed in pancreatic cancer and related to tumor survival and immune escape. We observed that inhibiting STAT3 signaling post-STING activation effectively suppressed tumor growth through signal transducer and activator of transcription 1 (STAT1)-mediated apoptosis but led to a potential risk of immune-related adverse events (irAEs). To address this issue, we designed a tumor-penetrating liposome for the codelivery of STING agonist and STAT3 inhibitor. These nanoparticles regulated the STING/STAT3 signaling axis and effectively inhibited the proliferation and survival of tumor. Simultaneously, we found a significant increase in the activation of NK cells and CD8+ T cells after treatment, leading to robust innate immunity and adaptive immune response. We highlight the potential of regulating the STING/STAT3 axis as a promising treatment for improving clinical outcomes in PDAC patients.
Collapse
Affiliation(s)
- Rui Li
- Department of Biomedical Engineering, College of Future Technology, National Biomedical Imaging Canter, Peking University, Beijing 100871, China
| | - Renfa Liu
- Department of Biomedical Engineering, College of Future Technology, National Biomedical Imaging Canter, Peking University, Beijing 100871, China
| | - Yunxue Xu
- Department of Biomedical Engineering, College of Future Technology, National Biomedical Imaging Canter, Peking University, Beijing 100871, China
| | - Shuhao Zhang
- Department of Biomedical Engineering, College of Future Technology, National Biomedical Imaging Canter, Peking University, Beijing 100871, China
| | - Peipei Yang
- Department of Ultrasound, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Wenlong Zeng
- Department of Biomedical Engineering, College of Future Technology, National Biomedical Imaging Canter, Peking University, Beijing 100871, China
| | - Huiyang Wang
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310027, China
| | - Yijia Liu
- Department of Biomedical Engineering, College of Future Technology, National Biomedical Imaging Canter, Peking University, Beijing 100871, China
| | - Huajing Yang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, China
| | - Xiuli Yue
- School of Environment, Harbin Institute of Technology, Harbin 150001, China
| | - Zhifei Dai
- Department of Biomedical Engineering, College of Future Technology, National Biomedical Imaging Canter, Peking University, Beijing 100871, China
| |
Collapse
|
24
|
Fang Y, Liu X, Ren J, Wang X, Zhou F, Huang S, You L, Zhao Y. Integrated analysis of microbiome and metabolome reveals signatures in PDAC tumorigenesis and prognosis. Microbiol Spectr 2024; 12:e0096224. [PMID: 39387592 PMCID: PMC11540152 DOI: 10.1128/spectrum.00962-24] [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: 04/29/2024] [Accepted: 09/16/2024] [Indexed: 10/15/2024] Open
Abstract
Pancreatic cancer, predominantly pancreatic ductal adenocarcinoma (PDAC), is one of the most malignant tumors of the digestive system. Emerging evidence suggests the involvement of the microbiome and metabolic substances in the development of PDAC, yet the results remain contradictory. This study aims to identify the alterations and relationships in intratumoral microbiome and metabolites in PDAC. We collected matched tumor and normal adjacent tissue (NAT) samples from 105 PDAC patients and performed a 6-year follow-up. 2bRAD-M sequencing, untargeted liquid chromatography-tandem mass spectrometry, and untargeted gas chromatography-mass spectrometry were performed. Compared with NATs, microbial α-diversity decreased in PDAC tumors. The relative abundance of Staphylococcus aureus, Cutibacterium acnes, and Cutibacterium granulosum was higher in PDAC tumor after adjusting for confounding factors body mass index and M stage, and the presence of Ralstonia pickettii_B was found associated with a worse overall survival. Metabolomic analysis revealed distinctive differences in composition between PDAC and NAT, with 553 discriminative metabolites identified. Differential metabolites were revealed to originate from the microbiota and showed significant interactions with shifted bacterial species through KO (KEGG Orthology) genes. These findings suggest that the PDAC microenvironment harbors unique microbial-derived enzymatic reactions, potentially influencing the occurrence and development of PDAC by modulating the levels of glycerol-3-phosphate, succinate, carbonate, and beta-alanine. IMPORTANCE We conducted a large sample-size pancreatic adenocarcinoma microbiome study using a novel microbiome sequencing method and two metabolomic assays. Two significant outcomes of our analysis are: (i) commensal opportunistic pathogens Staphylococcus aureus, Cutibacterium acnes, and Cutibacterium granulosum were enriched in pancreatic ductal adenocarcinoma (PDAC) tumors compared with normal adjacent tissues, and (ii) worse overall survival was found related to the presence of Ralstonia pickettii_B. Microbial species affect the tumorigenesis, metastasis, and prognosis of PDAC via unique microbe-enzyme-metabolite interaction. Thus, our study highlights the need for further investigation of the potential associations between pancreatic microbiota-derived omics signatures, which may drive the clinical transformation of microbiome-derived strategies toward therapy-targeted bacteria.
Collapse
Affiliation(s)
- Yuan Fang
- Department of General
Surgery, Peking Union Medical College Hospital, Peking Union Medical
College, Chinese Academy of Medical
Sciences, Beijing,
China
- Key Laboratory of
Research in Pancreatic Tumor, Chinese Academy of Medical
Sciences, Beijing,
China
- National Science and
Technology Key Infrastructure on Translational Medicine in Peking Union
Medical College Hospital,
Beijing, China
- 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,
China
| | - Xiaohong Liu
- Department of General
Surgery, Peking Union Medical College Hospital, Peking Union Medical
College, Chinese Academy of Medical
Sciences, Beijing,
China
- Key Laboratory of
Research in Pancreatic Tumor, Chinese Academy of Medical
Sciences, Beijing,
China
- National Science and
Technology Key Infrastructure on Translational Medicine in Peking Union
Medical College Hospital,
Beijing, China
- 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,
China
| | - Jie Ren
- Department of General
Surgery, Peking Union Medical College Hospital, Peking Union Medical
College, Chinese Academy of Medical
Sciences, Beijing,
China
- Key Laboratory of
Research in Pancreatic Tumor, Chinese Academy of Medical
Sciences, Beijing,
China
- National Science and
Technology Key Infrastructure on Translational Medicine in Peking Union
Medical College Hospital,
Beijing, China
- 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,
China
| | - Xing Wang
- Department of General
Surgery, Peking Union Medical College Hospital, Peking Union Medical
College, Chinese Academy of Medical
Sciences, Beijing,
China
- Key Laboratory of
Research in Pancreatic Tumor, Chinese Academy of Medical
Sciences, Beijing,
China
- National Science and
Technology Key Infrastructure on Translational Medicine in Peking Union
Medical College Hospital,
Beijing, China
- 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,
China
| | - Feihan Zhou
- Department of General
Surgery, Peking Union Medical College Hospital, Peking Union Medical
College, Chinese Academy of Medical
Sciences, Beijing,
China
- Key Laboratory of
Research in Pancreatic Tumor, Chinese Academy of Medical
Sciences, Beijing,
China
- National Science and
Technology Key Infrastructure on Translational Medicine in Peking Union
Medical College Hospital,
Beijing, China
- 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,
China
| | - Shi Huang
- Faculty of Dentistry,
The University of Hong Kong, Hong
Kong SAR, China
| | - Lei You
- Department of General
Surgery, Peking Union Medical College Hospital, Peking Union Medical
College, Chinese Academy of Medical
Sciences, Beijing,
China
- Key Laboratory of
Research in Pancreatic Tumor, Chinese Academy of Medical
Sciences, Beijing,
China
- National Science and
Technology Key Infrastructure on Translational Medicine in Peking Union
Medical College Hospital,
Beijing, China
- 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,
China
| | - Yupei Zhao
- Department of General
Surgery, Peking Union Medical College Hospital, Peking Union Medical
College, Chinese Academy of Medical
Sciences, Beijing,
China
- Key Laboratory of
Research in Pancreatic Tumor, Chinese Academy of Medical
Sciences, Beijing,
China
- National Science and
Technology Key Infrastructure on Translational Medicine in Peking Union
Medical College Hospital,
Beijing, China
- 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,
China
| |
Collapse
|
25
|
Becker JH, Metropulos AE, Spaulding C, Marinelarena AM, Shields MA, Principe DR, Pham TD, Munshi HG. Targeting BCL2 with Venetoclax Enhances the Efficacy of the KRASG12D Inhibitor MRTX1133 in Pancreatic Cancer. Cancer Res 2024; 84:3629-3639. [PMID: 39137400 PMCID: PMC11532783 DOI: 10.1158/0008-5472.can-23-3574] [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: 05/22/2024] [Accepted: 08/08/2024] [Indexed: 08/15/2024]
Abstract
MRTX1133 is currently being evaluated in patients with pancreatic ductal adenocarcinoma (PDAC) tumors harboring a KRASG12D mutation. Combination strategies have the potential to enhance the efficacy of MRTX1133 to further promote cell death and tumor regression. In this study, we demonstrated that MRTX1133 increased the levels of the proapoptotic protein BIM in PDAC cells and conferred sensitivity to the FDA-approved BCL2 inhibitor venetoclax. Combined treatment with MRTX1133 and venetoclax resulted in cell death and growth suppression in 3D cultures. BIM was required for apoptosis induced by the combination treatment. Consistently, BIM was induced in tumors treated with MRTX1133, and venetoclax enhanced the efficacy of MRTX1133 in vivo. Venetoclax could also resensitize MRTX1133-resistant PDAC cells to MRTX1133 in 3D cultures, and tumors established from resistant cells responded to the combination of MRTX1133 and venetoclax. These results provide a rationale for the clinical testing of MRTX1133 and venetoclax in patients with PDAC. Significance: The combination of MRTX1133 and the FDA-approved drug venetoclax promotes cancer cell death and tumor regression in pancreatic ductal adenocarcinoma, providing rationale for testing venetoclax with KRASG12D inhibitors in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Jeffrey H. Becker
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Jesse Brown VA Medical Center, Chicago, Illinois
| | - Anastasia E. Metropulos
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Jesse Brown VA Medical Center, Chicago, Illinois
| | - Christina Spaulding
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Jesse Brown VA Medical Center, Chicago, Illinois
| | | | - Mario A. Shields
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- The Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois
| | - Daniel R. Principe
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Thao D. Pham
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- The Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois
| | - Hidayatullah G. Munshi
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Jesse Brown VA Medical Center, Chicago, Illinois
- The Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois
| |
Collapse
|
26
|
Gicquel T, Marchiano F, Reyes-Castellanos G, Audebert S, Camoin L, Habermann BH, Giannesini B, Carrier A. Integrative study of skeletal muscle mitochondrial dysfunction in a murine pancreatic cancer-induced cachexia model. eLife 2024; 13:RP93312. [PMID: 39422661 PMCID: PMC11488855 DOI: 10.7554/elife.93312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), the most common pancreatic cancer, is a deadly cancer, often diagnosed late and resistant to current therapies. PDAC patients are frequently affected by cachexia characterized by muscle mass and strength loss (sarcopenia) contributing to patient frailty and poor therapeutic response. This study assesses the mechanisms underlying mitochondrial remodeling in the cachectic skeletal muscle, through an integrative exploration combining functional, morphological, and omics-based evaluation of gastrocnemius muscle from KIC genetically engineered mice developing autochthonous pancreatic tumor and cachexia. Cachectic PDAC KIC mice exhibit severe sarcopenia with loss of muscle mass and strength associated with reduced muscle fiber's size and induction of protein degradation processes. Mitochondria in PDAC atrophied muscles show reduced respiratory capacities and structural alterations, associated with deregulation of oxidative phosphorylation and mitochondrial dynamics pathways. Beyond the metabolic pathways known to be altered in sarcopenic muscle (carbohydrates, proteins, and redox), lipid and nucleic acid metabolisms are also affected. Although the number of mitochondria per cell is not altered, mitochondrial mass shows a twofold decrease and the mitochondrial DNA threefold, suggesting a defect in mitochondrial genome homeostasis. In conclusion, this work provides a framework to guide toward the most relevant targets in the clinic to limit PDAC-induced cachexia.
