1
|
Rademaker G, Hernandez GA, Seo Y, Dahal S, Miller-Phillips L, Li AL, Peng XL, Luan C, Qiu L, Liegeois MA, Wang B, Wen KW, Kim GE, Collisson EA, Kruger SF, Boeck S, Ormanns S, Guenther M, Heinemann V, Haas M, Looney MR, Yeh JJ, Zoncu R, Perera RM. PCSK9 drives sterol-dependent metastatic organ choice in pancreatic cancer. Nature 2025:10.1038/s41586-025-09017-8. [PMID: 40399683 DOI: 10.1038/s41586-025-09017-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 04/14/2025] [Indexed: 05/23/2025]
Abstract
To grow at distant sites, metastatic cells must overcome major challenges posed by the unique cellular and metabolic composition of secondary organs1. Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease that metastasizes to the liver and lungs. Despite evidence of metabolic reprogramming away from the primary site, the key drivers that dictate the ability of PDAC cells to colonize the liver or lungs and survive there remain undefined. Here we identified PCSK9 as predictive of liver versus lung colonization by integrating metastatic tropism data of human PDAC cell lines2, in vivo metastasis modelling in mice and gene expression correlation analysis. PCSK9 negatively regulates low density lipoprotein (LDL)-cholesterol import and, accordingly, PCSK9-low PDAC cells preferentially colonize LDL-rich liver tissue. LDL-cholesterol taken up by liver-avid PCSK9-low cells supports activation of pro-growth mTORC1 activation at the lysosome, and through conversion into the signalling oxysterol, 24(S)-hydroxycholesterol, reprogrammes the microenvironment to release nutrients from neighbouring hepatocytes. Conversely, PCSK9-high, lung-avid PDAC cells rely on transcriptional upregulation of the distal cholesterol synthesis pathway to generate intermediates-7-dehydrocholesterol and 7-dehydrodesmosterol-with protective action against ferroptosis, a vulnerability in the oxygen-rich microenvironment of the lung. Increasing the amount of PCSK9 redirected liver-avid cells to the lung whereas ablating PCSK9 drove lung-avid cells to the liver, thereby establishing PCSK9 as necessary and sufficient for secondary organ site preference. Our studies reveal PCSK9-driven differential utilization of the distal cholesterol synthesis pathway as a key and potentially actionable driver of metastatic growth in PDAC.
Collapse
Affiliation(s)
- Gilles Rademaker
- Department of Anatomy, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Grace A Hernandez
- Department of Anatomy, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Yurim Seo
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Sumena Dahal
- Department of Anatomy, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Lisa Miller-Phillips
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
- Department of Hematology/Oncology, LMU Klinikum, University of Munich Comprehensive Cancer Center, Munich, Germany
| | - Alexander L Li
- Department of Anatomy, University of California San Francisco, San Francisco, CA, USA
| | - Xianlu Laura Peng
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Changfei Luan
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Longhui Qiu
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Maude A Liegeois
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Bruce Wang
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Kwun W Wen
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Grace E Kim
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | | | - Stephan F Kruger
- Department of Hematology/Oncology, LMU Klinikum, University of Munich Comprehensive Cancer Center, Munich, Germany
| | - Stefan Boeck
- Department of Hematology/Oncology, LMU Klinikum, University of Munich Comprehensive Cancer Center, Munich, Germany
- Department of Hematology and Oncology, München Klinik Neuperlach, Munich, Germany
- German Cancer Consortium (DKTK), Munich, Germany
| | - Steffen Ormanns
- Institute of General Pathology, Medical University Innsbruck, Innsbruck, Austria
- Innpath Institute for Pathology, Tirol Kliniken, Innsbruck, Austria
| | - Michael Guenther
- Institute of General Pathology, Medical University Innsbruck, Innsbruck, Austria
- Innpath Institute for Pathology, Tirol Kliniken, Innsbruck, Austria
| | - Volker Heinemann
- Department of Hematology/Oncology, LMU Klinikum, University of Munich Comprehensive Cancer Center, Munich, Germany
| | - Michael Haas
- Department of Hematology/Oncology, LMU Klinikum, University of Munich Comprehensive Cancer Center, Munich, Germany
- Department of Hematology and Oncology, München Klinik Neuperlach, Munich, Germany
| | - Mark R Looney
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Jen Jen Yeh
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Departments of Surgery and Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Roberto Zoncu
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA
| | - Rushika M Perera
- Department of Anatomy, University of California San Francisco, San Francisco, CA, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA.
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
2
|
Zhang J, Kong X, Zhou B, Li R, Yu Z, Zhu J, Xi Q, Li Y, Zhao Z, Zhang R. Metabolic reprogramming of drug resistance in pancreatic cancer: mechanisms and effects. Mol Aspects Med 2025; 103:101368. [PMID: 40398192 DOI: 10.1016/j.mam.2025.101368] [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: 01/19/2025] [Revised: 05/10/2025] [Accepted: 05/10/2025] [Indexed: 05/23/2025]
Abstract
Pancreatic cancer is a highly aggressive gastrointestinal malignancy, often termed the "king of cancers" due to its notoriously high mortality rate. Its clinical characteristics, including late diagnosis, low surgical resectability, high recurrence rates, significant chemoresistance, and poor prognosis have collectively driven the persistent rise in incidence and mortality. Despite ongoing advancements in therapeutic strategies, the management of pancreatic cancer, particularly at advanced stages, remains challenging. Chemotherapy remains the mainstay of current treatment. However, the prevalent problem of chemotherapy resistance poses a significant obstacle to effective treatment. Metabolic reprogramming, characterized by alterations in glucose metabolism, lipid biosynthesis, and amino acid utilization, supports the high energy demands and rapid proliferation of cancer cells. Emerging evidence suggests that these metabolic changes, possibly mediated by epigenetic mechanisms, also contribute to tumorigenesis and metastasis. These findings highlight the critical role of metabolic alterations in pancreatic cancer pathogenesis. This review explores the relationship between metabolic reprogramming and chemotherapy resistance, discussing underlying mechanisms and summarizing preclinical studies and drug development targeting metabolism. The aim is to provide a comprehensive perspective on potential therapeutic strategies for pancreatic cancer.
Collapse
Affiliation(s)
- Jinyi Zhang
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Xueqing Kong
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Boyan Zhou
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Rui Li
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhaoan Yu
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Jinrong Zhu
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Qing Xi
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Yan Li
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Zichao Zhao
- Department of Emergency Medicine, Shaodong People's Hospital, Shaodong City, Hunan Province, China.
| | - Rongxin Zhang
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
3
|
Tsyben A, Dannhorn A, Hamm G, Pitoulias M, Couturier DL, Sawle A, Briggs M, Wright AJ, Brodie C, Mendil L, Miller JL, Williams EC, Franzén L, De Jong G, Gracia T, Memi F, Bayraktar OA, Adapa R, Rao J, González-Fernández A, Bunch J, Takats Z, Barry ST, Goodwin RJA, Mair R, Brindle KM. Cell-intrinsic metabolic phenotypes identified in patients with glioblastoma, using mass spectrometry imaging of 13C-labelled glucose metabolism. Nat Metab 2025:10.1038/s42255-025-01293-y. [PMID: 40389678 DOI: 10.1038/s42255-025-01293-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 03/27/2025] [Indexed: 05/21/2025]
Abstract
Transcriptomic studies have attempted to classify glioblastoma (GB) into subtypes that predict survival and have different therapeutic vulnerabilities1-3. Here we identified three metabolic subtypes: glycolytic, oxidative and a mix of glycolytic and oxidative, using mass spectrometry imaging of rapidly excised tumour sections from two patients with GB who were infused with [U-13C]glucose and from spatial transcriptomic analysis of contiguous sections. The phenotypes are not correlated with microenvironmental features, including proliferation rate, immune cell infiltration and vascularization, are retained when patient-derived cells are grown in vitro or as orthotopically implanted xenografts and are robust to changes in oxygen concentration, demonstrating their cell-intrinsic nature. The spatial extent of the regions occupied by cells displaying these distinct metabolic phenotypes is large enough to be detected using clinically applicable metabolic imaging techniques. A limitation of the study is that it is based on only two patient tumours, albeit on multiple sections, and therefore represents a proof-of-concept study.
Collapse
Affiliation(s)
- Anastasia Tsyben
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Andreas Dannhorn
- Integrated BioAnalysis, Clinical Pharmacology & Safety Sciences R&D, AstraZeneca, Cambridge, UK
| | - Gregory Hamm
- Integrated BioAnalysis, Clinical Pharmacology & Safety Sciences R&D, AstraZeneca, Cambridge, UK
| | - Manthos Pitoulias
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | | | - Ashley Sawle
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Mayen Briggs
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Alan J Wright
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Cara Brodie
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Lee Mendil
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Jodi L Miller
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Eleanor C Williams
- AstraZeneca, Cambridge Biomedical Campus, Cambridge, UK
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Lovisa Franzén
- Safety Sciences, Clinical Pharmacology & Safety Sciences R&D, AstraZeneca, Gothenburg, Sweden
- Department of Gene Technology, KTH Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden
| | - Grand De Jong
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Tannia Gracia
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Fani Memi
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Omer Ali Bayraktar
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Ram Adapa
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Jyotsna Rao
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | | | - Josephine Bunch
- National Physical Laboratory, Teddington, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College, London, UK
| | - Zoltan Takats
- Department of Metabolism, Digestion and Reproduction, Imperial College, London, UK
| | - Simon T Barry
- AstraZeneca, Cambridge Biomedical Campus, Cambridge, UK
| | - Richard J A Goodwin
- Integrated BioAnalysis, Clinical Pharmacology & Safety Sciences R&D, AstraZeneca, Cambridge, UK
| | - Richard Mair
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
- Department of Clinical Neurosciences, Cambridge Biomedical Campus, Cambridge, UK.
| | - Kevin M Brindle
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
- Department of Biochemistry, University of Cambridge, Cambridge, UK.
| |
Collapse
|
4
|
Wang P, Wang J, Fang Z, Chen Q, Zhang Y, Qiu X, Bao Z. Novel metabolic subtypes in IDH-mutant gliomas: implications for prognosis and therapy. BMC Cancer 2025; 25:815. [PMID: 40307749 PMCID: PMC12044917 DOI: 10.1186/s12885-025-14176-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Although IDH-mutant glioma generally has a better prognosis than their IDH-wildtype counterparts, considerable prognostic heterogeneity persists among patients with the same IDH mutation. Current study has primarily focused on the different IDH statuses or grades, while the metabolic heterogeneity within IDH-mutant gliomas remains insufficiently characterized. This study aims to identify transcriptomic metabolic subtypes and associated immune microenvironment differences to better understand survival variability and potential therapeutic targets in IDH-mutant glioma. METHODS Patients with IDH-mutant gliomas were included from four public datasets (TCGA, n = 373; CGGA325, n = 167; CGGA693, n = 333; GLASS, n = 100), supplemented by 22 cases from Beijing Tiantan Hospital as an independent cohort. Consensus clustering was used to define novel metabolic subtypes. Clinical features were assessed using chi-square tests and Kaplan-Meier analysis. Metabolic profiles were characterized through enrichment analysis and GSVA; immune infiltration was analyzed using CIBERSORTx and ESTIMATE. Tumor samples from the independent cohort underwent untargeted metabolomics for validation. LASSO regression was applied to select metabolic signatures, and the CGP2014 drug library was used for drug screening. RESULTS Three metabolic subtypes (C1-C3) with distinct prognoses (p < 0.05) were identified. C1 exhibited enhanced carbohydrate and nucleotide metabolism; C2 displayed upregulated amino acid and lipid metabolism; and C3 demonstrated elevated lipid, nucleotide, and vitamin metabolism. These patterns were validated in the independent cohort. Subtypes were also correlated with immune infiltration. A 13-gene metabolic signature was established to stratify prognostic risk and suggest subtype-specific drug sensitivities. CONCLUSIONS Our study provided a novel metabolic subtype for IDH-mutant glioma and highlighted these patients' metabolic heterogeneity and potential therapeutic strategies.
Collapse
Affiliation(s)
- Peng Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Radiation Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jiayi Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zheng Fang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Qiaodong Chen
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Ying Zhang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| | - Xiaoguang Qiu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
- Department of Radiation Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Zhaoshi Bao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| |
Collapse
|
5
|
Fan Z, Xiao Y, Du Y, Zhang Y, Zhou W. Pancreatic cancer subtyping - the keystone of precision treatment. Front Immunol 2025; 16:1563725. [PMID: 40264765 PMCID: PMC12011869 DOI: 10.3389/fimmu.2025.1563725] [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: 01/20/2025] [Accepted: 03/17/2025] [Indexed: 04/24/2025] Open
Abstract
In recent years, the incidence and mortality rates of pancreatic cancer have been rising, posing a severe threat to human health. Tumor heterogeneity remains a critical barrier to advancing diagnosis and treatment efforts. The lack of specific early symptoms, limited early diagnostic methods, high biological complexity, and restricted therapeutic options contribute to the poor outcomes and prognosis of pancreatic cancer. Therefore, there is an urgent need to explore the different subtypes in-depth and develop personalized therapeutic strategies tailored to each subtype. Increasing evidence highlights the pivotal role of molecular subtyping in treating pancreatic cancer. This review focuses on recent advancements in classifying molecular subtypes and therapeutic approaches, discussed from the perspectives of gene mutations, genomics, transcriptomics, proteomics, metabolomics, and immunomics.
Collapse
Affiliation(s)
- Zeyang Fan
- The Second Clinical Medical School, Lanzhou University,
Lanzhou, China
| | - Yao Xiao
- The Second Clinical Medical School, Lanzhou University,
Lanzhou, China
| | - Yan Du
- The Second Clinical Medical School, Lanzhou University,
Lanzhou, China
| | - Yan Zhang
- The Second Clinical Medical School, Lanzhou University,
Lanzhou, China
| | - Wence Zhou
- Department of General Surgery , The Second Hospital of Lanzhou University & The Second Clinical Medical School, Lanzhou University, Lanzhou, China
| |
Collapse
|
6
|
Shangguan F, Ma N, Chen Y, Zheng Y, Ma T, An J, Lin J, Yang H. Fucoxanthin suppresses pancreatic cancer progression by inducing bioenergetics metabolism crisis and promoting SLC31A1‑mediated sensitivity to DDP. Int J Oncol 2025; 66:31. [PMID: 40052552 PMCID: PMC11900939 DOI: 10.3892/ijo.2025.5737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 01/16/2025] [Indexed: 03/14/2025] Open
Abstract
Pancreatic cancer (PC) is one of the most malignant tumors, with a 5‑year survival rate <10%. Chemosynthetic drugs are widely used in the treatment of PC; however, their toxicity and side effects often reduce the quality of life for patients. MTT and colony formation assay were performed to detect cell growth and viability in PC cells. Levels of ROS in whole cell and mitochondria were analyzed through flow cytometry. ATP production was evaluated using an ATP Assay Kit. Cellular bioenergetics were analyzed with a Seahorse XFe96 Analyzer, and changes in target molecules were monitored by western blotting. The present study reports that fucoxanthin (FX), a carotenoid derived from aquatic brown seaweed, significantly inhibits PC by inhibiting cell proliferation and inducing cell death via the non‑classical pathway. FX switches mitochondrial respiration to aerobic glycolysis in PC cells. Furthermore, FX decreases whole‑cell ATP levels, which indicates that promotion of glycolysis does not compensate for FX‑induced ATP depletion in mitochondria. Moreover, FX decreased the reduced glutathione/oxidized glutathione ratio observed under glucose‑limited conditions. These alterations caused by FX may decrease metabolic flexibility, indicating higher sensitivity to glucose‑limited (GL) conditions. FX increased the cytotoxicity of cisplatin (DDP) and the expression of solute carrier family 31 member 1 (SLC31A1) in PC cells. Furthermore, the knockdown of SLC31A1 can attenuate cytotoxicity caused by the combination of FX and DDP. It was inferred that FX increased the sensitivity of PC cells to DDP), potentially by upregulating SLC31A1 expression. In conclusion, FX exhibited potent antitumor effects by reprogramming energy metabolism and inducing a distinct form of regulated cell death. Therefore, combining FX with GL treatment or DDP presents a promising therapeutic strategy for PC.