Collapse
Affiliation(s)
- Tristan Gicquel
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCMMarseilleFrance
- Nutrition And Cancer Research Network (NACRe Network)Jouy-en-JosasFrance
| | | | - Gabriela Reyes-Castellanos
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCMMarseilleFrance
- Nutrition And Cancer Research Network (NACRe Network)Jouy-en-JosasFrance
| | - Stephane Audebert
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCMMarseilleFrance
| | - Luc Camoin
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCMMarseilleFrance
| | | | | | - Alice Carrier
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCMMarseilleFrance
- Nutrition And Cancer Research Network (NACRe Network)Jouy-en-JosasFrance
| |
Collapse
|
27
|
Yao W, Wang Y, Zhang X, Lin Y. B3GNT5 is a novel marker correlated with malignant phenotype and poor outcome in pancreatic cancer. iScience 2024; 27:110889. [PMID: 39319269 PMCID: PMC11421285 DOI: 10.1016/j.isci.2024.110889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 06/23/2024] [Accepted: 09/03/2024] [Indexed: 09/26/2024] Open
Abstract
Pancreatic cancer (PC) is one of the most lethal malignancies and new therapeutic strategies are urgently needed. β1,3-N-acetylglucosaminyltransferase V (B3GNT5) may be a potential option for cancer treatment, but its role in PC remains unknown. In this study, we first demonstrated through bioinformatics analysis that B3GNT5 was high expression in PC and predicted poor prognosis. We further constructed B3GNT5 overexpression or knockdown cell lines by employing lentivirus packaging techniques and confirmed that B3GNT5 could promote tumor cell viability and autonomous growth using cultured cells and vivo xenograft models. In addition, we found that knockdown of B3GNT5 in PC cells inhibited cell migration, invasion, and angiogenesis, as well as stemness of cancer stem cells and enhanced chemotherapy sensitivity to gemcitabine. Mechanistically, overexpression of the transcription factor STAT5B in PC cells enhanced the transcriptional activity of the B3GNT5 promoter. Our work confirmed a tumor-promotive role of B3GNT5 in PC pathogenesis.
Collapse
Affiliation(s)
- Wei Yao
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, P.R. China
| | - Yihui Wang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, P.R. China
| | - Xin Zhang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, P.R. China
| | - Yuhe Lin
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, P.R. China
| |
Collapse
|
28
|
Jiang W, Liu L, Wang M, Li X, Zhou T, Hou X, Qiao L, Chen C, Zuo D, Liu J, Ren L. KLF15 suppresses stemness of pancreatic cancer by decreasing USP21-mediated Nanog stability. Cell Mol Life Sci 2024; 81:417. [PMID: 39367978 PMCID: PMC11455850 DOI: 10.1007/s00018-024-05442-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/08/2024] [Accepted: 08/22/2024] [Indexed: 10/07/2024]
Abstract
The existence of cancer stem cells (CSCs) in pancreatic ductal adenocarcinoma (PDAC) is considered to be the key factor for metastasis and chemoresistance. Thus, novel therapeutic strategies for eradicating CSCs are urgently needed. Here we aimed to explore the role of KLF15 in stemness and the feasibility of using KLF15 to inhibit CSCs and improve chemotherapy sensitivity in PDAC. In this study, we report that KLF15 is negatively associated with poor survival and advanced pathological staging of PDAC. Moreover, tumorous KLF15 suppresses the stemness of PDAC by promoting the degradation of Nanog, and KLF15 directly interacts with Nanog, inhibiting interaction between Nanog with USP21. We also demonstrate that the KLF15/Nanog complex inhibit the stemness in vivo and in PDX cells. Tazemetostat suppresses stemness and sensitizes PDAC cells to gemcitabine by promoting KLF15 expression in PDAC. In summary, the findings of our study confirm the value of KLF15 level in diagnosis and prognosis of PDAC, it is the first time to explore the inhibition role of KLF15 in stemness of PDAC and the regulation mechanism of Nanog, contributing to provide a new therapeutic strategy that using Tazemetostat sensitizes PDAC cells to gemcitabine by promoting KLF15 expression for PDAC.
Collapse
Affiliation(s)
- Wenna Jiang
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Hexi District, Huanhuxi Road, Tianjin, 300060, China
| | - Lin Liu
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Hexi District, Huanhuxi Road, Tianjin, 300060, China
| | - Meng Wang
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Hexi District, Huanhuxi Road, Tianjin, 300060, China
| | - Xueyang Li
- Department of Pancreatic Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Tianxing Zhou
- Department of Pancreatic Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Xupeng Hou
- Department of Pancreatic Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Lu Qiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences of Tianjin Medical University, Tianjin, China
| | - Chong Chen
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Hexi District, Huanhuxi Road, Tianjin, 300060, China
| | - Duo Zuo
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Hexi District, Huanhuxi Road, Tianjin, 300060, China
| | - Jing Liu
- Department of Breast Oncoplastic Surgery, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Li Ren
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Hexi District, Huanhuxi Road, Tianjin, 300060, China.
| |
Collapse
|
29
|
Lu S, Wang C, Ma J, Wang Y. Metabolic mediators: microbial-derived metabolites as key regulators of anti-tumor immunity, immunotherapy, and chemotherapy. Front Immunol 2024; 15:1456030. [PMID: 39351241 PMCID: PMC11439727 DOI: 10.3389/fimmu.2024.1456030] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/27/2024] [Indexed: 10/04/2024] Open
Abstract
The human microbiome has recently emerged as a focal point in cancer research, specifically in anti-tumor immunity, immunotherapy, and chemotherapy. This review explores microbial-derived metabolites, emphasizing their crucial roles in shaping fundamental aspects of cancer treatment. Metabolites such as short-chain fatty acids (SCFAs), Trimethylamine N-Oxide (TMAO), and Tryptophan Metabolites take the spotlight, underscoring their diverse origins and functions and their profound impact on the host immune system. The focus is on SCFAs' remarkable ability to modulate immune responses, reduce inflammation, and enhance anti-tumor immunity within the intricate tumor microenvironment (TME). The review critically evaluates TMAO, intricately tied to dietary choices and gut microbiota composition, assessing its implications for cancer susceptibility, progression, and immunosuppression. Additionally, the involvement of tryptophan and other amino acid metabolites in shaping immune responses is discussed, highlighting their influence on immune checkpoints, immunosuppression, and immunotherapy effectiveness. The examination extends to their dynamic interaction with chemotherapy, emphasizing the potential of microbial-derived metabolites to alter treatment protocols and optimize outcomes for cancer patients. A comprehensive understanding of their role in cancer therapy is attained by exploring their impacts on drug metabolism, therapeutic responses, and resistance development. In conclusion, this review underscores the pivotal contributions of microbial-derived metabolites in regulating anti-tumor immunity, immunotherapy responses, and chemotherapy outcomes. By illuminating the intricate interactions between these metabolites and cancer therapy, the article enhances our understanding of cancer biology, paving the way for the development of more effective treatment options in the ongoing battle against cancer.
Collapse
Affiliation(s)
- Shan Lu
- Department of General Practice, The Second Hospital of Jilin University, Changchun, China
| | - Chunling Wang
- Medical Affairs Department, The Second Hospital of Jilin University, Changchun, China
| | - Jingru Ma
- Department of Clinical Laboratory, the Second Hospital of Jilin University, Changchun, China
| | - Yichao Wang
- Department of Obstetrics and Gynecology, the Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
30
|
DePuccio MJ, Shiu-Yee K, Kurien NA, Sarna A, Waterman BL, Rush LJ, McAlearney AS, Ejaz A. A qualitative study of providers' perspectives on cross-institutional care coordination for pancreatic cancer: challenges and opportunities. BMC Health Serv Res 2024; 24:1041. [PMID: 39251999 PMCID: PMC11382481 DOI: 10.1186/s12913-024-11483-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 08/22/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Despite calls for regionalizing pancreatic cancer (PC) care to high-volume centers (HVCs), many patients with PC elect to receive therapy closer to their home or at multiple institutions. In the context of cross-institutional PC care, the challenges associated with coordinating care are poorly understood. METHODS In this qualitative study we conducted semi-structured interviews with oncology clinicians from a HVC (n = 9) and community-based hospitals (n = 11) to assess their perspectives related to coordinating the care of and treating PC patients across their respective institutions. Interviews were transcribed, coded, and analyzed using deductive and inductive approaches to identify themes related to cross-institutional coordination challenges and to note improvement opportunities. RESULTS Clinicians identified challenges associated with closed-loop communication due, in part, to not having access to a shared electronic health record. Challenges with patient co-management were attributed to patients receiving inconsistent recommendations from different clinicians. To address these challenges, participants suggested several improvement opportunities such as building rapport with clinicians across institutions and updating tumor board processes. The opportunity to update tumor board processes was reportedly multi-dimensional and could involve: (1) designating a tumor board coordinator; (2) documenting and disseminating tumor board recommendations; and (3) using teleconferencing to facilitate community-based clinician engagement during tumor board meetings. CONCLUSIONS In light of communication barriers and challenges associated with patient co-management, enabling the development of relationships among PC clinicians and improving the practices of multidisciplinary tumor boards could potentially foster cross-institutional coordination. Research examining how multidisciplinary tumor board coordinators and teleconferencing platforms could enhance cross-institutional communication and thereby improve patient outcomes is warranted.
Collapse
Affiliation(s)
- Matthew J DePuccio
- The Center for the Advancement of Team Science, Analytics, and Systems Thinking in Health Services and Implementation Science Research (CATALYST), College of Medicine, The Ohio State University, 700 Ackerman Rd, Suite 4000, Columbus, OH, 43202, USA
- Department of Health Systems Management, Rush University, Chicago, IL, USA
| | - Karen Shiu-Yee
- The Center for the Advancement of Team Science, Analytics, and Systems Thinking in Health Services and Implementation Science Research (CATALYST), College of Medicine, The Ohio State University, 700 Ackerman Rd, Suite 4000, Columbus, OH, 43202, USA
| | - Natasha A Kurien
- The Center for the Advancement of Team Science, Analytics, and Systems Thinking in Health Services and Implementation Science Research (CATALYST), College of Medicine, The Ohio State University, 700 Ackerman Rd, Suite 4000, Columbus, OH, 43202, USA
| | - Angela Sarna
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center, 320 W. 10th Avenue, M-260 Starling Loving Hall, Columbus, OH, 43210, USA
| | - Brittany L Waterman
- Division of Palliative Medicine, College of Medicine, The Ohio State University, McCampbell Hall, 5th floor, 1581 Dodd Dr, Columbus, OH, 43210, USA
| | - Laura J Rush
- The Center for the Advancement of Team Science, Analytics, and Systems Thinking in Health Services and Implementation Science Research (CATALYST), College of Medicine, The Ohio State University, 700 Ackerman Rd, Suite 4000, Columbus, OH, 43202, USA
| | - Ann Scheck McAlearney
- The Center for the Advancement of Team Science, Analytics, and Systems Thinking in Health Services and Implementation Science Research (CATALYST), College of Medicine, The Ohio State University, 700 Ackerman Rd, Suite 4000, Columbus, OH, 43202, USA
- Department of Family and Community Medicine, College of Medicine, The Ohio State University, 700 Ackerman Rd, Suite 4000, Columbus, OH, 43210, USA
| | - Aslam Ejaz
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center, 320 W. 10th Avenue, M-260 Starling Loving Hall, Columbus, OH, 43210, USA.
- Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
31
|
Li S, Liu Y, Sui X, Zhuo Y, Siqi H, Sijia Z, Hui Z, Dihua L, Dapeng Z, Lei Y. Novel Tubeimoside I liposomal drug delivery system in combination with gemcitabine for the treatment of pancreatic cancer. Nanomedicine (Lond) 2024; 19:1977-1993. [PMID: 39225145 PMCID: PMC11485868 DOI: 10.1080/17435889.2024.2382076] [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: 04/16/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
Aim: To evaluate the anti-pancreatic cancer effect of novel Tubeimoside I multifunctional liposomes combined with gemcitabine.Methods: Liposomes were prepared through the thin film hydration method, with evaluations conducted on parameters including encapsulation efficiency (EE%), particle size, polydispersity index (PDI), zeta potential (ZP), storage stability, and release over a 7-day period. The cellular uptake rate, therapeutic efficacy in vitro and in vivo and the role of immune microenvironment modulation were evaluated.Results: The novel Tubeimoside I multifunctional liposomal exhibited good stability, significant anti-cancer activity, and immune microenvironment remodeling effects. Furthermore, it showed a safety profile.Conclusion: This study underscores the potential of Novel Tubeimoside I multifunctional liposomal as a promising treatment option for pancreatic cancer.
Collapse
Affiliation(s)
- Shuhui Li
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yuansheng Liu
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, 300000, China
| | - Xiaojun Sui
- Tianjin Key Laboratory of Organ Injury and ITCWM Repair Associated with Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, China
- Graduate School ofTianjin Medical University, Tianjin, 300270, China
| | - Yuzhen Zhuo
- Tianjin Key Laboratory of Organ Injury and ITCWM Repair Associated with Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, China
| | - He Siqi
- Graduate School ofTianjin Medical University, Tianjin, 300270, China
| | - Zhang Sijia
- Graduate School ofTianjin Medical University, Tianjin, 300270, China
| | - Zhang Hui
- Tianjin Key Laboratory of Organ Injury and ITCWM Repair Associated with Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, China
| | - Li Dihua
- Tianjin Key Laboratory of Organ Injury and ITCWM Repair Associated with Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, China
| | - Zhang Dapeng
- Tianjin Key Laboratory of Organ Injury and ITCWM Repair Associated with Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, China
| | - Yang Lei
- Tianjin Key Laboratory of Organ Injury and ITCWM Repair Associated with Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, China
| |
Collapse
|
32
|
Sarfraz Z, Sarfraz A, Farooq MD, Khalid M, Cheema K, Javad F, Khan T, Pervaiz Z, Sarfraz M, Jaan A, Sadiq S, Anwar J. The Current Landscape of Clinical Trials for Immunotherapy in Pancreatic Cancer: A State-of-the-Art Review. J Gastrointest Cancer 2024; 55:1026-1057. [PMID: 38976079 DOI: 10.1007/s12029-024-01078-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2024] [Indexed: 07/09/2024]
Abstract
BACKGROUND Pancreatic cancer remains a lethal malignancy with a 5-year survival rate below 6% and about 500,000 deaths annually worldwide. Pancreatic adenocarcinoma, the most prevalent form, is commonly associated with diabetes, chronic pancreatitis, obesity, and smoking, mainly affecting individuals aged 60 to 80 years. This systematic review aims to evaluate the efficacy of immunotherapeutic approaches in the treatment of pancreatic cancer. METHODS A systematic search was conducted to identify clinical trials (Phases I-III) assessing immunotherapy in pancreatic cancer in PubMed/Medline, CINAHL, Scopus, and Web of Science, adhering to PRISMA Statement 2020 guidelines. The final search was completed on May 25, 2024. Ongoing trials were sourced from ClinicalTrials.gov and the World Health Organization's International Clinical Trials Registry Platform (ICTRP). Keywords such as "pancreatic," "immunotherapy," "cancer," and "clinical trial" were used across databases. Gray literature was excluded. RESULTS Phase I trials, involving 337 patients, reported a median overall survival (OS) of 13.6 months (IQR: 5-62.5 months) and a median progression-free survival (PFS) of 5.1 months (IQR: 1.9-11.7 months). Phase II/III trials pooled in a total of 1463 participants had a median OS of 12.2 months (IQR: 2.5-35.55 months) and a median PFS of 8.8 months (IQR: 1.4-33.51 months). CONCLUSIONS Immunotherapy shows potential for extending survival among pancreatic cancer patients, though results vary. The immunosuppressive nature of the tumor microenvironment and diverse patient responses underline the need for further research to optimize these therapeutic strategies.