Collapse
Affiliation(s)
- Fugen Shangguan
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Nengfang Ma
- College of Life and Environmental Science, Wenzhou University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yang Chen
- College of Life and Environmental Science, Wenzhou University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yuansi Zheng
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Ting Ma
- College of Life and Environmental Science, Wenzhou University, Wenzhou, Zhejiang 325000, P.R. China
| | - Jing An
- Division of Infectious Diseases and Global Health, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - Jianhu Lin
- Department of Trauma Surgery and Emergency Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Hailong Yang
- College of Life and Environmental Science, Wenzhou University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
7
|
Miao J, Chen B, Zhang L, Lu Z, Wang R, Wang C, Jiang X, Shen Q, Li Y, Shi D, Ouyang Y, Chen X, Deng X, Zhang S, Zou H, Chen S. Metabolic expression profiling analysis reveals pyruvate-mediated EPHB2 upregulation promotes lymphatic metastasis in head and neck squamous cell carcinomas. J Transl Med 2025; 23:316. [PMID: 40075431 PMCID: PMC11899055 DOI: 10.1186/s12967-025-06305-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
Lymphatic metastasis is a well-known factor for initiating distant metastasis of head and neck squamous cell carcinoma (HNSCC), which caused major death in most patients with cancer. Meanwhile, metabolic reprogramming to support metastasis is regarded as a prominent hallmark of cancers. However, how metabolic disorders drive in HNSCC remains unclear. We firstly established a new classification of HNSCC patients based on metabolism gene expression profiles from the TCGA and GEO database, and identified an enriched carbohydrate metabolism subgroup which was significantly associated with lymphatic metastasis and worse clinical outcome. Moreover, we found that highly activated pyruvate metabolism endowed tumors with EPHB2 upregulation and promoted tumor lymphangiogenesis independently of VEGF-C/VEGFR3 signaling pathway. Mechanically, high nuclear acetyl-CoA production from pyruvate metabolism promoted histone acetylation, which in turn transcriptionally upregulated EPHB2 expression and secretion in tumor cells. EPHB2 bound with EFNB1 in lymphatic endothelial cells promoted YAP/TAZ cytoplasmic retention, which alleviated YAP/TAZ-mediated prospero homeobox protein 1 (PROX1) transcriptional repression, and then triggered tumor lymphangiogenesis. Importantly, combined treatment with EFNB1-Fc and VEGFR3 inhibitor synergistic abrogated lymphangiogenesis in vitro and in vivo, suggesting that targeting EPHB2 might be a potential strategy to patients with no or slight response to VEGFR3 inhibitor. These findings uncover the mechanism by which pyruvate metabolism is linked to lymphatic metastasis of tumor and provides a promising therapeutic strategy for the prevention of HNSCC metastasis.
Collapse
Affiliation(s)
- Jingjing Miao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Boyu Chen
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, P. R. China
| | - Lu Zhang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Zhongming Lu
- Department of Otolaryngology Head and Neck Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, P. R. China
| | - Rui Wang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Chunyang Wang
- Guanghua School of Stomatology, Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510060, P. R. China
| | - Xingyu Jiang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Qi Shen
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Zhejiang, 311402, P. R. China
| | - Yue Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Dongni Shi
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Ying Ouyang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xiangfu Chen
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xiaowu Deng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Siyi Zhang
- Department of Otolaryngology Head and Neck Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, P. R. China.
| | - Hequn Zou
- Medical School, The Chinese University of Hong Kong, Shenzhen, 518172, P. R. China.
| | - Shuwei Chen
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| |
Collapse
|
8
|
Seo R, de Guzman ACV, Park S, Lee JY, Kang SJ. Cancer-intrinsic Cxcl5 orchestrates a global metabolic reprogramming for resistance to oxidative cell death in 3D. Cell Death Differ 2025:10.1038/s41418-025-01466-y. [PMID: 40050422 DOI: 10.1038/s41418-025-01466-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 01/10/2025] [Accepted: 02/19/2025] [Indexed: 03/09/2025] Open
Abstract
Pancreatic ductal adenocarcinoma is characterized by a three-dimensional (3D) tumor microenvironment devoid of oxygen and nutrients but enriched in extracellular matrix, which acts as a physical and chemical barrier. In 3D, cancer cells reprogram their metabolic pathways in ways that help them survive hostile conditions. However, little is known about the metabolic phenotypes of cancer cells in 3D and the intrinsic cues that modulate them. We found that Cxcl5 deletion restricted pancreatic tumor growth in a 3D spheroid-in-Matrigel culture system without affecting cancer cell growth in 2D culture. Cxcl5 deletion impaired 3D-specific global metabolic reprogramming, resistance to hypoxia-induced cell death, and upregulation of Hif1α and Myc. Overexpression of Hif1α and Myc, however, effectively restored 3D culture-induced metabolic reconfiguration, growth, redox homeostasis, and mitochondrial function in Cxcl5-/- cells, reducing ferroptosis. We also found that pancreatic cancer patients with higher expression of hypoxia and metabolism-related genes whose expression is well-correlated with CXCL5 generally have poorer prognosis. Together, our findings identify an unanticipated role of Cxcl5 in orchestrating the cancer metabolic reprogramming in 3D culture that is required for energy and biomass maintenance and that restricts oxidative cell death. Thus, our results provide a rationale for targeting CXCL5 as a promising therapeutic strategy.
Collapse
Affiliation(s)
- Ramin Seo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Arvie Camille V de Guzman
- College of Pharmacy, Natural Product Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sunghyouk Park
- College of Pharmacy, Natural Product Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ji Youn Lee
- Biometrology Group, Division of Biomedical Metrology, Korea Research Institute of Standards and Science, Daejeon, 34113, Republic of Korea
| | - Suk-Jo Kang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
9
|
Chaudhary S, Siddiqui JA, Pothuraju R, Bhatia R. Ribosome biogenesis, altered metabolism and ribotoxic stress response in pancreatic ductal adenocarcinoma tumor microenvironment. Cancer Lett 2025; 612:217484. [PMID: 39842499 DOI: 10.1016/j.canlet.2025.217484] [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: 11/06/2024] [Revised: 01/17/2025] [Accepted: 01/18/2025] [Indexed: 01/24/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy with a poor overall survival rate. Cellular stress response pathways promoting cancer cell fitness in harsh tumor microenvironment (TME) play a critical role in cancer growth and survival. The influence of oncogenic Kras, multi-functional heterogeneous cancer-associated fibroblasts (CAFs), and immunosuppressive TME on cancer cells makes the disease more complex and difficult to treat. The desmoplastic reaction by CAFs comprises approximately 90 % of the tumor, with only 10 % of cancer cells making things even more complicated, resulting in therapy resistance. Consistently increasing fibrosis creates a hypoxic environment and elevated interstitial fluid pressure inside the tumor constraining vascular supply. Stress conditions in TME alter translation efficiency and metabolism to fulfill the energy requirements of rapidly growing cancer cells. Extensive research has been conducted on multiple molecular and metabolic regulators in PDAC TME. However, the role of TME in influencing translation programs, a prerequisite for cell cycle progression and functional/growth requirements for cancer cells, remains elusive. This review highlights the recent advancements in understanding altered translational programs in PDAC TME. We emphasize the role of ribosome biogenesis, ribosome-induced stress response, and the concept of specialized ribosomes and their probable role in mutationally rewiring the pancreatic TME.
Collapse
Affiliation(s)
- Sanjib Chaudhary
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, 781039, Guwahati, Assam, India
| | - Jawed Akhtar Siddiqui
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA; Cancer Center Research Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - Ramesh Pothuraju
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thiruvananthapuram, Kerala, India.
| | - Rakesh Bhatia
- Amity School of Biological Sciences, Amity University Punjab, 82A, Mohali, Punjab, 140306, India.
| |
Collapse
|
10
|
Chia ML, Bulat F, Gaunt A, Ros S, Wright AJ, Sawle A, Porcu L, Vias M, Brenton JD, Brindle KM. Metabolic imaging distinguishes ovarian cancer subtypes and detects their early and variable responses to treatment. Oncogene 2025; 44:563-574. [PMID: 39639170 PMCID: PMC11850285 DOI: 10.1038/s41388-024-03231-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024]
Abstract
High grade serous ovarian cancer displays two metabolic subtypes; a high OXPHOS subtype that shows increased expression of genes encoding electron transport chain components, increased oxygen consumption, and increased chemosensitivity, and a low OXPHOS subtype that exhibits glycolytic metabolism and is more drug resistant. We show here in patient-derived organoids and in the xenografts obtained by their subcutaneous implantation that the low OXPHOS subtype shows higher lactate dehydrogenase activity and monocarboxylate transporter 4 expression than the high OXPHOS subtype and increased lactate labeling in 13C magnetic resonance spectroscopy (MRS) measurements of hyperpolarized [1-13C]pyruvate metabolism. There was no difference between the subtypes in PET measurements of 2-deoxy-2-[fluorine-18]fluoro-D-glucose ([18F]FDG) uptake. Both metabolic imaging techniques could detect the early response to Carboplatin treatment in drug-sensitive high OXPHOS xenografts and no response in drug-resistant in low OXPHOS xenografts. 13C magnetic resonance spectroscopic imaging of hyperpolarized [1-13C]pyruvate metabolism has the potential to be used clinically to distinguish low OXPHOS and high OXPHOS tumor deposits in HGSOC patients and to detect their differential responses to treatment.
Collapse
Affiliation(s)
- Ming Li Chia
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Flaviu Bulat
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Adam Gaunt
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Susana Ros
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Alan J Wright
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
- Guy's and St Thomas's NHS Foundation Trust, St Thomas' Hospital, London, UK
| | - Ashley Sawle
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Luca Porcu
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Maria Vias
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - James D Brenton
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Kevin M Brindle
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK.
- Department of Biochemistry, University of Cambridge, Cambridge, UK.
| |
Collapse
|
11
|
Bathe OF. Tumor metabolism as a factor affecting diversity in cancer cachexia. Am J Physiol Cell Physiol 2025; 328:C908-C920. [PMID: 39870605 DOI: 10.1152/ajpcell.00677.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 09/21/2024] [Accepted: 01/20/2025] [Indexed: 01/29/2025]
Abstract
Cancer cachexia is a multifaceted metabolic syndrome characterized by muscle wasting, fat redistribution, and metabolic dysregulation, commonly associated with advanced cancer but sometimes also evident in early-stage disease. More subtle body composition changes have also been reported in association with cancer, including sarcopenia, myosteatosis, and increased fat radiodensity. Emerging evidence reveals that body composition changes including sarcopenia, myosteatosis, and increased fat radiodensity, arise from distinct biological mechanisms and significantly impact survival outcomes. Importantly, these features often occur independently, with their combined presence exacerbating poor prognoses. Tumor plays a pivotal role in driving these host changes, either by acting as a metabolic parasite or by releasing mediators that disrupt normal tissue function. This review explores the diversity of tumor metabolism. It highlights the potential for tumor-specific metabolic phenotypes to influence systemic effects, including fat redistribution and sarcopenia. Addressing this tumor-host metabolic interplay requires personalized approaches that disrupt tumor metabolism while preserving host health. Promising strategies include targeted pharmacological interventions and anticachexia agents like growth differentiation factor 15 (GDF-15) inhibitors. Nutritional modifications such as ketogenic diets and omega-3 fatty acid supplementation also merit further investigation. In addition to preserving muscle, these therapies will need to be evaluated for their capability to improve survival and quality of life. This review underscores the need for further research into tumor-driven metabolic effects on the host and the development of integrative treatment strategies to address the interconnected challenges of cancer progression and cachexia.
Collapse
Affiliation(s)
- Oliver F Bathe
- Department of Surgery and Oncology, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
12
|
Jiang J, Chen Y, Zheng Y, Ding Y, Wang H, Zhou Q, Teng L, Zhang X. Sialic acid metabolism-based classification reveals novel metabolic subtypes with distinct characteristics of tumor microenvironment and clinical outcomes in gastric cancer. Cancer Cell Int 2025; 25:61. [PMID: 39987095 PMCID: PMC11847363 DOI: 10.1186/s12935-025-03695-0] [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: 12/18/2024] [Accepted: 02/13/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND High heterogeneity in gastric cancer (GC) remains a challenge for standard treatments and prognosis prediction. Dysregulation of sialic acid metabolism (SiaM) is recognized as a key metabolic hallmark of tumor immune evasion and metastasis. Herein, we aimed to develop a SiaM-based metabolic classification in GC. METHODS SiaM-related genes were obtained from the MsigDB database. Bulk and single-cell transcriptional data of 956 GC patients were acquired from the GEO, TCGA, and MEDLINE databases. Proteomic profiles of 20 GC samples were derived from our institution. The consensus clustering algorithm was applied to identify SiaM-based clusters. The SiaM-based model was established via LASSO regression and evaluated via Kaplan‒Meier curve and ROC curve analyses. In vitro and in vivo experiments were conducted to explore the function of ST3GAL1 in GC. RESULTS Three SiaM clusters presented distinct patterns of clinicopathological features, transcriptomic alterations, and tumor immune microenvironment landscapes in GC. Compared with clusters A and B, cluster C presented elevated SiaM activity, higher metastatic potential, more abundant immunosuppressive features, and a worse prognosis. Based on the differentially expressed genes between these clusters, a risk model for six genes (ARHGAP6, ST3GAL1, ADAM28, C7, PLCL1, and TTC28) was then constructed. The model exhibited robust performance in predicting peritoneal metastasis and prognosis in four independent cohorts. As a hub gene in the model, ST3GAL1 promoted GC cell migration and invasion in vitro and in vivo. CONCLUSIONS Our study proposed a novel SiaM-based classification that identified three metabolic subtypes with distinct characteristics of tumor microenvironment and clinical outcomes in GC.
Collapse
Affiliation(s)
- Junjie Jiang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Gastroenterology, Affiliated Hangzhou First People'S Hospital, Westlake University School of Medicine, 261 Huansha Road, Hangzhou, 310006, Zhejiang, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou, Zhejiang, China
- Hangzhou Institute of Digestive Disease, Hangzhou, Zhejiang, China
| | - Yiran Chen
- Department of Surgical Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yangyang Zheng
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yongfeng Ding
- Department of Medical Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Haiyong Wang
- Department of Surgical Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Quan Zhou
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Surgical Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lisong Teng
- Department of Surgical Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaofeng Zhang
- Department of Gastroenterology, Affiliated Hangzhou First People'S Hospital, Westlake University School of Medicine, 261 Huansha Road, Hangzhou, 310006, Zhejiang, China.
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China.
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou, Zhejiang, China.
- Hangzhou Institute of Digestive Disease, Hangzhou, Zhejiang, China.
| |
Collapse
|
13
|
Link JM, Eng JR, Pelz C, MacPherson-Hawthorne K, Worth PJ, Sivagnanam S, Keith DJ, Owen S, Langer EM, Grossblatt-Wait A, Salgado-Garza G, Creason AL, Protzek S, Egger J, Holly H, Heskett MB, Chin K, Kirchberger N, Betre K, Bucher E, Kilburn D, Hu Z, Munks MW, English IA, Tsuda M, Goecks J, Demir E, Adey AC, Kardosh A, Lopez CD, Sheppard BC, Guimaraes A, Brinkerhoff B, Morgan TK, Mills GB, Coussens LM, Brody JR, Sears RC. Ongoing replication stress tolerance and clonal T cell responses distinguish liver and lung recurrence and outcomes in pancreatic cancer. NATURE CANCER 2025; 6:123-144. [PMID: 39789181 PMCID: PMC11779630 DOI: 10.1038/s43018-024-00881-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 11/15/2024] [Indexed: 01/12/2025]
Abstract
Patients with metastatic pancreatic ductal adenocarcinoma survive longer if disease spreads to the lung but not the liver. Here we generated overlapping, multi-omic datasets to identify molecular and cellular features that distinguish patients whose disease develops liver metastasis (liver cohort) from those whose disease develops lung metastasis without liver metastases (lung cohort). Lung cohort patients survived longer than liver cohort patients, despite sharing the same tumor subtype. We developed a primary organotropism (pORG) gene set enriched in liver cohort versus lung cohort primary tumors. We identified ongoing replication stress response pathways in high pORG/liver cohort tumors, whereas low pORG/lung cohort tumors had greater densities of lymphocytes and shared T cell clonal responses. Our study demonstrates that liver-avid pancreatic ductal adenocarcinoma is associated with tolerance to ongoing replication stress, limited tumor immunity and less-favorable outcomes, whereas low replication stress, lung-avid/liver-averse tumors are associated with active tumor immunity that may account for favorable outcomes.
Collapse
Affiliation(s)
- Jason M Link
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA.
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA.
| | - Jennifer R Eng
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Carl Pelz
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA
| | | | - Patrick J Worth
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA
- Knight Cancer Institute, Portland, OR, USA
- Department of Surgery, Oregon Health and Science University, Portland, OR, USA
| | - Shamaline Sivagnanam
- Department of Cell, Development and Cancer Biology, Oregon Health and Science University, Portland, OR, USA
| | - Dove J Keith
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA
| | - Sydney Owen
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA
| | - Ellen M Langer
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA
- Knight Cancer Institute, Portland, OR, USA
- Center for Early Detection Advanced Research, Oregon Health and Science University, Portland, OR, USA
| | - Alison Grossblatt-Wait
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA
- Center for Early Detection Advanced Research, Oregon Health and Science University, Portland, OR, USA
| | | | - Allison L Creason
- Knight Cancer Institute, Portland, OR, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA
| | - Sara Protzek
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA
| | - Julian Egger
- Knight Cancer Institute, Portland, OR, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA
| | - Hannah Holly
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | | | - Koei Chin
- Knight Cancer Institute, Portland, OR, USA
- Center for Early Detection Advanced Research, Oregon Health and Science University, Portland, OR, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA
| | - Nell Kirchberger
- Department of Cell, Development and Cancer Biology, Oregon Health and Science University, Portland, OR, USA
| | - Konjit Betre
- Department of Cell, Development and Cancer Biology, Oregon Health and Science University, Portland, OR, USA
| | - Elmar Bucher
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA
| | - David Kilburn
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA
| | - Zhi Hu
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA
| | - Michael W Munks
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA
| | - Isabel A English
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Motoyuki Tsuda
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Jeremy Goecks
- Knight Cancer Institute, Portland, OR, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA
| | - Emek Demir
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA
- Knight Cancer Institute, Portland, OR, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA
| | - Andrew C Adey
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
- Knight Cancer Institute, Portland, OR, USA
| | - Adel Kardosh
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA
- Knight Cancer Institute, Portland, OR, USA
- Department of Hematology and Oncology, Oregon Health and Science University, Portland, OR, USA
| | - Charles D Lopez
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA
- Knight Cancer Institute, Portland, OR, USA
- Department of Hematology and Oncology, Oregon Health and Science University, Portland, OR, USA
| | - Brett C Sheppard
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA
- Knight Cancer Institute, Portland, OR, USA
- Department of Surgery, Oregon Health and Science University, Portland, OR, USA
| | - Alex Guimaraes
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA
- Knight Cancer Institute, Portland, OR, USA
- Department of Radiology, Oregon Health and Science University, Portland, OR, USA
| | - Brian Brinkerhoff
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA
- Department of Pathology and Laboratory Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Terry K Morgan
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA
- Knight Cancer Institute, Portland, OR, USA
- Center for Early Detection Advanced Research, Oregon Health and Science University, Portland, OR, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA
- Department of Pathology and Laboratory Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Gordon B Mills
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA
- Knight Cancer Institute, Portland, OR, USA
- Department of Oncological Sciences, Oregon Health and Science University, Portland, OR, USA
| | - Lisa M Coussens
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA
- Knight Cancer Institute, Portland, OR, USA
- Department of Cell, Development and Cancer Biology, Oregon Health and Science University, Portland, OR, USA
| | - Jonathan R Brody
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA
- Knight Cancer Institute, Portland, OR, USA
- Department of Surgery, Oregon Health and Science University, Portland, OR, USA
- Department of Cell, Development and Cancer Biology, Oregon Health and Science University, Portland, OR, USA
| | - Rosalie C Sears
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA.
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA.
- Knight Cancer Institute, Portland, OR, USA.
| |
Collapse
|
14
|
Salu P, Tuvin D, Reindl KM. AGR2 knockdown induces ER stress and mitochondria fission to facilitate pancreatic cancer cell death. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119854. [PMID: 39353469 DOI: 10.1016/j.bbamcr.2024.119854] [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: 03/13/2024] [Revised: 09/10/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024]
Abstract
Anterior gradient 2 (AGR2) is often overexpressed in many human cancers, including pancreatic ductal adenocarcinoma (PDAC). Elevated AGR2 expression is known to play a critical role in tumor development, progression, and metastasis and positively correlates with poor patient survival. However, the relationship between AGR2 expression and tumor growth is not fully understood. Our study aims to investigate the impact of AGR2 knockdown on the survival of two pancreatic cancer cell lines, HPAF-II and PANC-1, that exhibit high AGR2 expression. This study revealed that the knockdown of AGR2 expression through an inducible shRNA-mediated approach reduced the proliferative ability and colony-forming potential of PDAC cells compared to scramble controls. Significantly, knocking down AGR2 led to the inhibition of multiple protein biosynthesis pathways and induced ER stress through unfolded protein response (UPR) activation. AGR2 knockdown induced ER stress and increased mitochondrial fission, while mitochondrial fusion remained unaffected. Ultimately, apoptotic cell death was heightened in AGR2 knockdown PDAC cells compared to the controls. Overall, these data reveal a new axis involving AGR2-ER stress-associated mitochondrial fission that could be targeted to improve PDAC patient outcomes.
Collapse
Affiliation(s)
- Philip Salu
- North Dakota State University, Department of Biological Sciences, Fargo, ND, United States of America
| | - Daniel Tuvin
- Roger Maris Cancer Center, Sanford Health, Fargo, ND, United States of America
| | - Katie M Reindl
- North Dakota State University, Department of Biological Sciences, Fargo, ND, United States of America.
| |
Collapse
|
15
|
Mantini G, Agostini A, Tufo M, Rossi S, Kulesko M, Carbone C, Salvatore L, Tortora G, Scambia G, Giacò L. Weighted gene co-expression network analysis reveals key stromal prognostic markers in pancreatic cancer. Sci Rep 2024; 14:31749. [PMID: 39738404 PMCID: PMC11685961 DOI: 10.1038/s41598-024-82563-9] [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: 08/30/2024] [Accepted: 12/06/2024] [Indexed: 01/02/2025] Open
Abstract
In recent years, it has been shown that stroma compartment can favor tumor proliferation and aggressiveness. Although extensive research with network analyses such as Weighted Gene Co-expression Network Analysis (WGCNA) has been conducted on pancreatic cancer and its stromal components, WGCNA has not previously been applied to isolate and identify genes associated with the abundance of stroma and survival outcome from bulk RNA data. We investigated the gene expression profile and clinical information of 140 pancreatic ductal adenocarcinoma patients from TCGA. Network analysis was performed using WGCNA and four modules were found to be associated to patients' clinical traits. Specifically, one module of 2459 genes, was associated to stromal sample content. Subsequently, those genes were further analyzed for survival association through log-rank test and Cox regression. HPGDS and ITGA9-AS1 emerged as significant indicators of favorable prognosis while KCMF1 and YARS1 were implicated in poorer prognostic outcomes. Importantly, HPGDS was found to be stromal-specific in the TMA cohort of Human Protein Atlas. Single sample GSEA showed that the stromal module is enriched for stromal signature of Moffitt and Puleo. These findings suggest that we uncovered a stromal specific signature through WGCNA and found putative prognostic markers.
Collapse
Affiliation(s)
- G Mantini
- Bioinformatics Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.
| | - A Agostini
- Bioinformatics Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - M Tufo
- Bioinformatics Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - S Rossi
- Bioinformatics Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - M Kulesko
- Bioinformatics Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - C Carbone
- Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - L Salvatore
- Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Medical Oncology, Catholic University of the Sacred Heart, Rome, Italy
| | - G Tortora
- Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Medical Oncology, Catholic University of the Sacred Heart, Rome, Italy
| | - G Scambia
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Institute of Obstetrics and Gynecology, Catholic University of the Sacred Heart, Rome, Italy
| | - L Giacò
- Bioinformatics Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
16
|
Peng H, Dou H, He S, Xie YA, Zhang Q, Zheng J. The role of GOT1 in cancer metabolism. Front Oncol 2024; 14:1519046. [PMID: 39777342 PMCID: PMC11703747 DOI: 10.3389/fonc.2024.1519046] [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: 10/29/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
GOT1, a cytoplasmic glutamic oxaloacetic transaminase, plays a critical role in various metabolic pathways essential for cellular homeostasis and dysregulated metabolism. Recent studies have highlighted the significant plasticity and roles of GOT1 in metabolic reprogramming through participating in both classical and non-classical glutamine metabolism, glycolytic metabolism, and other metabolic pathways. This review summarizes emerging insights on the metabolic roles of GOT1 in cancer cells and emphasizes the response of cancer cells to altered metabolism when the expression of GOT1 is altered. We review how cancer cells repurpose cell intrinsic metabolism and their flexibility when GOT1 is inhibited and delineate the molecular mechanisms of GOT1's interaction with specific oncogenes and regulators at multiple levels, including transcriptional and epigenetic regulation, which govern cellular growth and metabolism. These insights may provide new directions for cancer metabolism research and novel targets for cancer treatment.
Collapse
Affiliation(s)
- Huan Peng
- Birth Defects Prevention and Control Institute, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Key Laboratory of Reproductive Health and Birth Defect Prevention, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Clinical Research Center for Pediatric Diseases, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Key Laboratory of Birth Defects and Stem Cell Biobank, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Huihong Dou
- Birth Defects Prevention and Control Institute, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Key Laboratory of Reproductive Health and Birth Defect Prevention, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Clinical Research Center for Pediatric Diseases, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Sheng He
- Birth Defects Prevention and Control Institute, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Key Laboratory of Reproductive Health and Birth Defect Prevention, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Clinical Research Center for Pediatric Diseases, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Key Laboratory of Birth Defects Research and Prevention, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yu-an Xie
- Birth Defects Prevention and Control Institute, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Key Laboratory of Reproductive Health and Birth Defect Prevention, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Clinical Research Center for Pediatric Diseases, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Key Laboratory of Birth Defects and Stem Cell Biobank, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Qinle Zhang
- Birth Defects Prevention and Control Institute, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Key Laboratory of Reproductive Health and Birth Defect Prevention, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Clinical Research Center for Pediatric Diseases, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Key Laboratory of Birth Defects and Stem Cell Biobank, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jianqiu Zheng
- Birth Defects Prevention and Control Institute, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Key Laboratory of Reproductive Health and Birth Defect Prevention, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Clinical Research Center for Pediatric Diseases, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
17
|
Vitale F, Zileri Dal Verme L, Paratore M, Negri M, Nista EC, Ainora ME, Esposto G, Mignini I, Borriello R, Galasso L, Alfieri S, Gasbarrini A, Zocco MA, Nicoletti A. The Past, Present, and Future of Biomarkers for the Early Diagnosis of Pancreatic Cancer. Biomedicines 2024; 12:2840. [PMID: 39767746 PMCID: PMC11673965 DOI: 10.3390/biomedicines12122840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/30/2024] [Accepted: 12/11/2024] [Indexed: 01/04/2025] Open
Abstract
Pancreatic cancer is one of the most aggressive cancers with a very poor 5-year survival rate and reduced therapeutic options when diagnosed in an advanced stage. The dismal prognosis of pancreatic cancer has guided significant efforts to discover novel biomarkers in order to anticipate diagnosis, increasing the population of patients who can benefit from curative surgical treatment. CA 19-9 is the reference biomarker that supports the diagnosis and guides the response to treatments. However, it has significant limitations, a low specificity, and is inefficient as a screening tool. Several potential biomarkers have been discovered in the serum, urine, feces, and pancreatic juice of patients. However, most of this evidence needs further validation in larger cohorts. The advent of advanced omics sciences and liquid biopsy techniques has further enhanced this field of research. The aim of this review is to analyze the historical evolution of the research on novel biomarkers for the early diagnosis of pancreatic cancer, focusing on the current evidence for the most promising biomarkers from different body fluids and the novel trends in research, such as omics sciences and liquid biopsy, in order to favor the application of modern personalized medicine.
Collapse
Affiliation(s)
- Federica Vitale
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Lorenzo Zileri Dal Verme
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Mattia Paratore
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Marcantonio Negri
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Enrico Celestino Nista
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Maria Elena Ainora
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Giorgio Esposto
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Irene Mignini
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Raffaele Borriello
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Linda Galasso
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Sergio Alfieri
- Centro Pancreas, Chirurgia Digestiva, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy;
| | - Antonio Gasbarrini
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Maria Assunta Zocco
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Alberto Nicoletti
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| |
Collapse
|
18
|
Hashimoto A, Hashimoto S. Plasticity and Tumor Microenvironment in Pancreatic Cancer: Genetic, Metabolic, and Immune Perspectives. Cancers (Basel) 2024; 16:4094. [PMID: 39682280 DOI: 10.3390/cancers16234094] [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: 11/09/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Cancer has long been believed to be a genetic disease caused by the accumulation of mutations in key genes involved in cellular processes. However, recent advances in sequencing technology have demonstrated that cells with cancer driver mutations are also present in normal tissues in response to aging, environmental damage, and chronic inflammation, suggesting that not only intrinsic factors within cancer cells, but also environmental alterations are important key factors in cancer development and progression. Pancreatic cancer tissue is mostly comprised of stromal cells and immune cells. The desmoplasmic microenvironment characteristic of pancreatic cancer is hypoxic and hypotrophic. Pancreatic cancer cells may adapt to this environment by rewiring their metabolism through epigenomic changes, enhancing intrinsic plasticity, creating an acidic and immunosuppressive tumor microenvironment, and inducing noncancerous cells to become tumor-promoting. In addition, pancreatic cancer has often metastasized to local and distant sites by the time of diagnosis, suggesting that a similar mechanism is operating from the precancerous stage. Here, we review key recent findings on how pancreatic cancers acquire plasticity, undergo metabolic reprogramming, and promote immunosuppressive microenvironment formation during their evolution. Furthermore, we present the following two signaling pathways that we have identified: one based on the small G-protein ARF6 driven by KRAS/TP53 mutations, and the other based on the RNA-binding protein Arid5a mediated by inflammatory cytokines, which promote both metabolic reprogramming and immune evasion in pancreatic cancer. Finally, the striking diversity among pancreatic cancers in the relative importance of mutational burden and the tumor microenvironment, their clinical relevance, and the potential for novel therapeutic strategies will be discussed.
Collapse
Affiliation(s)
- Ari Hashimoto
- Department of Molecular Biology, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Shigeru Hashimoto
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0818, Japan
| |
Collapse
|
19
|
Wu B, Wang Z, Liu J, Li N, Wang X, Bai H, Wang C, Shi J, Zhang S, Song J, Li Y, Nie G. Dual rectification of metabolism abnormality in pancreatic cancer by a programmed nanomedicine. Nat Commun 2024; 15:10526. [PMID: 39627234 PMCID: PMC11615375 DOI: 10.1038/s41467-024-54963-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 11/19/2024] [Indexed: 12/06/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive and lethal malignancy characterized by dysregulated energy and stromal metabolism. It is strongly supported by activated pancreatic stellate cells (PSC) which drive excessive desmoplasia and tumor growth via metabolic crosstalk. Herein, a programmed nanosystem is designed to dual rectify the metabolism abnormalities of the PDAC cells, which overexpress glucose transporter 1(GLUT1) and CD71, and the PSC for oncotherapy. The nanosystem is based on a tumor microenvironment-responsive liposome encapsulating an NF-κB inhibitor (TPCA-1) and a CD71 aptamer-linked Glut1 siRNA. TPCA-1 reverses the activated PSC to quiescence, which hampers metabolic support of the PSC to PDAC cells and bolsters the PDAC cell-targeting delivery of the siRNA. Aerobic glycolysis and the following enhancement of oxidative phosphorylation are restrained by the nano-modulation so as to amplify anti-PDAC efficacy in an orthotopic xenograft mouse model, which implies more personalized PDAC treatment based on different energy metabolic profiles.
Collapse
MESH Headings
- Animals
- Humans
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/genetics
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/genetics
- Cell Line, Tumor
- Mice
- Nanomedicine/methods
- Liposomes/metabolism
- Pancreatic Stellate Cells/metabolism
- Pancreatic Stellate Cells/pathology
- Tumor Microenvironment
- Glucose Transporter Type 1/metabolism
- Glucose Transporter Type 1/genetics
- RNA, Small Interfering/metabolism
- RNA, Small Interfering/genetics
- NF-kappa B/metabolism
- Xenograft Model Antitumor Assays
- Receptors, Transferrin/metabolism
- Receptors, Transferrin/genetics
- Oxidative Phosphorylation
- Glycolysis
- Mice, Nude
- Aptamers, Nucleotide/metabolism
Collapse
Affiliation(s)
- Bowen Wu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, PR China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, PR China
- Henan Institute of Advanced Technology, Henan, PR China
| | - Zhiqin Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, PR China
- College of Pharmaceutical Science, Jilin University, Changchun, PR China
| | - Jingyuan Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, PR China
| | - Naishi Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, PR China
| | - Xudong Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, PR China
| | - HaoChen Bai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, PR China
| | - Chunling Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, PR China
| | - Jian Shi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, PR China
| | - Saiyang Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Jian Song
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Yiye Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, PR China.
- College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, PR China.
| | - Guangjun Nie
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, PR China.
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, PR China.
- Henan Institute of Advanced Technology, Henan, PR China.
- College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, PR China.
| |
Collapse
|
20
|
Carigga Gutierrez NM, Clainche TL, Bulin A, Leo S, Kadri M, Abdelhamid AGA, Pujol‐Solé N, Obaid G, Hograindleur M, Gardette V, Busser B, Motto‐Ros V, Josserand V, Henry M, Sancey L, Hurbin A, Elleaume H, Kandiah E, Guével XL, Coll J, Broekgaarden M. Engineering Radiocatalytic Nanoliposomes with Hydrophobic Gold Nanoclusters for Radiotherapy Enhancement. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404605. [PMID: 39473330 PMCID: PMC11636064 DOI: 10.1002/adma.202404605] [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: 03/29/2024] [Revised: 09/21/2024] [Indexed: 12/13/2024]
Abstract
Chemoradiation therapy is on the forefront of pancreatic cancer care, and there is a continued effort to improve its safety and efficacy. Liposomes are widely used to improve chemotherapy safety, and may accurately deliver high-Z element- radiocatalytic nanomaterials to cancer tissues. In this study, the interaction between X-rays and long-circulating nanoliposome formulations loaded with gold nanoclusters is explored in the context of oxaliplatin chemotherapy for desmoplastic pancreatic cancer. Hydrophobic gold nanoclusters stabilized with dodecanethiol (AuDDT) are efficiently incorporated in nanoliposomal bilayers. AuDDT-nanoliposomes significantly augmented radiation-induced •OH production, which is most effective with monochromatic X-rays at energies that exceed the K-shell electron binding energy of Au (81.7 keV). Cargo release assays reveal that AuDDT-nanoliposomes can permeabilize lipid bilayers in an X-ray dose- and formulation-dependent manner. The radiocatalytic effect of AuDDT-nanoliposomes significantly augments radiotherapy and oxaliplatin-chemoradiotherapy outcomes in 3D pancreatic microtumors. The PEGylated AuDDT-nanoliposomes display high tumor accumulation in an orthotopic mouse model of pancreatic cancer, showing promise for nanoliposomes as carriers for radiocatalytic nanomaterials. Altogether, compelling proof for chemo-radiation dose-enhancement using AuDDT-nanoliposomes is presented. Further improving the nanoliposomal loading of high-Z elements will advance the safety, efficacy, and translatability of such chemoradiation dose-enhancement approaches.