Collapse
Affiliation(s)
- Zouina Sarfraz
- Department of Medicine, Fatima Jinnah Medical University, Queen's Road, Mozang Chungi, Lahore, Pakistan.
| | | | | | - Musfira Khalid
- Department of Medicine, Fatima Jinnah Medical University, Queen's Road, Mozang Chungi, Lahore, Pakistan
| | | | | | - Taleah Khan
- CMH Lahore Medical College and Institute of Dentistry, Lahore, Pakistan
| | - Zainab Pervaiz
- CMH Lahore Medical College and Institute of Dentistry, Lahore, Pakistan
| | | | - Ali Jaan
- Rochester General Hospital, Rochester, NY, USA
| | | | - Junaid Anwar
- Baptist Hospitals of Southeast Texas, Beaumont, TX, USA
| |
Collapse
|
33
|
Ba Q, Wang X, Lu Y. Establishment of a prognostic model for pancreatic cancer based on mitochondrial metabolism related genes. Discov Oncol 2024; 15:376. [PMID: 39196457 PMCID: PMC11358576 DOI: 10.1007/s12672-024-01255-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 08/20/2024] [Indexed: 08/29/2024] Open
Abstract
AIM Pancreatic ductal adenocarcinoma (PAAD) is recognized as an exceptionally aggressive cancer that both highly lethal and unfavorable prognosis. The mitochondrial metabolism pathway is intimately involved in oncogenesis and tumor progression, however, much remains unknown in this area. In this study, the bioinformatic tools have been used to construct a prognostic model with mitochondrial metabolism-related genes (MMRGs) to evaluate the survival, immune status, mutation profile, and drug sensitivity of PAAD patients. METHOD Univariate Cox regression and LASSO regression were used to screen the differentially expressed genes (DEGs), and multivariate Cox regression was used to develop the risk model. Kaplan-Meier estimator was employed to identify MMRGs signatures associated with overall survival (OS). ROC curves were utilized to evaluate the model's performance. Maftools, immunedeconv and CIBERSORT R packages were applied to analyze the gene mutation profiles and immune status. The corresponding sensitivity to pharmaceutical agents was assessed using oncoPredict R packages. RESULTS A prognostic model with five MMRGs was developed, which defined the patients as high-risk showed lower survival rates. There was good consistency among individuals categorized as high-risk, showing elevated rates of genetic alterations, particularly in the TP53 and KRAS genes. Furthermore, these patients exhibited increased levels of immunosuppression, characterized by an increased presence of macrophages, neutrophils, Th2 cells, and regulatory T cells. Additionally, high-risk patients showed increased sensitivity to Sabutoclax and Venetoclax. CONCLUSION By utilizing a gene signature associated with mitochondrial metabolism, a prognostic model has been established which could be a highly efficient method for predicting the outcomes of PAAD patients.
Collapse
Affiliation(s)
- Qinwen Ba
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiong Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yanjun Lu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
34
|
Vogt M, Dudvarski Stankovic N, Cruz Garcia Y, Hofstetter J, Schneider K, Kuybu F, Hauck T, Adhikari B, Hamann A, Rocca Y, Grysczyk L, Martin B, Gebhardt-Wolf A, Wiegering A, Diefenbacher M, Gasteiger G, Knapp S, Saur D, Eilers M, Rosenfeldt M, Erhard F, Vos SM, Wolf E. Targeting MYC effector functions in pancreatic cancer by inhibiting the ATPase RUVBL1/2. Gut 2024; 73:1509-1528. [PMID: 38821858 PMCID: PMC11347226 DOI: 10.1136/gutjnl-2023-331519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/15/2024] [Indexed: 06/02/2024]
Abstract
OBJECTIVE The hallmark oncogene MYC drives the progression of most tumours, but direct inhibition of MYC by a small-molecule drug has not reached clinical testing. MYC is a transcription factor that depends on several binding partners to function. We therefore explored the possibility of targeting MYC via its interactome in pancreatic ductal adenocarcinoma (PDAC). DESIGN To identify the most suitable targets among all MYC binding partners, we constructed a targeted shRNA library and performed screens in cultured PDAC cells and tumours in mice. RESULTS Unexpectedly, many MYC binding partners were found to be important for cultured PDAC cells but dispensable in vivo. However, some were also essential for tumours in their natural environment and, among these, the ATPases RUVBL1 and RUVBL2 ranked first. Degradation of RUVBL1 by the auxin-degron system led to the arrest of cultured PDAC cells but not untransformed cells and to complete tumour regression in mice, which was preceded by immune cell infiltration. Mechanistically, RUVBL1 was required for MYC to establish oncogenic and immunoevasive gene expression identifying the RUVBL1/2 complex as a druggable vulnerability in MYC-driven cancer. CONCLUSION One implication of our study is that PDAC cell dependencies are strongly influenced by the environment, so genetic screens should be performed in vitro and in vivo. Moreover, the auxin-degron system can be applied in a PDAC model, allowing target validation in living mice. Finally, by revealing the nuclear functions of the RUVBL1/2 complex, our study presents a pharmaceutical strategy to render pancreatic cancers potentially susceptible to immunotherapy.
Collapse
Affiliation(s)
- Markus Vogt
- Cancer Systems Biology Group, Chair of Biochemistry and Molecular Biology, Theodor Boveri Institute, University of Würzburg, Würzburg, Germany
- Institute of Biochemistry, University of Kiel, Kiel, Germany
| | - Nevenka Dudvarski Stankovic
- Cancer Systems Biology Group, Chair of Biochemistry and Molecular Biology, Theodor Boveri Institute, University of Würzburg, Würzburg, Germany
- Institute of Biochemistry, University of Kiel, Kiel, Germany
| | - Yiliam Cruz Garcia
- Cancer Systems Biology Group, Chair of Biochemistry and Molecular Biology, Theodor Boveri Institute, University of Würzburg, Würzburg, Germany
- Institute of Biochemistry, University of Kiel, Kiel, Germany
| | - Julia Hofstetter
- Cancer Systems Biology Group, Chair of Biochemistry and Molecular Biology, Theodor Boveri Institute, University of Würzburg, Würzburg, Germany
| | - Katharina Schneider
- Cancer Systems Biology Group, Chair of Biochemistry and Molecular Biology, Theodor Boveri Institute, University of Würzburg, Würzburg, Germany
- Institute of Biochemistry, University of Kiel, Kiel, Germany
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Filiz Kuybu
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Theresa Hauck
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Bikash Adhikari
- Cancer Systems Biology Group, Chair of Biochemistry and Molecular Biology, Theodor Boveri Institute, University of Würzburg, Würzburg, Germany
- Institute of Biochemistry, University of Kiel, Kiel, Germany
| | - Anton Hamann
- Institute for Pharmaceutical Chemistry, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Yamila Rocca
- Max Planck Research Group and Institute of Systems Immunology, University of Würzburg, Würzburg, Germany
| | - Lara Grysczyk
- Cancer Systems Biology Group, Chair of Biochemistry and Molecular Biology, Theodor Boveri Institute, University of Würzburg, Würzburg, Germany
| | - Benedikt Martin
- Cancer Systems Biology Group, Chair of Biochemistry and Molecular Biology, Theodor Boveri Institute, University of Würzburg, Würzburg, Germany
| | - Anneli Gebhardt-Wolf
- Chair of Biochemistry and Molecular Biology, Theodor Boveri Institute, University of Würzburg, Würzburg, Germany
| | - Armin Wiegering
- Chair of Biochemistry and Molecular Biology, Theodor Boveri Institute, University of Würzburg, Würzburg, Germany
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Markus Diefenbacher
- Comprehensive Pneumology Center (CPC)/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL/CPC-M), Munich, Germany
- Ludwig-Maximilian-Universität München (LMU), Munich, Germany
| | - Georg Gasteiger
- Max Planck Research Group and Institute of Systems Immunology, University of Würzburg, Würzburg, Germany
| | - Stefan Knapp
- Institute for Pharmaceutical Chemistry, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Dieter Saur
- Institute of Translational Cancer Research, TUM School of Medicine and Health, Munich, Germany
| | - Martin Eilers
- Chair of Biochemistry and Molecular Biology, Theodor Boveri Institute, University of Würzburg, Würzburg, Germany
| | | | - Florian Erhard
- Computational Systems Virology and Bioinformatics, Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Seychelle M Vos
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Elmar Wolf
- Cancer Systems Biology Group, Chair of Biochemistry and Molecular Biology, Theodor Boveri Institute, University of Würzburg, Würzburg, Germany
- Institute of Biochemistry, University of Kiel, Kiel, Germany
| |
Collapse
|
35
|
Gordon JW, Chen HY, Nickles T, Lee PM, Bok R, Ohliger MA, Okamoto K, Ko AH, Larson PEZ, Wang ZJ. Hyperpolarized 13C Metabolic MRI of Patients with Pancreatic Ductal Adenocarcinoma. J Magn Reson Imaging 2024; 60:741-749. [PMID: 38041836 PMCID: PMC11144260 DOI: 10.1002/jmri.29162] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 12/04/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDA) is the third leading cause of cancer-related death in the United States. However, early response assessment using the current approach of measuring changes in tumor size on computed tomography (CT) or MRI is challenging. PURPOSE To investigate the feasibility of hyperpolarized (HP) [1-13C]pyruvate MRI to quantify metabolism in the normal appearing pancreas and PDA, and to assess changes in PDA metabolism following systemic chemotherapy. STUDY TYPE Prospective. SUBJECTS Six patients (65.0 ± 7.6 years, 2 females) with locally advanced or metastatic PDA enrolled prior to starting a new line of systemic chemotherapy. FIELD STRENGTH/SEQUENCE 3-T, T1-weighted gradient echo, metabolite-selective 13C echoplanar imaging. ASSESSMENT Time-resolved HP [1-13C]pyruvate data were acquired before (N = 6) and 4-weeks after (N = 3) treatment initiation. Pyruvate metabolism, as quantified by pharmacokinetic modeling and metabolite area-under-the-curve ratios, was assessed in manually segmented PDA and normal appearing pancreas ROIs (N = 5). The change in tumor metabolism before and 4-weeks after treatment initiation was assessed in primary PDA (N = 2) and liver metastases (N = 1), and was compared to objective tumor response defined by response evaluation criteria in solid tumors (RECIST) on subsequent CTs. STATISTICAL TESTS Descriptive tests (mean ± standard deviation), model fit error for pharmacokinetic rate constants. RESULTS Primary PDA showed reduced alanine-to-lactate ratios when compared to normal pancreas, due to increased lactate-to-pyruvate or reduced alanine-to-pyruvate ratios. Of the three patients who received HP [1-13C]pyruvate MRI before and 4-weeks after treatment initiation, one patient had a primary tumor with early metabolic response (increase in alanine-to-lactate) and subsequent partial response according to RECIST, one patient had a primary tumor with relatively stable metabolism and subsequent stable disease by RECIST, and one patient had metastatic PDA with increase in lactate-to-pyruvate of the liver metastases and corresponding progressive disease according to RECIST. DATA CONCLUSION Altered pyruvate metabolism with increased lactate or reduced alanine was observed in the primary tumor. Early metabolic response assessed at 4-weeks after treatment initiation correlated with subsequent objective tumor response assessed using RECIST. LEVEL OF EVIDENCE 2 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Jeremy W Gordon
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Hsin-Yu Chen
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Tanner Nickles
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Philip M Lee
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Robert Bok
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Michael A Ohliger
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Kimberly Okamoto
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Andrew H Ko
- Department of Medicine, University of California, San Francisco, California, USA
| | - Peder E Z Larson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, California, USA
| | - Zhen J Wang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, California, USA
| |
Collapse
|
36
|
Sun Y, Jiang W, Liao X, Wang D. Hallmarks of perineural invasion in pancreatic ductal adenocarcinoma: new biological dimensions. Front Oncol 2024; 14:1421067. [PMID: 39119085 PMCID: PMC11307098 DOI: 10.3389/fonc.2024.1421067] [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: 04/21/2024] [Accepted: 07/01/2024] [Indexed: 08/10/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignant tumor with a high metastatic potential. Perineural invasion (PNI) occurs in the early stages of PDAC with a high incidence rate and is directly associated with a poor prognosis. It involves close interaction among PDAC cells, nerves and the tumor microenvironment. In this review, we detailed discuss PNI-related pain, six specific steps of PNI, and treatment of PDAC with PNI and emphasize the importance of novel technologies for further investigation.