Collapse
Affiliation(s)
| | - Tristan Le Clainche
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
| | - Anne‐Laure Bulin
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
| | - Sofia Leo
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
- Porphychem SASLongvic21600France
| | - Malika Kadri
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
| | - Ahmed Gamal Ali Abdelhamid
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
| | - Núria Pujol‐Solé
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
| | - Girgis Obaid
- Department of BioengineeringUniversity of Texas at DallasRichardsonTX75080USA
| | - Marc‐André Hograindleur
- European Synchrotron Radiation FacilityCM01 Beamline71 Avenue des MartyrsGrenoble38000France
| | - Vincent Gardette
- Université Lyon 1Institut Lumière Matière, CNRS UMR 5306Université de LyonVilleurbanneFrance
- University Hospital of Grenoble AlpesGrenoble38000France
| | - Benoit Busser
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
- University Hospital of Grenoble AlpesGrenoble38000France
| | - Vincent Motto‐Ros
- Université Lyon 1Institut Lumière Matière, CNRS UMR 5306Université de LyonVilleurbanneFrance
- University Hospital of Grenoble AlpesGrenoble38000France
| | - Véronique Josserand
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
| | - Maxime Henry
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
| | - Lucie Sancey
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
| | - Amandine Hurbin
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
| | - Hélène Elleaume
- Université de Grenoble‐AlpesSynchrotron Radiation for Biomedicine, Inserm UA072280 Rue de la PiscineSaint Martin d'Hères38400France
| | - Eaazhisai Kandiah
- Department of BioengineeringUniversity of Texas at DallasRichardsonTX75080USA
| | - Xavier Le Guével
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
| | - Jean‐Luc Coll
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
| | - Mans Broekgaarden
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
| |
Collapse
|
21
|
Reyes-Oliveras A, Ellis AE, Sheldon RD, Haab B. Metabolomics and 13C labelled glucose tracing to identify carbon incorporation into aberrant cell membrane glycans in cancer. Commun Biol 2024; 7:1576. [PMID: 39592729 PMCID: PMC11599571 DOI: 10.1038/s42003-024-07277-0] [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: 05/10/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Cell membrane glycans contribute to immune recognition, signaling, and cellular adhesion and migration, and altered membrane glycosylation is a feature of cancer cells that contributes to cancer progression. The uptake and metabolism of glucose and other nutrients essential for glycan synthesis could underlie altered membrane glycosylation, but the relationship between shifts in nutrient metabolism and the effects on glycans have not been directly examined. We developed a method that combines stable isotope tracing with metabolomics to enable direct observations of glucose allocation to nucleotide sugars and cell-membrane glycans. We compared the glucose allocation to membrane glycans of two pancreatic cancer cell lines that are genetically identical but have differing energy requirements. The 8988-S cells had higher glucose allocation to membrane glycans and intracellular pathways relating to glycan synthesis, but the 8988-T cells had higher glucose uptake and commitment of glucose to non-glycosylation pathways. The cell lines differed in the requirements of glucose for energy production, resulting in differences in glucose bioavailability for glycan synthesis. The workflow demonstrated here enables studies on the effects of metabolic shifts on the commitment of nutrients to cell-membrane glycans. The results suggest that cell-membrane glycans are remodeled through shifts in glucose commitment to non-glycosylation pathways.
Collapse
Affiliation(s)
- Alfredo Reyes-Oliveras
- Department of Cell Biology, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI, USA
| | - Abigail E Ellis
- Mass Spectrometry Core, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI, USA
| | - Ryan D Sheldon
- Mass Spectrometry Core, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI, USA
| | - Brian Haab
- Department of Cell Biology, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI, USA.
| |
Collapse
|
22
|
Han X, Burrows M, Kim LC, Xu JP, Vostrejs W, Van Le TN, Poltorack C, Jiang Y, Cukierman E, Stanger BZ, Reiss KA, Shaffer SM, Mesaros C, Keith B, Simon MC. Cancer-associated fibroblasts maintain critical pancreatic cancer cell lipid homeostasis in the tumor microenvironment. Cell Rep 2024; 43:114972. [PMID: 39535921 PMCID: PMC11648993 DOI: 10.1016/j.celrep.2024.114972] [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: 04/04/2024] [Revised: 08/06/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with abundant cancer-associated fibroblasts (CAFs) creating hallmark desmoplasia that limits oxygen and nutrient delivery. This study explores the importance of lipid homeostasis under stress. Exogenous unsaturated lipids, rather than de novo synthesis, sustain PDAC cell viability by relieving endoplasmic reticulum (ER) stress under nutrient scarcity. Furthermore, CAFs are less hypoxic than adjacent malignant cells in vivo, nominating them as a potential source of unsaturated lipids. CAF-conditioned medium promotes PDAC cell survival upon nutrient and oxygen deprivation, an effect reversed by delipidation. Lysophosphatidylcholines (LPCs) are particularly enriched in CAF-conditioned medium and preferentially taken up by PDAC cells, where they are converted to phosphatidylcholine (PC) to sustain membrane integrity. Blocking LPC-to-PC conversion inhibits PDAC cell survival and increases ER stress. These findings show a critical lipid "cross-feeding" mechanism that promotes PDAC cell survival, offering a potential metabolic target for treatment.
Collapse
Affiliation(s)
- Xu Han
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michelle Burrows
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Laura C Kim
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jimmy P Xu
- Centers for Cancer Pharmacology and Excellence in Environmental Toxicology, Department of Pharmacology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Will Vostrejs
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tran Ngoc Van Le
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Carson Poltorack
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yanqing Jiang
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edna Cukierman
- Cancer Signaling & Microenvironment Program, Marvin & Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Temple Health, Philadelphia, PA 19111, USA
| | - Ben Z Stanger
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kim A Reiss
- Division of Hematology-Oncology, Penn Medicine Abramson Cancer Center, Philadelphia, PA 19104, USA
| | - Sydney M Shaffer
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Clementina Mesaros
- Centers for Cancer Pharmacology and Excellence in Environmental Toxicology, Department of Pharmacology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brian Keith
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
23
|
Moreno P, Ohara Y, Craig AJ, Liu H, Yang S, Dorsey TH, Zhang L, Panigrahi G, Cawley H, Azizian A, Gaedcke J, Ghadimi M, Hanna N, Hussain SP. ADRA2A promotes the classical/progenitor subtype and reduces disease aggressiveness of pancreatic cancer. Carcinogenesis 2024; 45:845-856. [PMID: 39136088 PMCID: PMC11584292 DOI: 10.1093/carcin/bgae056] [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: 03/14/2024] [Revised: 07/09/2024] [Accepted: 08/12/2024] [Indexed: 08/21/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) manifests diverse molecular subtypes, including the classical/progenitor and basal-like/squamous subtypes, with the latter known for its aggressiveness. We employed integrative transcriptome and metabolome analyses to identify potential genes contributing to the molecular subtype differentiation and its metabolic features. Our comprehensive analysis revealed that adrenoceptor alpha 2A (ADRA2A) was downregulated in the basal-like/squamous subtype, suggesting its potential role as a candidate suppressor of this subtype. Reduced ADRA2A expression was significantly associated with a high frequency of lymph node metastasis, higher pathological grade, advanced disease stage, and decreased survival among PDAC patients. In vitro experiments demonstrated that ADRA2A transgene expression and ADRA2A agonist inhibited PDAC cell invasion. Additionally, ADRA2A-high condition downregulated the basal-like/squamous gene expression signature, while upregulating the classical/progenitor gene expression signature in our PDAC patient cohort and PDAC cell lines. Metabolome analysis conducted on the PDAC cohort and cell lines revealed that elevated ADRA2A levels were associated with suppressed amino acid and carnitine/acylcarnitine metabolism, which are characteristic metabolic profiles of the classical/progenitor subtype. Collectively, our findings suggest that heightened ADRA2A expression induces transcriptome and metabolome characteristics indicative of classical/progenitor subtype with decreased disease aggressiveness in PDAC patients. These observations introduce ADRA2A as a candidate for diagnostic and therapeutic targeting in PDAC.
Collapse
Affiliation(s)
- Paloma Moreno
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Yuuki Ohara
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Amanda J Craig
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Huaitian Liu
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Shouhui Yang
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Tiffany H Dorsey
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Lin Zhang
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Gatikrushna Panigrahi
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Helen Cawley
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Azadeh Azizian
- Städtisches Klinikum Karlsruhe, Moltkestraße 90, 76133 Karlsruhe, Germany
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Jochen Gaedcke
- Städtisches Klinikum Karlsruhe, Moltkestraße 90, 76133 Karlsruhe, Germany
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Michael Ghadimi
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Nader Hanna
- Division of General and Oncologic Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, United States
- Division of Surgical Oncology, Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - S Perwez Hussain
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| |
Collapse
|
24
|
Eze-Odurukwe A, Rehman A, Ayinla L, Anika NN, Shahid R, Ugwuoru AL, Mansoor M, Kamran M. Metabolite Biomarkers for Early Detection of Pancreatic Ductal Adenocarcinoma: A Systematic Review. Cureus 2024; 16:e74528. [PMID: 39726485 PMCID: PMC11671176 DOI: 10.7759/cureus.74528] [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] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal malignancies, with a poor prognosis. This poor prognosis is largely attributed to a late-stage diagnosis. Recent advancements in metabolomics have emerged as a promising avenue for biomarker discovery in PDAC. This systematic review evaluates the potential of metabolite biomarkers for early detection of PDAC. Four studies meeting the inclusion criteria were analyzed, encompassing experimental, case-control, and prospective cohort designs. Key findings include the identification of distinct metabolic subtypes in PDAC with varying sensitivities to metabolic inhibitors. A biomarker signature comprising nine metabolites plus CA19-9 showed high accuracy in distinguishing PDAC from chronic pancreatitis, outperforming CA19-9 alone. Another study identified a five-metabolite signature demonstrating high diagnostic accuracy for pancreatic cancer, differentiating it from type 2 diabetes mellitus. A two-metabolite model (isoleucine and adrenic acid) showed superior performance in detecting stage-I PDAC compared to CA19-9. These studies consistently demonstrate altered metabolic pathways in PDAC patients compared to healthy controls and those with benign pancreatic conditions. Integrating metabolomic data with other molecular profiling approaches has become a powerful strategy for improving diagnostic accuracy. However, challenges remain, including the influence of confounding factors, the need for large-scale validation studies, and the standardization of metabolomic methods. The potential of artificial intelligence in interpreting complex metabolomic data offers promising avenues for future research. This review highlights the significant potential of metabolite biomarkers in early PDAC detection while emphasizing the need for further validation and refinement of these approaches.
Collapse
Affiliation(s)
| | | | - Lois Ayinla
- General Medicine, All Saints University School of Medicine, Roseau, DMA
| | - Nabila N Anika
- Surgery, Baylor College of Medicine, Houston, USA
- Medicine and Surgery, Holy Family Red Crescent Medical College and Hospital, Dhaka, BGD
| | - Ramsha Shahid
- Physiology, Akhtar Saeed Medical and Dental College, Islamabad, PAK
| | | | - Muzafar Mansoor
- Internal Medicine, Allama Iqbal Medical College, Lahore, PAK
| | | |
Collapse
|
25
|
Xie R, Luo Y, Bao B, Wu X, Guo J, Wang J, Qu X, Che X, Zheng C. The Role of Fatty Acid Metabolism, the Related Potential Biomarkers, and Targeted Therapeutic Strategies in Gastrointestinal Cancers. Drug Dev Res 2024; 85:e70014. [PMID: 39527665 DOI: 10.1002/ddr.70014] [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: 05/17/2024] [Revised: 10/12/2024] [Accepted: 10/13/2024] [Indexed: 11/16/2024]
Abstract
Gastrointestinal cancer has emerged as a significant global health concern due to its high incidence and mortality, limited effectiveness of early detection, suboptimal treatment outcomes, and poor prognosis. Metabolic reprogramming is a prominent feature of cancer, and fatty acid metabolism assumes a pivotal role in bridging glucose metabolism and lipid metabolism. Fatty acids play important roles in cellular structural composition, energy supply, signal transduction, and other lipid-related processes. Changes in the levels of fatty acid metabolite may indicate the malignant transformation of gastrointestinal cells, which have an impact on the prognosis of patients and can be used as a marker to monitor the efficacy of anticancer therapy. Therefore, targeting key enzymes involved in fatty acid metabolism, either as monotherapy or in combination with other agents, is a promising strategy for anticancer treatment. This article reviews the potential mechanisms of fatty acid metabolism disorders in the occurrence and development of gastrointestinal tumors, and summarizes the related potential biomarkers and anticancer strategies.
Collapse
Affiliation(s)
- Ruixi Xie
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ying Luo
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Bowen Bao
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xinshu Wu
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jia Guo
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jin Wang
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiujuan Qu
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaofang Che
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chunlei Zheng
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Oncology, Shanghai Electric Power Hospital, Shanghai, China
| |
Collapse
|
26
|
Wang X, Yang J, Ren B, Yang G, Liu X, Xiao R, Ren J, Zhou F, You L, Zhao Y. Comprehensive multi-omics profiling identifies novel molecular subtypes of pancreatic ductal adenocarcinoma. Genes Dis 2024; 11:101143. [PMID: 39253579 PMCID: PMC11382047 DOI: 10.1016/j.gendis.2023.101143] [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: 05/19/2023] [Revised: 09/04/2023] [Accepted: 09/10/2023] [Indexed: 09/11/2024] Open
Abstract
Pancreatic cancer, a highly fatal malignancy, is predicted to rank as the second leading cause of cancer-related death in the next decade. This highlights the urgent need for new insights into personalized diagnosis and treatment. Although molecular subtypes of pancreatic cancer were well established in genomics and transcriptomics, few known molecular classifications are translated to guide clinical strategies and require a paradigm shift. Notably, chronically developing and continuously improving high-throughput technologies and systems serve as an important driving force to further portray the molecular landscape of pancreatic cancer in terms of epigenomics, proteomics, metabonomics, and metagenomics. Therefore, a more comprehensive understanding of molecular classifications at multiple levels using an integrated multi-omics approach holds great promise to exploit more potential therapeutic options. In this review, we recapitulated the molecular spectrum from different omics levels, discussed various subtypes on multi-omics means to move one step forward towards bench-to-beside translation of pancreatic cancer with clinical impact, and proposed some methodological and scientific challenges in store.
Collapse
Affiliation(s)
- Xing Wang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, China
| | - Jinshou Yang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, China
| | - Bo Ren
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, China
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, China
| | - Xiaohong Liu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, China
| | - Ruiling Xiao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, China
| | - Jie Ren
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, China
| | - Feihan Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, China
| |
Collapse
|
27
|
Ali HA, Karasinska JM, Topham JT, Johal D, Kalloger S, Metcalfe A, Warren CS, Miyagi A, Tao LV, Kevorkova M, Chafe SC, McDonald PC, Dedhar S, Parker SJ, Renouf DJ, Schaeffer DF. Pancreatic cancer tumor organoids exhibit subtype-specific differences in metabolic profiles. Cancer Metab 2024; 12:28. [PMID: 39363341 PMCID: PMC11448267 DOI: 10.1186/s40170-024-00357-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 09/26/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive disease characterized by complex metabolic rewiring that enables growth in changing nutrient availability and oxygen conditions. Transcriptome-based prognostic PDAC tumor subtypes, known as 'basal-like' and 'classical' subtypes are associated with differences in metabolic gene expression including genes involved in glycolysis. Tumor subtype-specific metabolism phenotypes may provide new targets for treatment development in PDAC, but their functional relevance has not been fully elucidated. We aimed to investigate differences in metabolic profiles and transcriptomes in tumor models derived from patients with basal-like and classical tumors. METHODS Patient-derived organoids (PDOs) were established from tumor biopsies collected from patients with metastatic PDAC, including three PDOs from basal-like and five PDOs from classical tumors. Metabolic analyses included assessment of differences in metabolic activity using Seahorse Glycolysis and Mito Stress tests and 13C-glucose metabolites tracing analysis. In order to investigate the influence of mitochondrial pyruvate transport on metabolic differences, PDOs were treated with the mitochondrial pyruvate carrier 1 (MPC1) inhibitor UK-5099. Prognostic relevance of MPC1 was determined using a tumor tissue microarray (TMA) in resectable, and proteomics profiling in metastatic PDAC datasets. Whole genome and transcriptome sequencing, differential gene expression and gene set enrichment analyses were performed in PDOs. RESULTS Metastatic PDAC PDOs showed subtype-specific differences in glycolysis and oxidative phosphorylation (OXPHOS). Basal-like tumor-derived PDOs had a lower baseline extracellular acidification rate, but higher glycolytic reserves and oxygen consumption rate (OCR) than classical tumor-derived PDOs. OCR difference was eliminated following treatment with UK-5099. In the 13C-glucose metabolites tracing experiment, a basal-like tumor PDO showed lower fractions of some M + 2 metabolites but higher sensitivity to UK-5099 mediated reduction in M + 2 metabolites than a classical tumor PDO. Protein level analyses revealed lower MPC1 protein levels in basal-like PDAC cases and association of low MPC1 levels with clinicopathologic parameters of tumor aggressiveness in PDAC. PDO differential gene expression analyses identified additional subtype-specific cellular pathways and potential disease outcome biomarkers. CONCLUSIONS Our findings point to distinct metabolic profiles in PDAC subtypes with basal-like tumor PDOs showing higher OXPHOS and sensitivity to MPC1 inhibition. Subtypes-specific metabolic vulnerabilities may be exploited for selective therapeutic targeting.
Collapse
Affiliation(s)
| | | | | | - Danisha Johal
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | | | | | | | | | - Lan V Tao
- Pancreas Centre BC, Vancouver, BC, Canada
| | | | - Shawn C Chafe
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Paul C McDonald
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Shoukat Dedhar
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Seth J Parker
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Daniel J Renouf
- Pancreas Centre BC, Vancouver, BC, Canada
- Division of Medical Oncology, BC Cancer, Vancouver, BC, Canada
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - David F Schaeffer
- Pancreas Centre BC, Vancouver, BC, Canada.
- Division of Anatomic Pathology, Vancouver General Hospital, Vancouver, BC, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
28
|
Gardner GL, Stuart JA. Tumor microenvironment-like conditions alter pancreatic cancer cell metabolism and behavior. Am J Physiol Cell Physiol 2024; 327:C959-C978. [PMID: 39183564 DOI: 10.1152/ajpcell.00452.2024] [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: 07/03/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
The tumor microenvironment is complex and dynamic, characterized by poor vascularization, limited nutrient availability, hypoxia, and an acidic pH. This environment plays a critical role in driving cancer progression. However, standard cell culture conditions used to study cancer cell biology in vitro fail to replicate the in vivo environment of tumors. Recently, "physiological" cell culture media that closely resemble human plasma have been developed (e.g., Plasmax, HPLM), along with more frequent adoption of physiological oxygen conditions (1%-8% O2). Nonetheless, further refinement of tumor-specific culture conditions may be needed. In this study, we describe the development of a tumor microenvironment medium (TMEM) based on murine pancreatic ductal adenocarcinoma (PDAC) tumor interstitial fluid. Using RNA-sequencing, we show that murine PDAC cells (KPCY) cultured in tumor-like conditions (TMEM, pH 7.0, 1.5% O2) exhibit profound differences in gene expression compared with plasma-like conditions (mouse plasma medium, pH 7.4, 5% O2). Specifically, the expression of genes and pathways associated with cell migration, biosynthesis, angiogenesis, and epithelial-to-mesenchymal transition were altered, suggesting tumor-like conditions promote metastatic phenotypes and metabolic remodeling. Using functional assays to validate RNA-seq data, we confirmed increased motility at 1.5% O2/TMEM, despite reduced cell proliferation. Moreover, a hallmark shift to glycolytic metabolism was identified via measurement of glucose uptake/lactate production and mitochondrial respiration. Taken together, these findings demonstrate that growth in 1.5% O2/TMEM alters several biological responses in ways relevant to cancer biology, and more closely models hallmark cancerous phenotypes in culture. This highlights the importance of establishing tumor microenvironment-like conditions in standard cancer research. NEW & NOTEWORTHY Standard cell culture conditions do not replicate the complex tumor microenvironment experienced by cells in vivo. Although currently available plasma-like media are superior to traditional supraphysiological media, they fail to model tumor-like conditions. Using RNA-seq analysis and functional metabolic and migratory assays, we show that tumor microenvironment medium (TMEM), used with representative tumor hypoxia, better models cancerous phenotypes in culture. This emphasizes the critical importance of accurately modeling the tumor microenvironment in cancer research.
Collapse
Affiliation(s)
| | - Jeffrey Alan Stuart
- Department of Biological Sciences, Brock University, St. Catharines, Ontario, Canada
| |
Collapse
|
29
|
Bareham B, Dibble M, Parsons M. Defining and modeling dynamic spatial heterogeneity within tumor microenvironments. Curr Opin Cell Biol 2024; 90:102422. [PMID: 39216233 PMCID: PMC11846781 DOI: 10.1016/j.ceb.2024.102422] [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: 05/08/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024]
Abstract
Many solid tumors exhibit significant genetic, cellular, and biophysical heterogeneity which dynamically evolves during disease progression and after treatment. This constant flux in cell composition, phenotype, spatial relationships, and tissue properties poses significant challenges in accurately diagnosing and treating patients. Much of the complexity lies in unraveling the molecular changes in different tumor compartments, how they influence one another in space and time and where vulnerabilities exist that might be appropriate to target therapeutically. Recent advances in spatial profiling tools and technologies are enabling new insight into the underlying biology of complex tumors, creating a greater understanding of the intricate relationship between cell types, states, and the microenvironment. Here we reflect on some recent discoveries in this area, where the key knowledge and technology gaps lie, and the advancements in spatial measurements and in vitro models for the study of spatial intratumoral heterogeneity.
Collapse
Affiliation(s)
- Bethany Bareham
- Randall Centre for Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
| | - Matthew Dibble
- Randall Centre for Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
| | - Maddy Parsons
- Randall Centre for Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK.
| |
Collapse
|
30
|
Bai R, Cui J. Regulation of fatty acid synthase on tumor and progress in the development of related therapies. Chin Med J (Engl) 2024; 137:1894-1902. [PMID: 38273440 PMCID: PMC11332710 DOI: 10.1097/cm9.0000000000002880] [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: 05/19/2023] [Indexed: 01/27/2024] Open
Abstract
ABSTRACT Fatty acid synthase (FASN) is an essential molecule in lipid metabolic pathways, which are crucial for cancer-related studies. Recent studies have focused on a comprehensive understanding of the novel and important regulatory effects of FASN on malignant biological behavior and immune-cell infiltration, which are closely related to tumor occurrence and development, immune escape, and immune response. FASN-targeting antitumor treatment strategies are being developed. Therefore, in this review, we focused on the effects of FASN on tumor and immune-cell infiltration and reviewed the progress of related anti-tumor therapy development.
Collapse
Affiliation(s)
| | - Jiuwei Cui
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
31
|
Ohara Y, Liu H, Craig AJ, Yang S, Moreno P, Dorsey TH, Cawley H, Azizian A, Gaedcke J, Ghadimi M, Hanna N, Ambs S, Hussain SP. ELAPOR1 induces the classical/progenitor subtype and contributes to reduced disease aggressiveness through metabolic reprogramming in pancreatic cancer. Int J Cancer 2024; 155:569-581. [PMID: 38630934 DOI: 10.1002/ijc.34960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/21/2023] [Accepted: 01/16/2024] [Indexed: 04/19/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a heterogeneous disease with distinct molecular subtypes described as classical/progenitor and basal-like/squamous PDAC. We hypothesized that integrative transcriptome and metabolome approaches can identify candidate genes whose inactivation contributes to the development of the aggressive basal-like/squamous subtype. Using our integrated approach, we identified endosome-lysosome associated apoptosis and autophagy regulator 1 (ELAPOR1/KIAA1324) as a candidate tumor suppressor in both our NCI-UMD-German cohort and additional validation cohorts. Diminished ELAPOR1 expression was linked to high histological grade, advanced disease stage, the basal-like/squamous subtype, and reduced patient survival in PDAC. In vitro experiments demonstrated that ELAPOR1 transgene expression not only inhibited the migration and invasion of PDAC cells but also induced gene expression characteristics associated with the classical/progenitor subtype. Metabolome analysis of patient tumors and PDAC cells revealed a metabolic program associated with both upregulated ELAPOR1 and the classical/progenitor subtype, encompassing upregulated lipogenesis and downregulated amino acid metabolism. 1-Methylnicotinamide, a known oncometabolite derived from S-adenosylmethionine, was inversely associated with ELAPOR1 expression and promoted migration and invasion of PDAC cells in vitro. Taken together, our data suggest that enhanced ELAPOR1 expression promotes transcriptome and metabolome characteristics that are indicative of the classical/progenitor subtype, whereas its reduction associates with basal-like/squamous tumors with increased disease aggressiveness in PDAC patients. These findings position ELAPOR1 as a promising candidate for diagnostic and therapeutic targeting in PDAC.
Collapse
Affiliation(s)
- Yuuki Ohara
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Huaitian Liu
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Amanda J Craig
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Shouhui Yang
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Paloma Moreno
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Tiffany H Dorsey
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Helen Cawley
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | - Michael Ghadimi
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Nader Hanna
- Division of General & Oncologic Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Division of Surgical Oncology, Department of Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - S Perwez Hussain
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
32
|
Schneider C, Hilbert J, Genevaux F, Höfer S, Krauß L, Schicktanz F, Contreras CT, Jansari S, Papargyriou A, Richter T, Alfayomy AM, Falcomatà C, Schneeweis C, Orben F, Öllinger R, Wegwitz F, Boshnakovska A, Rehling P, Müller D, Ströbel P, Ellenrieder V, Conradi L, Hessmann E, Ghadimi M, Grade M, Wirth M, Steiger K, Rad R, Kuster B, Sippl W, Reichert M, Saur D, Schneider G. A Novel AMPK Inhibitor Sensitizes Pancreatic Cancer Cells to Ferroptosis Induction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307695. [PMID: 38885414 PMCID: PMC11336956 DOI: 10.1002/advs.202307695] [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: 10/13/2023] [Revised: 04/12/2024] [Indexed: 06/20/2024]
Abstract
Cancer cells must develop strategies to adapt to the dynamically changing stresses caused by intrinsic or extrinsic processes, or therapeutic agents. Metabolic adaptability is crucial to mitigate such challenges. Considering metabolism as a central node of adaptability, it is focused on an energy sensor, the AMP-activated protein kinase (AMPK). In a subtype of pancreatic ductal adenocarcinoma (PDAC) elevated AMPK expression and phosphorylation is identified. Using drug repurposing that combined screening experiments and chemoproteomic affinity profiling, it is identified and characterized PF-3758309, initially developed as an inhibitor of PAK4, as an AMPK inhibitor. PF-3758309 shows activity in pre-clinical PDAC models, including primary patient-derived organoids. Genetic loss-of-function experiments showed that AMPK limits the induction of ferroptosis, and consequently, PF-3758309 treatment restores the sensitivity toward ferroptosis inducers. The work established a chemical scaffold for the development of specific AMPK-targeting compounds and deciphered the framework for the development of AMPK inhibitor-based combination therapies tailored for PDAC.
Collapse
Affiliation(s)
- Carolin Schneider
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center Göttingen37075GöttingenGermany
| | - Jorina Hilbert
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center Göttingen37075GöttingenGermany
| | - Franziska Genevaux
- Medical Clinic and Polyclinic IIKlinikum rechts der IsarTechnical University of Munich81675MunichGermany
| | - Stefanie Höfer
- Proteomics and BioanalyticsDepartment of Molecular Life SciencesSchool of Life SciencesTechnical University of Munich85354FreisingGermany
| | - Lukas Krauß
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center Göttingen37075GöttingenGermany
| | - Felix Schicktanz
- Institute of PathologyTechnical University of Munich81675MunichGermany
| | - Constanza Tapia Contreras
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center Göttingen37075GöttingenGermany
| | - Shaishavi Jansari
- Department of Gynecology and ObstetricsUniversity Medical Center GöttingenGöttingenGermany
| | - Aristeidis Papargyriou
- Medical Clinic and Polyclinic IIKlinikum rechts der IsarTechnical University of Munich81675MunichGermany
- Institute of Stem Cell ResearchHelmholtz Zentrum MuenchenD‐85764NeuherbergGermany
- Translational Pancreatic Research Cancer CenterMedical Clinic and Polyclinic IIKlinikum rechts der IsarTechnical University of Munich81675MunichGermany
- Center for Organoid Systems (COS)Technical University of Munich85747GarchingGermany
| | - Thorsten Richter
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center Göttingen37075GöttingenGermany
| | - Abdallah M. Alfayomy
- Department of Medicinal ChemistryInstitute of PharmacyMartin‐Luther University Halle‐Wittenberg06120Halle (Saale)Germany
- Department of Pharmaceutical ChemistryAl‐Azhar UniversityAssiut71524Egypt
| | - Chiara Falcomatà
- Institute for Translational Cancer Research and Experimental Cancer TherapyTechnical University Munich81675MunichGermany
- Precision Immunology InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Christian Schneeweis
- Institute for Translational Cancer Research and Experimental Cancer TherapyTechnical University Munich81675MunichGermany
| | - Felix Orben
- Medical Clinic and Polyclinic IIKlinikum rechts der IsarTechnical University of Munich81675MunichGermany
| | - Ruppert Öllinger
- Institute of Molecular Oncology and Functional GenomicsTUM School of MedicineTechnical University of Munich81675MunichGermany
| | - Florian Wegwitz
- Department of Gynecology and ObstetricsUniversity Medical Center GöttingenGöttingenGermany
| | - Angela Boshnakovska
- Department of Cellular BiochemistryUniversity Medical Center37073GöttingenGermany
| | - Peter Rehling
- Department of Cellular BiochemistryUniversity Medical Center37073GöttingenGermany
- Max Planck Institute for Biophysical Chemistry37077GöttingenGermany
| | - Denise Müller
- Institute of PathologyUniversity Medical Center37075GöttingenGermany
| | - Philipp Ströbel
- Institute of PathologyUniversity Medical Center37075GöttingenGermany
- Clinical Research Unit 5002KFO5002University Medical Center Göttingen37075GöttingenGermany
- CCC‐N (Comprehensive Cancer Center Lower Saxony)37075GöttingenGermany
| | - Volker Ellenrieder
- Clinical Research Unit 5002KFO5002University Medical Center Göttingen37075GöttingenGermany
- CCC‐N (Comprehensive Cancer Center Lower Saxony)37075GöttingenGermany
- Department of GastroenterologyGastrointestinal Oncology and EndocrinologyUniversity Medical Center Göttingen37075GöttingenGermany
| | - Lena Conradi
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center Göttingen37075GöttingenGermany
- Clinical Research Unit 5002KFO5002University Medical Center Göttingen37075GöttingenGermany
- CCC‐N (Comprehensive Cancer Center Lower Saxony)37075GöttingenGermany
| | - Elisabeth Hessmann
- Clinical Research Unit 5002KFO5002University Medical Center Göttingen37075GöttingenGermany
- CCC‐N (Comprehensive Cancer Center Lower Saxony)37075GöttingenGermany
- Department of GastroenterologyGastrointestinal Oncology and EndocrinologyUniversity Medical Center Göttingen37075GöttingenGermany
| | - Michael Ghadimi
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center Göttingen37075GöttingenGermany
- CCC‐N (Comprehensive Cancer Center Lower Saxony)37075GöttingenGermany
| | - Marian Grade
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center Göttingen37075GöttingenGermany
- CCC‐N (Comprehensive Cancer Center Lower Saxony)37075GöttingenGermany
| | - Matthias Wirth
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center Göttingen37075GöttingenGermany
- Department of HematologyOncology and Cancer ImmunologyCampus Benjamin FranklinCharité – Universitätsmedizin BerlinCorporate Member of Freie Universität Berlin and Humboldt‐Universität zu Berlin12203BerlinGermany
| | - Katja Steiger
- Institute of PathologyTechnical University of Munich81675MunichGermany
- German Cancer Consortium (DKTK)partner site Municha partnership between DKFZ and University Hospital Klinikum rechts der Isar81675MünchenGermany
| | - Roland Rad
- Institute of Molecular Oncology and Functional GenomicsTUM School of MedicineTechnical University of Munich81675MunichGermany
- German Cancer Consortium (DKTK)partner site Municha partnership between DKFZ and University Hospital Klinikum rechts der Isar81675MünchenGermany
| | - Bernhard Kuster
- Proteomics and BioanalyticsDepartment of Molecular Life SciencesSchool of Life SciencesTechnical University of Munich85354FreisingGermany
- German Cancer Consortium (DKTK)partner site Municha partnership between DKFZ and University Hospital Klinikum rechts der Isar81675MünchenGermany
| | - Wolfgang Sippl
- Department of Medicinal ChemistryInstitute of PharmacyMartin‐Luther University Halle‐Wittenberg06120Halle (Saale)Germany
| | - Maximilian Reichert
- Medical Clinic and Polyclinic IIKlinikum rechts der IsarTechnical University of Munich81675MunichGermany
- Translational Pancreatic Research Cancer CenterMedical Clinic and Polyclinic IIKlinikum rechts der IsarTechnical University of Munich81675MunichGermany
- Center for Organoid Systems (COS)Technical University of Munich85747GarchingGermany
- German Cancer Consortium (DKTK)partner site Municha partnership between DKFZ and University Hospital Klinikum rechts der Isar81675MünchenGermany
- Center for Protein Assemblies (CPA)Technical University of Munich85747GarchingGermany
| | - Dieter Saur
- Institute for Translational Cancer Research and Experimental Cancer TherapyTechnical University Munich81675MunichGermany
- German Cancer Consortium (DKTK)partner site Municha partnership between DKFZ and University Hospital Klinikum rechts der Isar81675MünchenGermany
| | - Günter Schneider
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center Göttingen37075GöttingenGermany
- Institute for Translational Cancer Research and Experimental Cancer TherapyTechnical University Munich81675MunichGermany
- Clinical Research Unit 5002KFO5002University Medical Center Göttingen37075GöttingenGermany
- CCC‐N (Comprehensive Cancer Center Lower Saxony)37075GöttingenGermany
| |
Collapse
|
33
|
Vallés‐Martí A, de Goeij‐de Haas RR, Henneman AA, Piersma SR, Pham TV, Knol JC, Verheij J, Dijk F, Halfwerk H, Giovannetti E, Jiménez CR, Bijlsma MF. Kinase activities in pancreatic ductal adenocarcinoma with prognostic and therapeutic avenues. Mol Oncol 2024; 18:2020-2041. [PMID: 38650175 PMCID: PMC11306541 DOI: 10.1002/1878-0261.13625] [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: 07/19/2023] [Revised: 12/12/2023] [Accepted: 02/21/2024] [Indexed: 04/25/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease with a limited number of known driver mutations but considerable cancer cell heterogeneity. Phosphoproteomics provides a direct read-out of aberrant signaling and the resultant clinically relevant phenotype. Mass spectrometry (MS)-based proteomics and phosphoproteomics were applied to 42 PDAC tumors. Data encompassed over 19 936 phosphoserine or phosphothreonine (pS/T; in 5412 phosphoproteins) and 1208 phosphotyrosine (pY; in 501 phosphoproteins) sites and a total of 3756 proteins. Proteome data identified three distinct subtypes with tumor intrinsic and stromal features. Subsequently, three phospho-subtypes were apparent: two tumor intrinsic (Phos1/2) and one stromal (Phos3), resembling known PDAC molecular subtypes. Kinase activity was analyzed by the Integrative iNferred Kinase Activity (INKA) scoring. Phospho-subtypes displayed differential phosphorylation signals and kinase activity, such as FGR and GSK3 activation in Phos1, SRC kinase family and EPHA2 in Phos2, and EGFR, INSR, MET, ABL1, HIPK1, JAK, and PRKCD in Phos3. Kinase activity analysis of an external PDAC cohort supported our findings and underscored the importance of PI3K/AKT and ERK pathways, among others. Interestingly, unfavorable patient prognosis correlated with higher RTK, PAK2, STK10, and CDK7 activity and high proliferation, whereas long survival was associated with MYLK and PTK6 activity, which was previously unknown. Subtype-associated activity profiles can guide therapeutic combination approaches in tumor and stroma-enriched tissues, and emphasize the critical role of parallel signaling pathways. In addition, kinase activity profiling identifies potential disease markers with prognostic significance.