Collapse
Affiliation(s)
- Yaquan Sun
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, China
| | - Wei Jiang
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, China
| | - Xiang Liao
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, China
| | - Dongqing Wang
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, China
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
37
|
Mukund A, Afridi MA, Karolak A, Park MA, Permuth JB, Rasool G. Pancreatic Ductal Adenocarcinoma (PDAC): A Review of Recent Advancements Enabled by Artificial Intelligence. Cancers (Basel) 2024; 16:2240. [PMID: 38927945 PMCID: PMC11201559 DOI: 10.3390/cancers16122240] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/03/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) remains one of the most formidable challenges in oncology, characterized by its late detection and poor prognosis. Artificial intelligence (AI) and machine learning (ML) are emerging as pivotal tools in revolutionizing PDAC care across various dimensions. Consequently, many studies have focused on using AI to improve the standard of PDAC care. This review article attempts to consolidate the literature from the past five years to identify high-impact, novel, and meaningful studies focusing on their transformative potential in PDAC management. Our analysis spans a broad spectrum of applications, including but not limited to patient risk stratification, early detection, and prediction of treatment outcomes, thereby highlighting AI's potential role in enhancing the quality and precision of PDAC care. By categorizing the literature into discrete sections reflective of a patient's journey from screening and diagnosis through treatment and survivorship, this review offers a comprehensive examination of AI-driven methodologies in addressing the multifaceted challenges of PDAC. Each study is summarized by explaining the dataset, ML model, evaluation metrics, and impact the study has on improving PDAC-related outcomes. We also discuss prevailing obstacles and limitations inherent in the application of AI within the PDAC context, offering insightful perspectives on potential future directions and innovations.
Collapse
Affiliation(s)
- Ashwin Mukund
- Department of Machine Learning, Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA; (A.M.); (A.K.)
| | - Muhammad Ali Afridi
- School of Electrical Engineering and Computer Science (SEECS), National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan;
| | - Aleksandra Karolak
- Department of Machine Learning, Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA; (A.M.); (A.K.)
| | - Margaret A. Park
- Departments of Cancer Epidemiology and Gastrointestinal Oncology, Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA; (M.A.P.); (J.B.P.)
| | - Jennifer B. Permuth
- Departments of Cancer Epidemiology and Gastrointestinal Oncology, Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA; (M.A.P.); (J.B.P.)
| | - Ghulam Rasool
- Department of Machine Learning, Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA; (A.M.); (A.K.)
| |
Collapse
|
38
|
Qin Q, Yu R, Eriksson JE, Tsai HI, Zhu H. Cancer-associated fibroblasts in pancreatic ductal adenocarcinoma therapy: Challenges and opportunities. Cancer Lett 2024; 591:216859. [PMID: 38615928 DOI: 10.1016/j.canlet.2024.216859] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/16/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a solid organ malignancy with a high mortality rate. Statistics indicate that its incidence has been increasing as well as the associated deaths. Most patients with PDAC show poor response to therapies making the clinical management of this cancer difficult. Stromal cells in the tumor microenvironment (TME) contribute to the development of resistance to therapy in PDAC cancer cells. Cancer-associated fibroblasts (CAFs), the most prevalent stromal cells in the TME, promote a desmoplastic response, produce extracellular matrix proteins and cytokines, and directly influence the biological behavior of cancer cells. These multifaceted effects make it difficult to eradicate tumor cells from the body. As a result, CAF-targeting synergistic therapeutic strategies have gained increasing attention in recent years. However, due to the substantial heterogeneity in CAF origin, definition, and function, as well as high plasticity, majority of the available CAF-targeting therapeutic approaches are not effective, and in some cases, they exacerbate disease progression. This review primarily elucidates on the effect of CAFs on therapeutic efficiency of various treatment modalities, including chemotherapy, radiotherapy, immunotherapy, and targeted therapy. Strategies for CAF targeting therapies are also discussed.
Collapse
Affiliation(s)
- Qin Qin
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, 212001, China
| | - Rong Yu
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, 212001, China
| | - John E Eriksson
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, FI-20520 Finland
| | - Hsiang-I Tsai
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, 212001, China; Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| | - Haitao Zhu
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, 212001, China; Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| |
Collapse
|
39
|
Feineis D, Bringmann G. Structural variety and pharmacological potential of naphthylisoquinoline alkaloids. THE ALKALOIDS. CHEMISTRY AND BIOLOGY 2024; 91:1-410. [PMID: 38811064 DOI: 10.1016/bs.alkal.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Naphthylisoquinoline alkaloids are a fascinating class of natural biaryl compounds. They show characteristic mono- and dimeric scaffolds, with chiral axes and stereogenic centers. Since the appearance of the last comprehensive overview on these secondary plant metabolites in this series in 1995, the number of discovered representatives has tremendously increased to more than 280 examples known today. Many novel-type compounds have meanwhile been discovered, among them naphthylisoquinoline-related follow-up products like e.g., the first seco-type (i.e., ring-opened) and ring-contracted analogues. As highlighted in this review, the knowledge on the broad structural chemodiversity of naphthylisoquinoline alkaloids has been decisively driven forward by extensive phytochemical studies on the metabolite pattern of Ancistrocladus abbreviatus from Coastal West Africa, which is a particularly "creative" plant. These investigations furnished a considerable number of more than 80-mostly new-natural products from this single species, with promising antiplasmodial activities and with pronounced cytotoxic effects against human leukemia, pancreatic, cervical, and breast cancer cells. Another unique feature of naphthylisoquinoline alkaloids is their unprecedented biosynthetic origin from polyketidic precursors and not, as usual for isoquinoline alkaloids, from aromatic amino acids-a striking example of biosynthetic convergence in nature. Furthermore, remarkable botanical results are presented on the natural producers of naphthylisoquinoline alkaloids, the paleotropical Dioncophyllaceae and Ancistrocladaceae lianas, including first investigations on the chemoecological role of these plant metabolites and their storage and accumulation in particular plant organs.
Collapse
Affiliation(s)
- Doris Feineis
- Institute of Organic Chemistry, University of Würzburg, Würzburg, Germany
| | - Gerhard Bringmann
- Institute of Organic Chemistry, University of Würzburg, Würzburg, Germany.
| |
Collapse
|
40
|
Zheng R, Liu X, Zhang Y, Liu Y, Wang Y, Guo S, Jin X, Zhang J, Guan Y, Liu Y. Frontiers and future of immunotherapy for pancreatic cancer: from molecular mechanisms to clinical application. Front Immunol 2024; 15:1383978. [PMID: 38756774 PMCID: PMC11096556 DOI: 10.3389/fimmu.2024.1383978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/22/2024] [Indexed: 05/18/2024] Open
Abstract
Pancreatic cancer is a highly aggressive malignant tumor, that is becoming increasingly common in recent years. Despite advances in intensive treatment modalities including surgery, radiotherapy, biological therapy, and targeted therapy, the overall survival rate has not significantly improved in patients with pancreatic cancer. This may be attributed to the insidious onset, unknown pathophysiology, and poor prognosis of the disease. It is therefore essential to identify and develop more effective and safer treatments for pancreatic cancer. Tumor immunotherapy is the new and fourth pillar of anti-tumor therapy after surgery, radiotherapy, and chemotherapy. Significant progress has made in the use of immunotherapy for a wide variety of malignant tumors in recent years; a breakthrough has also been made in the treatment of pancreatic cancer. This review describes the advances in immune checkpoint inhibitors, cancer vaccines, adoptive cell therapy, oncolytic virus, and matrix-depletion therapies for the treatment of pancreatic cancer. At the same time, some new potential biomarkers and potential immunotherapy combinations for pancreatic cancer are discussed. The molecular mechanisms of various immunotherapies have also been elucidated, and their clinical applications have been highlighted. The current challenges associated with immunotherapy and proposed strategies that hold promise in overcoming these limitations have also been discussed, with the aim of offering new insights into immunotherapy for pancreatic cancer.