Collapse
Affiliation(s)
- Andrea Vallés‐Martí
- Department of Medical Oncology, Amsterdam University Medical CenterVU UniversityAmsterdamThe Netherlands
- OncoProteomics LaboratoryCancer Center AmsterdamThe Netherlands
- Cancer BiologyCancer Center AmsterdamThe Netherlands
- Pharmacology LaboratoryCancer Center AmsterdamThe Netherlands
| | - Richard R. de Goeij‐de Haas
- Department of Medical Oncology, Amsterdam University Medical CenterVU UniversityAmsterdamThe Netherlands
- OncoProteomics LaboratoryCancer Center AmsterdamThe Netherlands
| | - Alex A. Henneman
- Department of Medical Oncology, Amsterdam University Medical CenterVU UniversityAmsterdamThe Netherlands
- OncoProteomics LaboratoryCancer Center AmsterdamThe Netherlands
| | - Sander R. Piersma
- Department of Medical Oncology, Amsterdam University Medical CenterVU UniversityAmsterdamThe Netherlands
- OncoProteomics LaboratoryCancer Center AmsterdamThe Netherlands
| | - Thang V. Pham
- Department of Medical Oncology, Amsterdam University Medical CenterVU UniversityAmsterdamThe Netherlands
- OncoProteomics LaboratoryCancer Center AmsterdamThe Netherlands
| | - Jaco C. Knol
- Department of Medical Oncology, Amsterdam University Medical CenterVU UniversityAmsterdamThe Netherlands
- OncoProteomics LaboratoryCancer Center AmsterdamThe Netherlands
| | - Joanne Verheij
- Department of PathologyAmsterdam University Medical CenterThe Netherlands
| | - Frederike Dijk
- Department of PathologyAmsterdam University Medical CenterThe Netherlands
| | - Hans Halfwerk
- Department of PathologyAmsterdam University Medical CenterThe Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam University Medical CenterVU UniversityAmsterdamThe Netherlands
- Pharmacology LaboratoryCancer Center AmsterdamThe Netherlands
- Cancer Pharmacology Lab, AIRC Start‐Up UnitFondazione Pisana per la ScienzaSan Giuliano TermeItaly
| | - Connie R. Jiménez
- Department of Medical Oncology, Amsterdam University Medical CenterVU UniversityAmsterdamThe Netherlands
- OncoProteomics LaboratoryCancer Center AmsterdamThe Netherlands
| | - Maarten F. Bijlsma
- Cancer BiologyCancer Center AmsterdamThe Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam University Medical CenterUniversity of AmsterdamThe Netherlands
| |
Collapse
|
34
|
Huang Y, Zhu Q, Sun Y, Zhang W, Zou J. Alterations in genes involved in glycolysis and hypoxia affect the prognosis of pancreatic cancer. Heliyon 2024; 10:e34104. [PMID: 39100466 PMCID: PMC11295968 DOI: 10.1016/j.heliyon.2024.e34104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 08/06/2024] Open
Abstract
Purpose To construct a prognostic model for pancreatic cancer based on glycolytic and hypoxic metabolic subtypes. To analyze the biological characteristics of these subtypes and explore potential therapeutic options. Methods We obtained mRNA, simple nucleotide variation (SNP), and clinical data for pancreatic cancer from The Cancer Genome Atlas (TCGA). Patients were classified into four metabolic subtypes. We focused on glycolysis and hypoxia subtypes. Single-sample gene set enrichment analysis (ssGSEA) assessed immune cell infiltration. We evaluated the effects of immunotherapy and chemotherapy on these subtypes. Cox regression and random survival forest algorithms were used to build a prognostic model. Validation was performed using data from the International Cancer Genome Consortium (ICGC) and ArrayExpress database. Results We identified four subtypes. Kaplan-Meier survival analysis showed the glycolytic subtype had the longest survival, while the hypoxic subtype had the shortest. The glycolytic subtype exhibited higher immune cell infiltration. Immunotherapy and chemotherapy appeared more beneficial for the glycolytic subtype. KRAS mutations were more frequent in the hypoxic subtype. Our prognostic model indicated a worse prognosis for high-risk groups, validated by external data. Conclusion The glycolytic metabolic subtype of pancreatic cancer is associated with longer survival and better response to chemotherapy and immunotherapy compared to the hypoxic subtype.
Collapse
Affiliation(s)
- Yujie Huang
- Department of Emergency Medicine, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou, 215006, Jiangsu Province, China
| | - Qilu Zhu
- Institute: Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu Province, China
| | - Yizhang Sun
- Department of Urinary Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu Province, China
| | - Weigang Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou, 215006, Jiangsu Province, China
| | - Jiayue Zou
- Department of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou, 215006, Jiangsu Province, China
| |
Collapse
|
35
|
Nicoletti A, Paratore M, Vitale F, Negri M, Quero G, Esposto G, Mignini I, Alfieri S, Gasbarrini A, Zocco MA, Zileri Dal Verme L. Understanding the Conundrum of Pancreatic Cancer in the Omics Sciences Era. Int J Mol Sci 2024; 25:7623. [PMID: 39062863 PMCID: PMC11276793 DOI: 10.3390/ijms25147623] [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: 05/01/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Pancreatic cancer (PC) is an increasing cause of cancer-related death, with a dismal prognosis caused by its aggressive biology, the lack of clinical symptoms in the early phases of the disease, and the inefficacy of treatments. PC is characterized by a complex tumor microenvironment. The interaction of its cellular components plays a crucial role in tumor development and progression, contributing to the alteration of metabolism and cellular hyperproliferation, as well as to metastatic evolution and abnormal tumor-associated immunity. Furthermore, in response to intrinsic oncogenic alterations and the influence of the tumor microenvironment, cancer cells undergo a complex oncogene-directed metabolic reprogramming that includes changes in glucose utilization, lipid and amino acid metabolism, redox balance, and activation of recycling and scavenging pathways. The advent of omics sciences is revolutionizing the comprehension of the pathogenetic conundrum of pancreatic carcinogenesis. In particular, metabolomics and genomics has led to a more precise classification of PC into subtypes that show different biological behaviors and responses to treatments. The identification of molecular targets through the pharmacogenomic approach may help to personalize treatments. Novel specific biomarkers have been discovered using proteomics and metabolomics analyses. Radiomics allows for an earlier diagnosis through the computational analysis of imaging. However, the complexity, high expertise required, and costs of the omics approach are the main limitations for its use in clinical practice at present. In addition, the studies of extracellular vesicles (EVs), the use of organoids, the understanding of host-microbiota interactions, and more recently the advent of artificial intelligence are helping to make further steps towards precision and personalized medicine. This present review summarizes the main evidence for the application of omics sciences to the study of PC and the identification of future perspectives.
Collapse
Affiliation(s)
- Alberto Nicoletti
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (M.P.); (F.V.); (M.N.); (G.E.); (I.M.); (A.G.); (L.Z.D.V.)
| | - Mattia Paratore
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (M.P.); (F.V.); (M.N.); (G.E.); (I.M.); (A.G.); (L.Z.D.V.)
| | - Federica Vitale
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (M.P.); (F.V.); (M.N.); (G.E.); (I.M.); (A.G.); (L.Z.D.V.)
| | - Marcantonio Negri
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (M.P.); (F.V.); (M.N.); (G.E.); (I.M.); (A.G.); (L.Z.D.V.)
| | - Giuseppe Quero
- Centro Pancreas, Chirurgia Digestiva, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (G.Q.); (S.A.)
| | - Giorgio Esposto
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (M.P.); (F.V.); (M.N.); (G.E.); (I.M.); (A.G.); (L.Z.D.V.)
| | - Irene Mignini
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (M.P.); (F.V.); (M.N.); (G.E.); (I.M.); (A.G.); (L.Z.D.V.)
| | - Sergio Alfieri
- Centro Pancreas, Chirurgia Digestiva, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (G.Q.); (S.A.)
| | - Antonio Gasbarrini
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (M.P.); (F.V.); (M.N.); (G.E.); (I.M.); (A.G.); (L.Z.D.V.)
| | - Maria Assunta Zocco
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (M.P.); (F.V.); (M.N.); (G.E.); (I.M.); (A.G.); (L.Z.D.V.)
| | - Lorenzo Zileri Dal Verme
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (M.P.); (F.V.); (M.N.); (G.E.); (I.M.); (A.G.); (L.Z.D.V.)
| |
Collapse
|
36
|
Ohara Y, Craig AJ, Liu H, Yang S, Moreno P, Dorsey TH, Cawley H, Azizian A, Gaedcke J, Ghadimi M, Hanna N, Ambs S, Hussain SP. LMO3 is a suppressor of the basal-like/squamous subtype and reduces disease aggressiveness of pancreatic cancer through glycerol 3-phosphate metabolism. Carcinogenesis 2024; 45:475-486. [PMID: 38366633 PMCID: PMC11229528 DOI: 10.1093/carcin/bgae011] [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: 11/14/2023] [Revised: 01/17/2024] [Accepted: 02/13/2024] [Indexed: 02/18/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) encompasses diverse molecular subtypes, including the classical/progenitor and basal-like/squamous subtypes, each exhibiting distinct characteristics, with the latter known for its aggressiveness. We employed an integrative approach combining transcriptome and metabolome analyses to pinpoint potential genes contributing to the basal-like/squamous subtype differentiation. Applying this approach to our NCI-UMD-German and a validation cohort, we identified LIM Domain Only 3 (LMO3), a transcription co-factor, as a candidate suppressor of the basal-like/squamous subtype. Reduced LMO3 expression was significantly associated with higher pathological grade, advanced disease stage, induction of the basal-like/squamous subtype and decreased survival among PDAC patients. In vitro experiments demonstrated that LMO3 transgene expression inhibited PDAC cell proliferation and migration/invasion, concurrently downregulating the basal-like/squamous gene signature. Metabolome analysis of patient tumors and PDAC cells revealed a metabolic program linked to elevated LMO3 and the classical/progenitor subtype, characterized by enhanced lipogenesis and suppressed amino acid metabolism. Notably, glycerol 3-phosphate (G3P) levels positively correlated with LMO3 expression and associated with improved patient survival. Furthermore, glycerol-3-phosphate dehydrogenase 1 (GPD1), a crucial enzyme in G3P synthesis, showed upregulation in LMO3-high and classical/progenitor PDAC, suggesting its potential role in mitigating disease aggressiveness. Collectively, our findings suggest that heightened LMO3 expression reduces transcriptome and metabolome characteristics indicative of basal-like/squamous tumors with decreased disease aggressiveness in PDAC patients. The observations describe LMO3 as a candidate for diagnostic and therapeutic targeting in PDAC.
Collapse
Affiliation(s)
- Yuuki Ohara
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Amanda J Craig
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Huaitian Liu
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shouhui Yang
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Paloma Moreno
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tiffany H Dorsey
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Helen Cawley
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Azadeh Azizian
- Städtisches Klinikum Karlsruhe, Moltkestraße 90, 76133 Karlsruhe, Germany
| | - Jochen Gaedcke
- Städtisches Klinikum Karlsruhe, Moltkestraße 90, 76133 Karlsruhe, Germany
| | - Michael Ghadimi
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Nader Hanna
- Division of General and Oncologic Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of Surgical Oncology, Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - S Perwez Hussain
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
37
|
Smeets EMM, Trajkovic-Arsic M, Geijs D, Karakaya S, van Zanten M, Brosens LAA, Feuerecker B, Gotthardt M, Siveke JT, Braren R, Ciompi F, Aarntzen EHJG. Histology-Based Radiomics for [ 18F]FDG PET Identifies Tissue Heterogeneity in Pancreatic Cancer. J Nucl Med 2024; 65:1151-1159. [PMID: 38782455 DOI: 10.2967/jnumed.123.266262] [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: 07/01/2023] [Revised: 04/22/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
Radiomics features can reveal hidden patterns in a tumor but usually lack an underlying biologic rationale. In this work, we aimed to investigate whether there is a correlation between radiomics features extracted from [18F]FDG PET images and histologic expression patterns of a glycolytic marker, monocarboxylate transporter-4 (MCT4), in pancreatic cancer. Methods: A cohort of pancreatic ductal adenocarcinoma patients (n = 29) for whom both tumor cross sections and [18F]FDG PET/CT scans were available was used to develop an [18F]FDG PET radiomics signature. By using immunohistochemistry for MCT4, we computed density maps of MCT4 expression and extracted pathomics features. Cluster analysis identified 2 subgroups with distinct MCT4 expression patterns. From corresponding [18F]FDG PET scans, radiomics features that associate with the predefined MCT4 subgroups were identified. Results: Complex heat map visualization showed that the MCT4-high/heterogeneous subgroup was correlating with a higher MCT4 expression level and local variation. This pattern linked to a specific [18F]FDG PET signature, characterized by a higher SUVmean and SUVmax and second-order radiomics features, correlating with local variation. This MCT4-based [18F]FDG PET signature of 7 radiomics features demonstrated prognostic value in an independent cohort of pancreatic cancer patients (n = 71) and identified patients with worse survival. Conclusion: Our cross-modal pipeline allows the development of PET scan signatures based on immunohistochemical analysis of markers of a particular biologic feature, here demonstrated on pancreatic cancer using intratumoral MCT4 expression levels to select [18F]FDG PET radiomics features. This study demonstrated the potential of radiomics scores to noninvasively capture intratumoral marker heterogeneity and identify a subset of pancreatic ductal adenocarcinoma patients with a poor prognosis.