Collapse
Affiliation(s)
- Rui Zheng
- Department of Medical Immunology, Medical College of Yan’an University, Yanan, Shaanxi, China
| | - Xiaobin Liu
- Department of Medical Immunology, Medical College of Yan’an University, Yanan, Shaanxi, China
| | - Yufu Zhang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Yan’an University, Yan’an, Shaanxi, China
| | - Yongxian Liu
- Department of Medical Immunology, Medical College of Yan’an University, Yanan, Shaanxi, China
| | - Yaping Wang
- Department of Medical Immunology, Medical College of Yan’an University, Yanan, Shaanxi, China
| | - Shutong Guo
- Department of Medical Immunology, Medical College of Yan’an University, Yanan, Shaanxi, China
| | - Xiaoyan Jin
- Department of Medical Immunology, Medical College of Yan’an University, Yanan, Shaanxi, China
| | - Jing Zhang
- Department of Medical Immunology, Medical College of Yan’an University, Yanan, Shaanxi, China
| | - Yuehong Guan
- Department of Medical Immunology, Medical College of Yan’an University, Yanan, Shaanxi, China
| | - Yusi Liu
- Department of Medical Immunology, Medical College of Yan’an University, Yanan, Shaanxi, China
| |
Collapse
|
41
|
Ding M, Yang Y, Zhang Z, Liu H, Dai Y, Wang Z, Ma S, Liu Y, Wang Q. Structural characterization of the polysaccharide from the black crystal region of Inonotus obliquus and its effect on AsPC-1 and SW1990 pancreatic cancer cell apoptosis. Int J Biol Macromol 2024; 268:131891. [PMID: 38677687 DOI: 10.1016/j.ijbiomac.2024.131891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 04/09/2024] [Accepted: 04/24/2024] [Indexed: 04/29/2024]
Abstract
In this study, one water soluble polysaccharide (IOP1-1) with a weight average molecular weight of 6886 Da was obtained from the black crystal region of Inonotus obliquus by hot water extraction, DEAE-52 cellulose extraction and Sephadex-100 column chromatography purification. Structural analysis indicated that IOP1-1 was a glucan with a main chain composed of α-Glcp-(1 → 4)-α-Glcp-(1 → 4)-β-Glcp-(1 → 4)-β-Glcp-(1 → 4)-α-Glcp-(1 → 6)-β-Glcp-(1 → 4)-α-Glcp-(1 → 3)-β-Glcp-(1→. The CCK-8 assay results showed that IOP1-1 inhibited AsPC-1 and SW1990 pancreatic cancer cell proliferation in a concentration-dependent manner. Flow cytometric analysis revealed that IOP1-1 induced cell cycle arrest in AsPC-1 and SW1990 cells. Hoechst 33342 staining and Annexin V-FITC/PI double staining analysis showed that IOP1-1 could induce apoptosis in AsPC-1 and SW1990 cells. Furthermore, western blot analysis confirmed that IOP1-1 could induce apoptosis in AsPC-1 and SW1990 pancreatic cancer cells through three pathways: the mitochondrial pathway, the death receptor pathway, and endoplasmic reticulum stress. According to these research data, IOP1-1 may be utilized as an adjuvant treatment to anticancer medications, opening up new application prospects and opportunities.
Collapse
Affiliation(s)
- Miao Ding
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; College of Plant Protection, Jilin Agricultural University, Changchun 130118, China
| | - Yu Yang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; College of Life Science, Jilin Agricultural University, Changchun 130118, China
| | - Ziyang Zhang
- College of Plant Protection, Jilin Agricultural University, Changchun 130118, China
| | - Hongxiang Liu
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; College of Plant Protection, Jilin Agricultural University, Changchun 130118, China
| | - Yingdi Dai
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; College of Plant Protection, Jilin Agricultural University, Changchun 130118, China
| | - Zixuan Wang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; College of Plant Protection, Jilin Agricultural University, Changchun 130118, China
| | - Sijia Ma
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; College of Plant Protection, Jilin Agricultural University, Changchun 130118, China
| | - Yang Liu
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; College of Plant Protection, Jilin Agricultural University, Changchun 130118, China.
| | - Qi Wang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; College of Plant Protection, Jilin Agricultural University, Changchun 130118, China.
| |
Collapse
|
42
|
Zhu H, Xu J, Wang W, Zhang B, Liu J, Liang C, Hua J, Meng Q, Yu X, Shi S. Intratumoral CD38 +CD19 +B cells associate with poor clinical outcomes and immunosuppression in patients with pancreatic ductal adenocarcinoma. EBioMedicine 2024; 103:105098. [PMID: 38608514 PMCID: PMC11017281 DOI: 10.1016/j.ebiom.2024.105098] [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: 08/22/2023] [Revised: 03/13/2024] [Accepted: 03/19/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND The widespread involvement of tumor-infiltrating B cells highlights their potential role in tumor behavior. However, B cell heterogeneity in PDAC remains unexplored. Studying TIL-Bs in PDAC aims to identify new treatment strategies. METHODS We performed single-cell RNA sequencing to study the heterogeneity of B cells in PDAC. The prognostic and immunologic value of the identified CD38+ B cells was explored in FUSCC (n = 147) and TCGA (n = 176) cohorts. Flow cytometry was conducted to characterize the relationship between CD38+ B cells and other immune cells, as well as their phenotypic features. In vitro and in vivo experiments were performed to assess the putative effect of CD38+ B cells on antitumor immunity. FINDINGS The presence of CD38+ B cells in PDAC was associated with unfavorable clinicopathological features and poorer overall survival (p < 0.001). Increased infiltration of CD38+ B cells was accompanied by reduced natural killer (NK) cells (p = 0.021) and increased regulatory T cells (p = 0.016). Molecular profiling revealed high expression of IL-10, IL-35, TGF-β, GZMB, TIM-1, CD5 and CD21, confirming their putative regulatory B cell-like features. Co-culture experiments demonstrated suppression of NK cell cytotoxicity by CD38+ B cell-derived IL-10 (p < 0.001). Finally, in vivo experiments suggested adoptive transfer of CD38+ B cells reduced antitumor immunity and administration of a CD38 inhibitor hampered tumor growth (p < 0.001). INTERPRETATION We discovered regulatory B cell-like CD38+ B cell infiltration as an independent prognostic factor in PDAC. The use of CD38 inhibitor may provide new possibilities for PDAC immunotherapy. FUNDING This study was supported by the National Natural Science Foundation of China (U21A20374), Shanghai Municipal Science and Technology Major Project (21JC1401500), Scientific Innovation Project of Shanghai Education Committee (2019-01-07-00-07-E00057), Special Project for Clinical Research in the Health Industry of the Shanghai Health Commission (No. 20204Y0265) and Natural Science Foundation of Shanghai (23ZR1479300).
Collapse
MESH Headings
- Humans
- ADP-ribosyl Cyclase 1/metabolism
- Carcinoma, Pancreatic Ductal/immunology
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/mortality
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/therapy
- Animals
- Mice
- Prognosis
- Antigens, CD19/metabolism
- Antigens, CD19/immunology
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/mortality
- Female
- Male
- Membrane Glycoproteins/metabolism
- Membrane Glycoproteins/genetics
- Cell Line, Tumor
- Tumor Microenvironment/immunology
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Middle Aged
- B-Lymphocyte Subsets/immunology
- B-Lymphocyte Subsets/metabolism
- Immunosuppression Therapy
Collapse
Affiliation(s)
- Heng Zhu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
| | - Wei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
| | - Bo Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
| | - Jiang Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
| | - Chen Liang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
| | - Jie Hua
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
| | - Qingcai Meng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China; Shanghai Pancreatic Cancer Institute, 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; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
| |
Collapse
|
43
|
Xia Y, Ma J, Yang X, Liu D, Zhu Y, Zhao Y, Fei X, Xu D, Dai J. Identifying the Spatial Architecture That Restricts the Proximity of CD8 + T Cells to Tumor Cells in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2024; 16:1434. [PMID: 38611111 PMCID: PMC11010991 DOI: 10.3390/cancers16071434] [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: 03/10/2024] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/14/2024] Open
Abstract
The anti-tumor function of CD8+ T cells is dependent on their proximity to tumor cells. Current studies have focused on the infiltration level of CD8+ T cells in the tumor microenvironment, while further spatial information, such as spatial localization and inter-cellular communication, have not been defined. In this study, co-detection by indexing (CODEX) was designed to characterize PDAC tissue regions with seven protein markers in order to identify the spatial architecture that regulates CD8+ T cells in human pancreatic ductal adenocarcinoma (PDAC). The cellular neighborhood algorithm was used to identify a total of six conserved and distinct cellular neighborhoods. Among these, one unique spatial architecture of CD8+ T and CD4+ T cell-enriched neighborhoods enriched the majority of CD8+ T cells, but heralded a poor prognosis. The proximity analysis revealed that the CD8+ T cells in this spatial architecture were significantly closer to themselves and the CD4+ T cells than to the tumor cells. Collectively, we identified a unique spatial architecture that restricted the proximity of CD8+ T cells to tumor cells in the tumor microenvironment, indicating a novel immune evasion mechanism of pancreatic ductal adenocarcinoma in a topologically regulated manner and providing new insights into the biology of PDAC.
Collapse
Affiliation(s)
- Yihan Xia
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (Y.X.); (J.M.); (X.Y.); (D.L.); (Y.Z.); (Y.Z.); (X.F.)
- College of Health Sciences and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Junrui Ma
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (Y.X.); (J.M.); (X.Y.); (D.L.); (Y.Z.); (Y.Z.); (X.F.)
- College of Health Sciences and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaobao Yang
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (Y.X.); (J.M.); (X.Y.); (D.L.); (Y.Z.); (Y.Z.); (X.F.)
- College of Health Sciences and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Danping Liu
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (Y.X.); (J.M.); (X.Y.); (D.L.); (Y.Z.); (Y.Z.); (X.F.)
- College of Health Sciences and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yujie Zhu
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (Y.X.); (J.M.); (X.Y.); (D.L.); (Y.Z.); (Y.Z.); (X.F.)
- College of Health Sciences and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yanan Zhao
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (Y.X.); (J.M.); (X.Y.); (D.L.); (Y.Z.); (Y.Z.); (X.F.)
- College of Health Sciences and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xuefeng Fei
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (Y.X.); (J.M.); (X.Y.); (D.L.); (Y.Z.); (Y.Z.); (X.F.)
- College of Health Sciences and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Dakang Xu
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (Y.X.); (J.M.); (X.Y.); (D.L.); (Y.Z.); (Y.Z.); (X.F.)
- College of Health Sciences and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jing Dai
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (Y.X.); (J.M.); (X.Y.); (D.L.); (Y.Z.); (Y.Z.); (X.F.)
- College of Health Sciences and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
44
|
Brozos-Vázquez E, Toledano-Fonseca M, Costa-Fraga N, García-Ortiz MV, Díaz-Lagares Á, Rodríguez-Ariza A, Aranda E, López-López R. Pancreatic cancer biomarkers: A pathway to advance in personalized treatment selection. Cancer Treat Rev 2024; 125:102719. [PMID: 38490088 DOI: 10.1016/j.ctrv.2024.102719] [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: 01/08/2024] [Revised: 03/08/2024] [Accepted: 03/10/2024] [Indexed: 03/17/2024]
Abstract
Pancreatic cancer is one of the tumors with the worst prognosis, and unlike other cancers, few advances have been made in recent years. The only curative option is surgery, but only 15-20% of patients are candidates, with a high risk of relapse. In advanced pancreatic cancer there are few first-line treatment options and no validated biomarkers for better treatment selection. The development of targeted therapies in pancreatic cancer is increasingly feasible due to tumor-agnostic treatments, such as PARP inhibitors in patients with BRCA1, BRCA2 or PALB2 alterations or immunotherapies in patients with high microsatellite instability/tumor mutational burden. In addition, other therapeutic molecules have been developed for patients with KRAS G12C mutation or fusions in NTRK or NRG1. Consequently, there has been a growing interest in biomarkers that may help guide targeted therapy in pancreatic cancer. Therefore, this review aims to offer an updated perspective on biomarkers with therapeutic potential in pancreatic cancer.
Collapse
Affiliation(s)
- Elena Brozos-Vázquez
- Medical Oncology Department, University Hospital of A Coruña (CHUAC), A Coruña, Spain
| | - Marta Toledano-Fonseca
- Cancer Network Biomedical Research Center (CIBERONC), Madrid, Spain; Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
| | - Nicolás Costa-Fraga
- Epigenomics Unit, Cancer Epigenomics, Translational Medical Oncology Group (ONCOMET); Clinical University Hospital & Health Research Institute of Santiago de Compostela. CIBERONC; University of Santiago de Compostela, Santiago de Compostela, Spain
| | - María Victoria García-Ortiz
- Cancer Network Biomedical Research Center (CIBERONC), Madrid, Spain; Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
| | - Ángel Díaz-Lagares
- Epigenomics Unit, Cancer Epigenomics, Translational Medical Oncology Group (ONCOMET); Clinical University Hospital & Health Research Institute of Santiago de Compostela. CIBERONC; Department of Clinical Analysis, University Hospital Complex of Santiago de Compostela (CHUS), Santiago de Compostela, Spain
| | - Antonio Rodríguez-Ariza
- Cancer Network Biomedical Research Center (CIBERONC), Madrid, Spain; Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain; Medical Oncology Department, Reina Sofía University Hospital, Córdoba, Spain.
| | - Enrique Aranda
- Cancer Network Biomedical Research Center (CIBERONC), Madrid, Spain; Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain; Medical Oncology Department, Reina Sofía University Hospital, Córdoba, Spain; Department of Medicine, Faculty of Medicine, University of Córdoba, Córdoba, Spain
| | - Rafael López-López
- Clinical University Hospital & Health Research Institute of Santiago de Compostela. CIBERONC; Medical Oncology Department & Translational Medical Oncology Group-ONCOMET, Spain; Oncology at Santiago de Compostela School of Medicine, Spain
| |
Collapse
|
45
|
Guo S, Wang Z. Unveiling the immunosuppressive landscape of pancreatic ductal adenocarcinoma: implications for innovative immunotherapy strategies. Front Oncol 2024; 14:1349308. [PMID: 38590651 PMCID: PMC10999533 DOI: 10.3389/fonc.2024.1349308] [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: 12/04/2023] [Accepted: 03/12/2024] [Indexed: 04/10/2024] Open
Abstract
Pancreatic cancer, particularly pancreatic ductal adenocarcinoma (PDAC), stands as the fourth leading cause of cancer-related deaths in the United States, marked by challenging treatment and dismal prognoses. As immunotherapy emerges as a promising avenue for mitigating PDAC's malignant progression, a comprehensive understanding of the tumor's immunosuppressive characteristics becomes imperative. This paper systematically delves into the intricate immunosuppressive network within PDAC, spotlighting the significant crosstalk between immunosuppressive cells and factors in the hypoxic acidic pancreatic tumor microenvironment. By elucidating these mechanisms, we aim to provide insights into potential immunotherapy strategies and treatment targets, laying the groundwork for future studies on PDAC immunosuppression. Recognizing the profound impact of immunosuppression on PDAC invasion and metastasis, this discussion aims to catalyze the development of more effective and targeted immunotherapies for PDAC patients.
Collapse
Affiliation(s)
- Songyu Guo
- First Clinical Medical College, Inner Mongolia Medical University, Hohhot, China
- Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Zhenxia Wang
- Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
46
|
Xiao C, Li J, Hua A, Wang X, Li S, Li Z, Xu C, Zhang Z, Yang X, Li Z. Hyperbaric Oxygen Boosts Antitumor Efficacy of Copper-Diethyldithiocarbamate Nanoparticles against Pancreatic Ductal Adenocarcinoma by Regulating Cancer Stem Cell Metabolism. RESEARCH (WASHINGTON, D.C.) 2024; 7:0335. [PMID: 38766644 PMCID: PMC11100349 DOI: 10.34133/research.0335] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 02/16/2024] [Indexed: 05/22/2024]
Abstract
Cuproptosis-based cancer nanomedicine has received widespread attention recently. However, cuproptosis nanomedicine against pancreatic ductal adenocarcinoma (PDAC) is severely limited by cancer stem cells (CSCs), which reside in the hypoxic stroma and adopt glycolysis metabolism accordingly to resist cuproptosis-induced mitochondria damage. Here, we leverage hyperbaric oxygen (HBO) to regulate CSC metabolism by overcoming tumor hypoxia and to augment CSC elimination efficacy of polydopamine and hydroxyethyl starch stabilized copper-diethyldithiocarbamate nanoparticles (CuET@PH NPs). Mechanistically, while HBO and CuET@PH NPs inhibit glycolysis and oxidative phosphorylation, respectively, the combination of HBO and CuET@PH NPs potently suppresses energy metabolism of CSCs, thereby achieving robust tumor inhibition of PDAC and elongating mice survival importantly. This study reveals novel insights into the effects of cuproptosis nanomedicine on PDAC CSC metabolism and suggests that the combination of HBO with cuproptosis nanomedicine holds significant clinical translation potential for PDAC patients.
Collapse
Affiliation(s)
- Chen Xiao
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Jiayuan Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Ao Hua
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Xing Wang
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Shiyou Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Zheng Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Chen Xu
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Zhijie Zhang
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Xiangliang Yang
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- National Engineering Research Center for Nanomedicine,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Key Laboratory of Molecular Biophysics of Ministry of Education,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Hubei Bioinformatics and Molecular Imaging Key Laboratory,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Zifu Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- National Engineering Research Center for Nanomedicine,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Key Laboratory of Molecular Biophysics of Ministry of Education,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Hubei Bioinformatics and Molecular Imaging Key Laboratory,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| |
Collapse
|
47
|
Ye X, Yu Y, Zheng X, Ma H. Clinical immunotherapy in pancreatic cancer. Cancer Immunol Immunother 2024; 73:64. [PMID: 38430289 PMCID: PMC10908626 DOI: 10.1007/s00262-024-03632-6] [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: 10/07/2023] [Accepted: 01/09/2024] [Indexed: 03/03/2024]
Abstract
Pancreatic cancer remains a challenging disease with limited treatment options, resulting in high mortality rates. The predominant approach to managing pancreatic cancer patients continues to be systemic cytotoxic chemotherapy. Despite substantial advancements in immunotherapy strategies for various cancers, their clinical utility in pancreatic cancer has proven less effective and durable. Whether administered as monotherapy, employing immune checkpoint inhibitors, tumor vaccines, chimeric antigen receptors T cells, or in combination with conventional chemoradiotherapy, the clinical outcomes remain underwhelming. Extensive preclinical experiments and clinical trials in the realm of pancreatic cancer have provided valuable insights into the complexities of immunotherapy. Chief among the hurdles are the immunosuppressive tumor microenvironment, limited immunogenicity, and the inherent heterogeneity of pancreatic cancer. In this comprehensive review, we provide an overview and critical analysis of current clinical immunotherapy strategies for pancreatic cancer, emphasizing their endeavors to overcome immunotherapy resistance. Particular focus is placed on strategies aimed at reshaping the immunosuppressive microenvironment and enhancing T cell-mediated tumor cell killing. Ultimately, through deeper elucidation of the underlying pathogenic mechanisms of pancreatic cancer and the refinement of therapeutic approaches, we anticipate breakthroughs that will pave the way for more effective treatments in this challenging disease.