Collapse
Affiliation(s)
- Esther M M Smeets
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marija Trajkovic-Arsic
- German Cancer Consortium, partner site Essen, a partnership between DKFZ and University Hospital Essen, Essen, Germany
- Bridge Institute of Experimental Tumor Therapy and Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Daan Geijs
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sinan Karakaya
- German Cancer Consortium, partner site Essen, a partnership between DKFZ and University Hospital Essen, Essen, Germany
- Bridge Institute of Experimental Tumor Therapy and Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Monica van Zanten
- Department of Pathology, Canisius Wilhelmina Ziekenhuis, Nijmegen, The Netherlands
| | - Lodewijk A A Brosens
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Benedikt Feuerecker
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
- Department of Radiology, School of Medicine, Technical University of Munich, Munich, Germany
- German Cancer Consortium, partner site Munich, a partnership between DKFZ and Technical University of Munich, Munich, Germany
- Department of Radiology, Ludwig Maximilians University, Munich, Germany; and
| | - Martin Gotthardt
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jens T Siveke
- German Cancer Consortium, partner site Essen, a partnership between DKFZ and University Hospital Essen, Essen, Germany
- Bridge Institute of Experimental Tumor Therapy and Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- National Center for Tumor Diseases West, Campus Essen, Essen, Germany
| | - Rickmer Braren
- Department of Radiology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Francesco Ciompi
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Erik H J G Aarntzen
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands;
| |
Collapse
|
38
|
Pulvirenti A, Barbagallo M, Putignano AR, Pea A, Polidori R, Upstill-Goddard R, Cortese N, Kunderfranco P, Brunelli L, De Simone G, Pastorelli R, Spaggiari P, Nappo G, Jamieson NB, Zerbi A, Chang DK, Capretti G, Marchesi F. Integrating metabolic profiling of pancreatic juice with transcriptomic analysis of pancreatic cancer tissue identifies distinct clinical subgroups. Front Oncol 2024; 14:1405612. [PMID: 38988711 PMCID: PMC11234733 DOI: 10.3389/fonc.2024.1405612] [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: 03/23/2024] [Accepted: 05/31/2024] [Indexed: 07/12/2024] Open
Abstract
Introduction Metabolic reprogramming is a hallmark feature of pancreatic ductal adenocarcinoma (PDAC). A pancreatic juice (PJ) metabolic signature has been reported to be prognostic of oncological outcome for PDAC. Integration of PJ profiling with transcriptomic and spatial characterization of the tumor microenvironment would help in identifying PDACs with peculiar vulnerabilities. Methods We performed a transcriptomic analysis of 26 PDAC samples grouped into 3 metabolic clusters (M_CL) according to their PJ metabolic profile. We analyzed molecular subtypes and transcriptional differences. Validation was performed by multidimensional imaging on tumor slides. Results Pancreatic juice metabolic profiling was associated with PDAC transcriptomic molecular subtypes (p=0.004). Tumors identified as M_CL1 exhibited a non-squamous molecular phenotype and demonstrated longer survival. Enrichment analysis revealed the upregulation of immune genes and pathways in M_CL1 samples compared to M_CL2, the group with worse prognosis, a difference confirmed by immunofluorescence on tissue slides. Enrichment analysis of 39 immune signatures by xCell confirmed decreased immune signatures in M_CL2 compared to M_CL1 and allowed a stratification of patients associated with longer survival. Discussion PJ metabolic fingerprints reflect PDAC molecular subtypes and the immune microenvironment, confirming PJ as a promising source of biomarkers for personalized therapy.
Collapse
Affiliation(s)
- Alessandra Pulvirenti
- Section of Pancreatic Surgery, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy
- Department of Surgical Oncological and Gastroenterological Sciences (DiSCOG), University of Padua, Padua, Italy
| | - Marialuisa Barbagallo
- Department of Immunology and Inflammation, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy
| | - Anna Rita Putignano
- Department of Immunology and Inflammation, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy
| | - Antonio Pea
- Department of General and Pancreatic Surgery-The Pancreas Institute, Verona University Hospital Trust, Verona, Italy
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Rebecca Polidori
- Department of Immunology and Inflammation, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Rosie Upstill-Goddard
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Nina Cortese
- Department of Immunology and Inflammation, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy
| | - Paolo Kunderfranco
- Bioinformatics Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy
| | - Laura Brunelli
- Laboratory of Metabolites and Proteins in Translational Research, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Giulia De Simone
- Laboratory of Metabolites and Proteins in Translational Research, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Roberta Pastorelli
- Laboratory of Metabolites and Proteins in Translational Research, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Paola Spaggiari
- Pathology Department, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy
| | - Gennaro Nappo
- Section of Pancreatic Surgery, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Nigel B. Jamieson
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Alessandro Zerbi
- Section of Pancreatic Surgery, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - David K. Chang
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Giovanni Capretti
- Section of Pancreatic Surgery, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Federica Marchesi
- Department of Immunology and Inflammation, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| |
Collapse
|
39
|
Low JC, Cao J, Hesse F, Wright AJ, Tsyben A, Alshamleh I, Mair R, Brindle KM. Deuterium Metabolic Imaging Differentiates Glioblastoma Metabolic Subtypes and Detects Early Response to Chemoradiotherapy. Cancer Res 2024; 84:1996-2008. [PMID: 38635885 PMCID: PMC11176915 DOI: 10.1158/0008-5472.can-23-2552] [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: 08/27/2023] [Revised: 01/30/2024] [Accepted: 03/28/2024] [Indexed: 04/20/2024]
Abstract
Metabolic subtypes of glioblastoma (GBM) have different prognoses and responses to treatment. Deuterium metabolic imaging with 2H-labeled substrates is a potential approach to stratify patients into metabolic subtypes for targeted treatment. In this study, we used 2H magnetic resonance spectroscopy and magnetic resonance spectroscopic imaging (MRSI) measurements of [6,6'-2H2]glucose metabolism to identify metabolic subtypes and their responses to chemoradiotherapy in patient-derived GBM xenografts in vivo. The metabolism of patient-derived cells was first characterized in vitro by measuring the oxygen consumption rate, a marker of mitochondrial tricarboxylic acid cycle activity, as well as the extracellular acidification rate and 2H-labeled lactate production from [6,6'-2H2]glucose, which are markers of glycolytic activity. Two cell lines representative of a glycolytic subtype and two representative of a mitochondrial subtype were identified. 2H magnetic resonance spectroscopy and MRSI measurements showed similar concentrations of 2H-labeled glucose from [6,6'-2H2]glucose in all four tumor models when implanted orthotopically in mice. The glycolytic subtypes showed higher concentrations of 2H-labeled lactate than the mitochondrial subtypes and normal-appearing brain tissue, whereas the mitochondrial subtypes showed more glutamate/glutamine labeling, a surrogate for tricarboxylic acid cycle activity, than the glycolytic subtypes and normal-appearing brain tissue. The response of the tumors to chemoradiation could be detected within 24 hours of treatment completion, with the mitochondrial subtypes showing a decrease in both 2H-labeled glutamate/glutamine and lactate concentrations and glycolytic tumors showing a decrease in 2H-labeled lactate concentration. This technique has the potential to be used clinically for treatment selection and early detection of treatment response. SIGNIFICANCE Deuterium magnetic resonance spectroscopic imaging of glucose metabolism has the potential to differentiate between glycolytic and mitochondrial metabolic subtypes in glioblastoma and to evaluate early treatment responses, which could guide patient treatment.
Collapse
Affiliation(s)
- Jacob C.M. Low
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Jianbo Cao
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Friederike Hesse
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Alan J. Wright
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Anastasia Tsyben
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Islam Alshamleh
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Richard Mair
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Kevin M. Brindle
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| |
Collapse
|
40
|
Fuentes-Rodriguez A, Mitchell A, Guérin SL, Landreville S. Recent Advances in Molecular and Genetic Research on Uveal Melanoma. Cells 2024; 13:1023. [PMID: 38920653 PMCID: PMC11201764 DOI: 10.3390/cells13121023] [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: 04/10/2024] [Revised: 06/08/2024] [Accepted: 06/09/2024] [Indexed: 06/27/2024] Open
Abstract
Uveal melanoma (UM), a distinct subtype of melanoma, presents unique challenges in its clinical management due to its complex molecular landscape and tendency for liver metastasis. This review highlights recent advancements in understanding the molecular pathogenesis, genetic alterations, and immune microenvironment of UM, with a focus on pivotal genes, such as GNAQ/11, BAP1, and CYSLTR2, and delves into the distinctive genetic and chromosomal classifications of UM, emphasizing the role of mutations and chromosomal rearrangements in disease progression and metastatic risk. Novel diagnostic biomarkers, including circulating tumor cells, DNA and extracellular vesicles, are discussed, offering potential non-invasive approaches for early detection and monitoring. It also explores emerging prognostic markers and their implications for patient stratification and personalized treatment strategies. Therapeutic approaches, including histone deacetylase inhibitors, MAPK pathway inhibitors, and emerging trends and concepts like CAR T-cell therapy, are evaluated for their efficacy in UM treatment. This review identifies challenges in UM research, such as the limited treatment options for metastatic UM and the need for improved prognostic tools, and suggests future directions, including the discovery of novel therapeutic targets, immunotherapeutic strategies, and advanced drug delivery systems. The review concludes by emphasizing the importance of continued research and innovation in addressing the unique challenges of UM to improve patient outcomes and develop more effective treatment strategies.
Collapse
Affiliation(s)
- Aurélie Fuentes-Rodriguez
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada; (A.F.-R.); (A.M.); (S.L.G.)
- Hôpital du Saint-Sacrement, Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l‘Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
- Université Laval Cancer Research Center, Quebec City, QC G1R 3S3, Canada
| | - Andrew Mitchell
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada; (A.F.-R.); (A.M.); (S.L.G.)
- Hôpital du Saint-Sacrement, Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l‘Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
- Université Laval Cancer Research Center, Quebec City, QC G1R 3S3, Canada
| | - Sylvain L. Guérin
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada; (A.F.-R.); (A.M.); (S.L.G.)
- Hôpital du Saint-Sacrement, Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l‘Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
| | - Solange Landreville
- Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada; (A.F.-R.); (A.M.); (S.L.G.)
- Hôpital du Saint-Sacrement, Regenerative Medicine Division, CHU de Québec-Université Laval Research Centre, Quebec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l‘Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
- Université Laval Cancer Research Center, Quebec City, QC G1R 3S3, Canada
| |
Collapse
|
41
|
Caggiano EG, Taniguchi CM. UCP2 and pancreatic cancer: conscious uncoupling for therapeutic effect. Cancer Metastasis Rev 2024; 43:777-794. [PMID: 38194152 PMCID: PMC11156755 DOI: 10.1007/s10555-023-10157-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/13/2023] [Indexed: 01/10/2024]
Abstract
Pancreatic cancer has an exaggerated dependence on mitochondrial metabolism, but methods to specifically target the mitochondria without off target effects in normal tissues that rely on these organelles is a significant challenge. The mitochondrial uncoupling protein 2 (UCP2) has potential as a cancer-specific drug target, and thus, we will review the known biology of UCP2 and discuss its potential role in the pathobiology and future therapy of pancreatic cancer.
Collapse
Affiliation(s)
- Emily G Caggiano
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Cullen M Taniguchi
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
42
|
De Santis MC, Bockorny B, Hirsch E, Cappello P, Martini M. Exploiting pancreatic cancer metabolism: challenges and opportunities. Trends Mol Med 2024; 30:592-604. [PMID: 38604929 DOI: 10.1016/j.molmed.2024.03.008] [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: 01/15/2024] [Revised: 03/12/2024] [Accepted: 03/15/2024] [Indexed: 04/13/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive form of pancreatic cancer, known for its challenging diagnosis and limited treatment options. The focus on metabolic reprogramming as a key factor in tumor initiation, progression, and therapy resistance has gained prominence. In this review we focus on the impact of metabolic changes on the interplay among stromal, immune, and tumor cells, as glutamine and branched-chain amino acids (BCAAs) emerge as pivotal players in modulating immune cell functions and tumor growth. We also discuss ongoing clinical trials that explore metabolic modulation for PDAC, targeting mitochondrial metabolism, asparagine and glutamine addiction, and autophagy inhibition. Overcoming challenges in understanding nutrient effects on immune-stromal-tumor interactions holds promise for innovative therapeutic strategies.
Collapse
Affiliation(s)
- Maria Chiara De Santis
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy.
| | - Bruno Bockorny
- BIDMC Department of Medicine, Harvard Medical School, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy
| | - Paola Cappello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy
| | - Miriam Martini
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy.
| |
Collapse
|
43
|
Ji Y, Xu Q, Wang W. Single-cell transcriptome reveals the heterogeneity of malignant ductal cells and the prognostic value of REG4 and SPINK1 in primary pancreatic ductal adenocarcinoma. PeerJ 2024; 12:e17350. [PMID: 38827297 PMCID: PMC11141562 DOI: 10.7717/peerj.17350] [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: 12/12/2023] [Accepted: 04/17/2024] [Indexed: 06/04/2024] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer-related deaths, with very limited therapeutic options available. This study aims to comprehensively depict the heterogeneity and identify prognostic targets for PDAC with single-cell RNA sequencing (scRNA-seq) analysis. Methods ScRNA-seq analysis was performed on 16 primary PDAC and three adjacent lesions. A series of analytical methods were applied for analysis in cell clustering, gene profiling, lineage trajectory analysis and cell-to-cell interactions. In vitro experiments including colony formation, wound healing and sphere formation assay were performed to assess the role of makers. Results A total of 32,480 cells were clustered into six major populations, among which the ductal cell cluster expressing high copy number variants (CNVs) was defined as malignant cells. Malignant cells were further subtyped into five subgroups which exhibited specific features in immunologic and metabolic activities. Pseudotime trajectory analysis indicated that components of various oncogenic pathways were differentially expressed along tumor progression. Furthermore, intensive substantial crosstalk between ductal cells and stromal cells was identified. Finally, genes (REG4 and SPINK1) screened out of differentially expressed genes (DEGs) were upregulated in PDAC cell lines. Silencing either of them significantly impaired proliferation, invasion, migration and stemness of PDAC cells. Conclusions Our findings offer a valuable resource for deciphering the heterogeneity of malignant ductal cells in PDAC. REG4 and SPINK1 are expected to be promising targets for PDAC therapy.
Collapse
MESH Headings
- Female
- Humans
- Male
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Pancreatic Ductal/diagnosis
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/pathology
- Cell Line, Tumor
- Gene Expression Regulation, Neoplastic
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Pancreatic Neoplasms/diagnosis
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/pathology
- Pancreatitis-Associated Proteins
- Prognosis
- Single-Cell Analysis
- Transcriptome
- Trypsin Inhibitor, Kazal Pancreatic/genetics
- Trypsin Inhibitor, Kazal Pancreatic/metabolism
Collapse
Affiliation(s)
- Yutian Ji
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
| | | | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
| |
Collapse
|
44
|
Li F, Si W, Xia L, Yin D, Wei T, Tao M, Cui X, Yang J, Hong T, Wei R. Positive feedback regulation between glycolysis and histone lactylation drives oncogenesis in pancreatic ductal adenocarcinoma. Mol Cancer 2024; 23:90. [PMID: 38711083 PMCID: PMC11071201 DOI: 10.1186/s12943-024-02008-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 04/24/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Metabolic reprogramming and epigenetic alterations contribute to the aggressiveness of pancreatic ductal adenocarcinoma (PDAC). Lactate-dependent histone modification is a new type of histone mark, which links glycolysis metabolite to the epigenetic process of lactylation. However, the role of histone lactylation in PDAC remains unclear. METHODS The level of histone lactylation in PDAC was identified by western blot and immunohistochemistry, and its relationship with the overall survival was evaluated using a Kaplan-Meier survival plot. The participation of histone lactylation in the growth and progression of PDAC was confirmed through inhibition of histone lactylation by glycolysis inhibitors or lactate dehydrogenase A (LDHA) knockdown both in vitro and in vivo. The potential writers and erasers of histone lactylation in PDAC were identified by western blot and functional experiments. The potential target genes of H3K18 lactylation (H3K18la) were screened by CUT&Tag and RNA-seq analyses. The candidate target genes TTK protein kinase (TTK) and BUB1 mitotic checkpoint serine/threonine kinase B (BUB1B) were validated through ChIP-qPCR, RT-qPCR and western blot analyses. Next, the effects of these two genes in PDAC were confirmed by knockdown or overexpression. The interaction between TTK and LDHA was identified by Co-IP assay. RESULTS Histone lactylation, especially H3K18la level was elevated in PDAC, and the high level of H3K18la was associated with poor prognosis. The suppression of glycolytic activity by different kinds of inhibitors or LDHA knockdown contributed to the anti-tumor effects of PDAC in vitro and in vivo. E1A binding protein p300 (P300) and histone deacetylase 2 were the potential writer and eraser of histone lactylation in PDAC cells, respectively. H3K18la was enriched at the promoters and activated the transcription of mitotic checkpoint regulators TTK and BUB1B. Interestingly, TTK and BUB1B could elevate the expression of P300 which in turn increased glycolysis. Moreover, TTK phosphorylated LDHA at tyrosine 239 (Y239) and activated LDHA, and subsequently upregulated lactate and H3K18la levels. CONCLUSIONS The glycolysis-H3K18la-TTK/BUB1B positive feedback loop exacerbates dysfunction in PDAC. These findings delivered a new exploration and significant inter-relationship between lactate metabolic reprogramming and epigenetic regulation, which might pave the way toward novel lactylation treatment strategies in PDAC therapy.