Collapse
Affiliation(s)
- Xiaorong Ye
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui Province, People's Republic of China
| | - Yue Yu
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui Province, People's Republic of China.
| | - Xiaohu Zheng
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui Province, People's Republic of China.
- Hefei National Research Center for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People's Republic of China.
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People's Republic of China.
| | - Hongdi Ma
- Hefei National Research Center for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People's Republic of China.
- Department of Pediatrics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui Province, People's Republic of China.
| |
Collapse
|
48
|
Wang N, Jiang Y, Li M, Wang H, Pan J, Tang Y, Xie S, Xu Y, Li X, Zhou X, Xu P, Lin W, Wang X. Protein Kinase STK24 Promotes Tumor Immune Evasion via the AKT-PD-L1 Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304342. [PMID: 38229183 PMCID: PMC10966517 DOI: 10.1002/advs.202304342] [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: 06/28/2023] [Revised: 12/27/2023] [Indexed: 01/18/2024]
Abstract
Immunotherapy targeting PD-L1 is still ineffective for a wide variety of tumors with high unpredictability. Deploying combined immunotherapy with alternative targeting is practical to overcome this therapeutic resistance. Here, the deficiency of serine-threonine kinase STK24 is observed in tumor cells causing substantial attenuation of tumor growth in murine syngeneic models, a process relying on cytotoxic CD8+ T and NK cells. Mechanistically, STK24 in tumor cells associates with and directly phosphorylates AKT at Thr21, which promotes AKT activation and subsequent PD-L1 induction. Deletion or inhibition of STK24, by contrast, blocks IFN-γ-mediated PD-L1 expression. Various murine models indicate that in vivo silencing of STK24 can significantly enhance the efficacy of the anti-PD-1 blockade strategy. Elevated STK24 levels are observed in patient specimens in multiple tumor types and inversely correlated with intratumoral infiltration of cytotoxic CD8+ T cells and with patient survival. The study collectively identifies STK24 as a critical modulator of antitumor immunity, which engages in AKT and PD-L1/PD-1 signaling and is a promising target for combined immunotherapy.
Collapse
Affiliation(s)
- Ning Wang
- Institute of Immunology and Bone Marrow Transplantation CenterThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiang310058China
| | - Yu Jiang
- Department of Clinical LaboratorySecond Affiliated Hospital of Zhejiang UniversitySchool of MedicineHangzhouZhejiang310058China
| | - Mengjie Li
- Institute of Immunology and Bone Marrow Transplantation CenterThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiang310058China
| | - Haofei Wang
- Institute of Immunology and Bone Marrow Transplantation CenterThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiang310058China
| | - Jie Pan
- Institute of Immunology and Bone Marrow Transplantation CenterThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiang310058China
| | - Yang Tang
- Institute of Immunology and Bone Marrow Transplantation CenterThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiang310058China
| | - Shaofang Xie
- Westlake Laboratory of Life Sciences and BiomedicineSchool of Life SciencesWestlake UniversityHangzhouZhejiang310024China
| | - Yunyang Xu
- Institute of Immunology and Bone Marrow Transplantation CenterThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiang310058China
| | - Xu Li
- Westlake Laboratory of Life Sciences and BiomedicineSchool of Life SciencesWestlake UniversityHangzhouZhejiang310024China
| | - Xuefei Zhou
- Department of PharmacologySchool of MedicineZhejiang UniversityHangzhouZhejiang310058China
| | - Pinglong Xu
- Life Sciences InstituteZhejiang UniversityHangzhouZhejiang310058China
| | - Wenlong Lin
- Institute of Immunology and Bone Marrow Transplantation CenterThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiang310058China
| | - Xiaojian Wang
- Institute of Immunology and Bone Marrow Transplantation CenterThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiang310058China
| |
Collapse
|
49
|
Sun H, Ge Y, Liu J, Li Z, Li H, Zhao T, Wang X, Feng Y, Wang H, Gao S, Shi L, Yang S, Sun P, Chang A, Hao J, Huang C. Tumor-derived interleukin 35 mediates the dissemination of gemcitabine resistance in pancreatic adenocarcinoma. Oncogene 2024; 43:776-788. [PMID: 38243080 DOI: 10.1038/s41388-024-02938-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 12/28/2023] [Accepted: 01/04/2024] [Indexed: 01/21/2024]
Abstract
Rapid development of drug resistance after chemotherapy is a major cause of treatment failure in individuals with pancreatic ductal adenocarcinoma (PDAC). In this study, we illustrate that tumor-derived interleukin 35 (IL-35) mediates the accelerated resistance of PDAC to gemcitabine (GEM). We observe that GEM resistance can spread from GEM-resistant PDAC cells to GEM-sensitive cells, and that IL-35 is responsible for the propagation of chemoresistance, which is supported by sequencing and experimental data. Additionally, we discover that GEM-resistant cells have significantly higher levels of IL-35 expression. Mechanistically, aberrantly expressed IL-35 triggers transcriptional activation of SOD2 expression via GP130-STAT1 signaling, scavenging reactive oxygen species (ROS) and leading to GEM resistance. Furthermore, GEM treatment stimulates IL-35 expression through activation of the NF-κB pathway, resulting in acquired chemoresistance. In the mouse model, a neutralizing antibody against IL-35 enhances the tumor suppressive effect of GEM. Collectively, our data suggests that IL-35 is critical in mediating GEM resistance in pancreatic cancer, and therefore could be a valuable therapeutic target in overcoming PDAC chemoresistance.
Collapse
Affiliation(s)
- Huizhi Sun
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yi Ge
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Jing Liu
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Zengxun Li
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Hui Li
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Tiansuo Zhao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Xiuchao Wang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yukuan Feng
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Hongwei Wang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Song Gao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Lei Shi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Shengyu Yang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Department of Cellular and Molecular Physiology, the Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Peiqing Sun
- Department of Cancer Biology, Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Antao Chang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
| | - Jihui Hao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
| | - Chongbiao Huang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
| |
Collapse
|
50
|
Kern J, Schilling D, Schneeweis C, Schmid RM, Schneider G, Combs SE, Dobiasch S. Identification of the unfolded protein response pathway as target for radiosensitization in pancreatic cancer. Radiother Oncol 2024; 191:110059. [PMID: 38135186 DOI: 10.1016/j.radonc.2023.110059] [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: 07/31/2023] [Revised: 12/12/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND AND PURPOSE Due to the high intrinsic radioresistance of pancreatic ductal adenocarcinoma (PDAC), radiotherapy (RT) is only beneficial in 30% of patients. Therefore, this study aimed to identify targets to improve the efficacy of RT in PDAC. MATERIALS AND METHODS Alamar Blue proliferation and colony formation assay (CFA) were used to determine the radioresponse of a cohort of 38 murine PDAC cell lines. A gene set enrichment analysis was performed to reveal differentially expressed pathways. CFA, cell cycle distribution, γH2AX FACS analysis, and Caspase 3/7 SYTOX assay were used to examine the effect of a combination treatment using KIRA8 as an IRE1α-inhibitor and Ceapin-A7 as an inhibitor against ATF6. RESULTS The unfolded protein response (UPR) was identified as a pathway highly expressed in radioresistant cell lines. Using the IRE1α-inhibitor KIRA8 or the ATF6-inhibitor Ceapin-A7 in combination with radiation, a radiosensitizing effect was observed in radioresistant cell lines, but no substantial alteration of the radioresponse in radiosensitive cell lines. Mechanistically, increased apoptosis by KIRA8 in combination with radiation and a cell cycle arrest in the G1 phase after ATF6 inhibition and radiation have been observed in radioresistant cell lines. CONCLUSION So, our data show evidence that the UPR is involved in radioresistance of PDAC. Increased apoptosis and a G1 cell cycle arrest seem to be responsible for the radiosensitizing effect of UPR inhibition. These findings are supportive for developing novel combination treatment concepts in PDAC to overcome radioresistance.
Collapse
Affiliation(s)
- Jana Kern
- Department of Radiation Oncology, School of Medicine, Klinikum rechts der Isar, Technical University Munich (TUM), Munich, Germany
| | - Daniela Schilling
- Department of Radiation Oncology, School of Medicine, Klinikum rechts der Isar, Technical University Munich (TUM), Munich, Germany; Institute of Radiation Medicine (IRM), Department of Radiation Sciences, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Christian Schneeweis
- Department of Medicine II, School of Medicine, Klinikum rechts der Isar, Technical University Munich (TUM), Munich, Germany
| | - Roland M Schmid
- Department of Medicine II, School of Medicine, Klinikum rechts der Isar, Technical University Munich (TUM), Munich, Germany
| | - Günter Schneider
- Department of Medicine II, School of Medicine, Klinikum rechts der Isar, Technical University Munich (TUM), Munich, Germany; Department of General Visceral and Pediatric Surgery, University Medical Center Göttingen, Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, School of Medicine, Klinikum rechts der Isar, Technical University Munich (TUM), Munich, Germany; Institute of Radiation Medicine (IRM), Department of Radiation Sciences, Helmholtz Zentrum Munich, Neuherberg, Germany; German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Sophie Dobiasch
- Department of Radiation Oncology, School of Medicine, Klinikum rechts der Isar, Technical University Munich (TUM), Munich, Germany; Institute of Radiation Medicine (IRM), Department of Radiation Sciences, Helmholtz Zentrum Munich, Neuherberg, Germany; German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.
| |
Collapse
|