Collapse
Affiliation(s)
- Fei Li
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, 100191, China
| | - Wenzhe Si
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Li Xia
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, 100191, China
| | - Deshan Yin
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, 100191, China
| | - Tianjiao Wei
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, 100191, China
| | - Ming Tao
- Department of General Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Xiaona Cui
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, 100191, China
| | - Jin Yang
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, 100191, China
| | - Tianpei Hong
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, 100191, China.
| | - Rui Wei
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
45
|
Bhandari K, Ding WQ. Protein Arginine Methyltransferases in Pancreatic Ductal Adenocarcinoma: New Molecular Targets for Therapy. Int J Mol Sci 2024; 25:3958. [PMID: 38612768 PMCID: PMC11011826 DOI: 10.3390/ijms25073958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/28/2024] [Accepted: 03/30/2024] [Indexed: 04/14/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignant disease with a low 5-year overall survival rate. It is the third-leading cause of cancer-related deaths in the United States. The lack of robust therapeutics, absence of effective biomarkers for early detection, and aggressive nature of the tumor contribute to the high mortality rate of PDAC. Notably, the outcomes of recent immunotherapy and targeted therapy against PDAC remain unsatisfactory, indicating the need for novel therapeutic strategies. One of the newly described molecular features of PDAC is the altered expression of protein arginine methyltransferases (PRMTs). PRMTs are a group of enzymes known to methylate arginine residues in both histone and non-histone proteins, thereby mediating cellular homeostasis in biological systems. Some of the PRMT enzymes are known to be overexpressed in PDAC that promotes tumor progression and chemo-resistance via regulating gene transcription, cellular metabolic processes, RNA metabolism, and epithelial mesenchymal transition (EMT). Small-molecule inhibitors of PRMTs are currently under clinical trials and can potentially become a new generation of anti-cancer drugs. This review aims to provide an overview of the current understanding of PRMTs in PDAC, focusing on their pathological roles and their potential as new therapeutic targets.
Collapse
Affiliation(s)
| | - Wei-Qun Ding
- Department of Pathology, University of Oklahoma Health Sciences Center, BMSB401A, 940 Stanton L. Young Blvd., Oklahoma City, OK 73104, USA;
| |
Collapse
|
46
|
Moreno P, Ohara Y, Craig AJ, Liu H, Yang S, Zhang L, Panigrahi G, Dorsey TH, Cawley H, Azizian A, Gaedcke J, Ghadimi M, Hanna N, Perwez Hussain S. ADRA2A promotes the classical/progenitor subtype and reduces disease aggressiveness of pancreatic cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.12.584316. [PMID: 38903083 PMCID: PMC11188071 DOI: 10.1101/2024.03.12.584316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) manifests diverse molecular subtypes, including the classical/progenitor and basal-like/squamous subtypes, with the latter known for its aggressiveness. We employed integrative transcriptome and metabolome analyses to identify potential genes contributing to the molecular subtype differentiation and its metabolic features. Transcriptome analysis in PDAC patient cohorts revealed downregulation of adrenoceptor alpha 2A (ADRA2A) in the basal-like/squamous subtype, suggesting its potential role as a candidate suppressor of this subtype. Reduced ADRA2A expression was significantly associated with a high frequency of lymph node metastasis, higher pathological grade, advanced disease stage, and decreased survival among PDAC patients. In vitro experiments demonstrated that ADRA2A transgene expression and ADRA2A agonist inhibited PDAC cell invasion. Additionally, ADRA2A-high condition downregulated the basal-like/squamous gene expression signature, while upregulating the classical/progenitor gene expression signature in our PDAC patient cohort and PDAC cell lines. Metabolome analysis conducted on the PDAC cohort and cell lines revealed that elevated ADRA2A levels were associated with suppressed amino acid and carnitine/acylcarnitine metabolism, which are characteristic metabolic profiles of the classical/progenitor subtype. Collectively, our findings suggest that heightened ADRA2A expression induces transcriptome and metabolome characteristics indicative of classical/progenitor subtype with decreased disease aggressiveness in PDAC patients. These observations introduce ADRA2A as a candidate for diagnostic and therapeutic targeting in PDAC.
Collapse
Affiliation(s)
- Paloma Moreno
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yuuki Ohara
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Amanda J. Craig
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Huaitian Liu
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shouhui Yang
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lin Zhang
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gatikrushna Panigrahi
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tiffany H. Dorsey
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Helen Cawley
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Azadeh Azizian
- Städtisches Klinikum Karlsruhe, Moltkestraße 90, 76133 Karlsruhe, Germany
| | - Jochen Gaedcke
- Städtisches Klinikum Karlsruhe, Moltkestraße 90, 76133 Karlsruhe, Germany
| | - Michael Ghadimi
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Nader Hanna
- Division of General & Oncologic Surgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Division of Surgical Oncology, Department of Surgery, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - S. Perwez Hussain
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
47
|
Anania S, Farnir M, Peiffer R, Boumahd Y, Thiry M, Agirman F, Maloujahmoum N, Bellahcène A, Peulen O. Identification of myoferlin as a mitochondria-associated membranes component required for calcium signaling in PDAC cell lines. Cell Commun Signal 2024; 22:133. [PMID: 38368370 PMCID: PMC10874564 DOI: 10.1186/s12964-024-01514-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/01/2024] [Indexed: 02/19/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma is an aggressive cancer type with one of the lowest survival rates due to late diagnosis and the absence of effective treatments. A better understanding of PDAC biology will help researchers to discover the Achilles' heel of cancer cells. In that regard, our research team investigated the function of an emerging oncoprotein known as myoferlin. Myoferlin is overexpressed in PDAC and its silencing/targeting has been shown to affect cancer cell proliferation, migration, mitochondrial dynamics and metabolism. Nevertheless, our comprehension of myoferlin functions in cells remains limited. In this study, we aimed to understand the molecular mechanism linking myoferlin silencing to mitochondrial dynamics. METHODS Experiments were performed on two pancreas cancer cell lines, Panc-1 and MiaPaCa-2. Myoferlin localization on mitochondria was evaluated by immunofluorescence, proximity ligation assay, and cell fractionation. The presence of myoferlin in mitochondria-associated membranes was assessed by cell fractionation and its function in mitochondrial calcium transfer was evaluated using calcium flow experiments, proximity ligation assays, co-immunoprecipitation, and timelapse fluorescence microscopy in living cells. RESULTS Myoferlin localization on mitochondria was investigated. Our results suggest that myoferlin is unlikely to be located on mitochondria. Instead, we identified myoferlin as a new component of mitochondria-associated membranes. Its silencing significantly reduces the mitochondrial calcium level upon stimulation, probably through myoferlin interaction with the inositol 1,4,5-triphosphate receptors 3. CONCLUSIONS For the first time, myoferlin was specifically demonstrated to be located in mitochondria-associated membranes where it participates to calcium flow. We hypothesized that this function explains our previous results on mitochondrial dynamics. This study improves our comprehension of myoferlin localization and function in cancer biology.
Collapse
Affiliation(s)
- Sandy Anania
- Metastasis Research Laboratory, GIGA-Cancer, Pathology Institute B23, Université de Liège, Liège, B-4000, Belgium
| | - Martin Farnir
- STAR Institute, Université de Liège, Allée du 6 Août 19, Liège, B-4000, Belgium
| | - Raphaël Peiffer
- Metastasis Research Laboratory, GIGA-Cancer, Pathology Institute B23, Université de Liège, Liège, B-4000, Belgium
| | - Yasmine Boumahd
- Metastasis Research Laboratory, GIGA-Cancer, Pathology Institute B23, Université de Liège, Liège, B-4000, Belgium
| | - Marc Thiry
- Cellular and Tissular Biology, GIGA-Neurosciences, Cell Biology L3, Université de Liège, Liège, B-4000, Belgium
| | - Ferman Agirman
- Metastasis Research Laboratory, GIGA-Cancer, Pathology Institute B23, Université de Liège, Liège, B-4000, Belgium
| | - Naima Maloujahmoum
- Metastasis Research Laboratory, GIGA-Cancer, Pathology Institute B23, Université de Liège, Liège, B-4000, Belgium
| | - Akeila Bellahcène
- Metastasis Research Laboratory, GIGA-Cancer, Pathology Institute B23, Université de Liège, Liège, B-4000, Belgium
| | - Olivier Peulen
- Metastasis Research Laboratory, GIGA-Cancer, Pathology Institute B23, Université de Liège, Liège, B-4000, Belgium.
- Center for Interdisciplinary Research on Medicines (CIRM), Mitochondria Adaptation in Cancer Group, Pathology Institute B23, Université de Liège, Liège, B-4000, Belgium.
| |
Collapse
|
48
|
Szczepanski JM, Rudolf MA, Shi J. Clinical Evaluation of the Pancreatic Cancer Microenvironment: Opportunities and Challenges. Cancers (Basel) 2024; 16:794. [PMID: 38398185 PMCID: PMC10887250 DOI: 10.3390/cancers16040794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/10/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Advances in our understanding of pancreatic ductal adenocarcinoma (PDAC) and its tumor microenvironment (TME) have the potential to transform treatment for the hundreds of thousands of patients who are diagnosed each year. Whereas the clinical assessment of cancer cell genetics has grown increasingly sophisticated and personalized, current protocols to evaluate the TME have lagged, despite evidence that the TME can be heterogeneous within and between patients. Here, we outline current protocols for PDAC diagnosis and management, review novel biomarkers, and highlight potential opportunities and challenges when evaluating the PDAC TME as we prepare to translate emerging TME-directed therapies to the clinic.
Collapse
Affiliation(s)
| | | | - Jiaqi Shi
- Department of Pathology and Clinical Labs, University of Michigan, Ann Arbor, MI 48109, USA; (J.M.S.); (M.A.R.)
| |
Collapse
|
49
|
Dash S, Ueda T, Komuro A, Honda M, Sugisawa R, Okada H. Deoxycytidine kinase inactivation enhances gemcitabine resistance and sensitizes mitochondrial metabolism interference in pancreatic cancer. Cell Death Dis 2024; 15:131. [PMID: 38346958 PMCID: PMC10861559 DOI: 10.1038/s41419-024-06531-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/28/2024] [Accepted: 02/01/2024] [Indexed: 02/15/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is considered one of the most lethal forms of cancer. Although in the last decade, an increase in 5-year patient survival has been observed, the mortality rate remains high. As a first-line treatment for PDAC, gemcitabine alone or in combination (gemcitabine plus paclitaxel) has been used; however, drug resistance to this regimen is a growing issue. In our previous study, we reported MYC/glutamine dependency as a therapeutic target in gemcitabine-resistant PDAC secondary to deoxycytidine kinase (DCK) inactivation. Moreover, enrichment of oxidative phosphorylation (OXPHOS)-associated genes was a common property shared by PDAC cell lines, and patient clinical samples coupled with low DCK expression was also demonstrated, which implicates DCK in cancer metabolism. In this article, we reveal that the expression of most genes encoding mitochondrial complexes is remarkably upregulated in PDAC patients with low DCK expression. The DCK-knockout (DCK KO) CFPAC-1 PDAC cell line model reiterated this observation. Particularly, OXPHOS was functionally enhanced in DCK KO cells as shown by a higher oxygen consumption rate and mitochondrial ATP production. Electron microscopic observations revealed abnormal mitochondrial morphology in DCK KO cells. Furthermore, DCK inactivation exhibited reactive oxygen species (ROS) reduction accompanied with ROS-scavenging gene activation, such as SOD1 and SOD2. SOD2 inhibition in DCK KO cells clearly induced cell growth suppression. In combination with increased anti-apoptotic gene BCL2 expression in DCK KO cells, we finally reveal that venetoclax and a mitochondrial complex I inhibitor are therapeutically efficacious for DCK-inactivated CFPAC-1 cells in in vitro and xenograft models. Hence, our work provides insight into inhibition of mitochondrial metabolism as a novel therapeutic approach to overcome DCK inactivation-mediated gemcitabine resistance in PDAC patient treatment.
Collapse
Affiliation(s)
- Suman Dash
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan
- Graduate School of Medical Sciences, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan
| | - Takeshi Ueda
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan
- Graduate School of Medical Sciences, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan
| | - Akiyoshi Komuro
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan
| | - Masahiko Honda
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan
| | - Ryoichi Sugisawa
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan
| | - Hitoshi Okada
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan.
- Graduate School of Medical Sciences, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan.
- Anti-aging Center, Kindai University, Higashi-Osaka, Osaka, 577-8502, Japan.
| |
Collapse
|
50
|
Michiels E, Madhloum H, Van Lint S, Messaoudi N, Kunda R, Martens S, Giron P, Olsen C, Lefesvre P, Dusetti N, El Mohajer L, Tomasini R, Hawinkels LJ, Ahsayni F, Nicolle R, Arsenijevic T, Bouchart C, Van Laethem JL, Rooman I. High-resolution and quantitative spatial analysis reveal intra-ductal phenotypic and functional diversification in pancreatic cancer. J Pathol 2024; 262:76-89. [PMID: 37842959 DOI: 10.1002/path.6212] [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: 04/11/2023] [Revised: 08/23/2023] [Accepted: 09/02/2023] [Indexed: 10/17/2023]
Abstract
A 'classical' and a 'basal-like' subtype of pancreatic cancer have been reported, with differential expression of GATA6 and different dosages of mutant KRAS. We established in situ detection of KRAS point mutations and mRNA panels for the consensus subtypes aiming to project these findings to paraffin-embedded clinical tumour samples for spatial quantitative analysis. We unveiled that, next to inter-patient and intra-patient inter-ductal heterogeneity, intraductal spatial phenotypes exist with anti-correlating expression levels of GATA6 and KRASG12D . The basal-like mRNA panel better captured the basal-like cell states than widely used protein markers. The panels corroborated the co-existence of the classical and basal-like cell states in a single tumour duct with functional diversification, i.e. proliferation and epithelial-to-mesenchymal transition respectively. Mutant KRASG12D detection ascertained an epithelial origin of vimentin-positive cells in the tumour. Uneven spatial distribution of cancer-associated fibroblasts could recreate similar intra-organoid diversification. This extensive heterogeneity with functional cooperation of plastic tumour cells poses extra challenges to therapeutic approaches. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Ellis Michiels
- Laboratory of Medical & Molecular Oncology (LMMO), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Hediel Madhloum
- Laboratory of Medical & Molecular Oncology (LMMO), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Silke Van Lint
- Laboratory of Medical & Molecular Oncology (LMMO), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Nouredin Messaoudi
- Department of Surgery, Department of Gastroenterology-Hepatology, Department of Advanced Interventional Endoscopy, Universitair Ziekenhuis Brussel (UZB), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Rastislav Kunda
- Department of Surgery, Department of Gastroenterology-Hepatology, Department of Advanced Interventional Endoscopy, Universitair Ziekenhuis Brussel (UZB), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Sandrina Martens
- Laboratory of Medical & Molecular Oncology (LMMO), Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Department of Cardio and Organ Systems, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Philippe Giron
- Centre for Medical Genetics, Clinical Sciences, Reproduction and Genetics Research Group, Universitair Ziekenhuis Brussel (UZB), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Catharina Olsen
- Centre for Medical Genetics, Clinical Sciences, Reproduction and Genetics Research Group, Universitair Ziekenhuis Brussel (UZB), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Pierre Lefesvre
- Department of Anatomo-Pathology, Universitair Ziekenhuis Brussel (UZB), Brussels, Belgium
| | - Nelson Dusetti
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Institut Paoli-Calmettes, Aix-Marseille, Marseille University, Marseille, France
| | - Leila El Mohajer
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Institut Paoli-Calmettes, Aix-Marseille, Marseille University, Marseille, France
| | - Richard Tomasini
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Institut Paoli-Calmettes, Aix-Marseille, Marseille University, Marseille, France
| | - Lukas Jac Hawinkels
- Department of Gastroenterology and Hepatology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Farah Ahsayni
- Department of Surgery, Department of Gastroenterology-Hepatology, Department of Advanced Interventional Endoscopy, Universitair Ziekenhuis Brussel (UZB), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Rémy Nicolle
- Centre de Recherche sur l'inflammation (CRI), INSERM, Paris, France
| | - Tatjana Arsenijevic
- Laboratory of Experimental Gastroenterology, Faculty of Medicine, Université Libre de Bruxelles (ULB), Bruxelles, Belgium
- Department of Gastroenterology, Hepatology and Digestive Oncology, HUB Bordet Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Christelle Bouchart
- Department of Radiation-Oncology, Université Libre de Bruxelles (ULB), Hospital Universitaire de Bruxelles (HUB) Institut Jules Bordet, Brussels, Belgium
| | - Jean-Luc Van Laethem
- Laboratory of Experimental Gastroenterology, Faculty of Medicine, Université Libre de Bruxelles (ULB), Bruxelles, Belgium
- Department of Gastroenterology, Hepatology and Digestive Oncology, HUB Bordet Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Ilse Rooman
- Laboratory of Medical & Molecular Oncology (LMMO), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|